US20100124543A1 - Sulphonamide derivatives as prodrugs of aspartyl protease inhibitors - Google Patents

Sulphonamide derivatives as prodrugs of aspartyl protease inhibitors Download PDF

Info

Publication number
US20100124543A1
US20100124543A1 US12/504,243 US50424309A US2010124543A1 US 20100124543 A1 US20100124543 A1 US 20100124543A1 US 50424309 A US50424309 A US 50424309A US 2010124543 A1 US2010124543 A1 US 2010124543A1
Authority
US
United States
Prior art keywords
alkyl
compound
optionally substituted
alkenyl
independently selected
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US12/504,243
Inventor
Roger Dennis Tung
Michael Robin Hale
Christopher Todd Baker
Eric Steven Furfine
Istvan Kaldor
Wieslaw Mieczylaw Kazmierski
Andrew Spaltenstein
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Vertex Pharmaceuticals Inc
Original Assignee
Vertex Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=25544691&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20100124543(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Vertex Pharmaceuticals Inc filed Critical Vertex Pharmaceuticals Inc
Priority to US12/504,243 priority Critical patent/US20100124543A1/en
Assigned to VERTEX PHARMACEUTICALS INCORPORATED reassignment VERTEX PHARMACEUTICALS INCORPORATED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: KAZMIERSKI, WIESLAW MIECZYSLAW, FURFINE, ERIC STEVEN, HALE, MICHAEL ROBIN, KALDOR, ISTVAN, SPALTENSTEIN, ANDREW, TUNG, ROGER DENNIS, BAKER, CHRISTOPHER TODD
Publication of US20100124543A1 publication Critical patent/US20100124543A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • 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
    • 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
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C311/00Amides of sulfonic acids, i.e. compounds having singly-bound oxygen atoms of sulfo groups replaced by nitrogen atoms, not being part of nitro or nitroso groups
    • C07C311/15Sulfonamides having sulfur atoms of sulfonamide groups bound to carbon atoms of six-membered aromatic rings
    • C07C311/16Sulfonamides having sulfur atoms of sulfonamide groups bound to carbon atoms of six-membered aromatic rings having the nitrogen atom of at least one of the sulfonamide groups bound to hydrogen atoms or to an acyclic carbon atom
    • C07C311/18Sulfonamides having sulfur atoms of sulfonamide groups bound to carbon atoms of six-membered aromatic rings having the nitrogen atom of at least one of the sulfonamide groups bound to hydrogen atoms or to an acyclic carbon atom to an acyclic carbon atom of a hydrocarbon radical substituted by nitrogen atoms, not being part of nitro or nitroso groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D307/00Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
    • C07D307/02Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings
    • C07D307/04Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having no double bonds between ring members or between ring members and non-ring members
    • C07D307/18Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having no double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D307/20Oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/655Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having oxygen atoms, with or without sulfur, selenium, or tellurium atoms, as the only ring hetero atoms
    • C07F9/65515Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having oxygen atoms, with or without sulfur, selenium, or tellurium atoms, as the only ring hetero atoms the oxygen atom being part of a five-membered ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6564Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having phosphorus atoms, with or without nitrogen, oxygen, sulfur, selenium or tellurium atoms, as ring hetero atoms
    • C07F9/6581Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having phosphorus atoms, with or without nitrogen, oxygen, sulfur, selenium or tellurium atoms, as ring hetero atoms having phosphorus and nitrogen atoms with or without oxygen or sulfur atoms, as ring hetero atoms
    • C07F9/6584Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having phosphorus atoms, with or without nitrogen, oxygen, sulfur, selenium or tellurium atoms, as ring hetero atoms having phosphorus and nitrogen atoms with or without oxygen or sulfur atoms, as ring hetero atoms having one phosphorus atom as ring hetero atom
    • C07F9/65842Cyclic amide derivatives of acids of phosphorus, in which one nitrogen atom belongs to the ring
    • C07F9/65844Cyclic amide derivatives of acids of phosphorus, in which one nitrogen atom belongs to the ring the phosphorus atom being part of a five-membered ring which may be condensed with another ring system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H15/00Compounds containing hydrocarbon or substituted hydrocarbon radicals directly attached to hetero atoms of saccharide radicals
    • C07H15/02Acyclic radicals, not substituted by cyclic structures
    • C07H15/04Acyclic radicals, not substituted by cyclic structures attached to an oxygen atom of the saccharide radical

Definitions

  • the present invention relates to prodrugs of a class of sulfonamides which are aspartyl protease inhibitors.
  • this invention relates to a novel class of prodrugs of HIV aspartyl protease inhibitors characterized by favorable aqueous solubility, high oral bioavailability and facile in vivo generation of the active ingredient.
  • This invention also relates to pharmaceutical compositions comprising these prodrugs.
  • the prodrugs and pharmaceutical compositions of this invention are particularly well suited for decreasing the pill burden and increasing patient compliance. This invention also relates to methods of treating mammals with these prodrugs and pharmaceutical compositions.
  • Aspartyl protease inhibitors are considered the most effective current drug in the fight against HIV infection. These inhibitors, however, require certain physicochemical properties in order to achieve good potency against the enzyme. One of these properties is high hydrophobicity. Unfortunately, this property results in poor aqueous solubility and low oral bioavailability.
  • U.S. Pat. No. 5,585,397 describes a class of sulfonamide compounds that are inhibitors of the aspartyl protease enzyme. These compounds illustrate the drawbacks concomitant to pharmaceutical compositions comprising hydrophobic aspartyl protease inhibitors.
  • VX-478 (4-amino-N-((2-syn,3S)-2-hydroxy-4-phenyl-2((S)-tetrahydrofuran-3-yl-oxycarbonylamino)-butyl-N-isobutyl-benzenesulfonamide) is an aspartyl protease inhibitor disclosed in the '397 patent. It has a relatively low aqueous solubility.
  • VX-478 produces an upper limit of 150 mg of VX-478 in each capsule. Given a therapeutic dose of 2400 mg/day of VX-478, this formulation would require a patient to consume 16 capsules per day. Such a high pill burden would likely result in poor patient compliance, thus producing sub-optimal therapeutic benefit of the drug. The high pill burden is also a deterrent to increasing the amount of the drug administered per day to a patient. Another drawback of the pill burden and the concomitant patient compliance problem is in the treatment of children infected with HIV.
  • these “solution” formulations such as the mesylate formulation, are at a saturation solubility of VX-478. This creates the real potential of having the drug crystallize out of solution under various storage and/or shipping conditions. This, in turn, would likely result in a loss of some of the oral bioavailability achieved with VX-478.
  • the present invention provides novel prodrugs of a class of sulfonamide compounds that are inhibitors of aspartyl protease, in particular, HIV aspartyl protease. These prodrugs are characterized by excellent aqueous solubility, increased bioavailability and are readily metabolized into the active inhibitors in vivo.
  • the present invention also provides pharmaceutical compositions comprising these prodrugs and methods of treating HIV infection in mammals using these prodrugs and the pharmaceutical compositions thereof.
  • prodrugs can be used alone or in combination with other therapeutic or prophylactic agents, such as anti-virals, antibiotics, immunomodulators or vaccines, for the treatment or prophylaxis of viral infection.
  • This novel class of sulfonamides is represented by formula I:
  • A is selected from H; Ht; —R 1 —Ht; —R 1 —C 1 -C 6 alkyl, which is optionally substituted with one or more groups independently selected from hydroxy, C 1 -C 4 alkoxy, Ht, —O-Ht, —NR 2 —CO—N(R 2 ) 2 or —CO—N(R 2 ) 2 ; —R 1 —C 2 -C 6 alkenyl, which is optionally substituted with one or more groups independently selected from hydroxy, C 1 -C 4 alkoxy, Ht, —O-Ht, —NR 2 —CO—N(R 2 ) 2 or —CO—N(R 2 ) 2 ; or R 7 ;
  • each R 1 is independently selected from —C(O)—, —S(O) 2 —, —C(O)—C(O)—, —O—C(O)—, —O—S(O) 2 , —NR 2 —S(O) 2 —, —NR 2 —C(O)— or —NR 2 —C(O)—C(O)—;
  • each Ht is independently selected from C 3 -C 7 cycloalkyl; C 5 -C 7 cycloalkenyl; C 6 -C 10 aryl; or a 5-7 membered saturated or unsaturated heterocycle, containing one or more heteroatoms selected from N, N(R 2 ), O, S and S(O) n ; wherein said aryl or said heterocycle is optionally fused to Q; and wherein any member of said Ht is optionally substituted with one or more substituents independently selected from oxo, —OR 2 , SR 2 , —R 2 , —N(R 2 )(R 2 ), —R 2 —OH, —CN, —CO 2 R 2 , —C(O)—N(R 2 ) 2 , —S(O) 2 —N(R 2 ) 2 , —N(R 2 )—C(O)—R 2 , —C(O)—R 2 , —S(
  • each R 2 is independently selected from H, or C 1 -C 4 alkyl optionally substituted with Q;
  • B when present, is —N(R 2 )—C(R 3 ) 2 —C(O)—;
  • each x is independently 0 or 1;
  • each R 3 is independently selected from H, Ht, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 3 -C 6 cycloalkyl or C 5 -C 6 cycloalkenyl; wherein any member of said R 3 , except H, is optionally substituted with one or more substituents selected from —OR 2 , —C(O)—NH—R 2 , —S(O) n —N(R 2 )(R 2 ), Ht, —CN, —SR 2 , —CO 2 R 2 , NR 2 —C(O)—R 2 ;
  • each n is independently 1 or 2;
  • G when present, is selected from H, R 7 or C 1 -C 4 alkyl, or, when G is C 1 -C 4 alkyl, G and R′ are bound to one another either directly or through a C 1 -C 3 linker to form a heterocyclic ring; or
  • D and D′ are independently selected from Q; C 1 -C 6 alkyl, which is optionally substituted with one or more groups selected from C 3 -C 6 cycloalkyl, —OR 2 , —R 3 , —O-Q or Q; C 2 -C 4 alkenyl, which is optionally substituted with one or more groups selected from C 3 -C 6 cycloalkyl, —OR 2 , —R 3 , —O-Q or Q; C 3 -C 6 cycloalkyl, which is optionally substituted with or fused to Q; or C 5 -C 6 cycloalkenyl, which is optionally substituted with or fused to Q;
  • each Q is independently selected from a 3-7 membered saturated, partially saturated or unsaturated carbocyclic ring system; or a 5-7 membered saturated, partially saturated or unsaturated heterocyclic ring containing one or more heteroatoms selected from O, N, S, S(O), or N(R 2 ); wherein Q is optionally substituted with one or more groups selected from oxo, —OR 2 , —R 2 , —N(R 2 ) 2 , —N(R 2 )—C(O)—R 2 , —R 2 —OH, —CN, —CO 2 R 2 , —C(O)—N(R 2 ) 2 , halo or —CF 3 ;
  • E is selected from Ht; O-Ht; Ht-Ht; —O—R 3 ; —N(R 2 )(R 3 ); C 1 -C 6 alkyl, which is optionally substituted with one or more groups selected from R 4 or Ht; C 2 -C 6 alkenyl, which is optionally substituted with one or more groups selected from R 4 or Hit; C 3 -C 6 saturated carbocycle, which is optionally substituted with one or more groups selected from R 4 or Ht; or C 5 -C 6 unsaturated carbocycle, which is optionally substituted with one or more groups selected from R 4 or Ht;
  • each R 4 is independently selected from —OR 2 , —SR 2 , —C(O)—NHR 2 , —S(O) 2 —NHR 2 , halo, —NR 2 —C(O)—R 2 , —N(R 2 ) 2 or —CN;
  • each R 7 is independently selected from
  • each M is independently selected from H, Li, Na, K, Mg, Ca, Ba, —N(R 2 ) 4 , C 1 -C 12 -alkyl, C 2 -C 12 -alkenyl, or —R 6 ; wherein 1 to 4 —CH 2 radicals of the alkyl or alkenyl group, other than the —CH 2 that is bound to Z, is optionally replaced by a heteroatom group selected from O, S, S(O), S(O 2 ), or N(R 2 ); and wherein any hydrogen in said alkyl, alkenyl or R 6 is optionally replaced with a substituent selected from oxo, —OR 2 , —R 2 , N(R 2 ) 2 , N(R 2 ) 3 , R 2 OH, —CN, —CO 2 R 2 , —C(O)—N(R 2 ) 2 , S(O) 2 —N(R 2 ) 2 , N(R 2 )—
  • M′ is H, C 1 -C 12 -alkyl, C 2 -C 12 -alkenyl, or —R 6 ; wherein 1 to 4 —CH 2 radicals of the alkyl or alkenyl group is optionally replaced by a heteroatom group selected from O, S, S(O), S(O 2 ), or N(R 2 ); and wherein any hydrogen in said alkyl, alkenyl or R 6 is optionally replaced with a substituent selected from oxo, —OR 2 , —R 2 , —N(R 2 ) 2 , N(R 2 ) 3 , —R 2 OH, —CN, —CO 2 R 2 , —C(O)—N(R 2 ) 2 , —S(O) 2 —N(R 2 ) 2 , —N(R 2 )—C(O)—R 2 , —C(O)R 2 , —S(O) n —R 2 ,
  • Z is CH 2 , O, S, N(R 2 ) 2 , or, when M is absent, H;
  • Y is P or S
  • X is O or S
  • R 9 is C(R 2 ) 2 , O or N(R 2 ); and wherein when Y is S, Z is not S; and
  • R 6 is a 5-6 membered saturated, partially saturated or unsaturated carbocyclic or heterocyclic ring system, or an 8-10 membered saturated, partially saturated or unsaturated bicyclic ring system; wherein any of said heterocyclic ring systems contains one or more heteroatoms selected from O, N, S, S(O) n or N(R 2 ); and wherein any of said ring systems optionally contains 1 to 4 substituents independently selected from OH, C 1 -C 4 alkyl, O—C 1 -C 4 alkyl or OC(O)C 1 -C 4 alkyl.
  • compositions comprising the sulfonamide prodrugs of formula I and methods for their use as prodrugs of HIV aspartyl protease inhibitors.
  • —SO 2 — and “—S(O) 2 -” as used herein refer to a sulfone or sulfone derivative (i.e., both appended groups linked to the S), and not a sulfinate ester.
  • the stereochemistry of OR 7 is defined relative to D on the adjacent carbon atom, when the molecule is drawn in an extended zig-zag representation (such as that drawn for compounds of formula XI, XV, XXII, XXIII and XXXI). If both OR 7 and D reside on the same side of the plane defined by the extended backbone of the compound, the stereochemistry of OR 7 will be referred to as “syn”. If OR 7 and D reside on opposite sides of that plane, the stereochemistry of OR 7 will be referred to as “anti”.
  • aryl alone or in combination with any other term, refers to a carbocyclic aromatic radical containing the specified number of carbon atoms.
  • heterocyclic refers to a stable 5-7 membered monocycle or 8-11 membered bicyclic heterocycle which is either saturated or unsaturated, and which may be optionally benzofused if monocyclic.
  • Each heterocycle consists of carbon atoms and from one to four heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur.
  • nitrogen and sulfur heteroatoms include any oxidized form of nitrogen and sulfur, and the quaternized form of any basic nitrogen.
  • the heterocyclic ring may be attached by any heteroatom of the cycle which results in the creation of a stable structure.
  • Preferred heterocycles defined above include, for example, benzimidazolyl, imidazolyl, imidazolinoyl, imidazolidinyl, quinolyl, isoquinolyl, indolyl, pyridyl, pyrrolyl, pyrrolinyl, pyrazolyl, pyrazinyl, quinoxolyl, piperidinyl, morpholinyl, thiamorpholinyl, furyl, thienyl, triazolyl, thiazolyl, ⁇ -carbolinyl, tetrazolyl, thiazolidinyl, benzofuranoyl, thiamorpholinyl sulfone, benzoxazolyl, oxopiperidinyl, oxopyrroldinyl, oxoazepinyl, azepinyl, isoxazolyl, tetrahydropyranyl, tetrahydrofuranyl
  • HIV protease and “HIV aspartyl protease” are used interchangeably and refer to the aspartyl protease encoded by the human immunodeficiency virus type 1 or 2. In a preferred embodiment of this invention, these terms refer to the human immunodeficiency virus type 1 aspartyl protease.
  • pharmaceutically effective amount refers to an amount effective in treating HIV infection in a patient.
  • prophylactically effective amount refers to an amount effective in preventing HIV infection in a patient.
  • patient refers to a mammal, including a human.
  • pharmaceutically acceptable carrier or adjuvant refers to a non-toxic carrier or adjuvant that may be administered to a patient, together with a compound of this invention, and which does not destroy the pharmacological activity thereof.
  • Pharmaceutically acceptable salts of the compounds of this invention include those derived from pharmaceutically acceptable inorganic and organic acids and bases.
  • suitable acids include hydrochloric, hydrobromic, sulfuric, nitric, perchloric, fumaric, maleic, phosphoric, glycollic, lactic, salicylic, succinic, toluene-p-sulfonic, tartaric, acetic, citric, methanesulfonic, formic, benzoic, malonic, naphthalene-2-sulfonic and benzenesulfonic acids.
  • Other acids, such as oxalic while not in themselves pharmaceutically acceptable, may be employed in the preparation of salts useful as intermediates in obtaining the compounds of the invention and their pharmaceutically acceptable acid addition salts.
  • Salts derived from appropriate bases include alkali metal (e.g., sodium), alkaline earth metal (e.g., magnesium), ammonium and N—(C 1-4 alkyl) 4+ salts.
  • thiocarbamates refers to compounds containing the functional group N—SO 2 —O.
  • the compounds of this invention contain one or more asymmetric carbon atoms and thus occur as racemates and racemic mixtures, single enantiomers, diastereomeric mixtures and individual diastereomers. All such isomeric forms of these compounds are expressly included in the present invention.
  • Each stereogenic carbon may be of the R or S configuration.
  • the explicitly shown hydroxyl is also preferred to be syn to D, in the extended zigzag conformation between the nitrogens shown in compounds of formula I.
  • stable refers to compounds which possess stability sufficient to allow manufacture and administration to a mammal by methods known in the art. Typically, such compounds are stable at a temperature of 40° C. or less, in the absence of moisture or other chemically reactive conditions, for at least a week.
  • the compounds of the present invention may be used in the form of salts derived from inorganic or organic acids. Included among such acid salts, for example, are the following: acetate, adipate, alginate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxy-yethanesulfonate, lactate, maleate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, oxalate, pamoate, pectinate, persulf
  • This invention also envisions the quaternization of any basic nitrogen-containing groups of the compounds disclosed herein.
  • the basic nitrogen can be quaternized with any agents known to those of ordinary skill in the art including, for example, lower alkyl halides, such as methyl, ethyl, propyl and butyl chloride, bromides and iodides; dialkyl sulfates including dimethyl, diethyl, dibutyl and diamyl sulfates; long chain halides such as decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides; and aralkyl halides including benzyl and phenethyl bromides. Water or oil-soluble or dispersible products may be obtained by such quaternization.
  • novel sulfonamides of this invention are those of formula I:
  • A is selected from H; Ht; —R 1 —Ht; —R 1 —C 1 -C 6 alkyl, which is optionally substituted with one or more groups independently selected from hydroxy, C 1 -C 4 alkoxy, Ht, —O-Ht, —NR 2 —CO—N(R 2 ) 2 or —CO—N(R 2 ) 2 ; —R 1 —C 2 -C 6 alkenyl, which is optionally substituted with one or more groups independently selected from hydroxy, C 1 -C 4 alkoxy, Ht, —O-Ht, —NR 2 —CO—N(R 2 ) 2 or —CO—N(R 2 ) 2 ; or R 7 ;
  • each R 1 is independently selected from —C(O)—, —S(O) 2 —, —C(O)—C(O)—, —O—C(O)—, —O—S(O) 2 , —NR 2 —S(O) 2 —, —NR 2 —C(O)— or —NR 2 —C(O)—C(O)—;
  • each Ht is independently selected from C 3 -C 7 cycloalkyl; C 5 -C 7 cycloalkenyl; C 6 -C 10 aryl; or a 5-7 membered saturated or unsaturated heterocycle, containing one or more heteroatoms selected from N, N(R 2 ), O, S and S(O) n ; wherein said aryl or said heterocycle is optionally fused to Q; and wherein any member of said Ht is optionally substituted with one or more substituents independently selected from oxo, —OR 2 , SR 2 , —R 2 , —N(R 2 )(R 2 ), —R 2 —OH, —CN, —CO 2 R 2 , —C(O)—N(R 2 ) 2 , —S(O) 2 —N(R 2 ) 2 , —N(R 2 )—C(O)—R 2 , —C(O)—R 2 , —S(
  • each R 2 is independently selected from H, or C 1 -C 4 alkyl optionally substituted with Q;
  • B when present, is —N(R 2 )—C(R 3 ) 2 —C(O)—;
  • each x is independently 0 or 1;
  • each R 3 is independently selected from H, Ht, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 3 -C 6 cycloalkyl or C 5 -C 6 cycloalkenyl; wherein any member of said R 3 , except H, is optionally substituted with one or more substituents selected from —OR 2 , —C(O)—NH—R 2 , —S(O), —N(R 2 )(R 2 ), Ht, —CN, —SR 2 , —CO 2 R 2 , NR 2 —C(O)—R 2 ;
  • each n is independently 1 or 2;
  • G when present, is selected from H, R 7 or C 1 -C 4 alkyl, or, when G is C 1 -C 4 alkyl, G and R 7 are bound to one another either directly or through a C 1 -C 3 linker to form a heterocyclic ring; or
  • D and D′ are independently selected from Q; C 1 -C 6 alkyl, which is optionally substituted with one or more groups selected from C 3 -C 6 cycloalkyl, —OR 2 , —R 3 , —O-Q or Q; C 2 -C 4 alkenyl, which is optionally substituted with one or more groups selected from C 3 -C 6 cycloalkyl, —OR 2 , —R 3 , —O-Q or Q; C 3 -C 6 cycloalkyl, which is optionally substituted with or fused to Q; or C 5 -C 6 cycloalkenyl, which is optionally substituted with or fused to Q;
  • each Q is independently selected from a 3-7 membered saturated, partially saturated or unsaturated carbocyclic ring system; or a 5-7 membered saturated, partially saturated or unsaturated heterocyclic ring containing one or more heteroatoms selected from O, N, S, S(O) n or N(R 2 ); wherein Q is optionally substituted with one or more groups selected from oxo, —OR 2 , —R 2 , —N(R 2 ) 2 , —N(R 2 )—C(O)—R 2 , —R 2 —OH, —CN, —CO 2 R 2 , —C(O)—N(R 2 ) 2 , halo or —CF 3 ;
  • E is selected from Ht; O-Ht; Ht-Ht; —O—R 3 ; —N(R 2 )(R 3 ); C 1 -C 6 alkyl, which is optionally substituted with one or more groups selected from R 4 or Ht; C 2 -C 6 alkenyl, which is optionally substituted with one or more groups selected from R 4 or Ht; C 3 -C 6 saturated carbocycle, which is optionally substituted with one or more groups selected from R 4 or Ht; or C 5 -C 6 unsaturated carbocycle, which is optionally substituted with one or more groups selected from R 4 or Ht;
  • each R 4 is independently selected from —OR 2 , —SR 2 , —C(O)—NHR 2 , —S(O) 2 —NHR 2 , halo, —NR 2 —C(O)—R 2 , —N(R 2 ) 2 or —CN;
  • each R 7 is independently selected from
  • each M is independently selected from H, Li, Na, K, Mg, Ca, Ba, —N(R 2 ) 4 , C 1 -C 12 -alkyl, C 2 -C 12 -alkenyl, or —R 6 ; wherein 1 to 4 —CH 2 radicals of the alkyl or alkenyl group, other than the —CH 2 that is bound to Z, is optionally replaced by a heteroatom group selected from O, S, S(O), S(O 2 ), or N(R 2 ); and wherein any hydrogen in said alkyl, alkenyl or R 6 is optionally replaced with a substituent selected from oxo, —OR 2 , —R 2 , N(R 2 ) 2 , N(R 2 ) 3 , R 2 OH, —CN, —CO 2 R 2 , —C(O)—N(R 2 ) 2 , S(O) 2 —N(R 2 ) 2 , N(R 2 )—
  • M′ is H, C 1 -C 12 -alkyl, C 2 -C 12 -alkenyl, or —R 6 ; wherein 1 to 4 —CH 2 radicals of the alkyl or alkenyl group is optionally replaced by a heteroatom group selected from O, S, S(O), S(O 2 ), or N(R 2 ); and wherein any hydrogen in said alkyl, alkenyl or R 6 is optionally replaced with a substituent selected from oxo, —OR 2 , —R 2 , —N(R 2 ) 2 , N(R 2 ) 3 , —R 2 OH, —CN, —CO 2 R 2 , —C(O)—N(R 2 ) 2 , —S(O) 2 —N(R 2 ) 2 , —N(R 2 )—C(O)—R 2 , —C(O)R 2 , —S(O) n —R 2 ,
  • Z is CH 2 , O, S, N(R 2 ) 2 , or, when M is not present, H.
  • Y is P or S
  • X is O or S
  • R 9 is C(R 2 ) 2 , O or N(R 2 ); and wherein when Y is S, Z is not S; and
  • R 6 is a 5-6 membered saturated, partially saturated or unsaturated carbocyclic or heterocyclic ring system, or an 8-10 membered saturated, partially saturated or unsaturated bicyclic ring system; wherein any of said heterocyclic ring systems contains one or more heteroatoms selected from O, N, S, S(O) n or N(R 2 ); and wherein any of said ring systems optionally contains 1 to 4 substituents independently selected from OH, C 1 -C 4 alkyl, O—C 1 -C 4 alkyl or O—C(O)—C 1 -C 4 alkyl.
  • At least one R 7 is selected from:
  • component M or M′ in the formulae set forth herein will have either a covalent, a covalent/zwitterionic, or an ionic association with either Z or R 9 depending upon the actual choice for M or M′.
  • M or M′ is hydrogen, alkyl, alkenyl, or R 6
  • M or M′ is covalently bound to R 9 or Z.
  • M is a mono- or bivalent metal or other charged species (i.e., NH 4 + )
  • Z When x is 0 in (M) x , Z may be a charged species. When that occurs, the other M may be oppositely charged to produce a 0 net charge on the molecule. Alternatively, the counter ion may located elsewhere in the molecule.
  • variables A, R 1 -R 4 , R 6 -R 9 , Ht, B, x, n, D, D′, M, Q, X, Y, Z and E are to be taken as they are defined above for the compounds of formula I.
  • the compounds of this invention are those represented by formulas XXII, XXIII or XXXI:
  • R 3 , R 7 , Ht, D, D′, x, E are as defined above for compounds of formula I.
  • R 3 and R 3′ the two R 3 moieties present in formula XXXI have been labeled R 3 and R 3′ .
  • A is selected from 3-tetrahydrofuryl-O—C(O)—, 3-(1,5-dioxane)-O—C(O)—, or 3-hydroxy-hexahydrofura[2,3-b]-furanyl-O—C(O)—;
  • D′ is C 1 -C 4 alkyl which is optionally substituted with one or more groups selected from the group consisting of C 3 -C 6 cycloalkyl, —OR 2 , —R 3 , —O-Q and Q;
  • E is C 6 -C 10 aryl optionally substituted with one or more substituents selected from oxo, —OR 2 , SR 2 , —R 2 , —N(R 2 ) 2 , —R 2 —OH, —CN, —CO 2 R 2 , —C(O)—N(R 2 ) 2 , —S(O) 2 —N(R 2 ) 2 , —N(R 2 )—C(O)—R 2 , —C(O)—R 2 , —S(O), —R 2 , —OCF 3 , —S(O) n -Q, methylenedioxy, —N(R 2 )—S(O) 2 (R 2 ), halo, —CF 3 , —NO 2 , Q, —OQ, —OR 7 , —SR 7 , —R 7 , —N(R 2 )(R 7 ) or —N
  • Ht insofar as it is defined as part of R 3 , is defined as above except for the exclusion of heterocycles;
  • each M is independently selected from H, Li, Na, K, Mg, Ca, Ba, C 1 -C 4 alkyl or —N(R 2 ) 4 ; and each M′ is H or C 1 -C 4 alkyl.
  • E is a 5-membered heterocyclic ring containing one S and optionally containing N as an additional heteroatom, wherein said heterocyclic ring is optionally substituted with one to two groups independently selected from —CH 3 , R 4 , or Ht; and
  • R 7 in —OR 7 is —PO(OM) 2 or C(O)CH 2 OCH 2 CH 2 OCH 2 CH 2 OCH 3 and both R 7 in —N(R 7 ) 2 are H, wherein M is H, Li, Na, K or C 1 -C 4 alkyl; or wherein R 7 in —OR 7 is C(O)CH 2 OCH 2 CH 2 OCH 3 , one R 7 in —N(R 7 ) 2 is C(O)CH 2 OCH 2 CH 2 OCH 3 and the other is H.
  • R 3 is C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 5 -C 6 cycloalkyl, C 5 -C 6 cycloalkenyl or a 5-6 membered saturated or unsaturated heterocycle, wherein any member of said R 3 may be optionally substituted with one or more substituents selected from the group consisting of —OR 2 , —C(O)—NH—R 2 , —S(O) n N(R 2 )(R 2 ), Ht, —CN, —SR 2 , —C(O) 2 R 2 and NR 2 —C(O)—R 2 ; and
  • D′ is C 1 -C 3 alkyl or C 3 alkenyl, wherein said alkyl or alkenyl may optionally be substituted with one or more groups selected from the group consisting of C 3 -C 6 cycloalkyl, —OR 2 , —O-Q and Q (with all other variables being defined as above for compounds of formula I).
  • each R 3 is independently C 1 -C 6 alkyl which may be optionally substituted with a substituent selected from the group consisting of —OR 2 , —C(O)—NH—R 2 , —S(O) n N(R 2 )(R 2 ), Ht, —CN, —SR 2 , —CO 2 R 2 or —NR 2 —C(O)—R 2 ; and D′ is C 1 -C 4 alkyl, which may be optionally substituted with a group selected from the group consisting of C 3 -C 6 cycloalkyl, —OR 2 , —O-Q; and E is Ht, Ht-Ht and —NR 2 R 3 .
  • the invention provides compounds of the following formulae:
  • the prodrugs of the present invention may be synthesized using conventional synthetic techniques.
  • U.S. Pat. No. 5,585,397 discloses the synthesis of compounds of formula:
  • Prodrugs of formula (I) of the present invention can be readily synthesized from the '397 compounds using conventional techniques.
  • One of skill in the art would be well aware of conventional synthetic reagents to convert the —OH group of the '397 compounds to a desired —OR 7 functionality of the present invention, wherein R 7 is as defined above.
  • the relative ease with which the compounds of this invention can be synthesized represents an enormous advantage in the large scale production of these compounds.
  • VX-478 a compound disclosed in the '397 patent, can be readily converted to the corresponding bis-phosphate ester derivative, as shown below:
  • compositions of the present invention may be readily prepared using known techniques.
  • disodium salt of the mono-phosphate ester shown above can be prepared as shown below:
  • the compounds of this invention may be modified by appending appropriate functionalities to enhance selective biological properties.
  • modifications are known in the art and include those which increase biological penetration into a given biological system (e.g., blood, lymphatic system, central nervous system), increase oral availability, increase solubility to allow administration by injection, alter metabolism and alter rate of excretion.
  • the first mechanism involves the enzymatic or chemical transformation of the prodrug species into the active form.
  • the second mechanism involves the enzymatic or chemical cleavage of a functionality on the prodrug to produce the active compound.
  • the chemical or enzymatic transformation can involve to transfer of a functional group (i.e., R 7 ) from one heteroatom within the molecule to another heteroatom. This transfer is demonstrated in the chemical reactions shown below:
  • the cleavage mechanism is demonstrated by the reaction below where a phosphate ester-containing prodrug is converted into the active form of the drug by removal of the phosphate group.
  • protease inhibitors and their utility as inhibitors of aspartyl proteases are described in U.S. Pat. No. 5,585,397, the disclosure of which is incorporated herein by reference.
  • the prodrugs of the present invention are characterized by unexpectedly high aqueous solubility. This solubility facilitates administration of higher doses of the prodrug, resulting in a greater drug load per unit dosage.
  • the prodrugs of the present invention are also characterized by facile hydrolytic cleavage to release the active aspartyl protease inhibitor in vivo.
  • the high aqueous solubility and the facile in vivo metabolism result in a greater bioavailability of the drug. As a result, the pill burden on a patient is significantly reduced.
  • the prodrugs of this invention may be employed in a conventional manner for the treatment of viruses, such as HIV and HTLV, which depend on aspartyl proteases for obligatory events in their life cycle. Such methods of treatment, their dosage levels and requirements may be selected by those of ordinary skill in the art from available methods and techniques.
  • a prodrug of this invention may be combined with a pharmaceutically acceptable adjuvant for administration to a virally-infected patient in a pharmaceutically acceptable manner and in an amount effective to lessen the severity of the viral infection.
  • the prodrugs of this invention may be used in vaccines and methods for protecting individuals against viral infection over an extended period of time.
  • the prodrugs may be employed in such vaccines either alone or together with other compounds of this invention in a manner consistent with the conventional utilization of protease inhibitors in vaccines.
  • a prodrug of this invention may be combined with pharmaceutically acceptable adjuvants conventionally employed in vaccines and administered in prophylactically effective amounts to protect individuals over an extended period time against HIV infection.
  • the novel protease inhibitors of this invention can be administered as agents for treating or preventing HIV infection in a mammal.
  • the prodrugs of this invention may be administered to a healthy or HIV-infected patient either as a single agent or in combination with other anti-viral agents which interfere with the replication cycle of HIV.
  • the compounds of this invention may be administered with other anti-viral agents which target different events in the viral life cycle, the therapeutic effect of these compounds is potentiated.
  • the co-administered anti-viral agent can be one which targets early events in the life cycle of the virus, such as cell entry, reverse transcription and viral DNA integration into cellular DNA.
  • Anti-HIV agents targeting such early life cycle events include, didanosine (ddI), alcitabine (ddC), d4T, zidovudine (AZT), polysulfated polysaccharides, sT4 (soluble CD4), ganiclovir, dideoxycytidine, trisodium phosphonoformate, eflornithine, ribavirin, acyclovir, alpha interferon and trimenotrexate.
  • non-nucleoside inhibitors of reverse transcriptase such as TIBO or nevirapine may be used to potentiate the effect of the compounds of this invention, as may viral uncoating inhibitors, inhibitors of trans-activating proteins such as tat or rev, or inhibitors of the viral integrase.
  • Combination therapies according to this invention exert a synergistic effect in inhibiting HIV replication because each component agent of the combination acts on a different site of HIV replication.
  • the use of such combinations also advantageously reduces the dosage of a given conventional anti-retroviral agent which would be required for a desired therapeutic or prophylactic effect as compared to when that agent is administered as a monotherapy.
  • These combinations may reduce or eliminate the side effects of conventional single anti-retroviral agent therapies while not interfering with the anti-retroviral activity of those agents.
  • These combinations reduce potential of resistance to single agent therapies, while minimizing any associated toxicity.
  • These combinations may also increase the efficacy of the conventional agent without increasing the associated toxicity.
  • prodrugs act synergistically in preventing the replication of HIV in human T cells.
  • Preferred combination therapies include the administration of a prodrug of this invention with AZT, ddI, ddC or d4T.
  • the prodrugs of this invention may also be co-administered with other HIV protease inhibitors such as Ro 31-8959 (Roche), L-735,524 (Merck), XM 323 (Du-Pont Merck) and A-80,987 (Abbott) to increase the effect of therapy or prophylaxis against various viral mutants or members of other HIV quasi species.
  • HIV protease inhibitors such as Ro 31-8959 (Roche), L-735,524 (Merck), XM 323 (Du-Pont Merck) and A-80,987 (Abbott) to increase the effect of therapy or prophylaxis against various viral mutants or members of other HIV quasi species.
  • prodrugs of this invention we prefer administering the prodrugs of this invention as single agents or in combination with retroviral reverse transcriptase inhibitors, such as derivatives of AZT, or other HIV aspartyl protease inhibitors.
  • retroviral reverse transcriptase inhibitors such as derivatives of AZT, or other HIV aspartyl protease inhibitors.
  • retroviral reverse transcriptase inhibitors such as derivatives of AZT, or other HIV aspartyl protease inhibitors.
  • the prodrugs of this invention can also be administered in combination with immunomodulators (e.g., bropirimine, anti-human alpha interferon antibody, IL-2, GM-CSF, methionine enkephalin, interferon alpha, diethyldithiocarbamate, tumor necrosis factor, naltrexone and rEPO); and antibiotics (e.g., pentamidine isethiorate) to prevent or combat infection and disease associated with HIV infections, such as AIDS and ARC.
  • immunomodulators e.g., bropirimine, anti-human alpha interferon antibody, IL-2, GM-CSF, methionine enkephalin, interferon alpha, diethyldithiocarbamate, tumor necrosis factor, naltrexone and rEPO
  • antibiotics e.g., pentamidine isethiorate
  • prodrugs of this invention When the prodrugs of this invention are administered in combination therapies with other agents, they may be administered sequentially or concurrently to the patient.
  • pharmaceutical or prophylactic compositions according to this invention may be comprised of a combination of a prodrug of this invention and another therapeutic or prophylactic agent.
  • the compounds of this invention can also be used as inhibitory agents for other viruses which depend on similar aspartyl proteases for obligatory events in their life cycle. These viruses include, as well as other AIDS-like diseases caused by retroviruses, such as simian immunodeficiency viruses, but are not limited to, HTLV-I and HTLV-II.
  • viruses include, as well as other AIDS-like diseases caused by retroviruses, such as simian immunodeficiency viruses, but are not limited to, HTLV-I and HTLV-II.
  • the compounds of this invention may also be used to inhibit other aspartyl proteases, and in particular, other human aspartyl proteases, including renin and aspartyl proteases that process endothelin precursors.
  • compositions of this invention comprise any of the compounds of the present invention, and pharmaceutically acceptable salts thereof, with any pharmaceutically acceptable carrier, adjuvant or vehicle.
  • Pharmaceutically acceptable carriers, adjuvants and vehicles that may be used in the pharmaceutical compositions of this invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol and wool fat.
  • compositions of this invention may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir. We prefer oral administration or administration by injection.
  • the pharmaceutical compositions of this invention may contain any conventional non-toxic pharmaceutically-acceptable carriers, adjuvants or vehicles.
  • parenteral as used herein includes subcutaneous, intracutaneous, intravenous, intramuscular, intra-articular, intrasynovial, intrasternal, intrathecal, intralesional and intracranial injection or infusion techniques.
  • the pharmaceutical compositions may be in the form of a sterile injectable preparation, for example, as a sterile injectable aqueous or oleaginous suspension.
  • This suspension may be formulated according to techniques known in the art using suitable dispersing or wetting agents (such as, for example, Tween 80) and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example, as a solution in 1,3-butanediol.
  • suitable vehicles and solvents that may be employed are mannitol, 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 and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions.
  • These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant such as Ph. Helv or a similar alcohol.
  • compositions of this invention may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, and aqueous suspensions and solutions.
  • carriers which are commonly used include lactose and corn starch.
  • Lubricating agents such as magnesium stearate, are also typically added.
  • useful diluents include lactose and dried corn starch.
  • aqueous suspensions are administered orally, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening and/or flavoring and/or coloring agents may be added.
  • compositions of this invention may also be administered in the form of suppositories for rectal administration.
  • These compositions can be prepared by mixing a compound of this invention with a suitable non-irritating excipient which is solid at room temperature but liquid at the rectal temperature and therefore will melt in the rectum to release the active components.
  • suitable non-irritating excipient include, but are not limited to, cocoa butter, beeswax and polyethylene glycols.
  • Topical administration of the pharmaceutical compositions of this invention is especially useful when the desired treatment involves areas or organs readily accessible by topical application.
  • the pharmaceutical composition should be formulated with a suitable ointment containing the active components suspended or dissolved in a carrier.
  • Carriers for topical administration of the compounds of this invention include, but are not limited to, mineral oil, liquid petroleum, white petroleum, propylene glycol, polyoxyethylene polyoxypropylene compound, emulsifying wax and water.
  • the pharmaceutical composition can be formulated with a suitable lotion or cream containing the active compound suspended or dissolved in a carrier.
  • Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
  • the pharmaceutical compositions of this invention may also be topically applied to the lower intestinal tract by rectal suppository formulation or in a suitable enema formulation. Topically-transdermal patches are also included in this invention.
  • compositions of this invention may be administered by nasal aerosol or inhalation.
  • Such compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other solubilizing or dispersing agents known in the art.
  • Dosage levels of between about 0.01 and about 100 mg/kg body weight per day, preferably between about 0.5 and about 50 mg/kg body weight per day of the active ingredient compound are useful in the prevention and treatment of viral infection, including HIV infection.
  • the pharmaceutical compositions of this invention will be administered from about 1 to about 5 times per day or alternatively, as a continuous infusion. Such administration can be used as a chronic or acute therapy.
  • the amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration.
  • a typical preparation will contain from about 5% to about 95% active compound (w/w).
  • such preparations contain from about 20% to about 80% active compound.
  • a maintenance dose of a compound, composition or combination of this invention may be administered, if necessary. Subsequently, the dosage or frequency of administration, or both, may be reduced, as a function of the symptoms, to a level at which the improved condition is retained when the symptoms have been alleviated to the desired level, treatment should cease. Patients may, however, require intermittent treatment on a long-term basis upon any recurrence of disease symptoms.
  • 13C (d3-acetonitrile): 157.1, 157.0, 153.2, 139.6, +130.3, +130.2, +129.2, +127.2, 126.2, +114.2, +76.0, +75.4, ⁇ 73.6, ⁇ 67.4, ⁇ 58.2, +54.9, ⁇ 50.2, ⁇ 41.6, ⁇ 39.8, ⁇ 35.9, ⁇ 33.4, +27.3, +23.1, +20.4, +20.2.
  • 13C (dmso): 155.8, 155.7, 138.5, +129.1, +129.0, +128.0, +126.1, 122.9, +112.7, +74.6, +74.3, ⁇ 72.7, ⁇ 66.2, ⁇ 57.2, +53.6, ⁇ 49.5, ⁇ 37.4, ⁇ 36.7, ⁇ 35.5, ⁇ 32.1, ⁇ 27.6, +26.2, +20.0, +19.8.
  • 13C (DMSO): 169.3, 155.8, 153.1, 138.0, 129.1, 129.0, 128.1, 126.3, 122.6, 112.8, 94.3, 75.6, 74.6, 72.4, 66.1, 57.8, 52.7, 52.0, 49.3, 38.4, 34.7, 32.2, 29.1, 26.6, 21.4, 20.1, 20.0.
  • 13C (DMSO): 170.2, 169.8, 156.4, 143.4, 138.8, 129.5, 128.8, 128.5, 126.8, 119.7, 74.9, 74.2, 73.7, 71.6, 70.7, 70.3, 68.0, 67.2, 59.3, 57.6, 53.8, 49.6, 35.7, 33.8, 27.1, 20.4.
  • 227 can be synthesized directly from 197.
  • 197 was dissolved in pyridine (300 mL).
  • the resulting solution was concentrated in vacuo to about 150 ml at 50-55° C.
  • the solution was then cooled under N 2 to 5° C., and treated with POCl 3 (6.5 ml, 1.24 equiv.) over 2 minutes.
  • the cooling bath was removed and the reaction stirred at ambient temperature for 2.5 hrs.
  • the solution was then cooled to 5° C. and water (300 ml) was added over 30 minutes.
  • the resulting mixture was extracted with 4-methylpentan-2-one (MIBK, 2 ⁇ 150 ml).
  • the combined extracts were washed with 2N HCl (2 ⁇ 250 ml).
  • the acid washes were back extracted with MIBK (60 ml), then the combined MIBK solutions were treated with 2N HCl (150 ml).
  • the two phase mixture was stirred rapidly and heated to 50° C. for 2 hours.
  • the reaction mixture was cooled to 20° C., the phases were separated and the MIBK solution was washed with brine (150 ml).
  • the product, 227 was isolated by drying the solution with magnesium sulfate, filtering of the drying agent and concentrating in vacuo at 40° C. to give the product as a pale yellow foam (31 g, 90% yield).
  • Solid 228 (2.66 g, 4.54 mmol) was treated with ml aqueous sodium bicarbonate (Baker, 3.0 equiv., 13.63 mmol, 1.14 g) and loaded onto a resin column (Mitsubishi Kasei Corp., MCI-gel, CHP-20). Distilled water was run through until the eluent was neutral followed by product elution with 1% acetonitrile in water. Pure fractions were pooled and lyophilized to give 918 mg of pure bis-sodium salt 229.
  • the solution was evaporated en vacuo to 100 ml then lyophilized to yield the TEA salt (1.5 TEA equivalents).
  • the TEA salt was (5.8 g) was dissolved in 200 ml water, 300 ml of 1 N HCl was added and the mixture was extracted with EtOAc (3 ⁇ 200 ml).
  • the ethyl acetate solution was dried with MgSO 4 then evaporated en vacuo to yield 4 g of the free acid.
  • Two grams of the free acid was dissolved in 50 ml of acetonitrile and a solution of 573 mg NaHCO 3 in 200 ml water was added. The mixture was lyophilized yielding 2.1 g of the bis sodium salt (compound 229).
  • 13C (DMSO): 156.2, 150.1, 145.7, 140.0, +129.7, +129.2, +128.5, +126.3, +125.0, +71.8, ⁇ 60.0, +56.2, ⁇ 56.0, ⁇ 51.8, ⁇ 36.0, +26.3, +20.3, +20.1, +14.6.
  • reaction mixture was then diluted with ethyl acetate and extracted with 1N hydrochloric acid, saturated sodium bicarbonate and water. Evaporation of the solvent and purification on silica gel (30% ethyl acetate-hexane) gave the desired product (500 mg) as a waxy solid.
  • Separate groups of male and female Sprague-Dawley rats received oral doses of compound 229 by oral gavage, in different vehicles at the same dose equivalent (40 mg/kg molar equivalent of VX-478).
  • the different vehicles for compound 229 were: 1) water; 2) 5/4/1; 3) PEG 400; 4) TPGS/PEG 400; and 5) PEG.
  • the vehicles for VX-478 were: 1) 33% TPGS/PEG400/PEG; and 2) 12.5% TPGS/PEG 400/PEG.

