US20070225317A1 - Rsv polymerase inhibitors - Google Patents

Rsv polymerase inhibitors Download PDF

Info

Publication number
US20070225317A1
US20070225317A1 US11/750,157 US75015707A US2007225317A1 US 20070225317 A1 US20070225317 A1 US 20070225317A1 US 75015707 A US75015707 A US 75015707A US 2007225317 A1 US2007225317 A1 US 2007225317A1
Authority
US
United States
Prior art keywords
alkyl
amino
optionally substituted
hydroxy
compound
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
US11/750,157
Inventor
Bruno Simoneau
Josee Bordeleau
Gulrez Fazal
Serge Landry
Steve Mason
Jean Rancourt
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.)
Individual
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 US11/750,157 priority Critical patent/US20070225317A1/en
Publication of US20070225317A1 publication Critical patent/US20070225317A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • 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
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the present invention relates to compounds, process for their synthesis, compositions and methods for the treatment or prevention of Respiratory Syncytial Virus (RSV) infection.
  • RSV Respiratory Syncytial Virus
  • the present invention provides novel compounds, pharmaceutical compositions containing such compounds and methods for using these compounds in the treatment or prevention of Respiratory Syncytial Virus infection.
  • Respiratory Syncytial Virus is the most common cause of severe lower respiratory tract infections (LRTI) in young infants, virtually all of whom suffer at least one RSV infection in the upper respiratory tract by the age of two. In very young infants, infection can progress to the lower respiratory tract causing bronchiolitis and pneumonia that may necessitate hospitalization.
  • RSV is an enveloped virus with a negative strand non-segmented RNA genome. It belongs to the Mononegavirales order, Paramyxoviridae family and Pneumovirus genus. RSV exhibits nucleocapsid-associated RNA dependent RNA polymerase (RdRp) activity for viral transcription and replication, which occurs in the cytoplasm of an infected cell.
  • RdRp nucleocapsid-associated RNA dependent RNA polymerase
  • the RSV polymerase is comprised of at least five viral components: the genomic RNA, and the L, N, P and M2-1 proteins. Together these components form a Ribonucleoprotein (RNP) complex with RdRp activity required for the synthesis of both viral genomic RNA (“replicase” activity) and sub-genomic mRNAs (“transcriptase” activity). As opposed to the replication of viral products, RSV mRNAs are co-transcriptionally capped (i.e. guanylated and methylated) at their 5′ ends and polyadenylated at their 3′ ends by the RNP complex. These modifications are necessary for translation of the viral mRNAs by the host protein synthesis machinery.
  • RNP Ribonucleoprotein
  • the RNP complex functions exclusively in the cytoplasm of the RSV-infected cells. Since the host proteins responsible for capping of mRNAs are located in the nucleus of the cell, capping of its mRNAs by the viral RNP complex is essential for the synthesis of RSV proteins. Thus, the RSV RNP complex is an attractive target in screening for potential antiviral therapeutics since it has multiple activities essential for viral replication.
  • U.S. Pat. No. 4,176,184 discloses 7,7-dimethyl-2-(2-methoxyphenyl)-5-[3-(2-(3,4-dimethoxyphenyl)ethylamino)propyl]-5H,7H-imidazo[4,5-h]isoquinoline-4,6-dione dihydrochloride (example 28) having anti-arrhythmic activity.
  • the present invention therefore provides novel compounds, compositions, uses and methods that inhibit Respiratory Syncytial virus polymerase essential for viral replication.
  • the invention provides a compound of Formula (Ia), or an enantiomer or a salt thereof: wherein R 1 is —(CH ⁇ CH) 0-1 —(C 6 or C 10 )aryl or —(CH ⁇ CH) 0-1 -5-, 6-, 9- or 10-membered heteroaryl, said aryl or heteroaryl being optionally substituted with one, two or three substituents, each independently selected from:
  • the invention provides the use of a compound of formula (I), or an enantiomer or a salt thereof: wherein R 1 is —(CH ⁇ CH) 0-1 —, —(C 6 or C 10 )aryl or —(CH ⁇ CH) 0-1 -5-, 6-, 9- or 10-membered heteroaryl, said aryl or heteroaryl being optionally substituted with one, two or three substituents, each independently selected from:
  • the invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective and acceptable amount of a compound of formula (I) or (Ia) in association with a pharmaceutically-acceptable carrier.
  • the invention provides a pharmaceutical composition for use in the treatment or prevention of respiratory syncytial virus infection, wherein the composition comprises a therapeutically effective and acceptable amount of a compound of formula (I) or (Ia) in association with a pharmaceutically-acceptable carrier.
  • the pharmaceutical composition according to this invention further comprises a therapeutically effective amount of at least one other antiviral agent.
  • the invention provides an anti-respiratory syncytial virus pharmaceutical composition
  • the invention provides a use of a compound of formula (I) or (Ia) to inhibit the replication of a respiratory syncytial virus.
  • the invention provides a use of a compound of formula (I) or (Ia), in the treatment or prevention of a respiratory syncytial virus infection in a mammal.
  • the invention provides a method of treating or preventing a respiratory syncytial virus infection in a mammal comprising administering to the mammal an anti-respiratory syncytial virally-effective and acceptable amount of a compound of formula (I) or (Ia) or a composition containing such a compound, alone or in combination with at least one other antiviral agent, administered together or separately.
  • the invention provides a method of inhibiting replication of a respiratory syncytial virus comprising exposing virus-infected cells to a anti-respiratory syncytial virally-effective and acceptable amount of at least one of a compound of formula (I) or (Ia).
  • the invention provides a packaged pharmaceutical comprising a pharmaceutical composition containing a compound of formula (I) or (Ia) and directions identifying an administration regimen.
  • the invention provides a packaged pharmaceutical for use for the treatment or prevention of respiratory syncytial virus infection in a mammal, wherein the packaged pharmaceutical comprises a pharmaceutical composition containing a compound of formula (I) or (Ia) and directions identifying an administration regimen.
  • the invention provides an article of manufacture comprising packaging material contained within which is a composition effective to inhibit a respiratory syncytial virus, the packaging material comprising a label which indicates that the composition can be used to treat or prevent infection by a respiratory syncytial virus, wherein said composition includes a compound of formula (I) or (Ia).
  • halo as used herein means a halogen radical selected from bromo, chloro, fluoro or iodo.
  • (C 1-6 )alkyl as used herein, either alone or in combination with another radical, means straight or branched-chain alkyl radicals containing up to six carbon atoms and includes, but is not limited to, methyl (Me), ethyl (Et), propyl (Pr), butyl (Bu), hexyl, 1-methylethyl (iPr), 1-methylpropyl (sec-Bu), 2-methylpropyl (iBu) and 1,1-dimethylethyl (tBu); wherein the abbreviations commonly used herein are given in parentheses.
  • (C 2-6 )alkynyl as used herein, either alone or in combination with another radical, means straight or branched-chain alkyne radicals containing from two to six carbon atoms, at least two of which are linked by a triple bond and includes, but is not limited to, —C ⁇ CH.
  • (C 3-7 )cycloalkyl as used herein, either alone or in combination with another radical, means saturated cyclic hydrocarbon radicals containing from three to seven carbon atoms and includes cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl.
  • (C 1-6 )alkoxy or “O—(C 1-6 )alkyl” as used herein interchangeably, mean a straight chain alkyl containing one to six carbon atoms linked through an oxygen atom or a branched chain alkyl radical containing three to six carbon atoms linked through an oxygen atom and includes but is not limited to methoxy, ethoxy, propoxy, 1-methylethoxy, butoxy and 1,1-dimethylethoxy. The latter radical is known commonly as tert-butoxy.
  • (C 1-6 )haloalkyl as used herein means an alkyl radical containing one to six carbon atoms wherein one or more hydrogen atoms are replaced by a halogen atom (including but not limited to trifluoromethyl).
  • (C 1-6 )alkylthio or “S—(C 1-6 )alkyl” as used herein interchangeably, mean a straight chain alkyl containing one to six carbon atoms linked through a sulfur atom, or a branched chain alkyl radical containing three to six carbon atoms linked through a sulfur atom.
  • amino as used herein means an amino radical of formula —NH 2 .
  • (C 1-4 )alkylamino” or “NH—(C 1-6 )alkyl” as used herein means an alkyl radical containing one to six carbon atoms linked through a nitrogen atom, and includes but is not limited to methylamino, propylamino, (1-methylethyl)amino and (2-methylbutyl)amino.
  • di((C 1-6 )alkyl)amino or “N((C 1-6 )alkyl) 2 ” means an amino radical having two (C 1-6 )alkyl substituents, each of which contains one to six carbon atoms and includes but is not limited to dimethylamino, diethylamino, ethylmethylamino and the like.
  • aryl or “(C 6 or C 10 or C 14 )aryl” used interchangeably, either alone or in combination with another radical, mean either an aromatic monocyclic system containing 6 carbon atoms or an aromatic bicyclic system containing 10 carbon atoms, or an aromatic tricyclic system containing 14 carbon atoms.
  • aryl includes but is not limited to phenyl, naphthyl or anthryl.
  • (C 7-16 )aralkyl or “(C 1-6 )alkyl-aryl” used interchangeably, either alone or in combination with another radical, means an aryl as defined above linked through an alkyl group, wherein alkyl is as defined above containing from 1 to 6 carbon atoms.
  • Aralkyl includes, but is not limited to, for example, benzyl, and phenylbutyl.
  • Het or “heterocycle” as used herein means a monovalent radical derived by removal of a hydrogen from a five- or six-membered, saturated or unsaturated (including aromatic) ring system containing from one to three heteroatoms each independently selected from nitrogen, oxygen and sulfur.
  • the heterocycle may bear one or two substituents; for example, N-oxido, (C 1-4 )alkyl, (C 1-3 )alkyl-phenyl, (C 1-6 )alkoxy, halo, amino or (C 1-6 )alkylamino.
  • the five- or six-membered heterocycle can be fused to a second cycloalkyl, an aryl (e.g. phenyl) or another heterocycle.
  • heterocycles and optionally substituted heterocycles include, but are not limited to, morpholine, thiadiazole, quinoline, isoquinoline, 3,4-methylene-dioxyphenyl, benzothiazole, benzofuran, benzothiophene, pyrrolidine, tetrahydrofuran, thiazolidine, pyrrole, indole, benzimidazole, 1H-imidazole, 1-methyl-1H-imidazole, pyrazole, furan, thiophene, oxazole, isoxazole, thiazole, 2-methylthiazole, 2-aminothiazole, 2-(methylamino)thiazole, piperidine, 1-methylpiperidine, 1-methylpiperazine, 1,4-dioxane, pyridine, pyridine N-oxide, pyrimidine, 2,4-dihydroxypyrimidine.
  • 2,4-dimethylpyrimidine 2,6-dimethylpyridine, 1-methyl-1H-tetrazole, 2-methyl-2H-tetrazole, benzoxazole, thiazolo[4,5-b]-pyridine, heterocycles selected from the following group: or each heterocycle exemplified in Tables 1 and 2 below.
  • heteroaryl as used herein means a 5- or 6-membered monocylic, or 9 or 10-membered bicyclic aromatic heterocycle as defined above.
  • heteroaryl includes but is not limited to: indole, benzimidazole, imidazole, furan, thiophene, oxazole, thiazole, pyrazole, pyrrole, pyridine, pyrimidine, benzofuran, benzothiophene, isoquinoline, quinoline, heteroaryl selected from the following group: or each heteroaryl exemplified in Tables 1 and 2 below.
  • pharmaceutically acceptable carrier means a non-toxic, generally inert vehicle for the active ingredient which does not adversely affect the ingredient.
  • mammal as it is used herein is meant to encompass humans, as well as non-human mammals which are susceptible to infection by respiratory syncytial virus including domestic animals, including but not limited to cattle, pigs, horses, sheep, dogs and cats, and non-domestic animals.
  • the term “effective amount” means a predetermined antiviral amount of the antiviral agent, i.e. an amount of the agent sufficient to be effective against the virus in vivo. It is understood that when an antiviral agent is administered in combination with another antiviral agent, the effective amount of each antiviral agent administered in combination may be lower or higher than the effective amount of each antiviral agent administered alone. Such a situation may arise, for example, when the antiviral agents administered in combination act synergistically with each other, so that the effective amount of either or both of the antiviral agents administered in combination is lower than the effective amount of either or both of the antiviral agents administered alone. In an alternative example, the antiviral agents may act antagonistically with each other, so that the effective amount of either or both of the antiviral agents administered in combination is higher than the effective amount of either or both of the antiviral agents administered alone.
  • salt thereof means any acid addition salt of a compound according to the invention; preferably a pharmaceutically acceptable salt thereof.
  • pharmaceutically acceptable salt means a salt of a compound of formula (I) or (Ia) which is, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, generally water or oil-soluble or dispersible, and effective for their intended use.
  • pharmaceutically-acceptable acid addition salts examples of suitable salts are found in, e.g., S. M. Birge et al., J. Pharm. Sci., 1977, 6, pp. 1-19.
  • pharmaceutically-acceptable acid addition salt means those salts which retain the biological effectiveness and properties of the free bases and which are not biologically or otherwise undesirable, formed with inorganic acids including but not limited to hydrochloric acid, hydrobromic acid, sulfuric acid, sulfamic acid, nitric acid, phosphoric acid, and the like, and organic acids including but not limited to acetic acid, trifluoroacetic acid, adipic acid, ascorbic acid, aspartic acid, benzenesulfonic acid, benzoic acid, butyric acid, camphoric acid, camphorsulfonic acid, cinnamic acid, citric acid, digluconic acid, ethanesulfonic acid, glutamic acid, glycolic acid, glycerophosphoric acid, hemisulfic acid, hexanoic acid, formic acid, fumaric acid, 2-hydroxyethanesulfonic acid (isethionic acid), lactic acid, hydroxymaleic acid, mal
  • antiviral agent means an agent (compound or biological) that is effective to inhibit the formation and/or replication of a virus in a mammal. This includes agents that interfere with either host or viral mechanisms necessary for the formation and/or replication of a virus in a mammal. Such agents can be selected from immunomodulatory agents, inhibitors of RSV polymerase or inhibitors of another target in the RSV life cycle.
  • Immunomodulatory agent means those agents (compounds or biologicals) that are effective to enhance or potentiate the immune system response in a mammal
  • Immunomodulatory agents include, for example, class I interferons (such as ⁇ -(alpha), ⁇ -(beta), ⁇ -(delta), ⁇ -(omega) and ⁇ -(tau) interferons, consensus interferons and asialo-interferons), class II interferons (such as ⁇ -(gamma) interferons) and pegylated interferons.
  • inhibitor of RSV polymerase means an agent (compound or biological) that is effective to inhibit the function of an RSV polymerase in a mammal.
  • inhibitor of another target in the RSV life cycle means an agent (compound or biological) that is effective to inhibit the formation and/or replication of RSV in a mammal other than by inhibiting the function of the RSV polymerase. This includes agents that interfere with either-host or RSV viral mechanisms necessary for the formation and/or replication of RSV in a mammal.
  • Inhibitors of another target in the RSV life cycle include but are not limited to fusion inhibitors, including but not limited to Synagis® (palivizumab), VP14637 (ViroPharma), R170591 (Janssen), RFI-641 (BMS) or BMS-43377 (BMS); and other inhibitors including but not limited to ribavirin.
  • treatment means the administration of a compound or composition according to the present invention to alleviate or eliminate symptoms of respiratory syncytial virus infection and/or to reduce or eliminate viral load in a patient and/or to reduce the incidence and/or length of time of hospitalization.
  • hospitalization means medical care in a hospital, clinic or other medical facility to treat respiratory syncytial virus infection.
  • prevention means the administration of a compound or composition according to the present invention pre-exposure of the individual to the virus or post-exposure of the individual to the virus but before the appearance of symptoms of the disease, and/or prior to the detection of viral infection, to prevent the appearance of symptoms of the disease.
  • the invention provides a compound of Formula (Ia) or an enantiomer thereof or a salt thereof, wherein R 1 is —(CH ⁇ CH) 0-1 —(C 6 or G 10 )aryl or 5-, 6-, 9- or 10-membered heteroaryl, said aryl or heteroaryl being optionally substituted with one, two or three substituents, each independently selected from:
  • compounds of formula (Ia) are those wherein:
  • R 1 is (CH ⁇ CH) 0-1 -phenyl, the phenyl being optionally substituted with one, two or three substituents, each independently selected from:
  • R 3 is a 5-, 6-, 9- or 10-membered heteroaryl optionally substituted with (C 1-6 )alkyl;
  • R 4 and R 5 are each independently Me or Et; or R 4 and R 5 are linked, together with the carbon atom to which they are attached, to form a (C 3-7 )cycloalkyl group; with the proviso that R 1 is not 2-methoxyphenyl, when R 2 is H, R 3 is 3,4-dimethoxyphenyl, R 4 is CH 3 and R 5 is CH 3 .
  • R 1 is phenyl, 2-pyridyl, quinolinyl or benzofuranyl, the phenyl and 2-pyridyl each being optionally substituted with one or two substituents, each independently selected from:
  • R 1 is phenyl, 2-pyridyl, quinolinyl or benzofuranyl, the phenyl and 2-pyridyl each being optionally substituted with one or two substituents, each independently selected from:
  • R 2 is H, NH 2 , bromo, chloro, or OH;
  • R 3 is phenyl optionally substituted with one, two or three substituents, each independently selected from:
  • R 4 and R 5 are each independently Me; or R 4 and R 5 are linked, together with the carbon atom to which they are attached, to form a cyclopentyl or cyclohexyl group; with the proviso that R 1 is not 2-nethoxyphenyl, when R 2 is H, R 3 is 3,4-dimethoxyphenyl, R 4 is CH 3 and R 5 is CH 3 .
  • R 1 is R 2 is NH 2 or bromo
  • R 3 is phenyl substituted with one or two substituents each independently selected from hydroxy, methoxy, and iodo
  • R 4 and R 5 are each independently Me; or R 4 and R 5 are linked, together with the carbon atom to which they are attached, to form a cyclohexyl group.
  • compounds of the present invention are useful in the treatment or prevention of respiratory syncytial virus infections.
  • the use of compounds of the formula (I) or enantiomers thereof or salts thereof are provided in the manufacture of a medicament for the treatment or prevention of respiratory syncytial virus infection in a mammal: wherein R 1 is —(CH ⁇ CH) 0-1 —(C 6 or C 10 )aryl or 5-, 6-, 9- or 10-membered heteroaryl, said aryl or heteroaryl being optionally substituted with one, two or three substituents, each independently selected from:
  • R 1 is (CH ⁇ CH) 0-1 —-phenyl, the phenyl being optionally substituted with one, two or three substituents, each independently selected from:
  • R 1 is a 5-, 6-, 9- or 10-membered heteroaryl optionally substituted with one, two or three substituents, each independently selected from nitro, (C 1-6 )alkyl optionally substituted with amino, (C 1-6 )haloalkyl, hydroxy, (C 1-6 )alkoxy, halo, (C 1-6 )alkylthio, cyano and heteroaryl;
  • R 2 is H, (C 1-6 )alkyl, hydroxy, halo, amino, (C 1-6 )alkylamino, or (C 2-4 )alkynyl;
  • R 3 is naphthyl, anthryl, or phenyl optionally substituted with one, two or three substituents, each independently selected from:
  • R 1 is phenyl, 2-pyridyl, quinolinyl or benzofuranyl, the phenyl and 2-pyridyl each being optionally substituted with one or two substituents, each independently selected from:
  • R 1 is phenyl, 2-pyridyl, quinolinyl or benzofuranyl, the phenyl and 2-pyridyl each being optionally substituted with one or two substituents, each independently selected from:
  • R 1 is R 2 is NH 2 or bromo
  • R 3 is phenyl substituted with one or two substituents each independently selected from hydroxy, methoxy, and iodo
  • R 4 and R 5 are each independently Me; or R 4 and R 5 are linked, together with the carbon atom to which they are attached, to form a cyclohexyl group.
  • the invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective and acceptable amount of a compound of formula (I) or (Ia) in association with a pharmaceutically-acceptable carrier.
  • the invention provides a pharmaceutical composition for use in the treatment or prevention of respiratory syncytial virus infection, wherein the composition comprises a therapeutically effective and acceptable amount of a compound of formula (I) or (Ia), alone or in combination with at least one other antiviral agent, in association with a pharmaceutically-acceptable carrier.
  • compositions according to this invention may additionally comprise one or more immunomodulatory agents.
  • compositions of this invention may additionally comprise one or more other inhibitors of RSV polymerase.
  • compositions of this invention may additionally comprise one or more inhibitors of other targets in the RSV life cycle, including but not limited to, fusion inhibitors, including but not limited to Synagis® (palivizumab), VP14637 (ViroPharma), R170591 (Janssen), RFI-641 (BMS) or BMS-43377 (BMS); and other inhibitors including but not limited to ribavirin.
  • fusion inhibitors including but not limited to Synagis® (palivizumab), VP14637 (ViroPharma), R170591 (Janssen), RFI-641 (BMS) or BMS-43377 (BMS); and other inhibitors including but not limited to ribavirin.
  • compositions of this invention may be administered orally, rectally, parenterally, via an implanted reservoir or topically by aerosol or nebulizer through the airways (nose or mouth). Oral or rectal administration or administration by injection is preferred.
  • the pharmaceutical compositions of this invention may contain any conventional non-toxic pharmaceutically-acceptable carriers, adjuvants or vehicles.
  • the pH of the formulation may be adjusted with pharmaceutically acceptable acids, bases or buffers to enhance the stability of the formulated compound or its delivery form.
  • parenteral as used herein includes but is not limited to subcutaneous, intracutaneous, intravenous, intramuscular, intra-articular, intrasynovial, intrasternal, intrathecal, and intralesional injection or infusion techniques.
  • compositions may be in the form of a sterile injectable preparation, for example, as a sterile injectable aqueous or oleaginous suspension.
  • This suspension may be formulated according to techniques known in the art using suitable dispersing or wetting agents (such as, for example Tween 80) and suspending agents.
  • compositions of this invention may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, and aqueous suspensions and solutions.
  • carriers which are commonly used include lactose and corn starch.
  • Lubricating agents such as magnesium stearate, are also typically added.
  • useful diluents include but are not limited to lactose and dried corn starch.
  • Dosage levels of between about 0.01 and about 100 mg/kg body weight per day, preferably between about 0.5 and about 75 mg/kg body weight per day of the polymerase inhibitor compounds described herein are useful in a monotherapy for the prevention and treatment of RSV mediated disease.
  • the pharmaceutical compositions of this invention will be administered from about 1 to about 5 times per day or alternatively, as a continuous infusion. Such administration can be used as a chronic or acute therapy.
  • the amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration.
  • a typical preparation will contain from about 5% to about 95% active compound (w/w).
  • such preparations contain from about 20% to about 80% active compound.
  • compositions of this invention comprise a combination of a compound of formula (I) or (Ia) and one or more additional therapeutic or prophylactic agent
  • both the compound and the additional agent should be present at dosage levels of between about 10 to 100%, and more preferably between about 10 and 80% of the dosage normally administered in a monotherapy regimen.
  • the resulting composition may be administered in vivo to mammals, such as man, to inhibit RSV polymerase or to treat or prevent RSV virus infection.
  • Such treatment may also be achieved using the compounds of this invention in combination with agents which include, but are not limited to: fusion inhibitors, including but not limited to Synagis® (palivizumab), VP14637 (ViroPharma), R170591 (Janssen), RFI-641 (BMS) or BMS-43377 (BMS); and other inhibitors including but not limited to ribavirin.
  • the additional agents may be combined with the compounds of this invention to create a single dosage form. Alternatively these additional agents may be separately administered to a mammal as part of a multiple dosage form.
  • another embodiment of this invention provides methods of inhibiting RSV polymerase activity in a mammal by administering a compound of the formula (I) or (Ia).
  • these methods are useful in decreasing RSV polymerase activity in a mammal.
  • the pharmaceutical composition comprises only a compound of this invention as the active component, such methods may additionally comprise the step of administering to said mammal an agent selected from an immunomodulatory agent, an antiviral agent, or another RSV polymerase inhibitor.
  • additional agent may be administered to the mammal prior to, concurrently with, or following the administration of the compositions of this invention.
  • these methods are useful for inhibiting viral replication in a mammal. Such methods are useful in treating or preventing RSV disease. If the pharmaceutical composition comprises only a compound of this invention as the active component, such methods may additionally comprise the step of administering to said mammal an agent selected from an immunomodulatory agent, an antiviral agent or another RSV polymerase inhibitor. Such additional agent may be administered to the mammal prior to, concurrently with, or following the administration of the composition according to this invention.
  • the compounds set forth herein may also be used as laboratory reagents.
  • the compounds of this invention may also be used to treat or prevent viral contamination of materials and therefore reduce the risk of viral infection of laboratory or medical personnel or patients who come in contact with such materials (e.g. blood, tissue, surgical instruments and garments, laboratory instruments and garments, and blood collection apparatuses and materials).
  • the compounds set forth herein may also be used as research reagents.
  • the compounds of this invention may also be used as positive control to validate cellular assays or in vitro or in vivo viral replication assays.
  • the compounds of this invention may be used as probes in displacement assays which measure displacement of such a probe from binding to an RSV polymerase as a means for identifying inhibitors of the RSV polymerase.
  • the side-chain on the isoquinoline-1,3(2H,4H)-dione 1(i) can be incorporated using a Mitsunobu reaction with alcohol 2(i) to give 2(ii).
  • Reduction of the nitro group on 2(ii) gave the diamino intermediate 2(iii) that was transformed to 1H-imidazol[4,5-h]isoquinoline-7,9(6H,8H)-dione 2(iv) using an aldehyde in presence of p-chloranil.
  • Alternative methods to generate 2(iv) from diamino 2(iii) are known to the skilled in the art.
  • BOC tert-butyloxycarbonyl; dba: dibenzylideneacetone; DIAD: diisopropyl azodicarboxylate; DMF: N,N-dimethylformamide; DMSO: dimethylsulfoxide; dppf: 1,1′-bis(diphenylphosphino)ferrocene; DTT: dithiothreitol; EDTA: ethylenediaminetetraacetate; EGTA: 1,2-di(2-aminoethoxy)ethane-N,N,N′,N′-tetraacetic acid; Et 2 O: diethyl ether; EtOAc: ethyl acetate; HPLC: high performance liquid chromatography; iPr: isopropyl; Me: methyl; MeOH: methanol; MeCN: acetonitrile; Ph: phenyl; SDS: sodium dodecyl sulfate; TFA: trifluor
  • reaction buffer 50 mM TrisAcetate (pH 7.5), 120 mM potassium acetate, 4.5 mM MgCl 2 , 5% glycerol, 2 mM EGTA, 3 mM DTT, 50 ⁇ g/ml BSA, 04 mM of each ATP, GTP and UTP, 4% DMSO). Reactions also contained 1 ⁇ Ci 3 H-CTP ( ⁇ 20 Cl/mmol).
  • Reactions were assembled in 96-well FlashPlateTM (PerkinElmer) to which 0.6 ⁇ mol oligo-dT 30 was previously immobilized in 10 mM Tris HCl (pH 7.5) 1 mM EDTA. 0.5 M LiCl, 0.5% SDS, 70 mM NaCl, 2.5 mM KCl. Following 2 h incubation at 30° C., the reaction was stopped with hybridization buffer (17 mM Tris HCl (pH 7.5), 1.7 mM EDTA, 0.85 M LiCl, 0.85% SDS, 120 mM NaCl, 3.75 mM KCl) which favors hybridization of RNA with DNA oligonucleotides.
  • hybridization buffer 17 mM Tris HCl (pH 7.5), 1.7 mM EDTA, 0.85 M LiCl, 0.85% SDS, 120 mM NaCl, 3.75 mM KCl
  • the radionucleotide that was incorporated into newly synthesized RNA was detected by proximity to scintillant embedded in the wells of the plate using a TopCountTM (Beckman) multi-well plate scintillation detector.
  • TopCountTM Beckman multi-well plate scintillation detector.
  • the compounds described in Tables 1 and 2 exhibited IC 50 ⁇ 5 ⁇ M in this assay.
  • HEp-2 cells were plated at 10,000 cells per 96 well in DMEM 10% FBS for 24 h.
  • the monolayer was infected with RSV-Long at an MOI of 0.1 pfu per cell in 50 ⁇ L DMEM 2% FBS.
  • the virus was removed after one hour adsorption and 100 ⁇ L media, with or without compound, was added. Incubation was continued for 48 h.
  • the monolayer was fixed with 0.063% glutaraldehyde.
  • the plates were blocked with 1% BSA in phosphate-buffered saline (PBS) for one hour.
  • PBS phosphate-buffered saline
  • Viral replication was detected with a monoclonal antibody directed against RSV F diluted 1/2000 in PBS1% BSA. After 1 h incubation, the plates were washed and Sheep anti mouse HRP diluted 1/6000 was added. One hour later, the plates were washed and were developed with o-phenylenediamine dihydrochloride (OPD) for thirty min. The absorbance was read at 450 nm. Percent inhibition was calculated using SAS program for non-linear regression analysis.
  • OPD o-phenylenediamine dihydrochloride
  • Tables 1 and 2 list compounds representative of the invention. All compounds described in Tables 1 and 2 exhibited IC 50 ⁇ 5 ⁇ M in the RSV polymerase assay of Example 4. In addition, many of the compounds described in Tables 1 and 2 are active in the RSV ELISA assay of Example 5.
  • MS Mass spectrometric data. TABLE 1 Cpd MS entry # R 1 R 2 R 3 (MH)+ 1001 H 557 1002 H 552 1003 H 595 1004 H 543 1005 H 595 1006 H 541 1007 H 606/608 1008 H 557 1009 H 586 1010 H 579/581 1011 H 581/563 1012 H 579/581 1013 H 571 1014 H 572 1015 H 595/597/599 1016 H 555 1017 H 602 1018 H 575 1019 H 615 1020 H 588 1021 H 618 1022 H 563 1023 H 563 1024 H 581 1025 H 559 1026 H 577 1027 H 567 1028 H 542 1029 H 578 1030 H 594 1031 H 600/602 1032 H 562 1033 H 566 1034 H 578 1035 H 579 1036 H 578 1037 H 578 1038 H 561 1039 H 578 1040 H 567 1031 H 600/602 1032 H 562 1033 H

