USRE38132E1 - Phenylpyrrolidines, phenylimidazolidines,3-phenyl-1,3-oxizolidines and 3-phenyl-1,3-thiazolidines and their use in the treatment of inflammatory disease - Google Patents

Phenylpyrrolidines, phenylimidazolidines,3-phenyl-1,3-oxizolidines and 3-phenyl-1,3-thiazolidines and their use in the treatment of inflammatory disease Download PDF

Info

Publication number
USRE38132E1
USRE38132E1 US10/167,732 US16773202A USRE38132E US RE38132 E1 USRE38132 E1 US RE38132E1 US 16773202 A US16773202 A US 16773202A US RE38132 E USRE38132 E US RE38132E
Authority
US
United States
Prior art keywords
group
carbon atoms
formula
alkyl
hydrogen atom
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.)
Expired - Lifetime
Application number
US10/167,732
Inventor
Terence A. Kelly
Barbara Jean Bormann
Leah Lynn Frye
Jiang-Ping Wu
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.)
Boehringer Ingelheim Pharmaceuticals Inc
Original Assignee
Boehringer Ingelheim Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from PCT/US1998/004254 external-priority patent/WO1998039303A1/en
Priority claimed from US09/375,010 external-priority patent/US6355664B1/en
Application filed by Boehringer Ingelheim Pharmaceuticals Inc filed Critical Boehringer Ingelheim Pharmaceuticals Inc
Priority to US10/167,732 priority Critical patent/USRE38132E1/en
Application granted granted Critical
Publication of USRE38132E1 publication Critical patent/USRE38132E1/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D207/00Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D207/02Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D207/30Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having two double bonds between ring members or between ring members and non-ring members
    • C07D207/34Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having two double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D207/36Oxygen or sulfur atoms
    • C07D207/402,5-Pyrrolidine-diones
    • C07D207/4042,5-Pyrrolidine-diones with only hydrogen atoms or radicals containing only hydrogen and carbon atoms directly attached to other ring carbon atoms, e.g. succinimide
    • C07D207/408Radicals containing only hydrogen and carbon atoms attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D207/00Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D207/02Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D207/30Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having two double bonds between ring members or between ring members and non-ring members
    • C07D207/34Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having two double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D207/36Oxygen or sulfur atoms
    • C07D207/402,5-Pyrrolidine-diones
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/02Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings
    • C07D231/10Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D231/12Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D233/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings
    • C07D233/54Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members
    • C07D233/56Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members with only hydrogen atoms or radicals containing only hydrogen and carbon atoms, attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D233/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings
    • C07D233/54Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members
    • C07D233/66Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D233/72Two oxygen atoms, e.g. hydantoin
    • C07D233/74Two oxygen atoms, e.g. hydantoin with only hydrogen atoms or radicals containing only hydrogen and carbon atoms, attached to other ring members
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D233/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings
    • C07D233/54Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members
    • C07D233/66Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D233/72Two oxygen atoms, e.g. hydantoin
    • C07D233/76Two oxygen atoms, e.g. hydantoin with substituted hydrocarbon radicals attached to the third ring carbon atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D233/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings
    • C07D233/54Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members
    • C07D233/66Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D233/72Two oxygen atoms, e.g. hydantoin
    • C07D233/76Two oxygen atoms, e.g. hydantoin with substituted hydrocarbon radicals attached to the third ring carbon atom
    • C07D233/78Radicals substituted by oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D233/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings
    • C07D233/54Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members
    • C07D233/66Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D233/72Two oxygen atoms, e.g. hydantoin
    • C07D233/80Two oxygen atoms, e.g. hydantoin with hetero atoms or acyl radicals directly attached to ring nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D233/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings
    • C07D233/54Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members
    • C07D233/66Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D233/84Sulfur atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D233/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings
    • C07D233/54Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members
    • C07D233/66Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D233/86Oxygen and sulfur atoms, e.g. thiohydantoin
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D249/00Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms
    • C07D249/02Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms not condensed with other rings
    • C07D249/081,2,4-Triazoles; Hydrogenated 1,2,4-triazoles
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D263/00Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings
    • C07D263/02Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings
    • C07D263/30Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D263/34Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D263/44Two oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/10Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/06Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/10Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/06Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/553Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having one nitrogen atom as the only ring hetero atom
    • C07F9/576Six-membered rings
    • C07F9/59Hydrogenated pyridine rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/645Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having two nitrogen atoms as the only ring hetero atoms
    • C07F9/6503Five-membered rings
    • C07F9/6506Five-membered rings having the nitrogen atoms in positions 1 and 3
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6558Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing at least two different or differently substituted hetero rings neither condensed among themselves nor condensed with a common carbocyclic ring or ring system
    • C07F9/65583Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing at least two different or differently substituted hetero rings neither condensed among themselves nor condensed with a common carbocyclic ring or ring system each of the hetero rings containing nitrogen as ring hetero atom

