US20050124623A1 - Diazaindole-dicarbonyl-piperazinyl antiviral agents - Google Patents

Diazaindole-dicarbonyl-piperazinyl antiviral agents Download PDF

Info

Publication number
US20050124623A1
US20050124623A1 US10/979,558 US97955804A US2005124623A1 US 20050124623 A1 US20050124623 A1 US 20050124623A1 US 97955804 A US97955804 A US 97955804A US 2005124623 A1 US2005124623 A1 US 2005124623A1
Authority
US
United States
Prior art keywords
group
phenyl
heteroaryl
alkyl
halogen
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/979,558
Inventor
John Bender
Zhong Yang
John Kadow
Nicholas Meanwell
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
ViiV Healthcare UK No 5 Ltd
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Priority to US10/979,558 priority Critical patent/US20050124623A1/en
Assigned to BRISTOL-MYERS SQUIBB COMPANY reassignment BRISTOL-MYERS SQUIBB COMPANY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: KADOW, JOHN F., YANG, ZHONG, BENDER, JOHN A., MEANWELL, NICHOLAS A.
Publication of US20050124623A1 publication Critical patent/US20050124623A1/en
Priority to US12/026,633 priority patent/US7902204B2/en
Assigned to VIIV HEALTHCARE (NO. 5) LIMITED reassignment VIIV HEALTHCARE (NO. 5) LIMITED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BRISTOL-MYERS SQUIBB COMPANY
Assigned to VIIV HEALTHCARE UK (NO. 5) LIMITED reassignment VIIV HEALTHCARE UK (NO. 5) LIMITED CORRECTIVE ASSIGNMENT TO CORRECT THE RECEIVING PARTY DATA PREVIOUSLY RECORDED AT REEL: 039374 FRAME: 0388. ASSIGNOR(S) HEREBY CONFIRMS THE ASSIGNMENT . Assignors: BRISTOL-MYERS SQUIBB COMPANY
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/50Pyridazines; Hydrogenated pyridazines
    • A61K31/5025Pyridazines; Hydrogenated pyridazines ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • 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
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators

Definitions

  • This invention provides compounds having drug and bio-affecting properties, their pharmaceutical compositions and method of use.
  • the invention is concerned with new diazaindole derivatives that possess unique antiviral activity.
  • the present invention relates to compounds useful for the treatment of HIV and AIDS.
  • HIV-1 human immunodeficiency virus-1
  • HIV-1 human immunodeficiency virus-1
  • RT nucleoside reverse transcriptase
  • AZT or Retrovir®
  • didanosine or Videx®
  • stavudine or Zerit®
  • lamivudine or 3TC or Epivir®
  • zalcitabine or DDC or Hivid®
  • abacavir succinate or Ziagen®
  • Tenofovir disoproxil fumarate salt or Viread®
  • Combivir® contains ⁇ 3TC plus AZT
  • Trizivir® contains abacavir, lamivudine, and zidovudine
  • Emtriva® emtricitabine
  • three non-nucleoside reverse transcriptase inhibitors nevirapine (or Viramune®), delavirdine (or Rescriptor®) and efavirenz (or Sustiva®), nine peptidomimetic protease
  • NNRTIs Non-nucleoside reverse transcriptase inhibitors
  • NNRTIs the major drawback to the development and application of NNRTIs is the propensity for rapid emergence of drug resistant strains, both in tissue cell culture and in treated individuals, particularly those subject to monotherapy.
  • Pedersen & Pedersen, Ref 15 there is considerable interest in the identification of NNRTIs less prone to the development of resistance.
  • a recent overview of non-nucleoside reverse transcriptase inhibitors perspectives on novel therapeutic compounds and strategies for the treatment of HIV infection. has appeared (Buckheit, reference 99).
  • a review covering both NRTI and NNRTIs has appeared (De Clercq, reference 100).
  • An overview of the current state of the HIV drugs has been published (De Clercq, reference 101).
  • indole derivatives including indole-3-sulfones, piperazino indoles, pyrazino indoles, and 5H-indolo[3,2-b][1,5]benzothiazepine derivatives have been reported as HIV-1 reverse transciptase inhibitors (Greenlee et al, Ref. 1; Williams et al, Ref. 2; Romero et al, Ref. 3; Font et al, Ref. 17; Romero et al, Ref. 18; Young et al, Ref. 19; Genin et al, Ref. 20; Silvestri et al, Ref. 21).
  • Indole 2-carboxamides have also been described as inhibitors of cell adhesion and HIV infection (Boschelli et al, U.S. Pat. No. 5,424,329, Ref. 4). 3-Substituted indole natural products (Semicochliodinol A and B, didemethylasterriquinone and isocochliodinol) were disclosed as inhibitors of HIV-1 protease (Fredenhagen et al, Ref. 22).
  • New drugs for the treatment of HIV are needed for the treatment of patients who become resistant to the currently approved drugs described above which target reverse transcriptase or the protease.
  • One approach to obtaining these drugs is to find molecules which inhibit new and different targets of the virus.
  • a general class of inhibitors which are under active study are HIV entry inhibitors. This general classification includes drugs aimed at several targets which include chemokine receptor (CCR5 or CXCR4) inhibitors, fusion inhibitors targeting viral gp41, and inhibitors which prevent attachment of the viral envelope, gp120, the its human cellular target CD4.
  • CCR5 or CXCR4 chemokine receptor
  • fusion inhibitors targeting viral gp41 fusion inhibitors targeting viral gp41
  • Indole, azaindole and other oxo amide containing derivatives have been disclosed in a number different PCT and issued U.S. patent applications (Reference 93-95, 106, 108,109, 110, 111,112, 113, and 114). None of these applications discloses diazindole compounds such as described in this invention.
  • the extra nitrogen of the diazaindole class of molecules provides altered properties especially in combination with specific substituents that are advantageous and not available from the azaindoles.
  • the diazaindoles are easier to access and thus offer the potential to provide patients with lower cost treatments.
  • a series of PCT International Patent applications Bernd Nickel et.al.
  • the present invention comprises compounds of Formula I, their pharmaceutical formulations, and their use in patients suffering from or susceptible to a virus such as HIV.
  • the compounds of Formula I which include nontoxic pharmaceutically acceptable salts thereof, have the formula and meaning as described below.
  • the present invention comprises compounds of Formula I, including pharmaceutically acceptable salts thereof, which are effective antiviral agents, particularly as inhibitors of HIV.
  • Another preferred embodiment of the invention are compounds of Formula I, including pharmaceutically acceptable salts thereof
  • Another preferred embodiment of the invention are compounds of Formula I, as above including pharmaceutically acceptable salts thereof,
  • Another preferred embodiment of the invention are compounds of Formula I, as above including pharmaceutically acceptable salts thereof, wherein:
  • Q is selected from group (A) or (B), and
  • Another preferred embodiment are compounds of Formula I, including pharmaceutically acceptable salts thereof,
  • Another embodiment of the present invention is a method for treating mammals infected with the HIV virus, comprising administering to said mammal an antiviral effective amount of a compound of Formula I, including pharmaceutically acceptable salts thereof, and one or more pharmaceutically acceptable carriers, excipients or diluents; optionally the compound of Formula I can be administered in combination with an antiviral effective amount of an AIDS treatment agent selected from the group consisting of: (a) an AIDS antiviral agent; (b) an anti-infective agent; (c) an immunomodulator; and (d) HIV entry inhibitors.
  • an AIDS treatment agent selected from the group consisting of: (a) an AIDS antiviral agent; (b) an anti-infective agent; (c) an immunomodulator; and (d) HIV entry inhibitors.
  • Another embodiment of the present invention is a pharmaceutical composition
  • a pharmaceutical composition comprising an antiviral effective amount of a compound of Formula I, including pharmaceutically acceptable salts thereof, and one or more pharmaceutically acceptable carriers, excipients, diluents and optionally in combination with an antiviral effective amount of an AIDS treatment agent selected from the group consisting of: (a) an AIDS antiviral agent; (b) an anti-infective agent; (c) an immunomodulator; and (d) HIV entry inhibitors.
  • an AIDS treatment agent selected from the group consisting of: (a) an AIDS antiviral agent; (b) an anti-infective agent; (c) an immunomodulator; and (d) HIV entry inhibitors.
  • the present invention includes the individual diastereoisomeric and enantiomeric forms of the compounds of Formula I in addition to the mixtures thereof.
  • C 1-6 alkyl as used herein and in the claims (unless specified otherwise) mean straight or branched chain alkyl groups such as methyl, ethyl, propyl, isopropyl, butyl, isobutyl, t-butyl, amyl, hexyl and the like.
  • Halogen refers to chlorine, bromine, iodine or fluorine.
  • aryl group refers to an all carbon monocyclic or fused-ring polycyclic (i.e., rings which share adjacent pairs of carbon atoms) groups having a completely conjugated pi-electron system. Examples, without limitation, of aryl groups are phenyl, napthalenyl and anthracenyl. The aryl group may be substituted or unsubstituted.
  • the substituted group(s) is preferably one or more selected from alkyl, cycloalkyl, aryl, heteroaryl, heteroalicyclic, hydroxy, alkoxy, aryloxy, heteroaryloxy, heteroalicycloxy, thiohydroxy, thioaryloxy, thioheteroaryloxy, thioheteroalicycloxy, cyano, halogen, nitro, carbonyl, O-carbamyl, N-carbamyl, C-amido, N-amido, C-carboxy, O-carboxy, sulfinyl, sulfonyl, sulfonamido, trihalomethyl, ureido, amino and —NR x R y , wherein R x and R y are independently selected from the group consisting of hydrogen, alkyl, cycloalkyl, aryl, carbonyl, C-carboxy, sulfonyl, trihalomethyl,
  • heteroaryl refers to a monocyclic or fused ring (i.e., rings which share an adjacent pair of atoms) group having in the ring(s) one or more atoms selected from the group consisting of nitrogen, oxygen and sulfur and, in addition, having a completely conjugated pi-electron system. Unless otherwise indicated, the heteroaryl group may be attached at either a carbon or nitrogen atom within the heteroaryl group. It should be noted that the term heteroaryl is intended to encompass an N-oxide of the parent heteroaryl if such an N-oxide is chemically feasible as is known in the art.
  • heteroaryl groups are furyl, thienyl, benzothienyl, thiazolyl, imidazolyl, oxazolyl, oxadiazolyl, thiadiazolyl, benzothiazolyl, triazolyl, tetrazolyl, isoxazolyl, isothiazolyl, pyrrolyl, pyranyl, tetrahydropyranyl, pyrazolyl, pyridyl, pyrimidinyl, quinolinyl, isoquinolinyl, purinyl, carbazolyl, benzoxazolyl, benzimidazolyl, indolyl, isoindolyl, pyrazinyl.
  • the substituted group(s) is preferably one or more selected from alkyl, cycloalkyl, aryl, heteroaryl, heteroalicyclic, hydroxy, alkoxy, aryloxy, heteroaryloxy, heteroalicycloxy, thiohydroxy, thioaryloxy, thioheteroaryloxy, thioheteroalicycloxy, cyano, halogen, nitro, carbonyl, O-carbamyl, N-carbamyl, C-amido, N-amido, C-carboxy, O-carboxy, sulfinyl, sulfonyl, sulfonamido, trihalomethyl, ureido, amino, and —NR x R y , wherein R x and R y are as defined above.
  • a “heteroalicyclic” group refers to a monocyclic or fused ring group having in the ring(s) one or more atoms selected from the group consisting of nitrogen, oxygen and sulfur. Rings are selected from those which provide stable arrangements of bonds and are not intended to encomplish systems which would not exist. The rings may also have one or more double bonds. However, the rings do not have a completely conjugated pi-electron system.
  • heteroalicyclic groups examples, without limitation, of heteroalicyclic groups are azetidinyl, piperidyl, piperazinyl, imidazolinyl, thiazolidinyl, 3-pyrrolidin-1-yl, morpholinyl, thiomorpholinyl and tetrahydropyranyl.
  • the substituted group(s) is preferably one or more selected from alkyl, cycloalkyl, aryl, heteroaryl, heteroalicyclic, hydroxy, alkoxy, aryloxy, heteroaryloxy, heteroalicycloxy, thiohydroxy, thioalkoxy, thioaryloxy, thioheteroaryloxy, thioheteroalicycloxy, cyano, halogen, nitro, carbonyl, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, C-thioamido, N-amido, C-carboxy, O-carboxy, sulfinyl, sulfonyl, sulfonamido, trihalomethanesulfonamido, trihalomethanesulfonyl, silyl, guanyl, guanidino,
  • alkyl group refers to a saturated aliphatic hydrocarbon including straight chain and branched chain groups.
  • the alkyl group has 1 to 20 carbon atoms (whenever a numerical range; e.g., “1-20”, is stated herein, it means that the group, in this case the alkyl group may contain 1 carbon atom, 2 carbon atoms, 3 carbon atoms, etc. up to and including 20 carbon atoms). More preferably, it is a medium size alkyl having 1 to 10 carbon atoms. Most preferably, it is a lower alkyl having 1 to 4 carbon atoms.
  • the alkyl group may be substituted or unsubstituted.
  • the substituent group(s) is preferably one or more individually selected from trihaloalkyl, cycloalkyl, aryl, heteroaryl, heteroalicyclic, hydroxy, alkoxy, aryloxy, heteroaryloxy, heteroalicycloxy, thiohydroxy, thioalkoxy, thioaryloxy, thioheteroaryloxy, thioheteroalicycloxy, cyano, halo, nitro, carbonyl, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, C-thioamido, N-amido, C-carboxy, O-carboxy, sulfinyl, sulfonyl, sulfonamido, trihalomethanesulfonamido, trihalomethanesulfonyl, and combined, a five- or six-member
  • a “cycloalkyl” group refers to an all-carbon monocyclic or fused ring (i.e., rings which share and adjacent pair of carbon atoms) group wherein one or more rings does not have a completely conjugated pi-electron system.
  • examples, without limitation, of cycloalkyl groups are cyclopropane, cyclobutane, cyclopentane, cyclopentene, cyclohexane, cyclohexadiene, cycloheptane, cycloheptatriene and adamantane.
  • a cycloalkyl group may be substituted or unsubstituted.
  • the substituent group(s) is preferably one or more individually selected from alkyl, aryl, heteroaryl, heteroalicyclic, hydroxy, alkoxy, aryloxy, heteroaryloxy, heteroalicycloxy, thiohydroxy, thioalkoxy, thioaryloxy, thioheteroaryloxy, thioheteroalicycloxy, cyano, halo, nitro, carbonyl, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, C-thioamido, N-amido, C-carboxy, O-carboxy, sulfinyl, sulfonyl, sulfonamido, trihalo-methanesulfonamido, trihalomethanesulfonyl, silyl, guanyl, guanidino, ureid
  • alkenyl refers to an alkyl group, as defined herein, consisting of at least two carbon atoms and at least one carbon-carbon double bond.
  • alkynyl group refers to an alkyl group, as defined herein, consisting of at least two carbon atoms and at least one carbon-carbon triple bond.
  • a “hydroxy” group refers to an —OH group.
  • alkoxy refers to both an —O-alkyl and an —O-cycloalkyl group as defined herein.
  • aryloxy refers to both an —O-aryl and an —O-heteroaryl group, as defined herein.
  • heteroaryloxy refers to a heteroaryl-O— group with heteroaryl as defined herein.
  • heteroalicycloxy refers to a heteroalicyclic-O— group with heteroalicyclic as defined herein.
  • a “thiohydroxy” group refers to an —SH group.
  • a “thioalkoxy” group refers to both an S-alkyl and an —S-cycloalkyl group, as defined herein.
  • a “thioaryloxy” group refers to both an —S-aryl and an —S-heteroaryl group, as defined herein.
  • a “thioheteroaryloxy” group refers to a heteroaryl-S— group with heteroaryl as defined herein.
  • a “thioheteroalicycloxy” group refers to a heteroalicyclic-S— group with heteroalicyclic as defined herein.
  • a “carbonyl” group refers to a —C( ⁇ O)—R′′ group, where R′′ is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl (bonded through a ring carbon) and heteroalicyclic (bonded through a ring carbon), as each is defined herein.
  • aldehyde refers to a carbonyl group where R′′ is hydrogen.
  • a “thiocarbonyl” group refers to a —C( ⁇ S)—R′′ group, with R′′ as defined herein.
  • a “Keto” group refers to a —CC( ⁇ O)C— group wherein the carbon on either or both sides of the C ⁇ O may be alkyl, cycloalkyl, aryl or a carbon of a heteroaryl or heteroaliacyclic group.
  • a “trihalomethanecarbonyl” group refers to a Z 3 CC( ⁇ O)— group with said Z being a halogen.
  • C-carboxy refers to a —C( ⁇ O)O—R′′ groups, with R′′ as defined herein.
  • O-carboxy refers to a R′′C( ⁇ O)O-group, with R′′ as defined herein.
  • a “carboxylic acid” group refers to a C-carboxy group in which R′′ is hydrogen.
  • a “trihalomethyl” group refers to a —CZ 3 , group wherein Z is a halogen group as defined herein.
  • a “trihalomethanesulfonyl” group refers to an Z 3 CS( ⁇ O) 2 — groups with Z as defined above.
  • a “trihalomethanesulfonamido” group refers to a Z 3 CS( ⁇ O) 2 NR x — group with Z and R X as defined herein.
  • a “sulfinyl” group refers to a —S( ⁇ O)—R′′ group, with R′′ as defined herein and, in addition, as a bond only; i.e., —S(O)—.
  • a “sulfonyl” group refers to a —S( ⁇ O) 2 R′′ group with R′′ as defined herein and, in addition as a bond only; i.e., —S(O) 2 —.
  • S-sulfonamido refers to a —S( ⁇ O) 2 NR X R Y , with R X and R Y as defined herein.
  • N-Sulfonamido refers to a R′′S( ⁇ O) 2 NR X — group with R x as defined herein.
  • a “O-carbamyl” group refers to a —OC( ⁇ O)NR x R y as defined herein.
  • N-carbamyl refers to a R x OC( ⁇ O)NR y group, with R x and R y as defined herein.
  • a “O-thiocarbamyl” group refers to a —OC( ⁇ S)NR x R y group with R x and R y as defined herein.
  • N-thiocarbamyl refers to a R x OC( ⁇ S)NR y — group with R x and R y as defined herein.
  • amino refers to an —NH 2 group.
  • a “C-amido” group refers to a —C( ⁇ O)NR x R y group with R x and R y as defined herein.
  • C-thioamido refers to a —C( ⁇ S)NR x R y group, with R x and R y as defined herein.
  • N-amido group refers to a R x C( ⁇ O)NR y — group, with R x and R y as defined herein.
  • an “ureido” group refers to a —NR x C( ⁇ O)NR y R y2 group with R x and R y as defined herein and R y2 defined the same as R x and R y .
  • An “thioureido” group refers to a —NR x C( ⁇ S)NR y R y2 group with R x and R y as defined herein and R y2 defined the same as R x and R y .
  • a “guanidino” group refers to a —R x NC( ⁇ N)NR y R y2 group, with R x , R y and R y2 as defined herein.
  • a “guanyl” group refers to a R x R y NC( ⁇ N)— group, with R x and R y as defined herein.
  • a “cyano” group refers to a —CN group.
  • a “silyl” group refers to a —Si(R′′) 3 , with R′′ as defined herein.
  • a “phosphonyl” group refers to a P( ⁇ O)(OR x ) 2 with R x as defined herein.
  • a “hydrazino” group refers to a —NR x NR y R y2 group with R x , R y and R y2 as defined herein.
  • Any two adjacent R groups may combine to form an additional aryl, cycloalkyl, heteroaryl or heterocyclic ring fused to the ring initially bearing those R groups.
  • nitogen atoms in heteroaryl systems can be “participating in a heteroaryl ring double bond”, and this refers to the form of double bonds in the two tautomeric structures which comprise five-member ring heteroaryl groups. This dictates whether nitrogens can be substituted as well understood by chemists in the art.
  • the disclosure and claims of the present invention are based on the known general principles of chemical bonding. It is understood that the claims do not encompass structures known to be unstable or not able to exist based on the literature.
  • Physiologically acceptable salts and prodrugs of compounds disclosed herein are within the scope of this invention.
  • pharmaceutically acceptable salt as used herein and in the claims is intended to include nontoxic base addition salts. Suitable salts include those derived from organic and inorganic acids such as, without limitation, hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid, methanesulfonic acid, acetic acid, tartaric acid, lactic acid, sulfinic acid, citric acid, maleic acid, fumaric acid, sorbic acid, aconitic acid, salicylic acid, phthalic acid, and the like.
  • salts of acidic groups such as a carboxylate
  • suitable organic bases such as lower alkylamines (methylamine, ethylamine, cyclohexylamine, and the like) or with substituted lower alkylamines (e.g. hydroxyl-substituted alkylamines such as diethanolamine, triethanolamine or tris(hydroxymethyl)-aminomethane), or with bases such as piperidine or morpholine.
  • the term “antiviral effective amount” means the total amount of each active component of the method that is sufficient to show a meaningful patient benefit, i.e., healing of acute conditions characterized by inhibition of the HIV infection.
  • a meaningful patient benefit i.e., healing of acute conditions characterized by inhibition of the HIV infection.
  • the term refers to that ingredient alone.
  • the term refers to combined amounts of the active ingredients that result in the therapeutic effect, whether administered in combination, serially or simultaneously.
  • the terms “treat, treating, treatment” as used herein and in the claims means preventing or ameliorating diseases associated with HIV infection.
  • the present invention is also directed to combinations of the compounds with one or more agents useful in the treatment of AIDS.
  • the compounds of this invention may be effectively administered, whether at periods of pre-exposure and/or post-exposure, in combination with effective amounts of the AIDS antivirals, immunomodulators, antiinfectives, or vaccines, such as those in the following table.
  • AIDS, ARC, HIV Sulfate Ind. Ltd. (Osaka, positive Japan) asymptomatic ddC Hoffman-La Roche HIV infection, AIDS, Dideoxycytidine ARC ddI Bristol-Myers Squibb HIV infection, AIDS, Dideoxyinosine ARC; combination with AZT/d4T DMP-450 AVID HIV infection, (Camden, NJ) AIDS, ARC (protease inhibitor) Efavirenz DuPont Merck HIV infection, (DMP 266) AIDS, ARC ( ⁇ )6-Chloro- (non-nucleoside RT 4-(S)-cyclo- inhibitor) propylethynyl- 4(S)-trifluoro- methyl-1,4- dihydro-2H-3,1- benzoxazin-2- one, STOCRINE EL10 Elan Corp, PLC HIV infection (Gainesville, GA) Famciclovir Smith Kline herpes zoster, herpes simplex
  • HIV infection HIV infection, AIDS, ARC Recombinant Triton Biosciences AIDS, Kaposi's Human (Almeda, CA) sarcoma, ARC Interferon Beta Interferon Interferon Sciences ARC, AIDS alfa-n3 Indinavir Merck HIV infection, AIDS, ARC, asymptomatic HIV positive, also in combination with AZT/ddI/ddC ISIS 2922 ISIS Pharmaceuticals CMV retinitis KNI-272 Nat'l Cancer Institute HIV-assoc.
  • Lamivudine Glaxo Wellcome HIV infection, 3TC AIDS, ARC (reverse transcriptase inhibitor); also with AZT Lobucavir Bristol-Myers Squibb CMV infection Nelfinavir Agouron HIV infection, Pharmaceuticals AIDS, ARC (protease inhibitor) Nevirapine Boeheringer HIV infection, Ingleheim AIDS, ARC (RT inhibitor) Novapren Novaferon Labs, Inc. HIV inhibitor (Akron, OH) Peptide T Peninsula Labs AIDS Octapeptide (Belmont, CA) Sequence Trisodium Astra Pharm. CMV retinitis, HIV Phosphono- Products, Inc.
  • HIV infection other CMV formate infections
  • AIDS other CMV formate infections
  • PNU-140690 Pharmacia Upjohn HIV infection, AIDS, ARC (protease inhibitor) Probucol Vyrex HIV infection, AIDS RBC-CD4 Sheffield Med. Tech HIV infection, (Houston, TX) AIDS, ARC Ritonavir Abbott HIV infection, AIDS, ARC (protease inhibitor) Saquinavir Hoffmann-LaRoche HIV infection, AIDS, ARC (protease inhibitor) Stavudine; d4T Bristol-Myers Squibb HIV infection, AIDS, Didehydro- ARC deoxythymidine Valaciclovir Glaxo Wellcome Genital HSV & CMV infections Virazole Viratek/ICN asymptomatic HIV Ribavirin (Costa Mesa, CA) positive, LAS, ARC VX-478 Vertex HIV infection, AIDS, ARC Zalcitabine Hoffmann-LaRoche HIV infection, AIDS, ARC, with AZT Zi
  • AIDS ARC (Irving, TX) CL246, 738 American Cyanamid AIDS, Kaposi's Lederle Labs sarcoma EL10 Elan Corp, PLC HIV infection (Gainesville, GA) FP-21399 Fuki ImmunoPharm Blocks HIV fusion with CD4+ cells
  • Gamma Genentech ARC in combination Interferon w/TNF (tumor necrosis factor) Granulocyte Genetics Institute AIDS Macrophage Sandoz Colony Stimulating Factor Granulocyte Hoechst-Roussel AIDS Macrophage Immunex Colony Stimulating Factor Granulocyte Schering-Plough AIDS, combination Macrophage w/AZT Colony Stimulating Factor HIV Core Rorer Seropositive HIV Particle Immuno- stimulant IL-2 Cetus AIDS, in Interleukin-2 combination w/AZT IL-2 Hoffman-LaRoche AIDS, ARC, HIV, in Interleukin-2 Immunex combination w/AZT
  • Kaposi's sarcoma Muramyl- Tripeptide Granulocyte Amgen AIDS, in Colony combination w/AZT Stimulating Factor Remune Immune Response Immunotherapeutic Corp.
  • rCD4 Genentech AIDS ARC Recombinant Soluble Human CD4 rCD4-IgG AIDS, ARC hybrids Recombinant Biogen AIDS, ARC Soluble Human CD4 Interferon Hoffman-La Roche Kaposi's sarcoma Alfa 2a AIDS, ARC, in combination w/AZT SK&F106528 Smith Kline HIV infection Soluble T4 Thymopentin Immunobiology HIV infection Research Institute (Annandale, NJ) Tumor Necrosis Genentech ARC, in combination Factor; TNF w/gamma Interferon ANTI-INFECTIVES Clindamycin Pharmacia Upjohn PCP with Primaquine Fluconazole Pfizer Cryptococcal meningitis, candidiasis Pastille Squib
  • the compounds of the invention herein may be used in combination with another class of agents for treating AIDS which are called HIV entry inhibitors.
  • HIV entry inhibitors are discussed in DRUGS OF THE FUTURE 1999, 24(12), pp. 1355-1362; CELL, Vol. 9, pp. 243-246, Oct. 29, 1999; and DRUG DISCOVERY TODAY, Vol. 5, No. 5, May 2000, pp. 183-194 and Inhibitors of the entry of HIV into host cells. Meanwell, Nicholas A.; Kadow, John F. Current Opinion in Drug Discovery & Development (2003), 6(4), 451-461.
  • the compounds can be utilized in combination with other attachment inhibitors, fusion inhibitors, and chemokine receptor antagonists aimed at either the CCR5 or CXCR4 coreceptor.
  • Preferred combinations are simultaneous or alternating treatments with a compound of the present invention and an inhibitor of HIV protease and/or a non-nucleoside inhibitor of HIV reverse transcriptase.
  • An optional fourth component in the combination is a nucleoside inhibitor of HIV reverse transcriptase, such as AZT, 3TC, ddC or ddI.
  • a preferred inhibitor of HIV protease is Reyataz® (active ingredient Atazanavir). Typically a dose of 300 to 600 mg is administered once a day. This may be co-administered with a low dose of Ritonavir (50 to 500 mgs).
  • Another preferred inhibitor of HIV protease is Kaletra®.
  • indinavir is the sulfate salt of N-(2(R)-hydroxy-1-(S)-indanyl)-2(R)-phenylmethyl-4-(S)-hydroxy-5-(1-(4-(3-pyridyl-methyl)-2(S)-N′-(t-butylcarboxamido)-piperazinyl))-pentaneamide ethanolate, and is synthesized according to U.S. Pat. No. 5,413,999.
  • Indinavir is generally administered at a dosage of 800 mg three times a day.
  • Other preferred protease inhibitors are nelfinavir and ritonavir.
  • HIV protease is saquinavir which is administered in a dosage of 600 or 1200 mg tid.
  • Preferred non-nucleoside inhibitors of HIV reverse transcriptase include efavirenz.
  • the preparation of ddC, ddI and AZT are also described in EPO 0,484,071. These combinations may have unexpected effects on limiting the spread and degree of infection of HIV.
  • Preferred combinations include those with the following (1) indinavir with efavirenz, and, optionally, AZT and/or 3TC and/or ddI and/or ddC; (2) indinavir, and any of AZT and/or ddI and/or ddC and/or 3TC, in particular, indinavir and AZT and 3TC; (3) stavudine and 3TC and/or zidovudine; (4) zidovudine and lamivudine and 141W94 and 1592U89; (5) zidovudine and lamivudine.
  • the compound of the present invention and other active agents may be administered separately or in conjunction.
  • the administration of one element may be prior to, concurrent to, or subsequent to the administration of other agent(s).
  • the present invention comprises compounds of Formula I, their pharmaceutical formulations, and their use in patients suffering from or susceptible to HIV infection.
  • the compounds of Formula I include pharmaceutically acceptable salts thereof.
  • Scheme A depicts one of the preferred methods for preparing the compounds of the invention.
  • a functionalized diazaindole which also has a carboxy ester appended to the three position is condensed with an acetonitrile anion functionalized with Y to provide the alpha cyano ketone examples of the invention.
  • Step A The carboxylic ester intermediates Z-CO 2 R or more preferably the acid chlorides Z-CO 2 Cl from Scheme A are condensed with a cyanomethyl intermediate YCH 2 CN under basic conditions to form the ⁇ -cyanoketo intermediate ZC(O)CH(CN)Y.
  • the base KHMDS in THF at r.t. is employed most often, but other amide bases such as NaHMDS could be utilized.
  • the typical solvent utilized is THF but DMF can be employed for less soluble molecules.
  • reaction with an acid chloride Z-CO 2 Cl is conducted with the reaction flask immersed in a dry ice acetone cooling bath ( ⁇ 78° C.) when THF is the solvent and an acetonitrile/acetone cooling bath ( ⁇ 42° C.) when DMF is the solvent but temperatures between ⁇ 78° and 50° C. could be employed in appropriate cases.
  • the reaction is stirred between 1 h and 1 day.
  • the reaction when judged to be complete by TLC or LC or LC/MS is maintained at the cold temperature and oxidant added directly to the reaction as described in Step B.
  • the reaction could be allowed to attain ambient temperature and either allowed to react further if necessary and then quenched or be immediately quenched with saturated aqueous sodium bicarbonate.
  • This is currently the preferred method for diazaindole esters of this invention.
  • the preparation of the acid chlorides from Z-CO 2 R— is accomplished by initial hydrolysis of the ester to the analogous carboxylic acid Z-CO 2 H. A typical procedure involves stirring the ester with LiOH in THF and water at 100° C. for 6 h to 2 days, concentrating the crude mixture and recrystallizing the carboxylic acid from water.
  • the carboxylic acid Z-CO 2 H is then dissolved or more typically suspended as a slurry in dichloromethane and stirred with oxalyl chloride and a catalytic amount of DMF from 4-24 h but typically overnight.
  • the solvents are removed in vacuo and the acid chloride used directly.
  • Possible alternative solvents are benzene or toluene.
  • a possible alternative method for conversion of the carboxylic acid to an acid chloride entails reacting thionyl choride in benzene at 100° C. between 2 h and 6 h with the acid in the presence of catalytic DMF followed by concentration in vacuo to yield the acid chloride Z-CO 2 Cl.
  • the acid chloride Z-CO 2 Cl is the preferred reactant for conducting step A for the preparation of diazindole compounds of formula I.
  • the acid may be converted to an acid anhydrides which may also find utility in the alkylation reaction.
  • Step B The preferred method for accomplishing step B, the conversion of the ⁇ -cyanoketo intermediate ZC(O)CH(CN)Y to the diacarbonyl compounds of formula I or ketoamide intermediates to prepare compounds of formula I is to to add 1-20 equivalents but most preferably 5 equivalents of a commercially available solution of 32% peracetic acid in dilute aq acetic acid tb the reaction flask containing the completed reaction described in Step A.
  • the reaction is typically stirred at the same temperature at which the alkylation reaction was conducted (for the Step A reactions with an acid chloride in THF ⁇ 78° and for the step A reactions in DMF ⁇ 42°) for a period of 1 h and then allowed to warm to ambient temperature if not already at that tmeperature.
  • the reaction mixture is then either allowed to react further or immediately diluted with saturated aq. ammonium chloride and EtOAc.
  • the resultant precipitate is isolated by filtration as the oxoacetyl product ZC(O)C(O)Y.
  • organic soluble acid products the acid is extracted into the organic layer and the layers separated.
  • the organic layer is concentrated in vacuo and the product purified via preparative HPLC.
  • the ⁇ -cyanoketo intermediate ZC(O)CH(CN)Y if isolated, can be oxidized to the oxoacetyl product ZC(O)C(O)Y using a variety of oxidants including mCPBA, NaOCl (bleach), peracetic acid, or nickel peroxide.
  • mCPBA mCPBA
  • NaOCl (bleach) peracetic acid
  • nickel peroxide nickel peroxide.
  • a solution of peracetic acid in acetic acid is added to a solution of ⁇ -cyanoketo intermediate ZC(O)CH(CN)Y in THF and the reaction is stirred at between r.t and ⁇ 70° C. for between 30 min and 2 h.
  • the reaction mixture is then diluted with saturated aq.
  • Step A and Step B can be combined into a one pot reaction by adding the oxidant directly to the reaction pot after the completion of step A without isolating the ⁇ -cyanoketo intermediate ZC(O)CH(CN)Y.
  • Scheme B depicts a typical method for preparing the cyanomethyl piperazine or piperidine analogues utilized in scheme A.
  • Two general literature references for some of the chemistry depicted in these initial schemes are Takahashi, K.; Shibasaki, K.; Ogura, K.; Iida, H.; Chem Lett. 1983, 859 or Yang; Z.; Zhang, Z.; Meanwell, N. A.; Kadow, J. F.; Wang, T.; Org. Lett. 2002, 4, 1103.
  • Step C The secondary amine of a functionalized piperazine or piperidine can be alkylated with a haloacetonitrile under basic conditions to yield a cyanomethyl piperazine or piperidine analogue.
  • N-benzoyl piperazine was added to a solution of chloroacetonitrile and TEA in THF and stirred at r.t. for between 2 and 5 days.
  • a resulting precipitate is removed by filtration, the filtrate is concentrated in vacuo, and the residue purified via chromatography to yield the cyanomethyl intermediate YCH 2 CN.
  • the alkylation with haloacetonitrile can also be carried out with an alternate base, such as 4-methylmorpholine or diisopropylethyl amine.
  • Step D The reaction of an (alkoxymethylene)cyanoacetate with an amino malonate under basic conditions is known to yield a 2,4-dicarboxylic ester-3-aminopyrrole.
  • step D is carried out by reacting an amino malonate with an 2-alkoxy 1-cyano acrylate in the presence of a base such as sodium ethoxide.
  • a base such as sodium ethoxide.
  • Step E The 3-Aminopyrrole 2-carboxylic ester resulting from step D can be cyclized to the desired 7-hydroxyl-4,6-diazaindole using a number of reagents including formamides, dialkyl acetal formamides, nitriles and formamidines.
  • 3-aminopyrrole-2,4-dicarboxylic acid diethyl ester and formidine acetate are heated at reflux in EtOH for 1 to 3 days. The reaction solution is filtered hot and the product usually crystallized upon cooling and is then rinsed with diethyl ether.
  • Step F A 3-carboxylic ester 7-hydroxyl-4,6-diazaindole can then be converted to a 7-chloro analogue by treatment with a chlorinating reagent such as POCl 3 or SOCl 2 .
  • a chlorinating reagent such as POCl 3 or SOCl 2 .
  • 3-ethylester-7-hydroxyl-4,6-diazaindole and POCl 3 are combined and heated at 105° C. for between 3 and 5 h, cooled to r.t. and diluted with diethyl ether.
  • the precipitate that forms is collected by filtration and was shown to be the 7-chloro-4,6-diazaindole.
  • the corresponding 7-bromo-4,6-diazindole may be prepared by substituting POBr 3 for the chlorinating agents described above.
  • Step G A 7-chloro-4,6-diazaindole can be displaced with a variety of nucleophiles to form the claimed R 5 substituents or intermediates from which the claimed R 5 substituents can be formed. Included in these are cyanide, alkoxides, amines, alcohols and various metallated species (cuprates, lithiates, zincates and Grignard reagents).
  • 3-ethylester-7-chloro-4,6-diazaindole and 3-methyl pyrazole in EtOH are heated at between 100° C. and 140° C. for 20 min to 1 h. Upon cooling the reaction is concentrated and purified by silica gel chromatography or by preparative HPLC.
  • This step may also be carried out after the initial coupling and oxidation steps (steps A and B) have been preformed on the 3-ethylester-7-chloro-4,6-diazaindole intermediate.
  • a cyano moiety could be introduced and converted to acids, esters, amides, imidates, or heteraromatics. Typical amide coupling methodology could be used to prepare amides from acids. It should also be noted that the halogen moiety may be carried through until compounds of the invention are realized and then the conditions described in Step G may frequently be used to prepare further compounds of the invention.
  • a 7-chloro or 7-bromo-4,6-diazaindole could be coupled to a heteroaryl stannane or boronic ester via Stille or Suzuki methodology respectively.
  • Other metal catalyzed methodology such as copper mediated displacements could also be used to prepare N linked heteraromatic or heteroalicyclic derivatives.
  • substituted diazaindoles containing a chloride, bromide, iodide, triflate, or phosphonate should undergo coupling reactions with a boronate (Suzuki type reactions) or a stannane to provide substituted diazaindoles.
  • Stannanes and boronates are prepared via standard literature procedures or as described in the experimental section of this application.
  • the vinyl bromides, chlorides, triflates, or phosphonates may undergo metal mediated coupling to provide compounds of formula W-H. Stille or Suzuki couplings are particularly useful.
  • a detailed discussion of the references and best conditions for these kinds of metal mediated coupling is described later in this application where the discussion is combined with a description of how these types of reactions may also be used to funtionalize diazaindoles.
  • applications incorporated in their entirety elsewhere in this application contain methods for preparing heteroaryls from funtional groups appended to indoles and azaindoles. This methodology is also applicable to diazaindoles.
  • Step H A TMS-isocyanide would be reacted with an acid fluoride in the presence of a dialkyl acetylene dicarboxylate to form a substituted pyrrole.
  • a dialkyl acetylene dicarboxylate For representative examples see: Livinghouse, T.; Smith, R.; J. Chem. Soc, Chem. Commun 1983, 5, 210.
  • trimethylsilylmethyl isocyanide (generated from the lithiation of methyl isocyanide, followed by silylation with TMSCl) would be stirred with an aryl acid fluoride and dimethyl acetylenedicarboxylate in toluene at 80° C.
  • TMSCl trimethylsilylmethyl isocyanide
  • Step I A mixture of the keto-diester-pyrrole and hydrazine dihydrochloride in ethanol heated at reflux should result in the formation of the desired 4-hydroxyl-5,6-diazaindole.
  • the keto-diester-pyrrole, hydrazine hydrate and a catalytic amount of p-toluenesulfonic acid could be heated to reflux in toluene or benzene in the presence of a Dean-Stark trap and upon dehydration, the desired 4-hydroxyl-5,6-diazaindole should form.
  • Step J, Step K and Step L A 4-hydroxyl-5,6-diazaindole intermediate could be converted to the intermediates in which R 3 is modified by direct functionalization of the hydroxyl group or by conversion of the hydroxyl group to a leaving group (halogen or triflate) followed by nucleophilic displacement or a metal (Pd or Cu) mediated coupling.
  • step(s) might also be carried out after the initial coupling and oxidation steps (steps A and B) have been preformed on the 4-hydroxyl-5,6-diazaindole intermediate.
  • the conditions described for step G could also be utilized for this system.
  • step DD1 Journal of the Chemical Society [Section] C: Organic (1970), (2), 285-90. could be reacted with hydrazine in ethanol between RT and reflux to provide the cyclized product of step DD1.
  • Reaction with phosphoryl chloride (2.2 to 5 equivalents should provide the dichloride as shown in step DD2.
  • step DD3 selective reaction of the C-7 chloride could occur by using benzyl alcohol and triethylamine in a cosolvent such as THF.
  • step DD4 the 4-chloro group might then be displaced with sodium or potassium methoxide in solvents such as methanol or toluene or a mixture. Stoichiometric copper I iodide could be added to speed slow reactions.
  • step DD5 selective hydrogenation of the benzyl group using 5 to 10% Pd/C in EtOH under a balloon pressure of hydrogen brovides the 7-hydroxy compound.
  • the benzyl group may be cleaved selectively with TMSI in acetonitrile at temperatures from 0 to 65° C. or using HBr in 1,2,dichloroethane at temps from ⁇ 20 to 50° C.
  • An alternate prep is to react the dichlorointemrediate above with methoxide rather than benzyl alcohol and then to selectivel cleave the C-7 ether using conditions described for the benzyl cleavage.
  • Step DD6 describes acylation of the functionalized intermediate and is done using the same procedures described in step O of Scheme F.
  • Step DD7 amide copuling with piperazine or piperidine is carried out according to the general procedures described in Step P of Scheme F to provide compounds of the invention. It should be understood that the order of steps DD5-DD7 could be switched to determine which order provides best yields.
  • Step N Nucleophilic or metal catalyzed substitution of the 4-chloro-5,7-diazaindole will yield the R 3 substituents of claim 1 .
  • the diazaindole core may be coupled to the functionalized piperidine or piperazine through an oxoacetate or through an acylation/amidation process as shown in Scheme F.
  • Step O Conversion of a specific 3H-diazaindole to the depicted ketoacid might be accomplished via several methods.
  • Method a for step O One successful method has been to use Fridel-Crafts acylation conditions mediated by an ionic liquid.
  • the ionic liquid 1-alkyl-3-alkylimidazolium chloroaluminate is generally useful in promoting the Friedel-Crafts type acylation and does work with some diazainoles.
  • the ionic liquid is generated by mixing 1-alkyl-3-alkylimidazolium chloride with aluminium chloride at room temperature with vigorous stirring.
  • glyoxyl ester could be hydrolyzed in situ at ambient temperature upon prolonged reaction time (typically overnight) to give the corresponding glyoxyl acid which would be ready for amide formation.
  • a representative experimental procedure is as follows: 1-ethyl-3-methylimidazolium chloride (2 equiv.; purchased from TCI; weighted under a stream of nitrogen) would be stirred in an oven-dried round bottom flask at r.t. under a nitrogen atmosphere, and then aluminium chloride (6 equiv.; anhydrous powder packaged under argon in ampules purchased from Aldrich preferred would be added; after weighing under a stream of nitrogen). The mixture would be vigorously stirred to form a liquid, to which would then be added diazaindole (1 equiv.) followed by stirring until a homogenous mixture resulted.
  • Step O method B The diazaindole could be treated with a Grignard reagent such as MeMgI (methyl magnesium iodide), methyl magnesium bromide or ethyl magnesium bromide and then a zinc halide, such as ZnCl 2 (zinc chloride) or zinc bromide, followed by the addition of an oxalyl chloride mono ester, such as ClCOCOOMe (methyl chlorooxoacetate) or another ester as above, to afford the diaza-indole glyoxyl ester.
  • Oxalic acid esters such as methyl oxalate, ethyl oxalate or as above are used.
