WO2002102315A2 - QUINAZOLINE AND PYRIDO[2,3-d]PYRIMIDINE INHIBITORS OF PHOSPHODIESTERASE (PDE) 7 - Google Patents

QUINAZOLINE AND PYRIDO[2,3-d]PYRIMIDINE INHIBITORS OF PHOSPHODIESTERASE (PDE) 7 Download PDF

Info

Publication number
WO2002102315A2
WO2002102315A2 PCT/US2002/019130 US0219130W WO02102315A2 WO 2002102315 A2 WO2002102315 A2 WO 2002102315A2 US 0219130 W US0219130 W US 0219130W WO 02102315 A2 WO02102315 A2 WO 02102315A2
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
optionally substituted
groups
heteroaryl
heterocyclo
Prior art date
Application number
PCT/US2002/019130
Other languages
French (fr)
Other versions
WO2002102315A3 (en
Inventor
William J. Pitts
Joseph Barbosa
Original Assignee
Bristol-Myers Squibb Company
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Bristol-Myers Squibb Company filed Critical Bristol-Myers Squibb Company
Priority to EP02742138A priority Critical patent/EP1404337A4/en
Priority to CA002450724A priority patent/CA2450724A1/en
Priority to JP2003504904A priority patent/JP2005506961A/en
Publication of WO2002102315A2 publication Critical patent/WO2002102315A2/en
Publication of WO2002102315A3 publication Critical patent/WO2002102315A3/en

Links

Classifications

    • 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
    • C07D403/12Heterocyclic 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 chain containing hetero atoms as chain links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/513Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim having oxo groups directly attached to the heterocyclic ring, e.g. cytosine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • A61K31/52Purines, e.g. adenine
    • A61K31/522Purines, e.g. adenine having oxo groups directly attached to the heterocyclic ring, e.g. hypoxanthine, guanine, acyclovir
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/04Drugs for disorders of the respiratory system for throat disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/04Antipruritics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/14Drugs for dermatological disorders for baldness or alopecia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/14Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/08Vasodilators for multiple indications
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • 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
    • C07D401/12Heterocyclic 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 chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D473/00Heterocyclic compounds containing purine ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D473/00Heterocyclic compounds containing purine ring systems
    • C07D473/02Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6
    • C07D473/16Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6 two nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/10Spiro-condensed systems

