WO2006054182A2 - Phosphoindoles as hiv inhibitors - Google Patents

Phosphoindoles as hiv inhibitors Download PDF

Info

Publication number
WO2006054182A2
WO2006054182A2 PCT/IB2005/004063 IB2005004063W WO2006054182A2 WO 2006054182 A2 WO2006054182 A2 WO 2006054182A2 IB 2005004063 W IB2005004063 W IB 2005004063W WO 2006054182 A2 WO2006054182 A2 WO 2006054182A2
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
aryl
heterocycle
compound
mhz
Prior art date
Application number
PCT/IB2005/004063
Other languages
French (fr)
Other versions
WO2006054182A3 (en
WO2006054182A9 (en
Inventor
Richard Storer
Cyril Dousson
François-René ALEXANDRE
Arlène ROLAND
Original Assignee
Idenix 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=36407947&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2006054182(A2) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to KR1020077008493A priority Critical patent/KR101242962B1/en
Priority to AU2005305609A priority patent/AU2005305609B2/en
Priority to CN2005800392187A priority patent/CN101084231B/en
Priority to BRPI0515344-1A priority patent/BRPI0515344A/en
Priority to RSP-2009/0057A priority patent/RS50981B/en
Priority to NZ554232A priority patent/NZ554232A/en
Priority to JP2007531877A priority patent/JP5031569B2/en
Priority to PL05850774T priority patent/PL1799696T3/en
Priority to DK05850774T priority patent/DK1799696T3/en
Application filed by Idenix Pharmaceuticals, Inc. filed Critical Idenix Pharmaceuticals, Inc.
Priority to CA2583409A priority patent/CA2583409C/en
Priority to MX2007003040A priority patent/MX2007003040A/en
Priority to DE602005011031T priority patent/DE602005011031D1/en
Priority to EP05850774A priority patent/EP1799696B1/en
Publication of WO2006054182A2 publication Critical patent/WO2006054182A2/en
Publication of WO2006054182A9 publication Critical patent/WO2006054182A9/en
Priority to TW095134413A priority patent/TW200804280A/en
Publication of WO2006054182A3 publication Critical patent/WO2006054182A3/en
Priority to IL181909A priority patent/IL181909A0/en
Priority to NO20071871A priority patent/NO20071871L/en
Priority to HK07108120.9A priority patent/HK1100142A1/en
Priority to HR20090086T priority patent/HRP20090086T3/en

Links

Classifications

    • 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 Table
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6561Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing systems of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring or ring system, with or without other non-condensed hetero rings
    • 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 Table
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6558Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing at least two different or differently substituted hetero rings neither condensed among themselves nor condensed with a common carbocyclic ring or ring system
    • C07F9/65586Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing at least two different or differently substituted hetero rings neither condensed among themselves nor condensed with a common carbocyclic ring or ring system at least one of the hetero rings does not contain nitrogen as ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • 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 Table
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/553Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having one nitrogen atom as the only ring hetero atom
    • 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 Table
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/553Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having one nitrogen atom as the only ring hetero atom
    • C07F9/572Five-membered rings
    • C07F9/5728Five-membered rings condensed with carbocyclic rings or carbocyclic ring systems
    • 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 Table
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6558Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing at least two different or differently substituted hetero rings neither condensed among themselves nor condensed with a common carbocyclic ring or ring system
    • C07F9/65583Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing at least two different or differently substituted hetero rings neither condensed among themselves nor condensed with a common carbocyclic ring or ring system each of the hetero rings containing nitrogen as ring hetero atom

Definitions

  • the present invention provides new human immunodeficiency virus (HIV) reverse transcriptase inhibitory compounds and their pharmaceutically acceptable salts, prodrugs, analogs and derivatives. Also included are methods of using these compounds for the prophylaxis and treatment of HIV infection and AIDS, and pharmaceutical compositions that contain the compounds.
  • HIV human immunodeficiency virus
  • HIV-I human immunodeficiency virus reverse transcriptase
  • Inhibitors include non-nucleoside reverse transcriptase inhibitors or NNRTIs that bind to a specific allosteric site of the HIV reverse transcriptase near the polymerase site and interfere with reverse transcription by either altering the conformation or the mobility of the reverse transcriptase, thus leading to noncompetitive inhibition of the enzyme (Kohlstaedt et al., Science (1992), 256:1783- 90).
  • NNRTIs non-nucleoside reverse transcriptase inhibitors
  • examples of these include:
  • DABO dihydroalkoxybenzyloxopyrimidine
  • PETT phenylethylthiazolylthiourea
  • alpha-anilinophenylacetamide (alpha-APA) derivatives (Pauwels et al., PNAS
  • Indole derivatives described in U.S. Patent No. 5,527,819 assigned to Merck & Co. have been shown to be inhibitors of HIV reverse transcriptase. Some of these compounds exhibited ICso values against HIV reverse transcriptase at concentrations of from 3-35 ⁇ M.
  • a process for synthesizing optionally substituted indoles by a palladium- catalyzed angulation between a ketone and an iodoaniline was also developed at Merck & Co. (Chen et al., J. Org. Chem. (1997), 62(9):2676-77).
  • the compounds disclosed in the '819 patent are generally represented by the following broad structural Formula (III):
  • U.S. Patent No. 5,124,327 to Merck & Co. discloses a class of optionally substituted sulfonylphenyl indole compounds. The patent reports that the compounds are active as reverse transcriptase inhibitors and could be useful in the treatment of HIV infection and AIDS.
  • PCT Publication No. WO 2004/014364 to Idenix Pharmaceuticals discloses another class of phenylindoles that displays enhanced anti-BTV activity. These compounds are also substituted with at least two moieties other than hydrogen on either or both rings, hi addition, these compounds incorporate a number of different substituents with a carboxamide functionality at position-2 on the indole, the position shown in formula (II) above as "Z". Typical placement of substituents is at the 3" and 5" positions on the phenyl ring, and at the 4' and 5', 5' and 6', or 5' and T positions on the benzo ring of the indole moiety.
  • WO 01/02388 to SmithKline Beecham S.P.A discloses optionally substituted phenylindoles with a carbamyl substituent that are alleged to have utility in the treatment of HIV 5 AIDS, osteoporosis, cancers, and Alzheimer's disease.
  • Warner-Lambert Company discloses various indole-thiazepinones, oxazepinones, diazepinones, benzothiophenes, benzofurans, and indole-2-carboxamides for the the treatment of HIV in U.S. 5,424,329; U.S. 5,565,446; U.S. 5,703,069; and WO 96/29077.
  • U.S. Patent No. 5,945,440 to Kleinschroth et al. discloses a class of indolocarbazole amides for the treatment of a variety of diseases including cancer, viral diseases (including HIV), cardiac and vascular diseases, bronchopulmonary diseases, inflammatory disorders, degenerative diseases of the central nervous system, and other diseases.
  • U.S. Patent No. 4,866,084 to Gunasekera et al. teaches certain bisindole alkaloid compounds that have antiviral and antitumor activity, including HSV (herpes simplex virus).
  • HSV herpes simplex virus
  • U.S. Patent 5,935,982 to Dykstra et al. reports a different class of bisindoles that have utility versus retroviral infections and especially HIV.
  • U.S. Patent No. 5,852,011 to Matsunaga et al. discloses a class of indole derivatives substituted by a heteroaryl function and an amide function. The compounds are said to possess antitumor, antiviral, and antimicrobial properties.
  • U.S. Patent No. 5,935,982 to Dykstra et al. discloses a class of bis-indoles and specifically propose their use for treating retroviral infections, and especially infection by
  • U.S. Patent No. 5,929,114 to Domagala et al. discloses a class of arylthio and bithiobisarylamide compounds, including indole derivative, that reportedly have antibacterial and antiviral activity.
  • U.S. Patent No. 5,830,894 to Pevear et al. discloses a class of triazinoindole derivatives that reportedly have anti-pestivirus activity, most notably BVDV activity.
  • 3-phosphoindole compounds dispay antiviral activity against HIV, in particular against strains of HIV that have developed cross resistance to other anti-HIV drugs.
  • Compounds, compositions and methods for treatment of HIV infection are disclosed that include the 3-phosphoindole compounds.
  • the 3-phosphoindoles can be in the form of a wide variety of moieties, including, but not limited to, phosphates, phosphonates, a phosphorthioate, including .thiophosphates, thiophosphonates, phosphate, and a phosphoramidate, including.iminiophosphates and iminophosphonates.
  • the compounds that can have anti-HIV activity are of Formula (A) below. These compounds include an phosphorus-linked substituent at position-3 and particular substituents at position-2 on the indole, and a monosubstitution at position R 5 or a disubstitution at positions R 4 and R 5 , R 5 and R 6 , or R 5 and R 7 on the benzo ring.
  • substituent "X" represents a phenyl ring that is unsubstituted or is substituted by one or more halogens or lower alkyl groups such as methyl or ethyl.
  • Particular substituents at position 2 on the indole moiety include, for example, hydrogen, hydroxy, halogen, alkyl, aryl, heteroaryl, and especially substituents having a carboxamide or carboxamide moiety shown as "Z" in Formula (A).
  • Substituents for the benzo ring of the indole moiety include but are not limited to chlorine, fluorine, bromine, iodine, CF 3 , CN, NO 2 , and methoxy.
  • the active compound may be a salt or prodrug that, upon administration, provides directly or indirectly the parent compound or that itself exhibits a desired activity.
  • the compound of Formula A includes a charged heteroatom, and in a particular embodiment, the compound includes an N-oxide group.
  • Figure 1 is an illustration of three (3) general structures of the present invention given as Formula (A), Formula (B), and Formula (C).
  • Figure 2 is an illustration of phosphorylated compounds (I)-(X) of the general Formulae (A) and (B).
  • Figure 3 is an illustration of 9- and 10-membered bicyclic phosphorylated compounds (XI)-(XX) of the general Formula (C).
  • compositions of matter, method of use and a pharmaceutical composition for the treatment of retroviral infections in mammals and, in particular, HIV in humans Included within the present invention are the following features:
  • compositions comprising an effective anti-HIV treatment amount of a 3-phosphoindole or its pharmaceutically acceptable salt or prodrug thereof, together with a pharmaceutically acceptable carrier or diluent;
  • compositions comprising an effective anti-HIV treatment amount of a 3-phosphoindole or its pharmaceutically acceptable salt or prodrug thereof, in combination with one or more other anti-HIV agents, optionally with a pharmaceutically acceptable carrier or diluent; • pharmaceutical compositions for the treatment or prophylaxis of an HIV infection that is resistant to one or more reverse transcriptase inhibitors, in a host, comprising an effective anti-HIV treatment amount of a 3-phosphoindole or its pharmaceutically acceptable salt or prodrug, optionally in combination with at least one other anti-HIV agent, optionally with a pharmaceutically acceptable carrier or diluent;
  • compositions for the treatment or prophylaxis of an HIV infection in a host as a form of salvage therapy comprising an effective anti-HIV treatment amount of a 3-phosphoindole or its pharmaceutically acceptable salt or prodrug, optionally in combination with at least one other anti-HIV agent, and optionally with a pharmaceutically acceptable carrier or diluent;
  • compositions for the treatment or prophylaxis of an HIV infection that is resistant to one or more reverse transcriptase inhibitors due to a reverse transcriptase mutation such as lysine 103 -> asparagine and/or tyrosine 181 - ⁇ cysteine, in a host comprising an effective anti-HIV treatment amount of a 3-phosphoindole or its pharmaceutically acceptable salt or prodrug, optionally in combination with at least one other anti-HIV agent, and optionally with a pharmaceutically acceptable carrier or diluent;
  • a 3-phosphoindole or a pharmaceutically acceptable salt, prodrug, N— oxide, quaternary amine, stereochemical isomer or tautomer thereof is provided.
  • the 3-phosphoindole can be in the form of a phosphate, phosphonate, thiophosphate, thiophosphonate, iminiophosphate or iminophosphonate.
  • the compound is represented generally by the following chemical Formula (A):
  • halogen F, Cl, Br, or I
  • F typically F
  • X and Y may come together to form an optionally substituted bicyclic or tricyclic phosphorylated heterocycle wherein each ring comprises 3-7 members;
  • the heteroaryl can optionally include a charged heteroatom, and in particular can include an N-oxide
  • each W is independently:
  • R 1 is:
  • each R 2 is independently:
  • alkyl optionally substituted, branched or unbranched, alkyl, such as a C 1-6 alkyl;
  • alkenyl such as a C 2-6 alkenyl
  • alkynyl such as a C 2-6 alkynyl
  • each R 3 is independently: a) H; b) OH; c) halogen (F, Cl, Br, or I);
  • alkyl optionally substituted, branched or uribranched, alkyl, such as a C 1-6 alkyl;
  • alkenyl such as a C 2-6 alkenyl
  • alkynyl optionally substituted, branched or unbranched, alkynyl, such as a C 2-6 alkynyl;
  • heterocycle substitution is selected from the group consisting of:
  • n independently is 0, 1 or 2;
  • each A is independently a disubstituted spacer selected from the group consisting of:
  • arylene optionally substituted arylene
  • O-alkylene optionally substituted arylene
  • aralkylene branched or unbranched, and optionally having one or more heteroatoms, aryl, cycloalkyl or heterocyclyl functions in or attached to the chain;
  • A may be joined by any desired linkage such as, for example, an ether, thioether, amino, carboxamido, ester or carbon-carbon linkage, or any combinatin thereof;
  • each R 4' , R 5' , R 6> and R 7' independently is:
  • X is an optionally substituted phenyl
  • Y is any of its definitions
  • R 1 , R 4 , R 6 , and R 7 are all hydrogen
  • R 5 is halogen
  • Z is a carboxamide moiety.
  • R 1 is acyl, alkyl, aryl, alkaryl, or aralkyl.
  • R 4 is fluoro, nitro or cyano
  • W is oxygen
  • Y is O-alkyl
  • X is an optionally substituted phenyl
  • Y is any of its definitions
  • R 1 , R 4 , R 6 , and R 7 are all hydrogen
  • R 5 is chlorine
  • Z is a carboxamide or carboxamido-heterocyclyl moiety.
  • X is tolyl, thiazolyl or pyridyl; Y is H, OH, or O-alkyl; R 1 , R 4' , R 6' , and R 7' are all hydrogen; R 5> is halogen; and Z is carboxamido, acyl, an alkyl-sulphonyl group or a carboxylic acid derivative.
  • X, Y, R 1 , R 4 , R 6 , R 7 and R 5 all are as defined above, and Z is a carboxamido-alkylene-heterocycle, typically carboxamido-alkylene-pyridyl; a thioamido-pyridyl wherein the pyridyl is unsubstituted or substituted by OH, OMe or lower alkyl; an imino-nitrile; or an alkylsulphonyl-aryl group.
  • Z is a carboxamido-alkylene-heterocycle wherein the heterocycle includes at least one N-oxide group.
  • the invention provides a phenylindole for use in the treatment of HIV represented by the following general Formula (B):
  • A, n, R »4 4 ' , r R»5 3 ' , ⁇ R- ⁇ 6 D ' and R >V' is as defined above for Formula (A);
  • each R 2" , R 3" , R 4 ", R 5" , and R 6" independently is:
  • R or R may be joined to Y to form an optionally substituted bicyclic or tricyclic phosphorylated heterocycle wherein each ring comprises 3-14 members.
  • Y is alkyl
  • W is O, S(O) n , or NH
  • R 1 , R 4' , R 6' , R 7' , R 2" , R 3" , R 4 ", R 5" , and R 6" are all hydrogen
  • R 5 is halogen
  • Z is carboxamide
  • Y is aryl; W is O, S(O) n , or NH; R 1 , R 4' , R 6> , R 7' , R 2" , R 3" , R 4 ", R 5" , and R 6" are all hydrogen; R 5 is amino-alkyl, thioamino-alkyl, or aminocarbonyl-alkyl; and Z is carboxamide; c) Y is -OH or -SR 2 ; W is O, S(O) n , or N-NH 2 ; R 1 , R 4' , R 6' , R 7' , R 2" , R 4 ", and R 6" are all hydrogen; R 5' is halogen; R 3" and R 5 ' are methyl; and Z is carboxamide;
  • Y is -OH;
  • W is O, S(O) n , or NH;
  • R 1 , R 6> , R 7' , R 2" , R 4 ", and R 6" are all hydrogen;
  • R 4' and R 5' are halogen;
  • R 3" and R 5 are methyl; and
  • Z is carboxamide;
  • Y is -OH;
  • W is O, S(O) n , or N-NR 2 R 2 ;
  • R 1 , R 4' , R 6' , R 7' , R 2" , R 3" , R 4 ", R 5" , and R 6' are all hydrogen;
  • R 5 is halogen; and
  • Z is carboxamide;
  • Y is alkenyl
  • W is O, S(O) n , or NH
  • R 5 is halogen
  • Z is carboxamide or carboxamide- heterocycle
  • Y is alkynyl or -NR 2 R 3 ;
  • W is O 5 S(O) n , or N-O-alkyl;
  • R 1 , R 4' , R 6' , R 7> , R 2" , R 3 , R 4 ", R 5 , and R 6 are all hydrogen;
  • R 5' is halogen; and
  • Z is carboxamide;
  • Y is alkenylene
  • W is O 5 S(O) n , or NH
  • R 1 , R 4' , R 6' , R 7' , R 2" , R 3" , R 4 ", R 5" , and R 6 are all hydrogen
  • R 5 is halogen
  • Z is carboxamide
  • Y is ethyl
  • W is O 5 S(O) n , or N-OH
  • R 1 , R 4' , R 6> , R 7' , R 2" , R 3" , R 4 ", R 5" , and R 6 are all hydrogen
  • R 5 is chloro
  • Z is carboxamide
  • Y is -O-methyl
  • W is O 5 S(O) n , or NH
  • R 1 , R 4> , R 6' 5 R 7' , R 2" , R 3" , R 4 ", R 5" , and R 6 are all hydrogen
  • R 5 is halogen
  • Z is carboxamide
  • Y is -O-ethyl
  • W is O 5 S(O) n , or N-NH 2
  • R 1 , R 4' , R 6' , R 7' , R 2" , R 3" , R 4 ", R 5" , and R 6 are all hydrogen
  • R 5 is halogen
  • Z is carboxamide -heterocycle
  • W is O, S(O) n , or NH;
  • R 1 , R 4' , R 6' , R 7' , R 2" , R 3" , R 4 ", R 5" , and R 6 are all hydrogen;
  • R 5 is chloro; and
  • Z is carboxamide;
  • Y is -0-H;
  • W is O, S(O) n , or NH;
  • R 1 , R 4' , R 6' , R 7> , R 2" , R 3" , R 4 " 5 R 5" , and R 6" are all hydrogen;
  • R 5 is halogen;
  • n) Y is -0-H;
  • W is O, S(O) n , or N-NH 2 ;
  • R 1 , R 4' , R 6> , R 7> , R 2" , R 3" , R 4 ", R 5" , and R 6 are all hydrogen;
  • R 5' is halogen;
  • Y is -O-H
  • W is O, S(O) n , or NH
  • R 1 , R 4' , R 6> , R 7' , R 2" , R 3" , R 4 ", R 5" , and R 6" are all hydrogen
  • R 5' is halogen
  • Z is carboxamide-cyclopropyl or carboxamide-cyclobutyl
  • Y is -O-methyl
  • W is O, S(O) n , or NH
  • R 1 , R 4' , R 6' , R 7' , R 2" , R 3" , R 4 ", R 5" , and R 6" are all hydrogen
  • R 5' is halogen
  • Z is a carboxamide-ethyl, carboxamide -ethanol, or carboxamide -ethyl-methoxy
  • Y is -OH or -NR 2 R 3 ;
  • W is O, S(O) n , or N-O-alkyl;
  • R 1 , R 4' , R 6' , R 7' , R 2" , R 3" , R 4 ", R 5 , and R 6 are all hydrogen;
  • R 5 is halogen;
  • Y is -OH or -SR 2 ;
  • W is O, S(O) n , or N-NR 2 R 2 ;
  • R 5 , and R 6 are all hydrogen; R 5 is halogen; and Z is an isopropanol carboxamide moiety; and
  • s) Y is -OH; W is O, S(O) n , or N-OH; R 1 , R 4' , R 6' , R 7' , R 2" , R 3" , R 4 ", R 5" , and R 6" are all hydrogen; R 5 is halogen; and Z is a thioacetamide group.
  • the present invention provides an optionally substituted 9-
  • each W 5 Z, R 1 , R 2 , R 3 , A 5 n, R 4' , R 5' , R 6' and R 7' is as defined above for Formula (A);
  • each R 3" , R 4 ", R 5" , and R 6" is as defined above for Formula (B);
  • each Y and T independently is:
  • each Y and T independently is:
  • W is O, Y is CR 2 , T is (CH 2 ) 2 , R 1 , R 4' , R 6' , R 7' , R 3" , R 4" , R 5" , and R 6" are all H; R 5 is halogen, and Z is carboxamide;
  • W is S, Y is O, T is CH 2 , R 1 , R 4' , R 6' , R 7' , R 3" , R 4" , R 5" , and R 6" are all H; R 5' is halogen, and Z is carboxamido-heterocycle wherein said heterocycle is optionally substituted furan, imidazole, thiazole, or pyridyl;
  • W is S 5 Y is NR 2 , T is CH 2 , R 1 , R 4' , R 6' , R 7' , R 3" , R 4" , R 5" , and R 6" are all H; R 5' is halogen, and Z is carboxamide;
  • W is O
  • Y is SR 2
  • T is CH 2
  • R 1 , R 4' , R 6' , R 7' , R 3" , R 4" , R 5" , and R 6" are all H
  • R 5' is halogen
  • Z is alkyl-carboxamide
  • W is S
  • Y is SR 2
  • T is CH 2
  • R 1 , R 4' , R 6' , R 7' , R 3" , R 4" , R 5" , and R 6" are all H
  • R 5' is halogen
  • Z is carboxamide
  • W is O
  • Y is CR 2
  • R 1 , R 4' , R 6' , R 7' , R 3" , R 4" , R 5" , and R 6" are all H
  • R 5 is halogen
  • Z is methoxymethyl-carboxamide
  • W is O
  • Y is SR 2
  • W is S
  • Y is NR 2
  • R 5 is chloro, and Z is carboxamide;
  • W is S
  • Y is N
  • R 1 , R 4' , R 6' , R 7' , R 3" , R 4" , R 5" , and R 6" are all
  • R 5 is fluoro, and Z is carboxamide
  • W is NH
  • Y is CR 2
  • T is NR 2
  • R 1 , R 4' , R 6' , R 7' , R 3" , R 4" , R 5" , and R 6" are all H
  • R 5 is amino-alkyl
  • Z is carboxamide
  • W is NR 2 , Y is O, T is (CH 2 ) 2 , R 1 , R 4' , R 6> , R 7' , R 3" , R 4" , R 5" , and R 6" are all H; R 5 is chloro, and Z is carboxamide;
  • W is N-OH, Y is O, T is CH 2 , R 1 , R 4> , R 6' , R 7> , R 3" , R 4" , R 5" , and R 6" are all H; R 5 is thioamino-alkyl, and Z is alkyl carboxamide ; n) W is S, Y is SR 2 , T is CH 2 , R 1 , R 4> , R 6' , R 7> , R 3" , R 4" , R 5" , and R 6" are all H; R 5' is halogen, and Z is a carboxamide-heterocyclyl group wherein the heterocycle is an optionally substituted pyridine, thiazole, imidazole or furan;
  • W is N-O-alkyl
  • Y is NR 2
  • T is CH 2
  • R 1 , R 4' , R 6' , R 7' , R 3" , R 4" , R 5" , and R 6" are all H
  • R 5' is halogen
  • W is NH
  • Y is SR 2
  • T is (CHb) 2
  • R 5 is halogen
  • Z is carboxamide-alkyl-phenyl wherein phenyl is optionally substituted by one or more halo, oxo, OH, NO 2 , MeOH, -NH-
  • W is NR 2
  • Y is (CBb) 2
  • T is NR 2
  • R 1 , R 4' , R 6' , R 7' , R 3" , R 4" , R 5" , and R 6" are all H
  • R 5 is halogen
  • Z is a carboxamide-cyclopropyl group
  • W is N-OH, Y is CH, T is O, R 1 , R 4' , R 6' , R 7' , R 3" , R 4" , R 5" , and R 6" are all H; R 5 is halogen, and Z is a carboxamide-cyclopropyl group;
  • W is N-O-alkyl
  • Y is O
  • T is (CBb) 2
  • R 1 , R 4' , R 6> , R 7> , R 3" , R 4" , R 5" , and R 6" are all H
  • R 5 is chloro
  • Z is carboxamide-methoxyethyl
  • W is O, Y is N, T is CH 2 , R 1 , R 4' , R 6' , R 7' , R 3" , R 4" , R 5" , and R 6" are all H; R 5' is halogen, and Z is carboxamide-ethanol;
  • W is N-NR 2 R 2 , Y is NR 2 , T is CH 2 , R 1 , R 4' , R 6' , R 7' , R 3" , R 4" , R 5" , and R 6" are all H; R 5 is halogen, and Z is carboxamide; and
  • W is O
  • Y is CR 2
  • T is CH 2
  • R 1 , R 4' , R 6' , R 7' , R 3" , R 4" , R 5" , and R 6" are all H
  • R 5 is aminocarbonyl-alkyl
  • Z is carboxamide.
  • the compound includes a charged heteroatom.
  • a charged nitrogen such as through an N-oxide, is part of the compound.
  • the charged heteroatom can be on a heteroaromatic ring that is attached to the indole, for example through C(O)NH-alkyl, or specifically through C(O)NH-CH 2 .
  • Z is C(O)-NH-R 2 wherein R 2 is an optionally substituted alkylheterocycle, wherein the heterocycle is of the formula
  • Rx-Rx Rx' RX Rx Rx , wherein each Rx is independently CH or TSf + -O " . In a particular embodiment, one of Rx is N + -O " .
  • the alkyl group can be C 1-10 alkyl, or C 1-4 alkyl, or can specifically be a methylene or ethylene.
  • R 2 is of the formula
  • n 0, 1 or 2.
  • the compound is:
  • Either substituent on the phenyl ring may be substituted by CN, Me, halo, alkyl, alkenyl, alkynyl, alkyl-CN or alkenyl-CN, as the most commonly synthesized and tested N-oxides in the series to date.
  • X and Y each independently, is: a) 3-14 membered carbocycle, aryl, heterocycle, any of which may comprise a monocyclic, bicyclic, tricyclic or spiro structure, or optionally may be substituted;
  • X and Y may be joined to form an optionally substituted bicyclic or tricyclic phosphorylated heterocycle wherein each ring comprises 3-7 members;
  • W is:
  • R 1 is:
  • R 2 is:
  • said optional substitution comprises one or more of: a) a substituted or unsubstituted heterocycle;
  • R 3 is:
  • substitution is selected from the group consisting of:
  • n independently is 0, 1 or 2;
  • each A is independently a disubstituted spacer selected from the group consisting of:
  • arylene optionally substituted arylene
  • O-alkylene branched or unbranched, and optionally having one or more heteroatoms, aryl, cycloalkyl or heterocyclyl functions in or attached to the chain;
  • aralkylene branched or unbranched, and optionally having one or more heteroatoms, aryl, cycloalkyl or heterocyclyl functions in or attached to the chain;
  • A may be joined by any desired linkage such as, for example, an ether, thioether, amino, carboxamido, ester or carbon-carbon linkage, or any combinatin thereof;
  • R 4' , R 5' , R 6' and R 7' independently is:
  • X is an optionally substituted phenyl
  • Y is any of its definitions
  • R 1 , R 4 , R 6 , and R 7 are all hydrogen
  • R 5 is halogen
  • Z is a carboxamide moiety.
  • R 1 is acyl, alkyl, aryl, alkaryl, or aralkyl.
  • R 4 is fluoro, nitro or cyano
  • W is oxygen
  • Y is O-alkyl
  • X is an optionally substituted phenyl
  • Y is any of its definitions
  • R 1 , R 4 , R 6 , and R 7 are all hydrogen
  • R 5 is chlorine
  • Z is a carboxamide or carboxamido-heterocyclyl moiety.
  • X is tolyl, thiazolyl or pyridyl; Y is H, OH, or O-alkyl; R 1 , R 4' , R 6' , and R 7' are all hydrogen; R 5' is halogen; and Z is carboxamido, acyl, an alkyl-sulphonyl group or a carboxylic acid derivative.
  • Z is a carboxamido-alkylene-heterocyclyl wherein the heterocycle typically is imidazole, furan, pyridine, pyrimidine, or thiazole that optionally may be substituted; a thioamido-pyridyl wherein the pyridyl is unsubstituted or substituted by OH, OMe or lower alkyl; an imino-nitrile, or an alkylsulphonyl-aryl group.
  • a first series of subembodiments of the present invention is given where the Formula (A) is as provided above, and W, X and Y are defined as: a) W is O, X is alkyl, and Y is -O-alkyl;
  • W is O, X is -O-aryl, and Y is alkyl;
  • W is O, X is -O-aryl, and Y is -NR 2 R 3 ;
  • W is O, X is -O-alkyl, and Y is alkyl;
  • W is O, X is -O-alkyl, and Y is halo;
  • W is O, X is -O-heterocycle, and Y is alkyl;
  • W is O, X is aryl, and Y is -O-alkyl;
  • W is O, X is heterocyclyl, and Y is -O-aryl;
  • W is O, X is alkyl, and Y is -O-heterocyclyl;
  • W is -NR 2 R 2 , X is heterocyclyl, and Y is -O-aryl;
  • W is -NR 2 R 2 , X is alkyl, and Y is halo;
  • W is S, X is alkyl, and Y is -O-alkyl;
  • W is S, X is alkyl, and Y is -NR 2 R 3 ;
  • n) W is S, X is -O-aryl, and Y is alkyl;
  • W is S, X is -O-aryl, and Y is C-halo;
  • W is S, X is -O-alkyl, and Y is alkyl;
  • W is S, X is -O-heterocycle, and Y is alkyl;
  • W is S, X is aryl, and Y is -O-alkyl;
  • W is S, X is heterocyclyl, and Y is -O-aryl;
  • W is S, X is alkyl, and Y is -O-heterocyclyl;
  • W is O 5
  • X is aryl, and Y is -O-aryl;
  • W is -NR 2
  • X is -O-alkyl
  • Y is -NR 2 R 3 ;
  • W is O, X is -O-aryl, and Y is -O-aryl;
  • x) W is O 3 X is alkyl, and Y is alkyl; and y) W is -NR 2 , X is -O-alkyl, and Y is alkyl.
  • R 1 is H, alkyl, acyl, aryl, aralkyl, or alkaryl; and Z is defined as:
  • R 2 is H, and R 3 is NR 2 R 2 , or R 2 is C 1-5 alkyl optionally substituted with OH and R 3 is -NH 2 ;
  • R 2 is H, and R 3 is alkyl substituted by optionally substituted aryl or heterocycle;
  • R 3 is an amino acid residue or -NH(CH 2 ) p -(amino acid residue);
  • R 2 is H or alkyl, and R 3 is aryl;
  • R 2 is as defined above, and R 3 is -OH;
  • R 2 is optionally substituted, branched chain alkyl
  • R 2 is NH or alkyl, and R 3 is NH 2 ;
  • R 2 and R 3 are as defined as above;
  • R 2 is as defined above;
  • R 2 is optionally substituted aryl, cycloalkyl or a heterocyclyl ring;
  • R 2 is as defined bove;
  • R 3 is as defined above;
  • R 2 is alkylene and R 3 is aryl or heteroaryl;
  • R 2 is alkylene and R 3 is optionally substituted alkyl, aryl or heteroaryl;
  • R 2 is -NH, p is 0-10, A is a divalent linker or an optionally substituted aryl or heteroaryl, and W is O or S;
  • R 2 is an optionally substituted alkylene, alkenylene, or alkynylene, and R 3 is any of its definitions as provided above;
  • R 2 is an ptionally substituted alkylene, alkenylene, or alkynylene, and R 3 is any of its definitions as provided above;
  • R 2 , R 3 and A each is as defined previously.
  • R4', R5', R6' and R7' are defined as:
  • R 4> and R 7' are both hydrogen, and R 5' and R 6' independently are halo; -C 1-6 alkyl, alkenyl or alkynyl optionally substituted with one or more halo;
  • R 4' and R 6' are both hydrogen, and R 5' and R 7' independently are halo, -C 1-6 alkyl, alkenyl or alkynyl optionally substituted with one or more halo;
  • R 6' and R 7> are both hydrogen, and R 4' and R 5' independently are Cl, F, Br, I, methyl, ethyl or CF 3 ;
  • R 4' and R 7 are both hydrogen, and R 5 and R 6 independently are Cl, F, Br, I, methyl, ethyl or CF 3 ;
  • R 4' and R 6' are both hydrogen, and R 5' and R 7' independently are Cl, F, Br, I, methyl, ethyl or CF 3 ;
  • a fourth series of subembodiments for Formula (A) is defined where W, X and Y are as defined in the first series of subembodiments, and Z is as defined in the second series of subembodiments.
  • a fifth series of subembodimentts for Formula (A) is defined where W, X and Y are as defined in the first series of subembodiments, and R 4 , R 5 , R 6' and R 7' are as defined in the third series of subembodiments
  • a sixth series of subembodimentts for Formula (A) is defined where Z is as defined in the second series of subembodiments, and R , R 5 , R 6 and R 7 are as defined in the third series of subembodiments.
  • Non-limiting species of the first embodiment as given by Formula (A) above are defined when:
  • R 4' is F or Cl
  • R 6' and R 7' are H
  • R 5' is F or Cl
  • X is H
  • Y is -O-alkynyl
  • R 6 and R 7 independently are F or Cl, R 6 and R 7 are H, X is carbocycle, and Y is H;
  • R 6 and R 7> are H
  • X is -C 2-6 alkenyl
  • Y is -OH
  • Y is -heterocycle
  • R 6' and R 7' are H, X is H, and Y is -O-alkyl;
  • R 4 and R 5 independently are H, F or Cl, R 6' and R 7' are H, X is optionally substituted phenyl, and Y is -O-alkyl;
  • W is as defined for Formula (A).
  • the invention provides a phenylindole for use in the treatment of HIV represented by the following general Formula (B):
  • each W, Y, Z, R 1 , R 2 , R 3 , A, n, R 4' , R 5> , R 6' and R 7' is as defined above for Formula (A);
  • each R 2" , R 3" , R 4 ", R 5" , and R 6" independently is:
  • R 2 or R 6 may be joined to Y to form an optionally substituted bicyclic or tricyclic phosphorylated heterocycle comprising 4 -14 members.
  • a first series of subembodiments of the Formula (B) include all the subembodiments provided for Formula (A) above.
  • R 2 , R 3 , R 4 , R 5 , and R 6 are defined as:
  • R 2 , R 4 , and R 6 are hydrogen, and R 3 and R 5 each independently is halogen; - NO 2 ; -CN; -OR 2 ; -NH-R 5 -C 1-6 alkyl; -NHCO-C 1-6 alkyl; oxime; hydrazine; -
  • R 2 , R 4 , and R 6 are hydrogen, and R 3 and R 5 each independently is halogen, or -C 1-6 alkyl, alkenyl, alkynyl optionally substituted with one or more halogen;
  • R 2" , R 3" , R 4" , R 5" and R 6" are hydrogen;
  • R 2 , R 4 , and R 6 are hydrogen, and R 3 and R 5 are methyl;
  • R 2 , R 4 , and R 6 are hydrogen, and R 3 and R 5 are chloro;
  • R 2 , R 4 , and R 6 are hydrogen, and R 3 and R 5 are fluoro;
  • R 2" , R 4" , and R 6" are hydrogen, R 3" is iodo and R 5" is bromo;
  • R 2 , R 4 , and R 6 are hydrogen, R 3 is methyl, and R 5 is chloro; and i) R 2" , R 4" , and R 6" are hydrogen, R 3" is chloro, and R 5" is methyl.
  • W is S
  • Y is OH
  • R 2" , R 4" , and R 6" are hydrogen
  • R 3" and R 5" each independently is halogen, or -C 1-6 alkyl, alkenyl, alkynyl optionally substituted with one or more halogen;
  • W is O, Y is C 1-6 alkyl, R 2" , R 3" , R 4" , R 5" and R 6" are hydrogen;
  • W is S, Y is C 1-6 alkylene, R 2" , R 4" , and R 6" are hydrogen, and R 3" and R 5" are methyl;
  • W is NH
  • Y is -O-alkyl
  • R 2" , R 4" , and R 6" are hydrogen
  • R 5 are chloro
  • W is S
  • Y is -O-alkenyl
  • R 2" , R 4" , and R 6" are hydrogen
  • R 5" are fluoro
  • W is O
  • Y is aryl
  • R 2" , R 4" , and R 6" are hydrogen
  • R 3" and R 5" are fluoro
  • W is NH
  • Y is -O-alkynyl
  • R 2" , R 4" , and R 6" are hydrogen
  • R 3" and R 5" are fluoro
  • W is S
  • Y is S
  • R 2" , R 4" , and R 6" are hydrogen
  • R 3" and R 5" are fluoro

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Oncology (AREA)
  • Virology (AREA)
  • Communicable Diseases (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • AIDS & HIV (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Indole Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

3-phosphoindole compounds for the treatment of retroviral infections, and particularly for HIV, are described. Also included are compositions comprising the 3-­phosphoindole derivatives alone or in combination with one or more other anti-retroviral agents, processes for their preparation, and methods of manufacturing a medicament incorporating these compounds.