Abstract

The present invention relates to prodrugs of a class of sulfonamides which are aspartyl protease inhibitors. In one embodiment, this invention relates to a novel class of prodrugs of HIV aspartyl protease inhibitors characterized by favorable aqueous solubility, high oral bioavailability and facile in vivo generation of the active ingredient. This invention also relates to pharmaceutical compositions comprising these prodrugs. The prodrugs and pharmaceutical compositions of this invention are particularly well suited for decreasing the pill burden and increasing patient compliance. This invention also relates to methods of treating mammals with these prodrugs and pharmaceutical compositions.

Description

    TECHNICAL FIELD OF THE INVENTION
  • The present invention relates to prodrugs of a class of sulfonamides which are aspartyl protease inhibitors. In one embodiment, this invention relates to a novel class of prodrugs of HIV aspartyl protease inhibitors characterized by favorable aqueous solubility, high oral bioavailability and facile in vivo generation of the active ingredient. This invention also relates to pharmaceutical compositions comprising these prodrugs. The prodrugs and pharmaceutical compositions of this invention are particularly well suited for decreasing the pill burden and increasing patient compliance. This invention also relates to methods of treating mammals with these prodrugs and pharmaceutical compositions.
  • BACKGROUND OF THE INVENTION
  • Aspartyl protease inhibitors are considered the most effective current drug in the fight against HIV infection. These inhibitors, however, require certain physicochemical properties in order to achieve good potency against the enzyme. One of these properties is high hydrophobicity. Unfortunately, this property results in poor aqueous solubility and low oral bioavailability.
  • U.S. Pat. No. 5,585,397 describes a class of sulfonamide compounds that are inhibitors of the aspartyl protease enzyme. These compounds illustrate the drawbacks concomitant to pharmaceutical compositions comprising hydrophobic aspartyl protease inhibitors. For example, VX-478 (4-amino-N-((2-syn,3S)-2-hydroxy-4-phenyl-2((S)-tetrahydrofuran-3-yl-oxycarbonylamino)-butyl-N-isobutyl-benzenesulfonamide) is an aspartyl protease inhibitor disclosed in the '397 patent. It has a relatively low aqueous solubility. While the oral bioavailability of this inhibitor in a “solution” formulation is excellent, the dosage of VX-478 in this form is severely limited by the amount of liquid present in the particular liquid dosage from, e.g., encapsulated into a soft gelatin capsule. A higher aqueous solubility would increase drug load per unit dosage of VX-478.
  • Currently, the solution formulation of VX-478 produces an upper limit of 150 mg of VX-478 in each capsule. Given a therapeutic dose of 2400 mg/day of VX-478, this formulation would require a patient to consume 16 capsules per day. Such a high pill burden would likely result in poor patient compliance, thus producing sub-optimal therapeutic benefit of the drug. The high pill burden is also a deterrent to increasing the amount of the drug administered per day to a patient. Another drawback of the pill burden and the concomitant patient compliance problem is in the treatment of children infected with HIV.
  • Furthermore, these “solution” formulations, such as the mesylate formulation, are at a saturation solubility of VX-478. This creates the real potential of having the drug crystallize out of solution under various storage and/or shipping conditions. This, in turn, would likely result in a loss of some of the oral bioavailability achieved with VX-478.
  • One way of overcoming these problems is to develop a standard solid dosage form, such as a tablet or a capsule or a suspension form. Unfortunately, such solid dosage forms have much lower oral bioavailability of the drug.
  • Thus, there is a need to improve the drug load per unit dosage form for aspartyl protease inhibitors. Such an improved dosage form would reduce the pill burden and increase patient compliance. It would also provide for the possibility of increasing the amounts of the drug administered per day to a patient.
  • SUMMARY OF THE INVENTION
  • The present invention provides novel prodrugs of a class of sulfonamide compounds that are inhibitors of aspartyl protease, in particular, HIV aspartyl protease. These prodrugs are characterized by excellent aqueous solubility, increased bioavailability and are readily metabolized into the active inhibitors in vivo. The present invention also provides pharmaceutical compositions comprising these prodrugs and methods of treating HIV infection in mammals using these prodrugs and the pharmaceutical compositions thereof.
  • These prodrugs can be used alone or in combination with other therapeutic or prophylactic agents, such as anti-virals, antibiotics, immunomodulators or vaccines, for the treatment or prophylaxis of viral infection.
  • It is a principal object of this invention to provide a novel class of prodrugs of sulfonamide compounds that are aspartyl protease inhibitors, and particularly, HIV aspartyl protease inhibitors. This novel class of sulfonamides is represented by formula I:
  • Figure US20100124543A1-20100520-C00001
  • wherein:
  • A is selected from H; Ht; —R1—Ht; —R1—C1-C6 alkyl, which is optionally substituted with one or more groups independently selected from hydroxy, C1-C4 alkoxy, Ht, —O-Ht, —NR2—CO—N(R2)2 or —CO—N(R2)2; —R1—C2-C6 alkenyl, which is optionally substituted with one or more groups independently selected from hydroxy, C1-C4 alkoxy, Ht, —O-Ht, —NR2—CO—N(R2)2 or —CO—N(R2)2; or R7;
  • each R1 is independently selected from —C(O)—, —S(O)2—, —C(O)—C(O)—, —O—C(O)—, —O—S(O)2, —NR2—S(O)2—, —NR2—C(O)— or —NR2—C(O)—C(O)—;
  • each Ht is independently selected from C3-C7 cycloalkyl; C5-C7 cycloalkenyl; C6-C10 aryl; or a 5-7 membered saturated or unsaturated heterocycle, containing one or more heteroatoms selected from N, N(R2), O, S and S(O)n; wherein said aryl or said heterocycle is optionally fused to Q; and wherein any member of said Ht is optionally substituted with one or more substituents independently selected from oxo, —OR2, SR2, —R2, —N(R2)(R2), —R2—OH, —CN, —CO2R2, —C(O)—N(R2)2, —S(O)2—N(R2)2, —N(R2)—C(O)—R2, —C(O)—R2, —S(O)n—R2, —OCF3, —S(O)n-Q, methylenedioxy, —N(R2)—S(O)2 (R2), halo, —CF3, —NO2, Q, —OQ, —OR7, —SR7, —R7, —N(R2)(R7) or —N(R7)2;
  • each R2 is independently selected from H, or C1-C4 alkyl optionally substituted with Q;
  • B, when present, is —N(R2)—C(R3)2—C(O)—;
  • each x is independently 0 or 1;
  • each R3 is independently selected from H, Ht, C1-C6 alkyl, C2-C6 alkenyl, C3-C6 cycloalkyl or C5-C6 cycloalkenyl; wherein any member of said R3, except H, is optionally substituted with one or more substituents selected from —OR2, —C(O)—NH—R2, —S(O)n—N(R2)(R2), Ht, —CN, —SR2, —CO2R2, NR2—C(O)—R2;
  • each n is independently 1 or 2;
  • G, when present, is selected from H, R7 or C1-C4 alkyl, or, when G is C1-C4 alkyl, G and R′ are bound to one another either directly or through a C1-C3 linker to form a heterocyclic ring; or
  • when G is not present (i.e., when x in (G)x is 0), then the nitrogen to which G is attached is bound directly to the R7 group on —OR7;
  • D and D′ are independently selected from Q; C1-C6 alkyl, which is optionally substituted with one or more groups selected from C3-C6 cycloalkyl, —OR2, —R3, —O-Q or Q; C2-C4 alkenyl, which is optionally substituted with one or more groups selected from C3-C6 cycloalkyl, —OR2, —R3, —O-Q or Q; C3-C6 cycloalkyl, which is optionally substituted with or fused to Q; or C5-C6 cycloalkenyl, which is optionally substituted with or fused to Q;
  • each Q is independently selected from a 3-7 membered saturated, partially saturated or unsaturated carbocyclic ring system; or a 5-7 membered saturated, partially saturated or unsaturated heterocyclic ring containing one or more heteroatoms selected from O, N, S, S(O), or N(R2); wherein Q is optionally substituted with one or more groups selected from oxo, —OR2, —R2, —N(R2)2, —N(R2)—C(O)—R2, —R2—OH, —CN, —CO2R2, —C(O)—N(R2)2, halo or —CF3;
  • E is selected from Ht; O-Ht; Ht-Ht; —O—R3; —N(R2)(R3); C1-C6 alkyl, which is optionally substituted with one or more groups selected from R4 or Ht; C2-C6 alkenyl, which is optionally substituted with one or more groups selected from R4 or Hit; C3-C6 saturated carbocycle, which is optionally substituted with one or more groups selected from R4 or Ht; or C5-C6 unsaturated carbocycle, which is optionally substituted with one or more groups selected from R4 or Ht;
  • each R4 is independently selected from —OR2, —SR2, —C(O)—NHR2, —S(O)2—NHR2, halo, —NR2—C(O)—R2, —N(R2)2 or —CN;
  • each R7 is independently selected from
  • Figure US20100124543A1-20100520-C00002
  • wherein each M is independently selected from H, Li, Na, K, Mg, Ca, Ba, —N(R2)4, C1-C12-alkyl, C2-C12-alkenyl, or —R6; wherein 1 to 4 —CH2 radicals of the alkyl or alkenyl group, other than the —CH2 that is bound to Z, is optionally replaced by a heteroatom group selected from O, S, S(O), S(O2), or N(R2); and wherein any hydrogen in said alkyl, alkenyl or R6 is optionally replaced with a substituent selected from oxo, —OR2, —R2, N(R2)2, N(R2)3, R2OH, —CN, —CO2R2, —C(O)—N(R2)2, S(O)2—N(R2)2, N(R2)—C(O)—R2, C(O)R2, —S(O)n—R2, OCF3, —S(O)n—R6, N(R2)—S(O)2 (R2), halo, —CF3, or —NO2;
  • M′ is H, C1-C12-alkyl, C2-C12-alkenyl, or —R6; wherein 1 to 4 —CH2 radicals of the alkyl or alkenyl group is optionally replaced by a heteroatom group selected from O, S, S(O), S(O2), or N(R2); and wherein any hydrogen in said alkyl, alkenyl or R6 is optionally replaced with a substituent selected from oxo, —OR2, —R2, —N(R2)2, N(R2)3, —R2OH, —CN, —CO2R2, —C(O)—N(R2)2, —S(O)2—N(R2)2, —N(R2)—C(O)—R2, —C(O)R2, —S(O)n—R2, —OCF3, —S(O)n—R6, —N(R2)—S(O)2 (R2), halo, —CF3, or —NO2;
  • Z is CH2, O, S, N(R2)2, or, when M is absent, H;
  • Y is P or S;
  • X is O or S; and
  • R9 is C(R2)2, O or N(R2); and wherein when Y is S, Z is not S; and
  • R6 is a 5-6 membered saturated, partially saturated or unsaturated carbocyclic or heterocyclic ring system, or an 8-10 membered saturated, partially saturated or unsaturated bicyclic ring system; wherein any of said heterocyclic ring systems contains one or more heteroatoms selected from O, N, S, S(O)n or N(R2); and wherein any of said ring systems optionally contains 1 to 4 substituents independently selected from OH, C1-C4 alkyl, O—C1-C4 alkyl or OC(O)C1-C4 alkyl.
  • It is a also an object of this invention to provide pharmaceutical compositions comprising the sulfonamide prodrugs of formula I and methods for their use as prodrugs of HIV aspartyl protease inhibitors.
  • DETAILED DESCRIPTION OF THE INVENTION
  • In order that the invention herein described may be more fully understood, the following detailed description is set forth. In the description, the following abbreviations are used:
  • Designation Reagent or Fragment
    Ac acetyl
    Me methyl
    Et ethyl
    Bzl benzyl
    Trityl triphenylmethyl
    Asn D- or L-asparagine
    Ile D- or L-isoleucine
    Phe D- or L-phenylalanine
    Val D- or L-valine
    Boc tert-butoxycarbonyl
    Cbz benzyloxycarbonyl (carbobenzyloxy)
    Fmoc 9-fluorenylmethoxycarbonyl
    DCC dicyclohexylcarbodiimide
    DIC diisopropylcarbodiimide
    EDC 1-(3-dimethylaminopropyl)-3-
    ethylcarbodiimide hydrochloride
    HOBt 1-hydroxybenzotriazole
    HOSu 1-hydroxysuccinimide
    TFA trifluoroacetic acid
    DIEA diisopropylethylamine
    DBU 1,8-diazabicyclo (5.4.0) undec-7-ene
    EtOAc ethyl acetate
  • The following terms are employed herein:
  • Unless expressly stated to the contrary, the terms “—SO2—” and “—S(O)2-” as used herein refer to a sulfone or sulfone derivative (i.e., both appended groups linked to the S), and not a sulfinate ester.
  • For the compounds of formula I, and intermediates thereof, the stereochemistry of OR7 is defined relative to D on the adjacent carbon atom, when the molecule is drawn in an extended zig-zag representation (such as that drawn for compounds of formula XI, XV, XXII, XXIII and XXXI). If both OR7 and D reside on the same side of the plane defined by the extended backbone of the compound, the stereochemistry of OR7 will be referred to as “syn”. If OR7 and D reside on opposite sides of that plane, the stereochemistry of OR7 will be referred to as “anti”.
  • The term “aryl”, alone or in combination with any other term, refers to a carbocyclic aromatic radical containing the specified number of carbon atoms.
  • The term “heterocyclic” refers to a stable 5-7 membered monocycle or 8-11 membered bicyclic heterocycle which is either saturated or unsaturated, and which may be optionally benzofused if monocyclic. Each heterocycle consists of carbon atoms and from one to four heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur. As used herein, the terms “nitrogen and sulfur heteroatoms” include any oxidized form of nitrogen and sulfur, and the quaternized form of any basic nitrogen. The heterocyclic ring may be attached by any heteroatom of the cycle which results in the creation of a stable structure. Preferred heterocycles defined above include, for example, benzimidazolyl, imidazolyl, imidazolinoyl, imidazolidinyl, quinolyl, isoquinolyl, indolyl, pyridyl, pyrrolyl, pyrrolinyl, pyrazolyl, pyrazinyl, quinoxolyl, piperidinyl, morpholinyl, thiamorpholinyl, furyl, thienyl, triazolyl, thiazolyl, β-carbolinyl, tetrazolyl, thiazolidinyl, benzofuranoyl, thiamorpholinyl sulfone, benzoxazolyl, oxopiperidinyl, oxopyrroldinyl, oxoazepinyl, azepinyl, isoxazolyl, tetrahydropyranyl, tetrahydrofuranyl, thiadiazoyl, benzodioxolyl, thiophenyl, tetrahydrothiophenyl and sulfolanyl.
  • The terms “HIV protease” and “HIV aspartyl protease” are used interchangeably and refer to the aspartyl protease encoded by the human immunodeficiency virus type 1 or 2. In a preferred embodiment of this invention, these terms refer to the human immunodeficiency virus type 1 aspartyl protease.
  • The term “pharmaceutically effective amount” refers to an amount effective in treating HIV infection in a patient. The term “prophylactically effective amount” refers to an amount effective in preventing HIV infection in a patient. As used herein, the term “patient” refers to a mammal, including a human.
  • The term “pharmaceutically acceptable carrier or adjuvant” refers to a non-toxic carrier or adjuvant that may be administered to a patient, together with a compound of this invention, and which does not destroy the pharmacological activity thereof.
  • Pharmaceutically acceptable salts of the compounds of this invention include those derived from pharmaceutically acceptable inorganic and organic acids and bases. Examples of suitable acids include hydrochloric, hydrobromic, sulfuric, nitric, perchloric, fumaric, maleic, phosphoric, glycollic, lactic, salicylic, succinic, toluene-p-sulfonic, tartaric, acetic, citric, methanesulfonic, formic, benzoic, malonic, naphthalene-2-sulfonic and benzenesulfonic acids. Other acids, such as oxalic, while not in themselves pharmaceutically acceptable, may be employed in the preparation of salts useful as intermediates in obtaining the compounds of the invention and their pharmaceutically acceptable acid addition salts.
  • Salts derived from appropriate bases include alkali metal (e.g., sodium), alkaline earth metal (e.g., magnesium), ammonium and N—(C1-4 alkyl)4+ salts.
  • The term “thiocarbamates” refers to compounds containing the functional group N—SO2—O.
  • The compounds of this invention contain one or more asymmetric carbon atoms and thus occur as racemates and racemic mixtures, single enantiomers, diastereomeric mixtures and individual diastereomers. All such isomeric forms of these compounds are expressly included in the present invention. Each stereogenic carbon may be of the R or S configuration. The explicitly shown hydroxyl is also preferred to be syn to D, in the extended zigzag conformation between the nitrogens shown in compounds of formula I.
  • Combinations of substituents and variables envisioned by this invention are only those that result in the formation of stable compounds. The term “stable”, as used herein, refers to compounds which possess stability sufficient to allow manufacture and administration to a mammal by methods known in the art. Typically, such compounds are stable at a temperature of 40° C. or less, in the absence of moisture or other chemically reactive conditions, for at least a week.
  • The compounds of the present invention may be used in the form of salts derived from inorganic or organic acids. Included among such acid salts, for example, are the following: acetate, adipate, alginate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxy-yethanesulfonate, lactate, maleate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, oxalate, pamoate, pectinate, persulfate, 3-phenylpropionate, picrate, pivalate, propionate, succinate, tartrate, thiocyanate, tosylate and undecanoate.
  • This invention also envisions the quaternization of any basic nitrogen-containing groups of the compounds disclosed herein. The basic nitrogen can be quaternized with any agents known to those of ordinary skill in the art including, for example, lower alkyl halides, such as methyl, ethyl, propyl and butyl chloride, bromides and iodides; dialkyl sulfates including dimethyl, diethyl, dibutyl and diamyl sulfates; long chain halides such as decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides; and aralkyl halides including benzyl and phenethyl bromides. Water or oil-soluble or dispersible products may be obtained by such quaternization.
  • The novel sulfonamides of this invention are those of formula I:
  • Figure US20100124543A1-20100520-C00003
  • wherein:
  • A is selected from H; Ht; —R1—Ht; —R1—C1-C6 alkyl, which is optionally substituted with one or more groups independently selected from hydroxy, C1-C4 alkoxy, Ht, —O-Ht, —NR2—CO—N(R2)2 or —CO—N(R2)2; —R1—C2-C6 alkenyl, which is optionally substituted with one or more groups independently selected from hydroxy, C1-C4 alkoxy, Ht, —O-Ht, —NR2—CO—N(R2)2 or —CO—N(R2)2; or R7;
  • each R1 is independently selected from —C(O)—, —S(O)2—, —C(O)—C(O)—, —O—C(O)—, —O—S(O)2, —NR2—S(O)2—, —NR2—C(O)— or —NR2—C(O)—C(O)—;
  • each Ht is independently selected from C3-C7 cycloalkyl; C5-C7 cycloalkenyl; C6-C10 aryl; or a 5-7 membered saturated or unsaturated heterocycle, containing one or more heteroatoms selected from N, N(R2), O, S and S(O)n; wherein said aryl or said heterocycle is optionally fused to Q; and wherein any member of said Ht is optionally substituted with one or more substituents independently selected from oxo, —OR2, SR2, —R2, —N(R2)(R2), —R2—OH, —CN, —CO2R2, —C(O)—N(R2)2, —S(O)2—N(R2)2, —N(R2)—C(O)—R2, —C(O)—R2, —S(O)n—R2, —OCF3, —S(O)n-Q, methylenedioxy, —N(R2)—S(O)2 (R2), halo, —CF3, —NO2, Q, —OQ, —OR7, —SR7, —R7, —N(R2)(R7) or —N(R7)2;
  • each R2 is independently selected from H, or C1-C4 alkyl optionally substituted with Q;
  • B, when present, is —N(R2)—C(R3)2—C(O)—;
  • each x is independently 0 or 1;
  • each R3 is independently selected from H, Ht, C1-C6 alkyl, C2-C6 alkenyl, C3-C6 cycloalkyl or C5-C6 cycloalkenyl; wherein any member of said R3, except H, is optionally substituted with one or more substituents selected from —OR2, —C(O)—NH—R2, —S(O), —N(R2)(R2), Ht, —CN, —SR2, —CO2R2, NR2—C(O)—R2;
  • each n is independently 1 or 2;
  • G, when present, is selected from H, R7 or C1-C4 alkyl, or, when G is C1-C4 alkyl, G and R7 are bound to one another either directly or through a C1-C3 linker to form a heterocyclic ring; or
  • when G is not present (i.e., when x in (G)x is 0), then the nitrogen to which G is attached is bound directly to the R7 group in —OR7 with the concomitant displacement of one -ZM group from R7;
  • D and D′ are independently selected from Q; C1-C6 alkyl, which is optionally substituted with one or more groups selected from C3-C6 cycloalkyl, —OR2, —R3, —O-Q or Q; C2-C4 alkenyl, which is optionally substituted with one or more groups selected from C3-C6 cycloalkyl, —OR2, —R3, —O-Q or Q; C3-C6 cycloalkyl, which is optionally substituted with or fused to Q; or C5-C6 cycloalkenyl, which is optionally substituted with or fused to Q;
  • each Q is independently selected from a 3-7 membered saturated, partially saturated or unsaturated carbocyclic ring system; or a 5-7 membered saturated, partially saturated or unsaturated heterocyclic ring containing one or more heteroatoms selected from O, N, S, S(O)n or N(R2); wherein Q is optionally substituted with one or more groups selected from oxo, —OR2, —R2, —N(R2)2, —N(R2)—C(O)—R2, —R2—OH, —CN, —CO2R2, —C(O)—N(R2)2, halo or —CF3;
  • E is selected from Ht; O-Ht; Ht-Ht; —O—R3; —N(R2)(R3); C1-C6 alkyl, which is optionally substituted with one or more groups selected from R4 or Ht; C2-C6 alkenyl, which is optionally substituted with one or more groups selected from R4 or Ht; C3-C6 saturated carbocycle, which is optionally substituted with one or more groups selected from R4 or Ht; or C5-C6 unsaturated carbocycle, which is optionally substituted with one or more groups selected from R4 or Ht;
  • each R4 is independently selected from —OR2, —SR2, —C(O)—NHR2, —S(O)2—NHR2, halo, —NR2—C(O)—R2, —N(R2)2 or —CN;
  • each R7 is independently selected from
  • Figure US20100124543A1-20100520-C00004
  • wherein each M is independently selected from H, Li, Na, K, Mg, Ca, Ba, —N(R2)4, C1-C12-alkyl, C2-C12-alkenyl, or —R6; wherein 1 to 4 —CH2 radicals of the alkyl or alkenyl group, other than the —CH2 that is bound to Z, is optionally replaced by a heteroatom group selected from O, S, S(O), S(O2), or N(R2); and wherein any hydrogen in said alkyl, alkenyl or R6 is optionally replaced with a substituent selected from oxo, —OR2, —R2, N(R2)2, N(R2)3, R2OH, —CN, —CO2R2, —C(O)—N(R2)2, S(O)2—N(R2)2, N(R2)—C(O)—R2, C(O)R2, —S(O)n—R2, OCF3, —S(O)n—R6, N(R2)—S(O)2 (R2), halo, —CF3, or —NO2;
  • M′ is H, C1-C12-alkyl, C2-C12-alkenyl, or —R6; wherein 1 to 4 —CH2 radicals of the alkyl or alkenyl group is optionally replaced by a heteroatom group selected from O, S, S(O), S(O2), or N(R2); and wherein any hydrogen in said alkyl, alkenyl or R6 is optionally replaced with a substituent selected from oxo, —OR2, —R2, —N(R2)2, N(R2)3, —R2OH, —CN, —CO2R2, —C(O)—N(R2)2, —S(O)2—N(R2)2, —N(R2)—C(O)—R2, —C(O)R2, —S(O)n—R2, —OCF3, —S(O)n—R6, —N(R2)—S(O)2 (R2), halo, —CF3, or —NO2;
  • Z is CH2, O, S, N(R2)2, or, when M is not present, H.
  • Y is P or S;
  • X is O or S; and
  • R9 is C(R2)2, O or N(R2); and wherein when Y is S, Z is not S; and
  • R6 is a 5-6 membered saturated, partially saturated or unsaturated carbocyclic or heterocyclic ring system, or an 8-10 membered saturated, partially saturated or unsaturated bicyclic ring system; wherein any of said heterocyclic ring systems contains one or more heteroatoms selected from O, N, S, S(O)n or N(R2); and wherein any of said ring systems optionally contains 1 to 4 substituents independently selected from OH, C1-C4 alkyl, O—C1-C4 alkyl or O—C(O)—C1-C4 alkyl.
  • Preferably, at least one R7 is selected from:
  • Figure US20100124543A1-20100520-C00005
    Figure US20100124543A1-20100520-C00006
  • It will be understood by those of skill in the art that component M or M′ in the formulae set forth herein will have either a covalent, a covalent/zwitterionic, or an ionic association with either Z or R9 depending upon the actual choice for M or M′. When M or M′ is hydrogen, alkyl, alkenyl, or R6, M or M′ is covalently bound to R9 or Z. If M is a mono- or bivalent metal or other charged species (i.e., NH4 +), there is an ionic interaction between M and Z and the resulting compound is a salt.
  • When x is 0 in (M)x, Z may be a charged species. When that occurs, the other M may be oppositely charged to produce a 0 net charge on the molecule. Alternatively, the counter ion may located elsewhere in the molecule.