Definitions

  • the present invention relates generally to a series of novel small molecules, their synthesis and their use in the treatment of inflammatory disease.
  • the invention further relates to the use of similar, but known, compounds in the treatment of inflammatory disease.
  • integrins constituitively expressed on leukocytes
  • LFA-1 integrin-1
  • ICAM-2 intercellular adhesion molecules
  • ICAM-4 distinct intercellular adhesion molecules
  • Immune processes such as antigen presentation, T-cell mediated cytotoxicity and leukocyte extravasation all require cellular adhesion mediated by ICAMs interacting with the Leukointegrins. See generally Kishimoto, T. K.; Rothlein; R. R. Adv. Pharmacol. 1994,25:117-138 and Diamond, M.; Springer, T. Current Biology, 1994, 4, 506-532.
  • Leukocyte Adhesion Deficiency a condition termed “Leukocyte Adhesion Deficiency” (Anderson, D. C.; et al., Fed. Proc. 1985, 44, 2671-2677 and Anderson, D. C.; et al., J. Infect. Dis. 1985, 152, 668-689).
  • These individuals are unable to mount a normal inflammatory and/or immune response(s) due to an inability of their cells to adhere to cellular substrates.
  • These data show that immune reactions are mitigated when lymphocytes are unable to adhere in a normal fashion due to the lack of functional adhesion molecules of the CD18 family.
  • antagonism of CD18, CD11 /ICAM-1 interactions will also inhibit an inflammatory response.
  • antagonism of the interaction between the CAMs and the Leukointegrins can be realized by agents directed against either component.
  • blocking of the CAMs, such as for example ICAM-1, or the Leukointegrins, such as for example LFA-1 by antibodies directed against either or both of these molecules effectively inhibits inflammatory responses.
  • In vitro models of inflammation and immune response inhibited by antibodies to CAMs or Leukointegrins include antigen or mitogen-induced lymphocyte proliferation, homotypic aggregation of lymphocytes, T-cell mediated cytolysis and antigen-specific induced tolerance. The relevance of the in vitro studies are supported by in vivo studies with antibodies directed against ICAM-1 or LFA-1.
  • antibodies directed against LFA-1 can prevent thyroid graft rejection and prolong heart allograft survival in mice (Gorski, A.; Immunology Today, 1994, 15,251-255).
  • ICAM-1 have shown efficacy in vivo as anti-inflammatory agents in human diseases such as renal allograft rejection and rheumatoid arthritis (Rothlein, R. R.; Scharschmidt, L., in: Adhesion Molecules; Wegner, C. D., Ed.; 1994, 1-38, Cosimi, C. B.; et al., J. Immunol. 1990, 144, 4604-4612 and Kavanaugh, A.; et al., Arthritis Rheum.
  • Soluble ICAM-1 acts as a direct antagonist of CD18, CD11 /ICAM-1 interactions on cells and shows inhibitory activity in in vitro models of immune response such as the human mixed lymphocyte response, cytotoxic T cell responses and T cell proliferation from diabetic patients in response to islet cells (Becker, J. C.; et al., J. Immunol. 1993, 151, 7224 and Roep, B. O.; et al., Lancet, 1994, 343, 1590).
  • a first aspect of the invention comprises a method for treating or preventing inflammatory and immune cell-mediated disease(s) by the administration of certain novel and known small molecules. These compounds act by inhibiting the interaction of cellular adhesion molecules, specifically by antagonizing the binding of human intercellular adhesion molecules (including, for example, ICAM-1, ICAM-2 and ICAM-3) to the Leukointegrins (including, for example, CD18/CD11a and CD18/CD11b).
  • a second aspect of the invention comprises novel small molecules having the above-noted therapeutic activities.
  • a third aspect of the invention comprises methods for making these novel compounds.
  • a final aspect of the invention comprises pharmaceutical compositions comprising the above-mentioned compounds suitable for the prevention or treatment of inflammatory and immune cell-mediated condition(s).
  • the invention comprises a method for treating or preventing inflammatory and immune cell-mediated diseases by the administration of certain novel and known small molecules of the formula I
  • Y is an oxygen or sulfur atom
  • Z is an oxygen or sulfur atom
  • X is a divalent group of the formula >CHR 1 , >NR 1 , >CHSO 2 R 1 , or >NSO 2 R 1 , or an oxygen or sulfur atom, wherein R 1 is:
  • aryl which is selected from the class consisting of phenyl, naphthyl, indolyl, thiophenyl, pyridyl, pyrimidinyl, furyl, pyrrolyl, oxazolyl, thiazolyl, pyrazolyl, isoxazolyl, imidazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl, pyridazinyl, pyrazinyl, triazinyl, indolyzinyl, isoindolyl, benzo[b]furanyl, benzo[b]thiophenyl, indazolyl, benzthiazolyl, benzimidazolyl, quinolinyl, isoquinolinyl, purinyl, quinolizinyl, cinnolinyl, pthalaninyl, quinoxalinyl, napthy
  • R 8 and R 9 are each independently a hydrogen atom, alkyl of 1 to 6 carbon atoms, cycloalkyl of 3 to 6 carbon atoms or acyl of 1 to 7 carbon atoms, or wherein R 8 and R 9 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring,
  • R 10 and R 11 are each independently a hydrogen atom, alkyl of 1 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, or wherein R 10 and R 11 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring,
  • R 13 , R 14 and R 15 are each, independently, a hydrogen atom or alkyl of 1 to 3 carbon atoms and wherein two of R 13 , R 14 and R 15 may additionally constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom(s) between them form a heterocyclic ring,
  • R 17 and R 18 are each, independently, a hydrogen atom, alkyl of 1 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, or wherein R 17 and R 18 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring,
  • R 21 and R 22 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring, or
  • R 24 , R 25 and R 26 are each, independently, a branched or unbranched alkyl group of 1 to 7 carbon atoms and Q ⁇ is a chlorine, bromine or iodine counterion,
  • R 27 , R 28 and R 29 are each, independently, a hydrogen atom or alkyl of I to 3 carbon atoms, and wherein two of R 27 , R 28 and R 29 may additionally constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom(s) between them form a heterocyclic ring,
  • R 30 , R 31 , R 32 and R 33 are each, independently, a hydrogen atom or alkyl of 1 to 3 carbon atoms, and wherein two of R 30 , R 31 , R 32 and R 33 may additionally constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom(s) between them form a heterocyclic ring,
  • aryl which is selected from the class consisting of phenyl, naphthyl, indolyl, thiophenyl, pyridyl, pyrimidinyl, furyl, pyrrolyl, oxazolyl, thiazolyl, pyrazolyl, isoxazolyl, imidazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl, pyridazinyl, pyrazinyl, triazinyl, indolyzinyl, isoindolyl, benzo[b]furanyl, benzo[b]thiophenyl, indazolyl, benzthiazolyl, benzimidazolyl, quinolinyl, isoquinolinyl, purinyl, quinolizinyl, cinnolinyl, pthalaninyl, quinoxalinyl, napthyridiny
  • R 8 and R 9 are each, independently, a hydrogen atom, alkyl of 1 to 6 carbon atoms, cycloalkyl of 3 to 6 carbon atoms or acyl of 1 to 7 carbon atoms, or wherein R 8 and R 9 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring,
  • R 10 and R 11 are each, independently, a hydrogen atom, alkyl of 1 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, or wherein R 10 and R 11 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring,
  • (ix) a group of the formula —SR 12b , wherein R 12b is a hydrogen atom, or an alkyl or acyl group of 1 to 7 carbon atoms,
  • R 13 , R 14 and R 15 are each, independently, a hydrogen atom or alkyl of 1 to 3 carbon atoms, and wherein two of R 13 , R 14 and R 15 may additionally constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom(s) between them form a heterocyclic ring;
  • R 2 is:
  • R 34 is a hydrogen atom, or an alkyl or acyl group of 1 to 7 carbon atoms, or
  • R 35 and R 36 are each, independently, a hydrogen atom, alkyl of 1 to 2 carbon atoms, or acyl of 1 to 2 carbon atoms;
  • R 3 is a group of the formula —(CR 375l R38 ) x (CR 39 R 40 ) y R 41 , wherein;
  • x and y are each independently 0 or 1
  • R 37 , R 38 and R 39 are each, independently:
  • (B) a group of the formula —OR 42 , wherein R 42 is a hydrogen atom, or an alkyl or acyl group of 1 to 7 carbon atoms, or
  • R 40 is:
  • (B) a group of the formula —OR 42 , wherein R 42 is a hydrogen atom, or an alkyl or acyl group of 1 to 7 carbon atoms,
  • (D) aryl which is selected from the class consisting of phenyl, 2-naphthyl, 2-, 3-, 5- or 6-indolyl, 2- or 3-thiophenyl, 2-, 3- or 4-pyridyl, 2-, 4- or 5-pyrimidinyl, 2- or 3-furyl, 1-, 2- or 3-pyrrolyl, 2-, 4- or 5-oxazolyl, 2-, 4- or 5-thiazolyl, 1-, 3-, 4- or 5-pyrazolyl, 3-, 4- or 5-isoxazolyl, 1-, 2-, 4- or 5-imidazolyl, 3-, 4- or 5-isothiazolyl, 4- or 5-oxadiazolyl, 1-, 4- or 5-triazolyl, 2-thiadiazolyl, 3- or 4-pyridazinyl, 2-pyrazinyl, 2-triazinyl, 2-, -3,6-or 7-indolyzinyl, 2-, 3-, 5- or 6-isoindolyl, 2-, 3-, 5- or 6-benzo
  • R 43 which is aryl selected from the class consisting of phenyl, 2-naphthyl, 2-, 3-, 5- or 6-indolyl, 2- or 3-thiophenyl, 2-, 3- or 4-pyridyl, 2-, 4- or 5-pyrimidinyl, 2- or 3-furyl, 1-, 2- or 3-pyrrolyl, 2-, 4- or 5-oxazolyl, 2-, 4- or 5-thiazolyl, 1-, 3-, 4- or 5-pyrazolyl, 3-, 4- or 5-isoxazolyl, 1-, 2-, 4- or 5-midazolyl, 3-, 4- or 5-isothiazolyl, 4- or 5-oxadiazolyl, 1-, 4- or 5-triazolyl, 2-thiadiazolyl, 3- or 4-pyridazinyl, 2-pyrazinyl, 2-triazinyl, 2-, -3, 6- or 7-indolyzinyl, 2-, 3-, 5- or 6-isoindolyl, 2-, 3-, 5- or 6-benz
  • R 44 is straight or branched alkyl of 1 to 5 carbon atoms or cycloalkyl of 3 to 5 carbon atoms,
  • R 45 and R 46 are each, independently, a hydrogen atom, alkyl or fluoroalkyl of 1 to 6 carbon atoms, cycloalkyl of 3 to 6 carbon atoms or acyl of 1 to 7 carbon atoms, or wherein R 45 and R 46 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring,
  • R 47 and R 48 are each independently a hydrogen atom, alkyl or fluoroalkyl of 1 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, or wherein R 47 and R 48 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring,
  • R 49 is a hydrogen atom, or an alkyl, fluoroalkyl or acyl group of 1 to 7 carbon atoms,
  • R 50 is a hydrogen atom, or an alkyl, fluoroalkyl or acyl group of 1 to 7 carbon atoms,
  • R 51 , R 52 and R 53 are each, independently, a hydrogen atom or alkyl of 1 to 3 carbon atoms, and wherein two of R 51 , R 52 and R 53 may additionally constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom(s) between them form a heterocyclic ring, or
  • R 55 and R 56 are each, independently, a hydrogen atom, alkyl or fluoroalkyl of 1 to 6 carbon atoms, cycloalkyl of 3 to 6 carbon atoms or acyl of 1 to 7 carbon atoms, or wherein R 55 and R 56 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring, and wherein one of R 55 and R 56 may additionally be the group R 43 ,
  • R 57 and R 58 are each, independently, a hydrogen atom, alkyl or fluoroalkyl of 1 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, or wherein R 57 and R 58 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring, and wherein one of R 57 and R 58 may additionally be the group R 43 ,
  • (ix) a group of the formula —SR 61 , wherein R 61 is a hydrogen atom, an alkyl, fluoroalkyl or acyl group of 1 to 7 carbon atoms, or R 43 ,
  • R 41 is:
  • aryl selected from the class consisting of phenyl, 2-naphthyl, 2-, 3-, 5- or 6-indolyl, 2- or 3-thiophenyl, 2-, 3- or 4-pyridyl, 2-, 4- or 5-pyrimidinyl, 2- or 3-furyl, 1-, 2- or 3-pyrrolyl, 2-, 4- or 5-oxazolyl, 2-, 4- or 5-thiazolyl, 1-, 3-, 4- or 5-pyrazolyl, 3-, 4- or 5-isoxazolyl, 1-, 2-, 4- or 5-imidazolyl, 3-, 4- or 5-isothiazolyl, 4- or 5-oxadiazolyl, 1-, 4- or 5-triazolyl, 2-thiadiazolyl, 3- or 4-pyridazinyl, 2-pyrazinyl, 2-triazinyl, 2-, -3, 6- or 7-indolyzinyl, 2-, 3-, 5- or 6-isoindolyl, 2-, 3-, 5- or 6-benzo[b]faranyl
  • R 62 which is aryl selected from the class consisting of phenyl, 2-naphthyl, 2-, 3-, 5- or 6-indolyl, 2- or 3-thiophenyl, 2-, 3- or 4-pyridyl, 2-, 4- or 5-pyrimidinyl, 2- or 3-furyl, 1-, 2- or 3-pyrrolyl, 2-, 4- or 5-oxazolyl, 2-, 4- or 5-thiazolyl, 1-, 3-, 4- or 5-pyrazolyl, 3-, 4- or 5-isoxazolyl, 1-, 2-, 4- or 5-imidazolyl, 3-, 4- or 5-isothiazolyl, 4- or 5-oxadiazolyl, 1-, 4- or 5-triazolyl, 2-thiadiazolyl, 3- or 4-pyridazinyl, 2-pyrazinyl, 2-triazinyl, 2-, -3, 6- or 7-indolyzinyl, 2-, 3-, 5- or 6-isoindolyl, 2-, 3-, 5- or
  • R 64 and R 65 are each, independently, a hydrogen atom, alkyl or fluoroalkyl of 1 to 6 carbon atoms, cycloalkyl of 3 to 6 carbon atoms or acyl of 1 to 7 carbon atoms, or wherein R 64 and R 65 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring,
  • R 66 and R 67 are each, independently, a hydrogen atom, alkyl or fluoroalkyl of 1 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, or wherein R 66 and R 67 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring,
  • (ix) a group of the formula —SR 69 , wherein R 69 is a hydrogen atom, or an alkyl, fluoroalkyl or acyl group of 1 to 7 carbon atoms,
  • R 70 , R 71 and R 72 are each, independently, a hydrogen atom or alkyl or fluoroalkyl of 1 to 3 carbon atoms, and wherein two of R 70 , R 71 and R 72 may additionally constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom(s) between them form a heterocyclic ring, or
  • (B) methyl which may be mono- or polysubstituted with fluorine atoms and additionally may be monosubstituted with R 62 ,
  • R 74 and R 75 are each, independently, a hydrogen atom, alkyl or fluoroalkyl of 1 to 6 carbon atoms, cycloalkyl of 3 to 6 carbon atoms or acyl of 1 to 7 carbon atoms, or wherein R 74 and R 75 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring, and wherein one of R 74 and R 75 may additionally be the group R 62 ,
  • R 76 and R 77 are each, independently, a hydrogen atom, alkyl or fluoroallyl of 1 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, or wherein R 76 and R 77 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring, and wherein one of R 76 and R 77 may additionally be the group R 62 ,
  • (G) a group of the formula —COR 78 , wherein R 78 is a hydrogen atom, straight or branched alkyl of 1 to 5 carbon atoms, cycloalkyl of 3 to 5 carbon atoms or R 62 ,
  • (H) a group of the formula —OR 79 , wherein R 79 is a hydrogen atom, an alkyl, fluoroalkyl or acyl group of 1 to 7 carbon atoms, or R 62 ,
  • (I) a group of the formula —SR 80 , wherein R 80 is a hydrogen atom, an alkyl, fluoroalkyl or acyl group of 1 to 7 carbon atoms, or R 62 ,
  • R 4 is Cl or trifluoromethyl; and, R 5 and R 6 are each, independently, a hydrogen, fluorine, chlorine, bromine or iodine atom, methyl or trifluoromethyl;
  • the invention comprises novel compounds of the formula I
  • R 62 which is aryl selected from the class consisting of phenyl, 2-naphthyl, 2-, 3-, 5- or 6-indolyl, 2- or 3-thiophenyl, 2-, 3- or 4-pyridyl, 2-, 4- or 5-pyrimidinyl, 2- or 3-furyl, 1-, 2- or 3-pyrrolyl, 2-, 4- or 5-oxazolyl, 2-, 4- or 5-thiazolyl, 1-, 3-, 4- or 5-pyrazolyl, 3-, 4- or 5-isoxazolyl, 1-, 2-, 4- or 5-imidazolyl, 3-, 4- or 5-isothiazolyl, 4- or 5-oxadiazolyl, 1-, 4- or 5-triazolyl, 2-thiadiazolyl, 3- or 4-pyridazinyl, 2-pyrazinyl, 2-triazinyl, 2-, -3, 6- or 7-indolyzinyl, 2-, 3-, 5- or 6-isoindolyl, 2-, 3-, 5- or
  • R 64 and R 65 are each, independently, a hydrogen atom, alkyl or fluoroalkyl of 1 to 6 carbon atoms, cycloalkyl of 3 to 6 carbon atoms or acyl of 1 to 7 carbon atoms, or wherein R 64 and R 65 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring,
  • R 66 and R 67 are each, independently, a hydrogen atom, alkyl or fluoroalkyl of 1 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, or wherein R 66 and R 67 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring,
  • (ix) a group of the formula —SR 69 , wherein R 69 is a hydrogen atom, or an alkyl , fluoroalkyl or acyl group of 1 to 7 carbon atoms,
  • R 70 , R 71 and R 72 are each, independently, a hydrogen atom or alkyl or fluoroalkyl of 1 to 3 carbon atoms, and wherein two of R 70 , R 71 and R 72 may additionally constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom(s) between them form a heterocyclic ring, or
  • (B) methyl which may be mono- or polysubstituted with fluorine atoms and additionally may be monosubstituted with R 62 ,
  • R 74 and R 75 are each, independently, a hydrogen atom, alkyl or fluoroalkyl of 1 to 6 carbon atoms, cycloalkyl of 3 to 6 carbon atoms or acyl of 1 to 7 carbon atoms, or wherein R 74 and R 75 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring, and wherein one of R 74 and R 75 may additionally be the group R 62 ,
  • R 76 and R 77 are each, independently, a hydrogen atom, alkyl or fluoroalkyl of 1 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, or wherein R 76 and R 77 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring, and wherein one of R 76 and R 77 may additionally be the group R 62 ,
  • (G) a group of the formula —COR 78 , wherein R 78 is a hydrogen atom, straight or branched alkyl of 1 to 5 carbon atoms, cycloalkyl of 3 to 5 carbon atoms or R 62 ,
  • (H) a group of the formula —OR 79 , wherein R 79 is a hydrogen atom, an alkyl fluoroalkyl or acyl group of 1 to 7 carbon atoms, or R 62 ,
  • (I) a group of the formula —SR 80 , wherein R 80 is a hydrogen atom, an alkyl, fluoroalkyl or acyl group of 1 to 7 carbon atoms, or R 62 ,
  • Preferred novel compounds of formula I are those wherein:
  • Y is an oxygen or sulfur atom
  • Z is an oxygen or sulfur atom
  • X is a divalent group of the formula ⁇ CHR 1 , ⁇ NR 1 , ⁇ CHSO 2 R 1 , or ⁇ NSO 2 R 1 , or an oxygen or sulfur atom, wherein R 1 is:
  • aryl selected from the class consisting of phenyl, naphthyl, indolyl, thiophenyl, pyridyl, pyrimidinyl, furyl, pyrrolyl, oxazolyl, thiazolyl, pyrazolyl, isoxazolyl, imidazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl, pyridazinyl, pyrazinyl, triazinyl, indolyzinyl, isoindolyl, benzo[b]furanyl, benzo[b]thiophenyl, indazolyl, benzthiazolyl, benzimidazolyl, quinolinyl, isoquinolinyl, purinyl, quinolizinyl, cinnolinyl, pthalaninyl, quinoxalinyl, napthyridiny
  • R 8 and R 9 are each, independently, a hydrogen atom, alkyl of 1 to 6 carbon atoms, cycloalkyl of 3 to 6 carbon atoms or acyl of 1 to 7 carbon atoms, or wherein R 8 and R 9 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring,
  • R 10 and R 11 are each, independently, a hydrogen atom, alkyl of 1 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, or wherein R 10 and R 11 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring,
  • R 13 , R 14 and R 15 are each, independently, a hydrogen atom or alkyl of 1 to 3 carbon atoms and wherein two of R 13 , R 14 and R 15 may additionally constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom(s) between them form a heterocyclic ring,
  • R 17 and R 18 are each, independently, a hydrogen atom, alkyl of 1 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, or wherein R 17 and R 18 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring,
  • R 24 , R 25 and R 26 are each, independently, a branched or unbranched alkyl group of 1 to 7 carbon atoms and Q ⁇ is a chlorine, bromine or iodine counterion,
  • R 27 , R 28 and R 29 are each, independently, a hydrogen atom or alkyl of 1 to 3 carbon atoms and wherein two of R 27 , R 28 and R 29 may additionally constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom(s) between them form a heterocyclic ring,
  • R 30 , R 31 , R 32 and R 33 are each independently a hydrogen atom or alkyl of 1 to 3 carbon atoms and wherein two of R 30 , R 31 , R 32 and R 33 may additionally constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom(s) between them form a heterocyclic ring, or
  • R 2 is:
  • R 3 is a group of the formula —CH 2 R 41 , wherein:
  • R 41 is:
  • aryl selected from the class consisting of phenyl, 2-naphthyl, 2-, 3-, 5- or 6-indolyl, 2- or 3-thiophenyl, 2-, 3- or 4-pyridyl, 2-, 4- or 5-pyrimidinyl, 2- or 3-furyl, 1-, 2- or 3-pyrrolyl, 2-, 4- or 5-oxazolyl, 2-, 4- or 5-thiazolyl, 1-, 3-, 4- or 5-pyrazolyl, 3-, 4- or 5-isoxazolyl, 1-, 2-, 4- or 5-imidazolyl, 3-, 4- or 5-isothiazolyl, 4- or 5-oxadiazolyl, 1-, 4- or 5-triazolyl, 2-thiadiazolyl, 3- or 4-pyridazinyl, 2-pyrazinyl, 2-triazinyl, 2-, - 3 , 6- or 7-indolyzinyl, 2-, 3-, 5- or 6-isoindolyl, 2-, 3-, 5- or 6-benzo[b]fur
  • R 62 which is aryl selected from the class consisting of phenyl, 2-naphthyl, 2-, 3-, 5- or 6-indolyl, 2- or 3-thiophenyl, 2-, 3- or 4-pyridyl, 2-, 4- or 5-pyrimidinyl, 2- or 3-furyl, 1-, 2- or 3-pyrrolyl, 2-, 4- or 5-oxoazolyl, 2-, 4- or 5-thiazolyl, 1-, 3-, 4- or 5-pyrazolyl, 3-, 4- or 5-isoxazolyl, 1-, 2-, 4- or 5-imidazolyl, 3-, 4- or 5-isothiazolyl, 4- or 5-oxadiazolyl, 1-, 4- or 5-triazolyl, 2-thiadiazolyl, 3- or 4-pyridazinyl, 2-pyrazinyl, 2-triazinyl, 2-, -3, 6- or 7-indolyzinyl, 2-, 3-, 5- or 6-isoindolyl, 2-, 3-, 5-
  • R 64 and R 65 are each, independently, a hydrogen atom, alkyl or fluooralkyl of 1 to 6 carbon atoms, cycloalkyl of 3 to 6 carbn atoms or acyl of 1 to 7 carbon atoms, or wherein R 64 and R 65 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring,
  • R 66 and R 67 are each, independently, a hydrogen atom, alkyl or fluoroalkyl of 1 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, or wherein R 66 and R 67 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring,
  • (ix) a group of the formula —SR 69 , wherein R 69 is a hydrogen atom, or an alkyl, fluoroalkyl or acyl group of 1 to 7 carbon atoms,
  • R 70 , R 71 and R 72 are each, independently, a hydrogen atom or alkyl or fluoroalkyl of 1 to 3 carbon atoms, and wherein two of R 70 , R 71 and R 72 may additionally constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom(s) between them form a heterocyclic ring, or
  • (B) methyl which may be mono- or polysubstituted with fluorine atoms and additionally may be monosubstituted with R 62 ,
  • R 74 and R 75 are each, independently, a hydrogen atom, alkyl or fluoroalkyl of 1 to 6 carbon atoms, cycloalkyl of 3 to 6 carbon atoms or acyl of 1 to 7 carbon atoms, or wherein R 74 and R 75 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring, and wherein one of R 74 and R 75 may additionally be the group, R 62 ,
  • R 76 and R 77 are each, independently, a hydrogen atom, alkyl or fluoroalkyl of 1 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, or wherein R 76 and R 77 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring, and wherein one of R 76 and R 77 may additionally be the group R 62 ,
  • (G) a group of the formula —COR 78 , wherein R 78 is a hydrogen atom, straight or branched alkyl of 1 to 5 carbon atoms, cycloalkyl of 3 to 5 carbon atoms or R 62 ,
  • (H) a group of the formula —OR 79 , wherein R 79 is a hydrogen atom, an alkyl, fluoroalkyl or acyl group of 1 to 7 carbon atoms, or R 62 ,
  • (I) a group of the formula —SR 80 , wherein R 80 is a hydrogen atom, an alkyl, fluoroalkyl or acyl group of 1 to 7 carbon atoms, or R 62 ,
  • R 4 is Cl or trifluoromethyl
  • R 5 and R 6 are each independently a hydrogen, fluorine, chlorine, bromine or iodine atom, methyl or trifluoromethyl;
  • Y is an oxygen atom
  • Z is an oxygen atom
  • X is a divalent group of the formula >CHR 1 or >NR 1 , wherein R 1 is:
  • aryl selected from the class consisting of phenyl, thiophenyl, pyridyl, pyrimidinyl, furyl, pyrrolyl, oxazolyl, thiazolyl, pyrazolyl, isoxazolyl, imidazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl, pyridazinyl, pyrazinyl and triazinyl,
  • R 7 is straight or branched alkyl of 1 to 5 carbon
  • R 12a is a hydrogen atom or a methyl
  • R 13 , R 14 and R 15 are each hydrogen atoms
  • (k) a group of the formula —OR 19 , wherein R 19 is a hydrogen atom, or an alkyl or acyl group of 1 to 7 carbon atoms, or
  • R 24 , R 25 and R 26 are each methyl and Q ⁇ is a chlorine, bromine or iodine counterion
  • R 27 , R 28 and R 29 are each hydrogen atoms
  • R 30 , R 31 , R 32 and R 33 are each hydrogen atoms, or
  • R 2 is:
  • R 3 is a group of the formula —CH 2 R 41 , wherein
  • aryl selected from the class consisting of phenyl, thiophenyl, pyridyl, pyrimidinyl, furyl, pyrrolyl, oxazolyl, thiazolyl, pyrazolyl, isoxazolyl, imidazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl, pyridazinyl, pyrazinyl and triazinyl, wherein one or more of the hydrogen atoms of said aryl group are necessarily and independently replaced with:
  • R 62 which is aryl selected from the class consisting of phenyl, thiophenyl, pyridyl, pyrimidinyl, furyl, pyrrolyl, oxazolyl, thiazolyl, pyrazolyl, isoxazolyl, imidazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl, pyridazinyl, pyrazinyl and triazinyl,
  • R 4 is Cl or trifluoromethyl
  • R 5 is a hydrogen atom
  • R 6 is Cl, or trifluoromethyl
  • Y is an oxygen atom
  • Z is an oxygen atom
  • X is a divalent group of the formula >CHR 1 or >NR 1 , wherein R 1 is:
  • aryl selected from the class consisting of phenyl, thiophenyl, pyridyl, pyrimidinyl, furyl, pyrrolyl, oxazolyl, thiazolyl, pyrazolyl, isoxazolyl, imidazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl, pyridazinyl, pyrazinyl and triazinyl, wherein one or more hydrogen atoms of said aryl group may be optionally and independently replaced with:
  • R 7 is straight or branched alkyl of 1 to 5 carbon
  • R 12a is a hydrogen atom or a methyl
  • R 13 , R 14 and R 15 are each hydrogen atoms
  • R 16 is straight or branched alkyl of 1 to 7
  • R 19 is a hydrogen atom, or an alkyl or acyl
  • R 24 , R 25 and R 26 are each methyl and
  • Q ⁇ is a chlorine, bromine or iodine counterion
  • R 27 , R 28 and R 29 are each hydrogen atoms
  • R 30 , R 31 , R 32 and R 33 are each hydrogen atoms, or
  • R 2 is:
  • R 3 is a group of the formula —CH 2 R 41 , wherein
  • aryl selected from the class consisting of phenyl, thiophenyl, pyridyl, pyrimidinyl, furyl, oxazolyl, thiazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, thiadiazolyl, pyridazinyl, and pyrazinyl, wherein one or more of the hydrogen atoms of said aryl group are necessarily and independently replaced with:
  • R 62 which is aryl selected from the class consisting of phenyl, thiophenyl, pyridyl, pyrimidinyl, furyl, oxazolyl, thiazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, thiadiazolyl, pyridazinyl, and pyrazinyl,
  • (B) methyl which may be mono- or polysubstituted with fluorine atoms or which may be monosubstituted with R 62 ;
  • R 4 is Cl or trifluoromethyl
  • R 5 is a hydrogen atom
  • R 6 is Cl, or trifluoromethyl
  • Y is an oxygen atom
  • Z is an oxygen atom
  • X is a divalent group of the formula >CHR 1 or >NR 1 , wherein R 1 is:
  • aryl selected from the class consisting of phenyl or pyridyl
  • R 12a is a hydrogen atom or a methyl
  • R 13 , R 14 and R 15 are each hydrogen atoms
  • R 24 , R 25 and R 26 are each methyl and Q ⁇ is a chlorine, bromine or iodine counterion
  • R 27 , R 28 and R 29 are each hydrogen atoms
  • R 30 , R 31 , R 32 and R 33 are each hydrogen atoms, or
  • R 2 is:
  • R 3 is a group of the formula —CH 2 R 41 , wherein
  • aryl selected from the class consisting of phenyl or pyridyl
  • (A) R 62 which is aryl selected from the class consisting of phenyl, or pyridyl,
  • R 4 is a chlorine atom or trifluoromethyl
  • R 5 is a hydrogen atom
  • R 6 is a chlorine atom, or trifluoromethyl
  • Y is an oxygen atom
  • Z is an oxygen atom
  • X is a divalent group of the formula >CHR 1 or >NR 1 , wherein R 1 is:
  • aryl selected from the class consisting of phenyl or pyridyl, wherein one hydrogen atom of said aryl group may be optionally replaced with:
  • R 12a is a hydrogen atom or a methyl, or
  • R 13 , R 14 and R 15 are each hydrogen atoms, or
  • R 27 , R 28 and R 29 are each hydrogen atoms, or
  • R 30 , R 31 , R 32 and R 33 are each hydrogen atoms
  • R 2 is:
  • R 3 is a group of the formula —CH 2 R 41 , wherein
  • (A) R 62 which is aryl selected from the class consisting of phenyl, or pyridyl,
  • R 4 is a chlorine atom or trifluoromethyl
  • R 5 is a hydrogen atom
  • R 6 is a chlorine atom, or trifluoromethyl
  • Y is an oxygen atom
  • Z is an oxygen atom
  • X is a divalent group of the formula >NR 1 , wherein R 1 is:
  • R 2 is:
  • R 3 is a group of the formula —CH 2 R 41 , wherein R 41 is:
  • (A) R 62 which is aryl selected from the class consisting of phenyl, or pyridyl,
  • R 4 is a chlorine atom or trifluoromethyl
  • R 5 is a hydrogen atom
  • R 6 is a chlorine atom, or trifluoromethyl
  • Y is an oxygen atom
  • Z is an oxygen atom
  • X is a divalent group of the formula >NR 1 , wherein R 1 is:
  • R 2 is:
  • R 3 is a group of the formula —CH 2 R 41 , wherein R 41 is:
  • (A) R 62 which is aryl selected from the class consisting of phenyl, or pyridyl,
  • R 4 is a chlorine atom
  • R 5 is a hydrogen atom
  • R 6 is a chlorine atom
  • Ethyl 2-(R)-amino-2-(4-bromobenzyl)-propanoate (the starting material for example 29) was produced from racemic ethyl 2-amino-2-(4-bromobenzyl)-propanoate by the following procedure: To 1.3 L of a buffer made from 13.69 g KH 2 PO 4 and 2 L of water was added 20 g of the commercially available enzyme Lipase L10 (Amino Enzyme USA Co., Ltd, Lombardi, Ill.) followed by 12 g of the HCl salt of the racemic amino ester.
  • Lipase L10 Amino Enzyme USA Co., Ltd, Lombardi, Ill.
  • aqueous base such as, for example, NaOH, KOH, Na 2 CO 3 , NaHCO 3 , K 2 CO 3 or KHCO 3
  • An appropriate isocyanate is added to this mixture and the resulting solution was stirred until the reaction essentially reaches completion.
  • the mixture is acidified and the resulting ureidoacetic acid is isolated by filtration or by extraction into organic solvent. Removal of solvent produces the intermediate ureidoacetic acid.
  • the intermediate ureidoacetic acid is cyclized by heating in the presence of a catalytic amount of acid (such as, for example, sulfuric acid, methanesulfonic acid, benzenesulfonic acid or hydrochloric acid) in an organic or aqueous solvent, to produce the desired hydantoin.
  • a catalytic amount of acid such as, for example, sulfuric acid, methanesulfonic acid, benzenesulfonic acid or hydrochloric acid
  • Workup consists of collection of the hydantoin by filtration and purification by, for example, silica gel chromatography or recrystallization.
  • aqueous base such as, for example, NaOH, KOH, Na 2 CO 3 , NaHCO 3 , K 2 CO 3 or KHCO 3
  • An appropriate isocyanate is added to this mixture and the resulting solution is stirred until the reaction essentially reaches completion.
  • the mixture is acidified and the resulting ureidoacetic acid is isolated by filtration or extraction into organic solvent. Removal of solvent produces the intermediate ureidoacetic acid.
  • the intermediate ureidoacetic acid is then cyclized to the desired hydantoin in organic solvent (such as, for example, DMF, NMP, or THF) using any of a number of dehydrating agents (such as, for example, dicyclohexylcarbodiimide (DCC) or 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide HCl (EDC)) in the presence of an ester activating agent (such as 1-hydroxybenzotriazole hydrate (HOBT)) and a non-nucleophilic base (such as, for example, triethylamine or N,N-diisopropylethylamine).
  • organic solvent such as, for example, DMF, NMP, or THF
  • dehydrating agents such as, for example, dicyclohexylcarbodiimide (DCC) or 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide
  • Method B is exemplified by the synthesis of the compound of Example 15 (see Table 2), which was carried out as follows: To a solution of (R)-phenylalanine (0.33 g, 2 mmol) in 1 mL of 2 N NaOH and 10 mL of water at 50° C. was added 3,5-dichlorophenyl isocyanate (0.38 g, 2 mmol). The resulting mixture was then stirred for 1 h. The solution was cooled and treated with concentrated HCl until a precipitate formed and the solution remained acidic. The precipitate was collected by filtration and dried in vacuo to produce the desired ureidoacetic acid (0.60 g, 85%).
  • the ureidoacetic acid (0.35 g, 1 mmol) was dissolved in 20 mL of DMF and treated with EDC (0.19 g, 1 mmol) and HOBT (0.14 g, 1 mmol) for 1 h at room temperature. After this period N,N-diisopropylethylamine (0.35 mL, 2 mmol) was added and the mixture stirred overnight. Workup consisted of trituration with water, collection of the hydantoin by filtration, and purification by silica gel chromatography. The yield in this example was 0.20 g (60%).
  • An appropriate amino ester or hydroxy ester and an appropriate isocyanate are dissolved in an organic solvent (such as, for example, DMF, TBF or DMSO) in the presence of a base (such as, for example, NaOH, KOH, Na 2 CO 3 , NaHCO 3 , K 2 CO 3 or KHCO 3 ) and warmed to between about room temperature and 60° C. After approximately 1 h, the temperature of the reaction mixture is raised to between about 50 and 100° C. until the reaction appears complete. The solution is then cooled and diluted with an organic solvent (such as, for example, EtOAc or CH 2 Cl 2 ). The organic phase is washed sequentially with dilute aqueous acid (e.g.
  • hydantoin 1 N HCl
  • water dried (e.g. over MgSO 4 ) and concentrated.
  • the desired hydantoin is purified, for example by silica gel chromatography or by recrystallization.
  • the ureidoacetic ester can be cyclized to the hydantoin by heating to between about 50 and 100° C. in the presence of an acid such as, for example, aqueous HCl until the reaction appears complete).
  • Method C is exemplified by the synthesis of the compound of Example 30 of Table 3, which is as follows: Methyl 2-amino-2-benzylbutyric acid (0.21 g, 1 mmol) and 3,5-dichlorophenyl isocyanate (0.19 g, 1 mmol) were dissolved in DMSO (5 mL) in the presence of approximately 0.2 g of Na 2 CO 3 and allowed to stir at 50° C. for 1 h. After this period the solution was heated to 90° C. for 2 hr. The solution was then cooled, diluted with EtOAc and washed with 0.1 N HCl and water. The organic layer was dried over MgSO 4 and concentrated to produce a crude product which was further purified by silica gel chromatography to yield 0.12 g (33%) of the compound of example 30.
  • Table 3 is as follows: Methyl 2-amino-2-benzylbutyric acid (0.21 g, 1 mmol) and 3,5-dichloroph
  • An amino acid attached to a solid phase resin through its carboxylic acid via an appropriate linker (for example the Wang resin: 4-benzyloxy-benzyl polystyrene) is protected on its nitrogen with a reagent that will allow for the alkylation of the alpha-carbon (for example, a benzaldehyde derivative that forms an imine with the nitrogen of the amino acid).
  • a reagent that will allow for the alkylation of the alpha-carbon (for example, a benzaldehyde derivative that forms an imine with the nitrogen of the amino acid).
  • the protected compound is then treated with a base and an alkylating agent to generate the new protected amino acid derivative.
  • the protecting group is removed using standard conditions (in the case of an imine this is accomplished, for example, with aqueous HCl) and the free amino group is reacted with an isocyanate to generate the intermediate urea.
  • This intermediate is treated with a reagent to catalyze the cyclization of the urea portion onto the carboxylate end of the molecule which forms the desired hydantoin and cleaves the product from the resin. Purification is via silica gel chromatography, reverse phase HPLC of recrystallization.
  • Method D is exemplified by the synthesis of the compound of Example 67, which was carried out as follows: A reaction vessel was charged with the commercially available Fmoc-Ala-Wang (300 mg, 0.150 mmol) and 3 mL of a 20% solution of piperidine in N-methyl pyrollidinone (NMP). The reaction vessel was agitated at room temperature on an orbital shaker for 45 minutes. The resin was filtered and washed with NMP (3 ⁇ 1 mL). The reaction vessel containing resin was equipped with a rubber septum, placed under argon, charged with 3,4-dichlorobenzaldehyde (394 mg, 2.25 mmol), trimethyl orthoformate (3.5 mL), and NMP (1.5 mL).
  • NMP N-methyl pyrollidinone
  • the resulting mixture was agitated at room temperature for 15 h.
  • the solid resin was isolated by filtration and washed sequentially with NMP (3 ⁇ 3 mL), tetrahydrofuran (3 ⁇ mL), and CH 2 Cl 2 (3 ⁇ mL). The resin was then dried under vacuum for approximately one hour to produce the imine-resin intermediate.
  • the imine-resin intermediate was alkylated with 2,3-difluoro-4-trifluormethylbenzyl bromide (123.8 mg, 0.45 mmol) by mixing these two reagents, 2-tert-butylimino-2-diethylamino-1,3-dimethylperhydro-1,3,2-diazaphosphorine (BEMP, 0.217 mL, 0.75 mmol), and NMP (3.5 mL) and agitating the mixture at room temperature on an orbital shaker for 15 h.
  • the solid was isolated by filtration and washed sequentially with NMP (3 ⁇ mL), THF (3 ⁇ mL), and CH 2 Cl 2 (3 ⁇ 3 mL) yielding the alkylated-imine-resin intermediate upon drying.
  • the imine was cleaved from the preceding intermediate by treatment with aqueous 1 N HCl (1.8 mL) and THF (3.6 mL) and agitating at room temperature for about 15 h.
  • the resin bound amino ester was isolated by filtration and washed sequentially with NMP (3 ⁇ 3 mL), THF (3 ⁇ 3 mL), and CH 2 Cl 2 (3 ⁇ 3 mL) and dried under vacuum.
  • the resin bound amino ester was converted to the hydantoin using a procedure that cleaves the final product from the resin.
  • the intermediate amino-ester was placed in a reaction vessel and treated with 3 mL of a 20% solution of N,N-diisopropylethylamine in NMP. After agitation at room temperature under argon for 1 h, the resin was filtered, washed with NMP (3 ⁇ 3 mL) and methanol (3 ⁇ 3 mL), and placed under vacuum. Subsequently, the vessel was opened under argon and charged with 2.5 mL of a 1.75 M solution of 3,5-dichlorphenylisocyanate in dimethylformamide (DMF, 0.45 mmol).
  • DMF dimethylformamide
  • an appropriate isocyante ester dissolved in an organic solvent (such as, for, example, methylene chloride) is added an appropriate aniline, and the mixture is stirred for between about 1 and 24 h, at about room temperature, under an inert atmosphere, such as argon. The organic solvent is is then removed in vacua. Excess aniline is removed (as by boiling the crude solid in hexanes and decanting off the liquid, or by flash chromatography over silica gel) leaving the solid ureidoacetic ester.
  • organic solvent such as, for, example, methylene chloride
  • the ureidoacetic ester is cyclized to the desired hydantoin by treatment with base (such as, for example, NaH, NaHMDS, Na 2 CO 3 , NaHCO 3 , K 2 CO 3 or KHCO 3 ) in an organic solvent (such as, for example, THF or DMF), followed by warming to approximately 60-90° C.
  • base such as, for example, NaH, NaHMDS, Na 2 CO 3 , NaHCO 3 , K 2 CO 3 or KHCO 3
  • organic solvent such as, for example, THF or DMF
  • EtOAc organic solvent
  • the organic solution is washed sequentially with dilute aqueous acid (such as 1 N HCl) and then water, dried (as with MgSO 4 ) and concentrated.
  • the desired hydantoin is purified, as by silica gel chromatography or recrystallization.
  • the intermediate ureidoacetic ester can be cyclized to the hydantoin by heating
  • Method E is exemplified by the synthesis of the compound from example 70, shown in Table 5, which was carried out as follows: To a solution of ethyl 2-isocyanato-3-phenylpropionate (99.0 mL, 0.110 g, 0.501 mmol) in dry CH 2 Cl 2 (5.0 mL) was added 3,4,5-trichloroaniline (0.1952 g, 0.994 mmol) as a solid. The mixture was stirred at room temperature under an argon atmosphere for 20 h. The solution was then concentrated in vacuo and the residue was recrystallized two times from ethyl acetate/hexanes to give 0.14 g (65%) of the pure intermediate urea as a white solid.
  • Equimolar amounts of the an appropriate starting diacid or anhydride and an appropriate starting aniline are refluxed in a solvent (such as xylene) in the presence of a catalytic amount of base (such as triethylamine) for between about 2 and 24 h.
  • a solvent such as xylene
  • base such as triethylamine
  • the solvent is removed in vacuo and the residue is dissolved in an organic solvent (such as EtOAc), washed sequentially with aqueous dilute base (such as NaHCO 3 ) and aqueous dilute acid (such as HCl), dried (for example over MgSO 4 ), and concentrated.
  • Purification is performed via, for example, recrystallization or chromatography over silica gel.
  • the starting diacids and anhydrides are available either commercially or via a number of known literature methods.
  • a procedure for the synthesis of 2-benzyl-3-carboxy-2-methylbutanoic acid (the starting material for example 74) is given.
  • the ester was subjected to a [3,3] sigmatropic rearrangement to produce the next intermediate. Under argon, at ⁇ 78° C., a solution of 560 mg of the intermediate ester (2.57 mmol) in THF (1 mL) was added to a THF solution of lithium di-isopropylamide (LDA, 3.25 mmol, generated from 1.3 mL of 2.5 M n-BuLi and 0.54 mL of iPr 2 NH in 3 mL of THF, ⁇ 10° C., 15 min) containing 500 microliter of DMPU. The mixture was stirred for 30 min before a solution of 480 mg of TBSCl (3.1 mmol) in 1 mL of THF was added.
  • LDA lithium di-isopropylamide
  • the mixture was stirred at ⁇ 78° C. for 30 min, at room temperature for 20 min and then heated at 60° C. for 10 h.
  • the mixture was cooled to 0° C., quenched with 2 N HCl (5 mL) and stirred at room temperature for 10 h.
  • the mixture was made basic with 2 N NaOH to pH 10, extracted with ether (5 mL).
  • the aqueous layer was separated, acidified to pH 1 with concentrated HCl, extracted with EtOAc and concentrated to give 500 mg (89%) of the intermediate: 2-benzyl-2,3-dimethyl-4-pentenoic acid.
  • the mono-acid was converted to the desired diacid by oxidation of the terminal alkene with ozone and the resulting intermediate further oxidation with a chromium reagent.
  • a solution of 500 mg of 2-benzyl-2,3-dimethyl-4-pentenoic acid (2.29 mmol) in MeOH (20 mL) and methylene chloride (10 mL) containing 120 microliter of pyridine was passed rapidly enough of a stream of O 3 at ⁇ 78° C., such that the solution turned slightly blue.
  • the mixture was treated with 1 mL of methyl sulfide and stirred at ⁇ 78° C. for 5 min.
  • the mixture was then warmed to room temperature, concentrated and passed through a silica gel column (with 10% MeOH in CH 2 Cl 2 as eluting solvent) and concentrated.
  • the crude material was dissolved in 5 mL of acetone and treated with Jones reagent (16 g CrO 3 16 g con. H 2 SO 4 in 100 mL of H 2 O) at room temperature until the orange color persisted. After addition of water (10 mL), the mixture was stirred for 1 h, washed with EtOAc and concentrated. The mixture was purified by silica gel chromatography with 3% AcOH-EtOAc to give 300 mg of the desired diacid (55%).
  • Method F is exemplified by the synthesis of the compounds of Examples 92 and 93 (see Table 6) which was carried out as follows: A mixture of isomers of the starting diacid of example 92 (0.58 g, 1.8 mmol, 3:1 mixture of isomers), 3,5-dichloroaniline (0.35 g, 2.2 mmol), Et 3 N (10 mL, 0.07 mmol) in xylene (5 mL) was refluxed under argon in a flask fitted with Dean-Stark trap for 24 h.
  • a typical sequence involves heating the substrate with a reagent such as P 2 S 3 in a high boiling solvent such as tetralin for between 1 and 48 h. Isolation of the product follows relatively standard conditions such as the dilution of the mixture into an organic solvent such as EtOAc and washing this mixture with water and saturated aqueous NaCl followed by drying and concentration. Purification is accomplished by silica gel chromatography or recrystallization, to afford the desired product.
  • a reagent such as P 2 S 3 in a high boiling solvent such as tetralin for between 1 and 48 h.
  • Isolation of the product follows relatively standard conditions such as the dilution of the mixture into an organic solvent such as EtOAc and washing this mixture with water and saturated aqueous NaCl followed by drying and concentration. Purification is accomplished by silica gel chromatography or recrystallization, to afford the desired product.
  • Method G is exemplified by the synthesis of the compound of Example 78, which was carried out as follows: The starting substrate (1.5 g, 3.5 mmol) was dissolved in 5 mL of tetralin, treated with P 2 S 3 (0.9 g, 5.7 mmol) and heated to 225° C. for 2 h. Upon cooling, the mixture was diluted with water and the product was extracted into EtOAc. The organic layer was washed with saturated aqueous NaCl, dried and concentrated. The residual oil was triturated with hexanes to produce a yellow solid which was isolated by filtration. This material was further purified by flash chromatography (1:4 EtOAc:Hexanes) to afford 1.13 g (70%%) of the desired compound.
  • the dithio-carbonyl containing compounds produced via Method G can be selectively hydrolyzed to each of the two monothio-carbonyl compounds depending on the choice of conditions.
  • the thio-carbonyl at the 4-position of the ring is more susceptible to nucleophilic conditions. As shown in Example 81, it can be converted to the 4-oxo-species by treatment with aqueous ethanolamine followed by acid hydrolysis.
  • the thio-carbonyl at the 2-position of the ring is more nucleophilic at sulfur and can be alkylated with methyl sulfate. This intermediate can then be hydrolyzed with mild acid. This affords the compound of Example 80. Purification of either class of compound is easily performed by silica gel chromatography or recrystallization.
  • Example 80 was prepared by treating a solution of the starting material (0.23 g, 0.49 mmol) in 3 mL of THF with aqueous with 10 mL of 50% aqueous ethanolamine and heating under reflux for 2 h. Upon cooling, the mixture was extracted with EtOAc and the organic layer was washed with water and saturated aqueous NaCl, dried and concentrated to give a brown solid. This solid was then treated with 20 mL of 6 N HCl and heated under reflux for 72 h. Upon cooling, the mixture was extracted with EtOAc and the organic layer was washed with saturated aqueous NaCl, dried and concentrated. The product was purified by preparative TLC over silica gel using 1:1 EtOAc:Hexanes as the solvent to produce the product in 34% yield.
  • Example 81 was prepared by treating a solution of the starting material (0.5 g, 1.09 mmol) in 1.6 mL of 2 N NaOH. As the compound did not initially dissolve, 1 mL of water and 1 mL of THF were added to aid solubility. This mixture was then cooled in an ice bath and Me 2 SO 4 (0.12 mL, 1.3 mmol) was added dropwise over 5 min. The mixture was stirred another 3 h at 0° C. and then for 45 min at room temperature. The reaction was quenched by the addition of enough 1 N HCl needed to lower the pH of the solution to 2. The mixture was extracted with EtOAc and the organic layer was washed with saturated aqueous NaCl, dried and concentrated to give a yellow oil.
  • hydantoin is dissolved in an aprotic solvent (such as, for example, DMF, THF or DMSO) and treated with one equivalent of a base (such as, for example, NaH, LDA, LiHMDS, KHMDS, KH or NaHMDS). After about 10 min to 1 h an appropriate alkylating agent is added and the mixture stirred at between about room temperature and 90° C. for up to about 24 h. (Progress of the reaction can be monitored using TLC). The solution is then cooled and diluted with an organic solvent (such as, for example, EtOAc or CH 2 Cl 2 ).
  • an organic solvent such as, for example, EtOAc or CH 2 Cl 2 ).
  • the organic phase is washed sequentially with a dilute acid (such as 1 N HCl) and water, dried (for example over MgSO 4 ) and concentrated.
  • a dilute acid such as 1 N HCl
  • the desired hydantoin is purified, as by silica gel chromatography or by recrystallization.
  • Method I is exemplified by the synthesis of the compound of Example 97 (see Table 9), which was carried out by dissolving the starting material (0.21 g, 0.5 mmol) in DMF (5 mL) and treating the solution sequentially a solution of 1 M NAHMDS (0.5 mL, 0.5 mmol) and EtI (0.04 mL, 0.5 mmol). After 1 h, the reaction mixture was partitioned between EtOAc and water, and the organic phase washed with water and dried over MgSO 4 . Column chromatography over silica gel produced 0.17 g (72%) of the desired product.
  • An appropriate heterocycle is dissolved in an aprotic solvent (such as DMF, TBF or DMSO) and treated with one equivalent of a base, (such as Et 3 N, LDA, KHMDS, LiHMDS or NaHMDS) at between about ⁇ 78° C. and room temperature. After about 10 min to 2 h an appropriate alkylating agent is added and the mixture stirred at between about 0 and 90° C. for up to about 24 h. (Progress of the reaction can be monitored using TLC). The solution is then cooled and diluted with an organic solvent (such as, for example, EtOAc).
  • an organic solvent such as, for example, EtOAc
  • the organic phase is washed sequentially with dilute aqueous acid (such as 1 NHCl), and with water, dried (for example, over MgSO 4 ) and concentrated.
  • dilute aqueous acid such as 1 NHCl
  • water dried (for example, over MgSO 4 ) and concentrated.
  • the desired hydantoin is purified, as by silica gel chromatography or by recrystallization.
  • Method J is exemplified by the synthesis of the compound of Example 148 (see Table 10), which was carried out is as follows.
  • the starting material (0.11 g, 0.40 mmol) was dissolved in THF (5.0 mL) and cooled in a dry-ice/acetone bath (approximately ⁇ 78° C.).
  • Lithium bis(trimethylsilyl) amide (LiHMDS, 405.0 ⁇ L, 0.40 mmol) was added dropwise.
  • the resultant yellow solution was stirred in the cold bath for 15 minutes, at which point 2-fluorobenzyl bromide was added to it.
  • the mixture was stirred at this temperature for an additional 30 minutes and then at 0° C. for 30 minutes.
  • hydantoins can be C-alkylated with alkyl halides using magnesium methyl carbonate (MMC).
  • MMC magnesium methyl carbonate
  • a solution of MMC in an organic solvent (such as DMF) is saturated at about 80° C. with CO 2 over a period of about 1 h.
  • An appropriate hydantoin is then added and heated with the MMC for about 1 to 2 h, at which point an appropriate alkyl halide is added.
  • the reaction mixture is then warmed to about 110° C. for between about 2 to 3 h, then cooled to about room temperature.
  • the mixture is then poured into concentrated aqueous acid (such as HCl) over ice and cooled.
  • the solid formed is collected by filtration and purified by silica gel chromatography and/or via recrystallization to afford the desired product.
  • the compound listed in the Table 11 was produced via this method.
  • Method K is exemplified by the synthesis of the compound of Example 181 (see Table 11), which was carried out as follows: A dry two-necked round bottom-flask was evacuated and charged with a CO 2 atmosphere. Magnesium methyl carbonate in DMF (860 ⁇ L, 2.0 M) was added to the flask and the solution was heated to 80° C. CO 2 was introduced from a dry-ice vessel via a cannula connected to the reaction vessel and was bubbled through the solution for 1.0 h at which point an argon line was attached and the cannula was removed. The starting material (0.21 g, 0.86 mmol) in DMF (4.0 mL) was added and the reaction mixture was heated at 80° C. for 1.5 h.
  • An appropriately substituted arylboronic acid or arylstanane is mixed with an aryl halide or aryl triflate and a catalytic amount of tetrakis(triphenylphosphine)palladium in an appropriate solvent system (such as benzene containing ethanol and aqueous Na 2 CO 3 , DMF, NMP or THF) under an inert atmosphere.
  • an appropriate solvent system such as benzene containing ethanol and aqueous Na 2 CO 3 , DMF, NMP or THF
  • Other components such as, for example LiCl and triethylamine, may be added as necessary.
  • the mixture is heated at between about 50 and 150° C. for between about 2 and 48 h.
  • the mixture is next cooled and diluted with an organic solvent (such as EtOAc).
  • EtOAc organic solvent
  • the organic phase was washed successively with water and saturated aqueous NaCl, dried (as with Na 2 SO 4 ) and concentrated
  • An appropriately substituted arylboronic acid or arylstanane is mixed with an aryl halide or aryl triflate and a catalytic amount of tetrakis (triphenylphosphine) palladium in an appropriate solvent system (such as benzene containing ethanol and aqueous Na 2 CO 3 , DMF, NMP or THF) under an atmosphere of carbon monoxide.
  • an appropriate solvent system such as benzene containing ethanol and aqueous Na 2 CO 3 , DMF, NMP or THF
  • Other components such as, for example LiCl and triethylamine, may be added as necessary.
  • the mixture is heated at between about 50 and 150° C. for between about 2 and 48 h.
  • the mixture is next cooled and diluted with an organic solvent (such as EtOAc).
  • EtOAc organic solvent
  • the organic phase was washed successively with water and saturated aqueous NaCl, dried (as with Na 2 SO 4 )
  • Method M is exemplified by the synthesis of the compound of Example 215 (see Table 13) which was carried out as follows: the starting material (0.23 g, 0.53 mmol) was mixed with phenyltributylstannane (0.86 mL, 2.64 mmol), bistriphenylphosphine-palladium (II) chloride (0.037 g, 0.05 mmol), DMF (10.0 mL), and LiCl (5.1 mg, 1.6 mmol), the reaction mixture was then purged with argon, charged with CO and stirred at 115° C. for 12 h.
  • Compounds containing aromatic rings can be modified by numerous reagents via electrophilic aromatic substitution. These include techniques for acylation, nitration, sulfonation and halogenation of these rings.
  • the compound listed in Table 14 was produced via this method.
  • the compound of Example 217 is one such case and was prepared by the following procedure.
  • a solution of the starting material (0.4 g, 1.1 mmol) in 1 mL of HOAc and 0.1 mL of H 2 S 4 was treated with NaIO 3 (0.05 g, 0.2 mmol) and I 2 (0.06 g, 0.5 mmol).
  • the mixture was then heated to 70° C. for 19 h after which point it was cooled to ambient temperature, extracted several times into EtOAc.
  • the EtOAc was concentrated and the product was isolated after purification by silica gel chromatography. Yield: 33 mg (30%)
  • Compounds having acid-labile protecting groups may be deprotected by treatment under acidic conditions, in a known per se manner. Generally this involves treating the substrate with TFA, cation exchange resin (H+), HCL or HBr in AcOH with or without heating. The compound thus formed is collected by filtration or extraction and purified, as by silica gel chromatography or recrystallization, to afford the desired product.
  • Method O is exemplified by the synthesis of the compound of Example 219, which was carried out as follows: A stirred solution of the starting material (0.10 g, 0.19 mmol) in 10 mL of methylene chloride was chilled to 0° C. before adding 2.0 mL of trifluoroacetic acid. Stirring at 0° C. was continued for 20 min and then the solution was allowed to warm slowly to ambient temperature. The solution was stirred for an additional 6 hat which point it was concentrated yielding an off-white solid which was further dried under vacuum for 16 h. The crude solid was next triturated with 10 mL of boiling hexanes and the mixture was allowed to cool to ambient temperature. The resulting white precipitate was collected via filtration, washed with 5 mL of hexanes and dried under high vacuum for 4 h to afford 0.06 g (68% yield) of the compound from example 219.
  • Certain compounds having carboxylic esters may be converted to carboxylic acids by treatment with saponifying reagents, in a known per se manner. Generally this involves treating the substrate with NaOH, KOH or LiOH in a solvent such as H 2 O sometimes containing a solubilizing agent such as THF. Purification generally involves extracting the unreacted starting material with an organic solvent such as EtOAc or CH 2 Cl 2 , acidification of the aqueous layer and purification of the acid by filtration or extraction into an organic solvent such as EtOAc or CH 2 Cl 2 . Further purification can be performed using recrystallization, silica gel chromatography or reverse-phase HPLC, to afford the desired product.
  • saponifying reagents in a known per se manner. Generally this involves treating the substrate with NaOH, KOH or LiOH in a solvent such as H 2 O sometimes containing a solubilizing agent such as THF.
  • Purification generally involves extracting the unreacted starting material with an organic solvent such as EtOAc or CH 2
  • the compound listed in Table 16 was produced via this general method.
  • Example 239 (table 16) was prepared by dissolving the starting material (0.38 g, 0.65 mmol) in 4 mL of H2O and 8 mL of MeOH containing LiOH (0.08 g, 1.95 mmol) and heating the mixture at 60° C. for 2.5 h. The MeOH was removed by concentration and the aqueous residue treated with 1 N HCl. The product was extracted into EtOAc from which it crystallized upon cooling. Yield 262 mg (72%).
  • Primary amines can be protected as their pthalimide derivatives. These derivatives are rapidly synthesized via method U using the potassium salt of pthalimide as the nucleophile.
  • the amine can be liberated from the pthalimide protecting group using nucleophilic reagents such as hydrazine or methyl amine in a solvent such as EtOH.
  • Purification generally involves acidification of the aqueous layer and extracting the unreacted starting material with an organic solvent such as EtOAc or CH 2 Cl 2 Basification of the aqueous layer produces the free base of the amine which is purified by filtration or extraction into an organic solvent such as EtOAc or CH 2 Cl 2 . Further purification can be performed using recrystallization, silica gel chromatography or reverse-phase HPLC, to afford the desired product.
  • the compound of Example 240 was prepared by dissolving the starting material (prepare via method U, 0.72 g, 1.2 mmol) in 73 mL of EtOH and treating it with 19.5 mL of a 33% solution of MeNH 2 in EtOH. The mixture was heated under reflux for 2.5 h and then cooled to ambient temperature. The reaction mixture was concentrated and the residue was dissolved in CH 2 Cl 2 which was further washed with H 2 O and saturated aqueous NaCl. The organic layer was dried (Na 2 SO 4 ) and concentrated. The yellow oil was dissolved in EtOH and treated with HCl gas. The amine hydrochloride of the compound of Example 240 was obtained in 69% yield (0.49 g).
  • Aromatic nitrites can be converted into amidine groups by several methods. Generally this conversion requires a two step process wherein the first step involves treatment with acid (such as, for example HCl) and an alcohol (such as, for example MeOH or EtOH) to generate an intermediate imino ether. This derivative is then converted to the amidine via treatment with an amine. Purification is usually be way of recrystallization of a derivative salt of the amidine. Further purification can be performed using recrystallization, silica gel chromatography or reverse-phase HPLC, to afford the desired product.
  • acid such as, for example HCl
  • alcohol such as, for example MeOH or EtOH
  • the compound of example 243 was prepared by dissolving the starting material (0.2 g, 0.4 mmol) in 7 mL of EtOH, cooling the mixture in an ice bath, and treating the mixture with dry HCl gas for 15 min. The mixture was stirred at room temperature for 1 h and concentrated to yield the crude imino ether hydrochloride. The intermediate was dissolved s in EtOH (10 mL), cooled in an ice bath and treated with anhydrous NH3 gas for 20 min. After 5 h, the reaction mixture was concentrated to provide the crude amidine hydrochloride. This material was purified via silica gel chromatography (1:9 MeOH:CH 2 Cl 2 ) to yield 0.08 g (38%) of the product.
  • Certain compounds having carboxylic acids may be converted to alcohols by treatment with reducing reagents, in a known per se manner. Generally this involves treating the substrate with LiAlH 4 or a BH 3 -based reagent in a solvent such as THF or ether. After careful quenching with an aqueous system, purification generally involves extracting the product into organic solvent such as EtOAc or CH 2 Cl 2 and purification using recrystallization, silica gel chromatography or reverse-phase HPLC, to afford the desired product.
  • organic solvent such as EtOAc or CH 2 Cl 2
  • the compound of example 244 was prepared by dissolving the starting material (prepare via methods J and O, 0.16 g, 0.32 mmol) in 1 mL of THF, cooling the mixture in an ice bath and treating it with 0.65 mL of a 1 M BH 3 -THF solution (0.65 mmol). The mixture was allowed to warm to ambient temperature and stir for 15 h. The reaction mixture was quenched by the slow and careful addition of water and the organic components were extracted into EtOAc. The EtOAc layer was washed with water, saturated aqueous NaCl and dried over Na 2 SO 4 . concentration and silica gel chromatography (1:1 EtOAc:Hexanes) produced the desired compound (0.06 g, 42%).
  • Certain compounds having methoxy protecting groups may be deprotected to the hydroxy derivative by treatment with certain nucleophilic reagents, in a known per se manner. Generally this involves treating the substrate with BBr 3 or TMSI in a solvent such as CH 2 Cl 2 , generally cooled in an ice bath and followed with or without heating. After between about 10 min and 8 h the reaction is quenched with a weak base such as aqueous NaHCO 3 and the organic component extracted into a solvent such as EtOAc and purified after concentration, as by silica gel chromatography or recrystallization, to afford the desired product.
  • a weak base such as aqueous NaHCO 3
  • Method T is exemplified by the synthesis of the compound of Example 246, which was carried out as follows: A stirred solution of the starting material (0.35 g, 0.64 mmol) in 9 mL of CH 2 Cl 2 was chilled to 0° C. before adding 1.0 mL of BBr 3 (1.07 mmol, 1 M CH 2 Cl 2 ). Stirring was continued and then the solution was allowed to warm slowly to ambient temperature. The solution was stirred for an additional 4 h at which point it saturated aqueous NaHCO 3 was added, the organic layer was removed and concentrated yielding the crude product. Purification was performed by silica gel chromatography (1:4 EtOAc:Hexanes) yielding 0.16 g (48% yield) of the desired compound.
  • An appropriate electrophilic agent is dissolved in an aprotic solvent (such as DMF, THF or DMSO) and treated with one to three equivalents of a nucleophile (such as Me 3 N, Na salt of imidazole, Na2SO 3 , NaCN, P(OEt) 3 , or the K salt of Pthalimide at between about room temperature and 100° C.
  • a nucleophile such as Me 3 N, Na salt of imidazole, Na2SO 3 , NaCN, P(OEt) 3 , or the K salt of Pthalimide
  • the mixture stirred at between about 0 and 100° C. for up to about 24 h. (Progress of the reaction can be monitored using TLC).
  • the solution is then cooled and diluted with an organic solvent (such as, for example, EtOAc).
  • the organic phase is washed sequentially with dilute aqueous acid (such as 1 N HCl), and with water, dried (for example, over MgSO 4 ) and concentrated.
  • dilute aqueous acid such as 1 N HCl
  • water dried (for example, over MgSO 4 ) and concentrated.
  • the desired compound is purified, as by silica gel chromatography, reverse-phase HPLC or by recrystallization.
  • Method U is exemplified by the synthesis of the compound of Example 262, which was carried out as follows: To the starting material (0.28 g, 0.54 mmol) was added 1-H-pyrazolecarboxamidine (0.08 g, 0.54 mmol) followed by 7 mL of DMF and 0.2 mL of N,N-diisopropyl-ethylamine. The resulting mixture was then stirred 15 h at room temperature. Next was added ether which caused the mixture to become turbid. As no crystals were forming, MeOH was added to re-dissolve the reaction components and the product was precipitated as its HCl salt by the addition of 1 N HCl. The solid was collected and washed with ether to yield 0.11 g (20%) of the guanidine hydrochloride.
  • This assay protocol is designed to study the direct antagonism, by a test compound, of the interaction of the CAM, ICAM-1 with the Leukointegrin CD18/CD11a (LFA-1).
  • LFA-1 is immunopurified using the TS2/4 antibody from a 20 g pellet of human JY or SKW3 cells, utilizing a protocol previously described (Dustin, M. J.; et al., J. Immunol. 1992, 148, 2654-2660).
  • the LFA-1 is purified from SKW3 lysates by immunoaffinity chromatography on TS2/4 LFA-1 mAb Sepharose and eluted at pH 11.5 in the presence of 2 mM MgCl 2 and 1% octylglucoside. After collection and neutralization of fractions from the TS2/4 column, samples are pooled and precleared with Protein G agarose.
  • ICAM-1 A soluble form of ICAM-1 is constructed, expressed, purified and characterized as previously described (Marlin, S.; et al., Nature, 1990, 344, 70-72 and see Arruda, A.; et al., Antimicrob. Agents Chemother. 1992, 36, 1186-1192). Briefly, isoleucine 454 which is located at the putative boundary between domain 5 of the ectodomain and the transmembrane domain, is changed to a stop codon using standard oligonucleotide-directed mutagenesis. This construction yields a molecule identical with the first 453 amino acids of membrane bound ICAM-1.
  • An expression vector is created with a hamster dihydrofolate reductase gene, a neomycin-resistance marker, and the coding region of the sICAM-1 construct described above, along with the promotor, splice signals, and polyadenylation signal of the SV40 early region.
  • the recombinant plasmid is transfected into CHO DUX cells using standard calcium phosphate methods. Cells are passaged in selective media (G418) and colonies secreting sICAM-1 are amplified using methotrexate.
  • sICAM-1 is purified from serum-free media using traditional non-affinity chromatographic techniques, including ion exchange and size exclusion chromatography.
  • LFA-1 binding to ICAM-1 is monitored by first incubating sICAM-1 at 40 ⁇ g/mL in Dulbecco's phosphate buffered saline with calcium and magnesium, additional 2 mM MgCl 2 and 0.1 mM PMSF (Diluting Buffer) in a 96-well plate for 30 amin at room temperature. Plates are then blocked by the addition of 2% (w/v) bovine serum albumin in Diluting Buffer for 37° C. for 1 h. Blocking solution is removed from wells, and test compounds are diluted and then added followed by the addition of approximately 25 ng of immunoaffinity purified LFA-1. The LFA-1 is incubated in the presence of test compound and ICAM-1 at 37° C.
  • This reagent is allowed to incubate for 20 min at 37° C., wells are washed as above and the substrate for the horse radish peroxidase is added to each well to develop a quantitative colorimetric signal proportional to the amount of LFA-1 bound to sICAM-1.
  • Soluble ICAM-1 60 ⁇ g/mL is used as a positive control for inhibition of the LFA-1/ICAM-1 interaction.
  • the lack of the addition of LFA-1 to the binding assay is used as a background control for all samples. A dose-response curve is obtained for all test compounds.
  • MTT assay In order to obtain meaningful data from cellular assays, compounds must first be first tested in an assay to measure cellular toxicity.
  • the MTT assay can be used for this purpose.
  • MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide), is a yellow substrate that is cleaved by cells with active mitochondria to yield a dark blue/purple formazan product.
  • This precipitate can be solublized and the amount of material quantitatified via spectrophotometric procedures (Gerlier, D.; Thomasset, N. J. Immunol. Methods, 1986, 94, 57-63). The amount of color is proportional to the number of viable cells.
  • This assay system is used to assess the effect of test compounds on cell viability in vitro.
  • SKW3 cells which express LFA-1, are used.
  • Cells used in each assay were adjusted to 1.25 ⁇ 106 cells/mL and 100 ⁇ L of this stock is dispersed into each well of a 96 well, flat bottom microtiter plate.
  • triplicate wells are set up. Serial dilutions of each test compound or vehicle alone are added to each well. Cells are incubated with compound for 4-24 h at 37° C. before cell viability is assessed. Next, 10 ⁇ L of filter sterilized MTT is added to each well. The MTT stock is made in phosphate buffered saline at a concentration of 5 mg/mL. Plates are then incubated for 1 h at 37° C., 5% CO 2 atmosphere. Periodically, the plates are examined for formazan crystal development.
  • This assay is used to test the selective ability of a test compound to antagonize the interaction of a cell bound form of LFA-1 with ICAM-1.
  • the assay uses a human T cell line, SKW3 cells which express CD18, CD11a and other integrins not related to CD18, CD11a and which can be “activated” by phorbol esters. Phorbol esters enhance the affinity of CD18, CD11a for ICAM-1.
  • sICAM-1 (40 ⁇ g/mL) or fibronectin (100 ⁇ g/mL) in Diluting buffer for 1 h at room temperature.
  • Added to the wells are 100 ⁇ L of the appropriately diluted test compound or 100 ⁇ L of RPMI with 15% fetal bovine serum as a control.
  • SKW3 cells which express CD18, CD11a and VLA4 (Dustin, M.; et al., J. Exp. Med. 1987, 165, 672-692) are washed and suspended to a concentration of 10 6 cells/mL in RPMI with 15% fetal bovine serum.
  • cells are stimulated with the phorbol ester 12-myristate 13-acetate (PMA) for a final concentration of 100 ⁇ g/mL. 100 ⁇ L of cells are then added to the wells resulting in a final concentration of 50 ⁇ g/mL PMA and 2 cells/well. The plates are incubated for 2 h at 37° C. Unbound or loosely bound cells are gently washed away with RPMI. Cells remaining and hence bound to ICAM-1 or to fibronectin are quantitated by the same reagent used above for the MTT experiment.
  • PMA phorbol ester 12-myristate 13-acetate
  • Epstein-Barr virus-transformed cells exhibit aggregation. This aggregation can be enhanced by the addition of phorbol esters. Such homotypic aggregation (i.e., aggregation involving only one cell type) was found to be blocked by anti-LFA-1 antibodies (Rothlein, R. R.; et al., J. Exp. Med. 1986, 163, 1132-1149). Thus, the extent of LFA-1-dependent binding may be determined by assessing the extent of spontaneous or phorbol ester-dependent aggregate formation.
  • An agent which interferes with LFA-1 -dependent aggregation can be identified through the use of an assay capable of determining whether the agent interferes with either spontaneous, or phorbol ester-dependent aggregation of Epstein-Barr virus transformed cells. It is preferable to employ cells of the JY cell line (Terhost, L; et aL, Proc. Natl. Acad. Sci. USA, 1976, 73, 910) for the homotypic aggregation assay.
  • This assay capable of measuring LFA-1 dependent aggregation, may be employed to identify agents which act as antagonists to the LFA-1 dependent aggregation. Such agents may act by impairing the ability of either LFA-1 or ICAM-1 to mediate aggregation. Thus, agents may be examined to directly determine if they are antagonists of LFA-1 aggregation.
  • JY cells are cultured in RPMI 1640 culture medium supplemented with 10% fetal calf serum and 50 ⁇ g/mL gentamycin. The cells are cultured at 37° C. in an atmosphere of 5% CO 2 at a relative humidity of 95%. JY cells used in this assay are washed two times with RPMI 1640 medium containing 5 mM HEPES buffer and resuspended to a concentration of 2 ⁇ 10 6 cells/mL.
  • 96-well microtiter plates are 50 ⁇ L of test compound diluted in complete medium, 50 ⁇ L of complete medium with or without purified monoclonal antibodies (negative and positive controls for inhibition, respectively), 50 ⁇ L of complete medium containing 200 ng/mL of the phorbol ester phorbol myristate acetate (PMA) and 100 ⁇ L of cells at a concentration of 2 ⁇ 10 6 cells/mL in complete medium. This yields a final concentration of 50 ng/nL PMA and 2 ⁇ 10 5 cells/well. Cells are allowed to settle spontaneously, and the degree of aggregation is scored at various time points.
  • PMA phorbol ester phorbol myristate acetate
  • Scores range from 0 to 4 where 0 indicates that essentially no cells are in clusters; 1 indicates that ⁇ 25% of the cells are in clusters; 2 indicates that ⁇ 50% of the cells are in clusters; 3 indicates that ⁇ 75% of the cells are in clusters and 4 indicates that 100% of the cells are aggregated.
  • This procedure has been described by Rothlein, R. R.; et al., J. Exp. Med. 1986, 163, 1132-1149.
  • This paper also reported that antibody to LFA-1 is capable of inhibiting the formation of aggregates. Whereas 100% of the cells form aggregates in the absence of LFA-1 antibody, less than 20% of the cells were found to be in aggregates when anti-LFA-1 antibody was added in the same paper.
  • ICAM-1 is necessary for effective cellular interactions during an immune response mediated through LFA-1-dependent cell adhesion.
  • lymphocytes from two unrelated individuals are cultured together, blast transformation and cell proliferation of the lymphocytes are observed.
  • This response is known as a mixed lymphocyte reaction (MLR) and is analogous to the response of lymphocytes to the 10 addition of antigens or mitogens (Immunology: The Science of Self-Nonself Discrimination; Klein, J., Ed.; John Wiley & Sons: NY, 1982, pp 453-458).
  • MLR mixed lymphocyte reaction
  • Monoclonal antibodies directed against ICAM-1 and LFA-1 were used as controls to demonstrate inhibition of cell adhesion-dependent lymphocyte stimulation and proliferation.
  • This assay protocol is used to to determine the effect of a test compound on the human MLR.
  • the ability of a test compound to inhibit the MLR and antigen-specific mononuclear cell responses shows that it has therapeutic utility in acute graft rejection., as well as in related immune mediated disorders dependent on CD18, CD11a/ICAM interactions.
  • Peripheral blood is obtained from normal, healthy donors by venipuncture.
  • the blood is collected in heparinized tubes and diluted 1:1 at room temperature with Puck's G (GIBCO) balanced salt solution (BSS).
  • GIS Puck's G
  • BSS balanced salt solution
  • RPMI 1640 culture medium containing 0.5% of gentamicin, 1 mM L-glutamine (GICO) and 5% heat inactivated (56° C., 30 min) human AB sera (Flow Laboratories) (hereafter referred to as RPMI-culture medium).
  • PBMC Peripheral blood mononuclear cells
  • Stimulator cells cells that have been treated with irradiation so that they are unable to proliferate
  • Test compound is added to wells at various concentrations.
  • the total volume per culture is 0.2 mL.
  • Controls include compound vehicle alone (DMSO), responder cells alone, and stimulator cells alone.
  • the culture plates are incubated at 37° C. in a 5% CO 2 -humidified air atmosphere for 5 days.
  • the wells are pulsed with 0.5 ⁇ Ci of tritiated thymidine ( 3 HT) (New England Nuclear) for the last 18 h of culture. In some cases a two-way MLR can be performed.
  • the protocol is the same except that the second donor's cells are not inactivated by irradiation.
  • the cells are harvested onto glass fiber filters using an automated multiple sample harvester (Skatron, Norway), and rinsed with water and methanol.
  • the filters are oven dried and counted in Aquasol in a Beckman (LS-3801) liquid scintillation counter.
  • the ability of cells to recognize other cells from self or from another genetically different individual (non-self) is an important property in maintaining the integrity of tissue and organ structure.
  • the allogeneic cell transplant response is an important model for studies of transplant rejection and immunocompetence.
  • This T-cell-mediated immune response can be induced in adult mice by the injection of lymphocytes from a histoincompatible mouse strain into the footpad .
  • This response is characterized by T-cell proliferation which is limited to the popliteal lymph node that receives drainage from the injected footpad area. No in vitro system can completely duplicate this in vivo response.
  • this animal model can be used to assess the ability of a test compound to suppress transplant rejection.
  • mice Male or female mice (20-26 grams). Any histoincompatible mouse strains suffice for donor and recipient populations. Typically DBA mice are used as donors and C57 1/6 mice are used as recipients. A minimum of 1 week stabilization and conditioning period is required before use during which time the animals are maintained in accordance with the Animal Resource Center S.O.P. Each study utilizes 36 recipient mice divided into groups of 6. The tests last approximately four days. Donor mice are sacrificed by CO 2 asphyxiation and spleens are removed and made into a cell suspension. The cells (1.0 ⁇ 10 7 /metatarsal in 0.05 mL) are injected intra dermal (according to standard protocol) into the dorsal metatarsal skin of recipient mice.
  • mice Four days later, the animals are sacrificed by CO 2 asphyxiation and the popliteal nodes are removed and weighed. Groups of mice receiving putative immunosuppressive agents are dosed subcutaneously, intraperitoneally or orally one hour prior to cell injection and daily thereafter according to standard protocol. Student's T-test was used to determine significant differences between popliteal lymph nodes of groups of untreated mice and those mice treated with putative immunosuppressive agents (see: Kroczek, R. A.; Black, C. D. V.; Barbet, J.; Shevach, E. M., J. Immunology, 1987, 139, 3597).
  • novel and known small molecules utilized in the method according to the invention inhibit the ICAM-1/LFA-1 dependent homotypic aggregation of human lymphocytes, human lymphocyte adherence to ICAM-1 and human lymphocyte responses to antigens.
  • These compounds have therapeutic utility in the modulation of immune cell activation/proliferation, e.g., as competitive inhibitors of intercellular ligand/receptor binding reactions involving CAMs and Leukointegrins.
  • Inflammatory conditions which may be treated with the compounds comprehended by the invention include conditions resulting from a response of the non-specific immune system in a mammal (e.g., adult respiratory distress syndrome, shock, oxygen toxicity, multiple organ injury syndrome secondary to septicemia, multiple organ injury syndrome secondary to trauma, reperfusion injury of tissue due to cardiopulmonary bypass, myocardial infarction or use with thrombolysis agents, acute glomerulonephritis, vasculitis, reactive arthritis, dermatosis with acute inflammatory components, stroke, thermal injury, hemodialysis, leukapheresis, ulcerative colitis, necrotizing enterocolitis and granulocyte transfusion associated syndrome) and conditions resulting from a response of the specific immune system in a mammal (e.g., psoriasis, organ/tissue transplant rejection, graft vs.
  • a mammal e.g., adult respiratory distress syndrome, shock, oxygen toxicity, multiple organ injury syndrome secondary to septicemia, multiple
  • these novel and known compounds may also be used in treating asthma or as an adjunct to minimize toxicity with cytokine therapy in the treatment of cancers. In general these compounds may be employed in the treatment of those diseases currently treatable through steroid therapy.
  • these novel and known compounds may be administered for either a “prophylactic” or “therapeutic” purpose either alone or with other immunosuppressive or antiinflammatory agents.
  • the immunosuppressive compound(s) are provided in advance of any inflammatory response or symptom (for example, prior to, at, or shortly after the time of an organ or tissue transplant but in advance of any symptoms of organ rejection).
  • the prophylactic administration of a compound of the formula I serves to prevent or attenuate any subsequent inflammatory response (such as, for example, rejection of a transplanted organ or tissue, etc.).
  • the therapeutic administration of a compound of the formula I serves to attenuate any actual inflammation (such as, for example, the rejection of a transplanted organ or tissue).
  • a compound of the formula I can be administered either prior to the onset of inflammation (so as to suppress an anticipated inflammation) or after the initiation of inflammation.
  • novel and known compounds of the formula I may, in accordance with the invention, be administered in single or divided doses by the oral, parenteral or topical routes.
  • a suitable oral dosage for a compound of formula I would be in the range of about 0.5 mg to 1 g per day.
  • a suitable dosage unit may contain from 0.1 to 250 mg of said compounds, whereas for topical administration, formulations containing 0.01 to 1% active ingredient are preferred. It should be understood, however, that the dosage administration from patient to patient will vary and the dosage for any particular patient will depend upon the clinician's judgement, who will use as criteria for fixing a proper dosage the size and condition of the patient as well as the patient's response to the drug.
  • the compounds of the present invention When the compounds of the present invention are to be administered by the oral route, they may be administered as medicaments in the form of pharmaceutical preparations which contain them in association with a compatible pharmaceutical carrier material.
  • a compatible pharmaceutical carrier material can be an inert organic or inorganic carrier material suitable for oral administration. Examples of such carrier materials are water, gelatin, talc, starch, magnesium stearate, gum arabic, vegetable oils, polyalkylene-glycols, petroleum jelly and the like.
  • the pharmaceutical preparations can be prepared in a conventional manner and finished dosage forms can be solid dosage forms, for example, tablets, dragees, capsules, and the like, or liquid dosage forms, for example solutions, suspensions, emulsions and the like.
  • the pharmaceutical preparations may be subjected to conventional pharmaceutical operations such as sterilization. Further, the pharmaceutical preparations may contain conventional adjuvants such as preservatives, stabilizers, emulsifiers, flavor-improvers, wetting agents, buffers, salts for varying the osmotic pressure and the like.
  • Solid carrier material which can be used include, for example, starch, lactose, mannitol, methyl cellulose, microcrystalline cellulose, talc, silica, dibasic calcium phosphate, and high molecular weight polymers (such as polyethylene glycol).
  • a compound of formula I can be administered in an aqueous or non-aqueous solution, suspension or emulsion in a pharmaceutically acceptable oil or a mixture of liquids, which may contain bacteriostatic agents, antioxidants, preservatives, buffers or other solutes to render the solution isotonic with the blood, thickening agents, suspending agents or other pharmaceutically acceptable additives.
  • Additives of this type include, for example, tartrate, citrate and acetate buffers, ethanol, propylene glycol, polyethylene glycol, complex formers (such as EDTA), antioxidants (such as sodium bisulfite, sodium metabisulfite, and ascorbic acid), high molecular weight polymers (such as liquid polyethylene oxides) for viscosity regulation and polyethylene derivatives of sorbitol anhydrides.
  • complex formers such as EDTA
  • antioxidants such as sodium bisulfite, sodium metabisulfite, and ascorbic acid
  • high molecular weight polymers such as liquid polyethylene oxides for viscosity regulation and polyethylene derivatives of sorbitol anhydrides.
  • Preservatives may also be added if necessary, such as benzoic acid, methyl or propyl paraben, benzalkonium chloride and other quaternary ammonium compounds.
  • the compounds of this invention may also be administered as solutions for nasal application and may contain in addition to the compounds of this invention suitable buffers, tonicity adjusters, microbial preservatives, antioxidants and viscosity-increasing agents in an aqueous vehicle.
  • suitable buffers tonicity adjusters
  • microbial preservatives antioxidants
  • viscosity-increasing agents in an aqueous vehicle.
  • agents used to increase viscosity are polyvinyl alcohol, cellulose derivatives, polyvinylpyrrolidone, polysorbates or glycerin.
  • Microbial preservatives added may include benzalkonium chloride, thimerosal, chloro-butanol or phenylethyl alcohol.
  • the compounds provided by the invention can be administered by suppository.
  • Example A-1 Compound of 250 mg formula I Starch 160 mg Microcrys. Cellulose 90 mg Sodium Starch 10 mg Glycolate Magnesium Stearate 2 mg Fumed colloidal silica 1 mg
  • Example A-2 Compound of 50 mg formula I Dicalcium Phosphate 160 mg Microcrys. Cellulose 90 mg Stearic acid 5 mg Sodium Starch 10 mg Glycolate Fumed colloidal silica 1 mg
  • the compound of formula I is blended into a powder mixture with the premixed excipient materials as identified above with the exception of the lubricant.
  • the lubricant is then blended in and the resulting blend compressed into tablets or filled into hard gelatin capsules.
  • excipient materials are mixed and then added to one of the compounds of formula I in such volume as is necessary for dissolution. Mixing is continued until the solution is clear. The solution then filtered into the appropriate vials or ampoules and sterilized by autoclaving.
  • the excipient materials are mixed with the water and thereafter one of the compounds of formula I is added and mixing is continued until the suspension is homogeneous. The suspension is then transferred into the appropriate vials or ampoules.