Description

PHOSPHO-INDOLES AS HIV INHIBITORS
CROSS REFERENCE TO RELATED APPLICATIONS
This application claims priority to U.S. Provisional Application Nos. 60/611,061, filed September 16, 2005, 60/711,445 filed August 25, 2005 and 60/711,565, filed August 26, 2005, all entitled "Phospho-Indoles as HIV Inhibitors."
FIELD OF THE INVENTION
The present invention provides new human immunodeficiency virus (HIV) reverse transcriptase inhibitory compounds and their pharmaceutically acceptable salts, prodrugs, analogs and derivatives. Also included are methods of using these compounds for the prophylaxis and treatment of HIV infection and AIDS, and pharmaceutical compositions that contain the compounds.
BACKGROUND OF THE INVENTION
Numerous compounds have been synthesized to combat the human immunodeficiency virus (HIV) since it was discovered to be the etiological cause of the acquired immunodeficiency syndrome (AIDS) in 1983. A focal point of AIDS research efforts has been and continues to be the development of inhibitors of human immunodeficiency virus (HIV-I) reverse transcriptase, the enzyme responsible for the reverse transcription of the retroviral RNA to proviral DNA (W.C. Greene, New England Journal of Medicine (1991), 324:308-17; Mitsuya et al, Science (1990), 249:1533-44; EJ. DeClercq, Retrovirus (1992), 8:119-34). Inhibitors include non-nucleoside reverse transcriptase inhibitors or NNRTIs that bind to a specific allosteric site of the HIV reverse transcriptase near the polymerase site and interfere with reverse transcription by either altering the conformation or the mobility of the reverse transcriptase, thus leading to noncompetitive inhibition of the enzyme (Kohlstaedt et al., Science (1992), 256:1783- 90). Several classes of compounds have been identified as NNRTIs of HIV. Examples of these include:
1) ^[(l-hydroxyethoxy^ethyη-ό-Cphenylthio^hymines (HEPT) (Tanaka et al., J. Med. Chem. (1991), 34:349-57; Pontikis et al., J. Med. Chem. (1997), 40:1845- 54; Danel et al., J. Med. Chem. (1996), 39:2427-31; Baba et al., Antiviral Res.
(1992), 17:245-64);
2) bis(heteroaryl)piperazines (BHAP) (Romero et al., J. Med. Chem. (1993), 36:1505-8);
3) dihydroalkoxybenzyloxopyrimidine (DABO) (Danel et al., Acta Chemica Scandinavica (1997), 51:426-30; Mai et al., J. Med. Chem. (1997), 40:1447-54);
4) 2', 5'-bis-O-(tertbutyldimethylsilyl)-3'-spiro-5"-(4"-amino-l", 2"-oxathiole-2", 2"-dioxide)pyrimidines (TSAO) (Balzarini et al., PNAS USA (1992), 89:4392- 96);
5) phenylethylthiazolylthiourea (PETT) derivatives (Bell et al., J. Med. Chem. (1995), 38:4929-36; Cantrell et al., J. Med. Chem. (1996), 39:4261-74);
6) tetrahydro-imidazo[4,5,l-jk][l,4]-benzodiazepine-2(lH)-one and -thione (TIBO) derivatives (Pauwels et al., Nature (1990), 343:470-4);
7) phosphorus-substituted imidazole derivatives (PCT Publication No. WO 03/091264 A2 to Gilead Sciences, Inc.);
8) alpha-anilinophenylacetamide (alpha-APA) derivatives (Pauwels et al., PNAS
USA (1993), 90:1711-15); and
9) indole derivatives (U.S. Patent No. 5,527,819 to Merck & Co. and counterpart PCT Publication No. WO 94/19321).
Indole derivatives described in U.S. Patent No. 5,527,819 assigned to Merck & Co. have been shown to be inhibitors of HIV reverse transcriptase. Some of these compounds exhibited ICso values against HIV reverse transcriptase at concentrations of from 3-35 ηM. A process for synthesizing optionally substituted indoles by a palladium- catalyzed angulation between a ketone and an iodoaniline was also developed at Merck & Co. (Chen et al., J. Org. Chem. (1997), 62(9):2676-77). The compounds disclosed in the '819 patent are generally represented by the following broad structural Formula (III):
Figure imgf000004_0001
(HI)
in which the variables X, Y, Z, R and R6 are broadly defined.
U.S. Patent No. 5,124,327 to Merck & Co. discloses a class of optionally substituted sulfonylphenyl indole compounds. The patent reports that the compounds are active as reverse transcriptase inhibitors and could be useful in the treatment of HIV infection and AIDS.
U.S. Patent No. 6,710,068 to Idenix Pharmaceuticals, Ltd., discloses a class of phenylindoles substituted with at least two moieties other than hydrogen on either or both rings. See also PCT Publication No. WO 02/083126.
PCT Publication No. WO 2004/014364 to Idenix Pharmaceuticals discloses another class of phenylindoles that displays enhanced anti-BTV activity. These compounds are also substituted with at least two moieties other than hydrogen on either or both rings, hi addition, these compounds incorporate a number of different substituents with a carboxamide functionality at position-2 on the indole, the position shown in formula (II) above as "Z". Typical placement of substituents is at the 3" and 5" positions on the phenyl ring, and at the 4' and 5', 5' and 6', or 5' and T positions on the benzo ring of the indole moiety.
Bristol Myers Squibb disclose various optionally substituted indoles, azaindoles, piperazines, and pyrrolidines for the treatment of HIV and/or AIDS in several U.S. patents and U.S. and PCT publications. See U.S. Publication Nos. 2004/0006090; 2004/0063746; 2003/0096825; 2003/0236277; and WO 03/068221.
WO 01/02388 to SmithKline Beecham S.P.A discloses optionally substituted phenylindoles with a carbamyl substituent that are alleged to have utility in the treatment of HIV5 AIDS, osteoporosis, cancers, and Alzheimer's disease. Warner-Lambert Company discloses various indole-thiazepinones, oxazepinones, diazepinones, benzothiophenes, benzofurans, and indole-2-carboxamides for the the treatment of HIV in U.S. 5,424,329; U.S. 5,565,446; U.S. 5,703,069; and WO 96/29077.
Shinogi & Co. report optionally substituted indole derivatives that are viral integrase inhibitors useful as anti-HIV drugs in U.S. Publication No. 2002/0019434 and
U.S. Patent Nos. 6,716,605 and 6,506,787.
U.S. Patent No. 5,945,440 to Kleinschroth et al. discloses a class of indolocarbazole amides for the treatment of a variety of diseases including cancer, viral diseases (including HIV), cardiac and vascular diseases, bronchopulmonary diseases, inflammatory disorders, degenerative diseases of the central nervous system, and other diseases.
U.S. Patent No. 4,866,084 to Gunasekera et al. teaches certain bisindole alkaloid compounds that have antiviral and antitumor activity, including HSV (herpes simplex virus). U.S. Patent 5,935,982 to Dykstra et al. reports a different class of bisindoles that have utility versus retroviral infections and especially HIV.
U.S. Patent No. 5,852,011 to Matsunaga et al. discloses a class of indole derivatives substituted by a heteroaryl function and an amide function. The compounds are said to possess antitumor, antiviral, and antimicrobial properties.
U.S. Patent No. 5,935,982 to Dykstra et al. discloses a class of bis-indoles and specifically propose their use for treating retroviral infections, and especially infection by
HIV.
U.S. Patent No. 5,929,114 to Domagala et al. discloses a class of arylthio and bithiobisarylamide compounds, including indole derivative, that reportedly have antibacterial and antiviral activity.
U.S. Patent No. 5,830,894 to Pevear et al. discloses a class of triazinoindole derivatives that reportedly have anti-pestivirus activity, most notably BVDV activity.
Indoles have been used in the treatment of diseases other than HIV. U.S. Patent
No. 5,981,525 to Farina et al. discloses a complex array of indoles for use in the treatment of osteoporosis based on their ability to inhibit osteoclast H+-ATPase and thus reduce bone resorption. U.S. Patent No. 6,025,390, also to Farina et al., teaches another group of indole derivatives, termed heteroaromatic pentadienoic acid derivatives, also for the treatment of osteoporosis. U.S. Patent No. 5,489,685 to Houpis et al. discloses a series of compounds that are furo(2,3-b) pyridine carboxylic acid esters, allegedly useful in the treatment of HIV.
It is known that over a period of time, antiviral agents that are active against HIV induce mutations in the virus that reduce the efficacy of the drug. This was apparently the problem exhibited by the Merck indoles in U.S. Patent No. 5,527,819 (Williams et al, J. Med. Chem., 1993, 36(9), 1291-94). Drug resistance most typically occurs by mutation of a gene that encodes an enzyme used in viral replication, and most typically in the case of HIV, reverse transcriptase, protease, or DNA integrase. It has been demonstrated that the efficacy of a drug against HIV infection can be prolonged, augmented, or restored by administering the compound in combination or alternation with a second, and perhaps third, antiviral compound that induces a different mutation from that caused by the principal drug. Alternatively, the pharmacokinetics, biodistribution, or other parameters of a drug can be altered by such combination or alternation therapy, hi general, combination therapy is typical rather than alternation therapy since combination therapy induces multiple simultaneous pressures on the virus. However, one cannot predict which mutations will be induced in the HIV-I genome by a given drug, whether the mutations are permanent or transient, or how an infected cell with a mutated HIV-I sequence will respond to therapy with other agents in combination or alternation. These factors are exacerbated by the fact that there is a paucity of data on the kinetics of drug resistance in long-term cell cultures treated with modern antiretroviral agents.
Therefore, there is a need to provide new compounds and methods for the treatment of HIV.
It is thus an object of the present invention to provide new compounds, compositions, methods and uses for the treatment of patients infected with HIV.
It is yet another object of the present invention to provide new compositions and methods for the treatment of patients infected with HIV that exhibit activity against drug-resistant forms of the virus. SUMMARY OF THE INVENTION
3-phosphoindole compounds dispay antiviral activity against HIV, in particular against strains of HIV that have developed cross resistance to other anti-HIV drugs. Compounds, compositions and methods for treatment of HIV infection are disclosed that include the 3-phosphoindole compounds. The 3-phosphoindoles can be in the form of a wide variety of moieties, including, but not limited to, phosphates, phosphonates, a phosphorthioate, including .thiophosphates, thiophosphonates, phosphate, and a phosphoramidate, including.iminiophosphates and iminophosphonates.
In one embodiment, the compounds that can have anti-HIV activity are of Formula (A) below. These compounds include an phosphorus-linked substituent at position-3 and particular substituents at position-2 on the indole, and a monosubstitution at position R5 or a disubstitution at positions R4 and R5 , R5 and R6 , or R5 and R7 on the benzo ring.
Figure imgf000007_0001
In one particular embodiment, substituent "X" represents a phenyl ring that is unsubstituted or is substituted by one or more halogens or lower alkyl groups such as methyl or ethyl. Particular substituents at position 2 on the indole moiety include, for example, hydrogen, hydroxy, halogen, alkyl, aryl, heteroaryl, and especially substituents having a carboxamide or carboxamide moiety shown as "Z" in Formula (A). Substituents for the benzo ring of the indole moiety include but are not limited to chlorine, fluorine, bromine, iodine, CF3, CN, NO2, and methoxy.
The active compound may be a salt or prodrug that, upon administration, provides directly or indirectly the parent compound or that itself exhibits a desired activity. In another embodiment, the compound of Formula A includes a charged heteroatom, and in a particular embodiment, the compound includes an N-oxide group.
Modifications affecting the biological activity of the compounds of the present invention also are included here, as are any changes that produce increased activity over that of the parent compounds.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 is an illustration of three (3) general structures of the present invention given as Formula (A), Formula (B), and Formula (C).
Figure 2 is an illustration of phosphorylated compounds (I)-(X) of the general Formulae (A) and (B).
Figure 3 is an illustration of 9- and 10-membered bicyclic phosphorylated compounds (XI)-(XX) of the general Formula (C).
DETAILED DESCRIPTION OF THE INVENTION
Provided are a composition of matter, method of use and a pharmaceutical composition for the treatment of retroviral infections in mammals and, in particular, HIV in humans. Included within the present invention are the following features:
• 3-phosphoindoles and pharmaceutically acceptable salts and prodrugs thereof as described herein, optionally substantially free of other chemical entities;
• 3-phosphoindoles of Formulas A-C and pharmaceutically acceptable salts and prodrugs thereof as described herein, optionally substantially free of other chemical entities;
• 3-phosphoindoles and pharmaceutically acceptable salts and prodrugs thereof as described herein that are effective against HTV in a host;
• 3-phosphoindoles and pharmaceutically acceptable salts and prodrugs thereof as described herein that are effective against drug-resistant strains of HIV in a host, and, in certain embodiments, where the drug-resistant strains of HIV is due to a reverse transcriptase mutation, such as lysine 103 -> asparagine and/or tyrosine 181 -> cysteine;
• 3-phosphoindoles and pharmaceutically acceptable salts and prodrugs thereof as described herein for use in the treatment or prophylaxis of an HIV infection in a host, or in the manufacture of a medicament for the treatment or prophylaxis of an HIV infection in a host, especially in individuals diagnosed as having an HIV infection or being at risk for such infection;
• 3-phosphoindoles and pharmaceutically acceptable salts and prodrugs thereof as described herein for use in the treatment or prophylaxis of an HIV infection, or in the manufacture of a medicament for the treatment or prophylaxis of an HIV infection, which is resistant to one or more reverse transcriptase inhibitors, in a host;
• 3-phosphoindoles and their pharmaceutically acceptable salts and prodrugs thereor as described herein for use in the treatment or prophylaxis of an HIV infection as a form of salvage therapy in a host, or in the manufacture of a medicament for the treatment or prophylaxis of an HIV infection as a form of salvage therapy in a host, especially in individuals diagnosed as having an HIV infection or being at risk for such infection;
• 3-phosphoindoles and their pharmaceutically acceptable salts and prodrugs thereor as described herein for use in the treatment or prophylaxis, or in the manufacture of a medicament for the treatment or prophylaxis, of an HIV infection that is resistant to one or more reverse transcriptase inhibitors due to a reverse transcriptase mutation, such as lysine 103 -> asparagine and/or tyrosine 181 -> cysteine, in a host, especially in individuals diagnosed as having an HIV infection or being at risk for such infection;
• processes for the preparation of 3-phosphoindoles, optionally substantially isolated from other chemical entities;
• pharmaceutical compositions comprising an effective anti-HIV treatment amount of a 3-phosphoindole or its pharmaceutically acceptable salt or prodrug thereof, together with a pharmaceutically acceptable carrier or diluent;
• pharmaceutical compositions comprising an effective anti-HIV treatment amount of a 3-phosphoindole or its pharmaceutically acceptable salt or prodrug thereof, in combination with one or more other anti-HIV agents, optionally with a pharmaceutically acceptable carrier or diluent; • pharmaceutical compositions for the treatment or prophylaxis of an HIV infection that is resistant to one or more reverse transcriptase inhibitors, in a host, comprising an effective anti-HIV treatment amount of a 3-phosphoindole or its pharmaceutically acceptable salt or prodrug, optionally in combination with at least one other anti-HIV agent, optionally with a pharmaceutically acceptable carrier or diluent;
• pharmaceutical compositions for the treatment or prophylaxis of an HIV infection in a host as a form of salvage therapy comprising an effective anti-HIV treatment amount of a 3-phosphoindole or its pharmaceutically acceptable salt or prodrug, optionally in combination with at least one other anti-HIV agent, and optionally with a pharmaceutically acceptable carrier or diluent;
• pharmaceutical compositions for the treatment or prophylaxis of an HIV infection that is resistant to one or more reverse transcriptase inhibitors due to a reverse transcriptase mutation, such as lysine 103 -> asparagine and/or tyrosine 181 -^ cysteine, in a host comprising an effective anti-HIV treatment amount of a 3-phosphoindole or its pharmaceutically acceptable salt or prodrug, optionally in combination with at least one other anti-HIV agent, and optionally with a pharmaceutically acceptable carrier or diluent;
• methods for the treatment or prophylaxis of an HIV infection in a host, wherein the HIV can be resistant to one or more reverse transcriptase inhibitors, comprising administering to said host an effective anti-HIV treatment amount of a 3-phosphoindole or its pharmaceutically acceptable salt or prodrug, optionally in combination or alternation with at least one other anti-HIV agent, and optionally with a pharmaceutically acceptable carrier or diluent;
• methods for the treatment or prophylaxis of an HIV infection in a host as a form of salvage therapy comprising administering to said host an effective anti-HIV treatment amount of a 3-phosphoindole or its pharmaceutically acceptable salt or prodrug, optionally in combination or alternation with at least one other anti- HIV agent, and optionally with a pharmaceutically acceptable carrier or diluent;
• methods for the treatment or prophylaxis of an HIV infection, which is resistant to one or more reverse transcriptase inhibitors due to a reverse transcriptase mutation, such as lysine 103 -^ asparagine and/or tyrosine 181 1^ cysteine, in a host comprising administering to said host an effective anti-HIV treatment amount of a 3-phosphoindole or its pharmaceutically acceptable salt or prodrug, optionally in combination or alternation with at least one other anti-HIV agent, and optionally with a pharmaceutically acceptable carrier or diluent;
• use of a 3-phosphoindole or its pharmaceutically acceptable salt or prodrug, optionally in combination or alternation with at least one other anti-HIV agent, and optionally with a pharmaceutically acceptable carrier or diluent, for the treatment or prophylaxis of an HIV infection in a host;
• use of a 3-phosphoindole or its pharmaceutically acceptable salt or prodrug, optionally in combination or alternation with at least one other anti-HIV agent, and optionally with a pharmaceutically acceptable carrier or diluent, for the treatment or prophylaxis of an HIV infection, or in the manufacture of a medicament for the treatment or prophylaxis of an HIV infection, which is resistant to one or more reverse transcriptase inhibitors, which can be be due to a reverse transcriptase mutation, such as lysine 103 -> asparagine and/or tyrosine 181 -> cysteine, in a host. This use can be a form of salvage therapy; and
• any or all of the foregoing in which the host is a human.
I. Active Compounds of the Present Invention
In a general embodiment of the invention, a 3-phosphoindole or a pharmaceutically acceptable salt, prodrug, N— oxide, quaternary amine, stereochemical isomer or tautomer thereof is provided. The 3-phosphoindole can be in the form of a phosphate, phosphonate, thiophosphate, thiophosphonate, iminiophosphate or iminophosphonate.
In a first embodiment of the present invention, the compound is represented generally by the following chemical Formula (A):
Figure imgf000012_0001
or a pharmaceutically acceptable salt, prodrug, N— oxide, quaternary amine, stereochemical isomer or tautomer thereof, wherein:
each X and Y independently is
a) H;
b) halogen (F, Cl, Br, or I), typically F;
c) R3;
d) CF3;
e) Ci-β alkyl;
f) C2-6 alkenyl;
g) C2-6 alkynyl;
h) alkylheterocycle;
i) 3-14 membered carbocycle, aryl, h monocyclic, bicyclic, tricyclic or spiro structure; j) OH;
k) OR2;
1) O-alkyl;
m) O-alkenyl;
n) O-alkynyl;
o) O-alkylaryl;
p) O-aryl;
q) O-heterocycle;
r) O-aralkyl;
s) O-carbocycle;
t) SH;
u) SR2;
v) S-alkyl;
w) S-alkenyl;
x) S-alkynyl; y) S-alkylaryl;
z) S-aryl;
aa) S-heterocycle;
bb) S-aralkyl;
cc) S-carbocycle;
dd) NH2;
ee) NHR2;
ff) NR2R2;
gg) NH-alkyl; hh) N-dialkyl;
ii) NH-aryl;
jj) N-alkaryl;
kk) N-aralkyl;
11) NH-heterocycle;
mm) N-alkyl-heterocycle;
nn) N-alkenyl-heterocycle;
00) N-alkynyl-heterocycle; or
alternatively, X and Y may come together to form an optionally substituted bicyclic or tricyclic phosphorylated heterocycle wherein each ring comprises 3-7 members;
Z is:
a) H;
b) CN;
c) NO2;
d) Ci-6 alkyl;
e) C2-6 alkenyl;
f) C2-6 alkynyl;
g) alkaryl;
h) aralkyl;
i) heterocycle;
j) alkyl-heterocycle;
k) aryl;
1) alkoxy;
m) OR2; n) SR2;
o) S(O)nR2;
p) S(O)n-NR2R3;
q) N(R2)(R3);
r) carboxamido;
s) amido;
t) acyl;
u) C(=W)-R3; v) C(=W)NH-C(R3)(R3)-C(=W)-N(R2)(R2);
w) C(=W)NH-P(=W)(R3)-A-R3;
x) C(=W)NH-A-S(O)n-NR2;
y) C(=W)NH-CR3R3-S(O)n -NR2R2;
z) C(=W)-NH-A-C(=W)-N(R2)(R2);
aa) C(=W)-N(R2)(R2);
bb) C(=W)-NH-A-R3;
cc) C(=W)-NH-NH-R3;
dd) C(=W)-NH-C(R3)(R3)-C(=W)NH-C(R3)(R3)C(=W)-N(R2)(R2);
ee) C(=W)-NH-R2;
ff) C(=W)-NH-A-C(=W)-NH-A-C(=W)-NH2;
gg) C(R2)(R3)(R3);
hh) C(R2)(R3)-NH-R2;
ii) A-S(O)n-R3;
U) C(=W)-A-C(=W)-A-C(=W)R3;
kk) A-R3; 11) C(=W)-(O)R2;
mm) C(=W)-A-C(=W)-NH2; nn) an amino acid residue;
oo) C(=W)-N(R2)-A-(amino acid residue);
pp) C(=W)-N(R2)-A-(amino acid residue)-C(=W)-R3;
qq) C(=W)-amino acid residue;
rr) C(=W)-N(R2)-A-(amino acid residue)-A-C(=W)-R3;
ss) C(=W)-OR2;
tt) C(=W)-S(R2);
uu) C(=W)-NH-NH-R2;
w) C(=W)-NH-N(R2)-A-C(=W)R3;
ww) C(=W)-N(R2)-C(=W)-R3;
xx) C(=W)-A-NH-C(=W)R3;
yy) C(=W)-A-NH-C(=W)OR2;
zz) C(=W)-A-R3;
aaa) C(=W)-NH-NH-CH2-C(=W)R3;
bbb) P(=W)(R3)(R3); or
ccc) A-P(=W)(R3)(R3).
ddd) C(=W)-NH-C1-10alkyl-heteroaryl
eee) C(=W)-NH-C1-4alkyl-heteroaryl
fff) C(=W)-NH-CH2-heteroaryl
Figure imgf000016_0001
?gg) wherein in embodiments (ddd), (eee) and (fff), the heteroaryl can optionally include a charged heteroatom, and in particular can include an N-oxide
wherein each X, Y and Z independently may be unsubstituted or substituted by one or more of C1-6 alkyl; alkoxy; OH; oxo; halo (F, Cl, Br, or I); NR2R2; optionally substituted aryl; optionally substituted heterocycle; O-C(=W)-alkyl; C(=W)-OR2; CN; NO2; NH-C(=W)-alkyl; NH-S(O)n-alkyl; NH-S(O)n-NR2R2; or C3-6 cycloalkyl;
each W is independently:
a) O;
b) S;
c) NH;
Φ N-N(R2)(R2);
e) N(R2);
f) N-OH;
g) N-O-alkyl; or
h) N-O-R2;
R1 is:
a) H;
b) -R2;
c) C(=W)-R3;
d) C(=W)-O(R2);
e) C(=W)-S(R2);
f) Q=W)-NH-R2;
g) C(=W)-N(R2)(R2);
h) C(=W)-NH-A-(amino acid residue);
i) A-(amino acid residue)-R3; j) S(O)n-R3; or
k) S(O)2-N(R2)(R2);
any of which optionally may be substituted by one or more of C1-6 alkyl; OH; alkoxy; aryl; halo; CN; NO2; or N(R2)(R2);
each R2 is independently:
a) H;
b) CF3;
c) CN;
d) optionally substituted, branched or unbranched, alkyl, such as a C1-6 alkyl;
e) optionally substituted, branched or unbranched, alkenyl, such as a C2-6 alkenyl;
f) optionally substituted, branched or unbranched, alkynyl, such as a C2-6 alkynyl;
g) 3-14 membered carbocycle;
h) optionally substituted aryl;
i) optionally substituted aralkyl;
j) optionally substituted alkylaryl;
k) optionally substituted heterocycle;
1) optionally substituted alkylheterocycle;
m) optionally substituted heterocycle-alkyl;
n) A-heterocycle;
o) acyl;
p) alkoxy;
q) CH2-S(O)nR3;
r) C(alkyl)2-S(O)nalkyl; s) CH(alkyl)-S(O)nalkyl;
t) CH2NB2; u) CH2NH(alkyl);
v) CH2N(alkyl)2;
w) CH(alkyl)-NH2;
x) CH(alkyl)-NH(alkyl);
y) CH(alkyl)-N(alkyl)2;
z) C(alkyl)2-NH2; aa) C(alkyl)2-NH(alkyl);
bb) C(alkyl)2-N(alkyl)2;
cc) CH2-C(=W)H;
dd) CH2-C(=W)alkyl;
ee) A-alkyl;
fit) C(alkyl)2-C(=W)alkyl;
gg) CH2-C(^W)H;
hh) CH2-C(=W)alkenyl;
ii) CH(alkenyl)-C(=W)H;
jj) A-S(O)alkyl;
kk) CH(NH)-S(O)nalkyl; or
U) A-N(NH)alkyl;
mm) C(R3)(R3)-S(O)nNH2;
nn) C(R3)(R3)-S(O)nCF3;
oo) C(R3)(R3)-NH2;
each R3 is independently: a) H; b) OH; c) halogen (F, Cl, Br, or I);
d) CF3;
e) CN;
f) optionally substituted, branched or uribranched, alkyl, such as a C1-6 alkyl;
g) optionally substituted, branched or unbranched, alkenyl, such as a C2-6 alkenyl;
h) optionally substituted, branched or unbranched, alkynyl, such as a C2-6 alkynyl;
i) 3-14 membered carbocycle;
j) optionally substituted aryl;
k) optionally substituted aralkyl;
1) optionally substituted alkylaryl;
m) optionally substituted heterocycle;
n) optionally substituted alkylheterocycle;
o) optionally substituted heterocycle-alkyl;
p) A-heterocycle;
q) acyl;
r) carboxamido;
s) carbamoyl;
t) alkoxy;
u) OH
v) OR2; w) O-alkyl;
x) O-alkenyl;
y) O-alkynyl;
z) O-alkaryl;
aa) O-aralkyl;
bb) O-carbocycle;
cc) O-heterocycle;
dd) O-aryl;
ee) SH
ff) SR2
gg) S-alkyl;
hh) S-alkenyl;
ii) S-alkynyl;
jj) S-alkaryl;
kk) S-aralkyl;
11) S-carbocycle;
mm) S-heterocycle;
nn) S-aryl;
oo) S(O)n-R2;
pp) amino;
qq) NH2; rr) NHR2;
ss) N(R2XR2);
tt) NH-S(O)n-R2; uu) NHC(=W)-aryl; w) NHC(=W)-alkyl;
ww) NH-C(=W)-heterocycle;
xx) CH2-S(O)nR2; yy) C(=W)R2;
zz) C(=W)-N(R2)-R2;
aaa) C(alkyl)2-S(O)nR2;
bbb) CH(alkyl)-S(O)nR2;
ccc) C(alkyl)2-NH2;
ddd) CH(alkyl)-N(alkyl)R2;
eee) C(R2)(R2)-NR2R2;
fff) CH2N(alkyl)R2;
ggg) CH(alkyl)-NHR2;
hhh) C(alkyl)2-NHR2;
iii) C(alkyl)2-N(alkyl)R2;
jjj) CH2-C(=W)H;
kkk) CH2-C(=W)alkyl;
111) CR2R2-C(=W)R2;
mmm)A-R ;
nnn) C(R2)2-C(=W)R2;
ooo) CH2-C(=W)H;
ppp) CH2-C(=W)alkenyl;
qqq) CH(alkenyl)-C(=W)H;
irr) A-S(O)R2; sss) CH(NH)-S(O)nR2; or
ttt) A-N(NH)R2;
uuu) C(R2)(R2)-S(O)nNH2;
VW) C(R2)(R2)-S(O)nCF3;
www) C(R2)(R2)-NH2;
wherein the optional substitution comprises one or more of
a) a substituted or unsubstituted heterocycle;
b) C(=W)O-aryl;
c) C(=W)O-alkyl;
d) C(=W)NH2;
e) C(=W)NH-alkyl;
f) C(=W)NΗ-aryl;
g) C(=W)N-di-alkyl;
h) C(=W)N(alkyl)-aryl;
i) α-amino acid;
j) α-amino ester;
k) α-amino-carboxamide;
1) β-amino acid;
m) β-amino ester; or
n) β-amino-carboxarnide;
wherein, when the optional substitution includes a substituted heterocycle, then the heterocycle substitution is selected from the group consisting of:
a) C(=W)O-aryl;
b) C(=W)O-alkyl; c) C(=W)NH2;
d) C(=W)NH-aryl;
e) C(=W)NH-alkyl;
f) C(=W)N-di-alkyl;
g) C(=W)N(alkyl)-aryl;
h) α-amino acid;
i) α-amino ester;
j) α-amino-carboxamide;
k) β-amino acid;
1) β-amino ester; and
m) β-amino-carboxamide;
n) halo; or
o) cyano,
alone or in any combination;
n independently is 0, 1 or 2;
each A is independently a disubstituted spacer selected from the group consisting of:
a) C1-6 alkylene, branched or uribranched, and optionally having one or more heteroatoms, aryl, cycloalkyl or heterocyclyl functions in or attached to the chain;
b) C2-12 alkenylene, branched or unbranced, and optionally having one or more heteroatoms, aryl, cycloalkyl or heterocyclyl functions in or attached to the chain;
c) C2-12 alkynylene, branched or unbranced, and optionally having one or more heteroatoms, aryl, cycloalkyl or heterocyclyl functions in or attached to the chain;
d) optionally substituted arylene; e) O-alkylene, branched or uribranched, and optionally having one or more heteroatoms, aryl, cycloalkyl or heterocyclyl functions in or attached to the chain;
f) aralkylene, branched or unbranched, and optionally having one or more heteroatoms, aryl, cycloalkyl or heterocyclyl functions in or attached to the chain;
g) optionally substituted cycloalkyl; and
h) optionally substituted heterocycle;
wherein "A" may be joined by any desired linkage such as, for example, an ether, thioether, amino, carboxamido, ester or carbon-carbon linkage, or any combinatin thereof;
each R4', R5', R6> and R7' independently is:
a) H;
V) halogen (F, Cl, Br, I);
c) NO2;
d) CN;
e) CF3;
f) OH
g) OR2;
h) SH
i) SR2;
j) NR2R2;
k) NHS(O)nR2;
1) NHCO-C1-3 alkyl;
m) S(O)nR2;
n) aryl; o) heterocycle;
p) C1-6 alkyl;
q) C2-6 alkenyl;
r) C2-6 alkynyl;
s) C(=W)-S(O)nR2;
t) C(=W)-S(O)n-NR2R2;
u) C(=W)-aryl;
v) C(=W)-alkyl;
w) C(=W)-heterocycle; or
x) C(=W)-NR2R2; each of which optionally may be substituted with one or more of :
a) OR2;
b) S(O)nR2;
c) C(=W)-S(O)nR2;
d) C(=W)-S(O)n-NR2R2;
e) C(=W)-aryl;
f) C(=W)-alkyl;
g) C(=W)-heterocycle;
h) C(=W)NR2R2;
i) NO2;
j) CN; k) CF3;
1) halogen (F5 Cl, Br, I);
m) NHS(O)nR2; n) NHCO-C1-3 alkyl;
o) aryl;
p) heterocycle;
q) C1-6 alkyl;
r) C2-6 alkenyl;
s) C2-6 alkynyl; or
t) NR2R2.
In one embodiment of Formula (A), X is an optionally substituted phenyl; Y is any of its definitions; R1, R4 , R6 , and R7 are all hydrogen; R5 is halogen; and Z is a carboxamide moiety.
In an alternative embodiment, R1 is acyl, alkyl, aryl, alkaryl, or aralkyl.
In yet another alternative embodiment, R4 is fluoro, nitro or cyano, W is oxygen, and Y is O-alkyl.
In a second embodiment of Formula (A), X is an optionally substituted phenyl; Y is any of its definitions; R1, R4 , R6 , and R7 are all hydrogen; R5 is chlorine; and Z is a carboxamide or carboxamido-heterocyclyl moiety.
In yet another embodiment of Formula (A), X is tolyl, thiazolyl or pyridyl; Y is H, OH, or O-alkyl; R1, R4', R6', and R7' are all hydrogen; R5> is halogen; and Z is carboxamido, acyl, an alkyl-sulphonyl group or a carboxylic acid derivative.
In an alternative to the preceding embodiment, X, Y, R1, R4 , R6 , R7 and R5 all are as defined above, and Z is a carboxamido-alkylene-heterocycle, typically carboxamido-alkylene-pyridyl; a thioamido-pyridyl wherein the pyridyl is unsubstituted or substituted by OH, OMe or lower alkyl; an imino-nitrile; or an alkylsulphonyl-aryl group.
In yet another embodiment, Z is a carboxamido-alkylene-heterocycle wherein the heterocycle includes at least one N-oxide group. In a second embodiment, the invention provides a phenylindole for use in the treatment of HIV represented by the following general Formula (B):
Figure imgf000028_0001
or a pharmaceutically acceptable salt, prodrug, N — oxide, quaternary amine,
stereochemical isomer or tautomer thereof, wherein:
each W, Y, Z, R , 11, τ R>2%
Figure imgf000028_0002
A, n, R »44' , r R»53' , τ R-ι 6D' and R >V' is as defined above for Formula (A); and
each R2", R3", R4", R5", and R6" independently is:
a) H;
b) halogen;
c) NO2;
d) CN;
e) OR2;
f) SR2;
g) NH2;
h) NR2R3;
i) N(R2)-C(=W)-C1-4 alkyl;
j) N(R2)-SO2-C1-4 alkyl;
k) C1-6 alkyl;
1) C2-6 alkenyl; m) C2-6 alkynyl; n) aryl;
o) CF3;
p) CR2R2-S(O)n-R3;
q) CR2R2NR2R3;
r) C-OH;
s) CR2R2-C(=W)R2;
t) acyl;
u) C(=W)R2;
v) C(=W)0R2;
w) C(=W)SR2;
x) C(=W)-MR2R3;
y)
Figure imgf000029_0001
acid residue);
z) amino residue; or
aa) A-(amino acid residue);
wherein any of the above optionally may be substituted; or
alternatively, R or R may be joined to Y to form an optionally substituted bicyclic or tricyclic phosphorylated heterocycle wherein each ring comprises 3-14 members.
The following are non-limiting examples of embodiments of Formula (B):
a) Y is alkyl; W is O, S(O)n, or NH; R1, R4', R6', R7', R2", R3", R4", R5", and R6" are all hydrogen; R5 is halogen; and Z is carboxamide;
b) Y is aryl; W is O, S(O)n, or NH; R1, R4', R6>, R7', R2", R3", R4", R5", and R6" are all hydrogen; R5 is amino-alkyl, thioamino-alkyl, or aminocarbonyl-alkyl; and Z is carboxamide; c) Y is -OH or -SR2; W is O, S(O)n, or N-NH2; R1, R4', R6', R7', R2", R4", and R6" are all hydrogen; R5' is halogen; R3" and R5 ' are methyl; and Z is carboxamide;
d) Y is -OH; W is O, S(O)n, or NH; R1, R6>, R7', R2", R4", and R6" are all hydrogen; R4' and R5' are halogen; R3" and R5 are methyl; and Z is carboxamide;
e) Y is -OH; W is O, S(O)n, or N-NR2R2; R1, R4', R6', R7', R2", R3", R4", R5", and R6' are all hydrogen; R5 is halogen; and Z is carboxamide;
f) Y is alkenyl; W is O, S(O)n, or NH; R1, R4>, R6', R7', R2", R3", R4", R5", and R6" a arree a allll h hyyddrr<ogen; R5 is halogen; and Z is carboxamide or carboxamide- heterocycle;
g) Y is alkynyl or -NR2R3; W is O5 S(O)n, or N-O-alkyl; R1, R4', R6', R7>, R2", R3 , R4", R5 , and R6 are all hydrogen; R5' is halogen; and Z is carboxamide;
h) Y is alkenylene; W is O5 S(O)n, or NH; R1, R4', R6', R7', R2", R3", R4", R5", and R6 are all hydrogen; R5 is halogen; and Z is carboxamide;
i) Y is ethyl; W is O5 S(O)n, or N-OH; R1, R4', R6>, R7', R2", R3", R4", R5", and R6 are all hydrogen; R5 is chloro; and Z is carboxamide;
j) Y is -O-methyl; W is O5 S(O)n, or NH; R1, R4>, R6' 5 R7', R2", R3", R4", R5", and R6 are all hydrogen; R5 is halogen; and Z is carboxamide;
k) Y is -O-ethyl; W is O5 S(O)n, or N-NH2; R1, R4', R6', R7', R2", R3", R4", R5", and R6 are all hydrogen; R5 is halogen; and Z is carboxamide -heterocycle;
1) Yis -O-ethyl; W is O, S(O)n, or NH; R1, R4', R6', R7', R2", R3", R4", R5", and R6 are all hydrogen; R5 is chloro; and Z is carboxamide;
m) Y is -0-H; W is O, S(O)n, or NH; R1, R4', R6', R7>, R2", R3", R4"5 R5", and R6" are all hydrogen; R5 is halogen; and Z is a carboxamide alkyl group that is unsubstituted or substituted by one or more -NO2, -NH-C(=O)-alkyl, or -NH- S(O)n-alkyl;
n) Y is -0-H; W is O, S(O)n, or N-NH2; R1, R4', R6>, R7>, R2", R3", R4", R5", and R6 are all hydrogen; R5' is halogen; and Z is a carboxamido-alkylene- heterocyclyl group wherein said heterocycle imidazole, thiazole, pyridyl or furan, and wherein the heterocycle is unsubstituted or further substituted by one or more halo, oxo, -OH, -NO2, -MeOH, -NH-C(=O)-alkyl, or -NH-S(O)n- alkyl;
o) Y is -O-H; W is O, S(O)n, or NH; R1, R4', R6>, R7', R2", R3", R4", R5", and R6" are all hydrogen; R5' is halogen; and Z is carboxamide-cyclopropyl or carboxamide-cyclobutyl;
p) Y is -O-methyl; W is O, S(O)n, or NH; R1, R4', R6', R7', R2", R3", R4", R5", and R6" are all hydrogen; R5' is halogen; and Z is a carboxamide-ethyl, carboxamide -ethanol, or carboxamide -ethyl-methoxy;
q) Y is -OH or -NR2R3; W is O, S(O)n, or N-O-alkyl; R1, R4', R6', R7', R2", R3", R4", R5 , and R6 are all hydrogen; R5 is halogen; and Z is a carboxamide alkyl-phenyl group wherein the phenyl is further substituted by one or more halo, oxo, -OH, -OCH3, -NO2, -MeOH, or -NH-C(=O)-alkyl groups;
r) Y is -OH or -SR2; W is O, S(O)n, or N-NR2R2; R1 , R4>, R6', R7', R2", R3", R4",
R5 , and R6 are all hydrogen; R5 is halogen; and Z is an isopropanol carboxamide moiety; and
s) Y is -OH; W is O, S(O)n, or N-OH; R1, R4', R6', R7', R2", R3", R4", R5", and R6" are all hydrogen; R5 is halogen; and Z is a thioacetamide group.
In a third embodiment, the present invention provides an optionally substituted 9-
11 membered bicyclic 3-phosphoindole for use in the treatment of HIV represented by the following general Formula (C):
Figure imgf000031_0001
(C) or a pharmaceutically acceptable salt, prodrug, N— oxide, quaternary amine, stereochemical isomer or tautomer thereof, wherein:
each W5 Z, R1, R2, R3, A5 n, R4', R5', R6' and R7' is as defined above for Formula (A);
each R3", R4", R5", and R6" is as defined above for Formula (B);
indicates the presence of a single or double bond, wherein:
when indicates the presence of a double bond, each Y and T independently is:
a) CR3;
b) N; or
c) S(=W); such that at least one of Y and T is CR3; and
when indicates the presence of a single bond, then each Y and T independently is:
a) CHR3;
b) C(R3)(R3);
c) O;
d) S; or
e) NR2;
such that at least one of Y and T is C(R3)(R3); and
m is 1 or 2, with the proviso that m can only be 2 for T or Y = CR2.
The following are non-limiting examples of embodiments of Formula (C):
a) W is O, Y is CR2, T is (CH2)2, R1, R4', R6', R7', R3", R4", R5", and R6" are all H; R5 is halogen, and Z is carboxamide; b) W is S, Y is O, T is CH2, R1, R4', R6', R7', R3", R4", R5", and R6" are all H; R5' is halogen, and Z is carboxamido-heterocycle wherein said heterocycle is optionally substituted furan, imidazole, thiazole, or pyridyl;
c) W is S5 Y is NR2, T is CH2, R1, R4', R6', R7', R3", R4", R5", and R6" are all H; R5' is halogen, and Z is carboxamide;
d) W is O, Y is SR2, T is CH2, R1, R4', R6', R7', R3", R4", R5", and R6" are all H; R5' is halogen, and Z is alkyl-carboxamide;
e) W is S, Y is SR2, T is CH2, R1, R4', R6', R7', R3", R4", R5", and R6" are all H; R5' is halogen, and Z is carboxamide;
f) W is O, Y is CR2, T is C-C(=W)R3, R1, R4', R6', R7', R3", R4", R5", and R6" are all H; R5 is halogen, and Z is methoxymethyl-carboxamide;
g) W is S, Y is O, T is C-C(=W)R3, R1, R4>, R6', R7', R3", R4", R5", and R6" are all H; R5' is halogen, and Z is carboxamide-cyclobutyl;
h) W is O, Y is SR2, T is C-C(=W)R3, R4>, R6', R7', R3", R4", R5", and R6" are all H; R5 is chloro, and Z is alkyl carboxamide wherein alkyl is optionally substituted by one or more halo, oxo, -OH, -NO2, -MeOH, -NH-C(=O)alkyl, or-NH-S(O)n-alkyl;
i) W is S, Y is NR2, T is CC(=W)R3, R4', R6', R7', R3", R4", R5", and R6" are all H; R5 is chloro, and Z is carboxamide;
j) W is S, Y is N, T is C-C(=W)R3, R1, R4', R6', R7', R3", R4", R5", and R6" are all
H; R5 is fluoro, and Z is carboxamide;
k) W is NH, Y is CR2, T is NR2, R1, R4', R6', R7', R3", R4", R5", and R6" are all H; R5 is amino-alkyl, and Z is carboxamide;
1) W is NR2, Y is O, T is (CH2)2, R1, R4', R6>, R7', R3", R4", R5", and R6" are all H; R5 is chloro, and Z is carboxamide;
m) W is N-OH, Y is O, T is CH2, R1, R4>, R6', R7>, R3", R4", R5", and R6" are all H; R5 is thioamino-alkyl, and Z is alkyl carboxamide ; n) W is S, Y is SR2, T is CH2, R1, R4>, R6', R7>, R3", R4", R5", and R6" are all H; R5' is halogen, and Z is a carboxamide-heterocyclyl group wherein the heterocycle is an optionally substituted pyridine, thiazole, imidazole or furan;
o) W is N-O-alkyl, Y is NR2, T is CH2, R1, R4', R6', R7', R3", R4", R5", and R6" are all H; R5' is halogen, and Z is carboxamide-alkyl group wherein the alkyl is optionally substituted by one or more -NO2, -NH2, -NH-C(=W)alkyl, or -NH- S(O)n-alkyl;
p) W is NH, Y is SR2, T is (CHb)2, R1, R4', R6>> R7'> R3"> R4"> R5"> mά R6" 3^ all H; R5 is halogen, and Z is carboxamide-alkyl-phenyl wherein phenyl is optionally substituted by one or more halo, oxo, OH, NO2, MeOH, -NH-
C(=O)alkyl or-NH-S(O)n-alkyl groups;
q) W is NR2, Y is (CBb)2, T is NR2, R1, R4', R6', R7', R3", R4", R5", and R6" are all H; R5 is halogen, and Z is a carboxamide-cyclopropyl group;
r) W is N-OH, Y is CH, T is O, R1, R4', R6', R7', R3", R4", R5", and R6" are all H; R5 is halogen, and Z is a carboxamide-cyclopropyl group;
s) W is N-O-alkyl, Y is O, T is (CBb)2, R1, R4', R6>, R7>, R3", R4", R5", and R6" are all H; R5 is chloro, and Z is carboxamide-methoxyethyl;
t) W is O, Y is N, T is CH2, R1, R4', R6', R7', R3", R4", R5", and R6" are all H; R5' is halogen, and Z is carboxamide-ethanol;
u) W is N-NR2R2, Y is NR2, T is CH2, R1 , R4', R6', R7', R3", R4", R5", and R6" are all H; R5 is halogen, and Z is carboxamide; and
v) W is O, Y is CR2, T is CH2, R1, R4', R6', R7', R3", R4", R5", and R6" are all H; R5 is aminocarbonyl-alkyl, and Z is carboxamide.
In one set of embodiments of Formula A, B or C, Z is C(=W)-R3; C(=W)-NH-A- C(=W)-N(R2)(R2); C(=W)-NH-A-R3; C(=W)-NH-R2; or C(=W)-A-R3.
In one embodiment of Formula A, B or C, the compound includes a charged heteroatom. hi particular, a charged nitrogen, such as through an N-oxide, is part of the compound. The charged heteroatom can be on a heteroaromatic ring that is attached to the indole, for example through C(O)NH-alkyl, or specifically through C(O)NH-CH2. In a particular embodiment of Formula A, B or C, Z is C(O)-NH-R2 wherein R2 is an optionally substituted alkylheterocycle, wherein the heterocycle is of the formula
Rx-Rx Rx' RX Rx=Rx , wherein each Rx is independently CH or TSf+-O". In a particular embodiment, one of Rx is N+-O". The alkyl group can be C1-10 alkyl, or C1-4 alkyl, or can specifically be a methylene or ethylene. In a specific embodiment, R2 is of the formula
Figure imgf000035_0001
where n is 0, 1 or 2.
In particular embodiments of Formula A, B or C, the compound is:
Figure imgf000035_0002
Figure imgf000036_0001
Additional examples of an N-oxide embodiment of a compound of Formula A, B :
Figure imgf000037_0001
. Either substituent on the phenyl ring may be substituted by CN, Me, halo, alkyl, alkenyl, alkynyl, alkyl-CN or alkenyl-CN, as the most commonly synthesized and tested N-oxides in the series to date.
II. Particular Sub-Embodiments of the Present Invention
In the first embodiment of the invention, the compounds are represented generally by the Formula (A):
Figure imgf000037_0002
or a pharmaceutically acceptable salt, prodrug, N — oxide, quaternary amine, stereochemical isomer or tautomer thereof, wherein:
X and Y, each independently, is: a) 3-14 membered carbocycle, aryl, heterocycle, any of which may comprise a monocyclic, bicyclic, tricyclic or spiro structure, or optionally may be substituted;
b) H; c) OH;
d) Cl5 Br5 I5 F;
e) CF3;
f) C1-6 alkyl;
g) C2-6 alkenyl;
h) C2-6 alkynyl;
i) alkylheterocycle;
j) NH2; k) NH-alkyl;
1) N-dialkyl;
m) NH-aryl;
n) N-alkaryl;
o) N-aralkyl;
p) NH-heterocycle;
q) N-alkyl-heterocycle;
r) N-alkenyl-heterocycle;
s) N-alkynyl-heterocycle;
t) O-alkyl;
u) O-alkenyl;
v) O-alkynyl; w) O-alkylaryl;
x) O-aryl;
y) O-heterocycle;
z) O-aralkyl;
aa) O-carbocycle;