  • Except where expressly provided to the contrary, as used herein, the definitions of variables A, R1-R4, R6-R9, Ht, B, x, n, D, D′, M, Q, X, Y, Z and E are to be taken as they are defined above for the compounds of formula I.
  • According to a preferred embodiment, the compounds of this invention are those represented by formulas XXII, XXIII or XXXI:
  • Figure US20100124543A1-20100520-C00007
  • wherein A, R3, R7, Ht, D, D′, x, E are as defined above for compounds of formula I. For ease of reference, the two R3 moieties present in formula XXXI have been labeled R3 and R3′.
  • For compounds of formula XXII, more preferred compounds are those wherein:
  • A is selected from 3-tetrahydrofuryl-O—C(O)—, 3-(1,5-dioxane)-O—C(O)—, or 3-hydroxy-hexahydrofura[2,3-b]-furanyl-O—C(O)—;
  • D′ is C1-C4 alkyl which is optionally substituted with one or more groups selected from the group consisting of C3-C6 cycloalkyl, —OR2, —R3, —O-Q and Q;
  • E is C6-C10 aryl optionally substituted with one or more substituents selected from oxo, —OR2, SR2, —R2, —N(R2)2, —R2—OH, —CN, —CO2R2, —C(O)—N(R2)2, —S(O)2—N(R2)2, —N(R2)—C(O)—R2, —C(O)—R2, —S(O), —R2, —OCF3, —S(O)n-Q, methylenedioxy, —N(R2)—S(O)2 (R2), halo, —CF3, —NO2, Q, —OQ, —OR7, —SR7, —R7, —N(R2)(R7) or —N(R7)2; or a 5-membered heterocyclic ring containing one S and optionally containing N as an additional heteroatom, wherein said heterocyclic ring is optionally substituted with one to two groups independently selected from —CH3, R4, or Ht.
  • Ht, insofar as it is defined as part of R3, is defined as above except for the exclusion of heterocycles; and
  • all other variables are as defined for formula I.
  • Even more preferred are compounds of formula XXII, wherein A is 3-tetrahydrofuryl-O—C(O)—; G is hydrogen; D′ is isobutyl; E is phenyl substituted with N(R7)2; each M is independently selected from H, Li, Na, K, Mg, Ca, Ba, C1-C4 alkyl or —N(R2)4; and each M′ is H or C1-C4 alkyl.
  • Another preferred embodiment for the formula XXII compounds are those wherein:
  • E is a 5-membered heterocyclic ring containing one S and optionally containing N as an additional heteroatom, wherein said heterocyclic ring is optionally substituted with one to two groups independently selected from —CH3, R4, or Ht; and
  • all other variables are as defined for formula I.
  • Even more preferred are any of the formula XXII compounds set forth above, wherein R7 in —OR7 is —PO(OM)2 or C(O)CH2OCH2CH2OCH2CH2OCH3 and both R7 in —N(R7)2 are H, wherein M is H, Li, Na, K or C1-C4 alkyl; or wherein R7 in —OR7 is C(O)CH2OCH2CH2OCH3, one R7 in —N(R7)2 is C(O)CH2OCH2CH2OCH3 and the other is H.
  • The most preferred compound of formula XXII has the structure:
  • Figure US20100124543A1-20100520-C00008
  • For compounds of formula XXIII, most preferred compounds are those wherein:
  • R3 is C1-C6 alkyl, C2-C6 alkenyl, C5-C6 cycloalkyl, C5-C6 cycloalkenyl or a 5-6 membered saturated or unsaturated heterocycle, wherein any member of said R3 may be optionally substituted with one or more substituents selected from the group consisting of —OR2, —C(O)—NH—R2, —S(O)nN(R2)(R2), Ht, —CN, —SR2, —C(O)2R2 and NR2—C(O)—R2; and
  • D′ is C1-C3 alkyl or C3 alkenyl, wherein said alkyl or alkenyl may optionally be substituted with one or more groups selected from the group consisting of C3-C6 cycloalkyl, —OR2, —O-Q and Q (with all other variables being defined as above for compounds of formula I).
  • Even more preferred are compounds of formula XXIII described above, wherein R7 is —PO(OM)2 or —C(O)-M′.
  • For compounds of formula XXXI, most preferred compounds are those wherein A is R1—Ht, each R3 is independently C1-C6 alkyl which may be optionally substituted with a substituent selected from the group consisting of —OR2, —C(O)—NH—R2, —S(O)nN(R2)(R2), Ht, —CN, —SR2, —CO2R2 or —NR2—C(O)—R2; and D′ is C1-C4 alkyl, which may be optionally substituted with a group selected from the group consisting of C3-C6 cycloalkyl, —OR2, —O-Q; and E is Ht, Ht-Ht and —NR2R3.
  • Even more preferred are those compounds of formula XXXI described above wherein R7 is —PO(OM)2 or —C(O)-M′.
  • TABLE I
    Figure US20100124543A1-20100520-C00009
    CMPD R7 W
    198
    Figure US20100124543A1-20100520-C00010
    —NO2
    199
    Figure US20100124543A1-20100520-C00011
    —NH2
    200
    Figure US20100124543A1-20100520-C00012
    —NH2
    201
    Figure US20100124543A1-20100520-C00013
    —NH2
    202
    Figure US20100124543A1-20100520-C00014
    —NH2
    203
    Figure US20100124543A1-20100520-C00015
    —NH2
    204
    Figure US20100124543A1-20100520-C00016
    —NH2
    205
    Figure US20100124543A1-20100520-C00017
    —NH2
    206
    Figure US20100124543A1-20100520-C00018
    —NH2
    207
    Figure US20100124543A1-20100520-C00019
    —NH2
    208
    Figure US20100124543A1-20100520-C00020
    —NO2
    209
    Figure US20100124543A1-20100520-C00021
    —NO2
    210
    Figure US20100124543A1-20100520-C00022
    —NH2
    211
    Figure US20100124543A1-20100520-C00023
    —NH2
    212
    Figure US20100124543A1-20100520-C00024
    —NH2
    213
    Figure US20100124543A1-20100520-C00025
    —NH2
    214
    Figure US20100124543A1-20100520-C00026
    —NH2
    215
    Figure US20100124543A1-20100520-C00027
    —NH2
    216
    Figure US20100124543A1-20100520-C00028
    —NH2
    217
    Figure US20100124543A1-20100520-C00029
    —NH2
    219 H
    Figure US20100124543A1-20100520-C00030
    220 H
    Figure US20100124543A1-20100520-C00031
    221 H
    Figure US20100124543A1-20100520-C00032
    222 H
    Figure US20100124543A1-20100520-C00033
    223 H
    Figure US20100124543A1-20100520-C00034
    224 H
    Figure US20100124543A1-20100520-C00035
    225
    Figure US20100124543A1-20100520-C00036
    Figure US20100124543A1-20100520-C00037
    226
    Figure US20100124543A1-20100520-C00038
    —NO2
    227
    Figure US20100124543A1-20100520-C00039
    —NO2
    228
    Figure US20100124543A1-20100520-C00040
    —NH2
    229
    Figure US20100124543A1-20100520-C00041
    —NH2
    230 H
    Figure US20100124543A1-20100520-C00042
    231
    Figure US20100124543A1-20100520-C00043
    Figure US20100124543A1-20100520-C00044
    237
    Figure US20100124543A1-20100520-C00045
    —NO2
    238
    Figure US20100124543A1-20100520-C00046
    —NO2
    239 —SO3H —NO2
    240 —SO3H —NH2
    241
    Figure US20100124543A1-20100520-C00047
    —NO2
    242
    Figure US20100124543A1-20100520-C00048
    —NH2
    245
    Figure US20100124543A1-20100520-C00049
    —NH2
    246
    Figure US20100124543A1-20100520-C00050
    —NH2
    247
    Figure US20100124543A1-20100520-C00051
    —NH2
    248
    Figure US20100124543A1-20100520-C00052
    —NH2
    249
    Figure US20100124543A1-20100520-C00053
    —NH2
    250
    Figure US20100124543A1-20100520-C00054
    —NH2
    251
    Figure US20100124543A1-20100520-C00055
    —NH2
    252
    Figure US20100124543A1-20100520-C00056
    —NH2
    253
    Figure US20100124543A1-20100520-C00057
    —NH2
    254
    Figure US20100124543A1-20100520-C00058
    —NH2
    255 H —NH—CHO
    256 H
    Figure US20100124543A1-20100520-C00059
    257 H
    Figure US20100124543A1-20100520-C00060
    258 H
    Figure US20100124543A1-20100520-C00061
    259 H
    Figure US20100124543A1-20100520-C00062
    260 H
    Figure US20100124543A1-20100520-C00063
    261
    Figure US20100124543A1-20100520-C00064
    Figure US20100124543A1-20100520-C00065
    262
    Figure US20100124543A1-20100520-C00066
    Figure US20100124543A1-20100520-C00067
    263
    Figure US20100124543A1-20100520-C00068
    Figure US20100124543A1-20100520-C00069
    264 PO3K2 —NH2
    265 PO3Ca —NH2
    266 PO3Mg —NH2
    267
    Figure US20100124543A1-20100520-C00070
    —NH2
    308
    Figure US20100124543A1-20100520-C00071
    —NH2
    402 H
    Figure US20100124543A1-20100520-C00072
    403 H
    Figure US20100124543A1-20100520-C00073
    404 H
    Figure US20100124543A1-20100520-C00074
    405 H
    Figure US20100124543A1-20100520-C00075
    406 H
    Figure US20100124543A1-20100520-C00076
    407 H
    Figure US20100124543A1-20100520-C00077
    408
    Figure US20100124543A1-20100520-C00078
    —NH2
  • TABLE II
    Figure US20100124543A1-20100520-C00079
    COMPOUND A R7
    232
    Figure US20100124543A1-20100520-C00080
    Figure US20100124543A1-20100520-C00081
    233 H
    Figure US20100124543A1-20100520-C00082
    234
    Figure US20100124543A1-20100520-C00083
    H
    235
    Figure US20100124543A1-20100520-C00084
    Figure US20100124543A1-20100520-C00085
    236
    Figure US20100124543A1-20100520-C00086
    Figure US20100124543A1-20100520-C00087
  • TABLE III
    Figure US20100124543A1-20100520-C00088
    COMPOUND R7 W
    243
    Figure US20100124543A1-20100520-C00089
    —NO2
    244
    Figure US20100124543A1-20100520-C00090
    —NH2
    400
    Figure US20100124543A1-20100520-C00091
    —NO2
    401
    Figure US20100124543A1-20100520-C00092
    —NH2
  • According to another embodiment, the invention provides compounds of the following formulae:
  • Figure US20100124543A1-20100520-C00093
    Figure US20100124543A1-20100520-C00094
  • wherein, in compound 1005, when R7 is PO3M, (G)x is not H; and wherein R10 is selected from isopropoyl or cyclopentyl; R11 is selected from NHR7 or OR; and x, R7 and G are as defined above.
  • The prodrugs of the present invention may be synthesized using conventional synthetic techniques. U.S. Pat. No. 5,585,397 discloses the synthesis of compounds of formula:
  • Figure US20100124543A1-20100520-C00095
  • wherein A, B, n, D, D′, and E are as defined above. Prodrugs of formula (I) of the present invention can be readily synthesized from the '397 compounds using conventional techniques. One of skill in the art would be well aware of conventional synthetic reagents to convert the —OH group of the '397 compounds to a desired —OR7 functionality of the present invention, wherein R7 is as defined above. The relative ease with which the compounds of this invention can be synthesized represents an enormous advantage in the large scale production of these compounds.
  • For example, VX-478, a compound disclosed in the '397 patent, can be readily converted to the corresponding bis-phosphate ester derivative, as shown below:
  • Figure US20100124543A1-20100520-C00096
  • Alternatively, if the monophosphate ester of VX-478 is desired, then the synthetic scheme can be readily adapted by beginning with the 4-nitrophenyl derivative of VX-478, as shown below:
  • Figure US20100124543A1-20100520-C00097
  • Examples of specific compounds in addition to VX-478 which may be converted to the prodrugs of this invention by similar techniques (and the syntheses of those intermediates to the compounds of the present invention) are disclosed in WO 94/05639 and WO 96/33184, the disclosures of which are herein incorporated by reference.
  • Pharmaceutically acceptable salts of the compounds of the present invention may be readily prepared using known techniques. For example, the disodium salt of the mono-phosphate ester shown above can be prepared as shown below:
  • Figure US20100124543A1-20100520-C00098
  • The compounds of this invention may be modified by appending appropriate functionalities to enhance selective biological properties. Such modifications are known in the art and include those which increase biological penetration into a given biological system (e.g., blood, lymphatic system, central nervous system), increase oral availability, increase solubility to allow administration by injection, alter metabolism and alter rate of excretion.
  • Without being bound by theory, we believe that two different mechanisms are involved in converting the prodrugs of this invention into the active drug, depending upon the structure of the prodrug. The first mechanism involves the enzymatic or chemical transformation of the prodrug species into the active form. The second mechanism involves the enzymatic or chemical cleavage of a functionality on the prodrug to produce the active compound.
  • The chemical or enzymatic transformation can involve to transfer of a functional group (i.e., R7) from one heteroatom within the molecule to another heteroatom. This transfer is demonstrated in the chemical reactions shown below:
  • Figure US20100124543A1-20100520-C00099
  • The cleavage mechanism is demonstrated by the reaction below where a phosphate ester-containing prodrug is converted into the active form of the drug by removal of the phosphate group.
  • Figure US20100124543A1-20100520-C00100
  • These protease inhibitors and their utility as inhibitors of aspartyl proteases are described in U.S. Pat. No. 5,585,397, the disclosure of which is incorporated herein by reference.
  • The prodrugs of the present invention are characterized by unexpectedly high aqueous solubility. This solubility facilitates administration of higher doses of the prodrug, resulting in a greater drug load per unit dosage. The prodrugs of the present invention are also characterized by facile hydrolytic cleavage to release the active aspartyl protease inhibitor in vivo. The high aqueous solubility and the facile in vivo metabolism result in a greater bioavailability of the drug. As a result, the pill burden on a patient is significantly reduced.
  • The prodrugs of this invention may be employed in a conventional manner for the treatment of viruses, such as HIV and HTLV, which depend on aspartyl proteases for obligatory events in their life cycle. Such methods of treatment, their dosage levels and requirements may be selected by those of ordinary skill in the art from available methods and techniques. For example, a prodrug of this invention may be combined with a pharmaceutically acceptable adjuvant for administration to a virally-infected patient in a pharmaceutically acceptable manner and in an amount effective to lessen the severity of the viral infection.
  • Alternatively, the prodrugs of this invention may be used in vaccines and methods for protecting individuals against viral infection over an extended period of time. The prodrugs may be employed in such vaccines either alone or together with other compounds of this invention in a manner consistent with the conventional utilization of protease inhibitors in vaccines. For example, a prodrug of this invention may be combined with pharmaceutically acceptable adjuvants conventionally employed in vaccines and administered in prophylactically effective amounts to protect individuals over an extended period time against HIV infection. As such, the novel protease inhibitors of this invention can be administered as agents for treating or preventing HIV infection in a mammal.
  • The prodrugs of this invention may be administered to a healthy or HIV-infected patient either as a single agent or in combination with other anti-viral agents which interfere with the replication cycle of HIV. By administering the compounds of this invention with other anti-viral agents which target different events in the viral life cycle, the therapeutic effect of these compounds is potentiated. For instance, the co-administered anti-viral agent can be one which targets early events in the life cycle of the virus, such as cell entry, reverse transcription and viral DNA integration into cellular DNA. Anti-HIV agents targeting such early life cycle events include, didanosine (ddI), alcitabine (ddC), d4T, zidovudine (AZT), polysulfated polysaccharides, sT4 (soluble CD4), ganiclovir, dideoxycytidine, trisodium phosphonoformate, eflornithine, ribavirin, acyclovir, alpha interferon and trimenotrexate. Additionally, non-nucleoside inhibitors of reverse transcriptase, such as TIBO or nevirapine, may be used to potentiate the effect of the compounds of this invention, as may viral uncoating inhibitors, inhibitors of trans-activating proteins such as tat or rev, or inhibitors of the viral integrase.
  • Combination therapies according to this invention exert a synergistic effect in inhibiting HIV replication because each component agent of the combination acts on a different site of HIV replication. The use of such combinations also advantageously reduces the dosage of a given conventional anti-retroviral agent which would be required for a desired therapeutic or prophylactic effect as compared to when that agent is administered as a monotherapy. These combinations may reduce or eliminate the side effects of conventional single anti-retroviral agent therapies while not interfering with the anti-retroviral activity of those agents. These combinations reduce potential of resistance to single agent therapies, while minimizing any associated toxicity. These combinations may also increase the efficacy of the conventional agent without increasing the associated toxicity. In particular, we have discovered that these prodrugs act synergistically in preventing the replication of HIV in human T cells. Preferred combination therapies include the administration of a prodrug of this invention with AZT, ddI, ddC or d4T.
  • Alternatively, the prodrugs of this invention may also be co-administered with other HIV protease inhibitors such as Ro 31-8959 (Roche), L-735,524 (Merck), XM 323 (Du-Pont Merck) and A-80,987 (Abbott) to increase the effect of therapy or prophylaxis against various viral mutants or members of other HIV quasi species.
  • We prefer administering the prodrugs of this invention as single agents or in combination with retroviral reverse transcriptase inhibitors, such as derivatives of AZT, or other HIV aspartyl protease inhibitors. We believe that the co-administration of the compounds of this invention with retroviral reverse transcriptase inhibitors or HIV aspartyl protease inhibitors may exert a substantial synergistic effect, thereby preventing, substantially reducing, or completely eliminating viral infectivity and its associated symptoms.
  • The prodrugs of this invention can also be administered in combination with immunomodulators (e.g., bropirimine, anti-human alpha interferon antibody, IL-2, GM-CSF, methionine enkephalin, interferon alpha, diethyldithiocarbamate, tumor necrosis factor, naltrexone and rEPO); and antibiotics (e.g., pentamidine isethiorate) to prevent or combat infection and disease associated with HIV infections, such as AIDS and ARC.
  • When the prodrugs of this invention are administered in combination therapies with other agents, they may be administered sequentially or concurrently to the patient. Alternatively, pharmaceutical or prophylactic compositions according to this invention may be comprised of a combination of a prodrug of this invention and another therapeutic or prophylactic agent.
  • Although this invention focuses on the use of the prodrugs disclosed herein for preventing and treating HIV infection, the compounds of this invention can also be used as inhibitory agents for other viruses which depend on similar aspartyl proteases for obligatory events in their life cycle. These viruses include, as well as other AIDS-like diseases caused by retroviruses, such as simian immunodeficiency viruses, but are not limited to, HTLV-I and HTLV-II. In addition, the compounds of this invention may also be used to inhibit other aspartyl proteases, and in particular, other human aspartyl proteases, including renin and aspartyl proteases that process endothelin precursors.
  • Pharmaceutical compositions of this invention comprise any of the compounds of the present invention, and pharmaceutically acceptable salts thereof, with any pharmaceutically acceptable carrier, adjuvant or vehicle. Pharmaceutically acceptable carriers, adjuvants and vehicles that may be used in the pharmaceutical compositions of this invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol and wool fat.
  • The pharmaceutical compositions of this invention may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir. We prefer oral administration or administration by injection. The pharmaceutical compositions of this invention may contain any conventional non-toxic pharmaceutically-acceptable carriers, adjuvants or vehicles. The term parenteral as used herein includes subcutaneous, intracutaneous, intravenous, intramuscular, intra-articular, intrasynovial, intrasternal, intrathecal, intralesional and intracranial injection or infusion techniques.
  • The pharmaceutical compositions may be in the form of a sterile injectable preparation, for example, as a sterile injectable aqueous or oleaginous suspension. This suspension may be formulated according to techniques known in the art using suitable dispersing or wetting agents (such as, for example, Tween 80) and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic 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 mannitol, 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. Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions. These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant such as Ph. Helv or a similar alcohol.
  • The pharmaceutical compositions of this invention may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, and aqueous suspensions and solutions. In the case of tablets for oral use, carriers which are commonly used include lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added. For oral administration in a capsule form, useful diluents include lactose and dried corn starch. When aqueous suspensions are administered orally, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening and/or flavoring and/or coloring agents may be added.
  • The pharmaceutical compositions of this invention may also be administered in the form of suppositories for rectal administration. These compositions can be prepared by mixing a compound of this invention with a suitable non-irritating excipient which is solid at room temperature but liquid at the rectal temperature and therefore will melt in the rectum to release the active components. Such materials include, but are not limited to, cocoa butter, beeswax and polyethylene glycols.
  • Topical administration of the pharmaceutical compositions of this invention is especially useful when the desired treatment involves areas or organs readily accessible by topical application. For application topically to the skin, the pharmaceutical composition should be formulated with a suitable ointment containing the active components suspended or dissolved in a carrier. Carriers for topical administration of the compounds of this invention include, but are not limited to, mineral oil, liquid petroleum, white petroleum, propylene glycol, polyoxyethylene polyoxypropylene compound, emulsifying wax and water. Alternatively, the pharmaceutical composition can be formulated with a suitable lotion or cream containing the active compound suspended or dissolved in a carrier. Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water. The pharmaceutical compositions of this invention may also be topically applied to the lower intestinal tract by rectal suppository formulation or in a suitable enema formulation. Topically-transdermal patches are also included in this invention.
  • The pharmaceutical compositions of this invention may be administered by nasal aerosol or inhalation. Such compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other solubilizing or dispersing agents known in the art.
  • Dosage levels of between about 0.01 and about 100 mg/kg body weight per day, preferably between about 0.5 and about 50 mg/kg body weight per day of the active ingredient compound are useful in the prevention and treatment of viral infection, including HIV infection. Typically, the pharmaceutical compositions of this invention will be administered from about 1 to about 5 times per day or alternatively, as a continuous infusion. Such administration can be used as a chronic or acute therapy. The amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. A typical preparation will contain from about 5% to about 95% active compound (w/w). Preferably, such preparations contain from about 20% to about 80% active compound.