Definitions

  • the present invention relates to quinazoline and pyrido[2,3-d]pyrimidine inhibitors of phosphodiesterase 7 (PDE 7) (including both selective inhibitors of PDE 7, and dual inhibitors of PDE 7 and phosphodiesterase 4), pharmaceutical compositions containing these inhibitors, and the use of these inhibitors in the treatment of leukocyte activation-associated or leukocyte-activation mediated disease and inflammatory diseases either alone or in combination with other therapeutic agents.
  • PDE 7 phosphodiesterase 7
  • PDEs Phosphodiesterases hydrolyze the second messenger molecules cAMP and cGMP to affect cellular signaling.
  • PDE3 ,4,7,8 are specific for cAMP, and others (PDE5,6,9) for cGMP.
  • Further family members (PDE 1,2, 10,11) have dual specificity.
  • a recent publication demonstrated a role for PDE7 in the activation and/or proliferation of T cells(Zi, Yee and Beavo, Science 283:848-851, 1999). Resting T lymphocytes express mainly PDE3 and PDE4. However, upon activation, T cells dramatically upregulate PDE7 and appear to rely on this isozyme for regulation of cAMP levels.
  • a PDE7 inhibitor is defined herein as a compound for which the IC 50 of the compound in a PDE7 inhibition assay is less than 20 micromolar (preferably less than 10 micromolar, more preferably less than 5 micromolar, most preferably less than 1 micromolar).
  • the PDE7 IC 50 of a selective PDE7 inhibitor should be less than one-tenth the IC50 of said compound in all of the following PDE assays: PDE1, PDE3 and PDE4 (more preferably the PDE7 IC 5 o of a selective PDE7 inhibitor should be less than one- twentieth the IC 50 of said compound in the following PDE assays: PDE1 and PDE3, most preferably the PDE7 IC 50 of a selective PDE7 inhibitor should be less than one-hundreth the IC 50 of said compound in a PDE3 assay).
  • PDE1 inhibitors have demonstrated potent vasodilator activity. Such activity would represent an undesirable side effect in a therapeutic agent with the utilities listed in this patent for a PDE7 inhibitor.
  • the PDE3 family of enzymes are distributed in several tissues including the heart liver, and platelets.
  • PDE3 inhibitors have demonstrated potent cardiac iotropic activity. Such activity would represent an undesirable side effect in a therapeutic agent with the utilities listed in this patent for a PDE7 inhibitor.
  • Several isoforms of PDE4 exist, and these are expressed in a wide variety of tissues including heart, kidney, brain, the gastrointestinal track and circulating blood cells.
  • PDE4 inhibitors have demonstrated clinical utility for COPD, and have also been suggested to have utility for rheumatoid arthritis, and multiple sclerosis, and to possess anti- inflammatory activity.
  • the utility of PDE4 inhibitors has been limited to some extent by their propensity to cause emesis. As such there are circumstances where it would be desirable to develop PDE7 inhibitors, which have a degree of selectivity against PDE.
  • a selective inhibitor of PDE7 is expected to have broad application as an immunosuppressant in T cell-mediated diseases. PDE7 inhibitors will act at a different stage of the T cell signaling process compared to current immunosuppressants by inhibiting a very early stage of the T cell activation cascade.
  • a selective inhibitor of PDE7 is also expected to have a decreased potential for clinically significant side effects compared to current immunosuppressants, therefore the primary disease indications are solid organ transplantation (SOT) and rheumatoid arthritis. Additional indications may include EBD, psoriasis, asthma and lupus.
  • SOT solid organ transplantation
  • Additional indications may include EBD, psoriasis, asthma and lupus.
  • a dual PDE7-PDE4 inhibitor (PDE4/7 or PDE7/4) is defined herein as any compound which has an IC50 in both a PDE7 and a PDE4 inhibition assay of less than 20 micromolar (preferably less than 10 micromolar, and more preferably less than 5 micromolar and most preferably less than 1 micromolar), and an IC50 in a PDE3 inhibition assay which is at least 10 times higher than the IC50 of the compound in the PDE7 assay (more preferably at least 20 times higher than the IC50 of the compound in the PDE7 assay, and most preferably at least 100 times higher than the IC50 of the compound in the PDE7 assay).
  • a dual PDE4/7 inhibitor should have a ratio of inhibition or PDE7 IC50 divided by PDE4 IC50 of between one-tenth and 100.
  • Inhibitors that exhibit such a ratio of inhibition include those that inhibit PDE3, PDE4 and PDE7 as described above, and further inhibit PDE1 at an IC50 at least 10 times higher than the
  • IC50 of the compound in a PDE7 assay (more preferably at least 20 times higher than the
  • Preferred dual PDE7-PDE4 inhibitors further include those compounds that inhibit PDE3, PDE4 and PDE7 as described above, and further suppress both T cell proliferation, and TNF-alpha secretion from either THP-1 monocytes or human peripheral blood mononuclear cells at a level of less than 20 micromolar.
  • Leukocyte activation is defined herein as any or all of leukocyte (T cell, monocyte macrophage, neutrophil etc.) cell proliferation, cytokine production, adhesion protein expression, and production of inflammatory mediators. This is mediated in part by the action of PDE4 and/or PDE7 depending on the particular leukocyte under consideration.
  • leukocyte activation associated or leukocyte activation mediated disorders include transplant rejection, graph verses host disease, and autoimmune disorders, such as rheumatoid arthritis, multiple sclerosis, juvenile diabetes, COPD, asthma, and inflammatory bowel disease, T-cell mediated hypersensitivity diseases, ischemic or reperfusion injury, and T-cell proliferative disorders.
  • Dual PDE4/7 inhibitors would be expected to block the T cell component of a disease as well as possess anti-inflammatory activity.
  • a dual PDE4/7 inhibitor which is not significantly limited by emesis, may be more effective than either a selective
  • PDE4 inhibitor or a selective PDE7 inhibitor in a variety of disease states such as rheumatoid arthritis, asthma, COPD and multiple sclerosis.
  • PDE7A (EC 3.1.4.17) has two isoforms generated by alternate splicing; PDE7A1 restricted mainly to T cells and the brain, and PDE7A2 for which mRNA is expressed in a number of cell types including muscle cells.
  • the isoforms have different sequence at the amino termini, and it is thought that this portion of each molecule is likely to be important for cellular localization of the enzyme.
  • the present invention provides quinazoline and pyrido[2,3-d]pyrimidine compounds of the following formula (I), their enantiomers, diastereomers, tautomers and pharmaceutically acceptable salts, prodrugs and solvates thereof, for use as PDE7 inhibitors and dual PDE4/7 inhibitors:
  • R 1 is H or alkyl
  • L is (a) -OR 4 , -C(0)R 4 , -C(0)OR 4 , -SR 4 , -NR 3 R 4 , -C(0)NR 3 R 4 , -NR 3 S0 2 R 4b halogen, nitro, haloalkyl; or (b) alkyl, aryl, heteroaryl, heterocyclo, or cycloalkyl any of which may be optionally substituted with one to three groups T la , T 2a T 3a ; Y 1 , Y 2 and Y 3 are independently (a) hydrogen, halo, -OR 4a , or
  • R 3 and R 4 are independently H, alkyl, alkenyl, aryl, (aryl)alkyl, heteroaryl,
  • heteroarylalkyl cycloalkyl, (cycloalkyl)alkyl, heterocylo or (heterocyclo)alkyl any of which may be optionally substituted with one to three groups T la , T 2a or
  • T 3a. or R 3 and R 4 together with the nitrogen atom to which they are attached may combine to form a 4 to 8 membered heterocyclo ring optionally substituted with one to three groups T la , T 2a or T 3a ;
  • R 4a is hydrogen, alkyl, alkenyl, aryl, heteroaryl, (aryl)alkyl, (heteroaryl)alkyl, heterocylo , (heterocyclo)alkyl, cycloalkyl or (cycloalkyl)alkyl any of which may be optionally substituted with one to three groups T lb , T 2b or T 3b ;
  • R 4b is alkyl, alkenyl, aryl, (aryl)alkyl, heteroaryl, (heteroaryl)alkyl, cycloalkyl, (cycloalkyl) lkyl, heterocylo or (heterocyclo)alkyl any of which may be optionally substituted with one to three groups T la , T 2a or T 3a ;
  • Z is N or CH;
  • T b , ⁇ 2"2b ; and T 3"3b are are each independently
  • (1) are each independently hydrogen or a group provided in the definition of T , or
  • T 7 and T 8 may together be alkylene or alkenylene, completing a 3- to 8- membered saturated or unsaturated ring together with the atoms to which they are attached, which ring is unsubstituted or substituted with one or more groups listed in the description of T l lb , T 2 2b and T 3 3b , or
  • T 7 or T 8 together with T 9 , may be alkylene or alkenylene completing a 3- to 8-membered saturated or unsaturated ring together with the nitrogen atoms to which they are attached, which ring is unsubstituted or substituted with one or more groups listed in the description of T “ , T " and T " , or
  • T 1 ' and T 12 are each independently (1) a single bond
  • Preferred compounds of Formula I include those wherein: L is
  • aryl or heteroaryl either of which may be optionally substituted with one or more T la , T 2a , T 3a (especially cyano, optionally substituted alkyl, (hydroxy)alkyl, -OH, -OT 6 , -ST 6 , -SO t T 6 , -CO t H, -CO t T 6 , -T 4 NT 7 T 8 , or -T 4 N(T 10 )-T 5 -T 6 ); (c) optionally substituted alkyl (especially substituted with one or more -OH, -CO,H, -CO t T 6 , -f-NTV, -T 4 -N(T 10 )-T 5 -H, or ; -T 4 -N(T 10 )-T 5 -T 6 ); Y 1 , Y 2 and Y 3 are independently (a) H, -OR 4a or (b) alkyl or alkenyl either of which may be
  • heteroaryl (more preferably thiazolyl or oxazolyl) optionally substituted with one to three groups T 1 , T 2 , T 3 , preferably including H, alkyl, haloalkyl, halo, heteroaryl, cyano, C(0) t T 6 , OT 6 , -T 4 NT 7 T 8 ;
  • aryl substituted with one to three groups T 1 , T 2 , T 3 preferably including heteroaryl (preferably, imidazolyl, oxazolyl, or thiazolyl any of which may be further optionally substituted), cyano, C(0) t T 6 , S(0) t N(T 9 )T 6 , halo alkyl, and haloalkyl); or
  • aryl fused to a heterocyclo ring e.g., 2,3-dihydro-lH-indole bound through the aryl ring, quinolyl bound through the aryl ring (especially quinol-6-yl), quinazolinyl bound through the aryl ring (especially quinazolin-7-yl), cinnolinyl bound through the aryl ring (especially cinnolin-6-yl), isoqinolinyl bound through the aryl ring (especially isoquinol-6-yl), and phthalazinyl bound through the aryl ring (especially phthalazin-6-yl)) wherein the combined ring system may be optionally substituted with one to three groups T 1 , T 2 , T 3 (especially halo, OH, OT 6 , alkyl, -CO t H, -CO t T 6 , or -C(0)NT 7 T 8 ); R 3 is H or optionally substituted
  • heteroaryl (heteroaryl)alky where the heteroaryl group is optionally independently substituted with one or more groups T la , T 2a , T 3a (especially optionally substituted alkyl, halo, cyano, nitro, (hydroxy)alkyl, -OH, -OT 6 , -ST 6 ,
  • heterocycloalkyl where the heterocyclo group is optionally independently substituted with one or more groups T la , T 2a , T 3a (especially optionally substituted alkyl, halo, cyano, nitro, oxo, (hydroxy)alkyl, -OH, -OT 6 ,
  • T 3a (especially -OH, -OT 6 , -CO t H, -CO,T 6 , -T 4 NT 7 T 8 or -T 4 -N(T 10 )-T 5 -T 6 ); (f) heterocyclo optionally independently substituted with one or more groups T la ,
  • T 2a , T 3a (especially optionally substituted alkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted aralkyl, optionaly subsituted heterocyclo, cyano, -OH, -OT 6 , -CO t H, -CO t T 6 , oxo, hydroxy(alkyl), (alkoxy)alkyl, -T 4 -N(T 10 )-T 5 -T 6 , or -T 4 -NT 7 T 8 ); or R and R 4 together with the nitrogen atom to which they are attached combine to form a 4 to 8-membered heterocyclo ring (especially pyrrolidinyl, piperadinyl, piperazinyl, morpholinyl, diazapanyl or l,4-dioxa-8-azaspiro[4.5]decan-8-yl) optionally substituted with one to three groups T la , T 2a , T 3a
  • More preferred compounds of the present invention include compounds wherein:
  • L is (a) halogen, alkoxy, haloalkyl, -NR 3 R 4 , -C(0)OR 4 , -C(0)NR 3 R 4 (especially
  • aryl or heteroaryl either of which may be optionally substituted with one or more T la , T 2a , T 3a selected from cyano, optionally substituted alkyl, (hydroxy)alkyl, -OH, -OT 6 , -ST 6 , -SO t T 6 , -CO t H, -CO t T 6 , -T 4 NT 7 T 8 , or
  • T 4 is a bond or -C(0)-;
  • T 5 is -C(0)-, or -C(0)0-;
  • T 6 is alkyl or haloalkyl;
  • T 7 and T 8 are independently H; alkyl optiontionally substituted with cycloalkyl, heteroaryl, hydroxy or -NT 7 T 8 ; cycloalkyl; or aryl optionally substituted with halogen; or T 7 and T 8 together with the nitrogen atom to which they are attached combine to form a heterocyclo ring optionally substituted with (hydroxy)alkyl, CO t H or CO t T 6 T 10 is hydrogen;
  • T 4 is -C(O)-;
  • T 5 is -alkylene-O-;
  • T 6 is alkyl
  • T and T are independently H, alkyl, cycloalkyl, aryl, (aryl)alkyl (optionally substituted as described in the definition of R 4 ), or heterocyclo (optionally substituted as described in the definition of R 3 and R 4 combining to form a heterocyclo ring); and T ,0 is H; Y 1 , Y 2 and Y 3 are independently H or -OR 4a ; R 1 is H or alkyl; R 2 is (a) heteroaryl (more preferably thiazolyl or oxazolyl) optionally substituted with one to three groups T 1 , T 2 , T 3 , preferably including H, alkyl, haloalkyl, halo, heteroaryl, cyano, C(0) t T 6 , OT 6 , -T 4 NT 7 T 8 ;
  • aryl substituted with one to three groups T 1 , T 2 , T 3 preferably including heteroaryl (preferably, imidazolyl, oxazolyl, or thiazolyl any of which may be further optionally substituted), cyano, C(0) t T 6 , S(0) t N(T 9 )T 6 , halo alkyl, and haloalkyl); or
  • aryl fused to a heterocyclo ring especially quinolinyl or quinazolinyl bound through the aryl ring
  • the combined ring system may be optionally substituted with one to three groups T 1 , T 2 , T 3 (especially halo, OH, OT 6 , alkyl, -CO,H, -CO.T 6 , or -C(0)NT 7 T 8 );
  • R 3 is H or optionally substituted alkyl (especially substituted with one or more -OH, or -
  • T 4 is a bond, -S0 2 -, or -C(O)-;
  • T 5 is -S0 2 -, or -alkylene-O-;
  • T 6 is alkyl, or cycloalkyl;
  • T 7 and T 8 are independently H or alkyl; and T y and T 1'0 u are hydrogen; (c) (heteroaryl)alky where the heteroaryl group is optionally independently substituted with one or more groups T la , T 2a , T 3a selected from optionally substituted alkyl, halo, cyano, nitro, oxo, (hydroxy)alkyl, -OH, -OT , -ST 6 , -CO t H, -CO t T 6 , -S0 3 H, -SO t T 6 , -SO t N(T 9 )(T 6 ), -T 4 NT 7 T 8 , -T 4 -N(T 10 )-T 5 -T 6 , heterocyclo, or heteroaryl) where
  • T 4 is a bond, -S0 2 -, or -C(O)-;
  • T 5 is -S0 2 -, or -alkylene-O-;
  • T 6 is alkyl, or cycloalkyl
  • T 7 and T 8 are independently H or alkyl
  • T 9 and T 10 are hydrogen
  • heterocyclo (heterocyclo)alkyl where the heterocyclo group is optionally independently substituted with one or more groups T la , T 2a , T 3a selected from optionally substituted alkyl, halo, cyano, nitro, (hydroxy)alkyl, -OH, -OT 6 , -ST 6 , -CO,H, -CO,T 6 , -S0 3 H, -SO t T 6 , -T 4 NT 7 T 8 , -T 4 -N(T 10 )-T 5 -T 6 , heterocyclo, or heteroaryl) where
  • T 4 is a bond, -S0 2 -, or -C(O)-;
  • T 5 is -S0 2 -, or -alkylene-O-;
  • T 6 is alkyl, or cycloalkyl;
  • T 7 and T 8 are independently H or alkyl
  • T 9 and T 10 are hydrogen
  • T 3a selected from -OH, -OT 6 , -CO t H, -CO t T 6 , -T 4 NT 7 T 8 or -T 4 -N(T 10 )-T 5 -T 6 ) where
  • T 4 is a bond
  • T 5 is -CO)-
  • T 6 is alkyl
  • T 7 and T 8 are independently H or alkyl
  • T 10 is hydrogen; (f) heterocyclo optionally independently substituted with one or more groups T la , T 2a , T 3a selected from optionally substituted alkyl (especially substituted with -T 4 NT 7 T 8 ), optionally substituted aryl (especially substituted with halogen or haloalkyl), cyano, -OH, -OT 6 , -CO t H, -CO t T 6 , oxo, hydroxy(alkyl), (alkoxy)alkyl, -T 4 -N(T 10 )-T 5 -T 6 , or -T 4 -NT 7 T 8 ) where
  • T 4 is a bond or -C(O)-;
  • T 5 is -C(O)-, -S0 2 -, or -alkylene-C(0)0-;
  • T 6 is alkyl, alkoxy, or heteroaryl;
  • T 7 and T 8 are independently H, alkyl, or cycloalkyl; or T 7 and T 8 together with the nitrogen atom to which they are attached combine to form a an optionally substituted heterocyclo ring;
  • Preferred compounds of the present invention include compounds of Formula
  • R 3 , R 4 , Y 1 , Y 2 , Y 3 and Z are as described above (including preferred groups); R 2 is
  • W is O or S, more preferably S;
  • X is heteroaryl (preferably, pyrimidinyl, imidazolyl, oxazolyl, or thiazolyl any of which may be further optionally substituted), cyano, C(0) t T 6 , or S(0) t NT 7 T 8 ; and X 4 is alkyl, haloalkyl, NHT 8 or OT 6 .
  • R 1 * is H or alkyl
  • R 2* is optionally substituted heteroaryl
  • R 3* is H or alkyl
  • R 4* is optionally substituted (aryl)alkyl
  • Y 1 , Y 2 and Y 3 are each H.
  • Preferred compounds within Formula III are those wherein: R 1* is H; R 2* is thiazolyl, oxazolyl, or isoxozolyl (preferably thiazolyl) any of which may be optionally substituted (preferably with one or more alkyl, or alkoxycarbonyl groups); R 3* is H; and R 4* is optionally substituted (pheny)alkyl, (preferably substituted with one or more group of the formula -S0 2 R 5 where R 5 is alkyl, amino, alkylamino or dialkylamino).
  • R ⁇ is H
  • W is O or S (preferably S), X 1 is alkoxy, and X is alkyl;
  • R 3* is H;
  • R is (pheny)alkyl substituted with one or more group of the formula -S0 2 R where R is amino or alkyl;
  • Y 1 , Y 2 and Y 3 are each H.
  • Preferred compounds within the scope of Formula IV include:
  • alk refers to straight or branched chain hydrocarbon groups having 1 to 12 carbon atoms, preferably 1 to 8 carbon atoms, such as methyl, ethyl, n- propyl, i-propyl, n-butyl, i-butyl, t-butyl, pentyl, hexyl, heptyl, octyl, etc.
  • Lower alkyl groups that is, alkyl groups of 1 to 6 carbon atoms, are generally most preferred.
  • substituted alkyl refers to alkyl groups substituted with one or more groups listed in the definition of T 1 , T 2 and T 3 , preferably selected from halo, cyano, O- R 7 , S-R 7 , NR 8 R 9 , nitro, cycloalkyl, substituted cycloalkyl, oxo, aryl, substituted aryl, heterocyclo, heteroaryl, C0 2 R 7 , S(0)R 7 , S0 2 R 7 , S0 3 R 7 , S0 2 NR 8 R 9 , C(0)NR 8 R 9 , C(0)alkyl, and C(0)H.
  • alkylene refers to a straight chain bridge of 1 to 4 carbon atoms connected by single bonds (e.g., -(CH2) ⁇ - wherein x is 1 to 5), which may be substituted with one or more groups listed in the definition of T 1 , T 2 and T 3 .
  • alkenyl refers to straight or branched chain hydrocarbon groups having 2 to 12 carbon atoms, preferably 2 to 4 carbon atoms, and at least one double carbon to carbon bond (either cis or trans), such as ethenyl.
  • substituted alkenyl refers to an alkenyl group as defined above substituted with one or more groups listed in the definition of T , T and T , preferably selected from halo, cyano, 0-R , S-R 7 , NR 8 R , nitro, cycloalkyl, substituted cycloalkyl, oxo, aryl, substituted aryl, heterocyclo, heteroaryl, C0 2 R 7 , S(0)R , S0 2 R 7 , S0 3 R 7 , S0 2 NR 8 R 9 , C(0)NR 8 R 9 , C(0)alkyl, and C(0)H.
  • alkynyl refers to straight or branched chain hydrocarbon group having 2 to 12 carbon atoms and one, two or three triple bonds, preferably 2 to 6 carbon atoms and one triple bond.
  • substituted alkynyl refers to an alkynyl group as defined above substituted with one or more groups listed in the definition of T 1 , T 2 and T 3 , preferably selected from halo, cyano, 0-R 7 , S-R 7 , NR 8 R 9 , nitro, cycloalkyl, substituted cycloalkyl, oxo, aryl, substituted aryl, heterocyclo, heteroaryl, C0 2 R 7 , S(0)R 7 , S0 2 R 7 , S0 3 R 7 , S0 2 NR 8 R 9 , C(0)NR 8 R 9 , C(0)alkyl, and C(0)H.
  • halo refers to chloro, bromo, fluoro, and iodo.
  • cycloalkyl refers to saturated and partially unsaturated (containing 1 or 2 double bonds) cyclic hydrocarbon groups containing 1 to 3 rings, including monocyclicalkyl, bicyclicalkyl and tricyclicalkyl, containing a total of 3 to 20 carbons forming the rings, preferably 3 to 7 carbons, forming the ring and which may be fused to 1 or 2 aromatic or heterocyclo rings, which include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclodecyl, cyclododecyl, cyclohexenyl,
  • substituted cycloalkyl refers to such cycloalkyl group as defined above substituted with one or more groups listed in the definition of T 1 , T 2 and T 3 , preferably selected from halogen, nitro, alkyl, substituted alkyl, alkenyl, cyano, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heterocyclo, heteroaryl, oxo, OR 7 , C0 2 R 7 , C(0)NR 8 R 9 , OC(0)R 7 , OC(0)OR 7 , OC(0)NR 8 R 9 , OCH 2 C0 2 R 7 , C(0)R 7 , NR 8 R 9 , NR 10 C(O)R 7 , NR 10 C(O)OR 7 , NR 10 C(O)C(O)OR 7 , NR 10 C(O)C(O)OR 7 , NR 10 C(O)C(O)NR 8 R 9 , NRioC(0)
  • aromatic homocyclic i.e., hydrocarbon
  • bi- or tricyclic ring-containing groups preferably having 6 to 12 members such as phenyl, naphthyl and biphenyl, as well as such rings fused to a cycloalkyl, cycloalkenyl, heterocyclo, or heteroaryl ring. Examples include:
  • substituted aryl refers to such aryl groups as defined above substituted with one or more groups listed in the definition of T 1 , T 2 and T 3 , preferably selected from halogen, nitro, alkyl, substituted alkyl, alkenyl, cyano, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heterocyclo, heteroaryl, OR 7 , C0 2 R 7 , C(0)NR 8 R 9 , OC(0)R 7 , OC(0)OR 7 , OC(0)NR 8 R 9 , OCH 2 C0 2 R 7 , C(0)R 7 , NR 8 R 9 , NR ⁇ 0 C(O)R 7 , NR ⁇ 0 C(O)OR 7 , NR ⁇ oC(0)C(0)OR 7 , NR 10 C(O)C(O)NR 8 R 9 , NR 10 C(O)C(O)alkyl, NR 10 C(NCN)OR 7
  • NR,oC( NC)(CR 12 R 13 )rR 7 , NR 10 CO(CR 12 R 13 )rNR 8 R 9 , NR 1 o(CR 12 R 13 )mOR 7 , NR 10 (CR 12 Ri 3 )rC0 2 R 7 , NR 1 o(CR 12 R 13 )mNR 8 R 9 , NR 10 (CR 12 R 13 )nS0 2 (CR 14 R 15 )qR 7 , CONR 10 (CR 12 R 13 )nSO 2 (CR 14 R ⁇ 5 )qR 7 , SO 2 NR 10 (CR ⁇ 2 R ⁇ 3 )nCO(CR ⁇ 4 R ⁇ 5 )qR 7 , and SO 2 NR 10 (CR ⁇ 2 R i3 )mOR 7 as well as pentafluorophenyl.
  • heterocycle refers to fully saturated or partially unsaturated cyclic groups (for example, 3 to 13 member monocyclic, 7 to 17 member bicyclic, or 10 to 20 member tricyclic ring systems, preferably containing a total of 3 to 10 ring atoms) which have at least one heteroatom in at least one carbon atom-containing ring.
  • Each ring of the heterocyclic group containing a heteroatom may have 1, 2, 3 or 4 heteroatoms selected from nitrogen atoms, oxygen atoms and/or sulfur atoms, where the nitrogen and sulfur heteroatoms may optionally be oxidized and the nitrogen heteroatoms may optionally be quaternized.
  • the heterocyclic group may be attached at any heteroatom or carbon atom of the ring or ring system.
  • the rings of multi-ring heterocycles may be either fused, bridged and/or joined through one or more spiro unions.
  • Exemplary heterocyclic groups include
  • substituted heterocycle or “substituted heterocyclo” and the like refer o such heterocylo groups as defined above substituted with one or more groups listed in the definition of T 1 , T 2 and T 3 , preferably selected from halogen, nitro, alkyl, substituted alkyl, alkenyl, cyano, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heterocyclo, heteroaryl,oxo, OR 7 , C0 2 R 7 , C(0)NR 8 R 9 , OC(0)R 7 , OC(0)OR 7 , OC(0)NR 8 R 9 , OCH 2 C0 2 R 7 , C(0)R 7 , NR 8 R 9 , NR 10 C(O)R 7 , NR 10 C(O)OR 7 , NR 10 C(O)C(O)OR 7 , NR, 0 C(O)C(O)NR 8 R 9 , NR, 0
  • heteroaryl refers to a 5- 6- or 7- membered aromatic rings containing from 1 to 4 nitrogen atoms and/or 1 or 2 oxygen or sulfur atoms provided that the ring contains at least 1 carbon atom and no more than 4 heteroatoms.
  • the heteroaryl ring is linked through an available carbon or nitrogen atom.
  • such rings fused to a cycloalkyl, aryl, cycloheteroalkyl, or another heteroaryl ring.
  • One, two, or three available carbon or nitrogen atoms in the heteroaryl ring can be optionally substituted with substituents listed in the description of T l 5 T 2 and T 3 .
  • substituted heteroaryl refers to such heteroaryl groups as defined above substituted on any available atom with one or more groups listed in the definition of T 1 , T 2 and T 3 , preferably selected from” refers to such heterocylo groups as defined above substituted with one or more groups listed in the definition of T 1 , T 2 and T 3 , preferably selected from halogen, nitro, alkyl, substituted alkyl, alkenyl, cyano, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heterocyclo, heteroaryl, OR 7 , C0 2 R 7 , C(0)NR 8 R 9 , OC(0)R 7 , OC(0)OR 7 , OC(0)NR 8 R 9 , OCH 2 C0 2 R 7 , C(0)R 7 , NR 8 R 9 , NR 10 C(O)R 7 , NR, 0 C(O)OR 7 , NR 10 C(O)C(O
  • R 7 , Rio, and Rn are independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, alkynyl, cycloalkyl, substituted cycloalkyl, C(0)alkyl, C(0)substituted alkyl, C(0)cycloalkyl, C(O) substituted cycloalkyl, C(0)aryl, C(0)substituted aryl, C(0)Oalkyl, C(0)Osubstituted alkyl, C(0)heterocyclo, C(0)heteroaryl, aryl, substituted aryl, heterocyclo and heteroaryl.
  • R 8 and R 9 are independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, alkenyl, alkynyl, C(0)alkyl, C(0)substituted alkyl, C(0)cycloalkyl, C(0)substituted cycloalkyl, C(0)aryl, C(0)substituted aryl, C(0)Oalkyl, C(0)Osustituted alkyl, C(0)heterocyclo, C(0)heteroaryl, S(0) 2 alkyl, S(0) 2 substituted alkyl, S(0) 2 cycloalkyl, S(0) 2 substituted cycloalkyl, S(0) 2 aryl, S(0) 2 substituted cycloalkyl, S(0) 2 aryl, S(0) 2 substituted aryl, S(0) 2 heterocyclo, S(0) 2 heteroaryl, aryl, substituted aryl, heterocyclo, and heteroaryl or R 8 and R
  • R ⁇ 2 and R ]4 are independently selected from hydrogen and alkyl or 1 to 4 carbons.
  • Ri 3 and R ⁇ 5 are independently selected from hydrogen, alkyl of 1 to 4 carbons, and substituted alkyl or 1 to 4 carbons.
  • n is zero or an integer from 1 to 4.
  • m is an integer from 2 to 6.
  • p is an integer from 1 to 3.
  • q is zero or an integer from 1 to 3.
  • r is zero or an integer from 1 to 6.
  • T 1 , T 2 , and T 3 are each independently (1) hydrogen or T 6 , where T 6 is (i) alkyl, (hydroxy)alkyl, (alkoxy)alkyl, alkenyl, alkynyl, cycloalkyl, (cycloalkyl)alkyl, cycloalkenyl, (cycloalkenyl)alkyl, aryl, (aryl)alkyl, heterocyclo, (heterocylco)alkyl, heteroaryl, or (heteroaryl)alkyl; (ii) a group (i) which is itself substituted by one or more of the same or different groups (i); or (iii) a group (i) or (ii) which is independently substituted by one or more (preferably 1 to 3) of the following groups (2) to (13) of the definition of T 1 , T 2 and T 3 ; (2) -OH or -OT 6 ,
  • T 7 and T 8 may together be alkylene or alkenylene, completing a 3- to 8- membered saturated or unsaturated ring together with the atoms to which they are attached, which ring is unsubstituted or substituted with one or more groups listed in the description of T 1 , T 2 and T 3 , or
  • T 7 or T 8 together with T 9 , may be alkylene or alkenylene completing a 3- to 8-membered saturated or unsaturated ring together with the nitrogen atoms to which they are attached, which ring is unsubstituted or substituted with one or more groups listed in the description of T 1 , T 2 and T 3 , or
  • T and T are each independently
  • T cell-mediated diseases refers to any disorder or disease state in which modulation of the activity of T cells is implicated in a process which results in either a pathophysiological state or a process where the normal function of T cells is intended to be suppressed for therapeutic benefit.
  • T cell mediated disorders include transplant rejection, graph verses host disease, and autoimmune disorders, such as rheumatoid arthritis, multiple sclerosis, juvenile diabetes, asthma, and inflammatory bowel disease, T-cell mediated hypersensitivity diseases, ischemic or reperfusion injury, and T-cell proliferative disorders.
  • PDE7 inhibitors in accordance with the present invention are employed, typically in the form of a pharmaceutical composition including a pharmaceutically acceptable carrier for the treatment of T-cell mediated disease.
  • the compounds employed for this purpose are typically administered in an amount from about 0.01 to 100 mg kg/day.
  • compositions comprising at least one PDE7 inhibitor may be formulated, for example, by employing conventional solid or liquid vehicles or diluents, as well as pharmaceutical additives of a type appropriate to the mode of desired administration (for example, excipients, binders, preservatives, stabilizers, flavors, etc.) according to techniques such as those well known in the art of pharmaceutical formulation.
  • the PDE7 inhibitors may be administered by any suitable means, for example, orally, such as in the form of tablets, capsules, granules or powders; sublingually; buccally; parenterally, such as by subcutaneous, intravenous, intramuscular, or intrasternal injection or infusion techniques (e.g., as sterile injectable aqueous or non-aqueous solutions or suspensions); nasally such as by inhalation spray; topically, such as in the form of a cream or ointment; or rectally such as in the form of suppositories; in dosage unit formulations containing non-toxic, pharmaceutically acceptable vehicles or diluents.