Landscapes

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

Abstract

Compounds or enantiomers of formula (I) or a salt thereof:
Figure US20070225317A1-20070927-C00001

wherein R1, R2, R3, R4 and R5 are as defined herein, are useful for the manufacture of a medicament for the treatment or prevention of respiratory syncytial virus infection in a mammal.

Description

    RELATED APPLICATIONS
  • This application claims benefit to U.S. provisional application No. 60/515,820 filed on Oct. 30, 2003.
  • FIELD OF THE INVENTION
  • The present invention relates to compounds, process for their synthesis, compositions and methods for the treatment or prevention of Respiratory Syncytial Virus (RSV) infection. In particular, the present invention provides novel compounds, pharmaceutical compositions containing such compounds and methods for using these compounds in the treatment or prevention of Respiratory Syncytial Virus infection.
  • BACKGROUND OF THE INVENTION
  • Respiratory Syncytial Virus (RSV) is the most common cause of severe lower respiratory tract infections (LRTI) in young infants, virtually all of whom suffer at least one RSV infection in the upper respiratory tract by the age of two. In very young infants, infection can progress to the lower respiratory tract causing bronchiolitis and pneumonia that may necessitate hospitalization.
  • RSV is an enveloped virus with a negative strand non-segmented RNA genome. It belongs to the Mononegavirales order, Paramyxoviridae family and Pneumovirus genus. RSV exhibits nucleocapsid-associated RNA dependent RNA polymerase (RdRp) activity for viral transcription and replication, which occurs in the cytoplasm of an infected cell.
  • The RSV polymerase is comprised of at least five viral components: the genomic RNA, and the L, N, P and M2-1 proteins. Together these components form a Ribonucleoprotein (RNP) complex with RdRp activity required for the synthesis of both viral genomic RNA (“replicase” activity) and sub-genomic mRNAs (“transcriptase” activity). As opposed to the replication of viral products, RSV mRNAs are co-transcriptionally capped (i.e. guanylated and methylated) at their 5′ ends and polyadenylated at their 3′ ends by the RNP complex. These modifications are necessary for translation of the viral mRNAs by the host protein synthesis machinery. The RNP complex functions exclusively in the cytoplasm of the RSV-infected cells. Since the host proteins responsible for capping of mRNAs are located in the nucleus of the cell, capping of its mRNAs by the viral RNP complex is essential for the synthesis of RSV proteins. Thus, the RSV RNP complex is an attractive target in screening for potential antiviral therapeutics since it has multiple activities essential for viral replication.
  • U.S. Pat. No. 4,176,184 discloses 7,7-dimethyl-2-(2-methoxyphenyl)-5-[3-(2-(3,4-dimethoxyphenyl)ethylamino)propyl]-5H,7H-imidazo[4,5-h]isoquinoline-4,6-dione dihydrochloride (example 28) having anti-arrhythmic activity.
  • The present invention therefore provides novel compounds, compositions, uses and methods that inhibit Respiratory Syncytial virus polymerase essential for viral replication.
  • SUMMARY OF THE INVENTION
  • In a first aspect, the invention provides a compound of Formula (Ia), or an enantiomer or a salt thereof:
    Figure US20070225317A1-20070927-C00002

    wherein R1 is —(CH═CH)0-1—(C6 or C10)aryl or —(CH═CH)0-1-5-, 6-, 9- or 10-membered heteroaryl, said aryl or heteroaryl being optionally substituted with one, two or three substituents, each independently selected from:
      • (C1-14)alkyl optionally substituted with amino, halo, (C1-6)haloalkyl, hydroxy, (C1-6)alkoxy, (C1-6)alkylthio, nitro, azido, cyano, amino, (C1-6)alkylamino, di((C1-6)alkyl)amino, aryl and heteroaryl;
        R2 is H, (C1-4)alkyl, hydroxy, halo, (C1-6)haloalkyl, amino, (C1-6)alkylamino, di((C1-6)alkyl)amino, or (C2-6)alkynyl;
        R3 is (C6, C10 or C14)aryl or 5-, 6-, 9- or 10-membered heteroaryl, each of which being optionally substituted with one, two or three substituents, each independently selected from:
      • (C1-4)alkyl, halo, (C1-6)haloalkyl, hydroxy, (C1-6)alkoxy, (C1-6)alkylthio, nitro, amino, (C1-8)alkylamino, di((C1-6)alkyl)amino and COO(C1-6)alkyl; and
        R4 and R6 are each independently H or (C1-8)alkyl; or R4 and R5 are linked, together with the carbon atom to which they are attached, to form a (C3-7)cycloalkyl group; with the proviso that R1 is not 2-methoxyphenyl, when R2 is H, R3 is 3,4-dimethoxyphenyl, R4 is CH3 and R5 is CH3.
  • In a second aspect, the invention provides the use of a compound of formula (I), or an enantiomer or a salt thereof:
    Figure US20070225317A1-20070927-C00003

    wherein R1 is —(CH═CH)0-1—, —(C6 or C10)aryl or —(CH═CH)0-1-5-, 6-, 9- or 10-membered heteroaryl, said aryl or heteroaryl being optionally substituted with one, two or three substituents, each independently selected from:
      • (C1-4)alkyl optionally substituted with amino, halo, (C1-6)haloalkyl, hydroxy, (C1-6)alkoxy, (C1-6)alkylthio, nitro, azido, cyano, amino, (C1-6)alkylamino, di((C1-6)alkyl)amino, aryl and heteroaryl;
        R2 is H, (C1-6)alkyl, hydroxy, halo, (C1-6)haloalkyl, amino, (C1-6)alkylamino, di((C1-6)alkyl)amino, or (C2-6)alkynyl;
        R3 is (C6, C10 or C14)aryl or 5-, 6-, 9- or 10-membered heteroaryl, each of which being optionally substituted with one, two or three substituents, each independently selected from:
      • (C1-6)alkyl, halo, (C1-6)haloalkyl, hydroxy, (C1-8)alkoxy, (C1-6)alkylthio, nitro, amino, (C1-6)alkylamino, di((C1-6)alkyl)amino and COO(C1-6)alkyl; and
        R4 and R5 are each independently H or (C1-6)alkyl; or R4 and R5 are linked, together with the carbon atom to which they are attached, to form a (C3-7)cycloalkyl group; in the manufacture of a medicament for the treatment or prevention of respiratory syncytial virus infection in a mammal.
  • In a third aspect, the invention provides a pharmaceutical composition comprising a therapeutically effective and acceptable amount of a compound of formula (I) or (Ia) in association with a pharmaceutically-acceptable carrier.
  • In a fourth aspect, the invention provides a pharmaceutical composition for use in the treatment or prevention of respiratory syncytial virus infection, wherein the composition comprises a therapeutically effective and acceptable amount of a compound of formula (I) or (Ia) in association with a pharmaceutically-acceptable carrier.
  • According to an alternate embodiment of this aspect, the pharmaceutical composition according to this invention further comprises a therapeutically effective amount of at least one other antiviral agent.
  • In a fifth aspect, the invention provides an anti-respiratory syncytial virus pharmaceutical composition comprising an anti-respiratory syncytial virally-effective amount of a compound of formula (I) or (Ia), alone or in combination with at least one other antiviral agent, in association with a pharmaceutically-acceptable carrier.
  • In a sixth aspect, the invention provides a use of a compound of formula (I) or (Ia) to inhibit the replication of a respiratory syncytial virus.
  • In a seventh aspect, the invention provides a use of a compound of formula (I) or (Ia), in the treatment or prevention of a respiratory syncytial virus infection in a mammal.
  • In an eighth aspect, the invention provides a method of treating or preventing a respiratory syncytial virus infection in a mammal comprising administering to the mammal an anti-respiratory syncytial virally-effective and acceptable amount of a compound of formula (I) or (Ia) or a composition containing such a compound, alone or in combination with at least one other antiviral agent, administered together or separately.
  • In a ninth aspect, the invention provides a method of inhibiting replication of a respiratory syncytial virus comprising exposing virus-infected cells to a anti-respiratory syncytial virally-effective and acceptable amount of at least one of a compound of formula (I) or (Ia).
  • In a tenth aspect, the invention provides a packaged pharmaceutical comprising a pharmaceutical composition containing a compound of formula (I) or (Ia) and directions identifying an administration regimen.
  • In an eleventh aspect, the invention provides a packaged pharmaceutical for use for the treatment or prevention of respiratory syncytial virus infection in a mammal, wherein the packaged pharmaceutical comprises a pharmaceutical composition containing a compound of formula (I) or (Ia) and directions identifying an administration regimen.
  • In a twelfth aspect, the invention provides an article of manufacture comprising packaging material contained within which is a composition effective to inhibit a respiratory syncytial virus, the packaging material comprising a label which indicates that the composition can be used to treat or prevent infection by a respiratory syncytial virus, wherein said composition includes a compound of formula (I) or (Ia).
  • DETAILED DESCRIPTION OF PREFERRED EMBODIMENTS
  • Definitions
  • As used herein, the following definitions apply unless otherwise noted:
  • The term “halo” as used herein means a halogen radical selected from bromo, chloro, fluoro or iodo.
  • The term “(C1-6)alkyl” as used herein, either alone or in combination with another radical, means straight or branched-chain alkyl radicals containing up to six carbon atoms and includes, but is not limited to, methyl (Me), ethyl (Et), propyl (Pr), butyl (Bu), hexyl, 1-methylethyl (iPr), 1-methylpropyl (sec-Bu), 2-methylpropyl (iBu) and 1,1-dimethylethyl (tBu); wherein the abbreviations commonly used herein are given in parentheses.
  • The term “(C2-6)alkynyl” as used herein, either alone or in combination with another radical, means straight or branched-chain alkyne radicals containing from two to six carbon atoms, at least two of which are linked by a triple bond and includes, but is not limited to, —C≡CH.
  • The term “(C3-7)cycloalkyl” as used herein, either alone or in combination with another radical, means saturated cyclic hydrocarbon radicals containing from three to seven carbon atoms and includes cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl.
  • The terms “(C1-6)alkoxy” or “O—(C1-6)alkyl” as used herein interchangeably, mean a straight chain alkyl containing one to six carbon atoms linked through an oxygen atom or a branched chain alkyl radical containing three to six carbon atoms linked through an oxygen atom and includes but is not limited to methoxy, ethoxy, propoxy, 1-methylethoxy, butoxy and 1,1-dimethylethoxy. The latter radical is known commonly as tert-butoxy.
  • The term “(C1-6)haloalkyl” as used herein means an alkyl radical containing one to six carbon atoms wherein one or more hydrogen atoms are replaced by a halogen atom (including but not limited to trifluoromethyl).
  • The terms “(C1-6)alkylthio” or “S—(C1-6)alkyl” as used herein interchangeably, mean a straight chain alkyl containing one to six carbon atoms linked through a sulfur atom, or a branched chain alkyl radical containing three to six carbon atoms linked through a sulfur atom.
  • The term “amino” as used herein means an amino radical of formula —NH2. The term “(C1-4)alkylamino” or “NH—(C1-6)alkyl” as used herein means an alkyl radical containing one to six carbon atoms linked through a nitrogen atom, and includes but is not limited to methylamino, propylamino, (1-methylethyl)amino and (2-methylbutyl)amino. The term “di((C1-6)alkyl)amino” or “N((C1-6)alkyl)2” means an amino radical having two (C1-6)alkyl substituents, each of which contains one to six carbon atoms and includes but is not limited to dimethylamino, diethylamino, ethylmethylamino and the like.
  • The terms “aryl” or “(C6 or C10 or C14)aryl” used interchangeably, either alone or in combination with another radical, mean either an aromatic monocyclic system containing 6 carbon atoms or an aromatic bicyclic system containing 10 carbon atoms, or an aromatic tricyclic system containing 14 carbon atoms. For example, aryl includes but is not limited to phenyl, naphthyl or anthryl.
  • The terms “(C7-16)aralkyl” or “(C1-6)alkyl-aryl” used interchangeably, either alone or in combination with another radical, means an aryl as defined above linked through an alkyl group, wherein alkyl is as defined above containing from 1 to 6 carbon atoms. Aralkyl includes, but is not limited to, for example, benzyl, and phenylbutyl.
  • The term “Het” or “heterocycle” as used herein means a monovalent radical derived by removal of a hydrogen from a five- or six-membered, saturated or unsaturated (including aromatic) ring system containing from one to three heteroatoms each independently selected from nitrogen, oxygen and sulfur. Optionally, the heterocycle may bear one or two substituents; for example, N-oxido, (C1-4)alkyl, (C1-3)alkyl-phenyl, (C1-6)alkoxy, halo, amino or (C1-6)alkylamino. Again optionally, the five- or six-membered heterocycle can be fused to a second cycloalkyl, an aryl (e.g. phenyl) or another heterocycle.
  • Examples of suitable heterocycles and optionally substituted heterocycles include, but are not limited to, morpholine, thiadiazole, quinoline, isoquinoline, 3,4-methylene-dioxyphenyl, benzothiazole, benzofuran, benzothiophene, pyrrolidine, tetrahydrofuran, thiazolidine, pyrrole, indole, benzimidazole, 1H-imidazole, 1-methyl-1H-imidazole, pyrazole, furan, thiophene, oxazole, isoxazole, thiazole, 2-methylthiazole, 2-aminothiazole, 2-(methylamino)thiazole, piperidine, 1-methylpiperidine, 1-methylpiperazine, 1,4-dioxane, pyridine, pyridine N-oxide, pyrimidine, 2,4-dihydroxypyrimidine. 2,4-dimethylpyrimidine, 2,6-dimethylpyridine, 1-methyl-1H-tetrazole, 2-methyl-2H-tetrazole, benzoxazole, thiazolo[4,5-b]-pyridine, heterocycles selected from the following group:
    Figure US20070225317A1-20070927-C00004

    or each heterocycle exemplified in Tables 1 and 2 below.
  • The term “heteroaryl” as used herein means a 5- or 6-membered monocylic, or 9 or 10-membered bicyclic aromatic heterocycle as defined above. Specifically, heteroaryl includes but is not limited to: indole, benzimidazole, imidazole, furan, thiophene, oxazole, thiazole, pyrazole, pyrrole, pyridine, pyrimidine, benzofuran, benzothiophene, isoquinoline, quinoline, heteroaryl selected from the following group:
    Figure US20070225317A1-20070927-C00005

    or each heteroaryl exemplified in Tables 1 and 2 below.
  • The term “pharmaceutically acceptable carrier” as used herein means a non-toxic, generally inert vehicle for the active ingredient which does not adversely affect the ingredient.
  • The term “mammal” as it is used herein is meant to encompass humans, as well as non-human mammals which are susceptible to infection by respiratory syncytial virus including domestic animals, including but not limited to cattle, pigs, horses, sheep, dogs and cats, and non-domestic animals.
  • The term “effective amount” means a predetermined antiviral amount of the antiviral agent, i.e. an amount of the agent sufficient to be effective against the virus in vivo. It is understood that when an antiviral agent is administered in combination with another antiviral agent, the effective amount of each antiviral agent administered in combination may be lower or higher than the effective amount of each antiviral agent administered alone. Such a situation may arise, for example, when the antiviral agents administered in combination act synergistically with each other, so that the effective amount of either or both of the antiviral agents administered in combination is lower than the effective amount of either or both of the antiviral agents administered alone. In an alternative example, the antiviral agents may act antagonistically with each other, so that the effective amount of either or both of the antiviral agents administered in combination is higher than the effective amount of either or both of the antiviral agents administered alone.
  • The term “salt thereof” means any acid addition salt of a compound according to the invention; preferably a pharmaceutically acceptable salt thereof.
  • The term “pharmaceutically acceptable salt” means a salt of a compound of formula (I) or (Ia) which is, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, generally water or oil-soluble or dispersible, and effective for their intended use. The term includes pharmaceutically-acceptable acid addition salts. Examples of suitable salts are found in, e.g., S. M. Birge et al., J. Pharm. Sci., 1977, 6, pp. 1-19.
  • The term “pharmaceutically-acceptable acid addition salt” means those salts which retain the biological effectiveness and properties of the free bases and which are not biologically or otherwise undesirable, formed with inorganic acids including but not limited to hydrochloric acid, hydrobromic acid, sulfuric acid, sulfamic acid, nitric acid, phosphoric acid, and the like, and organic acids including but not limited to acetic acid, trifluoroacetic acid, adipic acid, ascorbic acid, aspartic acid, benzenesulfonic acid, benzoic acid, butyric acid, camphoric acid, camphorsulfonic acid, cinnamic acid, citric acid, digluconic acid, ethanesulfonic acid, glutamic acid, glycolic acid, glycerophosphoric acid, hemisulfic acid, hexanoic acid, formic acid, fumaric acid, 2-hydroxyethanesulfonic acid (isethionic acid), lactic acid, hydroxymaleic acid, malic acid, malonic acid, mandelic acid, mesitylenesulfonic acid, methanesulfonic acid, naphthalenesulfonic acid, nicotinic acid, 2-naphthalenesulfonic acid, oxalic acid, pamoic acid, pectinic acid, phenylacetic acid, 3-phenylpropionic acid, pivalic acid, propionic acid, pyruvic acid, salicylic acid, stearic acid, succinic acid, sulfanilic acid, tartaric acid, p-toluenesulfonic acid, undecanoic acid, and the like.
  • The term “antiviral agent” as used herein means an agent (compound or biological) that is effective to inhibit the formation and/or replication of a virus in a mammal. This includes agents that interfere with either host or viral mechanisms necessary for the formation and/or replication of a virus in a mammal. Such agents can be selected from immunomodulatory agents, inhibitors of RSV polymerase or inhibitors of another target in the RSV life cycle.
  • The term “immunomodulatory agent” as used herein means those agents (compounds or biologicals) that are effective to enhance or potentiate the immune system response in a mammal, Immunomodulatory agents include, for example, class I interferons (such as α-(alpha), β-(beta), δ-(delta), ω-(omega) and τ-(tau) interferons, consensus interferons and asialo-interferons), class II interferons (such as γ-(gamma) interferons) and pegylated interferons.
  • The term “inhibitor of RSV polymerase” as used herein means an agent (compound or biological) that is effective to inhibit the function of an RSV polymerase in a mammal.
  • The term “inhibitor of another target in the RSV life cycle” as used herein means an agent (compound or biological) that is effective to inhibit the formation and/or replication of RSV in a mammal other than by inhibiting the function of the RSV polymerase. This includes agents that interfere with either-host or RSV viral mechanisms necessary for the formation and/or replication of RSV in a mammal. Inhibitors of another target in the RSV life cycle include but are not limited to fusion inhibitors, including but not limited to Synagis® (palivizumab), VP14637 (ViroPharma), R170591 (Janssen), RFI-641 (BMS) or BMS-43377 (BMS); and other inhibitors including but not limited to ribavirin.
  • As used herein, the term “treatment” means the administration of a compound or composition according to the present invention to alleviate or eliminate symptoms of respiratory syncytial virus infection and/or to reduce or eliminate viral load in a patient and/or to reduce the incidence and/or length of time of hospitalization.
  • As used herein, the term “hospitalization” means medical care in a hospital, clinic or other medical facility to treat respiratory syncytial virus infection.
  • As used herein, the term “prevention” or “prophylaxis”, used interchangeably, means the administration of a compound or composition according to the present invention pre-exposure of the individual to the virus or post-exposure of the individual to the virus but before the appearance of symptoms of the disease, and/or prior to the detection of viral infection, to prevent the appearance of symptoms of the disease.
  • The following sign
    Figure US20070225317A1-20070927-C00006

    is used in sub-formulas to indicate the bond which is connected to the rest of the molecule as defined.
  • Preferred Embodiments
  • Compounds
  • According to a first embodiment, the invention provides a compound of Formula (Ia) or an enantiomer thereof or a salt thereof,
    Figure US20070225317A1-20070927-C00007

    wherein R1 is —(CH═CH)0-1—(C6 or G10)aryl or 5-, 6-, 9- or 10-membered heteroaryl, said aryl or heteroaryl being optionally substituted with one, two or three substituents, each independently selected from:
      • (C1-4)alkyl optionally substituted with amino, halo, (C1-6)haloalkyl, hydroxy, (C1-6)alkoxy, (C1-6)alkylthio, nitro, azido, cyano, amino, (C1-6)alkylamino, di((C1-6)alkyl)amino, aryl and heteroaryl;
        R2 is H, (C1-6)alkyl, hydroxy, halo, (C1-6)haloalkyl, amino, (C1-6)alkylamino, di((C1-6)alkyl)amino, or (C2-4)alkynyl;
        R3 is (C6, C10 or C14)aryl or 5-, 6-, 9- or 10-membered heteroaryl, each of which being optionally substituted with one, two or three substituents, each independently selected from:
      • (C1-6)alkyl, halo, (C1-6)haloalkyl, hydroxy, (C1-6)alkoxy, (C1-6)alkylthio, amino, (C1-6)alkylamino and di((C1-6)alkyl)amino; and
        R4 and R5 are each independently (C1-6)alkyl; or R4 and R5 are linked, together with the carbon atom to which they are attached, to form a (C3-7)cycloalkyl group; with the proviso that R1 is not 2-methoxyphenyl, when R2 is H, R3 is 3,4-dimethoxyphenyl, R4 is CH3 and R5 is CH3.
  • Preferably, compounds of formula (Ia) are those wherein:
  • R1 is (CH═CH)0-1-phenyl, the phenyl being optionally substituted with one, two or three substituents, each independently selected from:
      • (C1-6)alkyl optionally substituted with amino, halo, (C1-6)haloalkyl, hydroxy, (C1-6)alkoxy, (C1-6)alkylthio, amino, nitro, cyano, azido, (C1-6)alkylamino, di((C1-6)alkyl)amino, and heteroaryl;
        or R1 is a 5-, 6-, 9- or 10-membered heteroaryl optionally substituted with one, two or three substituents, each independently selected from nitro, (C1-6)alkyl optionally substituted with amino, (C1-6)haloalkyl, hydroxy, (C1-6)alkoxy, halo, (C1-6)alkylthio, cyano and heteroaryl;
        R2 is H, (C1-6)alkyl, hydroxy, halo, amino, (C1-6)alkylamino, or (C2-6)alkynyl;
        R3 is naphthyl, anthryl, or phenyl optionally substituted with one, two or three substituents, each independently selected from:
  • (C1-6)alkyl, halo, (C1-6)haloalky, hydroxy, (C1-4)alkoxy, and (C1-8)alkylthio; or R3 is a 5-, 6-, 9- or 10-membered heteroaryl optionally substituted with (C1-6)alkyl; and
  • R4 and R5 are each independently Me or Et; or R4 and R5 are linked, together with the carbon atom to which they are attached, to form a (C3-7)cycloalkyl group; with the proviso that R1 is not 2-methoxyphenyl, when R2 is H, R3 is 3,4-dimethoxyphenyl, R4 is CH3 and R5 is CH3.
  • More preferred are compounds of formula (Ia) wherein:
  • R1 is phenyl, 2-pyridyl, quinolinyl or benzofuranyl, the phenyl and 2-pyridyl each being optionally substituted with one or two substituents, each independently selected from:
      • methyl, hydroxy, methoxy, nitro, cyano, methylthio, fluoro, 2-aminoethyl and CF3;
        R2 is H, NH2, bromo, chloro, or OH;
        R3 is naphthyl, or phenyl optionally substituted with one, two or three substituents, each independently selected from:
      • methyl, iodo, hydroxy, methoxy, ethoxy and methylthio; and
        R4 and R5 are each independently Me; or R4 and R5 are linked, together with the carbon atom to which they are attached, to form a cyclopentyl or cyclohexyl group; with the proviso that R1 is not 2-methoxyphenyl, when R2 is H, R3 is 3,4-dimethoxyphenyl, R4 is CH3 and R5 is CH3.
  • Even more preferred are compounds of formula (Ia) wherein:
  • R1 is phenyl, 2-pyridyl, quinolinyl or benzofuranyl, the phenyl and 2-pyridyl each being optionally substituted with one or two substituents, each independently selected from:
      • methyl, methoxy, fluoro, CF3, nitro, cyano, and methylthio;
  • R2 is H, NH2, bromo, chloro, or OH;
  • R3 is phenyl optionally substituted with one, two or three substituents, each independently selected from:
      • hydroxy, methoxy, ethoxy, methyl, iodo and methylthio; and
  • R4 and R5 are each independently Me; or R4 and R5 are linked, together with the carbon atom to which they are attached, to form a cyclopentyl or cyclohexyl group; with the proviso that R1 is not 2-nethoxyphenyl, when R2 is H, R3 is 3,4-dimethoxyphenyl, R4 is CH3 and R5 is CH3.
  • Most preferred are compounds of formula (Ia) wherein:
    R1 is
    Figure US20070225317A1-20070927-C00008

    R2 is NH2 or bromo;
    R3 is phenyl substituted with one or two substituents each independently selected from hydroxy, methoxy, and iodo; and
    R4 and R5 are each independently Me; or R4 and R5 are linked, together with the carbon atom to which they are attached, to form a cyclohexyl group.
  • Specific Embodiments
  • Included within the scope of this invention is each single compound presented in Tables 1 and 2 below with the proviso that R1 is not 2-methoxyphenyl, when R2 is H, R3 is 3,4-dimethoxyphenyl, R4 is CH3 and R5 is CH3,
  • Anti-Respiratory Syncytial Virus Activity
  • According to a second embodiment of the present invention, compounds of the present invention are useful in the treatment or prevention of respiratory syncytial virus infections.
  • According to this second embodiment of the present invention, the use of compounds of the formula (I) or enantiomers thereof or salts thereof, are provided in the manufacture of a medicament for the treatment or prevention of respiratory syncytial virus infection in a mammal:
    Figure US20070225317A1-20070927-C00009

    wherein R1 is —(CH═CH)0-1—(C6 or C10)aryl or 5-, 6-, 9- or 10-membered heteroaryl, said aryl or heteroaryl being optionally substituted with one, two or three substituents, each independently selected from:
      • (C1-6)alkyl optionally substituted with amino, halo, (C1-6)haloalkyl, hydroxy, (C1-6)alkoxy, (C1-6)alkylthio, nitro, azido, cyano, amino, (C1-6)alkylamino, di((C1-6)alkylamino, aryl and heteroaryl;
        R2 is H, (C1-4)alkyl, hydroxy, halo, (C1-6haloalkyl, amino, (C1-6)alkylamino, di((C1-6)alkyl)amino, or (C2-6)alkynyl;
        R3 is (C6, C10 or C14)aryl or 5-, 6-, 9- or 10-membered heteroaryl, each of which being optionally substituted with one, two or three substituents, each independently selected from:
      • (C1-6)alkyl, halo, (C1-6)haloalkyl, hydroxy, (C1-6)alkoxy, (C1-6)alkylthio, amino, (C1-6)alkylamino and di((C1-6)alkyl)amino; and
        R4 and R5 are each independently (C1-8)alkyl; or R4 and R5 are linked, together with the carbon atom to which they are attached, to form a (C3-7)cycloalkyl group.
  • Preferred is the use of compounds of formula (I) wherein:
  • R1 is (CH═CH)0-1—-phenyl, the phenyl being optionally substituted with one, two or three substituents, each independently selected from:
  • (C1-4)alkyl optionally substituted with amino, halo, (C1-6)haloalkyl, hydroxy,
  • (C1-4)alkoxy, (C1-4)alkylthio, amino, nitro, cyano, azido, (C1-8)alkylamino, di((C1-6)alkyl)amino, and heteroaryl; or R1 is a 5-, 6-, 9- or 10-membered heteroaryl optionally substituted with one, two or three substituents, each independently selected from nitro, (C1-6)alkyl optionally substituted with amino, (C1-6)haloalkyl, hydroxy, (C1-6)alkoxy, halo, (C1-6)alkylthio, cyano and heteroaryl;
  • R2 is H, (C1-6)alkyl, hydroxy, halo, amino, (C1-6)alkylamino, or (C2-4)alkynyl;
  • R3 is naphthyl, anthryl, or phenyl optionally substituted with one, two or three substituents, each independently selected from:
      • (C1-4)alkyl, halo, (C1-6)haloalkyl, hydroxy, (C1-6)alkoxy, and (C1-6)alkylthio;
        or R3 is a 5-, 6-, 9- or 10-membered heteroaryl optionally substituted with (C1-6)alkyl; and
        R4 and R5 are each independently Me or Et; or R4 and R5 are linked, together with the carbon atom to which they are attached, to form a (C3-7)cycloalkyl group.
  • More preferred is the use of compounds of formula (I) wherein:
  • R1 is phenyl, 2-pyridyl, quinolinyl or benzofuranyl, the phenyl and 2-pyridyl each being optionally substituted with one or two substituents, each independently selected from:
      • methyl, hydroxy, methoxy, nitro, cyano, methylthio, fluoro, 2-aminoethyl and CF3;
        R2 is H, NH2, bromo, chloro, or OH;
        R3 is naphthyl, or phenyl optionally substituted with one, two or three substituents, each independently selected from:
      • methyl, iodo, hydroxy, methoxy, ethoxy and methylthio; and
        R4 and R5 are each independently Me; or R4 and R5 are linked, together with the carbon atom to which they are attached, to form a cyclopentyl or cyclohexyl group.
  • Even more preferred is the use of compounds of formula (I) wherein:
  • R1 is phenyl, 2-pyridyl, quinolinyl or benzofuranyl, the phenyl and 2-pyridyl each being optionally substituted with one or two substituents, each independently selected from:
      • methyl, methoxy, fluoro, CF3, nitro, cyano, and methylthio;
        R2 is H, NH2, bromo, chloro, or OH;
        R3 is phenyl optionally substituted with one, two or three substituents, each independently selected from:
      • hydroxy, methoxy, ethoxy, methyl, iodo and methylthio; and
        R4 and R5 are each independently Me; or R4 and R5 are linked, together with the carbon atom to which they are attached, to form a cyclopentyl or cyclohexyl group.
  • Most preferred is the use of compounds of formula (I) wherein:
    R1 is
    Figure US20070225317A1-20070927-C00010

    R2 is NH2 or bromo;
    R3 is phenyl substituted with one or two substituents each independently selected from hydroxy, methoxy, and iodo; and
    R4 and R5 are each independently Me; or R4 and R5 are linked, together with the carbon atom to which they are attached, to form a cyclohexyl group.
  • Included within the scope of this invention is the use of each single compound presented in Tables 1 and 2 below for the manufacture of a medicament for the treatment or prevention of respiratory syncytial virus infection in a mammal.
  • According to a further embodiment the invention provides a pharmaceutical composition comprising a therapeutically effective and acceptable amount of a compound of formula (I) or (Ia) in association with a pharmaceutically-acceptable carrier.
  • In a further aspect, the invention provides a pharmaceutical composition for use in the treatment or prevention of respiratory syncytial virus infection, wherein the composition comprises a therapeutically effective and acceptable amount of a compound of formula (I) or (Ia), alone or in combination with at least one other antiviral agent, in association with a pharmaceutically-acceptable carrier.
  • According to an alternate embodiment of this aspect, the pharmaceutical compositions according to this invention may additionally comprise one or more immunomodulatory agents.
  • According to another alternate embodiment, the pharmaceutical compositions of this invention may additionally comprise one or more other inhibitors of RSV polymerase.
  • According to yet another alternate embodiment, the pharmaceutical compositions of this invention may additionally comprise one or more inhibitors of other targets in the RSV life cycle, including but not limited to, fusion inhibitors, including but not limited to Synagis® (palivizumab), VP14637 (ViroPharma), R170591 (Janssen), RFI-641 (BMS) or BMS-43377 (BMS); and other inhibitors including but not limited to ribavirin.
  • The pharmaceutical compositions of this invention may be administered orally, rectally, parenterally, via an implanted reservoir or topically by aerosol or nebulizer through the airways (nose or mouth). Oral or rectal administration or administration by injection is preferred. The pharmaceutical compositions of this invention may contain any conventional non-toxic pharmaceutically-acceptable carriers, adjuvants or vehicles. In some cases, the pH of the formulation may be adjusted with pharmaceutically acceptable acids, bases or buffers to enhance the stability of the formulated compound or its delivery form. The term parenteral as used herein includes but is not limited to subcutaneous, intracutaneous, intravenous, intramuscular, intra-articular, intrasynovial, intrasternal, intrathecal, and intralesional injection or infusion techniques.
  • The pharmaceutical compositions may be in the form of a sterile injectable preparation, for example, as a sterile injectable aqueous or oleaginous suspension. This suspension may be formulated according to techniques known in the art using suitable dispersing or wetting agents (such as, for example Tween 80) and suspending agents.
  • The pharmaceutical compositions of this invention may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, and aqueous suspensions and solutions. In the case of tablets for oral use, carriers which are commonly used include lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added. For oral administration in a capsule form, useful diluents include but are not limited to lactose and dried corn starch. When aqueous suspensions are administered orally, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening and/or flavoring and/or coloring agents may be added.
  • Other suitable vehicles or carriers for the above noted formulations and compositions can be found in standard pharmaceutical texts, e.g. in “Remington's Pharmaceutical Sciences”, The Science and Practice of Pharmacy, 19th Ed. Mack Publishing Company, Easton, Pa., (1995).
  • Dosage levels of between about 0.01 and about 100 mg/kg body weight per day, preferably between about 0.5 and about 75 mg/kg body weight per day of the polymerase inhibitor compounds described herein are useful in a monotherapy for the prevention and treatment of RSV mediated disease. Typically, the pharmaceutical compositions of this invention will be administered from about 1 to about 5 times per day or alternatively, as a continuous infusion. Such administration can be used as a chronic or acute therapy. The amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. A typical preparation will contain from about 5% to about 95% active compound (w/w). Preferably, such preparations contain from about 20% to about 80% active compound.
  • As the skilled artisan will appreciate, lower or higher doses than those recited above may be required. Specific dosage and treatment regimens for any particular patient will depend upon a variety of factors, including the activity of the specific compound employed, the age, body weight, general health status, sex, diet, time of administration, rate of excretion, drug combination, the severity and course of the infection, the patient's disposition to the infection and the judgment of the treating physician. Generally, treatment is initiated with small dosages substantially less than the optimum dose of the compound. Thereafter, the dosage is increased by small increments until the optimum effect under the circumstances is reached. In general, the compound is most desirably administered at a concentration level that will generally afford antivirally effective results without causing any harmful or deleterious side effects.
  • When the compositions of this invention comprise a combination of a compound of formula (I) or (Ia) and one or more additional therapeutic or prophylactic agent, both the compound and the additional agent should be present at dosage levels of between about 10 to 100%, and more preferably between about 10 and 80% of the dosage normally administered in a monotherapy regimen.
  • When these compounds or their pharmaceutically acceptable salts are formulated together with a pharmaceutically acceptable carrier, the resulting composition may be administered in vivo to mammals, such as man, to inhibit RSV polymerase or to treat or prevent RSV virus infection. Such treatment may also be achieved using the compounds of this invention in combination with agents which include, but are not limited to: fusion inhibitors, including but not limited to Synagis® (palivizumab), VP14637 (ViroPharma), R170591 (Janssen), RFI-641 (BMS) or BMS-43377 (BMS); and other inhibitors including but not limited to ribavirin. The additional agents may be combined with the compounds of this invention to create a single dosage form. Alternatively these additional agents may be separately administered to a mammal as part of a multiple dosage form.
  • Accordingly, another embodiment of this invention provides methods of inhibiting RSV polymerase activity in a mammal by administering a compound of the formula (I) or (Ia).
  • In a preferred embodiment, these methods are useful in decreasing RSV polymerase activity in a mammal. If the pharmaceutical composition comprises only a compound of this invention as the active component, such methods may additionally comprise the step of administering to said mammal an agent selected from an immunomodulatory agent, an antiviral agent, or another RSV polymerase inhibitor. Such additional agent may be administered to the mammal prior to, concurrently with, or following the administration of the compositions of this invention.
  • In an alternate preferred embodiment, these methods are useful for inhibiting viral replication in a mammal. Such methods are useful in treating or preventing RSV disease. If the pharmaceutical composition comprises only a compound of this invention as the active component, such methods may additionally comprise the step of administering to said mammal an agent selected from an immunomodulatory agent, an antiviral agent or another RSV polymerase inhibitor. Such additional agent may be administered to the mammal prior to, concurrently with, or following the administration of the composition according to this invention.
  • The compounds set forth herein may also be used as laboratory reagents. The compounds of this invention may also be used to treat or prevent viral contamination of materials and therefore reduce the risk of viral infection of laboratory or medical personnel or patients who come in contact with such materials (e.g. blood, tissue, surgical instruments and garments, laboratory instruments and garments, and blood collection apparatuses and materials).
  • The compounds set forth herein may also be used as research reagents. The compounds of this invention may also be used as positive control to validate cellular assays or in vitro or in vivo viral replication assays. In addition, the compounds of this invention may be used as probes in displacement assays which measure displacement of such a probe from binding to an RSV polymerase as a means for identifying inhibitors of the RSV polymerase.
  • Methodology
  • The compounds of the present invention were synthesized according to a general process as illustrated in scheme I (wherein R1, R2, R3, R4 and R5 are as defined hereinbefore and R1a, R2a and R3a are groups which may be converted to R1, R2 and R3, respectively):
    Figure US20070225317A1-20070927-C00011
  • The general synthesis described in Scheme 1 is similar to the one described in U.S. Pat. No. 4,176,184. Briefly, reduction of 8-nitroisoquinoline-1,3(2H,4H)-dione 1(i) (prepared according to the procedure described in DE 3410168) to the corresponding aniline 1(ii) followed by acylation with acyl chloride and cyclization of the resulting amide gave 1H-imidazol[4,5-h]isoquinoline-7,9(6H,8H)-dione 1 (iii). Alkylation of 1(iii) with 1-bromo-3-chloropropane gave 1(hv), which reacted with a primary amine to give 1(v). If necessary, the transformation of R1a and R3a to R1 and R3 by methods known to the skilled in the art (including, but not limited to, removal of protective group, alkylation, oxidation and reduction) gave the compounds of formula (I) or (Ia).
    Figure US20070225317A1-20070927-C00012
  • Alternatively, the side-chain on the isoquinoline-1,3(2H,4H)-dione 1(i) can be incorporated using a Mitsunobu reaction with alcohol 2(i) to give 2(ii). Reduction of the nitro group on 2(ii) gave the diamino intermediate 2(iii) that was transformed to 1H-imidazol[4,5-h]isoquinoline-7,9(6H,8H)-dione 2(iv) using an aldehyde in presence of p-chloranil. Alternative methods to generate 2(iv) from diamino 2(iii) are known to the skilled in the art. By inverting the sequence of steps, 1 (ii) can be easily converted to 1H-imidazol[4,5-h]isoquinoline-7,9(6H,8H)-dione 2(v) and finally to 2(iv). Transformation of R1a and R3a to R1 and R3 in 2(iv) by methods known to the skilled in the art (including but not limited to removal of protective group, alkylation, oxidation and reduction), if necessary, and removal of the protective group P on the amine gave compounds of formula (I) or (Ia).
    Figure US20070225317A1-20070927-C00013
  • The bromination of 1(i) gave the bromo derivative 3(i), which was transformed to 3(iv) using sequences analogous to the two described in Scheme 2. 4-Bromo intermediate 3(iv) can be transformed to 3(vii) by art-recognized chemistry. Transformation of R1a, R2a (if not Br) and R3a to R1, R2 and R3 by methods known to the skilled in the art (including but not limited to removal of protective group, alkylation, oxidation and reduction) in 3(iv) or 3(vii), if necessary, followed by removal of the protective group P on the amine gave compounds of formula (I) or (Ia).
    Figure US20070225317A1-20070927-C00014
  • Briefly, aromatic substitution of 4(i) with a protected form of ammonia produced 4(ii). Removal of the protecting group on the aniline 4(iii) followed by a second aromatic substitution with diethyl malonate gave 4(iii). As described in previous Schemes, 4(iii) can be transformed to benzimidazole 4(iv). Upon saponification of the ester groups and decarboxylation in acidic media, diacid 4(v) was obtained. Treatment of diacid 4(v) with amine 4(vi) at high temperature gave, after acidification and protection of the amine, 4(vii). Finally, alkylation of 4(vii) followed by transformation of R1a and R3a to R1 and R3, if necessary, and removal of the amine protective group gave the compounds of formula (I) or (Ia).
  • EXAMPLES
  • The present invention is illustrated in further detail by the following non-limiting examples. All reactions were performed in a nitrogen or argon atmosphere unless otherwise stated. Room temperature is 18-22° C. (degrees Celsius). Solution percentages or ratios express a volume to volume relationship, unless stated otherwise.
  • Abbreviations or symbols used herein include:
  • BOC: tert-butyloxycarbonyl; dba: dibenzylideneacetone; DIAD: diisopropyl azodicarboxylate; DMF: N,N-dimethylformamide; DMSO: dimethylsulfoxide; dppf: 1,1′-bis(diphenylphosphino)ferrocene; DTT: dithiothreitol; EDTA: ethylenediaminetetraacetate; EGTA: 1,2-di(2-aminoethoxy)ethane-N,N,N′,N′-tetraacetic acid; Et2O: diethyl ether; EtOAc: ethyl acetate; HPLC: high performance liquid chromatography; iPr: isopropyl; Me: methyl; MeOH: methanol; MeCN: acetonitrile; Ph: phenyl; SDS: sodium dodecyl sulfate; TFA: trifluoroacetic acid; THF: tetrahydrofuran; and TrisAcetate: 2-amino-2-hydroxymethyl-1,3-propanediol acetate.
  • Syntheses
  • The following examples illustrate methods for preparing compounds of the invention.
  • Example 1 Entry 1132 8-(3-{[(3,4-dimethoxyphenyl)ethyl]amino}propyl)-6,6-dimethyl-2-(3-nitrophenyl)-1H-imidazo[4,5-h]isoquinoline-7,9(6H,8M-dione a) tert-Butyl[3-hydroxypropyl][2-(3,4-dimethoxyphenyl)ethyl]carbamate
  • Figure US20070225317A1-20070927-C00015
  • 2-(3,4-Dimethoxyphenyl)ethylamine (10.0 mL, 59.3 mmol) was added to an ice-cold solution of methyl acrylate (5.40 mL, 59.3 mmol) in MeOH (60 mL). The reaction mixture was stirred at room temperature for 4 h then was concentrated under reduced pressure. Et3N (10.0 mL, 71.7 mmol) and (BOC)2O (14.3 g, 65.5 mmol) were added to a solution of the residue in THF (60 mL). The reaction mixture was stirred at room temperature for 1 h then was concentrated under reduced pressure. The residue was taken in Et2O and the resulting solution was successively washed with aqueous 10% citric acid solution (2×), water, aqueous saturated NaHCO3 solution, water (2×) and brine, dried (MgSO4), filtered and concentrated under reduced pressure. LiAlH4 (2.25 g, 59.3 mmol) was added to an ice-cold solution of the residue in Et2O (300 mL). The reaction mixture was stirred al 0° C. for 3 h, 1 h at room temperature and then cooled to 0° C. Water (2.8 mL), 10% aqueous NaOH solution (2.8 mL) and water (8.5 mL) were successively and carefully added to the mixture. The resulting suspension was filtered and the filtrate was concentrated under reduced pressure. The residue was purified by flash chromatography (40-63μ silica gel; EtOAc:hexane, 3:2) to give the title compound (15.9 g, 79% yield).
  • b) tert-Butyl[3-(7-amino-4,4-dimethyl-8-nitro-1,3-dioxo-3,4-dihydro-1H-isoquinolin-2-yl)propyl]-[2-(3,4-dimethoxyphenyl)ethyl]carbamate
  • Figure US20070225317A1-20070927-C00016
  • DIAD (1.92 mL, 9.75 mmol) was slowly added to an ice-cold solution of 7-amino-4,4-dimethyl-8-nitroisoquinoline-1,3(2H,4H)-dione (2.03 g, 8.14 mmol; prepared according to the method described in DE 3410168), tert-butyl[3-hydroxypropyl][2-(3,4-dimethoxyphenyl)ethyl]carbamate (3.31 g, 9.78 mmol), and PPh3 (2.56 g, 9.76 mmol) in THF (40 mL). The reaction mixture was allowed to warm to room temperature and was stirred at this temperature for 16 h. The mixture was concentrated under reduced pressure and the residue was purified by flash chromatography (40-63μ silica gel, hexane:EtOAc 1:1) to give the title compound (4.29 g, 92% yield).
  • c) tert-Butyl[3-(7,8-diamino-4,4-dimethyl-1,3-dioxo-3,4-dihydro-1H-isoquinolin-2-yl)propyl]-[2-(3,4-dimethoxyphenyl)ethyl]carbamate
  • Figure US20070225317A1-20070927-C00017
  • A mixture of tert-butyl[3-(7-amino-4,4-dimethyl-8-nitro-1,3-dioxo-3,4-dihydro-1H-Isoquinolin-2-yl)propyl]-[2-(3,4-dimethoxyphenyl)ethyl]carbamate (3.60 g, 6.31 mmol) and 20% Pd(OH)2/C (0.72 g) in EtOH (100 mL) and THF (10 mL) was stirred under H2 (balloon) for 16 h. The catalyst was removed by filtration through diatomaceous earth and the filtrate was concentrated under reduced pressure. The residue was purified by flash chromatography (40-63μ silica gel, hexane:EtOAc 1:1 to 1:2) to give the title compound (2.85 g, 83% yield).
  • d) 8-(3-{[(3,4-dimethoxyphenyl)ethyl]amino}propyl)-6,6-dimethyl-2-(3-nitrophenyl)-1H-imidazo[4,5-h]isoquinoline-7,9(6H,8H)-dione
  • Figure US20070225317A1-20070927-C00018
  • A solution of tert-butyl[3-(7,8-diamino-4,4-dimethyl-1,3-dioxo-3,4-dihydro-1H-isoquinolin-2-yl)propyl]-[2-(3,4-dimethoxyphenyl)ethyl]carbamate (25.0 mg, 46.2 μmol), 3-nitrobenzaldehyde (14.0 mg, 92.4 μmol) and p-chloranil (12.2 ng, 49.6 μmol) in MeCN (1.0 mL) was stirred at room temperature for 16 h. The reaction mixture was concentrated under reduced pressure. The residue was dissolved in CH2Cl2 (1.0 mL) and TFA (1.0 mL) was added. The mixture was stirred at room temperature for 1 h then was concentrated under reduced pressure. CHCl3 (1 mL) and 1N aqueous NaOH solution (1 mL) were added to the residue. The aqueous layer was absorbed on a short column containing Extrelute® (˜1 g). The organic phase was added after 10 min to the column. The column washed with CHCl3 and the organic solution was concentrated under reduced pressure. The residue was purified by reverse phase HPLC (CombiPrep ODS-AQ 50×20 mm, 5μ, 120μ, MOCN/water+0.1% TfA). The pure fractions were combined and lyophilized to give the TFA salt of compound 1132 (18.3 mg, 64% yield).
  • Example 2 Entry 1059 4-Amino-8-(3-{[(3,4-dimethoxyphenyl)ethyl]amino}propyl)-6,6-dimethyl-2-(4-methyl-3-nitrophenyl)-1H-imidazo[4,5-h]isoquinoline-7,9(6H,8H)-dione a) 7-Amino-6-bromo-4,4-dimethyl-8-nitroisoquinoline-1,3(2H,4M-dione
  • Figure US20070225317A1-20070927-C00019
  • Bromine (9.27 mL, 188.6 mmol) was added dropwise to a solution of 7-amino-4,4-dimethyl-8-nitroisoquinoline-1,3(2H,4H)-dione (4.70 g, 18.86 mmol) in AcOH (190 mL). The reaction mixture was stirred at 25° C. for 2 h then was concentrated under reduced pressure. The residue was diluted with EtOAc, and successively washed with water, aqueous saturated NaHCO3, aqueous saturated Na2S2O3, and brine, dried (MgSO4), filtered and concentrated under reduced pressure to give the title compound (6.10 g, 99% yield).
  • b) 7,8-Diamino-6-bromo-4,4-dimethylisoquinoline-1,3(2H,4H)-dione
  • Figure US20070225317A1-20070927-C00020
  • A mixture of 7-amino-6-bromo-4,4-dimethyl-8-nitroisoquinoline-1,3(2H,4H)-dione (2 g, 6.09 mmol) and iron powder (325 mesh, 2.38 g, 42.67 mmol) in AcOH (40 mL) was heated to 70° C. for 8 h. The excess iron was removed using a magnetic bar and the reaction mixture was concentrated under reduced pressure. The residue was diluted with EtOAc, and successively washed with water, aqueous saturated NaHCO3, and brine, dried (MgSO4), filtered and concentrated under reduced pressure to give the title compound (1.92 g, 100% yield).
  • c) 4-Bromo-6,6-dimethyl-2-(4-methyl-3-nitrophenyl)-1H-imidazo[4,5-h]isoquinoline-7,9(6H,8H)-dione
  • Figure US20070225317A1-20070927-C00021
  • To a solution of 7,8-diamino-6-bromo-4,4-dimethylisoquinoline-1,3(2H,4H)-dione (1.40 g, 4.70 mmol) in MeCN (47 mL) and DMF (3 mL) was added 4-methyl-3-nitro-benzaldehyde (814.2 mg, 4.93 mmol) and p-chloranil (1.21 g, 4.93 mmol). The reaction mixture was stirred at 60° C. for 8 h. The cooled mixture was diluted with MeCN (50 mL) and the suspension was filtered. The solid was dried to give the title compound (1.65 g, 79% yield),
  • d) tert-Butyl{3-[4-bromo-6,6-dimethyl-2-(4-methyl-3-nitrophenyl)-7,9-dioxo-1,6,7,9-tetrahydro-8H-imidazo[4,5-h]isoquinolin-8-yl]propyl}[2-(3,4-dimethoxyphenyl)ethyl]carbamate
  • Figure US20070225317A1-20070927-C00022
  • DIAD (1.15 mL, 5.87 mmol) was added dropwise to a solution of 4-bromo-6,6-dimethyl-2-(4-methyl-3-nitrophenyl)-1H-imidazo[4,5-h]isoquinoline-7,9(6H,8H)-dione (1.3 g, 2.93 mmol), tert-butyl[2-(3,4-dimethoxyphenyl)ethyl](3-hydroxypropyl)carbamate (from Example 1) (1.99 g, 5.87 mmol), and PPh3 (1.54 g, 5.87 mmol) in THF (14.7 mL). The reaction mixture was stirred at 25° C. for 30 min, then was concentrated under reduced pressure. The residue was purified twice by flash chromatography (40-63μ silica gel) first with hexane:EtOAc 1:1 and then with CH2Cl2:acetone 95:5 to give the title compound (789.6 mg, 35% yield).
  • e) tert-Butyl[2-(3,4-dimethoxyphenyl)ethyl]{3-[4-[(methoxybenzyl)amino]-6,6-dimethyl-2-(4-methyl-3-nitrophenyl)-7,9-dioxo-1,6,7,9-tetrahydro-8H-imidazo[4,5-h]isoquinalin-8-yl]propyl}carbamate
  • Figure US20070225317A1-20070927-C00023
  • Pd2(dba)3 (5.6 mg, 5.12 μmol) and dppf (2.83 mg, 5.09 μmol) were added to a degassed (N2 for 30 min) mixture of tert-butyl {3-[4-bromo-6,6-dimethyl-2-(4-methyl-3-nitrophenyl)-7,9-dioxo-1,6,7,9-tetrahydro-8H-imidazo[4,5-h]isoquinolin-8-yl]propyl}[2-(3,4-dimethoxyphenyl)ethyl]carbamate (780 mg, 1.02 mmol), 4-methoxybenzylamine (533 μL, 4.08 mmol) and NaOt-Bu (196 mg, 2.04 mmol) in THF (10.2 mL). The reaction mixture was stirred at 60° C. for 3 h, then was concentrated under reduced pressure. The residue was purified by flash chromatography (40-63μ silica gel; CH2Cl2:acetone 95:5) to give the title compound (542 mg, 65% yield).
  • f)-4-Amino-8-(3-{[(3,4-dimethoxyphenyl)ethyl]amino}propyl)-6,6-dimethyl-2-(4-methyl-4-nitrophenyl)-H-imidazo[4,5-h]isoquinoline-7,9(6H,8H)-dione
  • Figure US20070225317A1-20070927-C00024
  • A solution of tert-butyl[2-(3,4-dimethoxyphenyl)ethyl]{3-[4-[(methoxybenzyl)amino]-6,6-dimethyl-2-(4-methyl-3-nitrophenyl)-7,9-dioxo-1,6,7,9-tetrahydro-8H-imidazo[4,5-h]isoquinolin-8-yl]propyl}carbamate (87.9 mg, 107.1 μmol) and TFA (1 mL) in CH2Cl2 (3 mL) was stirred at 25° C. for 45 min. The reaction mixture was concentrated under reduced pressure. The residue was purified by reverse phase HPLC (CombiPrep ODS-AQ 50×20 mm, 5μ, 120 A, MeCN/water+0.1% TFA) to give compound 1059 (36.4 mg, 56% yield).
  • Example 3 Entry 2007 8′-(3-{[2-(3,4-Dimethoxyphenyl)ethyl]amino}propyl)-2′-(2-methoxyphenyl)spiro[cyclohexane-1,6′-imidazo[4,5-h]isoquinoline]-7′,9′(1′H,8′H)-dione a) Methyl 6-chloro-2-[(4-methoxybenzyl)amino]-3-nitrobenzoate
  • Figure US20070225317A1-20070927-C00025
  • A solution of methyl 2,6-dichloro-3-nitrobenzoate (11.9 g, 47.4 mmol), p-methoxybenzylamine (8.13 g, 59.3 mmol; additional amounts of 1.63 g, 11.9 mmol added after 5 and 6.5 h), and Et3N (7.20 g, 71.1 mmol; additional amount of 1.20 g, 11.9 mmol added after 6.5 h) in THF (190 mL) was stirred at room temperature for 15 min then was heated to reflux for 10 h. Water (100 mL) was added to the mixture and most of the THF was removed under reduced pressure. The residue was partitioned between aqueous 1 N HCl solution (350 mL) and EtOAc (500 mL). The organic layer washed with aqueous 1 N HCl solution (100 mL) and brine (100 mL), then was dried (MgSO4), filtered and concentrated under reduced pressure. The residue was purified by flash chromatography (10-40μ silica gel; hexane:EtOAc 6:1) to give the title compound (13.2 g, 79% yield) as a yellow solid.
  • b) Methyl 2-amino-6-chloro-3-nitrobenzoate
  • Figure US20070225317A1-20070927-C00026
  • A solution of methyl 6-chloro-2-[(4-methoxybenzyl)amino]-3-nitrobenzoate (13.2 g, 37.6 mmol) and TFA (75 mL) was stirred at room temperature for 1.7 h. The mixture was diluted with EtOAc (500 mL) and water (250 mL). Solid NaHCO3 (125 g) was slowly added to the heterogeneous mixture. Saturated aqueous NaHCO3 solution (200 mL) and EtOAc were added and the phases were separated. The organic layer washed with saturated aqueous NaHCO3 solution (150 mL) and brine (50 mL) then dried (MgSO4), filtered and concentrated under reduced pressure. The residue was purified by flash chromatography (10-40μ silica gel; hexane:EtOAc 5:1) to yield the title compound (7.03 g, 81% yield) as a yellow solid.
  • c) Diethyl[3-amino-2-(methoxycarbonyl)-4-nitrophenyl]malonate
  • Figure US20070225317A1-20070927-C00027
  • A mixture of methyl 2-amino-6-chloro-3-nitrobenzoate (4.11 g, 17.8 mmol), diethyl malonate (4.85 g, 30.3 mmol) and solid K2CO3 (powder, 9.11 g, 65.9 mmol) in DMF (71 mL) was heated at 60° C. for 10 h. The reaction mixture was poured into aqueous 1 N HCl solution (400 mL) and the resulting solution was extracted with EtOAc (2×250 mL). The combined organic layers were washed with water (3×150 mL) and brine (100 ml), dried (MgSO4), filtered and concentrated under reduced pressure. Purification by flash chromatography (10-40μ silica gel; hexane:EtOAc 3:1) provided the title compound (4.80 g, 76% yield) as a yellow solid.
  • d) Diethyl[3,4-d]amino-2-(methoxycarbonyl)phenyl)malonate
  • Figure US20070225317A1-20070927-C00028
  • A mixture of diethyl[3-amino-2-(methoxycarbonyl)-4-nitrophenyl]malonate (4.80 g, 13.6 mmol), 10% Pd/C (0.72 g) in MeOH (70 mL) and EtOAc (70 mL) was stirred under H2 atmosphere (balloon) at room temperature for 21 h. The catalyst was removed by filtration through diatomaceous earth. The cake washed with EtOAc and the filtrate was concentrated to give the title compound (4.39 g, 100% yield) as a brown solid.
  • e) Diethyl[7-(methoxycarbonyl)-2-(2-methoxyphenyl)-1H-benzimidazol-6-yl]malonate
  • Figure US20070225317A1-20070927-C00029
  • A solution of diethyl[3,4-diamino-2-(methoxycarbonyl)phenyl]malonate (4.39 g, 13.5 mmol), 2-methoxybenzaldehyde (1.97 g, 14.5 mmol) and p-chloranil (6.10 g, 14.5 mmol) in MeCN (135 mL) was stirred at room temperature for 22 h. The resulting suspension was filtered and the solid recovered washed with MeCN (filtrate 1 kept). The solid was dissolved in EtOAc (800 mL) and the resulting solution was successively washed with aqueous 1N NaOH solution (3×150 mL), water (150 mL) and brine (100 mL), dried (MgSO4), filtered and concentrated under reduced pressure. The resulting solid was triturated with hexane:EtOAc (1:1; 135 mL) to give the title compound (4.22 g) as a white solid (filtrate 2 kept). The filtrates 1 and 2 were combined and concentrated under reduced pressure. The residue was treated as the initial solid but the residue after washings was concentrated and purified by flash chromatography (10-40μ silica gel; hexane:EtOAc 1:1) before being triturated with hexane:EtOAc (3:2; 35 mL) to give an additional 0.72 g amount (total of 4.96 g, 83% yield) of the title compound.
  • f) 6-(Carboxymethyl)-2-(2-methoxyphenyl)-1H-benzimidazole-7-carboxylic acid
  • Figure US20070225317A1-20070927-C00030
  • A solution of diethyl[7-(methoxycarbonyl)-2-(2-methoxyphenyl)-1H-benzimidazol-6-yl]malonate (4.96 g, 11.3 mmol) and aqueous 2 N NaOH solution (50.0 mL, 100 mmol) in THF (112 mL) and MeOH (45 mL) was stirred at room temperature for 17 h. The mixture was concentrated under reduced pressure. The residual aqueous solution was cooled to 0° C. and aqueous 2 N HCl solution (50 mL) was slowly added (pH 4 obtained). The resulting suspension was heated at 70° C. for 15 h. The cooled suspension was filtered to give the title compound (3.72 g, 100% yield) as a white solid.
  • g) tert-Butyl[2-(3,4-dimethoxyphenyl)ethyl]{3-[2-(2-methoxyphenyl)-7,9-dioxo-1,6,7,9-tetrahydro-8H-imidazo[4,5-h]isoquinolin-8-yl]propyl}carbamate
  • Figure US20070225317A1-20070927-C00031
  • A solution of 6-(carboxymethyl)-2-(2-methoxyphenyl)-1H-benzimidazole-7-carboxylic acid (800 mg, 2.45 mmol) and tert-butyl (3-aminopropyl)[2-(3,4-dimethoxyphenyl)ethyl]carbamate (1.00 g, 2.94 mmol) in ethyleneglycol (6.1 mL) was heated at 180° C. for 1 h. The cooled mixture was dissolved in EtOAc (500 mL) and the resulting solution washed with water (2×100 mL) and brine (200 mL), dried (MgSO4) and concentrated under reduced pressure. A solution of the resulting yellow solid (1.04 g) and aqueous 1 N HCl solution (33 mL) in MeOH (100 mL) was heated at 70° C. for 16 h. The mixture was concentrated under reduced pressure. (BOC)2O (1.34 g, 6.12 mmol) and Et3N (2.06 g, 20.4 mmol) were successively added to a solution of the residue in CH2Cl2 (30 mL). The mixture was stirred at room temperature for 15 min then was diluted with CH2Cl2 (300 mL). The resulting solution washed with water (2×100 mL) and brine (100 mL), dried (MgSO4), filtered and concentrated under reduced pressure. The residue was purified by flash chromatography (10-40μ silica gel; hexane:EtOAc 1:2 to 1:3) to give the title compound (839 mg, 54% yield) as a yellow solid.
  • h) tert-Butyl[2-(3,4-dimethoxyphenyl)ethyl]{3-[2′-(2-methoxyphenyl)-7′,9′-dioxo-1′,9′-dihydrospiro[cyclohexane-1,6′-imidazo[4,5-h]isoquinolin]-8′(7′-yl]propyl}carbamate
  • Figure US20070225317A1-20070927-C00032
  • A solution of tert-butyl[2-(3,4-dimethoxyphenyl)ethyl]{3-[2-(2-methoxyphenyl)-7,9-dioxo-1,6,7,9-tetrahydro-8H-imidazo[4,5-h]isoquinolin-8-yl]propyl}carbamate (100.0 mg, 0.159 mmol), 1,3-dibromopropane (43.9 mg, 0.19 mmol) and aqueous 1 N NaOH solution (0.32 mL, 0.32 mmol) in water (1.0 mL) and EtOH (1 mL) was heated at 80° C. for 2 h. The cooled mixture was partitioned between EtOAc and water. The organic layer washed with brine, dried (MgSO4), filtered and concentrated under reduced pressure. The residue was purified by flash chromatography (40-63μ silica gel; hexane:EtOAc 1:1) to yield the title compound (47 mg, 43% yield) as a white foam.
  • i) 8′-(3-{[2-(3,4-Dimethoxyphenyl)ethyl]amino}propyl)-2′-(2-methoxyphenyl)spiro[cyclohexane-1,6′-imidazo[4,5-h]isoquinoline]-7′,9′(1′H,8′H)-dione
  • Figure US20070225317A1-20070927-C00033
  • A solution of tert-butyl[2-(3,4-dimethoxyphenyl)ethyl]{3-[2′-(2-methoxyphenyl)-7′,9′-dioxo-1,9′-dihydrospiro[cyclohexane-1,6′-imidazo[4,5-h]isoquinolin]-8′(7′H)-yl]propyl}carbamate (47.0 mg, 0.07 mmol) in 4 N HCl solution in 1,4-dioxane (3 mL) was stirred at room temperature for 1 h. The mixture was concentrated under reduced pressure and the residue was purified by reverse phase HPLC (CombiPrep ODS-AQ 50×20 mm, 5μ, 120 A, MeCN/water+0.10% TFA). The combined pure fractions were frozen and lyophilized to give the bis-trifluoroacetic acid salt of compound 2007 (51 mg, 92% yield).
  • Example 4
  • RSV Polymerase Assay
  • The procedure used was described by Mason, S. W. et al, Nucleic Acids Research (2004) 32: 4758-4767. Transcription reactions contained 0.1-1 μL RSV polymerase fraction, isolated from RSV infected HEp-2 cells using a method described previously for Newcastle Disease Virus (Hamaguchi, M., T. Yoshida, K. Nichikawa, H. Naruse & Y Nagai (1983) Virology 128, 105-117), in reaction buffer (50 mM TrisAcetate (pH 7.5), 120 mM potassium acetate, 4.5 mM MgCl2, 5% glycerol, 2 mM EGTA, 3 mM DTT, 50 μg/ml BSA, 04 mM of each ATP, GTP and UTP, 4% DMSO). Reactions also contained 1 μCi 3H-CTP (˜20 Cl/mmol). Reactions were assembled in 96-well FlashPlate™ (PerkinElmer) to which 0.6 μmol oligo-dT30 was previously immobilized in 10 mM Tris HCl (pH 7.5) 1 mM EDTA. 0.5 M LiCl, 0.5% SDS, 70 mM NaCl, 2.5 mM KCl. Following 2 h incubation at 30° C., the reaction was stopped with hybridization buffer (17 mM Tris HCl (pH 7.5), 1.7 mM EDTA, 0.85 M LiCl, 0.85% SDS, 120 mM NaCl, 3.75 mM KCl) which favors hybridization of RNA with DNA oligonucleotides. The radionucleotide that was incorporated into newly synthesized RNA was detected by proximity to scintillant embedded in the wells of the plate using a TopCount™ (Beckman) multi-well plate scintillation detector. The compounds described in Tables 1 and 2 exhibited IC50≦5 μM in this assay.
  • Example 5
  • RSV ELISA
  • The procedure used was described by Mason, S. W. et al., Nucleic Acids Research (2004) 32: 4758-4767. HEp-2 cells were plated at 10,000 cells per 96 well in DMEM 10% FBS for 24 h. The monolayer was infected with RSV-Long at an MOI of 0.1 pfu per cell in 50 μL DMEM 2% FBS. The virus was removed after one hour adsorption and 100 μL media, with or without compound, was added. Incubation was continued for 48 h. The monolayer was fixed with 0.063% glutaraldehyde. The plates were blocked with 1% BSA in phosphate-buffered saline (PBS) for one hour. Viral replication was detected with a monoclonal antibody directed against RSV F diluted 1/2000 in PBS1% BSA. After 1 h incubation, the plates were washed and Sheep anti mouse HRP diluted 1/6000 was added. One hour later, the plates were washed and were developed with o-phenylenediamine dihydrochloride (OPD) for thirty min. The absorbance was read at 450 nm. Percent inhibition was calculated using SAS program for non-linear regression analysis.
  • Tables of Compounds
  • The following tables 1 and 2 list compounds representative of the invention. All compounds described in Tables 1 and 2 exhibited IC50≦5 μM in the RSV polymerase assay of Example 4. In addition, many of the compounds described in Tables 1 and 2 are active in the RSV ELISA assay of Example 5.
  • The following abbreviation is used within the present tables: MS: Mass spectrometric data.
    TABLE 1
    Figure US20070225317A1-20070927-C00034
    Cpd MS
    entry # R1 R2 R3 (MH)+
    1001
    Figure US20070225317A1-20070927-C00035
    H
    Figure US20070225317A1-20070927-C00036
    557
    1002
    Figure US20070225317A1-20070927-C00037
    H
    Figure US20070225317A1-20070927-C00038
    552
    1003
    Figure US20070225317A1-20070927-C00039
    H
    Figure US20070225317A1-20070927-C00040
    595
    1004
    Figure US20070225317A1-20070927-C00041
    H
    Figure US20070225317A1-20070927-C00042
    543
    1005
    Figure US20070225317A1-20070927-C00043
    H
    Figure US20070225317A1-20070927-C00044
    595
    1006
    Figure US20070225317A1-20070927-C00045
    H
    Figure US20070225317A1-20070927-C00046
    541
    1007
    Figure US20070225317A1-20070927-C00047
    H
    Figure US20070225317A1-20070927-C00048
    606/608
    1008
    Figure US20070225317A1-20070927-C00049
    H
    Figure US20070225317A1-20070927-C00050
    557
    1009
    Figure US20070225317A1-20070927-C00051
    H
    Figure US20070225317A1-20070927-C00052
    586
    1010
    Figure US20070225317A1-20070927-C00053
    H
    Figure US20070225317A1-20070927-C00054
    579/581
    1011
    Figure US20070225317A1-20070927-C00055
    H
    Figure US20070225317A1-20070927-C00056
    581/563
    1012
    Figure US20070225317A1-20070927-C00057
    H
    Figure US20070225317A1-20070927-C00058
    579/581
    1013
    Figure US20070225317A1-20070927-C00059
    H
    Figure US20070225317A1-20070927-C00060
    571
    1014
    Figure US20070225317A1-20070927-C00061
    H
    Figure US20070225317A1-20070927-C00062
    572
    1015
    Figure US20070225317A1-20070927-C00063
    H
    Figure US20070225317A1-20070927-C00064
    595/597/599
    1016
    Figure US20070225317A1-20070927-C00065
    H
    Figure US20070225317A1-20070927-C00066
    555
    1017
    Figure US20070225317A1-20070927-C00067
    H
    Figure US20070225317A1-20070927-C00068
    602
    1018
    Figure US20070225317A1-20070927-C00069
    H
    Figure US20070225317A1-20070927-C00070
    575
    1019
    Figure US20070225317A1-20070927-C00071
    H
    Figure US20070225317A1-20070927-C00072
    615
    1020
    Figure US20070225317A1-20070927-C00073
    H
    Figure US20070225317A1-20070927-C00074
    588
    1021
    Figure US20070225317A1-20070927-C00075
    H
    Figure US20070225317A1-20070927-C00076
    618
    1022
    Figure US20070225317A1-20070927-C00077
    H
    Figure US20070225317A1-20070927-C00078
    563
    1023
    Figure US20070225317A1-20070927-C00079
    H
    Figure US20070225317A1-20070927-C00080
    563
    1024
    Figure US20070225317A1-20070927-C00081
    H
    Figure US20070225317A1-20070927-C00082
    581
    1025
    Figure US20070225317A1-20070927-C00083
    H
    Figure US20070225317A1-20070927-C00084
    559
    1026
    Figure US20070225317A1-20070927-C00085
    H
    Figure US20070225317A1-20070927-C00086
    577
    1027
    Figure US20070225317A1-20070927-C00087
    H
    Figure US20070225317A1-20070927-C00088
    567
    1028
    Figure US20070225317A1-20070927-C00089
    H
    Figure US20070225317A1-20070927-C00090
    542
    1029
    Figure US20070225317A1-20070927-C00091
    H
    Figure US20070225317A1-20070927-C00092
    578
    1030
    Figure US20070225317A1-20070927-C00093
    H
    Figure US20070225317A1-20070927-C00094
    594
    1031
    Figure US20070225317A1-20070927-C00095
    H
    Figure US20070225317A1-20070927-C00096
    600/602
    1032
    Figure US20070225317A1-20070927-C00097
    H
    Figure US20070225317A1-20070927-C00098
    562
    1033
    Figure US20070225317A1-20070927-C00099
    H
    Figure US20070225317A1-20070927-C00100
    566
    1034
    Figure US20070225317A1-20070927-C00101
    H
    Figure US20070225317A1-20070927-C00102
    578
    1035
    Figure US20070225317A1-20070927-C00103
    H
    Figure US20070225317A1-20070927-C00104
    579
    1036
    Figure US20070225317A1-20070927-C00105
    H
    Figure US20070225317A1-20070927-C00106
    578
    1037
    Figure US20070225317A1-20070927-C00107
    H
    Figure US20070225317A1-20070927-C00108
    578
    1038
    Figure US20070225317A1-20070927-C00109
    H
    Figure US20070225317A1-20070927-C00110
    561
    1039
    Figure US20070225317A1-20070927-C00111
    H
    Figure US20070225317A1-20070927-C00112
    578
    1040
    Figure US20070225317A1-20070927-C00113
    H
    Figure US20070225317A1-20070927-C00114
    567
    1041
    Figure US20070225317A1-20070927-C00115
    H
    Figure US20070225317A1-20070927-C00116
    598
    1042
    Figure US20070225317A1-20070927-C00117
    H
    Figure US20070225317A1-20070927-C00118
    543
    1043
    Figure US20070225317A1-20070927-C00119
    H
    Figure US20070225317A1-20070927-C00120
    513
    1044
    Figure US20070225317A1-20070927-C00121
    H
    Figure US20070225317A1-20070927-C00122
    572
    1045
    Figure US20070225317A1-20070927-C00123
    H
    Figure US20070225317A1-20070927-C00124
    558
    1046
    Figure US20070225317A1-20070927-C00125
    Br
    Figure US20070225317A1-20070927-C00126
    635/637
    1047
    Figure US20070225317A1-20070927-C00127
    H
    Figure US20070225317A1-20070927-C00128
    585
    1048
    Figure US20070225317A1-20070927-C00129
    NH3
    Figure US20070225317A1-20070927-C00130
    572
    1049
    Figure US20070225317A1-20070927-C00131
    C≡CH
    Figure US20070225317A1-20070927-C00132
    561
    1050
    Figure US20070225317A1-20070927-C00133
    NHMe
    Figure US20070225317A1-20070927-C00134
    586
    1051
    Figure US20070225317A1-20070927-C00135
    Me
    Figure US20070225317A1-20070927-C00136
    571
    1052
    Figure US20070225317A1-20070927-C00137
    Br
    Figure US20070225317A1-20070927-C00138
    607
    1053
    Figure US20070225317A1-20070927-C00139
    Br
    Figure US20070225317A1-20070927-C00140
    644/666
    1054
    Figure US20070225317A1-20070927-C00141
    Cl
    Figure US20070225317A1-20070927-C00142
    620/622
    1055
    Figure US20070225317A1-20070927-C00143
    OH
    Figure US20070225317A1-20070927-C00144
    602
    1056
    Figure US20070225317A1-20070927-C00145
    Br
    Figure US20070225317A1-20070927-C00146
    621/623
    1057
    Figure US20070225317A1-20070927-C00147
    Br
    Figure US20070225317A1-20070927-C00148
    650/652
    1058
    Figure US20070225317A1-20070927-C00149
    NH3
    Figure US20070225317A1-20070927-C00150
    587
    1059
    Figure US20070225317A1-20070927-C00151
    NH3
    Figure US20070225317A1-20070927-C00152
    601
    1060
    Figure US20070225317A1-20070927-C00153
    Br
    Figure US20070225317A1-20070927-C00154
    656/658
    1061
    Figure US20070225317A1-20070927-C00155
    Br
    Figure US20070225317A1-20070927-C00156
    645/647
    1062
    Figure US20070225317A1-20070927-C00157
    Br
    Figure US20070225317A1-20070927-C00158
    620/622
    1063
    Figure US20070225317A1-20070927-C00159
    Br
    Figure US20070225317A1-20070927-C00160
    630/632
    1064
    Figure US20070225317A1-20070927-C00161
    Br
    Figure US20070225317A1-20070927-C00162
    619/621
    1065
    Figure US20070225317A1-20070927-C00163
    Br
    Figure US20070225317A1-20070927-C00164
    673/675
    1066
    Figure US20070225317A1-20070927-C00165
    Br
    Figure US20070225317A1-20070927-C00166
    623/625
    1067
    Figure US20070225317A1-20070927-C00167
    Br
    Figure US20070225317A1-20070927-C00168
    633/635
    1068
    Figure US20070225317A1-20070927-C00169
    Br
    Figure US20070225317A1-20070927-C00170
    609/611
    1069
    Figure US20070225317A1-20070927-C00171
    Br
    Figure US20070225317A1-20070927-C00172
    636/638
    1070
    Figure US20070225317A1-20070927-C00173
    Br
    Figure US20070225317A1-20070927-C00174
    644/646
    1071
    Figure US20070225317A1-20070927-C00175
    Br
    Figure US20070225317A1-20070927-C00176
    624/626
    1072
    Figure US20070225317A1-20070927-C00177
    Br
    Figure US20070225317A1-20070927-C00178
    672/674
    1073
    Figure US20070225317A1-20070927-C00179
    Br
    Figure US20070225317A1-20070927-C00180
    652/654
    1074
    Figure US20070225317A1-20070927-C00181
    Br
    Figure US20070225317A1-20070927-C00182
    651/653
    1075
    Figure US20070225317A1-20070927-C00183
    NH3
    Figure US20070225317A1-20070927-C00184
    593
    1076
    Figure US20070225317A1-20070927-C00185
    NH3
    Figure US20070225317A1-20070927-C00186
    582
    1077
    Figure US20070225317A1-20070927-C00187
    NH3
    Figure US20070225317A1-20070927-C00188
    557
    1078
    Figure US20070225317A1-20070927-C00189
    NH3
    Figure US20070225317A1-20070927-C00190
    567
    1079
    Figure US20070225317A1-20070927-C00191
    NH3
    Figure US20070225317A1-20070927-C00192
    558
    1080
    Figure US20070225317A1-20070927-C00193
    NH3
    Figure US20070225317A1-20070927-C00194
    556
    1081
    Figure US20070225317A1-20070927-C00195
    NH3
    Figure US20070225317A1-20070927-C00196
    610
    1082
    Figure US20070225317A1-20070927-C00197
    NH3
    Figure US20070225317A1-20070927-C00198
    560
    1083
    Figure US20070225317A1-20070927-C00199
    NH3
    Figure US20070225317A1-20070927-C00200
    570
    1084
    Figure US20070225317A1-20070927-C00201
    NH3
    Figure US20070225317A1-20070927-C00202
    585
    1085
    Figure US20070225317A1-20070927-C00203
    NH3
    Figure US20070225317A1-20070927-C00204
    573
    1086
    Figure US20070225317A1-20070927-C00205
    NH3
    Figure US20070225317A1-20070927-C00206
    561
    1087
    Figure US20070225317A1-20070927-C00207
    NH3
    Figure US20070225317A1-20070927-C00208
    581
    1088
    Figure US20070225317A1-20070927-C00209
    NH3
    Figure US20070225317A1-20070927-C00210
    609
    1089
    Figure US20070225317A1-20070927-C00211
    NH3
    Figure US20070225317A1-20070927-C00212
    589
    1090
    Figure US20070225317A1-20070927-C00213
    Br
    Figure US20070225317A1-20070927-C00214
    648/650
    1091
    Figure US20070225317A1-20070927-C00215
    Br
    Figure US20070225317A1-20070927-C00216
    648/650
    1092
    Figure US20070225317A1-20070927-C00217
    Br
    Figure US20070225317A1-20070927-C00218
    682/684/686
    1093
    Figure US20070225317A1-20070927-C00219
    Br
    Figure US20070225317A1-20070927-C00220
    650/652
    1094
    Figure US20070225317A1-20070927-C00221
    Br
    Figure US20070225317A1-20070927-C00222
    650/652
    1095
    Figure US20070225317A1-20070927-C00223
    Br
    Figure US20070225317A1-20070927-C00224
    664/666
    1096
    Figure US20070225317A1-20070927-C00225
    Br
    Figure US20070225317A1-20070927-C00226
    664/666
    1097
    Figure US20070225317A1-20070927-C00227
    Br
    Figure US20070225317A1-20070927-C00228
    694/696
    1098
    Figure US20070225317A1-20070927-C00229
    Br
    Figure US20070225317A1-20070927-C00230
    694/696
    1099
    Figure US20070225317A1-20070927-C00231
    Br
    Figure US20070225317A1-20070927-C00232
    664/666
    1100
    Figure US20070225317A1-20070927-C00233
    Br
    Figure US20070225317A1-20070927-C00234
    682/684/686
    1101
    Figure US20070225317A1-20070927-C00235
    Br
    Figure US20070225317A1-20070927-C00236
    704/706
    1102
    Figure US20070225317A1-20070927-C00237
    Br
    Figure US20070225317A1-20070927-C00238
    654/656
    1103
    Figure US20070225317A1-20070927-C00239
    Br
    Figure US20070225317A1-20070927-C00240
    649/651
    1104
    Figure US20070225317A1-20070927-C00241
    Br
    Figure US20070225317A1-20070927-C00242
    649/651
    1105
    Figure US20070225317A1-20070927-C00243
    Br
    Figure US20070225317A1-20070927-C00244
    640/642
    1106
    Figure US20070225317A1-20070927-C00245
    Br
    Figure US20070225317A1-20070927-C00246
    730/732
    1107
    Figure US20070225317A1-20070927-C00247
    Br
    Figure US20070225317A1-20070927-C00248
    632/634
    1108
    Figure US20070225317A1-20070927-C00249
    Br
    Figure US20070225317A1-20070927-C00250
    662/664
    1109
    Figure US20070225317A1-20070927-C00251
    Br
    Figure US20070225317A1-20070927-C00252
    702/704
    1110
    Figure US20070225317A1-20070927-C00253
    Br
    Figure US20070225317A1-20070927-C00254
    640/642
    1111
    Figure US20070225317A1-20070927-C00255
    Br
    Figure US20070225317A1-20070927-C00256
    640/642
    1112
    Figure US20070225317A1-20070927-C00257
    Br
    Figure US20070225317A1-20070927-C00258
    640/642
    1113
    Figure US20070225317A1-20070927-C00259
    Br
    Figure US20070225317A1-20070927-C00260
    632/634
    1114
    Figure US20070225317A1-20070927-C00261
    Br
    Figure US20070225317A1-20070927-C00262
    632/634
    1115
    Figure US20070225317A1-20070927-C00263
    Br
    Figure US20070225317A1-20070927-C00264
    640/642
    1116
    Figure US20070225317A1-20070927-C00265
    Br
    Figure US20070225317A1-20070927-C00266
    656/658/660
    1117
    Figure US20070225317A1-20070927-C00267
    Br
    Figure US20070225317A1-20070927-C00268
    650/652
    1118
    Figure US20070225317A1-20070927-C00269
    Br
    Figure US20070225317A1-20070927-C00270
    636/638
    1119
    Figure US20070225317A1-20070927-C00271
    Br
    Figure US20070225317A1-20070927-C00272
    650/652
    1120
    Figure US20070225317A1-20070927-C00273
    Br
    Figure US20070225317A1-20070927-C00274
    620/622
    1121
    Figure US20070225317A1-20070927-C00275
    Br
    Figure US20070225317A1-20070927-C00276
    746/748
    1122
    Figure US20070225317A1-20070927-C00277
    Br
    Figure US20070225317A1-20070927-C00278
    872/874
    1123
    Figure US20070225317A1-20070927-C00279
    Br
    Figure US20070225317A1-20070927-C00280
    648/650
    1124
    Figure US20070225317A1-20070927-C00281
    Br
    Figure US20070225317A1-20070927-C00282
    662/664
    1124
    Figure US20070225317A1-20070927-C00283
    Br
    Figure US20070225317A1-20070927-C00284
    648/650
    1125
    Figure US20070225317A1-20070927-C00285
    Br
    Figure US20070225317A1-20070927-C00286
    604/606
    1126
    Figure US20070225317A1-20070927-C00287
    Br
    Figure US20070225317A1-20070927-C00288
    607/609
    1127
    Figure US20070225317A1-20070927-C00289
    Br
    Figure US20070225317A1-20070927-C00290
    610/612
    1128
    Figure US20070225317A1-20070927-C00291
    Br
    Figure US20070225317A1-20070927-C00292
    643/645
    1129
    Figure US20070225317A1-20070927-C00293
    Br
    Figure US20070225317A1-20070927-C00294
    660/662
    1130
    Figure US20070225317A1-20070927-C00295
    NH3
    Figure US20070225317A1-20070927-C00296
    571
    1131
    Figure US20070225317A1-20070927-C00297
    NH3
    Figure US20070225317A1-20070927-C00298
    583
    1132
    Figure US20070225317A1-20070927-C00299
    H
    Figure US20070225317A1-20070927-C00300
    572
  • TABLE 2
    Figure US20070225317A1-20070927-C00301
    Cpd entry # R1 R2
    Figure US20070225317A1-20070927-C00302
    R3 MS (MH)+
    2001
    Figure US20070225317A1-20070927-C00303
    H
    Figure US20070225317A1-20070927-C00304
    Figure US20070225317A1-20070927-C00305
    600
    2002
    Figure US20070225317A1-20070927-C00306
    H
    Figure US20070225317A1-20070927-C00307
    Figure US20070225317A1-20070927-C00308
    555
    2003
    Figure US20070225317A1-20070927-C00309
    H
    Figure US20070225317A1-20070927-C00310
    Figure US20070225317A1-20070927-C00311
    583
    2004
    Figure US20070225317A1-20070927-C00312
    H
    Figure US20070225317A1-20070927-C00313
    Figure US20070225317A1-20070927-C00314
    612
    2005
    Figure US20070225317A1-20070927-C00315
    Br
    Figure US20070225317A1-20070927-C00316
    Figure US20070225317A1-20070927-C00317
    690/692
    2006
    Figure US20070225317A1-20070927-C00318
    NH2
    Figure US20070225317A1-20070927-C00319
    Figure US20070225317A1-20070927-C00320
    627
    2007
    Figure US20070225317A1-20070927-C00321
    H
    Figure US20070225317A1-20070927-C00322
    Figure US20070225317A1-20070927-C00323
    597
    2008
    Figure US20070225317A1-20070927-C00324
    Br
    Figure US20070225317A1-20070927-C00325
    Figure US20070225317A1-20070927-C00326
    704/706
    2009
    Figure US20070225317A1-20070927-C00327
    NH2
    Figure US20070225317A1-20070927-C00328
    Figure US20070225317A1-20070927-C00329
    641
    2010
    Figure US20070225317A1-20070927-C00330
    NH2
    Figure US20070225317A1-20070927-C00331
    Figure US20070225317A1-20070927-C00332
    627
    2011
    Figure US20070225317A1-20070927-C00333
    NH2
    Figure US20070225317A1-20070927-C00334
    Figure US20070225317A1-20070927-C00335
    597

Claims (25)

1.-8. (canceled)
9. A pharmaceutical composition comprising a therapeutically effective amount of a compound of formula (Ia) below, or an enantiomer or a salt thereof, in association with at least one pharmaceutically-acceptable carrier:
Figure US20070225317A1-20070927-C00336
wherein R1 is —(CH═CH)0-1—(C6 or C10)aryl or —(CH═CH)0-1-5-, 6-, 9- or 10-membered heteroaryl, said aryl or heteroaryl being optionally substituted with one, two or three substituents, each independently selected from:
(C1-6)alkyl optionally substituted with amino, halo, (C1-6)haloalkyl, hydroxy, (C1-6)alkoxy, (C1-6)alkylthio, nitro, azido, cyano, amino, (C1-6)alkylamino di((C1-6)alkyl)amino, aryl and heteroaryl;
R2 is H, (C1-6)alkyl, hydroxy, halo, (C1-6)haloalkyl, amino, (C1-6)alkylamino di((C1-6)alkyl)amino, or (C2-6)alkynyl:
R3 is (C6, C10 or C14)aryl or 5-, 6-, 9- or 10-membered heteroaryl, each of which being optionally substituted with one, two or three substituents, each independently selected from:
(C1-6)alkyl, halo, (C1-6)haloalkyl, hydroxy, (C1-6)alkoxy, (C6-)alkylthio, nitro amino, (C1-6)alkylamino, di((C1-6)alkyl)amino and COO(C1-6)alkyl- and
R4 and R5 are each independently H or (C1-6)alkyl; or R4 and R5 are linked, together with the carbon atom to which they are attached, to form a (C3-7)cycloalkyl group; with the proviso that R1 is not 2-methoxyphenyl when R2 is H, R3 is 3,4-dimethoxyphenyl, R4 is CH3 and R5 is CH3.
10. The pharmaceutical composition according to claim 9 for use in the treatment or prevention of respiratory syncytial virus infection.
11. The pharmaceutical composition according to claim 10 additionally comprising a therapeutically effective amount of at least one other antiviral agent.
12. The pharmaceutical composition according to claim 11 wherein the antiviral agent is selected from an immunomodulatory agent, an inhibitor of RSV polymerase or an inhibitor of another target in the RSV life cycle.
13. A method of treating or preventing a respiratory syncytial virus infection in a mammal comprising administering to the mammal an anti-respiratory syncytial virally-effective amount of a composition according to claim 9.
14. The method according to claim 13 wherein the composition additionally comprises a therapeutically effective amount of at least one other antiviral agent.
15. The method according to claim 14 wherein the antiviral agent is selected from an immunomodulatory agent, an inhibitor of RSV polymerase or an inhibitor of another target in the RSV life cycle.
16. A method of inhibiting replication of a respiratory syncytial virus comprising exposing virus-infected cells to an anti-respiratory syncytial virally-effective amount of a composition according to claim 9.
17. A packaged pharmaceutical comprising a pharmaceutical composition according to claim 9 and directions identifying an administration regimen therefor.
18. The packaged pharmaceutical according to claim 17 for use for the treatment or prevention of respiratory syncytial virus infection in a mammal.
19. An article of manufacture comprising packaging material contained within which is a composition effective to inhibit a respiratory syncytial virus, the packaging material comprising a label which indicates that the composition can be used to treat or prevent infection by a respiratory syncytial virus, wherein said composition is a composition according to claim 9.
20. A method of treating or preventing a respiratory syncytial virus infection in a mammal comprising administering to the mammal an anti-respiratory syncytial virally-effective amount of a compound of formula (I), or an enantiomer or a salt thereof or a composition thereof:
Figure US20070225317A1-20070927-C00337
wherein R1 is —(CH═CH)0-1—(C6 or C10C)aryl or —(CH═CH)0-1-5-, 6-, 9- or 10-membered heteroaryl, said aryl or heteroaryl being optionally substituted with one, two or three substituents, each independently selected from:
(C1-6)alkyl optionally substituted with amino, halo, (C1-6)haloalkyl, hydroxy, (C1-6)alkoxy, (C1-6)alkylthio, nitro, azido, cyano, amino, (C1-6)alkylamino, di((C1-6)alkyl)amino, aryl and heteroaryl;
R2 is H, (C1-6)alkyl, hydroxy, halo, (C1-6)haloalkyl, amino, (C1-6)alkylamino, di((C1-6)alkyl)amino, or (C2-6)alkynyl;
R3 is (C6, C10 or C14)aryl or 5-, 6-, 9- or 10-membered heteroaryl, each of which being optionally substituted with one, two or three substituents, each independently selected from:
(C1-6)alkyl, halo, (C1-6)haloalkyl, hydroxy, (C1-6)alkoxy, (C1-6)alkylthio, nitro, amino, (C1-6)alkylamino, di((C1-6)alkyl)amino and COO(C1-6)alkyl; and
R4 and R5 are each independently H or (C1-6)alkyl; or R4 and R5 are linked, together with the carbon atom to which they are attached, to form a (C3-7)cycloalkyl group.
21. The method according to claim 20 wherein the composition additionally comprises a therapeutically effective amount of at least one other antiviral agent.
22. The method according to claim 21 wherein the antiviral agent is selected from an immunomodulatory agent, an inhibitor of RSV polymerase or an inhibitor of another target in the RSV life cycle.
23. A method of inhibiting replication of a respiratory syncytial virus comprising exposing virus-infected cells to an anti-respiratory syncytial virally-effective amount of a compound, wherein the compound is of formula (I) of claim 20 or an enantiomer or a salt thereof.
24. A packaged pharmaceutical for use for the treatment or prevention of respiratory syncytial virus infection in a mammal comprising a pharmaceutical composition containing a compound and directions identifying an administration regimen, wherein the compound is of formula (I) of claim 20 or an enantiomer or a salt thereof.
25. An article of manufacture comprising packaging material contained within which is a composition effective to inhibit a respiratory syncytial virus, the packaging material comprising a label which indicates that the composition can be used to treat or prevent infection by a respiratory syncytial virus, wherein said composition includes a compound of formula (I) of claim 20 or an enantiomer or a salt thereof.
26. The composition according to claim 9, wherein, in the compound of formula (Ia):
R1 is —(CH═CH)0-1—(C6 or C10)aryl or 5-, 6-, 9- or 10-membered heteroaryl, said aryl or heteroaryl being optionally substituted with one, two or three substituents, each independently selected from:
(C1-6)alkyl optionally substituted with amino, halo, (C1-6)haloalkyl, hydroxy, (C1-6)alkoxy, (C1-6)alkylthio, nitro, azido, cyano, amino, (C1-6)alkylamino, di((C1-6)alkyl)amino, aryl and heteroaryl;
R2 is H, (C1-6)alkyl, hydroxy, halo, (C1-6)haloalkyl, amino, (C1-6)alkylamino, di((C1-6)alkyl)amino, or (C2-6)alkynyl;
R3 is (C6, C10 or C14)aryl or 5-, 6-, 9- or 10-membered heteroaryl, each of which being optionally substituted with one, two or three substituents, each independently selected from:
(C1-6)alkyl, halo, (C1-6)haloalkyl, hydroxy, (C1-6)alkoxy, (C1-6)alkylthio, amino, (C1-6)alkylamino and di((C1-6)alkyl)amino; and
R4 and R5 are each independently (C1-6)alkyl; or R4 and R5 are linked, together with the carbon atom to which they are attached, to form a (C3-7)cycloalkyl group;
with the proviso that R1 is not 2-methoxyphenyl, when R2 is H, R3 is 3,4-dimethoxyphenyl, R4 is CH3 and R5 is CH3.
27. The composition according to claim 9, wherein, in the compound of formula (Ia):
R1 is (CH═CH)0-1-phenyl, the phenyl being optionally substituted with one, two or three substituents, each independently selected from:
(C1-6)alkyl optionally substituted with amino, halo, (C1-6)haloalkyl, hydroxy, (C1-6)alkoxy, (C1-6)alkylthio, amino, nitro, cyano, azido, (C1-6)alkylamino, di((C1-6)alkyl)amino, and heteroaryl;
or R1 is a 5-, 6-, 9- or 10-membered heteroaryl optionally substituted with one, two or three substituents, each independently selected from nitro, (C1-6)alkyl optionally substituted with amino, (C1-6)haloalkyl, hydroxy, (C1-6)alkoxy, halo, (C1-6)alkylthio, cyano and heteroaryl;
R2 is H, (C1-6)alkyl, hydroxy, halo, amino, (C1-6)alkylamino, or (C2-6)alkynyl;
R3 is naphthyl, anthryl, or phenyl optionally substituted with one, two or three substituents, each independently selected from:
(C1-6)alkyl, halo, (C1-6)haloalkyl, hydroxy, (C1-6)alkoxy, and (C1-6)alkylthio;
or R3 is a 5-, 6-, 9- or 10-membered heteroaryl optionally substituted with (C1-6)alkyl; and
R4 and R5 are each independently Me or Et; or R4 and R5 are linked, together with the carbon atom to which they are attached, to form a (C3-7)cycloalkyl group;
with the proviso that R1 is not 2-methoxyphenyl, when R2 is H, R3 is 3,4-dimethoxyphenyl, R4 is CH3 and R5 is CH3.
28. The composition according to claim 9, wherein, in the compound of formula (Ia):
R1 is phenyl, 2-pyridyl, quinolinyl or benzofuranyl, the phenyl and 2-pyridyl each being optionally substituted with one or two substituents, each independently selected from:
methyl, hydroxy, methoxy, nitro, cyano, methylthio, fluoro, 2-aminoethyl and CF3;
R2 is H, NH2, bromo, chloro, or OH;
R3 is naphthyl, or phenyl optionally substituted with one, two or three substituents, each independently selected from:
methyl, iodo, hydroxy, methoxy, ethoxy and methylthio; and
R4 and R5 are each independently Me; or R4 and R5 are linked, together with the carbon atom to which they are attached, to form a cyclopentyl or cyclohexyl group;
with the proviso that R1 is not 2-methoxyphenyl, when R2 is H, R3 is 3,4-dimethoxyphenyl, R4 is CH3 and R5 is CH3.
29. The composition according to claim 9, wherein, in the compound of formula (Ia):
R1 is phenyl, 2-pyridyl, quinolinyl or benzofuranyl, the phenyl and 2-pyridyl each being optionally substituted with one or two substituents, each independently selected from:
methyl, methoxy, fluoro, CF3, nitro, cyano, and methylthio;
R2 is H, NH2, bromo, chloro, or OH;
R3 is phenyl optionally substituted with one, two or three substituents, each independently selected from:
hydroxy, methoxy, ethoxy, methyl, iodo and methylthio; and
R4 and R5 are each independently Me; or R4 and R5 are linked, together with the carbon atom to which they are attached, to form a cyclopentyl or cyclohexyl group;
with the proviso that R1 is not 2-methoxyphenyl, when R2 is H, R3 is 3,4-dimethoxyphenyl, R4 is CH3 and R5 is CH3.
30. The composition according to claim 9, wherein, in the compound of formula (Ia):
R1 is
Figure US20070225317A1-20070927-C00338
R2 is NH2 or bromo;
R3 is phenyl substituted with one or two substituents each independently selected from hydroxy, methoxy, and iodo; and
R4 and R5 are each independently Me; or R4 and R5 are linked, together with the carbon atom to which they are attached, to form a cyclohexyl group.
31. The composition according to claim 9, wherein, the compound of formula (Ia) is selected from the compounds of the following formula:
Figure US20070225317A1-20070927-C00339
wherein R1, R2 and R3 are defined as in the table below:
Cpd entry # R1 R2 R3 1002
Figure US20070225317A1-20070927-C00340
H
Figure US20070225317A1-20070927-C00341
1003
Figure US20070225317A1-20070927-C00342
H
Figure US20070225317A1-20070927-C00343
1004
Figure US20070225317A1-20070927-C00344
H
Figure US20070225317A1-20070927-C00345
1005
Figure US20070225317A1-20070927-C00346
H
Figure US20070225317A1-20070927-C00347
1006
Figure US20070225317A1-20070927-C00348
H
Figure US20070225317A1-20070927-C00349
1007
Figure US20070225317A1-20070927-C00350
H
Figure US20070225317A1-20070927-C00351
1008
Figure US20070225317A1-20070927-C00352
H
Figure US20070225317A1-20070927-C00353
1009
Figure US20070225317A1-20070927-C00354
H
Figure US20070225317A1-20070927-C00355
1010
Figure US20070225317A1-20070927-C00356
H
Figure US20070225317A1-20070927-C00357
1011
Figure US20070225317A1-20070927-C00358
H
Figure US20070225317A1-20070927-C00359
1012
Figure US20070225317A1-20070927-C00360
H
Figure US20070225317A1-20070927-C00361
1013
Figure US20070225317A1-20070927-C00362
H
Figure US20070225317A1-20070927-C00363
1014
Figure US20070225317A1-20070927-C00364
H
Figure US20070225317A1-20070927-C00365
1015
Figure US20070225317A1-20070927-C00366
H
Figure US20070225317A1-20070927-C00367
1016
Figure US20070225317A1-20070927-C00368
H
Figure US20070225317A1-20070927-C00369
1017
Figure US20070225317A1-20070927-C00370
H
Figure US20070225317A1-20070927-C00371
1018
Figure US20070225317A1-20070927-C00372
H
Figure US20070225317A1-20070927-C00373
1019
Figure US20070225317A1-20070927-C00374
H
Figure US20070225317A1-20070927-C00375
1020
Figure US20070225317A1-20070927-C00376
H
Figure US20070225317A1-20070927-C00377
1021
Figure US20070225317A1-20070927-C00378
H
Figure US20070225317A1-20070927-C00379
1022
Figure US20070225317A1-20070927-C00380
H
Figure US20070225317A1-20070927-C00381
1023
Figure US20070225317A1-20070927-C00382
H
Figure US20070225317A1-20070927-C00383
1024
Figure US20070225317A1-20070927-C00384
H
Figure US20070225317A1-20070927-C00385
1025
Figure US20070225317A1-20070927-C00386
H
Figure US20070225317A1-20070927-C00387
1026
Figure US20070225317A1-20070927-C00388
H
Figure US20070225317A1-20070927-C00389
1027
Figure US20070225317A1-20070927-C00390
H
Figure US20070225317A1-20070927-C00391
1028
Figure US20070225317A1-20070927-C00392
H
Figure US20070225317A1-20070927-C00393
1029
Figure US20070225317A1-20070927-C00394
H
Figure US20070225317A1-20070927-C00395
1030
Figure US20070225317A1-20070927-C00396
H
Figure US20070225317A1-20070927-C00397
1031
Figure US20070225317A1-20070927-C00398
H
Figure US20070225317A1-20070927-C00399
1032
Figure US20070225317A1-20070927-C00400
H
Figure US20070225317A1-20070927-C00401
1033
Figure US20070225317A1-20070927-C00402
H
Figure US20070225317A1-20070927-C00403
1034
Figure US20070225317A1-20070927-C00404
H
Figure US20070225317A1-20070927-C00405
1035
Figure US20070225317A1-20070927-C00406
H
Figure US20070225317A1-20070927-C00407
1036
Figure US20070225317A1-20070927-C00408
H
Figure US20070225317A1-20070927-C00409
1037
Figure US20070225317A1-20070927-C00410
H
Figure US20070225317A1-20070927-C00411
1038
Figure US20070225317A1-20070927-C00412
H
Figure US20070225317A1-20070927-C00413
1039
Figure US20070225317A1-20070927-C00414
H
Figure US20070225317A1-20070927-C00415
1040
Figure US20070225317A1-20070927-C00416
H
Figure US20070225317A1-20070927-C00417
1041
Figure US20070225317A1-20070927-C00418
H
Figure US20070225317A1-20070927-C00419
1042
Figure US20070225317A1-20070927-C00420
H
Figure US20070225317A1-20070927-C00421
1043
Figure US20070225317A1-20070927-C00422
H
Figure US20070225317A1-20070927-C00423
1044
Figure US20070225317A1-20070927-C00424
H
Figure US20070225317A1-20070927-C00425
1045
Figure US20070225317A1-20070927-C00426
H
Figure US20070225317A1-20070927-C00427
1046
Figure US20070225317A1-20070927-C00428
Br
Figure US20070225317A1-20070927-C00429
1047
Figure US20070225317A1-20070927-C00430
H
Figure US20070225317A1-20070927-C00431
1048
Figure US20070225317A1-20070927-C00432
NH3
Figure US20070225317A1-20070927-C00433
1049
Figure US20070225317A1-20070927-C00434
C≡CH
Figure US20070225317A1-20070927-C00435
1050
Figure US20070225317A1-20070927-C00436
NHMe
Figure US20070225317A1-20070927-C00437
1051
Figure US20070225317A1-20070927-C00438
Me
Figure US20070225317A1-20070927-C00439
1052
Figure US20070225317A1-20070927-C00440
Br
Figure US20070225317A1-20070927-C00441
1053
Figure US20070225317A1-20070927-C00442
Br
Figure US20070225317A1-20070927-C00443
1054
Figure US20070225317A1-20070927-C00444
Cl
Figure US20070225317A1-20070927-C00445
1055
Figure US20070225317A1-20070927-C00446
OH
Figure US20070225317A1-20070927-C00447
1056
Figure US20070225317A1-20070927-C00448
Br
Figure US20070225317A1-20070927-C00449
1057
Figure US20070225317A1-20070927-C00450
Br
Figure US20070225317A1-20070927-C00451
1058
Figure US20070225317A1-20070927-C00452
NH3
Figure US20070225317A1-20070927-C00453
1059
Figure US20070225317A1-20070927-C00454
NH3
Figure US20070225317A1-20070927-C00455
1060
Figure US20070225317A1-20070927-C00456
Br
Figure US20070225317A1-20070927-C00457
1061
Figure US20070225317A1-20070927-C00458
Br
Figure US20070225317A1-20070927-C00459
1062
Figure US20070225317A1-20070927-C00460
Br
Figure US20070225317A1-20070927-C00461
1063
Figure US20070225317A1-20070927-C00462
Br
Figure US20070225317A1-20070927-C00463
1064
Figure US20070225317A1-20070927-C00464
Br
Figure US20070225317A1-20070927-C00465
1065
Figure US20070225317A1-20070927-C00466
Br
Figure US20070225317A1-20070927-C00467
1066
Figure US20070225317A1-20070927-C00468
Br
Figure US20070225317A1-20070927-C00469
1067
Figure US20070225317A1-20070927-C00470
Br
Figure US20070225317A1-20070927-C00471
1068
Figure US20070225317A1-20070927-C00472
Br
Figure US20070225317A1-20070927-C00473
1069
Figure US20070225317A1-20070927-C00474
Br
Figure US20070225317A1-20070927-C00475
1070
Figure US20070225317A1-20070927-C00476
Br
Figure US20070225317A1-20070927-C00477
1071
Figure US20070225317A1-20070927-C00478
Br
Figure US20070225317A1-20070927-C00479
1072
Figure US20070225317A1-20070927-C00480
Br
Figure US20070225317A1-20070927-C00481
1073
Figure US20070225317A1-20070927-C00482
Br
Figure US20070225317A1-20070927-C00483
1074
Figure US20070225317A1-20070927-C00484
Br
Figure US20070225317A1-20070927-C00485
1075
Figure US20070225317A1-20070927-C00486
NH3
Figure US20070225317A1-20070927-C00487
1076
Figure US20070225317A1-20070927-C00488
NH3
Figure US20070225317A1-20070927-C00489
1077
Figure US20070225317A1-20070927-C00490
NH3
Figure US20070225317A1-20070927-C00491
1078
Figure US20070225317A1-20070927-C00492
NH3
Figure US20070225317A1-20070927-C00493
1079
Figure US20070225317A1-20070927-C00494
NH3
Figure US20070225317A1-20070927-C00495
1080
Figure US20070225317A1-20070927-C00496
NH3
Figure US20070225317A1-20070927-C00497
1081
Figure US20070225317A1-20070927-C00498
NH3
Figure US20070225317A1-20070927-C00499
1082
Figure US20070225317A1-20070927-C00500
NH3
Figure US20070225317A1-20070927-C00501
1083
Figure US20070225317A1-20070927-C00502
NH3
Figure US20070225317A1-20070927-C00503
1084
Figure US20070225317A1-20070927-C00504
NH3
Figure US20070225317A1-20070927-C00505
1085
Figure US20070225317A1-20070927-C00506
NH3
Figure US20070225317A1-20070927-C00507
1086
Figure US20070225317A1-20070927-C00508
NH3
Figure US20070225317A1-20070927-C00509
1087
Figure US20070225317A1-20070927-C00510
NH3
Figure US20070225317A1-20070927-C00511
1088
Figure US20070225317A1-20070927-C00512
NH3
Figure US20070225317A1-20070927-C00513
1089
Figure US20070225317A1-20070927-C00514
NH3
Figure US20070225317A1-20070927-C00515
1090
Figure US20070225317A1-20070927-C00516
Br
Figure US20070225317A1-20070927-C00517
1091
Figure US20070225317A1-20070927-C00518
Br
Figure US20070225317A1-20070927-C00519
1092
Figure US20070225317A1-20070927-C00520
Br
Figure US20070225317A1-20070927-C00521
1093
Figure US20070225317A1-20070927-C00522
Br
Figure US20070225317A1-20070927-C00523
1094
Figure US20070225317A1-20070927-C00524
Br
Figure US20070225317A1-20070927-C00525
1095
Figure US20070225317A1-20070927-C00526
Br
Figure US20070225317A1-20070927-C00527
1096
Figure US20070225317A1-20070927-C00528
Br
Figure US20070225317A1-20070927-C00529
1097
Figure US20070225317A1-20070927-C00530
Br
Figure US20070225317A1-20070927-C00531
1098
Figure US20070225317A1-20070927-C00532
Br
Figure US20070225317A1-20070927-C00533
1099
Figure US20070225317A1-20070927-C00534
Br
Figure US20070225317A1-20070927-C00535
1100
Figure US20070225317A1-20070927-C00536
Br
Figure US20070225317A1-20070927-C00537
1101
Figure US20070225317A1-20070927-C00538
Br
Figure US20070225317A1-20070927-C00539
1102
Figure US20070225317A1-20070927-C00540
Br
Figure US20070225317A1-20070927-C00541
1103
Figure US20070225317A1-20070927-C00542
Br
Figure US20070225317A1-20070927-C00543
1104
Figure US20070225317A1-20070927-C00544
Br
Figure US20070225317A1-20070927-C00545
1105
Figure US20070225317A1-20070927-C00546
Br
Figure US20070225317A1-20070927-C00547
1106
Figure US20070225317A1-20070927-C00548
Br
Figure US20070225317A1-20070927-C00549
1107
Figure US20070225317A1-20070927-C00550
Br
Figure US20070225317A1-20070927-C00551
1108
Figure US20070225317A1-20070927-C00552
Br
Figure US20070225317A1-20070927-C00553
1109
Figure US20070225317A1-20070927-C00554
Br
Figure US20070225317A1-20070927-C00555
1110
Figure US20070225317A1-20070927-C00556
Br
Figure US20070225317A1-20070927-C00557
1111
Figure US20070225317A1-20070927-C00558
Br
Figure US20070225317A1-20070927-C00559
1112
Figure US20070225317A1-20070927-C00560
Br
Figure US20070225317A1-20070927-C00561
1113
Figure US20070225317A1-20070927-C00562
Br
Figure US20070225317A1-20070927-C00563
1114
Figure US20070225317A1-20070927-C00564
Br
Figure US20070225317A1-20070927-C00565
1115
Figure US20070225317A1-20070927-C00566
Br
Figure US20070225317A1-20070927-C00567
1116
Figure US20070225317A1-20070927-C00568
Br
Figure US20070225317A1-20070927-C00569
1117
Figure US20070225317A1-20070927-C00570
Br
Figure US20070225317A1-20070927-C00571
1118
Figure US20070225317A1-20070927-C00572
Br
Figure US20070225317A1-20070927-C00573
1119
Figure US20070225317A1-20070927-C00574
Br
Figure US20070225317A1-20070927-C00575
1120
Figure US20070225317A1-20070927-C00576
Br
Figure US20070225317A1-20070927-C00577
1121
Figure US20070225317A1-20070927-C00578
Br
Figure US20070225317A1-20070927-C00579
1122
Figure US20070225317A1-20070927-C00580
Br
Figure US20070225317A1-20070927-C00581
1123
Figure US20070225317A1-20070927-C00582
Br
Figure US20070225317A1-20070927-C00583
1124
Figure US20070225317A1-20070927-C00584
Br
Figure US20070225317A1-20070927-C00585
1124
Figure US20070225317A1-20070927-C00586
Br
Figure US20070225317A1-20070927-C00587
1125
Figure US20070225317A1-20070927-C00588
Br
Figure US20070225317A1-20070927-C00589
1126
Figure US20070225317A1-20070927-C00590
Br
Figure US20070225317A1-20070927-C00591
1127
Figure US20070225317A1-20070927-C00592
Br
Figure US20070225317A1-20070927-C00593
1128
Figure US20070225317A1-20070927-C00594
Br
Figure US20070225317A1-20070927-C00595
1129
Figure US20070225317A1-20070927-C00596
Br
Figure US20070225317A1-20070927-C00597
1130
Figure US20070225317A1-20070927-C00598
NH3
Figure US20070225317A1-20070927-C00599
1131
Figure US20070225317A1-20070927-C00600
NH3
Figure US20070225317A1-20070927-C00601
1132
Figure US20070225317A1-20070927-C00602
H
Figure US20070225317A1-20070927-C00603
32. The composition according to claim 9, wherein, the compound of formula (Ia) is selected from the compounds of the following formula:
Figure US20070225317A1-20070927-C00604
wherein R1, R2, R3, R4 and R5 are defined as in the table below:
Cpd entry # R1 R2
Figure US20070225317A1-20070927-C00605
R3
2001
Figure US20070225317A1-20070927-C00606
H
Figure US20070225317A1-20070927-C00607
Figure US20070225317A1-20070927-C00608
2002
Figure US20070225317A1-20070927-C00609
H
Figure US20070225317A1-20070927-C00610
Figure US20070225317A1-20070927-C00611
2003
Figure US20070225317A1-20070927-C00612
H
Figure US20070225317A1-20070927-C00613
Figure US20070225317A1-20070927-C00614
2004
Figure US20070225317A1-20070927-C00615
H
Figure US20070225317A1-20070927-C00616
Figure US20070225317A1-20070927-C00617
2005
Figure US20070225317A1-20070927-C00618
Br
Figure US20070225317A1-20070927-C00619
Figure US20070225317A1-20070927-C00620
2006
Figure US20070225317A1-20070927-C00621
NH2
Figure US20070225317A1-20070927-C00622
Figure US20070225317A1-20070927-C00623
2007
Figure US20070225317A1-20070927-C00624
H
Figure US20070225317A1-20070927-C00625
Figure US20070225317A1-20070927-C00626
2008
Figure US20070225317A1-20070927-C00627
Br
Figure US20070225317A1-20070927-C00628
Figure US20070225317A1-20070927-C00629
2009
Figure US20070225317A1-20070927-C00630
NH2
Figure US20070225317A1-20070927-C00631
Figure US20070225317A1-20070927-C00632
2010
Figure US20070225317A1-20070927-C00633
NH2
Figure US20070225317A1-20070927-C00634
Figure US20070225317A1-20070927-C00635
2011
Figure US20070225317A1-20070927-C00636
NH2
Figure US20070225317A1-20070927-C00637
Figure US20070225317A1-20070927-C00638
US11/750,157 2003-10-30 2007-05-17 Rsv polymerase inhibitors Abandoned US20070225317A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/750,157 US20070225317A1 (en) 2003-10-30 2007-05-17 Rsv polymerase inhibitors

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US51582003P 2003-10-30 2003-10-30
US10/975,562 US7323567B2 (en) 2003-10-30 2004-10-28 RSV polymerase inhibitors
US11/750,157 US20070225317A1 (en) 2003-10-30 2007-05-17 Rsv polymerase inhibitors

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/975,562 Division US7323567B2 (en) 2003-10-30 2004-10-28 RSV polymerase inhibitors

Publications (1)

Publication Number Publication Date
US20070225317A1 true US20070225317A1 (en) 2007-09-27

Family

ID=34549448

Family Applications (2)

Application Number Title Priority Date Filing Date
US10/975,562 Active 2026-03-10 US7323567B2 (en) 2003-10-30 2004-10-28 RSV polymerase inhibitors
US11/750,157 Abandoned US20070225317A1 (en) 2003-10-30 2007-05-17 Rsv polymerase inhibitors

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US10/975,562 Active 2026-03-10 US7323567B2 (en) 2003-10-30 2004-10-28 RSV polymerase inhibitors

Country Status (5)

Country Link
US (2) US7323567B2 (en)
EP (1) EP1682547B1 (en)
JP (1) JP4537407B2 (en)
CA (1) CA2541618C (en)
WO (1) WO2005042530A1 (en)

Families Citing this family (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2001480A4 (en) 2006-03-31 2011-06-15 Abbott Lab Indazole compounds
US8999969B2 (en) 2009-07-09 2015-04-07 Gilead Sciences, Inc. Anti-RSV compounds
JP2013531801A (en) * 2010-07-02 2013-08-08 ヴェンタナ メディカル システムズ, インク. Hapten conjugates for target detection
EP3338784B1 (en) 2010-12-31 2020-07-22 Abbott Laboratories Human milk oligosaccharides for modulating inflammation
NZ612472A (en) 2010-12-31 2015-02-27 Abbott Lab Nutritional compositions comprising human milk oligosaccharides and nucleotides and uses thereof for treating and/or preventing enteric viral infection
CN103391783A (en) 2010-12-31 2013-11-13 雅培制药有限公司 Methods of using human milk oligosaccharides for improving airway respiratory health
MY166945A (en) 2010-12-31 2018-07-25 Abbott Lab Nutritional formulations including human milk oligosaccharides and long chain polyunsaturated polyunsaturated fatty acids and uses thereof
SG11201600977XA (en) 2013-08-21 2016-03-30 Alios Biopharma Inc Antiviral compounds
MA41614A (en) 2015-02-25 2018-01-02 Alios Biopharma Inc ANTIVIRAL COMPOUNDS
JO3637B1 (en) 2015-04-28 2020-08-27 Janssen Sciences Ireland Uc Rsv antiviral pyrazolo- and triazolo-pyrimidine compounds
WO2017015449A1 (en) 2015-07-22 2017-01-26 Enanta Pharmaceuticals, Inc. Benzodiazepine derivatives as rsv inhibitors
WO2017123884A1 (en) 2016-01-15 2017-07-20 Enanta Pharmaceuticals, Inc. Heterocyclic compounds as rsv inhibitors
KR102573499B1 (en) 2017-02-16 2023-08-31 이난타 파마슈티칼스, 인코포레이티드 Method for producing benzodiazepine derivatives
US11091501B2 (en) 2017-06-30 2021-08-17 Enanta Pharmaceuticals, Inc. Heterocyclic compounds as RSV inhibitors
US10881666B2 (en) 2017-09-29 2021-01-05 Enanta Pharmaceuticals, Inc. Combination pharmaceutical agents as RSV inhibitors
WO2019094903A1 (en) 2017-11-13 2019-05-16 Enanta Pharmaceuticals, Inc. Processes for the resolution of benzodiazepin-2-one and benzoazepin-2-one derivatives
TW201932470A (en) 2017-11-29 2019-08-16 愛爾蘭商健生科學愛爾蘭無限公司 Pyrazolopyrimidines having activity against RSV
KR20200116099A (en) * 2018-01-31 2020-10-08 얀센 사이언시즈 아일랜드 언리미티드 컴퍼니 Cycloalkyl-substituted pyrazolopyrimidines with activity against RSV
MA52348A (en) 2018-04-23 2021-03-03 Janssen Sciences Ireland Unlimited Co HETEROAROMATIC COMPOUNDS WITH AN ACTIVITY AGAINST RSV
KR20210138684A (en) 2019-03-18 2021-11-19 이난타 파마슈티칼스, 인코포레이티드 Benzodiazepine derivatives as RSV inhibitors
US11505558B1 (en) 2019-10-04 2022-11-22 Enanta Pharmaceuticals, Inc. Antiviral heterocyclic compounds
EP4037688A4 (en) 2019-10-04 2023-09-06 Enanta Pharmaceuticals, Inc. Antiviral heterocyclic compounds
UY39032A (en) 2020-01-24 2021-07-30 Enanta Pharm Inc HETEROCYCLIC COMPOUNDS AS ANTIVIRAL AGENTS
WO2022010882A1 (en) 2020-07-07 2022-01-13 Enanta Pharmaceuticals, Inc, Dihydroquinoxaline and dihydropyridopyrazine derivatives as rsv inhibitors
US11945824B2 (en) 2020-10-19 2024-04-02 Enanta Pharmaceuticals, Inc. Heterocyclic compounds as anti-viral agents
UY39642A (en) 2021-02-26 2022-09-30 Enanta Pharm Inc ANTIVIRAL HETEROCYCLIC COMPOUNDS

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4176184A (en) * 1977-07-21 1979-11-27 Boehringer Ingelheim Gmbh Imidazoisoquinoline-diones and salts thereof

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE2732906C2 (en) * 1977-07-21 1986-09-25 Dr. Karl Thomae Gmbh, 7950 Biberach Imidazo-isoquinoline-diones substituted in the 5-position by an aminoalkyl radical, process for their preparation and pharmaceuticals
DE3410168A1 (en) * 1984-03-20 1985-09-26 Boehringer Mannheim Gmbh, 6800 Mannheim Novel isochinoline diones, process for their preparation, medicaments containing these compounds and intermediates
EP1051169A4 (en) 1998-01-29 2002-05-29 Viropharma Inc Compounds, compositions and methods for treating or preventing pneumovirus infection and associated diseases
WO2000004900A1 (en) 1998-07-20 2000-02-03 Bristol-Myers Squibb Company Substituted benzimidazole antiviral agents
CA2376785A1 (en) * 1999-06-28 2001-01-04 Janssen Pharmaceutica N.V. Benzimidazoles and imidazopyridines as respiratory syncytial virus replication inhibitors
SK18942001A3 (en) 1999-06-28 2002-10-08 Janssen Pharmaceutica N. V. Respiratory syncytial virus replication inhibitors
UA74787C2 (en) 1999-06-28 2006-02-15 Janssen Pharmaceutica Nv Respiratory syncytial virus replication inhibitors
EP1222187B1 (en) * 1999-10-06 2004-09-22 Boehringer Ingelheim Pharmaceuticals Inc. Heterocyclic compounds useful as inhibitors of tyrosine kinases
GB9924990D0 (en) 1999-10-21 1999-12-22 Glaxo Group Ltd Medicaments
US6489338B2 (en) 2000-06-13 2002-12-03 Bristol-Myers Squibb Company Imidazopyridine and imidazopyrimidine antiviral agents
US7030150B2 (en) 2001-05-11 2006-04-18 Trimeris, Inc. Benzimidazole compounds and antiviral uses thereof

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4176184A (en) * 1977-07-21 1979-11-27 Boehringer Ingelheim Gmbh Imidazoisoquinoline-diones and salts thereof

Also Published As

Publication number Publication date
EP1682547B1 (en) 2012-10-24
EP1682547A4 (en) 2008-12-31
US20050239814A1 (en) 2005-10-27
JP2007509860A (en) 2007-04-19
US7323567B2 (en) 2008-01-29
CA2541618C (en) 2011-12-13
WO2005042530A1 (en) 2005-05-12
CA2541618A1 (en) 2005-05-12
JP4537407B2 (en) 2010-09-01
EP1682547A1 (en) 2006-07-26

Similar Documents

Publication Publication Date Title
US20070225317A1 (en) Rsv polymerase inhibitors
US10604533B2 (en) Heterocycle amines and uses thereof
US8030318B2 (en) Fused bicyclic inhibitors of HCV
US20070066632A1 (en) Fused bicyclic inhibitors of TGFbeta
TWI397530B (en) Substituted triazolopyridines and analogs thereof
DK2609082T3 (en) IMIDAZO [4,5-C] QUINOLINES AS DNA-PK INHIBITORS
US11766435B2 (en) N3-substituted iminopyrimidinones as antimalarial agents
US8383630B2 (en) Azaindazole compounds and methods of use
US20160046616A1 (en) Nrf2 small molecule inhibitors for cancer therapy
DK2588457T3 (en) PYRAZOLOQUINOLINDERIVATER as DNA-PK inhibitors
SG183855A1 (en) Morpholinylquinazolines
BRPI0722411A2 (en) bicyclic pyrimidinones and their uses
CA2687625A1 (en) 3-(imidazolyl)-pyrazolo[3,4-b]pyridines
US9169248B2 (en) Diazole amides
CA2664376A1 (en) Polycyclic agents for the treatment of respiratory syncytial virus infections
US20220142975A1 (en) Pharmaceutical Combination and Use Thereof
US10342782B2 (en) Positive allosteric modulators of the muscarinic acetylcholine receptor M4
WO2019210880A1 (en) Dicyclourea nucleocapsid inhibitor and pharmaceutical use thereof
US6743795B1 (en) 3-nitropyridine derivaives and the pharmaceutical compositions containing said derivatives
US20230241025A1 (en) Antiviral use of fabp4 modulating compounds
US20190167688A1 (en) Positive allosteric modulators of the muscarinic acetylcholine receptor m4
WO2023185073A1 (en) Parp7 inhibitor and use thereof
US20230365525A1 (en) Antiviral compounds
TW202246259A (en) Pyrazoleamide derivatives

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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