  • Upon improvement of a patient's condition, a maintenance dose of a compound, composition or combination of this invention may be administered, if necessary. Subsequently, the dosage or frequency of administration, or both, may be reduced, as a function of the symptoms, to a level at which the improved condition is retained when the symptoms have been alleviated to the desired level, treatment should cease. Patients may, however, require intermittent treatment on a long-term basis upon any recurrence of disease symptoms.
  • As the skilled artisan will appreciate, lower or higher doses than those recited above may be required. Specific dosage and treatment regimens for any particular patient will depend upon a variety of factors, including the activity of the specific compound employed, the age, body weight, general health status, sex, diet, time of administration, rate of excretion, drug combination, the severity and course of the infection, the patient's disposition to the infection and the judgment of the treating physician.
  • In order that this invention be more fully understood, the following examples are set forth. These examples are for the purpose of illustration only and are not to be construed as limiting the scope of the invention in any way.
  • Example 1 General Conditions
  • (A) Analytical HPLC 0-100% B/30 min, 1.5 mL/min, A=0.1% TFA in water, B=0.1% TFA in acetonitrile. Detection at 254 and 220 nm, C18 reverse phase Vydac, t0=2.4 min.
  • (B) ⅓ v/v EtOAc/hexane
  • (C) ½ v/v EtOAc/hexane
  • (D) Analytical HPLC 0-100% B/10 min, 1.5 mL/min, A=0.1% TFA in water, B=0.1% TFA in acetonitrile. Detection at 254 and 220 nm, C18 reverse phase Vydac, t0=2.4 min.
  • Figure US20100124543A1-20100520-C00101
  • A mixture of 2.0 g (3.7 mMol) of 197 and 3.0 g (16 mMol) of di-p-nitrophenyl carbonate in 10 ml of dimethylformamide was treated at 25° with 4 ml (4 mMol) of P4-phosphazene base (Fluka, 1M in hexane). The mixture was stirred for 6 h at 25° until all of the starting alcohol was consumed. The reaction mixture was partitioned between ethyl acetate and 1N hydrochloric acid. The organic layer was washed with 1N sodium hydroxide and brine, dried over magnesium sulfate and concentrated in vacuo. Titration with dichloromethane gave the desired mixed carbonate (1.2 g crop 1 and 0.6 g crop 2) as a fine powder. Combined yield: 69%. Rf=0.13 (1/3 EtOAc/hexane, conditions B), Rf=0.40 (1/2 EtOAc/hexane, conditions C), tHPLC=23.83 min (A), MS (ES+) 701 (M+1).
  • 1H-NMR (CDCl3): 0.82 (6H, dd), 1.9 (2H, m), 2.15 (1H, m), 2.8 (1H, m), 3.0 (4H, m), 3.5 (2H, m), 3.6 (1H, m), 3.8 (4H, m), 4.3 (1H, bs), 4.8 (1H, m), 5.17 (2H, m), 7.7 (7H, m), 7.95 (2H, d), 8.35 (4H, m).
  • 13C (CDCl3): 155.2 152.2, 149.9, 145.6, 135.9, +129.0, +128.8, +128.5, +127.2, +125.4, +124.4, +121.8, +78.1, +75.8, −73.1, −66.9, −56.5, +52.7, −48.2, −35.9, −35.9, 32.6, −+26.4, +19.9, +19.8.
  • Example 2
  • Figure US20100124543A1-20100520-C00102
  • To 0.20 g (0.286 mM) of 198 dissolved in 3 ml of THF was added 0.11 g (1.14 mM) of 1-Methyl-piperidine and the mixture was stirred overnight at room temperature (“rt”). All the solvents were then evaporated and the solid residue partitioned between EtOAc and water. The volatiles were removed and, where appropriate, the residue was treated with 1:1 TFA/DCM over 30 min at rt to remove the Boc protecting group. The product was dissolved in 0.25 ml TFA and 1.5 ml THF. Hydrogenolysis for 10 hours in presence of 30 mg of 10% Pd/C gave the desired compound. The final purification was on preparative reversed phase C18 using conditions Example 1, except that the flow rate was 18 ml/min.
  • C,H,N: calc: 49.27, 5.57, 8.25, found 49.15, 5.76, 8.29. C33H45N5O7S1.1.9CF3COOH
  • LC/MS (ES+) 632 (M+1) 1 peak at 4.71 min
  • Analytical HPLC(A) t=N/A min
  • 1H, 0.71 (3H, d), 0.74 (3H, d), 1.80 (2H, m), 2.03 (1H, m), 2.63 (2H, m), 2.74 (1H, m), 2.82 (3H, s), 2.92 (2H, m), 3.20 (4H, m), 3.42 (3H, m), 3.62 (2H, m), 3.75 (1H, m), 4.05 (3H, m), 4.97 (2H, m), 6.2 (1H, bs), 6.60 (2H, m), 7.22 (5H, m), 7.40 (3H, m),
  • 13C (DMSO): 156.4, 154.0, 153.8, 138.8, 129.6, 129.5, 128.3, 126.5, 123.7, 112.7, 74.8, 72.9, 66.7, 58.2, 54.0, 53.1, 49.3, 42.3, 40.8, 36.0, 33.3, 25.8, 20.4, 20.3
  • Example 3
  • Figure US20100124543A1-20100520-C00103
  • The synthesis of compound 200 from compound 198 was carried as described in Example 1, except that N,N-dimethyl-aminoethanol was used in place of di-p-nitrophenyl carbonate.
  • 1HNMR (acetone-d6): 0.82 (6H, dd), 1.83 (2H, m), 2.07 (1H, m), 2.64 (2H, m), 2.82 (6H, s), 2.90 (2H, m), 3.19 (1H, m), 3.38 (4H, m), 3.63 (2H, m), 3.76 (1H, m), 4.17 (2YH, m), 4.40 (1H, m), 4.56 (1H, m), 4.96 (1H, m), 5.06 (1H, m), 6.06 (1H, d), 6.68 (2H, d), 7.23 (5H, m), 7.47 (2H, d).
  • 13CNMR (acetone d6): 20.2, 20.3, 27.5, 33.4, 35.6, 43.8, 50.1, 54.2, 56.4, 58.5, 63.1, 67.4, 73.6, 76.2, 79.9, 114.2, 118.3, 127.4, 129.2, 130.1, 130.3, 139.3, 153.4, 157.0.
  • LC/MS: 1 peak, 621 (MH+).
  • Example 4
  • Figure US20100124543A1-20100520-C00104
  • The synthesis of compound 201 from compound 198 was carried as described in Example 1, except that N-acetyl-ethylenediamine was used in place of di-p-nitrophenyl carbonate.
  • C,H,N: calc: 49.66, 5.64, 8.83, found 49.76, 5.98, 8.93. C30H43N5O8S1.1.4CF3COOH.
  • LC/MS (ES+) 634 (M+1) 1 peak at 5.08 min.
  • Analytical HPLC(A) t=15.92 min.
  • 1H: d-3 acetonitrile: 0.88 (6H, dd), 1.92 (3H, s), 1.94 (2H, m), 2.17 (1H, m), 2.72 (2H, m), 2.96 (2H, m), 3.07 (3H, m), 3.29 (1H, m), 3.42 (3H, m), 3.69 (1H, m), 3.77 (1H, m), 3.82 (1H, m), 4.133 (1H, m), 4.40 (1H, bs), 5.05 (2H, m), 5.80 (1H, m), 6.10 (1H, d), 6.78 (2H, d), 6.83 (1H, bs), 7.28 (5H, m), 7.58 (2H, d).
  • 13C (d3-acetonitrile): 157.1, 157.0, 153.2, 139.6, +130.3, +130.2, +129.2, +127.2, 126.2, +114.2, +76.0, +75.4, −73.6, −67.4, −58.2, +54.9, −50.2, −41.6, −39.8, −35.9, −33.4, +27.3, +23.1, +20.4, +20.2.
  • Example 5
  • Figure US20100124543A1-20100520-C00105
  • The synthesis of compound 202 from compound 198 was carried as described in Example 1, except that mono N-Boc-piperazine was used in place of di-p-nitrophenyl carbonate.
  • C,H,N: calc: 48.28, 5.68, 8.41, found 48.28, 5.36, 8.28. C30H43N5O7S1×2 CF3COOH
  • LC/MS (ES+) 618 (M+1) 1 peak at 4.36 min.
  • Analytical HPLC(A) t=14.84 min.
  • 1H: d6-DMSO: 0.72 (3H, d), 0.77 (3H, d), 1.78 (2H, m), 2.09 (1H, m), 2.64 (2H, m), 2.73 (1H, m), 2.80 (1H, m), 3.08 (4H, m), 3.32 (2H, m), 3.41 (1H, m), 3.50 (4H, m), 3.54 (1H, m), 3.63 (1H, m), 3.70 (1H, m), 3.98 (1H, m), 4.89 (1H, m), 4.97 (1H, m), 6.61 (2H, d), 7.23 (5H, m), 7.42 (3H, m), 8.88 (2H, bs).
  • 13C: (DMSO): 155.7, 153.6, 153.0, 138.4, +129.1, +129.0, +128.1, +126.1, 123.2, +112.7, +75.2, +74.4, −72.5, −66.2, −56.9, +53.1, −48.8, −42.5, −40.8, −35.0, −32.2, +26.2, +20.0, +19.8.
  • Example 6
  • Figure US20100124543A1-20100520-C00106
  • The synthesis of compound 203 from compound 198 was carried as described in Example 1, except that mono-N-Boc-ethylenediamine was used in place of di-p-nitrophenyl carbonate.
  • C,H,N: calc: 46.89, 5.29, 8.54, found 46.50, 5.51, 8.54. C28H41N5O7S1×2 CF3COOH.
  • LC/MS (ES+) 592 (M+1) 1 peak at 4.32 min.
  • Analytical HPLC(A) t=14.69 min.
  • 1H:d-6 DMSO: 0.77 (6H, d), 1.82 (2H, m), 2.06 (1H, m), 2.57 (2H, m), 2.82 (4H, m), 2.97 (1H, m), 3.30 (5H, m), 3.55 (1H, m), 3.65 (1H, m), 3.70 (1H, m), 3.95 (1H, m), 4.88 (1H, m), 4.95 (1H, m), 6.62 (2H, d), 7.20 (6H, m), 7.39 (3H, m), 7.78 (3H, bs).
  • 13C (dmso): 155.9, 152.9, 138.5, 129.2, 128.9, 128.1, 126.1, 122.9, 112.7, 74.7, 74.5, 72.6, 66.2, 57.2, 53.2, 49.4, 38.8, 37.94, 35.1, 32.1, 26.3, 20.0, 19.8.
  • Figure US20100124543A1-20100520-C00107
  • Example 7
  • The synthesis of compound 204 from compound 1.9 was carried as described in Example 1, except that mono-1,3-diamino-3-N-Boc-propane was used in place of di-p-nitrophenyl carbonate.
  • C,H,N: calc: 49.07, 5.64, 8.89, found 48.95, 6.00, 8.92. C29H43N5O7S1×1.6CF3COOH
  • LC/MS (ES+) 605 (M+1) 1 peak at 4.27 min.
  • Analytical HPLC(A) t=14.72 min.
  • 1H:d-6 DMSO: 0.78 (6H, dd), 1.64 (2H, m), 1.83 (2H, m), 2.03 (1H, m), 2.57 (1H, m), 2.78 (4H, m), 2.94 (1H, m), 3.03 (2H, m), 3.32 (2H, m), 3.58 (1H, m), 3.63 (1H, m), 3.73 (1H, m), 3.87 (1H, m), 4.84 (1H, m), 4.92 (1H, m), 6.61 (2H, d), 7.22 (6H.m), 7.36 (1H, d), 7.28 (2H, d), 7.76 (3H, ns).
  • 13C (dmso): 155.8, 155.7, 138.5, +129.1, +129.0, +128.0, +126.1, 122.9, +112.7, +74.6, +74.3, −72.7, −66.2, −57.2, +53.6, −49.5, −37.4, −36.7, −35.5, −32.1, −27.6, +26.2, +20.0, +19.8.
  • Example 8
  • Figure US20100124543A1-20100520-C00108
  • The synthesis of compound 205 from compound 198 was carried as described in Example 1, except that 1,4-diamino-4-N-Boc-butane was used in place of di-p-nitrophenyl carbonate.
  • C,H,N: calc: 48.17, 5.59, 8.26, found 48.02, 5.96, 8.24. C30H45N5O7S1.2CF3COOH
  • LC/MS (ES+) 620 (M+1) 1 peak at 4.36 min.
  • Analytical HPLC(A) t=14.93 min.
  • 1H: d-6 DMSO: 0.77 (6H, dd), 1.43 (4H, m), 1.82 (2H, m), 2.03 (1H, m), 2.77 (4H, m), 2.95 (3H, m), 3.31 (2H, m), 3.56 (1H, m), 3.63 (1H, m), 3.70 (1H, bq), 3.82 (1H, m), 4.85 (1H, m), 4.92 (1H, m), 6.62 (2H, d), 7.2 (7H, m), 7.38 (2H, d), 7.72 (3H, bs).
  • 13C: 155.7, 152.9, +138.6, +129.1, +129.0, +128.0, +126.1, +123.0, +112.7, +74.4, +74.3, −72.7, −66.2, −57.2, +53.7, −49.7, −38.6, −38.5, −35.4, −32.1, −26.3, +26.2, −24.4, +20.1, +19.9.
  • Example 9
  • Figure US20100124543A1-20100520-C00109
  • The synthesis of compound 206 from compound 198 was carried as described in Example 1, except that (3R)-(+)-3-Boc-aminopyrrolidine was used in place of di-p-nitrophenyl carbonate.
  • C,H,N: calc: 48.28, 5.36, 8.28, found 47.89, 5.53, 8.57. C30H43N5O7S1×2 TFA
  • LC/MS (ES+) 618 (M+1) 1 peak at 4.32 min.
  • Analytical HPLC(A) t=14.31 min.
  • 1H and 13C NMR: complex and overlapping mixtures of rotomers.
  • Example 10
  • Figure US20100124543A1-20100520-C00110
  • The synthesis of compound 207 from compound 198 was carried as described in Example 1, except that (3S)-(−)-3-Boc-aminopyrrolidine was used in place of di-p-nitrophenyl carbonate.
  • LC/MS (ES+) 618 (M+1) 1 peak at 4.19 min.
  • Analytical HPLC(A) t=14.75 min.
  • 1H and 13C NMR: complex and overlapping mixtures of rotomers.
  • Example 11
  • Figure US20100124543A1-20100520-C00111
  • The synthesis of compound 308 from compound 198 was carried as described in Example 1, except that N-triphenylmethyl-N,N′-dimethylethanediamine was used in place of di-p-nitrophenyl carbonate.
  • 1H-NMR: 0.76 (6H, dd), 1.65 (2H, m), 1.95 (1H, m), 2.07 (1H, m), 2.7 (2H, m), 2.75 (3H, s), 2.95 (3H, m), 3.45 (2H, m), 3.7 (4H, m), 4.2 (2H, bm), 5.05 (2H, bd), 6.62 (2H, d), 7.2 (5H, m), 7.5 (2H, d).
  • LC/MS: 1 peak, 620 (MH+).
  • Example 12 General Procedures Acylation:
  • Figure US20100124543A1-20100520-C00112
  • To 200 mg (0.37 mM) of 197 dissolved in 5 ml CH2Cl2 was added N-CBz-L-Benzyl tyrosine 183 mg (0.41 mM) followed by 231 mg (1.12 mM) DCC, followed by 29 mg (0.23 mM) DMAP. The reaction is stirred at rt for 24 hr. The precipitates present were removed by filtration. The filtrate was then concentrated in vacuo. The final compound was purified on preparative reversed phase C18 using purification by HPLC C18 Waters Delta Prep 3000 Column: YMC-Pack ODS AA 12S05-2520WT 250×20 mm I.D. S-5 mm, 120 Å, 0-100% B over ½ h, flow=18 ml/min, monitored at 220 nm, B=0.1% trifluoroacetic acid in acetonitrile, A=0.1% trifluoroacetic acid in water. Analytical Column: YMC-Pack ODS AA1 2S05-2520WT 250×4.6 mmI.D. S-5 mm, 120 Å, 0-100% B at 1.5 ml/min. over ½ h, monitored at 220 nm, B=0.1% trifluoroacetic acid in acetonitrile, A=0.1% trifluoroacetic acid in water.
  • The aqueous phase was lyophilized to give 59 mg, (16.3%) GW431896X, (U11484-72-10) tHPLC=11.71 min., MW=966.04, LC/MS=MH+967.
  • Reduction of the Nitro Functionality:
  • Figure US20100124543A1-20100520-C00113
  • A slurry of 209 (170 mg) and 10 mg of 10% Pd.C in 95% EtOH was flushed with hydrogen in a scintillation vial equipped with septum and a stir bar. Continuous overnight hydrogenolysis under hydrogen balloon resulted in a complete conversion. The crude preparation was then filtered off the catalyst, and purified on RP C18 HPLC (Prep Nova-Pack C186 um, 60 A, gradient 0-100% B over 30 min. The desired product was collected and lyophilized affording a white fluffy solid (50 mg, 30.8%).
  • Example 13
  • Figure US20100124543A1-20100520-C00114
  • Compound 211 was obtained following the acylation and reduction procedures of Example 12.
  • ES+ 669.2 (M+1), tHPLC=8.06 min (D), 13C NMR (DMSO) 168.9, 156.9, 155.7, 153.1, 138.1, 130.5, 129.2, 129.1, 128.1, 126.2, 124.7, 122.5, 112.8, 76.2, 74.5, 72.5, 66.1, 58.0, 53.6, 52.6, 49.2, 33.6, 32.1, 26.6, 25.3, 20.0.
  • tHPLC=11.71 min (D), ES+ 967 (M+1).
  • Example 14
  • Figure US20100124543A1-20100520-C00115
  • 212 was obtained following the procedures of Example 12.
  • tHPLC=9.45 min (D), ES+ 592.2 (M+1).
  • 13C NMR (DMSO) 171.5, 155.8, 148.9, 137.8, 129.5, 129.3, 128.5, 126.7, 115.2, 75.2, 73.8, 73.1, 68.3, 67.0, 58.7, 57.1, 53.3, 49.2, 35.4, 32.4, 26.7, 20.1, 19.8.
  • 1H (CDCl3, 399.42 KHz): 8.33 (2H, d, J=8.8), 7.95 (2H, d, J=8.8), 7.23 (5H, m) 5.22 (m, 2H), 5.08 (m, 1H), 4.08 (m, 1H), 3.80-3.45 (7H, m), 3.41 (3H, s), 2.98 (m, 3H), 2.66 (m, 1H), 2.57 (m, 2H), 2.10 (s, 1H), 1.93 (2H, m), 0.82 (3H, d), 0.78 (3H, d).
  • ES+ 622 (M+1), 644 (M+Na)
  • tHPLC=10.29 min (D).
  • 13C NMR (CDCl3): 171.3, 155.5, 149.9, 145.6, 136.9, 129.2, 128.6, 128.5, 126.8, 124.4, 76.7, 75.3, 73.2, 72.9, 68.2, 66.9, 58.7, 55.9, 53.1, 48.3, 35.3, 32.7, 26.3, 19.9, 19.8.
  • Example 15
  • Figure US20100124543A1-20100520-C00116
  • Compound 213 was obtained following the procedure of Example 12. tHPLC=9.21 min (D); ES+ 622 (M+1).
  • 13C NMR (CDCl3): 170.54, 156.2, 148.6, 136.8, 129.4, 129.2, 128.6, 126.6, 115.7, 76.7, 74.6, 73.2, 71.8, 70.6, 68.2, 6.6.9, 58.9, 57.3, 53.8, 49.4, 36.2, 33.1, 26.8, 19.8, 19.5.
  • Intermediate: t HPLC=10.05 min (D); ES+=652 (M+H) 674 (M+Na).
  • Example 16
  • Figure US20100124543A1-20100520-C00117
  • 214 was obtained following the procedure of Example 12.
  • ES+ 634.4 (M+1); t HPLC=7.17 min (D).
  • 13C (DMSO): 169.3, 155.8, 153.1, 138.0, 129.1, 129.0, 128.1, 126.3, 122.6, 112.8, 94.3, 75.6, 74.6, 72.4, 66.1, 57.8, 52.7, 52.0, 49.3, 38.4, 34.7, 32.2, 29.1, 26.6, 21.4, 20.1, 20.0.
  • Example 17
  • Figure US20100124543A1-20100520-C00118
  • 215 was obtained following the procedure of Example 12.
  • t HPLC=9.12 min (D)
  • 1H (DMSO) all signals broad: 7.38 (3H, br m), 7.20 (5H, br m), 6.62 (2H, br m), 5.15 (1H, br m), 4.92 (1H, br m), 4.00 (3H, m), 3.7-3.0 (16H, m), 2.78 (2H, m), 2.57 (3H, m), 2.04 (m, 1H), 1.78 (m, 2H), 0.77 (6H, m)
  • 13C (DMSO) 170.6, 156.3, 153.7, 139.1, 129.8, 128.4, 126.7, 123.7, 113.3, 79.8, 79.2, 77.3, 76.1, 75.4, 75.2, 73.0, 71.9, 52.3, 51.8, 48.2, 46.7, 39.9, 38.7, 25.8, 22.6.
  • Intermediate:
  • t HPLC=10.18 min (D); ES+ 696.3 (M+1).
  • Example 18
  • Figure US20100124543A1-20100520-C00119
  • 216 was obtained following the procedure of Example 12.
  • 1H-NMR: 0.97 (6H, t), 1.95 (2H, m), 2.20 (1H, m), 2.9 (2H, m), 2.96 (6H, s), 3.00 (3H, s), 3.38 (1H, m), 3.42 (3H, m), 3.36 (1H, m), 3.6 (2H, m), 3.7 (6H, m), 3.98 (2H, m), 4.2 (2H, dd), 5.1 (1H, bs), 5.4 (1H, m), 6.8 (2H, d), 7.4 (5H, m), 7.6 (2H, d).
  • LC-MS: 1 peak, 692 (MH+).
  • Example 19
  • Figure US20100124543A1-20100520-C00120
  • 217 was obtained following the procedure of Example 12.
  • 1H-NMR (CDCl3): 0.78 (6H, dd), 1.9 (2H, m), 2.1 (1H, m), 2.3 (3H, s), 2.9 (8H, m), 2.9 (2H, m), 3.15 (1H, m), 3.35 (1H, m), 3.5 (1H, m), 3.75 (4H, m), 4.06 (2H, s), 4.15 (2H, m), 4.9 (1H, dd), 5.05 (1H, bs), 5.2 (1H, bs), 6.63 (2H, d), 7.2 (5H, m), 7.55 (2H, d), 8.0 (2H, m).
  • ESMSP: 676 (MH+).
  • Example 20 General Procedure for N-Acylated Compounds
  • Figure US20100124543A1-20100520-C00121
  • A mixture of 0.5 g (1 mMol) of (3S)-Tetrahydro-3-furfuryl-N-((1S,2R)-1-benzyl-2-hydroxy-3-(N-isobutyl-4-aminobenzenesulfonamido)propyl) carbamate, 0.4 g (1.5 mMol) of Boc-(S)-3-pyridyl alanine, 0.29 g (1.5 mMol) EDCI and 0.1 g 4-dimethylamino pyridine in 10 ml of N,N-dimethylformamide was stirred at 25° for 12 hours. The volatiles were removed in vacuo and the residue was partitioned between ethyl acetate and 1N hydrochloric acid. The organic layer was washed with 1N sodium hydroxide and brine, dried over magnesium sulfate and concentrated in vacuo. The residue was chromatographed on a 2 inch plug of silica gel (1:1 ethyl acetate:hexane) to give the desired N-acylated material. Deprotection by treatment with 50 ml of trifluoroacetic acid, followed by co-evaporation of residual acid with methanol gave the desired prodrug as a white foam (0.2 g, 26%).
  • H1-NMR (acetonitrile-D3): 0.95 (6H, dd), 2.0 (2H, m), 2.25 (1 h, m), 2.8-3.1 (5H, m), 3.6-4.0 (7H, m), 4.25 (1H, m), 4.75 (1H, m), 5.18 (1H, m), 5.45 (1H, m), 7.0 (2H, d), 7.4 (5H, m), 7.75 (2H, d), 8.2 (1H, m), 8.8 (1H, d), 8.85 (1H, d), 9.15 (1H, s).
  • LC/MS: 1 peak, 654 (MH+).
  • Example 21
  • Figure US20100124543A1-20100520-C00122
  • 220 was obtained using the general procedure in Example 20.
  • 1H-NMR (acetone-d6/methanol-d4): 0.95 (6H, t), 2.0 (2H, m), 2.2 (1H, m), 2.90 (1H, dd), 2.95 (2H, d), 3.12 (1H, dd), 3.4 (2H, m), 6 (1H, d), 3.8 (5H, m), 4.4 (2H, bm), 6.82 (2H, d), 7.20 (1H, s), 7.4 (5H, m), 7.65 (2H, d), 8.0 (1H, s).
  • LC/MS: 1 peak, 643 (MH+).
  • Example 22
  • Figure US20100124543A1-20100520-C00123
  • 221 was obtained using the general procedure in Example 20.
  • 1H-NMR (DMSO d-6): 0.76 (6H, t), 1.80 (2H, m), 2.10 (1H, m), 3.7 (4H, m), 3.75 (3H, s), 3.2 (5H, m), 3.58 (2H, s), 3.7 (4H, m), 4.97 (1H, bm), 5.18 (1H, bs), 6.7 (2H, d), 7.22 (5H, m), 7.45 (2H, d).
  • LC/MS: 1 peak, 646 (MH+).
  • Example 23
  • Figure US20100124543A1-20100520-C00124
  • 222 was obtained using the general procedure in Example 20.
  • 1HNMR (acetonitrile d-3): 1.0 (6H, t), 2.0 (2H, m), 2.2 (1H, m), 3.00 (6H, s), 3.02 (3H, s), 3.1 (4H, m), 3.5 (3H, m), 3.8 (8H, m), 4.4 (2H, s), 5.15 (1H, bs), 7.4 (5H, m), 7.97 (2H, d), 8.04 (2H, d).
  • LC/MS: 1 peak, 692 (MH+).
  • Example 24
  • Figure US20100124543A1-20100520-C00125
  • 223 was obtained using the general procedure in Example 20.
  • t HPLC=9.22 min (D); ES+ 622 (M+1).
  • 1H NMR d6-DMSO: 0.76 (6H, dd), 1.0-1.8 (15H, m), 2.03 (1H, m), 2.58 (2H, m), 2.79 (2H, m), 3.11 (1H, m), 3.28 (3H, s), 3.3-3.5 (12H, m), 3.94 (1H, m), 4.08 (1H, m), 4.94 (1H, m), 5.14 (1H, m), 6.61 (2H, d), 7.22 (5H, m), 7.40 (3H, m).
  • 13C (DMSO) 169.7, 165.9, 152.9, 138.4, 129.2, 129.1, 128.1, 126.2, 123.1, 112.8, 74.4, 74.1, 72.5, 71.2, 69.8, 66.1, 58.1, 57.1, 52.9, 47.5, 33.4, 33.2, 26.3, 24.5, 18.9, 18.8.
  • Example 25
  • Figure US20100124543A1-20100520-C00126
  • 224 was obtained using the general procedure in Example 20.
  • Example 26 O,N-Diacylated Prodrugs
  • The general procedure for N,O-diacylated compounds followed the protocol outlined in Example 20, above, except that a five fold excess of reagents was used relative to the starting material.
  • Figure US20100124543A1-20100520-C00127
  • t HPLC 9.26 min (D); ES+ 738 (M+1) 760 (M+Na).
  • 13C (DMSO): 170.2, 169.8, 156.4, 143.4, 138.8, 129.5, 128.8, 128.5, 126.8, 119.7, 74.9, 74.2, 73.7, 71.6, 70.7, 70.3, 68.0, 67.2, 59.3, 57.6, 53.8, 49.6, 35.7, 33.8, 27.1, 20.4.
  • 1H (DMSO): 10.1 (1H, s), 7.84 (d, 2H, J=8.5), 7.76 (d, J=8.7, 2H), 7.40 (1H, d, J=9.2), 7.22 (m, 5H), 5.14 (1H, m), 4.95 (1H, m), 4.1 (m, 8H), 3.7-3.3 (m, 13H), 3.28 (s, 3H), 3.26 (s, 3H), 2.86 (m, 2H), 2.73 (m, 1H), 2.59 (m, 1H), 2.04 (m, 1H), 1.83 (m, 2H), 0.78 (m, 6H).
  • Example 27
  • Figure US20100124543A1-20100520-C00128
  • To a mixture of 197 (2.93 g, 5.47 mmol) and phosphorous acid (Aldrich, 2.2 equiv., 12.03 mmol, 987 mg) in 20 ml pyridine was added 1,3-dicyclohexylcarbodiimide (Aldrich, 2.1 equiv., 11.49 mmol, 2.37 g) and the reaction heated to 60° C. under nitrogen for 3 h. Solvent was removed in vacuo, the residue treated with 200 ml 0.1N aqueous sodium bicarbonate and stirred 1 h at ambient temperature. The mixture was filtered, the filtrate acidified to pH 1.5 by addition of conc. HCl and extracted with ethyl acetate (3×100 ml). The combined organic layers were dried over magnesium sulfate, filtered and concentrated in vacuo to give 3.15 g (96%) of desired product 226 which was used directly in the next reaction. HPLC: Rt=8.91 min (96%), MS (AP+) 600.5 (M+1).
  • Example 28
  • Figure US20100124543A1-20100520-C00129
  • A suspension of 226 (˜5.47 mmol) in 18 ml hexamethyldisilazane was stirred at 120° C. until homogeneous followed by addition of bis(trimethylsilyl) peroxide (Gelest, Inc., 2.3 equiv., 12.58 mmol, 2.24 g, 2.71 ml). After 1 h the mixture was cooled to ambient temperature, solvent removed in vacuo, the residue stirred with 100 ml methanol, solvent removed in vacuo, the residue stirred with 100 ml 0.1N aqueous sodium bicarbonate, acidified to pH 1.5 by addition of conc. HCl, saturated with brine and extracted with ethyl acetate (3×100 ml). The combined organic layers were dried over magnesium sulfate, filtered and concentrated in vacuo to give 2.98 g (88%) of desired product 227, which was used directly in the next reaction. HPLC: Rt=9.28 min (90%), MS (AP+) 616.5 (M+1).
  • Alternatively, 227 can be synthesized directly from 197. In this method, 197 was dissolved in pyridine (300 mL). The resulting solution was concentrated in vacuo to about 150 ml at 50-55° C. The solution was then cooled under N2 to 5° C., and treated with POCl3 (6.5 ml, 1.24 equiv.) over 2 minutes. The cooling bath was removed and the reaction stirred at ambient temperature for 2.5 hrs. The solution was then cooled to 5° C. and water (300 ml) was added over 30 minutes.
  • The resulting mixture was extracted with 4-methylpentan-2-one (MIBK, 2×150 ml). The combined extracts were washed with 2N HCl (2×250 ml). The acid washes were back extracted with MIBK (60 ml), then the combined MIBK solutions were treated with 2N HCl (150 ml). The two phase mixture was stirred rapidly and heated to 50° C. for 2 hours. The reaction mixture was cooled to 20° C., the phases were separated and the MIBK solution was washed with brine (150 ml). The product, 227, was isolated by drying the solution with magnesium sulfate, filtering of the drying agent and concentrating in vacuo at 40° C. to give the product as a pale yellow foam (31 g, 90% yield).
  • Example 29
  • Figure US20100124543A1-20100520-C00130
  • A solution of 227 (2.98 g, 4.84 mmol) in 50 ml ethyl acetate was treated with 10% palladium on carbon (Aldrich, 300 mg) and put under 35 psi of hydrogen on a Parr shaker for 15 h. Catalyst was removed by filtration and solvent removed in vacuo to give 2.66 g (94%) of desired product 228. HPLC: Rt=7.23 min (92%), MS (ES+) 586.3 (M+1).
  • Example 30
  • Figure US20100124543A1-20100520-C00131
  • Solid 228 (2.66 g, 4.54 mmol) was treated with ml aqueous sodium bicarbonate (Baker, 3.0 equiv., 13.63 mmol, 1.14 g) and loaded onto a resin column (Mitsubishi Kasei Corp., MCI-gel, CHP-20). Distilled water was run through until the eluent was neutral followed by product elution with 1% acetonitrile in water. Pure fractions were pooled and lyophilized to give 918 mg of pure bis-sodium salt 229.
  • Alternatively, 7 g of 228 was dissolved in 100 ml of EtOAc with warming and the solution was extracted with 100 ml of aqueous 250 mM triethylammonium bicarbonate (TEABC) (2×). The aqueous extracts were combined and diluted to 1500 ml with water. This solution was applied to a 300 ml DEAE-52 column (Whatman) which was equilibrated with 50 mM TEABC. The column was washed with 8 L of 50 mM TEABC and the TEA salt was eluted with 2 L of 250 mM TEABC. The solution was evaporated en vacuo to 100 ml then lyophilized to yield the TEA salt (1.5 TEA equivalents). The TEA salt was (5.8 g) was dissolved in 200 ml water, 300 ml of 1 N HCl was added and the mixture was extracted with EtOAc (3×200 ml). The ethyl acetate solution was dried with MgSO4 then evaporated en vacuo to yield 4 g of the free acid. Two grams of the free acid was dissolved in 50 ml of acetonitrile and a solution of 573 mg NaHCO3 in 200 ml water was added. The mixture was lyophilized yielding 2.1 g of the bis sodium salt (compound 229).