Abstract

A method treating or preventing inflammatory and immune cell-mediated diseases by the administration of certain novel and known small molecules. Exemplary of the novel compounds are those of the following structural formulas:
Figure USRE038132-20030603-C00001
Figure USRE038132-20030603-C00002

Description

RELATED APPLICATIONS
This application is a continuation-in-part of International Application No. PCT/US98/04254, filed Mar. 3, 1998, and U.S. application Ser. No. 09/033,148, filed Mar. 2, 1998 now abandoned. The benefit of prior provisional application Ser. No. 60/040,011, filed on Mar. 3, 1997, is hereby claimed.
FIELD OF THE INVENTION
The present invention relates generally to a series of novel small molecules, their synthesis and their use in the treatment of inflammatory disease. The invention further relates to the use of similar, but known, compounds in the treatment of inflammatory disease.
BACKGROUND OF THE INVENTION
Research spanning the last decade has helped to elucidate the molecular events attending cell-cell interactions in the body, especially those events involved in the movement and activation of cells in the immune system. See generally, Springer, T. Nature, 1990, 346, 425-434. Cell surface proteins, and especially the Cellular Adhesion Molecules (“CAMs”) and “Leukointegrins”, including LFA-1, MAC-1 and gp150.95 (referred to in WHO nomenclature as CD18/CD11a, CD18/CD11b, and CD18/CD11c, respectively) have correspondingly been the subject of pharmaceutical research and development having as its goal the intervention in the processes of leukocyte extravasation to sites of injury and leukocyte movement to distinct targets. For example, it is presently believed that prior to the leukocyte extravasation, which is a mandatory component of the inflammatory response, activation of integrins constituitively expressed on leukocytes occurs and is followed by a tight ligand/receptor interaction between integrins (e.g., LFA-1) and one or several distinct intercellular adhesion molecules (ICAMs) designated ICAM-1, ICAM-2, ICAM-3 or ICAM-4 which are expressed on blood vessel endothelial cell surfaces and on other leukocytes. The interaction of the CAMs with the Leukointegrins is a vital step in the normal functioning of the immune system. Immune processes such as antigen presentation, T-cell mediated cytotoxicity and leukocyte extravasation all require cellular adhesion mediated by ICAMs interacting with the Leukointegrins. See generally Kishimoto, T. K.; Rothlein; R. R. Adv. Pharmacol. 1994,25:117-138 and Diamond, M.; Springer, T. Current Biology, 1994, 4, 506-532.
A group of individuals has been identified which lack the appropriate expression of Leukointegrins, a condition termed “Leukocyte Adhesion Deficiency” (Anderson, D. C.; et al., Fed. Proc. 1985, 44, 2671-2677 and Anderson, D. C.; et al., J. Infect. Dis. 1985, 152, 668-689). These individuals are unable to mount a normal inflammatory and/or immune response(s) due to an inability of their cells to adhere to cellular substrates. These data show that immune reactions are mitigated when lymphocytes are unable to adhere in a normal fashion due to the lack of functional adhesion molecules of the CD18 family. By virtue of the fact that LAD patients who lack CD18 cannot mount an inflammatory response, it is believed that antagonism of CD18, CD11 /ICAM-1 interactions will also inhibit an inflammatory response.
It has been demonstrated that the antagonism of the interaction between the CAMs and the Leukointegrins can be realized by agents directed against either component. Specifically, blocking of the CAMs, such as for example ICAM-1, or the Leukointegrins, such as for example LFA-1, by antibodies directed against either or both of these molecules effectively inhibits inflammatory responses. In vitro models of inflammation and immune response inhibited by antibodies to CAMs or Leukointegrins include antigen or mitogen-induced lymphocyte proliferation, homotypic aggregation of lymphocytes, T-cell mediated cytolysis and antigen-specific induced tolerance. The relevance of the in vitro studies are supported by in vivo studies with antibodies directed against ICAM-1 or LFA-1. For example, antibodies directed against LFA-1 can prevent thyroid graft rejection and prolong heart allograft survival in mice (Gorski, A.; Immunology Today, 1994, 15,251-255). Of greater significance, antibodies directed against ICAM-1 have shown efficacy in vivo as anti-inflammatory agents in human diseases such as renal allograft rejection and rheumatoid arthritis (Rothlein, R. R.; Scharschmidt, L., in: Adhesion Molecules; Wegner, C. D., Ed.; 1994, 1-38, Cosimi, C. B.; et al., J. Immunol. 1990, 144, 4604-4612 and Kavanaugh, A.; et al., Arthritis Rheum. 1994, 37, 992-1004) and antibodies directed against LFA-1 have demonstrated immunosuppressive effects in bone marrow transplantation and in the prevention of early rejection of renal allografts (Fischer, A.; et al., Lancet, 1989, 2, 1058-1060 and Le Mauff, B.; et al., Transplantation, 1991, 52, 291-295).
It has also been demonstrated that a recombinant soluble form of ICAM-1 can act as an inhibitor of the ICAM-1 interaction with LFA-1. Soluble ICAM-1 acts as a direct antagonist of CD18, CD11 /ICAM-1 interactions on cells and shows inhibitory activity in in vitro models of immune response such as the human mixed lymphocyte response, cytotoxic T cell responses and T cell proliferation from diabetic patients in response to islet cells (Becker, J. C.; et al., J. Immunol. 1993, 151, 7224 and Roep, B. O.; et al., Lancet, 1994, 343, 1590).
Thus, the prior art has demonstrated that large protein molecules which antagonize the binding of the CAMs to the Leukointegrins have therapeutic potential in mitigating inflammatory and immunological responses often associated with the pathogenesis of many autoimmune or inflammatory diseases. However proteins have significant deficiencies as therapeutic agents, including the inability to be delivered orally and potential immunoreactivity which limits the utility of theses molecules for chronic administration. Furthermore, protein-based therapeutics are generally expensive to produce.
It follows that small molecules having the similar ability as large protein molecules to antagonize the binding of the CAMs to the Leukointegrins would make preferable therapeutic agents. To date, however, no small molecules acting as direct antagonists have been reported.
Several small molecules have been described in the literature which affect the interaction of CAMs and Leukointegrins. A natural product isolated from the root of Trichilia rubra was found to be inhibitory in an in vitro cell binding assay (Musza, L. L.; et al., Tetrahedron, 1994, 50, 11369-11378). One series of molecules (Boschelli, D. H.; et al., J. Med. Chem. 1994, 37, 717 and Boschelli, D. H.; et al., J. Med. Chem. 1995, 38, 4597-4614) was found to be orally active in a reverse passive Arthus reaction, an induced model of inflammation that is characterized by neutrophil accumulation (Chang, Y. H.; et al., Eur. J. Pharmacol. 1992, 69, 155-164). Another series of molecules was also found to be orally active in a delayed type hypersensitivity reaction in rats (Sanfilippo, P. J.; et al., J. Med. Chem. 1995, 38, 1057-1059). All of these molecules appear to act nonspecifically, either by inhibiting the transcription of ICAM-1 along with other proteins or act intracellularly to inhibit the activation of the Leukointegrins by an unknown mechanism. None of the molecules directly antagonize the interaction of the CAMs with the Leukointegrins. Due to lack of potency, lack of selectivity and lack of a specific mechanism of action, the described small molecules are not likely to be satisfactory for therapeutic use.
Based on the status of the prior art, there remains a clear need for therapeutically useful small molecules having the ability to antagonize the interaction of CAMs and Leukointegrins.
SUMMARY OF THE INVENTION
A first aspect of the invention comprises a method for treating or preventing inflammatory and immune cell-mediated disease(s) by the administration of certain novel and known small molecules. These compounds act by inhibiting the interaction of cellular adhesion molecules, specifically by antagonizing the binding of human intercellular adhesion molecules (including, for example, ICAM-1, ICAM-2 and ICAM-3) to the Leukointegrins (including, for example, CD18/CD11a and CD18/CD11b). A second aspect of the invention comprises novel small molecules having the above-noted therapeutic activities. A third aspect of the invention comprises methods for making these novel compounds. A final aspect of the invention comprises pharmaceutical compositions comprising the above-mentioned compounds suitable for the prevention or treatment of inflammatory and immune cell-mediated condition(s).
DETAILED DESCRIPTION OF THE INVENTION
In its first aspect, the invention comprises a method for treating or preventing inflammatory and immune cell-mediated diseases by the administration of certain novel and known small molecules of the formula I
Figure USRE038132-20030603-C00003
wherein:
Y is an oxygen or sulfur atom;
Z is an oxygen or sulfur atom;
X is a divalent group of the formula >CHR1, >NR1, >CHSO2R1, or >NSO2R1, or an oxygen or sulfur atom, wherein R1 is:
(A) a hydrogen atom,
(B) branched or unbranched alkyl of 1 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, which alkyl or cycloakyl group may be mono- or polysubstituted with:
(i) halogen,
(ii) oxo,
(iii) aryl, which is selected from the class consisting of phenyl, naphthyl, indolyl, thiophenyl, pyridyl, pyrimidinyl, furyl, pyrrolyl, oxazolyl, thiazolyl, pyrazolyl, isoxazolyl, imidazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl, pyridazinyl, pyrazinyl, triazinyl, indolyzinyl, isoindolyl, benzo[b]furanyl, benzo[b]thiophenyl, indazolyl, benzthiazolyl, benzimidazolyl, quinolinyl, isoquinolinyl, purinyl, quinolizinyl, cinnolinyl, pthalaninyl, quinoxalinyl, napthyridinyl, pteridinyl and quinazolinyl, wherein one or more hydrogen atoms of said aryl group may be optionally and independently replaced with:
(a) alkyl of 1 to 3 carbon atoms,
(b) —COOH,
(c) —SO2OH,
(d) —PO(OH)2,
(e) a group of the formula —COOR7, wherein R7 is straight or branched alkyl of 1 to 5 carbon atoms or cycloalkyl of 3 to 5 carbon atoms,
(f) a group of the formula —NR8R9, wherein R8 and R9 are each independently a hydrogen atom, alkyl of 1 to 6 carbon atoms, cycloalkyl of 3 to 6 carbon atoms or acyl of 1 to 7 carbon atoms, or wherein R8 and R9 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring,
(g) a group of the formula —CONR10R11, wherein R10 and R11 are each independently a hydrogen atom, alkyl of 1 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, or wherein R10 and R11 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring,
(h) a group of the formula —OR12a, wherein R12a is a hydrogen atom, or an alkyl or acyl group of 1 to 7 carbon atoms,
(i) a group of the formula —SR12b, wherein R12b is a hydrogen atom, or an alkyl or acyl group of 1 to 7 carbon atoms,
(j) cyano, or
(k) an amidino group of the formula
Figure USRE038132-20030603-C00004
wherein R13, R14 and R15 are each, independently, a hydrogen atom or alkyl of 1 to 3 carbon atoms and wherein two of R13, R14 and R15 may additionally constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom(s) between them form a heterocyclic ring,
(iv) a group of the formula —COOR16, wherein R16 is straight or branched alkyl of 1 to 7 carbon atoms or cycloalkyl of 3 to 6 carbon atoms,
(v) cyano,
(vi) a group of the formula —CONR17R18, wherein R17 and R18 are each, independently, a hydrogen atom, alkyl of 1 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, or wherein R17 and R18 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring,
(vii) a group of the formula —OR19, wherein R19 is a hydrogen atom, or an alkyl or acyl group of 1 to 7 carbon atoms,
(viii) a group of the formula —SR20, wherein R20 is a hydrogen atom, or an allyl or acyl group of 1 to 7 carbon atoms,
(ix) a group of the formula —NR21R22, wherein R21 and R22 are each, independently,
(a) a hydrogen atom,
(b) alkyl or acyl of 1 to 7 carbon atoms or cycloalkyl of 3 to 7 carbon atoms,
(c) a group of the formula —CH2)mCOOH, wherein m is 0, 1 or 2, or
(d) a group of the formula —CH2)nCOOR23, wherein n is 0, 1 or 2, wherein R23 is straight or branched alkyl of 1 to 6 carbon atoms,
or wherein R21 and R22 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring, or
(x) a quaternary group of the formula
Figure USRE038132-20030603-C00005
wherein R24, R25 and R26 are each, independently, a branched or unbranched alkyl group of 1 to 7 carbon atoms and Q is a chlorine, bromine or iodine counterion,
(C) a branched or unbranched carboxylic acid group of 3 to 6 carbon atoms,
(D) a branched or unbranched phosphonic acid group of 2 to 6 carbon atoms,
(E) a branched or unbranched sulfonic acid group of 2 to 6 carbon atoms,
(F) an anilino group of the formula
Figure USRE038132-20030603-C00006
wherein r is 2, 3, 4, 5 or 6, and
R27, R28 and R29 are each, independently, a hydrogen atom or alkyl of I to 3 carbon atoms, and wherein two of R27, R28 and R29 may additionally constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom(s) between them form a heterocyclic ring,
(G) an guanidino group of the formula
Figure USRE038132-20030603-C00007
wherein s is 2, 3, 4, 5 or 6, and
R30, R31, R32 and R33 are each, independently, a hydrogen atom or alkyl of 1 to 3 carbon atoms, and wherein two of R30, R31, R32 and R33 may additionally constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom(s) between them form a heterocyclic ring,
(H) piperidyl, wherein the nitrogen atom of said group is optionally substituted with:
(i) alkyl of 1 to 3 carbon atoms,
(ii) a carboxylic ester group of 2 to 7 carbon atoms,
(iii) a carboxylic acid group of 2 to 5 carbon atoms,
(iv) a phosphonic acid group of 1 to 6 carbon atoms, or
(v) a sulfonic acid groups of 1 to 6 carbon atoms, or
(I) aryl which is selected from the class consisting of phenyl, naphthyl, indolyl, thiophenyl, pyridyl, pyrimidinyl, furyl, pyrrolyl, oxazolyl, thiazolyl, pyrazolyl, isoxazolyl, imidazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl, pyridazinyl, pyrazinyl, triazinyl, indolyzinyl, isoindolyl, benzo[b]furanyl, benzo[b]thiophenyl, indazolyl, benzthiazolyl, benzimidazolyl, quinolinyl, isoquinolinyl, purinyl, quinolizinyl, cinnolinyl, pthalaninyl, quinoxalinyl, napthyridinyl, pteridinyl and quinazolinyl, wherein one or more hydrogen atoms of said aryl group may be optionally and independently replaced with:
(i) alkyl of 1 to 3 carbon atoms,
(ii) —COOH,
(iii) —SO2OH,
(iv) —PO(OH)2,
(v) a group of the formula —COOR7, wherein R7 is straight or branched alkyl of 1 to 5 carbon atoms or cycloalkyl of 3 to 5 carbon atoms,
(vi) a group of the formula —NR8R9, wherein R8 and R9 are each, independently, a hydrogen atom, alkyl of 1 to 6 carbon atoms, cycloalkyl of 3 to 6 carbon atoms or acyl of 1 to 7 carbon atoms, or wherein R8 and R9 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring,
(vii) a group of the formula —CONR10R11, wherein R10 and R11 are each, independently, a hydrogen atom, alkyl of 1 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, or wherein R10 and R11 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring,
(viii) a group of the formula —OR12a, wherein R12a is a hydrogen atom, or an alkyl or acyl group of 1 to 7 carbon atoms,
(ix) a group of the formula —SR12b, wherein R12b is a hydrogen atom, or an alkyl or acyl group of 1 to 7 carbon atoms,
(x) cyano, or
(xi) an amidino group of the formula
Figure USRE038132-20030603-C00008
wherein R13, R14 and R15 are each, independently, a hydrogen atom or alkyl of 1 to 3 carbon atoms, and wherein two of R 13, R 14 and R15 may additionally constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom(s) between them form a heterocyclic ring;
R2 is:
(A) a hydrogen atom, or
(B) branched or unbranched alkyl of 1 to 3 carbon atoms or cycloalkyl of 3 to 5 carbon atoms wherein said alkyl or cycloalkyl group may optionally be substituted with:
(i) a group of the formula —OR34, wherein R34 is a hydrogen atom, or an alkyl or acyl group of 1 to 7 carbon atoms, or
(ii) a group of the formula —NR35R36, wherein R35 and R36 are each, independently, a hydrogen atom, alkyl of 1 to 2 carbon atoms, or acyl of 1 to 2 carbon atoms;
R3 is a group of the formula —(CR375l R38)x(CR39R40)yR41, wherein;
x and y are each independently 0 or 1,
R37, R38 and R39 are each, independently:
(A) a hydrogen atom,
(B) a group of the formula —OR42, wherein R42 is a hydrogen atom, or an alkyl or acyl group of 1 to 7 carbon atoms, or
(C) branched or unbranched alkyl of 1 to 3 carbon atoms or cycloalkyl of 3 to 5 carbon atoms,
R40 is:
(A) a hydrogen atom,
(B) a group of the formula —OR42, wherein R42 is a hydrogen atom, or an alkyl or acyl group of 1 to 7 carbon atoms,
(C) branched or unbranched alkyl of 1 to 3 carbon atoms or cycloalkyl of 3 to 5 carbon atoms, or
(D) aryl which is selected from the class consisting of phenyl, 2-naphthyl, 2-, 3-, 5- or 6-indolyl, 2- or 3-thiophenyl, 2-, 3- or 4-pyridyl, 2-, 4- or 5-pyrimidinyl, 2- or 3-furyl, 1-, 2- or 3-pyrrolyl, 2-, 4- or 5-oxazolyl, 2-, 4- or 5-thiazolyl, 1-, 3-, 4- or 5-pyrazolyl, 3-, 4- or 5-isoxazolyl, 1-, 2-, 4- or 5-imidazolyl, 3-, 4- or 5-isothiazolyl, 4- or 5-oxadiazolyl, 1-, 4- or 5-triazolyl, 2-thiadiazolyl, 3- or 4-pyridazinyl, 2-pyrazinyl, 2-triazinyl, 2-, -3,6-or 7-indolyzinyl, 2-, 3-, 5- or 6-isoindolyl, 2-, 3-, 5- or 6-benzo[b]furanyl, 2-, 3-, 5- or 6-benzo[b]thiophenyl, 3-, 5- or 6-indazolyl, 2-, 5- or 6-benzthiazolyl, 2-, 5- or 6-benzimidazolyl, 2-, 3-, 6- or 7-quinolinyl, 3-, 6- or 7-isoquinolinyl, 2- or 8-purinyl, 2-, 3-, 7- or 8-quinolizinyl, 3-, 6- or 7-cinnolinyl, 6- or 7-pthalaninyl, 2-, 3-, 6- or 7-quinoxalinyl, 2-, 3-, 6- or 7-napthyridinyl, 2-, 6- or 7-pteridinyl and 2-, 6- or 7-quinazolinyl,
wherein one or more of the hydrogen atoms of said aryl group may be optionally and independently replaced with:
(i) R43, which is aryl selected from the class consisting of phenyl, 2-naphthyl, 2-, 3-, 5- or 6-indolyl, 2- or 3-thiophenyl, 2-, 3- or 4-pyridyl, 2-, 4- or 5-pyrimidinyl, 2- or 3-furyl, 1-, 2- or 3-pyrrolyl, 2-, 4- or 5-oxazolyl, 2-, 4- or 5-thiazolyl, 1-, 3-, 4- or 5-pyrazolyl, 3-, 4- or 5-isoxazolyl, 1-, 2-, 4- or 5-midazolyl, 3-, 4- or 5-isothiazolyl, 4- or 5-oxadiazolyl, 1-, 4- or 5-triazolyl, 2-thiadiazolyl, 3- or 4-pyridazinyl, 2-pyrazinyl, 2-triazinyl, 2-, -3, 6- or 7-indolyzinyl, 2-, 3-, 5- or 6-isoindolyl, 2-, 3-, 5- or 6-benzo[b]furanyl, 2-, 3-, 5- or 6-benzo[b]thiophenyl, 3-, 5- or 6-indazolyl, 2-, 5- or 6-benzthiazolyl, 2-, 5- or 6-benzimidazolyl, 2-, 3-, 6- or 7-quinolinyl, 3-, 6- or 7-isoquinolinyl, 2- or 8-purinyl, 2-, 3-, 7- or 8-quinolizinyl, 3-, 6- or 7-cinnolinyl, 6- or 7-pthalaninyl, 2-, 3-, 6- or 7-quinoxalinyl, 2-, 3-, 6- or 7-napthyridinyl, 2-, 6- or 7-pteridinyl and 2-, 6- or 7-quinazolinyl, wherein one or more of the hydrogen atoms of said aryl group may be optionally and independently replaced with:
(a) branched or unbranched alkyl of 1 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, which alkyl or cycloakyl group may be mono- or polysubstituted with halogen or oxo,
(b) —COOH,
(c) —SO2OH,
(d) —PO(OH)2,
(e) a group of the formula —COOR44, wherein R44 is straight or branched alkyl of 1 to 5 carbon atoms or cycloalkyl of 3 to 5 carbon atoms,
(f) a group of the formula —NR45R46, wherein R45 and R46 are each, independently, a hydrogen atom, alkyl or fluoroalkyl of 1 to 6 carbon atoms, cycloalkyl of 3 to 6 carbon atoms or acyl of 1 to 7 carbon atoms, or wherein R45 and R46 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring,
(g) a group of the formula —CONR47R48, wherein R47 and R48 are each independently a hydrogen atom, alkyl or fluoroalkyl of 1 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, or wherein R47 and R48 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring,
(h) a group of the formula —OR49, wherein R49 is a hydrogen atom, or an alkyl, fluoroalkyl or acyl group of 1 to 7 carbon atoms,
(i) a group of the formula —SR50, wherein R50 is a hydrogen atom, or an alkyl, fluoroalkyl or acyl group of 1 to 7 carbon atoms,
(j) cyano,
(k) nitro,
(l) an amidino group of the formula
Figure USRE038132-20030603-C00009
wherein R51, R52 and R53 are each, independently, a hydrogen atom or alkyl of 1 to 3 carbon atoms, and wherein two of R51, R52 and R53 may additionally constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom(s) between them form a heterocyclic ring, or
(m) halogen,
(ii) methyl, which may be mono- or polysubstituted with fluorine atoms and additionally may be monosubstituted with R43,
(iii) branched or unbranched alkyl of 2 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, which alkyl or cycloakyl group may be mono- or polysubstituted with halogen or oxo,
(iv) a group of the formula —COOR54, wherein R54 is straight or branched alkyl of 1 to 5 carbon atoms or cycloalkyl of 3 to 5 carbon atoms,
(v) a group of the formula —NR55R56, wherein R55 and R56 are each, independently, a hydrogen atom, alkyl or fluoroalkyl of 1 to 6 carbon atoms, cycloalkyl of 3 to 6 carbon atoms or acyl of 1 to 7 carbon atoms, or wherein R55 and R56 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring, and wherein one of R55 and R56 may additionally be the group R43,
(vi) a group of the formula —CONR57R58, wherein R57 and R58 are each, independently, a hydrogen atom, alkyl or fluoroalkyl of 1 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, or wherein R57 and R58 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring, and wherein one of R57 and R58 may additionally be the group R43,
(vii) a group of the formula —COR59, wherein R59 is a hydrogen atom, straight or branched alkyl of 1 to 5 carbon atoms, cycloalkyl of 3 to 5 carbon atoms or R43,
(viii) a group of the formula —OR60, wherein R60 is a hydrogen atom, an alkyl, fluoroalkyl or acyl group of 1 to 7 carbon atoms, or R43,
(ix) a group of the formula —SR61, wherein R61 is a hydrogen atom, an alkyl, fluoroalkyl or acyl group of 1 to 7 carbon atoms, or R43,
(x) cyano,
(xi) nitro, or
(xii) halogen,
R41 is:
aryl selected from the class consisting of phenyl, 2-naphthyl, 2-, 3-, 5- or 6-indolyl, 2- or 3-thiophenyl, 2-, 3- or 4-pyridyl, 2-, 4- or 5-pyrimidinyl, 2- or 3-furyl, 1-, 2- or 3-pyrrolyl, 2-, 4- or 5-oxazolyl, 2-, 4- or 5-thiazolyl, 1-, 3-, 4- or 5-pyrazolyl, 3-, 4- or 5-isoxazolyl, 1-, 2-, 4- or 5-imidazolyl, 3-, 4- or 5-isothiazolyl, 4- or 5-oxadiazolyl, 1-, 4- or 5-triazolyl, 2-thiadiazolyl, 3- or 4-pyridazinyl, 2-pyrazinyl, 2-triazinyl, 2-, -3, 6- or 7-indolyzinyl, 2-, 3-, 5- or 6-isoindolyl, 2-, 3-, 5- or 6-benzo[b]faranyl, 2-, 3-, 5- or 6-benzo[b]thiophenyl, 3-, 5- or 6-indazolyl, 2-, 5- or 6-benzthiazolyl, 2-, 5- or 6-benzimidazolyl, 2-, 3-, 6- or 7-quinolinyl, 3-, 6- or 7-isoquinolinyl, 2- or 8-purinyl, 2-, 3-, 7- or 8-quinolizinyl, 3-, 6- or 7-cinnolinyl, 6- or 7-pthalaninyl, 2-, 3-, 6- or 7-quinoxalinyl, 2-, 3-, 6- or 7-napthyridinyl, 2-, 6- or 7-pteridinyl and 2-, 6- or 7-quinazolinyl,
wherein one or more of the hydrogen atoms of said aryl group may be optionally and independently replaced with:
(A) R62, which is aryl selected from the class consisting of phenyl, 2-naphthyl, 2-, 3-, 5- or 6-indolyl, 2- or 3-thiophenyl, 2-, 3- or 4-pyridyl, 2-, 4- or 5-pyrimidinyl, 2- or 3-furyl, 1-, 2- or 3-pyrrolyl, 2-, 4- or 5-oxazolyl, 2-, 4- or 5-thiazolyl, 1-, 3-, 4- or 5-pyrazolyl, 3-, 4- or 5-isoxazolyl, 1-, 2-, 4- or 5-imidazolyl, 3-, 4- or 5-isothiazolyl, 4- or 5-oxadiazolyl, 1-, 4- or 5-triazolyl, 2-thiadiazolyl, 3- or 4-pyridazinyl, 2-pyrazinyl, 2-triazinyl, 2-, -3, 6- or 7-indolyzinyl, 2-, 3-, 5- or 6-isoindolyl, 2-, 3-, 5- or 6-benzo[b]furanyl, 2-, 3-, 5- or 6-benzo[b]thiophenyl, 3-, 5- or 6-indazolyl, 2-, 5- or 6-benzthiazolyl, 2-, 5- or 6-benzimidazolyl, 2-, 3-, 6- or 7-quinolinyl, 3-, 6- or 7-isoquinolinyl, 2- or 8-purinyl, 2-, 3-, 7- or 8-quinolizinyl, 3-, 6- or 7-cinnolinyl, 6- or 7-pthalaninyl, 2-, 3-, 6- or 7-quinoxalinyl, 2-, 3-, 6- or 7-napthyridinyl, 2-, 6- or 7-pteridinyl and 2-, 6- or 7-quinazolinyl,
wherein one or more of the hydrogen atoms of said aryl group may be optionally and independently replaced with:
(i) branched or unbranched alkyl of 1 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, which alkyl or cycloakyl group may be mono- or polysubstituted with halogen or oxo,
(ii) —COOH,
(iii) —SO2OH,
(iv) —PO(OH)2,
(v) a group of the formula —COOR63, wherein R63 is straight or branched alkyl of 1 to 5 carbon atoms or cycloalkyl of 3 to 5 carbon atoms,
(vi) a group of the formula —NR64R65, wherein R64 and R65 are each, independently, a hydrogen atom, alkyl or fluoroalkyl of 1 to 6 carbon atoms, cycloalkyl of 3 to 6 carbon atoms or acyl of 1 to 7 carbon atoms, or wherein R64 and R65 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring,
(vii) a group of the formula —CONR66R67, wherein R66 and R67 are each, independently, a hydrogen atom, alkyl or fluoroalkyl of 1 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, or wherein R66 and R67 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring,
(viii) a group of the formula —OR68, wherein R68 is a hydrogen atom, or an alkyl, fluoroalkyl or acyl group of 1 to 7 carbon atoms,
(ix) a group of the formula —SR69, wherein R69 is a hydrogen atom, or an alkyl, fluoroalkyl or acyl group of 1 to 7 carbon atoms,
(x) cyano,
(xi) nitro, or
(xii) an amidino group of the formula
Figure USRE038132-20030603-C00010
wherein R70, R71 and R72 are each, independently, a hydrogen atom or alkyl or fluoroalkyl of 1 to 3 carbon atoms, and wherein two of R70, R71 and R72 may additionally constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom(s) between them form a heterocyclic ring, or
(xiii) halogen,
(B) methyl, which may be mono- or polysubstituted with fluorine atoms and additionally may be monosubstituted with R62,
(C) branched or unbranched alkyl of 2 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, which alkyl or cycloakyl group may be mono- or polysubstituted with halogen or oxo,
(D) a group of the formula —COOR73, wherein R73 is straight or branched alkyl of 1 to 5 carbon atoms or cycloalkyl of 3 to 5 carbon atoms,
(E) a group of the formula —NR74R75, wherein R74 and R75 are each, independently, a hydrogen atom, alkyl or fluoroalkyl of 1 to 6 carbon atoms, cycloalkyl of 3 to 6 carbon atoms or acyl of 1 to 7 carbon atoms, or wherein R74 and R75 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring, and wherein one of R74 and R75 may additionally be the group R62,
(F) a group of the formula —CONR76R77, wherein R76 and R77 are each, independently, a hydrogen atom, alkyl or fluoroallyl of 1 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, or wherein R76 and R77 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring, and wherein one of R76 and R77 may additionally be the group R62,
(G) a group of the formula —COR78, wherein R78 is a hydrogen atom, straight or branched alkyl of 1 to 5 carbon atoms, cycloalkyl of 3 to 5 carbon atoms or R62,
(H) a group of the formula —OR79, wherein R79 is a hydrogen atom, an alkyl, fluoroalkyl or acyl group of 1 to 7 carbon atoms, or R62,
(I) a group of the formula —SR80, wherein R80 is a hydrogen atom, an alkyl, fluoroalkyl or acyl group of 1 to 7 carbon atoms, or R62,
(J) cyano,
(K) nitro, or
(L) halogen;
R4 is Cl or trifluoromethyl; and, R5 and R6 are each, independently, a hydrogen, fluorine, chlorine, bromine or iodine atom, methyl or trifluoromethyl;
or a pharmaceutically acceptable salt thereof.
As mentioned before, some of the compounds embraced by the above-described genus are known and have been described in U.S. Pat. No. 3,668,217; U.S. Pat. No. 4,944,791; U.S. Pat. No. 3,741,981; Li, W.-Y; et al., J. Pharm. Sci. 1984, 73, 553-558 and, Abd El Halim, M. S.; et al., Monatshefte fur Chemie, 1994, 125, 1437-1442.
In its second aspect, the invention comprises novel compounds of the formula I
Figure USRE038132-20030603-C00011
wherein X, Y, Z, R2, R3, R4, R5 and R6 are defined as above except that, in the moiety R3, at least one of the hydrogen atoms of the aryl group R41 is necessarily, rather than optionally, replaced by:
(A) R62, which is aryl selected from the class consisting of phenyl, 2-naphthyl, 2-, 3-, 5- or 6-indolyl, 2- or 3-thiophenyl, 2-, 3- or 4-pyridyl, 2-, 4- or 5-pyrimidinyl, 2- or 3-furyl, 1-, 2- or 3-pyrrolyl, 2-, 4- or 5-oxazolyl, 2-, 4- or 5-thiazolyl, 1-, 3-, 4- or 5-pyrazolyl, 3-, 4- or 5-isoxazolyl, 1-, 2-, 4- or 5-imidazolyl, 3-, 4- or 5-isothiazolyl, 4- or 5-oxadiazolyl, 1-, 4- or 5-triazolyl, 2-thiadiazolyl, 3- or 4-pyridazinyl, 2-pyrazinyl, 2-triazinyl, 2-, -3, 6- or 7-indolyzinyl, 2-, 3-, 5- or 6-isoindolyl, 2-, 3-, 5- or 6-benzo[b]furanyl, 2-, 3-, 5- or 6-benzo[b]thiophenyl, 3-, 5- or 6-indazolyl, 2-, 5- or 6-benzthiazolyl, 2-, 5- or 6-benzimidazolyl, 2-, 3-, 6- or 7-quinolinyl, 3-, 6- or 7-isoquinolinyl, 2- or 8-purinyl, 2-, 3-, 7- or 8-quinolizinyl, 3-, 6- or 7-cinnolinyl, 6- or 7-pthalaninyl, 2-, 3-, 6- or 7-quinoxalinyl, 2-, 3-, 6- or 7-napthyridinyl, 2-, 6- or 7-pteridinyl or 2-, 6- and 7-quinazolinyl,
wherein one or more of the hydrogen atoms of said aryl group may be optionally and independently replaced with:
(i) branched or unbranched alkyl of 1 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, which alkyl or cycloakyl group may be mono- or polysubstituted with halogen or oxo,
(ii) —COOH,
(iii) —SO2OH,
(iv) —PO(OH)2,
(v) a group of the formula COOR63, wherein R63 is straight or branched alkyl of 1 to 5 carbon atoms or cycloalkyl of 3 to 5 carbon atoms,
(vi) a group of the formula —NR64R65, wherein R64 and R65 are each, independently, a hydrogen atom, alkyl or fluoroalkyl of 1 to 6 carbon atoms, cycloalkyl of 3 to 6 carbon atoms or acyl of 1 to 7 carbon atoms, or wherein R64 and R65 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring,
(vii) a group of the formula —CONR66R67, wherein R66 and R67 are each, independently, a hydrogen atom, alkyl or fluoroalkyl of 1 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, or wherein R66 and R67 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring,
(viii) a group of the formula —OR68, wherein R68 is a hydrogen atom, or an alkyl, fluoroalkyl or acyl group of 1 to 7 carbon atoms,
(ix) a group of the formula —SR69, wherein R69 is a hydrogen atom, or an alkyl , fluoroalkyl or acyl group of 1 to 7 carbon atoms,
(x) cyano,
(xi) nitro,
(xii) an amidino group of the formula
Figure USRE038132-20030603-C00012
wherein R70, R71 and R72 are each, independently, a hydrogen atom or alkyl or fluoroalkyl of 1 to 3 carbon atoms, and wherein two of R70, R71 and R72 may additionally constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom(s) between them form a heterocyclic ring, or
(xiii) halogen,
(B) methyl, which may be mono- or polysubstituted with fluorine atoms and additionally may be monosubstituted with R62,
(C) branched or unbranched alkyl of 2 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, which alkyl or cycloakyl group may be mono- or polysubstituted with halogen or oxo,
(D) a group of the formula —COOR73, wherein R73 is straight or branched alkyl of 1 to 5 carbon atoms or cycloalkyl of 3 to 5 carbon atoms,
(E) a group of the formula —NR74R75, wherein R74 and R75 are each, independently, a hydrogen atom, alkyl or fluoroalkyl of 1 to 6 carbon atoms, cycloalkyl of 3 to 6 carbon atoms or acyl of 1 to 7 carbon atoms, or wherein R74 and R75 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring, and wherein one of R74 and R75 may additionally be the group R62,
(F) a group of the formula —CONR76 R77, wherein R76 and R77 are each, independently, a hydrogen atom, alkyl or fluoroalkyl of 1 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, or wherein R76 and R77 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring, and wherein one of R76 and R77 may additionally be the group R62,
(G) a group of the formula —COR78, wherein R78 is a hydrogen atom, straight or branched alkyl of 1 to 5 carbon atoms, cycloalkyl of 3 to 5 carbon atoms or R62,
(H) a group of the formula —OR79, wherein R79 is a hydrogen atom, an alkyl fluoroalkyl or acyl group of 1 to 7 carbon atoms, or R62,
(I) a group of the formula —SR80, wherein R80 is a hydrogen atom, an alkyl, fluoroalkyl or acyl group of 1 to 7 carbon atoms, or R62,
(J) cyano,
(K) nitro, or
(L) halogen;
or pharmaceutically acceptable salts thereof.
Preferred novel compounds of formula I are those wherein:
Y is an oxygen or sulfur atom;
Z is an oxygen or sulfur atom;
X is a divalent group of the formula <CHR1, <NR1, <CHSO2R1, or <NSO2R1, or an oxygen or sulfur atom, wherein R1 is:
(A) a hydrogen atom,
(B) branched or unbranched alkyl of 1 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, which alkyl or cycloakyl group may be monosubstituted with:
(i) halogen,
(ii) oxo,
(iii) aryl selected from the class consisting of phenyl, naphthyl, indolyl, thiophenyl, pyridyl, pyrimidinyl, furyl, pyrrolyl, oxazolyl, thiazolyl, pyrazolyl, isoxazolyl, imidazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl, pyridazinyl, pyrazinyl, triazinyl, indolyzinyl, isoindolyl, benzo[b]furanyl, benzo[b]thiophenyl, indazolyl, benzthiazolyl, benzimidazolyl, quinolinyl, isoquinolinyl, purinyl, quinolizinyl, cinnolinyl, pthalaninyl, quinoxalinyl, napthyridinyl, pteridinyl and quinazolinyl,
wherein one or more hydrogen atoms of said aryl group may be optionally and independently replaced with:
(a) alkyl of 1 to 3 carbon atoms,
(b) —COOH,
(c) —SO2OH,
(d) —PO(OH)2,
(e) a group of the formula —COOR7, wherein R7 is straight or branched alkyl of 1 to 5 carbon atoms or cycloalkyl of 3 to 5 carbon atoms,
(f) a group of the formula —NR8R9, wherein R8 and R9 are each, independently, a hydrogen atom, alkyl of 1 to 6 carbon atoms, cycloalkyl of 3 to 6 carbon atoms or acyl of 1 to 7 carbon atoms, or wherein R8 and R9 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring,
(g) a group of the formula —CONR10R11, wherein R10 and R11 are each, independently, a hydrogen atom, alkyl of 1 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, or wherein R10 and R11 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring,
(h) a group of the formula —OR12a, wherein R12a is a hydrogen atom, or an alkyl or acyl group of 1 to 7 carbon atoms,
(i) a group of the formula —SR12b, wherein R12b is a hydrogen atom, or an alkyl or acyl group of 1 to 7 carbon atoms,
(j) cyano, or
(k) an amidino group of the formula
Figure USRE038132-20030603-C00013
wherein R13, R14 and R15 are each, independently, a hydrogen atom or alkyl of 1 to 3 carbon atoms and wherein two of R13, R14 and R15 may additionally constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom(s) between them form a heterocyclic ring,
(iv) a group of the formula —COOR16, wherein R16 is straight or branched alkyl of 1 to 7 carbon atoms or cycloalkyl of 3 to 6 carbon atoms,
(v) cyano,
(vi) a group of the formula —CONR17R18, wherein R17 and R18 are each, independently, a hydrogen atom, alkyl of 1 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, or wherein R17 and R18 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring,
(vii) a group of the formula —OR19, wherein R19 is a hydrogen atom, or an alkyl or acyl group of 1 to 7 carbon atoms,
(viii) a group of the formula —SR20, wherein R20 is a hydrogen atom, or an alkyl or acyl group of 1 to 7 carbon atoms,
(ix) a group of the formula —NR21R22, wherein R21 and R22 are each, independently:
(a) a hydrogen atom,
(b) alkyl or acyl of 1 to 7 carbon atoms or cycloalkyl of 3 to 7 carbon atoms,
(c) a group of the formula {(CH2)mCOOH, wherein m is 0, 1 or 2, or
(d) a group of the formula —(CH2)nCOOR23, wherein n is 0, 1 or 2, wherein R23 is straight or branched alkyl of 1 to 6 carbon atoms, or wherein R21 and R22 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring, or
(x) a quaternary group of the formula
Figure USRE038132-20030603-C00014
wherein R24, R25 and R26 are each, independently, a branched or unbranched alkyl group of 1 to 7 carbon atoms and Q is a chlorine, bromine or iodine counterion,
(C) a branched or unbranched carboxylic acid group of 3 to 6 carbon atoms,
(D) a branched or unbranched phosphonic acid group of 2 to 6 carbon atoms,
(E) a branched or unbranched sulfonic acid group of 2 to 6 carbon atoms,
(F) an amidino group of the formula
Figure USRE038132-20030603-C00015
wherein r is 2, 3, 4, 5 or 6, and
R27, R28 and R29 are each, independently, a hydrogen atom or alkyl of 1 to 3 carbon atoms and wherein two of R27, R28 and R29 may additionally constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom(s) between them form a heterocyclic ring,
(G) an guanidino group of the formula
Figure USRE038132-20030603-C00016
wherein s is 2, 3, 4, 5 or 6, and R30, R31, R32 and R33 are each independently a hydrogen atom or alkyl of 1 to 3 carbon atoms and wherein two of R30, R31, R32 and R33 may additionally constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom(s) between them form a heterocyclic ring, or
(H) piperidyl, wherein the nitrogen atom of said group is optionally substituted with:
(i) alkyl of 1 to 3 carbon atoms,
(ii) a carboxylic ester group of 2 to 7 carbon atoms,
(iii) a carboxylic acid group of 2 to 5 carbon atoms,
(iv) a phosphonic acid group of 1 to 6 carbon atoms, or
(v) a sulfonic acid group of 1 to 6 carbon atoms;
R2 is:
(A) a hydrogen atom, or
(B) methyl;
R3 is a group of the formula —CH2R41, wherein:
R41 is:
aryl selected from the class consisting of phenyl, 2-naphthyl, 2-, 3-, 5- or 6-indolyl, 2- or 3-thiophenyl, 2-, 3- or 4-pyridyl, 2-, 4- or 5-pyrimidinyl, 2- or 3-furyl, 1-, 2- or 3-pyrrolyl, 2-, 4- or 5-oxazolyl, 2-, 4- or 5-thiazolyl, 1-, 3-, 4- or 5-pyrazolyl, 3-, 4- or 5-isoxazolyl, 1-, 2-, 4- or 5-imidazolyl, 3-, 4- or 5-isothiazolyl, 4- or 5-oxadiazolyl, 1-, 4- or 5-triazolyl, 2-thiadiazolyl, 3- or 4-pyridazinyl, 2-pyrazinyl, 2-triazinyl, 2-, -3, 6- or 7-indolyzinyl, 2-, 3-, 5- or 6-isoindolyl, 2-, 3-, 5- or 6-benzo[b]furanyl, 2-, 3-, 5- or 6-benzo[b]thiophenyl, 3-, 5- or 6-indazolyl, 2-, 5- or 6-benzthiazolyl, 2-, 5- or 6-benzimidazolyl, 2-, 3-, 6- or 7-quinolinyl, 3-, 6- or 7-isoquinolinyl, 2- or 8-purinyl, 2-, 3-, 7- or 8-quinolizinyl, 3-, 6- or 7-cinnolinyl, 6- or 7-pthalaninyl, 2,- 3-, 6- or 7-quinoxalinyl, 2-, 3-, 6- or 7-napthyridinyl, 2-, 6- or 7-pteridinyl and 2-, 6- or 7-quinazolinyl,
 wherein one or more of the hydrogen atoms of said aryl group are necessarily and independently replaced with:
(A) R62, which is aryl selected from the class consisting of phenyl, 2-naphthyl, 2-, 3-, 5- or 6-indolyl, 2- or 3-thiophenyl, 2-, 3- or 4-pyridyl, 2-, 4- or 5-pyrimidinyl, 2- or 3-furyl, 1-, 2- or 3-pyrrolyl, 2-, 4- or 5-oxoazolyl, 2-, 4- or 5-thiazolyl, 1-, 3-, 4- or 5-pyrazolyl, 3-, 4- or 5-isoxazolyl, 1-, 2-, 4- or 5-imidazolyl, 3-, 4- or 5-isothiazolyl, 4- or 5-oxadiazolyl, 1-, 4- or 5-triazolyl, 2-thiadiazolyl, 3- or 4-pyridazinyl, 2-pyrazinyl, 2-triazinyl, 2-, -3, 6- or 7-indolyzinyl, 2-, 3-, 5- or 6-isoindolyl, 2-, 3-, 5- or 6-benzo[b] furanyl, 2-, 3-, 5- or 6-benzo[b]thiophenyl, 3-, 5- or 6-indazolyl, 2-, 5- or 6-benzthiazolyl, 2-, 5- or 6-benzimidazolyl, 2-, 3-, 6- or 7-quinolinyl, 3-, 6- or 7-isoquinolinyl, 2- or 8-purinyl, 2-, 3-, 7- or 8-quinolizinyl, 3-, 6- or 7-cinnolinyl, 6- or 7-phthalaninyl, 2-, 3-, 6- or 7-quinoxalinyl, 2-, 3-, 6- or 7-naphthyridinyl, 2-, 6- or 7-pteridinyl and 2-, 6- or 7-quinazolinyl,
 wherein one or more of the hydrogen atoms of said aryl group may be optionally and independently replaced with:
(i) branched or unbranched alkyl of 1 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, which alkyl or cycloalkyl group may be mono- or polysubstituted with halogen or oxo,
(ii) —COOH,
(iii) —SO2OH,
(iv) —PO(OH)2,
(v) a group of the formula —COOR63, wherein R63 is straight or branched alkyl of 1 to 5 carbon atoms or cycloalkyl of 3 to 5 carbon atoms,
(vi) a group of the formula —NR64R65, wherein R64 and R65 are each, independently, a hydrogen atom, alkyl or fluooralkyl of 1 to 6 carbon atoms, cycloalkyl of 3 to 6 carbn atoms or acyl of 1 to 7 carbon atoms, or wherein R64 and R65 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring,
(vii) a group of the formula —CONR66R67, wherein R66 and R67 are each, independently, a hydrogen atom, alkyl or fluoroalkyl of 1 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, or wherein R66 and R67 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring,
(viii) a group of the formula —OR68, wherein R68 is a hydrogen atom, or an alkyl, fluoroalkyl or acyl group of 1 to 7 carbon atoms,
(ix) a group of the formula —SR69, wherein R69 is a hydrogen atom, or an alkyl, fluoroalkyl or acyl group of 1 to 7 carbon atoms,
(x) cyano,
(xi) nitro,
(xii) an amidino group of the formula
Figure USRE038132-20030603-C00017
 wherein R70, R71 and R72 are each, independently, a hydrogen atom or alkyl or fluoroalkyl of 1 to 3 carbon atoms, and wherein two of R70, R71 and R72 may additionally constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom(s) between them form a heterocyclic ring, or
(xiii) halogen,
(B) methyl, which may be mono- or polysubstituted with fluorine atoms and additionally may be monosubstituted with R62,
(C) branched or unbranched alkyl of 2 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, which alkyl or cycloalkyl group may be mono- or polysubstituted with halogen or oxo,
(D) a group of the formula —COOR73, wherein R73 is straight or branched alkyl of 1 to 5 carbon atoms or cycloalkyl of 3 to 5 carbon atoms,
(E) a group of the formula —NR74R75, wherein R74 and R75 are each, independently, a hydrogen atom, alkyl or fluoroalkyl of 1 to 6 carbon atoms, cycloalkyl of 3 to 6 carbon atoms or acyl of 1 to 7 carbon atoms, or wherein R74 and R75 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring, and wherein one of R74 and R75 may additionally be the group, R62,
(F) a group of the formula —CONR76R77, wherein R76 and R77 are each, independently, a hydrogen atom, alkyl or fluoroalkyl of 1 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, or wherein R76 and R77 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring, and wherein one of R76 and R77 may additionally be the group R62,
(G) a group of the formula —COR78, wherein R78 is a hydrogen atom, straight or branched alkyl of 1 to 5 carbon atoms, cycloalkyl of 3 to 5 carbon atoms or R62,
(H) a group of the formula —OR79, wherein R79 is a hydrogen atom, an alkyl, fluoroalkyl or acyl group of 1 to 7 carbon atoms, or R62,
(I) a group of the formula —SR80, wherein R80 is a hydrogen atom, an alkyl, fluoroalkyl or acyl group of 1 to 7 carbon atoms, or R62,
(J) cyano,
(K) nitro, or
(L) halogen;
R4 is Cl or trifluoromethyl; and
R5 and R6 are each independently a hydrogen, fluorine, chlorine, bromine or iodine atom, methyl or trifluoromethyl;
or a pharmaceutically acceptable salt thereof.
More preferred are those novel compounds of formula I wherein:
Y is an oxygen atom;
Z is an oxygen atom;
X is a divalent group of the formula >CHR1 or >NR1, wherein R1 is:
(A) a hydrogen atom,
(B) branched or unbranched alkyl of 1 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, which alkyl or cycloalkyl group may be monosubstituted with:
(i) oxo,
(ii) aryl selected from the class consisting of phenyl, thiophenyl, pyridyl, pyrimidinyl, furyl, pyrrolyl, oxazolyl, thiazolyl, pyrazolyl, isoxazolyl, imidazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl, pyridazinyl, pyrazinyl and triazinyl,
 wherein one or more hydrogen atoms of said aryl group may be optionally and independently replaced with:
 (a) alkyl of 1 to 3 carbon atoms,
 (b) —COOH,
 (c) —SO2OH,
 (d) —PO(OH)2,
 (e) a group of the formula —COOR7, wherein
 R7 is straight or branched alkyl of 1 to 5 carbon
 atoms or cycloalkyl of 3 to 5 carbon atoms,
 (f) a group of the formula —NH2,
 (g) a group of the formula —CONH2,
 (h) a group of the formula —OR12a, wherein
 R12a is a hydrogen atom or a methyl,
 (i) an amidino group of the formula
Figure USRE038132-20030603-C00018
 wherein R13, R14 and R15 are each hydrogen atoms,
(j) a group of the formula —COOR16, wherein R16 is straight or branched alkyl of 1 to 7 carbon atoms or cycloalkyl of 3 to 6 carbon atoms,
(k) a group of the formula —OR19, wherein R19 is a hydrogen atom, or an alkyl or acyl group of 1 to 7 carbon atoms, or
(l) a quaternary group of the formula
Figure USRE038132-20030603-C00019
 wherein R24, R25 and R26 are each methyl and Qis a chlorine, bromine or iodine counterion,
(C) a branched or unbranched carboxylic acid group of 3 to 6 carbon atoms,
(D) a branched or unbranched phosphonic acid group of 2 to 6 carbon atoms,
(E) a branched or unbranched sulfonic acid group of 2 to 6 carbon atoms,
(F) an amidino group of the formula
Figure USRE038132-20030603-C00020
 wherein r is 2, 3, 4, 5 or 6, and R27, R28 and R29 are each hydrogen atoms,
(G) an guanidino group of the formula
Figure USRE038132-20030603-C00021
 wherein s is 2, 3, 4, 5 or 6, R30, R31, R32 and R33 are each hydrogen atoms, or
(H) piperidyl, wherein the nitrogen atom of said group is optionally substituted with:
(i) alkyl of 1 to 3 carbon atoms,
(ii) a carboxylic ester group of 2 to 7 carbon atoms,
(iii) a carboxylic acid group of 2 to 5 carbon atoms,
(iv) a phosphonic acid group of 1 to 6 carbon atoms, or
(v) a sulfonic acid group of 1 to 6 carbon atoms;
R2 is:
(A) a hydrogen atom, or
(B) methyl;
R3 is a group of the formula —CH2R41, wherein
R41 is
 aryl selected from the class consisting of phenyl, thiophenyl, pyridyl, pyrimidinyl, furyl, pyrrolyl, oxazolyl, thiazolyl, pyrazolyl, isoxazolyl, imidazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl, pyridazinyl, pyrazinyl and triazinyl, wherein one or more of the hydrogen atoms of said aryl group are necessarily and independently replaced with:
(A) R62, which is aryl selected from the class consisting of phenyl, thiophenyl, pyridyl, pyrimidinyl, furyl, pyrrolyl, oxazolyl, thiazolyl, pyrazolyl, isoxazolyl, imidazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl, pyridazinyl, pyrazinyl and triazinyl,
 wherein one or more of the hydrogen atoms of said aryl group may be optionally and independently replaced with:
(i) methyl,
(ii) —COOH,
(iii) —SO2OH,
(iv) —PO(OH)2,
(v) a group of the formula —COOR63, wherein R63 is methyl,
(vi) a group of the formula —NR64R65, wherein R64 and R65 are each, independently, a hydrogen atom or methyl,
(vii) a group of the formula —CONR66R67, wherein R66 and R67 are each, independently, a hydrogen atom or methyl,
(viii) a group of the formula —OR68, wherein R68 is a hydrogen atom or methyl,
(ix) a group of the formula —SR69, wherein R69 is a hydrogen atom or methyl,
(x) cyano,
(xi) nitro, or
(xii) halogen,
(B) methyl, which may be mono- or polysubstituted with fluorine atoms and which additionally may be monosubstituted with R62,
(C) branched or unbranched alkyl of 2 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, which alkyl or cycloalkyl group may be mono- or polysubstituted with halogen or oxo,
(D) a group of the formula —COOR73, wherein R73 is methyl,
(E) a group of the formula —NR74R75, wherein R74 and R75 are each, independently, a hydrogen atom or methyl, and wherein one of R74 and R75 may additionally be the group R62,
(F) a group of the formula —CONR76R77, wherein R76 and R77 are each, independently, a hydrogen atom or methyl, and wherein one of R76 and R77 may additionally be the group R62,
(G) a group of the formula —COR78, wherein R78 is a hydrogen atom, methyl or R62,
(H) a group of the formula —OR79, wherein R79 is a hydrogen atom, methyl or R62,
(I) a group of the formula —SR80, wherein R80 is a hydrogen atom, methyl or R62,
(J) cyano,
(K) nitro, or
(L) halogen;
R4 is Cl or trifluoromethyl;
R5 is a hydrogen atom; and,
R6 is Cl, or trifluoromethyl;
or a pharmaceutically acceptable salt thereof.
Even more preferred are those novel compounds of formula I wherein:
Y is an oxygen atom;
Z is an oxygen atom;
X is a divalent group of the formula >CHR1 or >NR1, wherein R1 is:
(A) a hydrogen atom,
(B) branched or unbranched alkyl of 1 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, which alkyl or cycloalkyl group may be monosubstituted with:
(i) oxo,
(ii) aryl selected from the class consisting of phenyl, thiophenyl, pyridyl, pyrimidinyl, furyl, pyrrolyl, oxazolyl, thiazolyl, pyrazolyl, isoxazolyl, imidazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl, pyridazinyl, pyrazinyl and triazinyl, wherein one or more hydrogen atoms of said aryl group may be optionally and independently replaced with:
 (a) alkyl of 1 to 3 carbon atoms,
 (b) —COOH,
 (c) —SO2OH,
 (d) —PO(OH)2,
 (e) a group of the formula —COOR7, wherein
 R7 is straight or branched alkyl of 1 to 5 carbon
 atoms or cycloalkyl of 3 to 5 carbon atoms,
 (f) a group of the formula —NH2,
 (g) a group of the formula —CONH2,
 (h) a group of the formula —OR12a, wherein
 R12a is a hydrogen atom or a methyl,
 (i) an amidino group of the formula
Figure USRE038132-20030603-C00022
 wherein R13, R14 and R15 are each hydrogen atoms,
 (j) a group of the formula —COOR16, wherein
 R16 is straight or branched alkyl of 1 to 7
 carbon atoms or cycloalkyl of 3 to 6 carbon
 atoms,
 (k) a group of the formula —OR19, wherein
 R19 is a hydrogen atom, or an alkyl or acyl
 group of 1 to 7 carbon atoms, or
 (l) a quaternary group of the formula
Figure USRE038132-20030603-C00023
 wherein R24, R25 and R26 are each methyl and
 Qis a chlorine, bromine or iodine counterion,
(C) a branched or unbranched carboxylic acid group of 3 to 6 carbon atoms,
(D) a branched or unbranched phosphonic acid group of 2 to 6 carbon atoms,
(E) a branched or unbranched sulfonic acid group of 2 to 6 carbon atoms,
(F) an amidino group of the formula
Figure USRE038132-20030603-C00024
 wherein r is 2, 3, 4, 5 or 6, and R27, R28 and R29 are each hydrogen atoms,
(G) an guanidino group of the formula
Figure USRE038132-20030603-C00025
 wherein s is 2, 3, 4, 5 or 6, R30, R31, R32 and R33 are each hydrogen atoms, or
(H) piperidyl, wherein the nitrogen atom of said group is optionally substituted with:
(i) alkyl of 1 to 3 carbon atoms,
(ii) a carboxylic ester group of 2 to 7 carbon atoms,
(iii) a carboxylic acid group of 2 to 5 carbon atoms,
(iv) a phosphonic acid group of 1 to 6 carbon atoms, or
(v) a sulfonic acid group of 1 to 6 carbon atoms;
R2 is:
(A) a hydrogen atom, or
(B) methyl;
R3 is a group of the formula —CH2R41, wherein
R41 is
 aryl selected from the class consisting of phenyl, thiophenyl, pyridyl, pyrimidinyl, furyl, oxazolyl, thiazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, thiadiazolyl, pyridazinyl, and pyrazinyl, wherein one or more of the hydrogen atoms of said aryl group are necessarily and independently replaced with:
(A) R62, which is aryl selected from the class consisting of phenyl, thiophenyl, pyridyl, pyrimidinyl, furyl, oxazolyl, thiazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, thiadiazolyl, pyridazinyl, and pyrazinyl,
 wherein one or more of the hydrogen atoms of said aryl group may be optionally and independently replaced with:
(i) methyl,
(ii) —COOH,
(iii) a group of the formula —COOR63, wherein R63 is methyl,
(iv) a group of the formula —OR68, wherein R68 is a hydrogen atom or methyl, or
(v) halogen,
(B) methyl, which may be mono- or polysubstituted with fluorine atoms or which may be monosubstituted with R62;
(C) branched or unbranched alkyl of 2 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, which alkyl or cycloalkyl group may be mono- or polysubstituted with halogen or oxo,
(D) a group of the formula —COOR73, wherein R73 is methyl,
(E) a group of the formula —NR76R77, wherein R76 and R77 are each methyl, and wherein one of R76 and R77 is methyl and the other is the group R62,
(F) a group of the formula —CONR78, wherein R78 is a hydrogen atom, methyl or R62,
(G) a group of the formula —OR79, wherein R79 is a hydrogen atom, methyl or R62,
(H) cyano,
(I) nitro, or
(J) halogen;
R4 is Cl or trifluoromethyl;
R5 is a hydrogen atom; and,
R6 is Cl, or trifluoromethyl;
or a pharmaceutically acceptable salt thereof.
Still more preferred are those novel compounds of formula I wherein:
Y is an oxygen atom;
Z is an oxygen atom;
X is a divalent group of the formula >CHR1 or >NR1, wherein R1 is:
(A) a hydrogen atom,
(B) branched or unbranched alkyl of 1 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, which alkyl or cycloalkyl group may be monosubstituted with:
(i) oxo,
(ii) aryl selected from the class consisting of phenyl or pyridyl,
 wherein one or more hydrogen atoms of said aryl group may be optionally and independently replaced with:
 (a) alkyl of 1 to 3 carbon atoms,
 —COOH,
 (c) —SO2OH,
 (d) —PO(OH)2,
 (e) a group of the formula —OR12a, wherein
 R12a is a hydrogen atom or a methyl,
 (f) an amidino group of the formula
Figure USRE038132-20030603-C00026
 wherein R13, R14 and R15 are each hydrogen atoms,
(iii) a group of the formula —OR19, wherein R19 is a hydrogen atom, or an alkyl or acyl group of 1 to 7 carbon atoms, or
(iv) a quaternary group of the formula
Figure USRE038132-20030603-C00027
 wherein R24, R25 and R26 are each methyl and Qis a chlorine, bromine or iodine counterion,
(C) a branched or unbranched carboxylic acid group of 3 to 6 carbon atoms,
(D) a branched or unbranched phosphonic acid group of 2 to 6 carbon atoms,
(E) a branched or unbranched sulfonic acid group of 2 to 6 carbon atoms,
(F) an amidino group o f the formula
Figure USRE038132-20030603-C00028
 wherein r is 2, 3, 4, 5 or 6, and R27, R28 and R29 are each hydrogen atoms,
(G) an guanidino group of the formula
Figure USRE038132-20030603-C00029
 wherein s is 2, 3, 4, 5 or 6, R30, R31, R32 and R33 are each hydrogen atoms, or
(H) piperidyl, wherein the nitrogen atom of said group is optionally substituted with:
(i) alkyl of 1 to 3 carbon atoms,
(ii) a carboxylic ester group of 2 to 7 carbon atoms,
(iii) a carboxylic acid group of 2 to 5 carbon atoms,
(iv) a phosphonic acid group of 1 to 6 carbon atoms, or
(v) a sulfonic acid group of 1 to 6 carbon atoms;
R2is:
(A) a hydrogen atom, or
(B) methyl;
R3 is a group of the formula —CH2R41, wherein
R41 is
 aryl selected from the class consisting of phenyl or pyridyl,
 wherein one or more of the hydrogen atoms of said aryl group are necessarily and independently replaced with:
(A) R62, which is aryl selected from the class consisting of phenyl, or pyridyl,
 wherein one or more of the hydrogen atoms of said aryl group may be optionally and independently replaced with:
(i) methyl,
(ii) —COOH
(iii) a group of the formula —COOR63, wherein R63 is methyl,
(iv) a group of the formula —OR68, wherein R68 is a hydrogen atom or methyl, or
(v) halogen,
(B) methyl, which may be mono- or polysubstituted with fluorine atoms or which may be monosubstituted with R62,
(C) branched or unbranched alkyl of 2 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, which alkyl or cycloalkyl group may be mono- or polysubstituted with fluorine or oxo,
(D) a group of the formula —COOR73, wherein R73 is methyl,
(E) a group of the formula —CONR76R77, wherein R76 and R77 are each methyl, and wherein one of R76 and R77 is methyl and the other is the group R62,
(F) a group of the formula —COR78, wherein R78 is a hydrogen atom, methyl or R62,
(G) a group of the formula —OR79, wherein R79 is a hydrogen atom, methyl or R62,
(H) cyano,
(I) nitro, or
(J) halogen;
R4 is a chlorine atom or trifluoromethyl;
R5 is a hydrogen atom; and,
R6 is a chlorine atom, or trifluoromethyl;
or a pharmaceutically acceptable salt thereof.
Especially preferred novel compounds of formula I are those wherein:
Y is an oxygen atom;
Z is an oxygen atom;
X is a divalent group of the formula >CHR1 or >NR1, wherein R1 is:
(A) a hydrogen atom,
(B) alkyl of 1 to 2 carbon atoms which may be monosubstituted with:
(i) oxo,
(ii) aryl selected from the class consisting of phenyl or pyridyl, wherein one hydrogen atom of said aryl group may be optionally replaced with:
 (a) alkyl of 1 to 3 carbon atoms,
 (b) —COOH,
 (c) —SO2OH,
 (d) —PO(OH)2,
 (e) a group of the formula —OR12a, wherein
 R12a is a hydrogen atom or a methyl, or
 (f) an amidino group of the formula
Figure USRE038132-20030603-C00030
 wherein R13, R14 and R15 are each hydrogen atoms, or
(iii) a group of the formula —OR19, wherein R19 is a hydrogen atom or methyl,
(C) a branched or unbranched carboxylic acid group of 3 to 6 carbon atoms,
(D) a branched or unbranched phosphonic acid group of 2 to 6 carbon atoms,
(E) a branched or unbranched sulfonic acid group of 2 to 6 carbon atoms,
(F) an amidino group of the formula
Figure USRE038132-20030603-C00031
 wherein r is 2, 3, 4, 5 or 6, and R27, R28 and R29 are each hydrogen atoms, or
(G) an guanidino group of the formula
Figure USRE038132-20030603-C00032
 wherein s is 2, 3, 4, 5 or 6, R30, R31, R32 and R33 are each hydrogen atoms,
R2 is:
(A) a hydrogen atom, or
(B) methyl;
R3 is a group of the formula —CH2R41, wherein
R41 is
 phenyl
 wherein one or more of the hydrogen atoms of said phenyl group are necessarily and independently replaced with:
(A) R62, which is aryl selected from the class consisting of phenyl, or pyridyl,
 wherein one or more of the hydrogen atoms of said aryl group may be optionally and independently replaced with:
(i) methyl,
(ii) a group of the formula —COOR63, wherein R63 is methyl,
(iv) a group of the formula —OR68, wherein R68 is a hydrogen atom or methyl, or
(v) halogen,
(B) methyl, which may be mono- or polysubstituted with fluorine atoms or which may be monosubstituted with R62,
(C) a group of the formula —COOR73, wherein R73 is methyl,
(D) a group of the formula —COR78, wherein R78 is methyl or R62,
(E) a group of the formula —OR79, wherein R79 is a hydrogen atom, methyl or R62,
(F) cyano,
(G) nitro, or
(H) halogen;
R4 is a chlorine atom or trifluoromethyl;
R5 is a hydrogen atom; and,
R6 is a chlorine atom, or trifluoromethyl;
or a pharmaceutically acceptable salt thereof.
Even more especially preferred novel compounds of formula I are those wherein:
Y is an oxygen atom;
Z is an oxygen atom;
X is a divalent group of the formula >NR1, wherein R1 is:
(A) a hydrogen atom,
(B) methyl or ethyl, or
(C) —COOH3
 R2 is:
(A) a hydrogen atom, or
(B) methyl;
R3 is a group of the formula —CH2R41, wherein R41 is:
 phenyl,
 wherein one or more of the hydrogen atoms of said phenyl group are necessarily and independently replaced with:
(A) R62, which is aryl selected from the class consisting of phenyl, or pyridyl,
 wherein one or more of the hydrogen atoms of said aryl group may be optionally and independently replaced with:
(i) methyl,
(ii) a group of the formula —COOR63, wherein R63 is methyl,
(iii) a group of the formula —OR68, wherein R68 is a hydrogen atom or methyl, or
(iv) halogen,
(B) methyl, which may be mono- or polysubstituted with fluorine atoms or which may be monosubstituted with R62,
(C) a group of the formula —COOR73, wherein R73 is methyl,
(D) a group of the formula —COR78, wherein R78 is methyl or R62,
(E) a group of the formula —OR79, wherein R79 is a hydrogen atom, methyl or R62,
(F) cyano,
(G) nitro, or
(H) halogen;
R4 is a chlorine atom or trifluoromethyl;
R5 is a hydrogen atom; and,
R6 is a chlorine atom, or trifluoromethyl;
or a pharmaceutically acceptable salt thereof.
Penultimately preferred novel compounds of formula I are those written:
Y is an oxygen atom;
Z is an oxygen atom;
X is a divalent group of the formula >NR1, wherein R1 is:
(A) a hydrogen atom;
(B) methyl or ethyl, or
(C) —COOH3
R2 is:
(A) a hydrogen atom, or
(B) methyl;
R3 is a group of the formula —CH2R41, wherein R41 is:
phenyl
 wherein one or more of the hydrogen atoms of said phenyl group are necessarily and independently replaced with:
(A) R62, which is aryl selected from the class consisting of phenyl, or pyridyl,
 wherein one or more of the hydrogen atoms of said aryl group may be optionally and independently replaced with:
(i) methyl, or
(ii) halogen,
(B) methyl, which may be mono- or polysubstituted with fluorine atoms,
(C) a group of the formula —COR78, wherein R78 is methyl or R62,
(D) halogen;
R4 is a chlorine atom;
R5 is a hydrogen atom; and,
R6 is a chlorine atom;
or a pharmaceutically acceptable salt thereof.
Ultimately preferred novel compounds of formula I are those specific compounds having the following structures:
Figure USRE038132-20030603-C00033
Figure USRE038132-20030603-C00034
or a pharmaceutically acceptable salt thereof.
Synthesis of the Compounds of the Invention
The synthesis of similar compounds to those of the invention are well known in the prior art. Depending on one's purpose, some routes may be better for providing small amounts of a variety of compounds while other routes may be more amenable to the large scale synthesis of a specific compound. Below are illustrated several routes to these compounds and examples of compounds that have been synthesized by the respective routes.
The starting amino acids and their derivatives necessary for the synthesis of the hydantoin and thio-hydantoin structures are either commercially available or are produced by obvious modifications of known literature procedures (see e.g.: Williams, R. W., Synthesis of Optically Active α-Amino Acids; Pergamon: Oxford, 1989, α-Amino Acid Synthesis: O'Donnell, M. J., Ed.; Tetrahedron Symposium in Print; Pergamon: London, 1988: Vol. 44, Issue 17, Jung, M. J. Chemistry and Biochemistry of the Amino Acids; Barrett, G. C., Ed.: Chapman and Hall: New York, 1985; p.227, and Spero, D. M.; Kapadia, S. R. J. Org. Chem. 1996, 61: 7398-7401). The synthesis and resolution of ethyl 2-amino-2-(4-bromobenzyl)-propanoate (the starting material for example 39) is given by way of example.
A solution of alanine ethyl ester hydrochloride (15.3 g, 99.3 mmol) in 60 mL of water was treated with triethylamine (14.6 mL, 104.8 mmol) at room temperature for 3 min. The mixture was then extracted twice with 100 mL of methylene chloride. The organic layers were combined, dried over sodium sulfate, and concentrated in vacuo to afford 10.0 g of the free base of the amino ester (86% yield). The residue was re-dissolved in methylene chloride and cooled in an ice bath. Magnesium sulfate (11.3 g, 93.9 mmol) was added, followed by trimethyl acetaldehyde (9.3 mL, 85.6 mmol). The ice bath was removed, and the mixture was stirred overnight. The magnesium sulfate was removed by filtration, and the filtrate was concentrated in vacuo to afford 11.8 g of the imine intermediate (74.6% yield).
The imine from above (11.8 g, 63.7 mmol) was dissolved in toluene (90 mL). 4-bromobenzyl bromide (17.5 g, 70.1 mmol) was added, and the reaction was cooled to about −10° C. Potassium tert-butoxide (8.6 g, 76.5 mmol) was added at such a rate that the temperature did not exceed 0° C. The reaction stirred in the cold bath for two hours, then was diluted with ether and washed with water (150 mL). The organic layer was dried (sodium sulfate), filtered, and concentrated in vacuo to afford a clear yellow oil. This was treated with 1 N HCl (100 ml, 100 mmol) and stirred overnight. The reaction was extracted with ethyl acetate (100 mL), and the aqueous layer was to afford 14.1 g of the racemic amino ester hydrochloride (68.7% yield).
The racemic compounds can be resolved into their component enantiomers via a number of known techniques. Ethyl 2-(R)-amino-2-(4-bromobenzyl)-propanoate (the starting material for example 29) was produced from racemic ethyl 2-amino-2-(4-bromobenzyl)-propanoate by the following procedure: To 1.3 L of a buffer made from 13.69 g KH2PO4 and 2 L of water was added 20 g of the commercially available enzyme Lipase L10 (Amino Enzyme USA Co., Ltd, Lombardi, Ill.) followed by 12 g of the HCl salt of the racemic amino ester. The pH was monitored and 1 N KOH was added as needed to keep the pH of the mixture at 6.4. The course of the reaction was monitored with reverse phase HPLC and after 2 days, the HPLC analysis indicated that 50.4% of the starting material had been hydrolyzed. At this point enough solid NaHCO3 was added to adjust the pH to 8.1 and the mixture was extracted twice with toluene, ether and EtOAc. The combined organic layers was dried and concentrated and the crude product purified by silica gel chromatography (EtOAc: Hexanes) to yield 5.21 g (87%) of ethyl 2-(R)-amino-2-(4-bromobenzyl)-propanoate.
Method A. Starting with an Amino Acid and a Phenylisocyanate. Cyclization wit Acid.
An appropriate amino acid is dissolved in aqueous base (such as, for example, NaOH, KOH, Na2CO3, NaHCO3, K2CO3 or KHCO3) and warmed to between about 20 and 90° C. An appropriate isocyanate is added to this mixture and the resulting solution was stirred until the reaction essentially reaches completion. Upon cooling, the mixture is acidified and the resulting ureidoacetic acid is isolated by filtration or by extraction into organic solvent. Removal of solvent produces the intermediate ureidoacetic acid. In the manner reported by Sauli (U.S. Pat. No. 4,099,008), the intermediate ureidoacetic acid is cyclized by heating in the presence of a catalytic amount of acid (such as, for example, sulfuric acid, methanesulfonic acid, benzenesulfonic acid or hydrochloric acid) in an organic or aqueous solvent, to produce the desired hydantoin. Workup consists of collection of the hydantoin by filtration and purification by, for example, silica gel chromatography or recrystallization.
Compounds listed in Table 1 were produced via this general method.
TABLE 1
Examples of Compounds Synthesized by Method A.
STARTING M. P.
EX. STRUCTURE STARTING AMINO ACID ISOCYANATE (° C.)
1
Figure USRE038132-20030603-C00035
Figure USRE038132-20030603-C00036
Figure USRE038132-20030603-C00037
165-6
2
Figure USRE038132-20030603-C00038
Figure USRE038132-20030603-C00039
Figure USRE038132-20030603-C00040
145-6
3
Figure USRE038132-20030603-C00041
Figure USRE038132-20030603-C00042
Figure USRE038132-20030603-C00043
165-7
4
Figure USRE038132-20030603-C00044
Figure USRE038132-20030603-C00045
Figure USRE038132-20030603-C00046
201-2
5
Figure USRE038132-20030603-C00047
Figure USRE038132-20030603-C00048
Figure USRE038132-20030603-C00049
206-8
6
Figure USRE038132-20030603-C00050
Figure USRE038132-20030603-C00051
Figure USRE038132-20030603-C00052
197-8
7
Figure USRE038132-20030603-C00053
Figure USRE038132-20030603-C00054
Figure USRE038132-20030603-C00055
195-6
8
Figure USRE038132-20030603-C00056
Figure USRE038132-20030603-C00057
Figure USRE038132-20030603-C00058
146-8
9
Figure USRE038132-20030603-C00059
Figure USRE038132-20030603-C00060
Figure USRE038132-20030603-C00061
225-6
10
Figure USRE038132-20030603-C00062
Figure USRE038132-20030603-C00063
Figure USRE038132-20030603-C00064
122-3
11
Figure USRE038132-20030603-C00065
Figure USRE038132-20030603-C00066
Figure USRE038132-20030603-C00067
197-8
12
Figure USRE038132-20030603-C00068
Figure USRE038132-20030603-C00069
Figure USRE038132-20030603-C00070
145-6
13
Figure USRE038132-20030603-C00071
Figure USRE038132-20030603-C00072
Figure USRE038132-20030603-C00073
 65-6
Method A is exemplified by the synthesis of the compound of Example 12 (see Table 1), which was carried out as follows. Homophenylalanine (1.00 g, 5.58 mmol) was dissolved in a solution of NaOH (0.28 g, 6.69 mmol) in H2O(10.0 mL) and heated at 45° C. When the solution became homogeneous, 3,5-dichlorophenyl isocyanate (1.05 g, 5.58 mmol) was added, and the mixture was heated at 45° C. for 2 h more. The cooled reaction mixture was then acidified with concentrated HCl to pH=2-3. The precipitate was collected by filtration, washed with water, and dried in vacuo at 50° C. to afford 0.85 g of the intermediate ureidoacetic acid (42%, crude yield). The intermediate was then taken up in a solution of concentrated HCl (5.0 mL) and water (5.0 mL) and heated under reflux for 5 h. The reaction mixture was then cooled to room temperature and the white solid was collected by suction filtration, washed with water, and dried in vacuo at 50° C. to afford 0.52 g of the crude hydantoin. This material was purified by recrystallization from EtOH to afford 0.37 g (45%) of the compound from Example 12.
Method B. Starting with an amino acid and a phenylisocyanate. Cyclization with EDC.
An appropriate amino acid is dissolved in aqueous base (such as, for example, NaOH, KOH, Na2CO3, NaHCO3, K2CO3 or KHCO3) and warmed to between about 20 and 90° C. An appropriate isocyanate is added to this mixture and the resulting solution is stirred until the reaction essentially reaches completion. Upon cooling, the mixture is acidified and the resulting ureidoacetic acid is isolated by filtration or extraction into organic solvent. Removal of solvent produces the intermediate ureidoacetic acid. The intermediate ureidoacetic acid is then cyclized to the desired hydantoin in organic solvent (such as, for example, DMF, NMP, or THF) using any of a number of dehydrating agents (such as, for example, dicyclohexylcarbodiimide (DCC) or 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide HCl (EDC)) in the presence of an ester activating agent (such as 1-hydroxybenzotriazole hydrate (HOBT)) and a non-nucleophilic base (such as, for example, triethylamine or N,N-diisopropylethylamine). Work-up consists of extraction into an organic solvent followed by purification via, for example, silica gel chromatography or recrystallization.
Compounds listed in Table 2 were produced via this general method.
TABLE 2
Examples of Compounds Synthesized by Method B.
STARTING M. P.
EX. STRUCTURE STARTING AMINO ACID ISOCYANATE (° C.)
14
Figure USRE038132-20030603-C00074
Figure USRE038132-20030603-C00075
Figure USRE038132-20030603-C00076
113-4
15
Figure USRE038132-20030603-C00077
Figure USRE038132-20030603-C00078
Figure USRE038132-20030603-C00079
114-5
16
Figure USRE038132-20030603-C00080
Figure USRE038132-20030603-C00081
Figure USRE038132-20030603-C00082
 96-7
17
Figure USRE038132-20030603-C00083
Figure USRE038132-20030603-C00084
Figure USRE038132-20030603-C00085
195-7
18
Figure USRE038132-20030603-C00086
Figure USRE038132-20030603-C00087
Figure USRE038132-20030603-C00088
145-6
19
Figure USRE038132-20030603-C00089
Figure USRE038132-20030603-C00090
Figure USRE038132-20030603-C00091
190-1
20
Figure USRE038132-20030603-C00092
Figure USRE038132-20030603-C00093
Figure USRE038132-20030603-C00094
 128-30
21
Figure USRE038132-20030603-C00095
Figure USRE038132-20030603-C00096
Figure USRE038132-20030603-C00097
158-9
22
Figure USRE038132-20030603-C00098
Figure USRE038132-20030603-C00099
Figure USRE038132-20030603-C00100
 116-26
23
Figure USRE038132-20030603-C00101
Figure USRE038132-20030603-C00102
Figure USRE038132-20030603-C00103
278-9
24
Figure USRE038132-20030603-C00104
Figure USRE038132-20030603-C00105
Figure USRE038132-20030603-C00106
181-3
25
Figure USRE038132-20030603-C00107
Figure USRE038132-20030603-C00108
Figure USRE038132-20030603-C00109
153-4
26
Figure USRE038132-20030603-C00110
Figure USRE038132-20030603-C00111
Figure USRE038132-20030603-C00112
167-8
27
Figure USRE038132-20030603-C00113
Figure USRE038132-20030603-C00114
Figure USRE038132-20030603-C00115
173-5
Method B is exemplified by the synthesis of the compound of Example 15 (see Table 2), which was carried out as follows: To a solution of (R)-phenylalanine (0.33 g, 2 mmol) in 1 mL of 2 N NaOH and 10 mL of water at 50° C. was added 3,5-dichlorophenyl isocyanate (0.38 g, 2 mmol). The resulting mixture was then stirred for 1 h. The solution was cooled and treated with concentrated HCl until a precipitate formed and the solution remained acidic. The precipitate was collected by filtration and dried in vacuo to produce the desired ureidoacetic acid (0.60 g, 85%). The ureidoacetic acid (0.35 g, 1 mmol) was dissolved in 20 mL of DMF and treated with EDC (0.19 g, 1 mmol) and HOBT (0.14 g, 1 mmol) for 1 h at room temperature. After this period N,N-diisopropylethylamine (0.35 mL, 2 mmol) was added and the mixture stirred overnight. Workup consisted of trituration with water, collection of the hydantoin by filtration, and purification by silica gel chromatography. The yield in this example was 0.20 g (60%).
Method C. Starting with an Amino Ester or a Hydroxy Ester and a Phenyllsocyanate. Cyclization with Base or Acid.
An appropriate amino ester or hydroxy ester and an appropriate isocyanate are dissolved in an organic solvent (such as, for example, DMF, TBF or DMSO) in the presence of a base (such as, for example, NaOH, KOH, Na2CO3, NaHCO3, K2CO3 or KHCO3) and warmed to between about room temperature and 60° C. After approximately 1 h, the temperature of the reaction mixture is raised to between about 50 and 100° C. until the reaction appears complete. The solution is then cooled and diluted with an organic solvent (such as, for example, EtOAc or CH2Cl2). The organic phase is washed sequentially with dilute aqueous acid (e.g. 1 N HCl) and water, dried (e.g. over MgSO4) and concentrated. The desired hydantoin is purified, for example by silica gel chromatography or by recrystallization. (Alternatively the ureidoacetic ester can be cyclized to the hydantoin by heating to between about 50 and 100° C. in the presence of an acid such as, for example, aqueous HCl until the reaction appears complete).
Compounds listed in Table 3 were produced via this general method.
TABLE 3
Examples of Compounds Synthesized by Method C.
STARTING M. P.
EX. STRUCTURE STARTING AMINO ESTER ISOCYANATE (° C.)
28
Figure USRE038132-20030603-C00116
Figure USRE038132-20030603-C00117
Figure USRE038132-20030603-C00118
200-2
29
Figure USRE038132-20030603-C00119
Figure USRE038132-20030603-C00120
Figure USRE038132-20030603-C00121
 63-5
30
Figure USRE038132-20030603-C00122
Figure USRE038132-20030603-C00123
Figure USRE038132-20030603-C00124
162-4
31
Figure USRE038132-20030603-C00125
Figure USRE038132-20030603-C00126
Figure USRE038132-20030603-C00127
157-8
32
Figure USRE038132-20030603-C00128
Figure USRE038132-20030603-C00129
Figure USRE038132-20030603-C00130
203-5
33
Figure USRE038132-20030603-C00131
Figure USRE038132-20030603-C00132
Figure USRE038132-20030603-C00133
oil
34
Figure USRE038132-20030603-C00134
Figure USRE038132-20030603-C00135
Figure USRE038132-20030603-C00136
108-9
35
Figure USRE038132-20030603-C00137
Figure USRE038132-20030603-C00138
Figure USRE038132-20030603-C00139
105-6
36
Figure USRE038132-20030603-C00140
Figure USRE038132-20030603-C00141
Figure USRE038132-20030603-C00142
 58-60
37
Figure USRE038132-20030603-C00143
Figure USRE038132-20030603-C00144
Figure USRE038132-20030603-C00145
 92-3
38
Figure USRE038132-20030603-C00146
Figure USRE038132-20030603-C00147
Figure USRE038132-20030603-C00148
194-5
39
Figure USRE038132-20030603-C00149
Figure USRE038132-20030603-C00150
Figure USRE038132-20030603-C00151
135-6
40
Figure USRE038132-20030603-C00152
Figure USRE038132-20030603-C00153
Figure USRE038132-20030603-C00154
157-8
41
Figure USRE038132-20030603-C00155
Figure USRE038132-20030603-C00156
Figure USRE038132-20030603-C00157
 72-4
42
Figure USRE038132-20030603-C00158
Figure USRE038132-20030603-C00159
Figure USRE038132-20030603-C00160
143-4
43
Figure USRE038132-20030603-C00161
Figure USRE038132-20030603-C00162
Figure USRE038132-20030603-C00163
 73-4
44
Figure USRE038132-20030603-C00164
Figure USRE038132-20030603-C00165
Figure USRE038132-20030603-C00166
oil
45
Figure USRE038132-20030603-C00167
Figure USRE038132-20030603-C00168
Figure USRE038132-20030603-C00169
not determ.
Method C is exemplified by the synthesis of the compound of Example 30 of Table 3, which is as follows: Methyl 2-amino-2-benzylbutyric acid (0.21 g, 1 mmol) and 3,5-dichlorophenyl isocyanate (0.19 g, 1 mmol) were dissolved in DMSO (5 mL) in the presence of approximately 0.2 g of Na2CO3 and allowed to stir at 50° C. for 1 h. After this period the solution was heated to 90° C. for 2 hr. The solution was then cooled, diluted with EtOAc and washed with 0.1 N HCl and water. The organic layer was dried over MgSO4 and concentrated to produce a crude product which was further purified by silica gel chromatography to yield 0.12 g (33%) of the compound of example 30.
Method D. Solid Phase Synthesis.
There are several examples in the literature which demonstrate that the synthesis of hydantoins and their precursor amino acid derivatives can be performed in the solid phase which may make the synthesis of large varieties of these compounds amenable to an automated approach. Examples for the synthesis of the precursor amino acid derivatives are shown in the following citations: J. American Chemical Society, 1996, 118, 6070-1, Tetrahedron Letters, 1997, 38, 7163-7166, Tetrahedron Letters, 1997, 38, 8821. An literature citation which demonstrates the conversion of these amino acid derivatives to hydantoins is J. Organic Chemistry 1997, 62, 6060-2.
An amino acid attached to a solid phase resin through its carboxylic acid via an appropriate linker (for example the Wang resin: 4-benzyloxy-benzyl polystyrene) is protected on its nitrogen with a reagent that will allow for the alkylation of the alpha-carbon (for example, a benzaldehyde derivative that forms an imine with the nitrogen of the amino acid). The protected compound is then treated with a base and an alkylating agent to generate the new protected amino acid derivative. The protecting group is removed using standard conditions (in the case of an imine this is accomplished, for example, with aqueous HCl) and the free amino group is reacted with an isocyanate to generate the intermediate urea. This intermediate is treated with a reagent to catalyze the cyclization of the urea portion onto the carboxylate end of the molecule which forms the desired hydantoin and cleaves the product from the resin. Purification is via silica gel chromatography, reverse phase HPLC of recrystallization.
Compounds listed in Table 4 were produced via this general method.
TABLE 4
Examples of Compounds Synthesized by Method D.
STARTING M. P.
EX. STRUCTURE ALKYLATING AGENT ISOCYANATE (° C.)
46
Figure USRE038132-20030603-C00170
Figure USRE038132-20030603-C00171
Figure USRE038132-20030603-C00172
Not determ.
47
Figure USRE038132-20030603-C00173
Figure USRE038132-20030603-C00174
Figure USRE038132-20030603-C00175
Not determ.
48
Figure USRE038132-20030603-C00176
Figure USRE038132-20030603-C00177
Figure USRE038132-20030603-C00178
Not determ.
49
Figure USRE038132-20030603-C00179
Figure USRE038132-20030603-C00180
Figure USRE038132-20030603-C00181
 61-3
50
Figure USRE038132-20030603-C00182
Figure USRE038132-20030603-C00183
Figure USRE038132-20030603-C00184
>240
51
Figure USRE038132-20030603-C00185
Figure USRE038132-20030603-C00186
Figure USRE038132-20030603-C00187
Not determ.
52
Figure USRE038132-20030603-C00188
Figure USRE038132-20030603-C00189
Figure USRE038132-20030603-C00190
Not determ.
53
Figure USRE038132-20030603-C00191
Figure USRE038132-20030603-C00192
Figure USRE038132-20030603-C00193
Not determ.
54
Figure USRE038132-20030603-C00194
Figure USRE038132-20030603-C00195
Figure USRE038132-20030603-C00196
117-8
55
Figure USRE038132-20030603-C00197
Figure USRE038132-20030603-C00198
Figure USRE038132-20030603-C00199
150-1
56
Figure USRE038132-20030603-C00200
Figure USRE038132-20030603-C00201
Figure USRE038132-20030603-C00202
Not determ.
57
Figure USRE038132-20030603-C00203
Figure USRE038132-20030603-C00204
Figure USRE038132-20030603-C00205
Not determ.
58
Figure USRE038132-20030603-C00206
Figure USRE038132-20030603-C00207
Figure USRE038132-20030603-C00208
Not determ.
59
Figure USRE038132-20030603-C00209
Figure USRE038132-20030603-C00210
Figure USRE038132-20030603-C00211
173-4
60
Figure USRE038132-20030603-C00212
Figure USRE038132-20030603-C00213
Figure USRE038132-20030603-C00214
133-4
61
Figure USRE038132-20030603-C00215
Figure USRE038132-20030603-C00216
Figure USRE038132-20030603-C00217
122-3
62
Figure USRE038132-20030603-C00218
Figure USRE038132-20030603-C00219
Figure USRE038132-20030603-C00220
 67-9
63
Figure USRE038132-20030603-C00221
Figure USRE038132-20030603-C00222
Figure USRE038132-20030603-C00223
Not determ.
64
Figure USRE038132-20030603-C00224
Figure USRE038132-20030603-C00225
Figure USRE038132-20030603-C00226
 55-6
65
Figure USRE038132-20030603-C00227
Figure USRE038132-20030603-C00228
Figure USRE038132-20030603-C00229
170-1
66
Figure USRE038132-20030603-C00230
Figure USRE038132-20030603-C00231
Figure USRE038132-20030603-C00232
153-5
67
Figure USRE038132-20030603-C00233
Figure USRE038132-20030603-C00234
Figure USRE038132-20030603-C00235
163-4
68
Figure USRE038132-20030603-C00236
Figure USRE038132-20030603-C00237
Figure USRE038132-20030603-C00238
Not determ.
69
Figure USRE038132-20030603-C00239
Figure USRE038132-20030603-C00240
Figure USRE038132-20030603-C00241
 168-70
Method D is exemplified by the synthesis of the compound of Example 67, which was carried out as follows: A reaction vessel was charged with the commercially available Fmoc-Ala-Wang (300 mg, 0.150 mmol) and 3 mL of a 20% solution of piperidine in N-methyl pyrollidinone (NMP). The reaction vessel was agitated at room temperature on an orbital shaker for 45 minutes. The resin was filtered and washed with NMP (3×1 mL). The reaction vessel containing resin was equipped with a rubber septum, placed under argon, charged with 3,4-dichlorobenzaldehyde (394 mg, 2.25 mmol), trimethyl orthoformate (3.5 mL), and NMP (1.5 mL). The resulting mixture was agitated at room temperature for 15 h. The solid resin was isolated by filtration and washed sequentially with NMP (3×3 mL), tetrahydrofuran (3×mL), and CH2Cl2 (3×mL). The resin was then dried under vacuum for approximately one hour to produce the imine-resin intermediate.
The imine-resin intermediate was alkylated with 2,3-difluoro-4-trifluormethylbenzyl bromide (123.8 mg, 0.45 mmol) by mixing these two reagents, 2-tert-butylimino-2-diethylamino-1,3-dimethylperhydro-1,3,2-diazaphosphorine (BEMP, 0.217 mL, 0.75 mmol), and NMP (3.5 mL) and agitating the mixture at room temperature on an orbital shaker for 15 h. The solid was isolated by filtration and washed sequentially with NMP (3×mL), THF (3×mL), and CH2Cl2 (3×3 mL) yielding the alkylated-imine-resin intermediate upon drying.
The imine was cleaved from the preceding intermediate by treatment with aqueous 1 N HCl (1.8 mL) and THF (3.6 mL) and agitating at room temperature for about 15 h. The resin bound amino ester was isolated by filtration and washed sequentially with NMP (3×3 mL), THF (3×3 mL), and CH2Cl2 (3×3 mL) and dried under vacuum.
The resin bound amino ester was converted to the hydantoin using a procedure that cleaves the final product from the resin. The intermediate amino-ester was placed in a reaction vessel and treated with 3 mL of a 20% solution of N,N-diisopropylethylamine in NMP. After agitation at room temperature under argon for 1 h, the resin was filtered, washed with NMP (3×3 mL) and methanol (3×3 mL), and placed under vacuum. Subsequently, the vessel was opened under argon and charged with 2.5 mL of a 1.75 M solution of 3,5-dichlorphenylisocyanate in dimethylformamide (DMF, 0.45 mmol). The mixture was agitated at room temperature under argon overnight and the product removed from the resin by filtration. After the resin was washed ethyl acetate (6×2 mL), the combined organic solutions were diluted with water and then washed with water (3×3 mL) and saturated aqueous NaCl (2×3 mL), dried over sodium sulfate, filtered, and concentrated under a stream of nitrogen. Final purification was accomplished using reverse phase HPLC (acetonitrile-water gradient).
Method E. Starting with an Isocyanateester and an Aniline. Cyclization with base or Acid.
To an appropriate isocyante ester, dissolved in an organic solvent (such as, for, example, methylene chloride) is added an appropriate aniline, and the mixture is stirred for between about 1 and 24 h, at about room temperature, under an inert atmosphere, such as argon. The organic solvent is is then removed in vacua. Excess aniline is removed (as by boiling the crude solid in hexanes and decanting off the liquid, or by flash chromatography over silica gel) leaving the solid ureidoacetic ester. The ureidoacetic ester is cyclized to the desired hydantoin by treatment with base (such as, for example, NaH, NaHMDS, Na2CO3, NaHCO3, K2CO3 or KHCO3) in an organic solvent (such as, for example, THF or DMF), followed by warming to approximately 60-90° C. The solution is next cooled and diluted with an organic solvent (such as, for example, EtOAc). The organic solution is washed sequentially with dilute aqueous acid (such as 1 N HCl) and then water, dried (as with MgSO4) and concentrated. The desired hydantoin is purified, as by silica gel chromatography or recrystallization. (Alternatively the intermediate ureidoacetic ester can be cyclized to the hydantoin by heating to about 90° C. in the presence of an acid, such as aqueous HCl, as mentioned in method C).
The compounds listed in Table 5 were produced via this method.
TABLE 5
Example of Compound Synthesized by Method E.
STARTING STARTING M. P.
EX. STRUCTURE ISOCYANATE ESTER ANILINE (° C.)
70
Figure USRE038132-20030603-C00242
Figure USRE038132-20030603-C00243
Figure USRE038132-20030603-C00244
178-9
71
Figure USRE038132-20030603-C00245
Figure USRE038132-20030603-C00246
Figure USRE038132-20030603-C00247
145-6