bb) SR2; or
cc) NR2R3;
alternatively, X and Y may be joined to form an optionally substituted bicyclic or tricyclic phosphorylated heterocycle wherein each ring comprises 3-7 members;
Z is:
a) H; b) alkoxy;
c) NO2;
d) N(R2)(R3);
e) OR2;
f) carboxamido;
g) amido;
h) acyl;
i) S(O)nR2;
j) S(O)n-NR2R3;
k) C1-6 alkyl;
1) C2-6 alkenyl;
m) C2-6 alkynyl;
n) alkaryl; o) aralkyl;
p) heterocycle; q) alkyl-heterocycle;
r) aryl;
s) CN;
t) C(=W)-R2;
u) C(=W)NH-C(R2)(R2)-C(=W)-N(R2)(R2);
v) C(=W)NH-P(=W)(R2)-A-R2;
w) C(=W)NH-A-S(O)n-NR2;
x) C(=W)NH-CR2R3-S(O)n -NR2R3;
y) C(=W)-NH-A-C(=W)-N(R2)(R3);
z) C(=W)-N(R2)(R3);
aa) C(=W)-NH-A-R2;
bb) C(=W)-NH-NH-R2;
cc) C(-W)-NH-C(R2)(R2)-C(=W)NH-C(R2)(R3)C(=W)-N(R2)(R3);
dd) C(=W)-NH-R2;
ee) C(=W)-NH-A-C(=W)-NH-A-C(=W)-NH2;
ft) C(R2)(R2)(R3);
gg) C(R2)(R2)-NH-R2;
hh) A-S(O)n-R2;
ii) C(=W)-A-C(=W)-A-C(=W)R3;
Jj) A-R2; kk) C(=W)-(O)R2;
11) C(=W)-A-C(=W)-NH2; mm) an amino acid residue;
nn) C(=W)-N(R2)-A-(amino acid residue);
oo) C(=W)-N(R2)-A-(amino acid residue)-C(=W)-R2;
PP) C(=W)-amino acid residue;
qq) C(=W)-N(R2)-A-(amino acid residue)-A-C(=W)-R2;
IT) Q=W)-OR3;
ss) C(=W)-S(R2);
tt) C(=W)-NH-NH-R2;
uu) C(=W)-NH-N(R2)-A-C(=W)R2;
w) C(=W)-N(R2)-C(=W)-R3;
ww) C(=W)-A-NH-C(=W)R2;
xx) C(=W)-A-NH-C(=W)OR3;
yy) C(=W)-A-R3;
zz) C(=W)-NH-NH-CH2-C(=W)R2;
aaa) P(=W)(R2)(R2); or
bbb) A-P(=W)(R2)(R2);
wherein each of the foregoing X, Y and Z independently may be unsubstituted or substituted by one or more of:
a) H;
b) C1-6 alkyl;
c) alkoxy;
d) OH; e) oxo;
f) halo; g) NR2R2; h) optionally substituted aryl;
i) optionally substituted heterocyclyl;
j) O-C(=W)-alkyl;
k) C(=W)-0R2;
1) CN; m) NO2; n) NH-C(=W)alkyl;
o) NH-S(O)n-alkyl;
p) NH-S(O)n-NR2R2; or
q) C3-6 cycloalkyl;
W is:
a) O;
b) S;
c) NH;
d) N-N(R2)(R2);
e) N(R2);
f) N-OH; or
g) N-O-alkyl;
R1 is:
a) H;
b) R2;
c) CHV)-R2;
d) C(=W)-0(R2); e) C(=W)-S(R2); f) C(=W)-NH-R2;
g) C(=W)-N(R2)(R2);
h) C(=W)-NH-A-(amino acid residue);
i) A-(amino acid residue)-R2;
j) S(O)n -R3; or
k) S(O)2-N(R2XR2);
any of which optionally may be substituted by one or more:
a) C1-6 alkyl;
b) OH;
c) alkoxy;
d) aryl;
e) halo;
f> CN; g) NO2; or
h) N(R2XR2);
R2 is:
a) H;
b) OH;
c) halogen;
d) optionally substituted, branched or uribranched alkyl;
e) optionally substituted, branched or unbranched alkenyl;
f) optionally substituted, branched or unbranched alkynyl;
g) 3-14 membered carbocycle; h) alkylheterocycle;
i) acyl;
j) carboxamido;
k) carbamoyl;
1) alkoxy;
m) optionally substituted aryl;
n) optionally substituted aralkyl;
o) optionally substituted alkylaryl;
p) O-alkyl;
q) O-alkenyl;
r) O-alkynyl;
s) O-alkaryl;
t) O-aralkyl;
u) O-carbocycle;
v) O-heterocycle;
w) O-aryl;
x) CF3;
Y) CN; z) S(O)n-R3;
aa) N(R3)(R3);
bb) NH-S(O)n-R3;
cc) NHC(=W)-aryl;
dd) NHC(=W)-alkyl;
ee) NHC(=W)-heterocycle; ff) CH2-S(O)nR3;
gg) C(=W)R3;
hh) C(=W)NR3R3;
ii) C(alkyl)2-S(O)nR3;
jj) CH(alkyl)-S(O)nR3;
kk) C(alkyl)2-NH2;
U) CH(alkyl)-N(alkyl)R3;
mm) CR3R3-NR3R3;
nn) CH2N(alkyl)R3;
oo) CH(alkyl)-NHR3;
pp) C(alkyl)2-NHR3;
qq) C(alkyl)2-N(alkyl)R3;
rr) CH2-C(=W)H;
ss) CH2-C(=W)alkyl;
tt) CR3R3-C(=W)R3;
uu) A-R3;
w) C(R3)2-C(=W)R3;
ww) CH2-C(=W)H;
xx) CH2-C(=W)alkenyl;
yy) CH(alkenyl)-C(=W)H;
zz) A-S(O)R3;
aaa) CH(NH)-S(O)nR3; or
bbb) A-N(NH)R3;
wherein said optional substitution comprises one or more of: a) a substituted or unsubstituted heterocycle;
b) C(=W)O-aryl; c) C(=W)O-alkyl;
d) C(=W)NH2;
e) C(=W)NH-alkyl;
f) C(=W)NH-aryl;
g) C(=W)N-di-alkyl;
h) C(=W)N(alkyl)-aryl;
i) α-amino acid;
j) α-amino ester;
k) α-amino-carboxamide;
1) β-amino acid;
m) β-amino ester; or
n) β-amino carboxamide;
R3 is:
a) H;
t>) OH;
c) Ci-6alkyl;
d) C2-6 alkenyl;
e) C2-6alkynyl;
f) alkoxy;
g) CF3;
h) CN;
i) amino; j) NR2R2;
k) O-alkyl;
1) O-alkenyl;
m) O-alkynyl;
n) C(R2)(R2)-S(O)nNH2;
o) C(R2)(R2)-S(O)nCF3;
P) C(R2)(R2)-NH2;
q) A-heterocycle;
r) C(R2)(R2)-NR2R2;
s) C(R2)(R2)-C(=W)R2;
t) aryl;
u) carbocycle;
v) heterocycle;
W) cycloalkyl;
x) alkaryl;
y) alkylheterocycle;
z) aralkyl; or
aa) heterocycle-alkyl;
any of which may be unsubstituted or substituted with one or more of the following, taken in any combination:
a) halo;
b) OH;
c) OR2;
d) SR2; e) COOH;
f) carboxlic acid ester;
g) C(=W)R2;
h) C(=W)0R2;
i) C(=W)0R3;
j) C(=W)SR2;
k) A-C(=W)NH2;
1) C(=W)NR2R3;
m) NR2R2;
n) NR2R3;
o) NR2-S(O)nR3; p) NR2-C(=W)-C1-6alkyl;
q) S(O)nR3;
r) C1-6alkoxy;
s) C1-6 thioether;
t) amino acid residue;
u) NH-A-(amino acid residue);
v) C(=W)NH-A-(amino acid residue); and
wherein when said optional substitution comprises a substituted heterocycle, then substitution is selected from the group consisting of:
a) C(=W)O-aryl;
b) C(=W)O-alkyl;
c) C(=W)NH2;
d) C(=W)NH-aryl; e) C(=W)NH-alkyl;
f) C(=W)N-di-alkyl;
g) C(=W)N(alkyl)-aryl;
h) α-amino acid;
i) α-amino ester;
j) α-amino-carboxamide;
k) β-amino acid;
1) β-amino ester; or
m) β-amino-carboxamide;
n) halo; or
o) cyano,
taken alone or in any combination;
n independently is 0, 1 or 2;
each A is independently a disubstituted spacer selected from the group consisting of:
a) C1-6 alkylene, branched or unbranched, and optionally having one or more heteroatoms, aryl, cycloalkyl or heterocyclyl functions in or attached to the chain;
b) C2-12 alkenylene, branched or unbranced, and optionally having one or more heteroatoms, aryl, cycloalkyl or heterocyclyl functions in or attached to the chain;
c) C2-12 alkynylene, branched or unbranced, and optionally having one or more heteroatoms, aryl, cycloalkyl or heterocyclyl functions in or attached to the chain;
d) optionally substituted arylene; e) O-alkylene, branched or unbranched, and optionally having one or more heteroatoms, aryl, cycloalkyl or heterocyclyl functions in or attached to the chain;
f) aralkylene, branched or unbranched, and optionally having one or more heteroatoms, aryl, cycloalkyl or heterocyclyl functions in or attached to the chain;
g) optionally substituted cycloalkyl; and
h) optionally substituted heterocycle;
wherein "A" may be joined by any desired linkage such as, for example, an ether, thioether, amino, carboxamido, ester or carbon-carbon linkage, or any combinatin thereof;
Each R4', R5', R6' and R7' independently is:
a) H;
b) halogen;
c) NO\2;
d) CN;
e) CF3;
f) OR2;
g) NR2R2;
h) NHS(O)nR2;
i) NHCO-C1-3 alkyl;
j) S(O)nR2;
k) aryl;
1) heterocycle;
m) C1-6 alkyl;
n) C2-6 alkenyl; o) C2-6 alkynyl; p) C(=W)-S(O)nR2;
q) C(=W)-S(O)π-NR2R2;
r) C(=W)-aryl;
s) C(=W)-alkyl; t) C(=W)-heterocycle; or
u) C(=W)-NR2R2; each of which optionally may be substituted with one or more of :
a) OR2;
b) S(O)nR2
c) C(=W)-S(O)nR^;
d) C(=W)-S(0)n-NR 2zτR>2.
e) C(=W)-aryl;
f) C(=W)-alkyl;
g) C(=W)-heterocycle;
h) C(=W)NR2R2;
i) H;
j) NO2;
k) CN;
1) CF3;
m) halogen;
n) NHS(O)nR2;
o) NHCO-C1-3 alkyl;
p) aryl; q) heterocycle;
r) C1-6 alkyl; s) C2-6 alkenyl;
t) C2-6 alkynyl; or
u) NR2R2.
In one embodiment of Formula (A), X is an optionally substituted phenyl; Y is any of its definitions; R1, R4 , R6 , and R7 are all hydrogen; R5 is halogen; and Z is a carboxamide moiety.
In an alternative embodiment, R1 is acyl, alkyl, aryl, alkaryl, or aralkyl.
Li yet another alternative embodiment, R4 is fluoro, nitro or cyano, W is oxygen, and Y is O-alkyl.
In a second embodiment of Formula (A), X is an optionally substituted phenyl; Y is any of its definitions; R1, R4 , R6 , and R7 are all hydrogen; R5 is chlorine; and Z is a carboxamide or carboxamido-heterocyclyl moiety.
In yet another embodiment of Formula (A), X is tolyl, thiazolyl or pyridyl; Y is H, OH, or O-alkyl; R1, R4', R6', and R7' are all hydrogen; R5' is halogen; and Z is carboxamido, acyl, an alkyl-sulphonyl group or a carboxylic acid derivative.
In an alternative to the preceding embodiment, X, Y, R1, R4', R6', R7' and R5
all are as defined above, and Z is a carboxamido-alkylene-heterocyclyl wherein the heterocycle typically is imidazole, furan, pyridine, pyrimidine, or thiazole that optionally may be substituted; a thioamido-pyridyl wherein the pyridyl is unsubstituted or substituted by OH, OMe or lower alkyl; an imino-nitrile, or an alkylsulphonyl-aryl group.
A first series of subembodiments of the present invention is given where the Formula (A) is as provided above, and W, X and Y are defined as: a) W is O, X is alkyl, and Y is -O-alkyl;
b) W is O, X is -O-aryl, and Y is alkyl;
c) W is O, X is -O-aryl, and Y is -NR2R3;
d) W is O, X is -O-alkyl, and Y is alkyl;
e) W is O, X is -O-alkyl, and Y is halo;
f) W is O, X is -O-heterocycle, and Y is alkyl;
g) W is O, X is aryl, and Y is -O-alkyl;
h) W is O, X is heterocyclyl, and Y is -O-aryl;
i) W is O, X is alkyl, and Y is -O-heterocyclyl;
j) W is -NR2R2, X is heterocyclyl, and Y is -O-aryl;
k) W is -NR2R2, X is alkyl, and Y is halo;
1) W is S, X is alkyl, and Y is -O-alkyl;
m) W is S, X is alkyl, and Y is -NR2R3;
n) W is S, X is -O-aryl, and Y is alkyl;
o) W is S, X is -O-aryl, and Y is C-halo;
p) W is S, X is -O-alkyl, and Y is alkyl;
q) W is S, X is -O-heterocycle, and Y is alkyl;
r) W is S, X is aryl, and Y is -O-alkyl;
s) W is S, X is heterocyclyl, and Y is -O-aryl;
t) W is S, X is alkyl, and Y is -O-heterocyclyl;
u) W is O5 X is aryl, and Y is -O-aryl;
v) W is -NR2, X is -O-alkyl, and Y is -NR2R3;
w) W is O, X is -O-aryl, and Y is -O-aryl;
x) W is O3 X is alkyl, and Y is alkyl; and y) W is -NR2, X is -O-alkyl, and Y is alkyl.
A second series of subembodiments of the present invention is given where Formula (A) is as given, R1 is H, alkyl, acyl, aryl, aralkyl, or alkaryl; and Z is defined as:
a) -C(=W)NR2R3, R2 is H, and R3 is NR2R2;
b) -CHV)NR2R3, R2 is H, and R3 is NR2R2, or R2 is C1-5 alkyl optionally substituted with OH and R3 is -NH2;
c) -C(=W)NR2R3, R2 is H, and R3 is (CH2)mC(=W)NR2R2;
d) -C(=W)NR2R3, R2 is H, and R3 is alkyl substituted by optionally substituted aryl or heterocycle;
e) -C(=W)R3, R3 is an amino acid residue or -NH(CH2)p-(amino acid residue);
f) -C(=W)NHNHC2H5OH;
g) -C(=W)NHCH2C(=W)NH2;
h) -C(=W)NHCH2CONHNH2;
i) -C(=W)NHCH2CH2-(2-NO2, 5-methyl imidazole);
j) -C(=W)NHCH2NHCH(CH3)C(=W)OH;
k) -C(=W)NHCH=CHC(=W)NH2;
1) -C(=W)NR2R5NR2R3, R5 is (=0), R2 and R3 are as defined above;
m) -C(=W)NR2NR2-C(=W)R3, R2 is H or alkyl, and R3 is aryl;
n) -C(=W)N(-NR2R3)-N(-NR2R3)R3, R2 is H, R3 is R2 or alkoxy;
o) -C(=W)NHR2C(=W)-Q, Q is heterocycle, and R2 is as defined above;
p) -CHV)NR2R3, R2 is as defined above, and R3 is -OH;
q) -COR2R3, R2 is amino and R3 is a heterocycle;
r) -CHV)NHNHC(=W)R2 and R2 is NH2;
s) -C(=W)-R2-CH- A-C(=W)NH2, and R2 is NH; t) -C(=W)-R2-CH-A-C(=W)H, and R2 is NH;
u) -C(=W)-R2-CH-A-C(=W)OH, and R2 is NH;
v) -C(=W)-R2-CH-A-R3, R2 is NH, and R3 is CH3;
w) -C(=W)NHR2C(=W)NH2, and R2 is optionally substituted, branched chain alkyl;
x) -C(=W)R2R3, R2 is NH or alkyl, and R3 is NH2;
y) -C(=W)R2-C(=W)OR3, and R2 and R3 are as defined as above;
z) -C(=W)R2-NH-C(=W)C1-4 alkoxy, and R2 is as defined above;
aa) -C(=W)R2C(=W)C1-4 alkoxy, and R2 is as defined above;
bb) -C(=W)R2, and R2 is NH2;
cc) -C(=W)R2-NH-C(=W)OR3, and R2 and R3 are as defined as above;
dd) -C(=W)R2-C(=W)R2, and R2 is as defined above;
ee) -C(=W)NHR2 where R2 is optionally substituted aryl, cycloalkyl or a heterocyclyl ring;
ff) -C(=W)R2-W-R3, where R2 and R3 are as defined as above;
gg) -C(=W)-NH-CH(R2)-C(=W)-NH2, and R2 is as defined above;
hh) -C(^(W)-NH-NH2; ii) -C(=W)-NH-NH(R2), and R2 is as defined above;
jj) -C(=W)-NH-CH(C[=W]NH2XCH2-C[=W]-O-aryl);
kk) -C(=W)-NH-CH(-[CH2]4-NH-C[=W]-t-Bu0)(-C[=W]-NH2);
11) -C(=W)-NH-CH(-CH2-CH2-C[=W]-t-Bu0)(C[=W]-NH2);
mm) -C(=W)-NH-CH(R3)(-C[=W]-NH2), and R3 is as defined above;
nn) -C(=W)-NH-CH(-CH2-R3X-C[=W]-NH2), and R3 is as defined above;
oo) -C(=W)-NH-CH(-CH2OH)(-C[=W]-NH2); pp) -C(=W)-NH-CH(C[=W]-NH2)(C[=W]-NH2);
qq) -C(=W)-NHR2-C[=W]NH2, and R2 is as defined above;
ir) -C(=W)-NH-CH(-[CH2]4-NH-C[=W]-O-CH2-R3)(-C[=W]-MΪ2), and R3 is as defined above;
ss) -C(=W)-NH-CH(-CH2-C[=W]-NH2)(-C[=W]-NH2);
tt) -C(=W)-NH-CH(-CH2-R3)(-C[=W]-NH2), where R3 is as defined above;
uu) -C(=W)-NH-CH(-[CH2]4-NH2)(-C[=W]-NH2);
w) -C(=W)-NH-CH(-CH[R2][OH])(-C[=W]-NH2), and R2 is as defined above;
ww) ~C(=W)-NH-CH(-R2)(-C[=W]-NH2), and R2 is as defined above;
xx) -C(-W)-NH-CH(-R2-C[=W]-1S1H2)(-C[=W]-NH2), and R2 is asdefined above;
yy) -C(=W)-NH-CH(-R2-SCH3)(-C[=W]-NH2), and R2 is as defined bove;
zz) -C(=W)-NH-CH(-C[=N]-NH2)(-C[=W]-NH2);
aaa) -C(-W)-MΪ-CH(-R3)(-C[=W]-NH2), and R3 is as defined above;
bbb) -C(=W)-NH-CH(-CH2-R3)(-C[=W]-NH2), and R3 is as defined above;
ccc) -C(=W)-NH-CH(-R2)(-C[=W]-NH2), and R2 is as defined above;
ddd) -C(=W)-NH-R2-C[=W]R3, and R2 is alkylene and R3 is aryl or heteroaryl;
eee) -C(=W)-NH-R2-R3-C[=W]-NH2, where R2 is alkylene and R3 is ryl or heteroaryl;
fff) -C(=W)-NH-NH-R2-R3-C(=W)NH2, where R2 is alkylene and R3 is aryl or heteroaryl;
ggg) -C(=W)-NH-NH-CH(R3)-C(=W)R2, and R2 is NH2 and R3 is ptionally substituted aryl or heteroaryl;
hhh) -C(=W)MΪR2(-R3)-C(=W)NH2, where R2 is alkylene and R3 is optionally substituted alkyl, aryl or heteroaryl;
iii) -C(=W)NHR2(-R3)-C(=W)NH-R2-C(=W)OH, where R2 isalkylene and R3 is optionally substituted alkyl, aryl or heteroaryl; jjj) -C(=W)NHR2(-R3)-C(=W)NH-R2-NH2, where R2 is alkylene and R3 is optionally substituted alkyl, aryl or heteroaryl;
kkk) -C(=W)NHR2(-R3)-C(=W)NH-R3, where R2 is alkylene and R3 is optionally substituted alkyl, aryl or heteroaryl;
111)
Figure imgf000057_0001
where R2 is -NH, p is 0-10, A is a divalent linker or an optionally substituted aryl or heteroaryl, and W is O or S;
rnmm)-C(=W)NH-R3, where R3 is an optionally substituted heterocycle;
nnn) -C(=W)-NH-R2-R5-R3, where W is O or S, R2 is alkylene, alkenylene or alkynylene, R5 is -SO2, and R3 is -NH2;
ooo) -C(=W)-NB-NH-R2(R3)-R5-NH2, where W is O or S, R2 is alkylene, alkenylene or alkynylene, R3 is aryl, arylene, or heteroaryl, and R5 is -SO2;
ppp) -C(=W)-NH-R3(R5-NH2) where W is O or S, R3 is aryl, arylene or heteroaryl, and R5 is SO2;
qqq) -C(=W)-NH-R2-R3(R5-NB2) where W is O or S, R2 is alkylene, alkenylene or alkynylene, R is aryl, arylene or heteroaryl, and R is SO2;
rrr) -C(=W)-NH-R3(R2R5-NH2) where W is O or S, R2 is alkylene, alkenylene or alkynylene, R3 is aryl, arylene or heteroaryl, and R5 is SO2;
sss) -C(=W)-MΪR2(-R3)-C(=W)NH-R2-C(=W)OH, where R2 is an optionally substituted alkylene, alkenylene, or alkynylene, and R3 is any of its definitions as provided above;
ttt) -C(=W)-NHR2(-R3)-C(=W)Mi-R2-C(=W)NH2, where R2 is an ptionally substituted alkylene, alkenylene, or alkynylene, and R3 is any of its definitions as provided above;
uuu) -C(=W)-NHR2-C(=W)NH-R2-C(=W)OH, where R2 is an optionally substituted alkylene, alkenylene, or alkynylene, and R3 is any of its definitions as provided above; or vw) -C(=W)-NHR2-C(=W)NH-R2-C(=W)NH2, where R2 is an optionally substituted alkylene, alkenylene, or alkynylene, and R3 is any of its definitions as provided above; and
wherein R2, R3 and A each is as defined previously.
A third series of subembodiments of the present invention is given for the Formula (A) as provided above where R4', R5', R6' and R7' are defined as:
a) R6' and R7' are both hydrogen, and R4' and R5 independently are halo; -NO2; -N; -OR2, -NR2R2; -NH-R5-C1-3 alkyl; -NHCO-C1-3 alkyl; oxime; hydrazine; -H(SO2)C1-3alkyl; -NH-O-C1-3 alkyl; -NHOH; or C1-3 alkyl or alkenyl optionally substituted with one or more -OH, -SR, -C(=W)H, -C(=W)0H, halogen, -NR2R2, C1-3 alkoxy, or C1-3 thioether;
b) R4 and R7 are both hydrogen, and R5 and R6 independently are halo; -NO2; -N; -OR2; -NR2R2; -NHSO2-C1-6 alkyl; -NHCO-C1-6 alkyl; oxime; hydrazine; -H-O-C1-6 alkyl; -NH-OH; or C1-3 alkyl or alkenyl optionally substituted with one or more -OH, -SR, -C(=W)H, -(=W)0H, halogen, -NR2R2, C1-3 alkoxy, or C1-3 thioether;
c) R4 and R6 are both hydrogen, and R5 and R7 independently are halo; -NO2; -N; -OR2; -NR2R2; -NHSO2-C1-6 alkyl; -NHCO-C1-6 alkyl; oxime; hydrazine; -H-O-C1-6 alkyl; -NH-OH; or C1-6 alkyl or alkenyl optionally substituted with one or more -OH, -SR, -C(=W)H, -(=W)0H, halogen, -NR2R2, C1-3 alkoxy, or C1-3 thioether;
d) R4' and R7 are both hydrogen, and R5 and R6 independently are halo; -NO2; -N; -OR2; -NR2R2; -NH-O-C1-6 alkyl; -NHOH; or C1-6 alkyl or alkenyl optionally substituted with one or more -OH, -SR, -C(=W)H, -C(=W)0H, halogen or -NH2;
e) R4 and R6 are both hydrogen, and R5 and R7 independently are halo; -NO2; -N; -OR2; -NR2R2; -NH-O-C1-3 alkyl; -NHOH; or C1-6 alkyl or alkenyl optionally substituted with one or more -OH, -SR, -C(=W)H, -C(=W)0H, halogen or -NH2 ; f) R6> and R7' are both hydrogen, and R4' and R5' independently are halo; -C1-6 alkyl, alkenyl or alkynyl optionally substituted with one or more halo;
g) R4> and R7' are both hydrogen, and R5' and R6' independently are halo; -C1-6 alkyl, alkenyl or alkynyl optionally substituted with one or more halo;
h) R4' and R6' are both hydrogen, and R5' and R7' independently are halo, -C1-6 alkyl, alkenyl or alkynyl optionally substituted with one or more halo;
i) R6' and R7> are both hydrogen, and R4' and R5' independently are Cl, F, Br, I, methyl, ethyl or CF3;
j) R4' and R7 are both hydrogen, and R5 and R6 independently are Cl, F, Br, I, methyl, ethyl or CF3;
k) R4' and R6' are both hydrogen, and R5' and R7' independently are Cl, F, Br, I, methyl, ethyl or CF3; and
wherein A is as defined above.
A fourth series of subembodiments for Formula (A) is defined where W, X and Y are as defined in the first series of subembodiments, and Z is as defined in the second series of subembodiments.
A fifth series of subembodimentts for Formula (A) is defined where W, X and Y are as defined in the first series of subembodiments, and R4 , R5 , R6' and R7' are as defined in the third series of subembodiments
A sixth series of subembodimentts for Formula (A) is defined where Z is as defined in the second series of subembodiments, and R , R5 , R6 and R7 are as defined in the third series of subembodiments.
Non-limiting species of the first embodiment as given by Formula (A) above are defined when:
1) Z is -C(=W)NHNHC2H5OH, R4', R6> and R7' are H, R5' is Cl, X is H, and Y is H;
2) Z is -C(=W)NHCH2C(=W)NH2, R4> is F or Cl, R6> and R7' are H, R5' is F or Cl, X is H, and Y is OH; 3) Z is -C(=W)NHCH2CONHNH2, R4' is F or Cl, R6' and R7' are H, R5' is F or Cl, X is H, and Y is -O-alkyl;
4) Z is -C(=W)NHCH2CH2-(2-NO2, 5-Me-imidazole), R4' is F or Cl, R6' and R7' are H, R5' is F or Cl, X is H, and Y is -O-alkenyl;
5) Z is -C(=W)NHCH2NHCH(CH3)C(=W)OH, R4' is F or Cl, R6' and R7' are H, R5' is F or Cl, X is H, and Y is -O-alkynyl;
6) Z is -C(=W)CH=CHC(=W)NH2, R4' is F or Cl, R6' and R7' are H, R5' is F or Cl, X is H, and Y is -O-alkaryl;
7) Z is -C(=W)NHNHCH2C(=W)NH2, R4' is F or Cl, R6' and R7' are H, R5' is F or Cl, X is H, and Y is -O-aryl;
8) Z is -C(=W)NHCH2C(=W)R2, where R2 is NH2, R4' is F or Cl, R6' and R7' are H, R5' is F or Cl, X is H, and Y is -O-heterocycle;
9) Z is -C(=W)NHCH2-A-C(=W)NH2, where A is a divalent spacer, R4' is F or Cl, R6' and R7' are H, R5' is F or Cl, X is H, and Y is C1-6 alkyl;
10) Z is -C(=W)R2CH-A-C(=W)H, where A is a divalent spacer, R2 is NH, R4> is F or Cl, R6' and R7' are H, R5' is F or Cl, X is H, and Y is C2-6 alkynyl;
11) Z is -C(=W)R2CH-A-C(=W)OH, where A is a divalent spacer, R2 is NH, R4' is F or Cl, R6' and R7' are H, R5' is F or Cl, X is H, and Y is C2-6 alkynyl;
12) Z is -C(=W)R2-CH-A-R3, where A is a divalent spacer, R2 is NH, R3 is CH3, R4' is F or Cl, R6> and R7' are H, R5' is F or Cl, X is H, and Y is aryl;
13) Z is -C(=W)NHR2-C(=W)R2, where R2 is an optionally substituted, branched chain alkylene or NH, R4' is F or Cl, R6' and R7' are H, R5' is F or Cl, X is H, and Y is heterocycle;
14) Z is -C(=W)R2, where R2 is NH, R4' is F or Cl, R6' and R7' are H, R5' is F or Cl, X is OH, and Y is H;
15) Z is -C(=W)R2R3-heterocycle, where R2 is NH, R3 is CH2, heterocycle is optionally substituted morpholine, imidazole or pyrrole, R4 is F or Cl, R6 and R7 are H, R5' is F or Cl, X is C1-6 alkyl, and Y is H; 16) Z is -C(=W)R2C(=W)-O-R3, where R2 is optionally substituted alkyl, R3 is benzyloxy, R4' is F or Cl, R6' and R7' are H, R5' is F or Cl, X is -O-alkyl, and Y is H;
17) Z is -C(=W)R2-NH-C(==W)-C1-4 alkoxy, where R2 is an optionally substituted alkyl, R4> is F or Cl, R6' and R7' are H, R5' is F or Cl, X is -O-alkenyl, and Y is H;
18) Z is -C(=W)R3-C(=W)R2, where R2 is C1-4 alkoxy, R3 is optionally substituted alkyl, R4' is F or Cl5 R6' and R7> are H, R5' is F or Cl, X is -O-alkynyl, and Y is H;
19) Z is -C(=W)R2R3, where R2 is optionally substituted alkyl, R3 is optionally substituted phenyl, R4' is F or Cl, R6' and R7' are H, R5' is F or Cl, X is -O-alkaryl, and Y is H;
20) Z is -C(=W)R2-NH-C(=W)-O-R3, where R2 is optionally substituted alkyl, R3 is benzyl, R4' is F or Cl, R6' and R7' are H, R5' is F or Cl, X is -O-aralkyl, and Y is H;
21) Z is -C(=W)R2C(=W)-NH2, where R2 is optionally substituted alkyl, R4' is F or Cl, R6> and R7' are H, R5> is F or Cl, X is -O-aryl, and Y is H;
22) Z is -C(=W)R2-W-R3, where R2 and R3 are optionally substituted alkyl, R4' is F or Cl, R6> and R7' are H3 R5' is F or Cl, X is -O-heterocycle, and Y is H;
23) Z is -C(=W)R2C(=W)-O-R3, where R2 is optionally substituted alkyl, R3 is benzyloxy, R4' is F or Cl, R6' and R7' are H, R5' is F or Cl, X is -C2-6-alkenyl, and Y is H;
24) Z is -C(=W)-NH-CH(R2)-C(=W)-NH2, where R2 is NH2, R4' is F or Cl, R6' and R7' are H, R5' is F or Cl, X is -C2-6-alkynyl, and Y is H;
25) Z is -C(=W)-NH-NH2, where R4' is F or Cl, R6' and R7' are H, R5' is F or Cl5 X is aryl, and Y is H;
26) Z is -C(=W)-NH-NH(R2), where R2 is NH2, R4' is F or Cl, R6' and R7' are H, R5' is F or Cl, X is heterocycle, and Y is H;
27) Z is -C(=W)-NH-CH(C[-W]NH2)(CH2-C[=W]-O-CH2-aryl), where R4' and R5'
independently are F or Cl, R6 and R7 are H, X is carbocycle, and Y is H;
28) Z is -C(=W)-NH-CH(-[CH2]4-NH-C[=W]-t-Bu0)(-C[=W]-NH2)5 where R4> and R independently are F or Cl, R and R are H5 X is -O-alkaryl, and Y is C1-6 alkyl; 29) Z is -C(=W)-NH-CH(-CH2-CH2-C[=W]-t-Bu0)(-C[=W]-NH2), where R4' and R5' independently are F or Cl, R6' and R7' are H, X is -O-aiyl, and Y is -O-alkyl;
30) Z is -C(=W)-NH-CH(-CH2R3)(-C[=W]-NH2), where R3 is CF3, R4' and R5'
independently are F or Cl, R6 and R7> are H, X is -C2-6 alkenyl, and Y is -OH;
31) Z is -C(=W)-NH-CH(-CH2R3)(-C[=W]-NH2), whereR3 is as defined in the first embodiment, R4 and R5 independently are F or Cl, R6' and R7 are H, X is -C2-6 alkynyl, and Y is H;
32) Z is -C(=W)-NH-CH(-CH2OH)(-C[=W]-NH2), where R4' and R5' independently are F or Cl, R6' and R7' are H, X is -O-alkylaryl, and Y is -O-alkyl;
33) Z is -C(=W)-NH-CH(-C[=W]-NH2)(-C[=W]-NH2), where R4' and R5' independently are F or Cl, R6 and R7 are H, X is -heterocycle, and Y is -OH;
34) Z is -C(=W)-NH-CH(-R3)(-C[=W]-NH2), where R4' and R5' independently are F or Cl, R6' and R7' are H, X is -O-alkenyl, and Y is -O-alkyl;
35) Z is -C(=W)-NH-CH(-[CH2]4-NH-C[=W]-O-CH2-R3)(-C[=W]-MI2), where R3 is as defined in the first embodiment, R4 and R5 independently are F or Cl, R6 and R7 are
H, X is -OH, and Y is -O-alkylene;
36) Z is -C(=W)-MΪ-CH(-CH2-C[=W]-NH2)(-C[=W]-NH2), where R4' and R5' independently are F or Cl, R6 and R7 are H, X is -carbocycle, and Y is -O-alkyl;
37) Z is -C(=W)-NH-CH(-CH2-R3)(-C[=W]-NH2), where R3 is as defined in the first embodiment, R4 and R5 independently are F or Cl, R6 and R7 are H5 X is -O-alkyl, and
Y is -heterocycle;
38) Z is -C(=W)-NH-CH(-[CH2]4-NH2)(-C[=W]-NH2), where R4' and R5' independently are F or Cl, R6 and R7 are H, X is -O-alkenyl, and Y is -OH;
39) Z is -C(=W)-NH-CH(-CH[R2] [OH]X-Ct=W]-NH2), where R4' and R5' independently are F or Cl, R6' and R7' are H, X is -O-alkynyl, and Y is -O-alkyl;
40) Z is -C(=W)-NH-CH(R2)(-C[=W]-NH2), where R4' and R5> independently are F or Cl, R6' and R7' are H, X is -OH, and Y is -O-alkylene; 41) Z is -C(=W)-NH-CH(-R2-C[=W]-NH2)(-C[=W]-NH2), where R2 is NH2, R4> and R5' independently are F or Cl, R6' and R7' are H, X is -C2-6 alkylene, and Y is -OH;
42) Z is -C(=W)-NH-CH(C[=NH]-NH2)(-C[=W]-NH2), where R4' and R5> independently are F or Cl, R6 and R7 are H, X is -carbocycle, and Y is -O-alkyl;
43) Z is -C(=W)-NH-CH(-C[=NH]-NH2)(-C[=W]-NH2), where R4' and R5' independently are F or Cl, R6 and R7 are H, X is -OH, and Y is -O-alkylene;
44) Z is -C(=W)-NH-CH(-R3)(-C[=W]-NH2), where R3 is as defined in the first g geenneerraall e emmbbooddiimmeenntt,, R R4 ; and R5 independently are F or Cl, R6 and R7 are H, X is -O- aryl, and Y is -C1-6 alkyl;
45) Z is -C(=W)-NH-CH(-CH2-R3)(-C[=W]-NH2), where R3 is as defined in the first general embodiment, R4 and R5 independently are F or Cl, R6 and R7 are H, X is -OH, and Y is -O-alkylene;
46) Z is -C(=W)-NH-CH(-R2)(-C[=W]-NH2), where R2 is NH2, R4> and R5' independently are F or Cl, R6 and R7 are H, X is -C1-6 alkynyl, and Y is -O-aryl;
47) Z is -C(=W)-NH-NH-CH(R3)-C[=W]R2, where R2 is -NH2, R3 is optionally substituted aryl or heterocycle, R4' and R5 independently are F or Cl, R6 and R7 are H, X is -O-alkylaryl, and Y is -OH;
48) Z is -C(=W)-NHR2(-R3)-C[=W]NH2, where R2 is -alkylene, R3 is optionally substituted alkyl, aryl or heterocycle, R4 and R5 independently are F or Cl, R6 and R7 are H, X is -O-alkynyl, and Y is -OH;
49) Z is -C(=W)NHR2(-R3)-C(=W)NH-R2-C(=W)OH, where R2 is alkylene, R3 is optionally substituted alkyl, aryl or heterocycle, R4 and R5 independently are F or Cl, R6' and R7> are H, X is -O-alkenyl, and Y is -O-alkyl;
50) Z is -C(=W)NHR2(-R3)-C(=W)NH-R2-NH2, where R2 is alkylene, R3 is optionally substituted alkyl, aryl or heterocycle, R4' and R5 independently are F or Cl,
R6' and R7' are H, X is H, and Y is -O-alkyl;
51) Z is -C(=W)NHR2(-R3)-C(=W)NH-R3-C(=W)OH, where R2 is alkylene, R3 is optionally substituted alkyl, aryl or heterocycle, R4 and R5 independently are F or Cl, R6' and R7' are H, X is -OH, and Y is -O-alkylene; 52) Z is -C(=W)R2(CH2)P-A-C(=W)NH2, where R2 is -NH, p is 0-10, A is divalent, optionally substituted aryl or heterocycle, R3 is optionally substituted alkyl, aryl or heterocycle, R4 and R5' independently are F or Cl, R6' and R7 are H, X is -C1-6 alkyl, and Y is -O-alkenyl;
53) Z is -C(=W)NH-R3, where R3 is optionally substituted heterocycle, R4' and R5' independently are F or Cl, R6' and R7' are H, X is -O-alkyl, and Y is -OH;
54) Z is -C(=W)NH-R2-R5-R3, where R2 is alkylene, alkenylene or alkynylene, R5 is -SO2, R3 is -NH2, R4' and R5' independently are F or Cl, R6' and R7' are H, X is -O-aryl, and Y is -O-alkyl;
55) Z is -C(=W)NH-NH-R2(R3)-R5-NH2, where R2 is alkylene, alkenylene or alkynylene, R3 is aryl, arylene or heterocyclyl, R5 is -SO2, R4 and R5 independently are F or Cl, R6' and R7' are H, X is -O-alkynyl, and Y is -OH;
56) Z is -C(=W)NH-R3(R5-NH2), where R3 is aryl, arylene or heterocyclyl, R5 is - -SSOO22,, R R44' a annd R5 independently are F or Cl, R6 and R7 are H, X is —heterocyclyl, and Y is -O-alkyl;
57) Z is -C(=W)NH-R2R3(R5-NH2), where R2 is alkylene, alkenylene or alkynylene, R3 is aryl, arylene or heterocyclyl, R5 is -SO2, R4 and R5 independently are F or Cl, R6 and R7 are H, X is -aryl, and Y is -O-alkenyl;
58) Z is -C(=W)NH-R3(R2R5-NH2), where R2 is alkylene, alkenylene or alkynylene, R3 is aryl, arylene or heterocyclyl, R5 is -SO2, R4' and R5' independently are F or Cl, R6' and R7' are H, X is -OH, and Y is -O-alkylaryl;
59) Z is -C(=W)NHR2(-R3)-C(=W)NH-R2-C(=W)OH, where R2 is optionally substituted alkylene, alkenylene or alkynylene, R3 is any of the definitions provided in the first general embodiment, R4 and R5 independently are F or Cl, R6 and R7 are H, X is -C2-6 alkynyl, and Y is -O-alkenyl;
60) Z is -C(-W)NHR2(-R3)-C(=W)NH-R2-C(=W)NH2, where R2 is optionally substituted alkylene, alkenylene or alkynylene, R3 is any of the definitions provided in the first general embodiment, R4 and R5' independently are F or Cl, R6 and R7 are H, X is -C2-6 alkenyl, and Y is -H; 61) Z is -C(=W)NHR2-C(=W)NH-R2-C(=W)OH, where R2 is optionally substituted alkylene, alkenylene or alkynylene, R4> and R5' independently are F or Cl, R6' and R7 are H, X is -C1-6 alkyl, and Y is -H;
62) Z is -C(=W)NHR2-C(=W)NH-R2-C(=W)NH2, where R2 is optionally substituted alkylene, alkenylene or alkynylene, R4' and R5 independently are F or Cl, R6 and R7 are
H, X is -H, and Y is -O-alkyl;
63) Z is -C(=W)NH-R3 where W is O, and R3 is a heterocycle, and in particular pyridyl, pyrimidyl or imidazo that is optionally substituted by one or more halo, cyano, alkyl, alkenyl, alkynyl, or cyanoalkyl groups; R4 and R5 independently are H, F or Cl, R6' and R7' are H, X is optionally substituted phenyl, and Y is -O-alkyl; and
wherein W is as defined for Formula (A).
In a second embodiment, the invention provides a phenylindole for use in the treatment of HIV represented by the following general Formula (B):
Figure imgf000065_0001
(B) or a pharmaceutically acceptable salt, prodrug, N — oxide, quaternary amine, stereochemical isomer or tautomer thereof, wherein:
each W, Y, Z, R1, R2, R3, A, n, R4', R5>, R6' and R7' is as defined above for Formula (A); and
each R2", R3", R4", R5", and R6" independently is:
a) H; b) halogen; c) NO2;
d) CN;
e) OR2;
f) SR2;
g) NH2;
h) NR2R3;
i) N(R2)-C(=W)-C1-4 alkyl;
j) N(R2)-SO2-C1-4 alkyl;
k) C1-6 alkyl;
1) C2-6 alkenyl;
m) C2-6 alkynyl;
n) aryl;
o) CF3;
p) CR2R2-S(O)n-R3;
q) CR2R2NR2R3;
r) C-OH;
s) CR2R2-C(=W)R2;
t) acyl;
u) C(=W)R2;
v) C(=W)0R2;
w) C(=W)SR2;
x) CO=W)-NR2R3;
y) C(=W)NH(CH2)p-(amino acid residue);
z) amino residue; or aa) A-(amino acid residue);
bb) cyanoalkyl;
cc) cyanoalkenyl; or
dd) cyanoalkynyl,
wherein any of the above optionally may be substituted; or
alternatively, R2 or R6 may be joined to Y to form an optionally substituted bicyclic or tricyclic phosphorylated heterocycle comprising 4 -14 members.
A first series of subembodiments of the Formula (B) include all the subembodiments provided for Formula (A) above.
A second series of subembodiments of the present invention is given for the embodiment of the Formula (B) as provided above where R2 , R3 , R4 , R5 , and R6 are defined as:
a) R2 , R4 , and R6 are hydrogen, and R3 and R5 each independently is halogen; - NO2; -CN; -OR2; -NH-R5-C1-6 alkyl; -NHCO-C1-6 alkyl; oxime; hydrazine; -
N(OH)C1-6 alkyl; C1-6 alkoxy; -OH; -NR2R2; or -C1-6 alkyl, alkenyl or alkynyl optionally substituted with one or more of -OH, -SR, -CN, -halo, -C(=W)H, - C(=W)0H, halogen, NR2R2, -C1-6 thioether, or -C1-6alkoxy;
b) R2 , R4 , and R6 are hydrogen, and R3 and R5 each independently is halogen, or -C1-6 alkyl, alkenyl, alkynyl optionally substituted with one or more halogen;
c) R2", R3", R4", R5"and R6" are hydrogen;
d) R2 , R4 , and R6 are hydrogen, and R3 and R5 are methyl;
e) R2 , R4 , and R6 are hydrogen, and R3 and R5 are chloro;
f) R2 , R4 , and R6 are hydrogen, and R3 and R5 are fluoro;
g) R2", R4", and R6" are hydrogen, R3" is iodo and R5" is bromo;
h) R2 , R4 , and R6 are hydrogen, R3 is methyl, and R5 is chloro; and i) R2", R4", and R6" are hydrogen, R3" is chloro, and R5" is methyl.
A third series of subembodiments of the present invention is given for the embodiment of the Formula (B) as provided above where W, Y, R2 , R3 , R4 , R5 , and R6" are defined as:
a) W is O, Y is -O-alkyl, R2", R4", and R6" are hydrogen, and R3" and R5" each independently is halogen; -NO2; -CN; -OR2; -NH-R5-C1-6 alkyl; -NHCO-C1-6 alkyl; oxime; hydrazine; -N(OH)C1-6 alkyl; C1-6 alkoxy; -OH; -NR2R2; Or-C1-6 alkyl, alkenyl or alkynyl optionally substituted with one or more of -OH, -SR, -C(=W)H, -C(=W)0H, halogen, NR2R2, -C1-6 thioether, or -C1-6alkoxy;
b) W is S, Y is OH, R2", R4", and R6" are hydrogen, and R3" and R5" each independently is halogen, or -C1-6 alkyl, alkenyl, alkynyl optionally substituted with one or more halogen;
c) W is O, Y is C1-6 alkyl, R2", R3", R4", R5"and R6" are hydrogen;
d) W is S, Y is C1-6 alkylene, R2", R4", and R6" are hydrogen, and R3" and R5" are methyl;
e) W is NH, Y is -O-alkyl, R2", R4", and R6" are hydrogen, and R3" and
f) R5 are chloro;
g) W is S, Y is -O-alkenyl, R2", R4", and R6" are hydrogen, and R3" and
h) R5" are fluoro;
i) W is O, Y is aryl, R2", R4", and R6" are hydrogen, and R3" and R5" are fluoro;
j) W is NH, Y is -O-alkynyl, R2", R4", and R6" are hydrogen, and R3" and R5" are fluoro;
k) W is S, Y is S, R2", R4", and R6" are hydrogen, and R3" and R5" are fluoro;
1) W is O, Y is -O-alkyl, R2", R4", and R6" are hydrogen, R3" is iodo and R5" is bromo;
m) W is O, Y is -O-alkaryl, R2", R3" , R5" and R6" are hydrogen, and R4" is -OH; n) W is S3 Y is -NR2R3, R2", R3", R5"and R6" are hydrogen, and R4" is -NH2;
o) W is S, Y is -SR2, R2", R3", R5"and R6" are hydrogen, and R4" is -NO2; and
p) W is O, Y is -O-aralkyl, 2", R4", and R6" are hydrogen, R3" is chloro, and R5" is methyl.
A fourth series of subembodiments are defined when Z is as defined in any one of the second series of subembodiments for Formula (A), R4 , R5 , R6 and R7 are as defined in the third series of subembodiments for Formula (A), and R2 , R3 , R4 , R5 and R6 all are defined as in the second series of subembodiments for Formula (B).
A fifth series of subembodiments are defined where W and Y are as defined in the first series of subembodiments for Formula (A), Z is as defined in any one of the second series of subembodiments for Formula (A), R4 , R5 , R6 and R7 are as defined in the third series of subembodiments for Formula (A), and R2 , R3 , R4 , R5 and R6 all are defined as in the third series of subembodiments for Formula (B).
A sixth series of subembodiments are defined when Z is as defined in any one of the second series of subembodiments for Formula (A), R4', R5 , R6 and R7' are as defined in the third series of subembodiments for Formula (A), and W, Y, R2 , R3 , R4 , R5 and R6 all are defined as in the third series of subembodiments for Formula (B).
Non-limiting species of the second embodiment as given by Formula (B) above are defined when:
A) Z is -C(=W)NHNHC2H5OH, R4>, R6> and R7' are H, R5' is Cl3 Y is H, R2", R4" and R6" are H, and R3" and R5" are methyl;
B) Z is -C(=W)NHCH2C(=W)NH2, R4> is F or Cl3 R6' and R7' are H3 R5> is F or Cl3
Y is OH3 R2", R4" and R6" are H3 and R3" and R5" are methyl;
C) Z is -C(=W)NHCH2CONHNH2, R4' is F or Cl3 R6' and R7' are H3 R5' is F or Cl3
Y is -O-alkyl3 R2", R4" and R6" are H3 and R3" and R5" are methyl; D) Z is -C(=W)NHCH2CH2-(2-NO2, 5-Me-imidazole), R4' is F or Cl, R6' and R7' are H, R5' is F or Cl, Y is -O-alkenyl, R2", R4" and R6" are H, and R3" and R5" are methyl;
E) Z is -C(=W)NHCH2NHC(==W)OH5 R4' is F or Cl, R6' and R7' are H, R5' is F or Cl, Y is -O-alkynyl, R2", R4" and R6" are H, and R3" and R5" are methyl;
F) Z is -C(=W)CH=CHC(=W)NH2, R4' is F or Cl, R6' and R7' are H, R5' is F or Cl5
Y is -O-alkaryl, R2", R4" and R6" are H, and R3" and R5" are methyl;
G) Z is -C(==W)NHNHCH2C(=W)NH2, R4' is F or Cl, R6' and R7' are H, R5' is F or Cl, Y is -O-aiyl, R2", R4" and R6" are H, and R3" and R5" are methyl;
H) Z is -C(=W)NHCH2C(=W)R2, where R2 is NH2, R4> is F or Cl, R6' and R7' are H5 R5' is F or Cl, Y is -O-heterocycle, R2", R4" and R6" are H, and R3" and R5" are methyl;
I) Z is -C(=W)NHCH2-A-C(=W)NH2, where A is a divalent spacer, R4' is F or Cl, R6' and R7' are H5 R5' is F or Cl, Y is C1-6 alkyl, R2", R3", R5" and R6" are H5 and R4" is -NH2;
J) Z is -C(=W)R2CH-A-C(=W)H5 where A is a divalent spacer, R2 is NH5 R4' is F or Cl5 R6' and R7> are H5 R5' is F or Cl, Y is C2-6 alkynyl, R2" 5 R4" and R6" are H5 and R3" and R5 are methyl;
K) Z is -C(=W)R2CH-A-C(=W)OH5 where A is a divalent spacer, R2 is NH, R4' is F or Cl5 R6' and R7' are H5 R5' is F or Cl, Y is C2-6 alkynyl, R2", R3", R5" and R6" are H, and R4" is -NO2;
L) Z is -C(=W)R2-CH-A-R3, where A is a divalent spacer, R2 is NH, R3 is CH3,
R4' is F or Cl, R6' and R7> are H, R5' is F or Cl5 Y is aryl, R2", R4" and R6" are H, and R3" and R5 are methyl;
M) Z is -C(=W)NHR2-C(=W)R2, where R2 is an optionally substituted, branched chain alkylene or NH5 R4> is F or Cl, R6> and R7' are H, R5' is F or Cl5 Y is heterocycle, R2", R4" and R6" are H, and R3" and R5" are methyl;
N) Z is -C(=W)R2, where R2 is NH, R4> is F or Cl, R6' and R7' are H, R5> is F or Cl,
Y is H, R2", R3", R5" and R6" are H5 and R4" is -OH; O) Z is -C(=W)R2R3-heterocycle, where R2 is NH, R3 is CH2, heterocycle is optionally substituted morpholine, imidazole or pyrrole, R4 is F or Cl, R6 and R7 are H, R5> is F or Cl, Y is H, R2", R4" and R6" are H, and R3" and R5" are methyl;
P) Z is -C(=W)R2NH-C(=W)-O-R3, where R2 is optionally substituted alkyl, R3 is benzyloxy, R4' is F or Cl, R6' and R7' are H, R5> is F or Cl, Y is H, R2", R4" and R6" are H, and R3" and R5" are methyl;
Q) Z is -C(=W)R2-NH-C(=W)-C1-4 alkoxy, where R2 is an optionally substituted alkyl, R4> is F or Cl, R6' and R7' are H, R5' is F or Cl, Y is H, R2", R4" and R6" are H, and R3" and R5" are methyl;
R) Z is -C(=W)R3-C(=W)R2, where R2 is C1-4 alkoxy, R3 is optionally substituted alkyl, R4' is F or Cl, R6' and R7' are H, R5' is F or Cl, Y is H, R2", R4" and R6" are H, and R3 and R5 are methyl;
S) Z is -C(=W)R2R3, where R2 is optionally substituted alkyl, R3 is optionally substituted phenyl, R4' is F or Cl, R6' and R7' are H, R5' is F or Cl, Y is H, R2", R4" and R6" are H, and R3" and R5" are methyl;
T) Z is -C(=W)R2-NH-C(=W)-O-R3, where R2 is optionally substituted alkyl, R3 is benzyl, R4' is F or Cl, R6' and R7' are H, R5' is F or Cl, Y is H, R2", R4" and R6" are H, and R3 and R5 are methyl;
U) Z is -C(=W)R2C(=W)-NH2, where R2 is optionally substituted alkyl, R4' is F or Cl, R6' and R7' are H, R5' is F or Cl, Y is H, R2", R4" and R6" are H, and R3" and R5" are methyl;
V) Z is -C(=W)-A-R3, where R3 is an optionally substituted alkyl, A is an alkylene li innkkeerr,, R R44 i iis F or Cl, R0 and R7' are H, R5' is F or Cl, Y is H, R2", R4" and R6" are H, and R3" and R5" are methyl;
X) Z is -C(=W)R2C(=W)-O-R3, where R2 is optionally substituted alkyl, R3 is benzyloxy, R4' is F or Cl, R6> and R7' are H, R5' is F or Cl, Y is H, R2", R4" and R6" are H, and R3 and R5 are methyl;
Y) Z is -C(=W)-NH-CH(R>C(=W)-NH2, where RΛ is NH2, W 4' is F or Cl, Rb and R7' are H, R5' is F or Cl, Y is H, R2", R4" and R6" are H, and R3" and R5" are methyl; Z) Z is -C(=W)-NH-NH2, where R4' is F or Cl, R6' and R7' are H, R5' is F or Cl, Y is H, R2", R4" and R6" are H, and R3" and R5" are methyl;
AA) Z is -C(=W)-NH-1S1H(R2), where R2 is NH2, R4' is F or Cl, R6' and R7' are H, R5' is F or Cl, Y is H, R2", R4" and R6" are H, and R3" and R5" are methyl;
BB) ,
Figure imgf000072_0001
w,Λh,e~r.eΛ τR>44' and R
i innddeeppeennddeennttllyy a arree F F o orr C Cll,, '. R6' and R7' are H, R2 is alkyl or aryl, Y is H, R2", R3", R5" and R6" are H, and R4" is-NH2;
CC) Z is -C(=W)-NH-CH(-[CH2]4-NH-C[=W]-t-Bu0)(-C[=W]-NH2), where R4' and R5' independently are F or Cl, R6' and R7' are H, Y is C1-6 alkyl, R2", R4" and R6" are H, and R3" and R5" are methyl;
DD) Z is -C(=W)-NH-CH(-CH2-CH2-C[=W]-t-Bu0)(-C[=W]-NH2), where R4' and R5' independently are F or Cl, R6' and R7' are H, Y is -O-alkyl, R2", R4" and R6" are H, andR3 and R5 are methyl;
EE) Z is -C(=W)-NH-CH(-CH2R3)(-C[=W]-NH2), where R3 is CF3, R4' and R5> independently are F or Cl, R6' and R7' are H, Y is -OH, R2", R4" and R6" are H, and R3" and R5 are methyl;
FF) Z is -C(=W)-NH-CH(-CH2R3)(-C[=W]-NH2), whereR3 is as defined in the first e emmbbooddiimmeenntt,, R R44' a anndd R R55> i innddeeppeennddeently are F or Cl, R6' and R7> are H, Y is H, R2", R3", R5" and R6" are H, and R4" is -NO2;
GG) Z is -C(=W)-NH-CH(-acyl)(-C[=W]-NH2), where R4' and R5' independently are
F or Cl, R6' and R7' are H, Y is -O-alkyl, R2", R4" and R6" are H, and R3" and R5" are methyl;
HH) Z is -C(=W)-NH-CH(-C[=W]-NH2)(-C[=W]-NH2), where R4> and R5' independently are F or Cl3 R6' and R7' are H, Y is -OH, R2", R3", R5" and R6" are H, and R4" is -OH;
II) Z is -C(=W)-NH-CH(-R3)(-C[=W]-NH2), where R4> and R5' independently are F or Cl, R6' and R7> are H, Y is -O-alkyl, R2", R4" and R6" are H, and R3" and R5" are methyl; JJ) Z is -C(=W)-NH-CH(-CH2-R3)(-C[=W]-NH2), where R3 is as defined in the first embodiment, R4 and R5 independently are F or Cl, R6 and R7 are H, Y is -heterocycle, R2", R4" and R6" are H, and R3" and R5" are methyl;
KK) Z is -C(=W)-NH-CH(-[CH2]4-NH2)(-C[=W]-NH2), where R4' and R5' independently are F or Cl, R6' and R7' are H, Y is -OH, R2", R4" and R6" are H, and R3" and R5 are methyl;
LL) Z is -C(=W)-NH-CH(-CR2R2)(-C[=W]-NH2), where R4' and R5' independently are F or Cl, R6' and R7' are H, Y is -O-alkyl, R2", R4" and R6" are H, and R3" and R5" are methyl;
MM) Z is -C(=W)-NH-CH(R2)(-C[=W]-NH2), where R4' and R5' independently are F or Cl, R6' and R7' are H, Y is -O-alkylene, R2", R4" and R6" are H, and R3" and R5" are methyl;
NN) Z is -C(=W)-NH-CH(-R3)(-C[=W]-NH2), where R3 is as defined in the first general embodiment, R4 and R5 independently are F or Cl, R6 and R7 are H, Y is -C1-6 alkyl, R2", R4" and R6" are H, and R3" and R5" are methyl;
00) Z is -C(=W)-NH-CH(-CH2-R3)(-C[=W]-NH2), where R3 is as defined in the first general embodiment, R4 and R5 independently are F or Cl, R6 and R7 are H, Y is -O- alkylene, R2", R4" and R6" are H, and R3" and R5" are methyl;
PP) Z is -C(=W)-NH-CH(-R2)(-C[=W]-NH2), where R2 is NH2, R4' and R5' independently are F or Cl, R6' and R7' are H, Y is -O-aryl, R2", R3", R5" and R6" are H, and R4" is -NH2;