  • Example 31
  • Figure US20100124543A1-20100520-C00132
  • 0.53 g (3.0 mmol) 2-[2-(2-Methoxyethoxy)ethoxy]acetic acid was added to a stirred solution of 1.2 g (3.15 mmol) HATU 0.2 g (1.47 mmol) HOAt 0.4 g (4.0 mmol) NMM in 10 ml anhydrous N,N-dimethylformamide. The mixture was stirred at room temperature for 30 minutes, then 0.5 g (1 mmol) of (3S)-Tetrahydro-3-furfuryl-N-((1S,2R)-1-benzyl-2hydroxy-3-(N-isobutyl-4-aminobenzenesulfonamido)-propyl) carbamate was added to the solution in one portion. The mixture was stirred at 20° C. for an hour then at 50° C. for an additional 12 hours. It was then cooled to 20° C., 50 ml of ether was added, and the solution was washed with water three times. The aqueous phase was washed with ether, and then the combined organic phases were dried with anhydrous magnesium sulfate and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel chromatography to obtain the desired Mono-(N)acylated (102 mg, 15%) and Bis-(O,N) acylated (262 mg, 32%) compounds.
  • Mono-(N)-acylated: 1H-NMR (CDCl3): 0.85 (dd, 6H), 1.85 (m, 2H), 2.08 (m, 1H), 2.8-3.1 (m, 7H), 3.33 (s, 3H), 3.55 (m, 3H), 3.70-3.90 (m, 8H), 4.1 (s, 2H), 5.0 (d, 1H), 5.08 (s(br), 1H), 7.2 (m, 5H), 7.70 (d, 2H), 7.80 (d, 2H), 9.09 (s, 1H).
  • MS (FAB+): 666 (M+1).
  • Bis-(O,N)-acylated: 1H-NMR (CDCl3): 0.77 (m, 6H), 1.81 (m, 1H), 1.95 (m, 1H), 2.05 (m, 1H), 2.6-3.0 (m, 6H), 3.2 (m, 1H), 3.332 (s, 3H), 3.338 (s, 3H), 3.5-3.8 (m, 18H), 4.1 (s, 2H), 4.14 (s, 2H), 4.17 (m, 1H), 5.05 (m, 2H), 5.25 (s(br), 1H), 7.2 (m, 5H), 7.69 (d, 2H), 7.78 (d 2H), 9.06 (s, 1H).
  • MS (FAB+): 826 (M+1), 848 (M+Na).
  • Example 32
  • Figure US20100124543A1-20100520-C00133
  • We dissolved 0.521 g (1 mM) of 1273W94 in 5 ml THF, then cooled to −78° C. under nitrogen, and added 1.56 ml (2.5 mM) of a 1.6 M solution of nBuLi in hexane. After 20 min at −78° C., we added 105 μL (1.1 mM) of ethyl chlorocarbamate and warmed up the reaction to room temperature, followed by addition of another 105 μL of ethyl chlorocarbamate.
  • After stirring for additional 4 hrs, the reaction was quenched with water and the organic solvent evaporated. Part of the crude product was purified on a silica gel (Rf=0.69 (1:2 ethyl acetate:hexane)), yielding 0.131 g of the product.
  • C,H,N: calc: 46.06, 4.97, 5.88, found 45.90, 4.97, 5.88. C23H33N5O5S1. 2.2 TFA
  • LC/MS (ES+) 594 (M+1) 1 peak at 6.96 min.
  • Analytical HPLC(A) t=24.57 min. 13C (CDCl3): 155.8, 154.4, 149.9, 145.7, 136.8, +129.2, +128.7, +126.8, +124.2, 80.1, +76.9, −64.3, −56.2, −52.5, −48.7, −36.2, +28.1, +26.4, +20.0, +19.8, +14.3.
  • Example 33
  • Figure US20100124543A1-20100520-C00134
  • We dissolved 0.131 g of the above ethyl carbonate in 4 ml DCM, followed by 4 ml of TFA. Solvents were then removed after 45 min at room temperature, resulting in the title compound.
  • 1H (DMSO): 8.37 (2H, d, J=7.2), 8.15 (2H, m), 8.00 (2H, d, J=7.0), 7.37 (5H, m), 5.04 (1H, d, J=6.9), 4.06 (2H, q, J=7.0), 3.82 ((1H, m), 3.35 (2H, m), 2.95 (4H, m), 1.82 (1H, m), 1.20 (3H, t, J=7.0), 0.72 (overlapping doublets, 6H, J=6.2).
  • LC/MS 1 peak at 4.76 min.
  • ES+ 497.3 (M+1).
  • Example 34
  • Figure US20100124543A1-20100520-C00135
  • C,H,N: calc: 53.26, 6.14, 7.57, found 53.22, 6.14, 7.57. C23H33N5O5S1×0.8 TFA
  • LC/MS (ES+) 594 (M+1) 1 peak at 6.96 min.
  • Analytical HPLC(A) t=24.57 min. 1H (DMSO): 8.34 (2H, d, J=8.7), 8.02 (2H, d, J=8.0), 7.19 (5H, m), 6.98 (1H, d, J=7.2), 5.00 (1H, m), 3.83 (2H, q), 3.50 (2H, m), 3.06 (m, 2H), 2.96 (2H, m), 2.43 (1H, m), 1.97 (1H, m), 1.02 (3H, t), 0.84 (3H, d), 0.82 (3H, d).
  • 13C (DMSO): 156.2, 150.1, 145.7, 140.0, +129.7, +129.2, +128.5, +126.3, +125.0, +71.8, −60.0, +56.2, −56.0, −51.8, −36.0, +26.3, +20.3, +20.1, +14.6.
  • Example 35
  • Figure US20100124543A1-20100520-C00136
  • Synthesis of 235 was accomplished analogous to that set forth in Example 1.
  • Yield 15.2%; tHPLC=25.2 min (A).
  • Rf=0.54 (B); ES+ 687.3 (M+1).
  • 1H (CDCl3): 8.34 (overlapping d+d, 4H), 7.97 (d, 2H, J=8.9), 7.35 (7H, m), 5.09 (1H, m), 4.56 (1H, d, J=8.4), 4.20 (1H, m), 3.54 (1H, m), 3.00 (3H, m), 2.82 (1H, m), 1.84 (1H, m), 1.37 (9H, s), 0.84 (3H, d), 0.82 (3H, d).
  • Example 36
  • Figure US20100124543A1-20100520-C00137
  • We dissolved 150 mg of 235 in 3 ml of anhydrous dioxane, added 0.35 ml of S(+)-3-OH-THF and 0.14 ml triethyl amine. The mixture was refluxed gently under nitrogen for 2 days. Conversion to 236 was quantitative. Solvents were removed and the compound purified on silica (B).
  • tHPLC=22.98 min (A); ES+ 636.2 (M+1).
  • 1H NMR (CDCl3): 8.29 (2H, d), 7.91 (2H, d), 7.22 (5H, m), 5.13 (1H, m), 4.96 (1H, m), 4.52 (1H, d), 4.02 (1H, m), 3.84 (2H, m), 3.44 (1H, m), 3.36 (1H, m), 3.10 (3H, m, overlap), 2.88 (2H, m), 2.64 (1H, m), 2.14 (1H, m), 2.05 (1H, m), 1.84 (1H, m), 1.27 (9H, s), 0.78 (6H, two overl. d).
  • Example 37 Carbohydrate-Based Prodrugs
  • Figure US20100124543A1-20100520-C00138
  • A mixture of 0.54 g (1 mMol) of (3S)-Tetrahydro-3-furfuryl-N-((1S,2R)-1-benzyl-2-hydroxy-3-(N-isobutyl-4-aminobenzenesulfonamido)propyl) carbamate, 0.46 g (2 mMol) of 5-dimethyl-tert-butyosilyloxypentanoic acid, 0.346 g (1.8 mMol) of EDCI and 0.556 mL (4 mMol) of triethylamine in 10 ml of dimethyl formamide was stirred at rt for 24 h. Another 3 mMol each of acid, EDCI and triethylamine were added and stirring was continued for an additional 96 h. A third batch of acid and EDCI was added (3 mMol each) and the mixture was stirred 72 h to complete the reaction.
  • The reaction mixture was then diluted with ethyl acetate and extracted with 1N hydrochloric acid, saturated sodium bicarbonate and water. Evaporation of the solvent and purification on silica gel (30% ethyl acetate-hexane) gave the desired product (500 mg) as a waxy solid.
  • LCMS: 1 peak, 772.5 (M+Na)
  • 1H NMR (CDCL3): 0.01 (6H, s), 0.78 (6H, dd), 0.95 (9H, s), 1.4-1.8 (6H, m), 1.9 (2H, m), 2.05 (1H, m), 2.3 (2H, m), 2.65 (1H, m), 2.95 (2H, m), 3.22 (1H, m), 3.4 (1H, m), 3.6 (2H, m), 3.75 (3H, m), 4.8 (1H, d), 5.1 (1H, bs), 5.2 (1H, bs), 7.2 (5H, m), 7.95 (2H, d), 8.36 (2H, d).
  • 450 mg of the 238 was dissolved in 30 ml of tetrahydrofuran and treated with 20 ml of water and 50 ml of acetic acid. The mixture was stirred at rt for 2 h and evaporated. Titration with hexane gave the desired alcohol (290 mg) as a white solid.
  • A mixture of 0.15 g (0.24 mMol) of the alcohol produced above from the previous reaction, 0.205 g (0.5 mMol) of tetraacetylglucosylbromide and 0.191 g (0.7 mMol) of silver carbonate in 3 ml of dichloromethane was stirred at rt for 6 h. 150 mg of additional glucosyl bromide and 150 mg of silver carbonate were added and the mixture was stirred at rt overnight. The mixture was loaded onto a pad of silica gel and eluted with 30% ethylacetate-hexane to afford the desired protected carbohydrate pro-drug as a white foam (200 mg).
  • LCMS: 1 peak, 966 (M+H).
  • 1H-NMR (CDCl3): 0.78 (6H, dd), 1.9 (2H, m), 2.00 (3H, s), 2.02 (3H, s), 2.05 (3H, s), 2.06 (3H, s), 2.1 (2H, m), 2.3 (2H, m), 2.7 (1H, m), 2.94 (3H, bd), 3.35 (2H, m), 3.45 (2H.m), 3.8 (5H, m), 4.1 (3H, m), 4.5 (1H, d), 4.9 (1H, bs), 4.95 (1H, t), 5.08 (4H, m), 2H, d), 8.35 (2H, d).
  • Example 38
  • Figure US20100124543A1-20100520-C00139
  • 1.5 g (9.4 mmol) SO3.py complex was added to a stirred solution of 1 g (1.87 mmol) of 197 in 25 mL anhydrous tetrahydrofurane. The mixture was stirred at 20° C. for 12 hours, then filtered. The filtrate was concentrated at reduced pressure, and the residue was transferred to a silica gel column and eluted with EtOAc (neat), followed by EtOAc:EtOH (4:1) to obtain 471 mg (47%) 239 as a colorless foam.
  • 1H-NMR (CDCl3): 0.80 (m, 6H), 1.8-2.1 (m, 3H), 4.15 (s(br), 1H), 4.8 (t, 1H), 5.04 (s (br), 1H).
  • MS (ES−): 614 (M−1).
  • Figure US20100124543A1-20100520-C00140
  • 100 mg (0.162 mmol) 239 dissolved in 15 ml anhydrous tetrahydrofuran and 200 mg Pd/BaSO4 (5%) was added to the solution. The mixture was stirred under atmospheric pressure of hydrogen for 8 hours, and then the catalyst was filtered. The filtrate was concentrated under reduced pressure then dried under vacuum (˜1 Hg mm, 48 hrs.) to produce 80 mg (81%) 240 as a colorless foam.
  • 1H-NMR (DMSO-d6): 0.85 (dd, 6H), 0.90 (m, 1H), 2.05 (m, 2H), 2.58 (m, 3H), 2.84 (dd, 1H), 3.05 (m, 2H), 3.55-3.80 (m, 6H), 4.20 (t, 1H), 4.42 (m, 1H), 4.93 (s(br), 1H), 6.09 (s, 2H), 6.70 (d, 2H), 6.80 (d, 1H), 7.15-7.40 (m, 4H), 7.51 (d, 2H).
  • MS (ES−): 584 (M−1).
  • Example 39
  • Figure US20100124543A1-20100520-C00141
  • 780 mg (3 mmol) 2-Chloro-1,3,2-dioxaphospholane was added to a stirred solution of 1.07 g (2 mmol) 197 and 0.7 ml (4 mmol) N,N-Diisopropylethylamine in 25 ml dichloromethane at 0° C. The mixture was allowed to warm up to room temperature and it was stirred for 2 hours. The mixture was then cooled to 0° C. and 1.5 g (9.3 mmol) bromine was added in 5 ml dichloromethane. The mixture was stirred for 1 hour at 20° C., followed by evaporation under reduced pressure. An aqueous solution (50%) of 15 ml trimethylamine was added to the residue, and the mixture was stirred at 20° C. for 12 hours.
  • Solvents were removed under reduced pressure and 50 ml EtOAc:EtOH (9:1) was added to the residue. The solid was filtered, washed with EtOAc:EtOH (9:1) then the filtrate was concentrated under reduced pressure. The residue was chromatographed on a 3 inch plug of silica gel using ethyl acetate (neat), then methanol (neat), as eluents to obtain 1.15 g (82%) 241 as an off-white solid.
  • 1H-NMR (CDCl3): 0.60 (dd, 6H), 1.70 (m, 1H), 1.95 (m, 1H), 2.10 (m, 1H), 2.8-3.2 (m, 6H), 3.4 (s (br), 9H), 5.09 (s(br), 1H), 7.25 (m, 5H), 7.83 (d, 2H), 8.28 (d, 2H).
  • MS (ES+): 701 (M+1), 184 (phosphatidyl choline+).
  • Example 40
  • Figure US20100124543A1-20100520-C00142
  • 250 mg Pd/C (10%) was added to a solution of 250 mg (0.35 mmol) 241 in 10 ml methanol, and the mixture was stirred under atmospheric pressure of hydrogen for 4 hours at 20° C. The mixture was filtered, and the filtrate was concentrated under reduced pressure. The residue was then dissolved in 10 ml water and lyophilized to obtain 174 mg (74%) 242 as white solid.
  • 1H-NMR (DMSO-d6): 0.82 (dd, 6H), 1.80-2.00 (m, 2H), 2.10 (m, 1H), 2.80 (m, 3H), 3.00 (m, 2H), 3.2 (s (br), 9H), 4.0-4.3 (m, 4H), 4.91 (s(br), 1H), 6.08 (s(br), 2H), 6.67 (d, 2H), 7.30 (m, 5H), 7.48 (d, 2H), 8.12 (d, 1H).
  • MS (ES+): 671 (M+1), 184 (phosphatidyl choline+).
  • Example 41
  • Figure US20100124543A1-20100520-C00143
  • 0.175 ml (2 mmol) phosphorus trichloride was added to a stirred solution of 1.07 g (2 mmol) 197 and 0.35 ml (2 mmol) N,N-Diisopropylethylamine in 25 ml dichloromethane at 20° C. The mixture was stirred for 4 hours at 20° C., then 1 ml water was added and stirred for an additional 12 hours at 20° C. 3 g anhydrous magnesium sulfate was added to the mixture and it was stirred for minutes, then filtered. The filtrate was concentrated under reduced pressure and purified by silica gel chromatography using EtOAc:Hexane (4:1), then EtOAc:EtOH (1:1), to obtain 402 mg (48%) 226 and 427 mg (36%) 243. 226:
  • 1H-NMR (DMSO-d6): 0.82 (dd, 6H), 1.84 (m, 1H), 1.98 (m, 1H), 2.10 (m, 1H), 2.68 (dd, 1H), 2.9-3.2 (m, 4H), 3.6-3.8 (m, 3H), 3.94 (t, 1H), 4.30, (s(br), 1H), 4.97 (s(br), 1H), 7.30 (m, 5H), 8.14 (d, 2H), 8.43 (d, 2H).
  • MS (ES−): 598 (M−1).
  • 243: (1:1 mix of diastereomers):
  • 1H-NMR (CDCl3): 0.80 (m, 6H), 1.8-2.1 (m, 4H), 2.8-3.2 (m, 6H), 3.7-3.9 (m, 4H), 4.15 (m, 1H), 4.8-5.15 (m, 2H), 5.57, 5.72 ((d, d), 1H), 7.25 (m, 5H), 7.95 (dd, 2H), 8.35 (m, 2H).
  • MS (ES−): 580 (M−1), 598 ((M+H2O)−1).
  • Example 42
  • Figure US20100124543A1-20100520-C00144
  • The reduction was carried out as described in Example 40; (Yield: 79%).
  • 1H-NMR (DMSO-d6): 0.81 (dd, 6H), 1.82 (m, 1H), 1.95 (m, 1H), 2.08 (m, 1H), 2.6-3.15 (m, 6H), 3.6-3.75 (m, 3H), 4.03 (t, 1H), 4.28, (m, 1H), 4.96 (s(br), 1H), 6.07 (s, 2H), 6.65 (d, 2H), 7.25 (m, 5H), 7.42 (d, 2H).
  • MS (ES−): 568 (M−1).
  • Example 43
  • Figure US20100124543A1-20100520-C00145
  • The reduction was carried out as described in Example 40; (Yield: 98%).
  • (1:1 mix of diastereomers):
  • 1H-NMR (DMSO-d6): 0.82 (m, 6H), 1.75-2.0 (m, 2H), 2.05 (m, 1H), 2.6-3.2 (m, 6H), 3.55-3.8 (m, 4H), 4.02, 4.22 (m, t, 1H), 4.75 (m, 1H), 4.90, 5.01 ((d, d), 1H), 6.12 (s, 1H), 6.68 (d, 2H), 7.30 (m, 5H), 7.49 (d, 2H).
  • MS (ES−): 550 (M−1), 568 ((M+H2O)−1).
  • Example 44 Pharmacokinetics in Sprague-Dawley Rats Following Single Oral Dose
  • In order to study the pharmacokinetics of the prodrugs of this invention, we administered single oral doses of a series of prodrugs of this invention, as well as VX-478, to male and female Sprague-Dawley rats. Administration of molar equivalents of a series of prodrugs of this invention in a variety of pharmaceutical vehicles was tested.
  • Separate groups of male and female Sprague-Dawley rats (3/sex/group) received oral doses of compound 229 by oral gavage, in different vehicles at the same dose equivalent (40 mg/kg molar equivalent of VX-478). The different vehicles for compound 229 were: 1) water; 2) 5/4/1; 3) PEG 400; 4) TPGS/PEG 400; and 5) PEG. The vehicles for VX-478 were: 1) 33% TPGS/PEG400/PEG; and 2) 12.5% TPGS/PEG 400/PEG.
  • Blood samples were collected following administration at various time intervals and analyzed for the presence of both compound 229 and its metabolite, VX-478, by HPLC and MS methods. The results of this study are tabulated below (Table IV).
  • TABLE IV
    Compound 229 229 229 229 VX-478 VX-478
    vehicle H2O H2O:PG:EtOH PEG 400 TPGS/PEG 33% TPGS/ 12.5% TPGS/
    5:4:1 400/PG PEG 400/PG PEG 400/PG
    number of rats 3 3 3 3 6
    Figure US20100124543A1-20100520-P00001
    3
    Molar equiv. 40 PO 40 PO 40 PO 40 PO 41 PO 50 PO
    dose/478 Dose
    (mg/Kg)
    AUC 11.7 ± 4.8 10.6 ± 7.4 7.4 ± 1.8 8.2 ± 1.6 29.6 ± 5.8 16.2 ± 1.8 
    (ug*hr/ml)
    Cmax (μM)  7.1 ± 1.7  3.3 ± 0.6 3.1 ± 0.3 3.0 ± 0.7 14.0 ± 2.2 6.0 ± 1.0
    half life (hr) 1.7* 3.4* 2.8* 2.8*  2.5 ± 0.9 2.2 ± 1.0
    Relative Avail. of 39.5† 35.8† 25.0† 27.7† reference reference
    VX-478 90.2†† 81.8†† 57.1†† 63.3††
    a dose of 50 mg/kg of compound 229 is equal to 40 mg/Kg of VX-478.
    no compound 229 was detected in plasma at 15 min. (first data point).
    *Represents the harmonic mean
    †Relative availability of VX-478 when compared to a prototype clinical formulation
    ††Relative availability of VX-478 when compared to a prototype toxicology formulation
  • We performed a similar study on dogs using both a solid capsule formulation of compound 229 and an ethanolic/methyl cellulose solution formulation, as compared to a TPGS-containing solution formulation of VX-478. The results from this study are presented below in Table V.
  • TABLE V
    Compound 229 229 VX-478
    vehicle solid methyl 22%
    capsule cellulose in 5% TPGS/PEG
    EtOH/water 400/PG
    number of dogs 2  2  >2
    Molar equiv. dose/478 17 PO 17 PO 17 PO
    Dose (mg/Kg)
    AUC 16.7 ± 2.7  14.2 ± 3.2  23.5 ± 7.4 
    (ug*hr/ml)
    Cmax (μg/ml) 6.1 ± 1.7 6.3 ± 0.3 6.8 ± 1.1
    Tmax (hr) 2.3 ± 0.6 0.5 ± 0.5 1.0 ± 0.8
    Relative Avail. 71.1 60.4 reference
    of VX-478 (%)
  • The results demonstrate that oral administration of compound 229 as an aqueous solution resulted in improved bioavailability in comparison to the other vehicles studied. Also, following administration of compound 229, none of that compound was detected in the first time point blood sample (or later samples), suggesting first pass metabolism to VX-478. Comparison of the aqueous dose of compound 229 with the two non-aqueous formulations used for VX-478 indicated equivalence in delivery as illustrated by the range found for the bioavailability.
  • Example 45
  • Figure US20100124543A1-20100520-C00146
  • We added 0.28 ml (3.0 mmol) POCl3 to a stirred solution of 1.07 g (2.0 mmol) of compound 197 in 10 ml anhydrous pyridine at 5° C. The mixture was allowed to warm up to room temperature and stirred at 20° C. for 3 hours. The mixture was cooled to 0° C., and quenched with 10 ml water. The solvents were removed under reduced pressure, the residue was dissolved in 100 ml ethyl acetate and washed with 20 ml 1M sodium bicarbonate solution. The organic phase was dried with anhydrous magnesium sulfate, filtered then concentrated. Chromatographic purification (SiO2, EtOAc) produce 280 mg of compound 400 (Yield=23%).
  • 1H-NMR (DMSO-d6): 0.86 (dd, 6H), 2.05 (m, 2H), 2.84 (d, 2H), 2.95 (dd, 1H), 3.06 (m, 1H), 3.25 (dd, 1H), 3.50-3.70 (m, 4H), 4.20 (m, 1H), 4.35 (m, 1H), 7.2-7.4 (m, 5H), 7.9-8.1 (m, 2H), 8.40 (m, 2H).
  • MS (ES−): 596 (M−1).
  • Figure US20100124543A1-20100520-C00147
  • Compound 400 was converted to compound 401 using the standard hydrogenation method described above employing H2/PdC (10%), atmospheric pressure, 4 hours at room temperature, solvent: MeOH—H2O (5:1). Yield of 401=68%.
  • 1H-NMR (DMSO-d6): 0.85 (dd, 6H), 2.0 (m, 2H), 2.6-3.1 (m, 4H), 4.15 (m, 1H), 4.40 (m, 1H), 6.1 (s(br), 1H), 6.61 m (2H), 7.2-7.5 (m, 7H).
  • MS (ES−): 566 (M−1).
  • Example 46
  • Figure US20100124543A1-20100520-C00148
  • We added 1.0 g (2.8) mmol Na-t-Boc-nd-Cbz-L-Ornithine was added to stirred solution of 1.2 g (3.15 mmol) HATU, 0.2 g (1.47 mmol) HOAt, 0.4 g (4.0 mmol) NMM in 10 ml DMF. The mixture was stirred at room temperature for 2 hrs. then 0.5 g (1.0 mmol) of compound 218 was added and the solution was stirred at 50° C. for 12 hours. The mixture was cooled to room temperature, 100 ml ether was added and extracted with 5×50 ml water. The organic phase was dried with anhydrous magnesium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography (Hexane-EtOAc (1:1) then EtOAc (neat)) to yield 410 mg (48%) of compound 350.
  • Compound 350 A
  • 1H-NMR (CDCl3): 0.85 (dd, 6H), 1.41 (s, 3H), 1.45 (s, 6H), 1.60 (m, 4H), 1.90 (m, 2H), 2.1 (m, 1H), 2.75-3.25 (m, 6H), 3.60-3.90 (m, 6H), 5.15 (dd, 2H), 7.2-7.4 (m, 10H), 7.68 (dd, 4H).
  • MS (ES−): 852 (M−1).
  • MS (ES+): 854 (M+1).
  • Compound 350 B
  • 1H-NMR (CDCl3): 0.81 (dd, 6H), 1.39 (s, 9H), 1.40-2.10 (m, 9H), 2.70-3.20 (m, 8H), 3.60-3.90 (m, 6H), 4.10 (m, 1H), 4.80 (d, 1H), 5.04 (s(br), 2H), 7.1-7.3 (m, 10H), 7.61 (s, 4H).
  • MS (ES−): 866 (M−1).
  • MS (ES+): 868 (M+1).
  • Compound 350 C
  • 1H-NMR (CDCl3): 0.86 (dd, 6H), 1.40 (s, 3H), 1.46 (s, 6H), 1.60-2.10 (m, 7H), 2.70-3.15 (m, 6H), 3.60 (d, 1H), 3.70-4.10 (m, 6H), 4.81 (d, 1H), 5.05-5.30 (m, 7H), 7.18-7.4 (m, 17H), 7.55 (d, 2H).
  • Figure US20100124543A1-20100520-C00149
  • Compounds 350A, 350B and 350C were converted to Compounds 402, 403, and 404, respectively, using the standard hydrogenation method set forth above:
  • H2/PdC (10%), atmospheric pressure, 4 hours, room temperature, solvent: EtOH, Yield: 81%.
  • Compound 402
  • 1H-NMR (CDCl3): 0.80 (dd, 6H), 1.38 (s, 9H), 1.8 (m, 6H), 2.10 (m, 2H), 2.75-3.30 (m, 8H), 3.50-4.00 (m, 7H), 4.55 (s(br), 1H), 7.2 (m, 5H), 7.60 (d, 2H), 7.81 (d, 2H).
  • MS (ES+): 720 (M+1).
  • Compound 403
  • 1H-NMR (CDCl3): 0.87 (dd, 6H), 1.45 (s, 9H), 1.50-2.00 (m, 8H), 2.08 (m, 1H), 2.75-3.15 (m, 8H), 3.60 (d, 1H), 3.75-3.90 (m, 5H), 4.28 (s(br), 1H), 4.92 (d, 1H), 5.11 (m, 1H), 5.27 (s(br), 1H), 7.28-7.35 (m, 5H), 7.70 (s, 4H).
  • MS (ES+): 734 (M+1).
  • Compound 404
  • 1H-NMR (CDCl3): 0.80 (dd, 6H), 1.32 (s, 9H), 1.50-2.10 (m, 7H), 2.60-3.20 (m, 8H), 3.40-3.80 (m, 5H), 5.0 (s(br), 1H), 7.05-7.2 (m, 5H), 7.50-7.80 (m, 4H).
  • MS (ES+): 762 (M+1).
  • Example 47
  • Figure US20100124543A1-20100520-C00150
  • We added 5 ml TFA to a stirred solution of 260 mg (0.3 mmol) Compound 350A, 350B, or 350C in 20 ml chloroform. The mixture was stirred for 5 hours at room temperature, and then the solvents were removed under reduced pressure. The residue was dissolved in 20 ml dichloromethane, 2 ml (11 mmol) N,N-diisopropylethylamine and 1 ml (10 mmol) acetic anhydride was added to the reaction mixture. The solution was stirred for 1 hour, then the solvents were removed. The residue was purified by silica gel chromatography (eluant: EtOAc-EtOH (9:1)) to obtain 170 mg (71%) of compound 351A, 351B or 351C, respectively.
  • Compound 351A
  • 1H-NMR (CDCl3): 0.85 (dd, 6H), 1.60 (m, 3H), 1.80-2.00 (m, 3H), 2.06 (2, 3H), 2.75 (dd, 1H), 2.80-3.20 (m, 5H), 3.60-3.90 (m, 7H), 4.85 (d, 2H), 5.10 (m, 3H), 6.46 (d, 1H), 7.25 (m, 10H), 7.67 (s, 4H), 9.30 (s, 1H).
  • MS (ES+): 796 (M+1), 818 (M+Na).
  • Compound 351B
  • 1H-NMR (CDCl3): 0.80 (dd, 6H), 1.38 (m, 2H), 1.50 (m, 2H), 1.70 (m, 0.2H), 1.85 (m, 2H), 2.00 (s, 3H), 2.70 (dd, 1H), 2.75-3.20 (m, 7H), 3.55 (d, 1H), 3.75 (m, 6H), 4.45 (q, 1H), 4.83 (d, 1H), 4.95 (t, 1H), 5.03 (s(br), 3H), 6.46 (d, 1H), 7.20 (m, 10H), 7.61 (s, 4H), 9.29 (s, 1H).
  • MS (ES+): 810 (M+1), 832 (M+Na).
  • Compound 351C
  • 1H-NMR (CDCl3): 0.85 (dd, 6H), 1.70-2.00 (m, 6H), 2.07 (s, 3H), 2.70 (dd, 1H), 2.80-3.00 (m, 3H), 3.10 (dd, 1H), 3.60 (d, 1H), 3.65-4.00 (m, 6H), 4.1 (m, 1H), 4.62 (q, 1H), 4.82 (d, 1H), 5.00-5.30 (m, 5H), 7.10-7.40 (m, 15H), 7.55 (d, 2H), 7.65 (m, 3H) 9.18 (s(br), 1H), 9.45 (s(br), 1H), 9.56 (s(br), 1H).
  • MS (FAB+): 972 (M+1), 994 (M+Na).
  • Figure US20100124543A1-20100520-C00151
  • The conversion of compounds 351A, 351C, and 351C to 405, 406, and 407, respectively was achieved by standard hydrogenation using H2/PdC (10%), atmospheric pressure, 4 hours at room temperature, solvent: EtOH,
  • Yield=46%.
  • Compound 405
  • 1H-NMR (DMSO-d6): 0.85 (dd, 6H), 1.62 (m, 3H), 1.81 (m, 2H), 1.94 (s, 3H), 2.00-2.2 (m, 2H), 2.75-3.00 (m, 5H), 3.10 (m, 2H), 3.50-3.80 (m, 5H), 4.54 (m, 1H), 5.00 (m, 1H), 5.11 (d, 1H), 7.2-7.4 (m, 5H), 7.80-8.00 (m, 5H), 10.72 (s, 1H).
  • MS (ES+): 662 (M+1).
  • Compound 406
  • 1H-NMR (DMSO-d6): 0.80 (dd, 6H), 1.30-1.80 (m, 7H), 1.85 (s, 3H), 1.95-2.10 (m, 2H), 2.70 (m, 4H), 2.99 (m, 2H), 3.30 (m, 5H), 3.40-3.80 (m, 4H), 4.35 (m, 1H), 4.90 (s, 1H), 5.00 (d, 1H), 7.08-7.25 (m, 5H), 7.50 (s(br), 1H), 7.71 (d, 2H), 7.79 (d, 2H), 10.54 (s, 1H).
  • MS (ES+): 676 (M+1).
  • Compound 407
  • 1H-NMR (DMSO-d6): 0.80 (dd, 6H), 1.40-1.60 (m, 4H), 1.75 (m, 2H), 1.86 (s, 3H), 2.00 (m, 2H), 2.75 (dt, 2H), 3.00 (m, 2H), 3.10 (q, 2H), 3.40-3.70 (m, 5H), 4.39 (q, 1H), 4.92 (s (br), 1H), 5.01 (d, 1H), 7.20 (m, 5H), 7.70 (d+m, 3H), 7.81 (d, 2H), 8.30 (d, 1H), 10.60 (s, 1H).
  • MS (ES+): 704 (M+1).
  • Example 48
  • Figure US20100124543A1-20100520-C00152
  • We added 1.0 g (7.5 mmol) methanephosphonyl dichloride to a stirred solution of 2.14 g (4.00 mmol) of compound 197 in 20 ml toluene, containing 10% pyridine. The mixture was stirred at 100° C. for 5 hours, then cooled to 40° C., 2 g (18.5 mmol) benzyl alcohol was added to the reaction, and the mixture was stirred at 20° C. for 12 hours. The solid was filtered, washed with 2×10 ml toluene and the filtrate was concentrated under reduced pressure. The residue was purified using silica gel chromatography (eluants: Hexane-EtOAc (1:1), then EtOAc (neat)) to yield 550 mg (20%) of compound 352.
  • 1H-NMR (CDCl3): 0.67 (dd, 6H), 1.53 (d, 3H), 1.70 (m, 1H), 1.90-2.10 (m, 2H), 2.65-3.20 (m, 6H), 3.55 (d, 1H), 3.80 (m, 3H), 4.10 (m, 1H), 4.70 (q, 1H), 4.90-5.20 (m, 4H), 6.37 (d, 1H), 7.2-7.4 (m, 10H), 7.90 (d, 2H), 8.30 (d, 2H).
  • MS (ES+): 704 (M+1), 726 (M+Na).
  • Figure US20100124543A1-20100520-C00153
  • Compound 352 was converted to compound 408 using standard hydrogenation method: H2/PdC (10%), atmospheric pressure, 2 hours, room temperature, solvent: MeOH; Yield: 78%.
  • 1H-NMR (DMSO-d6): 0.84 (dd, 6H), 1:44 (d, 3H), 1.82 (m, 1H), 1.90-2.10 (m, 2H), 2.62 (m, 2H), 2.95 (m, 2H), 3.10 (d, 1H), 3.39 (d, 1H), 3.45-3.80 (m, 4H), 4.14 (t, 1H), 4.53 (m, 1H), 5.00 (s (br), 1H), 6.68 (d, 2H), 7.2-7.4 (m, 5H), 7.50 (d, 2H).
  • MS (ES−): 582 (M−1).
  • While we have described a number of embodiments of this invention, it is apparent that our basic constructions may be altered to provide other embodiments which utilize the products and processes of this invention. Therefore, it will be appreciated that the scope of this invention is to be defined by the appended claims, rather than by the specific embodiments which have been presented by way of example.