  • suitable means for example, orally, such as in the form of tablets, capsules, granules or powders; sublingually; buccally; parenterally, such as by subcutaneous, intravenous, intramuscular, or intrasternal injection or infusion techniques (e.g., as sterile injectable aqueous or non
  • the present compounds may, for example, be administered in a form suitable for immediate release or extended release. Immediate release or extended release may be achieved by the use of suitable pharmaceutical compositions comprising the present compounds, or, particularly in the case of extended release, by the use of devices such as subcutaneous implants or osmotic pumps.
  • the present compounds may also be administered in the form of liposomes.
  • compositions for oral administration include suspensions which may contain, for example, microcrystalline cellulose for imparting bulk, alginic acid or sodium alginate as a suspending agent, methylcellulose as a viscosity enhancer, and sweeteners or flavoring agents such as those known in the art; and immediate release tablets which may contain, for example, microcrystalline cellulose, dicalcium phosphate, starch, magnesium stearate and/or lactose and/or other excipients, binders, extenders, disintegrants, diluents and lubricants such as those known in the art.
  • the present compounds may also be delivered through the oral cavity by sublingual and/or buccal administration. Molded tablets, compressed tablets or freeze-dried tablets are exemplary forms which may be used.
  • compositions include those formulating the present compound(s) with fast dissolving diluents such as mannitol, lactose, sucrose and/or cyclodextrins. Also included in such formulations may be high molecular weight excipients such as celluloses (avicel) or polyethylene glycols (PEG). Such formulations may also include an excipient to aid mucosal adhesion such as hydroxy propyl cellulose (HPC), hydroxy propyl methyl cellulose (HPMC), sodium carboxy methyl cellulose (SCMC), maleic anhydride copolymer (e.g., Gantrez), and agents to control release such as polyacrylic copolymer (e.g., Carbopol 934). Lubricants, glidants, flavors, coloring agents and stabilizers may also be added for ease of fabrication and use.
  • fast dissolving diluents such as mannitol, lactose, sucrose and/or cyclodextrins
  • compositions for nasal aerosol or inhalation administration include solutions in saline which may contain, for example, benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, and or other solubilizing or dispersing agents such as those known in the art.
  • compositions for parenteral administration include injectable solutions or suspensions which may contain, for example, suitable non-toxic, parenterally acceptable diluents or solvents, such as mannitol, 1,3-butanediol, water, Ringer's solution, an isotonic sodium chloride solution, or other suitable dispersing or wetting and suspending agents, including synthetic mono- or diglycerides, and fatty acids, including oleic acid.
  • suitable non-toxic, parenterally acceptable diluents or solvents such as mannitol, 1,3-butanediol, water, Ringer's solution, an isotonic sodium chloride solution, or other suitable dispersing or wetting and suspending agents, including synthetic mono- or diglycerides, and fatty acids, including oleic acid.
  • compositions for rectal administration include suppositories which may contain, for example, a suitable non-irritating excipient, such as cocoa butter, synthetic glyceride esters or polyethylene glycols, which are solid at ordinary temperatures, but liquefy and/or dissolve in the rectal cavity to release the drug.
  • a suitable non-irritating excipient such as cocoa butter, synthetic glyceride esters or polyethylene glycols, which are solid at ordinary temperatures, but liquefy and/or dissolve in the rectal cavity to release the drug.
  • compositions for topical administration include a topical carrier such as Plastibase (mineral oil gelled with polyethylene).
  • a topical carrier such as Plastibase (mineral oil gelled with polyethylene).
  • the effective amount of a compound employed in the present invention may be determined by one of ordinary skill in the art, and includes exemplary dosage amounts for an adult human of from about 0.01 to 100 mg/kg of body weight of active compound per day, which may be administered in a single dose or in the form of individual divided doses, such as from 1 to 4 times per day.
  • the specific dose level and frequency of dosage for any particular subject may be varied and will depend upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the species, age, body weight, general health, sex and diet of the subject, the mode and time of administration, rate of excretion, drug combination, and severity of the particular condition.
  • Preferred subjects for treatment include animals, most preferably mammalian species such as humans, and domestic animals such as dogs, cats and the like, subject to inflammatory, immunological, or respiratory cell-associated disorders.
  • PDE7 inhibitors for use in the treatment of various T-cell mediated diseases are those covered by Formula I
  • Compounds of Formula I include salts, prodrugs and solvates.
  • salt(s) denotes acidic and/or basic salts formed with inorganic and/or organic acids and bases. Zwitterions (internal or inner salts) are included within the term “salt(s)” as used herein (and may be formed, for example, where the R substituents comprise an acid moiety such as a carboxyl group). Also included herein are quaternary ammonium salts such as alkylammonium salts. Pharmaceutically acceptable (i.e., non-toxic, physiologically acceptable) salts are preferred, although other salts are useful, for example, in isolation or purification steps which may be employed during preparation.
  • Salts of the compounds of the formula I may be formed, for example, by reacting a compound I with an amount of acid or base, such as an equivalent amount, in a medium such as one in which the salt precipitates or in an aqueous medium followed by lyophilization.
  • Exemplary acid addition salts include acetates (such as those formed with acetic acid or trihaloacetic acid, for example, trifluoroacetic acid), adipates, alginates, ascorbates, aspartates, benzoates, benzenesulfonates, bisulfates, borates, butyrates, citrates, camphorates, camphorsulfonates, cyclopentanepropionates, digluconates, dodecylsulfates, ethanesulfonates, fumarates, glucoheptanoates, glycerophosphates, hemisulfates, heptanoates, hexanoates, hydrochlorides, hydrobromides, hydroiodides, 2-hydroxyethanesulfonates, lactates, maleates, methanesulfonates, 2-naphthalenesulfonates, nicotinates, nitrates, oxal
  • 3-phenylpropionates phosphates, picrates, pivalates, propionates, salicylates, succinates, sulfates (such as those formed with sulfuric acid), sulfonates (such as those mentioned herein), tartrates, thiocyanates, toluenesulfonates, undecanoates, and the like.
  • Exemplary basic salts include ammonium salts, alkali metal salts such as sodium, lithium, and potassium salts, alkaline earth metal salts such as calcium and magnesium salts, salts with organic bases (for example, organic amines) such as benzathines, dicyclohexylamines, hydrabamines, N-methyl-D-glucamines, N-methyl-D- glucamides, t-butyl amines, and salts with amino acids such as arginine, lysine and the like.
  • the basic nitrogen-containing groups may be quatemized with agents such as lower alkyl halides (e.g.
  • dialkyl sulfates e.g. dimethyl, diethyl, dibutyl, and diamyl sulfates
  • long chain halides e.g. decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides
  • aralkyl halides e.g. benzyl and phenethyl bromides
  • Prodrugs and solvates of the compounds of the invention are also contemplated herein.
  • the term "prodrug”, as employed herein, denotes a compound which, upon administration to a subject, undergoes chemical conversion by metabolic or chemical processes to yield a compound of the Formula I, or a salt and/or solvate thereof.
  • Solvates of the compounds of Formula I are preferably hydrates.
  • All stereoisomers of the present compounds are contemplated within the scope of this invention.
  • Individual stereoisomers of the compounds of the invention may, for example, be substantially free of other isomers, or may be admixed, for example, as racemates or with all other, or other selected, stereoisomers.
  • the chiral centers of the present invention can have the S or R configuration as defined by the IUPAC 1974 Recommendations.
  • the compounds of Formula I are typically employed as part of a pharmaceutical composition including a pharmaceutically acceptable carrier for the treatment of respiratory and non-respiratory diseases.
  • the compounds employed for this purpose are typically administered in an amount of from about 0.01 to 100 mg/kg/day.
  • the compounds of Formula I are especially effective in inhibiting the PDE7 enzyme. Additionally a subset of compounds are also effective at inhibiting PDE4.
  • the pharmaceutical composition comprising at least one compound of Formula I may be formulated, for example, by employing conventional solid or liquid vehicles or diluents, as well as pharmaceutical additives of a type appropriate to the mode of desired administration (for example, excipients, binders, preservatives, stabilizers, flavors, etc.) according to techniques such as those well known in the art of pharmaceutical formulation.
  • the compounds of Formula I may be administered by any suitable means, for example, orally, such as in the form of tablets, capsules, granules or powders; sublingually; bucally; parenterally, such as by subcutaneous, intravenous, intramuscular, or intrasternal injection or infusion techniques (e.g., as sterile injectable aqueous or non- aqueous solutions or suspensions); nasally such as by inhalation spray; topically, such as in the form of a cream or ointment; or rectally such as in the form of suppositories; in dosage unit formulations containing non-toxic, pharmaceutically acceptable vehicles or diluents.
  • suitable means for example, orally, such as in the form of tablets, capsules, granules or powders; sublingually; bucally; parenterally, such as by subcutaneous, intravenous, intramuscular, or intrasternal injection or infusion techniques (e.g., as sterile injectable aqueous or non-
  • the present compounds may be based for immediate release or extended release by the use of suitable pharmaceutical compositions comprising the present compounds, or, particularly in the case of extended release, by the use of devices such as subcutaneous implants or osmotic pumps.
  • suitable pharmaceutical compositions comprising the present compounds, or, particularly in the case of extended release, by the use of devices such as subcutaneous implants or osmotic pumps.
  • the present compounds may also be administered liposomally.
  • compositions for oral administration include suspensions which may contain, for example, microcrystalline cellulose for imparting bulk, alginic acid or sodium alginate as a suspending agent, methylcellulose as a viscosity enhancer, and sweeteners or flavoring agents such as those known in the art; and immediate release tablets which may contain, for example, microcrystalline cellulose, dicalcium phosphate, starch, magnesium stearate and/or lactose and/or other excipients, binders, extenders, disintegrants, diluents and lubricants such as those known in the art.
  • the present compounds may also be delivered through the oral cavity by sublingual and/or buccal administration. Molded tablets, compressed tablets or freeze-dried tablets are exemplary forms which may be used.
  • compositions include those formulating the present compound(s) with fast dissolving diluents such as mannitol, lactose, sucrose and/or cyclodextrins. Also included in such formulations may be high molecular weight excipients such as celluloses (avicel) or polyethylene glycols (PEG). Such formulations may also include an excipient to aid mucosal adhesion such as hydroxy propyl cellulose (HPC), hydroxy propyl methyl cellulose (HPMC), sodium carboxy methyl cellulose (SCMC), maleic anhydride copolymer (e.g., Gantrez), and agents to control release such as polyacrylic copolymer (e.g., Carbopol 934). Lubricants, glidants, flavors, coloring agents and stabilizers may also be added for ease of fabrication and use.
  • fast dissolving diluents such as mannitol, lactose, sucrose and/or cyclodextrins
  • compositions for nasal aerosol or inhalation administration include solutions in saline which may contain, for example, benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, and/or other solubilizing or dispersing agents such as those known in the art.
  • compositions for parenteral administration include injectable solutions or suspensions which may contain, for example, suitable non-toxic, parenterally acceptable diluents or solvents, such as mannitol, 1,3-butanediol, water, Ringer's solution, an isotonic sodium chloride solution, or other suitable dispersing or wetting and suspending agents, including synthetic mono- or diglycerides, and fatty acids, including oleic acid.
  • suitable non-toxic, parenterally acceptable diluents or solvents such as mannitol, 1,3-butanediol, water, Ringer's solution, an isotonic sodium chloride solution, or other suitable dispersing or wetting and suspending agents, including synthetic mono- or diglycerides, and fatty acids, including oleic acid.
  • compositions for rectal administration include suppositories which may contain, for example, a suitable non-irritating excipient, such as cocoa butter, synthetic glyceride esters or polyethylene glycols, which are solid at ordinary temperatures, but liquefy and/or dissolve in the rectal cavity to release the drug.
  • a suitable non-irritating excipient such as cocoa butter, synthetic glyceride esters or polyethylene glycols, which are solid at ordinary temperatures, but liquefy and/or dissolve in the rectal cavity to release the drug.
  • exemplary compositions for topical administration include a topical carrier such as Plastibase (mineral oil gelled with polyethylene).
  • the effective amount of a compound of the present invention may be determined by one of ordinary skill in the art, and includes exemplary dosage amounts for an adult human from about 0.01 to 100 mg/kg of body weight of active compound per day, which may be administered in a single dose or in the form of individual divided doses, such as from 1 to 4 times per day. It will be understood that the specific dose level and frequency of dosage for any particular subject may be varied and will depend upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the species, age, body weight, general health, sex and diet of the subject, the mode and time of administration, rate of excretion, drug combination, and severity of the particular condition.
  • Preferred subjects for treatment include animals, most preferably mammalian species such as humans, and domestic animals such as dogs, cats and the like, subject to leukocyte activation or respiratory cell-associated disorders.
  • Utility Selective PDE7 inhibitors or dual PDE7-PDE4 inhibitors including compounds of formulas I are useful in the treatment (including prevention, partial alleviation or cure) of leukocyte activation-associated disorders, which include (but are not limited to) disorders such as: transplant rejection (such as organ transplant, acute transplant, xenotransplant or heterograft or homograft such as is employed in burn treatment); protection from ischemic or reperfusion injury such as ischemic or reperfusion injury incurred during organ transplantation, myocardial infarction, stroke or other causes; transplantation tolerance induction; arthritis (such as rheumatoid arthritis, psoriatic arthritis or osteoarthritis); multiple sclerosis; respiratory and pulmonary diseases including but not limited to asthma, exercise induced asthma, chronic obstructive pulmonary disease (COPD), emphysema, bronchitis, and acute respiratory distress syndrome (ARDS); inflammatory bowel disease, including ulcerative colitis and Crohn's disease; lupus (systemic
  • T-cell mediated hypersensitivity diseases including contact hypersensitivity, delayed-type hypersensitivity, and gluten- sensitive enteropathy (Celiac disease); psoriasis; contact dermatitis (including that due to poison ivy); Hashimoto's thyroiditis; Sjogren's syndrome; Autoimmune Hyperthyroidism, such as Graves' Disease; Addison's disease (autoimmune disease of the adrenal glands); Autoimmune polyglandular disease (also known as autoimmune polyglandular syndrome); autoimmune alopecia; pernicious anemia; vitiligo; autoimmune hypopituatarism; Guillain-Barre syndrome; other autoimmune diseases; glomerulonephritis; serum sickness; uticaria; allergic diseases such as respiratory allergies (e.g., asthma, hayfever, allergic rhinitis) or skin allergies; scleracierma; mycosis fungoides; acute inflammatory and respiratory responses (such as acute respiratory distress syndrome and ishchemia/reper
  • leukocyte activation-associated disorder or "leukocyte activation- mediated disorder” as used herein includes each of the above referenced diseases or disorders.
  • the compounds of the present invention are useful for treating the aforementioned exemplary disorders irrespective of their etiology.
  • Those present compounds which are dual PDE7/4 inhibitors may be more effective than either a selective PDE4 inhibitor or a selective PDE7 inhibitor in the above mentioned disease states, as a result of either additive or synergistic activity resulting from the combined inhibition of PDE7 and PDE4.
  • the present invention thus provides methods for the treatment of disorders as discussed above comprising the step of administering to a subject in need thereof of at least one selective PDE7 inhibitor or at least one dual PDE7-PDE4 inhibitor for the treatment of leukocyte activation-associated or leukocyte-activation mediated disease.
  • Other therapeutic agents such as those described below may be employed with the compounds of the present invention.
  • such other therapeutic agent(s) may be administered prior to, simultaneously with or following the administration of the compound(s) of the present invention.
  • the methods of treating diseases which would benefit from the inhibition of PDE7 or the inhibition of both PDE7-PDE4 by a dual agent may comprise administering compounds of Formula (I) alone or in combination with each other and/or other suitable therapeutic agents useful in treating such conditions such as: immunosuppressants such as, cyclosporins (e.g., cyclosporin A), anti-EL-l agents, such as Anakinra, the IL-1 receptor antagonist, CTLA4-Ig, antibodies such as anti-ICAM-3, anti-IL-2 receptor (Anti-Tac), anti-CD45RB, anti-CD2, anti-CD3, anti-CD4, anti-CD80, anti-CD86, monoclonal antibody OKT3, agents blocking the interaction between CD40 and CD 154, such as antibodies specific for CD40 and/or CD154 (i.e., CD40L), fusion proteins constructed from CD40 and CD 154 (CD40Ig and CD8-CD154), interferon beta, interferon gamma, methotrexate, FK
  • Patents and Applications incorporated herein by reference in their entirety: U.S Patent No. 6,235,740, U.S. Patent No. 6,239,133, U.S. Application Serial No. 60/065,042, filed 11/10/97 (Attorney Docket No. QA207*), U.S. Application Serial No. 09/173,413, filed 10/15/98 (Attorney Docket No. QA 207a), and U.S. Patent No. 5,990,109.
  • the human T cell antigen gp39 a member of the TNF gene family, is a ligand for the CD40 receptor: expression of a soluble form of gp39 with B cell co-stimulatory activity", EMBO J (England), 11(12), p 4313-4321 (Dec 1992); and Moreland, L.W.
  • PDE 4 inhibitors may also be employed in combination with PDE 4 inhibitors.
  • selective PDE4 inhibitors currently in development which can be used in combination with compounds of the present invention include Arofyline, Cilomilast, Roflumilast, C-11294A, CDC- 801, BAY- 19-8004, Cipamfylline, SCH351591, YM-976, PD- 189659, Mesiopram, Pumafentrine, CDC-998, IC-485, and KW-4490.
  • transplant such as organ transplant, acute transplant, xenotransplant or heterograft or homograft (such as is employed in burn treatment)
  • protection from ischemic or reperfusion injury such as ischemic or reperfusion injury incurred during organ transplantation, myocardial infarction, stroke or other causes
  • transplantation tolerance induction arthritis (such as rheumatoid arthritis, psoriatic arthritis or osteoarthritis); multiple sclerosis
  • respiratory and pulmonary diseases including but not limited to asthma, exercise induced asthma, chronic obstructive pulmonary disease (COPD), emphysema, bronchitis, and acute respiratory distress syndrome (ARDS); inflammatory bowel disease, including ulcerative colitis and Crohn's disease; lupus (systemic lupus erythematosis); graft
  • COPD chronic obstructive pulmonary disease
  • ARDS acute respiratory distress syndrome
  • T-cell mediated hypersensitivity diseases including contact hypersensitivity, delayed-type hypersensitivity, and gluten-sensitive enteropathy (Celiac disease); psoriasis; contact dermatitis (including that due to poison ivy); Hashimoto's thyroiditis; Sjogren's syndrome; Autoimmune Hyperthyroidism, such as Graves' Disease; Addison's disease (autoimmune disease of the adrenal glands); Autoimmune polyglandular disease (also known as autoimmune polyglandular syndrome); autoimmune alopecia; pernicious anemia; vitiligo; autoimmune hypopituatarism; Guillain-Barre syndrome; other autoimmune diseases; glomerulonephritis; serum sickness; uticaria; allergic diseases such as respiratory allergies (asthma, hayfever, allergic rhinitis) or skin allergies; scleracierma; mycosis fungoides; acute inflammatory and respiratory responses (such as acute respiratory distress syndrome and ishchemia/reperfusion
  • the combined activity of the present compounds towards T-cells and other PDE7-expressing cells may be of value in the treatment of any of the aforementioned disorders. Additionally those present compounds which are dual PDE4/7 inhibitors may be more effective than either a selective PDE4 inhibitor or a selective PDE7 inhibitor in the above mentioned disease states.
  • the compounds of the present invention are useful for the treatment of the aforementioned exemplary disorders irrespective of their etiology, for example, for the treatment of transplant rejection, rheumatoid arthritis, multiple sclerosis, chronic obstructive pulmonary disease, inflammatory bowel disease, lupus, graft v. host disease, T-cell mediated hypersensitivity disease, psoriasis, Hashimoto's thyroiditis, Guillain-Barre syndrome, cancer, contact dermatitis, allergic disease such as allergic rhinitis, asthma, ischemic or reperfusion injury, respiratory diseases such as asthma, COPD and bronchitis or atopic dermatitis whether or not associated with leukocyte activation.
  • Hut78 cells were grown in 10% FCS in Iscoves Modified Dulbecco's Medium (Gibco BRL-Life Technologies, Grand Island, NY) with antibiotics. Cells were centrifuged and resuspended in four volumes of [40 mM Tris (pH 7.5)/50 ⁇ M EDTA/200uM PMSF with a cocktail of Protease inhibitors (Boehringher Mannheim, Indianapolis, IN)] at 4C. Cells were homogenized using aVirtis homogenizer, and the lysate was centrifuged twice for 15 min at 15,000 x g. Glycerol was added to a final volume of 50% for storage at -20C.
  • SPA assay Inhibition of PDE activity in Hut78 cell lysate was determined using an SPA specific for cAMP (Amersham Pharmacia Biotech, Buckinghamshire, UK) according to the manufacturers instructions with minor modifications. Enzyme assays were performed at room temperature in the presence of 50mM Tris HCI, pH7.5, containing 8.3mM MgCl 2 , 1.7mM EGTA and 0.5mg/mL BSA.
  • Each assay was performed in a lOO ⁇ L reaction volume in 96 well microtitre plates containing the above buffer, 0.3ul of Hut78 cell lysate treated with 2 uM Zardaverine to inhibit PDE3 and PDE4, 0.05 uCi of [5',8- 3 H] Adenosine 3 ',5 '-cyclic phosphate as an ammonium salt for 20 min.
  • the reaction was terminated by the addition of 50 ⁇ l PDE SPA beads (lmg) water with lOmM cold cAMP (Sigma, St. Louis MO). The reaction mix was allowed to settle for 20 minutes before counting in a Top Count-NXT scintillation counter (Packard BioScience, Meriden, CT).
  • PDE1 For individual PDE enzymes other than PDE7, the assay was essentially unchanged except that 3 H-cyclic GMP was used as the substrate for PDE1, PDE5 and PDE6.
  • PDEs/activators and enzyme sources were used: PDE1, bovine (Sigma St Louis), calmodulin; PDE2, rat kidney, cGMP; PDE3, human platelet; PDE4, rat kidney; PDE5, human platelet, and PDE6, bovine retina.
  • PBMC Peripheral blood mononuclear cells
  • TNF secretion assay The ability of compounds to inhibit the production and secretion of TNF ⁇ from leukocytes was performed using either PBMC (obtained as described above) or the THP- 1 cell line as a source of monocytes. Compounds were diluted in RPMI 1640 supplemented with 10% FBS and DMSO at a final concentration of 0.2%. Cells (2xl0 5 /well in U-bottom 96 well plates) were pre-incubated with compounds for 30 min at 37 C prior to addition of lipopolysaccharide (LPS) at a final concentration of 6.25 ng/ml in a total volume of 200 ⁇ L. After 4h at 37C, 50 ⁇ L of supernatant was carefully aspirated for detection of soluble TNF ⁇ . Soluble TNF ⁇ was detected by ELISA developed by R&D Systems (Minneapolis, MN) according to the manufacturers instructions.
  • LPS lipopolysaccharide
  • Examples A2 to A13 were prepared in a similar manner to that used for Example Al utilizing the appropriate amines in step Al.l.
  • A14 was prepared in an manner analogous to example Al with the exception that in step Al.l 4-aminosulfonylbenzylamine hydrochloride was substituted for 4- methylsulfonylbenzylamine hydrochloride, and in step A1.2 ethyl 2-amino-4- trifluoromethyl-5-thiazole carboxylate was substituted for ethyl 2-amino-4-methyl-5- thiazole carboxylate.
  • LCMS Ret.
  • Example A 15 2-[[4-rr[4-(AminosuIfonyl)phenyllmethyl1amino1-2-quinazolinyllamino1-4-methyl-5- thiazolecarboxylic acid, ethyl ester
  • the vial was purged with N 2 , sealed and heated in a 105 °C oil bath for 2.25 h.
  • the reaction mixture was cooled to rt, filtered and concentrated in vacuo.
  • the residue was treated with methanol (ca. 1 mL) and the precipitated solid was collected by filtration, washed with methanol and dried to afford 41 mg (37%) of A15 as a tan solid.
  • A16 was prepared in an manner analogous to example A15 with the exception that in step A 15.1, 4-methylsulfonylbenzylamine hydrochloride was substituted for 4- aminosulfonylbenzylamine hydrochloride.
  • A17.1 A17.1 was prepared as described in the literature (Curd, et. al. J. Chem. Soc; 1948, 1759-1766. A17 was prepared in an manner analogous to example A15 starting with quinazoline A17.1 .
  • a 17 was isolated as a tan solid; LC/MS: 529.33 [M+H] + ; HPLC: >95 % at 1.34 min (Xterra 5 ⁇ m C18 S5 column 4.6 x 30 mm, 10-90 % aqueous methanol over 2 min containing 0.2% phosphoric acid, 5 mL/min, monitoring at 254 nm).
  • A18 A18 was prepared in an manner analogous to example Al with the exception that in step Al.l 4-aminosulfonylbenzylamine hydrochloride was substituted for 4- methylsulfonylbenzylamine hydrochloride, and in step A1.2 ethyl 4-aminobenzoate was substituted for ethyl 2-amino-4-methyl-5-thiazole carboxylate.
  • the product was purified by preparatory reverse phase HPLC to yield A18 in 21% yield.
  • A19 was prepared in an manner analogous to example Al with the exception that in step Al.l 4-aminosulfonylbenzylamine hydrochloride was substituted for 4- methylsulfonylbenzylamine hydrochloride, and in step A 1.2, 6-aminoquinoline was substituted for ethyl 2-amino-4-methyl-5-thiazole carboxylate.
  • Bl.l Bl.l was prepared from commercially available 2-aminonicotinic acid following the procedure reported in the literature (Robins, et. al. J. Am. Chem. Soc. 1955, 77, 2256- 2260.)
  • B1.2 A mixture of Bl.l (50 mg, 0.250 mmol, 1 eq), 4-aminosulfonylbenzylamine hydrochloride (58.5 mg, 0.262 mmol, 1.05 eq) and diisopropylethylamine (0.13 mL, 0.75 mmol, 3 eq) in absolute ethanol (1 mL) was stirred at rt for 24 h. The reaction mixture was then cooled in an ice/water bath and the solid was collected by filtration, washed with water and cold ethanol and dried to provide 77 mg (88%) of B1.2 as an off-white solid.
  • B2 was prepared in an manner analogous to example Bl with the exception that in step B1.2, 4-methylsulfonylbenzylamine hydrochloride was substituted for 4- aminosulfonylbenzylamine hydrochloride.
  • B2 was isolated as a yellow solid; LC/MS: 499.33 [M+H] + ; HPLC: >85 % at 1.19 min (Xterra 5 ⁇ m C18 S5 column 4.6 x 30 mm, 10-90 % aqueous methanol over 2 min containing 0.2% phosphoric acid, 5 mL/min, monitoring at 254 nm).