UU) Z is -C(=W)-NH-NH-CH(R3)-C[=W]R2, where R2 is -NH2, R3 is optionally substituted aryl or heterocycle, R4' and R5 independently are F or Cl, R6 and R7 are H, Y is -OH, R2", R4" and R6" are H, and R3" and R5" are methyl;
W) Z is -C(=W)-NHR2(-R3)-C[=W]NH2, where R2 is -alkylene, R3 is optionally substituted alkyl, aryl or heterocycle, R4' and R5' independently are F or Cl, R6 and R7 are H, Y is -OH, R2", R4" and R6" are H, and R3" and R5" are methyl; WW) Z is -C(=W)NHR2(-R3)-C(=W)NH-R2-C(=W)OH, where R2 is alkylene, R3 is optionally substituted alkyl, aryl or heterocycle, R4' and R5 independently are F or Cl, R6' and R7' are H, Y is -O-alkyl, R2", R4" and R6" are H, and R3" and R5" are methyl;
XX) Z is -C(=W)NHR2(-R3)-C(=W)NH-R2-NH2, where R2 is alkylene, R3 is optionally substituted alkyl, aryl or heterocycle, R4' and R5 independently are F or Cl,
R6> and R7> are H, Y is -O-alkyl, R2", R4" and R6" are H, and R3" and R5" are methyl;
YY) Z is -C(=W)NHR2(-R3)-C(=W)NH-R3-C(=W)OH, where R2 is alkylene, R3 is optionally substituted alkyl, aryl or heterocycle, R4' and R5 independently are F or Cl, R6' and R7' are H, Y is -O-alkylene, R2", R4" and R6" are H, and R3" and R5" are methyl;
ZZ) Z is
Figure imgf000074_0001
where R2 is -NH, p is 0-10, A is divalent, optionally substituted aryl or heterocycle, R3 is optionally substituted alkyl, aryl or h heetteerrooccyyccllee,, R R44 a anndd R R55 i innddeeppeennddeennttllyy a arree I F or Cl, R6 and R7 are H, Y is -O-alkenyl, R2", R3", R5" and R6" are H, and R4" is -NO2;
AAA) Z is -C(=W)NH-R3, where R3 is optionally substituted heterocycle, R4' and R5> independently are F or Cl, R6' and R7' are H, Y is -OH, R2", R4" and R6" are H, and R3" and R5 are methyl;
BBB) Z is -C(=W)NH- A-S(O)n-R2, where A is alkylene, alkenylene or alkynylene, R2 is -NH2, R4' and R5' independently are F or Cl, R6' and R7' are H, Y is -O-alkyl, R2", R4" and R6" are H, and R3" and R5 are methyl;
CCC) Z is -C(=W)NH-NH-R2(R3)-A-C(=W)NH2, where R2 is alkyl, A is alkylene, alkenylene or alkynylene, R3 is aryl, arylene or heterocyclyl, R4 and R5 independently are F or Cl, R6' and R7' are H, Y is -OH, R2", R4" and R6" are H, and R3" and R5" are methyl;
DDD) Z is -CC=W)NH-A-[S(O)2-NH2], where A is alkylene, alkenylene or alkynylene, R4' and R5' independently are F or Cl, R6' and R7' are H, Y is -O-alkenyl, R2", R4" and R6" are H, and R3 and R5 are methyl;
EEE) Z is -C(=W)NH-A-C(=W)NH-R2-C(-W)OH, where R2 and R3 are any of the definitions provided in the first general embodiment, R4' and R5' independently are F or Cl, R6' and R7' are H, Y is -O-alkenyl, R2", R4" and R6" are H, and R3" and R5" are methyl; FFF) Z is -C(=W)NHR2(-R3)-C(=W)NH-R2-C(=W)NH2, where R2 is optionally substituted alkylene, alkenylene or alkynylene, R3 is any of the definitions provided in the first general embodiment, R4' and R5' independently are F or Cl, R6 and R7' are H, Y is -H, R2", R4" and R6" are H, and R3" and R5" are methyl;
GGG) Z is -C(=W)NHR2-C(=W)NH-R2-C(=W)OH, where R2 is optionally substituted alkylene, alkenylene or alkynylene, R4 and R5' independently are F or Cl, R6 and R7 are H, Y is -H, R2", R4" and R6" are H, and R3" and R5" are methyl;
HHH) Z is -C(=W)NHR2-C(=W)NH-R2-C(=W)NH2, where R2 is optionally substituted alkylene, alkenylene or alkynylene, R4' and R5' independently are F or Cl, R6 and R7 are H, Y is -O-alkyl, R2", R4" and R6" are H, and R3" and R5" are methyl; and
wherein W is as defined for Formula (A).
hi a third embodiment, the present invention provides an optionally substituted 9- 11 membered bicyclic 3-phosphoindole for use in the treatment of HIV represented by the following general Formula (C):
Figure imgf000075_0001
(C)
or a pharmaceutically acceptable salt, prodrug, N— oxide, quaternary amine, stereochemical isomer or tautomer thereof, wherein:
each W, Z, R1, R2, R3, A, n, R4', R5', R6' and R7' is as defined above for Formula (A);
each R3", R4", R5", and R6" is as defined above for Formula (B);
indicates the presence of a single or double bond, where:
when indicates the presence of a double bond, each Y and T independently is: a) CR2;
b) N; or
c) SR2;
with the caveat that one of Y and T must be CR2;
when indicates the presence of a single bond, then each Y and T independently is:
a) CR2;
b) O; c) NR2; or
d) SR2;
with the caveat that one of Y and T must be CR2; and
m is 1 or 2, with the proviso that m can only be 2 for T or Y = CR2.
A first series of subembodiments of the Formula (C) include all the subembodiments provided for Formula (A) above in which substituent "X" is aryl, heterocyclyl, O-aryl, and O-heterocyclyl.
A second series of subembodiments of the present invention is given for the embodiment of Formula (C) as provided above where Y and T are defined as:
j) T is -CH2 and Y is -O-;
k) T is -C-C(=O)-OCH3 andY is -O-;
1) T is -O and Y is -CR2;
m) T is (-CH2)2 and Y is -NR2;
n) T is -CH2 and Y is -SR2;
o) T is -O and Y is -C-aryl;
p) T is -NR2 and Y is -C-halo; q) T is -SR2 and Y is -C-heterocycle;
r) T is -C-C1-6 alkyl and Y is -O-;
s) T is -C-C(=S)CH2 and Y is -CH2-;
t) T is -CH-OCH3 and Y is -SR2;
u) T is -C-OH and Y is -CH2-;
v) T is -C-O-C1-6 alkyl and Y is -CH2-;
w) T is -C-NH2 and Y is -CH2-;
x) T is -C-NH-C1-6 alkyl and Y is -CH2-; and
y) T is (-CH2)2 and Y is (-CH2)2.
A third series of subembodiments of the present invention is given for the embodiment of the Formula (C) as provided above where R3 , R4 , R5 , and R6 are defined as:
a) R4" and R6" are hydrogen, and R3 and R5 each independently is halogen; - NO2; -CN; -OR2; -NH-R5-C1-6 alkyl; -NHCO-C1-6 alkyl; hydrazine; -N(OH)C1- β alkyl; C1-6 alkoxy; -OH; -NR2R2; or -C1-6 alkyl, alkenyl or alkynyl optionally substituted with one or more of -OH, -SR, -C(=W)H, -C(=W)OH, halogen, NR2R2, -C1-6 thioether, or -C1-6alkoxy;
b) R4 and R6 are hydrogen, and R3 and R5 each independently is halogen, or - C1-6 alkyl, alkenyl, alkynyl optionally substituted with one or more halogen;
c) R3", R4", R5"and R6" are hydrogen;
d) R4 and R6 are hydrogen, and R3 and R5 are methyl;
e) R4 and R6 are hydrogen, and R3 and R5 are chloro;
f) R4 and R6 are hydrogen, and R3 and R5 are fluoro;
g) R4 and R6 are hydrogen, R3 is iodo and R5 is bromo;
h) R3", R4" and R6" are hydrogen, and R5" is chloro; i) R4 and R6 are hydrogen, R3" is chloro, and R5 is methyl.
i) R3", R5" and R6" are hydrogen, and R4" is halogen; -NO2; -CN; -OR2; -NH-R5-
C1-6 alkyl; -NHCO-C1-6 alkyl; hydrazine; -N(OH)Ci-6 alkyl; C1-6 alkoxy; -OH;
-NR R ; or -Ci-6 alkyl, alkenyl or alkynyl optionally substituted with one or more of -OH, -SR, -C(=W)H, -C(=W)0H, halogen, NR2R2, -C1-6 thioether, or
-Ci-6alkoxy;
j) R3", R5" and R6" are hydrogen, and R4" is -NO2;
k) R3", R5" and R6" are hydrogen, and R4" is -OR2;
1) R3", R5" and R6" are hydrogen, and R4" is halogen; and
m) R3", R5" and R6" are hydrogen, and R4" is -NH2.
A fourth series of subembodiments for Formula (C) are defined when Z is as defined in any one of the second series of subembodiments for Formula (A), R4 , R5 , R6 and R7' are as defined in the third series of subembodiments for Formula (A), and R3 , R4 , R5 and R6 all are defined as in the third series of subembodiments for Formula (C).
A fifth series of subembodiments are defined when Z is as defined in any one of the second series of subembodiments for Formula (A), R4 , R5 , R6 and R7 are as defined in the third series of subembodiments for Formula (A), and Y and T are as defined as in the second series of subembodiments for Formula (C).
A sixth series of subembodiments for Formula (C) are defined when Z is as defined in any one of the second series of subembodiments for Formula (A), R4 , R5 , R6 and R7> are as defined in the third series of subembodiments for Formula (A), and R3 , R , R5 and R6 all are defined as in the second series of subembodiments for Formula (B).
Species of the third embodiment as given by Formula (C) above are defined when:
a) Z is -C(=W)N(R2R3)C(=W)NH2, where R2 is NH and R3 is alkyl; R1, R4', R6' and R7> are H; R5> is halo or -NO2; R4"and R6" are H; R3" and R5" independently are -OH or -NR2R2; Y is O; and T is CH2; b) Z is -C(=W)-NH-CH(-C[=W]NH2)(-CH2-C[=W]-O-CH3-aryl); R1, R4', R6' and R7' are H; R5' is halo or -CN; R4"and R6" are H; R3" and R5" independently are -C1-5 alkoxy or -C1-5 alkyl optionally substituted with -C1-5 alkoxy; Y is SR2; and T is CH2;
c) Z is -C(=W)NHNH2; R1, R4',R6' and R7' are H; R5' is halo or -NR2R2; R4"and R6" are H; R3" and R5" independently are -NO2 or -CN; Y is CH2; and T-R2-R3 is CH2;
d) Z is -C(=W)NH-CH2-C(=W)NHNH2; R1, R4', R6' and R7' are H; R5' is halo or -NH-R5-C1-6 alkyl, where R5 is -C(O) or -S(O)n and n is 0, 1 or 2; R4"and R6" are H; R3" and R5" independently are -OR2 or -CN; Y is C-C1-6 alkyl; and is T is N;
e) Z is -C(=W)NH-CH2-C(=W)NHNH2; R1, R4', R6' and R7' are H; R5' is halo or -NH-R5-C1-6 alkyl, where R5 is -C(O) or -S(O)n and n is 0, 1 or 2; R4"and R6" are H; R3" and R5" independently are -OR2 or -CN; Y is C-C1-6 alkyl; and is T is SR2 and R2 is as defined for Formula (A);
f) Z is -C(=W)NH-CH(R2)-CHV)NH2, where R2 is -NH2 or alkyl; R1, R4' , R6' and R7' are H; R5> is halo or -NHCO-C1-6 alkyl; R4"and R6" are H; R3" and R5" independently are -NHOH; Y is C-alkenyl; and T is (CH2)2;
g) Z is -C(=W)NR2-C(=W)R3, where R2 is optionally substituted alkyl, alkenyl or alkynyl; R3 is NH2; R1, R4>,R6' and R7' are H; R5' is halo or oxime; R4"and R6" are H; R3" and R5 independently are H or ~NH-R5-C1-6 alkyl, where R5 is -C(O) or -S(O)n and n is 0, 1 or 2; Y is C-alkynyl; and T is -O;
h) Z is -C(=W)NH-R2-SR2 where R2 is optionally substituted alkyl, alkenyl or alkynyl; R1, R4' ,R6> and R7' are H; R5' is halo or hydrazine; R4"and R6" are H; R3" and R5" independently are H or -NHCO-C1-6 alkyl;Y is C-carbocycle; and T is NR2 where R2 is as defined for Formula (A);
i) Z is -C(=W)-NH-N(R3)(R2), where R2 is hydroxy or alkoxy, and R3 is H or alkyl; R1, R4', R6' and R7' are H; R5' is halo or C1-6 alkyl or alkenyl optionally substituted with one or more -OH, -SR, -C(=W)H, -C(=W)0H, halogen, -NR2R2, C1-3 alkoxy, or C1-3 thioether; R4"and R6" are H; R3" and R5" independently are H or -CN; Y is C-aryl; and T is N; j) Z is -C(=W)NH-CH2NH-CH(CH3)C(=W)OH; R1, R4', R6' and R7' are H; R5> is halo or CF3; R4"and R6" are H; R3" and R5" independently are H or -NH-O-C1-3 alkyl; Y is O; and T is CH2;
k) Z is -C(=W)-NH-N(R2)-CH(R2)-C(=W)R2, where R2 is H or NH2; R1, R4', R6' and R7' are H; R5' is halo or H; R4"and R6" are H; R3" and R5" independently are H or halogen; Y is -NR2 and T is CR2 where R2 is as defined in the first general embodiment;
1) Z is -C(=W)-N(R2)-C(=W)R3, where R2 is NH and R3 is CH3; R1, R4>, R6' and R7' are H; R5' is halo or H; R4"and R6" are H; R3" and R5" independently are H or halogen; Y is -O; and T is (C-O-alkyl);
m) Z is -C(=W)-NH-CH=CH-C(=W)R2, where R2 is NH2; R1, R6' and R7' are H; R4' and R5 are halo or H; R4 and R6 are H; R3 and R5 independently are H or -OR2 where R2 is as defined in the first general embodiment; Y is -O; and T is (C-CH2-CH3);
n) Z is -C(=W)-NH-CH=CH-C(=W)R2, where R2 is -NH2; R1, R6' and R7> are H; R4' and R5' are halo or -NO2; R4"and R6" are H; R3" and R5" independently are H or -NHOH; Y is -O and T is C-CH3;
o) Z is -C(=W)R2(CH)2-C(=W)R2, where R2 is NH or NH2; R1, R6' and R7' are H; R4 and R5' are halo or -OR2 where R2 is as defined in the first general embodiment; R4 and R6 are H; R3 and R5 independently are H or -C1-3 alkyl or alkenyl optionally substituted with one or more -OH, -SR, -C(=W)0H, halogen, or -NH2; Y is -SR2; and T is CH2;
p) Z is -C(=W)-R2-CH2-A-C(=W)R2, where R2 is as defined in the first general embodiment, and A is a divalent linker as defined in the first general embodiment; R1, R6' and R7' are H; R4' and R5> are halo or -CN; R4"and R6" are H; R3" and R5" independently are H or -C1-3 alkoxy; Y is -NR2 where R2 is as defined in the first general embodiment; and T is (CH2)2;
q) Z is -C(=W)-R2-CH2-A-C(=W)R2, where R2 is alkyl or -NH2, and A is a divalent linker; R1, R6' and R7' are H; R4' and R5' are halo or -NR2R2, where R2 is as defined in the first general embodiment; R4"and R6" are H; R3" and R5" independently are H or -OH; Y is -CH2 and T is (CH2)2; r) Z is -C(=W)-A-R2-C(=W)OR3, where R2 is -NH and R3 is -H or alkyl; R1, R6> and R7' are H; R4> and R5 are halo or -NHR5-C1-3 alkyl, where R5 is as defined in the first general embodiment; R4"and R6" are H; R3" and R5 independently are H or -NR2R2, where R2 is as defined in the first general embodiment; Y is -C-C1-6 alkyl; and T is CH2;
s) Z is -C(=W)-NH-C(=W)OR3, wherein R3 is as defined for Formula (A); R1, R6' and R7' are H; R4' and R5> are halo or -NHCO-C1-3 alkyl; R4" and R6" are H; R3" and R5" independently are halo or H; Y is -C-alkenyl; and T is O;
t) Z is -C(=W)R3-NH-C(=W)-R2, where R2 is -NH2 and R3 is -NH; R1, R6' and R7' are H; R4' and R5' are halo or oxime; R4"and R6" are H; R3 and R5" independently are H or -C1-3 alkyl or alkenyl optionally substituted with one or more halogen; Y is -C- alkynyl; and T is (CH2)2;
u) Z is -C(=W)-N(C=O)-N(R2)-R3, where R2 is H or alkyl, R3 is -NH2, R1, R6' and R7 are H; R4 and R5 are halo or hydrazine; R4 and R6 are H; R3 and R5 independently are H; Y is -C-aryl; and T is (CH2)2;
v) Z is -C(=W)-N(R2)-N(R2)-C(=W)R3, where R2 is H or alkyl, and R3 is NH2; R1,
R6' and R7' are H; R4' and R5' are halo or -NHS(O)2-C1-3 alkyl; R4"and R6" are H; R3" and R5 independently are H or -methyl; Y is -C-heterocycle; and T is O;
w) Z is -C(=W)-N(-N[R2][R3])-R3, where R2 is H or alkyl, and R3 is NH2; R1, R6' and R7 are H; R4' and R5' are halo or -C1-6 alkyl or alkenyl optionally substituted with one or more -OH, -SR, -C(=W)H, -C(=W)0H, halogen, NR2R2, C1-3 alkoxy, or C1-3 thioether, where R2 is as defined in the first general embodiment; R4 and R6 are H; R3 and R5 independently are H or -NR2R2, where R2 is as defined in the first general embodiment; Y is -C-carbocycle; and T is NR2 where R2 is as provided above;
x) Z is -C(=W)R2-C(=W)NH2, where R2 is alkyl; R1, R6' and R7' are H; R4' and R5' are halo or -H; R4 "and R6 are H; R3 and R5 independently are H or chlorine; Y is -SR2 and T is CH2;
y) Z is -C(=W)R2-SR2, where R2 is -NH or alkyl; R1 , R6' and R7' are H; R4> and R5' are halo or -CF3; R4 and R6 are H; R3 and R5" independently are H or fluorine; Y is -O; and T is (CH2)2; z) Z is -C(=W)-CH(R2)-C(=NH)R2, where R2 is H or NH2; R1, R4' and R7' are H; R5' and R6' are halo or -H; R4"and R6" are H; R3" and R5" independently are H or -CF3; Y is -NR2; and T is CR2; where R2 is as defined for Formula (A);
aa) Z is -C(=W)-NH-A-NH-C(=W)-A-C-(=W)-R2 where R2 is NH2 and A is a divalent linker; R1, R4' and R7' are H; R5' and R6> are -CN or -NO2; R4"and R6" are H; R3" and R5" independently are H or -NO2; Y is -CH2; and T is NR2, where R2 is as defined for Formula (A);
bb) Z is -C(=W)-R2-CH-(-A-C[=W]R2)(-C[=W]-NH2), where R2 is H, alkyl, or NH; A is a divalent spacer linkage as defined for Formula (A); R1, R4 , and R7 are H; R5 and R6' are H or -NHCO-C1-6 alkyl; R4" and R6" are H; R3" and R5" independently are H or hydrazinyl; Y is (CH2)2; and T is N;
cc) Z is -C(=W)-NH-A(-C[=W]-NH2), where A is an alkylene or arylene divalent spacer linkage; R1, R4' and R7' are H ; R5' and R6' are H or -NH(SO2)C1-6 alkyl; R4"and R6" are H; R3" and R5" independently are H or -NHOH; Y is -C-alkenyl; and T is N;
dd) Z is -C(=W)-NH-CH(-A-R3)(-C[=W]-NH2), where A is an alkylene or alkenylene spacer; R3 is OH; R1, R4' and R7' are H ; R5' and R6' are H or CF3; R4"and R6" are H; R3 and R5 independently are H or -NR2R2 where R2 is as defined for Formula (A); Y is -C-carbocycle; and T is SR2;
ee) Z is -C(=W)-NH-CH(R2)(R3), where R2 and R3 each is C(=W)NH2; R1, R6' and R7' are H ; R5' and R4' are H or halo; R4"and R6" are H; R3" and R5" independently are H or -C1-5 alkoxy; Y is -O; and T is CH2;
S) Z is -C(=W)-A-CH(-R2-C[=W]-NH2)(-C[=W]-NH2), where A is a spacer linkage as defined for Formula (A); R2 is NH; R1, R6' and R7' are H ; R5' and R4' are H or -NO2; R4"and R6" are H; R3" and R5" independently are H or -OH; Y is -N; and T is CH2;
gg) Z is -C(=W)-NH-CH-(-CH-R2-OH)(-C[=W]-NH2), where R2 is NH or alkylene;
R1, R6> and R7' are H ; R5' and R4' are H or -halogen; R4"and R6" are H; R3" and R5" independently are H or -halogen; Y is -SR2; and T is CH-OCH3 ;
hh) Z is -C(=W)-NH-CH(-R2X-C[=W]-NH2), where R2 is alkyl; R1 , R6' and R7' are H ; R5' and R4' are H or -NO2; R4"and R6" are H; R3" and R5" independently are H or -CN; Y is -C-aryl; and T is N; ii) Z is -C(=W)-NH-CH(-R2)(-C[=W]-NH2), where R2 is alkyl; R1, R6' and R7' are H; R5' and R4> are H or -OH; R4"and R6" are H; R3" and R5" independently are H or -CN; Y is -C-heterocycle; and T is (CH2)2;
jj) Z is -C(=W)-NH-CH(-R2-C[=W]-NH2)(-C[=W]-NH2, where R2 is alkylene; R1, R6' and R7' are H; R4' and R5'are H or -NO2; R4"and R6" are H; R3" and R5" independently are H or -NHOH; Y is -O and T is CH2;
fck) Z is -C(=W)-NH-CH(-R2)(-C[=W]-NH2), where R2 is -S(O)R3 and R3 is as defined for Formula (A); R1, R6' and R7' are H; R4> and R5'are H or -OR2, R4"and R6" are H; R3 and R5 independently are H or -C1-3 alkoxy; Y is -NR2, where R2 is as defined in the first general embodiment, and T is CH2;
11) Z is -C(=W)-NH-CH(-C[=NH]-NH2)(-C[=W]-NH2); R1, R6> and R7' are H; R4' and R5 are H or -NR2R2, where R2 is as defined in the first general embodiment; R4 and R6" are H; R3" and R5" independently are H or -OH; Y is -SR2, and T is (CH2)2;
mm) Z is -C(=W)-NH-NH-CH(R3)-C[=W]R2, where R2 is -NH2 and R3 is optionally substituted aryl or heteroaryl; R1, R6' and R7' are H; R4' and R5'are H or -CN; R4"and R6" are H; R3 and R5 independently are H or -NR2R2, where R2 is as defined in the first general embodiment; Y is -C-C1-6 alkyl; and T is N;
nn) Z is -C(=W)-NHR2(-R3)-C[=W]NH2, where R2 is alkyl, and R3 is optionally substituted alkyl, aryl or heteroaryl; R1, R6' and R7' are H; R4' and R5'are H or -NH-R5- C1-3 alkyl, where R5 is as defined in the first general embodiment; R4 and R6 are H; R3 and R5 independently are H or -C1-6 alkyl or alkenyl optionally substituted with one or more -OH, -SR, -C(=W)0H, halogen, or NH2; Y is -C-alkenyl; and T is SR2 where R2 is as defined for Formula (A);
oo) Z is -C(=W)-NHR2(-R3)-C[=W]NH-R2-C(=W)OH, where R2 is alkyl, and R3 is optionally substituted alkyl, aryl or heteroaryl; R1, R6 and R7 are H; R4 and R5 are H or
-NHCO-C1-3 alkyl; R4"and R6" are H; R3" and R5" independently are H or -C1-6 alkyl or alkenyl optionally substituted with one or more halogen; Y is -C-alkynyl; and T is CH2;
pp) Z is -C(=W)-NHR2(-R3)-C[=W]NH-R2 NH2, where R2 is alkyl, and R3 is optionally substituted alkyl, aryl or heteroaryl; R1, R6 and R7 are H; R4' and R5'are H or -oxime; R4"and R6" are H; R3" and R5" independently are H or -halogen; Y is -CH2; and T-NR2 where R2 is as defined for Formula (A);
qq) Z is -C(=W)-NHR2(-R3)-C(=W)NH-R3, where R2 is alkyl, and R3 is optionally substituted alkyl, aryl or heteroaryl; R1, R6 and R7 are H; R4 and R5 are H or -hydrazine; R4"and R6" are H; R3" and R5" independently are H; Y is -NR2 where R2 is as defined in the first general embodiment; and T is (CH2)2;
rr) Z is -C(=W)-R2-A-C(=W)-NH2, where R2 is -NH, and A is a divalent spacer linkage that is optionally substituted aryl or heteroaryl; R1, R6 and R7 are H; R4 and R5'are H or -NH(SO2)C1-6 alkyl; R4"and R6" are H; R3" and R5" independently are H or methyl; Y is -C-C1-6 alkyl; and T is N;
ss) Z is -C(=W)NH-R3, where R3 is an optionally heterocycle; R1, R6' and R7' are H; R4 and R5 are H or -C1-6 alkyl or alkenyl optionally substituted with one or more -OH, -SR, C(=W)H, C(=W)0H, halogen, NR2R2, C1-3 alkoxy, or C1-3 thioether; R4"and R6" are H; R3 and R5 independently are H or chloro; Y is -C-alkenyl; and T is NR2 where R2 is as defined for Formula (A);
tt) Z is -C(=W)NH-A-S(O)2-R3, where A is an alkylene, alkenylene or alkynylene divalent spacer linkage, R3 is -NH2, and R5 is -SO2; R1, R6' and R7' are H; R4' and R5'are H; R4"and R6" are H; R3" and R5" independently are H or fluoro; Y is -(CH2)2; and T is (CH2)2;
uu) Z is -C(=W)NH-NH-A-(R3)-S(O)2-NH2, where A is an alkylene, alkenylene or alkynylene divalent spacer linkage, R3 is aryl, arylene or heteroaryl, and R5 is -SO2; R1, R6' and R7' are H; R4' and R5'are H or -CF3; R4"and R6" are H; R3" and R5" independently are H or CF3; Y is -O; and T is CH2;
w) Z is -C(=W)NH-R3(SO2-NH2), where R3 is aryl, arylene or heteroaryl; R1, R4' and R7' are H; R5' and R6' are H or -halo; R4"and R6" are H; R3" and R5" independently are
H or -NO2; Y is -O; and T is CR2 where R2 is as defined for Formula (A);
ww) Z is -C(=W)NH-A-(NH2), where A is an alkylene, alkenylene or alkynylene divalent spacer linkage; R1, R4' and R7' are H; R5' and R6'are H or -NO2; R4"and R6" are H; R3 and R5 independently are H or -halo; Y is -0-; and T is C-carbocycle; xx) Z is -C(=W)NH-R3(-A-SO2-NH2), where A is an alkylene, alkenylene or alkynylene divalent spacer linkage; R3 is aryl, arylene or heteroaryl; R1, R4 and R7 are H; R5' and R6'are H or -CN; R4"and R6" are H; R3" and R5" independently are H or -NH- R5-C1-3 alkyl, where R5 is as defined in the first general embodiment; Y is -C- carbocycle; and T is NR2 where R2 is as defined for Formula (A);
yy) Z is -C(-W)NH(-R3)-C(-W)NH-A-C(=W)OH, where A is an optionally substituted alkylene, alkenylene or alkynylene divalent spacer linkage, R3 is as defined in the first general embodiment; R1, R4' and R7' are H; R5' and R6'are H or -OR2 where R2 is as defined in the first general embodiment; R4 and R6 are H; R3 and R5 independently are H or -CN; Y is -C-aryl; and T is CH2;
zz) Z is -C(=W)NH(-R3)-C(=W)NH-A-C(=W)NH2, where A is an optionally substituted divalent spacer linkage as defined for Formula (A); R3 is as defined in the first general embodiment; R1, R4' and R7' are H; R5' and R6'are H Or-NR2R2, where R2 is as defined in the first general embodiment; R4 and R6 are H; R3 and R5 independently are H or -NH-R5-C1-6 alkyl, where R5 is as defined in the first general embodiment; Y is
-C-heterocycle; and T is SR2;
aaa) Z is -C(=W)NHR2-C(=W)NH-R2-C(=W)OH, where R2 is as defined for Formula (A); A is an optionally substituted divalent spacer linkage; R1, R4' and R7' are H; R5' and R6'are H or -NHSO2-C1-6 alkyl; R4"and R6" are H; R3" and R5" independently are H or hydrazine; Y is -O and T is (CH2)2; and
bbb) Z is -C(=W)NHR2-C(=W)NH-R2-C(=W)NH2, where R2 is as defined for Formula (A); A is an optionally substituted divalent spacer linkage; R1, R4 and R7 are H; R5' and R6'are H or -NHCO-C1-6 alkyl; R4"and R6" are H; R3" and R5" independently are H or -OH; Y is -NR2 where R2 is as defined in the first general embodiment, and T is CH2;
wherein W is as defined above for the first general embodiment of Formula (A).
For all embodiments, subembodiments are defined when: 1) Z is -C(=W)-NH-R2; -C(=W)-NR2R3; -C-R2R3; -CR2-C(=W)R3; -R2- C(=W)R3; -R2-C(=W)R2; -R2R3; R3; or C(=W)-NH-CR2R2-C(=W)-NH-CR2R3-C(=W)- NR2R3;
2) R4', R6 and R7 are H, and R5 is i) halogen, and chlorine in particular; ii) hydrazine; iii) C1-6 alkyl, alkenyl, alkynyl, amino-alkyl, thioamino-alkyl, or aminocarbonyl-alkyl, each of which optionally may be substituted with one or more -OH, -C(=W)H, -C(=W)OH, -NR2R3, -Ci-3 alkoxy, or -C1-3 thioether; or one of the following combinations:
a) R5', R6' and R7 are hydrogen, and R4' is halogen;
b) R4>, R5' and R7' are hydrogen, and R6' is halogen;
c) R4 , R5 and R6 are hydrogen, and R7' is halogen;
d) R5', R6' and R7' are hydrogen, and R4' is CF3;
e) R4 , R5 and R7 are hydrogen, and R6 is CF3; and
f) R4', R5' and R6' are hydrogen, and R7' is CF3.
III. Definitions
The following definitions and term construction are intended, unless otherwise indicated.
Ranges, specific values, and typical values listed for radicals, substituents and derivatives are for illustration only, and do not exclude other defined values or values within defined ranges for the radicals, substituents and derivatives. Whenever a range is described herein, the range independently includes each member of the range. As an illustrative example, when C1-6-alkyl are refered to, this listing independently includes Ci -alkyl, C2-alkyl, C3-alkyl, C4-alkyl, C5-alkyl and C6-alkyl.
"Halo" is fluoro, chloro, bromo or iodo. "Alkyl", "alkoxy", "alkenyl", "alkynyl", etc., includes both straight chain and branched groups. However, reference to an individual radical such as "propyl" embraces only that straight-chain radical, whereas a branched chain isomer such as "isopropyl" is specifically termed such.
"Alkyl" as used herein and unless otherwise specified, is a saturated, straight, branched, or cyclic, primary, secondary, or tertiary hydrocarbon of, for example, C1-10, and specifically includes methyl, ethyl, propyl, isopropyl, cyclopropyl, butyl, isobutyl, t- butyl, pentyl, cyclopentyl, isopentyl, neopentyl, hexyl, isohexyl, cyclohexyl, cyclohexylmethyl, 3-methylpentyl, 2,2-dimethylbutyl, and 2,3-dimethylbutyl. When the context of this document allows alkyl to be substituted, the moieties with which the alkyl group may be substituted include but not limited to hydroxyl, amino, alkylamino, arylamino, alkoxy, aryloxy, aryl, heterocyclyl, halo, carboxy, acyl, acyloxy, amido, nitro, cyano, sulfonic acid, sulfate, phosphonic acid, phosphate, or phosphonate, either protected or unprotected as needed, as known to those skilled in the art and as taught, for example, in Greene et al., Protective Groups in Organic Synthesis, John Wiley and Sons,
Third Ed., 1999.
The term "lower alkyl" as used herein and unless otherwise specified, includes a C1-4 saturated, straight, branched, or if appropriate, cyclic (for example, cyclopropyl) alkyl group, including both substituted and unsubstituted forms. Unless otherwise specifically stated in this application, when alkyl is a suitable moiety, lower alkyl is typical. Similarly, when alkyl or lower alkyl is a suitable moiety, unsubstituted alkyl or lower alkyl is typical.
The terms "alkenyl" and "alkynyl" refer to alkyl moieties, including both substituted and unsubstituted forms wherein at least one saturated C-C bond is replaced by a double or triple bond. Thus, C2-6 alkenyl may be vinyl, allyl, 1-propenyl, 2- propenyl, 1-butenyl, 2-butenyl, 3-butenyl, 1-pentenyl, 2-pentenyl, 3-ρentenyl, 4- pentenyl, 1-hexenyl, 2-hexenyl, 3-hexenyl, 4-hexenyl, or 5-hexenyl. Similarly, C2-6 alkynyl may be ethynyl, 1-ρropynyl, 2-propynyl, 1-butynyl, 2-butynyl, 3-butynyl, 1- pentynyl, 2-pentynyl, 3-pentynyl, 4-pentynyl, 1-hexynyl, 2-hexynyl, 3-hexynyl, 4- hexynyl, or 5-hexynyl. The term "alkylene" refers to a saturated, straight chain, divalent alkyl radical of the formula -(CH2)n-, wherein "n" maybe any whole integer from 1 to 10.
As used herein with exceptions as noted, "aryl" is intended to mean any stable monocyclic, bicyclic or tricyclic carbon ring of up to 8 members in each ring, wherein at least one ring is aromatic as defined by the Huckel 4n+2 rule. Examples of aryl ring systems include phenyl, naphthyl, tetrahydronaphthyl, and biphenyl. The aryl group may be substituted with one or more moieties including but not limited to hydroxyl, amino, alkylamino, arylamino, alkoxy, aryloxy, alkyl, heterocyclyl, halo, carboxy, acyl, acyloxy, amido, nitro, cyano, sulfonamido, sulfonic acid, sulfate, phosphonic acid, phosphate, or phosphonate, either protected or unprotected as needed, as known to those skilled in the art and as taught, for example, in Greene et al., Protective Groups in Organic Synthesis.
John Wiley and Sons, Third Ed., 1999.
The term "heterocycle" or "heterocyclic" as used herein except where noted, refers to a stable 5- to 7-membered monocyclic or stable 8- to 11-membered bicyclic heterocyclic ring which is either saturated or unsaturated, including heteroaryl, and which consists of carbon atom(s) and from one to four heteroatoms including but not limited to O, S, N and P; and wherein the nitrogen and sulfur heteroatoms may optionally be oxidized, and/or the nitrogen heteroatom quaternized, and including any bicyclic group in which any of the above-identified heterocyclic rings is fused to a benzene ring. The heterocyclic ring may be attached at any heteroatom or carbon atom which results in the creation of a stable structure. The heteroaromatic ring may be partially or totally hydrogenated, as desired. For example, dihydropyridine may be used in place of pyridine. Functional oxygen and nitrogen groups on a heteroaryl may be protected as necessary or desired. Suitable protecting groups for oxygen or nitrogen include trimethylsilyl, dimethylhexylsilyl, t-butyldimethylsilyl, t-butyldiphenylsilyl, trityl, substituted trityl, alkyl, methanesulfonyl, p-toluenesulfonyl, or acyl groups such as acetyl and propionyl.
Non-limiting examples of heteroaryl and heterocyclic groups include furyl, pyridyl, pyrimidyl, pyridazinyl, pyrazinyl, piperidinyl, piperazinyl, thienyl, pyrrolyl, pyrrolinyl, pyrrolidinyl, imidazolyl, tetrazolyl, triazolyl, triazinyl, thiazinyl, oxazolyl, purinyl, carbazolyl, quinolinyl, pyrazolyl, morpholinyl, benzimidazolyl, and the like.
Any of the heteroaromatic and heterocyclic moieties may be optionally substituted as described above for aryl, including substitution(s) with one or more hydroyxl, amino, alkylamino, arylamino, alkoxy, aryloxy, alkyl, heterocyclyl, halo, carboxy, acyl, acyloxy, amido, nitro, cyano, sulfonic acid, sulfate, phosphonic acid, phosphate, or phosphonate, either protected or unprotected as needed, as known to those skilled in the art and as taught, for example, in Greene et al., Protective Groups in Organic Synthesis. John
Wiley and Sons, Third Ed., 1999.
The term "acyl" refers to a compound of the formula "RC(O)-", wherein R is substituted or unsubstituted alkyl or aryl as defined herein.
The term "carboxyl" refers to a compound of the formula "RCOOH", wherein R is substituted or unsubstituted alkyl or aryl as defined herein.
The term "aralkyl" as used herein unless otherwise specified, refers to an aryl group as defined above linked to the molecule through an alkyl group as defined above.
The term "alkaryl" as used herein unless otherwise specified, refers to an alkyl group as defined above linked to the molecule through an aryl group as defined above.
The term "alkoxy" as used herein unless otherwise specified, refers to a moiety of the structure "-O-alkyl", where alkyl is as defined above.
The term "amino" as used herein unless otherwise specified, refers to a moiety represented by the structure "-NR2", and includes primary amines, and secondary and tertiary amines optionally substituted by alkyl, aryl, heterocyclyl, and/or sulfonyl groups. Thus, R2 may represent two hydrogens, two alkyl moieties, or one hydrogen and one alkyl moiety.
The term "amido" as used herein unless otherwise specified, refers to a moiety represented by the structure "-C(O)NR2", wherein R2 is an H, alkyl, aryl, acyl, heterocyclyl and/or a sulfonyl group.
As used herein, an "amino acid" or an "amino acid residue" is a natural amino acid or some portion thereof (i.e., Ala, Arg, Asn, Asp, Cys, GIu, GIn, GIy5 His, HyI, Hyp, lie, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr and VaI) in D or L form, or an unnatural amino acid having one or more open valences such as, for example, t-butylglycine, ornithine, hippuric acid and phosphothreonine. Other unnatural amino acids are those represented by the formula "NH2(CH2)yCOOH", wherein y is 2-12, and includes aminoalkanoic acids such as ε-amino caproic acid (H2N-(CH2)5-COOH). The term also comprises natural and unnatural amino acids bearing amino-protecting groups such as acyl, trifluoroacetyl and benzyloxycarbonyl, as well as natural and unnatural aminot acids protected at carboxy moieties by protecting groups such as C1-6 alkyl, phenyl or benzyl ester and amide, and protecting groups known to those of skill in the art. In all instances where natural and unnatural amino acids contain one or more chiral centers, all possible stereochemical configurations, including both "D" and "L" forms and mixtures thereof, including racemic mixtures, are contained herein.
The term "quaternary amine" as used herein includes quaternary ammonium salts that have a positively charged nitrogen. They are formed by the reaction between a basic nitrogen in the compound of interest and an appropriate quaternizing agent such as, for example, methyliodide or benzyliodide. Appropriate counterions accompanying a quaternary amine include acetate, trifluoroacetate, chloro, bromo and iodo ions.
As used herein, the term "iV-oxides" denotes a state of the compounds of the present invention in which one or more nitrogen atoms are oxidized with an oxygen atom.
As used herein, a "retrovirus" includes any virus that expresses reverse transcriptase. Examples of a retrovirus include but are not limited to, HIV-I, HTV-2, HTLV-I, HTLV-II, FeLV, FIV, SIV, AMV, MMTV, and MoMuLV.
As used herein, "reverse transcriptase" or "RT" refers to an enzyme having a non-nucleoside inhibitory binding site similar to that of HIV-I RT, and to which ligands, which bind the composite binding pocket of the compounds of the present invention, also will bind. One measure of RT activity is viral replication. A measure of HIV-I viral replication is the automated assay that utilizes MTT, as described earlier in this specification. Another measure is the p24 core antigen enzyme immunoassay, such as, for example, the assay commercially available from Coulter Corporation/mimunotech, Inc.® (Westbrook, MI). Another means for measuring RT activity is by assaying recombinant HIV-I reverse transcriptase activity, such as, for example, by using the Quan-T-RT™ assay system commercially available from Amersham® (Arlington Heights, IL) and as described by Bosworth et al., Nature, 1989, 347:167-168. As used herein, a compound that "inhibits replication of human immunodeficiency virus (HIV)" means a compound that, when contacted with HIV-I, for example, via HIV-infected cells, effects a reduction in the amount of HIV-I as compared with an untreated control. Inhibition of replication of HIV-I may be measured by any means known to those skilled in the art, such as, for example, by the p24 assay disclosed above.
The reagent denoted "mCPBA" in the synthesis schemes is meta-chloro- peroxybenzoic acid.
The term "salvage therapy" as used herein means a compound that can be taken with any regimen after a patient' s initial treatment regimen has failed.
As used herein, the term "host" refers to a multicellular or unicellular organism in which the virus can replicate. Thus, "host" includes a cell line, an mammal and, preferably, a human. Alternatively, a host can be carrying a part of the HIV genome whose replication or function may be altered by the compounds of the present invention. The term host specifically refers to infected cells, cells transfected with all or part of the
HIV genome, and mammals, especially primates including chimpanzees and humans. In most mammal applications of the present invention, the host is a human patient. Veterinary applications, however, are clearly anticipated by the present invention, such as, for example, in chimpanzees.
IV. Pharmaceutically Acceptable Salts, Prodrugs, Stereoisomers and Tautomers
An active compound may be administered as a salt or prodrug that, upon administration to the recipient, is capable of providing directly or indirectly the parent compound, or that exhibits activity itself. Nonlimiting examples include a pharmaceuticaUy-acceptable salt, alternatively referred to as a "physiologically- acceptable salt". In addition, modifications made to a compound can affect its biologic activity, in some cases increasing the activity over the parent compound. This activity can be assessed by preparing a salt or prodrug form of the compound, and testing its antiviral activity by using methods described herein or other methods known to those of skill in the art of NNRTIs. The phrase "pharmaceutically acceptable salt or prodrug" is used throughout the specification to describe any pharmaceutically acceptable form (such as an ester, amide, salt of an ester, salt of an amide or related group) of a compound that, upon administration to a patient, provides an active compound of the invention. The terms "stereoisomer" and "tautomer" as used herein include all possible stereoisomeric and tautomeric forms of the compounds of the present invention, as well as their quaternary amine, salt, solvate, prodrug, derivative, and JV-oxide forms. Where the compounds of the general formulae (I) and (II) contain one or more chiral centers, all possible enantiomeric and diastereomeric forms are included.
The term "pharmaceutically acceptable salt" refers to the state of a compound in which the compound carries a counterion that is pharmaceutically acceptable, and wherein the salt retains the desired biological activity of the herein-identified compounds while exhibiting minimal undesired toxicological effects. Such salts are non-toxic, therapeutically useful forms of the compounds of the present invention. Any salt that retains the desired biological activity of the compounds contained herein and that exhibits minimal or no undesired or toxicological effects is intended for inclusion here. Pharmaceutically acceptable salts include those derived from pharmaceutically acceptable organic or inorganic acids and bases. Non-pharmaceutically acceptable acids and bases also find use herein, as for example, in the synthesis and/or purification of the compounds of interest. Thus, all "salts" are intended for inclusion here.
Non-limiting examples of suitable salts include those derived from inorganic acids, such as, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, nitric acid, bicarbonic acid, carbonic acid; and salts formed with organic acids, such as, for example, formic acid, acetic acid, oxalic acid, tartaric acid, succinic acid, malic acid, malonic acid, ascorbic acid, citric acid, benzoic acid, tannic acid, palmoic acid, alginic acid, polyglutamic acid, tosic acid, methanesulfonic acid, naphthalenesulfonic acid, naphthalenedisulfonic acid, α-ketoglutaric acid, α- glycerophosphoric acid and polygalacturonic acid. Suitable salts include those derived from alkali metals such as lithium, potassium and sodium, from alkaline earth metals such as calcium and magnesium, as well as from other bases well known to those of skill in the pharmaceutical art. Other suitable salts include those derived from metal cations such as zinc, bismuth, barium, or aluminum, or with a cation formed from an amine, such as ammonia, NN-dibenzylethylene-diamine, D-glucosamine,tetraethylammonium, or ethylenediamine. Moreover, suitable salts include those derived from a combination of acids and bases, such as, for example, a zinc tannate salt.
A pharmaceutically acceptable prodrug refers to a compound that is metabolized (i.e., hydrolyzed or oxidized, for example) in the host to form a compound of the present invention. Typical examples of prodrugs include compounds that have biologically labile protecting groups on a functional moiety of the active compound. Prodrugs include compounds that can be oxidized, reduced, animated, deaminated, hydroxylated, dehydroxylated, hydrolyzed, dehydrolyzed, alkylated, dealkylated, acylated, deacylated, phosphorylated, and/or dephosphorylated to produce the active compound.
The compounds of the present invention either possess antiviral activity against retroviruses and HIV in particular, or are metabolized to a compound that exhibits such activity.
Any of the oxo-pyrimidine compounds described herein may be administered as a prodrug to increase the activity, bioavailability, stability, or otherwise alter the properties of the oxo-pyrimidine. A number of prodrug ligands also are known. In general, acylation, alkylation or other lipophilic modifications of a heteroatom of the oxo- pyrimidine will increase the stability of the compound. Examples of substituent groups that can replace one or more hydrogens on a heterocycle include, but are not limited to, alkyl, aryl, steroidal, carbohydrate including sugars, 1,2-diacylglycerol, phospholipid, phosphotidylcholine, phosphocholine, and/or alcohol. Any of these may be used in combination with the disclosed oxo-pyrimidine compound to achieve a desired effect.
V. Methods of Treatment
In one embodiment, a method of treatment or prophylaxis of an HTV infection in a host is provided, comprising administering a 3-phosphoindole compound to a host in need thereof. In one particular embodiment, the comound is administered orally, parenterally, enterally, intravenously, intradermally, subcutaneously, percutaneously, transdermally, intranasally, topically or by inhalation therapy. The compound may be administered to a host that has been diagnosed with an HIV infection by measurement of a viral load in the host blood or tissue. In other embodiments the host can have been diagnosed by measurement of an anti-HIV antibody titer in blood. In another embodiment, the compounds are administered to reduce or prevent symptoms of AIDS (acquired immune deficiency syndrome) in a host. In yet another embodiment the compounds of the invention are administered to a host at risk of infection with HIV.