Claims (19)

1. A compound of formula I:
Figure US20100124543A1-20100520-C00154
wherein:
A is selected from Ht; —R1—Ht;
each R1 is independently selected from —C(O)—, —S(O)2—, —C(O)—C(O)—, —O—C(O)—, —O—S(O)2, —N(R2)—S(O)2—, —N(R2)—C(O)— or —N(R2)—C(O)—C(O)—;
each Ht is independently selected from C3-C7 cycloalkyl; C5-C7 cycloalkenyl; C6-C10 aryl; or a 5-7 membered saturated or unsaturated heterocycle, containing one or more heteroatoms selected from N, N(R2), O, S and S(O)n; wherein said aryl or said heterocycle is optionally fused to Q; and wherein any member of said Ht is optionally substituted with one or more substituents independently selected from oxo, —OR2, SR2, —R2, —N(R2)2, —R2—OH, —CN, —C(O)O—R2, —C(O)—N(R2)2, —S(O)2—N(R2)2, —N(R2)—C(O)—R2, —C(O)—R2, —S(O)n—R2, —OCF3, —S(O)n-Q, methylenedioxy, —N(R2)—S(O)2—R2, halo, —CF3, —NO2, Q, —OQ, —OR7, —SR7, —R7, —N(R2)(R7) or —N(R7)2;
each R2 is independently selected from H, or (C1-C4)-alkyl optionally substituted with Q;
B, when present, is —N(R2)—C(R3)2—C(O)—;
each x is independently 0 or 1;
each R3 is independently selected from H, Ht, (C1-C6)-alkyl, (C2-C6)-alkenyl, (C3-C6)-cycloalkyl or (C5-C6)-cycloalkenyl; wherein any member of said R3, except H, is optionally substituted with one or more substituents selected from —OR2, —C(O)—NH—R2, —S(O)n—N(R2)2, Ht, —CN, —SR2, —CO2R2, N(R2)—C(O)—R2;
each n is independently 1 or 2;
G, when present, is selected from H, R7 or (C1-C4)-alkyl, or, when G is (C1-C4)-alkyl, G and R7 are bound to one another either directly or through a C1-C3 linker to form a heterocyclic ring; or when G is absent, the atom to which G is attached is bound directly to the R7 group in —OR7 with the concomitant displacement of one -ZM group from R7;
D and D′ are independently selected from Q; (C1-C6)-alkyl, which is optionally substituted with one or more groups selected from (C3-C6)-cycloalkyl, —OR2, —R3, —O-Q or Q; (C2-C4)-alkenyl, which is optionally substituted with one or more groups selected from (C3-C6)-cycloalkyl, —OR2, —R3, —O-Q or Q; (C3-C6)-cycloalkyl, which is optionally substituted with or fused to Q; or (C5-C6)-cycloalkenyl, which is optionally substituted with or fused to Q;
each Q is independently selected from a 3-7 membered saturated, partially saturated or unsaturated carbocyclic ring system; or a 5-7 membered saturated, partially saturated or unsaturated heterocyclic ring containing one or more heteroatoms selected from O, N, S, S(O)n or N(R2); wherein any ring in Q is optionally substituted with one or more groups selected from oxo, —OR2, —R2, —N(R2)2, —N(R2)—C(O)—R2, —R2—OH, —CN, —C(O)OR2, —C(O)—N(R2)2, halo or —CF3;
E is selected from —O—R3; —N(R2)(R3); (C1-C6)-alkyl, which is optionally substituted with one or more groups selected from R4 or Ht; (C2-C6)-alkenyl, which is optionally substituted with one or more groups selected from R4 or Ht;
each R4 is independently selected from —OR2, —SR2, —C(O)—NHR2, —S(O)2—NHR2, halo, —N(R2)—C(O)—R2, —N(R2)2 or —CN;
each R7 is independently selected from
Figure US20100124543A1-20100520-C00155
wherein each M is independently selected
from H, Li, Na, K, Mg, Ca, Ba, —N(R2)4, (C1-C12)-alkyl, (C2-C12)-alkenyl, or —R6; wherein 1 to 4 —CH2 radicals of the alkyl or alkenyl group, other than the —CH2 that is bound to Z, is optionally replaced by a heteroatom group selected from O, S, S(O), S(O)2, or N(R2); and wherein any hydrogen in said alkyl, alkenyl or R6 is optionally replaced with a substituent selected from oxo, —OR2, —R2, N(R2)2, N(R2)3, R2OH, —CN, —C(O)OR2, —C(O)—N(R2)2, S(O)2—N(R2)2, N(R2)—C(O)—R2, C(O)R2, —S(O)n—R2, OCF3, —S(O)n—R6, N(R2)—S(O)2—R2, halo, —CF3, or —NO2;
M′ is H, (C1-C12)-alkyl, (C2-C12)-alkenyl, or —R6; wherein 1 to 4 —CH2 radicals of the alkyl or alkenyl group is optionally replaced by a heteroatom group selected from O, S, S(O), S(O)2, or N(R2); and wherein any hydrogen in said alkyl, alkenyl or R6 is optionally replaced with a substituent selected from oxo, —OR2, —R2, —N(R2)2, N(R2)3, —R2OH, —CN, —CO2R2, —C(O)—N(R2)2, —S(O)2—N(R2)2, —N(R2)—C(O)—R2, —C(O)R2, —S(O)n—R2, —OCF3, —S(O)n—R6, —N(R2)—S(O)2—R2, halo, —CF3, or —NO2;
Z is CH3, O, S, N(R2)2, or, when M is not present, H.
Y is P or S;
X is O or S; and
R9 is C(R2)2, O or N(R2); and wherein when Y is S, Z is not S; and
R6 is a 5-6 membered saturated, partially saturated or unsaturated carbocyclic or heterocyclic ring system, or an 8-10 membered saturated, partially saturated or unsaturated bicyclic ring system; wherein any of said heterocyclic ring systems contains one or more heteroatoms selected from O, N, S, S(O)n or N(R2); and wherein any of said ring systems optionally contains 1 to 4 substituents independently selected from OH, C1-C4 alkyl, O—(C1-C4)-alkyl or O—C(O)—(C1-C4)-alkyl.
2. The compound according to claim 1, wherein at least one R7 is selected from:
Figure US20100124543A1-20100520-C00156
Figure US20100124543A1-20100520-C00157
3. The compound according to claim 2, wherein said compound has formula XXII:
Figure US20100124543A1-20100520-C00158
wherein A, D′, R7 and E are as defined in claim 1.
4-12. (canceled)
13. The compound according to claim 2, wherein said compound has formula XXXI:
Figure US20100124543A1-20100520-C00159
14. The compound according to claim 13, wherein:
A is R1-Ht;
each R3 is independently (C1-C6)-alkyl which is optionally substituted with —OR2, —C(O)—NH—R2, —S(O)nN(R2)2, —Ht, —CN, —SR2, —CO2R2 or —N(R2)—C(O)—R2; and
D′ is (C1-C4)-alkyl, which is optionally substituted with (C3-C6)-cycloalkyl, —OR2, —O-Q; and E is —N(R2)(R3).
15. The compound according to claim 14, wherein R7 in the —OR7 group depicted in formula XXXI is —PO(OM)2 or —C(O)-M′.
16. (canceled)
17. A compound selected from:
Figure US20100124543A1-20100520-C00160
Figure US20100124543A1-20100520-C00161
wherein
R10 is selected from isopropoyl or cyclopentyl;
R11 is selected from NHR7 or OR7;
in compound 1005, when R7 is PO3M, (G)x is not H; and x, R7 and G are as defined in claim 1.
18. A pharmaceutical composition, comprising a compound according to any one of claims 1 in an amount effective to treat infection by a virus that is characterized by an aspartyl protease; and a pharmaceutically acceptable carrier, adjuvant or vehicle.
19. The pharmaceutical composition according to claim 18, wherein said virus is HIV.
20. The pharmaceutical composition according to claim 18, wherein said pharmaceutical composition is formulated for oral administration.
21. The pharmaceutical composition according to claim 18, further comprising one or more agents selected from an anti-viral agent, an HIV protease inhibitor other than a compound according to claim 1, and an immunostimulator.
22. The pharmaceutical composition according to claim 21, further comprising one or more agents selected from zidovudine (AZT), zalcitabine (ddC), didanosine (ddI), stavudine (d4T), 3TC, 935U83, 1592U89, 524W91, saquinavir (Ro 31-8959), L-735,524, SC-52151, ABT 538 (A84538), AG 1343, XM 412, XM 450, CGP 53,437, tucaresol, polysulfated polysaccharides, ganciclovir, dideoxycytidine, ribavirin, acyclovir, TIBO, nevirapine, IL-2, GM-CSF, interferon alpha, or erythropoietin (EPO).
23. A method for inhibiting aspartyl protease activity in a mammal, comprising the step of administering to said mammal a pharmaceutical composition according to claim 18.
24. A method for treating HIV infection in a mammal comprising the step of administering to said mammal a pharmaceutical composition according to claim 18.
25. The method according to claim 24, wherein said mammal is additionally administered one or more additional agents selected from an anti-viral agent, an HIV protease inhibitor other than a compound according to claim 1, and an immunostimulator either as a part of a single dosage form with said pharmaceutical composition or as a separate dosage form.
26. The method according to claim 25, wherein said additional agent is selected from zidovudine (AZT), zalcitabine (ddC), didanosine (ddI), stavudine (d4T), 3TC, 935U83, 1592U89, 524W91, saquinavir (Ro 31-8959), L-735,524, SC-52151, ABT 538 (A84538), AG 1343, XM 412, XM 450, CGP 53,437, tucaresol, polysulfated polysaccharides, ganciclovir, dideoxycytidine, ribavirin, acyclovir, TIBO, nevirapine, IL-2, GM-CSF, interferon alpha, or erythropoietin (EPO).
27. The method according to claim 24, wherein said step of administering comprises oral administration.
US12/504,243 1997-12-24 2009-07-16 Sulphonamide derivatives as prodrugs of aspartyl protease inhibitors Abandoned US20100124543A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/504,243 US20100124543A1 (en) 1997-12-24 2009-07-16 Sulphonamide derivatives as prodrugs of aspartyl protease inhibitors