  • Aprotic solvents such as CH 2 Cl 2 , Et 2 O, benzene, toluene, DCE, THF, dioxane or the like could potentially be used alone or in combination for this sequence.
  • Step O method c A Lewis acid catalyzed Friedel-Crafts reaction under standard conditions with an alkyl chloroacetoacetate might be utilized. This could be followed by in situ by hydrolysis of the ester my the method described below to form the diazaindole ketocarboxylic acid (cite previous patent(s)).
  • the diazindole ketoester precursors to the depicted acid could be prepared by reaction of diazaindoles with an excess of ClCOCOOMe in the presence of AlCl 3 (aluminum chloride).
  • oxalate esters such as ethyl or benzyl mono esters of oxalic acid could also suffice for either method shown above. More lipophilic esters ease isolation during aqueous extractions.
  • Lewis acid catalysts such as tin tetrachloride, titanium IV chloride, and aluminum chloride could be employed with this transformation with aluminum chloride being most preferred.
  • Step O Hydrolysis methods for Step O. Hydrolysis of a diazindole keto methyl ester would afford a potassium salt of the acid product shown as the product for Step O in Scheme F and this would then be ready for coupling with amines as shown in the next step. Acidification during workup, typically with aqueous HCl would provide the acid products from Step O as shown. Some typical conditions employ methanolic or ethanolic sodium hydroxide followed by careful acidification with aqueous hydrochloric acid of varying molarity but 1M HCl is preferred. The acidification is not utilized in many cases as described above for the preferred conditions. Lithium hydroxide or potassium hydroxide could also be employed and varying amounts of water could be added to the alcohols.
  • Propanols or butanols could also be used as solvents. Elevated temperatures up to the boiling points of the solvents may be utilized if ambient temperatures do not suffice. Alternatively, the hydrolysis may be carried out in a non polar solvent such as CH 2 Cl 2 or THF in the presence of Triton B. Temperatures of ⁇ 78° C. to the boiling point of the solvent may be employed but ⁇ 10° C. is preferred. Other conditions for ester hydrolysis are listed in reference 41 and both this reference and many of the conditions for ester hydrolysis are well known to chemists of average skill in the art.
  • the ketocarboxylic acid may be coupled with functionalized piperidines or piperazines using a number of standard amide bond or peptide bond forming coupling reagents.
  • the acid intermediate Z-C(O)(O)OH from Scheme F could be coupled with either a substituted piperazine or piperidine, H—Y using standard amide bond or peptide bond forming coupling reagents.
  • the combination of EDAC and triethylamine in tetrahydrofuran or BOPCl and diisopropyl ethyl amine in chloroform could be utilized but DEPBT, or other coupling reagents such as PyBop could be utilized.
  • Another useful coupling condition employs HATU (L. A. Carpino et.
  • DEPBT (3-(diethoxyphosphoryloxy)-1,2,3-benzotriazin-4(3H)-one) and N,N-diisopropylethylamine, commonly known as Hunig's base, represents another efficient method to form the amide bond (step P).
  • DEPBT is either purchased from Adrich or prepared according to the procedure of Ref. 28, Li, H.; Jiang, X.; Ye, Y.-H.; Fan, C.; Romoff, T.; Goodman, M. Organic Lett., 1999, 1, 91-93.
  • an inert solvent such as DMF or THF is used but other aprotic solvents could be used.
  • the amide bond construction reaction could be carried out using the preferred conditions described above, the EDC conditions described below, other coupling conditions described in this application, or alternatively by applying the conditions or coupling reagents for amide bond construction described in incorporated applications for construction of substituents R 2 —R 5 on indoles or azaindoles. Some specific nonlimiting examples are given in this application.
  • the acid could be converted to a methyl ester using excess diazomethane in THF/ether.
  • the methyl ester in dry THF could be reacted with the lithium amide of intermediate H—Y.
  • the lithium amide of H—Y, Li—Y is formed by reacting H—Y with lithium bistrimethylsilylamide in THF for 30 minutes in an ice water cooling bath.
  • Sodium or potassium amides could be formed similarly and utilized if additional reactivity is desired.
  • Other esters such as ethyl, phenyl, or pentafluorophenyl could be utilized and would be formed using standard methodology.
  • the acid can be converted to the acid chloride using oxalyl chloride in a solvent such as benzene or thionyl chloride either neat or containing a catalystic amount of DMF. Temperatures between 0° C. and reflux may be utilized depending on the substrate.
  • Compounds of Formula I can be obtained from the resultant compounds of formula Z-C(O)(O)Cl by reaction with the appropriate H—Y in the presence of a tertiary amine (3-10 eq.) such as triethylamine or diisopropylethylamine in an anhydrous aprotic solvent such as dichloromethane, dichloroethane, diethyl ether, dioxane, THF, acetonitrile, DMF or the like at temperatures ranging from 0° C. to reflux. Most preferred are dichloromethane, dichloroethane, or THF.
  • the reaction can be monitored by LC/MS.
  • the 3H-diazaindoles may also be prepared under Bartoli or Liemgruber-Batchko reaction conditions as shown in schemeG. Conditions for carrying out these reactions were contained in the incorporated patent applications.
  • Step Q in Scheme G depicts a potential synthesis of a diazaindole intermediate, via the well known Bartoli reaction in which vinyl magnesium bromide reacts with an aryl or heteroaryl nitro groups, to form a five-membered nitrogen containing ring as shown.
  • Some references for the above transformation to form an indole ring include: Bartoli et al. a) Tetrahedron Lett. 1989, 30, 2129. b) J. Chem. Soc. Perkin Trans. 1 1991, 2757. c) J. Chem. Soc. Perkin Trans. II 1991, 657. d) SynLett (1999), 1594.
  • a solution of vinyl Magnesium bromide in THF (typically 1.0M but from 0.25 to 3.0M) is added dropwise to a solution of the nitro pyridine in THF at ⁇ 78° under an inert atmosphere of either nitrogen or Argon. After addition is completed, the reaction temperature is allowed to warm to ⁇ 20° and then is stirred for approximately 12 h before quenching with 20% aq ammonium chloride solution. The reaction is extracted with ethyl acetate and then worked up in a typical manner using a drying agent such as anhydrous magnesium sulfate or sodium sulfate. Products are generally purified using chromatography over Silica gel. Best results are generally achieved using freshly prepared vinyl Magnesium bromide. In some cases, vinyl Magnesium chloride may be substituted for vinyl Magnesium bromide.
  • Step R Reaction with dimethylformamide dimethyl acetal in an inert solvent or neat under conditions for forming Batcho-Leimgruber precursors would provide the cyclization precursor, 33, as shown.
  • a typical condition would employ 20% DMF dimethyl acetal in DMF heated to 105-110 degrees C.
  • the pyridine may be oxidized to the N-oxide prior to the reaction using a peracid such as MCPBA or a more potent oxidant like meta-trifluoromethyl or meta nitro peroxy benzoic acids.
  • Step S Reduction of the nitro group using for example hydrogenation over Pt on/C catalyst in a solvent such as MeOH, EtOH, or EtOAc could provide the cyclized product. Generally only a slight positive pressure of hydrogen would be required (a stream) but higher pressures may be needed (1.5 atm). Alternatively the reduction may be carried out using tin dichloride and HCl, hydrogenation over Raney nickel or other catalysts, or by using other methods for nitro reduction such as described elsewhere in this application.
  • a solvent such as MeOH, EtOH, or EtOAc
  • Step T 1,2,3,4-Tetrazines have been shown to react with pyrrole and substituted pyrroles to form 5,6-diazaindole products.
  • This reaction proceeds through a [4+2]-cycloaddition followed by a retro-[4+2]-cycloaddition to release nitrogen gas and a subsequent oxidation to establish aromaticity.
  • Gonzalez, J. C. Lobo-Antunes, J; Perez-Lourido, P.; Santana, L.; Uriate, E. Synthesis 2002, 4, 475-478.
  • Step U The starting pyridazine N-oxide would initially be nitrated and the resulting nitro group then would be reduced under standard conditions to an amine. The chloro would then be removed under hydrogentation conditions. Alternatively, the chloro could remain in the molecule and be carried through the subsequent steps. This should allow for the formation of a 4-chloro-5,6-diazaindole. The chloro could then be converted to a methoxy or an amino group by nucleophilic displacement or copper catalyzed assisted coupling. This would result in an intermediate that could be converted to molecules claimed within this application via previously described amide bond coupling.
  • Step V The amine could then be functionalized with ethyl orthoformate under acidic conditions to form an ethoxyimine.
  • the amine and triethyl orthoformate were dissolved into a solution of DMF and ethanol that had been adjusted to pH 1 with anhydrous hydrogen chloride. The reaction was then heated to 150-160° C. and ethanol was collected by distillation resulting in the formation of the desired ethoxyimine.
  • Step W Deprotonation of the methyl group followed by acylation with diethyl oxalate would yield a ketoester intermediate that could be used to form a 3-oxoacetate-5,6-diazaindole (Step X) or could be used to make a 2-carboxylate-5,6-diazaindole by hydrolysis of the imine, followed by condensation of the amine onto the ketone five centers away.
  • Step X The ketoester could then be cyclized onto the ethoxyimine under basic conditions to arrive at the 3-oxoacetate-5,6-diazaindole.
  • functionalized piperazine or piperidones could be coupled to the ester through standard amide bond forming reactions.
  • This general scheme should also allow for the preparation of other 5,6-diazaindole intermediates with different R 3 and R 5 substituents by displacement or coupling to the chloro or displacement of the methoxy at some point in the sequence.
  • Step F15 substituted diazaindoles containing a chloride, bromide, iodide, triflate, or phosphonate could undergo coupling reactions with a boronate (Suzuki type reactions) or a stannane to provide substituted diazaindoles.
  • boronate Sudzuki type reactions
  • stannane a stannane to provide substituted diazaindoles.
  • Stannanes and boronates are prepared via standard literature procedures or as described in the experimental section of this application.
  • the substitututed diazindoles may undergo metal mediated coupling to provide compounds of Formula I wherein R 4 is aryl, heteroaryl, or heteroalicyclic for example.
  • Preferred procedures for coupling of a chloro diazaindole and a stannane employ dioxane, stoichiometric or an excess of the tin reagent (up to 5 equivalents), 0.1 to 1 eq of Palladium (0) tetrakis triphenyl phosphine in dioxane heated for 5 to 15 h at 110 to 120°.
  • Other solvents such as DMF, THF, toluene, or benzene could be employed.
  • the boronate or stannane could potentially be formed on the diazaindole via methods known in the art and the coupling performed in the reverse manner with aryl or heteroaryl based halogens or triflates.
  • boronate or stannane agents could be either purchased from commercial resources or prepared following disclosed documents. Additional examples for the preparation of tin reagents or boronate reagents are contained in the experimental section.
  • Novel stannane agents could be prepared from one of the following routes.
  • Boronate reagents are prepared as described in reference 71. Reaction of lithium or Grignard reagents with trialkyl borates generates boronates. Alternatively, Palladium catalyzed couplings of alkoxy diboron or alkyl diboron reagents with aryl or heteroaryl halides can provide boron reagents for use in Suzuki type couplings. Some example conditions for coupling a halide with (MeO)BB(OMe)2 utilize PdCl2 (dppf), KOAc, DMSO, at 80° C. until reaction is complete when followed by TLC or HPLC analysis.
  • dppf PdCl2
  • KOAc KOAc
  • DMSO DMSO
  • the installation of amines or N linked heteroaryls could be carried out by heating 1 to 40 equivalents of the appropriate amine and an equivalent of the appropriate diazaindole chloride, bromide or iodide with copper bronze (from 0.1 to 10 equivalents (preferably about 2 equivalents) and from 1 to 10 equivalents of finely pulverized potassium hydroxide (preferably about 2 equivalents). Temperatures of 120° to 200° might be employed with 140-160° generally preferred. For volatile starting materials a sealed reactor may be employed. The reaction would most often be applicable when the halogen being displaced is at the 7-position of a diazaindole but the method could work when the halogen is at a different position (4-7 position possible). As shown above the reaction could be employed on diazaindoles unsubstituted at position 3 or intermediates which contain the dicarbonyl or the intact dicarbonyl piperazine urea or thioureas contained in compounds of formula I.
  • a possible preparation of a key aldehyde intermediate, 43, using a procedure adapted from the method of Gilmore et. Al. Synlett 1992, 79-80 is shown in Scheme 16 above.
  • the aldehyde substituent is shown only at one position for the sake of clarity, and should not be considered as a limitation of the methodology.
  • the bromide or iodide intermediate would be converted into an aldehyde intermediate, 43, by metal-halogen exchange and subsequent reaction with dimethylformamide in an appropriate aprotic solvent.
  • Typical bases used could include, but would not be limited to, alkyl lithium bases such as n-butyl lithium, sec butyl lithium or tert butyl lithium or a metal such as lithium metal.
  • a preferred aprotic solvent is THF.
  • the transmetallation would be initiated at ⁇ 78° C. and allowed to react with dimethylformamide (allowing the reaction to warm may be required to enable complete reaction) to provide an aldehyde which is elaborated to compounds of Formula I.
  • Other methods for introduction of an aldehyde group to form intermediates of formula 43 include transition metal catalyzed carbonylation reactions of suitable bromo, trifluoromethane sulfonyl, or stannyl diazaindoles.
  • the pieces HW—R 18 can be prepared by a number of different methods.
  • One useful way is by reacting a mono protected piperazine with a heteroaryl chloride, bromide, iodide, or triflate. This reaction is typically carried out at elevated temperature (50 to 250 degrees celsius) in a solvent such as ethylene glycol, DME, dioxane, NMP, or DMF.
  • a tertiary amine base such as triethyl amide or diisopropyl ethyl amine is typically employed and usually 2 to 4 equivalents are employed. At least 2 equivalents are used if a salt of HW R 18 is utilized.
  • the piperazine is typically monoprotected with a BOC group since this material is commercially available. Removal of the Boc group is typically done using HCl (typically 1 to 6N) in dioxane to provide the HCl salt. TFA may also be used to generate the TFA salt.
  • the conditions for coupling heterocycles using copper catalysis discussed earlier in Scheme 12 may be used to couple W to R 18 via displacement of X in X—R 18 .
  • Palladium catalysis in the presence of a bidentate catalyst via the procedures of Buchwald or the use of a ferrocenyl catalyst via the methods of Hartwig could be used to couple the piperazine to the heteroaryl (R 18 ).
  • Scheme 53 describes how a protected piperazine can be coupled to Q-COOH via standard methodology. Conditions for removal of the amine protecting group which could be tBoc or other groups is protecting group specific. As shown in Scheme 53 where tBoc is the preferred protecting group used to exemplify the strategy, standard conditions for removal such as TFA in dichloromethane or alternatively aqueous HCl can provide the free amine. The free amine is coupled to heteraromatic R 18 using the conditions described in Scheme 52 for step F′′′′.
  • Scheme D1 describes a possible method for preparing the compounds described by H—W where Y is as defined in the description and claims of the invention. Typically, this methodology will work best when D is a group which lowers the PKA of the hydrogens on the adjacecent methylene moiety.
  • D is a group which lowers the PKA of the hydrogens on the adjacecent methylene moiety.
  • cyano, sulfonyl, amido and the like as specified in the claim.
  • A preferably could be aryl or heteroaryl moieties as described in claim 1 .
  • A could also be other groups described in claim 1 .
  • Alkoxide bases of C1 to C4 alcohols can be utilzed but other bases such as lithium, sodium, or potassium dialkyl amides or the corresponding bistrimethylsilyl amides could also be utilized.
  • an organometallic reagent to a ketone can provide an intermediate tertiary alkoxide which undergoes protonation and acid catalyzed elimination to form the desired double bond.
  • a number of organo metallic reagents could suffice as shown but an extra equivalent (at least two total) could be needed to compensate for deprotection of the amine nitrogen in many cases.
  • phosphinate or phosphine oxide based reagents could be used with similar bases or with sodium or postassium methoxide or ethoxide in the corresponding alcohol solvents.
  • a chloride, bromide, iodide, triflate, or phosphonate undergo coupling reactions with a boronate (Suzuki type reactions).
  • Stannanes and boronates are prepared via standard literature procedures or as described in the experimental section of this application.
  • the vinyl bromides, chlorides, triflates, or phosphonates may undergo metal mediated coupling to provide compounds of formula W—H. Stille or Suzuki couplings are particularly useful. A detailed discussion of the references and best conditions for these kinds of metal mediated coupling is described later in this application where the discussion is combined with a description of how these types of reactions may aslo be used to funtionalize diazaindoles.
  • the compounds Y—H could potentially be prepared via olefin metathesis using highly active Rhodium catalysts.
  • the methylene starting material can be prepared via simple Wittig methylenation of the precursor ketone which is prepared via literature methods.
  • the olefin metathesis is preferably carried out using 1% of the imadazoylidene ruthenium benzylidene catalyst described in the following reference. The reaction is carried out starting at low temperatures ( ⁇ 40°) or similar. Starting methylene material is mixed with excess olefin (5 to 100 equivalents) and the reaction is warmed to 40° C. Synthesis of Symmetrical Trisubstituted Olefins by Cross Metathesis. Chatterjee, Arnab K.; Sanders, Daniel P.; Grubbs, Robert H. Organic Letters ACS ASAP.
  • Scheme K1 shows a sequence in which a piperidone is coverted to a monofuntionalized olefin via Wittig olefination. Bromination and dehydrobromination provides a versatile vinyl bromide intermediate. This intermediate is coupled to the QC(O)C(O)OH acid with BOPCl to provide a compound of formula I. This intermediate is then functionalized using palladium mediated couplings to either boronates or stannanes. Conditions for these couplings are described in this application.
  • Scheme L1 shows specific examples of general Scheme K1 which are some of those described in the experimental section.
  • Scheme M1 shows how a protected vinyl bromide can be converted to a carboxylic acid via lithium bromide exchange and reaction with carbon dioxide.
  • carboxylic acids are excellent precursors to many heterocyles or amides.
  • the rest of Scheme M1 shows conversion to funtionalized oxadiazoles.
  • Other chemistry described in this application depicts other methods for converting acids to groups of other compounds of the invention.
  • Scheme N1 depicts a more specific example of Scheme M1.
  • Scheme P depicts methods for functionalizing the vinyl bromide to install groups D (or A). Either a modified Stille coupling or a zinc mediated coupling are depicted. Details of these tranformations are discussed later in the section on metal couplings.
  • Scheme Q depicts some specific examples of Scheme P.
  • Scheme R depicts methods for functionalizing the vinyl bromide to install groups D (or A). Either a modified Stille coupling, zinc mediated coupling, or a Suzuki boronic acid coupling are depicted. A method for converting the vinyl bromide to vinyl idodide is shown. If the vinyl bromide fails to undergo efficient reaction, the more reactive iodide can be prepared as a better partner. Details of these tranformations are discussed later in the section on metal couplings.
  • Scheme S provides specific examples of Scheme R.
  • Scheme T shows methods for converting the vinyl bromide into more funtionalized groups D (or A).
  • a key aldehyde intermediate is generated from the vinyl bromide and can be used to generate heteroaryls such as the oxazole via reaction with Tosmic.
  • Scheme U shows how a hydrazide (gnerated from the acid) can be used to prepare oxadiazoles with diffferent substituents.
  • Scheme V provides more specific examples of Scheme U.
  • Scheme W shows some other methods for installing D (or A).
  • Scheme X shows a particular example where a functionalized heteroaryl or in this case aryl are coupled and then further functionalization can occurr (in this case redcution of an ester to an alcohol).
  • Scheme Y provides more specific examples of Scheme X.
  • Scheme 54a depict a general method suitable for the synthesis of many of the compounds of formula I.
  • a suitable protected piperazine derivative, PG-YH, of Formula VI (wherein PG is an appropriate amine protecting group) is acylated with an appropriate acylating agent, R 18 C(O)L, (wherein L is a suitable leaving group) to provide the protected acylated piperazine derivative of Formula V.
  • Compound V is then deprotected using standard methods to provide the acylated piperazine derivative of Formula IV.
  • the compound of Formula V when PG represents tertiary-butoxycarbonyl the compound of Formula V can be deprotected to provide a compound of Formula IV by treatment with a strong acid, such as neat cold trifluoroacetic acid or aqueous hydrochloric acid, in an appropriate solvent such as dichloromethane.
  • a strong acid such as neat cold trifluoroacetic acid or aqueous hydrochloric acid
  • an appropriate solvent such as dichloromethane.
  • PG represents benzyl the deprotection may be effected by hydrogenation.
  • Examples containing substituted piperazines are prepared using the general procedures outlined in Schemes 55-38.
  • Substituted piperazines are either commercially available from Aldrich, Co. or prepared according to literature procedures (Behun et al, Ref. 88(a), Scheme 31, eq. 01).
  • Hydrogenation of alkyl substituted pyrazines under 40 to 50 psi pressure in EtOH afforded substituted piperazines.
  • the substituent was an ester or amide
  • the pyrazine systems could be partially reduced to the tetrahydropyrazine (Rossen et al, Ref. 88(b), Scheme 55, eq. 02).
  • the carbonyl substituted piperazines could be obtained under the same conditions described above by using commercially available dibenzyl piperazines (Scheme 55, eq. 03).
  • Piperazine intermediates could be prepared using standard chemistry as shown in Scheme 64.
  • Steps a16, a17, and a18 encompasses reactions and conditions for 1 0 , 2 0 and 3 0 amide bond formation as shown in Schemes 65 which provide compounds such as those of Formula 73.
  • Compounds of formula 73 represent intermediates for the preparation of Compounds I or compounds I depending on the identity of T and Y.
  • the reaction conditions for the formation of amide bonds encompass any reagents that generate a reactive intermediate for activation of the carboxylic acid to amide formation, for example (but not limited to), acyl halide, from carbodiimide, acyl iminium salt, symmetrical anhydrides, mixed anhydrides (including phosphonic/phosphinic mixed anhydrides), active esters (including silyl ester, methyl ester and thioester), acyl carbonate, acyl azide, acyl sulfonate and acyloxy N-phosphonium salt.
  • the reaction of the diazaindole carboxylic acids with amines to form amides may be mediated by standard amide bond forming conditions described in the art.
  • amide bond formation Some examples for amide bond formation are listed in references 41-53 but this list is not limiting.
  • carboxylic acid to amine coupling reagents which are applicable are EDC, Diisopropylcarbodiimide or other carbodiimides, PyBop (benzotriazolyloxytris(dimethylamino) phosphonium hexafluorophosphate), 2-(1H-benzotriazole-1-yl)-1,1,3,3-tetramethyl uronium hexafluorophosphate (HBTU).
  • a particularly useful method for azaindole 7-carboxylic acid to amide reactions is the use of carbonyl imidazole as the coupling reagent as described in reference 53.
  • the temperature of this reaction may be lower than in the cited reference, from 80° C. (or possibly lower) to 150° C. or higher.
  • An example of more specific conditions which are likely to be successful are depicted in Scheme 66.
  • a mixture of an acid intermediate, such as 74, (0.047 mmol) and 8.5 mg (0.052 mmol) of 1,1-carbonyldiimidazole in anhydrous THF (2 mL) could be heated to reflux under nitrogen. After 2.5 h, 0.052 mmol of amine could be added and heating continued. After an additional period of 3 ⁇ 20 h at reflux, the reaction mixture could be cooled and concentrated in vacuo. The residue could be purified by chromatography on silica gel to provide a compound of Formula I.
  • the carboxylic acid could be converted to an acid chloride using reagents such as thionyl chloride (neat or in an inert solvent) or oxalyl chloride in a solvent such as benzene, toluene, THF, or CH 2 Cl 2 .
  • reagents such as thionyl chloride (neat or in an inert solvent) or oxalyl chloride in a solvent such as benzene, toluene, THF, or CH 2 Cl 2 .
  • the amides could alternatively, be formed by reaction of the acid chloride with an excess of ammonia, primary, or secondary amine in an inert solvent such as benzene, toluene, THF, or CH 2 Cl 2 or with stoichiometric amounts of amines in the presence of a tertiary amine such as triethylamine or a base such as pyridine or 2,6-lutidine.
  • the acid chloride could be reacted with an amine under basic conditions (usually sodium or potassium hydroxide) in solvent mixtures containing water and possibly a miscible co solvent such as dioxane or THF.
  • Scheme 25B depicts a typical preparation of an acid chloride and derivatization to an amide of Formula I.
  • the carboxylic acid could be converted to an ester preferably a methyl or ethyl ester and then reacted with an amine.
  • the ester could be formed by reaction with diazomethane or alternatively trimethylsilyl diazomethane using standard conditions which are well known in the art. References and procedures for using these or other ester forming reactions can be found in reference 52 or 54.
  • Scheme 67 shows possible synthetic transformations on a chloro diazazaindole.
  • Step F-1 of Scheme 31 could be carried out according to the following procedures: Yamaguchi, S.; Yoshida, M.; Miyajima, I.; Araki, T.; Hirai, Y.; J. Heterocycl. Chem. 1995, 32(5), 1517-1519 in which they use 1 eq of Chloride, 1.9 eq Cu(I)CN, in dry DMF and reflux for 48 h.
  • the concentration of chloride in DMF is preferably 0.094 mmol per mL of solvent. Reaction times of 1-48 h may be approrpiate depending on substrate and reaction temperatures between 80° C. and reflux (156° C.) may be employed.
  • step F-1 An alternate procedure for carrying out step F-1 as described in the experimental section for Example 12 occurrs via reaction of the chloride intermediate with potassium cyanide (0.9 to 5 eqs, preferably 1.5 eqs) in a solvent such as DMF in the presence of catalytic sodium 4-toluene sulfinate at an elevate temperature such as 100° C. for 3 h.
  • Reaction temperature may vary between 50 and 200° C. depending on substrate and reaction time from 30 min to 48 h. Reactions may be conducted in a sealed tube to minimize escape of volatiles if necessary.
  • Transformation step I the hydrolysis of the nitrile to the acid may be carried out using acidic conditions such as MeOH and HCl at ambient temp followed by heating the intermediate imididate in the Methanol which provides the intermediate methyl ester which can then be hydrolyzed using potassium carbonate MeOH or LiOH or KOH in Methanol. This method is preferred to produce intact Compounds I. Alternatively, KOH/in ethanol or methanol may be utilized to achieve this transformation in step I. Other methods for this tranformations are well known in the literature or in the references incorporated in the experimental section.
  • stirring with MeOH and HCl at room temperature followed by a hydrolytic workup water and theyl acetate or dichloromethane, may produce the same product.
  • Step J the amide coupling is carried out as described above in the discussions for Scheme 65 and 66.
  • dicarbonyl intermediate 10 50 mg, 0.13 mmol
  • 4-methylpyrazole 31 mg, 0.38 mmol
  • ethanol 1 mL
  • the reaction was diluted with MeOH (2 mL), filtered and the filtrate was purified by preparative HPLC to yield 12 (34 mg, 0.08 mmol, 61%) as a white solid.
  • dicarbonyl intermediate 10 50 mg, 0.13 mmol
  • pyrazole 26 mg, 0.38 mmol
  • ethanol 1 mL
  • the reaction was diluted with MeOH (3 mL), filtered and the filtrate was purified by preparative HPLC to yield 14 (13 mg, 0.03 mmol, 24%) as a white solid.
  • dicarbonyl intermediate 10 50 mg, 0.13 mmol
  • 3-methyl-1,2,4-triazole 52 mg, 0.75 mmol
  • copper powder 16 mg, 0.25 mmol
  • the reaction was diluted with MeOH (3 mL), filtered through celite and the filtrate was purified by preparative HPLC to yield 16 (3 mg, 0.007 mmol, 5%) as a yellow solid.
  • dicarbonyl intermediate 10 600 mg, 1.5 mmol
  • tributyl(1-ethoxyvinyl)stannane 1.5 mL 4.5 mmol
  • tetrakis(triphenylphosphine)palladium(O) 350 mg, 0.30 mmol
  • 1,4-dioxane 15 mL
  • the reaction mixture was divided and 25% v/v was concentrated diluted with MeOH/CH 2 Cl 2 (2:1, 1.5 mL) and IN aqueous HCl (0.5 mL).
  • the reaction was stirred overnight, neutralized with 1N aqueous NaOH (0.5 mL) and concentrated.
  • dicarbonyl intermediate 10 40 mg, 0.10 mmol
  • potassium cyanide 10 mg, 0.15 mmol
  • sodium 4-toluenesulfinate 20 mg, 0.11 mmol
  • DMF 0.8 mL
  • the crude reaction mixture was partitioned between aqueous 5% Na 2 CO 3 (0.5 mL) and EtOAc (4 mL).
  • the organic layer was washed with aqueous 5% Na 2 CO 3 (0.7 mL), concentrated, dissolved into MeOH/DMSO (3:1, 2 mL) and purified by preparative HPLC to yield 20 (10 mg, 0.03 mmol, 26%) as a white solid.
  • dicarbonyl intermediate 10 50 mg, 0.13 mmol
  • N,N-dimethyl-1H-pyrazole-3-carboxamide 53 mg, 0.38 mmol
  • copper(0) 10 mg
  • 1,4-dioxane 0.8 mL
  • the reaction mixture was concentrated, diluted with MeOH and DMSO, filtered and purified by preparative HPLC to yield 21 (2.3 mg, 0.005 mmol, 4%) as a yellow solid.
  • Carboxylic acid 17 (34 mg, 0.73 mmol), N-methylpiperazine (15 mg, 0.15 mmol), N,N-diisopropylethylamine (0.13 mL, 94 mg, 0.73 mmol) and bis(2-oxo-3-oaxzolidinyl)phosphinic chloride (41 mg, 0.16 mmol) were dissolved into CH 2 Cl 2 (0.5 mL) and stirred for 20 h.
  • dicarbonyl intermediate 10 50 mg, 0.13 mmol
  • 3-phenyl-1H-pyrazole 80 mg, 0.56 mmol
  • 1,4-dioxane 2.5 mL
  • the reaction was concentrated and triturated with MeOH (3 mL).
  • the resulting solids were washed with MeOH and with Et 2 O to yield 23 (33 mg, 0.07 mmol, 50%) as a tan solid.
  • dicarbonyl intermediate 10 50 mg, 0.13 mmol
  • 1,2,4-triazole 26 mg, 0.44 mmol
  • copper(0) 8 mg, 0.13 mmol
  • K 2 CO 3 23 mg, 0.17 mmol
  • 1,4-dioxane 0.8 mL
  • the reaction mixture was diluted with MeOH/CH 2 Cl 2 (1:1, 2 mL), filtered, concentrated, dissolved into MeOH/DMSO (5:4, 1.8 mL) and purified by preparative HPLC.
  • the resulting yellow solid was triturated with MeOH to yield 28 (15 mg, 0.03 mmol, 28%) as a light yellow solid.
  • dicarbonyl intermediate 10 60 mg, 0.15 mmol
  • 1,2,3-triazole 95 mg, 1.4 mmol
  • 1,4-dioxane 3 mL
  • dicarbonyl intermediate 10 50 mg, 0.13 mmol
  • 3-(tributylstannyl)pyrazole 188 mg, 0.52 mmol
  • tetrakis(triphenylphosphine)-palladium(0) 72 mg, 0.06 mmol
  • 1,4-dioxane 0.8 mL
  • the reaction mixture was diluted with MeOH/CH 2 Cl 2 (1:1, 2 mL) and filtered to collect solids. The solids were dissolved into DMSO and purified by preparative HPLC to yield 30 (28 mg, 0.07 mmol, 52%) as a white solid.
  • dicarbonyl intermediate 10 50 mg, 0.13 mmol
  • 3-methyl-5-(tributylstannyl)isoxazole 141 mg, 0.38 mmol
  • tetrakis(triphenylphosphine)-palladium(0) 30 mg, 0.03 mmol
  • 1,4-dioxane 1 mL
  • dicarbonyl intermediate 10 50 mg, 0.13 mmol
  • 2-(tributylstannyl)pyridine 140 mg, 0.38 mmol
  • tetrakis(triphenylphosphine)-palladium(0) 30 mg, 0.03 mmol
  • 1,4-dioxane 0.8 mL
  • dicarbonyl intermediate 10 50 mg, 0.13 mmol
  • 3-(tributylstannyl)pyridine 160 mg, 0.43 mmol
  • tetrakis(triphenylphosphine)-palladium(0) 40 mg, 0.03 mmol
  • 1,4-dioxane 0.8 mL
  • the reaction mixture was concentrated to dryness and partitioned between EtOAc (5 mL) and saturated aqueous NaHCO 3 with cesium fluoride.
  • the biphasic suspension was filtered, separated and the aqueous layer was concentrated to dryness.
  • dicarbonyl intermediate 10 50 mg, 0.13 mmol
  • 4-(tributylstannyl)pyridine 140 mg, 0.38 mmol
  • tetrakis(triphenylphosphine)-palladium(0) 30 mg, 0.03 mmol
  • 1,4-dioxane 0.8 mL
  • dicarbonyl intermediate 10 50 mg, 0.13 mmol
  • 2-(tributylstannyl)pyrazine 160 mg, 0.43 mmol
  • tetrakis(triphenylphosphine)-palladium(0) 30 mg, 0.03 mmol
  • 1,4-dioxane 0.8 mL
  • the reaction mixture was concentrated to dryness, diluted with MeOH (2.5 mL) and DMSO (0.5 mL), filtered and purified by preparative HPLC.
  • dicarbonyl intermediate 10 50 mg, 0.13 mmol
  • 5-(tributylstannyl)pyrimidine 160 mg, 0.43 mmol
  • tetrakis(triphenylphosphine)-palladium(0) 30 mg, 0.03 mmol
  • 1,4-dioxane 0.8 mL
  • dicarbonyl intermediate 10 50 mg, 0.13 mmol
  • 4-(tributylstannyl)pyridazine 160 mg, 0.43 mmol
  • tetrakis(triphenylphosphine)-palladium(0) 30 mg, 0.03 mmol
  • 1,4-dioxane 0.8 mL
  • the reaction mixture was concentrated, diluted with MeOH, filtered and purified by preparative HPLC.
  • the resulting black oil was repurified by preparative HPLC and the resulting yellow solid was triturated with acetone to yield 37 (18.7 mg, 0.04 mmol, 33%) as an off-white solid.
  • reaction mixture was diluted with EtOAc (10 mL) and saturated aqueous NH 4 Cl (10 mL) and filtered. The layers were separated and the aqueous layer extracted with EtOAc (25 mL). The combined organic layers were concentrated and purified by preparative HPLC to yield 39 (102 mg, 0.24 mmol, 45%) as a bright yellow solid.
  • dicarbonyl intermediate 39 40 mg, 0.10 mmol
  • 2-(tributylstannyl)pyrazine 105 mg, 0.28 mmol
  • tetrakis(triphenylphosphine)-palladium(0) 30 mg, 0.03 mmol
  • 1,4-dioxane 0.8 mL
  • the reaction mixture was diluted with MeOH (1 mL) and DMSO (1 mL), filtered through celite and purified by preparative HPLC to yield 40 (12 mg, 0.03 mmol, 27%) as a yellow solid.
  • dicarbonyl intermediate 39 41 mg, 0.10 mmol
  • pyrazole 26 mg, 0.38 mmol
  • copper(0) 10 mg
  • 1,4-dioxane 0.8 mL
  • the reaction mixture was diluted with MeOH (1 mL) and DMSO (1 mL), filtered through celite and purified by preparative HPLC to yield 41 (11 mg, 0.02 mmol, 23%) as a light yellow solid.
  • reaction mixture was diluted with EtOAc (15 mL) and saturated aqueous NH 4 Cl (10 mL). The layers were separated and the aqueous layer extracted with EtOAc (2 ⁇ 20 mL). The combined organic layers were concentrated, the residue was purified by preparative HPLC and the resulting yellow solid was triturated with MeOH to yield 43 (18.6 mg, 0.04 mmol, 10%) as a white solid.
  • dicarbonyl intermediate 33 (30 mg, 0.074 mmol), 5-(tributylstannyl)pyrimidine (82 mg, 0.22 mmol), tetrakis(triphenylphosphine)-palladium(0) (20 mg, 0.02 mmol) and 1,4-dioxane (0.8 mL) were combined and heated at 130° C. with microwaves for 2 h.
  • the reaction mixture was diluted with MeOH/DMSO, filtered and purified by preparative HPLC to yield 44 (8 mg, 0.02 mmol, 30%) as a yellow solid.
  • reaction mixture was diluted with EtOAc (30 mL) and brine (25 mL) and filtered. The layers were separated and the organic layer concentrated. The residue was triturated with Et 2 O to yield 46 (340 mg, 0.76 mmol, 27%) as an orange/yellow solid.
  • dicarbonyl intermediate 46 (30 mg, 0.07 mmol), 1,2,4-triazole (28 mg, 0.41 mmol), copper(0) (8 mg, 0.13 mmol), K 2 CO 3 (20 mg, 0.14 mmol) and 1,4-dioxane (1 mL) were combined and heated at 140° C. with microwaves for 2 h.
  • the reaction mixture was diluted with MeOH/DMSO (2:3, 1 mL) and purified by preparative HPLC to yield 47 (4 mg, 0.008 mmol, 12%) as a yellow solid.
  • dicarbonyl intermediate 46 (30 mg, 0.07 mmol), pyrazole (34 mg, 0.5 mmol) and 1,4-dioxane (0.7 mL) were combined and heated at 140° C. with microwaves for 50 min.
  • the reaction mixture was diluted with MeOH/DMSO (1:1, 1.2 mL) and purified by preparative HPLC to yield 48 (10 mg, 0.02 mmol, 32%) as a yellow solid.
  • Oxalyl chloride (4.5 mL, 51 mmol) was added to a solution of diazaindole carboxylic acid 50 (1.08 g, 4.7 mmol) in CH 2 Cl 2 (8 mL) and the reaction mixture was stirred 14 h. Catalytic DMF (3 drops) was added to the reaction mixture and after 3 h the reaction was quenched with MeOH. The crude reaction mixture was concentrated to dryness to yield 51 (1.21 g, 49 mmol, 96%) as a tan solid with was used without further purification.
  • reaction mixture was diluted with H 2 O (5 mL) and saturated aqueous NH 4 Cl (5 mL) and extracted with EtOAc (3 ⁇ 20 mL). The layers were separated and the aqueous layer extracted with EtOAc (2 ⁇ 20 mL). The combined organic layers were concentrated, the residue was purified by preparative HPLC to yield 53 (7.3 mg, 0.02 mmol, 4%) as a yellow solid.
  • reaction mixture was diluted with saturated aqueous NH 4 Cl (5 mL) and extracted with EtOAc (3 ⁇ 20 mL). The combined organic layers were concentrated, the residue was purified by preparative HPLC to yield 56 (33 mg, 0.07 mmol, 18%) as a yellow solid.
  • dicarbonyl intermediate 33 31 mg, 0.077 mmol
  • pyrazole 20 mg, 0.29 mmol
  • 1,4-dioxane 0.8 mL
  • the reaction mixture was diluted with MeOH/DMSO (2:1, 1.5 mL), filtered and purified by preparative HPLC to yield 58 (10 mg, 0.024 mmol, 31%) as an orange solid.
  • dicarbonyl intermediate 33 33 mg, 0.081 mmol
  • 3-(tributylstannyl)-1H-pyrazole 73 mg, 0.20 mmol
  • tetrakis(triphenylphosphine)-palladium(0) 20 mg, 0.02 mmol
  • 1,4-dioxane 0.8 mL
  • the reaction mixture was diluted with MeOH/DMSO (2:1, 1.5 mL), filtered and purified by preparative HPLC to yield 59 (3.4 mg, 0.008 mmol, 10%) as a yellow solid.
  • Cytoxicity assays were conducted with the same HeLa using methodology well known in the art. This method has been described in the literature (S Weislow, R Kiser, D L Fine, J Bader, R H Shoemaker and M R Boyd: New soluble-formazan assay for HIV-1 cytopathic effects: application to high-flux screening of synthetic and natural products for AIDS-antiviral activity. Journal of the National Cancer Institute. 81(8):577-586, 1989.
  • MTT dye reduction assay
  • Example 10 Example 11 19 H A Example 12 20 H A Example 13 21 H A Example 14 22 H A Example 15 23 H A Example 16 24 H A Example 17 25 H A Example 18 26 H A Example 19 27 H A Example 20 28 H A Example 21 29 H A Example 22 30 H A Example 22 30 H A Example 24 32 H A Example 25 33 H A Example 26 34 H A Example 27 35 H A Example 28 36 H A Example 29 37 H A Example 30 39 H Cl A Example 31 40 H A Example 32 41 H A Example 33 43 H Cl A Example 34 44 H A Example 35 46 H Cl A Example 36 47 H A Example 37 48 H A Example 38 53 H A Example 39 54 H A Example 40 56 H A Example 41 57 H A Example 42 58 H A Example 43 59 H A
  • Table 4 shows other compounds of the invention which could be prepared by the methodology described herein and which are expected to have antiviral activity.
  • TABLE 4 EC 50 Group Example Cmpd. from No. No. R3 R5 Y Table 1
  • the compounds of the present invention may be administered orally, parenterally (including subcutaneous injections, intravenous, intramuscular, intrasternal injection or infusion techniques), by inhalation spray, or rectally, in dosage unit formulations containing conventional non-toxic pharmaceutically-acceptable carriers, adjuvants and vehicles.
  • a method of treating and a pharmaceutical composition for treating viral infections such as HIV infection and AIDS involves administering to a patient in need of such treatment a pharmaceutical composition comprising a pharmaceutical carrier and a therapeutically-effective amount of a compound of the present invention.
  • the pharmaceutical composition may be in the form of orally-administrable suspensions or tablets; nasal sprays, sterile injectable preparations, for example, as sterile injectable aqueous or oleagenous suspensions or suppositories.
  • these compositions When administered orally as a suspension, these compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may contain microcrystalline cellulose for imparting bulk, alginic acid or sodium alginate as a suspending agent, methylcellulose as a viscosity enhancer, and sweetners/flavoring agents known in the art.
  • these compositions may contain microcrystalline cellulose, dicalcium phosphate, starch, magnesium stearate and lactose and/or other excipients, binders, extenders, disintegrants, diluents and lubricants known in the art.
  • the injectable solutions or suspensions may be formulated according to known art, using suitable non-toxic, parenterally-acceptable diluents or solvents, such as mannitol, 1,3-butanediol, water, Ringer's solution or isotonic sodium chloride solution, or suitable dispersing or wetting and suspending agents, such as sterile, bland, fixed oils, including synthetic mono- or diglycerides, and fatty acids, including oleic acid.
  • suitable non-toxic, parenterally-acceptable diluents or solvents such as mannitol, 1,3-butanediol, water, Ringer's solution or isotonic sodium chloride solution, or suitable dispersing or wetting and suspending agents, such as sterile, bland, fixed oils, including synthetic mono- or diglycerides, and fatty acids, including oleic acid.
  • the compounds of this invention can be administered orally to humans in a dosage range of 1 to 100 mg/kg body weight in divided doses.
  • One preferred dosage range is 1 to 10 mg/kg body weight orally in divided doses.
  • Another preferred dosage range is 1 to 20 mg/kg body weight orally in divided doses.
  • the specific dose level and frequency of dosage for any particular patient may be varied and will depend upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the host undergoing therapy.

Abstract

The invention comprises substituted diazaindole-dicarbonyl-piperazinyl derivatives of general Formula I
Figure US20050124623A1-20050609-C00001
wherein:
Q is selected from the group consisting of
Figure US20050124623A1-20050609-C00002
— may represent a bond; T is —C(O)— or —CH(CN)—; and
—Y— is selected from the group consisting of
Figure US20050124623A1-20050609-C00003
compositions thereof and their use for treating HIV infection.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application claims the benefit of U.S. Provisional Application Ser. No. 60/525,624 filed Nov. 26, 2003.
  • BACKGROUND OF THE INVENTION
  • 1. Field of the Invention
  • This invention provides compounds having drug and bio-affecting properties, their pharmaceutical compositions and method of use. In particular, the invention is concerned with new diazaindole derivatives that possess unique antiviral activity. More particularly, the present invention relates to compounds useful for the treatment of HIV and AIDS.
  • 2. Background Art
  • HIV-1 (human immunodeficiency virus-1) infection remains a major medical problem, with an estimated 42 million people infected worldwide at the end of 2002. The number of cases of HIV and AIDS (acquired immunodeficiency syndrome) has risen rapidly. In 2002, ˜5.0 million new infections were reported, and 3.1 million people died from AIDS. Currently available drugs for the treatment of HIV include ten nucleoside reverse transcriptase (RT) inhibitors or approved single pill combinations (zidovudine or AZT (or Retrovir®), didanosine (or Videx®), stavudine (or Zerit®), lamivudine (or 3TC or Epivir®), zalcitabine (or DDC or Hivid®), abacavir succinate (or Ziagen®), Tenofovir disoproxil fumarate salt (or Viread®), Combivir® (contains −3TC plus AZT), Trizivir® (contains abacavir, lamivudine, and zidovudine) and Emtriva® (emtricitabine); three non-nucleoside reverse transcriptase inhibitors: nevirapine (or Viramune®), delavirdine (or Rescriptor®) and efavirenz (or Sustiva®), nine peptidomimetic protease inhibitors or approved formulations: saquinavir, indinavir, ritonavir, nelfinavir, amprenavir, lopinavir, Kaletra® (lopinavir and Ritonavir), Atazanavir (Reyataz®), Fosamprenavir® and one fusion inhibitor which targets viral gp41 T-20 (FUZEON®). Each of these drugs can only transiently restrain viral replication if used alone. However, when used in combination, these drugs have a profound effect on viremia and disease progression. In fact, significant reductions in death rates among AIDS patients have been recently documented as a consequence of the widespread application of combination therapy. However, despite these impressive results, 30 to 50% of patients ultimately fail combination drug therapies. Insufficient drug potency, non-compliance, restricted tissue penetration and drug-specific limitations within certain cell types (e.g. most nucleoside analogs cannot be phosphorylated in resting cells) may account for the incomplete suppression of sensitive viruses. Furthermore, the high replication rate and rapid turnover of HIV-1 combined with the frequent incorporation of mutations, leads to the appearance of drug-resistant variants and treatment failures when sub-optimal drug concentrations are present (Larder and Kemp; Gulick; Kuritzkes; Morris-Jones et al; Schinazi et al; Vacca and Condra; Flexner; Berkhout and Ren et al; (Ref. 6-14)). Therefore, novel anti-HIV agents exhibiting distinct resistance patterns, and favorable pharmacokinetic as well as safety profiles are needed to provide more treatment options.
  • Currently marketed HIV-1 drugs are dominated by either nucleoside reverse transcriptase inhibitors or peptidomimetic protease inhibitors. Non-nucleoside reverse transcriptase inhibitors (NNRTIs) have recently gained an increasingly important role in the therapy of HIV infections (Pedersen & Pedersen, Ref 15). At least 30 different classes of NNRTI have been described in the literature (De Clercq, Ref. 16) and several NNRTIs have been evaluated in clinical trials. Dipyridodiazepinone (nevirapine), benzoxazinone (efavirenz) and bis(heteroaryl) piperazine derivatives (delavirdine) have been approved for clinical use. However, the major drawback to the development and application of NNRTIs is the propensity for rapid emergence of drug resistant strains, both in tissue cell culture and in treated individuals, particularly those subject to monotherapy. As a consequence, there is considerable interest in the identification of NNRTIs less prone to the development of resistance (Pedersen & Pedersen, Ref 15). A recent overview of non-nucleoside reverse transcriptase inhibitors: perspectives on novel therapeutic compounds and strategies for the treatment of HIV infection. has appeared (Buckheit, reference 99). A review covering both NRTI and NNRTIs has appeared (De Clercq, reference 100). An overview of the current state of the HIV drugs has been published (De Clercq, reference 101).
  • Several indole derivatives including indole-3-sulfones, piperazino indoles, pyrazino indoles, and 5H-indolo[3,2-b][1,5]benzothiazepine derivatives have been reported as HIV-1 reverse transciptase inhibitors (Greenlee et al, Ref. 1; Williams et al, Ref. 2; Romero et al, Ref. 3; Font et al, Ref. 17; Romero et al, Ref. 18; Young et al, Ref. 19; Genin et al, Ref. 20; Silvestri et al, Ref. 21). Indole 2-carboxamides have also been described as inhibitors of cell adhesion and HIV infection (Boschelli et al, U.S. Pat. No. 5,424,329, Ref. 4). 3-Substituted indole natural products (Semicochliodinol A and B, didemethylasterriquinone and isocochliodinol) were disclosed as inhibitors of HIV-1 protease (Fredenhagen et al, Ref. 22).
  • Structurally related aza-indole amide derivatives have been disclosed previously (Kato et al, Ref. 23(a); Levacher et al, Ref. 23(b); Dompe Spa, WO-09504742, Ref. 5(a); SmithKline Beecham PLC, WO-09611929, Ref. 5(b); Schering Corp., U.S. Pat. No. 05,023,265, Ref. 5(c)). However, these structures differ from those claimed herein in that they are monoaza-indole mono-amide rather than oxoacetamide derivatives, and there is no mention of the use of these compounds for treating viral infections, particularly HIV.
  • New drugs for the treatment of HIV are needed for the treatment of patients who become resistant to the currently approved drugs described above which target reverse transcriptase or the protease. One approach to obtaining these drugs is to find molecules which inhibit new and different targets of the virus. A general class of inhibitors which are under active study are HIV entry inhibitors. This general classification includes drugs aimed at several targets which include chemokine receptor (CCR5 or CXCR4) inhibitors, fusion inhibitors targeting viral gp41, and inhibitors which prevent attachment of the viral envelope, gp120, the its human cellular target CD4. A number of reviews or general papers on viral entry inhibitors have recently appeared and some selected references are:
    • Chemokine receptor antagonists as HIV entry inhibitors. Expert Opinion on Therapeutic Patents (2004), 14(2), 251-255.
    • Inhibitors of the entry of HIV into host cells. Meanwell, Nicholas A.; Kadow, John F. Current Opinion in Drug Discovery & Development (2003), 6(4), 451-461.
    • Virus entry as a target for anti-HIV intervention. Este, Jose A. Retrovirology Laboratory irsiCaixa, Hospital Universitari Germans Trias i Pujol, Universitat Autonoma de Barcelona, Badalona, Spain. Current Medicinal Chemistry (2003), 10(17), 1617-1632.
    • New antiretroviral agents. Rachline, A.; Joly, V. Service de Maladies Infectieuses et Tropicales A, Hopital Bichat-Claude Bernard, Paris, Fr. Antibiotiques (2003), 5(2), 77-82.
    • New antiretroviral drugs. Gulick, R. M. Cornell HIV Clinical Trials Unit, Division of International Medicine and Infectious Diseases, Weill Medical College of Cornell University, New York, N.Y., USA. Clinical Microbiology and Infection (2003), 9(3), 186-193.
    • Sensitivity of HIV-1 to entry inhibitors correlates with envelope/coreceptor affinity, receptor density, and fusion kinetics. Reeves, Jacqueline D.; Gallo, Stephen A.; Ahmad, Navid; Miamidian, John L.; Harvey, Phoebe E.; Sharron, Matthew; Pohlmann, Stefan; Sfakianos, Jeffrey N.; Derdeyn, Cynthia A.; Blumenthal, Robert; Hunter, Eric; Doms, Robert W. Department of Microbiology, University of Pennsylvania, Philadelphia, Pa., USA. Proceedings of the National Academy of Sciences of the United States of America (2002), 99(25), 16249-16254. CODEN: PNASA6 ISSN: 0027-8424.
    • Opportunities and challenges in targeting HIV entry. Biscone, Mark J.; Pierson, Theodore C.; Doms, Robert W. Department of Microbiology, University of Pennsylvania, Philadelphia, Pa., USA. Current Opinion in Pharmacology (2002), 2(5), 529-533.
    • HIV entry inhibitors in clinical development. O'Hara, Bryan M.; Olson, William C. Progenics Pharmaceuticals, Inc., Tarrytown, N.Y., USA. Current Opinion in Pharmacology (2002), 2(5), 523-528.
    • Resistance mutation in HIV entry inhibitors. Hanna, Sheri L.; Yang, Chunfu; Owen, Sherry M.; Lal, Renu B. HIV Immunology and Diagnostics Branch, Division of AIDS, STD, Atlanta, Ga., USA. AIDS (London, United Kingdom) (2002), 16(12), 1603-1608.
    • HIV entry: are all receptors created equal? Goldsmith, Mark A.; Doms, Robert W. Genencor International, Inc., Palo Alto, Calif., USA. Nature Immunology (2002), 3(8), 709-710. CODEN: NIAMCZ ISSN: 1529-2908.
    • Peptide and non-peptide HIV fusion inhibitors. Jiang, Shibo; Zhao, Qian; Debnath, Asim K. The New York Blood Center, Lindsley F. Kimball Research Institute, New York, N.Y., USA. Current Pharmaceutical Design (2002), 8(8), 563-580.
    • A series of recent publications and disclosures characterize and describe a compound labelled as BMS-806, an initial member of a class of viral entry inhibitors which target viral gp-120 and prevent attachment of virus to host CD4.
    • A small molecule HIV-1 inhibitor that targets the HIV-1 envelope and inhibits CD4 receptor binding. Lin, Pin-Fang; Blair, Wade; Wang, Tao; Spicer, Timothy; Guo, Qi; Zhou, Nannan; Gong, Yi-Fei; Wang, H. -G. Heidi; Rose, Ronald; Yamanaka, Gregory; Robinson, Brett; Li, Chang-Ben; Fridell, Robert; Deminie, Carol; Demers, Gwendeline; Yang, Zheng; Zadjura, Lisa; Meanwell, Nicholas; Colonno, Richard. Proceedings of the National Academy of Sciences of the United States of America (2003), 100(19), 11013-11018.
    • Biochemical and genetic characterizations of a novel human immunodeficiency virus type 1 inhibitor that blocks gp120-CD4 interactions. Guo, Qi; Ho, Hsu-Tso; Dicker, Ira; Fan, Li; Zhou, Nannan; Friborg, Jacques; Wang, Tao; McAuliffe, Brian V.; Wang, Hwei-gene Heidi; Rose, Ronald E.; Fang, Hua; Scarnati, Helen T.; Langley, David R.; Meanwell, Nicholas A.; Abraham, Ralph; Colonno, Richard J.; Lin, Pin-fang. Journal of Virology (2003), 77(19), 10528-10536.
    • Method using small heterocyclic compounds for treating HIV infection by preventing interaction of CD4 and gp120. Ho, Hsu-Tso; Dalterio, Richard A.; Guo, Qi; Lin, Pin-Fang. PCT Int. Appl. (2003), WO 2003072028A2.
    • Discovery of 4-benzoyl-1-[(4-methoxy-1H-pyrrolo[2,3-b]pyridin-3-yl)oxoacetyl]-2-(R)-methylpiperazine (BMS-378806): A Novel HIV-1 Attachment Inhibitor That Interferes with CD4-gp120 Interactions. Wang, Tao; Zhang, Zhongxing; Wallace, Owen B.; Deshpande, Milind; Fang, Haiquan; Yang, Zheng; Zadjura, Lisa M.; Tweedie, Donald L.; Huang, Stella; Zhao, Fang; Ranadive, Sunanda; Robinson, Brett S.; Gong, Yi-Fei; Ricarrdi, Keith; Spicer, Timothy P.; Deminie, Carol; Rose, Ronald; Wang, Hwei-Gene Heidi; Blair, Wade S.; Shi, Pei-Yong; Lin, Pin-fang; Colonno, Richard J.; Meanwell, Nicholas A. Journal of Medicinal Chemistry (2003), 46(20), 4236-4239.
  • Indole, azaindole and other oxo amide containing derivatives have been disclosed in a number different PCT and issued U.S. patent applications (Reference 93-95, 106, 108,109, 110, 111,112, 113, and 114). None of these applications discloses diazindole compounds such as described in this invention. The extra nitrogen of the diazaindole class of molecules provides altered properties especially in combination with specific substituents that are advantageous and not available from the azaindoles. The diazaindoles are easier to access and thus offer the potential to provide patients with lower cost treatments. A series of PCT International Patent applications Bernd Nickel et.al. (reference 107a,b, and c) describes N-indolylglyoxamides for the treatment of cancer. Although some of these compounds contain N-heteroaryl or N-aryl piperazines, the substitution patterns at the other positions are outside the scope of this invention.
  • Structurally similar diazaindoles with a C-3 oxoacetyl group have also been previously disclosed (Hutchison et al, Ref 5(d); Resnyanskaya et al, Ref 24 (a); Cook et al, Ref 24(b)). However, these molecules differ from those claimed in that they do not contain the piperazine or piperidine moieties and there is no mention of the use of these molecules as antiviral agents, particularly against HIV.
  • Nothing in these references can be construed to disclose or suggest the novel compounds of this invention and their use to inhibit HIV infection.
  • References Cited
  • Patent Documents
    • 1. Greenlee, W. J.; Srinivasan, P. C. Indole reverse transcriptase inhibitors. U.S. Pat. No. 5,124,327.
    • 2. Williams, T. M.; Ciccarone, T. M.; Saari, W. S.; Wai, J. S.; Greenlee, W. J.; Balani, S. K.; Goldman, M. E.; Theohrides, A. D. Indoles as inhibitors of HIV reverse transcriptase. European Patent 530907.
    • 3. Romero, D. L.; Thomas, R. C.; Preparation of substituted indoles as anti-AIDS pharmaceuticals. PCT WO 93/01181.
    • 4. Boschelli, D. H.; Connor, D. T.; Unangst, P. C. Indole-2-carboxamides as inhibitors of cell adhesion. U.S. Pat. No. 5,424,329.
    • 5. (a) Mantovanini, M.; Melillo, G.; Daffonchio, L. Tropyl 7-azaindol-3-ylcarboxyamides as antitussive agents. PCT WO 95/04742 (Dompe Spa). (b) Cassidy, F.; Hughes, I.; Rahman, S.; Hunter, D. J. Bisheteroaryl-carbonyl and carboxamide derivatives with 5HT 2C/2B antagonists activity. PCT WO 96/11929. (c) Scherlock, M. H.; Tom, W. C. Substituted 1H-pyrrolopyridine-3-carboxamides. U.S. Pat. No. 5,023,265. (d) Hutchison, D. R.; Martinelli, M. J.; Wilson, T. M. Preparation or pyrrolo[2,3-d]pyrimidines as sPLA2 inhibitors PCT WO 00/00201.
      Other Publications
    • 6. Larder, B. A.; Kemp, S. D. Multiple mutations in the HIV-1 reverse transcriptase confer high-level resistance to zidovudine (AZT). Science, 1989, 246,1155-1158.
    • 7. Gulick, R. M. Current antiretroviral therapy: An overview. Quality of Life Research, 1997, 6, 471-474.
    • 8. Kuritzkes, D. R. HIV resistance to current therapies. Antiviral Therapy, 1997, 2 (Supplement 3), 61-67.
    • 9. Morris-Jones, S.; Moyle, G.; Easterbrook, P. J. Antiretroviral therapies in HIV-1 infection. Expert Opinion on Investigational Drugs, 1997, 6(8),1049-1061.
    • 10. Schinazi, R. F.; Larder, B. A.; Mellors, J. W. Mutations in retroviral genes associated with drug resistance. International Antiviral News, 1997, 5,129-142.
    • 11. Vacca, J. P.; Condra, J. H. Clinically effective HIV-1 protease inhibitors. Drug Discovery Today, 1997, 2, 261-272.
    • 12. Flexner, D. HIV-protease inhibitors. Drug Therapy, 1998, 338, 1281-1292.
    • 13. Berkhout, B. HIV-1 evolution under pressure of protease inhibitors: Climbing the stairs of viral fitness. J. Biomed. Sci., 1999, 6, 298-305.
    • 14. Ren, S.; Lien, E. J. Development of HIV protease inhibitors: A survey. Prog. Drug Res., 1998, 51, 1-31.
    • 15. Pedersen, O. S.; Pedersen, E. B. Non-nucleoside reverse transcriptase inhibitors: the NNRTI boom. Antiviral Chem. Chemother. 1999, 10, 285-314.
    • 16. (a) De Clercq, E. The role of non-nucleoside reverse transcriptase inhibitors (NNRTIs) in the therapy of HIV-1 infection. Antiviral Research, 1998, 38, 153-179. (b) De Clercq, E. Perspectives of non-nucleoside reverse transcriptase inhibitors (NNRTIs) in the therapy of HIV infection. IL. Farmaco, 1999, 54, 26-45.
    • 17. Font, M.; Monge, A.; Cuartero, A.; Elorriaga, A.; Martinez-Irujo, J. J.; Alberdi, E.; Santiago, E.; Prieto, I.; Lasarte, J. J.; Sarobe, P. and Borras, F. Indoles and pyrazino[4,5-b]indoles as nonnucleoside analog inhibitors of HIV-1 reverse transcriptase. Eur. J. Med. Chem., 1995, 30, 963-971.
    • 18. Romero, D. L.; Morge, R. A.; Genin, M. J.; Biles, C.; Busso, M,; Resnick, L.; Althaus, I. W.; Reusser, F.; Thomas, R. C and Tarpley, W. G. Bis(heteroaryl)piperazine (BHAP) reverse transcriptase inhibitors: structure-activity relationships of novel substituted indole analogues and the identification of 1-[(5-methanesulfonamido-1H-indol-2-yl)-carbonyl]-4-[3-[1-methylethyl)amino]-pyridinyl]piperazine momomethansulfonate (U-90152S), a second generation clinical candidate. J. Med. Chem., 1993, 36, 1505-1508.
    • 19. Young, S. D.; Amblard, M. C.; Britcher, S. F.; Grey, V. E.; Tran, L. O.; Lumma, W. C.; Huff, J. R.; Schleif, W. A.; Emini, E. E.; O'Brien, J. A.; Pettibone, D. J. 2-Heterocyclic indole-3-sulfones as inhibitors of HIV-reverse transcriptase. Bioorg. Med. Chem. Lett., 1995, 5, 491-496.
    • 20. Genin, M. J.; Poel, T. J.; Yagi, Y.; Biles, C.; Althaus, I.; Keiser, B. J.; Kopta, L. A.; Friis, J. M.; Reusser, F.; Adams, W. J.; Olmsted, R. A.; Voorman, R. L.; Thomas, R. C. and Romero, D. L. Synthesis and bioactivity of novel bis(heteroaryl)piperazine (BHAP) reverse transcriptase inhibitors: structure-activity relationships and increased metabolic stability of novel substituted pyridine analogs. J. Med. Chem., 1996, 39, 5267-5275.
    • 21. Silvestri, R.; Artico, M.; Bruno, B.; Massa, S.; Novellino, E.; Greco, G.; Marongiu, M. E.; Pani, A.; De Montis, A and La Colla, P. Synthesis and biological evaluation of 5H-indolo[3,2-b][1,5]benzothiazepine derivatives, designed as conformationally constrained analogues of the human immunodeficiency virus type 1 reverse transcriptase inhibitor L-737,126. Antiviral Chem. Chemother. 1998, 9, 139-148.
    • 22. Fredenhagen, A.; Petersen, F.; Tintelnot-Blomley, M.; Rosel, J.; Mett, H and Hug, P. J. Semicochliodinol A and B: Inhibitors of HIV-1 protease and EGF-R protein Tyrosine Kinase related to Asterriquinones produced by the fungus Chrysosporium nerdarium. Antibiotics, 1997, 50, 395-401.
    • 23. (a) Kato, M.; Ito, K.; Nishino, S.; Yamakuni, H.; Takasugi, H. New 5-HT3 (Serotonin-3) receptor antagonists. IV. Synthesis and structure-activity relationships of azabicycloalkaneacetamide derivatives. Chem. Pharm. Bull., 1995, 43, 1351-1357. (b) Levacher, V.; Benoit, R.; Duflos, J; Dupas, G.; Bourguignon, J.; Queguiner, G. Broadening the scope of NADH models by using chiral and non chiral pyrrolo[2,3-b]pyridine derivatives. Tetrahedron, 1991, 47, 429-440.
    • 24. (a) Resnyanskaya, E. V.; Tverdokhlebov, A. V.; Volovenko, Y. M.; Shishkin, O. V.; Zubatyuk, R. I. A simple synthesis of 1-acyl-3-aryl-3H-pyrrolo[2′,3′, :4,5]pyrimido[6,1-b]benzothiazol-6-ium-2-olates: Betainic derivatives of a novel heterocyclic system. Synthesis, 2002, 18, 2717-2724. (b) Cook, P. D.; Castle, R. N. Pyrrolopyridazines. 1. Synthesis and reactivity of [2,3-d]pyridazine 5-oxides. J. Het. Chem. 1973, 10(4), 551-557.
    • 25. Shadrina, L. P.; Dormidontov, Yu. P.; Ponomarev, V, G.; Lapkin, I. I. Reactions of organomagnesium derivatives of 7-aza- and benzoindoles with diethyl oxalate and the reactivity of ethoxalylindoles. Khim. Geterotsikl. Soedin., 1987, 1206-1209.
    • 26. Sycheva, T. V.; Rubtsov, N. M.; Sheinker, Yu. N.; Yakhontov, L. N. Some reactions of 5-cyano-6-chloro-7-azaindoles and lactam-lactim tautomerism in 5-cyano-6-hydroxy-7-azaindolines. Khim. Geterotsikl. Soedin., 1987, 100-106.
    • 27. (a) Desai, M.; Watthey, J. W. H.; Zuckerman, M. A convenient preparation of 1-aroylpiperazines. Org. Prep. Proced. Int., 1976, 8, 85-86. (b) Adamczyk, M.; Fino, J. R. Synthesis of procainamide metabolites. N-acetyl desethylprocainamide and desethylprocainamide. Org. Prep. Proced. Int. 1996, 28, 470-474. (c) Rossen, K.; Weissman, S. A.; Sager, J.; Reamer, R. A.; Askin, D.; Volante, R. P.; Reider, P. J. Asymmetric Hydrogenation of tetrahydropyrazines: Synthesis of (S)-piperazine 2-tert-butylcarboxamide, an intermediate in the preparation of the HIV protease inhibitor Indinavir. Tetrahedron Lett., 1995, 36, 6419-6422. (d) Wang, T.; Zhang, Z.; Meanwell, N. A. Benzoylation of Dianions: Preparation of mono-Benzoylated Symmetric Secondary Diamines. J. Org. Chem., 1999, 64, 7661-7662.
    • 28. Li, H.; Jiang, X.; Ye, Y.-H.; Fan, C.; Romoff, T.; Goodman, M. 3-(Diethoxyphosphoryloxy)-1,2,3-benzotriazin-4(3H)-one (DEPBT): A new coupling reagent with remarkable resistance to racemization. Organic Lett., 1999, 1, 91-93.
    • 29. Harada, N.; Kawaguchi, T.; Inoue, I.; Ohashi, M.; Oda, K.; Hashiyama, T.; Tsujihara, K. Synthesis and antitumor activity of quaternary salts of 2-(2′-oxoalkoxy)-9-hydroxyellipticines. Chem. Pharm. Bull., 1997, 45, 134-137.
    • 30. Schneller, S. W.; Luo, J.-K. Synthesis of 4-amino-1H-pyrrolo[2,3-b]pyridine (1,7-Dideazaadenine) and 1H-pyrrolo[2,3-b]pyridin-4-ol (1,7-Dideazahypoxanthine). J. Org. Chem., 1980, 45, 4045-4048.
    • 31. Shiotani, S.; Tanigochi, K. Furopyridines. XXII [1]. Elaboration of the C-substitutents alpha to the heteronitrogen atom of furo[2,3-b]-, -[3.2-b]-, -[2.3-c]- and -[3,2-c]pyridine. J. Het. Chem., 1997, 34, 901-907.
    • 32. Minakata, S.; Komatsu, M.; Ohshiro, Y. Regioselective functionalization of 1H-pyrrolo[2,3-b]pyridine via its N-oxide. Synthesis, 1992, 661-663.
    • 33. Klemm, L. H.; Hartling, R. Chemistry of thienopyridines. XXIV. Two transformations of thieno[2,3-b]pyridine 7-oxide (1). J. Het. Chem., 1976, 13, 1197-1200.
    • 34. Antonini, I.; Claudi, F.; Cristalli, G.; Franchetti, P.; Crifantini, M.; Martelli, S. Synthesis of 4-amino-1-β-D-ribofuranosyl-1H-pyrrolo[2,3-b]pyridine (1-Deazatubercidin) as a potential antitumor agent. J. Med. Chem., 1982, 25, 1258-1261.
    • 35. (a) Regnouf De Vains, J. B.; Papet, A. L.; Marsura, A. New symmetric and unsymmetric polyfunctionalized 2,2′-bipyridines. J. Het. Chem., 1994, 31, 1069-1077. (b) Miura, Y.; Yoshida, M.; Hamana, M. Synthesis of 2,3-fused quinolines from 3-substituted quinoline 1-oxides. Part II, Heterocycles, 1993, 36, 1005-1016. (c) Profft, V. E.; Rolle, W. Uber 4-merkaptoverbindungendes 2-methylpyridins. J. Prakt. Chem., 1960, 283 (11), 22-34.
    • 36. Nesi, R.; Giomi, D.; Turchi, S.; Tedeschi, P., Ponticelli, F. A new one step synthetic approach to the isoxazolo[4,5-b]pyridine system. Synth. Comm., 1992, 22, 2349-2355.
    • 37. (a) Walser, A.; Zenchoff, G.; Fryer, R. I. Quinazolines and 1,4-benzodiazepines. 75. 7-Hydroxyaminobenzodiazepines and derivatives. J. Med. Chem., 1976, 19, 1378-1381. (b) Barker, G.; Ellis, G. P. Benzopyrone. Part I. 6-Amino- and 6-hydroxy-2-subtituted chromones. J. Chem. Soc., 1970, 2230-2233.
    • 38. Ayyangar, N. R.; Lahoti, R J.; Daniel, T. An alternate synthesis of 3,4-diaminobenzophenone and mebendazole. Org. Prep. Proced. Int., 1991, 23, 627-631.
    • 39. Mahadevan, I.; Rasmussen, M. Ambident heterocyclic reactivity: The alkylation of pyrrolopyridines (azaindoles, diazaindenes). Tetrahedron, 1993, 49, 7337-7352.
    • 40. Chen, B. K.; Saksela, K.; Andino, R.; Baltimore, D. Distinct modes of human immunodeficiency type 1 proviral latency revealed by superinfection of nonproductively infected cell lines with recombinant luciferase-encoding viruses. J. Virol., 1994, 68, 654-660.
    • 41. Bodanszky, M.; Bodanszky, A. “The Practice of Peptide Synthesis” 2nd Ed., Springer-Verlag: Berlin Heidelberg, Germany, 1994.
    • 42. Albericio, F. et al. J. Org. Chem. 1998, 63, 9678.
    • 43. Knorr, R. et al. Tetrahedron Lett. 1989, 30, 1927.
    • 44. (a) Jaszay Z. M. et al. Synth. Commun., 1998 28, 2761 and references cited therein; (b) Bernasconi, S. et al. Synthesis, 1980, 385.
    • 45. (a) Jaszay Z. M. et al. Synthesis, 1989, 745 and references cited therein; (b) Nicolaou, K. C. et al. Angew. Chem. Int. Ed. 1999, 38, 1669.
    • 46. Ooi, T. et al. Synlett. 1999, 729.
    • 47. Ford, R. E. et al. J. Med. Chem. 1986, 29, 538.
    • 48. (a) Yeung, K.-S. et al. Bristol-Myers Squibb Unpublished Results. (b) Wang, W. et al. Tetrahedron Lett. 1999, 40, 2501.
    • 49. Brook, M. A. et al. Synthesis, 1983, 201.
    • 50. Yamazaki, N. et al. Tetrahedron Lett. 1972, 5047.
    • 51. Barry A. Bunin “The Combinatorial Index” 1998 Academic Press, San Diego/London pages 78-82.
    • 52. Richard C. Larock Comprehensive Organic Transormations 2nd Ed. 1999, John Wiley and Sons New York.
    • 53. M. D. Mullican et.al. J.Med. Chem. 1991, 34, 2186-2194.
    • 54. Protective groups in organic synthesis 3rd ed./Theodora W. Greene and Peter G. M. Wuts. New York: Wiley, 1999.
    • 55. Katritzky, Alan R. Lagowski, Jeanne M. The principles of heterocyclic ChemistryNew York: Academic Press, 1968.
    • 56. Paquette, Leo A. Principles of modern heterocyclic chemistry New York: Benjamin.
    • 57. Katritzky, Alan R.; Rees, Charles W.; Comprehensive heterocyclic chemistry: the structure, reactions, synthesis, and uses of heterocyclic compounds 1st ed.Oxford (Oxfordshire); New York: Pergamon Press, 1984. 8 v.
    • 58. Katritzky, Alan RHandbook of heterocyclic 1st edOxford (Oxfordshire); New York: Pergamon Press, 1985.
    • 59. Davies, David I Aromatic Heterocyclic Oxford; New York: Oxford University Press, 1991.
    • 60. Ellis, G. P. Synthesis of fused Chichester [Sussex]; New York: Wiley, c1987-c1992. Chemistry of heterocyclic compounds; v. 47.
    • 61. Joule, J. A Mills, K., Smith, G. F. Heterocyclic Chemistry, 3rd ed London; New York Chapman & Hall, 1995.
    • 62. Katritzky, Alan R., Rees, Charles W., Scriven, Eric F. V. Comprehensive heterocyclic chemistry II: a review of the literature 1982-1995.
    • 63. The structure, reactions, synthesis, and uses of heterocyclic compounds 1st ed. Oxford; New York: Pergamon, 1996. 11 v. in 12: ill.; 28 cm.
    • 64. Eicher, Theophil, Hauptmann, Siegfried. The chemistry of heterocycles: structure, reactions, syntheses, and applications Stuttgart; New York: G. Thieme, 1995.
    • 65. Grimmett, M. R. Imidazole and benzimidazole Synthesis London; San Diego: Academic Press, 1997.
    • 66. Advances in heterocyclic chemistry. Published in New York by Academic Press, starting in 1963-present.
    • 67. Gilchrist, T. L. (Thomas Lonsdale) Heterocyclic chemistry 3rd ed. Harlow, Essex: Longman, 1997, 414 p: ill.; 24 cm.
    • 68. Farina, Vittorio; Roth, Gregory P. Recent advances in the Stille reaction; Adv. Met.-Org. Chem. 1996, 5, 1-53.
    • 69. Farina, Vittorio; Krishnamurthy, Venkat; Scott, William J. The Stille reaction; Org. React. (N.Y.) (1997), 50, 1-652.
    • 70. Stille, J. K. Angew. Chem. Int. Ed. Engl. 1986, 25, 508-524.
    • 71. Norio Miyaura and Akiro Suzuki Chem Rev. 1995, 95, 2457.
    • 72. Home, D. A. Heterocycles 1994, 39, 139.
    • 73. Kamitori, Y. et.al. Heterocycles, 1994, 37(1), 153.
    • 74. Shawali, J. Heterocyclic Chem. 1976, 13, 989.
    • 75. a) Kende, A. S.et al. Org. Photochem. Synth. 1972, 1, 92. b) Hankes, L. V.; Biochem. Prep. 1966, 11, 63. c) Synth. Meth. 22, 837.
    • 76. Hulton et. al. Synth. Comm. 1979, 9, 789.
    • 77. Pattanayak, B. K. et.al. Indian J. Chem. 1978, 16, 1030.
    • 78. Chemische Berichte 1902, 35, 1545.
    • 79. Chemische Berichte Ibid 1911, 44, 493.
    • 80. Moubarak, I., Vessiere, R. Synthesis 1980, Vol. 1, 52-53.
    • 81. Ind J. Chem. 1973, 11, 1260.
    • 82. Roomi et.al. Can J. Chem. 1970, 48, 1689.
    • 83. Sorrel, T. N. J. Org. Chem. 1994, 59, 1589.
    • 84. Nitz, T. J. et. al. J. Org. Chem. 1994, 59, 5828-5832.
    • 85. Bowden, K. et.al. J. Chem. Soc. 1946, 953.
    • 86. Nitz, T. J. et. al. J. Org. Chem. 1994, 59, 5828-5832.
    • 87. Scholkopf et. al. Angew. Int. Ed. Engl. 1971, 10(5), 333.
    • 88. (a) Behun, J. D.; Levine, R. J. Org. Chem. 1961, 26, 3379. (b) Rossen, K.; Weissman, S. A.; Sager, J.; Reamer, R. A.; Askin, D.; Volante, R. P.; Reider, P. J. Asymmetric Hydrogenation of tetrahydropyrazines: Synthesis of (S)-piperazine 2-tert-butylcarboxamide, an intermediate in the preparation of the HIV protease inhibitor Indinavir. Tetrahedron Lett., 1995, 36, 6419-6422. (c) Jenneskens, L. W.; Mahy, J.; den Berg, E. M. M. de B.-v.; Van der Hoef, I.; Lugtenburg, J. Recl. Trav. Chim. Pays-Bas 1995, 114, 97.
    • 89. Wang, T.; Zhang, Z.; Meanwell, N. A. Benzoylation of Dianions: Preparation of mono-Benzoylated Symmetric Secondary Diamines. J. Org. Chem., 1999, 64, 7661-7662.
    • 90. (a) Adamczyk, M.; Fino, J. R. Synthesis of procainamide metabolites. N-acetyl desethylprocainamide and desethylprocainamide. Org. Prep. Proced. Int. 1996, 28, 470-474. (b) Wang, T.; Zhang, Z.; Meanwell, N. A. Regioselective mono-Benzoylation of Unsymmetrical Piperazines. J. Org. Chem., in press.
    • 91. Masuzawa, K.; Kitagawa, M.; Uchida, H. Bull Chem. Soc. Jpn. 1967, 40, 244-245.
    • 92. Furber, M.; Cooper, M. E.; Donald, D. K. Tetrahedron Lett. 1993, 34, 1351-1354.
    • 93. Blair, Wade S.; Deshpande, Milind; Fang, Haiquan; Lin, Pin-fang; Spicer, Timothy P.; Wallace, Owen B.; Wang, Hui; Wang, Tao; Zhang, Zhongxing; Yeung, Kap-sun. Preparation of antiviral indoleoxoacetyl piperazine derivatives U.S. Pat. No. 6,469,006. Preparation of antiviral indoleoxoacetyl piperazine derivatives. PCT Int. Appl. (PCT/US00/14359), WO 0076521 A1, filed May 24, 2000, published Dec. 21, 2000.
    • 94. Wang, Tao; Wallace, Owen B.; Zhang, Zhongxing; Meanwell, Nicholas A.; Bender, John A. Antiviral azaindole derivatives. U.S. Pat. No. 6,476,034 and Wang, Tao; Wallace, Owen B.; Zhang, Zhongxing; Meanwell, Nicholas A.; Bender, John A. Preparation of antiviral azaindole derivatives. PCT Int. Appl. (PCT/US01/02009), WO 0162255 A1, filed Jan. 19, 2001, published Aug. 30, 2001.
    • 95. Wallace, Owen B.; Wang, Tao; Yeung, Kap-Sun; Pearce, Bradley C.; Meanwell, Nicholas A.; Qiu, Zhilei; Fang, Haiquan; Xue, Qiufen May; Yin, Zhiwei. Composition and antiviral activity of substituted indoleoxoacetic piperazine derivatives. U.S. Pat. No. 6,573,262 which is a continuation-in-part application of U.S. Ser. No. 09/888,686 filed Jun. 25, 2001 (corresponding to PCT Int. Appl. (PCT/US01/20300), WO 0204440 A1, filed Jun. 26, 2001, published Jan. 17, 2002.
    • 96. J. L. Marco, S. T. Ingate, and P. M. Chinchon Tetrahedron 1999, 55, 7625-7644.
    • 97. C. Thomas, F. Orecher, and P.Gmeiner Synthesis 1998, 1491.
    • 98. M. P. Pavia, S. J. Lobbestael, C. P. Taylor, F. M. Hershenson, and D. W. Miskell.
    • 99. Buckheit, Robert W., Jr. Expert Opinion on Investigational Drugs 2001, 10(8), 1423-1442.
    • 100. Balzarini, J.; De Clercq, E.. Antiretroviral Therapy 2001, 31-62.
    • 101. E. De clercq Journal of Clinical Virology, 2001, 22, 73-89.
    • 102. Merour, Jean-Yves; Joseph, Benoit. Curr. Org. Chem. (2001), 5(5), 471-506.
    • 103. T. W. von Geldern et al. J. Med. Chem 1996, 39, 968.
    • 104. M. Abdaoui et al. Tetrahedron 2000, 56, 2427.
    • 105. W. J. Spillane et al. J. Chem. Soc., Perkin Trans. 1, 1982, 3, 677.
    • 106. Wang, Tao; Zhang, Zhongxing; Meanwell, Nicholas A.; Kadow, John F.; Yin, Zhiwei; Xue, Qiufen May. (USA). Composition and antiviral activity of substituted azaindoleoxoacetic piperazine derivatives. U.S. patent application Publ. (2003), US 20030207910 A1 published Nov. 6, 2003 which is U.S. patent application Ser. No. 10/214,982 filed Aug. 7, 2002, which is a continuation-in-part application of U.S. Ser. No. 10/038,306 filed Jan. 2, 2002 (corresponding to PCT Int. Appl. (PCT/US02/00455), WO 02/062423 A1, filed Jan. 2, 2002, published Aug. 15, 2002.
    • 107. a) Nickel, Bernd; Szelenyi, Istvan; Schmidt, Jurgen; Emig, Peter; Reichert, Dietmar; Gunther, Eckhard; Brune, Kay. Preparation of indolylglyoxylamides as antitumor agents. PCT Int. Appl. (1999), 47 pp. CODEN: PIXXD2 WO 9951224, b) Emig, Peter; Bacher, Gerald; Reichert, Dietmar; Baasner, Silke; Aue, Beate; Nickel, Bernd; Guenther, Eckhard. Preparation of N-(6-quinolinyl)-3-indolylglyoxylamides as antitumor agents. PCT Int. Appl. (2002), 4WO 2002010152A2, c) Nickel, Bernd; Klenner, Thomas; Bacher, Gerald; Beckers, Thomas; Emig, Peter; Engel, Juergen; Bruyneel, Erik; Kamp, Guenter; Peters, Kirsten. Indolyl-3-glyoxylic acid derivatives comprising therapeutically valuable properties. PCT Int. Appl. (2001), WO 2001022954A2.
    • 108. Wang, Tao; Wallace, Owen B.; Meanwell, Nicholas A.; Zhang, Zhongxing; Bender, John A.; Kadow, John F.; Yeung, Kap-Sun. Preparation of indole, azaindole, and related heterocyclic piperazinecarboxamides for treatment of AIDS. PCT Int. Appl. WO 2002085301A2, published Oct. 31, 2002; corresponding to U.S. patent Publication U.S. 20030096825A1, published May 22, 2003.
    • 109. Kadow, John F.; Xue, Qiufen May; Wang, Tao; Zhang, Zhongxing; Meanwell, Nicholas A. Preparation of indole, azaindole and related heterocyclic pyrrolidine derivatives as antiviral agents. PCT Int. Appl. WO 2003068221A1, published Aug. 21, 2003; corresponding to U.S. patent Publication U.S. 20030236277A1.
    • 110. Wang, Tao; Wallace, Owen B.; Meanwell, Nicholas A.; Kadow, John F.; Zhang, Zhongxing; Yang, Zhong. Preparation of piperazine derivatives as antiviral agents. PCT Int. Appl. (2003), WO 2003092695 A1; corresponding to U.S. patent Publication U.S. 20040009985A1, published Jan. 15, 2004.
    • 111. Kadow, John F.; Regueiro-Ren, Alicia; Xue, Qiufen May. Preparation of indolyl-, azaindolyl-, and related heterocyclic sulfonylureidopiperazines for treatment of HIV and AIDS. PCT Int. Appl. (2003), WO 2004000210 A2; corresponding to U.S. patent Publication U.S. 20040006090A1, published Jan. 8, 2004.
    • 112. Regueiro-Ren, Alicia; Xue, Qiufen May; Kadow, John F.; Taylor, Malcolm. Preparation of indolyl-, azaindolyl-, and related heterocyclic ureido and thioureido piperazines for treatment of HIV and AIDS. PCT Int. Appl. (2004), WO 2004011425 A2; corresponding to U.S. patent Publication U.S. 20040063746A1, published Apr. 1, 2004.
    • 113. Wang, Tao; Zhang, Zhongxing; Meanwell, Nicholas A.; Kadow, John F.; Yin, Zhiwei; Xue, Qiufen May; Regueiro-Ren, Alicia; Matiskella, John D.; Ueda, Yasutsugu. Composition and antiviral activity of substituted azaindoleoxoacetic piperazine derivatives. U.S. patent application Publ. (2004), US 2004110785 A1; published Jun. 10, 2004.
    • 114. Wang, Tao; Kadow, John F.; Meanwell, Nicholas A.; Yeung, Kap-Sun; Zhang, Zhongxing; Yin, Zhiwei; Qiu, Zhilei; Deon, Daniel H.; James, Clint A.; Ruediger, Edward H.; Bachand, Carol. Preparation and pharmaceutical compositions of indole, azaindole and related heterocyclic 4-alkenyl piperidine amides. U.S. patent application Publ. (2004), US 20040063744 A1; published Apr. 1, 2004; corresponding to PCT Intl. Appln. (2004), WO 2004/04337.
    SUMMARY OF THE INVENTION
  • The present invention comprises compounds of Formula I, their pharmaceutical formulations, and their use in patients suffering from or susceptible to a virus such as HIV. The compounds of Formula I, which include nontoxic pharmaceutically acceptable salts thereof, have the formula and meaning as described below.
  • The present invention comprises compounds of Formula I, including pharmaceutically acceptable salts thereof, which are effective antiviral agents, particularly as inhibitors of HIV.
    Figure US20050124623A1-20050609-C00004
      • wherein:
      • Q is selected from the group consisting of
        Figure US20050124623A1-20050609-C00005
      • T is —C(O)— or —CH(CN)—;
      • R1 is hydrogen or methyl;
      • R3 and R5 are independently selected from the group consisting of hydrogen, halogen, cyano, nitro, COOR8, XR9 and B;
      • R2 and R4 are independently O or do not exist with the proviso that only one of R2 and R4 are O;
      • R6 is (CH2)nH, wherein n is 0-1;
      • — represents a carbon-carbon bond or does not exist;
      • —Y— is selected from the group consisting of
        Figure US20050124623A1-20050609-C00006
      • R10, R11, R12, R13, R14,R15, R16 and R17 are each independently H or (C1-6)alkyl; wherein said (C1-6)alkyl may optionally be substituted with one to three same or different halogen, OH or CN;
      • R18 is a member selected from the group consisting of C(O)-phenyl, C(O)-heteroaryl, pyridinyl, pyrimidinyl, quinolyl, isoquinolyl, quinazolyl, quinoxalinyl, napthyridinyl, pthalazinyl, azabenzofuryl and azaindolyl; wherein said member is optionally substituted with from one to two substituents selected from the group consisting of methyl, -amino, —NHMe, —NMe2, methoxy, hydroxymethyl and halogen;
      • D is selected from the group consisting of hydrogen, cyano, S(O)2R24, halogen, COOR20, C(O)NR21R22, phenyl and heteroaryl; wherein said phenyl or heteroaryl is independently optionally substituted with one to three same or different halogens or from one to three same or different substituents selected from F (as defined below);
      • A is selected from the group consisting of phenyl, pyridinyl, furanyl, thienyl, isoxazole and oxazole; wherein said phenyl, pyridinyl, furanyl, thienyl, isoxazole or oxazole is independently optionally substituted with one to three same or different halogens or from one to three same or different substituents selected from F;
      • B is selected from the group consisting of (C1-6)alkyl, C(O)NR21R22, —C(O)CH3, —N(CH2CH2)2NC(O)pyrazolyl, phenyl and heteroaryl; wherein said (C1-6)alkyl, phenyl and heteroaryl are independently optionally substituted with one to three same or different halogens or from one to three same or different substituents selected from F;
      • heteroaryl is selected from the group consisting of pyridinyl, pyrazinyl, pyridazinyl, pyrimidinyl, furanyl, thienyl, benzothienyl, thiazolyl, oxazolyl, isoxazolyl, imidazolyl, oxadiazolyl, thiadiazolyl, pyrazolyl, tetrazolyl and triazolyl;
      • F is selected from the group consisting of (C1-6)alkyl, (C1-6)alkenyl, phenyl, pyridinyl, hydroxy, (C1-6)alkoxy, halogen, benzyl, —NR23C(O)—(C1-6)alkyl, —NR24R25, —S(O)2NR24R25, COOR26, —COR27, and —CONR24R25; wherein said (C1-6)alkyl or phenyl are each optionally substituted with hydroxy, (C1-6)alkoxy, (C1-6)alkyl, CF3, dimethylamino or from one to three same or different halogen;
      • R8, R9 and R26 are each independently selected from the group consisting of hydrogen and (C1-6)alkyl;
      • X is selected from the group consisting of NR26, O and S;
      • R20, R21,R22, R23, R24 and R25 are independently selected from the group consisting of hydrogen, (C1-6)alkyl, phenyl and heteroaryl; wherein said phenyl and heteroaryl are each independently optionally substituted with one to three same or different halogen or methyl; and
      • R27 is piperazinyl, N-methyl piperazinyl, or 3-pyrazolyl.
  • A preferred embodiment includes compounds where T is
    Figure US20050124623A1-20050609-C00007
  • Another preferred embodiment of the invention are compounds of Formula I, including pharmaceutically acceptable salts thereof
      • wherein:
      • R1 is hydrogen;
      • — represents a carbon-carbon bond; and
      • R2 and R4 do not exist.
      • D is selected from the group consisting of hydrogen, cyano, S(O)2R24, halogen, COOR24, C(O)NH2, phenyl and heteroaryl; wherein said phenyl or heteroaryl is independently optionally substituted with one to three same or different halogens or a member selected from the group consisting of (C1-6)alkyl, (C1-6)alkenyl, hydroxy, (C1-6)alkoxy, halogen, —NR24R25, —S(O)2NR24R25, COOR26 and —CONR24R25; wherein said (C1-6)alkyl is optionally substituted with one to three same or different halogen or a hydroxy; and
      • A is selected from the group consisting of phenyl, pyridinyl, furanyl, thienyl, isoxazole and oxazole; wherein said phenyl, pyridinyl, furanyl, thienyl, isoxazole or oxazole are independently optionally substituted with one to three same or different halogens or a member selected from the group consisting of (C1-4)alkyl, (C1-4)alkenyl, (C1-3)alkoxy, halogen and —NH2; wherein said (C1-3)alkyl is optionally substituted with one to three same or different halogens.
  • Another preferred embodiment are compounds I wherein:
      • R6 is hydrogen; and
      • R10, R11, R12, R13, R14,R15, R16, R17 are each independently H or methyl with the proviso that a maximum of two of R10—R17 is a methyl.
  • Another preferred embodiment of the invention are compounds of Formula I, as above including pharmaceutically acceptable salts thereof,
      • wherein:
      • Q is a member selected from groups (A), (B), and (C) consisting of
      • (A)
        Figure US20050124623A1-20050609-C00008
      • wherein R3 is hydrogen, C1-C3 alkoxy, —NR26R9 or halogen;
      • (B)
        Figure US20050124623A1-20050609-C00009
      • wherein R3 is hydrogen, methoxy or halogen; and
      • (C)
        Figure US20050124623A1-20050609-C00010
      • wherein R3 is hydrogen, methoxy or halogen.
  • Another preferred embodiment of the invention are compounds of Formula I, as above including pharmaceutically acceptable salts thereof, wherein:
      • group(A) of Q is:
      • (A)
        Figure US20050124623A1-20050609-C00011
      • wherein R3 is hydrogen; and
      • group (C) of Q is:
        Figure US20050124623A1-20050609-C00012
      • wherein:
      • R5 is hydrogen.
  • In another preferred embodiment of the invention, Q is selected from group (A) or (B), and
      • R5 is selected from the group consisting of hydrogen, halogen, heteroaryl, phenyl, cyano, methoxy, COOR8, C(O)NH2, C(O)NHheteroaryl, and C(O)NHCH3; wherein said C(O)NHheteroaryl, phenyl, and heteroaryl are independently optionally substituted with one to three same or different halogens or from one to three same or different substituents selected from F.
  • Other preferred embodiments are compounds I:
      • wherein heteroaryl is selected from the group consisting of pyridinyl, pyrazinyl, pyridazinyl, pyrimidinyl, furanyl, thienyl, thiazolyl, oxazolyl, isoxazolyl, imidazolyl, oxadiazolyl, thiadiazoyl, pyrazolyl, tetrazolyl and triazolyl; wherein said heteroaryl is independently optionally substituted with one to three same or different halogens or from one to three same or different substituents selected from F;
      • R18 is —C(O)phenyl or —C(O)heteroaryl; wherein said heteroaryl is pyridinyl, furanyl or thienyl; wherein heteroaryl is independently optionally substituted with a member selected from the group consisting of halogen, methyl, -amino, —NHMe, NMe2 and hydroxymethyl;
      • —W— is
        Figure US20050124623A1-20050609-C00013
      • R10, R11, R12, R13, R14,R15, R16 and R17 are each independently H or methyl with the proviso that not more than one is methyl; and
      • R18 is selected from the group consisting of C(O)-phenyl or C(O)-heteroaryl wherein each of C(O)-phenyl or —C(O)-heteroaryl may be optionally substituted with from one to two methyl, -amino, —NHMe, —NMe2, methoxy, hydroxymethyl or halogen groups; or
      • R18 is selected from the group consisting of pyridyl, pyrimidinyl, quinolyl, isoquinolyl, quinazolyl, quinoxalinyl, napthyridinyl, pthalazinyl, azabenzofuryl and azaindolyl, each of which may be optionally substituted with from one to two methyl, -amino, —NHMe, —NMe2, methoxy, hydroxymethyl or halogen groups.
  • In another preferred embodiment:
      • —W— is selected from the group consisting of
        Figure US20050124623A1-20050609-C00014
      • R10, R11, R12, R13, R14,R15, R16 and R17 are each independently H or methyl, with the proviso that one is methyl;
      • D is selected from the group consisting of hydrogen, cyano, S(O)2R24, halogen, COOR20, C(O)NH2, phenyl, or heteroaryl; wherein said phenyl or heteroaryl is independently optionally substituted with one to three same or different halogens or from one to three same or different substituents selected from the group consisting of (C1-6)alkyl, (C1-6)alkenyl, hydroxy, (C1-6)alkoxy, halogen, —NR24R25, —S(O)2NR24R25, COOR26 and —CONR24R25; wherein said (C1-6)alkyl is optionally substituted with one to three same or different halogen or a hydroxy; and
      • A is selected from the group consisting of phenyl, pyridinyl, furanyl, thienyl, isoxazole and oxazole; wherein said phenyl, pyridinyl, furanyl, thienyl, isoxazole or oxazole is independently optionally substituted with one to three same or different halogens or from one to three same or different substituents selected from the group consisting of (C1-4)alkyl, (C1-4)alkenyl, (C1-3)alkoxy, halogen and —NH2; wherein said (C1-4)alkyl is optionally substituted with one to three same or different halogens.
  • In another preferred embodiment:
      • Q is selected from Group (A).
  • Another preferred embodiment are compounds of Formula I, including pharmaceutically acceptable salts thereof,
    Figure US20050124623A1-20050609-C00015
      • wherein:
      • Q is selected from the group consisting of
        Figure US20050124623A1-20050609-C00016
      • R1 is hydrogen or methyl;
      • R3 and R5 are independently selected from the group consisting of hydrogen, halogen, cyano, nitro, COOR8, XR9 and B;
      • R2 and R4 are independently O or do not exist, with the proviso that only one of R2 and R4 are O;
      • R6 is (CH2)nH, wherein n is 0-1;
      • — represents a carbon-carbon bond or does not exist;
      • —Y— is selected from the group consisting of
        Figure US20050124623A1-20050609-C00017
      • R10, R11, R12, R13, R14,R15, R16 and R17 are each independently H or (C1-6)alkyl; wherein said (C1-6)alkyl may optionally be substituted with one to three same or different halogen, OH or CN;
      • R18 is a member selected from the group consisting of C(O)-phenyl, C(O)-heteroaryl, pyridinyl, pyrimidinyl, quinolyl, isoquinolyl, quinazolyl, quinoxalinyl, napthyridinyl, pthalazinyl, azabenzofuryl and azaindolyl; wherein said member is optionally substituted with from one to two substituents selected from the group consisting of methyl, -amino, —NHMe, —NMe2, methoxy, hydroxymethyl and halogen;
      • D is selected from the group consisting of hydrogen, cyano, S(O)2R24, halogen, COOR20, C(O)NR21R22, phenyl and heteroaryl; wherein said phenyl or heteroaryl is independently optionally substituted with one to three same or different halogens or from one to three same or different substituents selected from F;
      • A is selected from the group consisting of phenyl, pyridinyl, furanyl, thienyl, isoxazole and oxazole; wherein said phenyl, pyridinyl, furanyl, thienyl, isoxazole or oxazole is independently optionally substituted with one to three same or different halogens or from one to three same or different substituents selected from F;
      • B is selected from the group consisting of (C1-6)alkyl, (C3-6)cycloalkyl, C(O)NR21R22, —C(O)CH3, —N(CH2CH2)2NC(O)pyrazolyl, phenyl and heteroaryl; wherein said (C1-6)alkyl, phenyl and heteroaryl are independently optionally substituted with one to three same or different halogens or from one to three same or different substituents selected from F;
      • heteroaryl is selected from the group consisting of pyridinyl, pyrazinyl, pyridazinyl, pyrimidinyl, furanyl, thienyl, thiazolyl, oxazolyl, isoxazolyl, imidazolyl, oxadiazolyl, thiadiazolyl, pyrazolyl, tetrazolyl and triazolyl;
      • F is selected from the group consisting of (C1-6)alkyl, (C1-6)alkenyl, phenyl, pyridinyl, hydroxy, (C1-6)alkoxy, halogen, benzyl, —NR23C(O)—(C1-6)alkyl, —NR24R25, —S(O)2NR24R25, COOR26, —COR27, and —CONR24R25; wherein said (C1-6)alkyl or phenyl are each optionally substituted with hydroxy, (C1-6)alkoxy, dimethylamino or from one to three same or different halogen;
      • R8, R9 and R26 are each independently selected from the group consisting of hydrogen and (C1-6)alkyl;
      • X is selected from the group consisting of NR26, O and S;
      • R20, R21,R22, R23,R24 and R25 are independently selected from the group consisting of hydrogen, (C1-6)alkyl, phenyl and heteroaryl; wherein said phenyl and heteroaryl are each independently optionally substituted with one to three same or different halogen or methyl; and
      • R27 is piperazinyl, N-methylpiperazinyl or 3-pyrazolyl.
  • In another embodiment are Compounds I, including pharmaceutically acceptable salts, wherein:
      • Q is
        Figure US20050124623A1-20050609-C00018
      • R5 is selected from the group consisting of hydrogen, halogen, cyano, XR9, heteroaryl, —N(CH2CH2)2NC(O)pyrazolyl, and —C(O)CH3, wherein said heteroaryl is optionally substituted with one substituent selected from F;
      • heteroaryl is selected from the group consisting of pyridinyl, pyrazinyl, pyridazinyl, pyrimidinyl, isoxazolyl, isoxazolyl, pyrazolyl, and triazolyl;
      • —Y— is selected from the group consisting of
        Figure US20050124623A1-20050609-C00019
      • R10, R11, R12, R13, R14,R15, R16 and R17 are each hydrogen;
      • A is phenyl or pyridinyl;
      • R18 is C(O)-phenyl, isoquinolyl or quinazolyl;
      • D is cyano or oxadiazolyl;
      • F is selected from the group consisting of (C1-6)alkyl, phenyl, pyridinyl, (C1-2)alkoxy, —COOR26—COR27 and —CONR24R25; wherein said phenyl is optionally substituted with one group selected from methyl, methoxy, fluoro, or trifluoromethyl;
      • X is selected from the group consisting of O;
      • R9 is (C1-2)alkyl;
      • R26 is hydrogen, methyl, or ethyl;
      • R24 and R25 are independently selected from the group consisting of hydrogen and methyl; and
      • R27 is piperazinyl, N-methyl piperazinyl, or 3-pyrazolyl.
  • Another embodiment of the present invention is a method for treating mammals infected with the HIV virus, comprising administering to said mammal an antiviral effective amount of a compound of Formula I, including pharmaceutically acceptable salts thereof, and one or more pharmaceutically acceptable carriers, excipients or diluents; optionally the compound of Formula I can be administered in combination with an antiviral effective amount of an AIDS treatment agent selected from the group consisting of: (a) an AIDS antiviral agent; (b) an anti-infective agent; (c) an immunomodulator; and (d) HIV entry inhibitors.
  • Another embodiment of the present invention is a pharmaceutical composition comprising an antiviral effective amount of a compound of Formula I, including pharmaceutically acceptable salts thereof, and one or more pharmaceutically acceptable carriers, excipients, diluents and optionally in combination with an antiviral effective amount of an AIDS treatment agent selected from the group consisting of: (a) an AIDS antiviral agent; (b) an anti-infective agent; (c) an immunomodulator; and (d) HIV entry inhibitors.
  • DETAILED DESCRIPTION OF THE INVENTION
  • Since the compounds of the present invention, may possess asymmetric centers and therefore occur as mixtures of diastereomers and enantiomers, the present invention includes the individual diastereoisomeric and enantiomeric forms of the compounds of Formula I in addition to the mixtures thereof.
  • Definitions
  • The term “C1-6 alkyl” as used herein and in the claims (unless specified otherwise) mean straight or branched chain alkyl groups such as methyl, ethyl, propyl, isopropyl, butyl, isobutyl, t-butyl, amyl, hexyl and the like.
  • “Halogen” refers to chlorine, bromine, iodine or fluorine.
  • An “aryl” group refers to an all carbon monocyclic or fused-ring polycyclic (i.e., rings which share adjacent pairs of carbon atoms) groups having a completely conjugated pi-electron system. Examples, without limitation, of aryl groups are phenyl, napthalenyl and anthracenyl. The aryl group may be substituted or unsubstituted. When substituted the substituted group(s) is preferably one or more selected from alkyl, cycloalkyl, aryl, heteroaryl, heteroalicyclic, hydroxy, alkoxy, aryloxy, heteroaryloxy, heteroalicycloxy, thiohydroxy, thioaryloxy, thioheteroaryloxy, thioheteroalicycloxy, cyano, halogen, nitro, carbonyl, O-carbamyl, N-carbamyl, C-amido, N-amido, C-carboxy, O-carboxy, sulfinyl, sulfonyl, sulfonamido, trihalomethyl, ureido, amino and —NRxRy, wherein Rx and Ry are independently selected from the group consisting of hydrogen, alkyl, cycloalkyl, aryl, carbonyl, C-carboxy, sulfonyl, trihalomethyl, and, combined, a five- or six-member heteroalicyclic ring.
  • As used herein, a “heteroaryl” group refers to a monocyclic or fused ring (i.e., rings which share an adjacent pair of atoms) group having in the ring(s) one or more atoms selected from the group consisting of nitrogen, oxygen and sulfur and, in addition, having a completely conjugated pi-electron system. Unless otherwise indicated, the heteroaryl group may be attached at either a carbon or nitrogen atom within the heteroaryl group. It should be noted that the term heteroaryl is intended to encompass an N-oxide of the parent heteroaryl if such an N-oxide is chemically feasible as is known in the art. Examples, without limitation, of heteroaryl groups are furyl, thienyl, benzothienyl, thiazolyl, imidazolyl, oxazolyl, oxadiazolyl, thiadiazolyl, benzothiazolyl, triazolyl, tetrazolyl, isoxazolyl, isothiazolyl, pyrrolyl, pyranyl, tetrahydropyranyl, pyrazolyl, pyridyl, pyrimidinyl, quinolinyl, isoquinolinyl, purinyl, carbazolyl, benzoxazolyl, benzimidazolyl, indolyl, isoindolyl, pyrazinyl. diazinyl, pyrazine, triazinyltriazine, tetrazinyl, and tetrazolyl. When substituted the substituted group(s) is preferably one or more selected from alkyl, cycloalkyl, aryl, heteroaryl, heteroalicyclic, hydroxy, alkoxy, aryloxy, heteroaryloxy, heteroalicycloxy, thiohydroxy, thioaryloxy, thioheteroaryloxy, thioheteroalicycloxy, cyano, halogen, nitro, carbonyl, O-carbamyl, N-carbamyl, C-amido, N-amido, C-carboxy, O-carboxy, sulfinyl, sulfonyl, sulfonamido, trihalomethyl, ureido, amino, and —NRxRy, wherein Rx and Ry are as defined above.
  • As used herein, a “heteroalicyclic” group refers to a monocyclic or fused ring group having in the ring(s) one or more atoms selected from the group consisting of nitrogen, oxygen and sulfur. Rings are selected from those which provide stable arrangements of bonds and are not intended to encomplish systems which would not exist. The rings may also have one or more double bonds. However, the rings do not have a completely conjugated pi-electron system. Examples, without limitation, of heteroalicyclic groups are azetidinyl, piperidyl, piperazinyl, imidazolinyl, thiazolidinyl, 3-pyrrolidin-1-yl, morpholinyl, thiomorpholinyl and tetrahydropyranyl. When substituted the substituted group(s) is preferably one or more selected from alkyl, cycloalkyl, aryl, heteroaryl, heteroalicyclic, hydroxy, alkoxy, aryloxy, heteroaryloxy, heteroalicycloxy, thiohydroxy, thioalkoxy, thioaryloxy, thioheteroaryloxy, thioheteroalicycloxy, cyano, halogen, nitro, carbonyl, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, C-thioamido, N-amido, C-carboxy, O-carboxy, sulfinyl, sulfonyl, sulfonamido, trihalomethanesulfonamido, trihalomethanesulfonyl, silyl, guanyl, guanidino, ureido, phosphonyl, amino and —NRxRy, wherein Rx and Ry are as defined above.
  • An “alkyl” group refers to a saturated aliphatic hydrocarbon including straight chain and branched chain groups. Preferably, the alkyl group has 1 to 20 carbon atoms (whenever a numerical range; e.g., “1-20”, is stated herein, it means that the group, in this case the alkyl group may contain 1 carbon atom, 2 carbon atoms, 3 carbon atoms, etc. up to and including 20 carbon atoms). More preferably, it is a medium size alkyl having 1 to 10 carbon atoms. Most preferably, it is a lower alkyl having 1 to 4 carbon atoms. The alkyl group may be substituted or unsubstituted. When substituted, the substituent group(s) is preferably one or more individually selected from trihaloalkyl, cycloalkyl, aryl, heteroaryl, heteroalicyclic, hydroxy, alkoxy, aryloxy, heteroaryloxy, heteroalicycloxy, thiohydroxy, thioalkoxy, thioaryloxy, thioheteroaryloxy, thioheteroalicycloxy, cyano, halo, nitro, carbonyl, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, C-thioamido, N-amido, C-carboxy, O-carboxy, sulfinyl, sulfonyl, sulfonamido, trihalomethanesulfonamido, trihalomethanesulfonyl, and combined, a five- or six-member heteroalicyclic ring.
  • A “cycloalkyl” group refers to an all-carbon monocyclic or fused ring (i.e., rings which share and adjacent pair of carbon atoms) group wherein one or more rings does not have a completely conjugated pi-electron system. Examples, without limitation, of cycloalkyl groups are cyclopropane, cyclobutane, cyclopentane, cyclopentene, cyclohexane, cyclohexadiene, cycloheptane, cycloheptatriene and adamantane. A cycloalkyl group may be substituted or unsubstituted. When substituted, the substituent group(s) is preferably one or more individually selected from alkyl, aryl, heteroaryl, heteroalicyclic, hydroxy, alkoxy, aryloxy, heteroaryloxy, heteroalicycloxy, thiohydroxy, thioalkoxy, thioaryloxy, thioheteroaryloxy, thioheteroalicycloxy, cyano, halo, nitro, carbonyl, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, C-thioamido, N-amido, C-carboxy, O-carboxy, sulfinyl, sulfonyl, sulfonamido, trihalo-methanesulfonamido, trihalomethanesulfonyl, silyl, guanyl, guanidino, ureido, phosphonyl, amino and —NRxRy with Rx and Ry as defined above.
  • An “alkenyl” group refers to an alkyl group, as defined herein, consisting of at least two carbon atoms and at least one carbon-carbon double bond.
  • An “alkynyl” group refers to an alkyl group, as defined herein, consisting of at least two carbon atoms and at least one carbon-carbon triple bond.
  • A “hydroxy” group refers to an —OH group.
  • An “alkoxy” group refers to both an —O-alkyl and an —O-cycloalkyl group as defined herein.
  • An “aryloxy” group refers to both an —O-aryl and an —O-heteroaryl group, as defined herein.
  • A “heteroaryloxy” group refers to a heteroaryl-O— group with heteroaryl as defined herein.
  • A “heteroalicycloxy” group refers to a heteroalicyclic-O— group with heteroalicyclic as defined herein.
  • A “thiohydroxy” group refers to an —SH group.
  • A “thioalkoxy” group refers to both an S-alkyl and an —S-cycloalkyl group, as defined herein.
  • A “thioaryloxy” group refers to both an —S-aryl and an —S-heteroaryl group, as defined herein.
  • A “thioheteroaryloxy” group refers to a heteroaryl-S— group with heteroaryl as defined herein.
  • A “thioheteroalicycloxy” group refers to a heteroalicyclic-S— group with heteroalicyclic as defined herein.
  • A “carbonyl” group refers to a —C(═O)—R″ group, where R″ is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl (bonded through a ring carbon) and heteroalicyclic (bonded through a ring carbon), as each is defined herein.
  • An “aldehyde” group refers to a carbonyl group where R″ is hydrogen.
  • A “thiocarbonyl” group refers to a —C(═S)—R″ group, with R″ as defined herein.
  • A “Keto” group refers to a —CC(═O)C— group wherein the carbon on either or both sides of the C═O may be alkyl, cycloalkyl, aryl or a carbon of a heteroaryl or heteroaliacyclic group.
  • A “trihalomethanecarbonyl” group refers to a Z3CC(═O)— group with said Z being a halogen.
  • A “C-carboxy” group refers to a —C(═O)O—R″ groups, with R″ as defined herein.
  • An “O-carboxy” group refers to a R″C(═O)O-group, with R″ as defined herein.
  • A “carboxylic acid” group refers to a C-carboxy group in which R″ is hydrogen.
  • A “trihalomethyl” group refers to a —CZ3, group wherein Z is a halogen group as defined herein.
  • A “trihalomethanesulfonyl” group refers to an Z3CS(═O)2— groups with Z as defined above.
  • A “trihalomethanesulfonamido” group refers to a Z3CS(═O)2NRx— group with Z and RX as defined herein.
  • A “sulfinyl” group refers to a —S(═O)—R″ group, with R″ as defined herein and, in addition, as a bond only; i.e., —S(O)—.
  • A “sulfonyl” group refers to a —S(═O)2R″ group with R″ as defined herein and, in addition as a bond only; i.e., —S(O)2—.
  • A “S-sulfonamido” group refers to a —S(═O)2NRXRY, with RX and RY as defined herein.
  • A “N-Sulfonamido” group refers to a R″S(═O)2NRX— group with Rx as defined herein.
  • A “O-carbamyl” group refers to a —OC(═O)NRxRy as defined herein.
  • A “N-carbamyl” group refers to a RxOC(═O)NRy group, with Rx and Ry as defined herein.
  • A “O-thiocarbamyl” group refers to a —OC(═S)NRxRy group with Rx and Ry as defined herein.
  • A “N-thiocarbamyl” group refers to a RxOC(═S)NRy— group with Rx and Ry as defined herein.
  • An “amino” group refers to an —NH2 group.
  • A “C-amido” group refers to a —C(═O)NRxRy group with Rx and Ry as defined herein.
  • A “C-thioamido” group refers to a —C(═S)NRxRy group, with Rx and Ry as defined herein.
  • A “N-amido” group refers to a RxC(═O)NRy— group, with Rx and Ry as defined herein.
  • An “ureido” group refers to a —NRxC(═O)NRyRy2 group with Rx and Ry as defined herein and Ry2 defined the same as Rx and Ry.
  • An “thioureido” group refers to a —NRxC(═S)NRyRy2 group with Rx and Ry as defined herein and Ry2 defined the same as Rx and Ry.
  • A “guanidino” group refers to a —RxNC(═N)NRyRy2 group, with Rx, Ry and Ry2 as defined herein.
  • A “guanyl” group refers to a RxRyNC(═N)— group, with Rx and Ry as defined herein.
  • A “cyano” group refers to a —CN group.
  • A “silyl” group refers to a —Si(R″)3, with R″ as defined herein.
  • A “phosphonyl” group refers to a P(═O)(ORx)2 with Rx as defined herein.
  • A “hydrazino” group refers to a —NRxNRyRy2 group with Rx, Ry and Ry2 as defined herein.
  • Any two adjacent R groups may combine to form an additional aryl, cycloalkyl, heteroaryl or heterocyclic ring fused to the ring initially bearing those R groups.
  • It is known in the art that nitogen atoms in heteroaryl systems can be “participating in a heteroaryl ring double bond”, and this refers to the form of double bonds in the two tautomeric structures which comprise five-member ring heteroaryl groups. This dictates whether nitrogens can be substituted as well understood by chemists in the art. The disclosure and claims of the present invention are based on the known general principles of chemical bonding. It is understood that the claims do not encompass structures known to be unstable or not able to exist based on the literature.
  • Physiologically acceptable salts and prodrugs of compounds disclosed herein are within the scope of this invention. The term “pharmaceutically acceptable salt” as used herein and in the claims is intended to include nontoxic base addition salts. Suitable salts include those derived from organic and inorganic acids such as, without limitation, hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid, methanesulfonic acid, acetic acid, tartaric acid, lactic acid, sulfinic acid, citric acid, maleic acid, fumaric acid, sorbic acid, aconitic acid, salicylic acid, phthalic acid, and the like. The term “pharmaceutically acceptable salt” as used herein is also intended to include salts of acidic groups, such as a carboxylate, with such counterions as ammonium, alkali metal salts, particularly sodium or potassium, alkaline earth metal salts, particularly calcium or magnesium, and salts with suitable organic bases such as lower alkylamines (methylamine, ethylamine, cyclohexylamine, and the like) or with substituted lower alkylamines (e.g. hydroxyl-substituted alkylamines such as diethanolamine, triethanolamine or tris(hydroxymethyl)-aminomethane), or with bases such as piperidine or morpholine.
  • In the method of the present invention, the term “antiviral effective amount” means the total amount of each active component of the method that is sufficient to show a meaningful patient benefit, i.e., healing of acute conditions characterized by inhibition of the HIV infection. When applied to an individual active ingredient, administered alone, the term refers to that ingredient alone. When applied to a combination, the term refers to combined amounts of the active ingredients that result in the therapeutic effect, whether administered in combination, serially or simultaneously. The terms “treat, treating, treatment” as used herein and in the claims means preventing or ameliorating diseases associated with HIV infection.
  • The present invention is also directed to combinations of the compounds with one or more agents useful in the treatment of AIDS. For example, the compounds of this invention may be effectively administered, whether at periods of pre-exposure and/or post-exposure, in combination with effective amounts of the AIDS antivirals, immunomodulators, antiinfectives, or vaccines, such as those in the following table.
    Drug Name Manufacturer Indication
    ANTIVIRALS
    097 Hoechst/Bayer HIV infection,
    AIDS, ARC
    (non-nucleoside
    reverse trans-
    criptase (RT)
    inhibitor)
    Amprenivir Glaxo Wellcome HIV infection,
    141 W94 AIDS, ARC
    GW 141 (protease inhibitor)
    Abacavir Glaxo Wellcome HIV infection,
    (1592U89) AIDS, ARC
    GW 1592 (RT inhibitor)
    Acemannan Carrington Labs ARC
    (Irving, TX)
    Acyclovir Burroughs Wellcome HIV infection, AIDS,
    ARC, in combination
    with AZT
    AD-439 Tanox Biosystems HIV infection, AIDS,
    ARC
    AD-519 Tanox Biosystems HIV infection, AIDS,
    ARC
    Adefovir Gilead Sciences HIV infection
    dipivoxil Ethigen ARC, PGL
    AL-721 (Los Angeles, CA) HIV positive, AIDS
    Alpha Glaxo Wellcome Kaposi's sarcoma,
    Interferon HIV in combination
    w/Retrovir
    Ansamycin Adria Laboratories ARC
    LM 427 (Dublin, OH)
    Erbamont
    (Stamford, CT)
    Antibody which Advanced Biotherapy AIDS, ARC
    Neutralizes pH Concepts
    Labile alpha (Rockville, MD)
    aberrant
    Interferon
    AR177 Aronex Pharm HIV infection, AIDS,
    ARC
    Beta-fluoro- Nat'l Cancer Institute AIDS-associated
    ddA diseases
    BMS-232623 Bristol-Myers Squibb/ HIV infection,
    (CGP-73547) Novartis AIDS, ARC
    (protease inhibitor)
    BMS-234475 Bristol-Myers Squibb/ HIV infection,
    (CGP-61755) Novartis AIDS, ARC
    (protease inhibitor)
    CI-1012 Warner-Lambert HIV-1 infection
    Cidofovir Gilead Science CMV retinitis,
    herpes, papillomavirus
    Curdlan AJI Pharma USA HIV infection
    sulfate
    Cytomegalovirus MedImmune CMV retinitis
    Immune globin
    Cytovene Syntex Sight threatening
    Ganciclovir CMV
    peripheral CMV
    retinitis
    Delaviridine Pharmacia-Upjohn HIV infection,
    AIDS, ARC
    (RT inhibitor)
    Dextran Ueno Fine Chem. AIDS, ARC, HIV
    Sulfate Ind. Ltd. (Osaka, positive
    Japan) asymptomatic
    ddC Hoffman-La Roche HIV infection, AIDS,
    Dideoxycytidine ARC
    ddI Bristol-Myers Squibb HIV infection, AIDS,
    Dideoxyinosine ARC; combination
    with AZT/d4T
    DMP-450 AVID HIV infection,
    (Camden, NJ) AIDS, ARC
    (protease inhibitor)
    Efavirenz DuPont Merck HIV infection,
    (DMP 266) AIDS, ARC
    (−)6-Chloro- (non-nucleoside RT
    4-(S)-cyclo- inhibitor)
    propylethynyl-
    4(S)-trifluoro-
    methyl-1,4-
    dihydro-2H-3,1-
    benzoxazin-2-
    one, STOCRINE
    EL10 Elan Corp, PLC HIV infection
    (Gainesville, GA)
    Famciclovir Smith Kline herpes zoster,
    herpes simplex
    FTC Emory University HIV infection,
    AIDS, ARC
    (reverse transcriptase
    inhibitor)
    GS 840 Gilead HIV infection,
    AIDS, ARC
    (reverse transcriptase
    inhibitor)
    HBY097 Hoechst Marion HIV infection,
    Roussel AIDS, ARC
    (non-nucleoside
    reverse transcriptase
    inhibitor)
    Hypericin VIMRx Pharm. HIV infection, AIDS,
    ARC
    Recombinant Triton Biosciences AIDS, Kaposi's
    Human (Almeda, CA) sarcoma, ARC
    Interferon
    Beta
    Interferon Interferon Sciences ARC, AIDS
    alfa-n3
    Indinavir Merck HIV infection, AIDS,
    ARC, asymptomatic
    HIV positive, also in
    combination with
    AZT/ddI/ddC
    ISIS 2922 ISIS Pharmaceuticals CMV retinitis
    KNI-272 Nat'l Cancer Institute HIV-assoc. diseases
    Lamivudine, Glaxo Wellcome HIV infection,
    3TC AIDS, ARC
    (reverse
    transcriptase
    inhibitor); also
    with AZT
    Lobucavir Bristol-Myers Squibb CMV infection
    Nelfinavir Agouron HIV infection,
    Pharmaceuticals AIDS, ARC
    (protease inhibitor)
    Nevirapine Boeheringer HIV infection,
    Ingleheim AIDS, ARC
    (RT inhibitor)
    Novapren Novaferon Labs, Inc. HIV inhibitor
    (Akron, OH)
    Peptide T Peninsula Labs AIDS
    Octapeptide (Belmont, CA)
    Sequence
    Trisodium Astra Pharm. CMV retinitis, HIV
    Phosphono- Products, Inc. infection, other CMV
    formate infections
    PNU-140690 Pharmacia Upjohn HIV infection,
    AIDS, ARC
    (protease inhibitor)
    Probucol Vyrex HIV infection, AIDS
    RBC-CD4 Sheffield Med. Tech HIV infection,
    (Houston, TX) AIDS, ARC
    Ritonavir Abbott HIV infection,
    AIDS, ARC
    (protease inhibitor)
    Saquinavir Hoffmann-LaRoche HIV infection,
    AIDS, ARC
    (protease inhibitor)
    Stavudine; d4T Bristol-Myers Squibb HIV infection, AIDS,
    Didehydro- ARC
    deoxythymidine
    Valaciclovir Glaxo Wellcome Genital HSV & CMV
    infections
    Virazole Viratek/ICN asymptomatic HIV
    Ribavirin (Costa Mesa, CA) positive, LAS, ARC
    VX-478 Vertex HIV infection, AIDS,
    ARC
    Zalcitabine Hoffmann-LaRoche HIV infection, AIDS,
    ARC, with AZT
    Zidovudine; Glaxo Wellcome HIV infection, AIDS,
    AZT ARC, Kaposi's
    sarcoma, in
    combination with
    other therapies
    Tenofovir Gilead HIV infection, AIDS,
    disoproxil, (reverse transcriptase
    fumarate salt inhibitor)
    (Viread ®)
    Combivir ® GSK HIV infection, AIDS,
    (reverse transcriptase
    inhibitor)
    abacavir GSK HIV infection, AIDS,
    succinate (reverse transcriptase
    (or Ziagen ®) inhibitor)
    Reyataz ® (or Bristol-Myers Squibb HIV infection AIDs,
    atazanavir) protease inhibitor
    Fuzeon ® Roche/Trimeris HIV infection AIDs,
    (or T-20) viral Fusion
    inhibitor
    IMMUNOMODULATORS
    AS-101 Wyeth-Ayerst AIDS
    Bropirimine Pharmacia Upjohn Advanced AIDS
    Acemannan Carrington Labs, Inc. AIDS, ARC
    (Irving, TX)
    CL246, 738 American Cyanamid AIDS, Kaposi's
    Lederle Labs sarcoma
    EL10 Elan Corp, PLC HIV infection
    (Gainesville, GA)
    FP-21399 Fuki ImmunoPharm Blocks HIV fusion
    with CD4+ cells
    Gamma Genentech ARC, in combination
    Interferon w/TNF (tumor
    necrosis factor)
    Granulocyte Genetics Institute AIDS
    Macrophage Sandoz
    Colony
    Stimulating
    Factor
    Granulocyte Hoechst-Roussel AIDS
    Macrophage Immunex
    Colony
    Stimulating
    Factor
    Granulocyte Schering-Plough AIDS, combination
    Macrophage w/AZT
    Colony
    Stimulating
    Factor
    HIV Core Rorer Seropositive HIV
    Particle
    Immuno-
    stimulant
    IL-2 Cetus AIDS, in
    Interleukin-2 combination w/AZT
    IL-2 Hoffman-LaRoche AIDS, ARC, HIV, in
    Interleukin-2 Immunex combination w/AZT
    IL-2 Chiron AIDS, increase in
    Interleukin-2 CD4 cell counts
    (aldeslukin)
    Immune Cutter Biological Pediatric AIDS, in
    Globulin (Berkeley, CA) combination w/AZT
    Intravenous
    (human)
    IMREG-1 Imreg AIDS, Kaposi's
    (New Orleans, LA) sarcoma, ARC, PGL
    IMREG-2 Imreg AIDS, Kaposi's
    (New Orleans, LA) sarcoma, ARC, PGL
    Imuthiol Merieux Institute AIDS, ARC
    Diethyl
    Dithio
    Carbamate
    Alpha-2 Schering Plough Kaposi's sarcoma
    Interferon w/AZT, AIDS
    Methionine- TNI Pharmaceutical AIDS, ARC
    Enkephalin (Chicago, IL)
    MTP-PE Ciba-Geigy Corp. Kaposi's sarcoma
    Muramyl-
    Tripeptide
    Granulocyte Amgen AIDS, in
    Colony combination w/AZT
    Stimulating
    Factor
    Remune Immune Response Immunotherapeutic
    Corp.
    rCD4 Genentech AIDS, ARC
    Recombinant
    Soluble Human
    CD4
    rCD4-IgG AIDS, ARC
    hybrids
    Recombinant Biogen AIDS, ARC
    Soluble Human
    CD4
    Interferon Hoffman-La Roche Kaposi's sarcoma
    Alfa 2a AIDS, ARC, in
    combination w/AZT
    SK&F106528 Smith Kline HIV infection
    Soluble T4
    Thymopentin Immunobiology HIV infection
    Research Institute
    (Annandale, NJ)
    Tumor Necrosis Genentech ARC, in combination
    Factor; TNF w/gamma Interferon
    ANTI-INFECTIVES
    Clindamycin Pharmacia Upjohn PCP
    with Primaquine
    Fluconazole Pfizer Cryptococcal
    meningitis,
    candidiasis
    Pastille Squibb Corp. Prevention of
    Nystatin oral candidiasis
    Pastille
    Ornidyl Merrell Dow PCP
    Eflornithine
    Pentamidine LyphoMed PCP treatment
    Isethionate (Rosemont, IL)
    (IM & IV)
    Trimethoprim Antibacterial
    Trimethoprim/ Antibacterial
    sulfa
    Piritrexim Burroughs Wellcome PCP treatment
    Pentamidine Fisons Corporation PCP prophylaxis
    Isethionate for
    Inhalation
    Spiramycin Rhone-Poulenc Cryptosporidial
    diarrhea
    Intraconazole- Janssen-Pharm. Histoplasmosis;
    R51211 cryptococcal
    meningitis
    Trimetrexate Warner-Lambert PCP
    Daunorubicin NeXstar, Sequus Kaposi's sarcoma
    Recombinant Ortho Pharm. Corp. Severe anemia
    Human assoc. with AZT
    Erythropoietin therapy
    Recombinant Serono AIDS-related
    Human wasting, cachexia
    Growth Hormone
    Megestrol Bristol-Myers Squibb Treatment of
    Acetate anorexia assoc.
    W/AIDS
    Testosterone Alza, Smith Kline AIDS-related wasting
    Total Enteral Norwich Eaton Diarrhea and
    Nutrition Pharmaceuticals malabsorption
    related to AIDS
  • Additionally, the compounds of the invention herein may be used in combination with another class of agents for treating AIDS which are called HIV entry inhibitors. Examples of such HIV entry inhibitors are discussed in DRUGS OF THE FUTURE 1999, 24(12), pp. 1355-1362; CELL, Vol. 9, pp. 243-246, Oct. 29, 1999; and DRUG DISCOVERY TODAY, Vol. 5, No. 5, May 2000, pp. 183-194 and Inhibitors of the entry of HIV into host cells. Meanwell, Nicholas A.; Kadow, John F. Current Opinion in Drug Discovery & Development (2003), 6(4), 451-461. Specifically the compounds can be utilized in combination with other attachment inhibitors, fusion inhibitors, and chemokine receptor antagonists aimed at either the CCR5 or CXCR4 coreceptor.
  • It will be understood that the scope of combinations of the compounds of this invention with AIDS antivirals, immunomodulators, anti-infectives, HIV entry inhibitors or vaccines is not limited to the list in the above Table but includes, in principle, any combination with any pharmaceutical composition useful for the treatment of AIDS.
  • Preferred combinations are simultaneous or alternating treatments with a compound of the present invention and an inhibitor of HIV protease and/or a non-nucleoside inhibitor of HIV reverse transcriptase. An optional fourth component in the combination is a nucleoside inhibitor of HIV reverse transcriptase, such as AZT, 3TC, ddC or ddI. A preferred inhibitor of HIV protease is Reyataz® (active ingredient Atazanavir). Typically a dose of 300 to 600 mg is administered once a day. This may be co-administered with a low dose of Ritonavir (50 to 500 mgs). Another preferred inhibitor of HIV protease is Kaletra®. Another useful inhibitor of HIV protease is indinavir, which is the sulfate salt of N-(2(R)-hydroxy-1-(S)-indanyl)-2(R)-phenylmethyl-4-(S)-hydroxy-5-(1-(4-(3-pyridyl-methyl)-2(S)-N′-(t-butylcarboxamido)-piperazinyl))-pentaneamide ethanolate, and is synthesized according to U.S. Pat. No. 5,413,999. Indinavir is generally administered at a dosage of 800 mg three times a day. Other preferred protease inhibitors are nelfinavir and ritonavir. Another preferred inhibitor of HIV protease is saquinavir which is administered in a dosage of 600 or 1200 mg tid. Preferred non-nucleoside inhibitors of HIV reverse transcriptase include efavirenz. The preparation of ddC, ddI and AZT are also described in EPO 0,484,071. These combinations may have unexpected effects on limiting the spread and degree of infection of HIV. Preferred combinations include those with the following (1) indinavir with efavirenz, and, optionally, AZT and/or 3TC and/or ddI and/or ddC; (2) indinavir, and any of AZT and/or ddI and/or ddC and/or 3TC, in particular, indinavir and AZT and 3TC; (3) stavudine and 3TC and/or zidovudine; (4) zidovudine and lamivudine and 141W94 and 1592U89; (5) zidovudine and lamivudine.
  • In such combinations the compound of the present invention and other active agents may be administered separately or in conjunction. In addition, the administration of one element may be prior to, concurrent to, or subsequent to the administration of other agent(s).
  • Abbreviations
  • The following abbreviations, most of which are conventional abbreviations well known to those skilled in the art, are used throughout the description of the invention and the examples. Some of the abbreviations used are as follows:
  • h=hour(s)
      • r.t.=room temperature
      • mol=mole(s)
      • mmol=millimole(s)
      • g=gram(s)
      • mg=milligram(s)
      • mL=milliliter(s)
      • TFA=Trifluoroacetic Acid
      • DCE=1,2-Dichloroethane
      • CH2Cl2=Dichloromethane
      • TPAP=tetrapropylammonium perruthenate
      • THF=Tetrahydofuran
      • DEPBT=3-(Diethoxyphosphoryloxy)-1,2,3-benzotriazin-4(3H)-one
      • DMAP=4-dimethylaminopyridine
      • P-EDC=Polymer supported 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide
      • EDC=1-(3-dimethylaminopropyl)-3-ethylcarbodiimide
      • DMF=N,N-dimethylformamide
      • Hunig's Base=N,N-Diisopropylethylamine
      • MCPBA=meta-Chloroperbenzoic Acid
      • azaindole=1H-Pyrrolo-pyridine
      • 4-azaindole=1H-pyrrolo[3,2-b]pyridine
      • 5-azaindole=1H-Pyrrolo[3,2-c]pyridine
      • 6-azaindole=1H-pyrrolo[2,3-c]pyridine
      • 7-azaindole=1H-Pyrrolo[2,3-b]pyridine
      • 4,6-diazaindole=5H-Pyrrolo[3,2-d]pyrimidine
      • 5,6-diazaindole=1H-Pyrrolo[2,3-d]pyridazine
      • 5,7-diazaindole=7H-Pyrrolo[2,3-d]pyrimidine
      • PMB=4-Methoxybenzyl
      • DDQ=2,3-Dichloro-5,6-dicyano-1,4-benzoquinone
      • OTf=Trifluoromethanesulfonoxy
      • NMM=4-Methylmorpholine
      • PIP-COPh=1-Benzoylpiperazine
      • NaHMDS=Sodium hexamethyldisilazide
      • EDAC=1-(3-Dimethylaminopropyl)-3-ethylcarbodiimide
      • TMS=Trimethylsilyl
      • DCM=Dichloromethane
      • DCE=Dichloroethane
      • MeOH=Methanol
      • THF=Tetrahydrofuran
      • EtOAc=Ethyl Acetate
      • LDA=Lithium diisopropylamide
      • TMP-Li=2,2,6,6-tetramethylpiperidinyl lithium
      • DME=Dimethoxyethane
      • DIBALH=Diisobutylaluminum hydride
      • HOBT=1-hydroxybenzotriazole
      • CBZ=Benzyloxycarbonyl
      • PCC=Pyridinium chlorochromate
        Chemistry
  • The present invention comprises compounds of Formula I, their pharmaceutical formulations, and their use in patients suffering from or susceptible to HIV infection. The compounds of Formula I include pharmaceutically acceptable salts thereof.
  • The synthesis procedures and anti-HIV-1 activities of substituted diazaindole oxoacetic and piperidine containing analogs are described below.
  • Scheme A depicts one of the preferred methods for preparing the compounds of the invention. In this method, as shown in Step A, a functionalized diazaindole which also has a carboxy ester appended to the three position is condensed with an acetonitrile anion functionalized with Y to provide the alpha cyano ketone examples of the invention. Oxidation of these compounds as shown in Step B, provides further compounds of the invention.
    Figure US20050124623A1-20050609-C00020
  • Step A. The carboxylic ester intermediates Z-CO2R or more preferably the acid chlorides Z-CO2Cl from Scheme A are condensed with a cyanomethyl intermediate YCH2CN under basic conditions to form the α-cyanoketo intermediate ZC(O)CH(CN)Y. The base KHMDS in THF at r.t. is employed most often, but other amide bases such as NaHMDS could be utilized. The typical solvent utilized is THF but DMF can be employed for less soluble molecules. Typically the reaction with an acid chloride Z-CO2Cl is conducted with the reaction flask immersed in a dry ice acetone cooling bath (˜−78° C.) when THF is the solvent and an acetonitrile/acetone cooling bath (˜−42° C.) when DMF is the solvent but temperatures between −78° and 50° C. could be employed in appropriate cases. The reaction is stirred between 1 h and 1 day. Typically the reaction when judged to be complete by TLC or LC or LC/MS is maintained at the cold temperature and oxidant added directly to the reaction as described in Step B. Alternatively the reaction could be allowed to attain ambient temperature and either allowed to react further if necessary and then quenched or be immediately quenched with saturated aqueous sodium bicarbonate. The mixture could then be extracted with EtOAc, concentrated and the α-cyanoketo intermediate ZC(O)CH(CN)Y could be purified by preparative HPLC. When the same reaction is carried out with an ester Z-CO2R as the reactant, the alkylation reaction is initiated and then is usually allowed to warm to ambient temperature for further reaction. Typically R is methyl or ethyl or less ideally another lower alkyl group. Phenoxy, pentafluorophenoxy, or Weinreb esters (R═—NH2OMe) might also be employed. As mentioned above, in the event that the carboxylic ester intermediates Z-CO2R are less reactive than desired under the standard condensation conditions, they may be activated by the intial conversion to an acid chloride Z-COCl (OR where R=H converted to Cl). This is currently the preferred method for diazaindole esters of this invention. The preparation of the acid chlorides from Z-CO2R— is accomplished by initial hydrolysis of the ester to the analogous carboxylic acid Z-CO2H. A typical procedure involves stirring the ester with LiOH in THF and water at 100° C. for 6 h to 2 days, concentrating the crude mixture and recrystallizing the carboxylic acid from water. The carboxylic acid Z-CO2H is then dissolved or more typically suspended as a slurry in dichloromethane and stirred with oxalyl chloride and a catalytic amount of DMF from 4-24 h but typically overnight. The solvents are removed in vacuo and the acid chloride used directly. Possible alternative solvents are benzene or toluene. A possible alternative method for conversion of the carboxylic acid to an acid chloride entails reacting thionyl choride in benzene at 100° C. between 2 h and 6 h with the acid in the presence of catalytic DMF followed by concentration in vacuo to yield the acid chloride Z-CO2Cl. As mentioned above, the acid chloride Z-CO2Cl is the preferred reactant for conducting step A for the preparation of diazindole compounds of formula I. Alternatively, the acid may be converted to an acid anhydrides which may also find utility in the alkylation reaction.
  • Step B. The preferred method for accomplishing step B, the conversion of the α-cyanoketo intermediate ZC(O)CH(CN)Y to the diacarbonyl compounds of formula I or ketoamide intermediates to prepare compounds of formula I is to to add 1-20 equivalents but most preferably 5 equivalents of a commercially available solution of 32% peracetic acid in dilute aq acetic acid tb the reaction flask containing the completed reaction described in Step A. The reaction is typically stirred at the same temperature at which the alkylation reaction was conducted (for the Step A reactions with an acid chloride in THF ˜−78° and for the step A reactions in DMF ˜−42°) for a period of 1 h and then allowed to warm to ambient temperature if not already at that tmeperature. The reaction mixture is then either allowed to react further or immediately diluted with saturated aq. ammonium chloride and EtOAc. For relatively insoluble acid products which precipatate, the resultant precipitate is isolated by filtration as the oxoacetyl product ZC(O)C(O)Y. For organic soluble acid products, the acid is extracted into the organic layer and the layers separated. The organic layer is concentrated in vacuo and the product purified via preparative HPLC. The α-cyanoketo intermediate ZC(O)CH(CN)Y, if isolated, can be oxidized to the oxoacetyl product ZC(O)C(O)Y using a variety of oxidants including mCPBA, NaOCl (bleach), peracetic acid, or nickel peroxide. In a typical procedure a solution of peracetic acid in acetic acid is added to a solution of α-cyanoketo intermediate ZC(O)CH(CN)Y in THF and the reaction is stirred at between r.t and −70° C. for between 30 min and 2 h. The reaction mixture is then diluted with saturated aq. ammonium chloride and EtOAc and the resultant precipitate is isolated by filtration as the oxoacetyl product ZC(O)C(O)Y. Step A and Step B can be combined into a one pot reaction by adding the oxidant directly to the reaction pot after the completion of step A without isolating the α-cyanoketo intermediate ZC(O)CH(CN)Y.
  • Scheme B depicts a typical method for preparing the cyanomethyl piperazine or piperidine analogues utilized in scheme A. Two general literature references for some of the chemistry depicted in these initial schemes are Takahashi, K.; Shibasaki, K.; Ogura, K.; Iida, H.; Chem Lett. 1983, 859 or Yang; Z.; Zhang, Z.; Meanwell, N. A.; Kadow, J. F.; Wang, T.; Org. Lett. 2002, 4, 1103.
    Figure US20050124623A1-20050609-C00021
  • Step C. The secondary amine of a functionalized piperazine or piperidine can be alkylated with a haloacetonitrile under basic conditions to yield a cyanomethyl piperazine or piperidine analogue. In a typical procedure N-benzoyl piperazine was added to a solution of chloroacetonitrile and TEA in THF and stirred at r.t. for between 2 and 5 days. A resulting precipitate is removed by filtration, the filtrate is concentrated in vacuo, and the residue purified via chromatography to yield the cyanomethyl intermediate YCH2CN. The alkylation with haloacetonitrile can also be carried out with an alternate base, such as 4-methylmorpholine or diisopropylethyl amine.
  • The diazaindole carboxylic ester condensation partners Z-C(O)OR utilized in Scheme A can be prepared as shown in the following schemes:
  • One preferred method for preparing 4,6 diazaindole is shown in Scheme C.
    Figure US20050124623A1-20050609-C00022
  • Step D. The reaction of an (alkoxymethylene)cyanoacetate with an amino malonate under basic conditions is known to yield a 2,4-dicarboxylic ester-3-aminopyrrole. As shown in Scheme C, step D is carried out by reacting an amino malonate with an 2-alkoxy 1-cyano acrylate in the presence of a base such as sodium ethoxide. For a representive example see; Elliot, A. J.; Montgomery, J. A., and Walsh, D. A. Tetrahedron Lett, 1996, 37(25), 4339-4340. A typical procedure and conditions is is described in the experimental section.
  • Step E. The 3-Aminopyrrole 2-carboxylic ester resulting from step D can be cyclized to the desired 7-hydroxyl-4,6-diazaindole using a number of reagents including formamides, dialkyl acetal formamides, nitriles and formamidines. In a typical procedure 3-aminopyrrole-2,4-dicarboxylic acid diethyl ester and formidine acetate are heated at reflux in EtOH for 1 to 3 days. The reaction solution is filtered hot and the product usually crystallized upon cooling and is then rinsed with diethyl ether.
  • Step F. A 3-carboxylic ester 7-hydroxyl-4,6-diazaindole can then be converted to a 7-chloro analogue by treatment with a chlorinating reagent such as POCl3 or SOCl2. In a typical reaction procedure 3-ethylester-7-hydroxyl-4,6-diazaindole and POCl3 are combined and heated at 105° C. for between 3 and 5 h, cooled to r.t. and diluted with diethyl ether. The precipitate that forms is collected by filtration and was shown to be the 7-chloro-4,6-diazaindole. Alternatively, when greater reactivity is desired for further functionalization and for carrying out step G, the corresponding 7-bromo-4,6-diazindole may be prepared by substituting POBr3 for the chlorinating agents described above.
  • Step G. A 7-chloro-4,6-diazaindole can be displaced with a variety of nucleophiles to form the claimed R5 substituents or intermediates from which the claimed R5 substituents can be formed. Included in these are cyanide, alkoxides, amines, alcohols and various metallated species (cuprates, lithiates, zincates and Grignard reagents). In a typical procedure 3-ethylester-7-chloro-4,6-diazaindole and 3-methyl pyrazole in EtOH are heated at between 100° C. and 140° C. for 20 min to 1 h. Upon cooling the reaction is concentrated and purified by silica gel chromatography or by preparative HPLC. This step may also be carried out after the initial coupling and oxidation steps (steps A and B) have been preformed on the 3-ethylester-7-chloro-4,6-diazaindole intermediate. A cyano moiety could be introduced and converted to acids, esters, amides, imidates, or heteraromatics. Typical amide coupling methodology could be used to prepare amides from acids. It should also be noted that the halogen moiety may be carried through until compounds of the invention are realized and then the conditions described in Step G may frequently be used to prepare further compounds of the invention.
  • Alternatively, a 7-chloro or 7-bromo-4,6-diazaindole could be coupled to a heteroaryl stannane or boronic ester via Stille or Suzuki methodology respectively. Other metal catalyzed methodology such as copper mediated displacements could also be used to prepare N linked heteraromatic or heteroalicyclic derivatives. In general, substituted diazaindoles containing a chloride, bromide, iodide, triflate, or phosphonate should undergo coupling reactions with a boronate (Suzuki type reactions) or a stannane to provide substituted diazaindoles. Stannanes and boronates are prepared via standard literature procedures or as described in the experimental section of this application. The vinyl bromides, chlorides, triflates, or phosphonates may undergo metal mediated coupling to provide compounds of formula W-H. Stille or Suzuki couplings are particularly useful. A detailed discussion of the references and best conditions for these kinds of metal mediated coupling is described later in this application where the discussion is combined with a description of how these types of reactions may also be used to funtionalize diazaindoles. In additions, applications incorporated in their entirety elsewhere in this application contain methods for preparing heteroaryls from funtional groups appended to indoles and azaindoles. This methodology is also applicable to diazaindoles.
  • One potential method for preparing the 5,6-diazaindoles QCOOR is shown in Scheme D.
    Figure US20050124623A1-20050609-C00023
  • Step H. A TMS-isocyanide would be reacted with an acid fluoride in the presence of a dialkyl acetylene dicarboxylate to form a substituted pyrrole. For representative examples see: Livinghouse, T.; Smith, R.; J. Chem. Soc, Chem. Commun 1983, 5, 210. In a typical procedure, trimethylsilylmethyl isocyanide (generated from the lithiation of methyl isocyanide, followed by silylation with TMSCl) would be stirred with an aryl acid fluoride and dimethyl acetylenedicarboxylate in toluene at 80° C. After a standard workup a functionalized pyrrole where R1 is hydrogen and R5 is aryl would be realized.
  • Step I. A mixture of the keto-diester-pyrrole and hydrazine dihydrochloride in ethanol heated at reflux should result in the formation of the desired 4-hydroxyl-5,6-diazaindole. Alternatively, the keto-diester-pyrrole, hydrazine hydrate and a catalytic amount of p-toluenesulfonic acid could be heated to reflux in toluene or benzene in the presence of a Dean-Stark trap and upon dehydration, the desired 4-hydroxyl-5,6-diazaindole should form.
  • Step J, Step K and Step L. A 4-hydroxyl-5,6-diazaindole intermediate could be converted to the intermediates in which R3 is modified by direct functionalization of the hydroxyl group or by conversion of the hydroxyl group to a leaving group (halogen or triflate) followed by nucleophilic displacement or a metal (Pd or Cu) mediated coupling. These step(s) might also be carried out after the initial coupling and oxidation steps (steps A and B) have been preformed on the 4-hydroxyl-5,6-diazaindole intermediate. The conditions described for step G could also be utilized for this system.
    Figure US20050124623A1-20050609-C00024
    Figure US20050124623A1-20050609-C00025
  • As shown in Scheme DD pyrrole 2,3 di-carboxylic ethyl ester prepared as in either of the following two references: Roeder, Erhard; Wiedenfeld, Helmut; Bourauel, Thomas. Synthesis of ethyl 2,3-bis(ethoxycarbonyl)-1H-pyrrole-1-propionate. Liebigs Annalen der Chemie (1987), (12), 1117-19. and Swan, George A.; Waggott, A. Chemistry of melanins. VI. Syntheses of 3-carboxypyrrole-2-acetic acid, 3,5-dicarboxypyrrole-2-acetic acid, and related compounds. Journal of the Chemical Society [Section] C: Organic (1970), (2), 285-90. could be reacted with hydrazine in ethanol between RT and reflux to provide the cyclized product of step DD1. Reaction with phosphoryl chloride (2.2 to 5 equivalents should provide the dichloride as shown in step DD2. In step DD3, selective reaction of the C-7 chloride could occur by using benzyl alcohol and triethylamine in a cosolvent such as THF. In step DD4, the 4-chloro group might then be displaced with sodium or potassium methoxide in solvents such as methanol or toluene or a mixture. Stoichiometric copper I iodide could be added to speed slow reactions. In step DD5, selective hydrogenation of the benzyl group using 5 to 10% Pd/C in EtOH under a balloon pressure of hydrogen brovides the 7-hydroxy compound. Alternatively the benzyl group may be cleaved selectively with TMSI in acetonitrile at temperatures from 0 to 65° C. or using HBr in 1,2,dichloroethane at temps from −20 to 50° C. An alternate prep is to react the dichlorointemrediate above with methoxide rather than benzyl alcohol and then to selectivel cleave the C-7 ether using conditions described for the benzyl cleavage. Reacting the C-7 hydroxy group /amide tautomer with POCl3 or POBr3 would generate the chloride or bromide selectively which may be functionalized as described in step G of Scheme C for the 4,6-diazindoles. Step DD6 describes acylation of the functionalized intermediate and is done using the same procedures described in step O of Scheme F. Step DD7, amide copuling with piperazine or piperidine is carried out according to the general procedures described in Step P of Scheme F to provide compounds of the invention. It should be understood that the order of steps DD5-DD7 could be switched to determine which order provides best yields.
  • The 5,7-diazaindole could be prepared as shown in Scheme E. Intermediate M1 is a known compound whose synthesis has been described in the literature in the following references: Olsen, David B.; Lafemina, Robert L.; Eldrup, Anne B.; Bera, Sanjib. Methods of inhibiting orthopoxvirus replication with nucleoside compounds. PCT Int. Appl. (2003), 99 pp. WO 2003068244A1 Mekouar, Khalid; Deziel, Robert; Mounir, Samir; Iyer, Radhakrishnan P. Preparation of 7-deaza L-nucleosides as antiviral agents against the hepatitis B virus. PCT Int. Appl. (2003), WO 2003055896A2 Carroll, Steven S.; Lafemina, Robert L.; Hall, Dawn L.; Himmelberger, Amy L.; Kuo, Lawrence C.; Maccoss, Malcolm; Olsen, David B.; Rutkowski, Carrie A.; Tomassini, Joanne E.; An, Haoyun; Bhat, Balkrishen; Bhat, Neelima; Cook, Phillip Dan; Eldrup, Anne B.; Guinosso, Charles J.; Prhavc, Marija; Prakash, Thazha P. Preparation of nucleoside derivatives as inhibitors of RNA-dependent RNA viral polymerase. PCT Int. Appl. (2002), 235 pp. CODEN: PIXXD2 WO 2002057425A2 Carroll, Steven S.; Maccoss, Malcolm; Olsen, David B.; Bhat, Balkrishen; Bhat, Neelima; Cook, Phillip Dan; Eldrup, Anne B.; Prakash, Thazha P.; Prhavc, Marija; Song, Quanlai. Preparation of nucleoside derivatives as inhibitors of RNA-dependent RNA viral polymerase. PCT Int. Appl. (2002), WO 2002057287A2.
    Figure US20050124623A1-20050609-C00026
  • Intermediate M2 where R is ethyl is a known compound which could be prepared as described in the following three literature references:
    • Ugarkar, Bheemarao G.; DaRe, Jay M.; Kopcho, Joseph J.; Browne, Clinton E., III; Schanzer, Juergen M.; Wiesner, James B.; Erion, Mark D. Adenosine Kinase Inhibitors. 1. Synthesis, Enzyme Inhibition, and Anti-seizure Activity of 5-Iodotubercidin Analogues. Journal of Medicinal Chemistry (2000), 43(15), 2883-2893.
    • Firestein, Gary Steven; Ugarkar, Bheemarao Ganapatrao; Miller, Leonard Paul; Gruber, Harry Edward; Bullough, David Andrew; Erion, Mark David; Castellino, Angelo John. Preparation of adenosine kinase-inhibiting purine nucleoside analogs as antiinflammatory agents. PCT Int. Appl. WO 9417803A1.
    • Browne, Clinton E.; Ugarkar, Bheemarao G.; Mullane, Kevin M.; Gruber, Harry E.; Bullough, David A.; Erion, Mark D.; Castellino, Angelo. Adenosine kinase inhibitors. Eur. Pat. Appl. EP 496617A1.
  • Step N. Nucleophilic or metal catalyzed substitution of the 4-chloro-5,7-diazaindole will yield the R3 substituents of claim 1.
  • The diazaindole core may be coupled to the functionalized piperidine or piperazine through an oxoacetate or through an acylation/amidation process as shown in Scheme F.
    Figure US20050124623A1-20050609-C00027
  • Step O. Conversion of a specific 3H-diazaindole to the depicted ketoacid might be accomplished via several methods. Method a for step O: One successful method has been to use Fridel-Crafts acylation conditions mediated by an ionic liquid. In particular the ionic liquid 1-alkyl-3-alkylimidazolium chloroaluminate is generally useful in promoting the Friedel-Crafts type acylation and does work with some diazainoles. The ionic liquid is generated by mixing 1-alkyl-3-alkylimidazolium chloride with aluminium chloride at room temperature with vigorous stirring. 1:2 or 1:3 molar ratio of 1-alkyl-3-alkylimidazolium chloride to aluminium chloride is preferred. One particular useful imidazolium chloroaluminate for the acylation of diazaindoles with methyl or ethyl chlorooxoacetate would be the 1-ethyl-3-methylimidazolium chloroaluminate. The reaction would typically be performed at ambient temperature and the diazaindoleglyoxyl ester would be expected to be isolated. The resulting ester could then be hydrolyzed using the hydrolysis methods for Step O described below.
  • More conveniently, it is probable that the glyoxyl ester could be hydrolyzed in situ at ambient temperature upon prolonged reaction time (typically overnight) to give the corresponding glyoxyl acid which would be ready for amide formation.
  • A representative experimental procedure is as follows: 1-ethyl-3-methylimidazolium chloride (2 equiv.; purchased from TCI; weighted under a stream of nitrogen) would be stirred in an oven-dried round bottom flask at r.t. under a nitrogen atmosphere, and then aluminium chloride (6 equiv.; anhydrous powder packaged under argon in ampules purchased from Aldrich preferred would be added; after weighing under a stream of nitrogen). The mixture would be vigorously stirred to form a liquid, to which would then be added diazaindole (1 equiv.) followed by stirring until a homogenous mixture resulted. To the reaction mixture would then be added dropwise ethyl or methyl chlorooxoacetate (2 equiv.) and then stirring would be continued at r.t. for 2 to 24 h, probably approximately 16 h. After stirring was completed, the mixture an ice-water bath and the reaction would be quenched by carefully adding excess water. The precipitates would be filtered, washed with water and dried under high vacuum to give the diazaindoleglyoxylic acid. For some examples, 3 or even equivalents of 1-ethyl-3-methylimidazolium chloride and chlorooxoacetate may be required. A more comprehensive reference with analogous examples with non diazaindoles but with conditions that could be utilized with diazaindoles is contained in: Yeung, Kap-Sun; Farkas, Michelle E.; Qiu, Zhilei; Yang, Zhong. Friedel-Crafts acylation of indoles in acidic imidazolium chloroaluminate ionic liquid at room temperature. Tetrahedron Letters (2002), 43(33), 5793-5795.4 Related references: (1) Welton, T. Chem Rev. 1999, 99, 2071; (2) Surette, J. K. D.; Green, L.; Singer, R. D. Chem. Commun. 1996, 2753; (3) Saleh, R. Y. WO 00/15594.
  • Step O method B. The diazaindole could be treated with a Grignard reagent such as MeMgI (methyl magnesium iodide), methyl magnesium bromide or ethyl magnesium bromide and then a zinc halide, such as ZnCl2 (zinc chloride) or zinc bromide, followed by the addition of an oxalyl chloride mono ester, such as ClCOCOOMe (methyl chlorooxoacetate) or another ester as above, to afford the diaza-indole glyoxyl ester. Oxalic acid esters such as methyl oxalate, ethyl oxalate or as above are used. Aprotic solvents such as CH2Cl2, Et2O, benzene, toluene, DCE, THF, dioxane or the like could potentially be used alone or in combination for this sequence.
  • Step O method c: A Lewis acid catalyzed Friedel-Crafts reaction under standard conditions with an alkyl chloroacetoacetate might be utilized. This could be followed by in situ by hydrolysis of the ester my the method described below to form the diazaindole ketocarboxylic acid (cite previous patent(s)). Thus the diazindole ketoester precursors to the depicted acid could be prepared by reaction of diazaindoles with an excess of ClCOCOOMe in the presence of AlCl3 (aluminum chloride). Some further descriptions of the exact procedures to carry out this reaction but on indoles or azaindoles are contained in a) Zhang, Zhongxing; Yang, Zhong; Wong, Henry; Zhu, Juliang; Meanwell, Nicholas A.; Kadow, John F.; Wang, Tao. “An Effective Procedure for the Acylation of Azaindoles at C-3.” J. Org. Chem. 2002, 67(17), 6226-6227; b) Tao Wang et. al. U.S. Pat. No. 6,476,034 B2 “Antiviral Azaindole derivatives” published Nov. 5, 2002; c) W. Blair et al. PCT patent application WO 00/76521 A1 published Dec. 21,2000; d) O. Wallace et. al. PCT application WO 02/04440A1 published Jan. 17, 2002. Some reactions of 5-cyano-6-chloro-7-azaindoles and lactam-lactim tautomerism in 5-cyano-6-hydroxy-7-azaindolines. Khim. Geterotsikl. Soedin., 1987, 100-106). Typically an inert solvent such as CH2Cl2 would be used but others such as THF, Et2O, DCE, dioxane, benzene, or toluene may find applicability either alone or in mixtures. Other oxalate esters such as ethyl or benzyl mono esters of oxalic acid could also suffice for either method shown above. More lipophilic esters ease isolation during aqueous extractions. Lewis acid catalysts, such as tin tetrachloride, titanium IV chloride, and aluminum chloride could be employed with this transformation with aluminum chloride being most preferred.
  • Hydrolysis methods for Step O. Hydrolysis of a diazindole keto methyl ester would afford a potassium salt of the acid product shown as the product for Step O in Scheme F and this would then be ready for coupling with amines as shown in the next step. Acidification during workup, typically with aqueous HCl would provide the acid products from Step O as shown. Some typical conditions employ methanolic or ethanolic sodium hydroxide followed by careful acidification with aqueous hydrochloric acid of varying molarity but 1M HCl is preferred. The acidification is not utilized in many cases as described above for the preferred conditions. Lithium hydroxide or potassium hydroxide could also be employed and varying amounts of water could be added to the alcohols. Propanols or butanols could also be used as solvents. Elevated temperatures up to the boiling points of the solvents may be utilized if ambient temperatures do not suffice. Alternatively, the hydrolysis may be carried out in a non polar solvent such as CH2Cl2 or THF in the presence of Triton B. Temperatures of −78° C. to the boiling point of the solvent may be employed but −10° C. is preferred. Other conditions for ester hydrolysis are listed in reference 41 and both this reference and many of the conditions for ester hydrolysis are well known to chemists of average skill in the art.
  • Step P. The ketocarboxylic acid may be coupled with functionalized piperidines or piperazines using a number of standard amide bond or peptide bond forming coupling reagents. The acid intermediate Z-C(O)(O)OH from Scheme F could be coupled with either a substituted piperazine or piperidine, H—Y using standard amide bond or peptide bond forming coupling reagents. The combination of EDAC and triethylamine in tetrahydrofuran or BOPCl and diisopropyl ethyl amine in chloroform could be utilized but DEPBT, or other coupling reagents such as PyBop could be utilized. Another useful coupling condition employs HATU (L. A. Carpino et. al. J.Chem.Soc. Chem Comm. 1994, 201-203; A. Virgilio et.al. J.Am. Chem. Soc. 1994, 116,11580-11581). A general procedure for using this reagent is Acid (1 eq) and H—Y or H—W-Boc or HCl salt (2 eq) in DMF are stirred at rt for between 1 h and 2 days. HATU (2 eq) is added in one portion and then DMAP(3 eq). The reaction could be stirred at rt for 2 to 15 h (reaction progress monitored by standard methods ie TLC, LC/MS). The mixture is filtered through filter paper to collect the solid. The filtrate is concentrated and water is added. The mixture is filtered again and the solid is washed with water. The solid is conbined and washed with water. Many reagents for amide bond couplings are known by an organic chemist skilled in the art and nearly all of these are applicable for realizing coupled amide products.
  • DEPBT (3-(diethoxyphosphoryloxy)-1,2,3-benzotriazin-4(3H)-one) and N,N-diisopropylethylamine, commonly known as Hunig's base, represents another efficient method to form the amide bond (step P). DEPBT is either purchased from Adrich or prepared according to the procedure of Ref. 28, Li, H.; Jiang, X.; Ye, Y.-H.; Fan, C.; Romoff, T.; Goodman, M. Organic Lett., 1999, 1, 91-93. Typically an inert solvent such as DMF or THF is used but other aprotic solvents could be used.
  • The amide bond construction reaction could be carried out using the preferred conditions described above, the EDC conditions described below, other coupling conditions described in this application, or alternatively by applying the conditions or coupling reagents for amide bond construction described in incorporated applications for construction of substituents R2—R5 on indoles or azaindoles. Some specific nonlimiting examples are given in this application.
  • Alternatively, the acid could be converted to a methyl ester using excess diazomethane in THF/ether. The methyl ester in dry THF could be reacted with the lithium amide of intermediate H—Y. The lithium amide of H—Y, Li—Y is formed by reacting H—Y with lithium bistrimethylsilylamide in THF for 30 minutes in an ice water cooling bath. Sodium or potassium amides could be formed similarly and utilized if additional reactivity is desired. Other esters such as ethyl, phenyl, or pentafluorophenyl could be utilized and would be formed using standard methodology.
  • In addition, the acid can be converted to the acid chloride using oxalyl chloride in a solvent such as benzene or thionyl chloride either neat or containing a catalystic amount of DMF. Temperatures between 0° C. and reflux may be utilized depending on the substrate. Compounds of Formula I can be obtained from the resultant compounds of formula Z-C(O)(O)Cl by reaction with the appropriate H—Y in the presence of a tertiary amine (3-10 eq.) such as triethylamine or diisopropylethylamine in an anhydrous aprotic solvent such as dichloromethane, dichloroethane, diethyl ether, dioxane, THF, acetonitrile, DMF or the like at temperatures ranging from 0° C. to reflux. Most preferred are dichloromethane, dichloroethane, or THF. The reaction can be monitored by LC/MS. The 3H-diazaindoles may also be prepared under Bartoli or Liemgruber-Batchko reaction conditions as shown in schemeG. Conditions for carrying out these reactions were contained in the incorporated patent applications.
  • Note: For the purposes of brevity, the following symbol is taken to represent the following systems:
    Figure US20050124623A1-20050609-C00028
    Figure US20050124623A1-20050609-C00029
  • Step Q. Step Q in Scheme G depicts a potential synthesis of a diazaindole intermediate, via the well known Bartoli reaction in which vinyl magnesium bromide reacts with an aryl or heteroaryl nitro groups, to form a five-membered nitrogen containing ring as shown. Some references for the above transformation to form an indole ring include: Bartoli et al. a) Tetrahedron Lett. 1989, 30, 2129. b) J. Chem. Soc. Perkin Trans. 1 1991, 2757. c) J. Chem. Soc. Perkin Trans. II 1991, 657. d) SynLett (1999), 1594. In the preferred procedure, which could be applied to diazaindole synthesis, a solution of vinyl Magnesium bromide in THF (typically 1.0M but from 0.25 to 3.0M) is added dropwise to a solution of the nitro pyridine in THF at −78° under an inert atmosphere of either nitrogen or Argon. After addition is completed, the reaction temperature is allowed to warm to −20° and then is stirred for approximately 12 h before quenching with 20% aq ammonium chloride solution. The reaction is extracted with ethyl acetate and then worked up in a typical manner using a drying agent such as anhydrous magnesium sulfate or sodium sulfate. Products are generally purified using chromatography over Silica gel. Best results are generally achieved using freshly prepared vinyl Magnesium bromide. In some cases, vinyl Magnesium chloride may be substituted for vinyl Magnesium bromide.
  • Step R. Reaction with dimethylformamide dimethyl acetal in an inert solvent or neat under conditions for forming Batcho-Leimgruber precursors would provide the cyclization precursor, 33, as shown. A typical condition would employ 20% DMF dimethyl acetal in DMF heated to 105-110 degrees C. Although the step is anticipated to work as shown, the pyridine may be oxidized to the N-oxide prior to the reaction using a peracid such as MCPBA or a more potent oxidant like meta-trifluoromethyl or meta nitro peroxy benzoic acids.
  • Step S. Reduction of the nitro group using for example hydrogenation over Pt on/C catalyst in a solvent such as MeOH, EtOH, or EtOAc could provide the cyclized product. Generally only a slight positive pressure of hydrogen would be required (a stream) but higher pressures may be needed (1.5 atm). Alternatively the reduction may be carried out using tin dichloride and HCl, hydrogenation over Raney nickel or other catalysts, or by using other methods for nitro reduction such as described elsewhere in this application.
  • Another possible method for preparation of 5,6-diazaindoles is shown in scheme H.
    Figure US20050124623A1-20050609-C00030
  • Step T. 1,2,3,4-Tetrazines have been shown to react with pyrrole and substituted pyrroles to form 5,6-diazaindole products. This reaction proceeds through a [4+2]-cycloaddition followed by a retro-[4+2]-cycloaddition to release nitrogen gas and a subsequent oxidation to establish aromaticity. For representative examples see: Seitz, Z.; Kaempchen, T.; Arch. Pharm. 1978, 311, 728. Takahashi, M; Ishida, H.; Kohmoto, M. Bull. Chem. Soc. Japan 1976, 49, 1725. Benson, S. C.; Palabrica, C. A.; Snyder, J. K. J. Org. Chem. 1987, 52, 4610. Gonzalez, J. C.; Lobo-Antunes, J; Perez-Lourido, P.; Santana, L.; Uriate, E. Synthesis 2002, 4, 475-478.
  • Another possible method for preparing a 5,6-diazaindole with a C-3 oxoacetate is shown in Scheme I (Cook, P. D.; Castle, R. N. J. Het. Chem. 1973, 10, 551.
    Figure US20050124623A1-20050609-C00031
  • Step U. The starting pyridazine N-oxide would initially be nitrated and the resulting nitro group then would be reduced under standard conditions to an amine. The chloro would then be removed under hydrogentation conditions. Alternatively, the chloro could remain in the molecule and be carried through the subsequent steps. This should allow for the formation of a 4-chloro-5,6-diazaindole. The chloro could then be converted to a methoxy or an amino group by nucleophilic displacement or copper catalyzed assisted coupling. This would result in an intermediate that could be converted to molecules claimed within this application via previously described amide bond coupling.
  • Step V The amine could then be functionalized with ethyl orthoformate under acidic conditions to form an ethoxyimine. In a typical procedure the amine and triethyl orthoformate were dissolved into a solution of DMF and ethanol that had been adjusted to pH 1 with anhydrous hydrogen chloride. The reaction was then heated to 150-160° C. and ethanol was collected by distillation resulting in the formation of the desired ethoxyimine.
  • Step W Deprotonation of the methyl group followed by acylation with diethyl oxalate would yield a ketoester intermediate that could be used to form a 3-oxoacetate-5,6-diazaindole (Step X) or could be used to make a 2-carboxylate-5,6-diazaindole by hydrolysis of the imine, followed by condensation of the amine onto the ketone five centers away.
  • Step X. The ketoester could then be cyclized onto the ethoxyimine under basic conditions to arrive at the 3-oxoacetate-5,6-diazaindole. To form the molecules of this claim, functionalized piperazine or piperidones could be coupled to the ester through standard amide bond forming reactions. This general scheme should also allow for the preparation of other 5,6-diazaindole intermediates with different R3 and R5 substituents by displacement or coupling to the chloro or displacement of the methoxy at some point in the sequence.
  • Preparations of Functionalized Piperazines and Piperidines are Described later in the Application.
  • Figure US20050124623A1-20050609-C00032
  • As shown above in Scheme 15, Step F15, substituted diazaindoles containing a chloride, bromide, iodide, triflate, or phosphonate could undergo coupling reactions with a boronate (Suzuki type reactions) or a stannane to provide substituted diazaindoles. Stannanes and boronates are prepared via standard literature procedures or as described in the experimental section of this application. The substitututed diazindoles may undergo metal mediated coupling to provide compounds of Formula I wherein R4 is aryl, heteroaryl, or heteroalicyclic for example. The bromo or chloro diazaindole intermediates, (or diazaindole triflates or iodides) may undergo Stille-type coupling with heteroarylstannanes as shown in Scheme 15. Conditions for this reaction are well known in the art and the following are three example references a) Farina, V.; Roth, G. P. Recent advances in the Stille reaction; Adv. Met.-Org. Chem. 1996, 5, 1-53. b) Farina, V.; Krishnamurthy, V.; Scott, W. J. The Stille reaction; Org. React. (N.Y.) 1997, 50, 1-652. and c) Stille, J. K. Angew. Chem. Int. Ed. Engl. 1986, 25, 508-524. Other references for general coupling conditions are also in the reference by Richard C. Larock Comprehensive Organic Transformations 2nd Ed. 1999, John Wiley and Sons New York. All of these references provide numerous conditions at the disposal of those skilled in the art in addition to the specific examples provided in Scheme 15 and in the specific embodiments. It can be well recognized that an diazaindole stannane could also couple to a heterocyclic or aryl halide or triflate to construct compounds of Formula I. Suzuki coupling (Norio Miyaura and Akiro Suzuki Chem Rev. 1995, 95, 2457.) between a triflate, bromo, or chloro diazaindole intermediate and a suitable boronate could also be employed and some specific examples are contained in this application. Palladium catalyzed couplings of stannanes and boronates between chloro diazaindole intermediates are also feasible and have been utilized extensively for this invention. Preferred procedures for coupling of a chloro diazaindole and a stannane employ dioxane, stoichiometric or an excess of the tin reagent (up to 5 equivalents), 0.1 to 1 eq of Palladium (0) tetrakis triphenyl phosphine in dioxane heated for 5 to 15 h at 110 to 120°. Other solvents such as DMF, THF, toluene, or benzene could be employed. Preferred procedures for Suzuki coupling of a chloro diazaindole and a boronate employ 1:1 DMF water as solvent, 2 equivalents of potassium carbonate as base stoichiometric or an excess of the boron reagent (up to 5 equivalents), 0.1 to 1 eq of Palladium (O) tetrakis triphenyl phosphine heated for 5 to 15 h at 110 to 120°. Some references (and the references therein) describing catalysts which are useful for coupling with aryl and heteroaryl chlorides are:
    • Littke, A. F.; Dai, C.; Fu, G. C. J. Am. Chem. Soc. 2000, 122(17), 4020-4028;
    • Varma, R. S.; Naicker, K. P. Tetrahedron Lett. 1999, 40(3), 439-442; Wallow, T. I.;
    • Novak, B. M. J. Org. Chem. 1994, 59(17), 5034-7; Buchwald, S.; Old, D. W.;
    • Wolfe, J. P.; Palucki, M.; Kamikawa, K.; Chieffi, A.; Sadighi, J. P.; Singer, R. A.;
    • Ahman, J. PCT Int. Appl. WO 0002887 2000; Wolfe, J. P.; Buchwald, S. L. Angew. Chem., Int. Ed. 1999, 38(23), 3415; Wolfe, J. P.; Singer, R. A.; Yang, B. H.;
    • Buchwald, S. L. J. Am. Chem. Soc. 1999, 121(41), 9550-9561; Wolfe, J. P.;
    • Buchwald, S. L. Angew. Chem., Int. Ed. 1999, 38(16), 2413-2416; Bracher, F.;
    • Hildebrand, D.; Liebigs Ann. Chem. 1992, 12, 1315-1319; and Bracher, F.;
    • Hildebrand, D.; Liebigs Ann. Chem. 1993, 8, 837-839.
  • Alternatively, the boronate or stannane could potentially be formed on the diazaindole via methods known in the art and the coupling performed in the reverse manner with aryl or heteroaryl based halogens or triflates.
  • Known boronate or stannane agents could be either purchased from commercial resources or prepared following disclosed documents. Additional examples for the preparation of tin reagents or boronate reagents are contained in the experimental section.
  • Novel stannane agents could be prepared from one of the following routes.
    Figure US20050124623A1-20050609-C00033
    Figure US20050124623A1-20050609-C00034
    Figure US20050124623A1-20050609-C00035
    Figure US20050124623A1-20050609-C00036
    Figure US20050124623A1-20050609-C00037
  • Boronate reagents are prepared as described in reference 71. Reaction of lithium or Grignard reagents with trialkyl borates generates boronates. Alternatively, Palladium catalyzed couplings of alkoxy diboron or alkyl diboron reagents with aryl or heteroaryl halides can provide boron reagents for use in Suzuki type couplings. Some example conditions for coupling a halide with (MeO)BB(OMe)2 utilize PdCl2 (dppf), KOAc, DMSO, at 80° C. until reaction is complete when followed by TLC or HPLC analysis.
  • Related examples are provided in the following experimental section.
  • Methods for direct addition of aryl or heteroaryl organometallic reagents to alpha chloro nitrogen containing heterocyles or the N-oxides of nitrogen containing heterocycles are known and applicable to the diazaindoles. Some examples are Shiotani et. Al. J. Heterocyclic Chem. 1997, 34(3), 901-907; Fourmigue et.al. J.Org. Chem. 1991, 56(16), 4858-4864.
    Figure US20050124623A1-20050609-C00038
    Figure US20050124623A1-20050609-C00039
  • Direct displacements to install amine or N linked heteroaryl substituents could also be used to prepare compounds of Formula I. As shown in Schemes 15aa and 15bb, a mixture of halo-diazaindole intermediate, 1-2 equivalents of copper powder; 1-2 equivalents of potassium carbonate, and a 2-30 equivalents of the corresponding heterocyclic reagent, with 10 equivalents preferred; was heated at 135-160° C. for 4 to 9 hours. The reaction mixture was cooled to room temperature and filtered through filter paper. The filtrate was diluted with methanol and purified either by preparative HPLC or silica gel. In many cases no chromatography is necessary, the product can be obtained by crystallization with methanol.
  • Alternatively, the installation of amines or N linked heteroaryls could be carried out by heating 1 to 40 equivalents of the appropriate amine and an equivalent of the appropriate diazaindole chloride, bromide or iodide with copper bronze (from 0.1 to 10 equivalents (preferably about 2 equivalents) and from 1 to 10 equivalents of finely pulverized potassium hydroxide (preferably about 2 equivalents). Temperatures of 120° to 200° might be employed with 140-160° generally preferred. For volatile starting materials a sealed reactor may be employed. The reaction would most often be applicable when the halogen being displaced is at the 7-position of a diazaindole but the method could work when the halogen is at a different position (4-7 position possible). As shown above the reaction could be employed on diazaindoles unsubstituted at position 3 or intermediates which contain the dicarbonyl or the intact dicarbonyl piperazine urea or thioureas contained in compounds of formula I.
    Figure US20050124623A1-20050609-C00040
  • A possible preparation of a key aldehyde intermediate, 43, using a procedure adapted from the method of Gilmore et. Al. Synlett 1992, 79-80 is shown in Scheme 16 above. The aldehyde substituent is shown only at one position for the sake of clarity, and should not be considered as a limitation of the methodology. The bromide or iodide intermediate would be converted into an aldehyde intermediate, 43, by metal-halogen exchange and subsequent reaction with dimethylformamide in an appropriate aprotic solvent. Typical bases used could include, but would not be limited to, alkyl lithium bases such as n-butyl lithium, sec butyl lithium or tert butyl lithium or a metal such as lithium metal. A preferred aprotic solvent is THF. Typically the transmetallation would be initiated at −78° C. and allowed to react with dimethylformamide (allowing the reaction to warm may be required to enable complete reaction) to provide an aldehyde which is elaborated to compounds of Formula I. Other methods for introduction of an aldehyde group to form intermediates of formula 43 include transition metal catalyzed carbonylation reactions of suitable bromo, trifluoromethane sulfonyl, or stannyl diazaindoles.
  • As shown in Scheme 52, the pieces HW—R18 can be prepared by a number of different methods. One useful way is by reacting a mono protected piperazine with a heteroaryl chloride, bromide, iodide, or triflate. This reaction is typically carried out at elevated temperature (50 to 250 degrees celsius) in a solvent such as ethylene glycol, DME, dioxane, NMP, or DMF. A tertiary amine base such as triethyl amide or diisopropyl ethyl amine is typically employed and usually 2 to 4 equivalents are employed. At least 2 equivalents are used if a salt of HW R18 is utilized. The piperazine is typically monoprotected with a BOC group since this material is commercially available. Removal of the Boc group is typically done using HCl (typically 1 to 6N) in dioxane to provide the HCl salt. TFA may also be used to generate the TFA salt. Alternatively, the conditions for coupling heterocycles using copper catalysis discussed earlier in Scheme 12 may be used to couple W to R18 via displacement of X in X—R18. Alternatively Palladium catalysis in the presence of a bidentate catalyst via the procedures of Buchwald or the use of a ferrocenyl catalyst via the methods of Hartwig could be used to couple the piperazine to the heteroaryl (R18).
    Figure US20050124623A1-20050609-C00041
    Figure US20050124623A1-20050609-C00042
  • Scheme 53 describes how a protected piperazine can be coupled to Q-COOH via standard methodology. Conditions for removal of the amine protecting group which could be tBoc or other groups is protecting group specific. As shown in Scheme 53 where tBoc is the preferred protecting group used to exemplify the strategy, standard conditions for removal such as TFA in dichloromethane or alternatively aqueous HCl can provide the free amine. The free amine is coupled to heteraromatic R18 using the conditions described in Scheme 52 for step F″″.
  • General Schemes:
  • Scheme D1 describes a possible method for preparing the compounds described by H—W where Y is as defined in the description and claims of the invention. Typically, this methodology will work best when D is a group which lowers the PKA of the hydrogens on the adjacecent methylene moiety. For example cyano, sulfonyl, amido and the like as specified in the claim. A preferably could be aryl or heteroaryl moieties as described in claim 1. A could also be other groups described in claim 1. Alkoxide bases of C1 to C4 alcohols can be utilzed but other bases such as lithium, sodium, or potassium dialkyl amides or the corresponding bistrimethylsilyl amides could also be utilized.
  • Preparation of Intermediates:
  • Figure US20050124623A1-20050609-C00043

    Note as shown in Scheme D1, the piperazine or piperidine moiety of Y may be substituted as defined by the invention. In the interest of clarity, unsubstituted piperidines and piperazines are used in the Schemes to keep them readable. It is understood substituents could be incorporated.
    Figure US20050124623A1-20050609-C00044
  • As shown in Scheme E1, addition of an organometallic reagent to a ketone can provide an intermediate tertiary alkoxide which undergoes protonation and acid catalyzed elimination to form the desired double bond. A number of organo metallic reagents could suffice as shown but an extra equivalent (at least two total) could be needed to compensate for deprotection of the amine nitrogen in many cases.
    Figure US20050124623A1-20050609-C00045
  • Standard olefination conditions such as Wittig, Horner Emmons, Petersen or Arsenic based could be used to convert the ketone to the desired products. Some general reviews of this methodology and directions for use are contained in the following references: Wadsworth, W. S, Jr., in “Organic Reactions”, Dauben, W. G., Ed., Wiley, New York, 1977, 25, 73. McMurry, J. E. Acct. Chem. Res. 1983, 16, 405. Cushman, M., et al. Bioorg. Med. Chem. 2002, 10, 2807. When Z=triphenyl phosphine, butyl lithium or LDA could be used to generate the phosphorus ylide in THF and then the ylide reacted with the ketone ot provide the desired product. The phosphinate or phosphine oxide based reagents could be used with similar bases or with sodium or postassium methoxide or ethoxide in the corresponding alcohol solvents.
    Figure US20050124623A1-20050609-C00046
    Figure US20050124623A1-20050609-C00047
  • As shown above in Scheme H1, a chloride, bromide, iodide, triflate, or phosphonate undergo coupling reactions with a boronate (Suzuki type reactions). Stannanes and boronates are prepared via standard literature procedures or as described in the experimental section of this application. The vinyl bromides, chlorides, triflates, or phosphonates may undergo metal mediated coupling to provide compounds of formula W—H. Stille or Suzuki couplings are particularly useful. A detailed discussion of the references and best conditions for these kinds of metal mediated coupling is described later in this application where the discussion is combined with a description of how these types of reactions may aslo be used to funtionalize diazaindoles.
  • When Ar is benzene, starting materials are commerially available
    Figure US20050124623A1-20050609-C00048
  • Alternatively, the compounds Y—H could potentially be prepared via olefin metathesis using highly active Rhodium catalysts. The methylene starting material can be prepared via simple Wittig methylenation of the precursor ketone which is prepared via literature methods. The olefin metathesis is preferably carried out using 1% of the imadazoylidene ruthenium benzylidene catalyst described in the following reference. The reaction is carried out starting at low temperatures (−40°) or similar. Starting methylene material is mixed with excess olefin (5 to 100 equivalents) and the reaction is warmed to 40° C. Synthesis of Symmetrical Trisubstituted Olefins by Cross Metathesis. Chatterjee, Arnab K.; Sanders, Daniel P.; Grubbs, Robert H. Organic Letters ACS ASAP.
  • Additional references are listed below which show additional conditions and substrates which could be used with this catalyst.
  • Functional group diversity by ruthenium-catalyzed olefin cross-metathesis. Toste, F. Dean; Chatterjee, Arnab K.; Grubbs, Robert H. The Arnold and Mabel Beckman Laboratory of Chemical Synthesis, Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, Calif., USA. Pure and Applied Chemistry (2002), 74(1), 7-10. A Versatile Precursor for the Synthesis of New Ruthenium Olefin Metathesis Catalysts. Sanford, Melanie S.; Love, Jennifer A.; Grubbs, Robert H.. Arnold and Mabel Beckman Laboratories for Chemical Synthesis Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, Calif., USA. Organometallics (2001), 20(25), 5314-5318. Olefin metathesis with 1,1-difluoroethylene. Trnka, Tina M.; Day, Michael W.; Grubbs, Robert H.. Arnold and Mabef Beckman Lab. of Chemical Synthesis, California Institute of Technology, Pasadena, Calif., USA. Angewandte Chemie, International Edition (2001), 40(18), 3441-3444.
  • Scheme K1 shows a sequence in which a piperidone is coverted to a monofuntionalized olefin via Wittig olefination. Bromination and dehydrobromination provides a versatile vinyl bromide intermediate. This intermediate is coupled to the QC(O)C(O)OH acid with BOPCl to provide a compound of formula I. This intermediate is then functionalized using palladium mediated couplings to either boronates or stannanes. Conditions for these couplings are described in this application.
    Figure US20050124623A1-20050609-C00049
  • Scheme L1 shows specific examples of general Scheme K1 which are some of those described in the experimental section.
    Figure US20050124623A1-20050609-C00050
  • Scheme M1 shows how a protected vinyl bromide can be converted to a carboxylic acid via lithium bromide exchange and reaction with carbon dioxide. As described in this application and the incorporated ones, carboxylic acids are excellent precursors to many heterocyles or amides. The rest of Scheme M1 shows conversion to funtionalized oxadiazoles. Other chemistry described in this application depicts other methods for converting acids to groups of other compounds of the invention.
    Figure US20050124623A1-20050609-C00051
  • Scheme N1 depicts a more specific example of Scheme M1.
    Figure US20050124623A1-20050609-C00052
  • Scheme P depicts methods for functionalizing the vinyl bromide to install groups D (or A). Either a modified Stille coupling or a zinc mediated coupling are depicted. Details of these tranformations are discussed later in the section on metal couplings.
    Figure US20050124623A1-20050609-C00053
  • Scheme Q depicts some specific examples of Scheme P.
    Figure US20050124623A1-20050609-C00054
  • Scheme R depicts methods for functionalizing the vinyl bromide to install groups D (or A). Either a modified Stille coupling, zinc mediated coupling, or a Suzuki boronic acid coupling are depicted. A method for converting the vinyl bromide to vinyl idodide is shown. If the vinyl bromide fails to undergo efficient reaction, the more reactive iodide can be prepared as a better partner. Details of these tranformations are discussed later in the section on metal couplings.
    Figure US20050124623A1-20050609-C00055
  • Scheme S provides specific examples of Scheme R.
    Figure US20050124623A1-20050609-C00056
  • Scheme T shows methods for converting the vinyl bromide into more funtionalized groups D (or A). A key aldehyde intermediate is generated from the vinyl bromide and can be used to generate heteroaryls such as the oxazole via reaction with Tosmic.
    Figure US20050124623A1-20050609-C00057
  • Scheme U shows how a hydrazide (gnerated from the acid) can be used to prepare oxadiazoles with diffferent substituents.
    Figure US20050124623A1-20050609-C00058
  • Scheme V provides more specific examples of Scheme U.
    Figure US20050124623A1-20050609-C00059
  • Scheme W shows some other methods for installing D (or A).
    Figure US20050124623A1-20050609-C00060
  • Scheme X shows a particular example where a functionalized heteroaryl or in this case aryl are coupled and then further functionalization can occurr (in this case redcution of an ester to an alcohol).
    Figure US20050124623A1-20050609-C00061
  • Scheme Y provides more specific examples of Scheme X.
    Figure US20050124623A1-20050609-C00062
  • Procedures for coupling piperazine amides to oxoacetyl derivatives are described in the Blair, Wang, Wallace, or Wang references 93-95 and 106 respectively. In addition, these applications describe preparations of heteroaryls and methods for funtionalizing heteraromatic systems in the presence of oxoacetyl amides. The entire disclosures in U.S. Pat. No. 6,469,006 granted Oct. 22, 2002; U.S. Pat. No. 6,476,034 granted Nov. 5, 2002; U.S. patent application Ser. No. 10/027,612 filed Dec. 19, 2001, which is a continuation-in-part of U.S. Ser. No. 09/888,686 filed Jun. 25, 2001 (corresponding to PCT WO 02/04440, published Jan. 17, 2002); and U.S. patent application Ser. No. 10/214,982 filed Aug. 7, 2002, which is a continuation-in-part of U.S. Ser. No. 10/038,306 filed Jan. 2, 2002 (corresponding to PCT WO 02/62423 published Aug. 15, 2002) are incorporated by reference herein. The procedures used to couple diazaindole oxoacetic acids to piperazine amides in these references could potentially be used analogously to form the compounds of this invention except the H—Y are used in place of the piperazine benzamides.
    Figure US20050124623A1-20050609-C00063
  • Scheme 54a depict a general method suitable for the synthesis of many of the compounds of formula I. As shown in these schemes, a suitable protected piperazine derivative, PG-YH, of Formula VI, (wherein PG is an appropriate amine protecting group) is acylated with an appropriate acylating agent, R18C(O)L, (wherein L is a suitable leaving group) to provide the protected acylated piperazine derivative of Formula V. Compound V is then deprotected using standard methods to provide the acylated piperazine derivative of Formula IV. For example, when PG represents tertiary-butoxycarbonyl the compound of Formula V can be deprotected to provide a compound of Formula IV by treatment with a strong acid, such as neat cold trifluoroacetic acid or aqueous hydrochloric acid, in an appropriate solvent such as dichloromethane. Alternatively, when PG represents benzyl the deprotection may be effected by hydrogenation.
  • Examples containing substituted piperazines are prepared using the general procedures outlined in Schemes 55-38. Substituted piperazines are either commercially available from Aldrich, Co. or prepared according to literature procedures (Behun et al, Ref. 88(a), Scheme 31, eq. 01). Hydrogenation of alkyl substituted pyrazines under 40 to 50 psi pressure in EtOH afforded substituted piperazines. When the substituent was an ester or amide, the pyrazine systems could be partially reduced to the tetrahydropyrazine (Rossen et al, Ref. 88(b), Scheme 55, eq. 02). The carbonyl substituted piperazines could be obtained under the same conditions described above by using commercially available dibenzyl piperazines (Scheme 55, eq. 03).
    Figure US20050124623A1-20050609-C00064
  • Mono-benzoylation of symmetric substituted piperazines could be achieved by using one of the following procedures (Scheme 57). (a) Treatment of a solution of piperazine in acetic acid with acetyl chloride afforded the desired mon-benzoylated piperazine (Desai et al. Ref. 27, Scheme 57, eq. 04). (b) Symmetric piperazines were treated with 2 equivalents of n-butyllithium, followed by the addition of benzoyl chloride at room temperature (Wang et al, Ref. 89, Scheme 57, eq. 05).
    Figure US20050124623A1-20050609-C00065
  • Mono-benzoylation of unsymmetric substituted piperazines could be achieved by using one of the following procedures (Scheme 57), in which all the methods were exemplified by mono-alkyl substituted piperazines. (a) Unsymmetric piperazines were treated with 2 equivalents of n-butyllithium, followed by the addition of benzoyl chloride at room temperature to afford a mixture of two regioisomers, which could be separated by chromatography (Wang et al, Ref. 89 and 90(b), Scheme 58 eq. 06); (b) Benzoic acid was converted to its pentafluorophenyl ester, and then further reaction with 2-alkylpiperazine to provide the mono-benzoylpiperazines with the benzoyl group at the less hindered nitrogen (Adamczyk et al, Ref. 90(a), Scheme 58, eq. 07); (c) A mixture of piperazine and methyl benzoate was treated with dialkylaluminum chloride in methylene chloride for 2-4 days to yield the mono-benzoylpiperazine with the benzoyl group at the less hindered nitrogen (Scheme 58 eq. 08); (d) Unsymmetric piperazines were treated with 2 equivalents of n-butyllithium, followed by subsequent addition of triethylsilyl chloride and benzoyl chloride in THF at room temperature to afford mono-benzoylpiperazines with the benzoyl group at the more hindered nitrogen (Wang et al, Ref. 90(b), Scheme 58, eq. 09).
    Figure US20050124623A1-20050609-C00066
  • Piperazine intermediates could be prepared using standard chemistry as shown in Scheme 64.
    Figure US20050124623A1-20050609-C00067
    Figure US20050124623A1-20050609-C00068
  • Steps a16, a17, and a18 encompasses reactions and conditions for 10, 20 and 30 amide bond formation as shown in Schemes 65 which provide compounds such as those of Formula 73. Compounds of formula 73 represent intermediates for the preparation of Compounds I or compounds I depending on the identity of T and Y.
  • The reaction conditions for the formation of amide bonds encompass any reagents that generate a reactive intermediate for activation of the carboxylic acid to amide formation, for example (but not limited to), acyl halide, from carbodiimide, acyl iminium salt, symmetrical anhydrides, mixed anhydrides (including phosphonic/phosphinic mixed anhydrides), active esters (including silyl ester, methyl ester and thioester), acyl carbonate, acyl azide, acyl sulfonate and acyloxy N-phosphonium salt. The reaction of the diazaindole carboxylic acids with amines to form amides may be mediated by standard amide bond forming conditions described in the art. Some examples for amide bond formation are listed in references 41-53 but this list is not limiting. Some carboxylic acid to amine coupling reagents which are applicable are EDC, Diisopropylcarbodiimide or other carbodiimides, PyBop (benzotriazolyloxytris(dimethylamino) phosphonium hexafluorophosphate), 2-(1H-benzotriazole-1-yl)-1,1,3,3-tetramethyl uronium hexafluorophosphate (HBTU). A particularly useful method for azaindole 7-carboxylic acid to amide reactions is the use of carbonyl imidazole as the coupling reagent as described in reference 53. The temperature of this reaction may be lower than in the cited reference, from 80° C. (or possibly lower) to 150° C. or higher. An example of more specific conditions which are likely to be successful are depicted in Scheme 66.
    Figure US20050124623A1-20050609-C00069
  • The following four general methods provide a more detailed description of procedures potentially useful for the preparation of diazindoleindolecarboxamides and these methods could potentially be employed for the synthesis of intermediates 73 useful for the preparation of compounds I or for the preparation of compounds of Formula I themselves.
  • Method 1:
  • To a mixture of an acid intermediate, such as 74, (1 equiv., 0.48 mmol), an appropriate amine (4 equiv.) and DMAP (58 mg, 0.47 mmol) dissolved CH2Cl2 (1 mL) should be added EDC (90 mg, 0.47 mmol). The resulting mixture should be shaken at rt for 12 h, and then evaporated in vacuo. The residue could be dissolved in MeOH, and subjected to preparative reverse phase HPLC purification.
  • Method 2:
  • To a mixture of an appropriate amine (4 equiv.) and HOBT (16 mg, 0.12 mmol) in THF (0.5 mL) could be added an acid intermediate, such as 74, (25 mg, 0.06 mmol) and NMM (50 μl, 0.45 mmol), followed by EDC (23 mg, 0.12 mmol). The reaction mixture could be shaken at rt for 12 h. The volatiles could be evaporated in vacuo; and the residue dissolved in MeOH and subjected to preparative reverse phase HPLC purification.
  • Method 3:
  • To a mixture of an acid intermediate, such as 74, (0.047 mmol), amine (4 equiv.) and DEPBT (prepared according to Li, H.; Jiang, X. Ye, Y.; Fan, C.; Todd, R.; Goodman, M. Organic Letters 1999, 1, 91; 21 mg, 0.071 mmol) in DMF (0.5 mL) could be added TEA (0.03 mL, 0.22 mmol). The resulting mixture could be shaken at rt for 12 h; and then diluted with MeOH (2 mL) and purified by preparative reverse phase HPLC.
  • Method 4:
  • A mixture of an acid intermediate, such as 74, (0.047 mmol) and 8.5 mg (0.052 mmol) of 1,1-carbonyldiimidazole in anhydrous THF (2 mL) could be heated to reflux under nitrogen. After 2.5 h, 0.052 mmol of amine could be added and heating continued. After an additional period of 3˜20 h at reflux, the reaction mixture could be cooled and concentrated in vacuo. The residue could be purified by chromatography on silica gel to provide a compound of Formula I.
  • In addition, the carboxylic acid could be converted to an acid chloride using reagents such as thionyl chloride (neat or in an inert solvent) or oxalyl chloride in a solvent such as benzene, toluene, THF, or CH2Cl2. The amides could alternatively, be formed by reaction of the acid chloride with an excess of ammonia, primary, or secondary amine in an inert solvent such as benzene, toluene, THF, or CH2Cl2 or with stoichiometric amounts of amines in the presence of a tertiary amine such as triethylamine or a base such as pyridine or 2,6-lutidine. Alternatively, the acid chloride could be reacted with an amine under basic conditions (usually sodium or potassium hydroxide) in solvent mixtures containing water and possibly a miscible co solvent such as dioxane or THF. Scheme 25B depicts a typical preparation of an acid chloride and derivatization to an amide of Formula I. Additionally, the carboxylic acid could be converted to an ester preferably a methyl or ethyl ester and then reacted with an amine. The ester could be formed by reaction with diazomethane or alternatively trimethylsilyl diazomethane using standard conditions which are well known in the art. References and procedures for using these or other ester forming reactions can be found in reference 52 or 54.
  • Additional references for the formation of amides from acids are: Norman, M. H.; Navas, F. III; Thompson, J. B.; Rigdon, G. C.; J. Med. Chem. 1996, 39(24), 4692-4703; Hong, F.; Pang, Y.-P.; Cusack, B.; Richelson, E.; J. Chem. Soc., Perkin Trans 1 1997, 14, 2083-2088; Langry, K. C.; Org. Prep. Proc. Int. 1994, 26(4), 429-438; Romero, D. L.; Morge, R. A.; Biles, C.; Berrios-Pena, N.; May, P. D.; Palmer, J. R.; Johnson, P. D.; Smith, H. W.; Busso, M.; Tan, C.-K.; Voorman, R. L.; Reusser, F.; Althaus, I. W.; Downey, K. M.; et al.; J. Med. Chem. 1994, 37(7), 999-1014; Bhattacharjee, A.; Mukhopadhyay, R.; Bhattacharjya, A.; Indian J. Chem., Sect B 1994, 33(7), 679-682.
    Figure US20050124623A1-20050609-C00070
  • Scheme 67 shows possible synthetic transformations on a chloro diazazaindole. Step F-1 of Scheme 31 could be carried out according to the following procedures: Yamaguchi, S.; Yoshida, M.; Miyajima, I.; Araki, T.; Hirai, Y.; J. Heterocycl. Chem. 1995, 32(5), 1517-1519 in which they use 1 eq of Chloride, 1.9 eq Cu(I)CN, in dry DMF and reflux for 48 h. The concentration of chloride in DMF is preferably 0.094 mmol per mL of solvent. Reaction times of 1-48 h may be approrpiate depending on substrate and reaction temperatures between 80° C. and reflux (156° C.) may be employed. An alternate procedure for carrying out step F-1 as described in the experimental section for Example 12 occurrs via reaction of the chloride intermediate with potassium cyanide (0.9 to 5 eqs, preferably 1.5 eqs) in a solvent such as DMF in the presence of catalytic sodium 4-toluene sulfinate at an elevate temperature such as 100° C. for 3 h. Reaction temperature may vary between 50 and 200° C. depending on substrate and reaction time from 30 min to 48 h. Reactions may be conducted in a sealed tube to minimize escape of volatiles if necessary.
  • Transformation step I, the hydrolysis of the nitrile to the acid may be carried out using acidic conditions such as MeOH and HCl at ambient temp followed by heating the intermediate imididate in the Methanol which provides the intermediate methyl ester which can then be hydrolyzed using potassium carbonate MeOH or LiOH or KOH in Methanol. This method is preferred to produce intact Compounds I. Alternatively, KOH/in ethanol or methanol may be utilized to achieve this transformation in step I. Other methods for this tranformations are well known in the literature or in the references incorporated in the experimental section.
  • Transformation step H can be used to directly produce unsubstituted carboxamides (R1═R2=hydrogen)via stirring in cold concentrated sulfuric acid or at ambient tmeperature for 0.5 to 15 days. Alternatively stirring with MeOH and HCl at room temperature followed by a hydrolytic workup (water and theyl acetate or dichloromethane, may produce the same product.
  • Step J the amide coupling is carried out as described above in the discussions for Scheme 65 and 66.
  • Chemistry
  • General:
  • Additional preparations of starting materials and precursors particulalry those for appending heteroaryls or carboxamides and for construction of substituted piperazines and alkenyl piperidines have been disclosed in a number different PCT and issued U.S. patents/applications (Reference 93-95, 106, 108,109, 110, 111,112, 113 and 114) and U.S. application Ser. No. 10/871,931 filed Jun. 18, 2004, which are hereby incorporated by reference.
  • All Liquid Chromatography (LC) data were recorded on a Shimadzu LC-10AS liquid chromatograph using a SPD-10AV UV-Vis detector with Mass Spectrometry (MS) data determined using a Micromass Platform for LC in electrospray mode.
  • LC/MS Method (Compound Identification)
    • Column A: YMC ODS-A S7 3.0×50 mm column
    • Column B: PHX-LUNA C18 4.6×30 mm column
    • Column C: XTERRA ms C18 4.6×30 mm column
    • Column D: YMC ODS-A C18 4.6×30 mm column
    • Column E: YMC ODS-A C18 4.6×33 mm column
    • Column F: YMC C18 S5 4.6×50 mm column
    • Column G: XTERRA C18 S7 3.0×50 mm column
    • Column H: YMC C18 S5 4.6×33 mm column
    • Column I: YMC ODS-A C18 S7 3.0×50 mm column
    • Column J: XTERRA C-18 S5 4.6×50 mm column
    • Column K: YMC ODS-A C18 4.6×33 mm column
    • Column L: Xterra MS C18 5 uM 4.6×30 mm column
    • Column M: YMC ODS-A C18 S3 4.6×33 mm column
    • Column N: XTERRA MS C18 7 u 3.0×50 mm column
    • Column O: Phenomenex 10 u 4.6×50 mm column
    • Column P: Waters Atlantis 4.6×50 mm C18 Sum column
    • Column Q: Phenomenex 5 u 4.6×50 mm C18 column
    • Column R: Phenomenex Lina C18 5 um 3.0×50 mm column
    • Column S: Phenomenex C18 10 u 3.0×50 mm column
      Standard LC Run Conditions A (used Unless Otherwise Noted):
    • Gradient: 100% Solvent A/0% Solvent B to 0% Solvent A/100% Solvent B
    • Gradient time: 2 minutes
    • Hold time: 1 minute
    • Flow rate: 5 mL/min
    • Detector Wavelength: 220 nm
    • Solvent A: 10% MeOH/90% H2O/0.1% Trifluoroacetic Acid
    • Solvent B: 10% H2O/90% MeOH/0.1% Trifluoroacetic Acid
      Alternate LC Run Conditions B:
    • Gradient: 100% Solvent A/0% Solvent B to 0% Solvent A/100% Solvent B
    • Gradient time: 2 minutes
    • Hold time: 1 minute
    • Flow rate: 5 ml/min
    • Detector Wavelength: 220 nm
    • Solvent A: 5% CH3CN /95% H2O/10 mM Ammonium Acetate
    • Solvent B: 95% CH3CN/55% H2O/10 mM Ammonium Acetate
  • Compounds purified by preparative HPLC were diluted in MeOH and/or DMSO (1-2 mL) and purified using the following methods on a Shimadzu LC-10A automated preparative HPLC system or on a Shimadzu LC-8A automated preparative HPLC system with detector (SPD-10AV UV-VIS) wavelength and solvent systems (A and B) the same as above.
  • Preparative HPLC Method (i.e., Compound Purification)
  • Purification Method: Initial gradient (10% B, 90% A) ramp to final gradient (100% B, 0% A) over 20 minutes, hold for 3 minutes (100% B, 0% A)
    • Solvent A: 10% MeOH/90% H2O/0.1% Trifluoroacetic Acid
    • Solvent B: 10% H2O/90% MeOH/0.1% Trifluoroacetic Acid
    • Column: YMC C18 S5 20×100 mm column
    • Detector Wavelength: 220 nm
  • Starting materials, can be purchased from commercial sources or prepared using literature procedures.
    Figure US20050124623A1-20050609-C00071
  • To a mixture of diethyl aminomalonate hydrochloride (8.0 g, 47.3 mmol) and ethyl (ethoxymethylene)cyanoacetate (10.0 g, 47.2 mmol) in ethanol (60 ml) at r.t. was added a 21 wt. % solution of sodium ethoxide in ethanol (62 ml, 165.4 mmol). The reaction mixture was then stirred at reflux for 20 h. After cooling to r.t., the mixture was neutralized with AcOH (6.75 ml, 118 mmol), concentrated, diluted with H2O (250 mL) and extracted with CHCl3 (3×250 mL). The combined organics were dried (MgSO4), filtered, concentrated and purified by flushing through a pad of silical gel (10 g, EtOAc) to yield amino pyrrole 3 (10.2 g, 45.1 mmol, 95%) as a yellow solid. 1H NMR: (500 MHz, DMSO-d6) δ 11.55 (br s, 1H), 7.21 (d, J=4.0, 1H,), 5.57 (s, 2H), 4.21 (q, J=7.5 Hz, 2H), 4.18 (q, J=7.8 Hz, 2H), 1.27 (t, J=7.5 Hz, 2H), 1.25 (t, J=7.8 Hz, 2H); LC/MS: (ES+) m/z (M-OEt)+=181; HPLC Rt=0.96, column N.
  • Preparation of 4-hydroxy-5H-pyrrolo[3,2-d]pyrimidine-7-carboxylic acid ethyl ester 4
  • Figure US20050124623A1-20050609-C00072
  • A mixture of amino pyrrole 3. (10.0 g, 44.2 mmol) and formamidine acetate (13.8 g, 133 mmol) in ethanol (100 ml) was heated at 105° C. for 20 h. The reaction mixture was filtered while still hot to collect solids that were rinsed with EtOH. The filtrate was allowed to cool to r.t., filtered to collect solids that were washed with EtOH. The combined solids were slurried with Et2O, filtered and dried under vacuum to yield 4,6-diazaindole 4 (6.60 g 31.9 mmol, 72%) as a pale yellow solid which was used without further purification. 1H NMR: (500 MHz, DMSO-d6) δ 7.91 (s, 1H), 7.89 (s, 1H), 4.22 (q, J=7.2 Hz, 2H), 3.17 (s, 2H), 1.27 (t, J=7.2 Hz, 3H); LC/MS: (ES+) m/z (M+H)+=208; HPLC Rt=0.55 min., column N.
  • Preparation of 4-chloro-5H-pyrrolo[3,2-d]pyrimidine-7-carboxylic acid ethyl ester 5
  • Figure US20050124623A1-20050609-C00073
  • 4,6-Diazaindole 4 (2.74 g, 13.2 mmol) was slurried in POCl3 (37 mL, 400 mmol) and heated at 105° C. for 3.5 h. The reaction mixture was cooled, diluted with Et2O (150 mL) and the resulting precipitate was collected by filtration, rinsed with EtOAc and Et2O and dried under vacuum to yield 7-chloro-4,6-diazaindole 5 (2.48 g, 11.0 mmol, 83%) as a yellow powder which was used without further purification. LC/MS: (ES+) m/z (M+H)+=226; HPLC Rt=0.84 min., column N.
  • Preparation of 4-chloro-5H-pyrrolo[3,2-d]pyrimidine-7-carboxylic acid 6
  • Figure US20050124623A1-20050609-C00074
  • To a solution of 7-chloro-4,6-diazaindole 5 (4.0 g, 18 mmol) in THF (90 mL) was added a solution of LiOH.H2O (2.5 g, 59 mmol) in H2O (60 mL). The reaction mixture was stirred at 100° C. for 1 d, concentrated and recrystallized from H2O (20 mL). The crystals were collected by filtration, washed with H2O and dried under high vacuum. The off-white solid was shown to be the lithium salt of the diazaindole carboxylic acid 6 (quantitative), which was used without further purification. 1H NMR: (500 MHz, DMSO-d6) δ 8.28 (s, 1H), 8.14 (s, 1H); LC/MS: (ES+) m/z (M+H)+=198; HPLC Rt=0.47 min., column G.
  • Preparation of 4-chloro-5H-pyrrolo[3,2-d]pyrimidine-7-carbonyl chloride 7
  • Figure US20050124623A1-20050609-C00075
  • A solution of the lithium salt of the diazaindole carboxylic acid 6 (1.3 g, 6.6 mmol) in thionyl chloride (22 mL, 300 mmol) and benzene (30 mL) was heated at 105° C. for 3 h. The reaction was cooled and concentrated under vacuum. The light yellow solid residue was shown to be acid chloride 7 and was used without further purification. The acid chloride 7 was identified by quenching a small amount with methanol to make the analogous methyl ester or with aniline to make the phenyl amide, each of which could be verified by LCMS. Methyl ester: LC/MS: (ES+) m/z (M+H)+=212; HPLC Rt=0.90 min., column G. Phenyl amide: LC/MS: (ES+) m/z (M+H)+=269; HPLC Rt=01.56 min., column G.
  • PREPARATION OF EXAMPLE 1 2-(4-Benzoyl-piperazin-1-yl)-3-(4-chloro-5H-pyrrolo[3,2-d]pyrimidin-7-yl)-3-oxo-propionitrile (Compound 9)
  • Figure US20050124623A1-20050609-C00076
  • EXAMPLE 1
  • To a solution of acid chloride 7 (0.5 mmol) and cyanomethylpiperazine 8 (150 mg, 0.66 mmol) in THF (4 mL) stirring at −35° C. was slowly added a solution of 0.5 M KHMDS in toluene (3.2 mL, 1.6 mmol). The reaction mixture was stirred at −35° C. for 1 h, quenched with sat. aqueous NaHCO3 (50 mL) and extracted with EtOAc (2×50 mL). The combined organics were concentrated and the residue purified by prep HPLC to yield the ketocyano intermediate 9 (39 mg, 0.96 mmol, 19%) as a yellow solid. 1H NMR: (500 MHz, DMSO-d6) δ 8.80 (s, 0.5H), 8.80 (s, 0.5H), 8.65 (s, 0.5H), 8.64 (s, 0.5H), 7.50-7.36 (m, 5H), 4.82-4.77 (m, 1H) 3.80-3.25 (m, 4H) 3.02-2.55 (m, 4H); LC/MS: (ES+) m/z (M+H)+=407; HPLC Rt=0.94 min., column G, conditions B.
  • PREPARATION OF EXAMPLE 2 1-(4-Benzoyl-piperazin-1-yl)-2-(4-chloro-5H-pyrrolo[3,2-d]pyrimidin-7-yl)-ethane-1,2-dione (Compound 10)
  • Figure US20050124623A1-20050609-C00077
  • EXAMPLE 2
  • To a solution of acid chloride 7 (6.6 mmol) and cyanomethylpiperazine 8 (1.96 g, 8.58 mmol) in THF (45 mL) stirring at −78° C. was slowly added a solution of 0.5 M KHMDS in toluene (42 mL, 21 mmol). The reaction mixture was stirred at −78° C. for 2 h and then a solution of 32 wt. % peracetic acid in dilute AcOH (12 ml, 57 mmol) was added and the reaction was stirred at r.t. for 1 h. The reaction mixture was quenched with sat. aqueous NH4Cl (150 mL) and stirred with EtOAc (200 mL). The resultant precipitate was collected by filtration, rinsed with H2O and EtOAc, dried under vacuum and shown to be dicarbonyl intermediate 10 (813 mg, 2.05 mmol, 24%) as an off-white solid. 1H NMR: (500 MHz, DMSO-d6) δ 13.71 (s, 1H), 8.85 (s, 1H), 8.76 (s, 1H), 7.51-7.25 (m, 5H), 3.89-3.20 (m, 8H); LC/MS: (ES+) m/z (M+H)+=398; HPLC Rt=0.80 min., column G, conditions B.
  • PREPARATION OF EXAMPLE 3 1-(4-Benzoyl-piperazin-1-yl)-2-[4-(3-methyl-pyrazol-1-yl)-5H-pyrrolo[3,2-d]pyrimidin-7-yl]-ethane-1,2-dione (Compound 11) EXAMPLE 3
  • Figure US20050124623A1-20050609-C00078
  • In a sealed tube dicarbonyl intermediate 10 (50 mg, 0.13 mmol), 3-methylpyrazole (31 mg, 0.38 mmol) and ethanol (1 mL) were combined and heated to 140° C. with microwaves for 45 min. The reaction was diluted with MeOH (2 mL), filtered and the filtrate was purified by preparative HPLC to yield 11 (36 mg, 0.08 mmol, 65%) as a light yellow solid. 1H NMR: (300 MHz, CD3OD) δ 8.83 (s, 1H), 8.83 (d, J=2.8 Hz, 1H), 8.52 (s, 1H), 7.50-7.41 (m, 5H), 6.50 (d, J=2.8 Hz, 1H), 3.95-3.45 (m, 8H), 2.45 (s, 3H); LC/MS: (ES+) m/z (M+H)+=444; HPLC Rt=0.97 min., column G, conditions B.
  • PREPARATION OF EXAMPLE 4 1-(4-Benzoyl-piperazin-1-yl)-2-[4-(4-methyl-pyrazol-1-yl)-5H-pyrrolo[3,2-d]pyrimidin-7-yl]-ethane-1,2-dione (Compound 12) EXAMPLE 4
  • Figure US20050124623A1-20050609-C00079
  • In a sealed tube dicarbonyl intermediate 10 (50 mg, 0.13 mmol), 4-methylpyrazole (31 mg, 0.38 mmol) and ethanol (1 mL) were combined and heated to 140° C. with microwaves for 30 min. The reaction was diluted with MeOH (2 mL), filtered and the filtrate was purified by preparative HPLC to yield 12 (34 mg, 0.08 mmol, 61%) as a white solid. 1H NMR: (300 MHz, CDCl3) δ 12.13 (br s), 9.17 (s, 1H), 8.74 (s, 1H), 8.40 (s, 1H), 7.85 (s, 1H), 7.46-7.36 (m, 5H), 3.85-3.55 (m, 8H), 2.20 (s, 3H); LC/MS: (ES+) m/z (M+H)+=444; HPLC Rt=0.97 min., column G, conditions B.
  • PREPARATION OF EXAMPLE 5 1-(4-Benzoyl-piperazin-1-yl)-2-(4-methoxy-5H-pyrrolo[3,2-d]pyrimidin-7-yl)-ethane-1,2-dione (Compound 13) EXAMPLE 5
  • Figure US20050124623A1-20050609-C00080
  • To a solution of dicarbonyl intermediate 10 (50 mg, 0.13 mmol) in MeOH (1 mL) was added KOMe (79 mg, 1.1 mmol). The reaction was heated at 90° C. for 0.5 h, cooled, diluted with MeOH (2 mL) and H2O (1 mL) and purified by preparative HPLC to yield 13 (24 mg, 0.06 mmol, 48%) as a white solid. 1H NMR: (500 MHz, CD3OD) δ 8.93 (s, 1H), 8.62 (s, 1H), 7.53-7.40 (m, 5H), 4.39 (s, 3H) 3.98-3.46 (m, 8H); LC/MS: (ES+) m/z (M+H)+=394; HPLC Rt=0.79 min., column G, conditions B.
  • PREPARATION OF EXAMPLE 6 1-(4-Benzoyl-piperazin-1-yl)-2-(4-pyrazol-1-yl-5H-pyrrolo[3,2-d]pyrimidin-7-yl)-ethane-1,2-dione (Compound 14) EAXAMPLE 6
  • Figure US20050124623A1-20050609-C00081
  • In a sealed tube dicarbonyl intermediate 10 (50 mg, 0.13 mmol), pyrazole (26 mg, 0.38 mmol) and ethanol (1 mL) were combined and heated at 140° C. with microwaves for 45 min. The reaction was diluted with MeOH (3 mL), filtered and the filtrate was purified by preparative HPLC to yield 14 (13 mg, 0.03 mmol, 24%) as a white solid. 1H NMR: (500 MHz, CD3OD) δ 8.87 (s, 1H), 8.81 (s, 1H), 8.53 (s, 1H), 8.04 (s, 1H), 7.53-7.39 (m, 5H), 6.69 (s, 1H), 3.98-3.46 (m, 8H); LC/MS: (ES+) m/z (M+H)+=430; HPLC Rt=0.95 min., column G, conditions B.
  • PREPARATION OF EXAMPLE 7 1-(4-Benzoyl-piperazin-1-yl)-2-[4-(3-methyl-[1,2,4]triazol-1-yl)-5H-pyrrolo[3,2-d]pyrimidin-7-yl]-ethane-1,2-dione (Compound 15) EXAMPLE 7
  • Figure US20050124623A1-20050609-C00082
  • In a sealed tube dicarbonyl intermediate 10 (50 mg, 0.13 mmol), 3-methyl-1,2,4-triazole (22 mg, 0.38 mmol) and ethanol (1 mL) were combined and heated at 140° C. with microwaves for 45 min. The reaction was diluted with MeOH/DMF (1:1, 3 mL), filtered and the filtrate was purified by preparative HPLC to yield 15 (9 mg, 0.02 mmol, 17%) as a white solid. 1H NMR: (500 MHz, DMSO-d6) δ 12.74 (br s, 1H), 9.54 (s, 1H), 8.94 (s, 1H), 8.54 (s, 1H), 7.55-7.35 (m, 5H), 3.90-3.22 (m, 8H), 2.53 (s, 3H); LC/MS: (ES+) m/z (M+H)+=445; HPLC Rt=0.88 min., column G, conditions B.
  • PREPARATION OF EXAMPLE 8 1-(4-(2H-1,2,3-Triazol-2-yl)-5H-pyrrolo[3,2-d]pyrimidin-7-yl)-2-(4-benzoylpiperazin-1-yl)ethane-1,2-dione (Compound 16) EXAMPLE 8
  • Figure US20050124623A1-20050609-C00083
  • In a sealed tube dicarbonyl intermediate 10 (50 mg, 0.13 mmol), 3-methyl-1,2,4-triazole (52 mg, 0.75 mmol) and copper powder (16 mg, 0.25 mmol) were combined and heated at 140° C. with microwaves for 1 h. The reaction was diluted with MeOH (3 mL), filtered through celite and the filtrate was purified by preparative HPLC to yield 16 (3 mg, 0.007 mmol, 5%) as a yellow solid. 1H NMR: (500 MHz, CD3OD) δ 8.97 (s, 1H), 8.61 (s, 1H), 8.28 (s, 1H), 8.27 (s, 1H), 7.52-7.40 (m, 5H), 4.02-3.44 (m, 8H); LC/MS: (ES+) m/z (M+H)+=431; HPLC Rt=0.82 min., column G, conditions B.
  • PREPARATION OF EXAMPLE 9 1-{7-[2-(4-Benzoyl-piperazin-1-yl)-2-oxo-acetyl]-5H-pyrrolo[3,2-d]pyrimidin-4-yl}-1H-pyrazole-3-carboxylic acid (Compound 17) EXAMPLE 9
  • Figure US20050124623A1-20050609-C00084
  • In a sealed tube dicarbonyl intermediate 10 (30 mg, 0.076 mmol) and 3-pyrazolecarboxylic acid (26 mg, 0.23 mmol), copper powder (10 mg, 0.16 mmol) and K2CO3 (52 mg, 0.38 mmol) were combined and heated at 150° C. with microwaves for 2 h. The reaction was diluted with MeOH (3 mL), filtered through celite, concentrated, dissolved into DMSO and purified by preparative HPLC to yield 17 (5 mg, 0.01 mmol, 14%) as a white solid. 1H NMR: (500 MHz, DMSO-d6) δ 13.32 (br s, 1H), 12.55 (br s, 1H), 8.98-8.96 (m, 2H), 8.71 (s, 1H), 7.51-7.38 (m, 5H), 7.13 (d, J=2.4 Hz, 1 H), 3.90-3.20 (m, 8H); LC/MS: (ES+) m/z (M+H)+=474; HPLC Rt=0.76 min., column G, conditions B.
  • PREPARATION OF EXAMPLE 10 1-(4-Benzoylpiperazin-1-yl)-2-(4-ethoxy-5H-pyrrolo[3,2-d]pyrimidin-7-yl)ethane-1,2-dione (Compound 18) EXAMPLE 10
  • Figure US20050124623A1-20050609-C00085
  • In a sealed tube dicarbonyl intermediate 10 (30 mg, 0.076 mmol) and 3-pyrazolecarboxylic acid (26 mg, 0.23 mmol), copper powder (10 mg, 0.16 mmol) and K2CO3 (52 mg, 0.38 mmol) in EtOH (1.0 mL) were combined and heated at 150° C. with microwaves for 2 h. The reaction was diluted with MeOH (3 mL), filtered through celite, concentrated, dissolved into DMSO and purified by preparative HPLC to yield 18 (1 mg, 0.002 mmol, 3%) as a white solid. 1H NMR: (500 MHz, CD3OD) δ 8.73 (s, 1H), 8.45 (s, 1H), 7.57-7.40 (m, 5H), 4.77 (q, J=7.3 Hz, 2H), 4.04-3.42 (m, 8H), 1.53 (t, J=7.3 Hz, 3H); LC/MS: (ES+) m/z (M+H)+=408; HPLC Rt=0.85 min., column G, conditions B.
  • PREPARATION OF EXAMPLE 11 1-(4-Acetyl-5H-pyrrolo[3,2-d]pyrimidin-7-yl)-2-(4-benzoylpiperazin-1-yl)ethane-1,2-dione (Compound 19) EXAMPLE 11
  • Figure US20050124623A1-20050609-C00086
  • In a sealed tube dicarbonyl intermediate 10 (600 mg, 1.5 mmol), tributyl(1-ethoxyvinyl)stannane (1.5 mL 4.5 mmol), tetrakis(triphenylphosphine)palladium(O) (350 mg, 0.30 mmol) and 1,4-dioxane (15 mL) were combined and heated at 120° C. with microwaves for 2 h. The reaction mixture was divided and 25% v/v was concentrated diluted with MeOH/CH2Cl2 (2:1, 1.5 mL) and IN aqueous HCl (0.5 mL). The reaction was stirred overnight, neutralized with 1N aqueous NaOH (0.5 mL) and concentrated. The residue was dissolved into DMSO, filtered and purified by preparative HPLC to yield 19 (79 mg, 0.20 mmol, 53%) as a pink solid. 1H NMR: (500 MHz, CD3OD) δ 9.19 (s, 1H), 8.62 (s, 1H), 7.52-7.40 (m, 5H), 3.92-3.41 (m, 8H), 2.80 (s, 3H); LC/MS: (ES+) m/z (M+H)+=406; HPLC Rt=0.79 min., column N.
  • PREPARATION OF EXAMPLE 12 7-(2-(4-Benzoylpiperazin-1-yl)-2-oxoacetyl)-5H-pyrrolo[3,2-d]pyrimidine-4-carbonitrile (Compound 20) EXAMPLE 12
  • Figure US20050124623A1-20050609-C00087
  • In a sealed tube dicarbonyl intermediate 10 (40 mg, 0.10 mmol), potassium cyanide (10 mg, 0.15 mmol), sodium 4-toluenesulfinate (20 mg, 0.11 mmol) and DMF (0.8 mL) were combined and heated at 100° C. for 3 h. The crude reaction mixture was partitioned between aqueous 5% Na2CO3 (0.5 mL) and EtOAc (4 mL). The organic layer was washed with aqueous 5% Na2CO3 (0.7 mL), concentrated, dissolved into MeOH/DMSO (3:1, 2 mL) and purified by preparative HPLC to yield 20 (10 mg, 0.03 mmol, 26%) as a white solid. 1H NMR: (500 MHz, DMSO-d6) δ 14.16 (br s, 1H), 9.20 (s, 1H), 8.96 (s, 1H), 7.52-7.36 (m, 5H), 3.90-3.28 (m, 8H); LC/MS: (ES+) m/z (M+H)+=389; HPLC Rt=0.92 min., column P, conditions B.
  • PREPARATION OF EXAMPLE 13 1-(7-(2-(4-Benzoylpiperazin-1-yl)-2-oxoacetyl)-5H-pyrrolo[3,2-d]pyrimidin-4-yl)-N,N-dimethyl-1H-pyrazole-3-carboxamide (Compound 21) EXAMPLE 13
  • Figure US20050124623A1-20050609-C00088
  • In a sealed tube dicarbonyl intermediate 10 (50 mg, 0.13 mmol), N,N-dimethyl-1H-pyrazole-3-carboxamide (53 mg, 0.38 mmol), copper(0) (10 mg) and 1,4-dioxane (0.8 mL) were combined and heated at 150° C. with microwaves for 2 h. The reaction mixture was concentrated, diluted with MeOH and DMSO, filtered and purified by preparative HPLC to yield 21 (2.3 mg, 0.005 mmol, 4%) as a yellow solid. 1H NMR: (500 MHz, CD3OD) δ 8.94-8.87 (m, 2H), 8.59 (s, 1H), 7.56-7.36 (m, 5H), 6.94 (d, J=2.8 Hz, 1H), 4.07-3.43 (m, 8H), 3.29 (s, 3H), 3.19 (s, 3H); LC/MS: (ES+) m/z (M+H)+=501; HPLC Rt=1.16 min., column N.
  • PREPARATION OF EXAMPLE 14 1-(4-Benzoylpiperazin-1-yl)-2-(4-(3-(1-methylpiperazine-4-carbonyl)-1H-pyrazol-1-yl)-5H-pyrrolo[3,2-d]pyrimidin-7-yl)ethane-1,2-dione (Compound 22) EXAMPLE 14
  • Figure US20050124623A1-20050609-C00089
  • Carboxylic acid 17 (34 mg, 0.73 mmol), N-methylpiperazine (15 mg, 0.15 mmol), N,N-diisopropylethylamine (0.13 mL, 94 mg, 0.73 mmol) and bis(2-oxo-3-oaxzolidinyl)phosphinic chloride (41 mg, 0.16 mmol) were dissolved into CH2Cl2 (0.5 mL) and stirred for 20 h. The reaction mixture was concentrated, diluted with MeOH (1.7 mL) filtered and purified by preparative HPLC to yield 22 (41 mg, 0.73 mmol, 99%) as a white solid 1H NMR: (500 MHz, CD3OD) δ 8.93 (d, J=2.8 Hz, 1H), 8.90 (s, 1H), 8.60 (s, 1H), 7.54-7.37 (m, 5H), 6.98 (d, J=2.8 Hz, 1H), 4.06-3.12 (m, 16H), 2.97 (s, 3H); LC/MS: (ES+) m/z (M+H)+=556; HPLC Rt=1.03 min., column P., conditions B.
  • PREPARATION OF EXAMPLE 15 1-(4-Benzoylpiperazin-1-yl)-2-(4-(3-phenyl-1H-pyrazol-1-yl)-5H-pyrrolo[3,2-d]pyrimidin-7-yl)ethane-1,2-dione (Compound 23) EAXMAPLE 15
  • Figure US20050124623A1-20050609-C00090
  • In a sealed tube dicarbonyl intermediate 10 (50 mg, 0.13 mmol), 3-phenyl-1H-pyrazole (80 mg, 0.56 mmol) and 1,4-dioxane (2.5 mL) were combined and heated at 170° C. with microwaves for 20 min. The reaction was concentrated and triturated with MeOH (3 mL). The resulting solids were washed with MeOH and with Et2O to yield 23 (33 mg, 0.07 mmol, 50%) as a tan solid. 1H NMR: (500 MHz, DMSO-d6) δ 12.42 (br s, 1H), 8.95 (d, J=2.8 Hz, 1H), 8.92 (s, 1H), 8.57 (br d, J=3.1 Hz, 1H), 8.27 (d, J=7.3 Hz, 2H), 7.55 (dd, J=7.3, 7.3 Hz, 2H), 7.48 (t, J=7.3 Hz, 1H), 7.51-7.38 (m, 5H), 7.30 (d, J=2.8 Hz, 1H), 3.92-3.21 (m, 8H); LC/MS: (ES+) m/z (M+H)+=505; HPLC Rt=1.40 min., column Q, conditions B.
  • PREPARATION OF EXAMPLE 16 1-(4-Benzoylpiperazin-1-yl)-2-(4-(3-(4-fluorophenyl)-1H-pyrazol-1-yl)-5H-pyrrolo[3,2-d]pyrimidin-7-yl)ethane-1,2-dione (Compound 24) EXAMPLE 16
  • Figure US20050124623A1-20050609-C00091
  • In a sealed tube dicarbonyl intermediate 10 (50 mg, 0.13 mmol), 3-(4-fluorophenyl)-1H-pyrazole (150 mg, 0.93 mmol) and 1,4-dioxane (2 mL) were combined and heated at 160° C. with microwaves for 20 min. The reaction was concentrated, diluted with MeOH (3 mL), neutralized with saturated aqueous NaHCO3 and 24 (15 mg, 0.03 mmol, 22%) was collected by filtration as a tan solid. 1H NMR: (500 MHz, DMSO-d6) δ 12.48 (br s, 1H), 8.96 (d, J=2.8 Hz, 1H), 8.91 (s, 1H), 8.58 (br s, 1H), 8.34 (dd, J=8.7, 5.6 Hz, 2H), 7.50-7.39 (m, 5H), 7.88 (t, J=8.7 Hz, 2H), 7.80 (d, J=2.8 Hz, 1H), 3.91-3.23 (m, 8H); LC/MS: (ES+) m/z (M+H)+=524; HPLC Rt=1.42 min., column Q, conditions B.
  • PREPARATION OF EXAMPLE 17 1-(4-Benzoylpiperazin-1-yl)-2-(4-(3-(4-methoxyphenyl)-1H-pyrazol-1-yl)-5H-pyrrolo[3,2-d]pyrimidin-7-yl)ethane-1,2-dione (Compound 25) EAXMAPLE 17
  • Figure US20050124623A1-20050609-C00092
  • In a sealed tube dicarbonyl intermediate 10 (50 mg, 0.13 mmol), 3-(4-methoxyphenyl)-1H-pyrazole (105 mg, 0.60 mmol) and 1,4-dioxane (2.5 mL) were combined and heated at 170° C. with microwaves for 20 min. The reaction was concentrated and triturated with MeOH (3 mL). The resulting solids were washed with MeOH and with Et2O to yield 25 (43 mg, 0.08 mmol, 61%) as a tan solid. 1H NMR: (500 MHz, DMSO-d6) δ 12.41 (br s, 1H), 8.91 (d, J=2.8 Hz, 1H), 8.89 (s, 1H), 8.55 (br d, J=3.4 Hz, 1H), 8.20 (d, J=8.7 Hz, 2H), 7.51-7.39 (m, 5H), 7.23 (d, J=2.8 Hz, 1H), 7.08 (d, J=8.7 Hz, 2H), 3.85 (s, 3H), 3.92-3.21 (m, 8H); LC/MS: (ES+) m/z (M+H)+=535; HPLC Rt=1.40 min., column Q, conditions B.
  • PREPARATION OF EXAMPLE 18 1-(4-Benzoylpiperazin-1-yl)-2-(4-(3-(2-methoxyphenyl)-1H-pyrazol-1-yl)-5H-pyrrolo[3,2-d]pyrimidin-7-yl)ethane-1,2-dione (Compound 26) EAXMAPLE 18
  • Figure US20050124623A1-20050609-C00093
  • In a sealed tube dicarbonyl intermediate 10 (51 mg, 0.13 mmol), 3-(2-methoxyphenyl)-1H-pyrazole (109 mg, 0.60 mmol) and 1,4-dioxane (2 mL) were combined and heated at 170° C. with microwaves for 20 min. The reaction was concentrated and triturated with MeOH (3 mL). The resulting solids were washed with MeOH and with Et2O to yield 26 (32 mg, 0.06 mmol, 47%) as a tan solid. 1H NMR: (500 MHz, DMSO-d6) δ 12.34 (br s, 1H), 8.91 (s, 1H), 8.90 (d, J=2.8 Hz, 1H), 8.55 (br d, J=3.4 Hz, 1H), 8.37 (dd, J=7.8, 1.5 Hz, 1H), 7.50-7.39 (m, 5H), 7.47 (ddd, J=8.2, 7.0, 1.5 Hz, 1H), 7.21 (d, J=2.8 Hz, 1H), 7.20 (d, J=8.2 Hz, 1H), 7.14 (dd, J=7.8, 7.0 Hz, 1H), 3.94 (s, 3H), 3.91-3.23 (m, 8H); LC/MS: (ES+) m/z (M+H)+=535; HPLC Rt=1.42 min., column Q, conditions B.
  • PREPARATION OF EXAMPLE 19 1-(4-Benzoylpiperazin-1-yl)-2-(4-(3-(3-(trifluoromethyl)phenyl)-1H-pyrazol-1-yl)-5H-pyrrolo[3,2-d]pyrimidin-7-yl)ethane-1,2-dione (Compound 27) EAXMAPLE 19
  • Figure US20050124623A1-20050609-C00094
  • In a sealed tube dicarbonyl intermediate 10 (50 mg, 0.13 mmol), 3-(3-(trifluoromethyl)phenyl)-1H-pyrazole (150 mg, 0.70 mmol) and 1,4-dioxane (2 mL) were combined and heated at 160° C. with microwaves for 20 min. The reaction was concentrated, dissolved into MeOH (3 mL) and purified by preparative HPLC to yield 27 (34 mg, 0.06 mmol, 46%) as a white solid. 1H NMR: (500 MHz, DMSO-d6) δ 12.63 (br s, 1H), 9.00 (d, J=2.8 Hz, 1H), 8.94 (s, 1H), 8.64 (s, 1H), 8.62 (d, J=7.9 Hz, 1H), 8.54 (s, 1H), 7.83 (d, J=7.6 Hz, 1H), 7.79 (dd, J=7.9, 7.6 Hz, 1H), 7.52-7.36 (m, 5H), 7.47 (d, J=2.8 Hz, 1H), 3.90-3.25 (m, 8H); LC/MS: (ES+) m/z (M+H)+=574; HPLC Rt=1.77 min., column Q, conditions B.
  • PREPARATION OF EXAMPLE 20 1-(4-(1H-1,2,4-Triazol-1-yl)-5H-pyrrolo[3,2-d]pyrimidin-7-yl)-2-(4-benzoylpiperazin-1-yl)ethane-1,2-dione (Compound 28) EXAMPLE 20
  • Figure US20050124623A1-20050609-C00095
  • In a sealed tube dicarbonyl intermediate 10 (50 mg, 0.13 mmol), 1,2,4-triazole (26 mg, 0.44 mmol), copper(0) (8 mg, 0.13 mmol), K2CO3 (23 mg, 0.17 mmol) and 1,4-dioxane (0.8 mL) were combined and heated at 140° C. with microwaves for 6 h. The reaction mixture was diluted with MeOH/CH2Cl2 (1:1, 2 mL), filtered, concentrated, dissolved into MeOH/DMSO (5:4, 1.8 mL) and purified by preparative HPLC. The resulting yellow solid was triturated with MeOH to yield 28 (15 mg, 0.03 mmol, 28%) as a light yellow solid. 1H NMR: (500 MHz, DMSO-d6) δ 12.95 (s, 1H), 9.71 (s, 1H), 8.98 (s, 1H), 8.61 (s, 1H), 8.56 (br s, 1H), 7.52-7.36 (m, 5H), 3.92-3.23 (m, 8H); LC/MS: (ES+) m/z (M+H)+=431; HPLC Rt=0.98 min., column C.
  • PREPARATION OF EXAMPLE 21 1-(4-(1H-1,2,3-Triazol-1-yl)-5H-pyrrolo[3,2-d]pyrimidin-7-yl)-2-(4-benzoylpiperazin-1-yl)ethane-1,2-dione (Compound 29) EXAMPLE 21
  • Figure US20050124623A1-20050609-C00096
  • In a sealed tube dicarbonyl intermediate 10 (60 mg, 0.15 mmol) and 1,2,3-triazole (95 mg, 1.4 mmol) in 1,4-dioxane (3 mL) were combined and heated at 170° C. with microwaves for 20 min. The reaction was concentrated and the residue was triturated with MeOH to yield 29 (10 mg, 0.023 mmol, 16%) as a yellow solid. 1H NMR: (500 MHz, CD3OD) δ 9.08 (s, 1H), 9.04 (s, 1H), 8.66 (s, 1H), 8.07 (s, 1H), 7.57-7.41 (m, 5H), 4.08-3.43 (m, 8H); LC/MS: (ES+) m/z (M+H)+=431; HPLC Rt=0.95 min., column O, conditions B.
  • PREPARATION OF EXAMPLE 22 1-(4-(1H-Pyrazol-3-yl)-5H-pyrrolo[3,2-d]pyrimidin-7-yl)-2-(4-benzoylpiperazin-1-yl)ethane-1,2-dione (Compound 30) EXAMPLE 22
  • Figure US20050124623A1-20050609-C00097
  • In a sealed tube dicarbonyl intermediate 10 (50 mg, 0.13 mmol), 3-(tributylstannyl)pyrazole (188 mg, 0.52 mmol), tetrakis(triphenylphosphine)-palladium(0) (72 mg, 0.06 mmol) and 1,4-dioxane (0.8 mL) were combined and heated at 110° C. with microwaves for 1 h. The reaction mixture was diluted with MeOH/CH2Cl2 (1:1, 2 mL) and filtered to collect solids. The solids were dissolved into DMSO and purified by preparative HPLC to yield 30 (28 mg, 0.07 mmol, 52%) as a white solid. 1H NMR: (500 MHz, DMSO-d6) δ 13.63 (br s, 1H), 12.41 (s, 1H), 9.03 (s, 1H), 8.44 (d, J=2.3 Hz, 1H), 8.03 (d, J=2.3 Hz, 1H), 7.50-7.37 (m, 5H), 7.13 (d, J=1.8 Hz, 1H), 3.90-3.23 (m, 8H); LC/MS: (ES+) m/z (M+H)+=430; HPLC Rt=0.87 min., column R, conditions B.
  • PREPARATION OF EXAMPLE 23 1-(4-Benzoylpiperazin-1-yl)-2-(4-(3-methylisoxazol-5-yl)-5H-pyrrolo[3,2-d]pyrimidin-7-yl)ethane-1,2-dione (Compound 31) EXAMPLE 23
  • Figure US20050124623A1-20050609-C00098
  • In a sealed tube dicarbonyl intermediate 10 (50 mg, 0.13 mmol), 3-methyl-5-(tributylstannyl)isoxazole (141 mg, 0.38 mmol), tetrakis(triphenylphosphine)-palladium(0) (30 mg, 0.03 mmol) and 1,4-dioxane (1 mL) were combined and heated at 110° C. with microwaves for 2 h, and then heated at 120° C. for 2 h. The reaction was repeated as described above and the reaction solution was heated at 110° C. with microwaves for 5 h. The two reactions were combined, diluted with MeOH/DMSO, filtered and purified by preparative HPLC. The resulting yellow solid was triturated with MeOH to yield 31 (4 mg, 0.008 mmol, 3%) as a white solid. 1H NMR: (500 MHz, CD3OD) δ 9.11 (s, 1H), 8.66 (s, 1H), 7.56-7.42 (m, 5H), 7.27 (s, 1H), 4.06-3.45 (m, 8H), 2.47 (s, 3H); LC/MS: (ES+) m/z (M+H)+=445; HPLC Rt=1.23 min., column L.
  • PREPARATION OF EXAMPLE 24 1-(4-Benzoylpiperazin-1-yl)-2-(4-(pyridin-2-yl)-5H-pyrrolo[3,2-d]pyrimidin-7-yl)ethane-1,2-dione (Compound 32) EXAMPLE 24
  • Figure US20050124623A1-20050609-C00099
  • In a sealed tube dicarbonyl intermediate 10 (50 mg, 0.13 mmol), 2-(tributylstannyl)pyridine (140 mg, 0.38 mmol), tetrakis(triphenylphosphine)-palladium(0) (30 mg, 0.03 mmol) and 1,4-dioxane (0.8 mL) were combined and heated at 110° C. with microwaves for 1 h. The reaction mixture was concentrated, diluted with MeOH, filtered and purified by preparative HPLC to yield 32 (8 mg, 0.02 mmol, 14%) as a yellow waxy solid. 1H NMR: (500 MHz, CD3OD) δ 9.15-9.10 (m, 1H), 8.89 (br s, 1H), 8.70 (d, J=7.9 Hz, 1H), 8.62 (s, 1H), 8.09-8.05 (m, 1H), 7.61-7.55 (m, 1H), 7.54-7.37 (m, 5H), 4.05-3.42 (m, 8H); LC/MS: (ES+) m/z (M+H)+=441; HPLC Rt=1.54 min., column P.
  • PREPARATION OF EXAMPLE 25 1-(4-Benzoylpiperazin-1-yl)-2-(4-(pyridin-3-yl)-5H-pyrrolo[3,2-d]pyrimidin-7-yl)ethane-1,2-dione (Compound 33) EXAMPLE 25
  • Figure US20050124623A1-20050609-C00100
  • In a sealed tube dicarbonyl intermediate 10 (50 mg, 0.13 mmol), 3-(tributylstannyl)pyridine (160 mg, 0.43 mmol), tetrakis(triphenylphosphine)-palladium(0) (40 mg, 0.03 mmol) and 1,4-dioxane (0.8 mL) were combined and heated at 130° C. for 4 h. The reaction mixture was concentrated to dryness and partitioned between EtOAc (5 mL) and saturated aqueous NaHCO3 with cesium fluoride. The biphasic suspension was filtered, separated and the aqueous layer was concentrated to dryness. The residue was diluted with MeOH (2 mL) and DMSO (0.5 mL), filtered and purified by preparative HPLC to yield 33 (7 mg, 0.02 mmol, 12%) as a yellow solid. 1H NMR: (500 MHz, CD3OD) δ 9.41 (s, 1H), 9.20 (s, 1H), 8.96 (d, J=5.5 Hz, 1H), 8.91 (d, J=7.6 Hz, 1H), 8.73 (s, 1H), 8.05 (dd, J=7.6, 5.5 Hz, 1H), 7.56-7.42 (m, 5H), 4.10-3.43 (m, 8H); LC/MS: (ES+) m/z (M+H)+=441; HPLC Rt=0.92 min., column S.
  • PREPARATION OF EXAMPLE 26 1-(4-Benzoylpiperazin-1-yl)-2-(4-(pyridin-4-yl)-5H-pyrrolo[3,2-d]pyrimidin-7-yl)ethane-1,2-dione (Compound 34) EXAMPLE 26
  • Figure US20050124623A1-20050609-C00101
  • In a sealed tube dicarbonyl intermediate 10 (50 mg, 0.13 mmol), 4-(tributylstannyl)pyridine (140 mg, 0.38 mmol), tetrakis(triphenylphosphine)-palladium(0) (30 mg, 0.03 mmol) and 1,4-dioxane (0.8 mL) were combined and heated at 110° C. with microwaves for 2 h, and then at 120° C. for 2 h. The reaction mixture was concentrated, diluted with MeOH/DMSO, filtered and purified by preparative HPLC to yield 34 (22 mg, 0.08 mmol, 38%) as a yellow solid. 1H NMR: (500 MHz, CD3OD) δ 9.24 (s, 1H), 9.04 (d, J=5.8 Hz, 2H), 8.75 (s, 1H), 8.52 (d, J=5.8 Hz, 2H), 7.57-7.40 (m, 5H), 4.12-3.44 (m, 8H); LC/MS: (ES+) m/z (M+H)+=441; HPLC Rt=0.78 min., column L.
  • PREPARATION OF EXAMPLE 27 1-(4-Benzoylpiperazin-1-yl)-2-(4-(pyrazin-2-yl)-5H-pyrrolo[3,2-d]pyrimidin-7-yl)ethane-1,2-dione (Compound 35) EXAMPLE 27
  • Figure US20050124623A1-20050609-C00102
  • In a sealed tube dicarbonyl intermediate 10 (50 mg, 0.13 mmol), 2-(tributylstannyl)pyrazine (160 mg, 0.43 mmol), tetrakis(triphenylphosphine)-palladium(0) (30 mg, 0.03 mmol) and 1,4-dioxane (0.8 mL) were combined and heated at 120° C. with microwaves for 2 h, and then at 130° C. for 2 h. The reaction mixture was concentrated to dryness, diluted with MeOH (2.5 mL) and DMSO (0.5 mL), filtered and purified by preparative HPLC. The resulting yellow solid was triturated with MeOH to yield 35 (8 mg, 0.02 mmol, 15%) as a light yellow solid. 1H NMR: (500 MHz, DMSO-d6) δ 12.93 (br s, 1H), 9.75 (d, J=1.2 Hz, 1H), 9.21 (s, 1H), 8.93 (dd, J=2.4, 1.2 Hz, 1H), 8.90 (d, J=2.4 Hz, 1H), 8.64 (br s, 1H), 7.53-7.35 (m, 5H), 3.96-3.21 (m, 8H); LC/MS: (ES+) m/z (M+H)+=442; HPLC Rt=1.15 min., column S.
  • PREPARATION OF EXAMPLE 28 1-(4-Benzoylpiperazin-1-yl)-2-(4-(pyrimidin-5-yl)-5H-pyrrolo[3,2-d]pyrimidin-7-yl)ethane-1,2-dione (Compound 36) EXAMPLE 28
  • Figure US20050124623A1-20050609-C00103
  • In a sealed tube dicarbonyl intermediate 10 (50 mg, 0.13 mmol), 5-(tributylstannyl)pyrimidine (160 mg, 0.43 mmol), tetrakis(triphenylphosphine)-palladium(0) (30 mg, 0.03 mmol) and 1,4-dioxane (0.8 mL) were combined and heated at 130° C. with microwaves for 2 h. The reaction mixture was concentrated, diluted with MeOH/DMSO, filtered and purified by preparative HPLC. The resulting orange solid was triturated with acetone to yield 36 (36.3 mg, 0.08 mmol, 63%) as a yellow solid. 1H NMR: (500 MHz, DMSO-d6) δ 13.48 (br s, 1H), 9.42 (s, 1H), 9.41 (s, 2H), 9.18 (s 1H), 7.52-7.39 (m, 5H), 3.91-3.24 (m, 8H); LC/MS: (ES+) m/z (M+H)+=442; HPLC Rt=0.90 min., column S.
  • PREPARATION OF EXAMPLE 29 1-(4-Benzoylpiperazin-1-yl)-2-(4-(pyridazin-4-yl)-5H-pyrrolo[3,2-d]pyrimidin-7-yl)ethane-1,2-dione (Compound 37) EXAMPLE 29
  • Figure US20050124623A1-20050609-C00104
  • In a sealed tube dicarbonyl intermediate 10 (50 mg, 0.13 mmol), 4-(tributylstannyl)pyridazine (160 mg, 0.43 mmol), tetrakis(triphenylphosphine)-palladium(0) (30 mg, 0.03 mmol) and 1,4-dioxane (0.8 mL) were combined and heated at 130° C. with microwaves for 2 h. The reaction mixture was concentrated, diluted with MeOH, filtered and purified by preparative HPLC. The resulting black oil was repurified by preparative HPLC and the resulting yellow solid was triturated with acetone to yield 37 (18.7 mg, 0.04 mmol, 33%) as an off-white solid. 1H NMR: (500 MHz, DMSO-d6) δ 13.48 (br s, 1H), 9.82 (br s, 1H), 9.54 (dd, J=5.2, 1.1 Hz, 1H), 9.22 (s 1H), 8.85 (br s, 1H), 8.29 (dd, J=5.2, 2.0 Hz, 1H), 7.54-7.37 (m, 5H), 3.91-3.25 (m, 8H); LC/MS: (ES+) m/z (M+H)+=442; HPLC Rt=0.89 min., column S.
  • Preparation of 2-(4-(isoquinolin-1-yl)piperazin-1-yl)acetonitrile 38
  • Figure US20050124623A1-20050609-C00105
  • To a solution of 1-(piperazin-1-yl)isoquinoline (459 mg, 2.15 mmol) in THF (15 mL) was added NEt3 (3.6 mL, 27 mmol) and chloroacetonitrile (1.8 mL, 28 mmol) and the reaction was stirred 3 h. The reaction mixture was filtered, concentrated and the residue purified by silica gel chromatography (Biotage 25-short, 25% EtOAc/hexanes to 100% EtOAc/hex) to yield 38 (188 mg, 0.75 mmol, 35%) as a white solid. LC/MS: (ES+) m/z (M+H)+=253; HPLC Rt=1.23 min., column Q, conditions B.
  • PREPARATION OF EXAMPLE 30 1-(4-Chloro-5H-pyrrolo[3,2-d]pyrimidin-7-yl)-2-(4-(isoquinolin-1-yl)piperazin-1-yl)ethane-1,2-dione (Compound 39) EXAMPLE 30
  • Figure US20050124623A1-20050609-C00106
  • To a slurry of acid chloride intermediate 7 (115 mg, 0.53 mmol) and 2-(4-(isoquinolin-1-yl)piperazin-1-yl)acetonitrile 38 (188 mg, 0.75 mmol) in THF (5 mL), at −78° C., was added 0.5 M KHMDS in toluene (3.4 mL, 1.7 mmol). The reaction was stirred 2 h. and the presence of the desired cyanoketone intermediate was verified by LCMS. A solution of 32% peracetic acid in dilute aqueous acetic acid (0.5 mL, 2.4 mmol) was added and the reaction mixture was allowed to warm to ambient temperature overnight. The reaction mixture was diluted with EtOAc (10 mL) and saturated aqueous NH4Cl (10 mL) and filtered. The layers were separated and the aqueous layer extracted with EtOAc (25 mL). The combined organic layers were concentrated and purified by preparative HPLC to yield 39 (102 mg, 0.24 mmol, 45%) as a bright yellow solid. 1H NMR: (500 MHz, CD3OD) δ 8.84 (s, 1H), 8.70 (s, 1H), 8.38 (d, J=7.9 Hz, 1H), 8.06 (d, J=7.2 Hz, 1H), 7.99 (dd, J=7.9, 7.6 Hz, 1H), 7.89 (d, J=6.7 Hz, 1H), 7.83 (dd, J=7.6, 7.2 Hz, 1H), 7.62 (d, J=6.7 Hz, 1H), 4.20-3.79 (m, 8H); LC/MS: (ES+) m/z (M+H)+=421; HPLC Rt=0.83 min., column S.
  • PREPARATION OF EXAMPLE 31 1-(4-(Isoquinolin-1-yl)piperazin-1-yl)-2-(4-(pyrazin-2-yl)-5H-pyrrolo[3,2-d]pyrimidin-7-yl)ethane-1,2-dione (Compound 40) EXAMPLE 31
  • Figure US20050124623A1-20050609-C00107
  • In a sealed tube dicarbonyl intermediate 39 (40 mg, 0.10 mmol), 2-(tributylstannyl)pyrazine (105 mg, 0.28 mmol), tetrakis(triphenylphosphine)-palladium(0) (30 mg, 0.03 mmol) and 1,4-dioxane (0.8 mL) were combined and heated at 130° C. with microwaves for 2 h. The reaction mixture was diluted with MeOH (1 mL) and DMSO (1 mL), filtered through celite and purified by preparative HPLC to yield 40 (12 mg, 0.03 mmol, 27%) as a yellow solid. 1H NMR: (500 MHz, CD3OD) δ 9.85 (s, 1H), 9.21 (s, 1H), 8.93 (br s, 1H), 8.83 (d, J=2.4 Hz, 1H), 8.72 (s, 1H), 8.42 (d, J=8.9 Hz, 1H), 8.09 (d, J=7.9 Hz, 1H), 8.05 (dd, J=8.0, 7.0 Hz, 1H), 7.87 (dd, J=8.9, 8.0 Hz, 1H), 7.85 (d, J=7.0 Hz, 1H), 7.66(d, J=6.7 Hz, 1H), 4.24-4.20 (m, 2H), 4.14-4.04 (m, 2H), 3.99-3.91 (m, 4H); LC/MS: (ES+) m/z (M+H)+=465; HPLC Rt=0.92 min., column S.
  • PREPARATION OF EXAMPLE 32 1-(4-(1H-Pyrazol-1-yl)-5H-pyrrolo[3,2-d]pyrimidin-7-yl)-2-(4-(isoquinolin-1-yl)piperazin-1-yl)ethane-1,2-dione (Compound 41) EXAMPLE 32
  • Figure US20050124623A1-20050609-C00108
  • In a sealed tube dicarbonyl intermediate 39 (41 mg, 0.10 mmol), pyrazole (26 mg, 0.38 mmol), copper(0) (10 mg) and 1,4-dioxane (0.8 mL) were combined and heated at 140° C. with microwaves for 50 min. The reaction mixture was diluted with MeOH (1 mL) and DMSO (1 mL), filtered through celite and purified by preparative HPLC to yield 41 (11 mg, 0.02 mmol, 23%) as a light yellow solid. 1H NMR: (500 MHz, CD3OD) δ 8.90 (br s, 1H), 8.85 (d, J=2.5 Hz, 1H), 8.61 (s, 1H), 8.42 (d, J=8.5 Hz, 1H), 8.09 (d, J=7.9 Hz, 1H), 8.08 (s, 1H), 8.04 (dd, J=7.6, 7.3 Hz, 1H), 7.89-7.85 (m, 1H), 7.85 (d, J=6.7 Hz, 1H), 7.66 (d, J=7.0 Hz, 1H), 6.72 (dd, J=2.5, 1.6 Hz, 1H), 4.22-4.18 (m, 2H), 4.12-4.07 (m, 2H), 3.97-3.89 (m, 4H); LC/MS: (ES+) m/z (M+H)+=453; HPLC Rt=0.97 min., column S.
  • Preperation of 2-(1-(cyanomethyl)piperidin-4-ylidene)-2-phenylacetonitrile 42
  • Figure US20050124623A1-20050609-C00109
  • To a solution of 2-phenyl-2-(piperidin-4-ylidene)acetonitrile (6.8 g, 34 mmol) in THF (150 mL) was added NEt3 (40 mL, 300 mmol) and chloroacetonitrile (20 mL, 315 mmol) and the reaction was stirred 16 h. The precipitates was filtered away and the filtrate concentrated to dryness. The residues was purified by silica gel chromatography (Biotage 40-short, 20% EtOAc/Hex to 50% EtOAc/Hex) to yield 42 (1.7 g, 7.2 mmol, 21%) as a yellow waxy solid. 1H NMR: (500 MHz, CDCl3) δ 7.44-7.34 (m, 3H), 7.30-7.27 (m, 2H), 3.65 (s, 2H), 2.96 (t, J=5.3 Hz, 2H), 2.90 (t, J=5.3 Hz, 2H), 2.70 (t, J=5.6 Hz, 2H), 2.62 (t, J=5.6 Hz, 2H); LC/MS: (ES+) m/z (M+H)+=238; HPLC Rt=1.33 min., column O, conditions B.
  • PREPARATION OF EXAMPLE 33 2-(1-(2-(4-Chloro-5H-pyrrolo[3,2-d]pyrimidin-7-yl)-2-oxoacetyl)piperidin-4-ylidene)-2-phenylacetonitrile (Compound 43) EXAMPLE 33
  • Figure US20050124623A1-20050609-C00110
  • To a slurry of acid chloride intermediate 7 (100 mg, 0.46 mmol) and phenylcyanoalkene intermediate 42 (143 mg, 0.60 mmol) in THF (4 mL) at −78° C. was added a solution of 0.5 M KHMDS in toluene (3.0 mL, 1.5 mmol). The reaction was stirred 2 h and the presence of the desired cyanoketone intermediate was verified by LCMS. A solution of 32% peracetic acid in dilute aqueous acetic acid (0.44 mL, 2.1 mmol) was added to the reaction mixture and then allowed to warm to ambient temperature overnight. The reaction mixture was diluted with EtOAc (15 mL) and saturated aqueous NH4Cl (10 mL). The layers were separated and the aqueous layer extracted with EtOAc (2×20 mL). The combined organic layers were concentrated, the residue was purified by preparative HPLC and the resulting yellow solid was triturated with MeOH to yield 43 (18.6 mg, 0.04 mmol, 10%) as a white solid. 1H NMR: (500 MHz, DMSO-d6) δ 13.73 (s, 1H), 8.85 (s, 0.5H), 8.84 (s, 0.5H), 8.79 (s, 0.5H), 8.76 (s, 0.5H), 7.54-7.30 (m, 5H), 3.86 (t, J=5.8 Hz, 1H), 3.70 (t, J=5.8 Hz, 1H), 3.56 (t, J=5.8 Hz, 1H), 3.38 (t, J=5.8 Hz, 1H), 2.93 (t, J=5.8 Hz, 1H), 2.65 (t, J=5.8 Hz, 1H), 2.63 (dd, J=5.8 Hz, 1H), 2.36 (t, J=5.8 Hz, 1H); LC/MS: (ES+) m/z (M+H)+=406; HPLC Rt=1.28 min., column S.
  • PREPARATION OF EXAMPLE 34 2-(1-(2-Oxo-2-(4-(pyrimidin-5-yl)-5H-pyrrolo[3,2-d]pyrimidin-7-yl)acetyl)piperidin-4-ylidene)-2-phenylacetonitrile (Compound 44) EXAMPLE 34
  • Figure US20050124623A1-20050609-C00111
  • In a sealed tube dicarbonyl intermediate 33 (30 mg, 0.074 mmol), 5-(tributylstannyl)pyrimidine (82 mg, 0.22 mmol), tetrakis(triphenylphosphine)-palladium(0) (20 mg, 0.02 mmol) and 1,4-dioxane (0.8 mL) were combined and heated at 130° C. with microwaves for 2 h. The reaction mixture was diluted with MeOH/DMSO, filtered and purified by preparative HPLC to yield 44 (8 mg, 0.02 mmol, 30%) as a yellow solid. 1H NMR: (500 MHz, CD3OD) δ 9.47 (s, 1H), 9.45 (s, 1H), 9.40 (s, 0.5H), 9.39 (s, 0.5H), 9.19 (s, 0.5H), 9.18 (s, 0.5H), 8.72 (s, 0.5H), 8.69 (s, 0.5H), 7.53-7.31 (m, 5H), 4.02 (dd, J=6.1, 5.8 Hz, 1H), 3.84 (dd, J=6.1, 5.8 Hz, 1H), 3.75 (dd, J=5.8, 5.8 Hz, 1H), 3.57 (dd, J=6.1, 5.8 Hz, 1H), 3.07 (dd, J=6.1, 5.8 Hz, 1H), 2.86 (dd, J=5.8, 5.8 Hz, 1H), 2.74 (dd, J=6.1, 5.8 Hz, 1H), 2.54 (dd, J=6.1, 5.8 Hz, 1H); LC/MS: (ES+) m/z (M+H)+=450; HPLC Rt=1.48 min., column O.
  • Preparation of 2-(4-((1,3,4-oxadiazol-2-yl)(phenyl)methylene)piperidin-1-yl)acetonitrile 45
  • Figure US20050124623A1-20050609-C00112
  • tert-Butyl 4-((1,3,4-oxadiazol-2-yl)(phenyl)methylene)piperidine-1-carboxylate (100 mg, 0.29 mmol) was diluted with 4 M HCl in 1,4-dioxane (1.2 mL, 4.8 mmol) and stirred 1 h. The reaction was concentrated and the residue diluted with THF (1.5 mL), triethylamine (0.5 mL, 3.8 mmol) and chloroacetonitrile (0.25 mL, 3.9 mmol). The reaction was stirred 3 d, concentrated, diluted with MeOH, filtered and purified by preparative HPLC to yield 45 (43 mg, 0.15 mmol, 53%) as a white solid. 1H NMR: (500 MHz, CDCl3) δ 8.27 (s, 1H), 7.41-7.31 (m, 3H), 7.20-7.16 (m, 2H), 3.55 (s, 2H), 3.02 (t, J=5.8 Hz, 2H), 2.75 (t, J=5.8 Hz, 2H), 2.61 (t, J=5.8 Hz, 2H), 2.39 (t, J=5.8 Hz, 2H); LC/MS: (ES+) m/z (M+H)+=281; HPLC Rt=0.97 min., column G, conditions B.
  • PREPARATION OF EXAMPLE 35 1-(4-((1,3,4-Oxadiazol-2-yl)(phenyl)methylene)piperidin-1-yl)-2-(4-(1H-1,2,4-triazol-1-yl)-5H-pyrrolo[3,2-d]pyrimidin-7-yl)ethane-1,2-dione (Compound 46) EXAMPLE 35
  • Figure US20050124623A1-20050609-C00113
  • To a slurry of acid chloride intermediate 7 (600 mg, 2.8 mmol) and 2-(4-((1,3,4-oxadiazol-2-yl)(phenyl)methylene)piperidin-1-yl)acetonitrile 45 (800 mg, 2.9 mmol) in THF (5 mL), at −78° C. was added a solution of 0.5 M KHMDS in toluene (17.2 mL, 8.6 mmol). The reaction was stirred 2 h. A solution of 32% peracetic acid in dilute aqueous acetic acid (2.8 mL, 13 mmol) was added and the reaction mixture was allowed to warm to ambient temperature over 1 h. The reaction mixture was diluted with EtOAc (30 mL) and brine (25 mL) and filtered. The layers were separated and the organic layer concentrated. The residue was triturated with Et2O to yield 46 (340 mg, 0.76 mmol, 27%) as an orange/yellow solid. 1H NMR: (500 MHz, DMSO-d6) δ 13.73 (s, 1H), 9.17 (s, 0.5H), 9.10 (s, 0.5H), 8.83 (s, 0.5H), 8.82 (s, 0.5H), 8.76 (s, 0.5H), 8.74 (s, 0.5H), 7.49-7.15 (m, 5H), 3.80 (t, J=5.8 Hz, 1H), 3.71 (t, J=5.8 Hz, 1H), 3.49 (t, J=5.8 Hz, 1H), 3.40 (t, J=5.8 Hz, 1H), 3.02 (t, J=5.8 Hz, 1H), 2.75 (t, J=5.8 Hz, 1H), 2.52 (t, J=5.8 Hz, 1H), 2.25 (t, J=5.8 Hz, 1H); LC/MS: (ES+) m/z (M+H)+=449; HPLC Rt=1.10 min., column P, conditions B.
  • PREPARATION OF EXAMPLE 36 1-(4-((1,3,4-Oxadiazol-2-yl)(phenyl)methylene)piperidin-1-yl)-2-(4-(1H-1,2,4-triazol-1-yl)-5H-pyrrolo[3,2-d]pyrimidin-7-yl)ethane-1,2-dione (Compound 47) EXAMPLE 36
  • Figure US20050124623A1-20050609-C00114
  • In a sealed tube dicarbonyl intermediate 46 (30 mg, 0.07 mmol), 1,2,4-triazole (28 mg, 0.41 mmol), copper(0) (8 mg, 0.13 mmol), K2CO3 (20 mg, 0.14 mmol) and 1,4-dioxane (1 mL) were combined and heated at 140° C. with microwaves for 2 h. The reaction mixture was diluted with MeOH/DMSO (2:3, 1 mL) and purified by preparative HPLC to yield 47 (4 mg, 0.008 mmol, 12%) as a yellow solid. 1H NMR: (500 MHz, CD3OD) δ 9.62 (s, 0.5H), 9.62 (s, 0.5H), 8.87 (s, 0.5H), 8.79 (s, 0.5H), 8.60 (s, 0.5H), 8.58 (s, 0.5H), 8.44 (s, 0.5H), 8.43 (s, 0.5H), 7.48-7.14 (m, 6H), 3.95 (dd, J=6.1, 5.8 Hz, 1H), 3.83 (dd, J=6.1, 5.8 Hz, 1H), 3.68 (dd, J=6.1, 5.5 Hz, 1H), 3.58 (dd, J=5.8, 5.8 Hz, 1H), 3.10 (dd, J=6.1, 5.8 Hz, 1H), 2.90 (dd, J=6.1, 5.5 Hz, 1H), 2.61 (dd, J=6.1, 5.8 Hz, 1H), 2.42 (dd, J=6.1, 5.5 Hz, 1H); LC/MS: (ES+) m/z (M+H)+=482; HPLC Rt=1.13 min., column P, conditions B.
  • PREPARATION OF EXAMPLE 37 1-(4-((1,3,4-Oxadiazol-2-yl)(phenyl)methylene)piperidin-1-yl)-2-(4-(1H-pyrazol-1-yl)-5H-pyrrolo[3,2-d]pyrimidin-7-yl)ethane-1,2-dione (Compound 48) EXAMPLE 37
  • Figure US20050124623A1-20050609-C00115
  • In a sealed tube dicarbonyl intermediate 46 (30 mg, 0.07 mmol), pyrazole (34 mg, 0.5 mmol) and 1,4-dioxane (0.7 mL) were combined and heated at 140° C. with microwaves for 50 min. The reaction mixture was diluted with MeOH/DMSO (1:1, 1.2 mL) and purified by preparative HPLC to yield 48 (10 mg, 0.02 mmol, 32%) as a yellow solid. 1H NMR: (500 MHz, CD3OD) δ 8.86 (s, 1H), 8.85 (s, 0.5H), 8.82 (d, J=2.8 Hz, 0.5H), 8.81 (d, J=2.8 Hz, 0.5H), 8.79 (s, 0.5H), 8.53 (s, 0.5H), 8.51 (s, 0.5H), 8.04 (d, J=1.5 Hz, 0.5H), 8.03 (d, J=1.5 Hz, 0.5H), 7.46-7.16 (m, 5H), 6.71-6.67 (m, 1H), 3.94 (dd, J=6.1, 5.8 Hz, 1), 3.81 (dd, J=6.1, 5.8 Hz, 1H), 3.69 (dd, J=6.1, 5.8 Hz, 1H), 3.57 (dd, J=5.8, 5.8 Hz, 1H), 3.08 (dd, J=5.8, 5.8 Hz, 1H), 2.91 (dd, J=6.1, 5.8 Hz, 1H), 2.59 (dd, J=6.1, 5.8 Hz, 1H), 2.43 (dd, J=6.1, 5.8 Hz, 1H); LC/MS: (ES+) m/z (M+H)+=481; HPLC Rt=1.27 min., column P, conditions B.
  • Preparation of ethyl 4-(1H-pyrazol-1-yl)-5H-pyrrolo[3,2-d]pyrimidine-7-carboxylate 49
  • Figure US20050124623A1-20050609-C00116
  • In a sealed tube 4-chloro-5H-pyrrolo[3,2-d]pyrimidine-7-carbonyl chloride 5 (2.0 g, 8.9 mmol), pyrazole (1.8 g, 26.5 mmol) and 1,4 dioxane (10 mL) were heated at 138° C. for 2 h. Upon cooling to ambient temperature a precipitate formed which was collected and washed with saturated aqueous NaHCO3 and diethyl ether to yield 49 (340 mg, 1.3 mmol) as a white solid. The filtrate was treated with saturated aqueous NaHCO3 (20 ml) and the resulting precipitate was washed with saturated aqueous NaHCO3 and diethyl ether to yield additional 49 (2.0 g, 7.8 mmol, 99% total yield). 1H NMR: (500 MHz, CD3OD) δ 8.99 (br s, 1H), 8.78 (br s, 1H), 8.29 (s, 1H), 8.06 (br s, 1H), 6.72 (br s, 1H), 4.30 (q, J=7.0 Hz, 2H), 1.33 (t, J=7.0 Hz, 3H); LC/MS: (ES+) m/z (M+H)+=258; HPLC Rt=1.20 min., column L.
  • Preparation of 4-(1H-pyrazol-1-yl)-5H-pyrrolo[3,2-d]pyrimidine-7-carboxylic acid 50
  • Figure US20050124623A1-20050609-C00117
  • To a solution of 4-(1H-pyrazol-1-yl)-5H-pyrrolo[3,2-d]pyrimidine-7-carboxylate 49 (2.0 g, 7.8 mmol) in THF (45 mL) was added a solution of LiOH.H2O (1.3 g, 31 mmol) in H2O (30 mL) and the reaction was stirred at 100° C. for 16 h. Additional LiOH.H2O (2.0 g, 48 mmol) was added, heating continued for 2 h, MeOH was added (10 mL) and heating continued at 100° C. for 1 d. The reaction mixture was cooled, filtered, concentrated to 20% volume and neutralized with ice and conc HCl. The white precipitate that formed was collected by filtration and washed with brine, H2O, EtOAc, and Et2O to yield 50 (quantitative), which was used without further purification. 1H NMR: (500 MHz, DMSO-d6) δ 12.07 (br s, 1H), 8.89 (br s, 1H), 8.75 (s, 1H), 8.14 (s, 1H), 8.09 (br s, 1H), 6.76-6.73 (m, 1H); LC/MS: (ES+) m/z (M+H)+=230; HPLC Rt=0.75 min., column S.
  • Preparation of 4-(1H-pyrazol-1-yl)-5H-pyrrolo[3,2-d]pyrimidine-7-carbonyl chloride 51
  • Figure US20050124623A1-20050609-C00118
  • Oxalyl chloride (4.5 mL, 51 mmol) was added to a solution of diazaindole carboxylic acid 50 (1.08 g, 4.7 mmol) in CH2Cl2 (8 mL) and the reaction mixture was stirred 14 h. Catalytic DMF (3 drops) was added to the reaction mixture and after 3 h the reaction was quenched with MeOH. The crude reaction mixture was concentrated to dryness to yield 51 (1.21 g, 49 mmol, 96%) as a tan solid with was used without further purification. 1H NMR: (500 MHz, DMSO-d6) δ 12.45 (br s, 1H), 8.87 (s, 1H), 8.87 (d, J=2.6 Hz, 1H), 8.32 (d, J=3.3 Hz, 1H), 8.14 (d, J=1.5 Hz, 1H), 6.77 (dd, J=2.6, 1.5 Hz, 1H); Methyl ester (obtained by stirring 51 in MeOH): LC/MS: (ES+) m/z (M+H)+=244; HPLC Rt=0.95 min., column O, conditions B.
  • Preparation of 2-(1-(cyanomethyl)piperidin-4-ylidene)-2-(pyridin-2-yl)acetonitrile 52
  • Figure US20050124623A1-20050609-C00119
  • To a solution of 2-(piperidin-4-ylidene)-2-(pyridin-2-yl)acetonitrile (560 mg, 2.8 mmol) in THF (20 mL) was added NEt3 (5 mL, 38 mmol) and chloroacetonitrile (3 mL, 47 mmol) and the reaction was stirred 16 h. The resulting precipitates were filtered away and the filtrate concentrated to dryness. The residues was purified by silica gel chromatography (Biotage 40-short, 50% EtOAc/hexanes to 100% EtOAc/hexanes) to yield 52 (470 mg, 2.0 mmol, 71%) as a yellow solid. 1H NMR: (500 MHz, CDCl3) δ 8.64 (br d, J=4.9 Hz, 1H), 7.77 (ddd, J=7.9, 7.6, 1.9 Hz, 1H), 7.50 (d, J=7.9 Hz, 1H), 7.27 (dd, J=7.6, 4.9 Hz, 1H), 3.58 (s, 2H), 2.94 (t, J=5.8 Hz, 2H), 2.91 (t, J=5.8 Hz, 2H), 2.83 (t, J=5.8 Hz, 2H), 2.67 (t, J=5.8 Hz, 2H); LC/MS: (ES+) m/z (M+H)+=239; HPLC Rt=0.99 min., column O, conditions B.
  • PREPARATION OF EXAMPLE 38 2-(1-(2-(4-(1H-pyrazol-1-yl)-5H-pyrrolo[3,2-d]pyrimidin-7-yl)-2-oxoacetyl)piperidin-4-ylidene)-2-(pyridin-2-yl)acetonitrile (Compound 53) EXAMPLE 38
  • Figure US20050124623A1-20050609-C00120
  • To a slurry of acid chloride pyrazole intermediate 51 (100 mg, 0.40 mmol) and 3-pyridinylcyanoalkene intermediate 52 (100 mg, 0.42 mmol) in THF (4 mL) at −78° C. was added a solution of 0.5 M KHMDS in toluene (3.0 mL, 1.5 mmol). The reaction mixture was stirred at −78° C. for 3 h and the presence of the desired cyanoketone intermediate was verified by LCMS. A solution of 32% peracetic acid in dilute aqueous acetic acid (0.44 mL, 2.1 mmol) was added to the reaction mixture and then allowed to warm to ambient temperature overnight. The reaction mixture was diluted with H2O (5 mL) and saturated aqueous NH4Cl (5 mL) and extracted with EtOAc (3×20 mL). The layers were separated and the aqueous layer extracted with EtOAc (2×20 mL). The combined organic layers were concentrated, the residue was purified by preparative HPLC to yield 53 (7.3 mg, 0.02 mmol, 4%) as a yellow solid. 1H NMR: (500 MHz, CD3OD) δ 8.95 (s, 0.5H), 8.93 (s, 0.5H), 8.88 (d, J=2.8 Hz, 0.5H), 8.86 (d, J=3.1 Hz, 0.5H), 8.71 (br d, J=4.9 Hz, 0.5H), 8.64-8.61 (m, 1H), 8.60 (s, 0.5H), 8.11 (br s, 0.5H), 8.10 (br s, 0.5H), 8.03 (ddd, J=7.9, 7.6, 1.5 Hz, 0.5H), 7.97 (ddd, J=7.9, 7.6, 1.5 Hz, 0.5H), 7.65 (d, J=7.9 Hz, 0.5H), 7.60 (d, J=7.9 Hz, 0.5H), 7.52 (dd, J=7.9, 4.9 Hz, 0.5H), 7.46 (dd, J=7.9, 4.9 Hz, 0.5H), 6.76-6.72 (m, 1H), 4.03 (dd, J=6.1, 6.1 Hz, 1H), 3.86 (dd, J=6.1, 5.8 Hz, 1H), 3.81 (dd, J=6.1, 5.5 Hz, 1H), 3.64 (dd, J=5.8, 5.8 Hz, 1H), 3.10 (dd, J=6.1, 5.8 Hz, 1H), 2.94 (dd, J=6.1, 5.5 Hz, 1H), 2.90 (dd, J=6.1, 5.8 Hz, 1H), 2.75 (dd, J=5.8, 5.8 Hz, 1H); LC/MS: (ES+) m/z (M+H)+=439; HPLC Rt=1.37 min., column O.
  • PREPARATION OF EXAMPLE 39 1-(4-Benzoylpiperazin-1-yl)-2-(4-(3-(pyridin-2-yl)-1H-pyrazol-1-yl)-5H-pyrrolo[3,2-d]pyrimidin-7-yl)ethane-1,2-dione (Compound 54) EXAMPLE 39
  • Figure US20050124623A1-20050609-C00121
  • In a sealed tube dicarbonyl intermediate 10 (52 mg, 0.13 mmol), 2-(1H-pyrazol-3-yl)pyridine (104 mg, 0.71 mmol) and 1,4-dioxane (2.0 mL) were combined and heated at 150° C. with microwaves for 1 h. The reaction mixture was concentrated, diluted with MeOH, filtered and purified by preparative HPLC to yield 54 (31 mg, 0.06 mmol, 47%) as an off-white solid. 1H NMR: (500 MHz, DMSO-d6) δ 12.60 (br s, 1H), 8.97 (d, J=1.8 Hz, 1H), 8.94 (s, 1H), 8.73-8.66 (m, 2H), 8.61 (br s, 1H), 8.02 (t, J=7.0 Hz, 1H), 7.54-7.38 (m, 6H), 7.31 (d, J=1.8 Hz, 1H), 3.92-3.13 (m, 8H); LC/MS: (ES+) m/z (M+H)+=507; HPLC Rt=1.19 min., column N.
  • Preparation of 2-(4-(quinazolin-4-yl)piperazin-1-yl)acetonitrile 55
  • Figure US20050124623A1-20050609-C00122
  • To a solution of 4-(piperazin-1-yl)quinazoline (1.8 g, 8.3 mmol) in THF (50 mL) was added NEt3 (20 mL, 150 mmol) and chloroacetonitrile (12 mL, 190 mmol) and the reaction was stirred 16 h. The reaction mixture was quenched with 50% saturated aqueous NaHCO3 and extracted with EtOAc (3×200 mL). The combined organics were purified by silica gel chromatography (50% EtOAc/hexanes to 80% EtOAc/hexanes) to yield 55 (1.6 g, 6.1 mmol, 73%) as an viscous yellow oil. 1H NMR: (500 MHz, CDCl3) δ 8.74 (s, 1H), 8.06 (d, J=8.6 Hz, 1H), 7.90 (d, J=8.5 Hz, 1H), 7.82-7.55 (m, 1H), 7.54-7.50 (m, 1H), 3.94-3.88 (m, 4H), 3.63 (s, 2H), 2.86-2.81 (m, 4H); LC/MS: (ES+) m/z (M+H)+=254; HPLC Rt=0.71 min., column O.
  • PREPARATION OF EXAMPLE 40 1-(4-(1H-Pyrazol-1-yl)-5H-pyrrolo[3,2-d]pyrimidin-7-yl)-2-(4-(quinazolin-4-yl)piperazin-1-yl)ethane-1,2-dione (Compound 56) EXAMPLE 40
  • Figure US20050124623A1-20050609-C00123
  • To a slurry of acid chloride pyrazole intermediate 51 (100 mg, 0.40 mmol) and 2-(4-(quinazolin-4-yl)piperazin-1-yl)acetonitrile 55 (98 mg, 0.39 mmol) in THF (4 mL), at −78° C. was added a solution of 0.5 M KHMDS in toluene (3.0 mL, 1.5 mmol). The reaction mixture was stirred 3 h and the presence of the desired cyanoketone intermediate was verified by LCMS. The reaction was treated with a solution of 32% peracetic acid in dilute aqueous acetic acid (0.40 mL, 1.9 mmol) and then allowed to warm to ambient temperature overnight. The reaction mixture was diluted with saturated aqueous NH4Cl (5 mL) and extracted with EtOAc (3×20 mL). The combined organic layers were concentrated, the residue was purified by preparative HPLC to yield 56 (33 mg, 0.07 mmol, 18%) as a yellow solid. 1H NMR: (500 MHz, CD3OD) δ 8.86 (s, 1H), 8.5 (d, J=2.7 Hz, 1H), 8.75 (s, 1H), 8.61 (s, 1H), 8.30 (d, J=8.2 Hz, 1H), 8.09-8.05 (m, 2H), 7.84 (d, J=8.5 Hz, 1H), 7.79 (dd, J=8.2, 7.6 Hz, 1H), 6.72 (br s, 1H), 4.62-4.56 (m, 2H), 4.43-4.38 (m, 2H), 4.12-4.07 (m, 2H), 3.91-3.86 (m, 2H); LC/MS: (ES+) m/z (M+H)+=439; HPLC Rt=1.06 min., column O.
  • PREPARATION OF EXAMPLE 41 1-(4-(4-(1H-Pyrazole-3-carbonyl)piperazin-1-yl)-5H-pyrrolo[3,2-d]pyrimidin-7-yl)-2-(4-benzoylpiperazin-1-yl)ethane-1,2-dione (Compound 57) EXAMPLE 41
  • Figure US20050124623A1-20050609-C00124
  • In a sealed tube dicarbonyl intermediate 10 (50 mg, 0.13 mmol), (4-methylpiperazin-1-yl)(1H-pyrazol-3-yl)methanone (75 mg, 0.39 mmol) and copper powder (10 mg, 0.16 mmol) were combined and heated at 150° C. with microwaves for 2 h. The reaction was purified by preparative HPLC to yield 57 (65 mg, 0.12 mmol, 92%) as a white solid. 1H NMR: (500 MHz, CD3COCD3) δ 10.31 (br s, 2H), 8.82 (s, 1H), 8.85 (s, 1H), 7.80 (s, 1H), 7.50-7.40 (m, 5H), 6.74 (s, 1H), 4.50-4.37 (m, 6H), 4.09-3.96 (m, 2H), 3.86-3.60 (m, 8H); LC/MS: (ES+) m/z (M+H)+=542; HPLC Rt=0.85 min., column G, conditions B.
  • PREPARATION OF EXAMPLE 42 2-(1-(2-(4-(1H-Pyrazol-1-yl)-5H-pyrrolo[3,2-d]pyrimidin-7-yl)-2-oxoacetyl)piperidin-4-ylidene)-2-phenylacetonitrile (Compound 58) EXAMPLE 42
  • Figure US20050124623A1-20050609-C00125
  • In a sealed tube dicarbonyl intermediate 33 (31 mg, 0.077 mmol), pyrazole (20 mg, 0.29 mmol) and 1,4-dioxane (0.8 mL) were combined and heated at 140° C. with microwaves for 50 min. The reaction mixture was diluted with MeOH/DMSO (2:1, 1.5 mL), filtered and purified by preparative HPLC to yield 58 (10 mg, 0.024 mmol, 31%) as an orange solid. 1H NMR: (500 MHz, CD3OD) δ 8.90 (s, 0.5H), 8.88 (s, 0.5H), 8.86 (d, J=2.8 Hz, 0.5H), 8.84 (d, J=2.8 Hz, 0.5H), 8.57 (s, 0.5H), 8.54 (s, 0.5H), 8.08 (br s, 0.5H), 8.06 (br s, 0.5H), 7.53-7.31 (m, 5H), 6.74-6.70 (m, 1H), 4.00 (t, J=5.8 Hz, 1H), 3.82 (t, J=5.8 Hz, 1H), 3.75 (t, J=5.8 Hz, 1H), 3.56 (t, J=5.8 Hz, 1H), 3.05 (t, J=5.8 Hz, 1H), 2.87 (t, J=5.8 Hz, 1H), 2.72 (t, J=5.8 Hz, 1H), 2.55 (t, J=5.8 Hz, 1H); LC/MS: (ES+) m/z (M+H)+=438; HPLC Rt=1.73 min., column O.
  • PREPARATION OF EXAMPLE 43 2-(1-(2-(4-(1H-pyrazol-3-yl)-5H-pyrrolo[3,2-d]pyrimidin-7-yl)-2-oxoacetyl)piperidin-4-ylidene)-2-phenylacetonitrile (Compound 59) EXAMPLE 43
  • Figure US20050124623A1-20050609-C00126
  • In a sealed tube dicarbonyl intermediate 33 (33 mg, 0.081 mmol), 3-(tributylstannyl)-1H-pyrazole (73 mg, 0.20 mmol), tetrakis(triphenylphosphine)-palladium(0) (20 mg, 0.02 mmol) and 1,4-dioxane (0.8 mL) were combined and heated at 130° C. with microwaves for 2 h. The reaction mixture was diluted with MeOH/DMSO (2:1, 1.5 mL), filtered and purified by preparative HPLC to yield 59 (3.4 mg, 0.008 mmol, 10%) as a yellow solid. 1H NMR: (500 MHz, CD3COCD3) δ 11.84 (br s, 1H), 9.06 (s, 0.5H), 9.05 (s, 0.5H), 8.58 (s, 0.5H), 8.55 (s, 0.5H), 8.01 (d, J=2.7 Hz, 0.5H), 8.01 (d, J=2.7 Hz, 0.5H), 7.55-7.32 (m, 5H), 7.24 (d, J=2.7 Hz, 0.5H), 7.23 (d, J=2.7 Hz, 0.5H), 4.01 (t, J=5.8 Hz, 1H), 3.84 (t, J=5.8 Hz, 1H), 3.74 (t, J=5.8 Hz, 1H), 3.56 (t, J=5.8 Hz, 1H), 3.07 (t, J=5.8 Hz, 1H), 2.84 (t, J=5.8 Hz, 1H), 2.78 (t, J=5.8 Hz, 1H), 2.54 (t, J=5.8 Hz, 1H); LC/MS: (ES+) m/z (M+H)+=438; HPLC Rt=1.29 min., column L.
  • Biology
    • ”μM” means micromolar;
    • “mL” means milliliter;
    • “μl” means microliter;
    • “mg” means milligram;
  • The materials and experimental procedures used to obtain the results reported in Tables 1-3 are described below.
  • Cells:
    • Virus production-Human embryonic Kidney cell line, 293, propagated in Dulbecco's Modified Eagle Medium (Life Technologies, Gaithersburg, Md.) containing 10% fetal Bovine serum (FBS, Sigma, St. Louis, Mo.).
    • Virus infection—Human epithelial cell line, HeLa, expressing the HIV-1 receptors CD4 and CCR5 was propagated in Dulbecco's Modified Eagle Medium (Life Technologies, Gaithersburg, Md.) containing 10% fetal Bovine serum (FBS, Sigma, St. Louis, Mo.) and supplemented with 0.2 mg/mL Geneticin (Life Technologies, Gaithersburg, Md.) and 0.4 mg/mL Zeocin (Invitrogen, Carlsbad, Calif.).
    • Virus-Single-round infectious reporter virus was produced by co-transfecting human embryonic Kidney 293 cells with an HIV-1 envelope DNA expression vector and a proviral cDNA containing an envelope deletion mutation and the luciferase reporter gene inserted in place of HIV-1 nef sequences (Chen et al, Ref. 41). Transfections were performed using lipofectAMINE PLUS reagent as described by the manufacturer (Life Technologies, Gaithersburg, Md.).
      Experiment
    • 1. Compound was added to HeLa CD4 CCR5 cells plated in 96 well plates at a cell density of 1×104 cells per well in 100 μl Dulbecco's Modified Eagle Medium containing 10% fetal Bovine serum at a concentration of <20 μM.
    • 2. 100 μl of single-round infectious reporter virus in Dulbecco's Modified Eagle Medium was then added to the plated cells and compound at an approximate multiplicity of infection (MOI) of 0.01, resulting in a final volume of 200 μl per well and a final compound concentration of <10 μM.
    • 3. Samples were harvested 72 h after infection.
    • 4. Viral infection was monitored by measuring luciferase expression from viral DNA in the infected cells using a luciferase reporter gene assay kit (Roche Molecular Biochemicals, Indianapolis, Ind.). Infected cell supernatants were removed and 50 μl of Dulbecco's Modified Eagle Medium (without phenol red) and 50 μl of luciferase assay reagent reconstituted as described by the manufacturer (Roche Molecular Biochemicals, Indianapolis, Ind.) was added per well. Luciferase activity was then quantified by measuring luminescence using a Wallac microbeta scintillation counter.
    • 5. The percent inhibition for each compound was calculated by quantifying the level of luciferase expression in cells infected in the presence of each compound as a percentage of that observed for cells infected in the absence of compound and subtracting such a determined value from 100.
    • 6. An EC50 provides a method for comparing the antiviral potency of the compounds of this invention. The effective concentration for fifty percent inhibition (EC50) was calculated with the Microsoft Excel Xlfit curve fitting software. For each compound, curves were generated from percent inhibition calculated at 10 different concentrations by using a four paramenter logistic model (model 205). The EC50 data for the compounds is shown in Tables 2-4. Table 1 is the key for the data in Tables 2-4.
  • Cytoxicity assays were conducted with the same HeLa using methodology well known in the art. This method has been described in the literature (S Weislow, R Kiser, D L Fine, J Bader, R H Shoemaker and M R Boyd: New soluble-formazan assay for HIV-1 cytopathic effects: application to high-flux screening of synthetic and natural products for AIDS-antiviral activity. Journal of the National Cancer Institute. 81(8):577-586, 1989.
  • Cells were incubated in the presence of drug for six days, after which cell viability was measured using a dye reduction assay (MTT) and determined as a CC50. This assay measures the intracellular reducing activity present in actively respiring cells.
  • Results
    TABLE 1
    Biological Data Key for EC50s
    Compounds with
    Compounds EC50 >50 nM
    Compounds* with but not yet Compounds
    with EC50s >1 μM tested at higher with
    EC50s >5 μM but <5 μM concentrations EC50 <1 μM
    Group C Group B Group A′ Group A

    *Some of these compounds may have been tested at a concentration lower than their EC50 but showed some ability to cause inhibition and thus should be evaluated at a higher concentration to determine the exact EC50.
  • TABLE 2
    Figure US20050124623A1-20050609-C00127
    Examples
    EC50
    Group
    from
    Example Compound Table
    Number Number R3 R5 Y 1
    Example 2 10 H Cl
    Figure US20050124623A1-20050609-C00128
    A
    Example 3 11 H
    Figure US20050124623A1-20050609-C00129
    Figure US20050124623A1-20050609-C00130
    A
    Example 4 12 H
    Figure US20050124623A1-20050609-C00131
    Figure US20050124623A1-20050609-C00132
    A
    Example 5 13 H
    Figure US20050124623A1-20050609-C00133
    Figure US20050124623A1-20050609-C00134
    A
    Example 6 14 H
    Figure US20050124623A1-20050609-C00135
    Figure US20050124623A1-20050609-C00136
    A
    Example 7 15 H
    Figure US20050124623A1-20050609-C00137
    Figure US20050124623A1-20050609-C00138
    A
    Example 8 16 H
    Figure US20050124623A1-20050609-C00139
    Figure US20050124623A1-20050609-C00140
    A
    Example 9 17 H
    Figure US20050124623A1-20050609-C00141
    Figure US20050124623A1-20050609-C00142
    A
  • TABLE 3
    Figure US20050124623A1-20050609-C00143
    Examples
    EC50
    Group
    Example Cmpd. from
    No. No. R3 R5 Y Table 1
    Example 1 9 H Cl
    Figure US20050124623A1-20050609-C00144
    A
    Example 10 18 H
    Figure US20050124623A1-20050609-C00145
    Figure US20050124623A1-20050609-C00146
    A
    Example 11 19 H
    Figure US20050124623A1-20050609-C00147
    Figure US20050124623A1-20050609-C00148
    A
    Example 12 20 H
    Figure US20050124623A1-20050609-C00149
    Figure US20050124623A1-20050609-C00150
    A
    Example 13 21 H
    Figure US20050124623A1-20050609-C00151
    Figure US20050124623A1-20050609-C00152
    A
    Example 14 22 H
    Figure US20050124623A1-20050609-C00153
    Figure US20050124623A1-20050609-C00154
    A
    Example 15 23 H
    Figure US20050124623A1-20050609-C00155
    Figure US20050124623A1-20050609-C00156
    A
    Example 16 24 H
    Figure US20050124623A1-20050609-C00157
    Figure US20050124623A1-20050609-C00158
    A
    Example 17 25 H
    Figure US20050124623A1-20050609-C00159
    Figure US20050124623A1-20050609-C00160
    A
    Example 18 26 H
    Figure US20050124623A1-20050609-C00161
    Figure US20050124623A1-20050609-C00162
    A
    Example 19 27 H
    Figure US20050124623A1-20050609-C00163
    Figure US20050124623A1-20050609-C00164
    A
    Example 20 28 H
    Figure US20050124623A1-20050609-C00165
    Figure US20050124623A1-20050609-C00166
    A
    Example 21 29 H
    Figure US20050124623A1-20050609-C00167
    Figure US20050124623A1-20050609-C00168
    A
    Example 22 30 H
    Figure US20050124623A1-20050609-C00169
    Figure US20050124623A1-20050609-C00170
    A
    Example 22 30 H
    Figure US20050124623A1-20050609-C00171
    Figure US20050124623A1-20050609-C00172
    A
    Example 24 32 H
    Figure US20050124623A1-20050609-C00173
    Figure US20050124623A1-20050609-C00174
    A
    Example 25 33 H
    Figure US20050124623A1-20050609-C00175
    Figure US20050124623A1-20050609-C00176
    A
    Example 26 34 H
    Figure US20050124623A1-20050609-C00177
    Figure US20050124623A1-20050609-C00178
    A
    Example 27 35 H
    Figure US20050124623A1-20050609-C00179
    Figure US20050124623A1-20050609-C00180
    A
    Example 28 36 H
    Figure US20050124623A1-20050609-C00181
    Figure US20050124623A1-20050609-C00182
    A
    Example 29 37 H
    Figure US20050124623A1-20050609-C00183
    Figure US20050124623A1-20050609-C00184
    A
    Example 30 39 H Cl
    Figure US20050124623A1-20050609-C00185
    A
    Example 31 40 H
    Figure US20050124623A1-20050609-C00186
    Figure US20050124623A1-20050609-C00187
    A
    Example 32 41 H
    Figure US20050124623A1-20050609-C00188
    Figure US20050124623A1-20050609-C00189
    A
    Example 33 43 H Cl
    Figure US20050124623A1-20050609-C00190
    A
    Example 34 44 H
    Figure US20050124623A1-20050609-C00191
    Figure US20050124623A1-20050609-C00192
    A
    Example 35 46 H Cl
    Figure US20050124623A1-20050609-C00193
    A
    Example 36 47 H
    Figure US20050124623A1-20050609-C00194
    Figure US20050124623A1-20050609-C00195
    A
    Example 37 48 H
    Figure US20050124623A1-20050609-C00196
    Figure US20050124623A1-20050609-C00197
    A
    Example 38 53 H
    Figure US20050124623A1-20050609-C00198
    Figure US20050124623A1-20050609-C00199
    A
    Example 39 54 H
    Figure US20050124623A1-20050609-C00200
    Figure US20050124623A1-20050609-C00201
    A
    Example 40 56 H
    Figure US20050124623A1-20050609-C00202
    Figure US20050124623A1-20050609-C00203
    A
    Example 41 57 H
    Figure US20050124623A1-20050609-C00204
    Figure US20050124623A1-20050609-C00205
    A
    Example 42 58 H
    Figure US20050124623A1-20050609-C00206
    Figure US20050124623A1-20050609-C00207
    A
    Example 43 59 H
    Figure US20050124623A1-20050609-C00208
    Figure US20050124623A1-20050609-C00209
    A
  • Table 4 shows other compounds of the invention which could be prepared by the methodology described herein and which are expected to have antiviral activity.
    TABLE 4
    Figure US20050124623A1-20050609-C00210
    EC50
    Group
    Example Cmpd. from
    No. No. R3 R5 Y Table 1
    Example # # H
    Figure US20050124623A1-20050609-C00211
    Figure US20050124623A1-20050609-C00212
    H
    Figure US20050124623A1-20050609-C00213
    Figure US20050124623A1-20050609-C00214
    H
    Figure US20050124623A1-20050609-C00215
    Figure US20050124623A1-20050609-C00216
    H
    Figure US20050124623A1-20050609-C00217
    Figure US20050124623A1-20050609-C00218
    H
    Figure US20050124623A1-20050609-C00219
    Figure US20050124623A1-20050609-C00220
    H
    Figure US20050124623A1-20050609-C00221
    Figure US20050124623A1-20050609-C00222
    H
    Figure US20050124623A1-20050609-C00223
    Figure US20050124623A1-20050609-C00224
    H
    Figure US20050124623A1-20050609-C00225
    Figure US20050124623A1-20050609-C00226
    Figure US20050124623A1-20050609-C00227
    Figure US20050124623A1-20050609-C00228
    H
    Figure US20050124623A1-20050609-C00229
    Figure US20050124623A1-20050609-C00230
    H
    Figure US20050124623A1-20050609-C00231
    Figure US20050124623A1-20050609-C00232
    H
    Figure US20050124623A1-20050609-C00233
    Figure US20050124623A1-20050609-C00234
    H
    Figure US20050124623A1-20050609-C00235
    Figure US20050124623A1-20050609-C00236
    H
    Figure US20050124623A1-20050609-C00237
    Figure US20050124623A1-20050609-C00238
    H
    Figure US20050124623A1-20050609-C00239
    Figure US20050124623A1-20050609-C00240
    H
    Figure US20050124623A1-20050609-C00241
    Figure US20050124623A1-20050609-C00242
    H
    Figure US20050124623A1-20050609-C00243
    Figure US20050124623A1-20050609-C00244
    H
    Figure US20050124623A1-20050609-C00245
    Figure US20050124623A1-20050609-C00246
    H
    Figure US20050124623A1-20050609-C00247
    Figure US20050124623A1-20050609-C00248
    H
    Figure US20050124623A1-20050609-C00249
    Figure US20050124623A1-20050609-C00250
    H
    Figure US20050124623A1-20050609-C00251
    Figure US20050124623A1-20050609-C00252
    H
    Figure US20050124623A1-20050609-C00253
    Figure US20050124623A1-20050609-C00254
    H
    Figure US20050124623A1-20050609-C00255
    Figure US20050124623A1-20050609-C00256
    H
    Figure US20050124623A1-20050609-C00257
    Figure US20050124623A1-20050609-C00258
    H
    Figure US20050124623A1-20050609-C00259
    Figure US20050124623A1-20050609-C00260
    H
    Figure US20050124623A1-20050609-C00261
    Figure US20050124623A1-20050609-C00262
    H
    Figure US20050124623A1-20050609-C00263
    Figure US20050124623A1-20050609-C00264
    H
    Figure US20050124623A1-20050609-C00265
    Figure US20050124623A1-20050609-C00266
    H
    Figure US20050124623A1-20050609-C00267
    Figure US20050124623A1-20050609-C00268
    H
    Figure US20050124623A1-20050609-C00269
    Figure US20050124623A1-20050609-C00270
    H
    Figure US20050124623A1-20050609-C00271
    Figure US20050124623A1-20050609-C00272
    H
    Figure US20050124623A1-20050609-C00273
    Figure US20050124623A1-20050609-C00274
    H
    Figure US20050124623A1-20050609-C00275
    Figure US20050124623A1-20050609-C00276
    H
    Figure US20050124623A1-20050609-C00277
    Figure US20050124623A1-20050609-C00278
    H
    Figure US20050124623A1-20050609-C00279
    Figure US20050124623A1-20050609-C00280
    H
    Figure US20050124623A1-20050609-C00281
    Figure US20050124623A1-20050609-C00282
    Figure US20050124623A1-20050609-C00283
    Figure US20050124623A1-20050609-C00284
    H
    Figure US20050124623A1-20050609-C00285
    Figure US20050124623A1-20050609-C00286
    H
    Figure US20050124623A1-20050609-C00287
    Figure US20050124623A1-20050609-C00288
    H
    Figure US20050124623A1-20050609-C00289
    Figure US20050124623A1-20050609-C00290
    H
    Figure US20050124623A1-20050609-C00291
    Figure US20050124623A1-20050609-C00292
    H
    Figure US20050124623A1-20050609-C00293
    Figure US20050124623A1-20050609-C00294
    H
    Figure US20050124623A1-20050609-C00295
    Figure US20050124623A1-20050609-C00296
    H
    Figure US20050124623A1-20050609-C00297
    Figure US20050124623A1-20050609-C00298
    H
    Figure US20050124623A1-20050609-C00299
    Figure US20050124623A1-20050609-C00300
    H
    Figure US20050124623A1-20050609-C00301
    Figure US20050124623A1-20050609-C00302
    H
    Figure US20050124623A1-20050609-C00303
    Figure US20050124623A1-20050609-C00304
    H
    Figure US20050124623A1-20050609-C00305
    Figure US20050124623A1-20050609-C00306
    H
    Figure US20050124623A1-20050609-C00307
    Figure US20050124623A1-20050609-C00308
    H
    Figure US20050124623A1-20050609-C00309
    Figure US20050124623A1-20050609-C00310
    H
    Figure US20050124623A1-20050609-C00311
    Figure US20050124623A1-20050609-C00312
    H
    Figure US20050124623A1-20050609-C00313
    Figure US20050124623A1-20050609-C00314
    H
    Figure US20050124623A1-20050609-C00315
    Figure US20050124623A1-20050609-C00316
    H
    Figure US20050124623A1-20050609-C00317
    Figure US20050124623A1-20050609-C00318
    H
    Figure US20050124623A1-20050609-C00319
    Figure US20050124623A1-20050609-C00320
    Figure US20050124623A1-20050609-C00321
    Figure US20050124623A1-20050609-C00322
    H
    Figure US20050124623A1-20050609-C00323
    Figure US20050124623A1-20050609-C00324
    H
    Figure US20050124623A1-20050609-C00325
    Figure US20050124623A1-20050609-C00326
    H
    Figure US20050124623A1-20050609-C00327
    Figure US20050124623A1-20050609-C00328
    H
    Figure US20050124623A1-20050609-C00329
    Figure US20050124623A1-20050609-C00330
    H
    Figure US20050124623A1-20050609-C00331
    Figure US20050124623A1-20050609-C00332
    H
    Figure US20050124623A1-20050609-C00333
    Figure US20050124623A1-20050609-C00334
    H
    Figure US20050124623A1-20050609-C00335
    Figure US20050124623A1-20050609-C00336
    H
    Figure US20050124623A1-20050609-C00337
    Figure US20050124623A1-20050609-C00338
    Figure US20050124623A1-20050609-C00339
    Figure US20050124623A1-20050609-C00340
    H
    Figure US20050124623A1-20050609-C00341
    Figure US20050124623A1-20050609-C00342
    H
    Figure US20050124623A1-20050609-C00343
    Figure US20050124623A1-20050609-C00344
    H
    Figure US20050124623A1-20050609-C00345
    Figure US20050124623A1-20050609-C00346
    H
    Figure US20050124623A1-20050609-C00347
    Figure US20050124623A1-20050609-C00348
    H
    Figure US20050124623A1-20050609-C00349
    Figure US20050124623A1-20050609-C00350
    H
    Figure US20050124623A1-20050609-C00351
    Figure US20050124623A1-20050609-C00352
    H
    Figure US20050124623A1-20050609-C00353
    Figure US20050124623A1-20050609-C00354
    H
    Figure US20050124623A1-20050609-C00355
    Figure US20050124623A1-20050609-C00356
    H
    Figure US20050124623A1-20050609-C00357
    Figure US20050124623A1-20050609-C00358
    H
    Figure US20050124623A1-20050609-C00359
    Figure US20050124623A1-20050609-C00360
    H
    Figure US20050124623A1-20050609-C00361
    Figure US20050124623A1-20050609-C00362
    H
    Figure US20050124623A1-20050609-C00363
    Figure US20050124623A1-20050609-C00364
    H
    Figure US20050124623A1-20050609-C00365
    Figure US20050124623A1-20050609-C00366
    H
    Figure US20050124623A1-20050609-C00367
    Figure US20050124623A1-20050609-C00368
    H
    Figure US20050124623A1-20050609-C00369
    Figure US20050124623A1-20050609-C00370
    H
    Figure US20050124623A1-20050609-C00371
    Figure US20050124623A1-20050609-C00372
    H
    Figure US20050124623A1-20050609-C00373
    Figure US20050124623A1-20050609-C00374
    H
    Figure US20050124623A1-20050609-C00375
    Figure US20050124623A1-20050609-C00376
    H
    Figure US20050124623A1-20050609-C00377
    Figure US20050124623A1-20050609-C00378
  • The compounds of the present invention may be administered orally, parenterally (including subcutaneous injections, intravenous, intramuscular, intrasternal injection or infusion techniques), by inhalation spray, or rectally, in dosage unit formulations containing conventional non-toxic pharmaceutically-acceptable carriers, adjuvants and vehicles.
  • Thus, in accordance with the present invention there is further provided a method of treating and a pharmaceutical composition for treating viral infections such as HIV infection and AIDS. The treatment involves administering to a patient in need of such treatment a pharmaceutical composition comprising a pharmaceutical carrier and a therapeutically-effective amount of a compound of the present invention.
  • The pharmaceutical composition may be in the form of orally-administrable suspensions or tablets; nasal sprays, sterile injectable preparations, for example, as sterile injectable aqueous or oleagenous suspensions or suppositories.
  • When administered orally as a suspension, these compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may contain microcrystalline cellulose for imparting bulk, alginic acid or sodium alginate as a suspending agent, methylcellulose as a viscosity enhancer, and sweetners/flavoring agents known in the art. As immediate release tablets, these compositions may contain microcrystalline cellulose, dicalcium phosphate, starch, magnesium stearate and lactose and/or other excipients, binders, extenders, disintegrants, diluents and lubricants known in the art.
  • The injectable solutions or suspensions may be formulated according to known art, using suitable non-toxic, parenterally-acceptable diluents or solvents, such as mannitol, 1,3-butanediol, water, Ringer's solution or isotonic sodium chloride solution, or suitable dispersing or wetting and suspending agents, such as sterile, bland, fixed oils, including synthetic mono- or diglycerides, and fatty acids, including oleic acid.
  • The compounds of this invention can be administered orally to humans in a dosage range of 1 to 100 mg/kg body weight in divided doses. One preferred dosage range is 1 to 10 mg/kg body weight orally in divided doses. Another preferred dosage range is 1 to 20 mg/kg body weight orally in divided doses. It will be understood, however, that the specific dose level and frequency of dosage for any particular patient may be varied and will depend upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the host undergoing therapy.

Claims (28)

1. A compound of Formula I, including pharmaceutically acceptable salts thereof,
Figure US20050124623A1-20050609-C00379
wherein:
Q is selected from the group consisting of
Figure US20050124623A1-20050609-C00380
T is —C(O)— or —CH(CN)—;
R1 is hydrogen or methyl;
R3 and R5 are independently selected from the group consisting of hydrogen, halogen, cyano, nitro, COOR8, XR9 and B;
R2 and R4 are independently O or do not exist with the proviso that only one of R2 and R4 are O;
R6 is (CH2)nH, wherein n is 0-1;
— represents a carbon-carbon bond or does not exist;
—Y— is selected from the group consisting of
Figure US20050124623A1-20050609-C00381
R10, R11, R12, R13, R14,R15, R16 and R17 are each independently H or (C1-6)alkyl; wherein said (C1-6)alkyl may optionally be substituted with one to three same or different halogen, OH or CN;
R18 is a member selected from the group consisting of C(O)-phenyl, C(O)-heteroaryl, pyridinyl, pyrimidinyl, quinolyl, isoquinolyl, quinazolyl, quinoxalinyl, napthyridinyl, pthalazinyl, azabenzofuryl and azaindolyl; wherein said member is optionally substituted with from one to two substituents selected from the group consisting of methyl, -amino, —NHMe, —NMe2, methoxy, hydroxymethyl and halogen;
D is selected from the group consisting of hydrogen, cyano, S(O)2R24, halogen, COOR20, C(O)NR21R22, phenyl and heteroaryl; wherein said phenyl or heteroaryl is independently optionally substituted with one to three same or different halogens or from one to three same or different substituents selected from F (as defined below);
A is selected from the group consisting of phenyl, pyridinyl, furanyl, thienyl, isoxazole and oxazole; wherein said phenyl, pyridinyl, furanyl, thienyl, isoxazole or oxazole is independently optionally substituted with one to three same or different halogens or from one to three same or different substituents selected from F;
B is selected from the group consisting of (C1-6)alkyl, C(O)NR21R22, —C(O)CH3, —N(CH2CH2)2NC(O)pyrazolyl, phenyl and heteroaryl; wherein said (C1-6)alkyl, phenyl and heteroaryl are independently optionally substituted with one to three same or different halogens or from one to three same or different substituents selected from F;
heteroaryl is selected from the group consisting of pyridinyl, pyrazinyl, pyridazinyl, pyrimidinyl, furanyl, thienyl, benzothienyl, thiazolyl, oxazolyl, isoxazolyl, imidazolyl, oxadiazolyl, thiadiazolyl, pyrazolyl, tetrazolyl and triazolyl;
F is selected from the group consisting of (C1-6)alkyl, (C1-6)alkenyl, phenyl, pyridinyl, hydroxy, (C1-6)alkoxy, halogen, benzyl, —NR23C(O)—(C1-6)alkyl, —NR24R25, —S(O)2NR24R25, COOR26, —COR27, and —CONR24R25; wherein said (C1-6)alkyl or phenyl are each optionally substituted with hydroxy, (C1-6)alkoxy, (C1-6)alkyl, CF3, dimethylamino or from one to three same or different halogen;
R8, R9 and R26 are each independently selected from the group consisting of hydrogen and (C1-6)alkyl;
X is selected from the group consisting of NR26, O and S;
R20, R21,R22, R23, R24 and R25 are independently selected from the group consisting of hydrogen, (C1-6)alkyl, phenyl and heteroaryl; wherein said phenyl and heteroaryl are each independently optionally substituted with one to three same or different halogen or methyl; and
R27 is piperazinyl, N-methyl piperazinyl, or 3-pyrazolyl.
2. A compound of claim 1 wherein T is
Figure US20050124623A1-20050609-C00382
3. A compound of claim 2, wherein:
R1 is hydrogen;
— represents a carbon-carbon bond; and
R2 and R4 do not exist.
D is selected from the group consisting of hydrogen, cyano, S(O)2R24, halogen, COOR20, C(O)NH2, phenyl and heteroaryl; wherein said phenyl or heteroaryl is independently optionally substituted with one to three same or different halogens or a member selected from the group consisting of (C1-6)alkyl, (C1-6)alkenyl, hydroxy, (C1-6)alkoxy, halogen, —NR24R25, —S(O)2NR24R25, COOR26 and —CONR24R25; wherein said (C1-6)alkyl is optionally substituted with one to three same or different halogen or a hydroxy; and
A is selected from the group consisting of phenyl, pyridinyl, furanyl, thienyl, isoxazole and oxazole; wherein said phenyl, pyridinyl, furanyl, thienyl, isoxazole or oxazole are independently optionally substituted with one to three same or different halogens or a member selected from the group consisting of (C1-4)alkyl, (C1-4)alkenyl, (C1-3)alkoxy, halogen and —NH2; wherein said (C1-4)alkyl is optionally substituted with one to three same or different halogens.
4. A compound of claim 3 wherein:
R6 is hydrogen; and
R10, R11, R12, R13, R14, R15, R16 and R17 are each independently H or methyl, with the proviso that a maximum of two of R10—R17 is a methyl.
5. A compound of claim 4 wherein:
Q is a member selected from groups (A), (B), and (C) consisting of
(A)
Figure US20050124623A1-20050609-C00383
wherein R3 is hydrogen, C1-C3 alkoxy, —NR26R9 or halogen;
(B)
Figure US20050124623A1-20050609-C00384
wherein R3 is hydrogen, methoxy or halogen; and
(C)
Figure US20050124623A1-20050609-C00385
wherein R3 is hydrogen, methoxy or halogen.
6. A compound of claim 5 wherein:
group(A) of Q is:
Figure US20050124623A1-20050609-C00386
wherein R3 is hydrogen; and
group (C) of Q is:
Figure US20050124623A1-20050609-C00387
wherein
R5 is hydrogen.
7. A compound of claim 5 wherein:
Q is selected from group (A) or (B); and
R5 is selected from the group consisting of hydrogen, halogen, heteroaryl, phenyl, cyano, methoxy, COOR8, C(O)NH2, C(O)NHheteroaryl, and C(O)NHCH3; wherein said C(O)NHheteroaryl, phenyl, and heteroaryl are independently optionally substituted with one to three same or different halogens or from one to three same or different substituents selected from F.
8. A compound of claim 7 wherein:
R5 is selected from the group consisting of C(O)NH2, C(O)NHCH3 and C(O)NHheteroaryl; wherein said C(O)NHheteroaryl is optionally substituted with one to three same or different halogens or from one to three same or different substituents selected from F.
9. A compound of claim 7 wherein:
R5 is selected from the group consisting of phenyl and heteroaryl; wherein said phenyl and heteroaryl are independently optionally substituted with one to three same or different halogens or from one to three same or different substituents selected from F.
10. A compound of claim 9 wherein:
heteroaryl is selected from the group consisting of pyridinyl, pyrazinyl, pyridazinyl, pyrimidinyl, furanyl, thienyl, thiazolyl, oxazolyl, isoxazolyl, imidazolyl, oxadiazolyl, thiadiazoyl, pyrazolyl, tetrazolyl and triazolyl; wherein said heteroaryl is independently optionally substituted with one to three same or different halogens or from one to three same or different substituents selected from F.
11. A compound of claim 5, wherein:
R18 is —C(O)phenyl or —C(O)heteroaryl; wherein said heteroaryl is pyridinyl, furanyl or thienyl; wherein heteroaryl is independently optionally substituted with a member selected from the group consisting of halogen, methyl, -amino, —NHMe, NMe2 and hydroxymethyl.
12. A compound of claim 5 wherein:
—W— is
Figure US20050124623A1-20050609-C00388
R10, R11, R12, R13, R14,R15, R16 and R17 are each independently H or methyl, with the proviso that not more than one is methyl; and
R18 is selected from the group consisting of C(O)-phenyl and C(O)-heteroaryl; wherein each of C(O)-phenyl or —C(O)-heteroaryl may be optionally substituted with from one to two methyl, -amino, —NHMe, —NMe2, methoxy, hydroxymethyl or halogen.
13. A compound of claim 5 wherein:
—W— is
Figure US20050124623A1-20050609-C00389
R10, R11, R12, R13, R14,R15, R16 and R17 are each independently H or methyl, with the proviso that not more than one is methyl; and
R18 is selected from the group consisting of pyridinyl, pyrimidinyl, quinolyl, isoquinolyl, quinazolyl, quinoxalinyl, napthyridinyl, pthalazinyl, azabenzofuryl and azaindolyl, each of which may be optionally substituted with from one to two substituents selected from the group consisting of methyl, -amino, —NHMe, —NMe2, methoxy, hydroxymethyl and halogen.
14. A compound of claim 5 wherein:
—W— is
Figure US20050124623A1-20050609-C00390
R10, R11, R12, R13, R14, R15, R16 and R17 are each independently H or methyl, with the proviso that one is methyl;
D is selected from the group consisting of hydrogen, cyano, S(O)2R24, halogen, COOR20, C(O)NH2, phenyl and heteroaryl; wherein said phenyl or heteroaryl is independently optionally substituted with one to three same or different halogens or from one to three same or different substituents selected from the group consisting of (C1-6)alkyl, (C1-6)alkenyl, hydroxy, (C1-6)alkoxy, halogen, —NR24R25, —S(O)2NR24R25, COOR26 and —CONR24R25; wherein said (C1-6)alkyl is optionally substituted with one to three same or different halogen or a hydroxy; and
A is selected from the group consisting of phenyl, pyridinyl, furanyl, thienyl, isoxazole and oxazole; wherein said phenyl, pyridinyl, furanyl, thienyl, isoxazole or oxazole is independently optionally substituted with one to three same or different halogens or from one to three same or different substituents selected from the group consisting of (C1-4)alkyl, (C1-4)alkenyl, (C1-3)alkoxy, halogen and —NH2; wherein said (C1-4)alkyl is optionally substituted with one to three same or different halogens.
15. A compound of claim 7 wherein:
Q is selected from Group (A).
16. A compound of claim 1 including pharmaceutically acceptable salts thereof,
Figure US20050124623A1-20050609-C00391
wherein:
Q is selected from the group consisting of
Figure US20050124623A1-20050609-C00392
R1 is hydrogen or methyl;
R3 and R5 are independently selected from the group consisting of hydrogen, halogen, cyano, nitro, COOR8, XR9 and B;
R2 and R4 are independently O or do not exist, with the proviso that only one of R2 and R4 are O;
R6 is (CH2)nH, wherein n is 0-1;
— represents a carbon-carbon bond or does not exist;
—Y— is selected from the group consisting of
Figure US20050124623A1-20050609-C00393
R10, R11, R12, R13, R14,R15, R16 and R17 are each independently H or (C1-6)alkyl; wherein said (C1-6)alkyl may optionally be substituted with one to three same or different halogen, OH or CN;
R18 is a member selected from the group consisting of C(O)-phenyl, C(O)-heteroaryl, pyridinyl, pyrimidinyl, quinolyl, isoquinolyl, quinazolyl, quinoxalinyl, napthyridinyl, pthalazinyl, azabenzofuryl and azaindolyl; wherein said member is optionally substituted with from one to two substituents selected from the group consisting of methyl, -amino, —NHMe, —NMe2, methoxy, hydroxymethyl and halogen;
D is selected from the group consisting of hydrogen, cyano, S(O)2R24, halogen, COOR20, C(O)NR21R22, phenyl and heteroaryl; wherein said phenyl or heteroaryl is independently optionally substituted with one to three same or different halogens or from one to three same or different substituents selected from F;
A is selected from the group consisting of phenyl, pyridinyl, furanyl, thienyl, isoxazole and oxazole; wherein said phenyl, pyridinyl, furanyl, thienyl, isoxazole or oxazole is independently optionally substituted with one to three same or different halogens or from one to three same or different substituents selected from F;
B is selected from the group consisting of (C1-6)alkyl, C(O)NR21R22, phenyl and heteroaryl; wherein said (C1-6)alkyl, phenyl and heteroaryl are independently optionally substituted with one to three same or different halogens or from one to three same or different substituents selected from F;
heteroaryl is selected from the group consisting of pyridinyl, pyrazinyl, pyridazinyl, pyrimidinyl, furanyl, thienyl, thiazolyl, oxazolyl, isoxazolyl, imidazolyl, oxadiazolyl, thiadiazolyl, pyrazolyl, tetrazolyl and triazolyl;
F is selected from the group consisting of (C1-6)alkyl, (C1-6)alkenyl, phenyl, pyridinyl, hydroxy, (C1-6)alkoxy, halogen, benzyl, —NR23C(O)—(C1-6)alkyl, —NR24R25, —S(O)2NR24R25, COOR26, —COR27, and —CONR24R25; wherein said (C1-6)alkyl or phenyl are each optionally substituted with hydroxy, (C1-6)alkoxy, dimethylamino or from one to three same or different halogen;
R8, R9 and R26 are each independently selected from the group consisting of hydrogen and (C1-6)alkyl;
X is selected from the group consisting of NR26, O and S;
R20, R21,R22, R23, R24 and R25 are independently selected from the group consisting of hydrogen, (C1-6)alkyl, phenyl and heteroaryl; wherein said phenyl and heteroaryl are each independently optionally substituted with one to three same or different halogen or methyl; and
R27 is piperazinyl, N-methylpiperazinyl or 3-pyrazolyl.
17. A compound of claim 6 wherein:
Q is
Figure US20050124623A1-20050609-C00394
R5 is selected from the group consisting of hydrogen, halogen, cyano, XR9, heteroaryl, —N(CH2CH2)2NC(O)pyrazolyl, and —C(O)CH3, wherein said heteroaryl is optionally substituted with one substituent selected from F;
heteroaryl is selected from the group consisting of pyridinyl, pyrazinyl, pyridazinyl, pyrimidinyl, isoxazolyl, isoxazolyl, pyrazolyl, and triazolyl;
—Y— is selected from the group consisting of
Figure US20050124623A1-20050609-C00395
R10, R11, R12, R13, R14,R15, R16 and R17 are each hydrogen;
A is phenyl or pyridinyl;
R18 is C(O)-phenyl, isoquinolyl or quinazolyl;
D is cyano or oxadiazolyl;
F is selected from the group consisting of (C1-6)alkyl, phenyl, pyridinyl, (C1-2)alkoxy, —COOR26—COR and —CONR24R25; wherein said phenyl is optionally substituted with one group selected from methyl, methoxy, fluoro, or trifluoromethyl;
X is selected from the group consisting of O;
R9 is (C1-2)alkyl;
R26 is hydrogen, methyl, or ethyl;
R24 and R25 are independently selected from the group consisting of hydrogen and methyl; and
R27 is piperazinyl, N-methyl piperazinyl, or 3-pyrazolyl.
18. A compound of claim 17 wherein:
R5 is heteroaryl optionally substituted with one halogen or one substituent selected from F;
—Y— is
Figure US20050124623A1-20050609-C00396
R18 is C(O)-phenyl, isoquinolyl or quinazolyl; and
F is (C1-6)alkyl.
19. A compound of claim 18 wherein R18 is C(O)-phenyl.
20. A compound of claim 18 wherein R18 is isoquinolyl or quinazolyl.
21. A compound of claim 17 wherein:
R5 is heteroaryl optionally substituted with one substituent selected from F;
heteroaryl is selected from the group consisting of pyridinyl, pyrazinyl, pyridazinyl, pyrimidinyl, isoxazolyl, isoxazolyl, pyrazolyl and triazolyl; and
—Y— is
Figure US20050124623A1-20050609-C00397
22. A compound of claim 17 wherein:
Q is
Figure US20050124623A1-20050609-C00398
R5 is heteroaryl optionally substituted with one substituent selected from F; and
heteroaryl is selected from the group consisting of pyrazolyl and triazolyl.
23. A compound of claim 22 wherein:
F is selected from the group consisting of methyl and —CONR24R25; and
R24 and R25 are independently selected from the group consisting of hydrogen and methyl.
24. A compound of claim 23 wherein:
F is methyl.
25. A pharmaceutical composition which comprises an antiviral effective amount of a compound of Formula I, including pharmaceutically acceptable salts thereof, as claimed in claim 1, and one or more pharmaceutically acceptable carriers, excipients or diluents.
26. The pharmaceutical composition of claim 25 which additionally comprises an antiviral effective amount of an AIDS treatment agent selected from the group consisting of:
(a) an AIDS antiviral agent;
(b) an anti-infective agent;
(c) an immunomodulator; and
(d) HIV entry inhibitors.
27. A method for treating a mammal infected with the HIV virus comprising administering to said mammal an antiviral effective amount of a compound of Formula I, including pharmaceutically accceptable salts thereof, as claimed in claim 1, and one or more pharmaceutically acceptable carriers, excipients or diluents.
28. The method of claim 27 comprising administering to said mammal an antiviral effective amount of a compound of Formula I, including pharmaceutically accceptable salts thereof, as claimed in claim 1, in combination with an antiviral effective amount of an AIDS treatment agent selected from the group consisting of an AIDS antiviral agent; an anti-infective agent; an immunomodulator; and an HIV entry inhibitor.
US10/979,558 2003-11-26 2004-11-02 Diazaindole-dicarbonyl-piperazinyl antiviral agents Abandoned US20050124623A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US10/979,558 US20050124623A1 (en) 2003-11-26 2004-11-02 Diazaindole-dicarbonyl-piperazinyl antiviral agents
US12/026,633 US7902204B2 (en) 2003-11-26 2008-02-06 Diazaindole-dicarbonyl-piperazinyl antiviral agents

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US52562403P 2003-11-26 2003-11-26
US10/979,558 US20050124623A1 (en) 2003-11-26 2004-11-02 Diazaindole-dicarbonyl-piperazinyl antiviral agents

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/026,633 Continuation US7902204B2 (en) 2003-11-26 2008-02-06 Diazaindole-dicarbonyl-piperazinyl antiviral agents

Publications (1)

Publication Number Publication Date
US20050124623A1 true US20050124623A1 (en) 2005-06-09

Family

ID=34652366

Family Applications (2)

Application Number Title Priority Date Filing Date
US10/979,558 Abandoned US20050124623A1 (en) 2003-11-26 2004-11-02 Diazaindole-dicarbonyl-piperazinyl antiviral agents
US12/026,633 Expired - Fee Related US7902204B2 (en) 2003-11-26 2008-02-06 Diazaindole-dicarbonyl-piperazinyl antiviral agents

Family Applications After (1)

Application Number Title Priority Date Filing Date
US12/026,633 Expired - Fee Related US7902204B2 (en) 2003-11-26 2008-02-06 Diazaindole-dicarbonyl-piperazinyl antiviral agents

Country Status (29)

Country Link
US (2) US20050124623A1 (en)
EP (1) EP1751161B1 (en)
JP (1) JP2007512332A (en)
KR (1) KR20070008526A (en)
CN (1) CN1906199A (en)
AR (1) AR046720A1 (en)
AT (1) ATE473981T1 (en)
AU (1) AU2004295309A1 (en)
BR (1) BRPI0417007A (en)
CA (1) CA2547347C (en)
CY (1) CY1110831T1 (en)
DE (1) DE602004028173D1 (en)
DK (1) DK1751161T3 (en)
ES (1) ES2347253T3 (en)
HR (1) HRP20100433T1 (en)
IL (1) IL175809A (en)
IS (1) IS2890B (en)
MX (1) MXPA06005730A (en)
NO (1) NO336261B1 (en)
NZ (1) NZ546904A (en)
PE (1) PE20050587A1 (en)
PL (1) PL1751161T3 (en)
PT (1) PT1751161E (en)
RU (1) RU2362777C2 (en)
SI (1) SI1751161T1 (en)
TW (1) TWI336253B (en)
UA (1) UA85502C2 (en)
WO (1) WO2005054247A1 (en)
ZA (1) ZA200604331B (en)

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060100209A1 (en) * 2004-11-09 2006-05-11 Chong-Hui Gu Formulations of 1-(4-benzoyl-piperazin-1-yl)-2-[4-methoxy-7-(3-methyl-[1,2,4]triazol-1-yl)-1H-pyrrolo[2,3-c]pyridin-3-yl]-ethane-1,2-dione
US20060100432A1 (en) * 2004-11-09 2006-05-11 Matiskella John D Crystalline materials of 1-(4-benzoyl-piperazin-1-yl)-2-[4-methoxy-7-(3-methyl-[1,2,4]triazol-1-yl)-1H-pyrrolo[2,3-c]pyridin-3-yl]-ethane-1,2-dione
US20070155702A1 (en) * 2005-12-14 2007-07-05 Bristol-Myers Squibb Company Crystalline forms of 1-benzoyl-4-[2-[4-methoxy-7-(3-methyl-1H-1,2,4-triazol-1-yl)-1-[(phosphonooxy)methyl]-1H-pyrrolo[2,3-c]pyridin-3-yl]-1,2-dioxoethyl]-piperazine
US20080139572A1 (en) * 2006-04-25 2008-06-12 Bristol-Myers Squibb Company Diketo-piperazine and piperidine derivatives as antiviral agents
US7449476B2 (en) 2004-05-26 2008-11-11 Bristol-Myers Squibb Company Tetrahydrocarboline antiviral agents
US20110021479A1 (en) * 2008-02-27 2011-01-27 Nycomed Gmbh Pyrrolopyrimidinecarboxamides
WO2011023693A1 (en) 2009-08-26 2011-03-03 Nycomed Gmbh Methylpyrrolopyrimidinecarboxamides
WO2012106522A2 (en) 2011-02-04 2012-08-09 Duquesne University Of The Holy Spirit Bicyclic and tricyclic pyrimidine tyrosine kinase inhibitors with antitubulin activity and methods of treating a patient
WO2016183093A1 (en) * 2015-05-11 2016-11-17 University Of Kansas Inhibitors of viruses
US10683293B2 (en) 2014-08-04 2020-06-16 Nuevolution A/S Optionally fused heterocyclyl-substituted derivatives of pyrimidine useful for the treatment of inflammatory, metabolic, oncologic and autoimmune diseases
US11447479B2 (en) 2019-12-20 2022-09-20 Nuevolution A/S Compounds active towards nuclear receptors
US11613532B2 (en) 2020-03-31 2023-03-28 Nuevolution A/S Compounds active towards nuclear receptors
US11780843B2 (en) 2020-03-31 2023-10-10 Nuevolution A/S Compounds active towards nuclear receptors

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011041713A2 (en) * 2009-10-02 2011-04-07 Glaxosmithkline Llc Piperazinyl antiviral agents
ES2667822T3 (en) * 2013-03-27 2018-05-14 VIIV Healthcare UK (No.5) Limited 2-keto amide derivatives as HIV binding inhibitors
EP2978762A1 (en) * 2013-03-27 2016-02-03 Bristol-Myers Squibb Company Piperazine and homopiperazine derivatives as hiv attachment inhibitors

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5023265A (en) * 1990-06-01 1991-06-11 Schering Corporation Substituted 1-H-pyrrolopyridine-3-carboxamides
US6469006B1 (en) * 1999-06-15 2002-10-22 Bristol-Myers Squibb Company Antiviral indoleoxoacetyl piperazine derivatives
US6476034B2 (en) * 2000-02-22 2002-11-05 Bristol-Myers Squibb Company Antiviral azaindole derivatives
US6573262B2 (en) * 2000-07-10 2003-06-03 Bristol-Myers Sqibb Company Composition and antiviral activity of substituted indoleoxoacetic piperazine derivatives
US20030207910A1 (en) * 2001-02-02 2003-11-06 Tao Wang Composition and antiviral activity of substituted azaindoleoxoacetic piperazine derivatives
US20040110785A1 (en) * 2001-02-02 2004-06-10 Tao Wang Composition and antiviral activity of substituted azaindoleoxoacetic piperazine derivatives

Family Cites Families (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IT1265057B1 (en) 1993-08-05 1996-10-28 Dompe Spa TROPIL 7-AZAINDOLIL-3-CARBOXYAMIDE
US5424329A (en) * 1993-08-18 1995-06-13 Warner-Lambert Company Indole-2-carboxamides as inhibitors of cell adhesion
GB9420521D0 (en) 1994-10-12 1994-11-30 Smithkline Beecham Plc Novel compounds
DE19814838C2 (en) 1998-04-02 2001-01-18 Asta Medica Ag Indolyl-3-glyoxylic acid derivatives with anti-tumor effects
AU4710699A (en) 1998-06-30 2000-01-17 Eli Lilly And Company Bicyclic sPLA2 inhibitors
TWI269654B (en) 1999-09-28 2007-01-01 Baxter Healthcare Sa N-substituted indole-3-glyoxylamide compounds having anti-tumor action
DE10037310A1 (en) 2000-07-28 2002-02-07 Asta Medica Ag New indole derivatives and their use as medicines
US20030069266A1 (en) * 2001-02-02 2003-04-10 Tao Wang Composition and antiviral activity of substituted azaindoleoxoacetic piperazine derivatives
US6825201B2 (en) 2001-04-25 2004-11-30 Bristol-Myers Squibb Company Indole, azaindole and related heterocyclic amidopiperazine derivatives
US20030236277A1 (en) 2002-02-14 2003-12-25 Kadow John F. Indole, azaindole and related heterocyclic pyrrolidine derivatives
US7037913B2 (en) 2002-05-01 2006-05-02 Bristol-Myers Squibb Company Bicyclo 4.4.0 antiviral derivatives
US20040063744A1 (en) 2002-05-28 2004-04-01 Tao Wang Indole, azaindole and related heterocyclic 4-alkenyl piperidine amides
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
US20050075364A1 (en) * 2003-07-01 2005-04-07 Kap-Sun Yeung Indole, azaindole and related heterocyclic N-substituted piperazine derivatives

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5023265A (en) * 1990-06-01 1991-06-11 Schering Corporation Substituted 1-H-pyrrolopyridine-3-carboxamides
US6469006B1 (en) * 1999-06-15 2002-10-22 Bristol-Myers Squibb Company Antiviral indoleoxoacetyl piperazine derivatives
US6476034B2 (en) * 2000-02-22 2002-11-05 Bristol-Myers Squibb Company Antiviral azaindole derivatives
US6573262B2 (en) * 2000-07-10 2003-06-03 Bristol-Myers Sqibb Company Composition and antiviral activity of substituted indoleoxoacetic piperazine derivatives
US20030207910A1 (en) * 2001-02-02 2003-11-06 Tao Wang Composition and antiviral activity of substituted azaindoleoxoacetic piperazine derivatives
US20040110785A1 (en) * 2001-02-02 2004-06-10 Tao Wang Composition and antiviral activity of substituted azaindoleoxoacetic piperazine derivatives

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7449476B2 (en) 2004-05-26 2008-11-11 Bristol-Myers Squibb Company Tetrahydrocarboline antiviral agents
US7829711B2 (en) 2004-11-09 2010-11-09 Bristol-Myers Squibb Company Crystalline materials of 1-(4-benzoyl-piperazin-1-yl)-2-[4-methoxy-7-(3-methyl-[1,2,4]triazol-1-yl)-1H-pyrrolo[2,3-C]pyridine-3-yl]-ethane-1,2-dione
US20060100432A1 (en) * 2004-11-09 2006-05-11 Matiskella John D Crystalline materials of 1-(4-benzoyl-piperazin-1-yl)-2-[4-methoxy-7-(3-methyl-[1,2,4]triazol-1-yl)-1H-pyrrolo[2,3-c]pyridin-3-yl]-ethane-1,2-dione
US20060100209A1 (en) * 2004-11-09 2006-05-11 Chong-Hui Gu Formulations of 1-(4-benzoyl-piperazin-1-yl)-2-[4-methoxy-7-(3-methyl-[1,2,4]triazol-1-yl)-1H-pyrrolo[2,3-c]pyridin-3-yl]-ethane-1,2-dione
US20090227794A1 (en) * 2004-11-09 2009-09-10 Bristol-Myers Squibb Company Crystalline materials of 1-(4-benzoyl-piperazin-1-yl)-2-[4-methoxy-7-(3-methyl-[1,2,4]triazol-1-yl)-1h-pyrrolo[2,3-c]pyridin-3-yl]-ethane-1,2-dione
US20100292246A1 (en) * 2004-11-09 2010-11-18 Bristol-Myers Squibb Company Formulations of 1-(4-benzoyl-piperazin-1-yl)-2-[4-methoxy-7-(3-methyl-[1,2,4]triazol-1-yl)-1h-pyrrolo[2,3-c]pyridin-3-yl]-ethane-1,2-dione
US20070155702A1 (en) * 2005-12-14 2007-07-05 Bristol-Myers Squibb Company Crystalline forms of 1-benzoyl-4-[2-[4-methoxy-7-(3-methyl-1H-1,2,4-triazol-1-yl)-1-[(phosphonooxy)methyl]-1H-pyrrolo[2,3-c]pyridin-3-yl]-1,2-dioxoethyl]-piperazine
US7851476B2 (en) 2005-12-14 2010-12-14 Bristol-Myers Squibb Company Crystalline forms of 1-benzoyl-4-[2-[4-methoxy-7-(3-methyl-1H-1,2,4-triazol-1-YL)-1-[(phosphonooxy)methyl]-1H-pyrrolo[2,3-C]pyridin-3-YL]-1,2-dioxoethyl]-piperazine
US20080139572A1 (en) * 2006-04-25 2008-06-12 Bristol-Myers Squibb Company Diketo-piperazine and piperidine derivatives as antiviral agents
US7807676B2 (en) 2006-04-25 2010-10-05 Bristol-Myers Squibb Company Diketo-Piperazine and Piperidine derivatives as antiviral agents
US7807671B2 (en) 2006-04-25 2010-10-05 Bristol-Myers Squibb Company Diketo-piperazine and piperidine derivatives as antiviral agents
US20110021479A1 (en) * 2008-02-27 2011-01-27 Nycomed Gmbh Pyrrolopyrimidinecarboxamides
US8445501B2 (en) 2008-02-27 2013-05-21 Takeda Gmbh Substituted 7-carboxamido-pyrrolo[3,2-d]pyrimidines
WO2011023693A1 (en) 2009-08-26 2011-03-03 Nycomed Gmbh Methylpyrrolopyrimidinecarboxamides
US8927557B2 (en) 2009-08-26 2015-01-06 Takeda Gmbh Methylpyrrolopyrimidinecarboxamides
US9376442B2 (en) 2009-08-26 2016-06-28 Takeda Gmbh Methylpyrrolopyrimidinecarboxamides
WO2012106522A2 (en) 2011-02-04 2012-08-09 Duquesne University Of The Holy Spirit Bicyclic and tricyclic pyrimidine tyrosine kinase inhibitors with antitubulin activity and methods of treating a patient
US10683293B2 (en) 2014-08-04 2020-06-16 Nuevolution A/S Optionally fused heterocyclyl-substituted derivatives of pyrimidine useful for the treatment of inflammatory, metabolic, oncologic and autoimmune diseases
US10689383B2 (en) 2014-08-04 2020-06-23 Nuevolution A/S Optionally fused heterocyclyl-substituted derivatives of pyrimidine useful for the treatment of inflammatory, metabolic, oncologic and autoimmune diseases
US11254681B2 (en) 2014-08-04 2022-02-22 Nuevolution A/S Optionally fused heterocyclyl-substituted derivatives of pyrimidine useful for the treatment of inflammatory, metabolic, oncologic and autoimmune diseases
WO2016183093A1 (en) * 2015-05-11 2016-11-17 University Of Kansas Inhibitors of viruses
US11447479B2 (en) 2019-12-20 2022-09-20 Nuevolution A/S Compounds active towards nuclear receptors
US11613532B2 (en) 2020-03-31 2023-03-28 Nuevolution A/S Compounds active towards nuclear receptors
US11780843B2 (en) 2020-03-31 2023-10-10 Nuevolution A/S Compounds active towards nuclear receptors

Also Published As

Publication number Publication date
IL175809A (en) 2012-06-28
ZA200604331B (en) 2007-10-31
NO20062474L (en) 2006-08-21
NZ546904A (en) 2010-01-29
ATE473981T1 (en) 2010-07-15
WO2005054247A1 (en) 2005-06-16
PL1751161T3 (en) 2010-12-31
SI1751161T1 (en) 2010-11-30
CN1906199A (en) 2007-01-31
TW200529825A (en) 2005-09-16
RU2006122661A (en) 2008-01-10
ES2347253T3 (en) 2010-10-27
IS8479A (en) 2006-05-23
EP1751161A1 (en) 2007-02-14
JP2007512332A (en) 2007-05-17
US20080125439A1 (en) 2008-05-29
CA2547347A1 (en) 2005-06-16
US7902204B2 (en) 2011-03-08
TWI336253B (en) 2011-01-21
DE602004028173D1 (en) 2010-08-26
MXPA06005730A (en) 2006-08-17
AR046720A1 (en) 2005-12-21
NO336261B1 (en) 2015-07-06
DK1751161T3 (en) 2010-11-08
UA85502C2 (en) 2009-01-26
HRP20100433T1 (en) 2010-09-30
IL175809A0 (en) 2006-10-05
PE20050587A1 (en) 2005-08-11
PT1751161E (en) 2010-09-08
IS2890B (en) 2014-09-15
AU2004295309A1 (en) 2005-06-16
RU2362777C2 (en) 2009-07-27
CA2547347C (en) 2012-05-15
EP1751161B1 (en) 2010-07-14
BRPI0417007A (en) 2007-01-16
CY1110831T1 (en) 2015-06-10
KR20070008526A (en) 2007-01-17

Similar Documents

Publication Publication Date Title
US7902204B2 (en) Diazaindole-dicarbonyl-piperazinyl antiviral agents
US7714019B2 (en) Indole, azaindole and related heterocyclic pyrrolidine derivatives
US8039486B2 (en) Indole, azaindole and related heterocyclic N-substituted piperazine derivatives
US7915283B2 (en) Indole, azaindole and related heterocyclic 4-alkenyl piperidine amides
EP1513832B1 (en) Indole, azaindole and related heterocyclic 4-alkenyl piperidine amides
EP1549313B1 (en) Composition and antiviral activity of substituted azaindoleoxoacetic piperazine derivatives
US20060094717A1 (en) Indole, azaindole and related heterocyclic ureido and thioureido piperazine derivatives
US6900206B2 (en) Indole, azaindole and related heterocyclic sulfonylureido piperazine derivatives
ES2368430T3 (en) INDOL, AZAINDOL AND RELATED HETEROCYCLIC N-SUBSTITUTED PIPERAZINE DERIVATIVES.

Legal Events

Date Code Title Description
AS Assignment

Owner name: BRISTOL-MYERS SQUIBB COMPANY, NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BENDER, JOHN A.;YANG, ZHONG;KADOW, JOHN F.;AND OTHERS;REEL/FRAME:015445/0208;SIGNING DATES FROM 20041130 TO 20041207

STCB Information on status: application discontinuation

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

AS Assignment

Owner name: VIIV HEALTHCARE (NO. 5) LIMITED, UNITED KINGDOM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:BRISTOL-MYERS SQUIBB COMPANY;REEL/FRAME:039374/0388

Effective date: 20160222

AS Assignment

Owner name: VIIV HEALTHCARE UK (NO. 5) LIMITED, UNITED KINGDOM

Free format text: CORRECTIVE ASSIGNMENT TO CORRECT THE RECEIVING PARTY DATA PREVIOUSLY RECORDED AT REEL: 039374 FRAME: 0388. ASSIGNOR(S) HEREBY CONFIRMS THE ASSIGNMENT;ASSIGNOR:BRISTOL-MYERS SQUIBB COMPANY;REEL/FRAME:039985/0093

Effective date: 20160222