In another embodiment, the active compound exhibits activity against drug- resistant forms of HIV, and thus exhibits decreased cross-resistance against currently approved antiviral therapies. The phrase " activity against a drug-resistant form of HIV means that a compound (or its prodrug or pharmaceutically acceptable salt) is active against the mutant strain with an EC50 of less than approximately 50, 25, 10 or 1 micromolar concentration. In one embodiment, the non-nucleoside reverse transcriptase inhibitor (NNRTI) displays an EC50 (in molar concentration) against a mutant HIV strain of less than approximately 5, 2.5, 1 or 0.1 micromolar. In one non-limiting embodiment, the HIV mutant strain has a reverse trancriptase mutation at lysine 103 -> asparagine and/or tyrosine 181 -^ cysteine.
The 3-phosphoindoles can be assessed for their ability to inhibit reverse transcriptase activity in vitro according to standard screening methods. The spectrum of activity exhibited by any particular compound is determined by evaluating the compound in assays described in this specification or with other confirmatory assays known to those skilled in the art of anti-HIV compounds. Compounds typically exhibit an EC50 of less than 10-15 μM.
In one embodiment, the efficacy of the 3-phosphoindoles is measured by the
HTV-specific enzyme-linked immunosorbent assay, p24 ELISA. Drug efficacy is expressed as percent inhibition of the HIV p24 antigen in this rapid and sensitive assay. In a related embodiment useful for specific experiments, the efficacy of the anti-HIV compound is determined by a "plaque reduction assay" which measures the concentration of compound necessary to reduce the plaque number of the virus in vitro, according to the methods set forth more particularly herein, by 50% (i.e., the EC50 of the compound). In some embodiments the compound exhibits an EC50 of less than 15, or less that 10 micromolar to nanomolar amounts in vitro.
VI. Combination or Alternation Therapy
In a certain embodiments, the 3-phosphoindole compound is administered in combination and/or alternation with one or more other anti-retroviral or anti-HIV agent. In one embodiment, the effect of administering two or more such agents in combination and/or alternation produces a synergistic effect in inhibiting HIV replication. In another embodiment, the effect of administering two or more such agents in combination and/or alternation produces an additive effect in inhibiting HTV replication.
In combination therapy, effective dosages of two or more agents are administered together, whereas during alternation therapy an effective dosage of each agent is administered serially. The dosages will depend upon absorption, inactivation, and excretion rates of the drugs as well as other factors known to those of skill in the art. Dosage values also will vary with the severity of the condition to be alleviated. For any particular individual, specific dosage regimens and schedules should be adjusted over time to meet the needs of the individual and the professional judgment of the person administering or supervising the administration of the compositions.
Drug resistance most typically occurs by mutation of a gene that encodes for an enzyme used in the viral replication cycle, and most typically in the case of HIV, in either the reverse transcriptase or protease genes. It has been demonstrated that the efficacy of an anti-HIV drug can be prolonged, augmented or restored by administering the compound in combination or alternation with a second, and perhaps third, antiviral compound that induces a different mutation(s) from that selected for by the principle drag. Such drug combinations simultaneously reduce the possibility of resistance to any single drug and any associated toxic effects. Alternatively, the pharmacokinetics, biodistribution, or other parameters of the drug can be altered by such combination or alternation therapy. For example, the use of a combination of drugs may permit an individual drug within that combination to be given at a dosage lower than what would be required when the drug is administered as a monotherapeutic. Likewise, when drugs that target different stages of the viral life cycle are combined, there exists the possibility for potentiating their effects. Moreover, use of combinations of drugs could lower or eliminate undesirable side-effects from a single drug while still producing anti-viral activity. IQ general, combination therapy is typical over alternation therapy because it places multiple, simultaneous pressures on the virus.
The second antiviral agent for the treatment of HIV can be, for example, a protease inhibitor, an HlV-integrase inhibitor, a chemokine inhibitor, or a reverse transcriptase inhibitor ("RTI"), the latter of which can either be a synthetic nucleoside reverse transcriptase inhibitor ("NRTI") or a non-nucleoside reverse transcriptase inhibitor ("NNRTI"). In other embodiments, a second or third compound may be a pyrophosphate analog or a fusion-binding inhibitor. A list compiling resistance data collected in vitro and in vivo for certain antiviral compounds is found in Schinazi et al., Mutations in retroviral genes associated with drug resistance, International Antiviral
News, 1997, 5(8).
In certain embodiments, the indole compound is administered in combination and/or alternation with FTC (2',3'-dideoxy-3'-thia-5-fluorocytidine); 141W94 (amprenavir, Glaxo Wellcome, Inc.); Viramune (nevirapine); Rescriptor (delavirdine); DMP-266 (efavirenz); DDI (2',3'-dideoxyinosine); 3TC (3'-thia-2',3'-dideoxycytidine);
DDC (2',3'-dideoxycytidine), abacavir (1592U89), which is (lS,4R)-4-[(2-amino-6- cyclopropyl-amino)-9H-purin-9-yl]-2-cyclopentene-l -methanol succinate, Tenofovir DF (Viread), D4T, or AZT.
Other examples of antiviral agents that can be used in combination and/or alternation with the compounds disclosed herein include, but are not limited to, foscarnet; carbovir; acyclovir; interferon; fusion inhibitors such as enfuvirtide; and β-D- dioxolane nucleosides such as β-D-dioxolanylguanine (DXG), β-D-dioxolanyl-2,6- diaminopurine (DAPD), and β-D-dioxolanyl-6-chloropurine (ACP). Interferons that may be used include Schering-Plough' s alpha interferon-2b products, Intron® A and PEG-Intron™; and Hoffman La Roche's Co-Pegasus and PEGASYS (pegylated interferon alfa-2a). Combinations with which the 3-phosphoindoles can be administered include Epzicom (ABC+3TC), Trizivir (ABC + 3TC + AZT), Truvada (FTC +Viread) and Combivir (AZT+3TC).
Examples of protease inhibitors that can be used in combination and/or alternation with the compounds disclosed herein include, but are not limited to indinavir
({l(lS,2R),5(S)}-2,3,5-trideoxy-N-(2,3-dihydro-2-hydroxy-lH-inden-l-yl)-5-[2-[[(l,l- dimethylethyl)amino]carbonyl]-4-(3-pyridinylmethyl)-l-piperazinyl]-2-(phenyhnethyl)- D-erythro-pentoamide sulfate; Merck & Co., Inc.); nelfmavir (Agouron); ritonavir (Abbott Labs), saquinavir (Roche); Amprenavir; Atazanavir; Fosamprenavir; Kaletra; and DMP-450 {[4R-4(r-a,5-a,6-b,7-6)-hexahydro-5,6-bis(hydroxy)-l,3-bis(3- amino)-phenyl]methyl-4,7-bis(phenyhnethyl)-2H-l,3-diazepin-2-one}-bismesylate (Triangle Pharmaceuticals, Inc.). Other compounds that can be administered in combination or alternation with the phenylindole to augment its anti- viral properties include (IS, 4R)-4-[2-amino-6- cyclopropyl-amino-9H-purin-9-yl]-2-cyclopentene-l-methanol succinate (1592U89, a carbovir analog, from GlaxoSmithKline); BILA 1906 (N-{lS-[[[3-[2S-{(l,l- dimethylethyl)amino] carbonyl} -4R-] 3 -pyrindinylmethyl)thio] - 1 -piperidinyl] -2R- hydroxy-lS-phenylmethyl)propyl]-amino]carbonyl]-2-methylpropyl}-2- quinolinecarboxamide) (Bio Mega/Boehringer Ingelheim); BILA 2185 (N-(I5I- dimethylethyl)-l-[2S-[[[2-2,6-dimethyl-phenoxy]-l-oxoethyl]amino]-2R-hydroxy-4- phenylbutyl]4R-pyridinylthio-2-piperidine-carboxamide) (Bio _ Mega/Boehringer Ingelheim); BM+51.0836 (triazoloiso-indolinone derivative) and BMS 186,318
(aminodiol derivative HIV-I protease inhibitor) (Bristol-Myers Squibb); d4API (9-[2,5- dihydro-5-(phosphonomethoxy)-2-furanyl]-adenine) (Gilead); HBY097 (S-4- isopropoxycarbonyl-6-methoxy-3 - [methylthio-methyl] -3 ,4-dihydroquinoxalin-2( 1 H)- thione); HEPT (l-[(2-hydroxy-ethoxy)methyl]6-[phenylthio3-thymine); KM-272 ((2S,3S)-3-amino-2-hydroxy-4-phenylbutyric acid-containing tripeptide); L-697,593 (5- ethyl-6-methyl-3-(2-phthalimido-ethyl)pyridin-2(lH)-one); L-732,524 (hydroxy- aminopentane amide HIV-I, protease inhibitor) (Merck & Co.); L-697,661 (3-{[(-4,7- dichloro-l,3-benzoxazol-2-yl)methyl]amino}-5-ethyl-6-methyl-pyridin-2(lH)-one); L- FDDC ((-)-β-L-5-fluoro-2',3'-dideoxycytidine); L-FDOC ((-)-β-L-5-fluoro-dioxolane cytosine); PFA (phosphonoformate; "foscarnet"; Astra); PMEA (9-(2- phosphonylmethoxyethyl)adenine) (Gilead); PMPA ((R)-9-(2-phosphonylrnethoxy- propyl)-adenine) (Gilead); Ro 31-8959 (hydroxyethylamine derivative HIV-I protease inhibitor) (Roche); RPI-3121 (peptidyl protease inhibitor, l-[(3S)-3-(n-alpha-benzyloxy- carbonyl)- 1 -asparginyl)-amino-2-hydroxy-4-phenylbutyryl] -n-tert-butyl- 1 -proline amide); 2720 (6-chloro-3,3-dimethyl-4-(isopropenyloxycarbonyl)-3,4-dihydro- quinoxalin-2(lH)thione); SC-52151 (hydroxyethylurea isostere protease inhibitor) (G.D. Searle); SC-55389A (hydroxyethyl-urea isostere protease inhibitor (G.D. Searle); TIBO R82150 ((+)-(5S)-4,5,6,7-tetrahydro-5-methyl-6-(3-methyl-2-butenyl)-imidazo-[4,5, 1 - jk]-[1.4]-benzodiazepin-2(lH)-thione) (Janssen Pharmaceuticals); TIBO 82913 ((+)- (5S)-4,5,6,7-tetrahydro-9-chloro-5-methyl-6-(3-methyl-2-butenyl)imidazo[4,5, 1 -Jk]-
[l,4]-benzo-diazepin-2(lH)-thione (Janssen Pharmaceuticals); TSAO-m3T ([2',5'-bis-O- (tert-butyldimethylsilyl)-3'-spiro-5'-(4'-amino-l',2'-oxathiole-2',2'-dioxide)]-β-D- pentofuranosyl-N3 -methyl-thymine) ; U90152 (l-[3-[(l -methylethyl-arnino] 2-pyridinyl] - 4-[[5 — [(methylsulphonyl)-amino]-lH-indol-2-yl]-carbonyl]-piperazine); UC (thio- carboxanilide derivatives) (Uniroyal); UC-781 (N-[4-chloro-3-(3-methyl-2- butenyloxy)phenyl] -2-methyl-3 -furancarbothioamide) ; UC- 82 (N- [4-chloro-3 -(3 -methyl- 2-butenyloxy)phenyl]-2-methyl-3-thiophenecarbothioamide); VB 11,328 (hydroxyethyl- sulphonamide protease inhibitor) (Vertex/Glaxo Wellcome); XM 323 (cyclic urea protease inhibitor) (Dupont Merck); and penciclovir. In yet another embodiment, the indole compound of the invention is administered in combination with the protease inhibitor LG 1350.
The following drugs have been approved by the FDA or are currently or have been in clinical trials for use in the treatment of HIV infection, and therefore in one embodiment, can be used in combination and/or alternation with the compounds of the present invention.
Figure imgf000098_0001
Figure imgf000099_0001
Figure imgf000100_0001
Figure imgf000101_0001
Figure imgf000102_0001
Additional drugs in clinical trials that can be used in combination and/or alternation with the 3-phosphoindoles include:
Figure imgf000103_0001
The following drugs have been approved by the FDA for use in the treatment of complications of HIV infection and AIDS, which can be used in combination and/or alternation with the compounds of the present invention.
Figure imgf000103_0002
Figure imgf000104_0001
Figure imgf000105_0001
Several products have been allowed to proceed as Investigational New Drugs (IND) by the FDA for the treatment of complications of HIV infection and AIDS. Therefore, the following drugs can be used in combination and/or alternation with the compounds of the present invention.
• Trimetrexate glucuronate for the treatment of Pneumocystis carinii pneumonia in AIDS patients who cannot tolerate standard forms of treatment.
• Ganciclovir for the treatment of cytomegalovirus retinitis in AIDS patients.
• Aerosolized pentamidine for the prevention of Pneumocystis carinii pneumonia in ADDS patients.
• Erythropoietin for the treatment of zidovudine-related anemia.
• Atovaquone for the treatment of AIDS patients with Pneumocystis carinii pneumonia who are intolerant or unresponsive to trimethoprim-sulfamethoxazole.
• Rifabutin for prophylaxis against Mycobacterium avium complex bacteremia in AIDS patients. » Vistide intravenous cidofovir for HIV-infected persons with relapsing cytomegalovirus (CMV) retinitis that has progressed despite treatment (Hoffmann-La Roche).
• Serostim, a mammalian derived recombinat human growth hormone, for the treatment of AIDS-related wasting (Serono Laboratories).
In general, during alternation therapy, an effective dosage of each agent is administered serially. During combination therapy, effective dosages of two or more agents are administered together. Dosages administered depend upon factors such as absorption, biodistribution, metabolism and excretion rates for each drug as well as other factors known to those skilled in the art. It is to be noted that dosage amounts will vary with the severity of the condition to be alleviated, the age, weight, and general physical condition of the subject who receives the drug. It is to be understood further that for any particular subject, specific dosage regimens and schedules should be adjusted over time according to the response of the subject to the drug, the needs of the subject, and the professional judgment of the person administering or supervising the administration of the compositions. Examples of suitable dosage ranges for anti-HIV compounds, including nucleoside derivatives such as, for example, D4T, DDI and 3TC, or protease inhibitors like nelfmavir and indinavir, are to be found in the scientific literature and Physicians' Desk Reference. Suggested ranges for effective dosages of the compounds of the present invention are guidelines only, and are not intended to limit the scope or use of the invention.
The disclosed combination and alternation regimen are useful in the treatment and prevention of retroviral infections and other related conditions, such as, for example, AIDS-related complex (ARC), persistent generalized lymphadenopathy (PGL), AIDS- related neurological conditions, anti-HIV antibody position and HIV-positive conditions, Kaposi's sarcoma, thrombocytopenia purpurea, and opportunistic infections. In addition, these compounds or formulations can be used prophylactically to prevent or retard the progression of clinical illness in individuals who are anti-HIV antibody or HFV-antigen positive, or who have been exposed to HIV. VII. Pharmaceutical Compositions
The indole compounds of the present invention can be administered to a subject in need thereof, optionally in combination or alternation with another anti-HIV or anti- retroviral agent, and/or with a pharmaceutically acceptable carrier, diluent or excipient. In one embodiment, a subject infected with HIV may be treated by administering to that subject an effective amount of an indole derivative, a salt, prodrug, stereoisomer or tautomer thereof, in the presence of a pharmaceutically acceptable carrier or diluent. For subjects with multiple drug resistance, the oxo-pyrimidine compound is administered either alone or in combination with one or more other anti-retroviral agents or anti-HIV agents. The active compounds may be administered by any appropriate route, for example, orally, parenterally, enterally, intravenously, intradermally, subcutaneously, percutaneously, transdermally, intranasally, topically or by inhalation therapy, and may be in solid, liquid or vapor form.
The active compound(s) are included within the pharmaceutically acceptable carrier, diluent or excipient in an amount sufficient to deliver to a patient a therapeutically effective amount of the active compound in order to inhibit viral replication in vivo, especially HIV replication, without causing serious toxic effects in a treated subject. By an "inhibitory amount" is meant an amount of active ingredient sufficient to halt viral replication as measured by, for example, an assay such as the ones referred to herein.
One dose of the indole compound for all the conditions mentioned is in the range of from about 0.1 to 100 mg/kg of body weight per day, or from about 1 to 75 mg/kg of body weight per day, and even more typically from about 1 to 20 mg/kg of body weight per day. The effective dosage range of the pharmaceutically acceptable derivatives is calculated based on the weight of the parent indole derivative compound to be delivered.
If the derivative compound itself exhibits activity, then the effective dosage can be estimated as above using the weight of the derivative, or by other means known to those of skill in the art.
The compounds are conveniently administered in units of any suitable dosage form, including but not limited to one containing from about 7 to 3000 mg, or from about
70 to 1400 mg, and even more typically from about 25 to 1000 mg of active ingredient per unit dosage form. For example, an oral dosage of from about 50 to 1000 mg is usually convenient.
Ideally, the active ingredient is administered to achieve peak plasma concentrations of the active compound of from about 0.02 to 70 μM, and typically of from about 0.5 to 10 μM. For example, this can be achieved by intravenous injection of a 0.1 to 25% solution of active ingredient, optionally in saline, or administered as a bolus of active ingredient. It is to be understood that for any particular subject, specific dosage regimens should be adjusted over time to meet individual needs. The concentrations set forth here are exemplary only and are not intended to limit the scope or practice of the claimed composition. The active ingredient may be administered all at once, or may be divided into a number of smaller doses to be administered at varying intervals of time.
One mode of administration of the active compound is oral. Oral compositions usually include an inert diluent or an edible carrier. They may be enclosed in gelatin capsules, compressed into tablets, or delivered in liquid form. For oral therapeutic administration, the active compound may be incorporated with excipients or formulated as solid dispersions or solid solutions, and used in the form of tablets, troches, or capsules. By a "solid dispersion" is meant a solid state comprising at least two components where one component is dispersed more or less evenly throughout the other component. By "solid solution" is meant a solid state comprising at least two components that are chemically and physically integrated to produce a homogeneous product. A solid solution is typical over a solid dispersion because it more easily forms a liquid solution upon contact with an appropriate liquid medium, thereby increasing the bioavailability of a drug. Pharmaceutically compatible binding agents and/or adjuvant materials also may be included as part of this composition.
The tablets, pills, capsules, troches and the like may contain any of the following ingredients or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose; a disintegrating agent such as alginic acid, Primogel, or cornstarch; a lubricant such as magnesium stearate or Sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent like sucrose of saccharin; and a flavoring agent, such as peppermint, methyl salicylate, or orange flavoring. When the dosage unit form is a capsule, it may contain a liquid carrier such as a fatty oil in addition to any material of the kinds given above. In addition, dosage unit forms may contain various other materials that modify the physical form of the dosage unit, such as, for example, coatings of sugar, shellac, or other enteric agents.
The indole compounds may be administered as a component of an elixir, suspension, syrup, wafer, chewing gum or the like. A syrup may contain sucrose as a sweetening agent, preservatives, dyes, colorings, and flavorings in addition to the active compounds.
The active compounds or their pharmaceutically acceptable salts or prodrugs can be mixed with other active materials that do not impair the desired action, or with materials that supplement the desired action, such as antibiotics, antifungals, anti¬ inflammatories, protease inhibitors, or other nucleoside or non-nucleoside antiviral agents. Solutions or suspensions used for parenteral, intradermal, subcutaneous, or topical application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates, or phosphates, and agents for the adjustment of tonicity such as sodium chloride or dextrose. A parenteral preparation normally will include sterile water and may be enclosed in ampoules, disposable syringes, or multiple dose vials made of glass or plastic.
If administered intravenously, typical carriers are physiological saline, phosphate buffered saline (PBS), a glucose solution, or a mixed solution comprising glucose and saline. If administration is percutaneous, such as, for example, through the use of a patch or ointment, the associated carrier may comprise a penetration-enhancing agent and/or a suitable wetting agent which are not harmful to the skin. If inhalation or insufflation is the desired route of administration, then the composition of the present invention includes the compound in the form of a solution, suspension or dry powder that can be delivered through the oral and/or nasal orifices.
Liposomal suspensions, which include liposomes targeted to infected cells with monoclonal antibodies to viral antigens, also are typical as pharmaceutically acceptable carriers. These may be prepared according to methods known to those skilled in the art, for example, as described in U.S. Patent No. 4,522,811, which is incorporated herein by reference in its entirety. For example, liposomal formulations may be prepared by dissolving appropriate lipid(s) such as stearoyl phosphatidyl ethanolamine, stearoyl phosphatidyl choline, arachadoyl phosphatidyl choline, and cholesterol, in an inorganic solvent that later is evaporated, leaving behind a thin film of dried lipid on the surface of the container. An aqueous solution of the active compound, or a salt or prodrug thereof, is then introduced into the container. The container is swirled to free lipid material from its sides and to disperse lipid aggregates, thereby forming the liposomal suspension.
VIII. Processes for Preparing the Active Compounds
General Schemes
Scheme 1:
Figure imgf000111_0001
Scheme 2: Method B
Figure imgf000112_0001
not isolated used crude
Figure imgf000112_0002
Scheme 3:
Figure imgf000113_0001
Scheme 4:
Figure imgf000113_0002
Scheme 5:
Figure imgf000113_0003
Scheme 6:
Figure imgf000114_0001
Scheme 7:
Figure imgf000114_0002
Scheme 8:
Figure imgf000114_0003
Scheme 9:
Figure imgf000115_0001
Scheme 10:
Figure imgf000115_0002
Particularized Schemes
Scheme 11 : N-branched Heterocycles
Figure imgf000116_0001
46
44 45
Scheme 12: Phosphinate Heterocycles
O O
Het-Br Method A Het— P-OEt Method B , Het-P-OEt Meth0d C, HeJ-OEt OEt OH
6l 49
47 48 not isolated used crude
Method H Method K
Figure imgf000116_0002
Figure imgf000116_0004
Figure imgf000116_0003
Scheme 13: Alkyl/Alkenyl/Alkynyl Phosphinates O O O
Method C
AIk-Br Method AA
AIk-P-OMe Method B . AIk-P-OMe AIk-P-OMe OMe OH 6l 55
53 54 not isolated used crude
Figure imgf000117_0001
Scheme 14: Alkyl Phosphinates
O
AnJLn Method D AIk-P-OMe
Cl 1 eq. MeOH Cl
58 55
Scheme 15: Alkyl Methyl Carboxamides
Method J
Figure imgf000117_0002
Figure imgf000117_0003
Scheme 16: Phosphinic Acids
Method AC
Figure imgf000117_0004
Figure imgf000117_0005
6 or 7 Methods
The following are descriptions of the methodologies used in the foregoing schemes. Numbers in bold print correspond to numbered compounds found in Schemes 1-16. Methods A, B, C, D, F, L, N, T, U and AA are known to those skilled in the art of making this general class of compounds, but have been adapted for preparing the specific compounds of the present invention. This also is true for Methods H, I, J, M, P, Q, Y, Z, AB and AC. Methods E, G, K, O, R, S, W, V and X are new processes for preparing the compounds of the invention.
METHOD A Bromobenzene 1 (1 eq) was stirred with diethylphosphite (1.1 eq.) and triethylamine (1.1 eq.) in a pressure tube and the mixture was degased with N2 for about 15 minutes. Then palladium tetrakis (0.05 eq.) was added and the mixture was stirred under pressure at about 85°C overnight. The next morning, Et2O was added to the reaction and the mixture was filtered. The filtrate was evaporated and the crude was purified by chromatography on silica gel (eluent: petroleum ether/EtOAc from 8/2 to
6/4). Alternatively, the reaction can be performed using 10 volumes of 1,2- diethoxyethane under the same conditions.
METHOD B
AU the reagents (1 eq. of diethylphosphonate 2 or dimethylphosphonate 19, 6 eq. of sodium hydroxide and ethanol or methanol (3ml/mmol)) were stirred at room temperature for about 5 hours. Then, ethanol (or methanol) was evaporated in vacuo and the mixture acidified with HCl 2.5N to reach pH = 1. Then the mixture was saturated with NaCl and extracted with ethyl acetate. Combined organic layers were dried over Na2SO4, filtered and concentrated to give the desired products 3 or 20. METHOD C
The compound 20 or 3 (1.2 eq.) was stirred at room temperature under N2 with thionyl chloride (3.6 eq.), dichloromethane (5ml/mmol) and a few drops of dimethylformamide for about 5 hours. An aliquot was taken and put in dry methanol and Et3N to monitor the reaction. When the reaction was complete, dichloromethane and thionyl chloride, co-evaporated with toluene, were evaporated in vacuo to give an oil which was stored under N2.
METHOD D
To a solution of arylphosphonyl dichloride 21 (1 mL, 6.35 mmol) in anhydrous dichloromethane (25 mL) ethanol or methanol (1,12 mL, 19.04 mmol) was added dropwise at about 00C followed by the addition of triethylamine (2,65 mL, 19.04 mmol).
The reaction mixture was stirred at room temperature for about 2 hours, and then was washed with a solution of HCl IN (50 mL). The aqueous layer was extracted with dichoromethane. Combined organic phases were dried and concentrated under reduced pressure. The crude oil was purified by chromatography on silica gel (eluent:
C6H12ZEtOAc 6/4) to afford dimethylarylphosphonate 19 or diethylarylphenylphosphonate 2.
METHOD E
The diethylarylphosphonate 2 (1 eq.) was stirred under N2 at room temperature with bromotrimethylsilane (10 eq.) and dichloromethane (5ml/mmol) for about 5 hours.
Then the bromotrimethylsilane and dichloromethane were evaporated to give a yellow oil. Next, oxalyl chloride, dimethylformamide and dichloromethane were added under N2 and the mixture was stirred at room temperature overnight, following which the oxalyl chloride and solvents were evaporated. The oil in dichloromethane solution was stirred and allowed to cool to about O0C, and ethanol or methanol (1,12 mL, 19.04 mmol) was added under N2, followed by the dropwise addition of triethylamine. The mixture then was warmed to room temperature. The solvent was evaporated and the resulting product was purified by chromatography on silica gel (eluent: CH2Cl2/Et0Ac : 8/2) to afford the compound 14. METHOD F
To a stirred solution of diethylarylphosphonate 2 (1 eq.) in dichloromethane (5ml/mmol) was added bromotrimethylsilane (5 eq.) at room temperature under N2. After about 3 hours of stirring at room temperature, the solvent was evaporated and dichloromethane (5ml/mmol), a few drops of dimethylformamide and oxalyl chloride (2,5 eq) were added. This mixture was stirred at room temperature overnight, and the solvent was evaporated to give an oil. A solution of the oil (1.2 eq.) in diethyl ether (3ml/rnmol) was stirred and cooled to about -17°C, after which anhydrous methanol (1.2 eq.) was added dropwise to remove the double addition components and triethylamine. The resulting mixture was warmed to room temperature, stirred for about 1 hour, and then filtered on autocup under N2 to remove triethylamine salts. The solvent was evaporated to give the compound 15.
METHOD G
«-BuLi (2.5M in hexane, 1.2 eq.) was added dropwise to a stirred and cooled
(to about -90 °C) solution of bromoindole 11 (1 eq.) in anhydrous THF (lOml/mrnol) under N2. After keeping the solution at about -900C for about 5 minutes, an appropriate chorophosphosphorus reagent 15, 4 or diphenylphosphonic chloride (1.2 eq.) was added dropwise to the solution at the same temperature. The reaction was allowed to warm slowly to about -40°C (TLC monitoring, eluent CH2Cl2/ ETOAc 9/1). Water then was added. Extraction with ethyl acetate, drying and evaporation led to a crude oil that was purified by chromatography on silica gel to give the compound 16, 5 or 26. METHOD H
A stirred and cooled (to about 0°C) solution of compound 5, 16, 26, 28 or 30 in methanol in a pressure tube was saturated with NH3 gas for about 10 minutes. Then the mixture was stirred at about 50° C overnight, and after TLC monitoring, excess ammonia and methanol were evaporated in vacuo and the crude purified by chromatography on silica gel to give the carboxamide 6, 7, 27, 29 or 31.
METHOD I
Lithium hydroxide (14 eq.) was added to a stirred solution of compound 16 or 5 in tetrahydrofuran (20ml/mmol) and water (20ml/mmol). This mixture next was stirred at room temperature and monitored by TLC. If necessary, equivalents of lithium hydroxide were added until the reaction was complete. Then the THF was evaporated and HCL
(IN) was added to reach pH 1. The aqueous layer was extracted with ethyl acetate and combined organic phases were dried, filtered and concentrated under reduced pressure to give the compound 17 or 8.
METHOD J The compound 17 or 8 (1 eq.) was stirred with dichloromethane (20ml/mmol) or DMF, and 1-hydroxybenzotriazole (1 eq.) was added followed by the addition of EDCI (1 eq.) followed by the amine (leq) were added. This mixture was stirred at room temperature overnight. The next morning the mixture was washed with water (to a pH of 5-6), and organic layers were dried, filtered and concentrated under reduced pressure.
The resulting oil was purified by chromatography on silica gel (eluent: CH2Cl2/Et0Ac) to give a powder 18 or 9.
METHOD K
In a microwave tube, the compound 5 or 6 (leq.) was stirred with DMF (5ml/mmol), and TMSBr (5 eq.) was added. The tube was heated under microwave irradiations under pressure at about 60 0C (maximum power input 10OW, CEM discover apparatus) for about 50 minutes. DMF was evaporated in vacuo, and the mixture was put in a pressure tube. Trimethylphosphite (4ml/mmol) was added and the mixture was stirred and heated at about 90 °C overnight. The mixture then was cooled in an ice water bath and HCl (IN) was added dropwise. The mixture was extracted with ethyl acetate, and combined organic layers were washed with HCl (IN) until no HP(OMe)2 remained. Next it was dried, filtered and concentrated under reduce pressure to provide an oil, and the resulting oil was purified by chromatography on silica gel to give the compound 16 or 7. METHOD L
To a stirred and cooled (to about 0°C) solution of ethyl indole-2-carboxylate 10 (1 eq.) in DMF (2ml/mmol) under N2, was added NaH (60% in oil, 1.2 eq.) portionwise. When gas evolution stopped, benzenesulfonyl chloride (1.2 eq.) was added. The reaction mixture was stirred for about 1 hour (TLC monitoring, eluent dichloromethane); a small amount of water then was added carefully and the DMF was evaporated. The crude residue was dissolved in ethyl acetate and washed with water and brine. After drying and evaporation of the solvents, the compound was purified by chromatography on silica gel (eluent: C6H12/Et0Ac 9/1 to 7/3) to give the ethyl l-phenylsulfonylindole-2- carboxylate.
To a stirred solution of ethyl l-phenylsulfonylindole-2-carboxylate (1 eq.) in
DMF (2.5ml/mmol) under N2, was added a solution of bromine (4 eq.) in DMF (0.5ml/mmol). This reaction mixture was stirred at room temperature for about 4 hours, following which water was added and the mixture was extracted with dichloromethane (x3). The organic layer was washed with a saturated solution of Na2SO5, dried and evaporated to give a crude yellow oil. Purification by chromatography on silica gel (eluent: C6H12ZEtOAc 9/1) afforded 3-brominated indole 11.
METHOD M
A mixture of 5 or 16 (1 eq.), acrylonitrile (10 eq.), palladium acetate (20% mol.), triethylamine (1 eq.) and tri-ort/jo-tolylphosphine (1 eq.) in degassed acetonitrile (30mL/mmol) was stirred and heated under microwave irradiation in a pressure tube for about 45 minutes. Next water was added and the aqueous layer was extracted with dichloromethane. The combined organic layers were dried and concentrated and purified by chromatography on silica gel (eluent: Petroleum ether/EtOAc 8/2) to give the compound 32 as a mixture of enantiomers E and Z (which are separated by preparative HPLC). METHOD N
Bromobenzene 1 (1 eq.), dimethylformamide (lml/mmol), triethylamine (3 eq.) and anilinium salt* (1.25 eq.) were put in a pressure tube and degassed with N2 for about 15 minutes. Then palladium tetrakis was added and this mixture was stirred at about 85°C overnight, following which the solvent was evaporated and water was added to achieve a pH of about 5-6. The mixture was basified with NaHCO3 until a pH 8 was reached, and then extracted with diethyl ether. The aqueous layer was acidified with HCl (IN) to reach a pH = 1, and extracted with ethyl acetate. Combined organic layers were dried, filtered and concentrated under reduced pressure to give the compound 14.
Anilinium salt was synthesised according to the procedure of Montchamp et al (J. Am. Chem. Soc, 2001, 123, 510-511).
METHOD O
Tetramethylorthosilicate (1.2 eq.) was added to a stirred solution of compound 14
(1.2 eq.) in toluene (4ml/mmol) under N2. This mixture was heated at reflux for about
1.5 hours, and then cooled to room temperature. Next the mixture was degased with N2 before adding triethylamine (3.3 eq.), bromoindole 11 (1 eq.), and palladium tetrakis
(0.05 eq.). The mixture was stirred at about 100°C over a week-end (about 48 hours), after which the reaction was cooled to room temperature and water was added to achieve a pH of about 8-9. The mixture next was extracted with ethyl acetate and the organic phases were washed with KHSO4 (IN) saturated with NaCl. It then was dried, filtered and concentrated under reduced pressure to give an oil, which was purified by chromatography on silica gel (eluent: CH2Cl2ZMeOH) to give the compound 12.
METHOD P
To a stirred solution of compound 12 (1 eq.) in methanol (25ml/mmol) was added trimethylsilyldiazomethane (13 eq.) in a several fractions at room temperature under N2. This mixture was stirred overnight, after which water was added and the methanol was evaporated under reduce pressure. Then NaHCO3 was added to reach pH 8, and the mixture was extracted with ethyl acetate. The organic phases were dried, filtered and concentrated under reduced pressure. The crude was purified by chromatography on silica gel (eluent: CeH12/EtOAc) to give the compound 16.
METHOD O The compound 18 was dissolved in chloroform (or in CH2Cl2) under stirring at room temperature; m-chloroperoxybenzoic acid was added and the reaction allowed to stir overnight (about 15 hours). Then the mixture was diluted with dichloromethane and extracted with a mixture of saturated K2CO3 / H2O (1/3). The aqueous layer was extracted three times with dichloromethane. The combined organic layers were dried over Na2SO4, filtered and concentrated under reduced pressure (TLC: dichloromethane/methanol = 9/1). The crude product was purified by chromatography on silica gel to give the compound 22.
METHOD R hi a microwaves sealed tube, the compounds 11 (1 eq.) and 23 (1.1 eq.) were added to toluene (8ml/mmol) and triethylarnine (3.3 eq.) and degased with N2 for about
10 minutes. Then Pd(PPh3)2 was added and the tube was heated under microwave irradiation under pressure at about 1200C (maximum power input 200W, CEM discover apparatus) for about 30 minutes. The reaction was monitored by TLC and the tube was heated for about 30 minutes if necessary. The solvent was evaporated and the crude product was purified by chromatography on silica gel (eluent: C6H12ZEtOAc : 8/2) to give the compound 5. METHOD S
In a microwave sealed tube, the compounds 11 (1 eq.) and 24 (2 eq.) were added to toluene (8ml/mmol) and then degased with N2 over about a 10 minute time period.
Next, Pd(OAc)2 in about 20% solution was added, and the tube was under microwave irradiation under pressure at about 150 0C (maximum power input 200W, CEM discover apparatus) for about 45 minutes. The reaction was monitored by TLC and if any starting material was present, the tube was heated for about another 45 minutes at approximately
17O0C. Next HCl (IN) (8ml/mmol) was added and the solution was extracted with ethyl acetate. The combined organic layers were dried, filtered and concentrated under reduced pressure. The crude product was purified by chromatography on silica gel
(eluent: petroleum ether/EtOAc : 1/1) to give the compound 5 or 16.
Alternatively the reaction is carried out at about 1500C in xylene for about 5 hours.
METHOD T Pyridine (1 eq.) was carefully added to a vigorously stirred solution of alkyl chloroformate (1 eq.) and arylphosphinic acid (1 eq.) in dichloromethane (2ml/mmol) at room temperature. Once effervescence stopped, the solution was refluxed for about 15 minutes and then allowed to cool to room temperature. The solution was poured into 0.1M hydrochloric acid (lml/mmol) and the organic layer was separated. After washing with water and drying over Na2SO4, the solvent was removed in vacuo to give the compound 23.
METHOD U
Alkyl or aryl bromide (0.15 mol) was added drop wise to a mixture of magnesium (3.6 g) and dry tetrahydrofuran (40 mL) under N2 atmosphere at about 5O0C. After this addition, the reaction mixture was stirred at about 500C. for an additional 1—2 hours to allow the reaction to run to completion. Then the mixture was added dropwise to a solution of triethyl phosphite (0.1 mol) and THF (25 mL) at between 40 0C. to 50 0C. in N2 atmosphere, and stirred for about 3 hours at about 500C.
After removal of the solvent under reduced pressure, the crude product was distilled from the semisolid residue in vacuo to give the compound 24.
METHOD V The compound 6 or 7 (1 eq.) and Lawesson's reagent (4 eq.) were heated in toluene (lOml/mmol) at about 90 °C. under N2 in a pressure tube. The reaction was monitored by TLC and heating continued until no starting material remained (about 5.5 hours). The crude solution was filtered and the filtrate was evaporated to dryness and purified by chromatography on silica gel to give the compound 25 and the compound
25'.
METHOD W
To a stirred and cooled (to about -90 0C.) solution of bromoindole 11 (1 eq.) was added dropwise n-butyllithium (1.2 eq.) under N2. After about 10 minutes, benzenephosphonyl dichloride 21 (1.1 eq.) in tetrahydrofuran (15ml/mmol) was added dropwise at a temperature of about -70 0C, and the temperature then was raised to about
-90 °C. and maintained for about 15 minutes. This was followed by the addition of methylmagnesium bromide (1.1 eq.), and the mixture was allowed to warm to about
-400C for about 1 hour, after which it was quenched with water and extracted with ethyl acetate then dichloromethane. The organic layer was dried over Na2SO4, filtered and concentrated under reduced pressure. The crude product was purified by chromatography on silica gel (eluent: CH2Cl2/Ac0Et : 9/1 to 7/3) to give the compound 28.
METHOD X
Dimethylamine (1 eq.) waas added dropwise to a stirred and cooled (to about -55 °C.) solution of phenyl phosphonic dichloride 21 (1 eq.) in diethyl ether (1.5ml/mmol).
Then triethylamine was added (1 eq.) and the mixture was allowed to warm to room temperature. The mixture next was filtered and the filtrate was evaporated to give an oil/mixture. To rid the oil/mixture of phenyl phosphonic dichloride, the oil/mixture was dissolved in EtOAc and washed twice with an HCl solution of pH 4-5. The organic phases were dried with Na2SO4, filtered and concentrated under reduced pressure to give an oil. Then, n-butyllithium (1.2 eq.) was added dropwise to a stirred and cooled (to about -80 0C.) solution of bromoindole 11 (1 eq.) in tetrahydrofuran (5ml/mmol) under
N2. At the end of the addition, the mixture was warmed to about -60 0C. and tetrahydrofuran (1.2 eq.; 3ml/mmol) was added dropwise to the the oil. Then the mixture was warmed slowly to about -10 0C, and the reaction was quenched with water
(8ml/mmol). HCl (IN) was added to achieve a pH of about 5, and the solvent was evaporated in vacuo. The aqueous phase was extracted with ethyl acetate and the combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated under reduced pressure. The crude product was purified by chromatography on silica gel (eluent: C6H12/Et0Ac : 9/1 to 0/10) to give the compound 30.
METHOD Y Suzuki cross-coupling
Pd(PPh3)4 (10% mol.), a solution OfNa2CO3 (6 eq.) in H2O (2M) and a solution of aryl, alkyl or heteroaryl boronic acid (2 eq.) in EtOH (0.3M) were added to a stirred solution of 5 or 16 (bearing an halogeno or inflate substituent) in degassed toluene under N2. The reaction tube was then heated under microwave irradiation at about 110 °C. until no starting material remained. Water was added and the reaction media was extracted with EtOAc, dried and concentrated. The crude residue was purified by column chromatography on silica gel (PE/EtOAc 8/2) to afford 37.