Method E is exemplified by the synthesis of the compound from example 70, shown in Table 5, which was carried out as follows: To a solution of ethyl 2-isocyanato-3-phenylpropionate (99.0 mL, 0.110 g, 0.501 mmol) in dry CH2Cl2 (5.0 mL) was added 3,4,5-trichloroaniline (0.1952 g, 0.994 mmol) as a solid. The mixture was stirred at room temperature under an argon atmosphere for 20 h. The solution was then concentrated in vacuo and the residue was recrystallized two times from ethyl acetate/hexanes to give 0.14 g (65%) of the pure intermediate urea as a white solid. A suspension of sodium hydride (0.06 g 60% dispersion in mineral oil, 1.52 mmol) in dry THF (4.0 mL) was treated with a solution of the above urea (0.108 g, 0.260 mmol) in dry THF (4.0 mL). The mixture was stirred at room temperature under an argon atmosphere for 1 h. The mixture was next poured into 100 mL 1 N aqueous HCl. The THF was removed under reduced pressure and the mixture was filtered. The solid was purified by preparative thin layer chromatography (SiO2, 1:1 hexanes/ethyl acetate) to give a white solid which was further purified by recrystallization from absolute EtOH to give 0.027 g of pure compound (28%).
Method F. Synthesis of Succinimides.
Equimolar amounts of the an appropriate starting diacid or anhydride and an appropriate starting aniline are refluxed in a solvent (such as xylene) in the presence of a catalytic amount of base (such as triethylamine) for between about 2 and 24 h. The solvent is removed in vacuo and the residue is dissolved in an organic solvent (such as EtOAc), washed sequentially with aqueous dilute base (such as NaHCO3) and aqueous dilute acid (such as HCl), dried (for example over MgSO4), and concentrated. Purification is performed via, for example, recrystallization or chromatography over silica gel.
The starting diacids and anhydrides are available either commercially or via a number of known literature methods. By way of example, a procedure for the synthesis of 2-benzyl-3-carboxy-2-methylbutanoic acid (the starting material for example 74) is given.
A solution of 2.0 g of 2-methyl-3-phenylpropanoic acid (12.2 mmol), 2.2 g of carbonyl-diimidazole (CDI, 13.56 mmol) in 20 mL of THF was refluxed under nitrogen for 1 h. The temperature was reduced to 50° C. and 1.2 mL of crotyl alcohol (14.1 mmol) was added followed by 20 mg of 4-(N,N-dimethylamino)-pyridine (DMAP). The mixture was heated at 50° C. for 3 h, concentrated and purified by silica gel chromatography to give 1.7 g of the intermediate ester: trans-2-butenyl 2-benzyl-3-carboxy-2-methylbutanoate (64%).
The ester was subjected to a [3,3] sigmatropic rearrangement to produce the next intermediate. Under argon, at −78° C., a solution of 560 mg of the intermediate ester (2.57 mmol) in THF (1 mL) was added to a THF solution of lithium di-isopropylamide (LDA, 3.25 mmol, generated from 1.3 mL of 2.5 M n-BuLi and 0.54 mL of iPr2NH in 3 mL of THF, −10° C., 15 min) containing 500 microliter of DMPU. The mixture was stirred for 30 min before a solution of 480 mg of TBSCl (3.1 mmol) in 1 mL of THF was added. The mixture was stirred at −78° C. for 30 min, at room temperature for 20 min and then heated at 60° C. for 10 h. The mixture was cooled to 0° C., quenched with 2 N HCl (5 mL) and stirred at room temperature for 10 h. The mixture was made basic with 2 N NaOH to pH 10, extracted with ether (5 mL). The aqueous layer was separated, acidified to pH 1 with concentrated HCl, extracted with EtOAc and concentrated to give 500 mg (89%) of the intermediate: 2-benzyl-2,3-dimethyl-4-pentenoic acid.
The mono-acid was converted to the desired diacid by oxidation of the terminal alkene with ozone and the resulting intermediate further oxidation with a chromium reagent. Through a solution of 500 mg of 2-benzyl-2,3-dimethyl-4-pentenoic acid (2.29 mmol) in MeOH (20 mL) and methylene chloride (10 mL) containing 120 microliter of pyridine was passed rapidly enough of a stream of O3 at −78° C., such that the solution turned slightly blue. The mixture was treated with 1 mL of methyl sulfide and stirred at −78° C. for 5 min. The mixture was then warmed to room temperature, concentrated and passed through a silica gel column (with 10% MeOH in CH2Cl2 as eluting solvent) and concentrated. The crude material was dissolved in 5 mL of acetone and treated with Jones reagent (16 g CrO3 16 g con. H2SO4 in 100 mL of H2O) at room temperature until the orange color persisted. After addition of water (10 mL), the mixture was stirred for 1 h, washed with EtOAc and concentrated. The mixture was purified by silica gel chromatography with 3% AcOH-EtOAc to give 300 mg of the desired diacid (55%).
Compounds listed in Table 6 were produced via this method.
TABLE 6
Examples of Compounds Synthesized by Method F.
MELTING POINT
EX. STRUCTURE STARTING MATERIAL (° C.)
72
Figure USRE038132-20030603-C00248
Figure USRE038132-20030603-C00249
oil
73
Figure USRE038132-20030603-C00250
Figure USRE038132-20030603-C00251
 139-40
74
Figure USRE038132-20030603-C00252
Figure USRE038132-20030603-C00253
111-2
75
Figure USRE038132-20030603-C00254
Figure USRE038132-20030603-C00255
104-5
76
Figure USRE038132-20030603-C00256
Figure USRE038132-20030603-C00257
112-3
77
Figure USRE038132-20030603-C00258
Figure USRE038132-20030603-C00259
not determined
Method F is exemplified by the synthesis of the compounds of Examples 92 and 93 (see Table 6) which was carried out as follows: A mixture of isomers of the starting diacid of example 92 (0.58 g, 1.8 mmol, 3:1 mixture of isomers), 3,5-dichloroaniline (0.35 g, 2.2 mmol), Et3N (10 mL, 0.07 mmol) in xylene (5 mL) was refluxed under argon in a flask fitted with Dean-Stark trap for 24 h. The mixture was cooled, concentrated and purified by silica gel chromatography (with 10% then with 15% ethyl acetate in hexanes as the eluting solvent) to give 0.45 g (52%) of trans-methyl isomer (example 73, mp 139-140° C.) and 15 mg (2%) of the cis-methyl isomer (example 72, mp=oil).
Method G. Conversion of Carbonyls to Thio-carbonyls
Several reagents are known in the literature which will convert carbonyls to thio carbonyls. A typical sequence involves heating the substrate with a reagent such as P2S3 in a high boiling solvent such as tetralin for between 1 and 48 h. Isolation of the product follows relatively standard conditions such as the dilution of the mixture into an organic solvent such as EtOAc and washing this mixture with water and saturated aqueous NaCl followed by drying and concentration. Purification is accomplished by silica gel chromatography or recrystallization, to afford the desired product.
Compounds listed in Table 7 were produced via this general method.
TABLE 7
Examples of Compounds Synthesized by Method G.
MELTING POINT
EXAMPLE STRUCTURE STARTING MATERIAL (° C.)
78
Figure USRE038132-20030603-C00260
Figure USRE038132-20030603-C00261
197-8
79
Figure USRE038132-20030603-C00262
Figure USRE038132-20030603-C00263
153-4
Method G is exemplified by the synthesis of the compound of Example 78, which was carried out as follows: The starting substrate (1.5 g, 3.5 mmol) was dissolved in 5 mL of tetralin, treated with P2S3 (0.9 g, 5.7 mmol) and heated to 225° C. for 2 h. Upon cooling, the mixture was diluted with water and the product was extracted into EtOAc. The organic layer was washed with saturated aqueous NaCl, dried and concentrated. The residual oil was triturated with hexanes to produce a yellow solid which was isolated by filtration. This material was further purified by flash chromatography (1:4 EtOAc:Hexanes) to afford 1.13 g (70%%) of the desired compound.
Method H. Selective Hydrolysis of Thio-carbonyls to Carbonyls
The dithio-carbonyl containing compounds produced via Method G can be selectively hydrolyzed to each of the two monothio-carbonyl compounds depending on the choice of conditions. In general the thio-carbonyl at the 4-position of the ring is more susceptible to nucleophilic conditions. As shown in Example 81, it can be converted to the 4-oxo-species by treatment with aqueous ethanolamine followed by acid hydrolysis. The thio-carbonyl at the 2-position of the ring is more nucleophilic at sulfur and can be alkylated with methyl sulfate. This intermediate can then be hydrolyzed with mild acid. This affords the compound of Example 80. Purification of either class of compound is easily performed by silica gel chromatography or recrystallization.
Compounds listed in Table 8 were produced via this general method.
TABLE 8
Examples of Compounds Synthesized by Method H.
REACTION M. P.
EX. STRUCTURE STARTING MATERIAL CONDITIONS (° C.)
80
Figure USRE038132-20030603-C00264
Figure USRE038132-20030603-C00265
1. H2N(CH2)2OH, THF, 100° C. 2. 6NHCl, 100° C. 153-4
81
Figure USRE038132-20030603-C00266
Figure USRE038132-20030603-C00267
1. NaOH, Me2Se4, 0° C. 2. 6NHCl, 100° C. 174-5
Example 80 was prepared by treating a solution of the starting material (0.23 g, 0.49 mmol) in 3 mL of THF with aqueous with 10 mL of 50% aqueous ethanolamine and heating under reflux for 2 h. Upon cooling, the mixture was extracted with EtOAc and the organic layer was washed with water and saturated aqueous NaCl, dried and concentrated to give a brown solid. This solid was then treated with 20 mL of 6 N HCl and heated under reflux for 72 h. Upon cooling, the mixture was extracted with EtOAc and the organic layer was washed with saturated aqueous NaCl, dried and concentrated. The product was purified by preparative TLC over silica gel using 1:1 EtOAc:Hexanes as the solvent to produce the product in 34% yield.
Example 81 was prepared by treating a solution of the starting material (0.5 g, 1.09 mmol) in 1.6 mL of 2 N NaOH. As the compound did not initially dissolve, 1 mL of water and 1 mL of THF were added to aid solubility. This mixture was then cooled in an ice bath and Me2SO4 (0.12 mL, 1.3 mmol) was added dropwise over 5 min. The mixture was stirred another 3 h at 0° C. and then for 45 min at room temperature. The reaction was quenched by the addition of enough 1 N HCl needed to lower the pH of the solution to 2. The mixture was extracted with EtOAc and the organic layer was washed with saturated aqueous NaCl, dried and concentrated to give a yellow oil. This oil was then treated with 10 mL of 6 N HCl and heated under reflux for 3 h. Upon cooling, the mixture was extracted with EtOAc and the organic layer was washed with saturated aqueous NaCl, dried and concentrated. The product was purified by column chromatography over silica gel using 1:1 EtOAc:Hexanes as the solvent to produce the product in 5% yield.
Method I. N-Alkylation of a Hydantoin.
An appropriate hydantoin is dissolved in an aprotic solvent (such as, for example, DMF, THF or DMSO) and treated with one equivalent of a base (such as, for example, NaH, LDA, LiHMDS, KHMDS, KH or NaHMDS). After about 10 min to 1 h an appropriate alkylating agent is added and the mixture stirred at between about room temperature and 90° C. for up to about 24 h. (Progress of the reaction can be monitored using TLC). The solution is then cooled and diluted with an organic solvent (such as, for example, EtOAc or CH2Cl2). The organic phase is washed sequentially with a dilute acid (such as 1 N HCl) and water, dried (for example over MgSO4) and concentrated. The desired hydantoin is purified, as by silica gel chromatography or by recrystallization.
Compounds listed in Table 9 were produced via this general method.
TABLE 9
Examples of Compounds Synthesized by Method I.
EX. STRUCTURE STARTING HYDANTOIN STARTING ALKYLATING AGENT M.P. (° C.)
82
Figure USRE038132-20030603-C00268
Figure USRE038132-20030603-C00269
CH3I 118-20
83
Figure USRE038132-20030603-C00270
Figure USRE038132-20030603-C00271
CH3I 113-4
84
Figure USRE038132-20030603-C00272
Figure USRE038132-20030603-C00273
PhCH2Br  38-40
85
Figure USRE038132-20030603-C00274
Figure USRE038132-20030603-C00275
CH3I oil
86
Figure USRE038132-20030603-C00276
Figure USRE038132-20030603-C00277
CH3I oil
87
Figure USRE038132-20030603-C00278
Figure USRE038132-20030603-C00279
CH3I 114-5
88
Figure USRE038132-20030603-C00280
Figure USRE038132-20030603-C00281
CH3I 112-4
89
Figure USRE038132-20030603-C00282
Figure USRE038132-20030603-C00283
CH3CH2I  107-11
90
Figure USRE038132-20030603-C00284
Figure USRE038132-20030603-C00285
CH3Ch2CH2I oil
91
Figure USRE038132-20030603-C00286
Figure USRE038132-20030603-C00287
Figure USRE038132-20030603-C00288
121-3
92
Figure USRE038132-20030603-C00289
Figure USRE038132-20030603-C00290
CH3I 143-5
93
Figure USRE038132-20030603-C00291
Figure USRE038132-20030603-C00292
CH3I 112-3
94
Figure USRE038132-20030603-C00293
Figure USRE038132-20030603-C00294
CH3I 102-4
95
Figure USRE038132-20030603-C00295
Figure USRE038132-20030603-C00296
CH3I  65-7
96
Figure USRE038132-20030603-C00297
Figure USRE038132-20030603-C00298
CH3COCl 110-1
97
Figure USRE038132-20030603-C00299
Figure USRE038132-20030603-C00300
CH3CH2I  57-9
98
Figure USRE038132-20030603-C00301
Figure USRE038132-20030603-C00302
CH3CH2CH2Br  51-3
99
Figure USRE038132-20030603-C00303
Figure USRE038132-20030603-C00304
Cl(CH2)2NMe2 oil
100
Figure USRE038132-20030603-C00305
Figure USRE038132-20030603-C00306
CH3I  58-60
101
Figure USRE038132-20030603-C00307
Figure USRE038132-20030603-C00308
CH3I  60-2
102
Figure USRE038132-20030603-C00309
Figure USRE038132-20030603-C00310
CH3COCl 111-3
103
Figure USRE038132-20030603-C00311
Figure USRE038132-20030603-C00312
Figure USRE038132-20030603-C00313
 78-9
104
Figure USRE038132-20030603-C00314
Figure USRE038132-20030603-C00315
Figure USRE038132-20030603-C00316
148-50
105
Figure USRE038132-20030603-C00317
Figure USRE038132-20030603-C00318
CH3CH2I 135-6
106
Figure USRE038132-20030603-C00319
Figure USRE038132-20030603-C00320
CH3CH2CH2Br 104-6
107
Figure USRE038132-20030603-C00321
Figure USRE038132-20030603-C00322
Figure USRE038132-20030603-C00323
 78-80
108
Figure USRE038132-20030603-C00324
Figure USRE038132-20030603-C00325
Cl(CH2)2NMe2 236-7
109
Figure USRE038132-20030603-C00326
Figure USRE038132-20030603-C00327
CD3I 136-8
110
Figure USRE038132-20030603-C00328
Figure USRE038132-20030603-C00329
Figure USRE038132-20030603-C00330
129-30
111
Figure USRE038132-20030603-C00331
Figure USRE038132-20030603-C00332
Figure USRE038132-20030603-C00333
 69-70
112
Figure USRE038132-20030603-C00334
Figure USRE038132-20030603-C00335
Figure USRE038132-20030603-C00336
 66-67
113
Figure USRE038132-20030603-C00337
Figure USRE038132-20030603-C00338
Figure USRE038132-20030603-C00339
104-6
114
Figure USRE038132-20030603-C00340
Figure USRE038132-20030603-C00341
Figure USRE038132-20030603-C00342
 84-5
115
Figure USRE038132-20030603-C00343
Figure USRE038132-20030603-C00344
Figure USRE038132-20030603-C00345
128-9
116
Figure USRE038132-20030603-C00346
Figure USRE038132-20030603-C00347
Figure USRE038132-20030603-C00348
 52-4
117
Figure USRE038132-20030603-C00349
Figure USRE038132-20030603-C00350
Figure USRE038132-20030603-C00351
 63-4
118
Figure USRE038132-20030603-C00352
Figure USRE038132-20030603-C00353
CH3I  94-5
119
Figure USRE038132-20030603-C00354
Figure USRE038132-20030603-C00355
CH3I 135-6
120
Figure USRE038132-20030603-C00356
Figure USRE038132-20030603-C00357
Cl(CH2)6Br 104-5
121
Figure USRE038132-20030603-C00358
Figure USRE038132-20030603-C00359
Cl(CH2)5Br 141-2
122
Figure USRE038132-20030603-C00360
Figure USRE038132-20030603-C00361
CH3I 128-32
123
Figure USRE038132-20030603-C00362
Figure USRE038132-20030603-C00363
CH3I  54-5
124
Figure USRE038132-20030603-C00364
Figure USRE038132-20030603-C00365
CH3I  71-5
125
Figure USRE038132-20030603-C00366
Figure USRE038132-20030603-C00367
CH3COCl  57-59
126
Figure USRE038132-20030603-C00368
Figure USRE038132-20030603-C00369
Figure USRE038132-20030603-C00370
126-7
127
Figure USRE038132-20030603-C00371
Figure USRE038132-20030603-C00372
CH3I 114-5
128
Figure USRE038132-20030603-C00373
Figure USRE038132-20030603-C00374
CH3COCl 153-4
129
Figure USRE038132-20030603-C00375
Figure USRE038132-20030603-C00376
Cl(CH2)2OCH3 oil
130
Figure USRE038132-20030603-C00377
Figure USRE038132-20030603-C00378
Figure USRE038132-20030603-C00379
 56-7
131
Figure USRE038132-20030603-C00380
Figure USRE038132-20030603-C00381
ClCH2OCH3 oil
132
Figure USRE038132-20030603-C00382
Figure USRE038132-20030603-C00383
Figure USRE038132-20030603-C00384
 76-78
133
Figure USRE038132-20030603-C00385
Figure USRE038132-20030603-C00386
Figure USRE038132-20030603-C00387
 76-78
134
Figure USRE038132-20030603-C00388
Figure USRE038132-20030603-C00389
CH3I oil
135
Figure USRE038132-20030603-C00390
Figure USRE038132-20030603-C00391
CH3COCl 105-7
136
Figure USRE038132-20030603-C00392
Figure USRE038132-20030603-C00393
CH3SO2Cl 141-3
137
Figure USRE038132-20030603-C00394
Figure USRE038132-20030603-C00395
CH3I  53-5
Method I is exemplified by the synthesis of the compound of Example 97 (see Table 9), which was carried out by dissolving the starting material (0.21 g, 0.5 mmol) in DMF (5 mL) and treating the solution sequentially a solution of 1 M NAHMDS (0.5 mL, 0.5 mmol) and EtI (0.04 mL, 0.5 mmol). After 1 h, the reaction mixture was partitioned between EtOAc and water, and the organic phase washed with water and dried over MgSO4. Column chromatography over silica gel produced 0.17 g (72%) of the desired product.
Method J. C-Alkylation of a Heterocycle.
An appropriate heterocycle is dissolved in an aprotic solvent (such as DMF, TBF or DMSO) and treated with one equivalent of a base, (such as Et3N, LDA, KHMDS, LiHMDS or NaHMDS) at between about −78° C. and room temperature. After about 10 min to 2 h an appropriate alkylating agent is added and the mixture stirred at between about 0 and 90° C. for up to about 24 h. (Progress of the reaction can be monitored using TLC). The solution is then cooled and diluted with an organic solvent (such as, for example, EtOAc). The organic phase is washed sequentially with dilute aqueous acid (such as 1 NHCl), and with water, dried (for example, over MgSO4) and concentrated. The desired hydantoin is purified, as by silica gel chromatography or by recrystallization.
Compounds listed in Table 10 were produced via this general method.
TABLE 10
Examples of Compounds Synthesized by Method J.
STARTING
AKLYLATING
EX. STRUCTURE STARTING HYDANTOIN AGENT M.P. (° C.)
138
Figure USRE038132-20030603-C00396
Figure USRE038132-20030603-C00397
CH3I not determined
139
Figure USRE038132-20030603-C00398
Figure USRE038132-20030603-C00399
CH3I oil
140
Figure USRE038132-20030603-C00400
Figure USRE038132-20030603-C00401
Figure USRE038132-20030603-C00402
 48-50
141
Figure USRE038132-20030603-C00403
Figure USRE038132-20030603-C00404
Figure USRE038132-20030603-C00405
125-7
142
Figure USRE038132-20030603-C00406
Figure USRE038132-20030603-C00407
Figure USRE038132-20030603-C00408
 95-6
143
Figure USRE038132-20030603-C00409
Figure USRE038132-20030603-C00410
Figure USRE038132-20030603-C00411
 97-8
144
Figure USRE038132-20030603-C00412
Figure USRE038132-20030603-C00413
Figure USRE038132-20030603-C00414
106-7
145
Figure USRE038132-20030603-C00415
Figure USRE038132-20030603-C00416
Figure USRE038132-20030603-C00417
105-6
146
Figure USRE038132-20030603-C00418
Figure USRE038132-20030603-C00419
Figure USRE038132-20030603-C00420
110-2
147
Figure USRE038132-20030603-C00421
Figure USRE038132-20030603-C00422
Figure USRE038132-20030603-C00423
106-8
148
Figure USRE038132-20030603-C00424
Figure USRE038132-20030603-C00425
Figure USRE038132-20030603-C00426
 82-3
149
Figure USRE038132-20030603-C00427
Figure USRE038132-20030603-C00428
Figure USRE038132-20030603-C00429
124-6
150
Figure USRE038132-20030603-C00430
Figure USRE038132-20030603-C00431
Figure USRE038132-20030603-C00432
oil
151
Figure USRE038132-20030603-C00433
Figure USRE038132-20030603-C00434
Figure USRE038132-20030603-C00435
166-8
152
Figure USRE038132-20030603-C00436
Figure USRE038132-20030603-C00437
Figure USRE038132-20030603-C00438
 62-4
153
Figure USRE038132-20030603-C00439
Figure USRE038132-20030603-C00440
Figure USRE038132-20030603-C00441
124-6
154
Figure USRE038132-20030603-C00442
Figure USRE038132-20030603-C00443
Figure USRE038132-20030603-C00444
 96-8
155
Figure USRE038132-20030603-C00445
Figure USRE038132-20030603-C00446
Figure USRE038132-20030603-C00447
105-7
156
Figure USRE038132-20030603-C00448
Figure USRE038132-20030603-C00449
Figure USRE038132-20030603-C00450
106-8
157
Figure USRE038132-20030603-C00451
Figure USRE038132-20030603-C00452
Figure USRE038132-20030603-C00453
 50-2
158
Figure USRE038132-20030603-C00454
Figure USRE038132-20030603-C00455
CH3 I not determ.
159
Figure USRE038132-20030603-C00456
Figure USRE038132-20030603-C00457
Figure USRE038132-20030603-C00458
160-1
160
Figure USRE038132-20030603-C00459
Figure USRE038132-20030603-C00460
Figure USRE038132-20030603-C00461
166-7
161
Figure USRE038132-20030603-C00462
Figure USRE038132-20030603-C00463
Figure USRE038132-20030603-C00464
 65-66
162
Figure USRE038132-20030603-C00465
Figure USRE038132-20030603-C00466
Figure USRE038132-20030603-C00467
143-4
163
Figure USRE038132-20030603-C00468
Figure USRE038132-20030603-C00469
Figure USRE038132-20030603-C00470
not determined
164
Figure USRE038132-20030603-C00471
Figure USRE038132-20030603-C00472
Figure USRE038132-20030603-C00473
138-9
165
Figure USRE038132-20030603-C00474
Figure USRE038132-20030603-C00475
Figure USRE038132-20030603-C00476
114-5
166
Figure USRE038132-20030603-C00477
Figure USRE038132-20030603-C00478
Figure USRE038132-20030603-C00479
125-6
167
Figure USRE038132-20030603-C00480
Figure USRE038132-20030603-C00481
Figure USRE038132-20030603-C00482
 37-9
168
Figure USRE038132-20030603-C00483
Figure USRE038132-20030603-C00484
CH3 I  53-5
169
Figure USRE038132-20030603-C00485
Figure USRE038132-20030603-C00486
Figure USRE038132-20030603-C00487
 98-100
170
Figure USRE038132-20030603-C00488
Figure USRE038132-20030603-C00489
Figure USRE038132-20030603-C00490
 65-66
171
Figure USRE038132-20030603-C00491
Figure USRE038132-20030603-C00492
Figure USRE038132-20030603-C00493
120-1
172
Figure USRE038132-20030603-C00494
Figure USRE038132-20030603-C00495
CH3 CH2I  55-6
173
Figure USRE038132-20030603-C00496
Figure USRE038132-20030603-C00497
CH3 CH2CH2Br  51-3
174
Figure USRE038132-20030603-C00498
Figure USRE038132-20030603-C00499
Figure USRE038132-20030603-C00500
127-8
175
Figure USRE038132-20030603-C00501
Figure USRE038132-20030603-C00502
ClCH2OCH3  54-5
176
Figure USRE038132-20030603-C00503
Figure USRE038132-20030603-C00504
Figure USRE038132-20030603-C00505
159-60
177
Figure USRE038132-20030603-C00506
Figure USRE038132-20030603-C00507
Figure USRE038132-20030603-C00508
 77-9
178
Figure USRE038132-20030603-C00509
Figure USRE038132-20030603-C00510
Figure USRE038132-20030603-C00511
 77-9
179
Figure USRE038132-20030603-C00512
Figure USRE038132-20030603-C00513
Figure USRE038132-20030603-C00514
oil
180
Figure USRE038132-20030603-C00515
Figure USRE038132-20030603-C00516
Figure USRE038132-20030603-C00517
120-1
Method J is exemplified by the synthesis of the compound of Example 148 (see Table 10), which was carried out is as follows. The starting material (0.11 g, 0.40 mmol) was dissolved in THF (5.0 mL) and cooled in a dry-ice/acetone bath (approximately −78° C.). Lithium bis(trimethylsilyl) amide (LiHMDS, 405.0 μL, 0.40 mmol) was added dropwise. The resultant yellow solution was stirred in the cold bath for 15 minutes, at which point 2-fluorobenzyl bromide was added to it. The mixture was stirred at this temperature for an additional 30 minutes and then at 0° C. for 30 minutes. The reaction mixture was next poured into 1 N HCl (40 mL) and extracted into EtOAc (50 mL). The organic layer was washed with saturated aqueous NaCl (35 mL), dried (Na2SO4), filtered, and concentrated in vacuo to afford 0.16 g of crude product. This material was purified by flash chromatography over silica gel (1:3 EtOAc/Hexanes) to afford 0.87 g (57.0%) of the compound of example 148.
Method K. C-Alkylation of Hydantoins using Methyl Magnesium Carbonate
As reported by Finkbeiner (J. Org. Chem. 1965, 30, 3414), hydantoins can be C-alkylated with alkyl halides using magnesium methyl carbonate (MMC). A solution of MMC in an organic solvent (such as DMF) is saturated at about 80° C. with CO2 over a period of about 1 h. An appropriate hydantoin is then added and heated with the MMC for about 1 to 2 h, at which point an appropriate alkyl halide is added. The reaction mixture is then warmed to about 110° C. for between about 2 to 3 h, then cooled to about room temperature. The mixture is then poured into concentrated aqueous acid (such as HCl) over ice and cooled. The solid formed is collected by filtration and purified by silica gel chromatography and/or via recrystallization to afford the desired product.
The compound listed in the Table 11 was produced via this method.
TABLE 11
Example of Compound Synthesized by Method K.
MELTING POINT
EX. STRUCTURE STARTING MATERIAL (° C.)
181
Figure USRE038132-20030603-C00518
Figure USRE038132-20030603-C00519
213-4
Method K is exemplified by the synthesis of the compound of Example 181 (see Table 11), which was carried out as follows: A dry two-necked round bottom-flask was evacuated and charged with a CO2 atmosphere. Magnesium methyl carbonate in DMF (860 μL, 2.0 M) was added to the flask and the solution was heated to 80° C. CO2 was introduced from a dry-ice vessel via a cannula connected to the reaction vessel and was bubbled through the solution for 1.0 h at which point an argon line was attached and the cannula was removed. The starting material (0.21 g, 0.86 mmol) in DMF (4.0 mL) was added and the reaction mixture was heated at 80° C. for 1.5 h. A solution of 3-picolyl chloride (0.12 g, 0.94 mmol-HCl salt was first free-based with NaOH) in DMF (1.0 mL) was then added dropwise. The temperature of the oil bath was increased to 110° C. and the mixture was heated at this temperature for 4.0 h. Upon cooling to room temperature the mixture was poured into a mixture 5 mL concentrated HCl and 10 g ice, then stored in a refrigerator overnight. The solution was next neutralized to pH 7-8 with 6 N NaOH and the resulting solid collected by suction filtration and washed with ice-water. Drying of the compound at 50° C. in vacuo afforded 0.20 g of crude product. This was purified by flash chromatography (5% MeOH/CH2Cl2) to afford 0.06 g of a material which was further purified by recrystallization with EtOH afforded 0.04 g (14.9%) of the compound of example 181. Method L. Synthesis of Compounds using Pd Catalyzed Cross Coupling
An appropriately substituted arylboronic acid or arylstanane is mixed with an aryl halide or aryl triflate and a catalytic amount of tetrakis(triphenylphosphine)palladium in an appropriate solvent system (such as benzene containing ethanol and aqueous Na2CO3, DMF, NMP or THF) under an inert atmosphere. Other components such as, for example LiCl and triethylamine, may be added as necessary. The mixture is heated at between about 50 and 150° C. for between about 2 and 48 h. The mixture is next cooled and diluted with an organic solvent (such as EtOAc). The organic phase was washed successively with water and saturated aqueous NaCl, dried (as with Na2SO4) and concentrated to give an impure mixture from which the desired material is isolated using silica gel chromatography.
The compounds listed in Table 12 were produced via this method.
TABLE 12
Example of Compound Synthesized by Method L
COUPLING MP
EX. STRUCTURE STARTING MATERIAL PARTNER (° C.)
182
Figure USRE038132-20030603-C00520
Figure USRE038132-20030603-C00521
Figure USRE038132-20030603-C00522
57-8 
183
Figure USRE038132-20030603-C00523
Figure USRE038132-20030603-C00524
Figure USRE038132-20030603-C00525
150-1 
184
Figure USRE038132-20030603-C00526
Figure USRE038132-20030603-C00527
Figure USRE038132-20030603-C00528
82-4 
185
Figure USRE038132-20030603-C00529
Figure USRE038132-20030603-C00530
Figure USRE038132-20030603-C00531
53-4 
186
Figure USRE038132-20030603-C00532
Figure USRE038132-20030603-C00533
Figure USRE038132-20030603-C00534
136-8 
187
Figure USRE038132-20030603-C00535
Figure USRE038132-20030603-C00536
Figure USRE038132-20030603-C00537
89-90
188
Figure USRE038132-20030603-C00538
Figure USRE038132-20030603-C00539
Figure USRE038132-20030603-C00540
56-57
189
Figure USRE038132-20030603-C00541
Figure USRE038132-20030603-C00542
Figure USRE038132-20030603-C00543
69-70
190
Figure USRE038132-20030603-C00544
Figure USRE038132-20030603-C00545
Figure USRE038132-20030603-C00546
77-9 
191
Figure USRE038132-20030603-C00547
Figure USRE038132-20030603-C00548
Figure USRE038132-20030603-C00549
76-8 
192
Figure USRE038132-20030603-C00550
Figure USRE038132-20030603-C00551
Figure USRE038132-20030603-C00552
201-2 
193
Figure USRE038132-20030603-C00553
Figure USRE038132-20030603-C00554
Figure USRE038132-20030603-C00555
70-3 
194
Figure USRE038132-20030603-C00556
Figure USRE038132-20030603-C00557
Figure USRE038132-20030603-C00558
77-80
195
Figure USRE038132-20030603-C00559
Figure USRE038132-20030603-C00560
Figure USRE038132-20030603-C00561
74-7 
196
Figure USRE038132-20030603-C00562
Figure USRE038132-20030603-C00563
Figure USRE038132-20030603-C00564
73-5 
197
Figure USRE038132-20030603-C00565
Figure USRE038132-20030603-C00566
Figure USRE038132-20030603-C00567
148-9 
198
Figure USRE038132-20030603-C00568
Figure USRE038132-20030603-C00569
Figure USRE038132-20030603-C00570
65-8 
199
Figure USRE038132-20030603-C00571
Figure USRE038132-20030603-C00572
Figure USRE038132-20030603-C00573
143-4 
200
Figure USRE038132-20030603-C00574
Figure USRE038132-20030603-C00575
Figure USRE038132-20030603-C00576
69-70
201
Figure USRE038132-20030603-C00577
Figure USRE038132-20030603-C00578
Figure USRE038132-20030603-C00579
97-99
202
Figure USRE038132-20030603-C00580
Figure USRE038132-20030603-C00581
Figure USRE038132-20030603-C00582
165-6 
203
Figure USRE038132-20030603-C00583
Figure USRE038132-20030603-C00584
Figure USRE038132-20030603-C00585
119-20 
204
Figure USRE038132-20030603-C00586
Figure USRE038132-20030603-C00587
Figure USRE038132-20030603-C00588
199-201
205
Figure USRE038132-20030603-C00589
Figure USRE038132-20030603-C00590
Figure USRE038132-20030603-C00591
168-170
206
Figure USRE038132-20030603-C00592
Figure USRE038132-20030603-C00593
Figure USRE038132-20030603-C00594
69-70
207
Figure USRE038132-20030603-C00595
Figure USRE038132-20030603-C00596
Figure USRE038132-20030603-C00597
70-2 
208
Figure USRE038132-20030603-C00598
Figure USRE038132-20030603-C00599
Figure USRE038132-20030603-C00600
163-4 
209
Figure USRE038132-20030603-C00601
Figure USRE038132-20030603-C00602
Figure USRE038132-20030603-C00603
110-111
210
Figure USRE038132-20030603-C00604
Figure USRE038132-20030603-C00605
Figure USRE038132-20030603-C00606
75-76
211
Figure USRE038132-20030603-C00607
Figure USRE038132-20030603-C00608
Figure USRE038132-20030603-C00609
80-1 
212
Figure USRE038132-20030603-C00610
Figure USRE038132-20030603-C00611
Figure USRE038132-20030603-C00612
66-68
213
Figure USRE038132-20030603-C00613
Figure USRE038132-20030603-C00614
Figure USRE038132-20030603-C00615
180-3 
214
Figure USRE038132-20030603-C00616
Figure USRE038132-20030603-C00617
Figure USRE038132-20030603-C00618
71-3 
Method L s exemplified by the synthesis of the compound of Example 182 (see Table 12) which was carried out as follows: In the manner taught by Miyaura, M; Yanagi, T; Suzuki, A. Synth. Commun. 1981, 11, 513, the starting material (0.24 g, 0.54 mmol) was mixed with phenylboric acid (0.73 g, 0.60 mmol), tetrakis-(triphenylphosphine) palladium(0)(0.31 g, 0.03 mmol), sodium carbonate (0.19 g, 1.79 mmol), benzene (3.0 mL), water (1.0 mL), and ethanol (1.0 mL) and stirred under reflux for 12 h. The reaction mixture was then poured into EtOAc (70 mL) and washed successively with water and saturated aqueous NaCl. The organic layer was dried (Na2SO4), filtered and concentrated in vacuo to afford 0.25 g of crude product. This material was purified by two successive silica gel chromatography columns (1:3 then 1:1 EtOAc/Hexanes) to produce 0.11 g (48%) of the compound of example 183.
Method M. Synthesis of Compounds using Carbonylative Pd Catalyzed Cross Coupling
An appropriately substituted arylboronic acid or arylstanane is mixed with an aryl halide or aryl triflate and a catalytic amount of tetrakis (triphenylphosphine) palladium in an appropriate solvent system (such as benzene containing ethanol and aqueous Na2CO3, DMF, NMP or THF) under an atmosphere of carbon monoxide. Other components such as, for example LiCl and triethylamine, may be added as necessary. The mixture is heated at between about 50 and 150° C. for between about 2 and 48 h. The mixture is next cooled and diluted with an organic solvent (such as EtOAc). The organic phase was washed successively with water and saturated aqueous NaCl, dried (as with Na2SO4) and concentrated to give an impure mixture from which the desired material is isolated using silica gel chromatography.
The compounds listed in Table 13 were produced via this method.
TABLE 13
Example of Compounds Synthesized by Method K
COUPLING MP
EX. STRUCTURE STARTING MATERIAL PARTNER (° C.)
215
Figure USRE038132-20030603-C00619
Figure USRE038132-20030603-C00620
Figure USRE038132-20030603-C00621
60-2
216
Figure USRE038132-20030603-C00622
Figure USRE038132-20030603-C00623
Figure USRE038132-20030603-C00624
99-100
Method M is exemplified by the synthesis of the compound of Example 215 (see Table 13) which was carried out as follows: the starting material (0.23 g, 0.53 mmol) was mixed with phenyltributylstannane (0.86 mL, 2.64 mmol), bistriphenylphosphine-palladium (II) chloride (0.037 g, 0.05 mmol), DMF (10.0 mL), and LiCl (5.1 mg, 1.6 mmol), the reaction mixture was then purged with argon, charged with CO and stirred at 115° C. for 12 h. The reaction mixture was then poured into 1 M tetrabutylammonium fluoride (10 mL), then diluted with 95 mL of EtOAc and washed successively with water and saturated aqueous NaCl. The organic layer was dried (Na2SO4), filtered and concentrated in vacuo to afford 1.2 g of crude product. This material was purified by silica gel chromatography (1:3 EtOAc/Hexanes) to produce 0.14 g (48%) of the compound of example 215.
Method N. Electrophilic Aromatic Substitution
Compounds containing aromatic rings can be modified by numerous reagents via electrophilic aromatic substitution. These include techniques for acylation, nitration, sulfonation and halogenation of these rings.
The compound listed in Table 14 was produced via this method.
TABLE 14
Example of Compound Synthesized by Method N
MELTING POINT
EXAMPLE STRUCTURE STARTING MATERIAL (° C.)
217
Figure USRE038132-20030603-C00625
Figure USRE038132-20030603-C00626
160-1
The compound of Example 217 is one such case and was prepared by the following procedure. A solution of the starting material (0.4 g, 1.1 mmol) in 1 mL of HOAc and 0.1 mL of H2S4 was treated with NaIO3 (0.05 g, 0.2 mmol) and I2 (0.06 g, 0.5 mmol). The mixture was then heated to 70° C. for 19 h after which point it was cooled to ambient temperature, extracted several times into EtOAc. The EtOAc was concentrated and the product was isolated after purification by silica gel chromatography. Yield: 33 mg (30%)
Method O. Deprotection of Compounds Protected with Acid-abile Groups
Compounds having acid-labile protecting groups may be deprotected by treatment under acidic conditions, in a known per se manner. Generally this involves treating the substrate with TFA, cation exchange resin (H+), HCL or HBr in AcOH with or without heating. The compound thus formed is collected by filtration or extraction and purified, as by silica gel chromatography or recrystallization, to afford the desired product.
Compounds listed in Table 15 were produced via this general method.
TABLE 15
Examples of Compounds Synthesized by Method O
MELT-
EX- ING
AM- POINT
PLE STRUCTURE STARTING MATERIAL (° C.)
218
Figure USRE038132-20030603-C00627
Figure USRE038132-20030603-C00628
195-6 
219
Figure USRE038132-20030603-C00629
Figure USRE038132-20030603-C00630
139-40
220
Figure USRE038132-20030603-C00631
Figure USRE038132-20030603-C00632
oil
221
Figure USRE038132-20030603-C00633
Figure USRE038132-20030603-C00634
oil
222
Figure USRE038132-20030603-C00635
Figure USRE038132-20030603-C00636
oil
223
Figure USRE038132-20030603-C00637
Figure USRE038132-20030603-C00638
84-5 
224
Figure USRE038132-20030603-C00639
Figure USRE038132-20030603-C00640
101-3 
225
Figure USRE038132-20030603-C00641
Figure USRE038132-20030603-C00642
76-7 
226
Figure USRE038132-20030603-C00643
Figure USRE038132-20030603-C00644
75-7 
227
Figure USRE038132-20030603-C00645
Figure USRE038132-20030603-C00646
75-7 
228
Figure USRE038132-20030603-C00647
Figure USRE038132-20030603-C00648
148-9 
229
Figure USRE038132-20030603-C00649
Figure USRE038132-20030603-C00650
174-5 
230
Figure USRE038132-20030603-C00651
Figure USRE038132-20030603-C00652
207-8 
231
Figure USRE038132-20030603-C00653
Figure USRE038132-20030603-C00654
88-9 
232
Figure USRE038132-20030603-C00655
Figure USRE038132-20030603-C00656
196-7 
233
Figure USRE038132-20030603-C00657
Figure USRE038132-20030603-C00658
209-10
234
Figure USRE038132-20030603-C00659
Figure USRE038132-20030603-C00660
133-4 
235
Figure USRE038132-20030603-C00661
Figure USRE038132-20030603-C00662
168-70
236
Figure USRE038132-20030603-C00663
Figure USRE038132-20030603-C00664
oil
237
Figure USRE038132-20030603-C00665
Figure USRE038132-20030603-C00666
not deter- mined
238
Figure USRE038132-20030603-C00667
Figure USRE038132-20030603-C00668
50-4 
Method O is exemplified by the synthesis of the compound of Example 219, which was carried out as follows: A stirred solution of the starting material (0.10 g, 0.19 mmol) in 10 mL of methylene chloride was chilled to 0° C. before adding 2.0 mL of trifluoroacetic acid. Stirring at 0° C. was continued for 20 min and then the solution was allowed to warm slowly to ambient temperature. The solution was stirred for an additional 6 hat which point it was concentrated yielding an off-white solid which was further dried under vacuum for 16 h. The crude solid was next triturated with 10 mL of boiling hexanes and the mixture was allowed to cool to ambient temperature. The resulting white precipitate was collected via filtration, washed with 5 mL of hexanes and dried under high vacuum for 4 h to afford 0.06 g (68% yield) of the compound from example 219.
Method P. Saponification of Esters to Acids with Hydroxide
Certain compounds having carboxylic esters may be converted to carboxylic acids by treatment with saponifying reagents, in a known per se manner. Generally this involves treating the substrate with NaOH, KOH or LiOH in a solvent such as H2O sometimes containing a solubilizing agent such as THF. Purification generally involves extracting the unreacted starting material with an organic solvent such as EtOAc or CH2Cl2, acidification of the aqueous layer and purification of the acid by filtration or extraction into an organic solvent such as EtOAc or CH2Cl2. Further purification can be performed using recrystallization, silica gel chromatography or reverse-phase HPLC, to afford the desired product.
The compound listed in Table 16 was produced via this general method.
TABLE 16
Examples of Compounds Synthesized by Method P.
MELTING POINT
EXAMPLE STRUCTURE STARTING MATERIAL (° C.)
239
Figure USRE038132-20030603-C00669
Figure USRE038132-20030603-C00670
138-40
Example 239 (table 16) was prepared by dissolving the starting material (0.38 g, 0.65 mmol) in 4 mL of H2O and 8 mL of MeOH containing LiOH (0.08 g, 1.95 mmol) and heating the mixture at 60° C. for 2.5 h. The MeOH was removed by concentration and the aqueous residue treated with 1 N HCl. The product was extracted into EtOAc from which it crystallized upon cooling. Yield 262 mg (72%).
Method Q. Cleavage of Pthalimide Protecting Group
Primary amines can be protected as their pthalimide derivatives. These derivatives are rapidly synthesized via method U using the potassium salt of pthalimide as the nucleophile. The amine can be liberated from the pthalimide protecting group using nucleophilic reagents such as hydrazine or methyl amine in a solvent such as EtOH. Purification generally involves acidification of the aqueous layer and extracting the unreacted starting material with an organic solvent such as EtOAc or CH2Cl2 Basification of the aqueous layer produces the free base of the amine which is purified by filtration or extraction into an organic solvent such as EtOAc or CH2Cl2. Further purification can be performed using recrystallization, silica gel chromatography or reverse-phase HPLC, to afford the desired product.
Compounds listed in Table 17 were produced via this general method.
TABLE 17
Examples of Compounds Synthesized by Method Q.
MELTING
EX- POINT
AMPLE STRUCTURE STARTING MATERIAL (° C.)
240
Figure USRE038132-20030603-C00671
Figure USRE038132-20030603-C00672
184-5
241
Figure USRE038132-20030603-C00673
Figure USRE038132-20030603-C00674
208-9
The compound of Example 240 was prepared by dissolving the starting material (prepare via method U, 0.72 g, 1.2 mmol) in 73 mL of EtOH and treating it with 19.5 mL of a 33% solution of MeNH2 in EtOH. The mixture was heated under reflux for 2.5 h and then cooled to ambient temperature. The reaction mixture was concentrated and the residue was dissolved in CH2Cl2 which was further washed with H2O and saturated aqueous NaCl. The organic layer was dried (Na2SO4) and concentrated. The yellow oil was dissolved in EtOH and treated with HCl gas. The amine hydrochloride of the compound of Example 240 was obtained in 69% yield (0.49 g).
Method R. Conversion of Nitriles into Amidines
Aromatic nitrites can be converted into amidine groups by several methods. Generally this conversion requires a two step process wherein the first step involves treatment with acid (such as, for example HCl) and an alcohol (such as, for example MeOH or EtOH) to generate an intermediate imino ether. This derivative is then converted to the amidine via treatment with an amine. Purification is usually be way of recrystallization of a derivative salt of the amidine. Further purification can be performed using recrystallization, silica gel chromatography or reverse-phase HPLC, to afford the desired product.
Compounds listed in Table 18 were produced via this general method.
TABLE 18
Examples of Compounds Synthesized by Method R.
MELTING POINT
EXAMPLE STRUCTURE STARTING MATERIAL (° C.)
242
Figure USRE038132-20030603-C00675
Figure USRE038132-20030603-C00676
210-2
243
Figure USRE038132-20030603-C00677
Figure USRE038132-20030603-C00678
180-1
The compound of example 243 was prepared by dissolving the starting material (0.2 g, 0.4 mmol) in 7 mL of EtOH, cooling the mixture in an ice bath, and treating the mixture with dry HCl gas for 15 min. The mixture was stirred at room temperature for 1 h and concentrated to yield the crude imino ether hydrochloride. The intermediate was dissolved s in EtOH (10 mL), cooled in an ice bath and treated with anhydrous NH3 gas for 20 min. After 5 h, the reaction mixture was concentrated to provide the crude amidine hydrochloride. This material was purified via silica gel chromatography (1:9 MeOH:CH2Cl2) to yield 0.08 g (38%) of the product.
Method S. Reduction of Carboxylic Acids to Alcohols
Certain compounds having carboxylic acids may be converted to alcohols by treatment with reducing reagents, in a known per se manner. Generally this involves treating the substrate with LiAlH4 or a BH3-based reagent in a solvent such as THF or ether. After careful quenching with an aqueous system, purification generally involves extracting the product into organic solvent such as EtOAc or CH2Cl2 and purification using recrystallization, silica gel chromatography or reverse-phase HPLC, to afford the desired product.
The compounds listed in Table 19 was produced via this general method.
TABLE 19
Examples of Compound Synthesized by Method S.
MELTING POINT
EXAMPLE STRUCTURE STARTING MATERIAL (° C.)
244
Figure USRE038132-20030603-C00679
Figure USRE038132-20030603-C00680
172-3
The compound of example 244 was prepared by dissolving the starting material (prepare via methods J and O, 0.16 g, 0.32 mmol) in 1 mL of THF, cooling the mixture in an ice bath and treating it with 0.65 mL of a 1 M BH3-THF solution (0.65 mmol). The mixture was allowed to warm to ambient temperature and stir for 15 h. The reaction mixture was quenched by the slow and careful addition of water and the organic components were extracted into EtOAc. The EtOAc layer was washed with water, saturated aqueous NaCl and dried over Na2SO4. concentration and silica gel chromatography (1:1 EtOAc:Hexanes) produced the desired compound (0.06 g, 42%).
Method T. Deprotection of Compounds with Nucleophilic Reagents
Certain compounds having methoxy protecting groups may be deprotected to the hydroxy derivative by treatment with certain nucleophilic reagents, in a known per se manner. Generally this involves treating the substrate with BBr3 or TMSI in a solvent such as CH2Cl2, generally cooled in an ice bath and followed with or without heating. After between about 10 min and 8 h the reaction is quenched with a weak base such as aqueous NaHCO3 and the organic component extracted into a solvent such as EtOAc and purified after concentration, as by silica gel chromatography or recrystallization, to afford the desired product.
Compounds listed in Table 20 were produced via this general method.
TABLE 20
Examples of Compounds Synthesized by Method T.
MELTING POINT
EXAMPLE STRUCTURE STARTING MATERIAL (° C.)
245
Figure USRE038132-20030603-C00681
Figure USRE038132-20030603-C00682
94-7
246
Figure USRE038132-20030603-C00683
Figure USRE038132-20030603-C00684
118-120
247
Figure USRE038132-20030603-C00685
Figure USRE038132-20030603-C00686
94-96
248
Figure USRE038132-20030603-C00687
Figure USRE038132-20030603-C00688
not determined
Method T is exemplified by the synthesis of the compound of Example 246, which was carried out as follows: A stirred solution of the starting material (0.35 g, 0.64 mmol) in 9 mL of CH2Cl2 was chilled to 0° C. before adding 1.0 mL of BBr3 (1.07 mmol, 1 M CH2Cl2). Stirring was continued and then the solution was allowed to warm slowly to ambient temperature. The solution was stirred for an additional 4 h at which point it saturated aqueous NaHCO3 was added, the organic layer was removed and concentrated yielding the crude product. Purification was performed by silica gel chromatography (1:4 EtOAc:Hexanes) yielding 0.16 g (48% yield) of the desired compound.
Method U. Nucleophilic Displacement
An appropriate electrophilic agent is dissolved in an aprotic solvent (such as DMF, THF or DMSO) and treated with one to three equivalents of a nucleophile (such as Me3N, Na salt of imidazole, Na2SO3, NaCN, P(OEt)3, or the K salt of Pthalimide at between about room temperature and 100° C. The mixture stirred at between about 0 and 100° C. for up to about 24 h. (Progress of the reaction can be monitored using TLC). The solution is then cooled and diluted with an organic solvent (such as, for example, EtOAc). The organic phase is washed sequentially with dilute aqueous acid (such as 1 N HCl), and with water, dried (for example, over MgSO4) and concentrated. The desired compound is purified, as by silica gel chromatography, reverse-phase HPLC or by recrystallization.
Compounds listed in Table 21 were produced via this general method.
TABLE 21
Examples of Compounds Synthesized by Method U.
EX. STRUCTURE STARTING ELECTROPHILE STARTING NUCLEOPHILE M. P. (° C.)
249
Figure USRE038132-20030603-C00689
Figure USRE038132-20030603-C00690
Na2SO3 200-2 
250
Figure USRE038132-20030603-C00691
Figure USRE038132-20030603-C00692
Na2SO3 193-4 
251
Figure USRE038132-20030603-C00693
Figure USRE038132-20030603-C00694
Na2SO3 >260
252
Figure USRE038132-20030603-C00695
Figure USRE038132-20030603-C00696
Figure USRE038132-20030603-C00697
195-6 
253
Figure USRE038132-20030603-C00698
Figure USRE038132-20030603-C00699
Me3N 55-6 
254
Figure USRE038132-20030603-C00700
Figure USRE038132-20030603-C00701
Me3N 54(d)
255
Figure USRE038132-20030603-C00702
Figure USRE038132-20030603-C00703
Me3N 42-3 
256
Figure USRE038132-20030603-C00704
Figure USRE038132-20030603-C00705
NaCN 152-3 
257
Figure USRE038132-20030603-C00706
Figure USRE038132-20030603-C00707
NaCN 149-50 
258
Figure USRE038132-20030603-C00708
Figure USRE038132-20030603-C00709
(CH3)2NH 68-70
259
Figure USRE038132-20030603-C00710
Figure USRE038132-20030603-C00711
Figure USRE038132-20030603-C00712
65-6 
260
Figure USRE038132-20030603-C00713
CH3COCl
Figure USRE038132-20030603-C00714
oil
261
Figure USRE038132-20030603-C00715
Figure USRE038132-20030603-C00716
Figure USRE038132-20030603-C00717
not determined
262
Figure USRE038132-20030603-C00718
Figure USRE038132-20030603-C00719
Figure USRE038132-20030603-C00720
188-90 
263
Figure USRE038132-20030603-C00721
Figure USRE038132-20030603-C00722
Figure USRE038132-20030603-C00723
118-21 
264
Figure USRE038132-20030603-C00724
Figure USRE038132-20030603-C00725
Figure USRE038132-20030603-C00726
113-5 
Method U is exemplified by the synthesis of the compound of Example 262, which was carried out as follows: To the starting material (0.28 g, 0.54 mmol) was added 1-H-pyrazolecarboxamidine (0.08 g, 0.54 mmol) followed by 7 mL of DMF and 0.2 mL of N,N-diisopropyl-ethylamine. The resulting mixture was then stirred 15 h at room temperature. Next was added ether which caused the mixture to become turbid. As no crystals were forming, MeOH was added to re-dissolve the reaction components and the product was precipitated as its HCl salt by the addition of 1 N HCl. The solid was collected and washed with ether to yield 0.11 g (20%) of the guanidine hydrochloride.
Method V. Resolution of a Mixture of Enantiomers
There are several ways to resolve the compounds of the invention into their enantiomerically pure forms. One such method is chiral HPLC. An exemplary column packing is Chiracel-OD (Diacel Chemistry Industries). An exemplary solvent system is 9:1 hexanes:iso-propyl alcohol. In general, the R-enantiomer is eluted first, but this should not be used as the sole criterion for the assignment of stereochemistry.
The compounds listed in Table 22 were resolved via this method.
TABLE 22
Examples of Compounds Obtained by Method V.
EXAMPLE STRUCTURE STARTING RACEMATE MELTING POINT (° C.)
265
Figure USRE038132-20030603-C00727
Figure USRE038132-20030603-C00728
not determined
266
Figure USRE038132-20030603-C00729
Figure USRE038132-20030603-C00730
not determined
267
Figure USRE038132-20030603-C00731
Figure USRE038132-20030603-C00732
oil
268
Figure USRE038132-20030603-C00733
Figure USRE038132-20030603-C00734
oil
269
Figure USRE038132-20030603-C00735
Figure USRE038132-20030603-C00736
oil
270
Figure USRE038132-20030603-C00737
Figure USRE038132-20030603-C00738
oil
271
Figure USRE038132-20030603-C00739
Figure USRE038132-20030603-C00740
52-54
272
Figure USRE038132-20030603-C00741
Figure USRE038132-20030603-C00742
136-7 
273
Figure USRE038132-20030603-C00743
Figure USRE038132-20030603-C00744
not determined
274
Figure USRE038132-20030603-C00745
Figure USRE038132-20030603-C00746
not determined
275
Figure USRE038132-20030603-C00747
Figure USRE038132-20030603-C00748
oil
276
Figure USRE038132-20030603-C00749
Figure USRE038132-20030603-C00750
oil
277
Figure USRE038132-20030603-C00751
Figure USRE038132-20030603-C00752
not determined
278
Figure USRE038132-20030603-C00753
Figure USRE038132-20030603-C00754
not determined
Description of Biological Properties
The biological properties of representative compounds of the formula I were investigated by way of the experimental protocols described below. The results of such testing are reported in Table 23, which appears below.
Assay to Determine Inhibition of LFA-1 Binding to ICAM-1
Purpose of Assay:
This assay protocol is designed to study the direct antagonism, by a test compound, of the interaction of the CAM, ICAM-1 with the Leukointegrin CD18/CD11a (LFA-1).
Description of Assay Protocol: LFA-1 is immunopurified using the TS2/4 antibody from a 20 g pellet of human JY or SKW3 cells, utilizing a protocol previously described (Dustin, M. J.; et al., J. Immunol. 1992, 148, 2654-2660). The LFA-1 is purified from SKW3 lysates by immunoaffinity chromatography on TS2/4 LFA-1 mAb Sepharose and eluted at pH 11.5 in the presence of 2 mM MgCl2 and 1% octylglucoside. After collection and neutralization of fractions from the TS2/4 column, samples are pooled and precleared with Protein G agarose.
A soluble form of ICAM-1 is constructed, expressed, purified and characterized as previously described (Marlin, S.; et al., Nature, 1990, 344, 70-72 and see Arruda, A.; et al., Antimicrob. Agents Chemother. 1992, 36, 1186-1192). Briefly, isoleucine 454 which is located at the putative boundary between domain 5 of the ectodomain and the transmembrane domain, is changed to a stop codon using standard oligonucleotide-directed mutagenesis. This construction yields a molecule identical with the first 453 amino acids of membrane bound ICAM-1. An expression vector is created with a hamster dihydrofolate reductase gene, a neomycin-resistance marker, and the coding region of the sICAM-1 construct described above, along with the promotor, splice signals, and polyadenylation signal of the SV40 early region. The recombinant plasmid is transfected into CHO DUX cells using standard calcium phosphate methods. Cells are passaged in selective media (G418) and colonies secreting sICAM-1 are amplified using methotrexate. sICAM-1 is purified from serum-free media using traditional non-affinity chromatographic techniques, including ion exchange and size exclusion chromatography.
LFA-1 binding to ICAM-1 is monitored by first incubating sICAM-1 at 40 μg/mL in Dulbecco's phosphate buffered saline with calcium and magnesium, additional 2 mM MgCl2 and 0.1 mM PMSF (Diluting Buffer) in a 96-well plate for 30 amin at room temperature. Plates are then blocked by the addition of 2% (w/v) bovine serum albumin in Diluting Buffer for 37° C. for 1 h. Blocking solution is removed from wells, and test compounds are diluted and then added followed by the addition of approximately 25 ng of immunoaffinity purified LFA-1. The LFA-1 is incubated in the presence of test compound and ICAM-1 at 37° C. for 1 h. Wells are washed 3 times with Diluting Buffer. The bound LFA-1 is detected by the addition of a polyclonal antibody directed against a peptide corresponding to the CD18 cytoplasmic tail in a 1:100 dilution with Diluting Buffer and 1% BSA and allowed to incubate for 45 min at 37° C. Wells are washed 3 times with Diluting Buffer and the bound polyclonal antibody is detected by the addition of a 1:4000 dilution of horse radish peroxidase conjugated to goat immunoglobulin directed against rabbit immunoglobulin. This reagent is allowed to incubate for 20 min at 37° C., wells are washed as above and the substrate for the horse radish peroxidase is added to each well to develop a quantitative colorimetric signal proportional to the amount of LFA-1 bound to sICAM-1. Soluble ICAM-1 (60 μg/mL) is used as a positive control for inhibition of the LFA-1/ICAM-1 interaction. The lack of the addition of LFA-1 to the binding assay is used as a background control for all samples. A dose-response curve is obtained for all test compounds.
Results of these tests are reported as Kd's in μM.
MMT Assay to Determine Cytotoxicity
Purpose of Assay:
In order to obtain meaningful data from cellular assays, compounds must first be first tested in an assay to measure cellular toxicity. The MTT assay can be used for this purpose.
Description of Assay Protocol:
MTT, (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide), is a yellow substrate that is cleaved by cells with active mitochondria to yield a dark blue/purple formazan product. This precipitate can be solublized and the amount of material quantitatified via spectrophotometric procedures (Gerlier, D.; Thomasset, N. J. Immunol. Methods, 1986, 94, 57-63). The amount of color is proportional to the number of viable cells. This assay system is used to assess the effect of test compounds on cell viability in vitro.
SKW3 cells, which express LFA-1, are used. Cells used in each assay were adjusted to 1.25·106 cells/mL and 100 μL of this stock is dispersed into each well of a 96 well, flat bottom microtiter plate. For each condition in a particular experiment, triplicate wells are set up. Serial dilutions of each test compound or vehicle alone are added to each well. Cells are incubated with compound for 4-24 h at 37° C. before cell viability is assessed. Next, 10 μL of filter sterilized MTT is added to each well. The MTT stock is made in phosphate buffered saline at a concentration of 5 mg/mL. Plates are then incubated for 1 h at 37° C., 5% CO2 atmosphere. Periodically, the plates are examined for formazan crystal development.
At the end of the incubation period, formazan crystals are dissolved by the addition of 100 μL of 0.04 N HCl in iso-propyl alcohol to each well. Each well is thoroughly mixed by repeated pipetting with a multichannel pipetter. The plates are allowed to sit at room temperature for 15-20 minutes and are then read with a spectrophotometer. Absorbance is measured at the test wavelength of 570 nm. Data are reported as the concentration range (in μM) wherein 50% of the cells are no longer viable.
Assay to Determine Inhibition of SKW3 cell binding to ICAM-1 vs. Binding to Fibronectin
Purpose of Assay:
This assay is used to test the selective ability of a test compound to antagonize the interaction of a cell bound form of LFA-1 with ICAM-1. The assay uses a human T cell line, SKW3 cells which express CD18, CD11a and other integrins not related to CD18, CD11a and which can be “activated” by phorbol esters. Phorbol esters enhance the affinity of CD18, CD11a for ICAM-1.
This same lymphocyte line, SKW3, also adheres to fibronectin in the presence of phorbol esters. This adhesion is mediated by membrane proteins independent of the LFA-1/ICAM-1 interaction. The SKW3 cells express another integrin, VLA4, which is the receptor for fibronectin. Therefore, as a preliminary indication of the selectivity of a test compounds to interfere with Leukointegrin/CAM interactions but not other integrin-ligand binding events, a compound can be tested for its ability to antagonize cell bound fibronectin receptor in its interaction with purified fibronectin. Compounds that inhibit this fibronectin adhesion are not specific antagonists of the CD18, CD11a/ICAM-1 binding.
Description of Assay Protocol:
Ninety-six well plates are coated with either sICAM-1(40 μg/mL) or fibronectin (100 μg/mL) in Diluting buffer for 1 h at room temperature. Added to the wells are 100 μL of the appropriately diluted test compound or 100 μL of RPMI with 15% fetal bovine serum as a control. SKW3 cells, which express CD18, CD11a and VLA4 (Dustin, M.; et al., J. Exp. Med. 1987, 165, 672-692) are washed and suspended to a concentration of 106 cells/mL in RPMI with 15% fetal bovine serum. Immediately before adding the cells to the wells, cells are stimulated with the phorbol ester 12-myristate 13-acetate (PMA) for a final concentration of 100 μg/mL. 100 μL of cells are then added to the wells resulting in a final concentration of 50 μg/mL PMA and 2 cells/well. The plates are incubated for 2 h at 37° C. Unbound or loosely bound cells are gently washed away with RPMI. Cells remaining and hence bound to ICAM-1 or to fibronectin are quantitated by the same reagent used above for the MTT experiment.
Data are reported as the concentration or concentration range (in μM) at which 50% of binding is inhibited.
Compounds Inhibit JY Cell Aggregation
Purpose of Assay:
This is an in vitro cell to cell adhesion assay which can be used to test the ability of a test compound to directly inhibit LFA-1 dependent aggregation at the cellular level.
Many Epstein-Barr virus-transformed cells exhibit aggregation. This aggregation can be enhanced by the addition of phorbol esters. Such homotypic aggregation (i.e., aggregation involving only one cell type) was found to be blocked by anti-LFA-1 antibodies (Rothlein, R. R.; et al., J. Exp. Med. 1986, 163, 1132-1149). Thus, the extent of LFA-1-dependent binding may be determined by assessing the extent of spontaneous or phorbol ester-dependent aggregate formation.
An agent which interferes with LFA-1 -dependent aggregation can be identified through the use of an assay capable of determining whether the agent interferes with either spontaneous, or phorbol ester-dependent aggregation of Epstein-Barr virus transformed cells. It is preferable to employ cells of the JY cell line (Terhost, L; et aL, Proc. Natl. Acad. Sci. USA, 1976, 73, 910) for the homotypic aggregation assay. This assay, capable of measuring LFA-1 dependent aggregation, may be employed to identify agents which act as antagonists to the LFA-1 dependent aggregation. Such agents may act by impairing the ability of either LFA-1 or ICAM-1 to mediate aggregation. Thus, agents may be examined to directly determine if they are antagonists of LFA-1 aggregation.
Description of Assay Protocol:
JY cells are cultured in RPMI 1640 culture medium supplemented with 10% fetal calf serum and 50 μg/mL gentamycin. The cells are cultured at 37° C. in an atmosphere of 5% CO2 at a relative humidity of 95%. JY cells used in this assay are washed two times with RPMI 1640 medium containing 5 mM HEPES buffer and resuspended to a concentration of 2·106 cells/mL. Added to flat-bottomed, 96-well microtiter plates are 50 μL of test compound diluted in complete medium, 50 μL of complete medium with or without purified monoclonal antibodies (negative and positive controls for inhibition, respectively), 50 μL of complete medium containing 200 ng/mL of the phorbol ester phorbol myristate acetate (PMA) and 100 μL of cells at a concentration of 2·106 cells/mL in complete medium. This yields a final concentration of 50 ng/nL PMA and 2·105 cells/well. Cells are allowed to settle spontaneously, and the degree of aggregation is scored at various time points. Scores range from 0 to 4 where 0 indicates that essentially no cells are in clusters; 1 indicates that <25% of the cells are in clusters; 2 indicates that <50% of the cells are in clusters; 3 indicates that <75% of the cells are in clusters and 4 indicates that 100% of the cells are aggregated. This procedure has been described by Rothlein, R. R.; et al., J. Exp. Med. 1986, 163, 1132-1149. This paper also reported that antibody to LFA-1 is capable of inhibiting the formation of aggregates. Whereas 100% of the cells form aggregates in the absence of LFA-1 antibody, less than 20% of the cells were found to be in aggregates when anti-LFA-1 antibody was added in the same paper.
Data are reported as the concentration or concentration range (in μM) at which 50% of binding is inhibited.
Assay to Determine Inhibition the Mixed Lymphocyte Reaction
Purpose of Assay:
As discussed above, ICAM-1 is necessary for effective cellular interactions during an immune response mediated through LFA-1-dependent cell adhesion. When lymphocytes from two unrelated individuals are cultured together, blast transformation and cell proliferation of the lymphocytes are observed. This response is known as a mixed lymphocyte reaction (MLR) and is analogous to the response of lymphocytes to the 10 addition of antigens or mitogens (Immunology: The Science of Self-Nonself Discrimination; Klein, J., Ed.; John Wiley & Sons: NY, 1982, pp 453-458). Monoclonal antibodies directed against ICAM-1 and LFA-1 were used as controls to demonstrate inhibition of cell adhesion-dependent lymphocyte stimulation and proliferation.
This assay protocol is used to to determine the effect of a test compound on the human MLR. The ability of a test compound to inhibit the MLR and antigen-specific mononuclear cell responses shows that it has therapeutic utility in acute graft rejection., as well as in related immune mediated disorders dependent on CD18, CD11a/ICAM interactions.
Description of Assay Protocol:
Peripheral blood is obtained from normal, healthy donors by venipuncture. The blood is collected in heparinized tubes and diluted 1:1 at room temperature with Puck's G (GIBCO) balanced salt solution (BSS). The blood mixture (20 mL) is layered over 15 mL of a Ficoll/Hypaque density gradient (Pharmacia, density=1.078, room temperature) and centrifuged at 1000·g for 20 minutes. The interface is then collected and washed 3 times in Puck's G. The cells are counted on a hemocytometer and resuspended in RPMI 1640 culture medium (GIBCO) containing 0.5% of gentamicin, 1 mM L-glutamine (GICO) and 5% heat inactivated (56° C., 30 min) human AB sera (Flow Laboratories) (hereafter referred to as RPMI-culture medium).
Peripheral blood mononuclear cells (PBMC) are cultured in medium at 6.25·105 cells/mL in a Linbro round-bottomed microtiter plate. Stimulator cells (cells that have been treated with irradiation so that they are unable to proliferate) from a separate donor are cultured with the responder cells at the same concentration. Test compound is added to wells at various concentrations. The total volume per culture is 0.2 mL. Controls include compound vehicle alone (DMSO), responder cells alone, and stimulator cells alone. The culture plates are incubated at 37° C. in a 5% CO2-humidified air atmosphere for 5 days.
The wells are pulsed with 0.5 μCi of tritiated thymidine (3HT) (New England Nuclear) for the last 18 h of culture. In some cases a two-way MLR can be performed. The protocol is the same except that the second donor's cells are not inactivated by irradiation.
The cells are harvested onto glass fiber filters using an automated multiple sample harvester (Skatron, Norway), and rinsed with water and methanol. The filters are oven dried and counted in Aquasol in a Beckman (LS-3801) liquid scintillation counter.
Data are reported as “+” or “−” at a given concentration (in μM).
In vivo: Allogeneic Cell Transplant Model
Purpose of Assay:
The ability of cells to recognize other cells from self or from another genetically different individual (non-self) is an important property in maintaining the integrity of tissue and organ structure. The allogeneic cell transplant response is an important model for studies of transplant rejection and immunocompetence. This T-cell-mediated immune response can be induced in adult mice by the injection of lymphocytes from a histoincompatible mouse strain into the footpad . This response is characterized by T-cell proliferation which is limited to the popliteal lymph node that receives drainage from the injected footpad area. No in vitro system can completely duplicate this in vivo response. Thus, this animal model can be used to assess the ability of a test compound to suppress transplant rejection.
Description of Assay Protocol:
Experiments are conducted using male or female mice (20-26 grams). Any histoincompatible mouse strains suffice for donor and recipient populations. Typically DBA mice are used as donors and C57 1/6 mice are used as recipients. A minimum of 1 week stabilization and conditioning period is required before use during which time the animals are maintained in accordance with the Animal Resource Center S.O.P. Each study utilizes 36 recipient mice divided into groups of 6. The tests last approximately four days. Donor mice are sacrificed by CO2 asphyxiation and spleens are removed and made into a cell suspension. The cells (1.0·107/metatarsal in 0.05 mL) are injected intra dermal (according to standard protocol) into the dorsal metatarsal skin of recipient mice. Four days later, the animals are sacrificed by CO2 asphyxiation and the popliteal nodes are removed and weighed. Groups of mice receiving putative immunosuppressive agents are dosed subcutaneously, intraperitoneally or orally one hour prior to cell injection and daily thereafter according to standard protocol. Student's T-test was used to determine significant differences between popliteal lymph nodes of groups of untreated mice and those mice treated with putative immunosuppressive agents (see: Kroczek, R. A.; Black, C. D. V.; Barbet, J.; Shevach, E. M., J. Immunology, 1987, 139, 3597).
Data are reported as the dose at which 50% inhibition is observed and the manner in which the compound was administered. In Table 23, the following legends are applicable: and=not determined. bpercent inhibition at 160 μg/mL. cno inhibition observed up to highest dose. Exact quantification not always possible due to intrinsic toxicity of compound (see MTT result). dpercent inhibition (concentration in μM ). eapproximated from incomplete dose-response curve. fnot determined; compound is a synthesis intermediate.
TABLE 23
Results of Biological Testing.a
Cellular JY Cell Mixed Allogeneic Cell
LFA-1/ICAM Toxicity Assay to Detect SKW3 Cells Aggregation Lymphocyte Transplata-
Cmpd. Binding Assay Binding to: Assay Reaction tion Assay
Of Assay MTT ICAM FIBRONECTIN JY Cell MLR ACT
Ex. Kd (μM) LD50 (μM) IC50 (μM) IC50 (μM) IC50 (μM) +/− (μM) ED50 (mg/kg)
1 1.64 nd nd nd nd nd nd
2 14.4 125-250 >100c >100c nd nd nd
3 3.00 125-250 nd nd nd nd nd
4 9.32 250-500 nd nd nd nd nd
5 15.4 250-500 nd nd nd nd nd
6 12.8 250-500 nd nd nd nd nd
7 1.33 250-500  50-100 No inhc nd nd nd
8 2.86 125-500 nd nd nd nd nd
9 7.87 250-500 nd nd nd nd nd
10 0.19 >500 50 No inhc 6.3 nd nd
11 6.30 nd nd nd nd nd nd
12 1.75 125-250 nd nd nd nd nd
13 1.96 nd nd nd nd nd nd
14 12.7 125-250  63-125 >125c nd nd nd
15 2.3 125-250 31-63 No Inhc 25-50 nd
16 0.85  63-125 19-38  >75c nd nd nd
17 3.01 125 >63c No inhc nd nd nd
18 0.76 125-250 32-63 >125c 6 nd
19 0.76 nd nd nd nd nd nd
20 1.76 125-250 16  63-125 nd nd nd
21 0.69 125-250 <16 125-250 nd nd
22 0.40 125-250 <16 No inhc nd nd
23 0.77 50-100 25-50 >100c 13 nd nd
24 0.85 >400 >60 No inhc nd nd nd
25 3.08 nd nd nd nd nd nd
26 2.75 nd nd nd nd nd nd
27 52e nd nd nd nd nd nd
28 2.59 100-200 100-200c No inhc nd nd nd
29 0.06  63-125 8 No inhc 0.1 +(50) ˜60 (per os)
30 1.48 nd nd nd nd nd nd
31 1.36 nd nd nd nd nd nd
32 12.1 nd nd nd nd nd nd
33 0.94 nd nd nd nd nd nd
34 0.13 200-400 6 No inhc 1.8 nd nd
35 0.10 38-46  5-10 No inhc 0.8 nd nd
36 4.05 nd nd nd nd nd nd
37 0.16  <13  5-10  >19 1.6 nd nd
38 1.15 nd nd nd nd nd nd
39 0.18 nd nd nd nd nd nd
40 0.48  50-100  50-100  50-100 25 nd nd
41 0.16 nd nd nd nd nd nd
42 0.60 nd nd nd nd nd nd
43 0.17 25-50 3-6 No inhc 1.8 nd nd
44 0.42  63-125  6-12 No inhc 0.3 nd nd
45 ndf nd nd nd nd nd nd
46 2.26 nd nd nd nd nd nd
47 4.31 nd nd nd nd nd nd
48 1.75 nd nd nd nd nd nd
49 0.26 nd nd nd nd nd nd
50 17e nd nd nd nd nd nd
51 0.43 nd nd nd nd nd nd
52 1.59 nd nd nd nd nd nd
53 1.47 nd nd nd nd nd nd
54 0.42 nd nd nd nd nd nd
55 0.20 nd nd nd nd nd nd
56 0.48 nd nd nd nd nd nd
57 0.36 nd nd nd nd nd nd
58 2.85 nd nd nd nd nd nd
59 0.33 nd nd nd nd nd nd
60 0.23 nd nd nd nd nd nd
61 2.35 nd nd nd nd nd nd
62 0.16 nd nd nd nd nd nd
63 3e nd nd nd nd nd nd
64 1.45 nd nd nd nd nd nd
65 1.32 nd nd nd nd nd nd
66 2.85 nd nd nd nd nd nd
67 3.54 nd nd nd nd nd nd
68 2.58 nd nd nd nd nd nd
69 70e nd nd nd nd nd nd
70 1.89 nd nd nd nd nd nd
71 4.38 nd nd nd nd nd nd
72 0.06 >363 20-40 No inhc 1.4 +(25) nd
73 0.34 nd nd nd nd nd nd
74 0.67 nd nd nd nd nd nd
75 0.29 nd nd nd nd nd nd
76 2.34 >500 150-300 >300c nd nd nd
77 ndf nd nd nd nd nd nd
78 0.34 nd nd nd nd nd nd
79 0.43 nd nd nd nd nd nd
80 0.19 >400 5 No inhc 3.2 nd nd
81 0.08 25-50 1-3 No inhc 6.3 nd nd
82 2.1 250-500  63-125 >250c nd nd nd
83 0.30 250-500 13-25 >200c 6 +(50) nd
84 0.42 nd nd nd nd nd nd
85 0.72 >500 19-38  >75c nd nd nd
86 0.53 >500 19-38  >75c nd nd nd
87 2.00 >500  50-100 >100c 50 nd nd
88 0.41 250-500 19-38  >75c nd nd nd
89 0.29  500 50 No inhc nd nd nd
90 0.73 >500  50-100 No inhc nd nd nd
91 0.43 nd nd nd nd nd nd
92 0.12  63-125 13-25 No inhc 3.1 +(25) nd
93 1.27 100-200 >100c No inhc 13 nd nd
94 0.74  200  50-100 No inhc 25 nd nd
95 0.26 nd nd nd nd nd nd
96 0.46 100-200 50 No inhc nd nd nd
97 0.023 >350  5-10 No inhc 2 +(25-50) nd
98 0.055 >340  5-10 No inhc 3 +(50) nd
99 0.52 nd nd nd nd nd nd
100 0.19 >400  8-15  >60 0.7 nd nd
101 0.68 nd nd nd nd nd nd
102 0.06 >400 25-50 >200 1.3 +(25) ˜25 per os
103 0.10 25-50 25  >25 0.13 nd nd
104 0.27 nd nd nd nd nd nd
105 0.12 nd nd nd nd nd nd
106 0.14 nd nd nd nd nd nd
107 0.49 nd nd nd nd nd nd
108 0.41 nd nd nd nd nd nd
109 0.03 100-200 3-6 No inhc 0.4 nd nd
110 0.15 200-400 25 No inhc 9 nd nd
111 0.19  36 3 No inhc 75 nd nd
112 0.39 nd nd nd nd nd nd
113 1.91 nd nd nd nd nd nd
114 0.26 nd nd nd nd nd nd
115 0.24 nd nd nd nd nd nd
116 0.19 nd nd nd nd nd nd
117 1.09  12 12  12 2.2 nd nd
118 0.46 200-400 >100 >100 3.4 nd nd
119 0.02 >400 <3 No inhc 0.05 nd nd
120 0.48 nd nd nd nd nd nd
121 0.21  <11 0.8-2   No inhc 25 nd nd
122 0.13 200-400 12-25 No inhc 1.8 nd nd
123 0.10  50-100 12-25  >25 0.6 +(50) nd
124 0.03 100-200 1.5 No inhc 0.1 nd nd
125 0.11 >400 1-3 No inhc 0.4 nd ˜90 per os
126 0.81 nd nd nd nd nd nd
127 0.05 125-250 0.8-1.6 No inhc 0.1 nd nd
128 0.16 125-250 1.5 No inhc 0.1 nd nd
129 0.10 nd nd nd nd nd nd
130 0.33 nd nd nd nd nd nd
131 0.20 >400 3-5 No inhc 1.1 nd nd
132 0.22 200-400 5 No inhc 2.4 nd nd
133 0.15 >400  5-10 No inhc 0.4 nd nd
134 0.05 >250 1.5 No inhc 0.1 nd nd
135 0.20 >400 25 No inhc 1.7 nd nd
136 0.19 150-300 0.6-1   No inhc nd nd nd
137 0.04 200-400 1-3 No inhc 0.4 nd nd
138 0.04 250-500 6 No inhc nd +(25-50) nd
139 0.79 nd nd nd nd nd nd
140 0.11 180-360 1.2  >10 0.2 nd nd
141 0.22  300 15  >15 0.1 nd
142 0.43 nd nd nd nd nd nd
143 1.20 nd nd nd nd nd nd
144 0.35 nd nd nd nd nd nd
145 1.05 nd nd nd nd nd nd
146 0.13  100 13-25 No inhc 0.4 +(50) nd
147 0.34 nd nd nd nd nd nd
148 0.50 nd nd nd nd nd nd
149 0.20 170-340 10  >10 2.9 nd nd
150 0.51 nd nd nd nd nd nd
151 0.046 >410  5-10 No inhc 0.2 +(25) nd
152 2.66 nd nd nd nd nd nd
153 2.19 nd nd nd nd nd nd
154 0.094  200  5-10 No inhc 0.4 nd nd
155 0.51 nd nd nd nd nd nd
156 0.69 nd nd nd nd nd nd
157 0.037 >390  5-10 No inhc 0.4 +(25) nd
158 0.69 nd nd nd nd nd nd
159 0.11  50-100 10-20 No inhc 2 nd nd
160 0.64 nd nd nd nd nd nd
161 4.56 nd nd nd nd nd nd
162 1.05 nd nd nd nd nd nd
163 0.16 200-400  9-19  >75c 2.4 nd nd
164 0.28 100-200 >100c No inhc 25 nd nd
165 1.07 nd nd nd nd nd nd
166 0.28 nd nd nd nd nd nd
167 0.60 nd nd nd nd nd nd
168 0.41 nd nd nd nd nd nd
169 0.05 160-230 12-25 No inhc 1.5 +(25) nd
170 0.02 nd nd nd nd nd nd
171 0.18 100-200 102 No inhc 0.2 nd nd
172 0.17  165 1.8 No inhc 0.7 nd nd
173 0.17 200-400 4.8 No inhc 2.2 nd nd
174 0.71 nd nd nd nd nd nd
175 0.23 nd nd nd nd nd nd
176 3.96 nd nd nd nd nd nd
177 0.25 155-310  7-14 No inhc 3.7 nd nd
178 1.17 nd nd nd nd nd nd
179 3.84 nd nd nd nd nd nd
180 0.19 200-400 1.5-3   No inhc 1.3 nd nd
181 47%b nd nd nd nd nd nd
182 0.05 200-400 3-6 No inhc 0.02 +(25) nd
183 0.94 nd nd nd nd nd nd
184 0.37  50-100 0.8-1.5 No inhc 0.3 nd nd
185 0.25 nd nd nd nd nd nd
186 1.11 nd nd nd nd nd nd
187 0.29  171 0.6 No inhc 0.6 nd nd
188 0.04  56-112 0.3-1   No inhc 0.03 nd nd
189 0.19  63 0.6-1.2 No inhc nd nd nd
190 0.43  50-100  6-12 No inhc 1.4 nd nd
191 0.14 nd nd nd nd nd nd
192 1e nd nd nd nd nd nd
193 0.29 nd nd nd nd nd nd
194 0.27 nd nd nd nd nd nd
195 0.30 nd nd nd nd nd nd
196 0.09 nd nd nd nd nd nd
197 0.19 nd nd nd nd nd nd
198 0.14 nd nd nd nd nd nd
199 0.27 nd nd nd nd nd nd
200 0.09 nd nd nd nd nd nd
201 0.68 nd nd nd nd nd nd
202 0.23 nd nd nd nd nd nd
203 0.34 nd nd nd nd nd nd
204 0.50 nd nd nd nd nd nd
205 0.56 nd nd nd nd nd nd
206 0.73 nd nd nd nd nd nd
207 1.28 nd nd nd nd nd nd
208 0.65 nd nd nd nd nd nd
209 0.94 nd nd nd nd nd nd
210 0.54 nd nd nd nd nd nd
211 2e nd nd nd nd nd nd
212 0.07 nd nd nd nd nd nd
213 2e nd nd nd nd nd nd
214 0.09 nd nd nd nd nd nd
215 0.20 100-200  50-100 >100 3.1 +(50) nd
216 0.50 nd nd nd nd nd nd
217 1.78 nd nd nd nd nd nd
218 0.80 nd nd nd nd nd nd
219 1.03 nd nd nd nd nd nd
220 0.27 nd nd nd nd nd nd
221 0.14 nd nd nd nd nd nd
222 0.06 nd nd nd nd nd nd
223 0.15 200-400 2-4 No inhc 0.2 nd nd
224 0.28  <25  5-10 No inhc 3.1 nd nd
225 0.63  50-100 3-4 No inhc 0.2 +(25) nd
226 0.07  100 3-6 No inhc 1.6 +(25) nd
227 0.18 200-400 25 No inhc 3.1 +(50) nd
228 0.41  50-100 12-25 No inhc 1.6 nd nd
229 0.30  50-100  6-13 No inhc 3.1 +(50) nd
230 2.20 nd nd nd nd nd nd
231 0.27 40-80 2-5 No inhc nd nd nd
232 1.24 nd nd nd nd nd nd
233 1.79 nd nd nd nd nd nd
234 0.41 100-200  5-10 No inhc 1.6 nd nd
235 0.33 100-200 9 No inhc 1.6 nd nd
236 0.33 nd nd nd nd nd nd
237 0.13  50-100 1-2 No inhc 0.1 nd nd
238 0.08 nd nd nd nd nd nd
239 0.28 nd nd nd nd nd nd
240 0.11 11-23 1-3    13.4 6.3 nd nd
241 0.10  9-18 0.8-1.6  13 nd nd nd
242 0.40 16-32 10-25 No inhc 2.8 nd nd
243 0.11 10-20 5-6 No inhc 3.1 nd nd
244 3.98 200-400 4-8 No inhc 2.2 nd nd
245 0.21 25-50 1-3 No inhc 0.7 nd nd
246 0.25 nd nd nd nd nd nd
247 0.23 40-80 1.5-3   No inhc 2.3 nd nd
248 0.37  50-100  9-18 No inhc 0.7 nd nd
249 0.16 200-400 <1 No inhc 0.4 nd nd
250 0.04 100-200 <1 No inhc 0.1 nd nd
251 0.06 100-200 <0.5 No inhc nd nd nd
252 0.11 17-20 0.4-0.8 No inhc 0.3 nd nd
253 0.05 100-200 1-2 No inhc 0.2 nd nd
254 0.08 100-200 1-2 No inhc 0.4 nd nd
255 0.14 100-200 1 No inhc 0.2 nd nd
256 0.12 150-300 0.8-2   No inhc 0.4 nd nd
257 0.08  300 1.6 No inhc 0.6 nd nd
258 1.66 nd nd nd nd nd nd
259 3.54 nd nd nd nd nd nd
260 0.07 200-400 19-38 No inhc 1.1 nd nd
261 ndf nd nd nd nd nd nd
262 0.08 25-75 1-2 No inhc 0.4 nd nd
263 0.09 20-40 0.8-1.6 No inhc 0.8 nd nd
264 0.12 12-24 2-5 No inhc 0.4 nd nd
265 0.28 130-250 19-38  >75c 3 nd nd
266 0.14 >500 25-50  >50c 3 +(25-50) nd
267 0.40  250 nd nd nd nd nd
268 1.59 250-500 nd nd nd nd nd
269 0.29 250-500 25 No inhc nd nd nd
270 0.65 250-500 nd nd nd nd nd
271 0.02 250-500 3 No inhc 0.1 +(13-25) ˜20 (per os)
272 0.56 250-500 50-100 No inhc nd nd nd
273 0.19 150-300 38  >75c 2 nd
274 0.14 200-300 25-50 >100c 3 nd
275 2.00 nd nd nd nd nd nd
276 1.11  375 10 No inhc 0.5 +(25) nd
277 0.07 nd nd nd nd nd nd
278 1.09 nd nd nd nd nd nd
Description of Therapeutic Use
The novel and known small molecules utilized in the method according to the invention inhibit the ICAM-1/LFA-1 dependent homotypic aggregation of human lymphocytes, human lymphocyte adherence to ICAM-1 and human lymphocyte responses to antigens. These compounds have therapeutic utility in the modulation of immune cell activation/proliferation, e.g., as competitive inhibitors of intercellular ligand/receptor binding reactions involving CAMs and Leukointegrins. Inflammatory conditions which may be treated with the compounds comprehended by the invention include conditions resulting from a response of the non-specific immune system in a mammal (e.g., adult respiratory distress syndrome, shock, oxygen toxicity, multiple organ injury syndrome secondary to septicemia, multiple organ injury syndrome secondary to trauma, reperfusion injury of tissue due to cardiopulmonary bypass, myocardial infarction or use with thrombolysis agents, acute glomerulonephritis, vasculitis, reactive arthritis, dermatosis with acute inflammatory components, stroke, thermal injury, hemodialysis, leukapheresis, ulcerative colitis, necrotizing enterocolitis and granulocyte transfusion associated syndrome) and conditions resulting from a response of the specific immune system in a mammal (e.g., psoriasis, organ/tissue transplant rejection, graft vs. host reactions and autoimmune diseases including Raynaud's syndrome, autoimmune thyroiditis, dermatitis, multiple sclerosis, rheumatoid arthritis, insulin-dependent diabetes mellitus, uveitis, inflammatory bowel disease including Crohn's disease and ulcerative colitis, and systemic lupus erythematosus). In accordance with the invention, these novel and known compounds may also be used in treating asthma or as an adjunct to minimize toxicity with cytokine therapy in the treatment of cancers. In general these compounds may be employed in the treatment of those diseases currently treatable through steroid therapy.
In accordance with the method provided by the invention, these novel and known compounds may be administered for either a “prophylactic” or “therapeutic” purpose either alone or with other immunosuppressive or antiinflammatory agents. When provided prophylactically, the immunosuppressive compound(s) are provided in advance of any inflammatory response or symptom (for example, prior to, at, or shortly after the time of an organ or tissue transplant but in advance of any symptoms of organ rejection). The prophylactic administration of a compound of the formula I serves to prevent or attenuate any subsequent inflammatory response (such as, for example, rejection of a transplanted organ or tissue, etc.). The therapeutic administration of a compound of the formula I serves to attenuate any actual inflammation (such as, for example, the rejection of a transplanted organ or tissue). Thus, in accordance with the invention, a compound of the formula I can be administered either prior to the onset of inflammation (so as to suppress an anticipated inflammation) or after the initiation of inflammation.
The novel and known compounds of the formula I may, in accordance with the invention, be administered in single or divided doses by the oral, parenteral or topical routes. A suitable oral dosage for a compound of formula I would be in the range of about 0.5 mg to 1 g per day. In parenteral formulations, a suitable dosage unit may contain from 0.1 to 250 mg of said compounds, whereas for topical administration, formulations containing 0.01 to 1% active ingredient are preferred. It should be understood, however, that the dosage administration from patient to patient will vary and the dosage for any particular patient will depend upon the clinician's judgement, who will use as criteria for fixing a proper dosage the size and condition of the patient as well as the patient's response to the drug.
When the compounds of the present invention are to be administered by the oral route, they may be administered as medicaments in the form of pharmaceutical preparations which contain them in association with a compatible pharmaceutical carrier material. Such carrier material can be an inert organic or inorganic carrier material suitable for oral administration. Examples of such carrier materials are water, gelatin, talc, starch, magnesium stearate, gum arabic, vegetable oils, polyalkylene-glycols, petroleum jelly and the like.
The pharmaceutical preparations can be prepared in a conventional manner and finished dosage forms can be solid dosage forms, for example, tablets, dragees, capsules, and the like, or liquid dosage forms, for example solutions, suspensions, emulsions and the like. The pharmaceutical preparations may be subjected to conventional pharmaceutical operations such as sterilization. Further, the pharmaceutical preparations may contain conventional adjuvants such as preservatives, stabilizers, emulsifiers, flavor-improvers, wetting agents, buffers, salts for varying the osmotic pressure and the like. Solid carrier material which can be used include, for example, starch, lactose, mannitol, methyl cellulose, microcrystalline cellulose, talc, silica, dibasic calcium phosphate, and high molecular weight polymers (such as polyethylene glycol).
For parenteral use, a compound of formula I can be administered in an aqueous or non-aqueous solution, suspension or emulsion in a pharmaceutically acceptable oil or a mixture of liquids, which may contain bacteriostatic agents, antioxidants, preservatives, buffers or other solutes to render the solution isotonic with the blood, thickening agents, suspending agents or other pharmaceutically acceptable additives. Additives of this type include, for example, tartrate, citrate and acetate buffers, ethanol, propylene glycol, polyethylene glycol, complex formers (such as EDTA), antioxidants (such as sodium bisulfite, sodium metabisulfite, and ascorbic acid), high molecular weight polymers (such as liquid polyethylene oxides) for viscosity regulation and polyethylene derivatives of sorbitol anhydrides. Preservatives may also be added if necessary, such as benzoic acid, methyl or propyl paraben, benzalkonium chloride and other quaternary ammonium compounds.
The compounds of this invention may also be administered as solutions for nasal application and may contain in addition to the compounds of this invention suitable buffers, tonicity adjusters, microbial preservatives, antioxidants and viscosity-increasing agents in an aqueous vehicle. Examples of agents used to increase viscosity are polyvinyl alcohol, cellulose derivatives, polyvinylpyrrolidone, polysorbates or glycerin. Microbial preservatives added may include benzalkonium chloride, thimerosal, chloro-butanol or phenylethyl alcohol.
Additionally, the compounds provided by the invention can be administered by suppository.
Formulations
Compounds of the formula I can be formulated for therapeutic administration in a number of ways. Descriptions of several exemplary formulations are given below.
Example A
Capsules or Tablets
Ingredients Quantity
Example A-1
Compound of 250 mg
formula I
Starch 160 mg
Microcrys. Cellulose 90 mg
Sodium Starch 10 mg
Glycolate
Magnesium Stearate 2 mg
Fumed colloidal silica 1 mg
Example A-2
Compound of 50 mg
formula I
Dicalcium Phosphate 160 mg
Microcrys. Cellulose 90 mg
Stearic acid 5 mg
Sodium Starch 10 mg
Glycolate
Fumed colloidal silica 1 mg
The compound of formula I is blended into a powder mixture with the premixed excipient materials as identified above with the exception of the lubricant. The lubricant is then blended in and the resulting blend compressed into tablets or filled into hard gelatin capsules.
Example B
Parenteral Solutions
Ingredients Quantity
Compound of formula I 500 mg
PEG 400 40% by volume
Ethyl Alcohol 5% by volume
Saline 55% by volume
The excipient materials are mixed and then added to one of the compounds of formula I in such volume as is necessary for dissolution. Mixing is continued until the solution is clear. The solution then filtered into the appropriate vials or ampoules and sterilized by autoclaving.
Example C
Suspension
Ingredients Quantity
Compound of formula I 100 mg
Citric acid 1.92 g
Benzalkonium chloride 0.025% by weight
EDTA 0.1% by weight
Polyvinylalcohol 10% by weight
Water q.s. to 100 mL
The excipient materials are mixed with the water and thereafter one of the compounds of formula I is added and mixing is continued until the suspension is homogeneous. The suspension is then transferred into the appropriate vials or ampoules.

Claims (14)

What is claimed is:
1. A compound of the formula I
Figure USRE038132-20030603-C00755
wherein:
Y is an oxygen atom;
Z is an oxygen atom;
X is a divalent group of the formula >CHR1 or >NR1, wherein R1 is:
(A) a hydrogen atom,
(B) branched or unbranched alkyl of 1 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, which alkyl or cycloakyl group is unsubstituted or monosubstituted with:
(i) oxo,
(ii) aryl selected from the class consisting of phenyl, thiophenyl, pyridyl, pyrimidinyl, furyl, pyrrolyl, oxazolyl, thiazolyl, pyrazolyl, isoxazolyl, imidazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl, pyridazinyl, pyrazinyl and triazinyl, wherein one or more hydrogen atoms of said aryl group are or are not independently replaced with:
(a) alkyl of 1 to 3 carbon atoms,
(b) —COOH,
(c) —SO2OH,
(d) —PO(OH)2,
(e) a group of the formula —COOR7, wherein R7 is straight or branched alkyl of 1 to 5 carbon atoms or cycloalkyl of 3 to 5 carbon atoms,
(f) a group of the formula —NH2,
(g) a group of the formula —CONH2,
(h) a group of the formula —OR12a, wherein R12a is a hydrogen atom or a methyl,
(i) an amidino group of the formula
Figure USRE038132-20030603-C00756
wherein R13, R14 and R15 are each hydrogen atoms,
(iii) a group of the formula —COOR16, wherein R16 is straight or branched alkyl of 1 to 7 carbon atoms or cycloalkyl of 3 to 6 carbon atoms,
(iv) a group of the formula OR19, wherein R19 is a hydrogen atom, or an alkyl or acyl group of 1 to 7 carbon atoms, or
(v) a quaternary group of the formula
Figure USRE038132-20030603-C00757
wherein R24, R25 and R26 are each methyl and Q is a chlorine, bromine or iodine counterion,
(C) a group of the formula —COOH connected via a branched or unbranched alkyl group of 2 to 5 carbon atoms,
(D) a group of the formula —PO(OH)2 connected via a branched or unbranched alkyl group of 2 to 6 carbon atoms,
(E) a group of the formula —SO2OH connected via a branched or unbranched alkyl group of 2 to 6 carbon atoms,
(F) an amidino group of the formula
Figure USRE038132-20030603-C00758
wherein r is 2, 3, 4, 5 or 6, and
R27, R28 and R29 are each hydrogen atoms,
(G) an guanidino group of the formula
Figure USRE038132-20030603-C00759
wherein s is 2, 3, 4, 5 or 6,
R30, R31, R32 and R33 are each hydrogen atoms, or
(H) piperidyl, wherein the nitrogen atom of said group is unsubtituted or substituted with:
(i) alkyl of 1 to 3 carbon atoms,
(ii) a group of the formula —COO(C1-6alkyl),
(iii) a group of the formula —COOH connected via an alkyl group of 1 to 4 carbon atoms,
(iv) a group of the formula —PO(OH)2 connected via an alkyl group of 1 to 6 carbon atoms, or
(v) a group of the formula —SO2OH connected via an alkyl group of 1 to 6 carbon atoms;
R2 is:
(A) a hydrogen atom, or
(B) methyl;
R3 is a group of the formula —CH2R41, wherein
R41 is
aryl selected from the class consisting of phenyl, thiophenyl, pyridyl, pyrimidinyl, furyl, oxazolyl, thiazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, thiadiazolyl, pyridazinyl, and pyrazinyl, wherein one or more of the hydrogen atoms of said aryl group are necessarily and independently replaced with:
(A) R62, which is aryl selected from the class consisting of phenyl, thiophenyl, pyridyl, pyrimidinyl, furyl, oxazolyl, thiazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, thiadiazolyl, pyridazinyl, and pyrazinyl, wherein one or more of the hydrogen atoms of said aryl group are or are not independently replaced with:
(i) methyl,
(ii) —COOH,
(iii) a group of the formula —COOR63, wherein R63 is methyl,
(iv) a group of the formula —OR68, wherein R68 is a hydrogen atom or methyl, or
(v) halogen,
(B) methyl, which is unsubstituted or mono- or polysubstituted with fluorine atoms or which is unsubstituted or monosubstituted with R62,
(C) branched or unbranched alkyl of 2 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, which alkyl or cycloakyl group is unsubstituted or mono- or polysubstituted with halogen or oxo,
(D) a group of the formula —COOR73, wherein R73 is methyl,
(E) a group of the formula —CONR76R77, wherein R76 and R77 are each methyl, and wherein one of R76 and R77 is methyl and the other is the group R62,
(F) a group of the formula —COR78, wherein R78 is a hydrogen atom, methyl or R62,
(G) a group of the formula —OR79, wherein R79 is a hydrogen atom, methyl or R62,
(H) cyano,
(I) nitro, or
(J) halogen;
R4 is Cl or trifluoromethyl;
R5 is a hydrogen atom; and,
R6 is Cl, or trifluoromethyl;
or a pharmaceutically acceptable salt thereof.
2. A compound of the formula I, in accordance with claim 1, wherein:
Y is an oxygen atom;
Z is an oxygen atom;
X is a divalent group of the formula >CHR1 or >NR1,
wherein R1 is:
(A) a hydrogen atom,
(B) branched or unbranched alkyl of 1 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, which alkyl or cycloakyl group is unsubstituted or monosubstituted with:
(i) oxo,
(ii) aryl selected from the class consisting of phenyl or pyridyl,
wherein one or more hydrogen atoms of said aryl group are or are not independently replaced with:
(a) alkyl of 1 to 3 carbon atoms,
(b) —COOH,
(c) —SO2OH,
(d) —PO(OH)2,
(e) a group of the formula —OR12a, wherein R12a is a hydrogen atom or a methyl,
(f) an amidino group of the formula
Figure USRE038132-20030603-C00760
wherein R13, R14 and R15 are each hydrogen atoms,
(iii) a group of the formula —OR19, wherein R19 is a hydrogen atom, or an alkyl or acyl group of 1 to 7 carbon atoms, or
(iv) a quaternary group of the formula
Figure USRE038132-20030603-C00761
wherein R24, R25 and R26 are each methyl and Q is a chlorine, bromine or iodine counterion,
(C) a group of the formula —COOH connected via a branched or unbranched alkyl group of 2 to 5 carbon atoms,
(D) a group of the formula —PO(OH)2 connected via a branched or unbranched alkyl group of 2 to 6 carbon atoms,
(E) a group of the formula —SO2 OH connected via a branched or unbranched alkyl group of 2 to 6 carbon atoms,
(F) an amidino group of the formula
Figure USRE038132-20030603-C00762
wherein r is 2, 3, 4, 5 or 6, and R27, R28 and R29 are each hydrogen atoms,
(G) an guanidino group of the formula
Figure USRE038132-20030603-C00763
wherein s is 2, 3, 4, 5 or 6, R30, R31, R32 and R33 are each hydrogen atoms, or
(H) piperidyl, wherein the nitrogen atom of said group is unsubstituted or substituted with:
(i) alkyl of 1 to 3 carbon atoms,
(ii) a group of the formula —COO(C1-6alkyl),
(iii) a group of the formula —COOH connected via an alkyl group of 1 to 4 carbon atoms,
(iv) a group of the formula —PO(OH)2 connected via an alkyl group of 1 to 6 carbon atoms, or
(v) a group of the formula —SO2 OH connected via an alkyl group of 1 to 6 carbon atoms;
R2 is:
(A) a hydrogen atom, or
(B) methyl;
R3 is a group of the formula —CH2 R41, wherein
R41 is
aryl selected from the class consisting of phenyl or pyridyl,
wherein one or more of the hydrogen atoms of said aryl group are necessarily and independently replaced with:
(A) R62, which is aryl selected from the class consisting of phenyl, or pyridyl,
wherein one or more of the hydrogen atoms of said aryl group are or are not independently replaced with:
(i) methyl,
(ii) —COOH
(iii) a group of the formula —COOR63, wherein R63 is methyl,
(iv) a group of the formula —OR68, wherein R68 is a hydrogen atom or methyl, or
(v) halogen,
(B) methyl, which is unsubstituted or mono- or polysubstituted with fluorine atoms or which is unsubstituted or monosubstituted with R62,
(C) branched or unbranched alkyl of 2 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, which alkyl or cycloakyl group is unsubstituted or mono- or polysubstituted with fluorine or oxo,
(D) a group of the formula —COOR73, wherein R73 is methyl,
(E) a group of the formula —CONR76R77, wherein R76 and R77 are each methyl, and wherein one of R76 and R77 is methyl and the other is the group R62,
(F) a group of the formula —COR78, wherein R78 is a hydrogen atom, methyl or R62,
(G) a group of the formula —OR79, wherein R79 is a hydrogen atom, methyl or R62,
(H) cyano,
(I) nitro, or
(J) halogen;
R4 is a chlorine atom or trifluoromethyl;
R5 is a hydrogen atom; and,
R6 is a chlorine atom, or trifluoromethyl;
or a pharmaceutically acceptable salt thereof.
3. A compound of the formula I, in accordance with claim 2, wherein:
Y is an oxygen atom;
Z is an oxygen atom;
X is a divalent group of the formula >CHR1 or >NR1, wherein R1 is:
(A) a hydrogen atom,
(B) alkyl of 1 to 2 carbon atoms which is unsubstituted or monosubstituted with:
(i) oxo,
(ii) aryl selected from the class consisting of phenyl or pyridyl, wherein one hydrogen atom of said aryl group is or is not replaced with:
(a) alkyl of 1 to 3 carbon atoms,
(b) —COOH,
(c) —SO2OH,
(d) —PO(OH)2,
(e) a group of the formula —OR12a, wherein R12a is a hydrogen atom or a methyl, or
(f) an amidino group of the formula
Figure USRE038132-20030603-C00764
wherein R13, R14 and R15 are each hydrogen atoms, or
(iii) a group of the formula —OR19, wherein R19 is a hydrogen atom or methyl,
a group of the formula —COOH connected via a branched or unbranched alkyl group of 2 to 5 carbon atoms,
(D) a group of the formula —PO(OH)2 connected via a branched or unbranched alkyl group of 2 to 6 carbon atoms,
(E) a group of the formula —SO2OH connected via a branched or unbranched alkyl group of 2 to 6 carbon atoms,
(F) an amidino group of the formula
Figure USRE038132-20030603-C00765
wherein r is 2, 3, 4, 5 or 6, and R27, R28 and R29 are each hydrogen atoms, or
(G) an guanidino group of the formula
Figure USRE038132-20030603-C00766
wherein s is 2, 3, 4, 5 or 6,
R30, R31, R32 and R33 are each hydrogen atoms,
R2 is:
(A) a hydrogen atom, or
(B) methyl;
R3 is a group of the formula —CH2 R41, wherein
R41 is
phenyl
wherein one or more of the hydrogen atoms of said phenyl group are necessarily and independently replaced with:
(A) R62, which is aryl selected from the class consisting of phenyl, or pyridyl,
wherein one or more of the hydrogen atoms of said aryl group are or are not independently replaced with:
(i) methyl,
(ii) a group of the formula —COOR63, wherein R63 is methyl,
(iii) a group of the formula —OR68, wherein R68 is a hydrogen atom or methyl, or
(iv) halogen,
(B) methyl, which is unsubstituted or mono- or polysubstituted with fluorine atoms or which is unsubstituted or monosubstituted with R62,
(C) a group of the formula —COOR73, wherein R73 is methyl,
(D) a group of the formula —COR78, wherein R78 is methyl or R62,
(E) a group of the formula —OR79, wherein R79 is a hydrogen atom, methyl or R62,
(F) cyano,
(G) nitro, or
(H) halogen;
R4 is a chlorine atom or trifluoromethyl;
R5 is a hydrogen atom; and,
R6 is a chlorine atom, or trifluoromethyl;
or a pharmaceutically acceptable salt thereof.
4. A compound of the formula I, in accordance with claims 3, wherein:
Y is an oxygen atom;
Z is an oxygen atom;
X is a divalent group of the formula >NR1,
wherein R1 is:
(A) a hydrogen atom,
(B) methyl or ethyl, or
(C) —COCH3
R2 is:
(A) a hydrogen atom, or
(B) methyl;
R3 is a group of the formula —CH2 R41, wherein
R41 is:
phenyl,
wherein one or more of the hydrogen atoms of said phenyl group are necessarily and independently replaced with:
(A) R62, which is aryl selected from the class consisting of phenyl, or pyridyl,
wherein one or more of the hydrogen atoms of said aryl group are or are not independently replaced with:
(i) methyl,
(ii) a group of the formula —COOR63, wherein R63 is methyl,
(iii) a group of the formula —OR68, wherein R68 is a hydrogen atom or methyl, or
(iv) halogen,
(B) methyl, which is unsubstituted or mono- or polysubstituted with fluorine atoms or which is unsubstituted or monosubstituted with R62,
(C) a group of the formula —COOR73, wherein R73 is methyl,
(D) a group of the formula —COR78, wherein R78 is methyl or R62,
(E) a group of the formula —OR79, wherein R79 is a hydrogen atom, methyl or R62,
(F) cyano,
(G) nitro, or
(H) halogen;
R4 is a chlorine atom or trifluoromethyl;
R5 is a hydrogen atom; and,
R6 is a chlorine atom, or trifluoromethyl;
or a pharmaceutically acceptable salt thereof.
5. A compound of the formula I, in accordance with claims 4, wherein:
Y is an oxygen atom;
Z is an oxygen atom;
X is a divalent group of the formula >NR1,
wherein R1 is:
(A) a hydrogen atom,
(B) methyl or ethyl, or
(C) —COCH3
R2 is:
(A) a hydrogen atom, or
(B) methyl;
R3 is a group of the formula —CH2 R41, wherein
R41 is
phenyl wherein one or more of the hydrogen atoms of said phenyl group are necessarily and independently replaced with:
(A) R62, which is aryl selected from the class consisting of phenyl, or pyridyl, wherein one or more of the hydrogen atoms of said aryl group are or are not independently replaced with:
(i) methyl, or
(ii) halogen,
(B) methyl, which is unsubstituted or mono- or polysubstituted with fluorine atoms,
(C) a group of the formula —COR78, wherein R78 is methyl or R62,
(D) halogen;
R4 is a chlorine atom;
R5 is a hydrogen atom; and,
R6 is a chlorine atom;
or a pharmaceutically acceptable salt thereof.
6. A compound selected from the group consisting of:
Figure USRE038132-20030603-C00767
Figure USRE038132-20030603-C00768
and pharmaceutically acceptable salts thereof.
7. A method for treating an inflammatory, immune cell-mediated disease or condition which comprises administering a prophylactic or therapeutic amount of a compound in accordance with claim 1, 2, 3, 4, 5 or 6.
8. The method of claim 7 wherein the disease or condition is selected from the group consisting of adult respiratory distress syndrome, shock, oxygen toxicity, multiple organ injury syndrome secondary to septicemia, multiple organ injury syndrome secondary to trauma, reperfusion injury of tissue due to cardiopulmonary bypass, myocardial infarction, acute glomerulonephritis, vasculitis, reactive arthritis, dermatosis with acute inflammatory components, stroke, thermal injury, hemodialysis, leukapheresis, ulcerative colitis, necrotizing enterocolitis and granulocyte transfusion associated syndrome.
9. The method of claim 1 or 7 wherein the disease or condition is selected from the group consisting of psoriasis, organ/tissue transplant rejection, graft vs. host reactions and autoimmune diseases.
10. The method of claim 7 wherein the disease or condition is asthma.
11. The method of claim 7 wherein the condition is toxicity associated with cytokine therapy.
12. A pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound in accordance with claim 1, 2, 3, 4, 5 or 6.
13. A method for treating an inflammatory or immune cell-mediated disease or condition, which method comprises administering a therapeutic amount of a compound of the formula I
Figure USRE038132-20030603-C00769
wherein:
Y is an oxygen or sulfur atom;
Z is an oxygen or sulfur atom;
X is a divalent group of the formula >CHR 1 , >NR 1 , >CHSO 2 R 1 , or >NSO 2 R 1 , or an oxygen or sulfur atom,
wherein R 1 is:
(A) a hydrogen atom,
(B) branched or unbranched alkyl of 1 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, which alkyl or cycloakyl group is unsubstituted or mono- or polysubstituted with:
(i) halogen,
(ii) oxo,
(iii) aryl which is selected from the class consisting of phenyl, naphthyl, indolyl, thiophenyl, pyridyl, pyrimidinyl, furyl, pyrrolyl, oxazolyl, thiazolyl, pyrazolyl, isoxazolyl, imidazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl, pyridazinyl, pyrazinyl, triazinyl, indolyzinyl, isoindolyl, benzo[b]furanyl, benzo[b]thiophenyl, indazolyl, benzthiazolyl, benzimidazolyl, quinolinyl, isoquinolinyl, purinyl, quinolizinyl, cinnolinyl, pthalaninyl, quinoxalinyl, napthyridinyl, pteridinyl and quinazolinyl,
wherein one or more hydrogen atoms of said aryl group are or are not independently replaced with:
(a) alkyl of 1 to 3 carbon atoms,
(b) —COOH,
(c) —SO 2 OH,
(d) —PO(OH)2,
(e) a group of the formula —COOR 7 , wherein R 7 is straight or branched alkyl of 1 to 5 carbon atoms or cycloalkyl of 3 to 5 carbon atoms,
(f) a group of the formula —NR 8 R 9 , wherein R 8 and R 9 are each independently a hydrogen atom, alkyl of 1 to 6 carbon atoms, cycloalkyl of 3 to 6 carbon atoms or acyl of 1 to 7 carbon atoms, or wherein R 8 and R 9 together form a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between R 8 and R 9 forms a heterocyclic ring,
(g) a group of the formula —CONR 10 R 11 , wherein R 10 and R 11 are each independently a hydrogen atom, alkyl of 1 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, or wherein R 10 and R 11 together form a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between R 10 and R 11 forms a heterocyclic ring,
(h) a group of the formula —OR 12a , wherein R 12a is a hydrogen atom, or an alkyl or acyl group of 1 to 7 carbon atoms,
(i) a group of the formula —SR 12b , wherein R 12b is a hydrogen atom, or an alkyl or acyl group of 1 to 7 carbon atoms,
(j) cyano, or
(k) an amidino group of the formula
Figure USRE038132-20030603-C00770
wherein R 13 , R 14 and R 15 are each, independently, a hydrogen atom or alkyl of 1 to 3 carbon atoms and wherein two of R 13 , R 14 and R 15 when additionally taken together form a saturated hydrocarbon bridge of 3 to 5 carbon atoms which bridge, together with the atom(s) between the two groups that form the bridge, forms a heterocyclic ring,
(iv) a group of the formula —COOR 16 , wherein R 16 is straight or branched alkyl of 1 to 7 carbon atoms or cycloalkyl of 3 to 6 carbon atoms,
(v) cyano,
(vi) a group of the formula —CONR 17 R 18 , wherein R 17 and R 18 are each, independently, a hydrogen atom, alkyl of 1 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, or wherein R 17 and R 18 together form a saturated hydrocarbon bridge of 3 to 5 carbon atoms which bridge, together with the nitrogen atom between R 17 and R 18 , forms a heterocyclic ring,
(vii) a group of the formula —OR 19 , wherein R 19 is a hydrogen atom, or an alkyl or acyl group of 1 to 7 carbon atoms,
(viii) a group of the formula —SR 20 , wherein R 20 is a hydrogen atom, or an alkyl or acyl group of 1 to 7 carbon atoms,
(ix) a group of the formula —NR 21 R 22 , wherein R 21 and R 22 are each, independently,
(a) a hydrogen atom,
(b) alkyl or acyl of 1 to 7 carbon atoms or cycloalkyl of 3 to 7 carbon atoms,
(c) a group of the formula —(CH 2)MCOOH, wherein m is 0, 1 or 2, or
(d) a group of the formula —(CH 2)nCOOR 23 , wherein n is 0, 1 or 2, wherein R 23 is straight or branched alkyl of 1 to 6 carbon atoms,
or wherein R 21 and R 22 together form a saturated hydrocarbon bridge of 3 to 5 carbon atoms which bridge, together with the nitrogen atom between R 21 and R 22 , forms a heterocyclic ring, or
(x) a quaternary group of the formula
Figure USRE038132-20030603-C00771
wherein R 24 , R 25 and R 26 are each, independently, a branched or unbranched alkyl group of 1 to 7 carbon atoms and Q is a chlorine, bromine or iodine counterion,
(C) a group of the formula —COOH connected via a branched or unbranched alkyl group of 2 to 5 carbon atoms,
(D) a group of the formula —PO(OH)2 connected via a branched or unbranched alkyl group of 2 to 6 carbon atoms,
(E) a group of the formula —SO 2 OH connected via a branched or unbranched alkyl group of 2 to 6 carbon atoms,
(F) an amidino group of the formula
Figure USRE038132-20030603-C00772
wherein r is 2, 3, 4, 5, or 6, and
R 27 , R 28 and R 29 are each, independently, a hydrogen atom or alkyl of 1 to 3 carbon atoms, and wherein two of R 27 , R 28 and R 29 when additionally taken together form a saturated hydrocarbon bridge of 3 to 5 carbon atoms which bridge, together with the [nitrogen] atom(s) between the two groups that form the bridge, forms a heterocyclic ring,
(G) an guanidino group of the formula
Figure USRE038132-20030603-C00773
wherein s is 2, 3, 4, 5 or 6, and
R 30 , R 31 , R 32 and R 33 are each, independently, a hydrogen atom or alkyl of 1 to 3 carbon atoms, and wherein two of R 30 , R 31 , R 32 and R 33 when additionally taken together form a saturated hydrocarbon bridge of 3 to 5 carbon atoms which bridge, together with the atom(s) between the two groups that form the bridge, forms a heterocyclic ring,
(H) piperidyl, wherein the nitrogen atom of said group is unsubstituted or substituted with:
(i) alkyl of 1 to 3 carbon atoms,
(ii) a group of the formula —COO(C 1-6 alkyl),
(iii) a group of the formula —COOH connected via an alkyl group of 1 to 4 carbon atoms,
(iv) a group of the formula —PO(OH)2 connected via an alkyl group of 1 to 6 carbon atoms, or
(v) a group of the formula —SO 2 OH connected via an alkyl group of 1 to 6 carbon atoms, or
(I) aryl which is selected from the class consisting of phenyl, naphthyl, indolyl, thiophenyl, pyridyl, pyrimidinyl, furyl, pyrrolyl, oxazolyl, thiazolyl, pyrazolyl, isoxazolyl, imidazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl, pyridazinyl, pyrazinyl, triazinyl, indolyzinyl, isoindolyl, benzo[b]furanyl, benzo[b]thiophenyl, indazolyl, benzthiazolyl, benzimidazolyl, quinolinyl, isoquinolinyl, purinyl, quinolizinyl, cinnolinyl, pthalaninyl, quinoxalinyl, napthyridinyl, pteridinyl and quinazolinyl,
wherein one or more hydrogen atoms of said aryl group are or are not independently replaced with:
(i) alkyl of 1 to 3 carbon atoms,
(ii) —COOH,
(iii) —SO 2 OH,
(iv) —PO(OH)2,
(v) a group of the formula —COOR 7 , wherein R 7 is straight or branched alkyl of 1 to 5 group atoms or cycloalkyl of 3 to 5 carbon atoms,
(vi) a group of the formula —NR 8 R 9 , wherein R 8 and R 9 are each, independently, a hydrogen atom, alkyl of 1 to 6 carbon atoms, cycloalkyl of 3 to 6 carbon atoms or acyl of 1 to 7 carbon atoms, or wherein R 8 and R 9 together form a saturated hydrocarbon bridge of 3 to 5 carbon atoms which bridge, together with the nitrogen atom between R 8 and R 9 , forms a heterocyclic ring,
(vii) a group of the formula —CONR 10 R 11 , wherein R 10 and R 11 are each, independently, a hydrogen atom, alkyl of 1 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, or wherein R 10 and R 11 together form a saturated hydrocarbon bridge of 3 to 5 carbon atoms which bridge, together with the nitrogen atom between R 10 and R 11 , forms a heterocyclic ring,
(viii) a group of the formula —OR 12a , wherein R 12a is a hydrogen atom, or an alkyl or acyl group of 1 to 7 carbon atoms,
(ix) a group of the formula —SR 12b , wherein R 12b is a hydrogen atom, or an alkyl or acyl group of 1 to 7 carbon atoms,
(x) cyano, or
(xi) an amidino group of the formula
Figure USRE038132-20030603-C00774
wherein R 13 , R 14 and R 15 are each, independently, a hydrogen atom or alkyl of 1 to 3 carbon atoms, and wherein two of R 13 , R 14 and R 15 when additionally taken together form a saturated hydrocarbon bridge of 3 to 5 carbon atoms which bridge, together with the atom(s) between the groups that form the bridge, forms a heterocyclic ring;
R 2 is:
(A) a hydrogen atom, or
(B) branched or unbranched alkyl of 1 to 3 carbon atoms or cycloalkyl of 3 to 5 carbon atoms wherein said alkyl or cycloalkyl group is unsubstituted or substituted with:
(i) a group of the formula —OR 34 , wherein R 34 is a hydrogen atom, or an alkyl or acyl group of 1 to 7 carbon atoms, or
(ii) a group of the formula —NR 35 R 36 , wherein R 35 and R 36 are each, independently, a hydrogen atom, alkyl of 1 to 2 carbon atoms, or acyl of 1 to 2 carbon atoms;
R 3 is a group of the formula —(CR 37 R 38)x(CR 39 R 40)y R 41 , wherein;
x and y are each independently 0 or 1,
R 37 , R 38 and R 39 are each, independently:
(A) a hydrogen atom,
(B) a group of the formula —OR 42 , wherein R 42 is a hydrogen atom, or an alkyl or acyl group of 1 to 7 carbon atoms, or
(C) branched or unbranched alkyl of 1 to 3 carbon atoms or cycloalkyl of 3 to 5 carbon atoms,
R 40 is:
(A) a hydrogen atom,
(B) a group of the formula —OR 42 , wherein R 42 is a hydrogen atom, or an alkyl or acyl group of 1 to 7 carbon atoms,
(C) branched or unbranched alkyl of 1 to 3 carbon atoms or cycloalkyl of 3 to 5 carbon atoms, or
(D) aryl which is selected from the class consisting of phenyl, 2 -naphthyl, 2 -, 3 -, 5 - or 6 -indolyl, 2 - or 3 -thiophenyl, 2 -, 3 - or 4 -pyridyl, 2 -, 4 - or 5 -pyrimidinyl, 2 - or 3 -furyl, 1 -, 2 - or 3 -pyrrolyl, 2 -, 4 - or 5 -oxazolyl, 2 -, 4 - or 5 -thiazolyl, 1 -, 3 -, 4 - or 5 -pyrazolyl, 3 -, 4 - or 5 -isoxazolyl, 1 -, 2 -, 4 - or 5 -imidazolyl, 3 -, 4 - or 5 -isothiazolyl, 4 - or 5 -oxadiazolyl, 1 -, 4 - or 5 -triazolyl, 2 -thiadiazolyl, 3 - or 4 -pyridazinyl, 2 -pyrazinyl, 2 -triazinyl, 2 -, - 3, 6 - or 7 -indolyzinyl, 2 -, 3 -, 5 - or 6 -isoindolyl, 2 -, 3 -, 5 - or 6 -benzo[b]furanyl, 2 -, 3 -, 5 - or 6 -benzo[b]thiophenyl, 3 -, 5 - or 6 -indazolyl, 2 -, 5 - or 6 -benzthiazolyl, 2 -, 5 - or 6 -benzimidazolyl, 2 -, 3 -, 6 - or 7 -quinolinyl, 3 -, 6 - or 7 -isoquinolinyl, 2 - or 8 -purinyl, 2 -, 3 -, 7 - or 8 -quinolizinyl, 3 -, 6 - or 7 -cinnolinyl, 6 - or 7 -pthalaninyl, 2 -, 3 -, 6 - or 7 -quinoxalinyl, 2 -, 3 -, 6 - or 7 -napthyridinyl, 2 -, 6 - or 7 -pteridinyl and 2 -, 6 - or 7 -quinazolinyl,
wherein one or more of the hydrogen atoms of said aryl group are or are not independently replaced with:
(i) R 43 , which is aryl selected from the class consisting of phenyl, 2 -naphthyl, 2 -, 3 -, 5 - or 6 -indolyl, 2 - or 3 -thiophenyl, 2 -, 3 - or 4 -pyridinyl, 2 -, 4 - or 5 -pyrimidinyl, 2 - or 3 -furyl, 1 -, 2 - or 3 -pyrrolyl, 2 -, 4 - or 5 -oxazolyl, 2 -, 4 - or 5 -thiazolyl, 1 -, 3 -, 4 - or 5 -pyrazolyl, 3 -, 4 - or 5 -isoxazolyl, 1 -, 2 -, 4 - or 5 -imidazolyl, 3 -, 4 - or 5 -isothiazolyl, 4 - or 5 -oxadiazolyl, 1 -, 4 - or 5 -triazolyl, 2 -thiadiazolyl, 3 - or 4 -pyridazinyl, 2 -pyrazinyl, 2 -triazinyl, 2 -, - 3, 6 - or 7 -indolyzinyl, 2 -, 3 -, 5 - or 6 -isoindolyl, 2 -, 3 -, 5 - or 6 -benzo[b]furanyl, 2 -, 3 -, 5 - or 6 -benzo[b]thiophenyl, 3 -, 5 - or 6 -indazolyl, 2 -, 5 - or 6 -benzthiazolyl, 2 -, 5 - or 6 -benzimidazolyl, 2 -, 3 -, 6 - or 7 -quinolinyl, 3 -, 6 - or 7 -isoquinolinyl, 2 - or 8 -purinyl, 2 -, 3 -, 7 - or 8 -quinolizinyl, 3 -, 6 - or 7 -cinnolinyl, 6 - or 7 -pthalaninyl, 2 -, 3 -, 6 - or 7 -quinoxalinyl, 2 -, 3 -, 6 - or 7 -napthyridinyl, 2 -, 6 - or 7 -pteridinyl and 2 -, 6 - or 7 -quinazolinyl,
wherein one or more of the hydrogen atoms of said aryl group are or are not independently replaced with:
(a) branched or unbranched alkyl of 1 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, which alkyl or cycloalkyl group is unsubstituted or mono- or polysubstituted with halogen or oxo,
(b) —COOH,
(c) —SO 2 OH,
(d) PO(OH)2,
(e) a group of the formula —COOR 44 , wherein R 44 is straight or branched alkyl of 1 to 5 carbon atoms or cycloalkyl of 3 to 5 carbon atoms,
(f) a group of the formula —NR 45 R 46 , wherein R 45 and R 46 are each, independently, a hydrogen atom, alkyl or fluoroalkyl of 1 to 6 carbon atoms, cycloalkyl of 3 to 6 carbon atoms or acyl of 1 to 7 carbon atoms, or wherein R 45 and R 46 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring,
(g) a group of the formula —CONR 47 R 48 , wherein R 47 and R 48 are each independently a hydrogen atom, alkyl or fluoroalkyl of 1 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, or wherein R 47 and R 48 constitute a saturated hydrocarbon bridge of 3 to 5 carbon atoms which together with the nitrogen atom between them form a heterocyclic ring,
(h) a group of the formula —OR 49 , wherein R 49 is a hydrogen atom, or an alkyl, fluoroalkyl or acyl group of 1 to 7 carbon atoms,
(i) a group of the formula —SR 50 , wherein R 50 is a hydrogen atom, or an alkyl, fluoroalkyl or acyl group of 1 to 7 carbon atoms,
(j) cyano,
(k) nitro,
(l) an amidino group of the formula
Figure USRE038132-20030603-C00775
wherein R 51 , R 52 and R 53 are each, independently, a hydrogen atom or alkyl of 1 to 3 carbon atoms, and wherein two of R 51 , R 52 and R 53 when additionally taken together form a saturated hydrocarbon bridge of 3 to 5 carbon atoms which bridge, together with the atom(s) between the two groups that form the bridge, forms a heterocyclic ring, or
(m) halogen,
(ii) methyl, which is unsubstituted or mono- or polysubstituted with fluorine atoms and additionally unsubstituted or monosubstituted with R 43,
(iii) branched or unbranched alkyl of 2 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, which alkyl or cycloalkyl group is unsubstituted or mono- or polysubstituted with halogen or oxo,
(iv) a group of the formula —COOR 54 , wherein R 54 is straight or branched alkyl of 1 to 5 carbon atoms or cycloalkyl of 3 to 5 carbon atoms,
(v) a group of the formula —NR 55 R 56 , wherein R 55 and R 56 are each, independently, a hydrogen atom, alkyl or fluoroalkyl of 1 to 6 carbon atoms, cycloalkyl of 3 to 6 carbon atoms or acyl of 1 to 7 carbon atoms, or wherein R 55 and R 56 together form a saturated hydrocarbon bridge of 3 to 5 carbon atoms which bridge, together with the nitrogen atom between R 55 and R 56 , forms a heterocyclic ring, and wherein one of R 55 and R 56 is or is not the group R 43,
(vi) a group of the formula —CONR 57 R 58 , wherein R 57 and R 58 are each, independently, a hydrogen, atom, alkyl or fluoroalkyl of 1 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, or wherein R 57 and R 58 together form a saturated hydrocarbon bridge of 3 to 5 carbon atoms which bridge, together with the nitrogen atom between R 57 and R 58 , forms a heterocyclic ring, and wherein one of R 57 and R 58 is or not the group R 43,
(vii) a group of the formula —COR 59 , wherein R 59 is a hydrogen atom, straight or branched alkyl of 1 to 5 carbon atoms, cycloalkyl of 3 to 5 carbon atoms or R43,
(viii) a group of the formula —OR 60 , wherein R 60 is a hydrogen atom, an alkyl, fluoroalkyl or acyl group of 1 to 7 carbon atoms, or R 43,
(ix) a group of the formula —SR 61 , wherein R 61 is a hydrogen atom, an alkyl, fluoroalkyl or acyl group of 1 to 7 carbon atoms, or R 43,
(x) cyano,
(xi) nitro, or
(xii) halogen,
R 41 is:
aryl selected from the class consisting of phenyl, 2 -naphthyl, 2 -, 3 -, 5 - or 6 -indolyl, 2 - or 3 -thiophenyl, 2 -, 3 - or 4 -pyridyl, 2 -, 4 - or 5 -pyrimidinyl, 2 - or 3 -furyl, 1 -, 2 - or 3 -pyrrolyl, 2 -, 4 - or 5 -oxazolyl, 2 -, 4 - or 5 -thiazolyl, 1 -, 3 -, 4 - or 5 -pyrazolyl, 3 -, 4 - or 5 -isoxazolyl, 1 -, 2 -, 4 - or 5 -imidazolyl, 3 -, 4 - or 5 -isothiazolyl, 4 - or 5 -oxadiazolyl, 1 -, 4 - or 5 -triazolyl, 2 -thiadiazolyl, 3 - or 4 -pyridazinyl, 2 -pyrazinyl, 2 -triazinyl, 2 -, 3 -, 6 - or 7 -indolyzinyl, 2 -, 3 -, 5 - or 6 -isoindolyl, 2 -, 3 -, 5 - or 6 -benzo[b]furanyl, 2 -, 3 -, 5 - or 6 -benzo[b]thiophenyl, 3 -, 5 - or 6 -indazolyl, 2 -, 5 - or 6 -benzthiazolyl, 2 -, 5 - or 6 -benzimidazolyl, 2 -, 3 -, 6 - or 7 -quinolinyl, 3 -, 6 - or 7 -isoquinolinyl, 2 - or 8 -purinyl, 2 -, 3 -, 7 - or 8 -quinolizinyl, 3 -, 6 - or 7 -cinnolinyl, 6 - or 7 -pthalaninyl, 2 -, 3 -, 6 - or 7 -quinoxalinyl, 2 -, 3 -, 6 - or 7 -napthyridinyl, 2 -, 6 - or 7 -pteridinyl and 2 -, 6 - or 7 -quinazolinyl,
wherein one or more of the hydrogen atoms of said aryl group are or are not independently replaced with:
(A) R 62 , which is aryl selected from the class consisting of phenyl, 2 -naphthyl, 2 -, 3 -, 5 - or 6 -indolyl, 2 - or 3 -thiophenyl, 2 -, 3 - or 4 -pyridyl, 2 -, 4 - or 5 -pyrimidinyl, 2 - or 3 -furyl, 1 -, 2 - or 3 -pyrrolyl, 2 -, 4 - or 5 -oxazolyl, 2 -, 4 - or 5 -thiazolyl, 1 -, 3 -, 4 - or 5 -pyrazolyl, 3 -, 4 - or 5 -isoxazolyl, 1 -, 2 -, 4 - or 5 -imidazolyl, 3 -, 4 - or 5 -isothiazolyl, 4 - or 5 -oxadiazolyl, 1 -, 4 - or 5 -triazolyl, 2 -thiadiazolyl, 3 - or 4 -pyridazinyl, 2 -pyrazinyl, 2 -triazinyl, 2 -, - 3, 6 - or 7 -indolyzinyl, 2 -, 3 -, 5 - or 6 -isoindolyl, 2 -, 3 -, 5 - or 6 -benzo[b]furanyl, 2 -, 3 -, 5 - or 6 -benzo[b]thiophenyl, 3 -, 5 - or 6 -indazolyl, 2 -, 5 - or 6 -benzthiazolyl, 2 -, 5 - or 6 -benzimidazolyl, 2 -, 3 -, 6 - or 7 -quinolinyl, 3 -, 6 - or 7 -isoquinolinyl, 2 - or 8 -purinyl, 2 -, 3 -, 7 - or 8 -quinolizinyl, 3 -, 6 - or 7 -cinnolinyl, 6 - or 7 -pthalaninyl, 2 -, 3 -, 6 - or 7 -quinoxalinyl, 2 -, 3 -, 6 - or 7 -napthyridinyl, 2 -, 6 - or 7 -pteridinyl and 2 -, 6 - or 7 -quinazolinyl,
wherein one or more of the hydrogen atoms of said aryl group are or are not independently replaced with:
(i) branched or unbranched alkyl of 1 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, which alkyl or cycloalkyl group is unsubstituted or mono- or polysubstituted with halogen or oxo,
(ii) —COOH,
(iii) SO 2 OH,
(iv) —PO(OH)2,
(v) a group of the formula —COOR 63 , wherein R 63 is straight or branched alkyl of 1 to 5 carbon atoms or cycloalkyl of 3 to 5 carbon atoms,
(vi) a group of the formula —NR 64 R 65 , wherein R 64 and R 65 are each, independently, a hydrogen atom, alkyl or fluoroalkyl of 1 to 6 carbon atoms, cycloalkyl of 3 to 6 carbon atoms or acyl of 1 to 7 carbon atoms, or wherein R 64 and R 65 together form a saturated hydrocarbon bridge of 3 to 5 carbon atoms which bridge, together with the nitrogen atom between R 64 and R 65 , forms a heterocyclic ring,
(vii) a group of the formula —CONR 66 R 67 , wherein R 66 and R 67 are each, independently, a hydrogen atom, alkyl or fluoroalkyl of 1 to 6 carbon atoms or cycloalkyl of 3 to 6 carbon atoms, or wherein R 66 and R 67 together form a saturated hydrocarbon bridge of 3 to 5 carbon atoms which bridge, together with the nitrogen atom between R 66 and R 67 , forms a heterocyclic ring,
(viii) a group of the formula —OR 68 , wherein R 68 is a hydrogen atom, or an alkyl, fluoroalkyl or acyl group of 1 to 7 carbon atoms,
(ix) a group of the formula —SR 69 , wherein R 69 is a hydrogen atom, or an alkyl, fluoroalkyl or acyl group of 1 to 7 carbon atoms,
(x) cyano,
(xi) nitro, or
(xii) an amidino group of the formula
Figure USRE038132-20030603-C00776
wherein R 70 , R 71 and R 72 are each, independently, a hydrogen atom or alkyl or fluoroalkyl of 1 to 3 carbon atoms, and wherein two of R 70 , R 71 and R 72 when additionally taken together form a saturated hydrocarbon bridge of 3 to 5 carbon atoms which bridge, together with the atom(s) between the two groups that form the bridge, forms a heterocyclic ring, or
(xiii) halogen,
(B) methyl, which is unsubstituted or mono- or polysubstituted with fluorine atoms and additionally is unsubstituted or monosubstituted with R 62,
(C) branched or unbranched alkyl of 2 to 6 carbon atoms, or cycloalkyl of 3 to 6 carbon atoms, which alkyl or cycloalkyl group is unsubstituted or mono- or polysubstituted with halogen or oxo,
(D) a group of the formula —COOR 73 , wherein R 73 is straight or branched alkyl of 1 to 5 carbon atoms or cycloalkyl of 3 to 5 carbon atoms,
(E) a group of the formula —NR 74 R 75 , wherein R 74 and R 75 are each, independently, a hydrogen atom, alkyl or fluoroalkyl of 1 to 6 carbon atoms, cycloalkyl of 3 to 6 carbon atoms or acyl of 1 to 7 carbon atoms, or wherein R 74 and R 75 together form a saturated hydrocarbon bridge of 3 to 5 carbon atoms which bridge, together with the nitrogen atom between R 74 and R 75 , forms a heterocyclic ring, and wherein one of R 74 and R 75 is or is not the group R 62,
(F) a group of the formula —CONR 76 R 77 , wherein R 76 and R 77 are each, independently, a hydrogen atom, alkyl or fluoroalkyl of 1 to 6 carbon atoms, or cycloalkyl of 3 to 6 carbon atoms, or wherein R 76 and R 77 together form a saturated hydrocarbon bridge of 3 to 5 carbon atoms which bridge, together with the nitrogen atom between R 76 and R 77 , forms a heterocyclic ring, and wherein one of R 76 and R 77 is or is not the group R 62,
(G) a group of the formula —COR 78 , wherein R 78 is a hydrogen atom, straight or branched alkyl of 1 to 5 carbon atoms, cycloalkyl of 3 to 5 carbon atoms or R 62,
(H) a group of the formula —OR 79 , wherein R 79 is a hydrogen atom, an alkyl, fluoroalkyl or acyl group of 1 to 7 carbon atoms, or R 62,
(I) a group of the formula —SR 80 , wherein R 80 is a hydrogen atom, an alkyl, fluoroalkyl or acyl group of 1 to 7 carbon atoms, or R 62,
(J) cyano,
(K) nitro, or
(L) halogen;
R 4 is Cl or trifluoromethyl; and,
R 5 and R 6 are each, independently, a hydrogen, fluorine, chlorine, bromine or iodine atom, methyl or trifluoromethyl;
or a pharmaceutically acceptable salt thereof.
14. The method of claim 13 wherein the disease or condition is selected from the group consisting of psoriasis, organ/tissue transplant rejection, graft vs. host reactions, and autoimmune diseases.
US10/167,732 1997-03-03 2002-06-12 Phenylpyrrolidines, phenylimidazolidines,3-phenyl-1,3-oxizolidines and 3-phenyl-1,3-thiazolidines and their use in the treatment of inflammatory disease Expired - Lifetime USRE38132E1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/167,732 USRE38132E1 (en) 1997-03-03 2002-06-12 Phenylpyrrolidines, phenylimidazolidines,3-phenyl-1,3-oxizolidines and 3-phenyl-1,3-thiazolidines and their use in the treatment of inflammatory disease

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US4001197P 1997-03-03 1997-03-03
US3314898A 1998-03-02 1998-03-02
PCT/US1998/004254 WO1998039303A1 (en) 1997-03-03 1998-03-03 Small molecules useful in the treatment of inflammatory disease
US09/375,010 US6355664B1 (en) 1997-03-03 1999-08-16 Phenylpyrrolidines, phenylimidazolidines, 3-phenyl-1,3-oxizolidines and 3-phenyl-1,3-thiazolidines and their use in the treatment of inflammatory disease
US10/167,732 USRE38132E1 (en) 1997-03-03 2002-06-12 Phenylpyrrolidines, phenylimidazolidines,3-phenyl-1,3-oxizolidines and 3-phenyl-1,3-thiazolidines and their use in the treatment of inflammatory disease

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/375,010 Reissue US6355664B1 (en) 1997-03-03 1999-08-16 Phenylpyrrolidines, phenylimidazolidines, 3-phenyl-1,3-oxizolidines and 3-phenyl-1,3-thiazolidines and their use in the treatment of inflammatory disease

Publications (1)

Publication Number Publication Date
USRE38132E1 true USRE38132E1 (en) 2003-06-03

Family

ID=27364323

Family Applications (1)

Application Number Title Priority Date Filing Date
US10/167,732 Expired - Lifetime USRE38132E1 (en) 1997-03-03 2002-06-12 Phenylpyrrolidines, phenylimidazolidines,3-phenyl-1,3-oxizolidines and 3-phenyl-1,3-thiazolidines and their use in the treatment of inflammatory disease

Country Status (1)

Country Link
US (1) USRE38132E1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8779157B2 (en) 2009-09-04 2014-07-15 Vanderbilt University MGLUR4 allosteric potentiators, compositions, and methods of treating neurological dysfunction

Citations (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE1940032A1 (en) 1968-08-12 1970-03-12 Sumitomo Chemical Co Substituted N-phenyl succinimides, processes for their preparation and their use for biocidal agents
DE1958183A1 (en) 1968-11-25 1970-06-04 Sumitomo Chemical Co Microbicidal imidazolidinedione erivs
DE2100800A1 (en) 1970-01-12 1971-07-15 Sumitomo Chemical Co Ltd , Osaka (Japan) Process for the preparation of cycli see arylimides
US3668217A (en) 1968-11-25 1972-06-06 Sumitomo Chemical Co 3-(3{40 ,5{40 -dihalogenophenyl)imidazolidine-2,4-dione derivatives
US3846441A (en) * 1970-12-09 1974-11-05 Sumitomo Chemical Co 1,5-disubstituted-3-substituted phenyl hydantoins
EP0091596A2 (en) 1982-04-08 1983-10-19 Shell Internationale Researchmaatschappij B.V. Hydantoins, their preparation and application
EP0343643A2 (en) 1988-05-25 1989-11-29 Warner-Lambert Company Arylmethylenyl derivatives of thiazolidnones, imidazolidinones and oxazolidinones useful as antiallergy agents and antiinflamatory agents
US4911748A (en) * 1988-12-22 1990-03-27 Ici Americas Inc. Herbicidal 5-substituted-3-phenyl-imidazolidine-2,4-diones
US4977270A (en) * 1988-12-01 1990-12-11 Ici Americas Inc. Substituted 2,4-dioxodiazolidines and -thiadiazolidines and their use as herbicides
US5208250A (en) 1988-05-25 1993-05-04 Warner-Lambert Company Known and selected novel arylmethylenyl derivatives of thiazolidinones, imidazolidinones and oxazolidinones useful as antiallergy agents and anti-inflammatory agents
EP0545478A1 (en) 1991-12-03 1993-06-09 MERCK SHARP &amp; DOHME LTD. Heterocyclic compounds as tachykinin antagonists
US5306822A (en) 1988-05-25 1994-04-26 Warner-Lambert Company Arylmethylenyl derivatives of oxazolidinone
WO1995018794A1 (en) 1994-01-05 1995-07-13 Roussel Uclaf Novel optionally substituted phenylimidazolidines, intermediates and process for their preparation, their use as drugs and pharmaceutical compositions containing them

Patent Citations (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE1940032A1 (en) 1968-08-12 1970-03-12 Sumitomo Chemical Co Substituted N-phenyl succinimides, processes for their preparation and their use for biocidal agents
US3741981A (en) 1968-08-12 1973-06-26 Sumitomo Chemical Co N-phenylsuccinimide derivatives
DE1958183A1 (en) 1968-11-25 1970-06-04 Sumitomo Chemical Co Microbicidal imidazolidinedione erivs
US3668217A (en) 1968-11-25 1972-06-06 Sumitomo Chemical Co 3-(3{40 ,5{40 -dihalogenophenyl)imidazolidine-2,4-dione derivatives
DE2100800A1 (en) 1970-01-12 1971-07-15 Sumitomo Chemical Co Ltd , Osaka (Japan) Process for the preparation of cycli see arylimides
US3846441A (en) * 1970-12-09 1974-11-05 Sumitomo Chemical Co 1,5-disubstituted-3-substituted phenyl hydantoins
EP0091596A2 (en) 1982-04-08 1983-10-19 Shell Internationale Researchmaatschappij B.V. Hydantoins, their preparation and application
US4944791A (en) 1982-04-08 1990-07-31 Shell Internationale Research Maatschappij B.V. Herbicidal hydantoins
EP0343643A2 (en) 1988-05-25 1989-11-29 Warner-Lambert Company Arylmethylenyl derivatives of thiazolidnones, imidazolidinones and oxazolidinones useful as antiallergy agents and antiinflamatory agents
US5306822A (en) 1988-05-25 1994-04-26 Warner-Lambert Company Arylmethylenyl derivatives of oxazolidinone
US5208250A (en) 1988-05-25 1993-05-04 Warner-Lambert Company Known and selected novel arylmethylenyl derivatives of thiazolidinones, imidazolidinones and oxazolidinones useful as antiallergy agents and anti-inflammatory agents
US5464856A (en) 1988-05-25 1995-11-07 Warner-Lambert Company Arylmethylenyl derivatives of imidazolidinones useful as antiinflammatory agents
US4977270A (en) * 1988-12-01 1990-12-11 Ici Americas Inc. Substituted 2,4-dioxodiazolidines and -thiadiazolidines and their use as herbicides
US4911748A (en) * 1988-12-22 1990-03-27 Ici Americas Inc. Herbicidal 5-substituted-3-phenyl-imidazolidine-2,4-diones
US5334606A (en) 1991-12-03 1994-08-02 Merck Sharpe & Dohme Ltd. Oxazolidindione substituted indole derivatives
EP0545478A1 (en) 1991-12-03 1993-06-09 MERCK SHARP &amp; DOHME LTD. Heterocyclic compounds as tachykinin antagonists
WO1995018794A1 (en) 1994-01-05 1995-07-13 Roussel Uclaf Novel optionally substituted phenylimidazolidines, intermediates and process for their preparation, their use as drugs and pharmaceutical compositions containing them
US5750553A (en) 1994-01-05 1998-05-12 Roussel Uclaf Optionally substituted phenylimidazolidines, their preparation process and intermediates, their use as medicaments and the pharmaceutical compositions containing them

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
Agric. Biol. Chem. 1982, 46, 2755-8 Takayama et al.
J. Pharm. Sci. 1984, 73, 553-8 Halim et al.
Monatshefte für Chemie, 1994, 125, 1437-1442 Li et al.

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8779157B2 (en) 2009-09-04 2014-07-15 Vanderbilt University MGLUR4 allosteric potentiators, compositions, and methods of treating neurological dysfunction

Similar Documents

Publication Publication Date Title
US6355664B1 (en) Phenylpyrrolidines, phenylimidazolidines, 3-phenyl-1,3-oxizolidines and 3-phenyl-1,3-thiazolidines and their use in the treatment of inflammatory disease
EP0966447B1 (en) Small molecules useful in the treatment of inflammatory disease
US6414153B1 (en) 1-phenylpydrrolidin-2-ones and -thiones and 1-(4-pyridyl)pydrrolidin-2-ones and -thiones which are useful in the treatment of inflammatory disease
US6350763B1 (en) Small molecules useful in the treatment of inflammation disease
Petersen et al. Synthesis and hypotensive activity of N-alkyl-N''-cyano-N'-pyridylguanidines
US6365615B1 (en) Small molecules useful in the treatment of inflammatory disease
US6624186B2 (en) Ion channel modulating agents comprising novel benzimidazolone derivatives
US7572921B2 (en) Substituted benzylimidazoles useful for the treatment of inflammatory diseases
US6353013B1 (en) Small molecules useful in the treatment of inflammatory disease
CS249544B2 (en) Method of new imidazolylphenylamidines production
US20030216390A1 (en) Melanin concentrating hormone receptor ligands: substituted benzoimidazole analogues
CA1337072C (en) 5-substituted [4,5-c] imidazopyridines
US20230183183A1 (en) Metabolites of [3-(4-(2-butyl-1-[4-(4-chloro-phenoxy)-phenyl]-1h-imidazol-4 yl)-phenoxy)-propyl]-diethyl-amine
USRE38132E1 (en) Phenylpyrrolidines, phenylimidazolidines,3-phenyl-1,3-oxizolidines and 3-phenyl-1,3-thiazolidines and their use in the treatment of inflammatory disease
KR20170012360A (en) Small molecule lfa-1 inhibitors
JP2003137872A (en) Substituted 2-cyclohexyl-4-phenyl-1h-imidazole derivative
US6509366B2 (en) Substituted imidazoles as selective modulators of Bradykinin B2 receptors
CZ246495A3 (en) Imidazolequinoline derivative as eaa antagonist
JP2002541145A (en) Aryl and heteroaryl fused aminoalkyl-imidazole derivatives: Bradykinin B2 receptor selective modulators
JPH0748369A (en) Substituted monopyridylmethyl and bipyridylmethyl derivatives
US20030232817A1 (en) Small molecules useful for the treatment of inflammatory disease
SE466104B (en) NEW TRIAZOLPYRIMIDINES AND PROCEDURES FOR THEIR PREPARATION
CZ314599A3 (en) Pyrrolidine derivatives and pharmaceutical preparation
MXPA99007583A (en) Small molecules useful in the treatment of inflammatory disease
HRP970150A2 (en) Machine for drilling lubrication holes and machine for milling lubrication grooves on the &#34;sulzer&#34; marine motor cylinder liners

Legal Events

Date Code Title Description
FPAY Fee payment

Year of fee payment: 4

FEPP Fee payment procedure

Free format text: PAYOR NUMBER ASSIGNED (ORIGINAL EVENT CODE: ASPN); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

FPAY Fee payment

Year of fee payment: 8

FPAY Fee payment

Year of fee payment: 12