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US08/998,050 US6436989B1 (en) 1997-12-24 1997-12-24 Prodrugs of aspartyl protease inhibitors
PCT/US1998/004595 WO1999033815A1 (en) 1997-12-24 1998-03-09 Sulphonamide derivatives as prodrugs of aspartyl protease inhibitors
US09/602,494 US6559137B1 (en) 1997-12-24 2000-06-23 Sulphonamide derivatives as prodrugs of aspartyl protease inhibitors
US10/370,171 US6838474B2 (en) 1997-12-24 2003-02-19 Sulphonamide derivatives as prodrugs of aspartyl protease inhibitors
US10/958,223 US7592368B2 (en) 1997-12-24 2004-10-04 Sulphonamide derivatives as prodrugs of aspartyl protease inhibitors
US12/504,243 US20100124543A1 (en) 1997-12-24 2009-07-16 Sulphonamide derivatives as prodrugs of aspartyl protease inhibitors

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/958,223 Continuation US7592368B2 (en) 1997-12-24 2004-10-04 Sulphonamide derivatives as prodrugs of aspartyl protease inhibitors

Publications (1)

Publication Number Publication Date
US20100124543A1 true US20100124543A1 (en) 2010-05-20

Family

ID=25544691

Family Applications (5)

Application Number Title Priority Date Filing Date
US08/998,050 Expired - Lifetime US6436989B1 (en) 1997-12-24 1997-12-24 Prodrugs of aspartyl protease inhibitors
US09/602,494 Expired - Lifetime US6559137B1 (en) 1997-12-24 2000-06-23 Sulphonamide derivatives as prodrugs of aspartyl protease inhibitors
US10/370,171 Expired - Lifetime US6838474B2 (en) 1997-12-24 2003-02-19 Sulphonamide derivatives as prodrugs of aspartyl protease inhibitors
US10/958,223 Expired - Fee Related US7592368B2 (en) 1997-12-24 2004-10-04 Sulphonamide derivatives as prodrugs of aspartyl protease inhibitors
US12/504,243 Abandoned US20100124543A1 (en) 1997-12-24 2009-07-16 Sulphonamide derivatives as prodrugs of aspartyl protease inhibitors

Family Applications Before (4)

Application Number Title Priority Date Filing Date
US08/998,050 Expired - Lifetime US6436989B1 (en) 1997-12-24 1997-12-24 Prodrugs of aspartyl protease inhibitors
US09/602,494 Expired - Lifetime US6559137B1 (en) 1997-12-24 2000-06-23 Sulphonamide derivatives as prodrugs of aspartyl protease inhibitors
US10/370,171 Expired - Lifetime US6838474B2 (en) 1997-12-24 2003-02-19 Sulphonamide derivatives as prodrugs of aspartyl protease inhibitors
US10/958,223 Expired - Fee Related US7592368B2 (en) 1997-12-24 2004-10-04 Sulphonamide derivatives as prodrugs of aspartyl protease inhibitors

Country Status (43)

Country Link
US (5) US6436989B1 (en)
EP (2) EP1944300A3 (en)
JP (3) JP3736964B2 (en)
KR (1) KR100520737B1 (en)
CN (2) CN101565412A (en)
AP (1) AP1172A (en)
AR (1) AR017965A1 (en)
AT (1) ATE382042T1 (en)
AU (1) AU755087B2 (en)
BG (1) BG64869B1 (en)
BR (1) BR9814480A (en)
CA (1) CA2231700C (en)
CO (1) CO4990992A1 (en)
CZ (1) CZ301653B6 (en)
DE (2) DE69838903T2 (en)
DK (1) DK0933372T3 (en)
EA (1) EA003509B1 (en)
EE (1) EE04466B1 (en)
ES (1) ES2299193T3 (en)
FR (1) FR08C0015I2 (en)
HK (1) HK1021737A1 (en)
HU (2) HU229596B1 (en)
ID (1) ID24962A (en)
IL (2) IL136941A0 (en)
IS (1) IS2817B (en)
LU (1) LU91426I2 (en)
ME (1) MEP82008A (en)
MY (1) MY131525A (en)
NL (1) NL300339I2 (en)
NO (2) NO326265B1 (en)
NZ (1) NZ505776A (en)
OA (1) OA11468A (en)
PE (1) PE20000048A1 (en)
PL (1) PL202845B1 (en)
PT (1) PT933372E (en)
RS (1) RS52483B (en)
SI (1) SI0933372T1 (en)
SK (1) SK287123B6 (en)
TR (1) TR200002615T2 (en)
TW (1) TW486474B (en)
UA (1) UA72733C2 (en)
WO (1) WO1999033815A1 (en)
ZA (1) ZA9811830B (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090274650A1 (en) * 1999-06-11 2009-11-05 Hale Michael R Inhibitors of aspartyl protease
WO2013009844A2 (en) * 2011-07-11 2013-01-17 Purdue Research Foundation C-3 substituted bicyclooctane based hiv protease inhibitors

Families Citing this family (154)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
UA59384C2 (en) * 1996-12-20 2003-09-15 Пфайзер, Інк. Preventing bone mass loss and recovery thereof by means of prostaglandin agonists
US6436989B1 (en) * 1997-12-24 2002-08-20 Vertex Pharmaceuticals, Incorporated Prodrugs of aspartyl protease inhibitors
US6686366B1 (en) 1998-06-02 2004-02-03 Osi Pharmaceuticals, Inc. Compounds specific to adenosine A3 receptor and uses thereof
US6878716B1 (en) 1998-06-02 2005-04-12 Osi Pharmaceuticals, Inc. Compounds specific to adenosine A1 receptor and uses thereof
BR9911612A (en) 1998-06-02 2001-02-06 Osi Pharm Inc Pyrrole [2,3d] pyrimidine compositions and their uses
GB9815567D0 (en) * 1998-07-18 1998-09-16 Glaxo Group Ltd Antiviral compound
AU6329599A (en) * 1998-09-28 2000-04-17 Glaxo Group Limited Antiviral combinations comprising (s)-2-ethyl -7-fluoro -3-oxo-3, 4-dihydro -2h-quinoxaline -1-carboxylic acid isopropyl ester
GB9914821D0 (en) * 1999-06-24 1999-08-25 Glaxo Group Ltd Compounds
US7160890B2 (en) 1999-12-02 2007-01-09 Osi Pharmaceuticals, Inc. Compounds specific to adenosine A3 receptor and uses thereof
US6664252B2 (en) 1999-12-02 2003-12-16 Osi Pharmaceuticals, Inc. 4-aminopyrrolo[2,3-d]pyrimidine compounds specific to adenosine A2a receptor and uses thereof
US6680322B2 (en) 1999-12-02 2004-01-20 Osi Pharmaceuticals, Inc. Compounds specific to adenosine A1 receptors and uses thereof
US6673802B2 (en) 2000-12-01 2004-01-06 Osi Pharmaceuticals, Inc. Compounds specific to adenosine A3 receptor and uses thereof
US6680324B2 (en) 2000-12-01 2004-01-20 Osi Pharmaceuticals, Inc. Compounds specific to adenosine A1 receptors and uses thereof
PL213327B1 (en) 2001-02-14 2013-02-28 Tibotec Pharm Ltd Broadspectrum 2-(substituted-amino)-benzothiazole sulfonamide hiv protease inhibitors
EE05384B1 (en) 2001-04-09 2011-02-15 Tibotec�Pharmaceuticals�Ltd. Broad-spectrum Á2- (Substituted Áamino) Benzoxazole Sulphonamides ÁkuiÁHIVÁProtease Inhibitors, Use, Pharmaceutical Formulations, and Method for Inhibiting Antiretroviral Replication
AP1652A (en) 2001-05-11 2006-08-11 Tibotec Pharm Ltd Broadspectrum 2-amino-benzoxazole sulfonamide HIV protease inhibitors.
US7576084B2 (en) * 2001-10-12 2009-08-18 Choongwae Pharma Corporation Reverse-turn mimetics and method relating thereto
EP2050751A1 (en) 2001-11-30 2009-04-22 OSI Pharmaceuticals, Inc. Compounds specific to adenosine A1 and A3 receptors and uses thereof
AR037797A1 (en) * 2001-12-12 2004-12-01 Tibotec Pharm Ltd COMBINATION OF PROTEASA INHIBITORS DEPENDENTS OF CITOCROMO P450
CN101973998A (en) 2001-12-20 2011-02-16 Osi药物公司 Pyrrolopyrimidine A2b selective antagonist compounds, their synthesis and use
WO2003053366A2 (en) 2001-12-20 2003-07-03 Osi Pharmaceuticals, Inc. Pyrimidine a2b selective antagonist compounds, their synthesis and use
CA2470964C (en) 2001-12-21 2013-07-02 Tibotec Pharmaceuticals Ltd. Broadspectrum heterocyclic substituted phenyl containing sulfonamide hiv protease inhibitors
MY142238A (en) 2002-03-12 2010-11-15 Tibotec Pharm Ltd Broadspectrum substituted benzimidazole sulfonamide hiv protease inhibitors
US7157489B2 (en) * 2002-03-12 2007-01-02 The Board Of Trustees Of The University Of Illinois HIV protease inhibitors
BR0309557A (en) * 2002-04-26 2005-03-01 Gilead Sciences Inc Non-Nucleoside Reverse Transcriptase Inhibitors
EP1517899B1 (en) 2002-05-17 2007-08-29 Tibotec Pharmaceuticals Ltd. Broadspectrum substituted benzisoxazole sulfonamide hiv protease inhibitors
EA010486B1 (en) 2002-08-14 2008-10-30 Тиботек Фармасьютикалз Лтд. Broadspectrum substituted oxindole sulfonamide hiv protease inhibitors
DE10303974A1 (en) 2003-01-31 2004-08-05 Abbott Gmbh & Co. Kg Amyloid β (1-42) oligomers, process for their preparation and their use
CA2425031A1 (en) * 2003-04-01 2004-10-01 Smithkline Beecham Corporation Pharmaceutical compositions
DK1628685T3 (en) 2003-04-25 2011-03-21 Gilead Sciences Inc Antiviral phosphonate analogues
US7470724B2 (en) 2003-04-25 2008-12-30 Gilead Sciences, Inc. Phosphonate compounds having immuno-modulatory activity
WO2004096234A2 (en) 2003-04-25 2004-11-11 Gilead Sciences, Inc. Kinase inhibitor phosphonate conjugates
WO2004096287A2 (en) 2003-04-25 2004-11-11 Gilead Sciences, Inc. Inosine monophosphate dehydrogenase inhibitory phosphonate compounds
US7452901B2 (en) 2003-04-25 2008-11-18 Gilead Sciences, Inc. Anti-cancer phosphonate analogs
US7432261B2 (en) 2003-04-25 2008-10-07 Gilead Sciences, Inc. Anti-inflammatory phosphonate compounds
US7407965B2 (en) 2003-04-25 2008-08-05 Gilead Sciences, Inc. Phosphonate analogs for treating metabolic diseases
WO2004096285A2 (en) 2003-04-25 2004-11-11 Gilead Sciences, Inc. Anti-infective phosphonate conjugates
WO2005002626A2 (en) 2003-04-25 2005-01-13 Gilead Sciences, Inc. Therapeutic phosphonate compounds
CN1845897A (en) 2003-07-09 2006-10-11 帕拉特克药品公司 Substituted tetracycline compounds
WO2005009944A1 (en) * 2003-07-09 2005-02-03 Paratek Pharmaceuticals, Inc. Prodrugs of 9-aminomethyl tetracycline compounds
US20050119163A1 (en) 2003-09-18 2005-06-02 The Government Of The United States Of America, As Represented By The Secretary, SH2 domain binding inhibitors
US7427624B2 (en) 2003-10-24 2008-09-23 Gilead Sciences, Inc. Purine nucleoside phosphorylase inhibitory phosphonate compounds
WO2005044308A1 (en) 2003-10-24 2005-05-19 Gilead Sciences, Inc. Phosphonate analogs of antimetabolites
US7834043B2 (en) * 2003-12-11 2010-11-16 Abbott Laboratories HIV protease inhibiting compounds
US8193227B2 (en) 2003-12-11 2012-06-05 Abbott Laboratories HIV protease inhibiting compounds
US20050131042A1 (en) * 2003-12-11 2005-06-16 Flentge Charles A. HIV protease inhibiting compounds
DK1699455T3 (en) * 2003-12-15 2013-08-05 Merck Sharp & Dohme HETEROCYCLIC ASPARTYL PROTEASE INHIBITORS
UA83881C2 (en) 2003-12-18 2008-08-26 Янссен Фармацевтика Н.В. Pyrido- and pyrimidopyrimidine derivatives as anti-proliferative agents
MXPA06006899A (en) 2003-12-22 2006-09-04 Gilead Sciences Inc 4aCO-SUBSTITUTED CARBOVIR-AND ABACAVIR-DERIVATIVES AS WELL AS RELATED COMPOUNDS WITH HIV AND HCV ANTIVIRAL ACTIVITY.
CA2571726C (en) 2004-07-06 2013-05-21 Abbott Laboratories Prodrugs of hiv protease inhibitors
CN101027062A (en) 2004-07-27 2007-08-29 吉里德科学公司 Phosphonate analogs of HIV inhibitor compounds
EP1773763B1 (en) * 2004-08-02 2008-12-17 Ambrilia Biopharma Inc. Lysine based compounds
US7388008B2 (en) * 2004-08-02 2008-06-17 Ambrilia Biopharma Inc. Lysine based compounds
WO2006024491A1 (en) 2004-08-30 2006-03-09 Interstitial Therapeutics Methods and compositions for the treatment of cell proliferation
CN101098698A (en) 2004-12-01 2008-01-02 德福根有限公司 5-carboxamido substitued thiazole derivatives that interact with ion channels, in particular with ion channels from the kv family
NI200700147A (en) 2004-12-08 2019-05-10 Janssen Pharmaceutica Nv QUINAZOLINE DERIVATIVES KINE INHIBITORS TARGETING MULTIP
EP1941799A3 (en) 2004-12-17 2008-09-17 Devgen NV Nematicidal compositions
RU2416616C2 (en) * 2005-01-19 2011-04-20 Райджел Фармасьютикалз, Инк. Prodrugs of 2,4-pyrimidine diamine compounds and use thereof
EP1855672A4 (en) * 2005-03-11 2011-11-30 Glaxosmithkline Llc Hiv protease inhibitors
AR053845A1 (en) 2005-04-15 2007-05-23 Tibotec Pharm Ltd 5-TIAZOLILMETIL [(1S, 2R) -3 - [[(2-AMINO-6-BENZOXAZOLIL) SULFONIL)] (2-METHYLPROPIL) AMINO] -2-HYDROXY-1- (PHENYLMETIL) PROPIL] CARBAMATE AS A PHARMACY IMPROVER METABOLIZED BY THE CYCROCHROME P450
WO2006114001A1 (en) 2005-04-27 2006-11-02 Ambrilia Biopharma Inc. Method for improving pharmacokinetics of protease inhibitors and protease inhibitor precursors
EP1940856B1 (en) 2005-10-21 2014-10-08 Universiteit Antwerpen Novel urokinase inhibitors
TWI385173B (en) 2005-11-28 2013-02-11 Tibotec Pharm Ltd Substituted aminophenylsulfonamide compounds as hiv protease inhibitor
TWI432438B (en) 2005-11-28 2014-04-01 Tibotec Pharm Ltd Substituted aminophenylsulfonamide compounds and derivatives as hiv protease inhibitor
SG10201706600VA (en) 2005-11-30 2017-09-28 Abbvie Inc Monoclonal antibodies and uses thereof
PT1954718E (en) 2005-11-30 2014-12-16 Abbvie Inc Anti-a globulomer antibodies, antigen-binding moieties thereof, corresponding hybridomas, nucleic acids, vectors, host cells, methods of producing said antibodies, compositions comprising said antibodies, uses of said antibodies and methods of using said antibodies
EP1971615B1 (en) * 2005-11-30 2014-01-01 TaiMed Biologics, Inc. Lysme-based prodrugs of aspartyl protease inhibitors and processes for their preparation
CN101346390B (en) * 2005-12-27 2011-11-09 大塚制药株式会社 Water-soluble benzoazepine compound and its pharmaceutical composition
JP2009542778A (en) 2006-07-13 2009-12-03 ジヤンセン・フアーマシユーチカ・ナームローゼ・フエンノートシヤツプ MTKI quinazoline derivative
EP2063918B1 (en) 2006-09-08 2014-02-26 Piramal Imaging SA Compounds and methods for 18f labeled agents
CA2664118A1 (en) 2006-09-21 2008-07-03 Ambrilia Biopharma Inc. Protease inhibitors
US8455626B2 (en) 2006-11-30 2013-06-04 Abbott Laboratories Aβ conformer selective anti-aβ globulomer monoclonal antibodies
US20100311767A1 (en) 2007-02-27 2010-12-09 Abbott Gmbh & Co. Kg Method for the treatment of amyloidoses
AU2008226823B2 (en) 2007-03-12 2014-03-13 Nektar Therapeutics Oligomer-protease inhibitor conjugates
EP2185562B1 (en) 2007-07-27 2015-12-02 Janssen Pharmaceutica, N.V. Pyrrolopyrimidines useful for the treatment of proliferative diseases
EP2053033A1 (en) 2007-10-26 2009-04-29 Bayer Schering Pharma AG Compounds for use in imaging, diagnosing and/or treatment of diseases of the central nervous system or of tumors
EP2100900A1 (en) 2008-03-07 2009-09-16 Universitätsspital Basel Bombesin analog peptide antagonist conjugates
DK2283024T3 (en) 2008-03-10 2013-08-05 Janssen Pharmaceutica Nv 4-aryl-2-aniline pyrimidines as PLK kinase inhibitors
US9095620B2 (en) * 2008-03-12 2015-08-04 Nektar Therapeutics Reagents
EP2116236A1 (en) 2008-04-21 2009-11-11 Université de Mons-Hainaut Bisbenzamidine derivatives for use as antioxidant
CA2729769C (en) 2008-07-08 2017-09-05 Gilead Sciences, Inc. Salts of hiv inhibitor compounds
CA2745295C (en) 2008-12-09 2017-01-10 Gilead Sciences, Inc. Modulators of toll-like receptors
EP2432788A1 (en) * 2009-05-20 2012-03-28 Ranbaxy Laboratories Limited Amorphous fosamprenavir calcium
EP2440249A2 (en) 2009-06-12 2012-04-18 Nektar Therapeutics Covalent conjugates comprising a protease inhibitor, a water-soluble, non-peptidic oligomer and a lipophilic moiety
US9085592B2 (en) 2009-09-16 2015-07-21 Ranbaxy Laboratories Limited Process for the preparation of fosamprenavir calcium
WO2011061590A1 (en) 2009-11-17 2011-05-26 Hetero Research Foundation Novel carboxamide derivatives as hiv inhibitors
EP2501431B1 (en) 2009-11-19 2020-01-08 Wellinq Medical B.V. Narrow profile composition-releasing expandable medical balloon catheter
WO2011085130A1 (en) 2010-01-07 2011-07-14 Pliva Hrvatska D.O.O. Solid state forms of fosamprenavir calcium salt and process for preparation thereof
MX367937B (en) 2010-01-27 2019-09-12 Viiv Healthcare Co Antiviral therapy.
US20110223131A1 (en) 2010-02-24 2011-09-15 Gilead Sciences, Inc. Antiviral compounds
US20110224443A1 (en) * 2010-03-15 2011-09-15 Venkata Naga Brahmeshwara Rao Mandava Preparation of fosamprenavir calcium
WO2011114212A1 (en) 2010-03-19 2011-09-22 Lupin Limited Ammonium, calcium and tris salts of fosamprenavir
MX336196B (en) 2010-04-15 2016-01-11 Abbvie Inc Amyloid-beta binding proteins.
WO2011141515A1 (en) 2010-05-14 2011-11-17 Bayer Pharma Aktiengesellschaft Diagnostic agents for amyloid beta imaging
WO2011158259A1 (en) 2010-06-18 2011-12-22 Matrix Laboratories Ltd Novel process for the preparation of (3s)-tetrahydrofuran-3-yl (is, 2r)-3-[[(4-aminophenyl) sulfonyl] (isobutyl) amino]-1-benzyl-2-(phosphonooxy) propylcarbamate and its pharmaceutically acceptable salts thereof
US8785648B1 (en) 2010-08-10 2014-07-22 The Regents Of The University Of California PKC-epsilon inhibitors
WO2012024187A1 (en) 2010-08-14 2012-02-23 Abbott Laboratories Amyloid-beta binding proteins
WO2012032389A2 (en) 2010-09-10 2012-03-15 Lupin Limited Process for preparation of substantially pure fosamprenavir calcium and its intermediates
GB201019043D0 (en) 2010-11-10 2010-12-22 Protea Biopharma N V Use of 2',5'-oligoadenylate derivative compounds
WO2012085625A1 (en) 2010-12-21 2012-06-28 Lupin Limited Process for the preparation of fosamprenavir calcium and intermediate used in its preparation
JP2014513044A (en) * 2011-02-10 2014-05-29 マイラン ラボラトリーズ リミテッド Phosamprenavir calcium crystals and method for preparing the same
KR102395085B1 (en) 2011-06-21 2022-05-09 알닐람 파마슈티칼스 인코포레이티드 Angiopoietin-like 3(angptl3) irna compostions and methods of use thereof
WO2012178033A2 (en) 2011-06-23 2012-12-27 Alnylam Pharmaceuticals, Inc. Serpina1 sirnas: compositions of matter and methods of treatment
WO2013011485A1 (en) 2011-07-20 2013-01-24 Ranbaxy Laboratories Limited Process for the preparation of sulfonamides useful as retroviral protease inhibitors
US9233943B2 (en) 2012-01-10 2016-01-12 Council Of Scientific & Industrial Research Process for synthesis of syn azido epdxide and its use as intermediate for the synthesis of amprenavir and saquinavir
US9127274B2 (en) 2012-04-26 2015-09-08 Alnylam Pharmaceuticals, Inc. Serpinc1 iRNA compositions and methods of use thereof
EP2700396A3 (en) 2012-06-20 2015-04-29 Sylphar Nv Strip for the delivery of oral care compositions
US9877981B2 (en) 2012-10-09 2018-01-30 President And Fellows Of Harvard College NAD biosynthesis and precursors for the treatment and prevention of cancer and proliferation
EP2912047B1 (en) 2012-10-29 2016-08-24 Cipla Limited Antiviral phosphonate analogues and process for preparation thereof
ES2657608T3 (en) 2012-12-05 2018-03-06 Alnylam Pharmaceuticals, Inc. Compositions of pcsk9 arni and methods of use thereof
KR102605775B1 (en) 2013-03-14 2023-11-29 알닐람 파마슈티칼스 인코포레이티드 Complement component c5 irna compositions and methods of use thereof
RS57418B1 (en) 2013-05-22 2018-09-28 Alnylam Pharmaceuticals Inc Serpina1 irna compositions and methods of use thereof
TWI727917B (en) 2013-05-22 2021-05-21 美商阿尼拉製藥公司 TMPRSS6 iRNA COMPOSITIONS AND METHODS OF USE THEREOF
IL282401B (en) 2013-12-12 2022-08-01 Alnylam Pharmaceuticals Inc Complement component irna compositions and methods of use thereof
WO2015123264A1 (en) 2014-02-11 2015-08-20 Alnylam Pharmaceuticals, Inc. Ketohexokinase (khk) irna compositions and methods of use thereof
WO2015175510A1 (en) 2014-05-12 2015-11-19 Alnylam Pharmaceuticals, Inc. Methods and compositions for treating a serpinc1-associated disorder
EA201692370A1 (en) 2014-05-22 2017-03-31 Элнилэм Фармасьютикалз, Инк. COMPOSITIONS of mRNA ANGIOTENZINOGENA (AGT) AND METHODS OF THEIR USE
EP3180003B1 (en) 2014-07-01 2022-01-12 The Regents of the University of California Pkc-epsilon inhibitors
WO2016001907A1 (en) 2014-07-02 2016-01-07 Prendergast Patrick T Mogroside iv and mogroside v as agonist/stimulator/un-blocking agent for toll-like receptor 4 and adjuvant for use in human/animal vaccine and to stimulate immunity against disease agents.
TWI733652B (en) 2014-07-11 2021-07-21 美商基利科學股份有限公司 Modulators of toll-like receptors for the treatment of hiv
EP3191591A1 (en) 2014-09-12 2017-07-19 Alnylam Pharmaceuticals, Inc. Polynucleotide agents targeting complement component c5 and methods of use thereof
EP3207138B1 (en) 2014-10-17 2020-07-15 Alnylam Pharmaceuticals, Inc. Polynucleotide agents targeting aminolevulinic acid synthase-1 (alas1) and uses thereof
WO2016069955A1 (en) 2014-10-29 2016-05-06 Wisconsin Alumni Research Foundation Boronic acid inhibitors of hiv protease
EP3212794B1 (en) 2014-10-30 2021-04-07 Genzyme Corporation Polynucleotide agents targeting serpinc1 (at3) and methods of use thereof
CN107250362B (en) 2014-11-17 2021-10-22 阿尔尼拉姆医药品有限公司 Apolipoprotein C3(APOC3) iRNA compositions and methods of use thereof
WO2016083490A1 (en) 2014-11-27 2016-06-02 Remynd Nv Compounds for the treatment of amyloid-associated diseases
CA2976445A1 (en) 2015-02-13 2016-08-18 Alnylam Pharmaceuticals, Inc. Patatin-like phospholipase domain containing 3 (pnpla3) irna compositions and methods of use thereof
WO2016168286A1 (en) 2015-04-13 2016-10-20 Alnylam Pharmaceuticals, Inc. Angiopoietin-like 3 (angptl3) irna compositions and methods of use thereof
US10603334B2 (en) 2015-04-28 2020-03-31 NewSouth Innovation Pty. Limited Targeting NAD+ to treat chemotherapy and radiotherapy induced cognitive impairment, neuropathies and inactivity
KR20180002688A (en) 2015-05-06 2018-01-08 알닐람 파마슈티칼스 인코포레이티드 (F12), calichein B, plasma (Fletcher factor) 1 (KLKB1) and kininogen 1 (KNG1) iRNA compositions and methods for their use
WO2016205323A1 (en) 2015-06-18 2016-12-22 Alnylam Pharmaceuticals, Inc. Polynucleotde agents targeting hydroxyacid oxidase (glycolate oxidase, hao1) and methods of use thereof
WO2017048620A1 (en) 2015-09-14 2017-03-23 Alnylam Pharmaceuticals, Inc. Polynucleotide agents targeting patatin-like phospholipase domain containing 3 (pnpla3) and methods of use thereof
WO2017048727A1 (en) 2015-09-15 2017-03-23 Gilead Sciences, Inc. Modulators of toll-like recptors for the treatment of hiv
CN108601795A (en) 2015-12-07 2018-09-28 建新公司 Method and composition for treating SERPINC1 associated diseases
JP2018536689A (en) 2015-12-10 2018-12-13 アルナイラム ファーマシューティカルズ, インコーポレイテッドAlnylam Pharmaceuticals, Inc. Sterol regulatory element binding protein (SREBP) chaperone (SCAP) iRNA compositions and methods of use thereof
US20190256845A1 (en) 2016-06-10 2019-08-22 Alnylam Pharmaceuticals, Inc. COMPLEMENT COMPONENT C5 iRNA COMPOSITIONS AND METHODS OF USE THEREOF FOR TREATING PAROXYSMAL NOCTURNAL HEMOGLOBINURIA (PNH)
TWI788312B (en) 2016-11-23 2023-01-01 美商阿尼拉製藥公司 SERPINA1 iRNA COMPOSITIONS AND METHODS OF USE THEREOF
KR20230166146A (en) 2016-12-16 2023-12-06 알닐람 파마슈티칼스 인코포레이티드 Methods for treating or preventing ttr-associated diseases using transthyretin(ttr) irna compositions
WO2018206760A1 (en) 2017-05-11 2018-11-15 Remynd N.V. Compounds for the treatment of epilepsy, neurodegenerative disorders and other cns disorders
KR20200031658A (en) 2017-07-21 2020-03-24 비이브 헬쓰케어 컴퍼니 Therapy to treat HIB infection and AIDS
US10851125B2 (en) 2017-08-01 2020-12-01 Gilead Sciences, Inc. Crystalline forms of ethyl ((S)-((((2R,5R)-5-(6-amino-9H-purin-9-yl)-4-fluoro-2,5-dihydrofuran-2-yl)oxy)methyl)(phenoxy)phosphoryl(-L-alaninate
WO2019089922A1 (en) 2017-11-01 2019-05-09 Alnylam Pharmaceuticals, Inc. Complement component c3 irna compositions and methods of use thereof
BR112021001613A2 (en) 2018-08-13 2021-05-04 Alnylam Pharmaceuticals, Inc. double-stranded ribonucleic acid agents, cell, pharmaceutical compositions, methods of inhibiting gene expression, inhibiting replication and treating a subject, methods for reducing the level of an antigen and for reducing viral load, and use of an agent dsrna
SG11202107669WA (en) 2019-01-16 2021-08-30 Genzyme Corp Serpinc1 irna compositions and methods of use thereof
WO2021154941A1 (en) 2020-01-31 2021-08-05 Alnylam Pharmaceuticals, Inc. Complement component c5 irna compositions for use in the treatment of amyotrophic lateral sclerosis (als)
MX2022010413A (en) 2020-02-24 2022-09-07 Univ Leuven Kath Pyrrolopyridine and imidazopyridine antiviral compounds.
WO2021209563A1 (en) 2020-04-16 2021-10-21 Som Innovation Biotech, S.A. Compounds for use in the treatment of viral infections by respiratory syndrome-related coronavirus
WO2022136486A1 (en) 2020-12-22 2022-06-30 Luxembourg Institute Of Health (Lih) Conolidine analogues as selective ackr3 modulators for the treatment of cancer and cardiovascular diseases
US20240002351A1 (en) 2021-03-04 2024-01-04 Universiteit Antwerpen Quinazolin-4-one and thieno[2,3-d]pyrimidin-4-one inhibitors of erbb4 (her4) for use in the treatment of cancer
CA3217589A1 (en) 2021-05-31 2022-12-08 Frederik Giesel Improved prostate-specific membrane antigen targeting radiopharmaceuticals and uses thereof
WO2023021132A1 (en) 2021-08-18 2023-02-23 Katholieke Universiteit Leuven 6-substituted- and 6,7-disubstituted-7-deazapurine ribonucleoside analogues
WO2023046900A1 (en) 2021-09-23 2023-03-30 Katholieke Universiteit Leuven Ribonucleoside analogues against -sars-cov-2
WO2023241799A1 (en) 2022-06-15 2023-12-21 Université Libre de Bruxelles Flavanols for use in the treatment of retroviral infections
WO2024062043A1 (en) 2022-09-21 2024-03-28 Universiteit Antwerpen Substituted phenothiazines as ferroptosis inhibitors