Abstract

Quinazoline and pyrido[2,3-d]pyrimidine phosphodiesterase 7 (PDE 7) inhibitors of the following formula [insert chemical structure here] wherein R1, R2, L, Y1, Y2, Y3 and Z are as described herein, are provided which are useful in treating T-cell mediated diseases.

Description

Ouinazoline and Pyridor2,3-dlpyrimidine Inhibitors of Phosphodiesterase (PDE) 7
Field of the Invention
The present invention relates to quinazoline and pyrido[2,3-d]pyrimidine inhibitors of phosphodiesterase 7 (PDE 7) (including both selective inhibitors of PDE 7, and dual inhibitors of PDE 7 and phosphodiesterase 4), pharmaceutical compositions containing these inhibitors, and the use of these inhibitors in the treatment of leukocyte activation-associated or leukocyte-activation mediated disease and inflammatory diseases either alone or in combination with other therapeutic agents.
Background of the Invention
Phosphodiesterases (PDEs) hydrolyze the second messenger molecules cAMP and cGMP to affect cellular signaling. At least 11 families of PDEs exist, some of which (PDE3 ,4,7,8) are specific for cAMP, and others (PDE5,6,9) for cGMP. Further family members (PDE 1,2, 10,11) have dual specificity. A recent publication demonstrated a role for PDE7 in the activation and/or proliferation of T cells(Zi, Yee and Beavo, Science 283:848-851, 1999). Resting T lymphocytes express mainly PDE3 and PDE4. However, upon activation, T cells dramatically upregulate PDE7 and appear to rely on this isozyme for regulation of cAMP levels. Removal of the ability to upregulate the production of PDE7 protein by anti-sense oligonucleotides inhibited the proliferation and IL-2 production along with the maintenance of high concentrations of intracellular cAMP in CD3xCD28 stimulated T cells. A PDE7 inhibitor is defined herein as a compound for which the IC50 of the compound in a PDE7 inhibition assay is less than 20 micromolar (preferably less than 10 micromolar, more preferably less than 5 micromolar, most preferably less than 1 micromolar). The PDE7 IC50 of a selective PDE7 inhibitor should be less than one-tenth the IC50 of said compound in all of the following PDE assays: PDE1, PDE3 and PDE4 (more preferably the PDE7 IC5o of a selective PDE7 inhibitor should be less than one- twentieth the IC50 of said compound in the following PDE assays: PDE1 and PDE3, most preferably the PDE7 IC50 of a selective PDE7 inhibitor should be less than one-hundreth the IC50 of said compound in a PDE3 assay).
Several isoforms of PDE 1 have been identified and are distributed in heart, lung, and kidney tissue, as well as in circulating blood cells and smooth muscle cells. PDE1 inhibitors have demonstrated potent vasodilator activity. Such activity would represent an undesirable side effect in a therapeutic agent with the utilities listed in this patent for a PDE7 inhibitor. The PDE3 family of enzymes are distributed in several tissues including the heart liver, and platelets. PDE3 inhibitors have demonstrated potent cardiac iotropic activity. Such activity would represent an undesirable side effect in a therapeutic agent with the utilities listed in this patent for a PDE7 inhibitor. Several isoforms of PDE4 exist, and these are expressed in a wide variety of tissues including heart, kidney, brain, the gastrointestinal track and circulating blood cells. PDE4 inhibitors have demonstrated clinical utility for COPD, and have also been suggested to have utility for rheumatoid arthritis, and multiple sclerosis, and to possess anti- inflammatory activity. The utility of PDE4 inhibitors has been limited to some extent by their propensity to cause emesis. As such there are circumstances where it would be desirable to develop PDE7 inhibitors, which have a degree of selectivity against PDE. A selective inhibitor of PDE7 is expected to have broad application as an immunosuppressant in T cell-mediated diseases. PDE7 inhibitors will act at a different stage of the T cell signaling process compared to current immunosuppressants by inhibiting a very early stage of the T cell activation cascade. A selective inhibitor of PDE7 is also expected to have a decreased potential for clinically significant side effects compared to current immunosuppressants, therefore the primary disease indications are solid organ transplantation (SOT) and rheumatoid arthritis. Additional indications may include EBD, psoriasis, asthma and lupus.
A dual PDE7-PDE4 inhibitor (PDE4/7 or PDE7/4) is defined herein as any compound which has an IC50 in both a PDE7 and a PDE4 inhibition assay of less than 20 micromolar (preferably less than 10 micromolar, and more preferably less than 5 micromolar and most preferably less than 1 micromolar), and an IC50 in a PDE3 inhibition assay which is at least 10 times higher than the IC50 of the compound in the PDE7 assay (more preferably at least 20 times higher than the IC50 of the compound in the PDE7 assay, and most preferably at least 100 times higher than the IC50 of the compound in the PDE7 assay). A dual PDE4/7 inhibitor should have a ratio of inhibition or PDE7 IC50 divided by PDE4 IC50 of between one-tenth and 100. Inhibitors that exhibit such a ratio of inhibition include those that inhibit PDE3, PDE4 and PDE7 as described above, and further inhibit PDE1 at an IC50 at least 10 times higher than the
IC50 of the compound in a PDE7 assay (more preferably at least 20 times higher than the
IC50 of the compound in the PDE7 assay, and most preferably at least 100 times higher than the IC50 of the compound in the PDE7 assay). Preferred dual PDE7-PDE4 inhibitors further include those compounds that inhibit PDE3, PDE4 and PDE7 as described above, and further suppress both T cell proliferation, and TNF-alpha secretion from either THP-1 monocytes or human peripheral blood mononuclear cells at a level of less than 20 micromolar.
"Leukocyte activation" is defined herein as any or all of leukocyte (T cell, monocyte macrophage, neutrophil etc.) cell proliferation, cytokine production, adhesion protein expression, and production of inflammatory mediators. This is mediated in part by the action of PDE4 and/or PDE7 depending on the particular leukocyte under consideration.
Examples of leukocyte activation associated or leukocyte activation mediated disorders include transplant rejection, graph verses host disease, and autoimmune disorders, such as rheumatoid arthritis, multiple sclerosis, juvenile diabetes, COPD, asthma, and inflammatory bowel disease, T-cell mediated hypersensitivity diseases, ischemic or reperfusion injury, and T-cell proliferative disorders.
Dual PDE4/7 inhibitors would be expected to block the T cell component of a disease as well as possess anti-inflammatory activity. Thus a dual PDE4/7 inhibitor which is not significantly limited by emesis, may be more effective than either a selective
PDE4 inhibitor or a selective PDE7 inhibitor in a variety of disease states such as rheumatoid arthritis, asthma, COPD and multiple sclerosis.
Development of either selective PDE7 inhibitors, or dual PDE7-PDE4 inhibitors will yield novel classes of therapeutics and have a novel mechanism of action by maintaining high levels of intracellular cAMP. These inhibitors would target a major unmet medical need in an area where current therapies possess significant toxicity. Two PDE7 genes have been identified. PDE7A (EC 3.1.4.17) has two isoforms generated by alternate splicing; PDE7A1 restricted mainly to T cells and the brain, and PDE7A2 for which mRNA is expressed in a number of cell types including muscle cells. The isoforms have different sequence at the amino termini, and it is thought that this portion of each molecule is likely to be important for cellular localization of the enzyme. However, the catalytic domain of each PDE7 A enzyme is identical (Han,P., Zhu,X. and Michaeli,T. Alternative splicing of the high affinity cAMP- specific phosphodiesterase (PDE7A) mRNA in human skeletal muscle and heart. J. Biol. Chem. 272 (26), 16152-16157 (1997)). Although abundant PDE7A2 mRNA has been identified, the presence of active enzyme in tissues is controversial, as no convincing data shows PDE7A2 protein in situ in the adult. PDE7B (EC 3.1.4.17), a second PDE7 gene family member, has approximately 70% homology to PDE7A in the enzymatic core (Sasaki.T., Kotera ., Yuasa,K. and Omori,K. Identification of human PDE7B, a cAMP- specific phosphodiesterase Biochem. Biophys. Res. Commun. 271 (3), 575-583 (2000)) . Two patents from Cold Spring Harbor Labs (US 5527896 and US 5977305) cover the methods of preparation and use of recombinant PDE7A protein. A recent publication describes moderately active PDE7 inhibitors (J. Med Chem. Vol. 43, 683 (2000)). WO 00/68230 discloses certain 1,9 dihydropurin-6-ones derivatives as PDE7 inhibitors.
Summary of the Invention
The present invention provides quinazoline and pyrido[2,3-d]pyrimidine compounds of the following formula (I), their enantiomers, diastereomers, tautomers and pharmaceutically acceptable salts, prodrugs and solvates thereof, for use as PDE7 inhibitors and dual PDE4/7 inhibitors:
Figure imgf000005_0001
wherein R1 is H or alkyl;
R2 is
(a) heteroaryl, or heterocyclo, either of which may be optionally substituted with one to three groups T1, T2, T3;
(b) aryl substituted with one to three groups T1, T2, T3 provided that at least one of T1, T2, T3 is other than H; or
(c) aryl fused to a heteroaryl or heterocyclo ring wherein the combined ring system may be optionally substituted with one to three groups T1, T2, T3;
L is (a) -OR4, -C(0)R4, -C(0)OR4, -SR4, -NR3R4, -C(0)NR3R4, -NR3S02R4b halogen, nitro, haloalkyl; or (b) alkyl, aryl, heteroaryl, heterocyclo, or cycloalkyl any of which may be optionally substituted with one to three groups Tla, T2a T3a; Y1 , Y2 and Y3 are independently (a) hydrogen, halo, -OR4a, or
(b) alkyl, alkenyl, or alkynyl any of which may be optionally substituted with one to three groups Tlb, T2b or T3b; R3 and R4 are independently H, alkyl, alkenyl, aryl, (aryl)alkyl, heteroaryl,
(heteroaryl)alkyl, cycloalkyl, (cycloalkyl)alkyl, heterocylo or (heterocyclo)alkyl any of which may be optionally substituted with one to three groups Tla, T2a or
T3a. or R3 and R4 together with the nitrogen atom to which they are attached may combine to form a 4 to 8 membered heterocyclo ring optionally substituted with one to three groups Tla, T2a or T3a; R4a is hydrogen, alkyl, alkenyl, aryl, heteroaryl, (aryl)alkyl, (heteroaryl)alkyl, heterocylo , (heterocyclo)alkyl, cycloalkyl or (cycloalkyl)alkyl any of which may be optionally substituted with one to three groups Tlb, T2b or T3b;
R4b is alkyl, alkenyl, aryl, (aryl)alkyl, heteroaryl, (heteroaryl)alkyl, cycloalkyl, (cycloalkyl) lkyl, heterocylo or (heterocyclo)alkyl any of which may be optionally substituted with one to three groups Tla, T2a or T3a; Z is N or CH;
T b, τ2"2b ; and T3"3b are are each independently
( 1 ) hydrogen or T6, where T6 is
(i) alkyl, (hydroxy)alkyl, (alkoxy)alkyl, alkenyl, alkynyl, cycloalkyl, (cycloalkyl)alkyl, cycloalkenyl,
(cycloalkenyl)alkyl, aryl, (aryl)alkyl, heterocyclo, (heterocylco)alkyl, heteroaryl, or (heteroaryl)alkyl; (ii) (ii) a group (i) which is itself substituted by one or more of the same or different groups (i); or (iii) (iii) a group (i) or (ii) which is independently substituted by one or more (preferably 1 to 3) of the following groups (2) to (13) of the definition of Tl lb, T2-2b and T3"3b,
(2) -OH or -OT6,
(3) -SH or -ST6, (4) -C(0)tH, -C(0)tT6, or -0-C(0)T6, where t is 1 or 2;
(5) -S03H, -S(0)tT6, or S(0)tN(T9)T6,
(6) halo,
(7) cyano,
(8) nitro, (9) -T4-NT7T8,
(10) -T4-N(T9)-T5-NT7T8,
(11) -T4-N(T10)-T5-T6,
(12) -T4-N(T10)-T5-H,
(13) oxo, T4 and T5 are each independently
(1) a single bond,
(2) -Tu-S(0)t-T12-,
(3) -T"-C(0)-T12-,
(4) _τu-C(S)-T12-, (5) -Tu-0-T12-,
(6) -Tn-S-T12-, (7) -Tn-0-C(0)-T12-,
(8) -Tn-C(0)-0-T12-,
(9) -Tπ-C(=NT9a)-T12-, or
(10) -Tu-C(0)-C(0)-T12- T7, T8, T9, T9a and T10
(1) are each independently hydrogen or a group provided in the definition of T , or
(2) T7 and T8 may together be alkylene or alkenylene, completing a 3- to 8- membered saturated or unsaturated ring together with the atoms to which they are attached, which ring is unsubstituted or substituted with one or more groups listed in the description of Tl lb, T2 2b and T3 3b, or
(3) T7 or T8, together with T9, may be alkylene or alkenylene completing a 3- to 8-membered saturated or unsaturated ring together with the nitrogen atoms to which they are attached, which ring is unsubstituted or substituted with one or more groups listed in the description of T " , T " and T " , or
(4) T7 and T8 or T9 and T10 together with the nitrogen atom to which they are attached may combine to form a group -N=CT13T14 where T13 and T14 are each independently H or a group provided in the definition of T6; and
T1 ' and T12 are each independently (1) a single bond,
(2) alkylene,
(3) alkenylene, or
(4) alkynylene.
Preferred compounds of Formula I include those wherein: L is
(a) halogen, alkoxy, haloalkyl, -NR3R4, -C(0)OR4, -C(0)NR3R4;
(b) aryl or heteroaryl either of which may be optionally substituted with one or more Tla, T2a, T3a (especially cyano, optionally substituted alkyl, (hydroxy)alkyl, -OH, -OT6, -ST6, -SOtT6, -COtH, -COtT6, -T4NT7T8, or -T4N(T10)-T5-T6); (c) optionally substituted alkyl (especially substituted with one or more -OH, -CO,H, -COtT6, -f-NTV, -T4-N(T10)-T5-H, or ; -T4-N(T10)-T5-T6); Y1, Y2 and Y3 are independently (a) H, -OR4a or (b) alkyl or alkenyl either of which may be optionally substituted (especially with one or more -OH, -OT6, -COtH, or -COtT6); R1 is H or alkyl; R2 is
(a) heteroaryl (more preferably thiazolyl or oxazolyl) optionally substituted with one to three groups T1, T2, T3, preferably including H, alkyl, haloalkyl, halo, heteroaryl, cyano, C(0)tT6, OT6, -T4NT7T8;
(b) aryl substituted with one to three groups T1, T2, T3 (preferably including heteroaryl (preferably, imidazolyl, oxazolyl, or thiazolyl any of which may be further optionally substituted), cyano, C(0)tT6, S(0)tN(T9)T6, halo alkyl, and haloalkyl); or
(c) aryl fused to a heterocyclo ring (e.g., 2,3-dihydro-lH-indole bound through the aryl ring, quinolyl bound through the aryl ring (especially quinol-6-yl), quinazolinyl bound through the aryl ring (especially quinazolin-7-yl), cinnolinyl bound through the aryl ring (especially cinnolin-6-yl), isoqinolinyl bound through the aryl ring (especially isoquinol-6-yl), and phthalazinyl bound through the aryl ring (especially phthalazin-6-yl)) wherein the combined ring system may be optionally substituted with one to three groups T1, T2, T3 (especially halo, OH, OT6, alkyl, -COtH, -COtT6, or -C(0)NT7T8); R3 is H or optionally substituted alkyl (especially substituted with one or more -OH, or -
OT6);
R4 is
(a) hydrogen;
(b) (aryl)alkyl where the aryl group is optionally independently substituted with one or more groups TIa, T2a, T3a (especially optionally substituted alkyl, halo, cyano, nitro, (hydroxy)alkyl, -OH, -OT6, -ST6, -COtH, -COtT6, -S03H, -SOtT6, -SOtN(T9)(T6), -T4NT7T8, -T4-N(T10)-T5-T6, heterocyclo, or heteroaryl);
(c) (heteroaryl)alky where the heteroaryl group is optionally independently substituted with one or more groups Tla, T2a, T3a (especially optionally substituted alkyl, halo, cyano, nitro, (hydroxy)alkyl, -OH, -OT6, -ST6,
-CO,H, -COtT6, -SO3H, -SO,T6, -SOtN(T9)(T6), -T4NT7T8, -T4-N(T10)-T5-T6, heterocyclo, or heteroaryl);
(d) (heterocyclo)alkyl where the heterocyclo group is optionally independently substituted with one or more groups Tla, T2a, T3a (especially optionally substituted alkyl, halo, cyano, nitro, oxo, (hydroxy)alkyl, -OH, -OT6,
-ST6, -CO,H, -COtT6, -S03H, -SO,T6, -SOtN(T9)(T6), -T4NT7T8, -T4-N(T10)-T5-T6, heterocyclo, or heteroaryl);
(e) alkyl optionally independently substituted with one or more groups Tl , T2a,
T3a (especially -OH, -OT6, -COtH, -CO,T6, -T4NT7T8 or -T4-N(T10)-T5-T6); (f) heterocyclo optionally independently substituted with one or more groups Tla,
T2a, T3a (especially optionally substituted alkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted aralkyl, optionaly subsituted heterocyclo, cyano, -OH, -OT6, -COtH, -COtT6, oxo, hydroxy(alkyl), (alkoxy)alkyl, -T4-N(T10)-T5-T6, or -T4-NT7T8); or R and R4 together with the nitrogen atom to which they are attached combine to form a 4 to 8-membered heterocyclo ring (especially pyrrolidinyl, piperadinyl, piperazinyl, morpholinyl, diazapanyl or l,4-dioxa-8-azaspiro[4.5]decan-8-yl) optionally substituted with one to three groups Tla, T2a, T3a (especially optionally substituted alkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted aralkyl, optionaly subsituted heterocyclo, cyano, -OH,
-OT6, -CO.H, -CO,T6, oxo, hydroxy(alkyl), (alkoxy)alkyl, -T4-N(T10)-T5-T6, or - T4-NT7T8);
More preferred compounds of the present invention include compounds wherein:
L is (a) halogen, alkoxy, haloalkyl, -NR3R4, -C(0)OR4, -C(0)NR3R4 (especially
-NR3R4);
(b) aryl or heteroaryl either of which may be optionally substituted with one or more Tla, T2a, T3a selected from cyano, optionally substituted alkyl, (hydroxy)alkyl, -OH, -OT6, -ST6, -SOtT6, -COtH, -COtT6, -T4NT7T8, or
-T4N(T10)-T5-T6, where
T4 is a bond or -C(0)-; T5 is -C(0)-, or -C(0)0-; T6 is alkyl or haloalkyl;
T7 and T8 are independently H; alkyl optiontionally substituted with cycloalkyl, heteroaryl, hydroxy or -NT7T8; cycloalkyl; or aryl optionally substituted with halogen; or T7 and T8 together with the nitrogen atom to which they are attached combine to form a heterocyclo ring optionally substituted with (hydroxy)alkyl, COtH or COtT6 T10 is hydrogen;
(c) alkyl optionally substituted with one or more -OH, -COtH, -COtT6,
-T4-NT7T8, -T4-N(T10)-T5-H, or ; -T4-N(T10)-T5-T6 where
T4 is -C(O)-; T5 is -alkylene-O-;
T6 is alkyl;
7 R
T and T are independently H, alkyl, cycloalkyl, aryl, (aryl)alkyl (optionally substituted as described in the definition of R4), or heterocyclo (optionally substituted as described in the definition of R3 and R4 combining to form a heterocyclo ring); and T,0 is H; Y1, Y2 and Y3 are independently H or -OR4a; R1 is H or alkyl; R2 is (a) heteroaryl (more preferably thiazolyl or oxazolyl) optionally substituted with one to three groups T1, T2, T3, preferably including H, alkyl, haloalkyl, halo, heteroaryl, cyano, C(0)tT6, OT6, -T4NT7T8;
(b) aryl substituted with one to three groups T1, T2, T3 (preferably including heteroaryl (preferably, imidazolyl, oxazolyl, or thiazolyl any of which may be further optionally substituted), cyano, C(0)tT6, S(0)tN(T9)T6, halo alkyl, and haloalkyl); or
(c) aryl fused to a heterocyclo ring (especially quinolinyl or quinazolinyl bound through the aryl ring) wherein the combined ring system may be optionally substituted with one to three groups T1, T2, T3 (especially halo, OH, OT6, alkyl, -CO,H, -CO.T6, or -C(0)NT7T8);
R3 is H or optionally substituted alkyl (especially substituted with one or more -OH, or -
OT6); R4 is
(a) hydrogen; (b) (aryl)alkyl where the aryl group is optionally independently substituted with one or more groups Tla, T23, T3a selected from optionally substituted alkyl, halo, cyano, nitro, (hydroxy)alkyl, -OH, -OT6, -ST6, -COtH, -COtT6, -S03H, -SOtT6, -SOtN(T9)(T6), -T4NT7T8, -T4-N(T10)-T5-T6, heterocyclo, or heteroaryl) where
T4 is a bond, -S02-, or -C(O)-; T5 is -S02-, or -alkylene-O-; T6 is alkyl, or cycloalkyl;
T7 and T8 are independently H or alkyl; and Ty and T 1'0u are hydrogen; (c) (heteroaryl)alky where the heteroaryl group is optionally independently substituted with one or more groups Tla, T2a, T3a selected from optionally substituted alkyl, halo, cyano, nitro, oxo, (hydroxy)alkyl, -OH, -OT , -ST6, -COtH, -COtT6, -S03H, -SOtT6, -SOtN(T9)(T6), -T4NT7T8, -T4-N(T10)-T5-T6, heterocyclo, or heteroaryl) where
T4 is a bond, -S02-, or -C(O)-;
T5 is -S02-, or -alkylene-O-;
T6 is alkyl, or cycloalkyl; T7 and T8 are independently H or alkyl; and
T9 and T10 are hydrogen;
(d) (heterocyclo)alkyl where the heterocyclo group is optionally independently substituted with one or more groups Tla, T2a, T3a selected from optionally substituted alkyl, halo, cyano, nitro, (hydroxy)alkyl, -OH, -OT6, -ST6, -CO,H, -CO,T6, -S03H, -SOtT6, -T4NT7T8, -T4-N(T10)-T5-T6, heterocyclo, or heteroaryl) where
T4 is a bond, -S02-, or -C(O)-;
T5 is -S02-, or -alkylene-O-; T6 is alkyl, or cycloalkyl;
T7 and T8 are independently H or alkyl; and
T9 and T10 are hydrogen;
(e) alkyl optionally independently substituted with one or more groups Tla, T2a,
T3a selected from -OH, -OT6, -COtH, -COtT6, -T4NT7T8 or -T4-N(T10)-T5-T6) where
T4 is a bond;
T5 is -CO)-;
T6 is alkyl; T7 and T8 are independently H or alkyl; and
T10 is hydrogen; (f) heterocyclo optionally independently substituted with one or more groups Tla, T2a, T3a selected from optionally substituted alkyl (especially substituted with -T4NT7T8), optionally substituted aryl (especially substituted with halogen or haloalkyl), cyano, -OH, -OT6, -COtH, -COtT6, oxo, hydroxy(alkyl), (alkoxy)alkyl, -T4-N(T10)-T5-T6, or -T4-NT7T8) where
T4 is a bond or -C(O)-; T5 is -C(O)-, -S02-, or -alkylene-C(0)0-; T6 is alkyl, alkoxy, or heteroaryl; T7 and T8 are independently H, alkyl, or cycloalkyl; or T7 and T8 together with the nitrogen atom to which they are attached combine to form a an optionally substituted heterocyclo ring; or R3 and R4 together with the nitrogen atom to which they are attached combine to form a heterocylco ring selected from pyrrolidinyl, piperadinyl, piperazinyl, morpholinyl, diazapanyl or l,4-dioxa-8-azaspiro[4.5]decan-8-yl), any of which are optionally independently substituted with one to three groups Tla, T2a, T3a selected from optionally substituted alkyl (especially substituted with -T4NT7T8), optionally substituted aryl (especially substituted with halogen or haloalkyl), cyano, -OH, -OT6, -COtH, -CO,T6, oxo, hydroxy(alkyl), (alkoxy)alkyl,
-T4-N(TI0)-T5-T6, or -T4-NT7T8) where
T4 is a bond or -C(O)-; T5 is -C(O)-, -S02-, or -alkylene-C(0)0-; T6 is alkyl, alkoxy, or heteroaryl;
T7 and T8 are independently H, alkyl, or cycloalkyl; or T7 and T8 together with the nitrogen atom to which they are attached combine to form a an optionally substituted heterocyclo ring; Preferred compounds of the present invention include compounds of Formula
(II),
Figure imgf000015_0001
II where:
R3, R4, Y1, Y2, Y3 and Z are as described above (including preferred groups); R2 is
Figure imgf000015_0002
wherein: W is O or S, more preferably S;
X "1' i :s NHTδ or OT°; X and X are independently hydrogen, halo, OT , alkyl, or haloalkyl;
X is heteroaryl (preferably, pyrimidinyl, imidazolyl, oxazolyl, or thiazolyl any of which may be further optionally substituted), cyano, C(0)tT6, or S(0)tNT7T8; and X4 is alkyl, haloalkyl, NHT8 or OT6.
Compounds within the scope of the Formulas I and II include dual PDE7-PDE4 inhibitors of the following Formula III:
Figure imgf000016_0001
III wherein
R1 * is H or alkyl;
R2* is optionally substituted heteroaryl;
R3* is H or alkyl;
R4* is optionally substituted (aryl)alkyl; and
Y1, Y2 and Y3 are each H.
Preferred compounds within Formula III are those wherein: R1* is H; R2* is thiazolyl, oxazolyl, or isoxozolyl (preferably thiazolyl) any of which may be optionally substituted (preferably with one or more alkyl, or alkoxycarbonyl groups); R3* is H; and R4* is optionally substituted (pheny)alkyl, (preferably substituted with one or more group of the formula -S02R5 where R5 is alkyl, amino, alkylamino or dialkylamino).
More preferred compounds within Formula III are those wherein
R^ is H;
R2 is
Figure imgf000016_0002
where W is O or S (preferably S), X1 is alkoxy, and X is alkyl;
R3* is H; R is (pheny)alkyl substituted with one or more group of the formula -S02R where R is amino or alkyl; and
Y1, Y2 and Y3 are each H.
Preferred compounds within the scope of Formula IV include:
Figure imgf000017_0001
The following are definitions of the terms as used throughout this specification and claims. The initial definition provided for a group or term herein applies to that group or term throughout the present specification, individually or as part of another group, unless otherwise indicated.
The terms "alk" or "alkyl" refer to straight or branched chain hydrocarbon groups having 1 to 12 carbon atoms, preferably 1 to 8 carbon atoms, such as methyl, ethyl, n- propyl, i-propyl, n-butyl, i-butyl, t-butyl, pentyl, hexyl, heptyl, octyl, etc. Lower alkyl groups, that is, alkyl groups of 1 to 6 carbon atoms, are generally most preferred.
The term "substituted alkyl" refers to alkyl groups substituted with one or more groups listed in the definition of T1, T2 and T3, preferably selected from halo, cyano, O- R7, S-R7, NR8R9, nitro, cycloalkyl, substituted cycloalkyl, oxo, aryl, substituted aryl, heterocyclo, heteroaryl, C02R7, S(0)R7, S02R7, S03R7, S02NR8R9, C(0)NR8R9, C(0)alkyl, and C(0)H.
The term "alkylene" refers to a straight chain bridge of 1 to 4 carbon atoms connected by single bonds (e.g., -(CH2)χ- wherein x is 1 to 5), which may be substituted with one or more groups listed in the definition of T1, T2 and T3. The term "alkenyl" refers to straight or branched chain hydrocarbon groups having 2 to 12 carbon atoms, preferably 2 to 4 carbon atoms, and at least one double carbon to carbon bond (either cis or trans), such as ethenyl.
The term "substituted alkenyl" refers to an alkenyl group as defined above substituted with one or more groups listed in the definition of T , T and T , preferably selected from halo, cyano, 0-R , S-R7, NR8R , nitro, cycloalkyl, substituted cycloalkyl, oxo, aryl, substituted aryl, heterocyclo, heteroaryl, C02R7, S(0)R , S02R7, S03R7, S02NR8R9, C(0)NR8R9, C(0)alkyl, and C(0)H.
The term "alkynyl" refers to straight or branched chain hydrocarbon group having 2 to 12 carbon atoms and one, two or three triple bonds, preferably 2 to 6 carbon atoms and one triple bond.
The term "substituted alkynyl" refers to an alkynyl group as defined above substituted with one or more groups listed in the definition of T1, T2 and T3, preferably selected from halo, cyano, 0-R7, S-R7, NR8R9, nitro, cycloalkyl, substituted cycloalkyl, oxo, aryl, substituted aryl, heterocyclo, heteroaryl, C02R7, S(0)R7, S02R7, S03R7, S02NR8R9, C(0)NR8R9, C(0)alkyl, and C(0)H.
The term "halo" refers to chloro, bromo, fluoro, and iodo.
The term "cycloalkyl" refers to saturated and partially unsaturated (containing 1 or 2 double bonds) cyclic hydrocarbon groups containing 1 to 3 rings, including monocyclicalkyl, bicyclicalkyl and tricyclicalkyl, containing a total of 3 to 20 carbons forming the rings, preferably 3 to 7 carbons, forming the ring and which may be fused to 1 or 2 aromatic or heterocyclo rings, which include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclodecyl, cyclododecyl, cyclohexenyl,
Figure imgf000018_0001
Figure imgf000019_0001
The term "substituted cycloalkyl" refers to such cycloalkyl group as defined above substituted with one or more groups listed in the definition of T1, T2 and T3, preferably selected from halogen, nitro, alkyl, substituted alkyl, alkenyl, cyano, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heterocyclo, heteroaryl, oxo, OR7, C02R7, C(0)NR8R9, OC(0)R7, OC(0)OR7, OC(0)NR8R9, OCH2C02R7, C(0)R7, NR8R9, NR10C(O)R7, NR10C(O)OR7, NR10C(O)C(O)OR7, NR10C(O)C(O)NR8R9, NRioC(0)C(0)alkyl, NR10C(NCN)OR7, NR]0C(O)NR8R9, NRι0C(NCN)NR8R9, NRιoC(NRn)NR8R9, NR10SO2NR8R9, NR10SO2R7, SR7, S(0)R7, S02R7, S03R7, S02NR8R9, NHOR7, NRι0NR8R9, N(COR7)ORι0, N(CO2R7)ORι0, C(O)NR10(CR12R13)rR7, CO(CR12R13)pO(CR,4R15)qCO2R7, CO(CR12R13)rOR7, CO(CR12R13)pO(CR14R,5)qR7, CO(CR12R13)rNR8R9, OC(0)0(CR,2R13)mNR8R9, OC(0)N(CR12R13)rR7, 0(CR12R13)mNR8R9, NR10C(O)(CR12R13)rR7, NR10C(O)(CR12R13)rOR7, NR10C(=NC)(CRι2R13)rR7, NR10CO(CR,23)rNR8R9, NR10(CR,2R13)mOR7, NR10(CR123)rCO2R7, NR10(CR123)mNR8R9, NR1o(CR12R13)nS02(CR,4R,5)qR7, CONR1o(CRι2R13)nS02(CR14R15)qR7, SO2NR10(CRι2R13)nCO(CRι4R15)qR7, and SO2NR10(CRι2R13)mOR7.
The terms "ar" or "aryl" refer to aromatic homocyclic (i.e., hydrocarbon) mono-, bi- or tricyclic ring-containing groups preferably having 6 to 12 members such as phenyl, naphthyl and biphenyl, as well as such rings fused to a cycloalkyl, cycloalkenyl, heterocyclo, or heteroaryl ring. Examples include:
Figure imgf000020_0001
The term "substituted aryl" refers to such aryl groups as defined above substituted with one or more groups listed in the definition of T1, T2 and T3, preferably selected from halogen, nitro, alkyl, substituted alkyl, alkenyl, cyano, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heterocyclo, heteroaryl, OR7, C02R7, C(0)NR8R9, OC(0)R7, OC(0)OR7, OC(0)NR8R9, OCH2C02R7, C(0)R7, NR8R9, NRι0C(O)R7, NRι0C(O)OR7, NRιoC(0)C(0)OR7, NR10C(O)C(O)NR8R9, NR10C(O)C(O)alkyl, NR10C(NCN)OR7, NR10C(O)NR8R9, NRι0C(NCN)NR8R9, NR10C(NRπ)NR8R9, NRι0SO2NR8R9, NR10SO2R7, SR7, S(0)R7, S02R7, S03R7, S02NR8R9, NHOR7, NR10NR8R9, N(COR7)OR10, N(CO2R7)OR10, C(O)NR10(CR12R13)rR7,
CO(CR12R13)pO(CR14R15)qC02R7, CO(CR12R13)rOR7, CO(CR123)pO(CRι45)qR7, CO(CR12R13)rNR8R9, OC(0)0(CR12R13)mNR8R9, OC(0)N(CR12R13)rR7, 0(CR12R13)mNR8R9, NR10C(O)(CRι2R13)rR7, NR10C(O)(CR123)rOR7,
NR,oC(=NC)(CR12R13)rR7, NR10CO(CR12R13)rNR8R9, NR1o(CR12R13)mOR7, NR10(CR12Ri3)rC02R7, NR1o(CR12R13)mNR8R9, NR10(CR12R13)nS02(CR14R15)qR7, CONR10(CR12R13)nSO2(CR145)qR7, SO2NR10(CRι23)nCO(CRι45)qR7, and SO2NR10(CRι2Ri3)mOR7 as well as pentafluorophenyl.
The terms "heterocycle", "heterocyclic", "heterocyclic group" or "heterocyclo" refer to fully saturated or partially unsaturated cyclic groups (for example, 3 to 13 member monocyclic, 7 to 17 member bicyclic, or 10 to 20 member tricyclic ring systems, preferably containing a total of 3 to 10 ring atoms) which have at least one heteroatom in at least one carbon atom-containing ring. Each ring of the heterocyclic group containing a heteroatom may have 1, 2, 3 or 4 heteroatoms selected from nitrogen atoms, oxygen atoms and/or sulfur atoms, where the nitrogen and sulfur heteroatoms may optionally be oxidized and the nitrogen heteroatoms may optionally be quaternized. The heterocyclic group may be attached at any heteroatom or carbon atom of the ring or ring system. The rings of multi-ring heterocycles may be either fused, bridged and/or joined through one or more spiro unions. Exemplary heterocyclic groups include
Figure imgf000021_0001
The terms "substituted heterocycle" or "substituted heterocyclo" and the like refer o such heterocylo groups as defined above substituted with one or more groups listed in the definition of T1, T2 and T3, preferably selected from halogen, nitro, alkyl, substituted alkyl, alkenyl, cyano, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heterocyclo, heteroaryl,oxo, OR7, C02R7, C(0)NR8R9, OC(0)R7, OC(0)OR7, OC(0)NR8R9, OCH2C02R7, C(0)R7, NR8R9, NR10C(O)R7, NR10C(O)OR7, NR10C(O)C(O)OR7, NR,0C(O)C(O)NR8R9, NR,0C(O)C(O)alkyl, NR10C(NCN)OR7, NR10C(O)NR8R9, NR10C(NCN)NR8R9, NRι0C(NRπ)NR8R9, NR10SO2NR8R9, NR10SO2R7, SR7, S(0)R7, S02R7, S03R7, S02NR8R9, NHOR7, NR10NR8R9, N(COR7)ORi0, N(CO2R7)OR10, C(O)NR10(CRι23)rR7, CO(CR12R13)pO(CR145)qC02R7, CO(CR12R13)rOR7, CO(CR12R13)pO(CR145)qR7, CO(CR12R13)rNR8R9, OC(0)0(CR123)mNR8R9, OC(0)N(CR12R13)rR7, 0(CRι2R13)mNR8R9, NR10C(O)(CR12RI3)rR7, NR10C(O)(CR12R13)rOR7, NR10C(=NC)(CR12R13)rR7, NR10CO(CR12R13)rNR8R9, NR10(CR12R13)mOR7, NR10(CR12Ri3)rC02R7, NR10(CRI2R13)mNR8R9, NR1o(CR12R13)nS02(CR14R15)qR7, CONR10(CR12R13)nSO2(CR14R15)qR7, SO2NR10(CR12R,3)nCO(CR145)qR7, and SO2NR10(CR123)mOR7.
The term "heteroaryl" as used herein alone or as part of another group refers to a 5- 6- or 7- membered aromatic rings containing from 1 to 4 nitrogen atoms and/or 1 or 2 oxygen or sulfur atoms provided that the ring contains at least 1 carbon atom and no more than 4 heteroatoms. The heteroaryl ring is linked through an available carbon or nitrogen atom. Also included within the definition of heteroaryl are such rings fused to a cycloalkyl, aryl, cycloheteroalkyl, or another heteroaryl ring. One, two, or three available carbon or nitrogen atoms in the heteroaryl ring can be optionally substituted with substituents listed in the description of Tl 5 T2 and T3. Also an available nitrogen or sulfur atom in the heteroaryl ring can be oxidized. Examples of heteroaryl rings include
Figure imgf000022_0001
Figure imgf000022_0002
Figure imgf000023_0001
Figure imgf000023_0002
Figure imgf000023_0003
Figure imgf000024_0001
The term "substituted heteroaryl" refers to such heteroaryl groups as defined above substituted on any available atom with one or more groups listed in the definition of T1, T2 and T3, preferably selected from" refers to such heterocylo groups as defined above substituted with one or more groups listed in the definition of T1, T2 and T3, preferably selected from halogen, nitro, alkyl, substituted alkyl, alkenyl, cyano, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heterocyclo, heteroaryl, OR7, C02R7, C(0)NR8R9, OC(0)R7, OC(0)OR7, OC(0)NR8R9, OCH2C02R7, C(0)R7, NR8R9, NR10C(O)R7, NR,0C(O)OR7, NR10C(O)C(O)OR7, NRI0C(O)C(O)NR8R9, NR10C(O)C(O)alkyl, NR10C(NCN)OR7, NR10C(O)NR8R9, NR10C(NCN)NR8R9, NR10C(NR11)NR8R9, NR10SO2NR8R9, NR10SO2R7, SR7, S(0)R7, S02R7, S03R7, S02NR8R9, NHOR7, NRι0NR8R9, N(COR7)ORιo, N(CO2R7)ORι0, C(O)NR10(CR12R13)rR7, CO(CR12R13)pO(CR145)qCO2R7, CO(CR12R13)rOR7, CO(CR,2RI3)pO(CR14R15)qR7, CO(CR12R13)rNR8R9, OC(0)0(CR12R13)mNR8R9, OC(0)N(CR12R,3)rR7, 0(CRι2R13)mNR8R9, NR10C(O)(CR123)rR7, NR10C(0)(CRI2R13)rOR7, NR1oC(=NC)(CR,2R13)rR7, NR,0CO(CR12RI3)rNR8R9, NR10(CRι23)mOR7, NR,0(CR12R13)rCO2R7, NR,0(CRι23)mNR8R9, NR10(CRι2R13)nS02(CR14R15)qR7, CONR1o(CR12R13)nS02(CR14R15)qR7, SO2NR,0(CRi2R13)nCO(CR,4R15)qR7, and SO2NRι0(CR12R13)mOR7.
R7, Rio, and Rn, are independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, alkynyl, cycloalkyl, substituted cycloalkyl, C(0)alkyl, C(0)substituted alkyl, C(0)cycloalkyl, C(O) substituted cycloalkyl, C(0)aryl, C(0)substituted aryl, C(0)Oalkyl, C(0)Osubstituted alkyl, C(0)heterocyclo, C(0)heteroaryl, aryl, substituted aryl, heterocyclo and heteroaryl.
R8 and R9 are independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, alkenyl, alkynyl, C(0)alkyl, C(0)substituted alkyl, C(0)cycloalkyl, C(0)substituted cycloalkyl, C(0)aryl, C(0)substituted aryl, C(0)Oalkyl, C(0)Osustituted alkyl, C(0)heterocyclo, C(0)heteroaryl, S(0)2alkyl, S(0)2substituted alkyl, S(0)2cycloalkyl, S(0)2substituted cycloalkyl, S(0)2aryl, S(0)2substituted aryl, S(0)2heterocyclo, S(0)2heteroaryl, aryl, substituted aryl, heterocyclo, and heteroaryl or R8 and R9 taken together with the nitrogen atom to which they are attached complete a heterocyclo or heteroaryl ring.
2 and R]4 are independently selected from hydrogen and alkyl or 1 to 4 carbons.
Ri3 and Rι5 are independently selected from hydrogen, alkyl of 1 to 4 carbons, and substituted alkyl or 1 to 4 carbons. n is zero or an integer from 1 to 4. m is an integer from 2 to 6. p is an integer from 1 to 3. q is zero or an integer from 1 to 3. r is zero or an integer from 1 to 6. T1, T2, and T3 are are each independently (1) hydrogen or T6, where T6 is (i) alkyl, (hydroxy)alkyl, (alkoxy)alkyl, alkenyl, alkynyl, cycloalkyl, (cycloalkyl)alkyl, cycloalkenyl, (cycloalkenyl)alkyl, aryl, (aryl)alkyl, heterocyclo, (heterocylco)alkyl, heteroaryl, or (heteroaryl)alkyl; (ii) a group (i) which is itself substituted by one or more of the same or different groups (i); or (iii) a group (i) or (ii) which is independently substituted by one or more (preferably 1 to 3) of the following groups (2) to (13) of the definition of T1, T2 and T3; (2) -OH or -OT6,
(3) -SH or -ST6,
(4) -C(0),H, -C(0)tT6, or -0-C(0)T6, where t is 1 or 2;
(5) -S03H, -S(0)tT6, or S(0)tN(T9)T6,
(6) halo, (7) cyano,
(8) nitro,
(9) -T4-NT7T8,
(10) -T4-N(T9)-T -NT7T8, (11) -T4-N(T10)-T5-T6, (12) -T -N(T10)-T5-H,
(13) oxo, T4 and T5 are each independently
(1) a single bond,
(2) -Tu-S(0),-T12-, (3) -Tn-C(0)-T12-,
(4) -TU-C(S)-T12-,
(5) -Tπ-0-T12-,
(6) -T"-S-T12-,
(7) -Tπ-0-C(0)-T12-, (8) -Tπ-C(0)-0-T12-,
(9) -T"-C(=NT9a)-TI2-, or (10) -Tn-C(O)-C(O)-T12- T7, T8, T9, T9a and T10
(1) are each independently hydrogen or a group provided in the definition of T6, or (2) T7 and T8 may together be alkylene or alkenylene, completing a 3- to 8- membered saturated or unsaturated ring together with the atoms to which they are attached, which ring is unsubstituted or substituted with one or more groups listed in the description of T1, T2 and T3, or
(3) T7 or T8, together with T9, may be alkylene or alkenylene completing a 3- to 8-membered saturated or unsaturated ring together with the nitrogen atoms to which they are attached, which ring is unsubstituted or substituted with one or more groups listed in the description of T1, T2 and T3, or
(4) T7 and T8 or T9 and T10 together with the nitrogen atom to which they are attached may combine to form a group -N=CT13T14 where T13 and T14 are each independently H or a group provided in the definition of T6; and
1 1 1
T and T are each independently
(1) a single bond,
(2) alkylene,
(3) alkenylene, or (4) alkynylene.
"T cell-mediated diseases" refers to any disorder or disease state in which modulation of the activity of T cells is implicated in a process which results in either a pathophysiological state or a process where the normal function of T cells is intended to be suppressed for therapeutic benefit. Examples of T cell mediated disorders include transplant rejection, graph verses host disease, and autoimmune disorders, such as rheumatoid arthritis, multiple sclerosis, juvenile diabetes, asthma, and inflammatory bowel disease, T-cell mediated hypersensitivity diseases, ischemic or reperfusion injury, and T-cell proliferative disorders.
PDE7 inhibitors in accordance with the present invention are employed, typically in the form of a pharmaceutical composition including a pharmaceutically acceptable carrier for the treatment of T-cell mediated disease. The compounds employed for this purpose are typically administered in an amount from about 0.01 to 100 mg kg/day.
The pharmaceutical compositions comprising at least one PDE7 inhibitor may be formulated, for example, by employing conventional solid or liquid vehicles or diluents, as well as pharmaceutical additives of a type appropriate to the mode of desired administration (for example, excipients, binders, preservatives, stabilizers, flavors, etc.) according to techniques such as those well known in the art of pharmaceutical formulation.
The PDE7 inhibitors may be administered by any suitable means, for example, orally, such as in the form of tablets, capsules, granules or powders; sublingually; buccally; parenterally, such as by subcutaneous, intravenous, intramuscular, or intrasternal injection or infusion techniques (e.g., as sterile injectable aqueous or non-aqueous solutions or suspensions); nasally such as by inhalation spray; topically, such as in the form of a cream or ointment; or rectally such as in the form of suppositories; in dosage unit formulations containing non-toxic, pharmaceutically acceptable vehicles or diluents. The present compounds may, for example, be administered in a form suitable for immediate release or extended release. Immediate release or extended release may be achieved by the use of suitable pharmaceutical compositions comprising the present compounds, or, particularly in the case of extended release, by the use of devices such as subcutaneous implants or osmotic pumps. The present compounds may also be administered in the form of liposomes.
Exemplary compositions for oral administration include suspensions which may contain, for example, microcrystalline cellulose for imparting bulk, alginic acid or sodium alginate as a suspending agent, methylcellulose as a viscosity enhancer, and sweeteners or flavoring agents such as those known in the art; and immediate release tablets which may contain, for example, microcrystalline cellulose, dicalcium phosphate, starch, magnesium stearate and/or lactose and/or other excipients, binders, extenders, disintegrants, diluents and lubricants such as those known in the art. The present compounds may also be delivered through the oral cavity by sublingual and/or buccal administration. Molded tablets, compressed tablets or freeze-dried tablets are exemplary forms which may be used. Exemplary compositions include those formulating the present compound(s) with fast dissolving diluents such as mannitol, lactose, sucrose and/or cyclodextrins. Also included in such formulations may be high molecular weight excipients such as celluloses (avicel) or polyethylene glycols (PEG). Such formulations may also include an excipient to aid mucosal adhesion such as hydroxy propyl cellulose (HPC), hydroxy propyl methyl cellulose (HPMC), sodium carboxy methyl cellulose (SCMC), maleic anhydride copolymer (e.g., Gantrez), and agents to control release such as polyacrylic copolymer (e.g., Carbopol 934). Lubricants, glidants, flavors, coloring agents and stabilizers may also be added for ease of fabrication and use.
Exemplary compositions for nasal aerosol or inhalation administration include solutions in saline which may contain, for example, benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, and or other solubilizing or dispersing agents such as those known in the art.
Exemplary compositions for parenteral administration include injectable solutions or suspensions which may contain, for example, suitable non-toxic, parenterally acceptable diluents or solvents, such as mannitol, 1,3-butanediol, water, Ringer's solution, an isotonic sodium chloride solution, or other suitable dispersing or wetting and suspending agents, including synthetic mono- or diglycerides, and fatty acids, including oleic acid.
Exemplary compositions for rectal administration include suppositories which may contain, for example, a suitable non-irritating excipient, such as cocoa butter, synthetic glyceride esters or polyethylene glycols, which are solid at ordinary temperatures, but liquefy and/or dissolve in the rectal cavity to release the drug.
Exemplary compositions for topical administration include a topical carrier such as Plastibase (mineral oil gelled with polyethylene). The effective amount of a compound employed in the present invention may be determined by one of ordinary skill in the art, and includes exemplary dosage amounts for an adult human of from about 0.01 to 100 mg/kg of body weight of active compound per day, which may be administered in a single dose or in the form of individual divided doses, such as from 1 to 4 times per day. It will be understood that the specific dose level and frequency of dosage for any particular subject may be varied and will depend upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the species, age, body weight, general health, sex and diet of the subject, the mode and time of administration, rate of excretion, drug combination, and severity of the particular condition. Preferred subjects for treatment include animals, most preferably mammalian species such as humans, and domestic animals such as dogs, cats and the like, subject to inflammatory, immunological, or respiratory cell-associated disorders.
PDE7 inhibitors for use in the treatment of various T-cell mediated diseases are those covered by Formula I Compounds of Formula I include salts, prodrugs and solvates. The term
"salt(s)", as employed herein, denotes acidic and/or basic salts formed with inorganic and/or organic acids and bases. Zwitterions (internal or inner salts) are included within the term "salt(s)" as used herein (and may be formed, for example, where the R substituents comprise an acid moiety such as a carboxyl group). Also included herein are quaternary ammonium salts such as alkylammonium salts. Pharmaceutically acceptable (i.e., non-toxic, physiologically acceptable) salts are preferred, although other salts are useful, for example, in isolation or purification steps which may be employed during preparation. Salts of the compounds of the formula I may be formed, for example, by reacting a compound I with an amount of acid or base, such as an equivalent amount, in a medium such as one in which the salt precipitates or in an aqueous medium followed by lyophilization.
Exemplary acid addition salts include acetates (such as those formed with acetic acid or trihaloacetic acid, for example, trifluoroacetic acid), adipates, alginates, ascorbates, aspartates, benzoates, benzenesulfonates, bisulfates, borates, butyrates, citrates, camphorates, camphorsulfonates, cyclopentanepropionates, digluconates, dodecylsulfates, ethanesulfonates, fumarates, glucoheptanoates, glycerophosphates, hemisulfates, heptanoates, hexanoates, hydrochlorides, hydrobromides, hydroiodides, 2-hydroxyethanesulfonates, lactates, maleates, methanesulfonates, 2-naphthalenesulfonates, nicotinates, nitrates, oxalates, pectinates, persulfates,
3-phenylpropionates, phosphates, picrates, pivalates, propionates, salicylates, succinates, sulfates (such as those formed with sulfuric acid), sulfonates (such as those mentioned herein), tartrates, thiocyanates, toluenesulfonates, undecanoates, and the like.
Exemplary basic salts (formed, for example, where the R substituents comprise an acidic moiety such as a carboxyl group) include ammonium salts, alkali metal salts such as sodium, lithium, and potassium salts, alkaline earth metal salts such as calcium and magnesium salts, salts with organic bases (for example, organic amines) such as benzathines, dicyclohexylamines, hydrabamines, N-methyl-D-glucamines, N-methyl-D- glucamides, t-butyl amines, and salts with amino acids such as arginine, lysine and the like. The basic nitrogen-containing groups may be quatemized with agents such as lower alkyl halides (e.g. methyl, ethyl, propyl, and butyl chlorides, bromides and iodides), dialkyl sulfates (e.g. dimethyl, diethyl, dibutyl, and diamyl sulfates), long chain halides (e.g. decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides), aralkyl halides (e.g. benzyl and phenethyl bromides), and others.
Prodrugs and solvates of the compounds of the invention are also contemplated herein. The term "prodrug", as employed herein, denotes a compound which, upon administration to a subject, undergoes chemical conversion by metabolic or chemical processes to yield a compound of the Formula I, or a salt and/or solvate thereof. Solvates of the compounds of Formula I are preferably hydrates.
All stereoisomers of the present compounds, such as those which may exist due to asymmetric carbons on the R substituents of the compound of the formula I, including enantiomeric and diastereomeric forms, are contemplated within the scope of this invention. Individual stereoisomers of the compounds of the invention may, for example, be substantially free of other isomers, or may be admixed, for example, as racemates or with all other, or other selected, stereoisomers. The chiral centers of the present invention can have the S or R configuration as defined by the IUPAC 1974 Recommendations. The compounds of Formula I are typically employed as part of a pharmaceutical composition including a pharmaceutically acceptable carrier for the treatment of respiratory and non-respiratory diseases. The compounds employed for this purpose are typically administered in an amount of from about 0.01 to 100 mg/kg/day. The compounds of Formula I are especially effective in inhibiting the PDE7 enzyme. Additionally a subset of compounds are also effective at inhibiting PDE4.
The pharmaceutical composition comprising at least one compound of Formula I may be formulated, for example, by employing conventional solid or liquid vehicles or diluents, as well as pharmaceutical additives of a type appropriate to the mode of desired administration (for example, excipients, binders, preservatives, stabilizers, flavors, etc.) according to techniques such as those well known in the art of pharmaceutical formulation.
The compounds of Formula I may be administered by any suitable means, for example, orally, such as in the form of tablets, capsules, granules or powders; sublingually; bucally; parenterally, such as by subcutaneous, intravenous, intramuscular, or intrasternal injection or infusion techniques (e.g., as sterile injectable aqueous or non- aqueous solutions or suspensions); nasally such as by inhalation spray; topically, such as in the form of a cream or ointment; or rectally such as in the form of suppositories; in dosage unit formulations containing non-toxic, pharmaceutically acceptable vehicles or diluents. The present compounds may be based for immediate release or extended release by the use of suitable pharmaceutical compositions comprising the present compounds, or, particularly in the case of extended release, by the use of devices such as subcutaneous implants or osmotic pumps. The present compounds may also be administered liposomally.
Exemplary compositions for oral administration include suspensions which may contain, for example, microcrystalline cellulose for imparting bulk, alginic acid or sodium alginate as a suspending agent, methylcellulose as a viscosity enhancer, and sweeteners or flavoring agents such as those known in the art; and immediate release tablets which may contain, for example, microcrystalline cellulose, dicalcium phosphate, starch, magnesium stearate and/or lactose and/or other excipients, binders, extenders, disintegrants, diluents and lubricants such as those known in the art. The present compounds may also be delivered through the oral cavity by sublingual and/or buccal administration. Molded tablets, compressed tablets or freeze-dried tablets are exemplary forms which may be used. Exemplary compositions include those formulating the present compound(s) with fast dissolving diluents such as mannitol, lactose, sucrose and/or cyclodextrins. Also included in such formulations may be high molecular weight excipients such as celluloses (avicel) or polyethylene glycols (PEG). Such formulations may also include an excipient to aid mucosal adhesion such as hydroxy propyl cellulose (HPC), hydroxy propyl methyl cellulose (HPMC), sodium carboxy methyl cellulose (SCMC), maleic anhydride copolymer (e.g., Gantrez), and agents to control release such as polyacrylic copolymer (e.g., Carbopol 934). Lubricants, glidants, flavors, coloring agents and stabilizers may also be added for ease of fabrication and use.
Exemplary compositions for nasal aerosol or inhalation administration include solutions in saline which may contain, for example, benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, and/or other solubilizing or dispersing agents such as those known in the art.
Exemplary compositions for parenteral administration include injectable solutions or suspensions which may contain, for example, suitable non-toxic, parenterally acceptable diluents or solvents, such as mannitol, 1,3-butanediol, water, Ringer's solution, an isotonic sodium chloride solution, or other suitable dispersing or wetting and suspending agents, including synthetic mono- or diglycerides, and fatty acids, including oleic acid.
Exemplary compositions for rectal administration include suppositories which may contain, for example, a suitable non-irritating excipient, such as cocoa butter, synthetic glyceride esters or polyethylene glycols, which are solid at ordinary temperatures, but liquefy and/or dissolve in the rectal cavity to release the drug. Exemplary compositions for topical administration include a topical carrier such as Plastibase (mineral oil gelled with polyethylene).
The effective amount of a compound of the present invention may be determined by one of ordinary skill in the art, and includes exemplary dosage amounts for an adult human from about 0.01 to 100 mg/kg of body weight of active compound per day, which may be administered in a single dose or in the form of individual divided doses, such as from 1 to 4 times per day. It will be understood that the specific dose level and frequency of dosage for any particular subject may be varied and will depend upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the species, age, body weight, general health, sex and diet of the subject, the mode and time of administration, rate of excretion, drug combination, and severity of the particular condition. Preferred subjects for treatment include animals, most preferably mammalian species such as humans, and domestic animals such as dogs, cats and the like, subject to leukocyte activation or respiratory cell-associated disorders.
Methods of Preparation
Compounds of Formula I may be prepared by reference to the methods illustrated in Schemes A and B. As shown therein the end product is a compound having the same structural formula as Formula I. It will be understood that any compound of Formula I may be produced by Scheme A by the suitable selection of appropriate substitution. Solvents, temperatures, pressures, and other reaction conditions may readily be selected by one of ordinary skill in the art. All documents cited are incorporated herein by reference in their entirety. Starting materials are commercially available or readily prepared by one of ordinary skill in the art. Constituents of compounds are as defined herein or elsewhere in the specification. Scheme A outlines the synthesis of compounds of Formula I. Compound I is readily available by several methods well known in the literature including the method of Curd et. al. (Z = CH) reported in J. Chem. Soc. 1948, 1759-1765, and the method of Robins et. al. (Z = N) reported in J. Am. Chem. Soc. 1955, 77, 2256-2259. Compound I is treated with reagent II, which may be an or an amine, alcohol, a thiol or a sulfonamide in the presence of a suitable base to provide intermediate III. Palladium catalyzed additions of amines to aryl and heteroaryl halides are a recent addition to organic methodology, which greatly simplify or permit the synthesis of compounds for which there was no satisfactory synthetic approach. For example see Wolfe, et. al in Ace. Chem. Res. 1998, 31, 803-818, and Wolfe, et. al. in J. Org. Chem. 2000, 65, 1158-1174. Use of this new "Buchwald-Hartwig amination" methodology allowed the conversion of III under palladium-catalysed coupling conditions in the presence of an amine IN to provide compound V, which is a compound of formula I. Scheme A
M C
Figure imgf000035_0001
I dioxane with base III
100°C Q= -NR3- , -0-, -S-, -S02NR3-
Figure imgf000035_0002
Tol.BINAP 0.3 Eq. f-BuONa 2.0 Eq.
Toluene / Dioxane or DMA at 105°C
An alternative synthesis of compound V, illustrated in Scheme B starts by condensation (for example of such condensations see Rajasekharan, et. al. Indian J. Chem Sect. B, 1983, 22, 76-77) of an anthranilic acid ester, or 2- aminonicotinic acid (VIII) which are either commercially available or readily prepared by a number of methods, with a cyanamide (VII) which is readily prepared by reaction of an amine (IV) with cyanogen bromide (VI) according to methods reported in the literature (for example see Joshua, et. al. J. Indian. Chem. Soc. 1961, 38, 979-987) to produce compound (LX). Treatment of (LX) with phosphorous oxychloride with our without the addition of a base such as hunigs base or N,N-dimethylaniline provides compound (X) which is a compound of Formula I. Compound (X) can react with a variety of nucleophiles under conventional heating or microwave heating to provide compound (V) which is also a compound of Formula I. Scheme B
VI BrCN
R -NH2 R -N CN
H IV VII
Figure imgf000036_0001
Cl
POCI3
IX
H
X
Figure imgf000036_0002
Q= -NR3- , -0-, -S-, -S02NR3
Utility Selective PDE7 inhibitors or dual PDE7-PDE4 inhibitors including compounds of formulas I, are useful in the treatment (including prevention, partial alleviation or cure) of leukocyte activation-associated disorders, which include (but are not limited to) disorders such as: transplant rejection (such as organ transplant, acute transplant, xenotransplant or heterograft or homograft such as is employed in burn treatment); protection from ischemic or reperfusion injury such as ischemic or reperfusion injury incurred during organ transplantation, myocardial infarction, stroke or other causes; transplantation tolerance induction; arthritis (such as rheumatoid arthritis, psoriatic arthritis or osteoarthritis); multiple sclerosis; respiratory and pulmonary diseases including but not limited to asthma, exercise induced asthma, chronic obstructive pulmonary disease (COPD), emphysema, bronchitis, and acute respiratory distress syndrome (ARDS); inflammatory bowel disease, including ulcerative colitis and Crohn's disease; lupus (systemic lupus erythematosis); graft vs. host disease; T-cell mediated hypersensitivity diseases, including contact hypersensitivity, delayed-type hypersensitivity, and gluten- sensitive enteropathy (Celiac disease); psoriasis; contact dermatitis (including that due to poison ivy); Hashimoto's thyroiditis; Sjogren's syndrome; Autoimmune Hyperthyroidism, such as Graves' Disease; Addison's disease (autoimmune disease of the adrenal glands); Autoimmune polyglandular disease (also known as autoimmune polyglandular syndrome); autoimmune alopecia; pernicious anemia; vitiligo; autoimmune hypopituatarism; Guillain-Barre syndrome; other autoimmune diseases; glomerulonephritis; serum sickness; uticaria; allergic diseases such as respiratory allergies (e.g., asthma, hayfever, allergic rhinitis) or skin allergies; scleracierma; mycosis fungoides; acute inflammatory and respiratory responses (such as acute respiratory distress syndrome and ishchemia/reperfusion injury); dermatomyositis; alopecia areata; chronic actinic dermatitis; eczema; Behcet's disease; Pustulosis palmoplanteris; Pyoderma gangrenum; Sezary's syndrome; atopic dermatitis; systemic schlerosis; and morphea.
The term "leukocyte activation-associated disorder" or "leukocyte activation- mediated disorder" as used herein includes each of the above referenced diseases or disorders. The compounds of the present invention are useful for treating the aforementioned exemplary disorders irrespective of their etiology.
Those present compounds which are dual PDE7/4 inhibitors may be more effective than either a selective PDE4 inhibitor or a selective PDE7 inhibitor in the above mentioned disease states, as a result of either additive or synergistic activity resulting from the combined inhibition of PDE7 and PDE4.
The present invention thus provides methods for the treatment of disorders as discussed above comprising the step of administering to a subject in need thereof of at least one selective PDE7 inhibitor or at least one dual PDE7-PDE4 inhibitor for the treatment of leukocyte activation-associated or leukocyte-activation mediated disease. Other therapeutic agents such as those described below may be employed with the compounds of the present invention. In the methods of the present invention, such other therapeutic agent(s) may be administered prior to, simultaneously with or following the administration of the compound(s) of the present invention.
The methods of treating diseases which would benefit from the inhibition of PDE7 or the inhibition of both PDE7-PDE4 by a dual agent may comprise administering compounds of Formula (I) alone or in combination with each other and/or other suitable therapeutic agents useful in treating such conditions such as: immunosuppressants such as, cyclosporins (e.g., cyclosporin A), anti-EL-l agents, such as Anakinra, the IL-1 receptor antagonist, CTLA4-Ig, antibodies such as anti-ICAM-3, anti-IL-2 receptor (Anti-Tac), anti-CD45RB, anti-CD2, anti-CD3, anti-CD4, anti-CD80, anti-CD86, monoclonal antibody OKT3, agents blocking the interaction between CD40 and CD 154, such as antibodies specific for CD40 and/or CD154 (i.e., CD40L), fusion proteins constructed from CD40 and CD 154 (CD40Ig and CD8-CD154), interferon beta, interferon gamma, methotrexate, FK506 (tacrolimus, Prograf), rapamycin (sirolimus or Rapamune)mycophenolate mofetil, leflunomide (Arava), azathioprine and cyclophosphamide, inhibitors, such as nuclear translocation inhibitors, of NF-kappa B function, such as deoxyspergualin (DSG), non-steroidal antiinflammatory drugs (NSAIDs) such as ibuprofen, cyclooxygenase-2 (COX-2) inhibitors such as celecoxib (Celebrex) and rofecoxib (Vioxx), or derivatives thereof, steroids such as prednisone or dexamethasone, gold compounds TNF-α inhibitors such as tenidap, anti-TNF antibodies or soluble TNF receptor such as etanercept (Enbrel), inhibitors of p-38 kinase such as BIRB-796, RO-3201195, VX-850, and VX-750, beta-2 agonists such as albuterol, levalbuterol (Xopenex), and salmeterol (Serevent), inhibitors of leukotriene synthesis such as montelukast (Singulair) and zariflukast (Accolate), and anticholinergic agents such as ipratropium bromide (Atrovent), PDE4 inhibitors such as Arofyline, Cilomilast, Roflumilast, C-11294A, CDC-801, BAY-19-8004, Cipamfylline, SCH351591, YM-976, PD-189659, Mesiopram, Pumafentrine, CDC-998, IC-485, and KW-4490, PDE7 inhibitors such as IC242, (Lee, et. al. PDE7A is expressed in human B-lymphocytes and is up-regulated by elevation of intracellular cAMP. Cell Signalling, 14, 277-284, (2002)) and also include compounds disclosed in the following patent documents: WO 0068230, WO 0129049, WO 0132618, WO 0134601, WO 0136425, WO 0174786, WO 0198274, WO 0228847, U.S. Provisional Application Serial No. 60/287,964, and U.S. Provisional Application Serial No. 60/355, 141anti-cytokines such as anti-IL-1 niAb or EL-l receptor agonist, anti-IL-4 or B -4 receptor fusion proteins and PTK inhibitors such as those disclosed in the following U.S. Patents and Applications, incorporated herein by reference in their entirety: U.S Patent No. 6,235,740, U.S. Patent No. 6,239,133, U.S. Application Serial No. 60/065,042, filed 11/10/97 (Attorney Docket No. QA207*), U.S. Application Serial No. 09/173,413, filed 10/15/98 (Attorney Docket No. QA 207a), and U.S. Patent No. 5,990,109.
See the following documents and references cited therein: Hollenbaugh, D., Douthwright, J., McDonald, V., and Aruffo, A., "Cleavable CD40Ig fusion proteins and the binding to sgp39", J. Immunol Methods (Netherlands), 188(1), p. 1-7 (Dec 15 1995); Hollenbaugh, D., Grosmaire, L.S., Kullas, CD., Chalupny, N.J., Braesch-Andersen, S., Noelle, R.J., Stamenkovic, I., Ledbetter, J.A., and Aruffo, A., "The human T cell antigen gp39, a member of the TNF gene family, is a ligand for the CD40 receptor: expression of a soluble form of gp39 with B cell co-stimulatory activity", EMBO J (England), 11(12), p 4313-4321 (Dec 1992); and Moreland, L.W. et al., "Treatment of rheumatoid arthritis with a recombinant human tumor necrosis factor receptor (p75)-Fc fusion protein, New England J. of Medicine, 337(3), p. 141-147 (1997).
Compounds present invention (especially selective PDE 7 inhibitors) may also be employed in combination with PDE 4 inhibitors. Examples of selective PDE4 inhibitors currently in development, which can be used in combination with compounds of the present invention include Arofyline, Cilomilast, Roflumilast, C-11294A, CDC- 801, BAY- 19-8004, Cipamfylline, SCH351591, YM-976, PD- 189659, Mesiopram, Pumafentrine, CDC-998, IC-485, and KW-4490.
The above other therapeutic agents, when employed in combination with the compounds of the present invention, may be used, for example, in those amounts indicated in the Physicians' Desk Reference (PDR) or as otherwise determined by one of ordinary skill in the art.
Use of the compounds of the present invention as encompassed by formula I in treating leukocyte activation-associated disorders is exemplified by, but is not limited to, treating a range of disorders such as: transplant (such as organ transplant, acute transplant, xenotransplant or heterograft or homograft (such as is employed in burn treatment)) rejection; protection from ischemic or reperfusion injury such as ischemic or reperfusion injury incurred during organ transplantation, myocardial infarction, stroke or other causes; transplantation tolerance induction; arthritis (such as rheumatoid arthritis, psoriatic arthritis or osteoarthritis); multiple sclerosis; respiratory and pulmonary diseases including but not limited to asthma, exercise induced asthma, chronic obstructive pulmonary disease (COPD), emphysema, bronchitis, and acute respiratory distress syndrome (ARDS); inflammatory bowel disease, including ulcerative colitis and Crohn's disease; lupus (systemic lupus erythematosis); graft vs. host disease; T-cell mediated hypersensitivity diseases, including contact hypersensitivity, delayed-type hypersensitivity, and gluten-sensitive enteropathy (Celiac disease); psoriasis; contact dermatitis (including that due to poison ivy); Hashimoto's thyroiditis; Sjogren's syndrome; Autoimmune Hyperthyroidism, such as Graves' Disease; Addison's disease (autoimmune disease of the adrenal glands); Autoimmune polyglandular disease (also known as autoimmune polyglandular syndrome); autoimmune alopecia; pernicious anemia; vitiligo; autoimmune hypopituatarism; Guillain-Barre syndrome; other autoimmune diseases; glomerulonephritis; serum sickness; uticaria; allergic diseases such as respiratory allergies (asthma, hayfever, allergic rhinitis) or skin allergies; scleracierma; mycosis fungoides; acute inflammatory and respiratory responses (such as acute respiratory distress syndrome and ishchemia/reperfusion injury); dermatomyositis; alopecia areata; chronic actinic dermatitis; eczema; Behcet's disease; Pustulosis palmoplanteris; Pyoderma gangrenum; Sezary's syndrome; atopic dermatitis; systemic schlerosis; and morphea.
The combined activity of the present compounds towards T-cells and other PDE7-expressing cells may be of value in the treatment of any of the aforementioned disorders. Additionally those present compounds which are dual PDE4/7 inhibitors may be more effective than either a selective PDE4 inhibitor or a selective PDE7 inhibitor in the above mentioned disease states.
In a particular embodiment, the compounds of the present invention are useful for the treatment of the aforementioned exemplary disorders irrespective of their etiology, for example, for the treatment of transplant rejection, rheumatoid arthritis, multiple sclerosis, chronic obstructive pulmonary disease, inflammatory bowel disease, lupus, graft v. host disease, T-cell mediated hypersensitivity disease, psoriasis, Hashimoto's thyroiditis, Guillain-Barre syndrome, cancer, contact dermatitis, allergic disease such as allergic rhinitis, asthma, ischemic or reperfusion injury, respiratory diseases such as asthma, COPD and bronchitis or atopic dermatitis whether or not associated with leukocyte activation.
PDE- containing cell lysates
Hut78 cells were grown in 10% FCS in Iscoves Modified Dulbecco's Medium (Gibco BRL-Life Technologies, Grand Island, NY) with antibiotics. Cells were centrifuged and resuspended in four volumes of [40 mM Tris (pH 7.5)/50 μM EDTA/200uM PMSF with a cocktail of Protease inhibitors (Boehringher Mannheim, Indianapolis, IN)] at 4C. Cells were homogenized using aVirtis homogenizer, and the lysate was centrifuged twice for 15 min at 15,000 x g. Glycerol was added to a final volume of 50% for storage at -20C.
SPA assay Inhibition of PDE activity in Hut78 cell lysate was determined using an SPA specific for cAMP (Amersham Pharmacia Biotech, Buckinghamshire, UK) according to the manufacturers instructions with minor modifications. Enzyme assays were performed at room temperature in the presence of 50mM Tris HCI, pH7.5, containing 8.3mM MgCl2, 1.7mM EGTA and 0.5mg/mL BSA. Each assay was performed in a lOOμL reaction volume in 96 well microtitre plates containing the above buffer, 0.3ul of Hut78 cell lysate treated with 2 uM Zardaverine to inhibit PDE3 and PDE4, 0.05 uCi of [5',8- 3H] Adenosine 3 ',5 '-cyclic phosphate as an ammonium salt for 20 min. The reaction was terminated by the addition of 50μl PDE SPA beads (lmg) water with lOmM cold cAMP (Sigma, St. Louis MO). The reaction mix was allowed to settle for 20 minutes before counting in a Top Count-NXT scintillation counter (Packard BioScience, Meriden, CT). For individual PDE enzymes other than PDE7, the assay was essentially unchanged except that 3H-cyclic GMP was used as the substrate for PDE1, PDE5 and PDE6. The following PDEs/activators and enzyme sources were used: PDE1, bovine (Sigma St Louis), calmodulin; PDE2, rat kidney, cGMP; PDE3, human platelet; PDE4, rat kidney; PDE5, human platelet, and PDE6, bovine retina.
T cell Proliferation Assay
Peripheral blood mononuclear cells (PBMC) were isolated from whole blood by density gradient centrifugation over Lymphoprep, 1.077. Cells were plated into 96 well U-bottom plates at 2.5xl05 cells/well in 10% FBS RPMI 1640 (Life Technologies/Gibco- BRL) containing lOug/ml anti-CD3 (G19-4, Bristol-Myers Squibb P.R.I., Princeton, NJ) and lug/ml anti-CD28 (9.3, Bristol-Myers Squibb P.R.I.) in the presence and absence of inhibitors. DMSO (used as a solvent for inhibitors) was added to the medium at 0.1% final concentration. The total volume per well was 200 μL. Cells were incubated at 37C 5% C02 for 3 days, at which time 0.5μCi of 3H-thymidine was added to each well. Six hours following the addition of 3H-thmidine, the plates were harvested onto filter plates, 30ul EcoLite scintillant (ICN, Costa Mesa, CA) was added per well, and plates read on a Top Count-NXT scintillation counter.
TNF secretion assay The ability of compounds to inhibit the production and secretion of TNFα from leukocytes was performed using either PBMC (obtained as described above) or the THP- 1 cell line as a source of monocytes. Compounds were diluted in RPMI 1640 supplemented with 10% FBS and DMSO at a final concentration of 0.2%. Cells (2xl05/well in U-bottom 96 well plates) were pre-incubated with compounds for 30 min at 37 C prior to addition of lipopolysaccharide (LPS) at a final concentration of 6.25 ng/ml in a total volume of 200 μL. After 4h at 37C, 50 μL of supernatant was carefully aspirated for detection of soluble TNFα. Soluble TNFα was detected by ELISA developed by R&D Systems (Minneapolis, MN) according to the manufacturers instructions.
Examples
The following examples illustrate preferred embodiments of the present invention and do not limit the scope of the present invention which is defined in the claims. Abbreviations employed in the Examples are defined below. Compounds of the Examples are identified by the example and step in which they are prepared (e.g., "Al.l" denotes the title compound of step 1 of Example Al), or by the example only where the compound is the title compound of the example (for example, "A2" denotes the title compound of Example A2).
Abbreviations
Ac Acetyl
AcOH Acetic acid aq. Aqueous
CDI Carbonyldiimidazole
Bn Benzyl
Bu Butyl
Boc tert-butoxycarbonyl
DIC 1,3-Diisopropyl carbodiimide
DMAP Dimethylaminopyridine
DMA N,N-Dimethylacetamide
DMF dimethylformamide
DMSO Dimethylsulfoxide EDC l-(3-Dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride
EtOAc Ethyl acetate
Et Ethyl
EtOH Ethanol
H Hydrogen h Hours i iso
HPLC High pressure liquid chromatography
HOAc Acetic acid
Lawesson's Reagent [2,4-bis(4-methoxyphenyl)- 1 ,3-dithia-2,4-diphosphetane-2-4- disufide
LC liquid chromatography
Me Methyl
MeOH Methanol min. Minutes
M+ (M+H)+
M+I (M+H)+
MS Mass spectrometry n normal
Pd/C Palladium on carbon
Ph Phenyl
Pr Propyl
Ret Time Retention time rt or RT Room temperature sat. Saturated
S-Tol-BINAP (S)-(-)-2,2 ' -Bis(di-p-tolylphosphino)- 1,1' -binapthyl
TFA Trifluoroacetic acid
THF Tetrahydrofuran
YMC YMC Inc, Wilmington, NC 28403 Unless otherwise noted HPLC conditions used to determine retention times; 4 min gradient 0-100%B in A(A; 0.1% TFA in 90/10 water/methanol; B; 0.1%TFA in 10/90 water/methanol) using a YMC turbopack column at 220 nm.
Example Al 2-rr6 -Dimethoxy-4-rrr4-(methylsulfonyl)phenvI1methyllaminol-2- quinazolinvnamino1-4-methyl-5-thiazolecarboxylic acid, ethyl ester
Figure imgf000045_0001
Al
Al.l: 2-Chloro-6,7-dimethoxy-4-(4-methylsulfonylbenzyl)quinazoline
Figure imgf000045_0002
Al.l A mixture of commercially available 2,4-dichloro-6,7-dimethoxyquinazoline (200 mg, 0.772 mmol, 1 eq), 4-methylsulfonylbenzylamine hydrochloride (180 mg, 0.810 mmol, 1.05 eq) and diisopropylethylamine (0.40 mL, 2.32 mmol, 3 eq) in tetrahydrofuran (7.7 mL) was heated at reflux for 15.25 h. The reaction mixture was then cooled to rt and concentrated in vacuo. The resultant solid was slurried in methanol (10 mL) collected by filtration, washed with methanol and dried to provide 282 mg (89%) of Al.l as an off- white solid. LC/MS: 408 [M+H]+; HPLC: 98 % at 3.19 min (Phenomenex 5 μm C18 column 4.6 x 50 mm, 10-90 % aqueous methanol over 4 min containing 0.2% phosphoric acid, 4 n L/min, monitoring at 254 nm); 1H NMR (400 MHz, DMSO-d6): 6 8.98 (m, 1 H), 7.91 (d, J = 8.3 Hz, 2 H), 7.69 (s, 1 H), 7.62 (d, J = 8.3 Hz, 2 H), 7.11 (s, 1 H), 4.84 (apparent d, I = 5.7 Hz, 2 H), 3.90 (s, 3 H), 3.89 (s, 3 H), 3.19 (s, 3 H).
A1.2: 2-rr6.7-Dimethoxy-4-rrr4-fmethylsulfonvnphenyllmethyllaminol-2- quinazolinyllamino1-4-methyl-5-thiazolecarboxylic acid, ethyl ester
To a mixture of Al.l (65.4 mg, 0.160 mmol, 1 eq) and ethyl 2-amino-4- methylthiazole-5-carboxylate (59.7 mg, 0320 mmol, 2 eq) in 1: 1 toluene/ 1,4-dioxane (1.4 mL) in a 2-dram vial was added tris(dibenzylideneacetone)dipalladium(0) (14.6 mg,
0.016 mmol, 0.1 eq), 2-(di-t-butylphosphino)biphenyl (14.3 mg, 0.048 mmol, 0.3 eq) and sodium t-butoxide (30.7 mg, 0.320 mmol, 2 eq). The vial was purged with N2, sealed and heated in a 105 °C oil bath for 29.5 h. The reaction mixture was cooled to rt, filtered through celite and concentrated in vacuo. The residue was treated with methanol (ca. 1 mL) and the precipitated solid was collected by filtration, washed with methanol and dried to afford 47.6 mg (53%) of Al as a tan solid. LC/MS: 558 [M+H]+; HPLC: >90 % at 3.27 min (Phenomenex 5 μm C18 column 4.6 x 50 mm, 10-90 % aqueous methanol over 4 min containing 0.2% phosphoric acid, 4 mL/min, monitoring at 254 nm); 1H NMR (400 MHz, DMSO- 6): 6 11.43 (s, 1 H), 8.81 (br s, 1 H), 7.88(d, J = 8.3 Hz, 2 H), 7.71 (d, J = 8.2 Hz, 2 H), 7.64 (s, 1 H), 6.93 (s, 1 H), 4.96 (br s, 2 H), 4.23 (q, J = 7.1 Hz, 2 H), 3.91 (s, 3 H), 3.87 (s, 3 H), 3.16 (s, 3 H), 1.27 (t, 7 = 7.1 Hz, 3 H).
Example A2-A13
Figure imgf000046_0001
Examples A2 to A13 were prepared in a similar manner to that used for Example Al utilizing the appropriate amines in step Al.l.
Table A
Figure imgf000047_0001
Figure imgf000048_0001
Example A14
2-rr4-rrr4-(Aminosulfonyl)phenyllmethyl]amino1-6,7-dimethoxy-2- quinazolinyl1amino]-4-trifluoromethyl-5-thiazolecarboxylic acid, ethyl ester
Figure imgf000049_0001
A14
A14 was prepared in an manner analogous to example Al with the exception that in step Al.l 4-aminosulfonylbenzylamine hydrochloride was substituted for 4- methylsulfonylbenzylamine hydrochloride, and in step A1.2 ethyl 2-amino-4- trifluoromethyl-5-thiazole carboxylate was substituted for ethyl 2-amino-4-methyl-5- thiazole carboxylate. LCMS = Ret. Time = l.όlmin*, M+ = 613.20 * HPLC conditions used to determine retention times; 2 min gradient 0-100%B in A(A; 0.1% TFA in 90/10 water/methanol; B; 0.1%TFA in 10/90 water/methanol) using a TMC turbopack column at 220 nm.
Example A 15 2-[[4-rr[4-(AminosuIfonyl)phenyllmethyl1amino1-2-quinazolinyllamino1-4-methyl-5- thiazolecarboxylic acid, ethyl ester
Figure imgf000049_0002
A 15 A 15.1 : 2-chloro-4-(4-aminosulfonylbenzyl)quinazoline
Figure imgf000050_0001
A15.1
A mixture of 2,4-dichloroquinazoline [prepared from benzoyleneurea and POCl3 by the method of Butler et al., J. Chem. Soc. 1959, 1512.] (100 mg, 0.502 mmol, 1 eq) ,
4-aminosulfonylbenzylamine hydrochloride (117.5 mg, 0.527 mmol, 1.05 eq) and diisopropylethylamine (0.26 mL, 1.506 mmol, 3 eq) in absolute ethanol (1.6 mL) was stirred at ambient temperature for 4 h. The precipitated solid was collected by filtration, washed with water and cold ethanol, and dried to afford 154 mg (88%) of 2-chloro-4-(4- aminosulfonylbenzyl)quinazoline as a white solid. LC/MS: 349 [M+H]+; HPLC: 96 % at
1.86 min (Primesphere 5 μm C18 column 4.6 x 30 mm, 10-90 % aqueous methanol over
2 min containing 0.2% phosphoric acid, 5 mL/min, monitoring at 254 nm); Η NMR (400
MHz, DMSC fe): δ 9.37 (t, J = 5.8 Hz, 1 H), 8.32 (d, J = 8.2 Hz, 1 H), 7.85-7.53 (m, 7
H), 7.32 (s, 2 H), 4.81 (d, J = 5.7 Hz, 2 H).
A15.2; 2-[r4-[r[4-(Aminosulfonyl)phenyllmethvnaminol-2-quinazolinynaminol-4- methyl-5-thiazolecarboxylic acid, ethyl ester
To a mixture of A15.1 (77 mg, 0.221 mmol, 1 eq) and ethyl 2-amino-4- methylthiazole-5-carboxylate (82 mg, 0.442 mmol, 2 eq) in N,N-dimethylacetamide (2.2 mL) in a 2-dram vial was added tris(dibenzylideneacetone)dipalladium(0) (20.2 mg, 0.022 mmol, 0.1 eq), 2-(di-t-butylphosphino)biphenyl (19.8 mg, 0.066 mmol, 0.3 eq) and sodium t-butoxide (42.5 mg, 0.442 mmol, 2 eq). The vial was purged with N2, sealed and heated in a 105 °C oil bath for 2.25 h. The reaction mixture was cooled to rt, filtered and concentrated in vacuo. The residue was treated with methanol (ca. 1 mL) and the precipitated solid was collected by filtration, washed with methanol and dried to afford 41 mg (37%) of A15 as a tan solid. LC/MS: 499 [M+H]+; HPLC: >95 % at 1.92 min (Primesphere 5 μm C18 column 4.6 x 30 mm, 10-90 % aqueous methanol over 2 min containing 0.2% phosphoric acid, 5 mL/min, monitoring at 254 nm); Η NMR (400 MHz, DMSO-< ): δ 11.55 (br s, 1 H), 9.12 (br s, 1 H), 8.23 (d, 7= 8.2 Hz, 1 H), 7.77-7.54 (m, 6 H), 7.36 (t, 7 = 7.5 Hz, 1 H), 7.28 (br s, 2 H), 4.93 (br s, 2 H), 4.24 (q, 7 = 7.1 Hz, 2 H), 2.50 (coincident with residual DMSO, 3 H), 1.29 (t, 7 = 7.1 Hz, 3 H).
Example A16
Figure imgf000051_0001
A16
A16 was prepared in an manner analogous to example A15 with the exception that in step A 15.1, 4-methylsulfonylbenzylamine hydrochloride was substituted for 4- aminosulfonylbenzylamine hydrochloride. A16 was isolated as a tan solid; LC/MS: 498.28 [M+H]+; HPLC: >90 % at 1.94 min (Primesphere 5 μm C18 column 4.6 x 30 mm, 10-90 % aqueous methanol over 2 min containing 0.2% phosphoric acid, 5 mL/min, monitoring at 254 nm); Η NMR (400 MHz, DMSO- ): δ 11.60 (br s, 1 H), 9.15 (br s, 1 H), 8.22 (d, 7 = 8.1 Hz, 1 H), 7.87 (d, 7 = 8.2 Hz, 2 H), 7.74 (m, 3H), 7.55 (d, 7 = 8.1 Hz, 1 H), 7.37 (m, 1 H), 4.96 (br s, 2 H), 4.24 (q, 7= 7.1 Hz, 2 H), 3.16 (s, 3 H), 2.50 (coincident with residual DMSO, 3 H), 1.28 (t, 7 = 7.1 Hz, 3 H).
Example A17
2-r[4-rrr4-(Aminosulfonyl)phenvnmethvnaminol-8-methoxy-2-quinazolinyl1amino]-
4-methyI-5-thiazolecarboxylic acid, ethyl ester
Figure imgf000052_0001
A17 A17.1: 2,4-Dichloro-8-methoxyquinazoline
Figure imgf000052_0002
A17.1 A17.1 was prepared as described in the literature (Curd, et. al. J. Chem. Soc; 1948, 1759-1766. A17 was prepared in an manner analogous to example A15 starting with quinazoline A17.1 . A 17 was isolated as a tan solid; LC/MS: 529.33 [M+H]+; HPLC: >95 % at 1.34 min (Xterra 5 μm C18 S5 column 4.6 x 30 mm, 10-90 % aqueous methanol over 2 min containing 0.2% phosphoric acid, 5 mL/min, monitoring at 254 nm).
Example A 18
4-[r4-[[[4-(Aminosulfonyl)phenyl1methyl1amino1-6.7-dimethoxy-2- quinazolinyllaminolbenzoic acid, ethyl ester
Figure imgf000052_0003
A18 A18 was prepared in an manner analogous to example Al with the exception that in step Al.l 4-aminosulfonylbenzylamine hydrochloride was substituted for 4- methylsulfonylbenzylamine hydrochloride, and in step A1.2 ethyl 4-aminobenzoate was substituted for ethyl 2-amino-4-methyl-5-thiazole carboxylate. The product was purified by preparatory reverse phase HPLC to yield A18 in 21% yield. LCMS = Ret. Time = 2.87min*, M+ = 538.40. * HPLC conditions used to determine retention times; 4 min gradient 0-100%B in A(A; 0.1% TFA in 90/10 water/methanol; B; 0.1%TFA in 10/90 water/methanol) using a YMC ODS S5 column at 220 nm.
Example A19
4-[[[6,7-Dimethoxy-2-(6-quinolinylamino)-4- q uinazolin yllaminol methyl] benzenesulfonamide
Figure imgf000053_0001
A19
A19 was prepared in an manner analogous to example Al with the exception that in step Al.l 4-aminosulfonylbenzylamine hydrochloride was substituted for 4- methylsulfonylbenzylamine hydrochloride, and in step A 1.2, 6-aminoquinoline was substituted for ethyl 2-amino-4-methyl-5-thiazole carboxylate. The product was purified by preparatory reverse phase HPLC to yield A19. Analytical HPLC ret. time = 1.09 min, [M+H]+ = 517.12. HPLC conditions : phenomenex primesphere 5 u C18 4.6 x 30 mm column, 5 mL/min, 2 min gradient, at 254 nm 0-100%B in A(A; 0.1% TFA in 90/10 water/methanol; B; 0.1%TFA in 10/90 water/methanol)
Example A20 4-πT6,7-Dimethoxy-2-(7-quinazolinylamino)-4- quinazolinyllaminolmethyllbenzenesulfonamide
Figure imgf000054_0001
A20 A20 was prepared in an manner analogous to example Al with the exception that in step Al.l 4-aminosulfonylbenzylamine hydrochloride was substituted for 4- methylsulfonylbenzylamine hydrochloride, and in step A 1.2, 7-aminoquinazoline (prepared according to the literature procedure of Naff, et. al. 7. Am. Chem. Soc. 1951, 73, 1372-1373.) was substituted for ethyl 2-amino-4-methyl-5-thiazole carboxylate. The product was purified by preparatory reverse phase HPLC to yield A20. Analytical HPLC ret. time = 2.17 min, [M+H]+ = 518.30. HPLC conditions: YMC ODS 5μ , 5 mL/min, 4 min gradient, at 254 nm 0-100%B in A(A; 0.1% TFA in 90/10 water/methanol; B; 0.1%TFA in 10/90 water/methanol)
Example Bl
2-r[4-rr[4-(Aminosulfonyl)phenyllmethyllaminolpyrido[2,3-dlpyrimidin-2- yl1aminol-4-methyl-5-thiazolecarboxyIic acid, ethyl ester
Figure imgf000054_0002
O H
Bl Bl.l; 2,4-Dichloropyridor2,3-d1pyrimidine
Figure imgf000055_0001
Bl.l Bl.l was prepared from commercially available 2-aminonicotinic acid following the procedure reported in the literature (Robins, et. al. J. Am. Chem. Soc. 1955, 77, 2256- 2260.)
B1.2: 4-[rr4-(Aminosulfonyl)phenyl1methyl1aminol-2-chIoropyridor2,3- dlpyrimidine
Figure imgf000055_0002
B1.2 A mixture of Bl.l (50 mg, 0.250 mmol, 1 eq), 4-aminosulfonylbenzylamine hydrochloride (58.5 mg, 0.262 mmol, 1.05 eq) and diisopropylethylamine (0.13 mL, 0.75 mmol, 3 eq) in absolute ethanol (1 mL) was stirred at rt for 24 h. The reaction mixture was then cooled in an ice/water bath and the solid was collected by filtration, washed with water and cold ethanol and dried to provide 77 mg (88%) of B1.2 as an off-white solid. LC/MS: 350.31 [M+H]+; HPLC: >95 % at 1.01 min (Xterra 5 μm C18 column 4.6 x 30 mm, 10-90 % aqueous methanol over 2 min containing 0.2% phosphoric acid, 5 mL/min, monitoring at 254 nm).
B1.3: 2-rr4-rr[4-(AminosulfonvI)phenyllmethyllamino1pyrido[2.3-d1pyrimidin-2- yllaminol-4-methyl-5-thiazolecarboxylic acid, ethyl ester
To a mixture of B1.2 (42 mg, 0.120 mmol, 1 eq) and ethyl 2-amino-4- methylthiazole-5-carboxylate (44.7 mg, 0.240 mmol, 2 eq) in N,N-dimethylacetamide (1.2 mL) in a 2-dram vial was added tris(dibenzylideneacetone)dipalladium(0) (11 mg, 0.012 mmol, 0.1 eq), 2-(di-t-butylphosphino)biphenyl (10.7 mg, 0.036 mmol, 0.3 eq) and sodium t-butoxide (23.1 mg, 0.240 mmol, 2 eq). The vial was purged with N2, sealed and heated in a 105 °C oil bath for 3.25 h. The reaction mixture was cooled to rt, filtered and concentrated in vacuo. The residue was treated with methanol (ca. 1 mL) and the precipitated solid was collected by filtration, washed with methanol and dried to afford 32.5 mg (54%) of product as an orange solid. LC/MS: 500.31 [M+H]+; HPLC: >95 % at 1.18 min (Xterra 5 μm C18 column 4.6 x 30 mm, 10-90 % aqueous methanol over 2 min containing 0.2% phosphoric acid, 5 mL/min, monitoring at 254 nm).
Example B2
4-Methyl-2-rr4-[[[4-(methylsulfonyl)phenyl]methvI1amino1pyrido[2.3-d1pyrimidin-2- yllaminol-5-thiazolecarboxylic acid, ethyl ester
Figure imgf000056_0001
B2
B2 was prepared in an manner analogous to example Bl with the exception that in step B1.2, 4-methylsulfonylbenzylamine hydrochloride was substituted for 4- aminosulfonylbenzylamine hydrochloride. B2 was isolated as a yellow solid; LC/MS: 499.33 [M+H]+; HPLC: >85 % at 1.19 min (Xterra 5 μm C18 S5 column 4.6 x 30 mm, 10-90 % aqueous methanol over 2 min containing 0.2% phosphoric acid, 5 mL/min, monitoring at 254 nm).

Claims

We claim:
1. A compound of Formula I
Figure imgf000057_0001
wherein
R1 is H or alkyl; R2is (a) heteroaryl, or heterocyclo, either of which may be optionally substituted with one to three groups T1, T2, T3; or (b) aryl fused to a heteroaryl or heterocyclo ring wherein the combined ring system may be optionally substituted with one to three groups T1, T2, T3; L is (a) -OR4, -C(0)R4, -C(0)OR4, -SR4, -NR3R4, -C(0)NR3R4, -NR3S02R4b halogen, nitro, haloalkyl; or (b) alkyl, aryl, heteroaryl, heterocyclo, or cycloalkyl any of which may be optionally substituted with one to three groups Tla, T2a T3a; Y1, Y2 and Y3 are independently (a) hydrogen, halo, -OR4a, or
(b) alkyl, alkenyl, or alkynyl any of which may be optionally substituted with one to three groups Tlb, T2b or T3b; R3 and R4 are independently H, alkyl, alkenyl, aryl, (aryl)alkyl, heteroaryl,
(heteroaryl)alkyl, cycloalkyl, (cycloalkyl)alkyl, heterocylo or (heterocyclo)alkyl any of which may be optionally substituted with one to three groups Tla, T2a or
T3a. or R3 and R4 together with the nitrogen atom to which they are attached may combine to form a 4 to 8 membered heterocyclo ring optionally substituted with one to three groups Tla, T2a or T3a;
R4a is hydrogen, alkyl, alkenyl, aryl, heteroaryl, (aryl)alkyl, (heteroaryl)alkyl, heterocylo , (heterocyclo)alkyl, cycloalkyl or (cycloalkyl)alkyl any of which may be optionally substituted with one to three groups Tlb, T b or T3b;
R4b is alkyl, alkenyl, aryl, (aryl)alkyl, heteroaryl, (heteroaryl)alkyl, cycloalkyl, (cycloalkyl)alkyl, heterocylo or (heterocyclo)alkyl any of which may be optionally substituted with one to three groups Tla, T2a or T3a;
Z is N or CH; Tl lb, T2 2b, and T3 3b are are each independently
(1) hydrogen or T6, where T6 is
(i) alkyl, (hydroxy)alkyl, (alkoxy)alkyl, alkenyl, alkynyl, cycloalkyl, (cycloalkyl)alkyl, cycloalkenyl, (cycloalkenyl)alkyl, aryl, (aryl)alkyl, heterocyclo, (heterocylco)alkyl, heteroaryl, or (heteroaryl)alkyl; (ii) a group (i) which is itself substituted by one or more of the same or different groups (i); or (iii) a group (i) or (ii) which is independently substituted by one or more (preferably 1 to 3) of the following groups (2) to (13) of the definition of T b, T2"2b and T3"3b,
(2) -OH or -OT6,
(3) -SH or -ST6, (4) -C(0),H, -C(0)tT6, or -0-C(0)T6, where t is 1 or 2;
(5) -S03H, -S(0)tT6, or S(0)tN(T9)T6,
(6) halo,
(7) cyano,
(8) nitro, (9) -T4-NT7T8,
(10) -T4-N(T9)-T5-NT7T8, (11) -T4-N(T10)-T5-T6,
(12) -T4-N(T,0)-T5-H,
(13) oxo, and T5 are each independently
(1) a single bond,
(2) -Tu-S(0)t-T12-,
(3) -Tu-C(0)-T12-,
(4) -TH-C(S)-T12-,
(5) _τu-o-τ12-,
(6) -τn-s-τ12-,
(7) -Tπ-0-C(0)-T12-,
(8) -Tu-C(0)-0-T12-,
(9) -Tn-C(=NT9a)-T12-, or
(10) -Tπ-C(0)-C(0)-T12-
T8, T9, T ' and T10
(1) are each independently hydrogen or a group provided in the definition of T6, or
(2) T7 and T8 may together be alkylene or alkenylene, completing a 3- to 8- membered saturated or unsaturated ring together with the atoms to which they are attached, which ring is unsubstituted or substituted with one or more groups listed in the description of T'"lb, T2"2b and T3 3b, or
(3) T7 or T8, together with T9, may be alkylene or alkenylene completing a 3- to 8-membered saturated or unsaturated ring together with the nitrogen atoms to which they are attached, which ring is unsubstituted or substituted with one or more groups listed in the description of T b, T2"2b and T3"3b, or
(4) T7 and T8 or T9 and T10 together with the nitrogen atom to which they are attached may combine to form a group -N=CT13T14 where T13 and T14 are each independently H or a group provided in the definition of T6; and and T are each independently (1) a single bond,
(2) alkylene, (3) alkenylene, or
(4) alkynylene provided said compound is other than
Figure imgf000060_0001
2. A compound of claim 1 wherein
L is (a) halogen, alkoxy, haloalkyl, -NR3R4, -C(0)OR4, -C(0)NR3R4;
(b) aryl or heteroaryl either of which may be optionally substituted with one or
« m,o«„re« r 1 la, -12a, T1-θa.;
(c) optionally substituted alkyl; Y1, Y2 and Y3 are independently
(a) H, -OR4a or
(b) alkyl or alkenyl either of which may be optionally substituted R1 is H or alkyl;
R2 is (a) heteroaryl optionally substituted with one to three groups T1, T2, T3; or
(b) aryl fused to a heterocyclo ring wherein the combined ring system may be optionally substituted with one to three groups T1, T2, T3; R3 is H or optionally substituted alkyl; R4 is (a) hydrogen;
(b) (aryl)alkyl where the aryl group is optionally independently substituted with one or more groups T1a, T2a, T3a;
(c) (heteroaryl)alky where the heteroaryl group is optionally independently substituted with one or more groups Tla, T2a, T3a; (d) (heterocyclo)alkyl where the heterocyclo group is optionally independently substituted with one or more groups Tla, T2a, T3a;
(e) alkyl optionally independently substituted with one or more groups Tla, T2a,
T3a.
(f) heterocyclo optionally independently substituted with one or more groups Tla, T2a, T3a; or R3 and R4 together with the nitrogen atom to which they are attached combine to form a 4 to 8-membered heterocyclo ring optionally substituted with one to three groups Tla, T2a, T3a.
3. A compound of claim 2 wherein
L is (a) halogen, alkoxy, haloalkyl, -NR3R4, -C(0)OR4, -C(0)NR3R4;
(b) aryl or heteroaryl either of which may be optionally substituted with one or more Tla, T2a, T3a selected from cyano, optionally substituted alkyl, (hydroxy)alkyl, -OH, -OT6, -ST6, -SOtT6, -COtH, -COtT6, -T4NT7T8, -T4N(T10)-T5-T6 or heteroaryl;
(c) alkyl optionally substituted with one or more -OH, -COtH, -COtT6, -T4-NT7T8,
-T4-N(T10)-T5-H, or ; -T4-N(T10)-T5-T6;
J is
(a) H, -OR 4a or (b) alkyl or alkenyl either of which may be optionally substituted with one or more -OH, -OT6, -COtH, or -COtT6;
R1 is H or alkyl;
R2 is
(a) thiazolyl or oxazolyl each optionally substituted with one to three groups T1, T2, T3, selected from alkyl, haloalkyl, halo, heteroaryl, cyano, C(0)tT6,
OT6, or -T4NT7T8; or
(b) aryl fused to a heterocyclo ring wherein the combined ring system may be optionally substituted with one to three groups T1, T2, T3 selected from halo, OH, OT6, alkyl, -COtH, -COtT6, or -C(0)NT7T8; R is H or alkyl optionally substituted with one or more -OH, or -OT ; R4 is
(a) hydrogen;
(b) (aryl)alkyl where the aryl group is optionally independently substituted with one or more groups Tla, T2a, T3a selected from optionally substituted alkyl, halo, cyano, nitro, (hydroxy)alkyl, -OH, -OT6, -ST6, -CO.H, -CO,T6,
-S03H, -SOtT6, -SOtN(T9)(T6), -T^NTV, -T4-N(T10)-T5-T6, heterocyclo, or heteroaryl;
(c) (heteroaryl)alky where the heteroaryl group is optionally independently substituted with one or more groups Tla, T2a, T3a selected from optionally substituted alkyl, halo, cyano, nitro, oxo, (hydroxy)alkyl, -OH, -OT6, -ST6, -COtH, -COtT6, -S03H, -SOtT6, -SOtN(T9)(T6), -T4NT7T8, -T4-N(T10)-T5-T6, heterocyclo, or heteroaryl;
(d) (heterocyclo)alkyl where the heterocyclo group is optionally independently substituted with one or more groups Tla, T2a, T3a selected from optionally substituted alkyl, halo, cyano, nitro, (hydroxy)alkyl, -OH, -OT6, -ST6,
-CO.H, -CO,T6, -S03H, -SOtT6, -T4NT7T8, -T4-N(T10)-T -T6, heterocyclo, or heteroaryl;
(e) alkyl optionally independently substituted with one or more groups Tla, T2a,
T3a selected from -OH, -OT6, -COtH, -COtT6, -T4NT7T8 or -T4-N(T10)-T5-T6;
(f) heterocyclo optionally independently substituted with one or more groups Tla,
T2a, T3a selected from optionally substituted alkyl, optionally substituted aryl, cyano, -OH, -OT6, -COtH, -COtT6, oxo, hydroxy(alkyl), (alkoxy)alkyl, -T4-N(T10)-T5-T6, or -T4-NT7T8; or R3 and R4 together with the nitrogen atom to which they are attached combine to form a heterocylco ring selected from pyrrolidinyl, piperadinyl, piperazinyl, morpholinyl, diazapanyl or l,4-dioxa-8-azaspiro[4.5]decan-8-yl), any of which are optionally independently substituted with one to three groups Tla, T a, T3a selected from optionally substituted alkyl, optionally substituted aryl, cyano, -OH, -OT6, -CO,H, -COtT6, oxo, hydroxy(alkyl), (alkoxy)alkyl, -T4-N(T10)-T5-T6, or -
T4-NT7T8.
4. A compound of claim 1 having formula II
Figure imgf000063_0001
II where: R2 is
Figure imgf000064_0001
wherein: W is O or S;
X1 is NHT8 or OT6;
X2 and X2a are independently hydrogen, halo, OT6, alkyl, or haloalkyl; X3 is heteroaryl (preferably, pyrimidinyl, imidazolyl, oxazolyl, or thiazolyl any of which may be further optionally substituted), cyano, C(0)tT6, or S(0)tNT7T8; and X4 is alkyl, haloalkyl, NHT8 or OT6.
5. A compound of claim 4 wherein R3 is H or alkyl optionally substituted with one or more -OH, or -OT6; R4 is
(a) hydrogen;
(b) (aryl)alkyl where the aryl group is optionally independently substituted with one or more groups Tla, T2a, T3a selected from optionally substituted alkyl, halo, cyano, nitro, (hydroxy)alkyl, -OH, -OT6, -ST6, -COtH, -CO,T6, -S03H, -SO,T6, -SO,N(T9)(T6), -T4NT7T8, -T4-N(T10)-T5-T6, heterocyclo, or heteroaryl where
T4 is a bond, -S02-, or -C(O)-;
T is -S02-, or -alkylene-O-; 6 .
T is alkyl, or cycloalkyl;
T and T are independently H or alkyl; and T9 and T10 are hydrogen;
(c) (heteroaryl)alky where the heteroaryl group is optionally independently substituted with one or more groups Tla, T2a, T3a selected from optionally substituted alkyl, halo, cyano, nitro, oxo, (hydroxy)alkyl, -OH, -OT6, -ST6, -CO,H, -COtT6, -S03H, -SOtT6, -SO,N(T9)(T6), -T4NT7T8,
-T4-N(T10)-T5-T6, heterocyclo, or heteroaryl where
T4 is a bond, -S02-, or -C(O)-;
T5 is -S02-, or -alkylene-O-; T6 is alkyl, or cycloalkyl;
T7 and T8 are independently H or alkyl; and
T9 and T10 are hydrogen;
(d) (heterocyclo)alkyl where the heterocyclo group is optionally independently substituted with one or more groups Tla, T2a, T3a selected from optionally substituted alkyl, halo, cyano, nitro, (hydroxy)alkyl, -OH, -OT6, -ST6,
-COtH, -COtT6, -S03H, -SO,T6, -T4NT7T8, -T4-N(T10)-T5-T6, heterocyclo, or heteroaryl where
T4 is a bond, -S02-, or -C(O)-; T5 is -S02-, or -alkylene-O-;
T6 is alkyl, or cycloalkyl;
T7 and T8 are independently H or alkyl; and
T9 and T10 are hydrogen;
(e) alkyl optionally independently substituted with one or more groups Tla, T2a, T3a selected from -OH, -OT6, -COtH, -COtT6, -T4NT7T8 or
-T4-N(T10)-T5-T6 where
T4 is a bond; T5 is -CO)-; T6 is alkyl;
T7 and T8 are independently H or alkyl; and T,0is hydrogen; (f) heterocyclo optionally independently substituted with one or more groups Tla, T2a, T3a selected from optionally substituted alkyl, optionally substituted aryl, cyano, -OH, -OT6, -COtH, -COtT6, oxo, hydroxy (alkyl), (alkoxy)alkyl, -T4-N(T10)-T5-T6, or -T4-NT7T8, where
T4 is a bond or -C(O)-; T5 is -C(O)-, -S02-, or -alkylene-C(0)0-; T is alkyl, alkoxy, or heteroaryl; T7 and T8 are independently H, alkyl, or cycloalkyl; or T7 and T8 together with the nitrogen atom to which they are attached combine to form a an optionally substituted heterocyclo ring; or R3 and R4 together with the nitrogen atom to which they are attached combine to form a heterocylco ring selected from pyrrolidinyl, piperadinyl, piperazinyl, morpholinyl, diazapanyl or l,4-dioxa-8-azaspiro[4.5]decan-8-yl), any of which are optionally independently substituted with one to three groups Tla, T2a, T3a selected from alkyl optionally substituted with -T4NT7T8, optionally substituted aryl, cyano, -OH, -OT6, -COtH, -COtT6, oxo, hydroxy(alkyl), (alkoxy)alkyl, -T4-N(T10)-T5-T6, or -T4-NT7T8, where
T4 is a bond or -C(O)-; T5 is -C(O)-, -S02-, or -alkylene-C(0)0-; T6 is alkyl, alkoxy, or heteroaryl; T7 and T8 are independently H, alkyl, or cycloalkyl; or T7 and T8 together with the nitrogen atom to which they are attached combine to form a an optionally substituted heterocyclo ring.
6. A compound of claim 1 of the following formula HI
Figure imgf000067_0001
III wherein
R1* is H or alkyl;
R >2* is optionally substituted heteroaryl;
R ,3J* is H or alkyl;
R >4* i •s optionally substituted (aryl)alkyl; and Y , Y and Y are each hydrogen.
7. A compound of claim 6 wherein
R1* is H;
R2* is thiazolyl, oxazolyl, or isoxozolyl (preferably thiazolyl) any of which may be optionally substituted (preferably with one or more alkyl, or alkoxycarbonyl groups); R3* is H; and
R4* is (pheny)alkyl, optionally substituted with one or more -S02R5 where R5 is alkyl, amino, alkylamino or dialkylamino.
8. A compound of claim 7 wherein
Figure imgf000067_0002
where W is O or S, X1 is alkoxy, and X2 is alkyl.
9. A pharmaceutical composition comprising at least one compound of claim 1 together with a pharmaceutically acceptable vehicle or carrier therefor.
10. A method of treating T-cell mediated diseases which comprises administering an effective amount of at least one compound claim 1 to a patient in need thereof.
11. A method of claim 10 wherein said T-cell mediated disorder is transplant rejection.
12. A method of claim 10 wherein said T-cell mediated disorder is graph verses host disease.
13. A method of claim 10 wherein said T-cell mediated disorder is rheumatoid arthritis.
14. A method of claim 10 wherein said T-cell mediated disorder is multiple sclerosis.
15. A method of claim 10 wherein said T-cell mediated disorder is juvenile diabetes.
16. A method of claim 10 wherein said T-cell mediated disorder is asthma.
17. A method of claim 10 wherein said T-cell mediated disorder is inflammatory bowel disease.
18. A method of claim 10 wherein said T-cell mediated disorder is ischemic or reperfusion injury.
19. A method of claim 10 wherein said T-cell mediated disorder is cell proliferation.
20. A method of claim 10 wherein the T-cell mediated disorder is psoriasis.
21. A pharmaceutical composition of claim 9 further comprising at least additional therapeutic agent selected from PDE 4 inhibitors, NSAIDs, COX-2 inhibitors, TNF-α inhibitors, beta-2 agonists, anti-cholinergic agents, and steriods.
PCT/US2002/019130 2001-06-19 2002-06-17 QUINAZOLINE AND PYRIDO[2,3-d]PYRIMIDINE INHIBITORS OF PHOSPHODIESTERASE (PDE) 7 WO2002102315A2 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
EP02742138A EP1404337A4 (en) 2001-06-19 2002-06-17 QUINAZOLINE AND PYRIDO 2,3-d|PYRIMIDINE INHIBITORS OF PHOSPHODIESTERASE (PDE) 7
CA002450724A CA2450724A1 (en) 2001-06-19 2002-06-17 Quinazoline and pyrido¬2,3-d|pyrimidine inhibitors of phosphodiesterase (pde) 7
JP2003504904A JP2005506961A (en) 2001-06-19 2002-06-17 Quinazoline and pyrido [2,3-d] pyrimidine derivative inhibitors for phosphodiesterase 7

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US29928701P 2001-06-19 2001-06-19
US60/299,287 2001-06-19
US36875202P 2002-03-29 2002-03-29
US60/368,752 2002-03-29

Publications (2)

Publication Number Publication Date
WO2002102315A2 true WO2002102315A2 (en) 2002-12-27
WO2002102315A3 WO2002102315A3 (en) 2003-11-20

Family

ID=26971149

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/US2002/019130 WO2002102315A2 (en) 2001-06-19 2002-06-17 QUINAZOLINE AND PYRIDO[2,3-d]PYRIMIDINE INHIBITORS OF PHOSPHODIESTERASE (PDE) 7
PCT/US2002/019126 WO2002102314A2 (en) 2001-06-19 2002-06-17 Purine inhibitors of phosphodiesterase (pde) 7

Family Applications After (1)

Application Number Title Priority Date Filing Date
PCT/US2002/019126 WO2002102314A2 (en) 2001-06-19 2002-06-17 Purine inhibitors of phosphodiesterase (pde) 7

Country Status (7)

Country Link
US (3) US7022849B2 (en)
EP (2) EP1404337A4 (en)
JP (2) JP2005500295A (en)
CA (2) CA2450724A1 (en)
HU (1) HUP0600228A2 (en)
PE (1) PE20030008A1 (en)
WO (2) WO2002102315A2 (en)

Cited By (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005061497A1 (en) * 2003-12-16 2005-07-07 Pfizer Products Inc. Pyrido[2,3-d]pyrimidine-2,4-diamines as pde 2 inhibitors
WO2005082883A2 (en) * 2004-02-18 2005-09-09 Pfizer Products Inc. Tetrahydroisoquinolinyl derivatives of quinazoline and isoquinoline
US7423042B2 (en) 2005-03-25 2008-09-09 Glaxo Group Limited Compounds
WO2008119057A2 (en) 2007-03-27 2008-10-02 Omeros Corporation The use of pde7 inhibitors for the treatment of movement disorders
US7479558B2 (en) 2005-03-25 2009-01-20 Glaxo Group Limited Process for preparing pyrido[2,3-d]pyrimidin-7-one and 3,4-dihydropyrimido[4,5-d]pyrimidin-2(1H)-one derivatives
US7566786B2 (en) 2003-05-21 2009-07-28 Glaxo Group Limited Quinoline derivatives as phosphodiesterase inhibitors
US7629462B2 (en) 2000-03-02 2009-12-08 Smithkline Beecham Corporation Tetrasubstituted pyrimidine compounds as chemical intermediates
US7629350B2 (en) 2002-04-19 2009-12-08 Smithkline Beecham Corporation Compounds
US7674789B2 (en) 2005-03-25 2010-03-09 Glaxo Group Limited Compounds
US7678801B2 (en) 2005-03-25 2010-03-16 Glaxo Group Limited Compounds
US7713972B2 (en) 2003-06-13 2010-05-11 Asubio Pharma Co., Ltd. Imidazotriazinone derivatives as PDE 7 (phosphodiesterase 7) inhibitors
US7759486B2 (en) 2000-10-23 2010-07-20 Glaxosmithkline Llc 2,4,5-Trisubstituted pyrimidine compounds
US7767687B2 (en) 2004-12-13 2010-08-03 Biogen Idec Ma Inc. Pyrido pyrimidinones, dihydro pyrimido pyrimidinones and pteridinones useful as RAF kinase inhibitors
WO2010138577A1 (en) * 2009-05-29 2010-12-02 Merck Sharp & Dohme Corp. Radiolabeled pde10 inhibitors
US7932250B2 (en) 2004-07-01 2011-04-26 Daiichi Sankyo Company, Limited Thienopyrazole derivative having PDE7 inhibitory activity
US7943624B2 (en) 2003-06-13 2011-05-17 Asubio Pharma Co. Ltd. Pyridinylpyrazolopyrimidinone derivatives as PDE 7 inhibitors
WO2012064667A2 (en) 2010-11-08 2012-05-18 Omeros Corporation Treatment of addiction and impulse-control disorders using pde7 inhibitors
US8222263B2 (en) 2007-03-14 2012-07-17 Exelixis Patent Company Llc Inhibitors of hedgehog pathway
US8637528B2 (en) 2007-03-27 2014-01-28 Omeros Corporation Use of PDE7 inhibitors for the treatment of movement disorders
US9220715B2 (en) 2010-11-08 2015-12-29 Omeros Corporation Treatment of addiction and impulse-control disorders using PDE7 inhibitors
WO2024038089A1 (en) 2022-08-18 2024-02-22 Mitodicure Gmbh Use of a therapeutic agent with phosphodiesterase-7 inhibitory activity for the treatment and prevention of diseases associated with chronic fatigue, exhaustion and/or exertional intolerance

Families Citing this family (60)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ATE448532T1 (en) * 1996-09-04 2009-11-15 Intertrust Tech Corp RELIABLE INFRASTRUCTURE SUPPORT SYSTEMS, METHODS AND TECHNIQUES FOR SECURE ELECTRONIC COMMERCIAL, ELECTRONIC TRANSACTIONS, TRADE PROCESS CONTROL AND AUTOMATION, DISTRIBUTED PROCESSING AND RIGHTS MANAGEMENT
US6455734B1 (en) * 2000-08-09 2002-09-24 Magnesium Diagnostics, Inc. Antagonists of the magnesium binding defect as therapeutic agents and methods for treatment of abnormal physiological states
US7115589B2 (en) * 1999-12-17 2006-10-03 Ariad Pharmaceuticals, Inc. Purine derivatives
EP1440072A4 (en) 2001-10-30 2005-02-02 Conforma Therapeutic Corp Purine analogs having hsp90-inhibiting activity
UA80871C2 (en) * 2003-04-29 2007-11-12 Pfizer Ltd 5,7-diaminopyrazolo[4,3-d]pyrimidines useful in the treatment of hypertension
EP1620437B1 (en) * 2003-04-29 2009-06-17 Pfizer Limited 5,7-diaminopyrazolo¬4,3-d pyrimidines useful in the traetment of hypertension
US20050065171A1 (en) * 2003-06-25 2005-03-24 Shakespeare William C. Substituted purine derivatives
AR045047A1 (en) * 2003-07-11 2005-10-12 Arena Pharm Inc ARILO AND HETEROARILO DERIVATIVES TRISUSTITUIDOS AS MODULATORS OF METABOLISM AND PROFILAXIS AND TREATMENT OF DISORDERS RELATED TO THEMSELVES
JP2007505933A (en) 2003-09-18 2007-03-15 コンフォーマ・セラピューティクス・コーポレイション Novel heterocyclic compounds as HSP90 inhibitors
WO2005030131A2 (en) * 2003-09-23 2005-04-07 Replidyne, Inc Bis-quinazoline compounds for the treatment of bacterial infections
CN101579529A (en) * 2003-09-29 2009-11-18 托皮根药品公司 Oligonucleotide compositions and method for treating disease including inflammatory conditions
GB0327323D0 (en) * 2003-11-24 2003-12-31 Pfizer Ltd Novel pharmaceuticals
US7572799B2 (en) 2003-11-24 2009-08-11 Pfizer Inc Pyrazolo[4,3-d]pyrimidines as Phosphodiesterase Inhibitors
GB0407723D0 (en) * 2004-04-05 2004-05-12 Novartis Ag Organic compounds
JP2007532526A (en) 2004-04-07 2007-11-15 ファイザー・インク Pyrazolo [4,3-d] pyrimidines
MX2007013595A (en) * 2005-05-04 2008-01-24 Renovis Inc Fused heterocyclic compounds, and compositions and uses thereof.
EP2258358A3 (en) 2005-08-26 2011-09-07 Braincells, Inc. Neurogenesis with acetylcholinesterase inhibitor
JP2009506069A (en) 2005-08-26 2009-02-12 ブレインセルス,インコーポレイティド Neurogenesis through modulation of muscarinic receptors
US20070105878A1 (en) * 2005-10-03 2007-05-10 Andrew Reaume Purine formulations and methods for managing disorders
JP2009512711A (en) 2005-10-21 2009-03-26 ブレインセルス,インコーポレイティド Regulation of neurogenesis by PDE inhibition
AU2006308889A1 (en) 2005-10-31 2007-05-10 Braincells, Inc. GABA receptor mediated modulation of neurogenesis
US8067433B2 (en) 2005-11-09 2011-11-29 Zalicus Inc. Methods, compositions, and kits for the treatment of ophthalmic disorders
CA2633035C (en) * 2005-12-15 2016-05-10 Rigel Pharmaceuticals, Inc. Kinase inhibitors and their uses
US20100216734A1 (en) * 2006-03-08 2010-08-26 Braincells, Inc. Modulation of neurogenesis by nootropic agents
AU2007223036A1 (en) * 2006-03-08 2007-09-13 Braincells, Inc. Modulation of neurogenesis by nootropic agents
US20100009983A1 (en) * 2006-05-09 2010-01-14 Braincells, Inc. 5 ht receptor mediated neurogenesis
AU2007249399A1 (en) 2006-05-09 2007-11-22 Braincells, Inc. Neurogenesis by modulating angiotensin
US7678808B2 (en) 2006-05-09 2010-03-16 Braincells, Inc. 5 HT receptor mediated neurogenesis
WO2007134451A1 (en) * 2006-05-19 2007-11-29 Topigen Pharmaceuticals Inc. Oligonucleotides affecting expression of phosphodiesterases
US7998971B2 (en) 2006-09-08 2011-08-16 Braincells Inc. Combinations containing a 4-acylaminopyridine derivative
EP2066355A2 (en) * 2006-09-19 2009-06-10 Braincells, Inc. Combination comprising a peroxisome proliferator activated receptor agent and a second neurogenic agent for treating a nervous system disorder, increasing neurodifferentiation and increasing neurogenesis
US20100184806A1 (en) 2006-09-19 2010-07-22 Braincells, Inc. Modulation of neurogenesis by ppar agents
ES2308916B1 (en) * 2007-03-22 2009-10-29 Consejo Superior De Investigaciones Cientificas DUAL INHIBITOR COMPOUND OF PDE7 AND / OR PDE4 ENZYMES, PHARMACEUTICAL COMPOSITIONS AND THEIR APPLICATIONS.
AU2008305294B2 (en) 2007-09-21 2012-11-29 Array Biopharma Inc. Pyridin-2 -yl-amino-1, 2, 4 -thiadiazole derivatives as glucokinase activators for the treatment of diabetes mellitus
ES2432821T3 (en) * 2008-07-31 2013-12-05 Genentech, Inc. Pyrimidine compounds, compositions and methods of use
US8703778B2 (en) 2008-09-26 2014-04-22 Intellikine Llc Heterocyclic kinase inhibitors
WO2010099217A1 (en) 2009-02-25 2010-09-02 Braincells, Inc. Modulation of neurogenesis using d-cycloserine combinations
US20100331305A1 (en) * 2009-06-24 2010-12-30 Genentech, Inc. Oxo-heterocycle fused pyrimidine compounds, compositions and methods of use
US8288381B2 (en) * 2009-11-12 2012-10-16 Genentech, Inc. N-9 substituted purine compounds, compositions and methods of use
CA2780018C (en) * 2009-11-12 2015-10-20 F. Hoffmann-La Roche Ag N-7 substituted purine and pyrazolopyrimidine compounds, compositions and methods of use
MY159257A (en) 2009-12-23 2016-12-30 Takeda Pharmaceuticals Co Fused heteroaromatic pyrrolidinones as syk inhibitors
CN102068434B (en) * 2011-01-25 2012-08-22 林治华 Application of 4-(cyclohexyl)-aminoquinazoline compounds
JP6026525B2 (en) 2011-06-22 2016-11-16 武田薬品工業株式会社 Substituted 6-aza-isoindoline-1-one derivatives
EP2804603A1 (en) 2012-01-10 2014-11-26 President and Fellows of Harvard College Beta-cell replication promoting compounds and methods of their use
JPWO2013146963A1 (en) 2012-03-28 2015-12-14 武田薬品工業株式会社 Heterocyclic compounds
WO2014123882A1 (en) * 2013-02-07 2014-08-14 Merck Sharp & Dohme Corp. 2,6,7 substituted purines as hdm2 inhibitors
JP6669499B2 (en) 2013-02-15 2020-03-18 カラ ファーマシューティカルズ インコーポレイテッド Therapeutic compounds
JP2016510000A (en) 2013-02-20 2016-04-04 カラ ファーマシューティカルズ インコーポレイテッド Therapeutic compounds and uses thereof
US9688688B2 (en) 2013-02-20 2017-06-27 Kala Pharmaceuticals, Inc. Crystalline forms of 4-((4-((4-fluoro-2-methyl-1H-indol-5-yl)oxy)-6-methoxyquinazolin-7-yl)oxy)-1-(2-oxa-7-azaspiro[3.5]nonan-7-yl)butan-1-one and uses thereof
KR20160099084A (en) 2013-11-01 2016-08-19 칼라 파마슈티컬스, 인크. Crystalline forms of therapeutic compounds and uses thereof
US9890173B2 (en) 2013-11-01 2018-02-13 Kala Pharmaceuticals, Inc. Crystalline forms of therapeutic compounds and uses thereof
WO2016015597A1 (en) 2014-07-26 2016-02-04 Sunshine Lake Pharma Co., Ltd. Compounds as cdk small-molecule inhibitors and uses thereof
US20190112317A1 (en) 2015-10-05 2019-04-18 The Trustees Of Columbia University In The City Of New York Activators of autophagic flux and phospholipase d and clearance of protein aggregates including tau and treatment of proteinopathies
CA3036340A1 (en) 2016-09-08 2018-03-15 Kala Pharmaceuticals, Inc. Crystalline forms of therapeutic compounds and uses thereof
AU2017324251A1 (en) 2016-09-08 2019-03-21 Kala Pharmaceuticals, Inc. Crystalline forms of therapeutic compounds and uses thereof
US10253036B2 (en) 2016-09-08 2019-04-09 Kala Pharmaceuticals, Inc. Crystalline forms of therapeutic compounds and uses thereof
PL3966207T3 (en) 2019-05-10 2024-03-04 Deciphera Pharmaceuticals, Llc Phenylaminopyrimidine amide autophagy inhibitors and methods of use thereof
SG11202112171XA (en) 2019-05-10 2021-12-30 Deciphera Pharmaceuticals Llc Heteroarylaminopyrimidine amide autophagy inhibitors and methods of use thereof
JP2022536540A (en) 2019-06-17 2022-08-17 デシフェラ・ファーマシューティカルズ,エルエルシー Aminopyrimidine amide autophagy inhibitors and methods of use thereof
JP2023542921A (en) 2020-09-21 2023-10-12 ランドス バイオファーマ インコーポレイテッド NLRX1 ligand

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5439895A (en) * 1992-07-15 1995-08-08 Ono Pharmaceutical Co., Ltd. 4-aminoquinazoline derivatives
WO1997020822A1 (en) * 1995-12-01 1997-06-12 Novartis Ag Quinazolin-2,4-diazirines as npy receptor antagonist
US5693652A (en) * 1991-09-30 1997-12-02 Eisai Co., Ltd. Benzimidazoles for ischemic heart conditions

Family Cites Families (44)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BE487239A (en) * 1948-02-09
DE2359536C2 (en) * 1972-12-08 1984-08-02 Takeda Chemical Industries, Ltd., Osaka 2,6-diaminonebularin derivatives
ES2009145A6 (en) * 1987-07-07 1989-09-01 Guillermo Tena Lab Morrith S A Prepn. of amino derivs. of di:hydro oxo pyrido-pyrimidine
US5240938A (en) 1991-02-13 1993-08-31 Merck & Co., Inc. Angiotensin II antagonists incorporating a substituted pyridoimidazolyl ring
DE4008858A1 (en) 1990-03-20 1991-09-26 Hoechst Ag New substd. purine antiviral cpds.
CA2044806A1 (en) 1990-07-18 1992-01-19 Roland Jaunin Purine derivatives
ATE124694T1 (en) 1990-11-06 1995-07-15 Pfizer CHINAZOLINE DERIVATIVES TO IMPROVE THE ANTITUMOR EFFECT.
US5177074A (en) 1991-03-26 1993-01-05 Merck & Co., Inc. Angiotensin ii antagonists incorporating a substituted thiophene or furan
JP3225545B2 (en) 1991-09-18 2001-11-05 味の素株式会社 Method for producing acyclic nucleosides
US5792868A (en) 1991-09-18 1998-08-11 Ajinomoto Co., Inc. Process for producing acyclic nucleosides and process for separating purine nucleosides
GB9125001D0 (en) 1991-11-25 1992-01-22 Ici Plc Heterocyclic compounds
EP0626964A1 (en) 1992-02-19 1994-12-07 Pfizer Inc. Heterocyclic compounds for enhancing antitumor activity
US5290801A (en) 1992-05-29 1994-03-01 The Du Pont Merck Pharmaceutical Company Benzimidazoles for the treatment of atherosclerosis
TW251284B (en) 1992-11-02 1995-07-11 Pfizer
GB9301000D0 (en) * 1993-01-20 1993-03-10 Glaxo Group Ltd Chemical compounds
CA2119315A1 (en) 1993-03-18 1994-09-19 Tsujiaki Hata Nucleoside derivatives and anti-herpes composition
JPH06321784A (en) 1993-03-18 1994-11-22 Nippon Chemiphar Co Ltd Anti-human immunodeficiency viral agent
US5374638A (en) 1993-03-19 1994-12-20 Merck & Co., Inc. Six membered ring fused imidazoles substituted with phenoxyphenylacetic acid derivatives used to treat asthma
JP3396924B2 (en) 1993-09-07 2003-04-14 日本曹達株式会社 Activated sludge bulking inhibitor
CA2185699A1 (en) 1994-04-04 1995-10-12 William R. Freeman Use of phosphonylmethoxyalkyl nucleosides for the treatment of raised intraocular pressure
US5646155A (en) * 1994-05-12 1997-07-08 University Of Massachusetts Medical Center Drugs to prevent recurrent herpes virus infections
JP3906488B2 (en) 1995-02-21 2007-04-18 味の素株式会社 Method for producing purine derivatives
DE69627195T2 (en) 1995-11-01 2004-01-29 Novartis Ag PURINE DERIVATIVES AND METHOD FOR THEIR PRODUCTION
WO1997020821A1 (en) 1995-12-01 1997-06-12 Novartis Ag Heteroaryl derivatives
US6107300A (en) 1996-03-27 2000-08-22 Dupont Pharmaceuticals Arylamino fused pyrimidines
US5753635A (en) 1996-08-16 1998-05-19 Berlex Laboratories, Inc. Purine derivatives and their use as anti-coagulants
EP0827960A1 (en) 1996-09-10 1998-03-11 Ajinomoto Co., Inc. Process for producing purine derivatives
CN1252054C (en) 1996-09-25 2006-04-19 曾尼卡有限公司 Qinoline derivatives inhibiting effect of growth factors such as VEGF
US5874438A (en) * 1996-10-11 1999-02-23 Bayer Aktiengesellschaft 2,2'-bridged bis-2,4-diaminoquinazolines
US5739127A (en) * 1996-11-08 1998-04-14 Bayer Aktiengesellschaft 2,4'-bridged bis-2,4-diaminoquinazolines
TW528755B (en) * 1996-12-24 2003-04-21 Glaxo Group Ltd 2-(purin-9-yl)-tetrahydrofuran-3,4-diol derivatives
CA2293400A1 (en) 1997-06-13 1998-12-17 Gerald Mcmahon Novel heteroaryl compounds for the modulation of protein tyrosine enzyme related cellular signal transduction
US6339089B2 (en) * 1997-08-13 2002-01-15 Fujirebio Inc. Pyrimidine nucleus-containing compound and a medicament containing the same for a blood oxygen partial pressure amelioration, and a method for preparing the same
CN1130363C (en) 1997-11-12 2003-12-10 三菱化学株式会社 Purine derivatives and medicine containing the same as the active ingredient
GB9802251D0 (en) 1998-02-03 1998-04-01 Ciba Geigy Ag Organic compounds
US6200976B1 (en) * 1998-04-17 2001-03-13 Boehringer Ingelheim Pharma Kg Antithrombotic quinoxazolines
AU2217200A (en) * 1998-12-23 2000-07-12 Neurogen Corporation 2-amino-9-alkylpurines: gaba brain receptor ligands
AU4589800A (en) 1999-05-05 2000-11-21 Darwin Discovery Limited 9-(1,2,3,4-tetrahydronaphthalen-1-yl)-1,9-dihydropurin-6-one derivatives as pde7inhibitors
EE200200405A (en) * 2000-01-25 2003-12-15 Warner-Lambert Company Pyrido [2,3-d] pyrimidine-2,7-diamines as kinase inhibitors
BR0017075A (en) 2000-01-27 2002-11-05 Warner Lambert Co Pyridopyrimidinone derivatives for the treatment of neurodegenerative disease
AU2001240150A1 (en) 2000-03-13 2001-09-24 Chemrx Advanced Technologies, Inc. Quinazoline synthesis
MXPA03002411A (en) 2000-09-20 2003-06-19 Merck Patent Gmbh 4-amino-quinazolines.
PL359920A1 (en) 2000-09-20 2004-09-06 Merck Patent Gmbh 4-amino-quinazolines
US20020173524A1 (en) 2000-10-11 2002-11-21 Tularik Inc. Modulation of CCR4 function

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5693652A (en) * 1991-09-30 1997-12-02 Eisai Co., Ltd. Benzimidazoles for ischemic heart conditions
US5439895A (en) * 1992-07-15 1995-08-08 Ono Pharmaceutical Co., Ltd. 4-aminoquinazoline derivatives
WO1997020822A1 (en) * 1995-12-01 1997-06-12 Novartis Ag Quinazolin-2,4-diazirines as npy receptor antagonist

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP1404337A2 *

Cited By (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7700768B2 (en) 2000-03-02 2010-04-20 Glaxosmithkline Llc Compounds
US7629462B2 (en) 2000-03-02 2009-12-08 Smithkline Beecham Corporation Tetrasubstituted pyrimidine compounds as chemical intermediates
US7759486B2 (en) 2000-10-23 2010-07-20 Glaxosmithkline Llc 2,4,5-Trisubstituted pyrimidine compounds
US7629350B2 (en) 2002-04-19 2009-12-08 Smithkline Beecham Corporation Compounds
US7566786B2 (en) 2003-05-21 2009-07-28 Glaxo Group Limited Quinoline derivatives as phosphodiesterase inhibitors
US7572915B2 (en) 2003-05-21 2009-08-11 Glaxo Group Limited Quinoline derivatives as phosphodiesterase inhibitors
US7713972B2 (en) 2003-06-13 2010-05-11 Asubio Pharma Co., Ltd. Imidazotriazinone derivatives as PDE 7 (phosphodiesterase 7) inhibitors
US7943624B2 (en) 2003-06-13 2011-05-17 Asubio Pharma Co. Ltd. Pyridinylpyrazolopyrimidinone derivatives as PDE 7 inhibitors
KR100816179B1 (en) * 2003-12-16 2008-03-28 화이자 프로덕츠 인코포레이티드 Pyrido[2,3-d]pyrimidine-2,4-diamines as pde 2 inhibitors
WO2005061497A1 (en) * 2003-12-16 2005-07-07 Pfizer Products Inc. Pyrido[2,3-d]pyrimidine-2,4-diamines as pde 2 inhibitors
CN100439366C (en) * 2003-12-16 2008-12-03 辉瑞产品公司 Pyrido[2,3-d]pyrimidine-2,4-diamines as PDE2 inhibitors
EA010424B1 (en) * 2003-12-16 2008-08-29 Пфайзер Продактс Инк. Pyrido[2,3-d]pyrimidine-2,4-diamines as pde 2 inhibitors
US7268142B2 (en) 2004-02-18 2007-09-11 Pfizer Inc. Tetrahydroisoquinolinyl derivatives of quinazoline and isoquinoline
WO2005082883A3 (en) * 2004-02-18 2007-04-26 Pfizer Prod Inc Tetrahydroisoquinolinyl derivatives of quinazoline and isoquinoline
WO2005082883A2 (en) * 2004-02-18 2005-09-09 Pfizer Products Inc. Tetrahydroisoquinolinyl derivatives of quinazoline and isoquinoline
EP2433943A1 (en) 2004-07-01 2012-03-28 Daiichi Sankyo Company, Limited Thienopyrazole derivatives having PDE7 inhibitory activity
US8901315B2 (en) 2004-07-01 2014-12-02 Daiichi Sankyo Company, Limited Thienopyrazole derivative having PDE7 inhibitory activity
US7932250B2 (en) 2004-07-01 2011-04-26 Daiichi Sankyo Company, Limited Thienopyrazole derivative having PDE7 inhibitory activity
US7767687B2 (en) 2004-12-13 2010-08-03 Biogen Idec Ma Inc. Pyrido pyrimidinones, dihydro pyrimido pyrimidinones and pteridinones useful as RAF kinase inhibitors
US7479558B2 (en) 2005-03-25 2009-01-20 Glaxo Group Limited Process for preparing pyrido[2,3-d]pyrimidin-7-one and 3,4-dihydropyrimido[4,5-d]pyrimidin-2(1H)-one derivatives
US7678801B2 (en) 2005-03-25 2010-03-16 Glaxo Group Limited Compounds
US7674789B2 (en) 2005-03-25 2010-03-09 Glaxo Group Limited Compounds
US7423042B2 (en) 2005-03-25 2008-09-09 Glaxo Group Limited Compounds
US8222263B2 (en) 2007-03-14 2012-07-17 Exelixis Patent Company Llc Inhibitors of hedgehog pathway
US8754092B2 (en) 2007-03-14 2014-06-17 Exelixis Patent Company Llc Inhibitors of the hedgehog pathway
US8796294B2 (en) 2007-03-14 2014-08-05 Exelixis Patent Company Llc Inhibitors of the hedgehog pathway
US9119822B2 (en) 2007-03-27 2015-09-01 Omeros Corporation Use of PDE7 inhibitors for the treatment of movement disorders
WO2008119057A2 (en) 2007-03-27 2008-10-02 Omeros Corporation The use of pde7 inhibitors for the treatment of movement disorders
US8637528B2 (en) 2007-03-27 2014-01-28 Omeros Corporation Use of PDE7 inhibitors for the treatment of movement disorders
WO2010138577A1 (en) * 2009-05-29 2010-12-02 Merck Sharp & Dohme Corp. Radiolabeled pde10 inhibitors
WO2012064667A2 (en) 2010-11-08 2012-05-18 Omeros Corporation Treatment of addiction and impulse-control disorders using pde7 inhibitors
US9220715B2 (en) 2010-11-08 2015-12-29 Omeros Corporation Treatment of addiction and impulse-control disorders using PDE7 inhibitors
US11207275B2 (en) 2010-11-08 2021-12-28 Omeros Corporation Treatment of addiction and impulse-control disorders using PDE7 inhibitors
US11464785B2 (en) 2010-11-08 2022-10-11 Omeros Corporation Treatment of addiction and impulse-control disorders using PDE7 inhibitors
EP4275752A2 (en) 2010-11-08 2023-11-15 Omeros Corporation Treatment of addiction and impulse-control disorders using pde7 inhibitors
WO2024038089A1 (en) 2022-08-18 2024-02-22 Mitodicure Gmbh Use of a therapeutic agent with phosphodiesterase-7 inhibitory activity for the treatment and prevention of diseases associated with chronic fatigue, exhaustion and/or exertional intolerance

Also Published As

Publication number Publication date
US6838559B2 (en) 2005-01-04
EP1404337A4 (en) 2006-04-12
EP1423390A4 (en) 2004-11-17
US20030100571A1 (en) 2003-05-29
CA2450724A1 (en) 2002-12-27
US20030092721A1 (en) 2003-05-15
CA2450936A1 (en) 2002-12-27
US20060116516A1 (en) 2006-06-01
US7022849B2 (en) 2006-04-04
WO2002102314A2 (en) 2002-12-27
WO2002102315A3 (en) 2003-11-20
EP1423390A2 (en) 2004-06-02
US7601836B2 (en) 2009-10-13
JP2005500295A (en) 2005-01-06
HUP0600228A2 (en) 2006-07-28
WO2002102314A3 (en) 2004-04-01
EP1404337A2 (en) 2004-04-07
PE20030008A1 (en) 2003-01-22
JP2005506961A (en) 2005-03-10

Similar Documents

Publication Publication Date Title
US7022849B2 (en) Quinazoline and pyrido[2,3-d]pyrimidine inhibitors of phosphodiesterase (PDE) 7
US20030092908A1 (en) Fused heterocyclic inhibitors of phosphodiesterase (PDE) 7
WO2002102313A2 (en) Pyrimidine inhibitors of phosphodiesterase (pde) 7
US7981881B2 (en) Fused heterocyclic compounds and use thereof
US7105667B2 (en) Fused heterocyclic compounds and use thereof
EP1066286B1 (en) Heterocyclo-substituted imidazopyrazine protein tyrosine kinase inhibitors
US6635626B1 (en) Imidazoquinoxaline protein tyrosine kinase inhibitors
US20040132750A1 (en) Acyl guanidine compounds and use thereof
AU2002315192A1 (en) Quinazoline and pyrido[2,3-d]pyrimidine inhibitors of phosphodiesterase (PDE) 7
AU2002303620A1 (en) Fused heterocyclic inhibitors of phosphodiesterase (PDE) 7
AU2002315190A1 (en) Purine inhibitors of phosphodiesterase (PDE) 7
AU2002344743A1 (en) Pyrimidine inhibitors of phosphodiesterase (PDE) 7
AU2002256419A1 (en) Dual inhibitors of PDE 7 and PDE 4

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

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

AL Designated countries for regional patents

Kind code of ref document: A2

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

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 2450724

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2002315192

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2003504904

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 2002742138

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2002742138

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWW Wipo information: withdrawn in national office

Ref document number: 2002742138

Country of ref document: EP