METHOD Z Under a nitrogen atmosphere, a dry reaction tube was charged with cuprous oxide
(10%mol), a ligand (20%mol), a nucleophile (1.5 eq.), cesium carbonate (2 eq.) and the aryl halide 5 or 16 (bearing an halogeno or triflate substituent) (1 eq.), followed by the addition of anhydrous and degassed acetonitrile (0.6 mL per mniol of aryl halide). The tube was sealed and stirring was applied at about 80 0C. until the reaction ran to completion. The reaction mixture then was cooled to room temperature, diluted with tert- butylmethyl ether and filtered through a plug of celite, the filter cake being further washed with butylmethyl ether. The filtrate was concentrated in vacuo to remove the acetonitrile and was redissolved in tert-butylmethyl ether. This organic layer filtrate was washed twice with water and once with brine before being dried on Na2SO4 and filtered. The solvent was removed in vacuo to yield the crude product, which was purified by chromatography on silica gel to give the compound 42.
METHOD AA
Alkyl halide was heated at about 90 °C. overnight in trimethylphosphite
(lOmL/mmol). The reaction media was cooled to about 0°C in an ice bath, and a solution of HCl (IN) was added carefully. The aqueous layer was extracted with EtOAc. The combined organic layers were washed with HCl (IN) and with water, and then dried and concentrated to give compound 53 as a colorless oil.
METHOD AB
In a microwaves sealed tube, the compounds 17 (1 eq.) , formaldehyde (37% wt in water, 1 eq.), and morpholine (1 eq.) were added to t-butyl alcohol (4ml/mmol). The tube was heated under microwave irradiation under pressure at about 170 0C (maximum power input 200W5 CEM discover apparatus) for about 60 minutes. The reaction was monitored by TLC and if any starting material remained, the tube was heated for about
45 minutes at approximately 170 °C. The solvents were evaporated in vacuo, and the crude residue was purified by chromatography on silica gel(MeOH/EtOAc : 2/98) to give the compound 18.
METHOD AC
Li a microwave tube, the compound 5 or 6 (1 eq.) was stirred with DMF
(5ml/mmol), and TMSBr (5 eq.) was added. The tube was heated under microwave irradiation under pressure at about 60 °C. (maximum power input 10OW, CEM discover apparatus) for about 50 minutes. After cooling, water was added and compound 60 was collected by filtration.
IX. Representative Examples of the Active Compounds
Tables 1 and 2 contain a non-limiting list of representative compounds that may be prepared by the methods and according to Schemes 1-16 provided above.
Table 1
Compound structure Description
2a
Figure imgf000127_0001
Colorless oil, 1H NMR (CDCl3, 250 MHz) δ 1,33 (t, J = 7.1 Hz,
6H), 4.05-4.25 (m, 4H), 7.46-7.57 (m, 3H), 7.78-7.87 (m, 2H) ,31P NMR (CDCl3, 101.256 MHz) δ 19.3. Compound structure Description
MHz) δ 1.26 (t, J = 7.5 Hz,
Figure imgf000128_0001
J= 7.5 Hz, 2H), 4.04-4.19 (ω, 4H), 7.37-7.4 (m, 2H), 7.58-7.69 (m, 2H), 31P NMR (CDCl3, 121.49 MHz) 5 19.53.
(t, J= 7.05 Hz,
2d
Figure imgf000128_0002
Hz, IH), 7.8- 7.83 (m, IH), 7.97-8.1 (m, 2H), 31P NMR (CDCl3, 121.49 MHz) δ 16.4, 19F NMR (CDCl3, 282.4 MHz) δ 9.27 (s, 3F), MS (ESI, EΫ) m/z = 283 (MH+).
White solid, 1H NMR (CDCl3, 300 MHz) δ 1.36 (t, J= 7.05 Hz,
2e
Figure imgf000128_0003
6H), 4.09-4.25 (m, 4H), 7.62 (td, J = 3.9 and 7.8 Hz, IH), 7.83- 7.85 (m, IH), 8.02-8.12 (m, 2H), 31P NMR (CDCl3, 121.49 MHz) δ 15.12, MS (ESI, El+) m/z = 240 (MH+).
Colorless oil, 1H NMR (CDCl3, 300 MHz) δ 1.34 (t, J= 7.05 Hz,
2f
Figure imgf000128_0004
6H), 4.05-4.21 (m, 4H), 7.32-7.38 (m, IH), 7.66-7.71 (m, IH), 7.73-7.78 (m, IH), 7.92-7.97 (m, IH), 31P NMR (CDCl3, 121.49 MHz) δ 16.37, MS (ES+) m/z = 292.9/294.8 (MH).
Colorless oil, 1H NMR (CDCl3, 300 MHz) δ 1.35 (t, J= 7.05 Hz,
2g 6H), 4.04-4.23 (m, 4H), 7.53 (t, J= 1.8 Hz, IH), 7.65 (d, J= 1.8
Figure imgf000128_0005
Hz, IH), 7.7 (d, J= 1.8 Hz, IH), 31P NMR (CDCl3, 121.49 MHz) δ 14.74.
Colorless oil, 1H NMR (CDCl3, 300 MHz) δ 1.34 (t, J= 7.05 Hz, 6H), 4.06-4.22 (m, 4H), 5.44 (d, J= 47.4 Hz, 2H), 7.51-7.6 (m,
Figure imgf000128_0006
2H), 7.78-7.85 (m, 2H), 19F NMR (CDCl3, 282.40 MHz) δ - 209.67 (t, J= 47.4 Hz, IF), MS (ES+) m/z = 247 (MH). Compound structure Description
Colorless oil, 1H NMR (CDCl3, 300 MHz) δ 1.34 (t, J = 7.05 Hz,
2i 6H), 4.04-4.2 (m, 8H), 5.86 (s, IH), 7.48-7.54 (m, IH), 7.68-7.7
Figure imgf000129_0001
(m, IH), 7.8-7.87 (m, IH), 7.93-7.98 (m, IH), 31P NMR (CDCl3, 121.49 MHz) δ 18.45, MS (ES+) m/z = 287 (MH).
Colorless oil, 1H NMR (CDCl3, 300 MHz) δ 1.35 (t, J = 7.05 Hz,
2J
Figure imgf000129_0002
6H), 2.39 (s, 3H), 4.05-4.21 (m, 4H), 7.34-7.36 (m, IH), 7.5-7.6
(m, 2H), 31P NMR (CDCl3, 121.49 MHz) δ 17.18, MS (ES+) m/z = 263.2 (MH).
J= 7.05 Hz,
2k (ro, 2H),
Figure imgf000129_0003
δ 18.84, MS
(ES+) m/z = 291.17 (MH).
(t, J= 7.05 Hz,
21 2H), 7.38-7.60 (m,
Figure imgf000129_0004
MHz) δ 17.99, MS (ESI, El+) m/z = 287 (MH+). δ 1.32 (t, J = 6.9 Hz, (s, 3H), 4.03-4.16 (m, 4H), 31P NMR (CDCl3, 121.49
Figure imgf000129_0005
MHz) δ 19.21, MS (ESI, El+) m/z = 245.13 (MH+). (t, J = 7.05 (m, q MHz) δ
Figure imgf000129_0006
Slight yellow oil, 1H NMR (CDCl3, 400 MHz) δ 1.36 (t, J = 7.07 Hz, 6H), 4.08-4.26 (m, 4H), 7.49-7.52 (m, IH), 7.67-7.73 (m, IH), 7.85-7.89 (m, IH), 31P NMR (CDCl3, 101.256 MHz) δ 14.73
Figure imgf000129_0007
Compound structure Description
(d, J= 7.8 Hz, IP), MS (ESI, El4) m/z = 301 (MH+). 300 MHz) δ 1.24 (d, J = 6.9 6H), 3.07 (heptuplet, J = 6.9 Hz, ,
2t (m, 2H), 8.04 (brs, IH), 31P δ 15.12, MS (ESI, El+) m/z =
Figure imgf000130_0001
δ 1.35 (t, J = 7.05 IH), 7.37-7.45 (m, 121.49 MHz)
Figure imgf000130_0002
= 267.11 (MH+). J= 7.5 Hz, Hz, 2H),
2v 2H), 7.58- MS (ES+)
Figure imgf000130_0003
(m, (m,
2w (m, 3H),
Figure imgf000130_0004
m/z = 255.06 (MH+). MHz) δ 1.23 (t, J= 7.05 7.49-7.57 (m, 2H), 7.69 MHz) δ 15.74, MS (ES+)
Figure imgf000130_0005
Colorless oil, 1H NMR tø-DMSO, 300 MHz) δ 1.23 (t, J= 7.05 Hz, 6H), 2.34 (s, 3H), 2.63 (q, J = 7.05 Hz, 2H), 3.94-4.05 (m, 4H), 7.29-7.31 (m, 2H), 7.44-7.45 (m, IH), 31P NMR (J6-DMSO,
Figure imgf000130_0006
121.49 MHz) δ 18.71, MS (ES+) m/z = 257.4 (MH+). 1H NMR (Jn-DMSO, 300 MHz) δ 0.88 (t, J = 7.2 (t, J = 7.05 Hz, 6H), 1.54-1.62 (m, 2H), 2.34 (s,
2z (m, 2H), 3.94-4.05 (m, 4H), 7.27-7.36 (m, 3H), 31P SO, 121.49 MHz) δ 18.73, MS (ES+) m/z = 271.36
Figure imgf000130_0007
Compound structure Description
Colorless oil, 1H NMR (CDCl3, 300 MHz) δ 1.28 (d, J = 7.0 Hz, 6H), 1.34 (t, J= 7.2 Hz, 6H), 2.97 (m, IH), 4.13 (m, 4H), 7.36- 7.45 (m, 2H), 7.58-7.75 (m, 2H), MS (ESI, El+) m/z = 257 (MH+).
Brown oil, 1H NMR ((I6-DMSO, 300 MHz) δ 1,22 (t, J = 7.2 Hz, 6H), 2.37 (s, 3H), 3.92-4.05 (m, 4H), 7.33-7.37 (m, 2H), 7.57-
Figure imgf000131_0001
7.64 (m, 2H), 31P NMR ((I6-DMSO, 101.256 MHz) δ 18.62. 1,22 (t, J = 13 Hz, (m, IH), 7.4-7.49 δ 19.86, MS (ESI,
Figure imgf000131_0002
Yellow oil, MS (ESI, El+) m/z = 243 (MH+).
Figure imgf000131_0003
300 MHz) δ 1,24 (t, J = 7.05 3H), 3.96-4.06 (m, 4H), 7.22 (td, J J= 7.5 Hz, IH), 7.62 (dd, J= 7.5
Figure imgf000131_0004
, 121.49 MHz) δ 20.29. = 7.05 Hz, 6H), (m, 2H), 19F
Figure imgf000131_0005
(CDCl3, 101.256 MHz) δ 18.33.
Pale yellow oil, 1H NMR (CDCl3, 300 MHz) δ 1.36 (t, J = 6.9 Hz, 6H), 4.08-4.23 (m, 4H), 6.96-7.04 (m, IH), 7.30-7.38 (m,
2ai 2H), 31P NMR (CDCl3, 101 MHz) δ 14.77-15.3 (m, IP)5 19F NMR
Figure imgf000131_0006
(CDCl3, 235.36 MHz) δ -107.56 (m, 2F), MS (ESI, El+) m/z = 251 (MH+). 7.5 Hz, 3.97-4.07
2ao (ddd, J= 1.2
Figure imgf000131_0007
MHz) δ 18.71, MS (ESI, El+) m/z = 243 (MH+). Compound structure Description (t, J = 7.05 (m, 3H), MHz) δ 17.71;
Figure imgf000132_0001
δ 1.22 (t, J= 7.05 Hz, 6H), 4.61 (s, 2H), 7.28-7.37 (m, 5H), , 121.49 MHz) δ 17.97;
Figure imgf000132_0002
(t, J = 7.2 (m, 2H), m/z =
Figure imgf000132_0003
(t, /= 7.05 Hz, 6H), (m, 3H), 31P NMR (J6- m/z = 254 (MH+)-
Figure imgf000132_0004
/= 7.2 Hz, (m, IH), δ 13.5 (d,
Figure imgf000132_0005
-108.43 (m, IF).
NC Orange oil, 1H NMR (J6-DMSO, 300 MHz) δ 1.25 (t, J= 7.2 Hz,
O
Il 6H), 4.04-4.13 (m, 4H), 7.82-7.90 (m, IH), 7.96-8.01 (m, IH),
2au \\ /r OprEt0Ei 8.15-8.18 (m, IH), 31P NMR (J6-DMSO, 121.49 MHz) δ 12.83 (d, J= 8.02 Hz, IP), 19F NMR (J6-DMSO, 282.4 MHz) - 108.7 (q, J= 8.2 Hz, IF),. MS (ES+) m/z = 258.2 (MH+).
Oil, 1H NMR (J6-DMSO, 300 MHz) δ 1.25 (t, J= 7.05 Hz, 6H), 3.97-4.13 (m, 4H), 7.46-7.53 (m, IH), 7.57-7.66 (m, 2H), 7.84-
Figure imgf000132_0006
7.96 (m, IH), 31P NMR (J6-DMSO, 121.49 MHz) δ 13.64, MS (ESI, El+) m/z = 249.05 (MH+).
Colorless oil, 1H NMR (CDCl3, 300 MHz) δ 1,32 (t, J = 7.3 Hz,
3a 3H), 4.08 (q, J = 13 Hz, 2H), 7.42-7.56 (m, 3H), 7.79-7.86 (m,
Figure imgf000132_0007
2H), 10.67 (brs, IH), 31P NMR (CDCl3, 101.256 MHz) δ 21.3. Compound structure Description
Colorless oil, 1H NMR (CDCl3, 300 MHz) δ 1,31 (t, J= 7.05 Hz, 3H), 2.32 (s, 6H), 4.03-4.15 (m, 2H), 7.15 (s, IH), 7.42 (d, J =
3b
Figure imgf000133_0001
13.8 Hz, 2H), 9.64 (brs, IH), 31P NMR (CDCl3, 101.256 MHz) δ 22.36, MS (ESI, El+) m/z = 215 (MH+). 7.05 Hz, (m, 2H),
Figure imgf000133_0002
(m, IF), 31P NMR (CDCl3, 101.256 MHz) δ 20, MS (ESI, El+) m/z = (MH+). J= 7.05 Hz, IH), 7.78-
3d
Figure imgf000133_0003
MHz) δ 18.09, 19F NMR (CDCl3, 282.4 MHz) δ -66.03, MS (ESI, El+) m/z = 255 (MH+).
Colorless oil, 1H NMR (CDCl3, 300 MHz) δ 1.33 (t, J= 7.05 Hz, 3H), 4.04 (quintuplet, J = 7.05 Hz, 2H), 7.27-8.09 (m, 5H), 31P
Figure imgf000133_0004
NMR (CDCl3, 101.256 MHz) δ 16.59, MS (ESI, El+) m/z = 212 (MH+).
Colorless oil, 1H NMR (CDCl3, 300 MHz) δ 1.33 (t, J= 7.05 Hz, 3H), 4.1 (quintuplet, J= 7.05 Hz, 2H), 7.51 (t, J= 1.8 Hz, IH),
3f 7.62 (d, J = 1.8 Hz, IH), 7.67 (d, J = 1.8 Hz, IH), 11.63 (brs,
Figure imgf000133_0005
IH), 31P NMR (CDCl3, 121.49 MHz) δ 15.74, MS (ESI, El+) m/z = 255 (MH+). (t, J = 7.5 Hz, 2H), 4.1
3g (m,
Figure imgf000133_0006
MHz) δ 21.63, MS (ESI, El+) m/z = 215 (MH+)
Colorless oil, 1H NMR (CDCl3, 300 MHz) δ 1.25 (d, J= 6.9 Hz,
O 6H), 1.31 (t, J = 7.05 Hz, 3H), 2.88-2.98 (m, IH), 4.03-4.13 (m,
Il
3h P-OEt 2H), 7.35-7.41 (m, 2H), 7.60-7.69 (m, 2H), 11.26 (brs, IH), 31P OH NMR (CDCl3, 121.49 MHz) δ 21.85, MS (ESI, El+) m/z = 229 (MH+) Compound structure Description
Colorless oil, 1H NMR (CDCl3, 300 MHz) δ 1.32 (d, J = 7.0 Hz, 6H), 2.35 (s, 3H), 4.04-4.14 (m, 2H), 7.32 (s, IH), 7.45-7.59 (m, 2H), 8.28 (brs, IH), 31P NMR (CDCl3, 121.49 MHz) δ 19.16.
Figure imgf000134_0001
J= 7.0 Hz, (m,
Figure imgf000134_0002
NMR (CDCl3, 121.49 MHz) δ 21.13, MS (ESI, El+) m/z = 263 (MH+). J= 7.0 Hz, 7.05-7.09 (m, 121.49
Figure imgf000134_0003
MHz) δ 20.36. MHz) δ 1.36 (t, J = 7.2 8.06 (brs, IH), 8.22 31P NMR (CDCl3,
Figure imgf000134_0004
= 322.99 (MH+).
Light orange oil, 1H NMR (CDCl3, 300 MHz) δ 1.34 (t, J= 7.05 Hz, 3H), 2.4 (s, 3H), 4.11 (quintuplet, J = 7.05 Hz, 2H), 7.04-
3q 7.08 (m, IH), 7.27-7.35 (m, IH), 7.4-7.43 (m, IH), 11.37 (brs,
Figure imgf000134_0005
IH), 31P NMR (CDCl3, 121.49 MHz) δ 19.37 (d, J= 9.6 Hz, IP), MS (ESI, El+) m/z = 219 (MH+).
Yellowish oil, 1H NMR (d6-DMSO, 300 MHz) δ 1.18 (t, J = 7.1 Hz, 3H), 3.87-3.97 (m, 2H), 7.55-7.67 (m, 3H), 7.98-8.18 (m, 3H), 8.53 (d, J = 8.4 Hz, IH), 31P NMR (dg-DMSO, 300 MHz) δ 14.76, MS (ESI, El+) m/z = 237 (MH+).
Yellowish oil, 1H NMR (CDCl3, 300 MHz) δ 1.25 (d, J= 6.9 Hz, 6H), 1.31 (t, J = 6.9 Hz, 3H), 2.70-3.00 (m, IH), 4.00-4.20 (m, 2H), 7.89 (d, J = 5.4 Hz, IH), 7.25-7.36 (m, IH), 7.47 (d, J = 13.5 Hz, IH), 12.22 (brs, IH) 31P NMR (CDCl3, 121.49 MHz) δ
Figure imgf000134_0006
17.93, 19F NMR (CDCl3, 282.40 MHz) δ -112.35 (m, IF). Compound structure Description J= 6.9 Hz, (m,
3t 31P NMR 254.3 (MH+).
Figure imgf000135_0001
J = 7.05 (m, IH), 31P NMR
Figure imgf000135_0002
(t, J = 7.2 (q, J = 7.5
3v IH), 7.34
Figure imgf000135_0003
MS (ES+) m/z = 229.3 (MH+). J= 6.9 Hz, (m, MHz) δ
Figure imgf000135_0004
Yellow pale oil, 1H NMR (dg-DMSO, 300 MHz) δ 1,19 (t, J = 6.9Hz, 3H), 2.30 (brs, 3H), 2.45 (brs, 3H), 3.87 (quintuplet, J = 7.5Hz, 2H), 7.18-7.27 (m, 2H), 7.57 (d, J = 14.4Hz, IH), 31P
Figure imgf000135_0005
NMR (d6-DMSO, 101 MHz) δ 15.79.
Yellow pale oil, 1H NMR (ds-DMSO, 300 MHz) δ 1,17 (t, J = 7.05 Hz, 3H), 2.27 (brs, 6H), 3.85 (quintuplet, J = 7.2Hz, 2H), 7.23-7.27 (m, IH), 131-1 Al (m, 2H), 31P NMR (dβ-DMSO, 101
Figure imgf000135_0006
MHz) δ 16.92, MS (ESI, El+) m/z = 215 (MH+). (t, J = 19F NMR (J6-
3ad
Figure imgf000135_0007
31P NMR (J6-DMSO, 101.256 MHz) δ 12.80 (d, J= 6.5 Hz, IP), MS (ESI, El+) m/z = 223 (MH+). Compound structure Description
Oil, 1H NMR (J6-DMSO, 300 MHz) δ 1.18 (t, / = 7.5 Hz, 3H), 1.19 (t, J = 7.05 Hz, 3H), 2.91 (q, J = 7.5 Hz, 2H), 3.89
3ae (quintuplet, J = 7.05 Hz, 2H), 7.24-7.36 (m, 2H), 7.46-7.52 (m,
Figure imgf000136_0001
IH), 7.74 (ddd, J = 1.2 and 7.5 and 13.8 Hz, IH), 31P NMR (J6- DMSO, 121.49 MHz) δ 15.66, MS (ESI, El+) m/z = 215 (MH+).
Oil, 1H NMR (J6-DMSO, 300 MHz) δ 1.18 (t, J = 6.9 Hz, 3H), 2.4 (s, 3H), 3.85-3.97 (m, 2H), 7.78-7.81 (m, IH), 7.82-7.84 (m, IH), 7.84-7.87 (m, IH), 31P NMR (4-DMSO, 121.49 MHz) δ 11.67, MS (ES+) m/z = 226.2 (MH+).
Oil, 1H NMR (J6-DMSO, 300 MHz) δ 1.17 (t, J = 7.1 Hz, 3H), 1.3 (s, 9H), 3.82-3.91 (m, 2H), 7.41-7.53 (m, 2H), 7.59-7.61 (m, IH), 7.66-7.71 (m, IH), MS (ES+) m/z = 243 (MH+).
Figure imgf000136_0002
Hz, 3H), 7.78-7.82 (q, J = 8.2
Hz, 3H), IH), 8.03- 11.94 (d, J=
Figure imgf000136_0003
(m, IF), MS (ES+) UiZz = 228.3 (MH+).
Oil, 1H NMR (J6-DMSO, 300 MHz) δ 1.20 (t, J= 7.05 Hz, 3H), 3.87-3.97 (m, 2H), 7.4-7.47 (m, IH), 7.51-7.59 (m, 2H), 7.83- 7.91 (m, IH), 31P NMR (J6-DMSO, 121.49 MHz) δ 11.26, MS (ESI, El+) m/z = 221.03 (MH+).
Oil, 1H NMR (J6-DMSO, 300 MHz) δ 1.19 (t, J = 7.2 Hz, 3H), 3.9-4 (m, 2H), 7.31-7.53 (m, 3H), 31P NMR (J6-DMSO, 101.256
3al MHz) δ 8.05 (d, J = 5.5 Hz, IP), 19F NMR (J6-DMSO, 282.4
Figure imgf000136_0005
MHz) -110.71 (d, J= 19.4 Hz, IF), -118.33 (dd, J= 5.9 and 19.2 Hz, IF), MS (ES+) m/z = 223.03 (MH+). Table 2
Co
MHz) δ Hz, 3H),
(d6- El+) m/z
δ 5-5.12 5H),
= 594
Figure imgf000137_0001
300 / = 7.1 4.44 (q, 7.42 (s,
3H), 23.6, MS
Figure imgf000137_0002
Compound structure Description
Thick yellow oil, 1H NMR (CDCl3, 300 MHz) δ
1.37 (t, J = 7.2 Hz, 3H), 1,44 (t, J = 7.2 Hz, 3H), 4.05-4.19 (m, 2H), 4.52 (q, J = 7.2 Hz, 2H), 7.11- 7.17 (m, 2H), 7.32-7.36 (m, IH), 7.48-7.54 (m, 2H), 7.60-7.63 (m, IH), 7.84-7.93 (m, 4H), 8.07- 8.09 (m, 2H), 31P NMR (CDCl3, 101.256 MHz) δ 23.6, 19F NMR (CDCl3, 282.4 MHz) δ -105.36 (m, IF), MS (ESI, El+) m/z = 550 (MH+).
Thick yellow oil, 1H NMR (CDCl3, 300 MHz) δ
1.38 (t, J= 7.05 Hz, 3H), 1,46 (t, J= 7.2 Hz, 3H), 4.09-4.2 (m, 2H), 4.53 (q, J = 12 Hz, 2H), 7.35 (dd, J = 2.1 and 9 Hz, IH), 7.37-7.43 (m, IH), 7.48-7.54 (m, 3H), 7.6-7.95 (m, 5H), 8.07-8.1 (m, 2H), 31P NMR (CDCl3, 101.256 MHz) δ 22.76, MS (ESI, El+) m/z = 566 (MH+).
Figure imgf000138_0001
δ Hz, 3H), Hz, 2H), 7.3-
5f (m, = 1.5
Figure imgf000138_0002
m/z = 560 (MH+).
Yellow solid, 1H NMR (CDCl3, 400 MHz) δ 1.39 (t, J = 7 Hz, 3H), 1,44 (t, J = 7.2 Hz, 3H), 4.09- 4.23 (m, 2H), 4.53 (qd, J = 2 and 7.2 Hz, 2H), 7.35 (dd, J = 2 and 9.2 Hz, IH), 7.48-7.53 (m,
5g 2H), 7.57-7.64 (m, 2H), 7.68-7.79 (m, IH), 7.88 (d, J = 2 Hz, IH), 7.94 (dd, J = 1.6 and 8.8 Hz, IH), 8.04-8.1 (m, 3H), 8.16-8.2 (m, IH), 31P
Figure imgf000138_0003
NMR (CDCl3, 101.256 MHz) δ 22.62, 19F NMR (CDCl3, 235.36 MHz) δ -63.2, MS (ESI, El+) m/z Compound structure Description
= 600 (MH+). δ 1.4 4.04- (dd, J 7.80 (dd, 2.1 Hz,
δ
Figure imgf000139_0001
δ 1.38 Hz, 3H), 2H), 7.3- (m,
5i Hz, IH),
Figure imgf000139_0002
(m, IH), 8.07-8.11 (m, 2H), 31P NMR (CDCl3, 121.49 MHz) δ 22.11, MS (ES+) m/z = 609.73/611.77 (MH+). MHz) 6.9 Hz, 7.2 Hz,
5j Hz, 2H), 7H),
Figure imgf000139_0003
(ES+) m/z = 573.8 (MH+). δ Hz, 3H), 5.56 NMR m/z
Figure imgf000139_0004
Compound structure Description
Thick yellow oil, 31P NMR (d6-OMSO, 121.49 MHz) δ 22.73, MS (ES+) tn/z = 580 (MH+).
White powder, 1H NMR (CDCl3, 300 MHz) δ
1.37 (t, J = 7.2 Hz, 3H)3 1.45 (t, J = 7.2 Hz, 3H), 3.81 (s, 3H), 4.05-4.18 (m, 2H), 4.53 (q, J = 7.2 Hz, 2H), 7.07 (dt, J = 1.2 Hz and 8.1 Hz, IH), 7.28-7.63 (m, 7H), 7.87 (d, J = 2.1, IH), 7.90 (dd,
Figure imgf000140_0001
J = 1.5 Hz and 9 Hz, IH), 8.08 (dd, J = 1.5 Hz and 7.5 Hz, 2H), 31P NMR (CDCl3, 121.49 MHz) δ 24.00 MHz) δ Hz, 3H), (q, J = Hz, 2H),
4H),
Figure imgf000140_0002
MHz) δ 24.36, MS (ESI, El+) m/z = 562 (MH+). MHz) δ Hz, 3H), J = 7.2 IH),
Sq
Figure imgf000140_0003
(m, 3H), 7.6-7.66 (m, IH), 7.9-7.98 (m, 2H), 8.03 (d, J = 2.1 Hz, IH), 8.09-8.12 (m, 2H), 31P NMR (CDCl3, 121.49 MHz) δ 21.9, MS (ESI, El+) mk = 562 (MH+). Co
δ -
(q, J (MH+).
MHz) δ (m, = 2.1 Hz J= 1.8 3H),
Figure imgf000141_0001
δ 24.31, MS (ESI, El+) m/z = 582 (MH+).
Yellowish powder, 1H NMR (CDCl3, 300 MHz) δ 1.23 (d, J = 7.2 Hz, 6H), 1.38 (t, J = 7.2 Hz, 3H), 1.46 (t, J = 7.2 Hz, 3H), 2.83-3.06 (m, IH), 4.05- 4.25 (m, 2H), 4.55 (q, J = 7.0 Hz, 2H), 7.05-7.10 (m, IH), 7.33 (dd, J = 2.1 Hz and 9.0 Hz, IH),
Figure imgf000141_0002
7.31-7.41 (m, IH), 7.45-7.64 (m, 4H), 7.86 (d, / = 1.8 Hz, IH), 7.94 (dd, J = 8.7 Hz and 1.8 Hz, IH), 8.06-8.11 (m, 2H), 31P NMR (CDCl3, 121.49 MHz) δ 22.83 (d, J = 7.65 Hz), MS (ES+) m/z = 592 (MH+). Compound δ 1.42 4.11-
5w IH), NMR m/z =
Figure imgf000142_0001
Oil, 1H NMR (J6-DMSO, 300 MHz) δ 1,27 (t, J = 6.9 Hz, 3H), 1,37 (t, J= 7.2 Hz, 3H), 2.34 (s, 3H), 3.97-4.02 (m, 2H), 4.44 (q, J = 7.2 Hz, 2H), 7.34-
Sx 7.38 (m, 2H), 7.51-7.55 (dd, J = 2.1 and 9 Hz, IH), 7.65-7.72 (m, 4H), 7.75-7.83 (m, 2H) 31P NMR (Jg-DMSO, 101.256 MHz) δ 23.57, MS
Figure imgf000142_0002
(ESI, El+) m/z = 546 (MH+). (t, J = 2.25 (2s, Hz, 2H),
5aa 3H), 23.64, MS
Figure imgf000142_0003
MHz) Hz, 3H), 7.44- (m,
5ad 3H), +
m/z =
Figure imgf000142_0004
Compound structure Description
δ (d, J = J= 9.3 (m, 13.91, 19F (ESI,
Figure imgf000143_0001
MHz) Hz, 3H), Hz, 2H),
4H), 8.06- MHz)
Figure imgf000143_0002
Oil, 1H NMR (J15-DMSO, 300 MHz) δ 1.25 (s, 9H), 1.25 (t, J= 7.2 Hz, 3H), 1.39 (t, J = 7.2 Hz, 3H), 3.99 (m, 2H), 4.46 (q, J= 7.2 Hz, 2H), 7.47- 7.55 (m, 2H), 7.6-7.7 (m, 4H), 7.75-7.83 (m, 3H), 8.05-8.1 (m, 3H), 31P NMR (J6-DMSO, 121.49 MHz) δ 22.6, MS (ES+) m/z = 588.17 (MH+).
Figure imgf000143_0003
J= 7.05 3H), 4- (dd, J 7.85-7.97
5aj 571.4
Figure imgf000143_0004
Compound structure Description (t, J= (q, / = (m,
5ak 7.92-8
Figure imgf000144_0001
565.93 (MH+).
Oil, 1H NMR (J6-DMSO, 300 MHz) δ 1.3 (t, J = 7.05 Hz3 3H), 1.37 (t, J= 7.05 Hz, 3H), 4-4.16 (m, 2H), 4.46 (q, J = 7.05 Hz, 2H), 7.56 (dd, J = 2.1
5al and 9 Hz, IH), 7.66-7.71 (m, 2H), 7.79-7.84 (m, IH), 7.91-7.99 (m, 2H), 8.08-8.18 (m, 5H), 19F NMR (J6-DMSO, 282.40 MHz) -112.68 (q, J =
Figure imgf000144_0002
8.2 Hz, IF), MS (ES+) m/z = 575 (MH+).
White solid, 1H NMR (J6-DMSO, 300 MHz) δ (m,
6a 10,27
Figure imgf000144_0003
= 363 (MH+). δ
7.29-
6b Hz, IH), (brs, 101.256
Figure imgf000144_0004
White Solid, 1H NMR (J6-DMSO, 300 MHz) δ
1.34 (t, J= 6.9 Hz, 3H), 4-4.04 (m, IH), 4.18-4.21 (m, IH), 7.31-7.4 (m, 3H), 7.57-7.62 (m, 2H),
6c 7.74-7.83 (m, 2H), 8.02 (brs, IH), 10.18 (brs, IH), 12.79 (brs, IH), 31P NMR (J6-DMSO, 121.49 MHz) δ 29.29, 19F NMR (J6-DMSO, 282.4 MHz)
Figure imgf000144_0005
Compound structure Description δ -106.3, MS (ESI, El+) m/z = 381 (MH+).
White solid, 1H NMR (J6-DMSO, 300 MHz) δ 1,35 (t, J = 7.05 Hz, 3H), 4-4.09 (m, IH), 4.17- 4.25 (m, IH), 7.3-7.35 (m, IH), 7.55-7.74 (m, 6H), 8.05 (brs, IH), 10.07 (brs, IH), 12.84 (brs, IH), 31P NMR (J6-DMSO, 121.49 MHz) δ 28.24,
Figure imgf000145_0001
MS (ESI, El+) m/z = 397 (MH+). δ Hz, 3H), 4.14-
6e Hz, IH), IH),
Figure imgf000145_0002
El+) m/z = 391 (MH+).
White solid, 31P NMR (CDCl3, 101.256 MHz) δ 29.02, 19F NMR (CDCl3, 235.36 MHz) δ -61.07,
6f MS (ESI, El+) m/z = 431 (MH+).
White solid, 1H NMR (J6-DMSO, 400 MHz) δ 1.36 (t, J = 7.05 Hz, 3H), 4-4.12 (m, IH), 4.17- 4.27 (m, IH), 7.33 (dd, J = 2 and 8.76 Hz, IH), 7.59 (dd, J= 1.56 and 8.78 Hz, IH), 7.66 (d, J =
6g 1.99 Hz, IH), 7.73 (td, J= 3.3 and 7.78 Hz, IH),
Figure imgf000145_0003
7.97-8.08 (m, 3H), 8.17-8.21 (m, IH), 9.98 (brs, IH), 12.83 (brs, IH), 31P NMR (J6-DMSO, 101.256 MHz) δ 28.62, MS (ESI, El+) m/z = 388 (MH+).
White solid, 1H NMR (ds-DMSO, 300 MHz) δ 1.16 (d, J= 6.9 Hz, 3H), 1.17 (d, J= 6.9 Hz, 3H),
6i 1.34 (t, J= 7.05 Hz, 3H), 2.88-2.97 (m, IH), 3.97- 4.06 (m, IH), 4.15-4.23 (m, IH), 7.31 (dd, J= 1.8 and 8.7 Hz, IH), 7.42-7.66 (m, 6H), 8.01 (brs,
Figure imgf000145_0004
IH), 10.31 (brs, IH), 12.74 (brs, IH), 31P NMR Compound structure Description
(J6-DMSO, 121.49 MHz) δ 30.22, MS (ES+) m/z = 404.8 (MH+). δ 4.25 7.35- IH), MS
Figure imgf000146_0001
δ 4.17- 7.50- IH), (J6- Hz), 19F
Figure imgf000146_0002
NMR (J6-DMSO, 282.4 MHz) δ -111.96 (q, J = 7.9 Hz, MS (ES+) m/z = 459/460 (MH+).
White solid; 31P NMR (J6-DMSO, 121.49 MHz) δ 28.35, MS (ES+) m/z = 411 (MH+).
White powder, 1H NMR (J6-DMSO, 300 MHz) δ 1,39 (t, J = 7.04 Hz, 3H), 3.77 (s, 3H), 4.25-3.95 (two m, 2H), 7.29-7.14 (m, 4H), 7.33 (dd, J= 8.8 and 1.98 Hz, IH), 7.49-7.42 (m, IH), 7.59 (dd, J= 8.8 and J< 1.5 Hz, IH), 7.66 (d, J= 1.93 Hz, IH),
Figure imgf000146_0003
8.00 (brs, IH), 10.20 (brs, IH), 12.75 (brs, IH), 31P NMR (J6-DMSO, 121.49 MHz) δ 30.85, MS (ESI, El+) m/z = 393 (MH+), 785 (2M+H1). Compound structure Description
MHz) δ 4.18- Hz, 7.74
Figure imgf000147_0001
31P NMR (J6-DMSO, 300 MHz) δ 27.25 (d, J = 6.57 Hz, IP), MS (ESI, El+) mk = 449 (MH+) δ 7.35 J = 1.5 8.08 (brs,
Figure imgf000147_0002
(J6- DMSO, 121.48 MHz) δ 26.63, MS (ESI, El+) mk = 499 (MH+).
White solid, 1H NMR (JrDMSO, 300 MHz) δ 1.35 (t, J = 6.9 Hz, 3H), 3.95-4.08 (m, IH), 4.26- 4.37 (m, IH), 7.27 (dd, J = 1.8 Hz and 8.7 Hz, IH), 7.38 (d, J= 1.8 Hz, IH), 7.56-7.65 (m, 4H), 7.86-7.94 (m, IH), 8.02-8.06 (m, IH), 8.12 (brs,
Figure imgf000147_0003
IH), 8.19 (d, J= 8.1 Hz, IH), 8.54-8.57 (m, IH), 10.45 (brs, IH), 12.83 (brs, IH), MS (ESI, El+) w/z = 413 (MH+). wder, 31P NMR (CDCl3, 121.49 (d, J = 8.0 Hz, IP), MS (ES+) m/z =
Figure imgf000147_0004
Co Description
Colourless oil, 1H NMR (CDCl3, 300 MHz) δ 1.48 (t, J= 7.05 Hz, 3H), 4.05-4.18 (m, IH), 4.28- 4.41 (m, IH), 6.18 (brs, IH), 7.22-7.26 (m, IH), 7.31-7.4 (m, 2H), 7.53-7.55 (m, IH), 7.57-7.58 (m, IH), 7.67-7.68 (m, IH), 10.90 (brs, IH), 11.13 (brs, IH), 31P NMR (CDCl3, 121.49 MHz) δ
Figure imgf000148_0001
27.7 (d, J = 7.8 Hz, IP), MS (ES+) m/z = 415.28 (MH+). 400 3.94- (m,
6w (brs,
= 377
Figure imgf000148_0002
Yellow powder, 1H NMR (J<rDMSO, 300 MHz) δ 1,29 (t, J = 7.05 Hz, 3H), 2.22 (s, 3H), 2.37 (s, 3H), 3.81-3.94 (m, IH), 4.11-4.24 (m, IH), 7.21- 7.26 (m,lH), 7.27 (dd, J = 2.1 and 8.7 Hz, IH), 7.32 (d, J = 1.8 Hz, IH), 7.4 (d, J= 7.5 Hz, IH), 7.48-7.6 (m, 2H), 8.01 (brs, IH), 10.42 (brs, IH),
Figure imgf000148_0003
12.75 (brs, IH), 31P NMR (J5-DMSO, 101.256 MHz) δ 31.55, MS (ESI, El+) m/z = 391 (MH+).
Off white powder; 1H NMR (J15-DMSO, 300 MHz) δ 1,33 (t, J = 6.9 Hz, 3H), 2.32 (s, 3H), 3.95-4.04 (m, IH), 4.12-4.24 (m, IH), 7.31 (dd, J
6ab = 1.5 and 8.7 Hz, IH), 7.41-7.61 (m, 6H), 8.00 (brs, IH), 10.28 (brs, IH), 12.74 (brs, IH), 31P
Figure imgf000148_0004
NMR (Jβ-DMSO, 101.256 MHz) δ 31.28, MS (ESI, El+) m/z = 311 (MH+). Compound structure Description 300 (m, 7.50- (brs, (J5- IP), 19F
Figure imgf000149_0001
NMR (Js-DMSO) δ -106.92 (m, 2F), MS (ESI, El+) m/z = 399 (MH+).
Pale yellow powder, 1H NMR (J5-DMSO, 300 MHz) δ 1,35 (t, J = 6.9 Hz, 3H), 3.97-4.11 (m, IH), 4.16-4.29 (m, IH), 7.22-7.42 (m, 3H), 7.56- 7.59 (m, 2H), 7.61-7.75 (m, IH), 7.77-7.88 (m, IH), 8.01 (brs, IH), 10.08 (brs, IH), 12.78 (brs,
Figure imgf000149_0002
IH), 31P NMR (J5-DMSO, 101 MHz) δ 25.2 (d, J = 11.3 Hz, IP), 19F NMR (J5-DMSO) δ -104.85 (m, IF), MS (ESI, El+) m/z = 381 (MH+). MHz) Hz, 3H),
7.32- (m, IH),
Figure imgf000149_0003
IH), 31P NMR (J5-DMSO, 121.69 MHz) δ 29.63, MS (ESI, EΫ) m/z = 391 (MH+). 300 (m, and
6ai 12.71
Figure imgf000149_0004
δ 25.31, MS (ESI, El+) m/z = 397 (MH+). Compound structure Description
δ 1.48 (t, (m, 8.7 Hz, 7.64-
6aj IH), MHz) δ
Figure imgf000150_0001
282.40 MHz) -107 (q, J= 7.34 Hz, IF), MS (ES+) m/z = 406.3 (MH+). δ 7.34 IH), 7.74 IH), IH), 31P MS
Figure imgf000150_0002
= 422 (MH+). 300 3H), (m, IH), 33.27, MS
Figure imgf000150_0003
δ 1.8 and IH), 7.67
7b 7.89 (t, IH),
Figure imgf000150_0004
(MH+). Compound structure Description
δ 1.8 and IH), (J6- El+) m/z
Figure imgf000151_0001
White solid, 1H NMR (J6-DMSO, 300 MHz) δ 1.14 (t, J = 7.5 Hz, 3H), 2.63 (q, J= 7.5 Hz, 2H), 3.75 (d, J = 11.4 Hz, 3H), 7.32 (dd, J= 1.95 and
7d 8.85 Hz, IH), 7.41-7.52 (m, 3H), 7.57-7.63 (m, 3H), 8.02 (brs, IH), 10.27 (brs, IH), 12.78 (brs,
Figure imgf000151_0002
IH), 31P NMR (CDCl3, 121.49 MHz) δ 32.2; MS (ESI, El+) m/z = 377 (MH+). δ 3.8 8.7 Hz, (d, J= IH), (brs, IH),
Figure imgf000151_0003
19F NMR (CDCl3, 282.4 MHz) δ 118.06 (s, 3F), MS (ESI, El+) m/z = All (MH+).
White solid, 1H NMR (J6-DMSO, 300 MHz) δ 3.79 (d, J = 11.7 Hz, 3H), 7.33 (dd, J = 1.95 and
8.7 Hz, IH), 7.59 (dd, J = 1.8 and 8.7 Hz, IH),
7f 7.65 (d, J= 1.8 Hz, IH), 7.69-7.75 (m, IH), 7.96-
8.08 (m, 3H), 8.17-8.21 (m, IH), 9.9 (brs, IH),
Figure imgf000151_0004
12.88 (brs, IH), 31P NMR (J6-DMSO, 121.49 MHz) δ 29.65, MS (ES+) m/z = 373.86 (MH+).
White solid, 1H NMR (J6-DMSO, 300 MHz) δ 1.16 (d, J= 6.9 Hz, 3H), 1.17 (d, J= 6.9 Hz, 3H), 2.88-2.98 (m, IH), 3.76 (d, J= 11.4 Hz, 3H), 7.32 (dd, J= 1.95 and 8.85 Hz, IH), 7.41-7.68 (m, 6H), 8.03 (brs, IH), 10.28 (brs, IH), 12.77 (brs, IH),
Figure imgf000151_0005
31P NMR (J6-DMSO, 121.49 MHz) δ 32.16, MS Compound structure Description
White solid, 1H NMR (J6-DMSO, 300 MHz) δ 3.77 (d, J = 11.7 Hz, 3H), 5.47 (d, J = 47.4 Hz, 2H), 7.33 (dd, J = 2.1 and 8.7 Hz, IH), 7.55-7.81 (m, 6H), 8.04 (brs, IH), 10.19 (brs, IH), 12.78 (brs, IH), 31P NMR (J6-DMSO, 121.49 MHz) δ
Figure imgf000152_0001
31.62, 19F NMR Cd6-DMSO, 282.40 MHz) δ - 207.99 (t, J = 47.4 Hz, IF), MS (ES+) m/z = 380.83 (MH+). MHz) δ 8.7 and (m, (brs, δ 31.74,
Figure imgf000152_0002
White solid; 1H NMR (J6-DMSO, 300 MHz) δ 3.79 (d, J = 11.7 Hz, 3H), 7.35 (dd, J = 8.7 and 2.1 Hz, IH), 7.51-7.72 (m, 9H), 7.83-7.87 (m,
7j 2H), 8.06 (brs, IH), 9.83 (brs, IH), 12.88 (brs, IH), 31P NMR (J6-DMSO, 121.49 MHz) δ 28.46 (d, J= 7.2 Hz), 19F NMR (J6-DMSO, 282.4 MHz)
Figure imgf000152_0003
δ -110.60 (m), MS (ES+) m/z = 445/447 (MH+). 300 MHz) δ 3H), 7.34 (dd, J (m, 5H), 8.04 IH), 31P 30.31, MS
Figure imgf000152_0004
White powder, 1H NMR (CDCl3, 300 MHz) δ 3.81 (s, 3H), 3.87 (d, J = 11.4 Hz, 3H), 6.02 (brs, IH), 7.06-7.08 (m, IH), 7.28-7.37 (m, 4H), 7.49- 7.52 (m, IH), 7.73 (brs, IH), 10.67 (brs, IH), 11.01 (brs, IH);, 31P NMR (CDCl3, 121.49 MHz)
Figure imgf000152_0005
δ 32.67, MS (ESI, El+) m/z = 379 (MH+). Compound structure Description
White powder, 1H NMR (CDCl3, 300 MHz) δ 3.92 (d, J= 11.7 Hz, 3H), 6.12 (brs, IH), 7.36 (dd, J = 1.8 Hz and 8.7 Hz, IH), 7.48-7.51 (m, IH), 7.55-7.65 (m, 3H), 7.9 (d, J= 12.9 Hz, IH), 10.81
7q (brs, IH), 10.97 (brs, IH), 31P NMR (CDCl3,
Figure imgf000153_0001
121.49 MHz) δ 29.27 (d, J = 6.8 Hz, IP), 19F NMR (CDCl3, 282.40 MHz) δ -62.89 (s, 3F), - 107.86 (q, J = 7.6 Hz, IF), MS (ESI, El+) m/z = 435 (MH4).
White powder, 1H NMR (CDCl3, 300 MHz) δ 3.94 (d, J= 11.7 Hz, 3H), 6.12 (brs, IH), 7.37 (dd, J = 2.1 Hz and 9 Hz, IH), 7.57 (dd, J = 1.8 Hz
7s and 8.7 Hz, IH), 7.63-7.637 (m, IH), 8.04 (brs, IH), 8.21 (brs, IH), 8.26 (brs, IH), 10.81 (brs,
Figure imgf000153_0002
IH), 10.94 (brs, IH), 19F NMR (CDCl3, 282.40 MHz) δ -62.96 , MS (ESI, El+) m/z = 485 (MH+). δ 7.3 (dd, 5H), 7.77 (m, IH), 10.58
Figure imgf000153_0003
121.49 MHz) δ 34.98, MS (ESI, El+) m/z = 399 (MH+). MHz) δ Hz, 3H), Hz,,
Figure imgf000153_0004
IH), 7.95 (dd, J = 9.6 Hz and 1.8 Hz, IH), 8.08-8.14 (m, 2H), 10.54 (brs, IH), 10.95 (brs, IH), 31P NMR (CDCl3, 121.49 MHz) δ 31.35 (d, J = 8.4 Hz, IP), MS (ES+) m/z = 409 (MH+). Compound Description
Off-white solid, 1H NMR (CDCl3, 300 MHz) δ 3.9 (d, J= 11.7 Hz, 3H), 6.09 (brs, IH), 7.24-7.28 (m, IH), 7.32-7.40 (m, 2H), 7.53-7.59 (m, 2H),
7v 7.65-7.66 (m, IH), 10.81 (brs, IH), 10.87 (brs, IH), 31P NMR (CDCl3, 121.49 MHz) δ 29.77 (d, J
Figure imgf000154_0001
= 7.78 Hz, IP), 19F NMR (CDCl3, 282.40 MHz) δ -111.22 (m, IF), MS (ES+) m/z = 401.29 (MH+). δ (q, J = (brs,
10.29
Figure imgf000154_0002
121.49 MHz) δ 32.21, MS (ES+) m/z = 391.3 (MH+). δ J = 7.2 J= 11.4 7.4- IH),
Figure imgf000154_0003
(d6- DMSO, 121.49 MHz) δ 32.17, MS (ES+) m/z = 391.26 (MH+). δ 2.40 (d, J = 7H), (CDCl3, 414
Figure imgf000154_0004
Compound structure Description
MHz) δ 7.20 (brs, J =
7z 8.7 Hz, (brs,
Figure imgf000155_0001
δ 31.74 , MS (ES+) m/z = 394.9 (MH+).
Off-white solid, 1H NMR ((CDCl3, 300 MHz) δ 0.69-0.71 (m, 2H), 0.98-1.02 (m, 2H), 1.85-1.95 (m, IH), 3.86 (d, J = 11.7 Hz, 3H), 5.98 (brs, IH),
7aa 7.2-7.22 (m, IH), 7.28 -7.35 (m, 3H), 7.47-7.58 (m, 3H), 7.71-7.72 (m, IH), 10.50 (brs, IH),
Figure imgf000155_0002
11.04 (brs, IH), 31P NMR (CDCl3, 121.49 MHz) δ 33.04 , MS (ES+) m/z = 389.08 (MH+).
MHz) 7.31-7.33 10.26
= 363
Figure imgf000155_0003
300 3H), 7.49 (d, J
7ae (brs, IH), NMR (J6- (ESI, El+)
Figure imgf000155_0004
Pale yellow powder, 1H NMR (CDCl3, 300 MHz) δ 3.85 (d, J = 11.4 Hz, 3H), 6.08 (brs, IH), 7.30 (dd, J = 2.0 and 9.0 Hz, IH), 7.36-7.56 (m, 4H), 7.68 (d, J= 1.8 Hz, IH), 7.73-7.81 (m, 2H), 10.78 (brs, IH), 10.03 (brs, IH), 31P NMR (CDCl3, 101
Figure imgf000155_0005
MHz) δ 33.3, MS (ESI, El+) m/z = 349 (MH+). Compound structure Description MHz) 7.18- (m, NMR m/z =
Figure imgf000156_0001
Orange powder, 1H NMR (J6-DMSO, 300 MHz) δ 3.79 (d, J = 11.4 Hz, 3H), 7.32-7.44 (m, 3H), 7.51-7.61 (m, 2H), 7.67 (d, J = 1.8 Hz, IH), 8.05
7aj (brs, IH), 9.86 (brs, IH), 12.86 (brs, IH), 31P NMR (J6-DMSO, 101 MHz) δ 29.99 (t, J = 8.3 Hz, IP), 19F NMR (J6-DMSO, 300MHz) δ -
Figure imgf000156_0002
106.93 (m, 2F), MS (ESI, El+) m/z = 385 (MH+). 300 (m, 8.01
7ak 31P NMR El+) m/z
Figure imgf000156_0003
Off white powder, 1H NMR (J6-DMSO, 300 MHz) δ 0.92 (t, J = 7.5 Hz, 3H), 2.78-2.92 (m, 2H), 3.7 (d, J= 11.7 Hz, 3H), 7.25-7.38 (m, 4H),
7an 7.52-7.59 (m, 2H), 7.67-7.75 (m, IH), 8.02 (brs, IH), 10.36 (brs, IH), 12.78 (brs, IH), 31P NMR.
Figure imgf000156_0004
(J6-DMSO, 101 MHz) δ 32.41, MS (ESI, El+) m/z = 377 (MH+). MHz) δ 2.1 and
IH), 12.8 δ
Figure imgf000156_0005
Compound structure Description
White powder, 1H NMR (J6-DMSO, 300 MHz) δ 3.75 (d, J = 11.7 Hz, 3H), 4.47 (s, 2H), 4.54 (s, 2H), 7.23-7.37 (m, 6H), 7.5-7.73 (m, 6H), 8.01 (brs, IH), 10.21 (tars, IH), 12.8 (bra, IH), 31P NMR (J6-DMSO, 121.49 MHz) δ 32.13, MS (ES+) HtZz = 468.86 (MH+).
Pale yellow powder, 1H NMR (CDCl3, 300 MHz) δ 3.88 (d, J = 11.7 Hz, 3H), 4.73 (s, 2H), 5.98 (brs, IH), 7.3-7.35 (m, IH), 7.42-7.49 (m, 2H), 7.56-7.59 (m, IH), 7.66-7.73 (m, 2H), 7.8-7.85 (m, IH), 10.34 (brs, IH), 10.99 (brs, IH), 31P
Figure imgf000157_0001
NMR (CDCl3, 101.256 MHz) δ 32.85, MS (ES+) m/z = 378.86 (MH+).
White powder, 1H NMR (J15-DMSO, 300 MHz) δ 1.24 (s, 9H), 3.76 (d, J = 11.4 Hz, 3H), 7.3-7.34 (m, IH), 7.4-7.65 (m, 5H), 7.76-7.81 (m, IH), 8.02 (brs, IH), 10.27 (brs, IH), 12.75 (brs, IH), 31P NMR (Jβ-DMSO, 121.49 MHz) δ 32.29, MS
Figure imgf000157_0002
(ES+) InZz = 405.11 (MH+). δ 2.4 IH), (m, 10.92 δ
Figure imgf000157_0003
White powder, 1H NMR (dβ-DMSO, 300 MHz) δ 3.78 (d, J = 11.4 Hz, 3H), 6.60 (d, J = 8.4 Hz, IH), 7.34 (d, J = 1.8 Hz, IH), 7.74 (dd, J = 1.7
7au and 8.4 Hz, IH), 8.03-8.18 (m, 3H), 8.29 (brs, IH), 9.67 (brs, IH), 12.89 (brs, IH), 31P NMR (
Figure imgf000157_0004
d6-DMSO, 121.49 MHz) δ 27.88, MS (ES") m/z = 406 (MH+). Compound structure Description
MHz) δ and (m,
7av (brs,
Figure imgf000158_0001
(MH+).
Figure imgf000158_0002
Compound structure Description
Figure imgf000159_0001
Compound structure Description
Figure imgf000160_0001
1,43 (t, J =
Hz, IH), (brs, MS (ESI,
Figure imgf000160_0002
δ 1,45 (t, J (m, J= 6.1
9a 6H), 7.75 IH),
Figure imgf000160_0003
MHz) δ 32.8, MS (ESI, El+) m/z = 482 (MH+).
J = 7.2 4H), 7.77 = 442-444
Figure imgf000160_0004
δ 4 (s, (d, J= 9 =
Figure imgf000160_0005
(CDCl3, 300 MHz) δ 1.46 (t, J= J = 7.1 Hz, 2H), 7.31-7.36 (m, IH), .04 (m, 3H), MS (ESI, El+) m/z =
Figure imgf000161_0001
MHz) δ 2.37 (s, J= 2.85 and 7.95
Figure imgf000161_0002
J = 13.5 Hz, IH), 31P NMR (CDCl3, 101.256 MHz) δ 23.8. 1.37 (t, J J = 7.2 8.07-8.24 δ 22.98,
Figure imgf000161_0003
1.46 (t, J J= 7.05 31P NMR =
Figure imgf000161_0004
δ 1.36 (q, J = J = 2.1 3H)5 31P m/z =
Figure imgf000161_0005
δ 1.26 (d, (t, J = 6.9 = 11.4
16d (m, IH), 7.37 IH), 7.49-
Figure imgf000161_0006
J = 9.6 Hz and 1.8 Hz, IH), 8.08-8.14 (m, 2H), MS (ES+) m/z = 578 (MH+). 0.83 (t, J J= 11.7 Hz, 8.05-8.12 24.36, MS
Figure imgf000162_0001
δ 2.29 7.25-7.34 7.9-7.94 f (MH+).
Figure imgf000162_0002
Off-white solid, 1H NMR (CDCl3, 300 MHz) δ 0.66-0.71 (m, 2H), 0.91-0.99 (m, 2H), 1.81-1.94 (m, IH), 3.69 (s, 3H), 3.79 (d, J = 11.4 Hz, 3H), 7.13-7.17 (m, IH), 7.28- 7.4 (m, 3H), 7.51-7.64 (m, 2H), 8.39-8.4 (m, IH), 9.96 (brs, IH), 31P NMR (CDCl3, 121.49 MHz) δ 31.29.
Thick yellow oil, 1H NMR (J6-DMSO, 300 MHz) δ 1.15 (t, J = 7.5 Hz, 3H), 1.39 (t, J = 12 Hz, 3H) 2.32 (s, 3H), 3.69 (d, J = 11.7 Hz, 3H), 4.46 (q, J = 7.5 Hz, 2H), 7.29 (brs, IH), 7.4-7.46 (m, 2H), 7.53-7.56 (m, IH), 7.63-7.81 (m, 4H), 8.03-8.12 (m, 3H), 31P NMR (J6-DMSO, 121.49 MHz) δ 24.57, MS (ES+) m/z = 560.5 (MH+).
Figure imgf000162_0003
δ 1.39 (t, J Hz, 3H), 3.71 7.53-7.85 121.49 i
Figure imgf000163_0001
Colorless oil, 1H NMR (CDCl3, 300 MHz) δ 1.45 (t, J = 7.2 Hz, 3H), 3.80 (d, J= 11.4 Hz, 3H), 4.54 (q, J= 7.2 Hz, 2H), 7.36 (dd, J= 2.1 and 9.0 Hz, IH), 7.47-7.67 (m, 6H), 7.84-7.96 (m, 4H), 8.09-8.12 (m, 2H), 31P NMR (CDCl3, 101 MHz) δ 26.7, MS (ESI, El+) m/z = 518 (MH+).
Figure imgf000163_0002
δ 1,36 (t, (q, J= 7.1 (dd, J = (m, k δ -105.46 MS
J = 7.05 4.46 (q, J 2H), (m, 3H), m
Figure imgf000163_0003
J= 7.1 Hz, (q, J = IH), 7.53 7.78-7.83
16n = 546
Figure imgf000164_0001
(t, J = J= 7.05 (m, 6H), (d6- = 638
Figure imgf000164_0002
(t, J = 3H), 4.45 IH), (m, 2H), lόq MHz) δ
Figure imgf000164_0003
Compound structure Description
White solid, 1H NMR (Jn-DMSO, 300 MHz) δ 2.33 (s, 3H), 3.73 (d, J= 11.7 Hz, 3H), 7.36-7.43 (m, 3H), 7.57-7.66 (m, 3H), 7.81- 7.82 (m, IH), 13.01 (brs, IH), 31P NMR (J15-DMSO, 121.49 MHz) δ 32.55, MS (ES+) m/z = 362 (M-H+).
Figure imgf000164_0004
Compound structure Description
(t, J= 7.5 (d, J = 11.7 (m, 2H), (brs, s, (ES+) m/z
Figure imgf000165_0001
Off-white solid, 1H NMR (J6-DMSO, 300 MHz) δ 2.29 (s, 6H), 3.78 (d, J = 11.7 Hz, 3H), 7.28 (brs, IH), 7.37 (brs, IH), 7.42 (brs, IH), 7.46-7.48 (m, 2H), 13.35 (s, IH), 31P NMR (J6-DMSO, 121.49 MHz) δ 35.15 , 19F NMR (J15-DMSO, 282.40 MHz) δ - 115.84 (s, IF) + MS (ES+) m/z = 395.9 (MH+).
Figure imgf000165_0002
1H NMR (CDCl3, 300 MHz) δ 3.88 (d, J= 12.0 J = 2.0 and 9.0 Hz, IH), 7.47-7.63 (m, 5H),
17d 10.33 (brs, IH), MS (ESI, El+) m/z = 350
Figure imgf000165_0003
Hz, 3H), 7.56 (dd, J = (brs, IH)5
17e δ 31.68, MS
Figure imgf000165_0004
Pale brown oil, 1H NMR (J6-DMSO, 300 MHz) δ 3.72 (d, J = 11.7 Hz, 3H), 7.25-7.64 (m, 6H), 8.06 (d, J = 2.1 Hz, IH), 12.99 (brs, IH), 31P NMR (J6-DMSO, 121.49 MHz) δ 29.23 (d, J= 7.3 Hz, IP), 19F NMR (J6-DMSO, 300 MHz) δ -112.01 (m, IF), MS (ESI, El+) m/z = 368 (MH+).
Figure imgf000165_0005
Compound structure Description
Oil, 1H NMR (J6-DMSO, 300 MHz) δ 2.28 (s, 6H), 3.71 (d, J = 11.7 Hz, 3H), 7.2-7.24 (m, IH), 7.36-7.45 (m, 3H), 7.57-7.6 (m, IH), 7.78-7.79 (m, IH), 13.05 (brs, IH); 14.8 (brs, IH), 31P NMR tørDMSO, 121.49 MHz) δ 32.87, MS (ES+) m/z = 378 (MH+).
Off-white solid, MS (ES+) m/z = 389 (MH+).
Figure imgf000166_0001
(s, 3H), 7.31-7.61
18a IH), MHz) δ
Figure imgf000166_0002
2.3 (s, 3H), 6.38-6.45
18b 12.84 δ 32.72, MS
Figure imgf000166_0003
White solid, 1H NMR (J6-DMSO, 300 MHz) δ 3.78 (d, J = 11.7 Hz, 3H), 4.58-4.74 (m, 2H), 5.46 (d, J = 47.4 Hz, 2H), 7.32-7.38 (m, 3H), 7.55-7.82 (m, 6H), 8.51-8.53 (m, 2H), 11.3 (t, J = 5.7 Hz, IH), 12.92 (d, J = 2.1 Hz, IH), 19F NMR (dg-DMSO, 282.40 MHz) δ -211.09 (t, J= 47.4
Figure imgf000166_0004
Hz, IF), MS (ES+) m/z = 472 (MH+). Compound Description
Off-white powder, 1H NMR (CDCl3, 300 MHz) δ 2.35 (s, 3H), 3.88 (d, J = 11.7 Hz, 3H), 4.77 (qd, J = 6 Hz and 15.9 Hz, 2H), 7.04-7.35 (m, 7H), 7.68 (brs, IH), 8.59-9.61
18d (m, 2H), 11.22 (brs, IH), 11..71 (t, J = 5.7 Hz, IH), 31P NMR (CDCl3, 121.49 MHz) δ 32.55 (d, J = 8.2 Hz, IP),
Figure imgf000167_0001
19F NMR (CDCl3, 282.40 MHz) δ -111.55 (q, J = 8.2 Hz, IF), MS (ES+) m/z = 471.92 (MH+).
Pale yellow powder, 1H NMR (CDCl3, 300 MHz) δ 3.88 (d, J= 11.7 Hz, 3H), 4.7-4.77 (m, 2H), 6.97 (tt, J= 2.4 Hz and 8.7 Hz, IH), 7.2-7.33 (m, 6H), 7.64 (brs, IH), 8.52-
18e 8.6 (m, 2H), 11.55-11.58 (m, 2H), 31P NMR (CDCl3, 121.49 MHz) δ 31.11 (t, J = 8.5 Hz, IP), 19F NMR
Figure imgf000167_0002
(CDCl3, 282.40 MHz) δ -106.21 (m, 2F), MS (ES+) m/z = 476.17 (MH+).
White powder, 1H NMR (CDCl3, 300 MHz) δ 2.28 (s, 6H), 3.82 (d, J= 11.7 Hz, 3H), 4.69 (dd, J= 6 Hz and 15 Hz, IH), 4.78 (dd, J = 6 Hz and 15 Hz, IH), 7.13 (brs, IH), 7.24-7.28 (m, 5H), 7.29-7.31 (m, IH), 7.38-7.39 (m, IH), 7.68-7.69 (m, IH), 7.76-7.8 (m, IH), 8.56 (dd, J =
Figure imgf000167_0003
1.5 Hz and 4.5 Hz, IH), 8.7 (d, J= 2.1 Hz, IH), 31P NMR (CDCl3, 121.49 MHz) δ 34.18, MS (ES+) m/z = 468.33 (MH+).
White powder, 1H NMR (CDCl3, 300 MHz) δ 2.3 (s, 6H), 3.03 (t, J = 7.2 Hz, 2H), 3.76-3.88 (m, 2H), 3.79 (d, J = 11.7 Hz, 3H), 7.16-7.3 (m, 6H), 7.47-7.5 (m, IH), 7.62- 7.63 (m, IH), 8.43-8.45 (m, 2H), 10.83 (brs, IH), 11.36-
Figure imgf000167_0004
11.38 (m, IH), 31P NMR (CDCl3, 121.49 MHz) δ 34.38, MS (ES+) m/z = 482.38 (MH+). Compound structure Description 2.26 (s, and 15.9 (brs, J= 1.8
18h J= 1.2 (t, J= 5.4 121.49
Figure imgf000168_0001
MHz) δ - 132.93 (d, J= 5.9 Hz, IF), MS (ES+) m/z = 486.3 (MH+). δ 2.26 (s, and 15.9 (brs, IH), 2H), 7.59
18i IH), 8.6 Hz, IH), 12.87
Figure imgf000168_0002
δ 32.69, MS
Figure imgf000168_0003
(MH+).
White solid, 1H NMR (Jn-DMSO, 300 MHz) δ 2.24 (s, 6H), 3.74 (d, J= 11.7 Hz, 3H), 4.51 (dd, J = 5.25 and 15 Hz, IH), 4.67 (dd, J = 6.0 and 15 Hz, IH), 7.22-7.38 (m, 6H), 7.53-7.64 (m, 4H), 11.34 (t, J= 5.7 Hz, IH), 12.66 (brs, IH), 31P NMR (J15-DMSO, 121.49 MHz) δ 32.79, MS (ES+) m/z = 545.33/547.31 (MH+).
Off-white solid, 1H NMR (J15-DMSO, 300 MHz) δ 2.28 (s, 6H), 3.77 (d, J = 11.4 Hz, 3H), 4.64-4.82 (m, 2H), 7.24 (brs, IH), 7.34 (dd, J = 1.8 and 8.7 Hz, IH), 7.36 (brs, IH), 7.4 (brs, IH), 7.52 (dd, J= 1.5 and 5.4 Hz, IH), 7.58- 7.63 (m, 2H), 8.76 (d, J= 5.1 Hz, IH), 9.14 (d, J= 1.5 Hz, IH), 11.48 (t, J= 6.0 Hz, IH), 12.85 (brs, IH), 31P NMR
Figure imgf000168_0004
(J6-DMSO, 121.49 MHz) δ 32.68, MS (ES+) m/z = 469.4 (MH+). Compound structure Description 1.19 (t, J = 3H), 3.84 6H), 7.70 (s, NMR
Figure imgf000169_0001
δ 2.24 (s, and 15.6 (brs,
18n 6.97-7.35
Figure imgf000169_0002
MHz) δ 8.51, MS (ES+) m/z = 482 (MH+).
Off-white powder, 1H NMR (J6-DMSO, 300 MHz) δ 1.12 (t, J= 7.5 Hz, 3H), 2.28 (s, 3H), 2.58 (q, J= 7.5 Hz, 2H), 3.77 (d, J = 11.7 Hz, 3H), 4.68 (dd, J = 5.4 and 17.1 Hz, IH), 4.78 (dd, J = 5.7 and 16.8 Hz, IH), 7.27 (brs, IH), 7.33 (dd, J= 2.1 and 8.7 Hz, IH), 7.36-7.46 (m, 2H), 7.52 (dd, J= 1.2 and 5.1 Hz, IH), 7.60 (dd, J= 1.8 and 8.7 Hz,
Figure imgf000169_0003
IH), 7.63 (d, J = 1.5 Hz, IH), 8.76 (d, J = 5.4 Hz, IH), 9.14 (d, J = 1.2 Hz, IH), 11.47 (t, J = 5.4 Hz, IH), 12.82 (brs, IH), 31P NMR (J6-DMSO, 121.49 MHz) δ 32.62, MS (ES+) m/z = 483 (MH+). δ 3.78 J= 2.1 IH), (m,
18q (ES+) m/z
Figure imgf000169_0004
Compound structure Description
δ 2.27 (s, 7.22 (m,
18r 13.08 32.16, MS
Figure imgf000170_0001
Figure imgf000170_0002
Compound structure Description
Figure imgf000171_0001
Co Description
Figure imgf000172_0001
Compound structure Description
Figure imgf000173_0001
Compound structure Description
Figure imgf000174_0001
Compound structure Description
Figure imgf000175_0001
Compound structure Description MHz) δ 3.77 (d, IH), 4.72
ISap 8.51- IH), 31P m/z =
Figure imgf000176_0001
Yellow pale powder, 1H NMR (J6-DMSO, 300 MHz) δ Hz, IH), 16.5 Hz, J = 2.1
18aq (m, 3H), J= 5.7 Hz,
Figure imgf000176_0002
MHz) δ 33.64, MS (ES+) m/z = 481.94 (MH+). δ 3.86 (d, 4.79 = 1.9 and
18ar 2H), 7.52- 2H), 10.76
Figure imgf000176_0003
101 MHz) δ 34.2, MS (ESI, El+) m/z = 440 (MH+). δ 3.62 (s, 6.9 (m, NMR El+) m/z =
Figure imgf000176_0004
Compound structure Description δ 3.76 Hz, IH), 2H), 8H), 11.21 (Jn- m/z = 429
Figure imgf000177_0001
3.76 (s, and 15.6
(m,
18au 2.1 Hz, 121.49
Figure imgf000177_0002
δ 3.76 = 5.7 and 7.03- 8.55-8.58
18av 31P El+) m/z =
Figure imgf000177_0003
300 MHz) δ 3.78 7.33-7.65 (m, 9H), J = 5.5 Hz, IH), 12.94 121.49 MHz) δ 30.76 (d, 300 MHz) δ -111.0 (MH+).
Figure imgf000177_0004
Compound structure Description
Yellow powder, 1H NMR (J6-DMSO, 300 MHz) δ 2.25 (s, 6H), 3.75 (d, J = 11.4 Hz, 3H), 4.6 (dd, J = 5.4 and 16.2 Hz, IH), 4.72 (dd, J= 5.7 and 16.2 Hz, IH), 7.24-7.39 (m, 6H), 7.59-7.63 (m, 2H), 8.52-8.54 (m, 2H), 11.41 (t, J = 5.7 Hz, IH), 12.86 (brs, IH), 31P NMR (J6-DMSO, 121.49 MHz) δ 32.88, MS (ES+) m/z = 467.9 (MH+).
White powder, 1H NMR (CDCl3, 300 MHz) δ 2.3 (s, 6H), 2.7-2.75 (m, 4H), 3.73-3.77 (m, 4H), 3.86 (d, J = 11.7 Hz, 3H), 4.43 (dd, J = 5.7 and 12.9 Hz, IH), 4.52 (dd, J = 6 and 12.9 Hz, IH), 7.17 (brs, IH), 7.32 (dd, J= 2.1 and 8.7 Hz, IH), 7.36 (brs, IH), 7.41 (brs, IH), 7.49 (dd, J = 2.1
Figure imgf000178_0001
and 8.7 Hz, IH), 7.71-7.72 (m, IH), 10.5 (brs, IH), 11.46 (t, J = 5.7 Hz, IH), 31P NMR (CDCl3, 101.256 MHz) δ 34.52, MS (ES+) m/z = 475.87 (MH+).
Pale yellow powder, 1H NMR (J6-DMSO, 300 MHz) δ 2.24 (s, 6H), 3.73 (d, J = 11.7 Hz, 3H), 4.56 (dd, J = 5.4 and 15.3 Hz, IH), 4.73 (dd, J = 5.4 and 15.3 Hz, IH), 7.15-7.4 (m, 7H), 7.47 (td, J = 1.8 and 7.5 Hz, IH), 7.59
18az (dd, J = 1.8 and 8,1 Hz, IH), 7.63 (d, J = 2.1 Hz, IH), 11.32 (t, J = 5.4 Hz, IH), 12.83 (brs, IH), 31P NMR (J6- DMSO, 121.49 MHz) δ 32.76, 19F NMR (J6-DMSO,
Figure imgf000178_0002
282.40 MHz) δ -118.4 (m, IF), MS (ES+) m/z = 485 (MH+).
White powder, 1H NMR (J6-DMSO, 300 MHz) δ 2.25 (s, 6H), 3.66 (s, 3H), 3.72 (d, J = 11.7 Hz, 3H), 4.52-4.7 (m, 2H), 6.9 (brs, IH), 7.22 (brs, 2H), 7.26 (brs, IH), 7.33 (dd, J = 2.1 and 9.3 Hz, IH), 7.59-7.61 (m, 3H), 11.17 (t, J = 5.1 Hz, IH), 12.83 (brs, IH), 31P NMR (J6-DMSO, 121.49 MHz) δ 32.93, MS (ES+) m/z = 471.36 (MH+).
Figure imgf000178_0003
Compound structure Description
δ 3.75 Hz, IH), 2H),
18aab J= 5.4 Hz, MHz)
Figure imgf000179_0001
-118.41 (m, IF), MS (ES+) m/z = 456.8 (MH+). δ 2.67- 11.7 Hz, Hz, IH), lSaac (m, 2H), NMR
Figure imgf000179_0002
= 447.92 (MH+).
Pale yellow powder, 1H NMR (4-DMSO, 300 MHz) δ 3.78 (d, J = 11.7 Hz, 3H), 4.54-4.74 (m, 2H), 7.16-7.28 (m, 2H), 7.28-7.65 (m, 7H), 7.74-7.75 (m, IH), 10.84 (brs,
18aad IH), 12.95 (brs, IH), 31P NMR (J5-DMSO, 121.49 MHz) δ 29.36, 19F NMR (J5-DMSO, 282.40 MHz) δ -138 (m, IF), -126.5 (m, 2F), MS (ES+) m/z = 493 (MH+).
Figure imgf000179_0003
δ 3.64 (s, 6.91 7.67-7.68
18aae NMR IP), 2F), MS
Figure imgf000179_0004
Compound structure Description
δ 2.39 5.4 Hz, (m, J = 5.4 MS
Figure imgf000180_0001
Pale yellow powder, 1H NMR (CDCl3, 300 MHz) δ 2.39 (s, 3H), 3.72 (s, 3H), 3.84 (d, J = 11.4 Hz, 3H), 4.71 (d, J = 5.4 Hz, 2H), 7.12 (s, IH), 7.32 (dd, J= 1.5 and 8.7 Hz, IH), 7.47-7.59 (m, 4H), 1.66-1.1 A (m, 2H), 11.21 (brs,
ISaag IH), 11.38 (t, J = 5.4 Hz, IH), 31P NMR (CDCl3, 121.49 MHz) δ 31.21, MS (ES+) m/z = 482.38 (MH+).
Figure imgf000180_0002
δ 2.24 (s, and 15.3 (m, (dd, J =
ISaaj (t, J= 121.49
Figure imgf000180_0003
δ 2.26 (s, and 15.3 (brs, (m, 2H), Hz, IH), (dg- = 501 (MH+)
Figure imgf000180_0004
Compound structure Description δ 2.24 = 4.8 and 7.22- 7.5-7.56
18aal J= 1.8 (m, 31P NMR
Figure imgf000181_0001
= 535 (MH+) δ 2.26 (s, and 15.9 (brs, IH), IH), 7.65 IH), (brs, MS
Figure imgf000181_0002
White powder, 1H NMR (dg-DMSO, 300 MHz) δ 2.26 (s, 6H), 3.69 (d, J= 11.7 Hz, 3H), 4.65 (ddd, J= 5.7 and 15.3 and 21.6 Hz, 2H), 7.21 (brs, IH), 7.25 (brs, IH), 7.3 (brs, IH), 7.37-7.45 (m, 2H), 7.46 (dd, J= 1.5 and 8.7 Hz, IH),
ISaan 7.82 (td, J = 1.8 and 7.8 Hz, IH), 8.49 (dd, J = 1.95 and 4.65 Hz, IH), 8.64 (d, J = 1.8 Hz, IH), 11.81 (t, J = 5.7 Hz, IH), 13.06 (brs, IH), 31P NMR (d6-DMSO, 121.49 MHz) δ 32.10, MS (ES+) m/z = 486 (MH+)
Light yellow solid, 1H NMR (d6-DMSO, 300 MHz) δ 3.75 (d, J = 11.4 Hz, 3H), 5.67 (d, J = 7.5 Hz, IH), 7.2 (brs, IH), 7.85 (dd, J= 1.8 and 8.7 Hz, IH), 7.85-7.95 (m, 3H),
ISaao 8.02-8.16 (m, 6H), 8.22 (d, J = 1.8 Hz, IH), 8.28 (brs, IH), 8.46-8.53 (m, 2H), 11.47 (d, J = 7.5 Hz, IH), 12.76
Figure imgf000181_0003
(brs, IH), 31P NMR (ds-DMSO, 121.49 MHz) δ 31.79, MS (ES+) PiZz = 482 (MH+) Compound structure Description
δ 3.80 (brs, (m, 5H), lδaap (brs, NMR
Figure imgf000182_0001
3.77 (d, J (d, J = 7.32
ISaaq 7.73 (dd, (brs,
Figure imgf000182_0002
121.49 MHz) δ 31.86, MS (ES+) m/z = 374.83 (MH+) δ 3.84 Hz, IH), and 8.7
ISaar 11.63 (brs, 121.49
Figure imgf000182_0003
ISaas
ISaat
Figure imgf000182_0004
Compound structure Description
Colorless oil, 1H NMR (CDCl3, 300 MHz) δ 3.76 (d, J = 11.1 Hz, 3H), 7.44-7.58 (m, 3H), 7.76-7.84 (m, 2H), 31P NMR (CDCl3, 101.256 MHz) δ 22.2.
Colorless oil, 1H NMR (CDCl3, 300 MHz) δ 3.81 (d, J = 11.1 Hz, 6H), 7.59-7.66 (m, IH), 7.84-7.87 (m, IH), 8-8.1
Figure imgf000183_0001
(m, 2H), 31P NMR (CDCl3, 121.49 MHz) δ 17.94, MS (ES+) TnZz = 212 (MH). (t, J = J = 11.1 MS (ES+)
Figure imgf000183_0002
MHz) δ 3.77 (d, J = 11.1 Hz,
19d 66 (m, 2H), MS (ES+)
Figure imgf000183_0003
Colorless oil, 1H NMR (CDCl3, 300 MHz) δ 3.78 (d, J = 11.4 Hz, 6H), 7.33-7.4 (m, IH), 7.69-7.77 (m, 2H), 7.91-
Figure imgf000183_0004
7.96 (m, IH), 31P NMR (CDCl3, 121.49 MHz) δ 19.2, MS (ES+) m/z = 265/267 (MH).
Colorless oil, 1H NMR (CDCl3, 300 MHz) δ 3.79 (d, J = 11.1 Hz, 6H), 5.44 (d, J= 47.4 Hz, 2H), 7.5-7.63 (m, 2H),
19f 7.78-7.85 (m, 2H), 31P NMR (CDCl3, 121.49 MHz) δ 22,
Figure imgf000183_0005
19F NMR (CDCl3, 282.40 MHz) δ -209.94 (t, J= 47.4 Hz, IF), MS (ES+) m/z = 219 (MH).
Yellowish oil, 1H NMR (CDCl3, 300 MHz) δ 2.41 (s, 3H), 3.77 (d, J = 11.1 Hz, 6H), 7.06-7.1 (m, IH), 7.24-
19g 7.32 (m, IH), 7.4-7.44 (m, IH), 31P NMR (CDCl3, 121.49
Figure imgf000183_0006
MHz) δ 20.13 (d, J = 9.7 Hz, IP), 19F NMR (CDCl3, 282.40 MHz) δ -112.5 (m, IF).
Colourles oil, 1H NMR (J5-DMSO, 300 MHz) δ 0.89 (t, J = 7.5 Hz, 3H), 1.6 (sextuplet, J= 7.5 Hz, 2H), 2.63 (t, J=
19h 7.5 Hz, 2H), 3.65 (d, J= 11.4 Hz, 3H), 3.654 (d, J= 11.1 Hz, 3H), 7.47-7.50 (m, 3H), 7.54-7.55 (m, IH), 31P NMR
Figure imgf000183_0007
(J6-DMSO, 121.49 MHz) δ 21.04, MS (ES+) m/z = 229.3 Compound structure Description
(MH+).
Colorless oil, 1H NMR ((I6-OMSO, 300 MHz) δ 1.18 (t, J
O
Il
P-OMe = 7.5 Hz, 3H), 2.35 (s, 3H), 2.63 (q, J= 7.5 Hz, 2H), 3.64
19i OMe (d, J = 11.1 Hz, 3H), 3.65 (d, J= 11.1 Hz, 3H), 7.31 (brs,
V 2H), 7.45-7.48 (m, IH), 31P NMR (J6-DMSO, 121.49 MHz) δ 21.40, MS (ES+) m/z = 229.3 (MH+).
Colorless oil, 1H NMR (J6-DMSO, 300 MHz) δ 0.68-0.74 (m, 2H), 0.97-1.03 (m, 2H), 2-2.07 (m, IH), 3.65 (d, J =
19j OMe 11.1 Hz , 3H), 7.29-7.33 (m, IH), 7.4-7.49 (m, 3H), 31P
Figure imgf000184_0001
NMR (J6-DMSO, 121.49 MHz) δ 21.05, MS (ES+) m/z = 227 (MH+). (J6-DMSO, 300 MHz) δ 2.37 (s, Hz, 3H), 7.51-7.72 (m, 3H), 31P 9 MHz) δ 18.44, MS (ES+) m/z =
Figure imgf000184_0002
(d, J= 6.9 (d, J = (m, 2H), 31P
Figure imgf000184_0003
Colorless oil, 1H NMR (CDCl3, 300 MHz) δ 3.8 (d, J = 11.1 Hz, 6H), 6.98-7.05 (m, IH), 7.27-7.36 (m, 2H), 31P NMR (CDCl3, 121.49 MHz) δ 17.87 (t, J= 9.96 Hz, IP),
Figure imgf000184_0004
19F NMR (CDCl3, 282.40 MHz) δ -107.27 (m, 2F). (d, J= 7.45-7.52 20.45, MS
Figure imgf000184_0005
Oil, 1H NMR (J6-DMSO, 300 MHz) δ 2.33 (s, 6H), 3.63 (d, J = 11.1 Hz, 6H), 7.28-7.29 (m, 2H), 7.32-7.34 (m, IH), 31P NMR (J6-DMSO, 121.49 MHz) δ 21.34, MS
Figure imgf000184_0006
(ESI, El+) m/z = 215 (MH+) Compound structure Description
Oil, 1H NMR (CDCl3, 300 MHz) δ 3.79 (d, J = 11.1 Hz, 6H), 7.25-7.3 (m, IH), 7.43-7.64 (m, 3H), 31P NMR
19q
Figure imgf000185_0001
(CDCl3, 101.256 MHz) δ 20 (d, J = 8.66 Hz, IP), 19F NMR (CDCl3, 282.4 MHz) δ -130.9 (m, IF), MS (ESI, Et) m/z = 205 (MH+). δ 3.65 (d, J = ILl 7.28-7.41 (m, 4H), 121.49 MHz) δ
Figure imgf000185_0002
Colorless oil; 1H NMR (CDCl3, 300 MHz) δ 3.71 and 3.74 (2s, 3H), 7.41-7.54 (m, 3H), 7.77-7.84 (m, 2H), 10.70 (brs, IH) ;31P NMR (CDCl3, 101 MHz) δ 22.3.
Colorless oil, 1H NMR (CDCl3, 300 MHz) δ 3.76 (d, J = 11.4 Hz, 3H), .7.57-7.64 (m, IH), 7.82-7.85 (m, IH),
Figure imgf000185_0003
7.99-8.1 (m, 2H), 8.9 (brs, IH), 31P NMR (CDCl3, 121.49 MHz) δ 18.13, MS (ES+) m/z = 197.9 (MH).
Colorless oil; 1H NMR (CDCl3, 300 MHz) δ 2.37 (s, 3H), 3.71 (d, J = 11.4 Hz, 3H), 7.32-7.35 (m, 2H), 7.58-7.64 (m, 2H), 10.73 (brs, IH), 3MS (ES+) m/z = 187 (MH).
Colorless oil, 1H NMR (CDCl3, 300 MHz) δ 3.71 (d, J = 11.1 Hz, 3H), 7.26-7.32 (m, IH), 7.65-7.95 (m, 4H), 31P
Figure imgf000185_0004
NMR (CDCl3, 121.49 MHz) δ 19.07, MS (ES+) m/z = 251/253 (MH+).
Colorless oil, 1H NMR (CDCl3, 300 MHz) δ 3.75 (d, J = 11.4 Hz, 3H), 5.42 (d, J= 47.4 Hz, 2H), 7.4-7.6 (m, 3H),
2Oe 7.8-7.85 (m, 2H), 31P NMR (CDCl3, 121.49 MHz) δ
Figure imgf000185_0005
21.42, 19F NMR (CDCl3, 282.40 MHz) δ -209.80 (t, J = 47.4 Hz, IF), MS (ES+) m/z = 205 (MH). (t, J = 2.6 (t, J =
2Of (m, 2H),
Figure imgf000185_0006
(MH+). Compound structure Description (J0-DMSO, 300 MHz) δ 2.36 (s, Hz, 3H), 7.47-7.64 (m, 3H), 31P 9 MHz) δ 14.79, MS (ES+) m/z =
Figure imgf000186_0001
Yellow pale oil, 1H NMR (CDCl3, 300 MHz) δ 1.25 (d, J = 6.9 Hz, 6H), 2.89-2.98 (m, IH), 3.72 (d, J = 11.4 Hz, 3H), 6.59 (brs, IH), 7.34-7.43 (m, 2H), 7.59-7.69 (m,
Figure imgf000186_0002
2H), 31P NMR (CDCl3, 121.49 MHz) δ 23.21. 1.24 (t, J J = 11.4
2Oi 31P NMR = 201
Figure imgf000186_0003
Yellow oil, 1H NMR (CDCl3, 300 MHz) δ 3.66 (d, J =
2Oj 11.4 Hz, 3H), 6.92-6.98 (m, IH), 7.24-7.31 (m, 2H), 7.64 (brs, IH), 31P NMR (CDCl3, 121.49 MHz) δ 16.92.
Figure imgf000186_0004
Oil, 1H NMR (CDCl3, 300 MHz) δ 2.37 (s, 3H), 3.71 (d, J = 11.4 Hz, 3H), 7.02-7.05 (m, IH), 7.24-7.32 (m, IH),
20k 7.36-7.41 (m, IH), 31P NMR (CDCl3, 121.49 MHz) δ 19.66 (d, J= 9.6 Hz, IP), 19F NMR (CDCl3, 282.40 MHz) δ -112.7 (q, J= 9.32 Hz, IF).
/ Yellow oil, 1H NMR (J5-DMSO, 300 MHz) δ 3.51 (d, J
O = 11.1 Hz, 3H), 3.79 (s, 3H), 7.12-7.28 (m, 3H), 7.39-
201 Il
P-OH 7.46 (m, IH), 31P NMR (J6-DMSO, 101.256 MHz) δ OMe 17.03, MS (ESI, El+) m/z = 203.2 (MH+).
Pale pink oil, 1H NMR (J15-DMSO, 300 MHz) δ 3.53 (d, J = 11.1 Hz, 3H), 7.37-7.62 (m, 4H), 31P NMR (Jn-DMSO,
20m
Figure imgf000186_0006
101.256 MHz) δ 15.11 (d, J= 8.77 Hz, IP), 19F NMR (J5- DMSO, 235.36 MHz) δ -111.8 (m, IF), MS (ESI, El+) m/z = 191 (MH+). Compound structure Description
Oil, 1H NMR (J6-DMSO, 300 MHz) δ 2.31 (s, 6H), 3.50
2On (d, J = 11.1 Hz, 3H), 7.20 (s, IH), 7.26 (s, IH), 7.31 (s, IH), 12.03 (brs, IH), MS (ESI, El+) m/z = 201 (MH4)
Figure imgf000187_0001
300 MHz) δ 3.51 (d, J = 11.4 4.59 (s, 2H), 7.3-7.71 (m, 9H), 31P MHz) δ 16.22, MS (ES+) m/z =
Figure imgf000187_0002
Compound structure Description
δ 3.87 (tt, J= 7.28-7.3 Hz and
22a (d, J = (brs,
Figure imgf000187_0003
(t, J =
8.5 Hz, IP), 19F NMR (CDCl3, 282.40 MHz) δ POSITIF, MS (ES+) m/z = 492.28 (MH+).
Light orange powder, 1H NMR (CDCl3, 300 MHz) δ 2.31 (s, 6H), 3.83 (d, J= 11.7 Hz, 3H), 4.6-4.77 (m, 2H), 7.16-7.37 (m, 6H), 7.46-7.49 (m, IH), 7.66- 7.67 (m, IH), 8.16-8.18 (m, IH), 8.48 (brs, IH), 11.31 (brs, IH), 31P NMR (CDCl3, 121.49 MHz) δ
Figure imgf000187_0004
34.5, MS (ES+) m/z = 484.43 (MH+). δ 2.33 (m, 2H), 7.26-
22c (m, IH),
Figure imgf000187_0005
(m, IH), 31P NMR (CDCl3, 121.49 MHz) δ 34.58, MS (ES+) Wz = 498.42 (MH+). Compound structure Description δ 2.26 J = 5.1 Hz, 7.57-
22d (m, IH), NMR (d6- (ES+)
Figure imgf000188_0001
2.34 (s, (m, 2H), Hz, IH), 1.8 and (m, (brs, 121.49
Figure imgf000188_0002
2.33 (s, = 5.4 Hz, Hz, IH), 2.1 and
22f (d, J = IH), (ES+)
Figure imgf000188_0003
δ 2.26 J = 5.1 Hz, (dd, J= 8.64 (t, J (t, J=
Figure imgf000188_0004
(MH+). Compound structure Description 1.20 (t, J Hz, 2H), and 15.5 7.19- (d, J
Figure imgf000189_0001
11.86 (t, J = 5.7 Hz, IH), 31P NMR (CDCl3, 121.49 MHz) δ 34.10, MS (ES+) m/z = 498 (MH+). MHz) δ (m, Hz, (m, 6.9 Hz, IH),
Figure imgf000189_0002
MHz) δ 32.67, MS (ES+) m/z = 485.35 (MH+). MHz) δ (m, IH),
22j 8.29 IH),
Figure imgf000189_0003
MS (ES+) m/z = 502.45 (MH+). δ 2.19 2H), 7.24 (d, J 11.79 δ 34.2,
Figure imgf000189_0004
Compound structure Description MHz) δ (s, 3H), (dd, J= IH),
221 9 Hz, 6.6 Hz, 3.6 Hz,
Figure imgf000190_0001
δ 32.64, MS (ES+) m/z = 482.43 (M+). δ 2.32 = 5.1 Hz,
22m Hz, IH), MS (ES+)
Figure imgf000190_0002
MHz) δ (dd, J= 15.9 Hz, (m, 6.9 Hz, 5.4 Hz,
Figure imgf000190_0003
121.49 MHz) δ 32.83, MS (ES+) m/z = 484.19 (MH+).
Off-white solid, 1H NMR (J6-DMSO, 300 MHz) δ 2.28 (s, 6H), 3.77 (d, J= 11.7 Hz, 3H), 4.64-4.74 (m, 2H), 7.24-7.47 (m, 7H), 7.57-7.64 (m, 2H), 8.34-
22p 8.36 (m, IH), 11.32-11.37 (m, IH), 12.86 (brs, IH), 31P NMR (J6-DMSO, 121.49 MHz) δ 32.67, MS (ES+) OT/-: = 484.5 (MH+).
Figure imgf000190_0004
Compound structure Description
Figure imgf000191_0001
Compound structure Description
Figure imgf000192_0001
Compound structure Description
Figure imgf000193_0001
Compound structure Description
Figure imgf000194_0001
Compound structure Description
Figure imgf000195_0001
Compound structure Description
Yellow powder, 1H NMR (CDCl3, 300 MHz) δ 2.4 (s, 3H), 3.86 (d, J = 12 Hz, 3H), 4.58 (dd, J = 5.4 and 15.6 Hz, IH), 4.73 (dd, J = 5.4 and 15.6 Hz, IH), 7.33-7.35 (m, 3H), 7.53-7.60 (m, 3H), 7.7-7.8 (m, 2H), 8.27-8.3 (m, 2H), 11.55 (t, J= 5.4 Hz, IH), 11.61 (brs, IH), 31P NMR (CDCl3, 121.49 MHz) δ 31.62, MS (ES+) m/z = 495.4 (MH+).
Colorless oil, 1H NMR (CDCl3, 300 MHz) δ 5.03- 5.2 (m, 2H), 7.3-7.47 (m, 5H), 7.48-7.63 (m, 3H),
Figure imgf000196_0001
23a 7.64 (d, J = 565.8 Hz, IH), 7.75-7.83 (m, 2H), 31P NMR (CDCl3, 101.256 MHz) δ 25.55, MS (ESI, El+) m/z = 233 (MH+). , 250 MHz) δ 1.59 (s, IH), 7.5-7.82 (m, 5H), 31P
Figure imgf000196_0002
δ 15.27, MS (ESI, El+) m/z = 198 (MH+). Compound structure Description
Colorless oil, 1H NMR Cd6-DMSO, 300 MHz) δ 2.38 (s, 3H), 3.68 (d, J= 12 Hz, 3H), 7.49 (d, J = 566 Hz,
Figure imgf000197_0001
IH), 7.4 (dd, J= 3 and 7.8 Hz, 2H), 7.62 (d, J= 13.8 Hz, IH), 7.64 (d, J= 13.5 Hz, IH)
Compound structure Description
J = 7.2 2H), 7.33 5H), 7.62- 2H), 31P El+) m/z =
Figure imgf000197_0002
(d, J = Hz and 9 (brs, MHz)
3.86 (d, J= IH), 7.42- 3H), 8.05
Figure imgf000197_0003
m/z = 365 (MH+).
Yellow powder, 1H NMR (CDCl3, 300 MHz) δ 1,45 (t, / = 7.2 Hz, 3H), 4.03-4.13 (m, IH), 4.25-4.36 (m, IH), 7.3 (dd, J= 1.95 Hz and 8.85 Hz, IH), 7.41-7.47 (m, 3H), 7.51-7.56 (m, IH), 7.72-7.79 (m, 3H), 8.01 (brs, IH), 10.59 (brs, IH),
Figure imgf000197_0004
12.59 (brs, IH), 31P NMR (CDCl3, 121.49 MHz) δ 32.32, MS (ESI, El+) m/z = 379 (MH+). Compound structure Description
(t, J = 7.2 2.1 and 9 8-8.04 (m, MS (ESI,
6.13 (d, J =
(brs, IH), (ESI,
Figure imgf000198_0001
1.36 (t, J= = 2.1 Hz 2H), 7.88- 101.256
2.24 (d, J = 8.7 Hz, (m, (J6-DMSO, (MH+).
(t, J = 6.5 6.7 Hz, 7.60 (m, NMR = 531
Figure imgf000198_0002
Compound structure Description
MHz) δ 8.7 Hz, (brs, IH), m/z = 384
Figure imgf000199_0001
δ 1.38- Hz, 2H), (m,
32a MS (ES+)
Figure imgf000199_0002
δ 1.38- Hz, 2H), (m,
32b MS (ES+)
Figure imgf000199_0003
300 MHz) δ 3.90 (d, J = (d, J = 16.5 Hz, IH), (brs, IH), 10.85 (brs, IH), δ 31.53, MS (ES+) m/z =
Figure imgf000199_0004
Figure imgf000199_0005
Compound structure Description
Figure imgf000200_0001
Compound structure Description
Figure imgf000201_0001
Compound structure Description
Figure imgf000202_0001
(t, /= 6.9 2H), 9.31
Figure imgf000202_0002
MHz) δ 12.37, MS (ESI, El+) m/z = 217 (MH+).
Yellow oil, 1H NMR (CDCl3, 300 MHz) δ 1.2 (t, J = 7.2 Hz, 6H), 3.95-4.1 (m, 4H), 7.25-7.3 (m, IH), 7.93-8 (m,
Figure imgf000202_0003
IH), 8.62-8.64 (m, IH), 8.83-8.85 (m, IH) 31P NMR (CDCl3, 121.49 MHz) δ 15.72.
O Yellow oil, 1H NMR (CDCl3, 300 MHz) δ 1.36 (t, / = 7.2
O Hz, 6H), 4.16-4.31 (m, 4H), 7.41-7.46 (m, IH), 7.77-7.85
47c " OHEt Et (m, IH), 7.96-8.01 (m, IH), 8.8-8.82 (m, IH) 31P NMR (CDCl3, 121.49 MHz) δ 7.16, MS (ESI, El+) m/z = 216 (MH+). J = 6.9
47d 8.67-8.71
Figure imgf000202_0004
MS Compound structure Description
(ESI, El+) m/z = 216 (MH+).
Oil, 1H NMR (CDCl3, 300 MHz) δ 1,33 (t, J= 7.2 Hz, 6H), 4.04-4.19 (m, 4H), 7.32-7.35 (m, IH), 7.41-7.45 (m, IH)5
Figure imgf000203_0001
7.97-8.01 (m, IH), 31P NMR (CDCl3, 121.49 MHz) δ 13.35, MS (ESI, El+) m/z = 221.11 (MH+).
Yellow pale oil, 1H NMR (CDCl3, 300 MHz) δ 1,34 (t, J = 7.05 Hz, 6H), 4.06-4.22 (m, 4H), 7.16-7.20 (m, IH), 7.65- 7.72 (m, 2H), 31P NMR (CDCl3, 121.49 MHz) δ 12.05, MS (ESI, El+) m/z = 221.11 (MH+).
Yellow oil, 1H NMR (CDCl3, 300 MHz) δ 1,27 (t, /= 7.05 Hz, 3H), 4.01-4.07 (m, 2H), 7.31-7.4 (m, 2H), 7.91-7.95
Figure imgf000203_0003
(m, IH), 12.73 (s, IH), 31P NMR (CDCl3, 101.256 MHz) δ 14.67.
Yellow oil, 1H NMR (CDCl3, 300 MHz) δ 1,31 (t, J= 7.05 Hz, 3H), 4.04-4.14 (m, 2H), 7.12-7.16 (m, IH), 7.61-7.68
Figure imgf000203_0004
(m, 2H), 9.12 (brs, IH), 31P NMR (CDCl3, 121.49 MHz) δ 14.7. δ 1.41 (t, J (m, 7.16-7.19 (m, (m, (m,
Figure imgf000203_0005
Yellowish powder, 1H NMR (CDCl3, 300 MHz) δ 1.38 (t, J = 7.05 Hz, 3H), 1.42 (t, J = 7.05 Hz, 3H), 4.08-4.26 (m, 2H), 4.52 (q, J= 7.05 Hz, 2H), 7.34 (dd, J = 2.1 Hz and 9
50b Hz, IH), 7.35-7.41 (m, IH), 7.47-7.53 (m, 2H), 7.58-7.64 (m, IH), 7.77-7.84 (m, IH), 7.88-7.92 (m, IH), 8.03-8.15
Figure imgf000203_0006
(m, 4H), 8.72-8.74 (m, IH), 31P NMR (CDCl3, 121.49 MHz) δ 19.72. Compound structure Description
Hz, 4.52 (q, J 7.39 (dd,
50c (m, 9 Hz, MHz) δ
Figure imgf000204_0001
White powder, 1H NMR (J6-DMSO, 300 MHz) δ 1.33 (t, J = 7.05 Hz, 3H), 4.03-4.09 (m, IH), 4.15-4.24 (m, IH), 7.21-7.23 (m, IH), 7.34 (dd, J= 2.1 and 8.7 Hz, IH), 7.55- 7.61 (m, 2H), 7.7 (d, J = 2.1 Hz, IH), 8-8.03 (m, 2H), 10.12 (brs, IH), 12.76 (brs, IH), 31P NMR (J6-DMSO,
Figure imgf000204_0002
121.49 MHz) δ 23.46, MS (ES+) m/z = 368.8 (MH+). (d, J = (m, J = 1.8 Hz, IH), 353.06
Figure imgf000204_0003
δ 3.77 (d, J and 8.7
52b IH), 8.01- (ES+) m/z
Figure imgf000204_0004
Colorless oil, 1H NMR (CDCl3, 300 MHz) δ 1.66 (d, J = 3.9 Hz, 3H), 1.76 (d, J = 5.4 Hz, 3H), 2.57 (dd, J = 7.65
53a
Figure imgf000204_0005
and 21.75 Hz, 2H), 3.74 (d, J= 10.5 Hz, 6H), 5.12-5.21 (m, IH), 31P NMR (CDCl3, 121.49 MHz) δ 7.32, MS (ESI, El+) m/z = 179 (MH+). Compound structure Description
Thick yellow oil, 1H NMR (CDCl3, 300 MHz) δ 1,24 (d, J = 3.9 Hz, 3H), 1,5 (t, J = 7.2 Hz, 3H), 1.54 (d, J = 6 Hz, 3H), 2.78 (dd, J = 8.1 and 19.5 Hz, 2H), 3.7 (d, J = 11.1 Hz, 3H), 4.57 (q, J = 7.2 Hz, 2H), 5.07-5.1 (m, IH), 7.37
56a (dd, J = 2.1 and 8.7 Hz, IH), 7.5-7.55 (m, 2H), 7.63-7.67 (m, IH), 7.87 (d, J= 1.8 Hz, IH), 7.96 (dd, J = 1.2 and 9 Hz, IH), 8.1-8.13 (m, 2H), 31P NMR (CDCl3, 121.49 MHz) δ 8.68, MS (ESI, El+) m/z = 511 (MH+).
Figure imgf000205_0001
1.03-1.38 3.59 (d, J = 7.59 (d, J (brs, MHz)
Figure imgf000205_0002
Pale yellow solid, 1H NMR (J15-DMSO, 300 MHz) δ 1.17 (d, J= 3.6 Hz, 3H), 1.55 (d, J= 5.7, Hz, 3H), 2.81 (m, 2H), 3.62 (d, J = 11.1 Hz, 3H), 5.0 (m, IH), 7.33 (dd, J = 2.0 and 8.7 Hz, IH), 7.57 (d, J= 8.7 Hz, IH), 7.68 (d, J = 2.0 Hz, IH), 7.85 (brs, IH), 7.87 (brs, 1H),1O.1O (brs, IH), 12.64 (brs, IH), MS (ESI, El+) m/z = 341 (MH+).
Yellow powder, 1H NMR (CDCl3, 300 MHz) δ 3.88 (d, J= 12 Hz, 3H), 4.4 (d, J= 5.4 Hz, 2H), 6.98 (tt, J= 2.4 Hz and 8.7 Hz, IH), 7.26-7.31 (m, 2H), 7.38 (dd, J = 1.8 Hz and 8.7 Hz, IH), 7.59 (d, J= 1.5 Hz, IH), 7.46 (dd, J= 1.8 Hz and 8.7 Hz, IH), 11.24 (brs, IH), 11.79 (t, J= 5.4 Hz, IH),
Figure imgf000205_0003
31P NMR (CDCl3, 121.49 MHz) δ 30.82 (t, J= 8.6 Hz, IP), 19F NMR (CDCl3, 282.40 MHz) δ -106.02 (m, 2F), MS (ES+) m/z = 424.04 (MH+). Compound structure Description 2.8 (t, J = Hz, 3H), (m, 2H),
59b 8.7 Hz, (t, J=
Figure imgf000206_0001
-106.25 (m, 2F), MS (ES+) m/z = 437.96 (MH+).
White solid, 1H NMR (J6-DMSO, 300 MHz) δ 7.14-7-17 (m, IH), 7.32-7.73 (m, 7H), 8.16 (brs, IH), 11.78 (brs, IH),
60a 12 (brs, IH), MS (ESI, El+) m/z 335 (MH+).
White powder, 1H NMR (J6-DMSO, 300 MHz) δ 3.72 (s, 3H), 6.9-6.93 (m, 2H), 7.19 (dd, J = 1.5 Hz and 8.7 Hz,
60b IH), 7.43-7.46 (m, IH), 7.58-7.69.(m, 3H), 8-8.03 (m, IH), 11.25 (brs, IH), 12.15 (brs, IH), 31P NMR (J6-DMSO, 121.49 MHz) δ 31.44, MS (ESI, El+) m/z = 365 (MH+).
Off white powder, 1H NMR (J6-DMSO, 400 MHz) δ 2.3 (s, 3H), 7.26 (dd, J = 2 and 8.8 Hz, IH), 7.3-7.37 (m, 2H),
6Od 7.46-7.54 (m, 3H), 7.82 (brs, IH), 7.88 (brs, IH), 10.69 (brs, IH), 12.49 (brs, IH), 31P NMR (J6-DMSO, 101.256
Figure imgf000206_0002
MHz) δ 23.81, MS (ESI, El+) m/z = 349 (MH+).
Off white powder, 1H NMR (J15-DMSO, 300 MHz) δ 2.21 (s, 6H), 7.2-7.28 (m, 2H), 7.37-7.53 (m, 3H), 7.79 (brs, IH), 7.87 (brs, IH), 10.68 (brs, IH), 12.47 (brs, IH), MS (ESI, El+) m/z = 363 (MH+).
Figure imgf000206_0003
Compound structure Description
δ 7.27 7.61- 10.54
6Og 101 MHz) -133.8 (m,
Figure imgf000207_0001
The following examples are provided to illustrate the present invention, and are in no way intended to limit the scope of the invention.
EXAMPLES
X. Compound synthesis
General Synthetic Method
Figure imgf000208_0001
Figure imgf000208_0002
Experimental :
The pyridine precursor was dissolved in chloroform (or in CH2Cl2) under stirring at room temperature; m-chloroperoxybenzoic acid was added and the reaction allowed to stir overnight (~15H).
The mixture was diluted with dichloromethane and extracted with a mixture of saturated K2CO3 / H2O (1/3).
The aqueous layer was extracted three times with dichloromethane. The combined organic layer was dried over sodium sulphate, filtered and concentrated under reduced pressure. (TLC: dichloromethane/methanol = 9/1). The crude product was then purified by chromatography.
Compounds synthesised:
Figure imgf000209_0001
Figure imgf000210_0001
Figure imgf000211_0003
Example 1. Ethyl S-bromo-S-chloro-l-CphenvIsuIfonylVlH-indole-Z-carboxylate
a) Synthesis of ethyl 5-chloro-l-(phenylsulfonyl)-lH-indole-2-carboxylate as starting material (based on work of Silvestri R., De Martino G., La Regina G., Artico M., Massa S., Vargiu L., Mura M., Loi A.-G., Marceddu T., La Colla P. J. Med. Chem. 2003, 4(5:2482-2493):
Figure imgf000211_0001
To a stirred and cooled (00C) solution of ethyl-5-chloroindole-2-carboxylate (1.052g, 4.70 mmol) in DMF (25 mL) under N2, was added NaH (60% in oil, 230 mg, 5.64 mmol) portionwise. After the end of gas evolution, phenylsulfonyl chloride (0.72 mL, 5.64 mmol) was added. The reaction mixture was stirred for 1 H (TLC monitoring, eluant dichloromethane). A little amount of water was added carefully and DMF was evaporated. Crude residue was dissolved in EtOAc and washed with water and brine. After drying and evaporation of solvents the compound was purified by chromatography on silica gel (eluant: cyclohexane/EtOAc 9/1 to 7/3) to afford protected indole (1.547 g,
90% yield. Off-white solid; 1H NMR tø-DMSO) δ 1,30 (t, J= 7.2 Hz, 3H), 4.35 (q, J = 7.2 Hz, 2H), 7.37 (s, IH), 7.53 (dd, J = 2.2 and 9.1 Hz), 7.62-7.77 (m, 3H), 7.80 (d, J = 2.2 Hz, IH), 7.99 (m, 2H), 8.06 (d, J= 9.1 Hz); MS (ESI, El+) m/z = 364 (MH+).
b) Synthesis of ethyl 3-bromo-5-chloro-l-(phenylsulfonyl)-lH"-indole-2- carboxylate intermediate:
Figure imgf000211_0002
To a stirred solution of ethyl 5-chloro-l-(phenylsulfonyl)-lH-indole-2- carboxylate (4.83 g, 13.27 mmol) in DMF (40 mL) under N2, was added a solution of bromine (1.3 mL, 26.54 mmol) in DMF (10 mL). Reaction media was stirred at RT 4Η, water was added (150 mL) and was extracted with dichloromethane (3x100 mL). Organic layer was washed with a saturated solution OfNa2SO5, dried and evaporated to give a crude yellow oil. Purification by chromatography on silica gel (eluant: cyclohexane/EtOAc 9/1) afforded 3-brominated indole (5.548 g, 93% yield). Off white solid; 1H NMR tø-DMSO) δ 1,37 (t, J= 7.2 Hz, 3H), 4.48 (q, J= 7.2 Hz, 2H), 7.59-7.68 (m, 4H), 7.77 (m, IH), 7.96-8.09 (m, 3H) ; MS (ESI, El+) m/z = 442-444 (MH+).
Method A: Typical procedure for the synthesis of Ethyl 5-chloro-3- (dialkoxyphosphoryl)-l-(phenylsulfonyl)-l-H-indole-2-carboxylate and Ethyl S- chloro-3-falkoxy(phenyI)phosphoryll-l-(phenylsuIfonyl)-lfl-indole-2-carboxylate
To a stirred and cooled (-900C) solution of ethyl 3-bromo-5-chloro-l- (phenylsulfonyl)-lH-indole-2-carboxylate ( 0.50 mmol) in anhydrous THF (2.5 mL) under N2, was added n-BuLi (2.5M in hexanes, 0.24 mL, 0.60 mmol) dropwise. After 5 min at -900C, appropriate chorophosphosphus reagent (0.60 mmol) was added dropwise at the same temperature. The reaction was allowed to warm up to RT over 3 H (TLC monitoring, eluant dichloromethane/ ETOAc 9/1). Water was then added (5 mL). Extraction with EtOAc (3x 20 mL) drying and evaporation led to a crude oil that was purified by chromatography on silica gel.
Example 2. Ethyl 5-chloro-3-(diethoxyphosphoryl)-l-(phenylsuIfonyl)-ljHr-indole-2- carboxylate
Figure imgf000212_0001
Method A: Purification by chromatography on silica gel (eluant: dichloromethane/EtOAc 9/1 to 8/2) afforded desired indole (176 mg, 71% yield). White solid; 1H NMR (J6-DMSO, 300 MHz) δ 1,19 (t, J= 7.1 Hz, 6H), 1,39 (t, J= 7.1 Hz, 3H), 3.99-4.09 (m, 4H), 4.46 (q, J= 7.1 Hz, 2H), 7.77 (dd, J= 2.1 and 8.7 Hz, IH), 7.67-7.82 (m, 4H), 8.07-8.12 (m, 2H); 31P NMR (J6-DMSO, 101 MHz) δ 9.7; MS (ESI, El+) m/z =
500 (MH+).
Example 3. Ethyl S-chloro-S-fethoxyCphenvDphosphoryli-l-rphenylsulfonvD-li?- mdoIe-2-carboxγlate a) Synthesis of ethyl hydrogen phenylphosphonochloridate intermediate was synthesized according to Smith A.B. HI, Ducry L., Corbett, R.M., Hirschmann R. Org.
Lett. 2000, 2:3887-3890:
i) Synthesis of diethyl phenylphosphonate:
Figure imgf000213_0001
Colorless oil; 1H NMR (CDCl3, 250 MHz) δ 1,33 (t, J= 7.1 Hz, 6H), 4.05-4.25 (m, 4H), 7.46-7.57 (m, 3H), 7.78-7.87 (m, 2H) ;31P NMR (CDCl3, 101 MHz) δ 19.3.
ii) Synthesis of ethyl hydrogen phenylphosphonate:
Figure imgf000213_0002
Colorless oil; 1H NMR (CDCl3, 300 MHz) δ 1,32 (t, J= 7.3 Hz, 3H), 4.08 (q, J= 7.3 Hz, 2H), 7.42-7.56 (m, 3H), 7.79-7.86 (m, 2H)5 10.67 (bra, IH) ;31P NMR (CDCl3, 101 MHz) δ 21.3;
iii) Synthesis of ethyl hydrogen phenylphosphonochloridate having the following physical characteristics:
Figure imgf000213_0003
31P NMR (CDCl3, 101 MHz) δ 10.20 and 10.24. b) Synthesis of final product Ethyl 5-chloro-3-[ethoxy(phenyl)phosphoryi]-l- (phenylsulfonyl)-lH-indole-2-carboxylate:
Figure imgf000214_0001
Method A: Purification by chromatography on silica gel (eluant: dichloromethane/EtOAc 9/1) afforded debrominated indole(318 mg) and then desired indole (326 mg, 41% yield). Colorless oil; 1H NMR (J6-DMSO, 300 MHz) δ 1,27 (t, J = 7.1 Hz, 3H), 1,36 (t, J= 7.1 Hz, 3H), 4.03 (m, 2H), 4.38 (q, J= 7.1 Hz, 2H), 7.51-7.83 (m, HH), 8.05-8.11 (m, 3H); 31P NMR (J6-DMSO, 101 MHz) δ 23.3; MS (ESI, El+) m/z = 532 (MH+).
Example 4. Ethyl 5-chloro-3-fethoxy(3,5-dimethyIphenγl))phosphorγIl-l-
(phenyIsulfonviyiffi-indole-2-carboxylate
a) Synthesis of diethyl 3,5-dimethylphenylphosphonate intermediate according to method described by Hirao T., Masunaga T., Oshiro Y., Agawa T. Synthesis 1981, 56-
57.
0
Il
Vi"! P-OEt OEt
Purification by chromatography on silica gel (eluant: cyclohexane/EtOAc 6/4) afforded product (1.625 g, 61% yield) as a colorless oil; 1H NMR (CDCl3, 300 MHz) δ 1,33 (t, J= 7.0 Hz, 6H), 2.35 (s, 6H), 4.02-4.18 (m, 4H), 7.18 (s, IH), 7.40 (s, IH), 7.45 (s, IH) ;31P
NMR (CDCl3, 101 MHz) δ 20.3. Other intermediates are synthesized according to Example 3 (i, ii and iii).
b) Synthesis of Ethyl 5-chloro-3-[ethoxy(3,5-dimethylphenyl))phosphoryl]-l- (phenylsulfonyl)-li7-indole-2-carboxylate:
Figure imgf000215_0001
Method A: Purification by chromatography on silica gel (eluant: dichloromethane/EtOAc 95/5) afforded product (750 mg, 56% yield) as a light yellow solid. 1H NMR (J15-DMSO, 300 MHz) δ 1,27 (t, J= 7.1 Hz, 3H), 1,36 (t, J= 7.1 Hz, 3H), 2.30 (s, 6H), 3.94-4.06 (m, 2H), 4.44 (q, J= 7.1 Hz, 2H), 7.25 (s, IH), 7.39 (s, IH), 7.42 (s, IH), 7.53 (dd, J= 2.1 and 9.0 Hz, IH), 7.65-7.71 (m, 2H), 7.77-7.82 (m, 2H), 8.05- 8.11 (m, 3H); 31P NMR (J6-DMSO, 101 MHz) δ 23.6; MS (ESI, El+) m/z = 560 (MH+).
Example 5. Ethyl S-chloro-S-fmethoxyfphenvπphosphoryn-l-CphenylsulfonvD-lg- indole-2-carboxyIate
a) synthesis of dimethyl phenylphosphonate:
Figure imgf000215_0002
To a solution of benzenephosphonyl dichloride (1 mL, 6.35 mmol) in anhydrous dichloromethane (25 mL) was added dropwise at 0°C ethanol (1,12 mL, 19.04 mmol) following by triethylamine (2,65 mL, 19.04 mmol). The raction mixture was stirred at RT 2h. The reaction mixture was washed with a solution of HCl IN (50 mL). Aqueous layer was extracted with dichoromethane. Combined organic phases were dried and concentrated under reduce pressure. The crude oil was purified by chromatography on silica gel(Eluant: cyclohexane/EtOAc 6/4) to afford dimethyl phenylphosphonate as colorless oil (1,11Og, 82% yield). 1H NMR (CDCl3, 300 MHz) δ 3.76 (d, J = 11.1 Hz, 3H), 7.44-7.58 (m, 3H), 7.76-7.84 (m, 2H) ;31P NMR (CDCl3, 101 MHz) δ 22.2.
Other intermediates are synthesised according to example 3 (i, ii and iii).
b) synthesis of final product Ethyl 5-chloro-3-[methoxy(phenyl)phosphoryl]-l- (phenylsulfonyl)-lH-indole-2-carboxylate:
Figure imgf000216_0001
Method A: Colorless oil; 1H NMR (CDCl3, 300 MHz) δ 1.45 (t, J= 7.2 Hz, 3H), 3.80 (d, J = 11.4 Hz,3H), 4.54 (q, J = 7.2 Hz, 2H), 7.36 (dd, J = 2.1 and 9.0 Hz, IH), 7.47-7.67 (m, 6H), 7.84-7.96 (m, 4H), 8.09-8.12 (m, 2H); 31P NMR (CDCl3, 101 MHz) δ
26.7; MS (ESI, El+) m/z = 518 (MH+).
Example 6. Ethyl 2-(aminocarbonyl)-5-chloro-liy-mdol-3-yl-(phenyI)phosphinate
Figure imgf000216_0002
Ethyl-5-chloro-3-[ethoxy(phenyl)phosphoryl]-l-(phenylsulfonyl)-lH-indole-2- carboxylate (268 mg, 0.50 rnmol) was dissolved in a saturated solution of ammonia in methanol (5 mL) in a pressure tube. The tube was heated under microwave irradiation under pressure at 65 °C (Maximum power input 10OW, CEM discover apparatus) for 2 Η. After evaporation of solvents, purification by chromatography on silica gel (eluant: dichloromethane/MeOΗ 95/5 to 9/1) afforded desired carboxamide indole (107 mg, 81% yield). White solid; 1H NMR (J6-DMSO, 300 MHz) δ 1,34 (t, J= 7.1 Hz, 3H), 4.05 (m, IH), 4.20 (m, IH), 7.32 (dd, J = 2.1 and 8.7Hz, IH), 7.49-7.61 (m, 5H)3 7.68-7.75 (m, 2H), 8.02 (brs, IH), 10,27 (brs, IH), 12.77 (brs, IH); 31P NMR (J6-DMSO, 101 MHz) δ 31.1; MS (ESI, El+) m/z = 363 (MH+).
Example 7. Ethyl Z-CaminocarbonvD-S-chloro-lff-indol-S-yl-O^-dimethylphenyl) phosphonate
Figure imgf000217_0001
Same procedure as described in exemple 5. white solid; 1H NMR (J6-DMSO, 300 MHz) δ 1,32 (t, J= 7.0 Hz, 3H), 2.26 (s, 6H), 3.90-4.03 (m, IH), 4.09-4.22 (m, IH), 7.21
(s, IH), 7.29-7.33 (m, 3H), 7.57 (dd, J = 1.8 and 9.0 Hz, IH), 7.60 (dd, J = 1.8 Hz,
IH), 7.99 (brs, IH), 10.3 (brs, 1H),12.7 (brs, IH); 31P NMR (J6-DMSO, 101 MHz) δ
31.3; MS (ESI, El+) m/z = 391 (MH+).
Example 8. Methyl 2-(aminocarbonyl)-5-chloro-lH-indol-3-yl-(phenyl)pliosphinate
Figure imgf000217_0002
Same procedure as described in exemple 5. Pale yellow powder; 1H NMR (CDCl3, 300 MHz) δ 3.85 (d, J= 11.4 Hz, 3H), 6.08 (broad s, IH), 7.30 (dd, J= 2.0 and 9.0 Hz, IH), 7.36-7.56 (m, 4H), 7.68 (d, J = 1.8 Hz, IH), 7.73-7.81 (m, 2H), 10.78 (broad s, IH), 10.03 (broad s, IH); 31P NMR (CDCl3, 101 MHz) δ 33.3; MS (ESI, El+) m/z = 349 (MH+). Example 9: Biological Activity Against Drug Resistant Strains of HIV
In one embodiment, the efficacy of an anti-HIV compound is measured in vitro by a rapid, sensitive, and automated assay that involves the reduction of 3-(4,5- dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT). An HIV-transformed cell line that is highly permissive and selective for HIV infection, such as, for example, the T-4 cell line, MT-4, is chosen as the target cell line (Koyanagi et al., Int. J. Cancer, 1985, 55:445-451). In situ reduction of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl- tetrazolium bromide (MTT) as assessed spectrophotometrically is the standard by which the viability of both mock-infected cells and HIV-infected cells is measured. Inhibition of the HlV-induced cytopathic effect serves as the end-point. A 50% cytotoxic concentration (CC50 in uM) is defined as the concentration of compound that reduces the absorbance of the mock-infected control sample by 50%. The percent efficacy of an anti-HIV compound is calculated by the formula (expressed as a %):
(ODHΓV test compound) — (ODContro0/(ODmock infected cells)" (ODcontrol) Here, (ODHIV test compound) is the optical density measured for a specific amount of a test compound in HIV-infected cells; (ODcontroi) is the optical density measured for untreated HIV-infected, control cells; and (ODm0Ck infected ceils) is the optical density measured for control, mock-infected cells that are untreated. Optical density values typically are assessed at 540 nm. The dosage of an anti-HIV test compound that provides 50% protection according to the preceding formula is defined as the 50% inhibitory concentration (IC50 in μM). The selectivity index (SI) is defined as the ratio of the CC50 to the IC50.
In another embodiment, the p24 ELISA assay is used to determine the efficacy of an anti-HIV compound. This viral replication immunoassay measures the amount of p24 viral capsid (core) antigen present, and is available commercially from sources such as, for example, Coulter Corporation/Immunotech, Inc.® (Westbrook, MI).
Still other embodiments include a reverse trancriptase assay in which the amount of viral replication is measured by utilizing a homopolymer poly rA:oligo dT template primer system that quantifies the incorporation into cells of tritiated thymidine monophosphate by scintillation counting methods (Southern Research Institute,
University of Alabama, Birmingham, AL); a syncytial inhibition assay that employs CEM-SS, HeLa-CD4, or HeLa-CD4-LTR-b-galactosidase cells having an immuno- fluorescent, chemiluminescent, or colorimetric endpoint; and an attachment-and fusion- inhibition assay that utilizes indicator cell lines and quantitation by chemiluminescent, colorimetric or microscopic evaluation (Southern Research Institute, University of Alabama, Birmingham, AL).
In one embodiment the indole compounds of the present invention do not exhibit cross resistance with other non-nucleoside reverse transcriptase inhibitors (NNRTIs), in that the compounds of the present invention display an EC50 (in molar concentration) in a mutant HIV strain of less than approximately 50, 25, 10 or 1 μM concentration. In a typical embodiment, the NNRTIs display an EC50 in a mutant HIV strain of less than approximately 5, 2.5, 1 or 0.1 μM concentration. The degree of cross-resistance against a drug resistant strain of HIV is measured by assessing the EC50 of the desired oxo- pyrimidine compound in the target mutated, i.e., drug resistant, virus.
Therefore, in another important embodiment of this invention, a method for treating a patient with a cross-resistant HIV is provided that includes administering an effective HlV-treatment amount of an indole compound, a salt, prodrug, stereoisomer or tautomer thereof.
Biological Activity Against Drug Resistant Strains of HIV
In one embodiment the phenylindoles of the present invention do not exhibit cross resistance with other non-nucleosides reverse transcriptase inhibitors (NNRTI), in that it displays an ECs0 (in molar concentrations) in a mutant HIV strain of less than approximately 50, 25, 10 or 1 micromolar concentration. In a typicial embodiment, the non-nucleosides reverse transcriptase inhibitors (NNRTI) displays an EC50 (in molar concentrations) in a mutant HIV strain of less than approximately 5, 2.5, 1 or 0.1 micromolar concentration. The degree of cross-resistance against a drug resistant strain of HIV can easily be measured by assessing the EC50 of the desired indole in the target mutated i.e., drug resistant, virus.
Therefore, in another important embodiment of this invention, a method for treating a patient with a cross-resistant HIV is provided that includes administering an effective HlV-treatment amount of a phenylindole or its prodrug or salt. MISSING OPEN FILING
Figure imgf000221_0001
TABLE 3 - ENZYME ASSAY DATA
κ> κ>
Figure imgf000221_0002
K)
Figure imgf000222_0001
TABLE 4 - CELL-BASED ASSAY DATA USING BH-IO CELLS
K) κ>
Figure imgf000223_0001
Figure imgf000224_0001
K)
Figure imgf000225_0001
K) Ul
Figure imgf000226_0001
K)
ON
Figure imgf000227_0001
K) -4
Figure imgf000228_0001
K)
OO
Figure imgf000229_0001
K)
Figure imgf000230_0001
O
Figure imgf000231_0001
Ph is phenyl; Me is methyl; and Et is ethyl. Relative solubilities (at OD 600nm) for the compounds ranged from about
75 μM/mL for Compound No. 5 to >1000 uM/ml for Compound Nos. 7-11. A cytochrome-P450 binding assay completed for Compound No. 3 indicated binding of 15.87 μM, and MDRC4 and CNDO cell-based assays for this same compound versus other mutant forms such as, for example, K103N/M184V/NAMS and K103N/Y181C/M184V/NAMS, gave EC50 values of from 0.0013-0.4466 μM.
TOXICOLOGY
1. pH dependent aqueous solubility
The aqueous solubility of each compound was determined by the conventional shake- flask method at a saturated concentration of ImM. The flask (vial) was shaken for 3 hrs at ambient temperature, and then centrifuged. The supernatant was analyzed by HPLC with UV detection for solubility determination. In general, a higher aqueous solubility is more desirable for drug candidates.
2. Human plasma protein binding
The human plasma protein binding was determined using the Equilibrium Dialysis method. Dialysis was conducted with pooled human plasma at 370C for approximately 6 hours at a drug concentration of lμM. At the end of the dialysis, buffer samples from the buffer-side of the dialysis cell were collected and analyzed by LC/MS/MS for free drug concentration. For NNRTIs, a lower protein binding is more desirable.
3. Bi-directional CACO-2 permeability
The objective of this assay is to determine the bi-directional permeability classification and efflux-limited absorption potential of a test compound in a Caco-2 cell monolayer system. Typically, the assay involves the measurement of non-specific binding to the Transwell apparatus in a pH7.4 assay buffer, the bi-directional permeability assessment of a test compound across Caco-2 cell monolayers, apical-to-basolateral transport assessment, basolateral-to-apical transport assessment, and monolayer integrity. High permeability and no efflux suggest that intestinal permeability is not expected to be a limiting factor for oral absorption in humans.
4. CYP450 inhibition
In vitro CYP450 inhibition screening allows for the prediction of potential drug-drug interactions. To determine whether a test compound inhibits a particular P450 enzyme activity, changes in the metabolism of a P450-specific substrate by human liver microsomes are monitored with varying concentrations of the test compound. Potency and rank order of the inhibition can be assessed by determination of the IC50 values for a particular isozyme. For NNRTIs, higher IC50 values would suggest less inhibition, and thus less potential for drug-drug interaction in patients. CYP3A4 inhibition was screened using the CYP3A4/BFC high throughput inhibitor screening kit (BD Biosciences), CYP2D6 inhibition was screened using the CYP2D6/AMMC high throughput inhibitor screening kit (BD Biosciences), and CYP2C9 inhibition was screened using the P450-Glo™ Assay kit (Promega).
5. In vitro metabolic stability in liver microsomes
The metabolic stability assay is to evaluate the stability of test compound in biological matrices. Data are useful in terms of understanding and predicting the elimination mechanisms of the test compound. CYP450-dependent metabolism of drugs can also vary widely from one species to another. Evaluating in vitro metabolism of a drug by liver microsomes from multiple species allows the comparison of metabolism in animal species to that of humans. This can help identify the most relevant animal model for PK and toxicology studies. The metabolic stability of a test compound was evaluated in vitro in liver microsomes of the rat, dog, monkey, and human. lOμM of the test compound was pre-incubated for 5 min at 370C with lmg/mL liver microsomes in 0.1M
Tris buffer, pH 7.4 containing 5mM MgC12 and O.lmM EDTA. Following pre¬ incubation, NADPH (final concentration of 3 niM) was added to start the reaction and samples incubated for 0 and 1 or 2 hours. After terminating the reaction, the supernatant was analyzed by HPLC-UV or LC/MS/MS for the disappearance of the parent and formation of metabolite(s). The %parent remaining was the peak area ratio of the 1 or 2 hr sample to the time 0 sample. In general, less metabolism (higher %parent values) is more desirable.
6. In vitro metabolism in liver microsomes - metabolic pathway
Phase I biotransformation of a test compound is evaluated in this assay. Samples from the metabolic stability experiment were analyzed by LC/MS/MS for metabolite profiling and identification. Structures of metabolites were elucidated based on multiple MS/MS experiments such as full scan, neutral loss scan, and product ion scan. Metabolic pathways were then postulated based on the structures of major metabolites. For compounds from the phosphinate series, N-oxidation of the terminal pyridine ring is the predominant pathway, followed by the oxidation of the dimethyl-phenyl group, where the methyl group(s) is hydroxylated, further oxidized to an aldehyde, and finally to a carboxylic acid. The elucidation of metabolic pathways is important in terms of understanding the elimination mechanism of a test compound and assisting the design of new molecules with improved DM-PK profiles.
CYP3A4
Protein binding induction,
(human CYP450 inhibition, human (IC50, human Metabolic stability in liver
Cat. Cmpd Aqueous solubility (uM) plasma) uM) hepatocytes microsomes c /o Parent after 2hrs pH1 pH3 pH7 pH9 %Free CYP3A4 CYP2D6 CYP2C9 (mRNA) Human Rat Dog Monke weak to 24%
Parent N >1000 231 2.1 1.7 0.2% 0.66 2.04 moderate 22%* 1%* * 1%*
N-
Oxide D 52 74.2 50.6 66.8 1.6% 1.07 >10 >10 76% 1% 82% 24%
U(
Parent O 820 590 9.1 10.1 0.9% 0.81 1.35 5.35 15% 2% 29% 1 %
N-
Oxide B 190 170 200 190 3.8% 1.04 >10 >10 94% 1% 85% 63%
Parent P >1000 724.3 24 33.8 <0.5% 0.77 2.01 1.68 3% 0% 20% 3%
N- 24
Oxide G 1.6% 1.14 >10 8.85 85% % 77% 41 %
*%Parent after 1hr incubation
Metabolic pathway (in
Cat. Cmpd vitro)
N-oxidation, hydroxylation to CH3OH- to CHO- to
Parent N COOH-
N-
Oxide D Hydroxylation to CH30H- to CHO- to COOH-
N-oxidation, hydroxylation to CH30H- to CHO- to
Parent O COOH-
N-
Oxide B Hydroxylation to CH30H- to CHO- to COOH-
N-oxidation, hydroxylation to CH30H- to CHO- to
Parent P COOH-
N-
Oxide G Hydroxylation to CH30H- to CHO- to COOH-
7. PK and oral bioavailability in the rat and dog.
The pharmacokinetics of a test compound was evaluated in Sprague-Dawley rats and Beagle dogs. A typical PK study involved dosing of 2 to 3 animals via a single IV bolus injection at 1 mg/kg and another 3 animals via a single oral gavage at 5 mg/kg. Blood samples were collected at various time points over a 24-hr period. Plasma was separated and analyzed for test compound and its metabolite(s) by LC/MS/MS. PK parameters were calculated from the plasma concentration - time profile using a noncompartmental method. Oral bioavailability (F) was calculated based on dose-normalized AUC values from oral and IV administration. Higher oral bioavailability values are better.
ON
-4
Figure imgf000238_0001

Claims

WE CLAIM:
1. An optionally substituted 3-phosphoindole compound.
2. The compound of claim 1 wherein the 3-phosphoindoles is in the form of a phosphate, a phosphonate, a thiophosphate, a thiophosphonate, an iminiophosphate or an iminophosphonate.
3. The compound of claim 1 wherein the compound is a phosphate or a phosphonate.
4. The compound of claim 1 wherein the compound is further substituted at the 2 position.
5. The compound of claim 1 wherein the compound is substituted at the nitrogen of the indole.
6. The compound of claim 1 wherein the compound is of Formula (A):
Figure imgf000239_0001
or a pharmaceutically acceptable salt, prodrug, N— oxide, quaternary amine, stereochemical isomer or tautomer thereof, wherein:
X and Y, each independently, is
b) 3-14 membered carbocycle, aryl, heterocycle, any of which may comprise a monocyclic, bicyclic, tricyclic or spiro structure, or optionally may be substituted;
b) H;
dd) OH;
ee) Cl, Br, I, F; ff) CF3; gg) C1-6 alkyl;
hh) C2-6 alkenyl;
ii) C2-6 alkynyl;
jj) alkylheterocycle;
kk)NH2;
11) Mϊ-alkyl;
mm) N-dialkyl;
nn) NH-aryl;
oo)N-alkaryl;
pp) N-aralkyl;
qq) NH-heterocycle;
rr) N-alkyl-heterocycle;
ss) N-alkenyl-heterocycle;
tt) N-alkynyl-heterocycle;
uu) O-alkyl;
w) O-alkenyl;
ww) O-alkynyl;
xx) O-alkylaryl;
yy) O-aryl;
zz) O-heterocycle;
aaa) O-aralkyl;
bbb) O-carbocycle;
ccc) SR2; or ddd) NR2R3;
alternatively, X and Y may be joined to form an optionally substituted bicyclic or tricyclic phosphorylated heterocycle wherein each ring comprises 3-7 members;
Z is:
mm) H;
nn) alkoxy;
oo)NO2;
pp)N(R2)(R3);
qq) OR2;
rr) carboxamido;
ss) amido;
tt) acyl;
uu) S(O)nR2;
W) S(O)n-NR2R3;
ww) C1-6 alkyl;
xx) C2-6 alkenyl;
yy) C2-6 alkynyl;
zz) alkaryl;
aaa) aralkyl;
bbb) heterocycle;
ccc) alkyl-heterocycle;
ddd) aryl;
eee) CN;
fff) C(=W)-R2; ggg) C(=W)NH-C(R2)(R2)-C(=W)-N(R2)(R2);
hhh) C(=W)NH-P(=W)(R2)-A-R2;
iii) C(=W)NH-A-S(O)n-NR2;
jjj) C(=W)NH-CR2R3-S(O)n -NR2R3;
kkk) C(=W)-NH-A-C(=W)-N(R2)(R3);
111) C(=W)-N(R2)(R3);
mmm) C(=W)-NH-A-R2;
nnn) C(=W)-NH-NH-R2;
ooo) C(=W)-NH-C(R2)(R2)-C(=W)NH-C(R2)(R3)C(=W)- N(R2)(R3);
ppp) C(=W)-NH-R2;
qqq) C(=W)-NH-A-C(=W)-NH-A-C(=W)-NH2;
nr) C(R2XR2XR3);
sss) C(R2)(R2)-NH-R2;
ttt) A-S(O)n-R2;
uuu) C(=W)-A-C(=W)-A-C(=W)R3;
vw) A-R2;
www) C(=W)-(O)R2;
xxx) C(=W)-A-C(=W)-NH2; mm)an amino acid residue;
aaa) C(=W)-N(R2)-A-(amino acid residue);
bbb) C(=W)-N(R2)-A-(amino acid residue)-C(=W)-R2;
ccc) C(=W)-amino acid residue;
ddd) C(=W)-N(R2)-A-(amino acid residue)- A-C(=W)-R2; eee) C(=W)-OR3;
fff) C(=W)-S(R2); ggg) C(^W)-NH-MI-R2;
hhh) C(=W)-NH-N(R2)-A-C(=W)R2;
iii) C(=W)-N(R2)-C(=W)-R3;
jjj) C(=W)-A-NH-C(=W)R2; kkk) C(=W)-A-NH-C(=W)OR3;
111) C(=W)-A-R3;
mum) C(=W)-NH-NH-CH2-C(=W)R2;
aaa) P(=W)(R2)(R2); or
bbb) A-P(=W)(R2)(R2);
hhh) A-P(=W)(R3)(R3);
iii) C(=W)-NH-C1-10alkyl-heteroaryl;
jjj) C(=W)-NH-C1-4alkyl-heteroaryl;
kkk) C(=W)-NH-CH2-heteroaryl; and
Figure imgf000243_0001
111) ; wherein in embodiments (ddd), (eee) and (fff), the heteroaryl can optionally include a charged heteroatom, and in particular can include an N-oxide
wherein each of the foregoing X, Y and Z independently may be unsubstituted or substituted by one or more of:
b) H;
ii) C1-6 alkyl;
iii) alkoxy; iv) OH;
V) oxo;
Vi) halo;
vii) NR2R2;
viii) optionally substituted aryl;
ix) optionally substituted heterocyclyl;
x) O-C(=W)-alkyl;
xi) C(=W)-OR2;
x) CN;
xi) NO2;
xii) NH-C(=W)-alkyl;
xiii) NH-S(O)n-alkyl;
xiv) NH-S(O)n-NR2R2; or
xv) C3-6 cycloalkyl;
W is:
h) O;
i) S;
j) NH;
k) N-N(R2XR2);
1) N(R2);
m) N-OH; or
n) N-O-alkyl;
R1 is:
1) H; m) R2;
n) C(=W)-R2;
o) C(=W)-O(R2);
p) C(=W)-S(R2);
q) C(=W)-NH-R2;
r) C(=W)-N(R2)(R2); s) C(=W)-NH-A-(amino acid residue);
t) A-(amino acid residue)-R2;
u) S(O)n-R3; or
v) S(O)2-N(R2)(R2);
any of which optionally may be substituted by one or more:
i. C1-6 alkyl;
ii. OH;
iii. alkoxy;
iv. aryl;
v. halo;
vi. CN;
vii. NO2; or
viii. N(R2XR2);
Each R2 is independently selected from:
aaa) H;
bbb) OH;
ccc) halogen;
ddd) optionally substituted, branched or unbranched alkyl; eee) optionally substituted, branched or unbranched alkenyl;
fff) optionally substituted, branched or unbranched alkynyl;
ggg) 3-14 membered carbocycle;
hhh) alkylheterocycle;
iii) acyl;
jjj) carboxamido;
kkk) carbamoyl;
111) alkoxy;
mmm) optionally substituted aryl;
nnn) optionally substituted aralkyl;
ooo) optionally substituted alkylaryl;
PPP) O-alkyl;
qqq) O-alkenyl;
rrr) O-alkynyl;
sss) O-alkaryl;
ttt) O-aralkyl;
uuu) O-carbocycle;
wv) O-heterocycle;
www) O-aryl;
xxx) CF3;
yyy) CN; zzz) S(O)n-R3;
aaaa) N(R3)(R3);
bbbb) NH-S(O)n-R3; cccc) NHC(=W)-aryl;
dddd) NHC(=W)-alkyl;
eeee) NHC(=W)-heterocycle;
ffff) CH2-S(O)nR3;
gggg) C(=W)R3;
hhhh) C(=W)NR3R3;
iiii)C(alkyl)2-S(O)nR3; jjjj)CH(alkyl)-S(O)nR3;
kkkk) C(alkyl)2-NH2;
Ull)CH(alkyl)-N(alkyl)R3;
mmmm) i CR3R2-NR3R3;
nnnn) CH2N(alkyl)R3;
oooo) CH(alkyl)-MϊR3;
PPPP) C(alkyl)2-NHR3;
qqqq) C(alkyl)2-N(alkyl)R3;
rrrr) CH2-C(=W)H;
ssss) CH2-C(=W)alkyl;
tttt)CR3] R3-C(=W)R3;
uuuu) A-R3; vwv) C(R3)2-C(=W)R3; www) CH2-C(=W)H; xxxx) CH2-C(=W)alkenyl; yyyy) CH(alkenyl)-C(=W)H; zzzz) A-S(O)R3; aaa) CH(NH)-S(O)nR3; or
bbb) A-N(NH)R3;
wherein said optional substitution comprises one or more of:
A) a substituted or unsubstituted heterocycle;
B) C(=W)O-aryl;
C) C(=W)O-alkyl;
D) C(=W)NH2;
E) C(=W)NH-alkyl;
F) C(=W)NH-aryl;
G) C(=W)N-di-alkyl;
H) C(=W)N(alkyl)-aryl;
I) α-amino acid;
J) α-amino ester;
K) α-amino-carboxamide;
L) β-amino acid;
M) β-amino ester; or
N) β-amino-carboxamide;
Each R3 is:
bb)H;
cc) OH;
dd) C1-6alkyl;
ee) C2-6 alkenyl;
ff) C2-6 alkynyl;
gg) alkoxy; hh) CF3;
ii) CN;
jj) amino;
kk)NR2R2;
11) O-alkyl;
mm) O-alkenyl;
nn) O-alkynyl;
oo)C(R2)(R2)-S(O)nNH2;
pp) C(R2)(R2)-S(O)nCF3;
qq) C(R2)(R2)-NH2;
rr) A-heterocycle;
ss) C(R2)(R2)-NR2R2;
tt) C(R2)(R2)-C(=W)R2;
uu) aryl;
w) carbocycle;
ww) heterocycle;
xx) cycloalkyl;
yy) alkaryl;
zz) alkylheterocycle;
aaa) aralkyl; or
bbb) heterocycle-alkyl;
any of which may be unsubstituted or substituted with one or more of the following taken in any combination:
i. halo; xxiv. OH;
xxv. OR2;
xxvi. SR2;
xxvii. COOH;
5 xxviii. carboxylic acid ester;
xxix. CHV)R2;
xxx. C(=W)0R2;
xxxi. C(=W)0R3;
xxxii. C(=W)SR2;
10 xxxiii. A-C(=W)NH2;
xxxiv. CC=W)NR2R3;
xxxv. NR2R2;
xxxvi. NR2R3;
xxxvii.NR2-S(O)nR3;
15 xxxviii. NR2-C(=W)-C1-6alkyl;
xxxix. S(O)nR3; xl. C1-6alkoxy;
xli. C1-6 thioether;
xlii. amino acid residue;
20 xliii. NH-A-(amino acid residue);
xliv. C(=W)NH- A-(amino acid residue); and
xlv. wherein when said optional substitution comprises a substituted heterocycle, then substitution is selected from the group consisting 25 of: b. C(=W)O-aryl; c. C(=W)O-alkyl;
d. C(=W)NH2;
e. C(=W)NH-aryl;
f. C(=W)NH-alkyl;
g. C(=W)N-di-alkyl;
h. C(=W)N(alkyl)-aryl;
i. α-amino acid;
j. α-amino ester;
k. α-amino-carboxamide;
k. β-amino acid;
1. β-amino ester; or
m. β-amino-carboxamide;
n. halo; or
o. cyano,
taken alone or in any combination;
n independently is 0, 1 or 2;
A is a disubstituted spacer selected from the group consisting of:
j. C1-6 alkylene, branched or unbranched, and optionally having one or more heteroatoms, aryl, cycloalkyl or heterocyclyl functions in or attached to the chain;
k. C2-12 alkenylene, branched or unbranced, and optionally having one or more heteroatoms, aryl, cycloalkyl or heterocyclyl functions in or attached to the chain; 1. C2-12 alkynylene, branched or unbranced, and optionally having one or more heteroatoms, aryl, cycloalkyl or heterocyclyl functions in or attached to the chain;
m. optionally substituted arylene;
n. O-alkylene, branched or unbranched, and optionally having one or more heteroatoms, aryl, cycloalkyl or heterocyclyl functions in or attached to the chain;
o. aralkylene, branched or unbranched, and optionally having one or more heteroatoms, aryl, cycloalkyl or heterocyclyl functions in or attached to the chain;
p. optionally substituted cycloalkyl; and
q. optionally substituted heterocycle;
wherein "A" may be joined by any desired linkage or any combination of desired linkages.
Each R4', R5', R6> and R7' independently is:
a. H;
b. halogen;
c. NO2;
d. CN;
e. CF3;
f. OR2
g. NR2R2;
h. NHS(O)nR2;
i. NHCO-Ci-3 alkyl;
j. S(O)nR2;
k. aryl; 1. heterocycle; m. C1-6 alkyl; n. C2-6 alkenyl; o. C2-6 alkynyl;
P- C(=W)-S(O)nR2;
q- CC=W)-S(O)n-NR2R2;
r. C(=W)-aryl;
s. C(=W)-alkyl;
t. C(=W)-heterocycle; or
u. C(=W)-NR2R2;
each of which optionally may be substituted with one or more of :
r. OR2;
ii. S(O)nR2;
iii. C(=W)-S(O)nR2;
iv. C(=W)-S(O)n-NR2R2;
xxi. C(=W)-aryl;
xxii. C(=W)-alkyl;
xxiii. C(=W)-heterocycle;
xxiv. C(=W)NR2R2;
xxv. H;
xx vi. NO2;
xxvii. CN;
xxviii. CF3;
xxix. halogen; xxx. NHS(O)nR2; xxxi. NHCO-C1-3 alkyl;
xxxii. aryl;
xxxiii. heterocycle;
xxxiv. C1-6 alkyl;
xxxv. C2-6 alkenyl;
xxxvi. C2-6 alkynyl; or
xxxvii.NR2R2.
7. The compound of claim 6 wherein R5 is halogen, CH3, CN, CF3 or NO2.
8. The compound of claim 7 wherein R5 is Cl.
9. The compound of claim 6 wherein W is O or S.
10. The compound of claim 6 wherein Z is optionally substituted amide or carboxamide.
11. The compound of claim 6 wherein X is substituted or unsubstituted phenyl.
12. The compound of claim 6 wherein Y is -0-C1-6 alkyl.
13. The compound of claim 6 wherein the compound is selected from the group consisting of:
Figure imgf000255_0001
Figure imgf000256_0001
Figure imgf000257_0001
14. The compound of claim 1 wherein the compound is of Formula (B):
Figure imgf000258_0001
or a pharmaceutically acceptable salt, prodrug, N— oxide, quaternary amine,
stereochemical isomer or tautomer thereof, wherein:
Y is:
a. 3-14 membered carbocycle, aryl, heterocycle, any of which may comprise a monocyclic, bicyclic, tricyclic or spiro structure, or any of which optionally may be substituted;
b. H;
c. OH;
d. halogen;
e. CF3;
f. C1-6 alkyl;
g. C2-6 alkenyl;
h. C2-6 alkynyl;
i. alkylheterocycle;
j. NH2; k. NH-alkyl;
1. N-dialkyl;
m. NH-aryl;
n. N-alkaryl; O. N-aralkyl;
P- NH-heterocycle;
q- N-alkyl-heterocycle;
r. N-alkenyl-heterocycle;
S. N-alkynyl-heterocycle;
t. O-alkyl;
U. O-alkenyl;
V. O-alkynyl;
W. O-alkylaryl;
X. O-aryl;
y- O-heterocycle;
Z. O-aralkyl;
aa. O-carbocycle;
bb. SR2; or
CC. NR2R3;
Z is: aaa) H;
bbb) alkoxy;
ccc) NO2;
ddd) N(R2XR3);
eee) OR2;
fff) carboxamido;
ggg) amido;
hhh) acyl; iii) S(O)nR2;
jjj) S(O)n-NR2R3;
kkk) C1-6 alkyl;
111) C2-6 alkenyl;
mnim) C2-6 alkynyl;
nnn) alkaryl;
ooo) aralkyl;
PPP) heterocycle;
qqq) alkyl-heterocycle;
rrr) aryl;
sss) CN;
ttt) C(=W)-R2;
uuu)
Figure imgf000260_0001
vw) C(=W)NH-P(=W)(R2)-A-R2;
www) C(=W)NH-A-S(O)n-NR2;
xxx) C(=W)NH-CR2R3-S(O)n -NR2R3;
yyy) C(=W)-NH-A-C(=W)-N(R2)(R3);
zzz) C(=W)-N(R2)(R3);
aaaa) C(=W)-NH-A-R2;
bbbb) C(-W)-NH-NH-R2;
cccc) C(=W)-NH-C(R2)(R2)-C(=W)NH-C(R2)(R3)C(=W)- N(R2)(R3);
dddd) C(=W)-NH-R2;
eeee) C(=W)-NH-A-C(=W)-NH-A-C(=W)-NH2; ffff) C(R2)(R2)(R3); gggg) C(R2)(R2)-NH-R2;
hhhh) A-S(O)n-R2;
iiii)C(=W)-A-C(=W)-A-C(=W)R3;
jjjj) A-R2; kkkk) C(=W)-(O)R2;
11U)C(=W)-A-C(=W)-NH2;
mmmm) an amino acid residue;
nnnn) C(=W)-N(R2)-A-(arnino acid residue);
oooo) C(=W)-N(R2)-A-(amino acid residue)-C(==W)-R2;
pppp) C(=W)-amino acid residue;
qqqq) C(=W)-N(R2)-A-(amino acid residue)-A-C(=W)-R2;
rrrr) C(=W)-OR3;
ssss) C(=W)-S(R2);
tttt)C(=W)-NH-MΪ-R2;
uuuu) C(=W)-NH-N(R2)-A-C(=W)R2;
ww) C(=W)-N(R2)-C(=W)-R3;
www) C(=W)-A-NH-C(=W)R2;
xxxx) C(=W)-A-MΪ-C(=W)OR3;
yyyy) C(=W)-A-R3;
zzzz) C(=W)-NH-MΪ-CH2-C(=W)R2;
aaa) P(=W)(R2)(R2); or
bbb) A-P(-W)(R2)(R2); wherein each of the foregoing Y and Z independently may be unsubstituted or substituted by one or more of:
r. H;
s. C1-6 alkyl;
t. alkoxy;
u. OH;
v. oxo;
w. halo;
x. NR2R2;
y. optionally substituted aryl;
z. optionally substituted heterocyclyl;
aa. O-C(=W)-alkyl;
bb. C(=W)-OR2;
cc. CN;
dd. NO2;
ee. NH-C(=W)-alkyl;
ff. NH-S(O)n-alkyl;
gg. NH-S(O)n-NR2R2; or
hh. C3-6 cycloalkyl;
W is:
h) O;
i) S;
j) NH; k) N-N(R2XR2); 1) N(R2);
m) N-OH; or
n) N-O-alkyl;
R1 is:
cc) H;
dd)R2;
ee) C(=W)-R2;
ff) C(=W)-O(R2);
gg) C(=W)-S(R2);
hh) C(=W)-NH-R2;
ii) C(=W)-N(R2)(R2);
jj) C(=W)-NH-A-(amino acid residue);
kk) A-(amino acid residue)-R ;
U) S(O)n-R3; or
mm) S(O)2-N(R2XR2);
any of which optionally may be substituted by one or more:
i. C1-6 alkyl;
ix. OH;
x. alkoxy;
xi. aryl;
xii. halo;
xiii. CN;
xiv. NO2; or
xv. N(R2)(R2); Each R2 is:
a) H; b) OH;
c) halogen;
ccc) optionally substituted, branched or unbranched alkyl;
ddd) optionally substituted, branched or unbranched alkenyl;
eee) optionally substituted, branched or unbranched alkynyl;
fff) 3-14 membered carbocycle;
ggg) alkylheterocycle;
hhh) acyl;
iii) carboxamido;
jjj) carbamoyl;
kkk) alkoxy;
111) optionally substituted aryl;
mmm) optionally substituted aralkyl;
nnn) optionally substituted alkaryl;
ooo) O-alkyl;
ppp) O-alkenyl;
qqq) O-alkynyl;
rrr) O-alkaryl;
sss) O-aralkyl;
ttt) O-carbocycle;
uuu) O-heterocycle;
vw) O-aryl; www) CF3;
xxx) CN;
yyy) S(O)n-R3;
zzz) N(R3)(R3);
5 aaaa) NH-S(O)n-R3; bbbb) NHC(=W)-aryl;
cccc) NHC(=W)-alkyl;
dddd) NHC(=W)-heterocycle;
eeee) CH2-S(O)nR3;
10 ffff) C(=W)R3;
gggg) C(=W)NR3R3;
hhhh) C(alkyl)2-S(O)nR3;
iiii)CH(alkyl)-S(O)nR3;
jjjj)C(alkyl)2-NH2;
15 kkkk) CH(alkyl)-N(alkyl)R3;
U11)CR3R2-NR3R3;
mmnxtn) CH2N(alkyl)R3;
nnnn) CH(alkyl)-NHR3;
oooo) C(alkyl)2-NHR3;
20 pppp) C(alkyl)2-N(alkyl)R3;
qqqq) CH2-C(^W)H;
rrrr) CH2-C(=W)alkyl;
ssss) CR3R3-C(=W)R3;
tttt)A-R3; uuuu) C(R3)2-C(=W)R3;
ww) CH2-C(=W)H; wwww) CH2-C(=W)alkenyl;
xxxx) CH(alkenyl)-C(=W)H;
yyyy) A-S(O)R3; zzzz) CH(NH)-S(O)nR3; or
aaaaa) A-N(NH)R3;
wherein said optional substitution comprises one or more of:
I. a substituted or unsubstituted heterocycle;
II. C(=W)O-aryl;
III. C(=W)O-alkyl;
IV. CHV)NH2;
V. C(=W)NH-alkyl;
VI. C(=W)NH-aryl;
VII. C(=W)N-di-alkyl;
Viπ. C(=W)N(alkyl)-aryl;
DC. α-amino acid;
X. α-amino ester;
XL α-amino-carboxamide;
Xn. β-amino acid;
Xπi. β-amino ester; or
XW. β-amino-carboxamide;
XV. halo; or
XVI. cyano, Each R3 is:
bb)H;
cc) OH;
dd) Cw alkyl;
ee) C2-6 alkenyl;
ff) C2-6alkynyl;
gg) alkoxy;
hh) CF3;
ii) CN;
jj) amino;
kk)NR2R2;
11) O-alkyl;
mm) O-alkenyl;
mi) O-alkynyl;
oo) C(R2)(R2)-S(O)πNH2;
pp) C(R2)(R2)-S(O)nCF3;
qq) C(R2)(R2)-NH2;
rr) A-heterocycle;
ss) C(R2)(R2)-NR2R2;
tt) C(R2)(R2)-C(=W)R2;
uu) aryl;
w) carbocycle;
ww) heterocycle;
xx) cycloalkyl; yy) alkaryl;
zz) alkyllieterocycle;
aaa) aralkyl; or
bbb) heterocycle-alkyl;
any of which may be unsubstituted or substituted with one or more of the following taken in any combination:
I. halo;
II. OH;
III. OR2;
IV. SR2;
V. COOH;
VI. carboxylic acid ester;
VII. C(=W)R2;
VIII. C(=W)OR2;
IX. C(=W)OR3;
X. C(=W)SR2;
XL A-C(=W)NH2;
XII. C(=W)NR2R3;
XIII. NR2R2;
XIV. NR2R3;
XV. NR2-S(O)nR3;
XVI. NR2-C(=W)-C1-6alkyl;
XVII. S(O)nR3;
XVIII. C1-6alkoxy; XIX. C1-6 thioether;
XX. amino acid residue;
XXI. NH-A-(amino acid residue);
XXII. C(=W)MΗ-A-(amino acid residue); and
XXIII. wherein when said optional substitution comprises a substituted heterocycle, then substitution is selected from the group consisting of:
i. C(=W)O-aryl;
ii. C(=W)O-alkyl;
iii. C(=W)NH2;
iv. C(=W)NH-aryl;
v. C(=W)NH-alkyl;
vi. C(=W)N-di-alkyl;
vii. C(=W)N(alkyl)-aryl;
viii. α-amino acid;
ix. α-amino ester;
x. α-amino-carboxamide;
xi. β-amino acid;
xii. β-amino ester; or
xiii. β-amino-carboxamide;
taken alone or in any combination;
n independently is 0, 1 or 2;
A is a disubstituted spacer selected from the group consisting of: i) C1-6 alkylene, branched or unbranched, and optionally having one or more heteroatoms, aryl, cycloalkyl or heterocyclyl functions in or attached to the chain;
j) C2-12 alkenylene, branched or unbranced, and optionally having one or more heteroatoms, aryl, cycloalkyl or heterocyclyl functions in or attached to the chain;
k) C2-12 alkynylene, branched or unbranced, and optionally having one or more heteroatoms, aryl, cycloalkyl or heterocyclyl functions in or attached to the chain;
1) optionally substituted arylene;
m) O-alkylene, branched or unbranched, and optionally having one or more heteroatoms, aryl, cycloalkyl or heterocyclyl functions in or attached to the chain;
n) aralkylene, branched or unbranched, and optionally having one or more heteroatoms, aryl, cycloalkyl or heterocyclyl functions in or attached to the chain;
o) optionally substituted cycloalkyl; and
p) optionally substituted heterocycle;
wherein "A" may be joined by any desired linkage or any combination of desired linkages.
Each R4', R5', R6' and R7' independently is:
a. H;
b. halogen;
c. NO2;
d. CN;
e. CF3;
f. OR2 g- NR2R2; h. NHS(O)nR2; i. NHCO-C1-3 alkyl;
j. S(O)nR2;
k. aryl;
1. heterocycle;
m. C1-6 alkyl;
n. C2-6 alkenyl;
o. C2-6 alkynyl;
P- C(=W)-S(0)nR2;
q- C(=W)-S(O)n-NR2R2;
r. C(=W)-aryl;
S. C(=W)-alkyl;
t. C(=W)-heterocycle; or
U. C(=W)-NR2R2;
each of which optionally may be substituted with one or more of :
I. OR2;
II. S(O)nR2;
III. C(=W)-S(0)nR2; IV. C(=W)-S(O)n-NR2R2;
V. C(=W)-aryl;
VI. C(=W)-alkyl;
VII. C(=W)-heterocycle;
VIII. C(^W)NR2R2; DC H;
X. NO2;
XL CN;
Xn. CF3;
Xiπ. halogen;
XrV. NHS(O)nR2;
XV. NHCO-C1-3 alkyl;
XVI. aryl;
XVII. heterocycle;
XVIII . C1-6 alkyl;
XIX. C2-6 alkenyl;
XX. C2-6 alkynyl; or
XXI. NR2R2; and
Each R2", R3", R4", ] X5 ", and R6' independently is
bb)H;
cc) halogen;
dd)NO2;
ee) CN;
ff) OR2;
gg) SR2; hh)NH2;
ii) NR2R3;
jj) N(R2)-C(=W)-C1-4 alkyl;
kk)N(R2)-SO2-C1-4 alkyl; U) Ci-β alkyl; mm) C2-6 alkenyl;
nn) C2-6 alkynyl; oo) aryl;
PP)CF3;
qq) CR2R2-S(O)n-R3;
rr) CR2R2NR2R3;
ss) C-OH;
tt) CR2R2-C(=W)R2; uu) acyl;
w) C(=W)R2;
ww) C(=W)OR2;
xx) C(=W)SR2;
yy) C(=W)-NR2R3;
zz) C(=W)NH(CH2)p-(amino acid residue);
aaa) amino residue; or
bbb) A-(amino acid residue);
wherein any of the above optionally may be substituted; or
alternatively, R2 or R6 may be joined to Y to form an optionally substituted bicyclic or tricyclic phosphorylated heterocycle comprising 4-14 members.
15. The compound of claim 14 wherein R5 is halogen, CH3> CN, CF3 or NO2.
22. The compound of claim 15 wherein R5' is Cl.
23. The compound of claim 14 wherein W is O or S. 24. The compound of claim 14 wherein Z is optionally substituted amido or carboxamide.
25. The compound of claim 14 wherein X is substituted or unsubstituted phenyl.
26. The compound of claim 14 wherein Y is -0-C1-6 alkyl.
27. The compound of claim 1 wherein the compound is of Formula (C):
Figure imgf000274_0001
(C)
or a pharmaceutically acceptable salt, prodrug, N — oxide, quaternary amine,
stereochemical isomer or tautomer thereof, wherein:
Y is:
a. 3-14 membered carbocycle, aryl, heterocycle, any of which may comprise a monocyclic, bicyclic, tricyclic or spiro structure;
b. H;
C. OH;
d. halogen;
e. CF3;
f. C1-6 alkyl;
g- C2-6 alkenyl;
h. C2-6 alkynyl;
i. alkylheterocycle;
j- NH2; k. NH-alkyl;
1. N-dialkyl;
m. NH-aryl;
n. N-alkaryl;
O. N-aralkyl;
P- Mϊ-heterocycle;
q- N-alkyl-heterocycle;
r. N-alkenyl-heterocycle;
S. N-alkynyl-heterocycle;
t. O-alkyl;
U. O-alkenyl;
V. O-alkynyl;
W. O-alkylaryl;
X. O-aryl;
y- O-heterocycle;
Z. O-aralkyl;
aa. O-carbocycle;
bb . SR2; or
CC. NR2R3;
Z is:
aaa) H;
bbb) alkoxy;
ccc) NO2;
ddd) N(R2XR3); eee) OR2;
fff) carboxamido;
ggg) amido;
hhh) acyl;
iii) S(O)nR2;
jjj) S(O)n-NR2R3;
kkk) C1-6 alkyl;
Ul) C2-6 J ilkenyl;
mmm) C2-6 alkynyl;
nnn) alkaryl;
ooo) aralkyl;
PPP) heterocycle;
qqq) alkyl-heterocycle;
rrr) aryl;
sss) CN;
ttt) C(=W)-R2;
uuu) C(=W)NH-C(R2)(R2)-C(=W)-N(R2)(R2);
vw) C(=W)NH-P(=W)(R2)-A-R2;
www) C(=W)NH-A-S(O)n-NR2;
xxx) C(=W)NH-CR2R3-S(O)n -NR2R3;
yyy) C(=W)-NH-A-C(=W)-N(R2)(R3);
zzz) C(=W)-N(R2)(R3);
aaaa) Q=W)-NH-A-R2;
bbbb) C(=W)-NH-NH-R2 cccc) C(=W)-NH-C(R2)(R2)-C(=W)NH-C(R2)(R3)C(=W)- N(R2)(R3);
dddd) C(=W)-NH-R2;
eeee) C(=W)-NH-A-C(=W)-NH-A-C(=W)-NH2;
ffff) C(R2)(R2)(R3);
gggg) C(R2)(R2)-NH-R2;
hhhh) A-S(O)n-R2;
iiii)C(=W)~A-C(=W)-A-C(=W)R3;
jjjj) A-R2; fckkk) C(=W)-(O)R2;
1111)C(=W)-A-C(=W)-NH2;
mrnmm) an amino acid residue;
nnnn) C(=W)-N(R2)-A-(amino acid residue);
oooo) C(=W)-N(R2)-A-(amino acid residue)-C(-W)-R2;
PPPP) C(=W)-amino acid residue;
qqqq) C(=W)-N(R2)-A-(amino acid residue)-A-C(=W)-R2;
rrrr) C(^W)-OR3;
ssss) C(=W)-S(R2);
tttt)C(=W)-NH-NH-R2;
uuuu) C(-W)-NH-N(R2)-A-C(=W)R2;
WW) C(-W)-N(R2)-C(=W)-R3;
www) C(=W)-A-NH-C(=W)R2;
xxxx) C(=W)-A-NH-C(=W)OR3;
yyyy) C(=W)-A-R3; zzzz) C(=W)-NH-NH-CH2-C(=W)R2;
aaa) P(=W)(R2)(R2); or
bbb) A-P(=W)(R2)(R2); wherein each of the foregoing Y and Z independently may be unsubstituted or substituted by one or more of:
nn)H;
oo) C1-6 alkyl;
pp) alkoxy;
qq) OH;
rr) oxo;
ss) halo;
tt) NR2R2;
uu) optionally substituted aryl;
w) optionally substituted heterocyclyl;
ww) O-C(=W)-alkyl;
xx) C(=W)-0R2;
yy) CN;
zz) NO2;
aaa) NH-C(=W)-alkyl;
bbb) NH-S(O)n-alkyl;
ccc) NH-S(O)n-NR2R2; or
ddd) C3-6 cycloalkyl;
W is:
h) O; i) S;
j) NH;
k) N-N(R2)(R2);
1) N(R2);
m) N-OH; or
n) N-O-alkyl;
Is:
1) H;
m) R2; n) C(=W)-R2;
o) C(=W)-0(R2);
P) C(=W)-S(R2);
q) C(=W)-NH-R2;
C(=W)-N(R2)(R2);
s) C(=W)-NH-A-(amino acid residue);
t) A-(amino acid residue)-R2;
u) S(O)n-R3; or
v) S(O)2-N(R2XR2);
any of which optionally may be substituted by one or more:
i. C1-6 alkyl;
ii. OH;
iii. alkoxy;
iv. aryl;
v. halo; vi. CN;
vii. NO2; or
viii. N(R2)(R2);
Each R2 is:
ccc) H; ddd) OH; eee) halogen;
fff) optionally substituted, branched or unbranched alkyl;
ggg) optionally substituted, branched or unbranched alkenyl;
hhh) optionally substituted, branched or unbranched alkynyl;
iii) 3-14 membered carbocycle;
jjj) alkylheterocycle;
kkk) acyl;
111) carboxamido;
mmm) carbamoyl;
nnn) alkoxy;
ooo) optionally substituted aryl;
ppp) optionally substituted aralkyl;
qqq) optionally substituted alkylaryl;
rrr) O-alkyl;
sss) O-alkenyl;
ttt) O-alkynyl;
uuu) O-alkaryl;
vw) O-aralkyl; www) O-carbocycle;
xxx) O-heterocycle;
yyy) O-aryl;
zzz) CF3;
aaaa) CN; bbbb) S(O)n-R3;
cccc) N(R3)(R3); dddd) NH-S(O)n-R3; eeee) NHC(=W)-aryl;
ffff) NHC(=W)alkyl;
gggg) NHC(=W)-heterocycle;
hhhh) CH2-S(O)nR3;
iiii)C(=W)R3;
jjjj)C(=W)NR3R3;
fckkk) C(alkyl)2-S(O)nR3;
llll)CH(alkyl)-S(O)nR3;
mmmm) C(alkyl)2-NH2;
nnnn) CH(alkyl)-N(alkyl)R3;
oooo) CR3R2-NR3R3;
PPPP) CH2N(alkyl)R3;
qqqq) CH(alkyl)-NHR3;
rrrr) C(alkyl)2-NHR3;
ssss) C(alkyl)2~N(alkyl)R3;
tttt)CH2-C(=W)H; uuuu) CH2-C(=W)alkyl;
ww) CR3R3-C(=W)R3;
wwww) A-R ;
xxxx) C(R3)2-C(=W)R3;
yyyy) CH2-C(=W)H;
zzzz) CH2-C(=W)alkenyl;
aaaaa) CH(alkenyl)-C(=W)H;
bbbbb) A-S(O)R3;
ccccc) CH(NH)-S(O)nR3; or
ddddd) A-N(NH)R3;
wherein said optional substitution comprises one or more of:
i. a substituted or unsubstituted heterocycle;
ii. C(=W)O-aryl;
iii. C(=W)O-alkyl;
iv. C(=W)NH2;
v. C(=W)NH-alkyl;
vi. C(=W)NH-aryl;
vii. C(=W)N-di-alkyl;
viii. C(=W)N(alkyl)-aryl;
ix. α-amino acid;
x. α-amino ester;
xi. α-amino-carboxarnide;
xii. β-amino acid;
xiii. β-amino ester; or xiv. β-amino-carboxamide;
Each R3 is:
bb)H;
cc) OH;
dd) C1-6 alkyl;
ee) C2-6 alkenyl;
ff) C2-6 alkynyl;
gg) alkoxy;
hh) CF3;
ii) CN;
jj) amino;
kk)NR2R2;
11) O-alkyl;
mm) O-alkenyl;
nn) O-alkynyl;
oo) C(R2)(R2)-S(O)nNH2;
pp) C(R2)(R2)-S(O)nCF3;
qq) C(R2)(R2)-NH2;
rr) A-heterocycle;
ss) C(R2)(R2)-NR2R2
tt) C(R2)(R2)-C(=W)R2;
uu) aryl;
w) carbocycle;
ww) heterocycle; xx) cycloalkyl;
yy) alkaryl;
zz) alkylheterocycle;
aaa) aralkyl; or
bbb) heterocycle-alkyl;
any of which may be unsubstituted or substituted with one or more of the following taken in any combination:
i) halo;
ϋ) OH;
iii) OR2;
iv) SR2;
v) COOH;
vi) carboxylic acid ester;
vii) C(=W)R2;
viii) C(=W)0R2;
ix) C(=W)0R3;
x) C(=W)SR2;
xi) A-C(=W)NH2;
xii) C(=W)NR2R3;
xiϋ) NR2R2; xiv) NR2R3;
xv) NR2-S(O)nR3;
xvi) NR2-C(=W)-C1-6alkyl;
xvii) S(O)nR3; xviii) C1-6alkoxy;
xix) C1-6 thioether;
xx) amino acid residue;
xxi) NH-A-(amino acid residue);
xxii) C(=W)NH-A-(amino acid residue); and
xxiii) wherein when said optional substitution comprises a substituted heterocycle, then substitution is selected from the group consisting of:
I. C(=W)O-aryl;
II. C(=W)O-alkyl;
III. C(=W)NH2;
IV. C(=W)NH-aryl;
V. C(=W)NH-alkyl;
VI. C(=W)N-di-alkyl;
VII. C(=W)N(alkyl)-aryl;
VIII. α-amino acid;
IX. α-amino ester;
X. α-amino-carboxamide;
XI. β-amino acid;
XII. β-amino ester; or
XIII. β-amino-carboxamide;
taken alone or in any combination;
n independently is 0, 1 or 2;
A is a disubstituted spacer selected from the group consisting of: a. C1-6 alkylene, branched or unbranched, and optionally having one or more heteroatoms, aryl, cycloalkyl or heterocyclyl functions in or attached to the chain;
b. C2-12 alkenylene, branched or unbranced, and optionally having one or more heteroatoms, aryl, cycloalkyl or heterocyclyl functions in or attached to the chain;
c. C2-12 alkynylene, branched or unbranced, and optionally having one or more heteroatoms, aryl, cycloalkyl or heterocyclyl functions in or attached to the chain;
d. optionally substituted arylene;
e. O-alkylene, branched or unbranched, and optionally having one or more heteroatoms, aryl, cycloalkyl or heterocyclyl functions in or attached to the chain;
f. aralkylene, branched or unbranched, and optionally having one or more heteroatoms, aryl, cycloalkyl or heterocyclyl functions in or attached to the chain;
g. optionally substituted cycloalkyl; and
h. optionally substituted heterocycle;
wherein "A" may be joined by any desired linkage or any combination of desired linkages.
Each R4', R5', R6' and R7' independently is:
a. H;
b. halogen;
C. NO2;
d. CN;
e. CF3;
f. OR2 g- NR2R2; h. NHS(O)nR2; i. NHCO-C1-3 alkyl;
j- S(O)nR2;
k. aryl;
1. heterocycle;
m. Ci-6 alkyl;
n. C2-6 alkenyl;
o. C2-6 alkynyl;
P- C(=W)-S(0)nR2;
q- C(=W)-S(O)n-NR2R2;
r. C(=W)-aryl;
S. C(=W)-alkyl;
t. C(=W)-heterocycle; or
U. C(=W)-NR2R2;
each of which optionally may be substituted with one or more of :
i. OR2;
ii. S(O)nR2;
iii. C(=W)-S(O)nR2;
iv. C(^W)-S(O)n-NR2R2;
v. C(=W)-aryl;
vi. C(=W)-alkyl;
vii. C(=W)-heterocycle;
viii. C(=W)NR2R2; ix. H;
x. NO2;
xi. CN;
xii. CF3;
xiii. halogen;
xiv. NHS(O)nR2;
xv. NHCO-C1-3 alkyl;
xvi. aryl;
xvii. heterocycle;
xviii. C1-6 alkyl;
xix. C2-6 alkenyl;
xx. C2-6 alkynyl; or
xxi. NR2R2.
Each R3", R4", R5", and R6" independently is:
a. H;
b. halogen;
c. NO2;
d. CN;
e. OR2;
f. SR2;
g. NH2;
h. NR2R3;
i. N(R2)-C(=W)-C1-4 alkyl;
j. N(R2)-SO2-C1-4 alkyl; k. C1-6 alkyl;
1. C2-6 alkenyl;
m. C2-6 alkynyl;
n. aryl;
o. CF3;
p. CR2R2-S(O)n-R3;
q. CR2R2NR2R3;
r. C-OH;
s. CR2R2-C(=W)R2;
t. acyl;
u. C(=W)R2;
v. C(^W)OR2;
w. C(=W)SR2;
x. C(=W)-NR2R3;
y. C(=W)NH(CH2)p-(amino acid residue);
z. amino residue; or
aa. A-(amino acid residue);
wherein any of the above optionally may be substituted;
wherein indicates the presence of a single or double bond, where:
when indicates the presence of a double bond, each Y and T independently is:
j- CR2;
k. N; or
1. SR2; with the caveat that one of Y and T must be CR2;
when indicates the presence of a single bond, then each Y and T
independently is:
m. CR2;
n. O;
o. NR2; or
p. SR2;
with the caveat that one of Y and T must be CR2; and
m is 1 or 2, with the proviso that m can only be 2 for T or Y = CR2.
22. The compound of claim 21 wherein R5' is halogen, CH3, CN, CF3 or NO2.
23. The compound of claim 22 wherein R5 is Cl.
24. The compound of claim 21 wherein Z is optionally substituted amido or carboxamide.
25. . The compound of claim 21 wherein W is O or S.
26. . The compound of claim 21 wherein Y is -0-C1-6 alkyl.
27. A pharmaceutical composition comprising an HlV-treatment effective amount of a 3-phosphoindole compound or its pharmaceutically acceptable salt, prodrug, stereoisomer, tautomer, iV-oxide or quaternary amine, with a pharmaceutically acceptable carrier or diluent
28. The composition of claim 27 wherein the 3-ρhosphoindoles is in the form of a phosphate, a phosphonate, a phosphorthioate, or a phosphoramidate.
29. The composition of claim 27 wherein the compound is a phosphate or a phosphonate.
30. The composition of claim 27 wherein the compound is further substituted at the 2 position. 31. The composition of claim 27 wherein the compound is substituted at the nitrogen of the indole.
32. The composition of claim 27 comprising an effective anti-HIV treatment amount of a compound of claims 1, 6, 14, or 21 or its pharmaceutically acceptable salt, prodrug, stereoisomer, tautomer, JV-oxide or quaternary amine, with a pharmaceutically acceptable carrier or diluent.
33. The composition of claim 27 wherein the compound is in combination with at least one other anti-HIV agent, optionally with a pharmaceutically acceptable carrier or diluent.
34. The pharmaceutical composition of claim 33 wherein the other anti-HIV agent is a reverse transcriptase inhibitor.
35. The pharmaceutical composition of claim 33 wherein the other anti-HIV agent is a reverse transcriptase inhibitor.
36. The pharmaceutical composition of claim 35 wherein the reverse transcriptase inhibitor induces a mutation lysine 103 -> asparagine and/or tyrosine 181 -^ cysteine in HW reverse transcriptase.
37. Use of an effective amount of a 3-phosphoindole compound or its pharmaceutically acceptable salt, prodrug, stereoisomer, tautomer, iV-oxide or quaternary amine, with a pharmaceutically acceptable carrier or diluent in the manufacture of a medicament for the treatment or prophylaxis of an HIV infection in a host.
38. The use of claim 37 wherein the 3-phosphoindoles is in the form of a phosphate, a phosphonate, a phosphorthioate, or a phosphoramidate.
39. The use of claim 37 wherein the compound is a phosphate or a phosphonate.
40. The use of claim 37 wherein the compound is further substituted at the 2 position.
41. The use of claim 37 wherein the compound is substituted at the nitrogen of the indole.
42. The use of claim 37 wherein the compound is a compound of one of claims 1, 9, or 16, or its pharmaceutically acceptable salt, prodrug, stereoisomer, tautomer, N- oxide or quaternary amine, optionally with a pharmaceutically acceptable carrier or diluent.
43. The use of claim 37 wherein the host is a human.
44. The use of claim 37 wherein the compound is administered in combination and/or alternation with at least one other anti-HFV agent, optionally with a pharmaceutically acceptable carrier or diluent.
45. The use of claim 44 wherein the other anti-HIV agent is a reverse transcriptase inhibitor.
46. The use of claim 45 wherein the reverse transcriptase inhibitor induces a mutation lysine 103 -^ asparagine and/or tyrosine 181 -^ cysteine in HIV reverse transcriptase.
47. The use of claim 37, wherein the HIV has a mutation at lysine 103 -> asparagine and/or tyrosine 181 -> cysteine in HIV reverse transcriptase.
48. The use of claim 37 wherein the HIV is resistant to one or more reverse transcriptase inhibitor(s) .
49. The use of claim 38 wherein the compound is administered in combination and/or alternation with at least one other anti-HIV agent, optionally with a pharmaceutically acceptable carrier or diluent.
50. Use of an anti-HIV effective treatment amount of a 3-phosphoindole compound or its pharmaceutically acceptable salt, prodrug, stereoisomer, tautomer, iV-oxide or quaternary amine, optionally with a pharmaceutically acceptable carrier or diluent in the manufacture of a medicament for for salvage therapy in the treatment or prophylaxis of an HIV infection in a host..
51. The use of claim 50 wherein the compound is a compound of one of claims 1, 9, or 16, or its pharmaceutically acceptable salt, prodrug, stereoisomer, tautomer, N- oxide or quaternary amine.
52. The use of claim 50 wherein the compound is administered in combination and/or alternation with at least one other anti-HIV agent, optionally with a pharmaceutically acceptable carrier or diluent.
PCT/IB2005/004063 2004-09-16 2005-09-16 Phosphoindoles as hiv inhibitors WO2006054182A2 (en)

Priority Applications (18)

Application Number Priority Date Filing Date Title
CA2583409A CA2583409C (en) 2004-09-17 2005-09-16 Phospho-indoles as hiv inhibitors
MX2007003040A MX2007003040A (en) 2004-09-17 2005-09-16 Phosphoindoles as hiv inhibitors.
AU2005305609A AU2005305609B2 (en) 2004-09-17 2005-09-16 Phosphoindoles as HIV inhibitors
BRPI0515344-1A BRPI0515344A (en) 2004-09-17 2005-09-16 compound, pharmaceutical composition, and uses of an effective amount of a 3-phosphoindol compound or pharmaceutically acceptable salt, prodrug, stereoisomer, tautomer, n-oxide or quaternary amine thereof and an anti-HIV effective treatment amount. a compound of 3-phosphoindole or pharmaceutically acceptable salt, prodrug, stereoisomer, tautomer, n-oxide or quaternary amine thereof
RSP-2009/0057A RS50981B (en) 2004-09-17 2005-09-16 Phosphoindoles as hiv inhibitors
NZ554232A NZ554232A (en) 2004-09-17 2005-09-16 Phosphoindoles as HIV inhibitors
JP2007531877A JP5031569B2 (en) 2004-09-17 2005-09-16 Phosphoindoles as HIV inhibitors
PL05850774T PL1799696T3 (en) 2004-09-17 2005-09-16 Phosphoindoles as hiv inhibitors
DE602005011031T DE602005011031D1 (en) 2004-09-17 2005-09-16 Phosphoindole als hiv-inhibitoren
KR1020077008493A KR101242962B1 (en) 2004-09-17 2005-09-16 Phosphoindoles as hiv inhibitors
EP05850774A EP1799696B1 (en) 2004-09-17 2005-09-16 Phosphoindoles as hiv inhibitors
CN2005800392187A CN101084231B (en) 2004-09-17 2005-09-16 Phosphoindoles as HIV inhibitors
DK05850774T DK1799696T3 (en) 2004-09-17 2005-09-16 Phosphoindoles as HIV inhibitors
TW095134413A TW200804280A (en) 2005-09-16 2006-09-18 Phospho-indoles as HIV inhibitors
IL181909A IL181909A0 (en) 2004-09-17 2007-03-13 Phosphoindoles as hiv inhibitors
NO20071871A NO20071871L (en) 2004-09-17 2007-04-12 Phosphoindoles as HIV inhibitors.
HK07108120.9A HK1100142A1 (en) 2004-09-17 2007-07-26 Phosphoindoles as hiv inhibitors
HR20090086T HRP20090086T3 (en) 2004-09-17 2009-02-12 Phosphoindoles as hiv inhibitors

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US61106104P 2004-09-17 2004-09-17
US60/611,061 2004-09-17
US71144505P 2005-08-25 2005-08-25
US60/711,445 2005-08-25
US71156505P 2005-08-26 2005-08-26
US60/711,565 2005-08-26

Publications (3)

Publication Number Publication Date
WO2006054182A2 true WO2006054182A2 (en) 2006-05-26
WO2006054182A9 WO2006054182A9 (en) 2006-08-17
WO2006054182A3 WO2006054182A3 (en) 2007-03-01

Family

ID=36407947

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2005/004063 WO2006054182A2 (en) 2004-09-16 2005-09-16 Phosphoindoles as hiv inhibitors

Country Status (25)

Country Link
US (2) US7534809B2 (en)
EP (3) EP1961757B1 (en)
JP (1) JP5031569B2 (en)
KR (1) KR101242962B1 (en)
CN (1) CN102304150B (en)
AT (2) ATE414092T1 (en)
AU (1) AU2005305609B2 (en)
BR (1) BRPI0515344A (en)
CA (1) CA2583409C (en)
CY (1) CY1109440T1 (en)
DE (2) DE602005023360D1 (en)
DK (1) DK1799696T3 (en)
ES (1) ES2320252T3 (en)
HK (1) HK1100142A1 (en)
HR (1) HRP20090086T3 (en)
IL (1) IL181909A0 (en)
MX (1) MX2007003040A (en)
NO (1) NO20071871L (en)
NZ (1) NZ554232A (en)
PL (1) PL1799696T3 (en)
PT (1) PT1799696E (en)
RS (1) RS50981B (en)
SG (1) SG155939A1 (en)
SI (1) SI1799696T1 (en)
WO (1) WO2006054182A2 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008042240A2 (en) 2006-09-29 2008-04-10 Idenix Pharmaceuticals, Inc. Enantiomerically pure phosphoindoles as hiv inhibitors
US7534809B2 (en) 2004-09-17 2009-05-19 Idenix Pharmaceuticals, Inc. Phospho-indoles as HIV inhibitors
EP2076492A2 (en) * 2006-10-06 2009-07-08 Merck & Co., Inc. Non-nucleoside reverse transcriptase inhibitors
WO2010101967A3 (en) * 2009-03-04 2010-11-11 Idenix Pharmaceuticals, Inc. Phosphothiophene and phosphothiazole hcv polymerase inhibitors
US8470870B2 (en) 2008-03-27 2013-06-25 Idenix Pharmaceuticals, Inc. Solid forms of an anti-HIV phosphoindole compound

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008145562A1 (en) 2007-05-30 2008-12-04 F. Hoffmann-La Roche Ag Non-nucleoside reverse transcriptase inhibitors
TW200944537A (en) * 2008-04-03 2009-11-01 Idenix Pharmaceuticals Inc Formulations comprising a phosphoindole compound and one or more second active agents
US8309737B2 (en) * 2009-02-03 2012-11-13 Idenix Pharmaceuticals, Inc. Phosphinate ruthenium complexes
US20120295898A1 (en) 2010-01-27 2012-11-22 Mark Richard Underwood Antiviral Therapy
CN103275126B (en) * 2013-05-28 2016-03-23 兰州大学 The method of the phosphonylation indoles in a kind of stereoselective syntheses 3-position
ITMI20131218A1 (en) * 2013-07-19 2015-01-20 Need Pharma Srl NEW UREIDS DERIVATIVES OF NAFTALENSOLFONIC ACIDS
WO2019075487A1 (en) * 2017-10-13 2019-04-18 Ursure, Inc. Harvard Life Lab Products and methods for monitoring adherence to nucleoside reverse transcriptase inhibitor therapy

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997048399A1 (en) * 1996-06-17 1997-12-24 Guilford Pharmaceuticals Inc. Certain phosphinyl derivatives useful as naaladase inhibitors
WO1997048409A1 (en) * 1996-06-17 1997-12-24 Guilford Pharmaceuticals Inc. Methods of cancer treatment using naaladase inhibitors
WO1997048400A1 (en) * 1996-06-17 1997-12-24 Guilford Pharmaceuticals Inc. Hydroxy-phosphinyl derivatives useful as naaladase inhibitors
WO1998038332A1 (en) * 1997-02-28 1998-09-03 Biosynth Ag Detection of microbial metabolites
WO1998053812A1 (en) * 1997-05-27 1998-12-03 Guilford Pharmaceuticals Inc. Inhibitors of naaladase enzyme activity
WO1999052915A1 (en) * 1998-04-10 1999-10-21 Chemi S.P.A. Chiral phosphorated ligands useful in catalysts
WO2003090690A2 (en) * 2002-04-26 2003-11-06 Gilead Sciences, Inc. Cellular accumulation of phosphonate analogs of hiv protease inhibitor compounds and the compounds as such

Family Cites Families (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4522811A (en) 1982-07-08 1985-06-11 Syntex (U.S.A.) Inc. Serial injection of muramyldipeptides and liposomes enhances the anti-infective activity of muramyldipeptides
US4866084A (en) * 1987-07-17 1989-09-12 Harbor Branch Oceanographic Institution, Inc. Topsentin compounds effective against viruses and certain tumors
US5527819A (en) * 1991-09-06 1996-06-18 Merck & Co., Inc. Inhibitors of HIV reverse transcriptase
US5124327A (en) * 1991-09-06 1992-06-23 Merck & Co., Inc. HIV reverse transcriptase
DE4217964A1 (en) 1992-05-30 1993-12-02 Goedecke Ag Indolocarbazole imides and their use
BR9405737A (en) 1993-02-24 1995-12-05 Merck & Co Inc Compound process of inhibiting HIV transcriptase, process for preventing HIV infection or treating HIV infection, pharmaceutical composition useful for inhibiting HIV transcriptase, and useful for preventing or treating HIV infection or for the treatment of AIDS or ARC
US5424329A (en) 1993-08-18 1995-06-13 Warner-Lambert Company Indole-2-carboxamides as inhibitors of cell adhesion
US5489586A (en) * 1994-03-07 1996-02-06 Warner-Lambert Company Method for treating inflammatory disease in humans
US5612330A (en) * 1994-03-07 1997-03-18 Warner-Lambert Company Methods for inhibiting and controlling viral growth
US5489685A (en) * 1994-05-12 1996-02-06 Merck & Co., Ltd. Method of synthesizing furo[2,3-b]pyridine carboxylic acid esters
DE69525418T2 (en) * 1994-05-31 2002-10-17 Mitsui Chemicals Inc Benzimidazole derivatives
US5734081A (en) 1994-08-05 1998-03-31 Warner-Lambert Company Arylthio compounds
AP648A (en) 1995-01-10 1998-05-25 Smithkline Beecham Farm S P A Indole derivatives useful in the treatment of osteoporosis.
US6017903A (en) * 1996-09-27 2000-01-25 Guilford Pharmaceuticals Inc. Pharmaceutical compositions and methods of treating a glutamate abnormality and effecting a neuronal activity in an animal using NAALADase inhibitors
US6025345A (en) * 1996-06-17 2000-02-15 Guilford Pharmaceuticals Inc. Inhibitors of NAALADase enzyme activity
GB9614347D0 (en) * 1996-07-09 1996-09-04 Smithkline Beecham Spa Novel compounds
HUP0001062A3 (en) 1996-09-27 2001-09-28 Guilford Pharmaceuticals Inc B Pharmaceutical compositions of phosphinic acid derivatives having naaladase inhibitor acivity
US5830894A (en) * 1997-02-21 1998-11-03 Viropharma Incorporated Methods for preventing and treating pestivirus infection and associated diseases
US5935982A (en) 1997-02-28 1999-08-10 The University Of North Carolina At Chapel Hill Methods of treating retroviral infection and compounds useful therefor
US6333323B1 (en) 1998-03-26 2001-12-25 Shionogi & Co., Ltd. Indole derivatives with antiviral activity
GB9914371D0 (en) 1999-06-18 1999-08-18 Smithkline Beecham Plc Novel compounds
ATE451921T1 (en) * 2001-04-11 2010-01-15 Idenix Cayman Ltd PHENYLINDOLES FOR THE TREATMENT OF HIV
US6825201B2 (en) 2001-04-25 2004-11-30 Bristol-Myers Squibb Company Indole, azaindole and related heterocyclic amidopiperazine derivatives
JP3368273B1 (en) * 2001-12-28 2003-01-20 株式会社トミー Driving device for moving toy and moving toy
US20030236277A1 (en) * 2002-02-14 2003-12-25 Kadow John F. Indole, azaindole and related heterocyclic pyrrolidine derivatives
US6900206B2 (en) * 2002-06-20 2005-05-31 Bristol-Myers Squibb Company Indole, azaindole and related heterocyclic sulfonylureido piperazine derivatives
US20040063746A1 (en) 2002-07-25 2004-04-01 Alicia Regueiro-Ren Indole, azaindole and related heterocyclic ureido and thioureido piperazine derivatives
WO2004014364A1 (en) * 2002-08-07 2004-02-19 Idenix (Cayman) Limited Substituted phenylindoles for the treatment of hiv
ES2320252T3 (en) 2004-09-17 2009-05-20 Idenix Pharmaceuticals, Inc. PHOSFOINDOLS AS HIV INHIBITORS.
BRPI0717511A2 (en) 2006-09-29 2013-11-19 Idenix Pharmaceuticals Inc PURE COMPOUND, PHARMACEUTICAL COMPOSITION, METHODS TO TREAT HIV INFECTION, PREVENT HIV INFECTION, INHIBIT HIV REPLICATION, AND USE OF COMPOUND

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997048399A1 (en) * 1996-06-17 1997-12-24 Guilford Pharmaceuticals Inc. Certain phosphinyl derivatives useful as naaladase inhibitors
WO1997048409A1 (en) * 1996-06-17 1997-12-24 Guilford Pharmaceuticals Inc. Methods of cancer treatment using naaladase inhibitors
WO1997048400A1 (en) * 1996-06-17 1997-12-24 Guilford Pharmaceuticals Inc. Hydroxy-phosphinyl derivatives useful as naaladase inhibitors
WO1998038332A1 (en) * 1997-02-28 1998-09-03 Biosynth Ag Detection of microbial metabolites
WO1998053812A1 (en) * 1997-05-27 1998-12-03 Guilford Pharmaceuticals Inc. Inhibitors of naaladase enzyme activity
WO1999052915A1 (en) * 1998-04-10 1999-10-21 Chemi S.P.A. Chiral phosphorated ligands useful in catalysts
WO2003090690A2 (en) * 2002-04-26 2003-11-06 Gilead Sciences, Inc. Cellular accumulation of phosphonate analogs of hiv protease inhibitor compounds and the compounds as such

Non-Patent Citations (10)

* Cited by examiner, † Cited by third party
Title
DATABASE BEILSTEIN [Online] BEILSTEIN INSTITUTE FOR ORGANIC CHEMISTRY, FRANKFURT-MAIN, DE; XP002404472 Database accession no. BRN 1520645, BRN 951126 & AOYAKI ET AL.: CHIBA, DAIGAKU KOGAKUBU KENKYU, vol. 26, no. 49, 1974, pages 185-187, *
HAIKAL A: "SYNTHESIS OF GUANOSINE-3'-(5-BROMO-4-CHLOROINDOL-3-YL)- PHOSPHATE (G-3'-BCIP)" COLLECTION OF CZECHOSLOVAK CHEMICAL COMMUNICATIONS, INSTITUTE OF ORGANIC CHEMISTRY & BIOCHEMISTRY, PRAGUE, CZ, vol. 61, no. 3, 1996, pages 427-431, XP002070952 ISSN: 0010-0765 *
HORWITZ J P ET AL: "SUBSTRATES FOR CYTOCHEMICAL DEMONSTRATION OF ENZYME ACTIVITY V. THYMIDINE 3'-AND 5'-(5-BROMO-4-CHLORO-3-INDOLYL)PHOSPHATES" JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY. WASHINGTON, US, vol. 13, no. 5, 1970, pages 1024-1025, XP000858147 ISSN: 0022-2623 *
HORWITZ J P ET AL: "SUBSTRSTES FOR CYTOCHEMICAL DEMONSTRATION OF ENZYME ACTIVITY II. SOME DIHALO-3-INDOLYL PHOSPHATES AND SULFATES" JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY. WASHINGTON, US, May 1966 (1966-05), page 447, XP000858148 ISSN: 0022-2623 *
MARCH L C ET AL: "THE SYNTHESIS OF RIBONUCLEOTIDE-5'-(5-IODOINDOL-3-OL) AND (4-METHYLCOUMARIN-7-OL) ESTERS FOR THE HISTOCHEMICAL DEMONSTRATION OF NUCLEASES (1A)" JOURNAL OF HETEROCYCLIC CHEMISTRY, HETEROCORPORATION. PROVO, US, vol. 7, no. 4, August 1970 (1970-08), pages 885-889, XP008062091 ISSN: 0022-152X *
RABIGER, D. ET AL.: "Synthesis of 5-Iodo and 5-Nitro-3-indolyl phosphates as Cytochemical Substrates for Acid Phosphatase" J. HETEROCYCLIC CHEMISTRY, no. 7, 1970, pages 307-311, XP002404470 *
See also references of EP1799696A2 *
TSOU K C ET AL: "INDIGOGENIC PHOSPHODIESTERS AS POTENTIAL CHROMOGENIC CANCER CHEMOTHERAPEUTIC AGENTS" JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY. WASHINGTON, US, vol. 15, no. 12, 1972, pages 1221-1224, XP000990197 ISSN: 0022-2623 *
TSOU K C ET AL: "SYNTHESIS OF 3-INDOLYL AND 5-BROMO-3-INDOLYL PHOSPHATE FOR HISTOCHEMICAL DEMONSTRATION OF ALKALINE PHOSPHATASE" JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY. WASHINGTON, US, vol. 10, no. 4, 1967, pages 662-664, XP008062064 ISSN: 0022-2623 *
TSOU K C ET AL: "SYNTHESIS OF 5-IODO-3-INDOLYLPHOSPHODIESTERS OF 5-FLUORODEOXYURIDINE AS POSSIBLE CHROMOGENIC CANCER CHEMOTHERAPEUTIC AGENTS" JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY. WASHINGTON, US, vol. 13, no. 4, 1970, pages 765-768, XP002070956 ISSN: 0022-2623 *

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7534809B2 (en) 2004-09-17 2009-05-19 Idenix Pharmaceuticals, Inc. Phospho-indoles as HIV inhibitors
US8044091B2 (en) 2004-09-17 2011-10-25 Idenix Pharmaceuticals, Inc. Phospho-indoles as HIV inhibitors
WO2008042240A2 (en) 2006-09-29 2008-04-10 Idenix Pharmaceuticals, Inc. Enantiomerically pure phosphoindoles as hiv inhibitors
JP2010504978A (en) * 2006-09-29 2010-02-18 アイデニクス ファーマシューティカルズ,インコーポレーテッド Enantiomerically pure phosphoindoles as HIV inhibitors
US7960428B2 (en) 2006-09-29 2011-06-14 Idenix Pharmaceuticals, Inc. Enantiomerically pure phosphoindoles as HIV inhibitors
US8486991B2 (en) 2006-09-29 2013-07-16 Idenix Pharmaceuticals, Inc. Enantiomerically pure phosphoindoles as HIV inhibitors
EP2076492A2 (en) * 2006-10-06 2009-07-08 Merck & Co., Inc. Non-nucleoside reverse transcriptase inhibitors
EP2076492A4 (en) * 2006-10-06 2010-12-22 Merck Sharp & Dohme Non-nucleoside reverse transcriptase inhibitors
US8470870B2 (en) 2008-03-27 2013-06-25 Idenix Pharmaceuticals, Inc. Solid forms of an anti-HIV phosphoindole compound
WO2010101967A3 (en) * 2009-03-04 2010-11-11 Idenix Pharmaceuticals, Inc. Phosphothiophene and phosphothiazole hcv polymerase inhibitors
US8193372B2 (en) 2009-03-04 2012-06-05 Idenix Pharmaceuticals, Inc. Phosphothiophene and phosphothiazole HCV polymerase inhibitors

Also Published As

Publication number Publication date
PL1799696T3 (en) 2009-04-30
EP1961757A1 (en) 2008-08-27
PT1799696E (en) 2009-02-16
SI1799696T1 (en) 2009-04-30
DE602005011031D1 (en) 2008-12-24
IL181909A0 (en) 2007-07-04
RS50981B (en) 2010-10-31
CN102304150B (en) 2014-06-18
WO2006054182A3 (en) 2007-03-01
EP1799696B1 (en) 2008-11-12
US20060074054A1 (en) 2006-04-06
KR20070063545A (en) 2007-06-19
AU2005305609B2 (en) 2011-10-06
JP2008515782A (en) 2008-05-15
HRP20090086T3 (en) 2009-03-31
CY1109440T1 (en) 2014-08-13
EP1799696A2 (en) 2007-06-27
CN102304150A (en) 2012-01-04
NZ554232A (en) 2010-09-30
ES2320252T3 (en) 2009-05-20
ATE479693T1 (en) 2010-09-15
US20090163444A1 (en) 2009-06-25
AU2005305609A1 (en) 2006-05-26
DK1799696T3 (en) 2009-03-09
SG155939A1 (en) 2009-10-29
JP5031569B2 (en) 2012-09-19
HK1100142A1 (en) 2007-09-07
CA2583409A1 (en) 2006-05-26
ATE414092T1 (en) 2008-11-15
CA2583409C (en) 2014-06-17
EP2256124A1 (en) 2010-12-01
KR101242962B1 (en) 2013-03-13
WO2006054182A9 (en) 2006-08-17
US8044091B2 (en) 2011-10-25
EP1961757B1 (en) 2010-09-01
US7534809B2 (en) 2009-05-19
BRPI0515344A (en) 2008-07-15
MX2007003040A (en) 2007-08-02
DE602005023360D1 (en) 2010-10-14
NO20071871L (en) 2007-05-15

Similar Documents

Publication Publication Date Title
WO2006054182A2 (en) Phosphoindoles as hiv inhibitors
US9522926B2 (en) Activators of glucokinase
JP5320284B2 (en) Metabolically stable alkoxyalkyl esters of antiviral or antiproliferative phosphonates, nucleoside phosphonates and nucleoside phosphates
Németh et al. Synthesis and evaluation of phosphorus containing, specific CDK9/CycT1 inhibitors
RU2598849C2 (en) Phosphorus containing compounds as protein kinase inhibitors
CA2757083A1 (en) Amino pyrimidine anticancer compounds
EP1888581A2 (en) Hiv-integrase inhibitor compounds
JP2006523669A (en) Phosphate / sulfate compounds and pharmaceutical compositions for inhibiting NIMA regulatory protein (PIN1)
MXPA06007906A (en) Pyrimidyl phosphonate antiviral compounds and methods of use.
HUT72459A (en) Unsaturated phosphonates derivatives of purines and pyrimidines
Uttaro et al. Synthesis of novel 3′-methyl-5′-norcarbocyclic nucleoside phosphonates as potential anti-HIV agents
ES2354338T3 (en) PHOSFOINDOLS AS HIV INHIBITORS.
RU2393163C2 (en) Phosphoindoles as hiv inhibitors
WO2003000251A1 (en) Novel idolinones and uses thereof
TW200804280A (en) Phospho-indoles as HIV inhibitors
WO2008003809A1 (en) Antitumour compounds
Chen Synthesis of fluorine-containing acyclic nucleoside phosphonates as potential antiviral agents

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KM KP KR KZ LC LK LR LS LT LU LV LY MA MD MG MK MN MW MX MZ NA NG NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SM SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): BW GH GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU LV MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

WWE Wipo information: entry into national phase

Ref document number: 181909

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: MX/a/2007/003040

Country of ref document: MX

Ref document number: 2583409

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2007531877

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 554232

Country of ref document: NZ

Ref document number: 2005305609

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 1020077008493

Country of ref document: KR

ENP Entry into the national phase

Ref document number: 2007114274

Country of ref document: RU

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2005850774

Country of ref document: EP

Ref document number: 1551/CHENP/2007

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2007114274

Country of ref document: RU

ENP Entry into the national phase

Ref document number: 2005305609

Country of ref document: AU

Date of ref document: 20050916

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2005305609

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 200580039218.7

Country of ref document: CN

WWP Wipo information: published in national office

Ref document number: 2005850774

Country of ref document: EP

ENP Entry into the national phase

Ref document number: PI0515344

Country of ref document: BR

WWE Wipo information: entry into national phase

Ref document number: P-2009/0057

Country of ref document: RS