Citations (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3743722A (en) * 1971-07-14 1973-07-03 Abbott Lab Anti-coagulant isolation
US4330542A (en) * 1979-06-14 1982-05-18 Sanofi N(Sulfonyl)anilines for treating angina pectoris
US4629724A (en) * 1984-12-03 1986-12-16 E. R. Squibb & Sons, Inc. Amino acid ester and amide renin inhibitors
US5196438A (en) * 1989-12-11 1993-03-23 Hoffmann-La Roche Inc. Amino acid derivatives
US5354866A (en) * 1989-05-23 1994-10-11 Abbott Laboratories Retroviral protease inhibiting compounds
US5585397A (en) * 1992-09-08 1996-12-17 Vertex Pharmaceuticals, Incorporated Sulfonamide inhibitors of aspartyl protease
US5646180A (en) * 1995-12-05 1997-07-08 Vertex Pharmaceuticals Incorporated Treatment of the CNS effects of HIV
US5723490A (en) * 1992-09-08 1998-03-03 Vertex Pharmaceuticals Incorporated THF-containing sulfonamide inhibitors of aspartyl protease
US5750493A (en) * 1995-08-30 1998-05-12 Raymond F. Schinazi Method to improve the biological and antiviral activity of protease inhibitors
US5783701A (en) * 1992-09-08 1998-07-21 Vertex Pharmaceuticals, Incorporated Sulfonamide inhibitors of aspartyl protease
US5843946A (en) * 1992-08-25 1998-12-01 G.D. Searle & Co. α-and β-amino acid hydroxyethylamino sulfonamides useful as retroviral protease inhibitors
US6180634B1 (en) * 1997-11-13 2001-01-30 Merck & Co., Inc. Combination therapy for the treatment of AIDS
US6319946B1 (en) * 1999-02-12 2001-11-20 Vertex Pharmaceuticals Incorporated Inhibitors of aspartyl protease
US6436989B1 (en) * 1997-12-24 2002-08-20 Vertex Pharmaceuticals, Incorporated Prodrugs of aspartyl protease inhibitors

Family Cites Families (37)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS5946252A (en) 1982-09-09 1984-03-15 Dainippon Ink & Chem Inc Fluorine-containing aminocarboxylate and its preparation
JPS5948449A (en) 1982-09-13 1984-03-19 Dainippon Ink & Chem Inc Straight-chain fluorine-containing anionic compound and its preparation
JPS6171830A (en) 1984-09-17 1986-04-12 Dainippon Ink & Chem Inc Cationic surfactant
US4616088A (en) 1984-10-29 1986-10-07 E. R. Squibb & Sons, Inc. Amino acid ester and amide renin inhibitor
DE3635907A1 (en) 1986-10-22 1988-04-28 Merck Patent Gmbh HYDROXY AMINO ACID DERIVATIVES
GB2200115B (en) 1987-01-21 1990-11-14 Sandoz Ltd Novel peptide derivatives, their production and use
CA1340588C (en) 1988-06-13 1999-06-08 Balraj Krishan Handa Amino acid derivatives
IL91780A (en) 1988-10-04 1995-08-31 Abbott Lab Renin inhibiting hexanoic acid amide derivatives, process for their preparation and pharmaceutical compositions containing them
WO1990007329A1 (en) 1989-01-06 1990-07-12 The Regents Of The University Of California Selection method for pharmacologically active compounds
US5151438A (en) 1989-05-23 1992-09-29 Abbott Laboratories Retroviral protease inhibiting compounds
IE902295A1 (en) 1989-07-07 1991-01-16 Abbott Lab Amino acid analog cck antagonists
WO1991018866A2 (en) 1990-06-01 1991-12-12 The Du Pont Merck Pharmaceutical Company 1,4-diamino-2,3-dihydroxybutanes
TW225540B (en) 1990-06-28 1994-06-21 Shionogi & Co
EP0554400B1 (en) 1990-11-19 1997-07-23 Monsanto Company Retroviral protease inhibitors
EP0813867B1 (en) 1990-11-19 2005-06-01 Monsanto Company Retroviral protease inhibitors
CA2096407C (en) 1990-11-19 2007-10-02 Kathryn Lea Reed Retroviral protease inhibitors
DK0558657T3 (en) 1990-11-19 1997-07-07 Monsanto Co Retroviral protease inhibitors
IE20010533A1 (en) 1990-11-20 2003-03-05 Abbott Lab Intermediates for preparing retroviral protease inhibiting compounds
ES2112880T3 (en) 1991-11-08 1998-04-16 Merck & Co Inc HIV PROTEASE INHIBITORS USEFUL FOR AIDS TREATMENT.
CA2131182C (en) 1992-05-20 2005-04-26 John S. Ng Method for making intermediates useful in synthesis of retroviral protease inhibitors
CA2136312A1 (en) 1992-05-21 1993-11-25 Michael Clare Retroviral protease inhibitors
DE69311810T2 (en) 1992-08-25 1997-11-27 Searle & Co N- (ALKANOYLAMINO-2-HYDROXYPROPYL) SULFONAMIDES CAN BE USED AS RETROVIRAL PROTEASE INHIBITORS
ES2112430T3 (en) 1992-08-25 1998-04-01 Searle & Co SULFONILALCANOYLAMINE HYDROXYETHYLAMINE SULFONAMIDES USEFUL AS RETROVIRAL PROTEASE INHIBITORS.
AU677792B2 (en) * 1992-09-03 1997-05-08 Boehringer Ingelheim Kg New aminoacid derivates, process for producing the same and pharmaceutical compositions containing these compounds
TW372972B (en) 1992-10-23 1999-11-01 Novartis Ag Antiretroviral acyl compounds
EP0666843B1 (en) 1992-10-30 1999-08-18 G.D. Searle & Co. Sulfonylalkanoylamino hydroxyethylamino sulfamic acids useful as retroviral protease inhibitors
EP0666842B1 (en) 1992-10-30 1998-06-24 G.D. Searle & Co. Hydroxyethylamino sulfamic acid derivatives useful as retroviral protease inhibitors
US5484926A (en) 1993-10-07 1996-01-16 Agouron Pharmaceuticals, Inc. HIV protease inhibitors
WO1994018192A1 (en) 1993-02-12 1994-08-18 Merck & Co., Inc. Piperazine derivatives as hiv protease inhibitors
MY128102A (en) 1993-02-17 2007-01-31 Chugai Pharmaceutical Co Ltd Indolin-2-one-derivatives
ES2127938T3 (en) 1993-08-24 1999-05-01 Searle & Co HYDROXYETHYLAMINE SULPHONAMIDES USEFUL AS RETROVIRAL PROTEASE INHIBITORS.
IL110898A0 (en) 1993-09-10 1994-11-28 Narhex Australia Pty Ltd Polar-substituted hydrocarbons
IL111584A0 (en) 1993-11-18 1995-01-24 Merck & Co Inc Prodrugs of an inhibitor of hiv protease and pharmaceutical compositions containing them
US5527829A (en) 1994-05-23 1996-06-18 Agouron Pharmaceuticals, Inc. HIV protease inhibitors
DE19506742A1 (en) 1995-02-27 1996-08-29 Bayer Ag Use of quinoxalines in combination with protease inhibitors as medicaments for the treatment of AIDS and / or HIV infections
US5691372A (en) 1995-04-19 1997-11-25 Vertex Pharmaceuticals Incorporated Oxygenated-Heterocycle containing sulfonamide inhibitors of aspartyl protease
JP2001527062A (en) * 1997-12-24 2001-12-25 バーテックス ファーマシューティカルズ インコーポレイテッド Prodrugs of aspartyl protease inhibitors

Patent Citations (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3743722A (en) * 1971-07-14 1973-07-03 Abbott Lab Anti-coagulant isolation
US4330542A (en) * 1979-06-14 1982-05-18 Sanofi N(Sulfonyl)anilines for treating angina pectoris
US4629724A (en) * 1984-12-03 1986-12-16 E. R. Squibb & Sons, Inc. Amino acid ester and amide renin inhibitors
US5354866A (en) * 1989-05-23 1994-10-11 Abbott Laboratories Retroviral protease inhibiting compounds
US5196438A (en) * 1989-12-11 1993-03-23 Hoffmann-La Roche Inc. Amino acid derivatives
US5843946A (en) * 1992-08-25 1998-12-01 G.D. Searle & Co. α-and β-amino acid hydroxyethylamino sulfonamides useful as retroviral protease inhibitors
US5977137A (en) * 1992-09-08 1999-11-02 Vertex Pharmaceuticals, Incorporated Sulfonamide inhibitors of aspartyl protease
US5723490A (en) * 1992-09-08 1998-03-03 Vertex Pharmaceuticals Incorporated THF-containing sulfonamide inhibitors of aspartyl protease
US5783701A (en) * 1992-09-08 1998-07-21 Vertex Pharmaceuticals, Incorporated Sulfonamide inhibitors of aspartyl protease
US5856353A (en) * 1992-09-08 1999-01-05 Vertex Pharmaceuticals, Incorporated Sulfonamide inhibitors of aspartyl protease
US5585397A (en) * 1992-09-08 1996-12-17 Vertex Pharmaceuticals, Incorporated Sulfonamide inhibitors of aspartyl protease
US5750493A (en) * 1995-08-30 1998-05-12 Raymond F. Schinazi Method to improve the biological and antiviral activity of protease inhibitors
US5646180A (en) * 1995-12-05 1997-07-08 Vertex Pharmaceuticals Incorporated Treatment of the CNS effects of HIV
US6180634B1 (en) * 1997-11-13 2001-01-30 Merck & Co., Inc. Combination therapy for the treatment of AIDS
US6436989B1 (en) * 1997-12-24 2002-08-20 Vertex Pharmaceuticals, Incorporated Prodrugs of aspartyl protease inhibitors
US6559137B1 (en) * 1997-12-24 2003-05-06 Vertex Pharmaceuticals Incorporated Sulphonamide derivatives as prodrugs of aspartyl protease inhibitors
US6838474B2 (en) * 1997-12-24 2005-01-04 Vertex Pharmaceuticals, Incorporated Sulphonamide derivatives as prodrugs of aspartyl protease inhibitors
US7592368B2 (en) * 1997-12-24 2009-09-22 Vertex Pharmaceuticals Incorporated Sulphonamide derivatives as prodrugs of aspartyl protease inhibitors
US6319946B1 (en) * 1999-02-12 2001-11-20 Vertex Pharmaceuticals Incorporated Inhibitors of aspartyl protease

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090274650A1 (en) * 1999-06-11 2009-11-05 Hale Michael R Inhibitors of aspartyl protease
US8455497B2 (en) * 1999-06-11 2013-06-04 Vertex Pharmaceuticals Incorporated Inhibitors of aspartyl protease
WO2013009844A2 (en) * 2011-07-11 2013-01-17 Purdue Research Foundation C-3 substituted bicyclooctane based hiv protease inhibitors
WO2013009844A3 (en) * 2011-07-11 2013-04-25 Purdue Research Foundation C-3 substituted bicyclooctane based hiv protease inhibitors
US9309213B2 (en) 2011-07-11 2016-04-12 Purdue Research Foundation C-3 substituted bicyclooctane based HIV protease inhibitors

Also Published As

Publication number Publication date
DK0933372T3 (en) 2008-04-28
JPH11209337A (en) 1999-08-03
DE122008000021I1 (en) 2008-08-14
PL342113A1 (en) 2001-05-21
NO20003304D0 (en) 2000-06-23
CZ20002363A3 (en) 2000-11-15
LU91426I9 (en) 2018-12-28
KR100520737B1 (en) 2005-10-12
NZ505776A (en) 2003-06-30
ID24962A (en) 2000-08-31
NO326265B1 (en) 2008-10-27
CO4990992A1 (en) 2000-12-26
DE69838903D1 (en) 2008-02-07
AP1172A (en) 2003-06-30
ATE382042T1 (en) 2008-01-15
EP0933372B1 (en) 2007-12-26
CZ301653B6 (en) 2010-05-12
LU91426I2 (en) 2008-06-02
EA200000703A1 (en) 2000-12-25
CA2231700C (en) 2005-08-09
YU39800A (en) 2004-03-12
US20030207871A1 (en) 2003-11-06
HUP0101831A3 (en) 2002-08-28
EE200000385A (en) 2001-12-17
FR08C0015I1 (en) 2008-05-30
CN101565412A (en) 2009-10-28
HUP0101831A2 (en) 2002-04-29
KR20010033600A (en) 2001-04-25
US6436989B1 (en) 2002-08-20
FR08C0015I2 (en) 2009-10-30
NL300339I2 (en) 2009-11-02
US7592368B2 (en) 2009-09-22
AU6546698A (en) 1999-07-19
DE69838903T2 (en) 2008-09-18
EP1944300A2 (en) 2008-07-16
JP2009102400A (en) 2009-05-14
HK1021737A1 (en) 2000-06-30
HUS1400042I1 (en) 2017-06-28
EP1944300A3 (en) 2008-11-05
ME00561B (en) 2011-10-10
SK287123B6 (en) 2009-12-07
OA11468A (en) 2004-05-05
IL136941A0 (en) 2001-06-14
WO1999033815A1 (en) 1999-07-08
SK9662000A3 (en) 2001-02-12
CN100503589C (en) 2009-06-24
DE122008000021I2 (en) 2010-02-04
US6838474B2 (en) 2005-01-04
IL136941A (en) 2006-06-11
UA72733C2 (en) 2005-04-15
IS5546A (en) 2000-06-22
EE04466B1 (en) 2005-04-15
ES2299193T3 (en) 2008-05-16
HU229596B1 (en) 2014-02-28
PE20000048A1 (en) 2000-02-08
MY131525A (en) 2007-08-30
NL300339I1 (en) 2008-06-02
TR200002615T2 (en) 2001-01-22
SI0933372T1 (en) 2008-06-30
TW486474B (en) 2002-05-11
ZA9811830B (en) 2000-06-23
JP2005350478A (en) 2005-12-22
CN1284071A (en) 2001-02-14
US6559137B1 (en) 2003-05-06
JP3736964B2 (en) 2006-01-18
NO2009008I2 (en) 2010-09-27
RS52483B (en) 2013-02-28
EA003509B1 (en) 2003-06-26
NO2009008I1 (en) 2009-05-04
JP4282639B2 (en) 2009-06-24
NO20003304L (en) 2000-08-21
BG104631A (en) 2001-02-28
PT933372E (en) 2008-03-31
BG64869B1 (en) 2006-07-31
US20050148548A1 (en) 2005-07-07
AU755087B2 (en) 2002-12-05
IS2817B (en) 2012-11-15
MEP82008A (en) 2011-12-20
PL202845B1 (en) 2009-07-31
AR017965A1 (en) 2001-10-24
AP2000001850A0 (en) 2000-06-30
CA2231700A1 (en) 1999-06-24
EP0933372A1 (en) 1999-08-04
BR9814480A (en) 2001-09-25

Similar Documents

Publication Publication Date Title
US7592368B2 (en) Sulphonamide derivatives as prodrugs of aspartyl protease inhibitors
US20020082249A1 (en) Prodrugs of aspartyle protease inhibitors
US8008297B2 (en) Lysine based compounds
WO1999033792A2 (en) Prodrugs os aspartyl protease inhibitors
AU2004322123B2 (en) Lysine based compounds
MXPA00006315A (en) Sulphonamide derivatives as prodrugs of aspartyl protease inhibitors
MXPA00006316A (en) Prodrugs of aspartyl protease inhibitors
CZ20002364A3 (en) Sulfonamide derivatives and pharmaceutical preparation, in which they are comprised
NZ552853A (en) Lysine based compounds

Legal Events

Date Code Title Description
AS Assignment

Owner name: VERTEX PHARMACEUTICALS INCORPORATED,MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:TUNG, ROGER DENNIS;HALE, MICHAEL ROBIN;BAKER, CHRISTOPHER TODD;AND OTHERS;SIGNING DATES FROM 19980305 TO 19981216;REEL/FRAME:023056/0738

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION