WO2004087053A9 - Dipeptidyl peptidase inhibitors - Google Patents

Dipeptidyl peptidase inhibitors

Info

Publication number
WO2004087053A9
WO2004087053A9 PCT/US2004/009217 US2004009217W WO2004087053A9 WO 2004087053 A9 WO2004087053 A9 WO 2004087053A9 US 2004009217 W US2004009217 W US 2004009217W WO 2004087053 A9 WO2004087053 A9 WO 2004087053A9
Authority
WO
WIPO (PCT)
Prior art keywords
group
compound according
ring
unsubstituted
phenyl
Prior art date
Application number
PCT/US2004/009217
Other languages
French (fr)
Other versions
WO2004087053A3 (en
WO2004087053A2 (en
Inventor
Jun Feng
Stephen L Gwaltney
Stephen W Kaldor
Jeffrey A Stafford
Michael B Wallace
Zhiyuan Zhang
Original Assignee
Syrrx Inc
Jun Feng
Stephen L Gwaltney
Stephen W Kaldor
Jeffrey A Stafford
Michael B Wallace
Zhiyuan Zhang
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 Syrrx Inc, Jun Feng, Stephen L Gwaltney, Stephen W Kaldor, Jeffrey A Stafford, Michael B Wallace, Zhiyuan Zhang filed Critical Syrrx Inc
Priority to EP04758366A priority Critical patent/EP1608317B1/en
Priority to CA002518465A priority patent/CA2518465A1/en
Priority to JP2006509315A priority patent/JP4887139B2/en
Publication of WO2004087053A2 publication Critical patent/WO2004087053A2/en
Publication of WO2004087053A9 publication Critical patent/WO2004087053A9/en
Publication of WO2004087053A3 publication Critical patent/WO2004087053A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/70Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings condensed with carbocyclic rings or ring systems
    • C07D239/72Quinazolines; Hydrogenated quinazolines
    • C07D239/95Quinazolines; Hydrogenated quinazolines with hetero atoms directly attached in positions 2 and 4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/08Drugs for genital or sexual disorders; Contraceptives for gonadal disorders or for enhancing fertility, e.g. inducers of ovulation or of spermatogenesis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/10Drugs for genital or sexual disorders; Contraceptives for impotence
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • 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
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • 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
    • 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
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • 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
    • C07D403/04Heterocyclic 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 directly linked by a ring-member-to-ring-member bond
    • 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
    • C07D473/18Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6 one oxygen and one nitrogen atom, e.g. guanine
    • 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

Definitions

  • the invention relates to compounds that may be used to inhibit dipeptidyl peptidases as well as compositions of matter and kits comprising these compounds.
  • the present invention also relates to methods for inhibiting dipeptidyl peptidases as well as treatment methods using compounds according to the present invention.
  • Dipeptidyl Peptidase IN (IUBMB Enzyme Nomenclature EC.3.4.14.5) is a type H membrane protein that has been referred to in the literature by a wide a variety of names including DPP4, DP4, DAPTV, FAP ⁇ , adenosine deaminase complexing protein 2, adenosine deaminase binding protein (ADAbp), dipeptidyl aminopeptidase IV; Xaa-Pro-dipeptidyl- aminopeptidase; Gly-Pro naphthylamidase; postproline dipeptidyl aminopeptidase IV; lymphocyte antigen CD26; glycoprotein GP110; dipeptidyl peptidase IV; glycylproline aminopeptidase; glycylproline aminopeptidase; X-prolyl dipeptidyl aminopeptidase; pep X; leukocyte antigen CD26; glyc
  • DPP-IV is a non-classical serine aminodipeptidase that removes Xaa-Pro dipeptides from the amino terminus (N-terminus) of polypeptides and proteins. DPP-IV dependent slow release of dipeptides of the type X-Gly or X-Ser has also been reported for some naturally occurring peptides.
  • DPP-IV is constitutively expressed on epithelial and endothelial cells of a variety of different tissues (intestine, liver, lung, kidney and placenta), and is also found in body fluids. DPP-IV is also expressed on circulating T-lymphocytes and has been shown to be synonymous with the cell-surface antigen, CD-26. DPP-IV has been implicated in a number of disease states, some of which are discussed below.
  • DPP-IV is responsible for the metabolic cleavage of certain endogenous peptides (GLP-1 (7-36), glucagon) in vivo and has demonstrated proteolytic activity against a variety of other peptides (GHRH, NPY, GLP-2, VIP) in vitro.
  • GLP-1 (7-36) is a 29 amino-acid peptide derived by post-translational processing of proglucagon in the small intestine; GLP-1 (7-36) has multiple actions in vivo including the stimulation of insulin secretion, inhibition of glucagon secretion, the promotion of satiety, and the slowing of gastric emptying.
  • DPP-IV has been shown to be the primary degrading enzyme of GLP-1 (7-36) in vivo.
  • GLP-1 (7-36) is degraded by DPP-IV efficiently to GLP-1 (9-36), which has been speculated to act as a physiological antagonist to GLP-1 (7-36).
  • Inhibiting DPP-IV in vivo is therefore believed to be useful for potentiating endogenous levels of GLP-1 (7-36) and attenuating the formation of its antagonist GLP-1 (9-36).
  • DPP-IV inhibitors are believed to be useful agents for the prevention, delay of progression, and/or treatment of conditions mediated by DPP-IV, in particular diabetes and more particularly, type 2 diabetes mellitus, diabetic dislipidemia, conditions of impaired glucose tolerance (IGT), conditions of impaired fasting plasma glucose (IFG), metabolic acidosis, ketosis, appetite regulation and obesity.
  • diabetes in particular diabetes and more particularly, type 2 diabetes mellitus, diabetic dislipidemia, conditions of impaired glucose tolerance (IGT), conditions of impaired fasting plasma glucose (IFG), metabolic acidosis, ketosis, appetite regulation and obesity.
  • ITT impaired glucose tolerance
  • IGF impaired fasting plasma glucose
  • metabolic acidosis ketosis
  • ketosis ketosis
  • appetite regulation and obesity are believed to be useful agents for the prevention, delay of progression, and/or treatment of conditions mediated by DPP-IV, in particular diabetes and more particularly, type 2 diabetes mellitus, diabetic dislipidemia, conditions of impaired glucose tolerance (IGT), conditions of impaired fasting plasma glucose
  • DPP-IV expression is increased in T-cells upon mitogenic or antigenic stimulation (Mattem, T submit et al., Scand. J. Immunol., 1991, 33, 737). It has been reported that inhibitors of DPP-IV and antibodies to DPP-IV suppress the proliferation of mitogen-stimulated and antigen-stimulated T-cells in a dose-dependant manner (Schon, E., et al., Biol. Chem., 1991, 372, 305). Various other functions of T-lymphocytes such as cytokine production, IL-2 mediated cell proliferation and B-cell helper activity have been shown to be dependent on DPP-IV activity (Schon, E., et al., Scand. J.
  • DPP-IV inhibitors based on boroProline, (Flentke, G. R., et al., Proc. Nat. Acad. Sci. USA, 1991, 88, 1556) although unstable, were effective at inhibiting antigen-induced lymphocyte proliferation and IL-2 production in murine CD4+ T-helper cells.
  • Such boronic acid inhibitors have been shown to have an effect in vivo in mice causing suppression of antibody production induced by immune challenge (Kubota, T. et al., Clin. Exp. ⁇ rnmun., 1992, 89, 192).
  • DPP-IV The role of DPP- IV in regulating T lymphocyte activation may also be attributed, in part, to its cell-surface association with the transmembrane phosphatase, CD45. DPP-IV inhibitors or non-active site ligands may possibly disrupt the CD45-DPP-IV association.
  • CD45 is known to be an integral component of the T-cell signaling apparatus. It has been reported that DPP-IV is essential for the penetration and infectivity of UPV-l and HTV-2 viruses in CD4+ T-cells (Wakselman, M., Nguyen, C, Mazaleyrat, J.-P., Callebaut, C, Krust, B., Hovanessian, A.
  • inhibitors of DPP-IV may be useful immunosuppressants (or cytokine release suppressant drugs) for the treatment of among other things: organ transplant rejection; autoimmune diseases such as inflammatory bowel disease, multiple sclerosis and rheumatoid arthritis; and the treatment of AIDS.
  • lung endothelial cell DPP-IV is an adhesion molecule for lung-metastatic rat breast and prostate carcinoma cells (Johnson, R. C, et al., J. Cell. Biol., 1993, 121, 1423). DPP-IV is known to bind to fibronectin and some metastatic tumor cells are known to carry large amounts of fibronectin on their surface. Potent DPP-IV inhibitors may be useful as drugs to prevent metastases of, for example, breast and prostrate tumors to the lungs.
  • DPP-IV inhibitors may be useful as agents to treat dermatological diseases such as psoriasis and lichen planus.
  • DPP-IV inhibitors may also act to suppress sperm motility and therefore act as a male contraceptive agent.
  • DPP-JN inhibitors have been implicated as novel for treatment of infertility, and particularly human female infertility due to Polycystic ovary syndrome (PCOS, Stein-Leventhal syndrome) which is a condition characterized by thickening of the ovarian capsule and formation of multiple follicular cysts. It results in infertility and amenorrhea.
  • PCOS Polycystic ovary syndrome
  • DPP-IV is thought to play a role in the cleavage of various cytokines (stimulating hematopoietic cells), growth factors and neuropeptides.
  • Stimulated hematopoietic cells are useful for the treatment of disorders that are characterized by a reduced number of hematopoietic cells or their precursors in vivo. Such conditions occur frequently in patients who are immunosuppressed, for example, as a consequence of chemotherapy and/or radiation therapy for cancer. It was discovered that inhibitors of dipeptidyl peptidase type IV are useful for stimulating the growth and differentiation of hematopoietic cells in the absence of exogenously added cytokines or other growth factors or stromal cells.
  • DPP-IV in human plasma has been shown to cleave N-terminal Tyr- Ala from growth hormone-releasing factor and cause motivation of this hormone. Therefore, inhibitors of DPP-IV may be useful in the treatment of short stature due to growth hormone deficiency (Dwarfism) and for promoting GH-dependent tissue growth or re-growth.
  • Dwarfism growth hormone deficiency
  • DPP-TV can also cleave neuropeptides and has been shown to modulate the activity of neuroactive peptides substance P, neuropeptide Y and CLP (Mentlein, R., Dahms, P., Grandt, D., Kruger, R., Proteolytic processing of neuropeptide Y and peptide YY by dipeptidyl peptidase IV, Regul. Pept, 49, 133, 1993; Wetzel, W., Wagner, T., Vogel, D., Demuth, H.-U., Balschun, D., Effects of the CLIP fragment ACTH 20-24 on the duration of REM sleep episodes, Neuropeptides, 31, 41, 1997).
  • DPP-IV inhibitors may also be useful agents for the regulation or normalization of neurological disorders.
  • Several compounds have been shown to inhibit DPP-JN. Nonetheless, a need still exists for new DPP-JN inhibitors that have advantageous potency, stability, selectivity, toxicity and or pharmacodynamics properties. In this regard, a novel class of DPP-JN inhibitors are provided herein.
  • the present invention relates to compounds that have activity for inhibiting DPP- JN. It is noted that these compounds may also have activity for inhibiting other S9 proteases and thus may be used against these other S9 proteases as well as DPP-JN.
  • the present invention also provides compositions, articles of manufacture and kits comprising these compounds.
  • a pharmaceutical composition that comprises a DPP-JN inhibitor according to the present invention as an active ingredient.
  • Pharmaceutical compositions according to the invention may optionally comprise 0.001 %-100% of one or more DPP-JN inhibitors of this invention.
  • compositions may be administered or coadministered by a wide variety of routes, including for example, orally, parenterally, intraperitoneally, intravenously, intraarterially, transdermally, sublingually, intramuscularly, rectally, transbuccally, intranasally, liposomally, via inhalation, vaginally, intraoccularly, via local delivery (for example by catheter or stent), subcutaneously, intraadiposally, intraarticularly, or intrathecally.
  • routes including for example, orally, parenterally, intraperitoneally, intravenously, intraarterially, transdermally, sublingually, intramuscularly, rectally, transbuccally, intranasally, liposomally, via inhalation, vaginally, intraoccularly, via local delivery (for example by catheter or stent), subcutaneously, intraadiposally, intraarticularly, or intrathecally.
  • routes including for example, orally, parenterally, intra
  • the invention is also directed to kits and other articles of manufacture for treating disease states associated with DPP-JN.
  • a kit comprising a composition comprising at least one DPP-JN inhibitor of the present invention in combination with instructions.
  • the instructions may indicate the disease state for which the composition is to be administered, storage information, dosing information and/or instructions regarding how to a ⁇ ninister the composition.
  • the kit may also comprise packaging materials.
  • the packaging material may comprise a container for housing the composition.
  • the kit may also optionally comprise additional components, such as syringes for administration of the composition.
  • the kit may comprise the composition in single or multiple dose forms.
  • an article of manufacture comprises a composition comprising at least one DPP-JN inhibitor of the present invention in combination with packaging materials.
  • the packaging material may comprise a container for housing the composition.
  • the container may optionally comprise a label indicating the disease state for which the composition is to be administered, storage information, dosing information and/or instructions regarding how to administer the composition.
  • the kit may also optionally comprise additional components, such as syringes for administration of the composition.
  • the kit may comprise the composition in single or multiple dose forms.
  • the compounds, compositions, kits and articles of manufacture are used to inhibit DPP-JN.
  • the compounds, compositions, kits and articles of manufacture are used to treat a disease state for which DPP-JN possesses activity that contributes to the pathology and/or symptomology of the disease state.
  • a compound is administered to a subject wherein DPP-JN activity within the subject is altered, preferably reduced.
  • a prodrug of a compound is administered to a subject that is converted to the compound in vivo where it inhibits DPP-JN.
  • a method of inhibiting DPP-JN comprises contacting DPP-JN with a compound according to the present invention.
  • a method of inhibiting DPP-IV comprises causing a compound according to the present invention to be present in a subject in order to inhibit DPP-JN in vivo.
  • a method of inhibiting DPP-JN comprises administering a first compound to a subject that is converted in vivo to a second compound wherein the second compound inhibits DPP-JN in vivo.
  • the compounds of the present invention may be the first or second compounds.
  • a therapeutic method comprises administering a compound according to the present invention.
  • a method of inhibiting cell proliferation comprises contacting a cell with an effective amount of a compound according to the present invention.
  • a method of inhibiting cell proliferation in a patient comprises administering to the patient a therapeutically effective amount of a compound according to the present invention.
  • a method of treating a condition in a patient which is known to be mediated by DPP-JN, or which is known to be treated by DPP-JN inhibitors comprising administering to the patient a therapeutically effective amount of a compound according to the present invention.
  • a method for using a compound according to the present invention in order to manufacture a medicament for use in the treatment of disease state which is known to be mediated by DPP-JN, or which is known to be treated by DPP-JN inhibitors.
  • DPP-JN possesses activity that contributes. to the pathology and/or symptomology of the disease state, the method comprising: causing a compound according to the present invention to be present in a subject in a therapeutically effective amount for the disease state.
  • DPP-JN possesses activity that contributes to the pathology and/or symptomology of the disease state, the method comprising: administering a first compound to a subject that is converted in vivo to a second compound such that the second compound is present in the subject in a therapeutically effective amount for the disease state.
  • the compounds of the present invention may be the first or second compounds.
  • DPP-IN possesses activity that contributes to the pathology and/or symptomology of the disease state, the method comprising: administering a compound according to the present invention to a subject such that the compound is present in the subject in a therapeutically effective amount for the disease state.
  • a method for treating a cell proliferative disease state comprising treating cells with a compound according to the present invention in combination with an anti-proliferative agent, wherein the cells are treated with the compound according to the present invention before, at the same time, and/or after the cells are treated with the anti-proliferative agent, referred to herein as combination therapy.
  • combination therapy is intended to cover when agents are administered before or after each other (sequential therapy) as well as when the agents are administered at the same time.
  • diabetes more particular type 2 diabetes mellitus, diabetic dislipidemia, conditions of impaired glucose tolerance (IGT), conditions of impaired fasting plasma glucose (TFG), metabolic acidosis, ketosis, appetite regulation, obesity, immunosuppressants or
  • the present invention is intended to encompass all pharmaceutically acceptable ionized forms (e.g., salts) and solvates (e.g., hydrates) of the compounds, regardless of whether such ionized forms and solvates are specified since it is well known in the art to administer pharmaceutical agents in an ionized or solvated form. It is also noted that unless a particular stereochemistry is specified, recitation of a compound is intended to encompass all possible stereoisomers (e.g., enantiomers or diastereomers depending on the number of chiral centers), independent of whether the compound is present as an individual isomer or a mixture of isomers.
  • pharmaceutically acceptable ionized forms e.g., salts
  • solvates e.g., hydrates
  • prodrugs may also be administered which are altered in vivo and become a compound according to the present invention.
  • the various methods of using the compounds of the present invention are intended, regardless of whether prodrug delivery is specified, to encompass the administration of a prodrug that is converted in vivo to a compound according to the present invention.
  • certain compounds of the present invention may be altered in vivo prior to inhibiting DPP-JN and thus may themselves be prodrugs for another compound. Such prodrugs of another compound may or may not themselves independently have DPP-IN inhibitory activity.
  • Figure 1 illustrates a ribbon diagram overview of the structure of DPP-JN, highlighting the secondary structural elements of the protein.
  • Figure 2 depicts different, representative 5 membered ring structures that may be formed when R3 and R4 are taken together to form a substituted or unsubstituted ring.
  • Figure 3 depicts different, representative 6 membered ring structures that may be formed when R3 and R4 are taken together to form a substituted or unsubstituted ring.
  • Figure 4 depict additional representative 5 or 6 membered ring structures that may be formed when R3 and R4 are taken together to form a substituted or unsubstituted ring.
  • the rings shown in Figures 2, 3 and 4 are unsubstituted. It is noted that the rings may optionally be further substituted by one or more substituents.
  • Alicyclic means a moiety comprising a non-aromatic ring structure. Alicyclic moieties may be saturated or partially unsaturated with one, two or more double or triple bonds. Alicyclic moieties may also optionally comprise heteroatoms such as nitrogen, oxygen and sulfur. The nitrogen atoms can be optionally quaternerized or oxidized and the sulfur atoms can be optionally oxidized.
  • alicyclic moieties include, but are not limited to moieties with C 3 - C 8 rings such as cyclopropyl, cyclohexane, cyclopentane, cyclopentene, cyclopentadiene, cyclohexane, cyclohexene, cyclohexadiene, cycloheptane, cycloheptene, cycloheptadiene, cyclooctane, cyclooctene, and cyclooctadiene.
  • "Aliphatic" means a moiety characterized by a straight or branched chain arrangement of constituent carbon atoms and may be saturated or partially unsaturated with one, two or more double or triple bonds.
  • Alkenyl represented by itself means a straight or branched, unsaturated, aliphatic radical having a chain of carbon atoms having at least one double bond between adjacent carbon atoms.
  • Cx alkenyl and C ⁇ . ⁇ alkenyl are typically used where X and Y indicate the number of carbon atoms in the chain.
  • C 2- alkenyl includes alkenyls that have a chain of between 2 and 6 carbons.
  • Alkoxy means an oxygen moiety having a further alkyl substituent.
  • the alkoxy groups of the present invention can be optionally substituted.
  • Alkyl represented by itself means a straight or branched, saturated or unsaturated, aliphatic radical having a chain of carbon atoms, optionally with oxygen (See “oxaalkyl”) or nitrogen atoms (See “aminoalkyl”) between the carbon atoms.
  • oxaalkyl oxygen
  • aminoalkyl nitrogen atoms between the carbon atoms.
  • Cx alkyl and C ⁇ . ⁇ alkyl are typically used where X and Y indicate the number of carbon atoms in the chain.
  • C ⁇ -6 alkyl includes alkyls that have a chain of between 1 and 6 carbons (e.g., methyl, ethyl, propyl, isopropyl, butyl, .fee-butyl, isobutyl, tert-butyl, vinyl, allyl, 1-propenyl, isopropenyl, 1 -butenyl, 2-butenyl, 3-butenyl, 2-methylalTyl, ethynyl, 1-propynyl, 2-propynyl, and the like).
  • 1 and 6 carbons e.g., methyl, ethyl, propyl, isopropyl, butyl, .fee-butyl, isobutyl, tert-butyl, vinyl, allyl, 1-propenyl, isopropenyl, 1 -butenyl, 2-butenyl, 3-butenyl, 2-methylalTyl
  • Alkyl represented along with another radical means a straight or branched, saturated or unsaturated aliphatic divalent radical having the number of atoms indicated or when no atoms are indicated means a bond (e.g., (C6- ⁇ o)aryl(C 1- 3)alkyl includes, benzyl, phenethyl, 1-phenylethyl, 3-phenylpropyl, 2- thienylmethyl, 2-pyridinylmethyl and the like).
  • Alkylene unless indicated otherwise, means a straight or branched, saturated or unsaturated, aliphatic, divalent radical.
  • Cx alkylene and C ⁇ . ⁇ alkylene are typically used where X and Y indicate the number of carbon atoms in the chain.
  • 2-methyltetramethylene (-CH 2 CH(CH 3 )CH 2 CH 2 -), pentamethylene (-CH 2 CH 2 CH 2 CH 2 CH 2 -) and the like).
  • Alkylidene means a straight or branched saturated or unsaturated, aliphatic radical connected to the parent molecule by a double bond.
  • Cx alkylidene and C ⁇ . ⁇ alkylidene are typically used where X and Y indicate the number of carbon atoms in the chain.
  • Alkynyl represented by itself means a straight or branched, unsaturated, aliphatic radical having a chain of carbon atoms having at least one triple bond between adjacent carbon atoms.
  • Cx alkynyl and C ⁇ . ⁇ alkynyl are typically used where X and Y indicate the number of carbon atoms in the chain.
  • C 2 ⁇ alkynyl includes alkynyls that have a chain of between 2 and 6 carbons.
  • amino means a nitrogen moiety having two further substituents where a hydrogen or carbon atom is attached to the nitrogen.
  • representative amino groups include -NH 2 , -NHCH 3 , -N(CH 3 ) 2 , -NHC 1-3 -alkyl, -N(C ⁇ . 3 -alkyl) 2 and the like.
  • the compounds of the invention containing amino moieties may include protected derivatives thereof. Suitable protecting groups for amino moieties include acetyl, tert-butoxycarbonyl, benzyloxycarbonyl, arid the like.
  • Aminoalkyl means an alkyl, as defined above, except where one or more substituted or unsubstituted nitrogen atoms (-N-) are positioned between carbon atoms of the alkyl.
  • an (C 2-6 ) aminoalkyl refers to a chain comprising between 2 and 6 carbons and one or more nitrogen atoms positioned between the carbon atoms.
  • "Animal” includes humans, non-human mammals (e.g. , dogs, cats, rabbits, cattle, horses, sheep, goats, swine, deer, and the like) and non-mammals (e.g., birds, and the like).
  • Aromatic means a moiety wherein the constituent atoms make up an unsaturated ring system, all atoms in the ring system are sp hybridized and the total number of pi electrons is equal to 4n+2.
  • An aromatic ring may be such that the ring atoms are only carbon atoms or may include carbon and non-carbon atoms (see Heteroaryl).
  • Aryl means a monocyclic or polycyclic ring assembly wherein each ring is aromatic or when fused with one or more rings forms an aromatic ring assembly. If one or more ring atoms is not carbon (e.g., N, S), the aryl is a heteroaryl.
  • Aryloxy means an oxygen moiety having a further aryl substituent.
  • the aryloxy groups of the present invention can be optionally substituted.
  • Bicycloalkyl means a saturated or partially unsaturated fused bicyclic or bridged polycyclic ring assembly.
  • Bicycloaryl means a bicyclic ring assembly wherein the rings are linked by a single bond or fused and at least one of the rings comprising the assembly is aromatic.
  • Cx bicycloaryl and C ⁇ . ⁇ bicycloaryl are typically used where X and Y indicate the number of carbon atoms in the bicyclic ring assembly and directly attached to the ring.
  • “Bridging ring” as used herein refers to a ring that is bonded to another ring to form a compound having a bicyclic structure where two ring atoms that are common to both rings are not directly bound to each other.
  • Non-exclusive examples of common compounds having a bridging ring include bor ⁇ eol, norbornane, 7-oxabicyclo[2.2. ljheptane, and the like.
  • One or both rings of the bicyclic system may also comprise heteroatoms.
  • Carbamoyl means the radical -OC(O)NR a Rb where R a and Rt, are each independently two further substituents where a hydrogen or carbon atom is attached to the nitrogen.
  • Carbocycle means a ring consisting of carbon atoms.
  • Carbocyclic ketone derivative means a carbocyclic derivative wherein the ring contains a -CO- moiety.
  • Carbonyl means the radical -CO-. It is noted that the carbonyl radical may be further substituted with a variety of substituents to form different carbonyl groups including acids, acid halides, aldehydes, amides, esters, and ketones.
  • Carboxy means the radical -CO 2 -. It is noted that compounds of the invention containing carboxy moieties may include protected derivatives thereof, i.e., where the oxygen is substituted with a protecting group. Suitable protecting groups for carboxy moieties include benzyl, tert-butyl, and the like.
  • Cyano means the radical -CN.
  • Cycloalkyl means a non-aromatic, saturated or partially unsaturated, monocyclic, fused bicyclic or bridged polycyclic ring assembly.
  • C x cycloalkyl and C ⁇ -Y cycloalkyl are typically used where X and Y indicate the number of carbon atoms in the ring assembly.
  • C3.10 cycloalkyl includes cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexenyl, 2,5-cyclohexadienyl, bicyclo[2.2.2]octyl, adamantan-1-yl, decahydronaphthyl, oxocyclohexyl, dioxocyclohexyl, thiocyclohexyl,
  • Cycloalkylene means a divalent saturated or partially unsaturated, monocyclic or polycyclic ring assembly. Cx cycloalkylene and C ⁇ . ⁇ cycloalkylene are typically used where
  • X and Y indicate the number of carbon atoms in the ring assembly.
  • Disease specifically includes any unhealthy condition of an animal or part thereof and includes an unhealthy condition that may be caused by, or incident to, medical or veterinary therapy applied to that animal, Le., the "side effects" of such therapy.
  • fused ring refers to a ring that is bonded to another ring to form a compound haying a bicyclic structure where the ring atoms that are common to both rings are directly bound to each other.
  • Non-exclusive examples of common fused rings include decalin, naphthalene, anthracene, phenanthrene, indole, furan, benzofuran, quinoline, and the like.
  • Compounds having fused ring systems may be saturated, partially saturated, carbocyclics, heterocyclics, aromatics, heteroaromatics, and the like.
  • Halo means fluoro, chloro, bromo or iodo.
  • Halo-substituted alkyl as an isolated group or part of a larger group, means
  • alkyl substituted by one or more "halo" atoms, as such terms are defined in this Application.
  • Halo-substituted alkyl includes haloalkyl, dihaloalkyl, trihaloalkyl, perhaloalkyl and the like
  • halo-substituted (C ⁇ alkyl includes chloromethyl, dichloromethyl, difluoromethyl, trifluoromethyl, 2,2,2-trifluoroethyl, perfluoroethyl, 2,2,2-trifluoro-l,l-dichloroethyl, and the like).
  • Heteroaryl means a cyclic aromatic group having five or six ring atoms, wherein at least one ring atom is a heteroatom and the remaining ring atoms are carbon.
  • the nitrogen atoms can be optionally quaternerized and the sulfur atoms can be optionally oxidized.
  • Heteroaryl groups of this invention include, but are not limited to, those derived from furan, imidazole, isothiazole, isoxazole, oxadiazole, oxazole, 1,2,3-oxadiazole, pyrazine, pyrazole, pyridazine, pyridine, pyrimidine, pyrroline, thiazole, 1,3,4-thiadiazole, triazole and tetrazole.
  • Heteroaryl also includes, but is not limited to, bicyclic or tricyclic rings, wherein the heteroaryl ring is fused to one or two rings independently selected from the group consisting of an aryl ring, a cycloalkyl ring, a cycloalkenyl ring, and another monocyclic heteroaryl or heterocycloalkyl ring.
  • bicyclic or tricyclic heteroaryls include, but are not limited to, those derived frombenzo[b]furan, benzo[b]thiophene, benzimidazole, imidazo[4,5-c]pyridine, quinazoline, thieno[2,3-c]pyridine, thieno[3,2-b]pyridine, thieno[2,3-b]pyridine, indolizine, imidazo[l,2a]pyridine, quinoline, isoquinoline, phthalazine, quinoxaline, naphthyridine, quinolizine, indole, isoindole, indazole, indoline, benzoxazole, benzopyrazole, benzothiazole, imidazo[l,5-a]pyridine, pyrazolo[l,5-a]pyridine, imidazo[l,2-a]pyrirmdine, imidazo[l,2- c]pyrimidine, imidazo[
  • the bicyclic or tricyclic heteroaryl rings can be attached to the parent molecule through either the heteroaryl group itself or the aryl, cycloalkyl, cycloalkenyl or heterocycloalkyl group to which it is fused.
  • the heteroaryl groups of this invention can be substituted or unsubstituted.
  • Heteroaryloxy means an oxygen moiety having a further heteroaryl substituent.
  • heteroaryloxy groups of the present invention can be optionally substituted.
  • Heteroatom refers to an atom that is not a carbon atom. Particular examples of heteroatoms include, but are not limited to nitrogen, oxygen, and sulfur.
  • Heteroatom moiety includes a moiety where the atom by which the moiety is attached is not a carbon.
  • Heterobicycloalkyl means bicycloalkyl, as defined in this Application, provided that one or more of the atoms within the ring is a heteroatom.
  • hetero(C 9-12 )bicycloalkyl as used in this application includes, but is not limited to, 3-aza- bicyclo[4.1.0]he ⁇ t-3-yl, 2-aza-bicyclo[3.1.0]hex-2-yl , 3-aza-bicyclo[3.1.0]hex-3-yl, and the like.
  • Heterobicycloaryl means bicycloaryl, as defined in this Application, provided that one or more of the atoms within the ring is a heteroatom.
  • hetero(C -1 o)bicycloaryl as used in this Application includes, but is not limited to, 2-amino-
  • Heterocycloalkyl means cycloalkyl, as defined in this AppHcation, provided that one or more of the atoms forming the ring is a heteroatom selected, independently from N, O, or S.
  • Non-exclusive examples of heterocycloalkyl include piperidyl, 4-morpholyl, 4- piperazinyl, pyrrolidinyl, perhydropyrrolizinyl, 1,4-diazaperhydroepinyl, 1,3-dioxanyl, 1,4- dioxanyl and the like.
  • Heterocycloalkylene means cycloalkylene, as defined in this Application, provided that one or more of the ring member carbon atoms is replaced by a heteroatom.
  • Heterocycloalkylene means cycloalkylene, as defined in this Application, provided that one or more of the ring member carbon atoms is replaced by a heteroatom.
  • “Hydroxy” means the radical -OH.
  • Jminoketone derivative means a derivative comprising the moiety -C(NR)-, wherein R comprises a hydrogen or carbon atom attached to the nitrogen.
  • “Isomers” mean any compound having an identical molecular formulae but differing in the nature or sequence of bonding of their atoms or in the arrangement of their atoms in space.
  • stereoisomers that differ in the arrangement of their atoms in space are termed “stereoisomers.”
  • stereoisomers that are not mirror images of one another are termed “diastereomers” and stereoisomers that are nonsuperimposable mirror images are termed “enantiomers” or sometimes "optical isomers.”
  • a carbon atom bonded to four nonidentical substituents is termed a “chiral center.”
  • a compound with one chiral center has two enantiomeric forms of opposite chirality.
  • a mixture of the two enantiomeric forms is termed a "racemic mixture.”
  • a compound that has more than one chiral center has 2 n ⁇ enantiomeric pairs, where n is the number of chiral centers.
  • Compounds with more than one chiral center may exist as ether an individual diastereomer or as a mixture of diastereomers, termed a "diastereomeric mixture.”
  • a stereoisomer may be characterized by the absolute configuration of that chiral center. Absolute configuration refers to the arrangement in space of the substituents attached to the chiral center.
  • Enantiomers are characterized by the absolute configuration of their chiral centers and described by the R- and S-sequencing rules of Cahn, Ingold and Prelog.
  • Oxaalkyl means an alkyl, as defined above, except where one or more oxygen atoms (-O-) are positioned between carbon atoms of the alkyl.
  • an (C 2- 6)oxaaikyl refers to a chain comprising between 2 and 6 carbons and one or more oxygen atoms positioned between the carbon atoms.
  • Oxoalkyl means an alkyl, further substituted with a carbonyl group.
  • the carbonyl group may be an aldehyde, ketone, ester, amide, acid or acid chloride.
  • “Pharmaceutically acceptable” means that which is useful in preparing a pharmaceutical composition that is generally safe, non-toxic and neither biologically nor otherwise undesirable and includes that which is acceptable for veterinary use as well as human pharmaceutical use.
  • “Pharmaceutically acceptable salts” means salts of inhibitors of the present invention which are pharmaceutically acceptable, as defined above, and which possess the desired pharmacological activity.
  • Such salts include acid addition salts formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or with organic acids such as acetic acid, propionic acid, hexanoic acid, heptanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartatic acid, citric acid, benzoic acid, ⁇ -(4-hydroxybenzoyl)benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1,2-ethanedisulfonic acid, 2-hydroxyethanesulfonic acid,
  • Pharmaceutically acceptable salts also include base addition salts which may be formed when acidic protons present are capable of reacting with inorganic or organic bases.
  • Acceptable inorganic bases include sodium hydroxide, sodium carbonate, potassium hydroxide, aluminum hydroxide and calcium hydroxide.
  • Acceptable organic bases include emanolamine, diethanolamine, triethanolamine, tromethamine, N-memylglucamine and the like.
  • Prodrug means a compound that is convertible in vivo metabolically into an inhibitor according to the present invention. The prodrug itself may or may not also have DPP-JN inhibitory activity.
  • an inhibitor comprising a hydroxy group may be administered as an ester that is converted by hydrolysis in vivo to the hydroxy compound.
  • Suitable esters that may be converted in vivo into hydroxy compounds include acetates, citrates, lactates, tartrates, malonates, oxalates, salicylates, propionates, succinates, fumarates, maleates, methylene-bis-b-hydroxynaphthoates, gentisates, isethionates, di-p-toluoyltartrates, methanesulfonates, ethanesulfonates, benzenesulfonates, p-toluenesulfonates, cyclohexylsulfamates, quinates, esters of amino acids, and the like.
  • an inhibitor comprising an amine group may be administered as an amide that is converted by hydrolysis in vivo to the amine compound.
  • Protected derivatives means derivatives of inhibitors in which a reactive site or sites are blocked with protecting groups. Protected derivatives are useful in the preparation of inhibitors or in themselves may be active as inhibitors. A comprehensive list of suitable protecting groups can be found in T.W. Greene, Protecting Groups in Organic Synthesis, 3rd edition, John Wiley & Sons, Inc. 1999.
  • Substituted or unsubstituted means that a given moiety may consist of only hydrogen substituents through available valencies (unsubstituted) or may further comprise one or more non-hydrogen substituents through available valencies (substituted) that are not otherwise specified by the name of the given moiety.
  • isopropyl is an example of an ethylene moiety that is substituted by -CH 3 .
  • a non-hydrogen substituent may be any substituent that may be bound to an atom of the given moiety that is specified to be substituted.
  • substituents include, but are not limited to, aldehyde, alicyclic, aliphatic, alkyl, alkylene, alkylidene, amide, amino, aminoalkyl, aromatic, aryl, bicycloalkyl, bicycloaryl, carbamoyl, carbocyclyl, carboxyl, carbonyl group, cycloalkyl, cycloalkylene, ester, halo, heterobicycloalkyl, heterocycloalkylene, heteroaryl, heterobicycloaryl, heterocycloalkyl, oxo, hydroxy, iminoketone, ketone, nitro, oxaalkyl, and oxoalkyl moieties, each of which may optionally also be substituted or unsubstituted.
  • Sulfinyl means the radical -SO-. It is noted that the sulfinyl radical may be further substituted with a variety of substituents to form different sulfinyl groups including sulfinic acids, sulfinamides, sulfinyl esters, and sulfoxides.
  • Sulfonyl means the radical -SO 2 -. It is noted that the sulfonyl radical may be further substituted with a variety of substituents to form different sulfonyl groups including sulfonic acids, sulfonamides, sulfonate esters, and sulfones.
  • “Therapeutically effective amount” means that amount which, when administered to an animal for treating a disease, is sufficient to effect such treatment for the disease.
  • Thiocarbonyl means the radical -CS-. It is noted that the thiocarbonyl radical may be further substituted with a variety of substituents to form different thiocarbonyl groups including thioacids, thioamides, thioesters, and thioketones.
  • Treatment or “treating” means any administration of a compound of the present invention and includes:
  • a alkyl indicates that there is one carbon atom but does not indicate what are the substituents on the carbon atom.
  • a alkyl comprises methyl (i.e., -CH 3 ) as well as -R a RbRc where R a , R b , and R c may each independently be hydrogen or any other substituent where the atom attached to the carbon is a heteroatom or cyano.
  • CF 3 , CH 2 OH and CH 2 CN for example, are all alkyls.
  • DPP- IV dipeptidyl peptidases JN
  • DPP-JN EC.3.4.14.5 also known as DPP4, DP4, DAP-TV, adenosine deaminase complexing protein 2, adenosine deaminase binding protein (ADAbp) or CD26
  • ADAbp adenosine deaminase binding protein
  • CD26 CD26
  • DPP-TV has a serine type mechanism of protease activity, cleaving off dipeptides from the ammo-terminus of peptides with proline or alanine at the penultimate position. In addition the slow release of dipeptides of the type X-Gly or X-Ser is reported for some naturally occurring peptides.
  • DPP-IV is constitutively expressed on epithelial and endothelial cells of a variety of different tissues (intestine, liver, lung, kidney and placenta), and is also found in body fluids.
  • DPP-JN is also expressed on circulating T-lymphocytes and has been shown to be synonymous with the cell-surface antigen, CD-26.
  • DPP-JN The wild-type form of full length DPP-JN is described in GenBank Accession Number NM_001935 ("Dipeptidyl peptidase TV (CD 26) gene expression in enterocyte-like colon cancer cell lines HT-29 and Caco-2. Cloning of the complete human coding sequence and changes of dipeptidyl peptidase TN mR ⁇ A levels during cell differentiation", Darmoul, D., Lacasa, M., Baricault, L., Marguet, D., Sapin, C, Trotot, P., Barbat, A. and Trugnan, G., J. Biol. Chem., 267 (7), 4824-4833, 1992).
  • DPP-JN is a member of the S9 family of serine proteases, more particularly the S9B family.
  • Other members of the S9 family include, but are not limited to:
  • Subfamily S9A Dipeptidyl-peptidase; Oligopeptidase B (EC 3.4.21.83): Oligopeptidase B; Prolyl oligopeptidase (EC 3.4.21.26): Subfamily S9B: Dipeptidyl aminopeptidase A; Dipeptidyl aminopeptidase B Dipeptidyl-peptidase TV (EC 3.4.14.5): Dipeptidyl-peptidase V Fibroblast activation protein alpha subunit; Seprase Subfamily S9C: Acvlaminoacvl-peptidase (EC 3.4.19.1) [0108] It is noted that the compounds of the present invention may also possess inhibitory activity for other S9 family members and thus maybe used to address disease states associated with these other family members.
  • FIG. 1 illustrates a ribbon diagram overview of the structure of DPP-TV, highlighting secondary structural elements of the protein.
  • DPP-TV is a cylindrical shaped molecule with an approximate height of 70 A and a diameter of 60 A.
  • the catalytic triad of DPP-IN (Ser642, Asp720 and His752) is illustrated in the center of the figure by a "ball and stick" representation. This triad of amino acids is located in the peptidase domain or catalytic domain of DPP-TV.
  • the catalytic domain is covalently linked to the ⁇ -propeller domain.
  • the catalytic domain of DPP-TV includes residues 1-67 and 511-778.
  • the catalytic domain of DPP-IN adopts a characteristic ⁇ / ⁇ hydrolase fold.
  • the core of this domain contains an 8- stranded ⁇ -sheet with all strands being parallel except one.
  • the ⁇ -sheet is significantly twisted and is flanked by three ⁇ -helices on one side and five ⁇ -helices on the other.
  • the topology of the ⁇ -strands is 1, 2, -lx, 2x and (lx) (J. S. Richardson: The anatomy and taxonomy of protein structure; (1981) Adv. Protein Chem.269, 15076-15084.).
  • a number of residues were identified that contribute to the shape and charge characteristics of the active site. Knowledge of these residues has been an important contribution to the design of DPP-JN inhibitors of the present invention.
  • DPP-TV inhibitors of the present invention comprise Formula I:
  • Ri is hydrogen or is selected from the group consisting of (C ⁇ - 1 o)alkyl, (C 3 -i 2 )cycloalkyl, (C3 -12 )cycloalkyl(C 1 - 5 )alkyl, hetero(C 3 -i 2 )cycloalkyl, aryl(C 1 - 10 )alkyl, (C 9- i 2 )bicycloaryl, hetero(C 4-12 )bicycloaryl, carbonyl (C ⁇ -3)alkyl, thiocarbonyl (C ⁇ -3 )alkyl, sulfonyl (C 1- 3)alkyl, sulfinyl amino, cyano, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;
  • R 2 is selected from the group consisting of halo, perhalo(C ⁇ - 1 o)alkyl, amino, cyano, thio, (C ⁇ -io)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, carbonyl (C ⁇ . 3 )alkyl, thiocarbonyl sulfinyl (C ⁇ . 3 )alkyl, imino alkoxy, aryloxy, heteroaryloxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, and a substituted or unsubstituted 4, 5, 6 or 7 membered ring;
  • R 3 and R 4 are taken together to form a substituted or unsubstituted 5 or 6 membered ring;
  • R 9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
  • DPP-JN inhibitors of the present invention comprise Formula JJ:
  • J, K, L, and M are each independently selected from the group of moieties where the ring atom is either C or N;
  • Ri is hydrogen or is selected from the group consisting of (C 1 - ⁇ o)alkyl, (C3-i2)cycloalkyl, (C3- 1 2)cycloalkyl(C 1 - 5 )alkyl, hetero(C3- 12 )cycloalkyl, aryl(C 1 - 1 o)alkyl, (Cg- ⁇ bicycloaryl, hetero(C -i 2 )bicycloaryl, carbonyl (C 1-3 )alkyl, thiocarbonyl (C 1-3 )a ⁇ kyl, sulfonyl (C 1 -3)alkyl, sulfinyl (Ci ⁇ alkyl, imino (C ⁇ .
  • R 2 is selected from the group consisting of halo, perhalo(C ⁇ - ⁇ o)alkyl, amino, cyano, thio, (C ⁇ - ⁇ o)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, carbonyl (C ⁇ - 3 )alkyl, thiocarbonyl (Ci ⁇ alkyl, sulfonyl (C ⁇ -3 )alkyl, sulfinyl (C ⁇ .
  • R is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
  • DPP-JN inhibitors of the present invention having particular substitution patterns have significantly improved stability in biological assays. Improvement in the stabilities of the compounds in rat liver microsomes, for example, as demonstrated by the increase in the half life of the test compound in microsomal assays, has been demonstrated in particular substitutions of the DPP-IN inhibitors of the present invention.
  • DPP-IV inhibitors comprising the Formula JJ with substitution at the 6 and/or the 7-positions result in enhanced stability as demonstrated in biological assays.
  • compounds of the Formula JJ comprise quinazolinones that are substituted at the 6-position, the 7-position, or both the 6 and 7 positions of the compound of Formula TJ. ⁇ on-limiting examples of the compounds of the Formula JI include quinazolinone derivatives, naphthyridine derivatives, 4H-pyrido[3,4- d]pyrimidine derivatives, etc.
  • DPP-JN inhibitors comprise compounds of the Formula JJ wherein K is CR12, where R12 is independently selected from the group consisting of halo, perhalo(Cl-10)alkyl, CF3, cyano, nitro, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, a carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
  • R12 is independently selected from the group consisting of halo, perhalo(Cl-10)alkyl, CF3, cyano, nitro, alkyl, aryl, heteroaryl, aminosulfon
  • K is CR12 where R12 is independently selected from the group consisting of halo, perhalo(Cl-10)alkyl, CF3, cyano, nitro, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, a carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, and L is nitrogen.
  • R12 is independently selected from the group consisting of halo, perhalo(Cl-10)alkyl, CF3, cyano, nitro, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl
  • K is CR 12 , where R 12 is independently selected from the group consisting of halo, perhalo(C 1 - 1 o)alkyl, CF 3 , cyano, nitro, alkyl, aryloxy, heteroaryloxy, amino, and alkoxy, each substituted or unsubstituted;
  • Ri is selected from the group consisting of -(CH 2 )-(2-cyano)phenyl, -(CH 2 )-(3-cyano)phenyl, -(CH 2 )-(2-hydroxy)phenyl, -(CH 2 )-(3-hydroxy)phenyl, -(CH 2 )-(2- alkenyl)phenyl, -(CH 2 )-(3-alkenyl)phenyl, -(CH 2 )-(2-alkynyl)phenyl, -(CH 2 )-(3- alkynyl)phenyl, -(CH 2 )-(2-nitro)phenyl, -(CH 2 )-(3-nitro)phenyl, -(CH 2 )-(2-carboxy)phenyl, -(CH 2 )-(3-carboxy)phenyl, -(CH 2 )-(2-carboxamido) ⁇ henyl, -(CH 2
  • R 2 is selected from the group consisting of 3-amino-piperidin-l-yl, 3-aminomethyl- pyrrolidin-1-yl, azetidin-1-yl, 3-aminoazetidin-l-yl, pyrrolidin- 1-yl, 3-aminocyclopent-l-yl, 3- aminomethylcyclopent-1-yl, 3-aminomethylcyclohex-l-yl, hexahydroazepin-1-yl, 3- aminohexahydroazepin-1-yl, 3-amino-cyclohex-l-yl, piperazin- 1-yl, homopiperazin- 1-yl, 3- amino-pyrrolidin-1-yl, and R-3-aminopiperidin-l-yl, each substituted or unsubstituted. [0118] In yet another variation,
  • K is CRi 2 , where R 12 is independently selected from the group consisting of halo, perhalo(C 1 - 1 o)alkyl, CF 3 , cyano, nitro, alkyl, aryloxy, heteroaryloxy, amino, and alkoxy, each substituted or unsubstituted;
  • Ri is selected from the group consisting of -(CH 2 )-(2-cyano)phenyl, -(CH 2 )-(3-cyano) ⁇ henyl, -(CH 2 )-(2-hydroxy) ⁇ henyl, -(CH 2 )-(3-hydroxy)phenyl, -(CH 2 )-(2- alkenyl) ⁇ henyl, -(CH 2 )-(3-alkenyl) ⁇ henyl, -(CH 2 )-(2-alkynyl)phenyl, -(CH 2 )-(3 ⁇ alkynyl)phenyl, -(CH 2 )-(2-nitro)phenyl, -(CH 2 )-(3-nitro) ⁇ henyl, -(CH 2 )-(2-carboxy)phenyl, -(CH 2 )-(3-carboxy)phenyl, -(CH 2 )-(2-carboxamido)phenyl, -
  • R 2 is selected from the group consisting of 3-amino-piperidin-l-yl, 3-aminomethyl- pyrrolidin-1-yl, azetidin-1-yl, 3-aminoazetidin-l-yl, pyrrolidin- 1-yl, 3-aminocyclopent-l-yl, 3- aminomethylcyclopent-1-yl, 3-aminomethylcyclohex-l-yl, hexahydroazepin-1-yl, 3- aminohexahydroazepin-1-yl, 3-amino-cyclohex-l-yl, piperazin- 1-yl, homopiperazin- 1-yl, 3- amino-pyrrolidin-1-yl, and R-3-aminopiperidin-l-yl, each substituted or unsubstituted.
  • DPP-IN inhibitors of the present invention comprise a member selected from the group consisting of Formulae HJa, JJIb, T
  • Ri is hydrogen or is selected from the group consisting of (C ⁇ -io)alkyl, (C3 -1 2)cycloalkyl, (C 3 . 12 )cycloalkyl(C 1 - 5 )alkyl, hetero(C 3 -i2)cycloalkyl, aryl(C ⁇ - 10 )alkyl, (Cg- ⁇ bicycloaryl, hetero(C -12 )bicycloaryl, carbonyl (C ⁇ alkyl, thiocarbonyl (C ⁇ alkyl, sulfonyl (C 1-3 )alkyl, sulfinyl (C ⁇ alkyl, imino (C 1-3 )alkyl, amino, cyano, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;
  • R 2 is selected from the group consisting of halo, perhalo(C ⁇ - 1 o)alkyl, amino, cyano, thio, (C 1 - 1 o)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, carbonyl ( ⁇ alkyl, thiocarbonyl sulfinyl (C ⁇ - 3 )alkyl, imino (C ⁇ - 3 )alkyl, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, and a substituted or unsubstituted 4, 5, 6 or 7 membered ring;
  • R 9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted; and each R ⁇ 9 is independently selected from the group consisting of hydrogen, halo, perhalo(C ⁇ - ⁇ o)alkyl, CF 3 , cyano, nitro, alkyl, alkene, alkyne, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or
  • DPP-JN inhibitors of the present invention comprise Formula TN:
  • W, X, and Y are each independently selected from the group of moieties where the ring atom is either C, N, O or S;
  • Ri is hydrogen or is selected from the group consisting of (C ⁇ - ⁇ o)alkyl, (C 3- i 2 )cycloalkyl, (C3-i 2 )cycloalkyl(C 1 - 5 )alkyl, hetero(C 3 - 12 )cycloalkyl, aryl(C 1 -io)alkyl, (C 9 - ⁇ 2 )bicycloaryl, hetero(C , 12 )bicycloaryl, carbonyl sulfonyl (C 1 -3)alkyl, sulfinyl ( ⁇ alkyl, imino (C ⁇ aHcyl, amino, cyano, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;
  • R 2 is selected from the group consisting of halo, perhalo(C 1 - 1 o)alkyl, amino, cyano, thio, (C 1 - 1 o)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, carbonyl (C ⁇ - 3 )alkyl, thiocarbonyl hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, and a substituted or unsubstituted 4, 5, 6 or 7 membered ring; and R is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
  • DPP-IV inhibitors of the present invention comprise Formulae Va, Vb or Vc:
  • Ri is hydrogen or is selected from the group consisting of (Ci-io)alkyl, (C 3 - 12 )cycloalkyl, (C 3- i 2 )cycloalkyl(C ⁇ - 5 )alkyl, hetero(C 3 - 12 )cycloalkyl, aryl(Ci-io)alkyl, (C 9-12 )bicycloaryl, hetero(C -12 )bicycloaryl, carbonyl (C ⁇ alkyl, thiocarbonyl ( ⁇ alkyl, sulfonyl (C ⁇ .
  • R 2 is selected from the group consisting of halo, perhalo(Ci- 1 o)alkyl, amino, cyano, thio, (C ⁇ - 1 o)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, carbonyl ( ⁇ alkyl, thiocarbonyl sulfinyl (C 1-3 )alkyl, imino (C 1-3 )alkyl, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, and a substituted or unsubstituted 4, 5, 6 or 7 membered ring;
  • R 9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted; and each R ⁇ 9 is independently selected from the group consisting of hydrogen, halo, ⁇ erhalo(C 1 - 1 o)alkyl, CF 3 , cyano, nitro, alkyl, alkene, alkyne, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group,
  • DPP-IV inhibitors of the present invention comprise Formula Via or Formula VJb:
  • W, X, and Y are each independently selected from the group of moieties where the ring atom is either C, N, O or S;
  • Ri is hydrogen or is selected from the group consisting of (Ci-io)alkyl, (C 3- i 2 )cycloalkyl, (C 3- i 2 )cycloalkyl(Ci- 5 )alkyl, hetero(C 3 - ⁇ 2 )cycloalkyl, aryl(C 1 - 1 o)alkyl, (C 9- ⁇ 2 )bicycloaryl, hetero(C -i 2 )bicycloaryl, carbonyl ( ⁇ alkyl, thiocarbonyl (C ⁇ -3 )alkyl, sulfonyl (Ci- 3 )alkyl, sulfinyl (Chalky!, imino ( ⁇ alkyl, amino, cyano, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or
  • R 2 is selected from the group consisting of halo, perhalo(Ci-io)alkyl, amino, cyano, thio, (Ci- 1 o)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, carbonyl (Ci ⁇ alkyl, thiocarbonyl hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, and a substituted or unsubstituted 4, 5, 6 or 7 membered ring; and
  • R 9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
  • DPP-IV inhibitors of the present invention comprise Formulae Vila, VHb, or VJJc: wherein
  • X is selected from the group of moieties where the ring atom is either C, N, O or S in Formula VTJa, or X is selected from the group of moieties where the ring atom is either C or N in Formula VTJb or Formula VTJc;
  • Ri is hydrogen or is selected from the group consisting of (Ci- 10 )alkyl, (C 3 -i 2 )cycloalkyl, (C 3 .i 2 )cycloalkyl(Ci- 5 )alkyl, hetero(C 3 - 12 )cycloaIkyl, aryl(C ⁇ - ⁇ o)alkyl, C 9- i 2 )bicycloaryl, hetero(C 4 .
  • R 2 is selected from the group consisting of halo, ⁇ erhalo(Ci-io)alkyl, amino, cyano, thio, (Cr ⁇ o)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, carbonyl (C 1- 3)alkyl, thiocarbonyl (Ci- 3 )alkyl, sulfonyl alkoxy, aryloxy, heteroaryloxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, and a substituted or unsubstituted 4, 5, 6 or 7 membered ring;
  • R is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted; and each R 19 is independently selected from the group consisting of hydrogen, halo, perhalo(Ci-io)alkyl, CF3, cyano, nitro, alkyl, alkene, alkyne, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstit
  • DPP-TV inhibitors of the present invention comprise Formulae Villa VTUb and VTTIc:
  • J, K, L, and M are each independently selected from the group of moieties where the ring atom is either C, N, O or S;
  • Ri is hydrogen or is selected from the group consisting of (C ⁇ -io)alkyl, (C 3 - ⁇ 2 )cycloalkyl, (C 3- i 2 )cycloalkyl(Ci- 5 )alkyl, hetero(C 3 - 12 )cycloalkyl, aryl(C ⁇ -i 0 )alkyl, (C 9- ⁇ 2 )bicycloaryl, hetero(C 4 .i 2 )bicycloaryl, carbonyl sulfonyl sulfinyl (Ci.
  • R 2 is selected from the group consisting of halo, perhalo(C ⁇ - ⁇ 0 )alkyl, amino, cyano, thio, (C ⁇ - ⁇ o)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, carbonyl thiocarbonyl sulfinyl (Ci_ 3 )alkyl, imino hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, and a substituted or unsubstituted 4, 5, 6 or 7 membered ring; and
  • R 9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
  • DPP-JV inhibitors of the present invention comprise Formula TX:
  • J, K, L, and M are each independently selected from the group of moieties where the ring atom is either C, N, O or S;
  • Ri is hydrogen or is selected from the group consisting of (Ci-io)alkyl, (C 3 -i 2 )cycloalkyl, (C 3- i 2 )cycloalkyl(Ci- 5 )alkyl, hetero(C 3 -i 2 )cycloalkyl, aryl(Ci-io)alkyl, (C 9 .i 2 )bicycloaryl, hetero(C 4-12 )bicycloaryl, carbonyl (Ci ⁇ alkyl, thiocarbonyl (C 1-3 )alkyl, sulfonyl (C ⁇ - 3 )alkyl, sulfinyl (Ci ⁇ alkyl, imino (Chalky!, amino, cyano, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsub
  • R 2 is selected from the group consisting of halo, perhalo(C ⁇ - ⁇ o)alkyl, amino, cyano, thio, (C ⁇ -io)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, carbonyl (Ci.3)alkyl, thiocarbonyl sulfonyl (C 1-3 )alkyl, sulfinyl (Ci_ 3 )alkyl, imino alkoxy, aryloxy, heteroaryloxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, and a substituted or unsubstituted 4, 5, 6 or 7 membered ring; and
  • R 9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
  • DPP-TV inhibitors of the present invention comprise Formula X:
  • Ri is -ZR m , where Z is a moiety providing 1-6 atom separation between R m and the ring to which Ri is attached, and R m is selected from the group consisting of a substituted or unsubstituted (C3- )cycloalkyl and aryl;
  • R2 is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring;
  • R 3 and R 4 are taken together to form a substituted or unsubstituted 5 or 6 membered ring;
  • R 9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
  • DPP-TV inhibitors of the present invention comprise Formula XI:
  • J, K, L, and M are each independently selected from the group of CR ⁇ 2 and N;
  • Ri is -ZR m , where Z is a moiety providing 1-6 atom separation between R m and the ring to which Ri is attached, and R m is selected from the group consisting of a substituted or unsubstituted (C 3 - 7 )cycloalkyl and aryl;
  • R 2 is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring;
  • R 9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted; and each R12 is hydrogen or is independently selected from the group consisting of halo, perhalo(Ci-io)alkyl, CF3, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, alkoxy, a carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
  • DPP-IV inhibitors of the present invention comprise
  • Ri is -ZR m , where Z is a moiety providing 1-6 atom separation between R ra and the ring to which Ri is attached, and R m is selected from the group consisting of a substituted or unsubstituted (C 3 - 7 )cycloalkyl and aryl;
  • R 2 is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring;
  • R 9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted;
  • each R 19 is independently selected from the group consisting of hydrogen, halo, perhalo(Ci-io)alkyl, CF 3 , cyano, nitro, alkyl, alkene, alkyne, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio,
  • DPP-IN inhibitors of the present invention comprise Formula XTJJ:
  • W, X, and Y are each independently selected from the group of moieties where the ring atom is either C, N, O or S;
  • Ri is -ZRm, where Z is a moiety providing 1-6 atom separation between R m and the ring to which Ri is attached, and R m is selected from the group consisting of a substituted or unsubstituted (C3- 7 )cycloalkyl and aryl;
  • R 2 is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring;
  • R 9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
  • DPP-TV inhibitors of the present invention comprise Formulae XTVa, XTVb, and XTVc:
  • Ri is -ZR m , where Z is a moiety providing 1-6 atom separation between R m and the ring to which Ri is attached, and R m is selected from the group consisting of a substituted or unsubstituted (C3- )cycloalkyl and aryl;
  • R 2 is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring;
  • R 9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted; and each R ⁇ 9 is independently selected from the group consisting of hydrogen, halo, perhalo(Ci-io)alkyl, CF3, cyano, nitro, alkyl, alkene, alkyne, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsub
  • DPP-JV inhibitors of the present invention comprise Formulae XVa and XVb:
  • W, X, and Y are each independently selected from the group of moieties where the ring atom is either C, N, O or S;
  • Ri is -ZR m , where Z is a moiety providing 1-6 atom separation between R m and the ring to which Ri is attached, and R m is selected from the group consisting of a substituted or unsubstituted (C 3 - )cycloalkyl and aryl;
  • R 2 is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring;
  • R 9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
  • DPP-TV inhibitors of the present invention comprise Formulae XVIa, XVTb, and XVIc:
  • X is selected from the group of moieties where the ring atom is either C, N, O or S in Formula XVIa, or X is selected from the group of moieties where the ring atom is either C or N in Formula XVTb or Formula XVTc;
  • Ri is -ZR m , where Z is a moiety providing 1-6 atom separation between R m and the ring to which Ri is attached, and R m is selected from the group consisting of a substituted or unsubstituted (C 3 - )cycloalkyl and aryl;
  • R 2 is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring;
  • Rg is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted;
  • each R 19 is independently selected from the group consisting of hydrogen, halo, perhalo(C ⁇ - ⁇ o)alkyl, CF 3 , cyano, nitro, alkyl, alkene, alkyne, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio,
  • DPP-IN inhibitors of the present invention comprise Formulae XVJJa, XVJJb, and XVHc:
  • J, K, L, and M are each independently selected from the group of moieties where the ring atom is either C, N, O or S;
  • Ri is -ZR m , where Z is a moiety providing 1-6 atom separation between R m and the ring to which Ri is attached, and R m is selected from the group consisting of a substituted or unsubstituted (C3- 7 )cycloalkyl and aryl; and
  • R 2 is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring.
  • DPP-I inhibitors of the present invention comprise Formula XV : xvm wherein
  • J, K, L, and M are each independently selected from the group of moieties where the ring atom is either C, N, O or S;
  • Ri is -ZR m , where Z is a moiety providing 1-6 atom separation between R m and the ring to which.Ri is attached, and R m is selected from the group consisting of a substituted or unsubstituted (C 3 - )cycloalkyl and aryl;
  • R 2 is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring;
  • R 9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
  • DPP-TV inhibitors of the present invention comprise Formula XIX:
  • Ri is -ZR m , where Z is a moiety providing 1-6 atom separation between R m and the ring to which Ri is attached, and -R m is selected from the group consisting of a substituted or unsubstituted (C 3 - )cycloalkyl and aryl;
  • R 2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R 2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein; R 3 and R 4 are taken together to form a substituted or unsubstituted 5 or 6 membered ring; and
  • R 9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
  • DPP-IN inhibitors of the present invention comprise Formula XX:
  • J, K, L, and M are each independently selected from the group of CR 12 and N;
  • Ri is -ZR m , where Z is a moiety providing 1-6 atom separation between R m and the ring to which Ri is attached, and -R m is selected from the group consisting of a substituted or unsubstituted (C 3 - )cycloalkyl and aryl;
  • R 2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R 2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein;
  • R 9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted; and each R ⁇ 2 is hydrogen or is independently selected from the group consisting of halo, perhalo(C ⁇ - ⁇ o)alkyl, CF3, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, alkoxy, a carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
  • DPP-IV inhibitors of the present invention comprise Formulae XXIa, XXIb, XXIc, XXId, XXIe, and XXJf:
  • Ri is -ZR m , where Z is a moiety providing 1-6 atom separation between R m and the ring to which Ri is attached, and -R m is selected from the group consisting of a substituted or unsubstituted (C 3 - )cycloalkyl and aryl;
  • R 2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R 2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein;
  • R 9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted; and each R 19 is independently selected from the group consisting of hydrogen, halo, perhalo(Ci-io)alkyl, CF 3 , cyano, nitro, alkyl, alkene, alkyne, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or un
  • DPP-TV inhibitors of the present invention comprise Formula XXII:
  • W, X, and Y are each independently selected from the group of moieties where the ring atom is either C, N, O or S;
  • Ri is -ZR m , where Z is a moiety providing 1-6 atom separation between R m and the ring to which Ri is attached, and -R m is selected from the group consisting of a substituted or unsubstituted (C3- )cycloalkyl and aryl;
  • R 2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R 2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein; and
  • R 9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
  • DPP-IV inhibitors of the present invention comprise Formulae XXTHa, XXJJIb, and XXTHc:
  • Ri is -ZR m , where Z is a moiety providing 1-6 atom separation between R m and the ring to which Ri is attached, and -R m is selected from the group consisting of a substituted or unsubstituted (C 3 - )cycloalkyl and aryl;
  • R 2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R 2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein;
  • R 9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted; and each R 19 is independently selected from the group consisting of hydrogen, halo, perhalo(C ⁇ - ⁇ o)alkyl, CF 3 , cyano, nitro, alkyl, alkene, alkyne, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or un
  • DPP-IN inhibitors of the present invention comprise Formulae XXINa and XXIVb:
  • W, X, and Y are each independently selected from the group of moieties where the ring atom is either C, N, O or S;
  • Ri is -ZR m , where Z is a moiety providing 1-6 atom separation between R m and the ring to which Ri is attached, and -R m is selected from the group consisting of a substituted or unsubstituted (C 3 - 7 )cycloalkyl and aryl;
  • R 2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R 2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein; and
  • R 9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
  • DPP-IV inhibitors of the present invention comprise Formulae XXVa, XXVb, and XXVc:
  • X is selected from the group of moieties where the ring atom is either C, N, O or S in Formula XXVa, or X is selected from the group of moieties where the ring atom is either C or N in Formula XXVb or Formula XXVc;
  • Ri is -ZR m , where Z is a moiety providing 1-6 atom separation between R m and the ring to which Ri is attached, and -R m is selected from the group consisting of a substituted or unsubstituted (C 3 - 7 )cycloalkyl and aryl;
  • R 2 is -UN, where U is a moiety providing 1-6 atom separation between V and the ring to which R 2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein;
  • Rg is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted; and each R 19 is independently selected from the group consisting of hydrogen, halo, perhalo(C ⁇ - ⁇ o)alkyl, CF3, cyano, nitro, alkyl, alkene, alkyne, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, carbonyl group, irnine group, sulfonyl group and sulfinyl group, each substituted or
  • DPP-TV inhibitors of the present invention comprise Formulae XXVIa, XXVIb, and XXVIc:
  • J, K, L, and M are each independently selected from the group of moieties where the ring atom is either C, N, O or S;
  • Ri is -ZR m , where Z is a moiety providing 1-6 atom separation between R m and the ring to which Ri is attached, and -R m is selected from the group consisting of a substituted or unsubstituted (C 3 - 7 )cycloalkyl and aryl; and R 2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R 2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein.
  • DPP-TV inhibitors of the present invention comprise Formula XXVTT:
  • J, K, L, and M are each independently selected from the group of moieties where the ring atom is either C, N, O or S;
  • Ri is -ZR m , where Z is a moiety providing 1-6 atom separation between R m and the ring to which Ri is attached, and -R m is selected from the group consisting of a substituted or unsubstituted (C 3 - )cycloalkyl and aryl;
  • R 2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R 2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein; and
  • R 9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
  • DPP-TV inhibitors of the present invention comprise Formula XXVTJJ:
  • Ri is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring;
  • R 2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R 2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein;
  • R 3 and R 4 are taken together to form a substituted or unsubstituted 5 or 6 membered ring;
  • R 9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
  • DPP-IV inhibitors of the present invention comprise Formula XXIX:
  • J, K, L, and M are each independently selected from the group of CR 12 and N;
  • Ri is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring;
  • R 2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein;
  • R 9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted; and each R 12 is hydrogen or is independently selected from the group consisting of halo, perhalo(Ci-io)alkyl, CF 3 , alkyl, aryl; heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, alkoxy, a carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
  • DPP-IN inhibitors of the present invention comprise Formulae XXXa, XXXb, XXXc, XXXd, XXXe, and XXXf:
  • Ri is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring;
  • R 2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R 2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein;
  • R 9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted; and each R 19 is independently selected from the group consisting of hydrogen, halo, perhalo(C ⁇ - ⁇ o)alkyl, CF 3 , cyano, nitro, alkyl, alkene, alkyne, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or un
  • DPP-IN inhibitors of the present invention comprise Formula XXXI:
  • W, X, and Y are each independently selected from the group of moieties where the ring atom is either C, N, O or S;
  • Ri is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring;
  • R 2 is -UN, where U is a moiety providing 1-6 atom separation between N and the ring to which R 2 is attached and N comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein; and R 9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
  • DPP-IN inhibitors of the present invention comprise Formulae XXXJJa, XXXTIb, and XXXIJc:
  • Ri is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring;
  • R 2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R 2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein;
  • Rg is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted; and each Rig is independently selected from the group consisting of hydrogen, halo, perhalo(Ci-io)alkyl, CF 3 , cyano, nitro, alkyl, alkene, alkyne, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or un
  • DPP-IV inhibitors of the present invention comprise Formulae XXXJJJa and XXXITJb:
  • W, X, and Y are each independently selected from the group of moieties where the ring atom is either C, N, O or S;
  • Ri is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring;
  • R 2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R 2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein; and
  • Rg is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
  • DPP-TV inhibitors of the present invention comprise Formulae XXXJNa, XXXIVb, and XXXTNc:
  • X is selected from the group of moieties where the ring atom is either C, N, O or S in Formula XXXTVa, or X is selected from the group of moieties where the ring atom is either C or N in Formula XXXIVb or Formula XXXTVc;
  • Ri is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring;
  • R 2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R 2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein;
  • R 9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted; and each R 1 9 is independently selected from the group consisting of hydrogen, halo, perhalo(C ⁇ - ⁇ o)alkyl, CF3, cyano, nitro, alkyl, alkene, alkyne, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsub
  • DPP-TV inhibitors of the present invention comprise Formulae XXXVa, XXXVb, and XXXVc:
  • J, K, L, and M are each independently selected from the group of moieties where the ring atom is either C, N, O or S;
  • Ri is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring; and R 2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R 2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein.
  • DPP-TV inhibitors of the present invention comprise Formula XXXVI:
  • J, K, L, and M are each independently selected from the group of moieties where the ring atom is either C, N, O or S;
  • Ri is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring;
  • R2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein; and
  • R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
  • DPP-TV inhibitors of the present invention comprise Formula XXXVII:
  • J, K, L, and M are each independently selected from the group of CR12 and N, provided that at least one of K and L is CR 12 where R ⁇ 2 is not hydrogen;
  • Ri is -ZR m , where Z is a moiety providing 1-6 atom separation between R m and the ring to which Ri is attached, and R m is selected from the group consisting of a substituted or unsubstituted (C3- )cycloalkyl and aryl;
  • R 2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R 2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein;
  • Rg is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted; and each R 12 is hydrogen or is independently selected from the group consisting of halo, perhalo(Ci-io)alkyl, CF 3 , alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, alkoxy, a carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
  • DPP-TV inhibitors of the present invention comprise Formula XXXVHI:
  • J, K, L, and M are each independently selected from the group of CR ⁇ 2 and N, provided that at least one of K and L is CR 12 where R 12 is not hydrogen;
  • Ri is -ZR m , where Z is a moiety providing 1-6 atom separation between R m and the ring to which Ri is attached, and R m is selected from the group consisting of a substituted or unsubstituted (C 3 - )cycloalkyl and aryl; R 2 is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring;
  • R 9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted; and each R 12 is hydrogen or is independently selected from the group consisting of halo, perhalo(C ⁇ - ⁇ o)alkyl, CF 3 , alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, alkoxy, a carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
  • DPP-IN inhibitors of the present invention comprise Formula XXXIX:
  • J, K, L, and M are each independently selected from the group of CR 12 and N, provided that at least one of K and L is CR 12 where R 12 is not hydrogen;
  • Ri is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring;
  • R 2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R 2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein; and
  • Rg is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted; and each R12 is hydrogen or is independently selected from the group consisting of halo, perhalo(Ci-io)alkyl, CF 3 , alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, alkoxy, a carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
  • R 3 and R 4 are taken together to form an unsubstituted or substituted 5 or 6 membered cycloalkyl or heterocycloalkyl ring.
  • R 3 and R 4 are taken together to form a substituted or unsubstituted phenyl ring.
  • R3 and R 4 are taken together to form a substituted or unsubstituted heteroaryl ring.
  • R3 and R 4 are taken together to form a substituted or unsubstituted heteroaryl selected from the group consisting of substituted or unsubstituted furan, thiophene, pyrrole, pyrazole, triazole, isoxazole, oxazole, thiazole, isothiazole, oxadiazole, pyridine, pyridazine, pyrimidine, pyrazine, triazine, benzofuran, isobenzofuran, benzothiophene, isobenzothiophene, imidazole, benzimidazole, indole, isoindole, quinoline, isoquinoline, cinnoline, quinazoline, naphthyridine, pyridopyridine, quinoxaline, phthalazine, and benzothiazole.
  • R3 and R are taken together to form a 5 or 6-membered ring where the ring comprises at least one CO group.
  • R 3 and R 4 are taken together to form a 5 or 6-membered ring comprising of 1-3 nitrogen ring atoms.
  • R 3 and j are taken together to form a 5 or 6-membered ring where the ring comprises a sulfur atom.
  • the ring sulfur atom is in an oxidized form as SO or SO 2 .
  • R 3 and R 4 are taken together to form a ring system such that the compound formed is selected from the group consisting of substituted or unsubstituted 4-oxo-4H-quinazoline, 3H-pvrido[2,3-d]pyrir din-4-one, 3H-pyrido[3,2-d]pyrimidin-4-one, 3H-pyrido[3,4-d]pyrimidin-4-one and 3H-pyrido[4,3-d]pyrimidin-4-one.
  • the ring formed by R3 and R4 comprises substituents that form a ring fused to the ring formed by R 3 and R 4 .
  • the fused ring may be further substituted or unsubstituted, and may be a saturated or unsaturated ring.
  • the fused ring is a heterocyclic ring.
  • the 5 or 6 membered ring is further substituted such that a 3 , 4, 5, 6 or 7 membered bridging ring is formed with the 5 or 6 membered ring.
  • the 3, 4, 5, 6 or 7 membered ring may be further substituted or unsubstituted, and may be a saturated or unsaturated ring.
  • the 3, 4, 5, 6 or 7 membered bridged ring may be a heterocyclic ring.
  • J, K, L and M each comprises a carbon ring atom.
  • J comprises a nitrogen ring atom.
  • K comprises a nitrogen ring atom.
  • L comprises a nitrogen ring atom.
  • M comprises a nitrogen ring atom.
  • J and L each comprise a nitrogen ring atom or J and K each comprise a nitrogen ring atom.
  • K and L each comprise a nitrogen ring atom.
  • K and M each comprise a nitrogen ring atom.
  • J and M each comprise a nitrogen ring atom, or L and M each comprise a nitrogen ring atom. ;
  • At least one of J, K, L and M comprise a nitrogen ring atom.
  • at least two of J, K, L and M comprise a nitrogen ring atom.
  • at least three of J, K, L and M comprise a nitrogen ring atom.
  • at least three of J, K, L and M comprise a nitrogen ring atom.
  • the ring formed by J, K, L, and M may comprise substituents that form a ring fused or bridged (e.g., through available valencies) to the ring formed by J, K, L, and M.
  • Jn particular with respect to the case of Formula VTJJib, J and M optionally form a fused or bridged ring relative to the ring formed by J, K, L, and M.
  • K is CR12, where R ⁇ is independently selected from the group consisting of halo, perhalo(C 1 -io)alkyl, CF 3 , alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, alkoxy, a carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
  • R ⁇ is independently selected from the group consisting of halo, perhalo(C 1 -io)alkyl, CF 3 , alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl,
  • K is CR ⁇ 2 , where R 12 is independently selected from the group consisting of halo, perhalo(C ⁇ - ⁇ o)alkyl, CF 3 , cyan ⁇ , nitro, alkyl, aryloxy, heteroaryloxy, amino, and alkoxy, each substituted or unsubstituted.
  • K is CR 12 , where R ⁇ 2 is independently selected from the group consisting of heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryl, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, thio, a carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
  • K is CR 12 , where R12 is independently selected from the group consisting of chloro, bromo, fluoro, iodo, methoxy, morpholin-4-yl, and pyrrolidin- 1-yl, each substituted or unsubstituted.
  • L comprises a nitrogen ring atom.
  • L is CRu, where R12 is independently selected from the group consisting of halo, perhalo(C 1 - 1 o)alkyl, CF3, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, alkoxy, a carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
  • R12 is independently selected from the group consisting of halo, perhalo(C 1 - 1 o)alkyl, CF3, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, ary
  • L is CR12, where R 12 is independently selected from the group consisting of halo, perhalo(C ⁇ - ⁇ o)alkyl, CF3, cyano, nitro, alkyl, aryloxy, heteroaryloxy, amino, morpholin-
  • J, K, L and M at least one of J, K, L and M is SO.
  • at least one of J, K, L and M is SO 2 .
  • J and M form a bridged ring relative to the ring formed by J, K, L, and M
  • K and L are independently CR 12 , where R 12 is independently selected from the group consisting of halo, perhalo(C 1 - 1 o)alkyl, CF 3 , cyano, nitro, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, a carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
  • R 12 is independently selected from the group consisting of halo, perhalo(C 1 - 1 o)alkyl, CF 3 , cyano, nitro, alkyl, aryl, heteroaryl, aminosulfon
  • K is CR 12 , where R 12 is independently selected from the group consisting of halo, perhalo(Ci-io)alkyl, CF 3 , cyano, nitro, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, a carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted; and
  • At least one R ⁇ 2 is fluorine.
  • At least one of W, X, and Y is CO. In another variation, at least one of W, X, and Y is SO. In another variation, at least one of W, X, and Y is SO 2 . In one variation of each of the above embodiments and variations comprising W, W is selected from the group consisting of CO, SO or SO 2 . In one variation of each of the above embodiments and variations comprising Y, Y is selected from the group consisting of CO, SO or SO 2 . [0181] In another variation, at least one of W, X, and Y comprises a ring nitrogen atom. In yet another variation, at least two of W, X, and Y comprises a ring nitrogen atom. In yet another variation, W comprises a ring nitrogen atom. In still variation, X comprises a ring nitrogen atom. In a further variation, Y comprises a ring nitrogen atom.
  • W and Y are taken together to form a substituted or unsubstituted bridged ring relative to the ring formed by W, X and Y.
  • at least two (t'.e., two or three) of W, X, and Y are taken together to form a substituted or unsubstituted ring fused or bridged (e.g., by substitution through available valencies) to the ring formed by W, X and Y.
  • W, X, and Y are selected such that the compound comprises a ring system selected from the group consisting of 4-oxo-4H-tMeno[3,2-d]pvrimidine, 7-oxo- l,2,3,7-tefrahydro-8-tMa-4,6-diaza-cyclopenta[a]mdene,7-memyl-6-oxo-6,7-dmydro-p ⁇ rm and 6-oxo-6,9-dihydro-purine, each substituted or unsubstituted.
  • a ring system selected from the group consisting of 4-oxo-4H-tMeno[3,2-d]pvrimidine, 7-oxo- l,2,3,7-tefrahydro-8-tMa-4,6-diaza-cyclopenta[a]mdene,7-memyl-6-oxo-6,7-dmydro-p ⁇ rm and 6-oxo-6,
  • Ri is selected from the group consisting of -NH2, -NHCH3, -N(CH 3 ) 2 , -NHC ⁇ -3-alkyl, and -N(C ⁇ -3-alkyl) 2 , each unsubstituted or substituted through available valencies.
  • Ri is selected from the group consisting of a ( .
  • alkyl an amino, alkyl, or alkoxy carbonyl (C ⁇ -3 )alkyl; an amino, alkyl, or alkoxy thiocarbonyl (Ci ⁇ alkyl; an amino, alkyl, or alkoxy sulfonyl (C ⁇ -3)alkyl; an amino, alkyl, or alkoxy sulfinyl (C ⁇ - 3 )alkyl; an amino, alkyl, or alkoxy imino (Ci ⁇ alkyl; and an aryl ( . 5 )alkyl, each unsubstituted or substituted through available valencies.
  • Ri is selected from the group consisting of a (C 2 - ⁇ o)alkenyl; an amino, alkyl, or alkoxy carbonyl (Ci ⁇ alkenyl; an amino, alkyl, or alkoxy thiocarbonyl ( . 3 )alkenyl; an amino, alkyl, or alkoxy sulfonyl (Ci ⁇ alkenyl; an amino, alkyl, or alkoxy sulfinyl and a heteroaryl ( . 5 ) alkenyl, each unsubstituted or substituted through available valencies.
  • Ri is selected from the group consisting of a (C 2- io)alkynyl; an amino, alkyl, or alkoxy carbonyl (Ci ⁇ alkynyl; an amino, alkyl, or alkoxy thiocarbonyl (C ⁇ . 3 )alkynyl; an amino, alkyl, or alkoxy sulfonyl an amino, alkyl, or alkoxy sulfinyl ( . 3 )alkynyl; an amino, alkyl, or alkoxy imino an aryl ( ..
  • Ri is substituted or unsubstituted (C 3 . 7 )cycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted phenyl.
  • Ri is hydroxy. Jn yet another variation, Ri is
  • Rn is selected from the group consisting of substituted or unsubstituted alkyl, cycloalkyl, aryl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl.
  • Ri is a carbonyl group.
  • Rl is selected from the group consisting of an aldehyde, acid, amide, and ester.
  • Ri is selected from the group consisting of py ⁇ olidin- 1 - ylcarbonyl, piperidin-1 -ylcarbonyl, and morpholin-4-ylcarbonyl, each unsubstituted or substituted through available valencies.
  • Ri is -Z-R m , where
  • Z is a moiety having a chain of 1-6 atoms selected from the group consisting of substituted or unsubstituted C, N, O, and S, attaching -R m to the ring, or Z is a unsubstituted or substituted -(Ci- 3 )alkylene optionally interrupted by O, N(O), -NH, SO, SO 2 , or carbonyl; and
  • -R m is selected from the group consisting of a substituted or unsubstituted (Ci- )alkyl, -(C 2 - 7 )alkene, -(C 2 - 7 )alkyne, -(C 3 - 7 )cycloalkyl, and aryl.
  • Z is a moiety providing 1-6 atom separation between R m and the ring to which Ri is attached, and
  • R m is selected from the group consisting of a substituted or unsubstituted (C 3 - 7 )cycloalkyl and aryl.
  • the 1-6 atoms of Z comprise of carbon atoms.
  • the 1-6 atoms of Z are selected from the group consisting of at least one oxygen, or at least one nitrogen atom.
  • Z comprises of 1 atom.
  • Z is selected from the group consisting of -CH 2 -, -CH 2 CH 2 -, -CH2CH2CH2-, -C(O)-, -CH 2 C(O)-, -C(O)CH 2 -, -CH 2 -C(O)CH 2 -, -C(O)CH 2 CH 2 -, -CH 2 CH 2 C(O)-, -O-, -OCH2-, -CH 2 O-, -CH 2 OCH 2 -, -OCH 2 CH 2 -, -CH 2 CH 2 O-, -N(CH 3 )-, -NHCH2-, -CH2NH-, -CH2NHCH2-, -NHCH 2 CH 2 -, -CH 2 CH 2 NH-, -NH-C(O)-, -NCH 3 -C(O)-, -C(O)NH-, -C(O)NCH 3 -, -
  • Z is selected from the group consisting of -CH 2 -, -C(O)-, -CH 2 C(O)-, -C(O)CH 2 -, -CH 2 -C(O)CH 2 -, -C(O)CH 2 CH 2 -, and -CH 2 CH 2 C(O)-, each substituted or unsubstituted.
  • Z provides 1-3 atom separation between R m and the ring.
  • Z provides 1 atom separation between R m and the ring.
  • the 1 atom separation is provided by an atom selected from the group consisting of C, N, O, and S.
  • the 1 atom separation is provided by a carbon atom.
  • the 1 atom separation is provided by an oxygen atom.
  • the 1 atom separation is provided by a nitrogen atom.
  • Z is selected from the group consisting of -CH 2 -, -CHR 9 -, -C(R 9 )(R 9 )-, -C(O)-, -C(S)-, -C(NH)-, -C(NR 9 )-, -O-, -N(H)-, -N(R 9 )-, and -S-.
  • Z is a carbonyl
  • R m is a substituted or unsubstituted (C3 -7 )cycloalkyl.
  • R m is a substituted or unsubstituted aryl.
  • R m is a substituted or unsubstituted phenyl.
  • R m is a ring having a non-hydrogen substituent at a 2 or 3 position of the ring.
  • R m has a non- hydrogen substituent at a 2 or 3 position of the ring selected from.the group consisting of ( - 10 )alkyl, (C 3- ⁇ 2 )cycloalkyl, hetero(C 3 - 12 )cycloalkyl, aryl(C ⁇ - ⁇ o)alkyl, heteroaryl(Ci-5)alkyl, (C 9- i 2 )bicycloaryl, hetero(C - ⁇ 2 )bicycloaryl, carbonyl (Ci ⁇ alkyl, thiocarbonyl (Ci ⁇ alkyl, sulfonyl (C ⁇ -3)alkyl, sulfinyl imino (C ⁇ .
  • R m is selected from the group consisting of alkyl, cycloalkyl, aryl, arylalkyl, heteroarylalkyl, and bicycloaryl, each substituted or unsubstituted.
  • R m is selected from the group consisting of (2-cyano)phenyl, (3-cyano)phenyl, (2- hydroxy)phenyl, (3-hydroxy)phenyl, (2-alkenyl)phenyl, (3-alkenyl)phenyl, (2-alkynyl)phenyl, (3-alkynyl)phenyl, (2-nitro)phenyl, (3-nitro)phenyl, (2-carboxy)phenyl, (3-carboxy)phenyl, (2-carboxamido)phenyl, (3-carboxamido) ⁇ henyl, (2-sulfonamido)phenyl,
  • Ri is -(CR 5 R 6 )o -2 -(R 7 ) 0 . 2 -R 8 , where
  • R5 and R ⁇ are each independently hydrogen, halo or substituted or unsubstituted (C ⁇ -8 )alkyl or R 5 and R 6 are taken together to form a (C 3 - 6 )cycloalkyl ring;
  • R 7 is selected from the group consisting of CO; CS; (C ⁇ - ⁇ o)alkylene, hetero(C3- ⁇ 2 )cycloalkylene, amino, arylene, heteroarylene, imine, sulfonyl, and sulfinyl, each substituted or unsubstituted; and
  • R 8 is hydrogen or is selected from the group consisting of (C ⁇ - 7 )alkyl, (C 2 - 7 )alkenyl, (C 2 - 7 )alkynyl, (C3- 7 )cycloalkyl, (C 3 - 7 )heterocycloalkyl, aryl, aryl(Ci- 3 )alkyl, heteroaryl(C ⁇ - 2 )alkyl, heteroaryl, -SH, (C ⁇ - 7 )alkyl-S-, (Ci- 7 )alkyl-SO-, (C ⁇ - 7 )alkyl-SO 2 -, CO, CS, NHCONNH2, -NHCSNH2, -NHCONH2, -NHCO(C ⁇ - 7 )alkyl, -NHS02(C ⁇ - 7 )alkyl, -OCO-(Q- 5 )alkyl, cyano, nitro, halo, hydroxy, perhalo(C ⁇ -
  • Ri is selected from the group consisting of -(CH 2 )-(2-cyano) ⁇ henyl, -(CH 2 )-(3-cyano)phenyl, -(CH 2 )-(2-hydroxy)phenyl, -(CH 2 )-(3- hydroxy) ⁇ henyl, -(CH 2 )-(2-alkenyl)phenyl, -(CH 2 )-(3-alkeny ⁇ ) ⁇ henyl, -(CH 2 )-(2- alkynyl)phenyl, -(CH2)-(3-alkynyl) ⁇ henyl, -(CH 2 )-(2-nitro)phenyl, -(CH 2 )-(3-nitro)phenyl, -(CH 2 )-(2-carboxy)phenyl, -(CH 2 )-(3-carboxy)phenyl, -(CH 2 )-(2-carboxamido)
  • -(CH 2 )-(3-CO 2 (C ⁇ - 7 )alkyl)phenyl -CH 2 -NH 2 , -CH 2 -OH, -CH 2 -(C 3 - 7 )alkyl, -CH 2 -alkene, -CH 2 -alkyne, -CH 2 -CCH, -CH 2 -(C 3 - 7 )cycloalkyl, and -CH 2 -aryl, each substituted or unsubstituted.
  • Ri is selected from the group consisting of -(C ⁇ )alkyl-aryl, -(C ⁇ )alkyl-bicycloaryl, -aminoaryl, -aminoheteroaryl, -aminobicycloaryl, -aminoheterobicycloaryl, -O-aryl, -O- heteroaryl, -O-bicycloaryl, -O-heterobicycloaryl, -(S)-aryl, -(S)-heteroaryl, -(S)-bicycloaryl, - S-heterobicycloaryl, -C(O)-aryl, -C(O)-heteroaryl, - ., C(O)-bicycloaryl, -C(O)- heterobicycloaryl, -C(S)-aryl, -C(S)-heteroaryl, - ., C(O)-bicycloaryl,
  • Ri is a substituted or unsubstituted aryl. In a further variation of each of the above embodiments and variations comprising Ri, Ri is a substituted or unsubstituted phenyl. In yet another variation of each of the above embodiments and variations comprising Ri, Ri is a substituted or unsubstituted heteroaryl.
  • R 2 is selected from the group consisting of halo, perhalo(C ⁇ - ⁇ o)alkyl, amino, cyano, thio, (Cr ⁇ o)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, carbonyl thiocarbonyl (C ⁇ . 3 )alkyl, sulfonyl (C ⁇ -3)alkyl, sulfinyl (Ci.
  • R 2 is selected from the group consisting of (C 3 -i 2 )cycloalkyl(C ⁇ .
  • R 2 is selected from the group consisting of I, Br, CI, and F.
  • R 2 is selected from the group consisting of -SH, -SCH3, and -S(C ⁇ . 3 )alkyl unsubstituted or substituted through available valencips.
  • R 2 is selected from the group consisting of -NH 2 , -NHCH 3 , -N(CH 3 ) 2 , -NH(C ⁇ -3 )alkyl, -N(C ⁇ .3-alkyl) 2 , -NR14R15, where R M is selected from the group consisting of a substituted or unsubstituted (Cs- ⁇ cycloalkyl, hetero(C 3 . ⁇ 2 )cycloalkyl, (C 6 - ⁇ 2 )aryl, hetero(C 5 - ⁇ 2 )aryl, (C 9- ⁇ 2 )bicycloaryl and hetero(C 4- ⁇ 2 )bicycloaryl, and R 15 is selected from the group consisting of hydrogen and a substituted or unsubstituted (C ⁇ -8 )alkyl, and -NR 1 0R11, where Rio and -Rn together are -(CEJ ⁇ s- optionally interrupted by one O,
  • R 2 is selected from the group consisting of a (Ci ⁇ alkyl; an amino, alkyl, or alkoxy carbonyl (Ci ⁇ alkyl; an amino, alkyl, or alkoxy thiocarbonyl (C ⁇ -3 )alkyl; an amino, alkyl, or alkoxy sulfonyl (C ⁇ .3)alkyl; an amino, alkyl, or alkoxy sulfinyl (C ⁇ .
  • R 2 is selected from the group consisting of a (C 2 - ⁇ o)alkenyl; an amino, alkyl, or alkoxy carbonyl (C ⁇ -3)alkenyl; an amino, alkyl, or alkoxy thiocarbonyl (C ⁇ -3 )alkenyl; an amino, alkyl, or alkoxy sulfonyl (Q.
  • alkenyl an amino, alkyl, or alkoxy sulfinyl (Ci ⁇ alkenyl; an amino, alkyl, or alkoxy imino (C ⁇ -3)alkenyl; an aryl (C ⁇ - 5 )alkenyl, and a heteroaryl (C ⁇ - 5 )alkenyl, each unsubstituted or substituted through available valencies.
  • R 2 is selected from the group consisting of a (C 2 - ⁇ o)alkynyl; an amino, alkyl, or alkoxy carbonyl (C ⁇ _3)alkynyl; an amino, alkyl, or alkoxy thiocarbonyl (C ⁇ . 3 )alkynyl; an amino, alkyl, or alkoxy sulfonyl ( . 3)alkynyl; an amino, alkyl, or alkoxy sulfinyl (Ci ⁇ alkynyl; an amino, alkyl, or alkoxy imino (C ⁇ . 3 )alkynyl; an aryl (C 1 - 5 ) alkynyl; and a heteroaryl (C ⁇ -5)alkynyl, each unsubstituted or substituted through available valencies.
  • a (C 2 - ⁇ o)alkynyl an amino, alkyl, or alkoxy carbonyl (C ⁇ _3)alky
  • R 2 is a substituted or unsubstituted 3, 4, 5, 6, or 7 membered ring.
  • R 2 is substituted or unsubstituted (C3 -7 )cycloalkyl (i.e., a substituted or unsubstituted 3, 4, 5, 6, or 7 membered cycloalkyl).
  • R 2 is substituted or unsubstituted (C 3-7 )heterocycloalkyl (e.g., a substituted or unsubstituted 4, 5,
  • R 2 is substituted or unsubstituted aryl. In another variation, R 2 is substituted or unsubstituted phenyl. In yet another variation,
  • R 2 is substituted or unsubstituted heteroaryl.
  • R 2 is hydroxy
  • R 2 is -ORn, where Rn is selected from the group consisting of substituted or unsubstituted alkyl, cycloalkyl, aryl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl.
  • R 2 is a carbonyl group.
  • R 2 is selected from the group consisting of an aldehyde, acid, amide, ketone and ester.
  • R 2 is selected from the group consisting of pyrrolidin- 1 -ylcarbonyl, piperidin-1 -ylcarbonyl, and morpholin-4- ylcarbonyl, each unsubstituted or substituted through available valencies.
  • R 2 is selected from the group consisting of 3-amino-piperidin-l-yl, 3-aminomethyl-pyrrolidin-l-yl, azetidin-1-yl, 3- aminoazetidin-1-yl, pyrrolidin- 1-yl, 3-aminocyclopent-l-yl, 3-aminomethylcyclopent-l-yl, 3- aminomethylcyclohex-1-yl, hexahydroazepin-1-yl, 3-aminohexahydroazepin-l-yl, 3-amino- cyclohex-1-yl, piperazin- 1-yl, homopiperazin- 1-yl, 3-amino-pyrrolidin-l-yl, and R-3- aminopiperidin-1-yl, each substituted or unsubstituted.
  • R 2 is MR a Rb, where
  • M is CH or N, R a is H or (C ⁇ -6 )alkyl, and R is amino(Co 4 )alkyl; or
  • R 2 comprises a primary or secondary amine where the nitrogen of the arnine is 1-6 atoms from the nearest ring atom.
  • R 2 comprises a primary or secondary amine where the nitrogen of the amine is 2-5 atoms from the nearest ring atom.
  • R 2 comprises a primary or secondary amine where the nitrogen of the amine is 3-4 atoms from the nearest ring atom.
  • R 2 is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring wherein at least one substituent is selected from the group consisting of a primary, secondary or tertiary amine, a heterocycloalkyl comprising a nitrogen ring atom, and a heteroaryl comprising a nitrogen ring atom.
  • R 2 is selected from the group consisting of
  • each R 8 is independently selected from the group consisting of halo, perhalo(C ⁇ - ⁇ o)alkyl, CF3, cyano, nitro, hydroxy, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
  • R 2 is selected from the group consisting of
  • each R 8 is independently selected from the group consisting of halo, perhalo(C ⁇ - ⁇ o)alkyl, CF3, cyano, nitro, hydroxy, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
  • R 2 , R 2 is a substituted or unsubstituted heteroaryl selected from the group consisting of furan, thiophene, pyrrole, pyrazole, triazole, isoxazole, oxazole, thiazole, isothiazole, oxadiazole, pyridine, pyridazine, pyrimidine, pyrazine, triazine, benzofuran, isobenzofuran, benzothiophene, isobenzothiophene, imidazole, benzimidazole, indole, isoindole, quinoline, isoquinoline, cinnoline, quinazoline, naphthyridine, pyridopyridine, quinoxaline, phthalazine, and benzothiazole, each substituted or unsubstituted.
  • R 2 is selected from the group consisting of (C 3 .. ⁇ 2 )cycloalkyl, hetero(C 3 -i 2 )cycloalkyl, aryl(C ⁇ - ⁇ o)alkyl, heteroaryl (C ⁇ -5)alkyl, (Cg- ⁇ bicycloaryl, and hetero(C 4 .i 2 )bicycloaryl, each substituted or unsubstituted.
  • R 2 , R 2 is a substituted or unsubstituted (C 3 - )cycloalkyl ring, optionally comprising O, N(O),
  • R 1 9, two Rig are taken together to form a substituted or unsubstituted fused or bridged ring.
  • U provides 1-4 atom separation between V and the ring. In another variation, U provides 1-3 atom separation between V and the ring.
  • U is selected from the group consisting of -CH 2 -,
  • U is selected from the group consisting of -CH 2 -, -CHR 9 -, -C(R 9 )(R 9 )-, -O-, -N(H)-,
  • V is selected from the group consisting of a primary, secondary or tertiary amine, a heterocycloalkyl comprising a nitrogen ring atom, and a heteroaryl comprising a nitrogen ring atom
  • V is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring wherein at least one substituent is selected from the group consisting of a primary, secondary or tertiary amine, a heterocycloalkyl comprising a nitrogen ring atom, and a heteroaryl comprising a nitrogen ring atom.
  • V the basic nitrogen of V forms part of a primary, secondary or tertiary amine.
  • the basic nitrogen of V is a nitrogen ring atom of a heterocycloalkyl comprising a nitrogen ring atom or a heteroaryl comprising a nitrogen ring atom.
  • -UV, -UV is selected from the group consisting of
  • each R 8 is independently selected from the group consisting of halo, perhalo(C ⁇ - ⁇ o)alkyl, CF 3 , cyano, nitro, hydroxy, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, with the proviso that at least one R 8 serves as V.
  • -UV is selected from the group consisting of
  • each R 8 is independently selected from the group consisting of halo, perhalo(C ⁇ - ⁇ o)alkyl, CF 3 , cyano, nitro, hydroxy, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, with the proviso that at least one R 8 serves as V.
  • At least one R 8 is a primary, secondary or tertiary amine.
  • R 8 at least one R 8 is a substituted or unsubstituted heterocycloalkyl comprising a nitrogen ring atom or a substituted or unsubstituted heteroaryl comprising a nitrogen ring atom.
  • At least one R 8 is selected from the group consisting of -NH 2 , -NH(C ⁇ _s alkyl), -
  • N(C ⁇ - 5 alkyl) 2 piperazine, imidazole, and pyridine.
  • Ri is selected from the group consisting of -(CH 2 )-(2-cyano)phenyl,
  • -(CH 2 )-(3-CO 2 (Ci- 7 )alkyl)phenyl -CH 2 -NH 2 , -CH 2 -OH, -CH 2 -(C 3 - 7 )alkyl, -CH 2 -alkene, -CH 2 -alkyne, -CH 2 -CCH, -CH 2 -(C 3 - 7 )cycloalkyl, and -CH 2 -aryl, each substituted or unsubstituted, and
  • R 2 is selected from the group consisting of 3-amino-piperidin-l-yl, 3-aminomethyl- pyrrolidin-1-yl, azetidin-1-yl, 3-aminoazetidin-l-yl, pyrrohdin-1-yl, 3-aminocyclopent-l-yl, 3- aminomethylcyclopent-1-yl, 3-aminomethylcyclohex-l-yl, hexahydroazepin-1-yl, 3- aminohexahydroazepin-1-yl, 3-amino-cyclohex-l-yl, piperazin- 1-yl, homopiperazin- 1-yl, 3- amino-pyrrolidin-1-yl, and R-3-aminopiperidin-l-yl, each substituted or unsubstituted. [0246] In another particular variation according to the present invention,
  • Ri is selected from the group consisting of -NH 2 , -NHCH 3 , -N(CH 3 )2, -NHCi- 3 -alkyl, and -N(C ⁇ -3-alkyl) 2 each unsubstituted or substituted through available valencies, and
  • R2 is selected from the group consisting of 3-amino-piperidin-l-yl, 3-aminomethyl- pyrrolidin-1-yl, azetidin-1-yl, 3-aminoazetidin-l-yl, pyrrolidin- 1-yl, 3-aminocyclopent-l-yl, 3- aminomethylcyclopent-1-yl, 3-aminomethylcyclohex-l-yl, hexahydroazepin-1-yl, 3- aminohexahydroazepin-1-yl, 3-amino-cyclohex-l-yl, piperazin- 1-yl, homopiperazin- 1-yl, 3- amino-pyrrolidin-1-yl, and R-3-aminopiperidin-l-yl, each substituted or unsubstituted. [0247] In another variation according to the present invention,
  • Ri is selected from the group consisting of -(CH 2 )-(2-cyano)phenyl, -(CH 2 )-(3-cyano)phenyl, -(CH )-(2-hydroxy)phenyl, -(CH 2 )-(3-hydroxy)phenyl, -(CH 2 )-(2- alkenyl)phenyl, -(CH 2 )-(3-alkenyl)phenyl, -(CH 2 )-(2-alkynyl)phenyl, -(CH 2 )-(3- alkynyl) ⁇ henyl, -(CH 2 )-(2-nitro)phenyl, -(CH 2 )-(3-nitro) ⁇ henyl, -(CH 2 )-(2-carboxy) ⁇ henyl, -(CH 2 )-(3-carboxy)phenyl, -(CH 2 )-(2-carboxamido)phenyl, -(CH 2
  • -(CH 2 )-(3-CO 2 (C ⁇ - 7 )alkyl)phenyl -CH 2 -NH 2 , -CH 2 -OH, -CH 2 -(C 3 - 7 )alkyl, -CH 2 -alkene, -CH 2 -alkyne, -CH 2 -CCH, -CH 2 -(C 3 - 7 )cycloalkyl, and -CH 2 -aryl, each substituted or unsubstituted, and
  • R 2 is selected from the group consisting of 3-amino-piperidin-l-yl, 3-aminomethyl- pyrrolidin-1-yl, azetidin-1-yl, 3-aminoazetidin-l-yl, pyrrolidin- 1-yl, 3-aminocyclopent-l-yl, 3- aminomethylcyclopent-1-yl, 3-aminomethylcyclohex-l-yl, hexahydroazepin-1-yl, 3- aminohexahydroazepin-1-yl, 3-amino-cyclohex-l-yl, piperazin- 1-yl, homopiperazin- 1-yl, 3- amino-py ⁇ Olidin-1-yl, and R-3-aminopiperidin-l-yl, each substituted or unsubstituted. [0248] In another variation according to the present invention,
  • Ri is selected from the group consisting of -(CH 2 )-(2-cyano)phenyl, -(CH 2 )-(3-cyano) ⁇ henyl, -(CH 2 )-(2-hydroxy) ⁇ henyl, -(CH 2 )-(3-hydroxy)phenyl, -(CH 2 )-(2- alkenyl)phenyl, -(CH 2 )-(3-alkenyl)phenyl, -(CH 2 )-(2-alkynyl)phenyl, -(CH 2 )-(3- alkynyl)phenyl, -(CH 2 )-(2-nitro)phenyl, -(CH 2 )-(3-nitro)phenyl, -(CH 2 )-(2-carboxy)phenyl, -(CH 2 )-(3-carboxy)phenyl, -(CH 2 )-(2-carboxamido)phenyl, -(CH 2
  • -(CH 2 )-(3-CO 2 (C ⁇ - 7 )alkyl)phenyl -CH 2 -NH 2 , -CH 2 -OH, -CH 2 -(C 3 - 7 )alkyl, -CH 2 -alkene, -CH 2 -alkyne, -CH 2 -CCH, -CH 2 -(C 3 - )cycloalkyl, and -CH 2 -aryl, each substituted or unsubstituted, and
  • R 2 is selected from the group consisting of 3-amino-piperidin-l-yl, 3-aminomethyl- pyrcolidin-1-yl, azetidin-1-yl, 3-ammoazetidin-l-yl, pyrrohdin-1-yl, 3-aminocyclopent-l-yl, 3- aminomethylcyclopent-1-yl, 3-aminomethylcyclohex-l-yl, hexahydroazepin-1-yl, 3- aminohexahydroazepin-1-yl, 3-amino-cyclohex-l-yl, piperazin- 1-yl, homopiperazin- 1-yl, 3- ammo-pyrrolidin-1-yl, and R-3-aminopiperidin-l-yl, each substituted or unsubstituted. [0249] In yet another variation according to the present invention,
  • Ri is selected from the group consisting of-(CH 2 )-(2-cyano)phenyl, -(CH 2 )-(3-cyano)phenyl, -(CH 2 )-(2-hydroxy)phenyl, -(CH 2 )-(3-hydroxy)phenyl, -(CH 2 )-(2- alkenyl) ⁇ henyl, -(CH 2 )-(3-alkenyl)phenyl, -(CH 2 )-(2-alkynyl)phenyl, -(CH 2 )-(3- alkynyl)phenyl, -(CH 2 )-(2-nitro)phenyl, -(CH 2 )-(3-nitro)phenyl, -(CH 2 )-(2-carboxy)phenyl, -(CH 2 )-(3-carboxy)phenyl, -(CH 2 )-(2-carboxarmdo)phenyl, -(CH 2 )
  • -(CH 2 )-(3-CO 2 (Ci- 7 )alkyl)phenyl -CH 2 -NH 2 , -CH 2 -OH, -CH 2 -(C 3 - 7 )alkyl, -CH 2 -alkene, -CH 2 -alkyne, -CH 2 -CCH, -CH 2 -(C 3 - )cycloalkyl, and -CH 2 -aryl, each substituted or unsubstituted, and
  • R 2 is selected from the group consisting of 3-amino-piperidin-l-yl, 3-aminomethyl- pyrrohdin-1-yl, azetidin-1-yl, 3-aminoazetidin-l-yl, pyrroUdin-1-yl, 3-aminocyclopent-l-yl, 3- aminomethylcyclopent-1-yl, 3-aminomethylcyclohex-l-yl, hexahydroazepin-1-yl, 3- aminohexahydroazepin-1-yl, 3-amino-cyclohex-l-yl, piperazin- 1-yl, homopiperazin- 1-yl, 3- amino-py ⁇ olidin-1-yl, and R-3-aminopiperidin-l-yl, each substituted or unsubstituted. [0250] In yet another variation according to the present invention,
  • Q is CO
  • Ri is selected from the group consisting of -NH 2 , -NHCH 3 , -N(CH 3 ) 2 , -NHC ⁇ . 3 -alkyl, and -N(C ⁇ - 3 -alkyl) 2 each unsubstituted or substituted through available valencies, and
  • R 2 is selected from the group consisting of 3-amino-piperidin-l-yl, 3-aminomethyl- pyrrohdin-1-yl, azetidin-1-yl, 3-aminoazetidin-l-yl, pyrrohdin-1-yl, 3-aminocyclopent-l-yl, 3- aminomethylcyclopent-1-yl, 3-aminomethylcyclohex-l-yl, hexahydroazepin-1-yl, 3- aminohexahydroazepin-1-yl, 3-amino-cyclohex-l-yl, piperazin- 1-yl, homopiperazin- 1-yl, 3- amino-pyrrolidin-1-yl, and R-3-aminopiperidin-l-yl, each substituted or unsubstituted. [0251] In another variation according to the present invention,
  • Ri is selected from the group consisting of -NH 2 , -NHCH3, -N(CH 3 ) 2 , -NHC 1 . 3 -a.kyl, and -N(Ci- 3 -alkyl) 2 each unsubstituted or substituted through available valencies, and
  • R 2 is selected from the group consisting of 3-amino-piperidin-l-yl, 3-aminomethyl- pyrcolidin-1-yl, azetidin-1-yl, 3-aminoazetidin-l-yl, pyrrolidin- 1-yl, 3-aminocyclopent-l-yl, 3- aminomethylcyclopent-1-yl, 3-aminomethylcyclohex-l-yl, hexahydroazepin-1-yl, 3- aminohexahydroazepin-1-yl, 3-amino-cyclohex-l-yl, piperazin- 1-yl, homopiperazin- 1-yl, 3- amino-pyrrolidin-1-yl, and R-3-aminopiperidin-l-yl, each substituted or unsubstituted. [0252] In another variation according to the present invention,
  • Ri is selected from the group consisting of -NH 2 , -NHCH 3 , -N(CH 3 ) 2 , -NHC ⁇ . 3 -alkyl, and -N(C ⁇ - 3 -alkyl) 2 each unsubstituted or substituted through available valencies, and
  • R 2 is selected from the group consisting of 3-amino-piperidin-l-yl, 3-aminomethyl- pyrcolidin-1-yl, azetidin-1-yl, 3-aminoazetidin-l-yl, pyrrolidin- 1-yl, 3-aminocyclopent-l-yl, 3- aminomethylcyclopent-1-yl, 3-aminomethylcyclohex-l-yl, hexahydroazepin-1-yl, 3- aminohexahydroazepin-1-yl, 3-amino-cyclohex-l-yl, piperazin- 1-yl, homopiperazin- 1-yl, 3- amino-pyrrolidin-1-yl, and R-3-aminopiperidin-l-yl, each substituted or unsubstituted.
  • DPP-TV inhibitors include:
  • the present invention provides the compounds in the form of a pharmaceutically acceptable salt.
  • the present invention provides the compounds present in a mixture of stereoisomers. In yet another embodiment, the present invention provides the compounds as a single stereoisomer.
  • the present invention provides pharmaceutical compositions comprising the compound as an active ingredient.
  • the present invention provides pharmaceutical compositions wherein the composition is a solid formulation adapted for oral administration.
  • the present invention provides pharmaceutical composition wherein the composition is a tablet.
  • the present invention provides the pharmaceutical composition wherein the composition is a liquid formulation adapted for oral administration.
  • the present invention provides pharmaceutical composition wherein the composition is a liquid formulation adapted for parenteral administration.
  • the present invention provides the pharmaceutical composition comprising the compound of the invention wherein the composition is adapted for administration by a route selected from the group consisting of orally, parenterally, intraperitoneally, intravenously, intraarterially, transdermally, sublingually, intramuscularly, rectally, transbuccally, intranasally, liposomally, via inhalation, vaginally, intraoccularly, via local delivery (for example by catheter or stent), subcutaneously, intraadiposally, intraarticularly, and intrathecally.
  • the present invention provides a kit comprising a compound of the present invention and instructions which comprise one or more forms of information selected from the group consisting of indicating a disease state for which the compound is to be administered, storage information for the compound, dosing information and instructions regarding how to administer the compound.
  • the present invention provides the kit that comprises the compound in a multiple dose form.
  • the present invention provides an article of manufacture comprising a compound of the present invention, and packaging materials.
  • the packaging material comprises a container for housing the compound.
  • the invention provides the article of manufacture wherein the container comprises a label indicating one or more members of the group consisting of a disease state for which the compound is to be administered, storage information, dosing information and/or instructions regarding how to administer the composition.
  • the present invention provides the article of manufacture wherein the article of manufacture comprises the compound in a multiple dose form.
  • the present invention provides a method of inhibiting DPP-IN comprising contacting DPP-IN with a compound according to the present invention.
  • the present invention provides a method of inhibiting DPP-IN comprising causing a compound according to the present invention to be present in a subject in order to inhibit DPP-IN in vivo.
  • the present invention provides a method of inhibiting DPP-IN comprising: administering a first compound to a subject that is converted in vivo to a second compound wherein the second compound inhibits DPP-IN in vivo, the second compound being a compound of the present invention.
  • the present invention provides therapeutic method comprising: administering a compound according to the present invention to a subject.
  • the present invention provides a method of treating a disease state for which DPP-IN possesses activity that contributes to the pathology and/or symptomology of the disease state, the method comprising causing a compound of the present invention to be present in a subject in a therapeutically effective amount for the disease state.
  • the present invention provides a method of treating cancer in a patient in need thereof, comprising administering to said patient a therapeutically effective amount of a compound according to the present invention.
  • the present invention provides a method of treating a disease where the disease is type I or type U diabetes.
  • the present invention provides a method of treating autoimmune disorders such as, but not limited to, rheumatoid arthritis, psoriasis, and multiple sclerosis in a patient in need thereof, comprising administering to said patient a therapeutically effective amount of a compound according to the present invention.
  • the present invention provides a method of treating cancer where the cancer treated is colorectal, prostate, breast, thyroid, skin, lung, or head and neck.
  • the present invention provides a method of treating a condition characterized by inadequate lymphocyte or hemapoietic cell activation or concentration in a patient in need thereof, comprising administering to said patient a therapeutically effective amount of a compound according to the present invention.
  • the present invention provides a method of treating HTV infection in a patient in need thereof, comprising admimstering to said patient a therapeutically effective amount of a compound according to the present invention.
  • the present invention provides a method of treating a condition characterized by inadequate lymphocyte or hemapoietic cell activation or concentration in a patient in need thereof, wherein the condition is a side effect of chemotherapy or radiation therapy.
  • the present invention provides a method of treating a condition characterized by inadequate lymphocyte or hemapoietic cell activation or concentration in a patient in need thereof, wherein the condition is a result of kidney failure.
  • the present invention provides a method of treating a condition characterized by inadequate lymphocyte or hemapoietic cell activation or concentration in a patient in need thereof, wherein the condition is a result of a bone marrow disorder.
  • the present invention provides a method of treating a condition characterized by immunodeficiency symptoms in a patient in need thereof, comprising adrninistering to said patient a therapeutically effective amount of a compound according to the present invention.
  • the compounds of the present invention may be present and optionally administered in the form of salts, hydrates and prodrugs that are converted in vivo into the compounds of the present invention.
  • the compounds of the present invention possess a free base form the compounds can be prepared as a pharmaceutically acceptable acid addition salt by reacting the free base form of the compound with a pharmaceutically acceptable inorganic or organic acid, e.g.
  • hydrohalides such as hydrochloride, hydrobromide, hydroiodide
  • other mineral acids and their conesponding salts such as sulfate, nitrate, phosphate, etc.
  • alkyl and monoarylsulfonates such as ethanesulfonate, toluenesulfonate andbenzenesulfonate
  • other organic acids and their corresponding salts such as acetate, tartrate, maleate, succinate, citrate, benzoate, salicylate and ascorbate.
  • Further acid addition salts of the present invention include, but are not limited to: adipate, alginate, arginate, aspartate, bisulfate, bisulfite, bromide, butyrate, camphorate, camphorsulfonate, caprylate, chloride, chlorobenzoate, cyclopentanepropionate, digluconate, dihydrogenphosphate, dinitrobenzoate, dodecylsulfate, fumarate, galacterate (frommucic acid), galacturonate, glucoheptaoate, gluconate, glutamate, glycerophosphate, hemisuccinate, hemisulfate, heptanoate, hexanoate, hippurate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethanesulfonate, iodide, isethionate, iso- butyrate, lactate, lactobionate, malate, malonate,
  • a pharmaceutically acceptable base addition salt can be prepared by reacting the free acid form of the compound with a pharmaceutically acceptable inorganic or organic base.
  • bases are alkali metal hydroxides including potassium, sodium and lithium hydroxides; alkaline earth metal hydroxides such as barium and calcium hydroxides; alkali metal alkoxides, e.g.
  • potassium ethanolate and sodium propanolate potassium ethanolate and sodium propanolate
  • various organic bases such as ammonium hydroxide, piperidine, diethanolamine and N-methylglutamine.
  • aluminum salts of the compounds of the present invention include, but are not limited to: copper, ferric, ferrous, lithium, magnesium, manganic, manganous, potassium, sodium and zinc salts.
  • Organic base salts include, but are not l mited to, salts of primary, secondary and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, e.g., arginine, betaine, caffeine, chloroprocaine, choline, N,N'-dibenzylethylenediamine (benzathine), chcyclohexylamine, diemylamine, 2-diethylaminoethanol, 2- dimethylaminoethanol, ethanolamine, ethylene ⁇ iamine, N-emylmo holine, N- ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, iso-propylamine, lidocaine, lysine, meglumine, N-methyl-D-glucamine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, tri
  • N-oxides of compounds according to the present invention can be prepared by methods known to those of ordinary skill in the art.
  • N-oxides can be prepared by treating an unoxidized form of the compound with an oxidizing agent (e.g., trifluoroperacetic acid, permaleic acid, perbenzoic acid, peracetic acid, met ⁇ -chloroperoxybenzoic acid, or the like) in a suitable inert organic solvent (e.g., a halogenated hydrocarbon such as dichloromethane) at approximately 0 °C.
  • an oxidizing agent e.g., trifluoroperacetic acid, permaleic acid, perbenzoic acid, peracetic acid, met ⁇ -chloroperoxybenzoic acid, or the like
  • a suitable inert organic solvent e.g., a halogenated hydrocarbon such as dichloromethane
  • the N-oxides of the compounds can be prepared from the N-oxide of an appropriate starting material.
  • Prodrug derivatives of compounds according to the present invention can be prepared by modifying substituents of compounds of the present invention that are then converted in vivo to a different substituent. It is noted that in many instances, the prodrugs themselves also fall within the scope of the range of compounds according to the present invention.
  • prodrugs can be prepared by reacting a compound with a carbamylating agent (e.g., l -acyloxyalkylcarbonochloridate.p ⁇ r ⁇ -nitrophenyl carbonate, or the like) or an acylating agent. Further examples of methods of making prodrugs are described in Saulnier et al.(1994), Bioorganic and Medicinal Chemistry Letters, Vol. 4, p. 1985.
  • Compounds of the present invention may also be conveniently prepared, or formed during the process of the invention, as solvates (e.g. hydrates). Hydrates of compounds of the present invention may be conveniently prepared by reciystallization from an aqueous/organic solvent mixture, using organic solvents such as dioxin, tetrahydrofuran or methanol.
  • a "pharmaceutically acceptable salt”, as used herein, is intended to encompass any compound according to the present invention that is utilized in the form of a salt thereof, especially where the salt confers on the compound improved pharmacokinetic properties as compared to the free form of compound or a different salt form of the compound.
  • the pharmaceutically acceptable salt form may also initially confer desirable pharmacokinetic properties on the compound that it did not previously possess, and may even positively affect the pharmacodynamics of the compound with respect to its therapeutic activity in the body.
  • An example of a pharmacokinetic property that may be favorably affected is the manner in which the compound is transported across cell membranes, which in turn may directly and positively affect the absorption, distribution, biotransformation and excretion of the compound.
  • the solubility of the compound is usually dependent upon the character of the particular salt form thereof, which it utilized.
  • an aqueous solution of the compound will provide the most rapid absorption of the compound into the body of a subject being treated, while lipid solutions and suspensions, as well as solid dosage forms, will result in less rapid adsorption of the compound.
  • DPP-TV is believed to contribute to the pathology and/or symptomology of several different diseases such that reduction of the activity of DPP-TV in a subject through inhibition maybe used to therapeutically address these disease states. Examples of various diseases that may be treated using the DPP-TV inhibitors of the present invention are described herein. It is noted that additional diseases beyond those disclosed herein may be later identified as the biological roles that DPP-TV plays in various pathways becomes more fully understood.
  • DPP-TV inhibitors of the present invention maybe used to treat are those involving the prevention and treatment of diabetes and obesity, in particular type 2 diabetes mellitus, diabetic dislipidemia, conditions of impaired glucose tolerance (IGT), conditions of impaired fasting plasma glucose (IFG), metabolic acidosis, ketosis, appetite regulation and obesity.
  • ITT impaired glucose tolerance
  • IGF impaired fasting plasma glucose
  • metabolic acidosis ketosis
  • appetite regulation and obesity are those involving the prevention and treatment of diabetes and obesity, in particular type 2 diabetes mellitus, diabetic dislipidemia, conditions of impaired glucose tolerance (IGT), conditions of impaired fasting plasma glucose (IFG), metabolic acidosis, ketosis, appetite regulation and obesity.
  • DPP-IN inhibitors of the present invention may also be used as immunosuppressants (or cytokine release suppressant drugs) for the treatment of among other things: organ transplant rejection; autoimmune diseases such as inflammatory bowel disease, multiple sclerosis and rheumatoid arthritis; and the treatment of AIDS.
  • DPP-TV inhibitors of the present invention may also be used for treating various cancers including breast cancer, lung cancer and prostate cancer.
  • DPP-IN inhibitors of the present invention may also be used to treat dermatological diseases such as psoriasis, rheumatoid arthritis (RA) and lichen planus. [0291] DPP-IN inhibitors of the present invention may also be used to treat infertility and amenorrhea.
  • DPP-IN inhibitors of the present invention may also be used to modulate cleavage of various cytokines (stimulating hematopoietic cells), growth factors and neuropeptides. For example, such conditions occur frequently in patients who are immunosuppressed, for example, as a consequence of chemotherapy and/or radiation therapy for cancer.
  • DPP-IN inhibitors of the present invention may also be used prevent or reduce cleavage of ⁇ -terminal Tyr- Ala from growth hormone-releasing factor. Accordingly, these inhibitors may be used in the treatment of short stature due to growth hormone deficiency (Dwarfism) and for promoting GH-dependent tissue growth or re-growth.
  • DPP-TV inhibitors of the present invention may also be used to address disease states associated with cleavage of neuropeptides and thus may be useful for the regulation or normalization of neurological disorders.
  • DPP-TV inhibitors of the present invention may be used in conjunction with other agents to inhibit undesirable and uncontrolled cell proliferation.
  • examples of other anti-cell proliferation agents that may be used in conjunction with the DPP- IN inhibitors of the present invention include, but are not limited to, retinoid acid and derivatives thereof, 2-methoxyestradiol, A ⁇ GIOSTATT ⁇ TM protein, E ⁇ DOSTATI ⁇ TM protein, suramin, squalamine, tissue inhibitor of metalloproteinase-I, tissue inhibitor of metalloprote ase-2, plasminogen activator inhibitor-1, plasminogen activator inhibitor-2, cartilage-derived inhibitor, paclitaxel, platelet factor 4, protamine sulfate (clupeine), sulfated chitin derivatives (prepared from queen crab shells), sulfated polysaccharide peptidoglycan complex (sp-pg), staurosporine, modulators of matrix metabolism, including for example, proline analogs ((l-
  • anti-angiogenesis agents include antibodies, preferably monoclonal antibodies against these angiogenic growth factors: bFGF, aFGF, FGF-5, VEGF isoforms, NEGF-C, HGF/SF and Ang- l/Ang-2.
  • Ferrara ⁇ . and Alitalo, K. “Clinical application of angiogenic growth factors and their inhibitors” (1999) Nature Medicine 5:1359-1364.
  • compositions and administration methods may be used in conjunction with the DPP-JN inhibitors of the present invention.
  • Such compositions may include, in addition to the DPP-IN inhibitors of the present invention, conventional pharmaceutical excipients, and other conventional, pharmaceutically inactive agents.
  • the compositions may include active agents in addition to the DPP-IN inhibitors of the present invention. These additional active agents may include additional compounds according to the invention, and/or one or more other pharmaceutically active agents.
  • the compositions may be in gaseous, liquid, semi-liquid or solid form, formulated in a manner suitable for the route of administration to be used. For oral aciministration, capsules and tablets are typically used.
  • compositions comprising DPP-IN inhibitors of the present invention may be administered or coadministered orally, parenterally, intraperitoneally, intravenously, intraarterially, transdermally, sublingually, intramuscularly, rectally, transbuccally, intranasally, liposomally, via inhalation, vaginally, intraoccularly, via local delivery (for example by catheter or stent), subcutaneously, intraadiposally, intraarticularly, or intrathecally.
  • the compounds and/or compositions according to the invention may also be administered or coadministered in slow release dosage forms.
  • the DPP-JN inhibitors and compositions comprising them may be administered or coadministered in any conventional dosage form.
  • Co-administration in the context of this invention is intended to mean the administration of more than one therapeutic agent, one of which includes a DPP-IN inhibitor, in the course of a coordinated treatment to achieve an improved clinical outcome.
  • Such co-administration may also be coextensive, that is, occurring during overlapping periods of time.
  • Solutions or suspensions used for parenteral, intradermal, subcutaneous, or topical application may optionally include one or more of the following components: a sterile diluent, such as water for injection, saline solution, fixed oil, polyethylene glycol, glycerine, propylene glycol or other synthetic solvent; antimicrobial agents, such as benzyl alcohol and methyl parabens; antioxidants, such as ascorbic acid and sodium bisulfite; chelating agents, such as ethylenediaminetetraacetic acid (EDTA); buffers, such as acetates, citrates and phosphates; agents for the adjustment of tonicity such as sodium chloride or dextrose, and agents for adjusting the acidity or alkalinity of the composition, such as alkaline or acidifying agents or buffers like carbonates, bicarbonates, phosphates, hydrochloric acid, and organic acids like acetic and citric acid.
  • Parenteral preparations may optionally be enclosed in ampules
  • DPP-TV inhibitors according to the present invention exhibit insufficient solubility
  • methods for solubilizing the compounds may be used. Such methods are known to those of skill in this art, and include, but are not limited to, using cosolvents, such as dimethylsulfoxide (DMSO), using surfactants, such as TWEE ⁇ , or dissolution in aqueous sodium bicarbonate. Derivatives of the compounds, such as prodrugs of the compounds may also be used in formulating effective pharmaceutical compositions.
  • DMSO dimethylsulfoxide
  • TWEE ⁇ surfactants
  • Derivatives of the compounds, such as prodrugs of the compounds may also be used in formulating effective pharmaceutical compositions.
  • a solution, suspension, emulsion or the like may be formed. The fonn of the resulting composition will depend upon a number of factors, including the intended mode of administration, and the solubility of the compound in the selected carrier or vehicle. The effective concentration needed to ameliorate the disease being treated may be empirically determined.
  • compositions according to the present invention are optionally provided for administration to humans and animals in unit dosage forms, such as tablets, capsules, pills, powders, dry powders for inhalers, granules, sterile parenteral solutions or suspensions, and oral solutions or suspensions, and oil-water emulsions containing suitable quantities of the compounds, particularly the pharmaceutically acceptable salts, preferably the sodium salts, thereof.
  • the pharmaceutically therapeutically active compounds and derivatives thereof are typically formulated and administered in unit-dosage forms or multiple-dosage forms.
  • Unit- dose forms refers to physically discrete units suitable for human and aiiimal subjects and packaged individually as is known in the art.
  • Each unit-dose contains a predetermined quantity of the therapeutically active compound sufficient to produce the desired therapeutic effect, in association with the required pharmaceutical carrier, vehicle or diluent.
  • unit-dose forms include ampoules and syringes individually packaged tablet or capsule.
  • Unit-dose forms may be administered in fractions or multiples thereof.
  • a multiple-dose form is a plurality of identical unit-dosage forms packaged in a single container to be administered in segregated unit-dose form.
  • Examples of multiple-dose forms include vials, bottles of tablets or capsules or bottles of pint or gallons.
  • multiple dose form is a multiple of unit-doses that are not segregated in packaging.
  • the composition may comprise: a diluent such as lactose, sucrose, dicalcium phosphate, or carboxymethylcellulose; a lubricant, such as magnesium stearate, calcium stearate and talc; and a binder such as starch, natural gums, such as gum acaciagelatin, glucose, molasses, polvinylpyrrolidine, celluloses and derivatives thereof, povidone, crospovidones and other such binders known to those of skill in the art.
  • a diluent such as lactose, sucrose, dicalcium phosphate, or carboxymethylcellulose
  • a lubricant such as magnesium stearate, calcium stearate and talc
  • a binder such as starch, natural gums, such as gum acaciagelatin, glucose, molasses, polvinylpyrrolidine, celluloses and derivatives thereof, povidone, crospovidones and other such binders
  • Liquid pharmaceutically administrable compositions can, for example, be prepared by dissolving, dispersing, or otherwise mixing an active compound as defined above and optional pharmaceutical adjuvants in a carrier, such as, for example, water, saline, aqueous dextrose, glycerol, glycols, ethanol, and the like, to form a solution or suspension.
  • a carrier such as, for example, water, saline, aqueous dextrose, glycerol, glycols, ethanol, and the like
  • the pharmaceutical composition to be administered may also contain minor amounts of auxiliary substances such as wetting agents, emulsifying agents, or solubilizing agents, pH buffering agents and the like, for example, acetate, sodium citrate, cyclodextrine derivatives, sorbitan monolaurate, triethanolamine sodium acetate, triethanolamine oleate, and other such agents.
  • composition or formulation to be administered will, in any event, contain a sufficient quantity of a DPP-JN inhibitor of the present invention to reduce DPP-JN activity in vivo, thereby treating the disease state of the subject.
  • Dosage forms or compositions may optionally comprise one or more DPP-TV inhibitors according to the present invention in the range of 0.005% to 100% (weight/weight) with the balance comprising additional substances such as those described herein.
  • a pharmaceutically acceptable composition may optionally comprise any one or more commonly employed excipients, such as, for example pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, talcum, cellulose derivatives, sodium crosscarmellose, glucose, sucrose, magnesium carbonate, sodium saccharin, talcum.
  • compositions include solutions, suspensions, tablets, capsules, powders, dry powders for inhalers and sustained release formulations, such as, but not limited to, implants and microencapsulated delivery systems, and biodegradable, biocompatible polymers, such as collagen, ethylene vinyl acetate, polyanhydrides, polyglycolic acid, polyorthoesters, polylactic acid and others. Methods for preparing these formulations are known to those skilled in the art.
  • the compositions may optionally contain 0.01%- 100% (weight/weight) of one or more DPP-IN inhibitors, optionally 0.1-95%, and optionally 1-95%.
  • Salts, preferably sodium salts, of the DPP-IN inhibitors may be prepared with carriers that protect the compound against rapid elimination from the body, such as time release formulations or coatings.
  • the formulations may further include other active compounds to obtain desired combinations of properties.
  • Oral pharmaceutical dosage fonns may be as a solid, gel or liquid.
  • solid dosage forms include, but are not limited to tablets, capsules, granules, and bulk powders. More specific examples of oral tablets include compressed, chewable lozenges and tablets that may be enteric-coated, sugar-coated or film-coated.
  • capsules include hard or soft gelatin capsules. Granules and powders may be provided in non-effervescent or effervescent forms. Each may be combined with other ingredients known to those skilled in the art.
  • DPP-IN inhibitors according to the present invention are provided as solid dosage forms, preferably capsules or tablets.
  • the tablets, pills, capsules, troches and the like may optionally contain one or more of the following ingredients, or compounds of a similar nature: a binder; a diluent; a disintegrating agent; a lubricant; a glidant; a sweetening agent; and a flavoring agent.
  • binders examples include, but are not limited to, microcrystalline cellulose, gum tragacanth, glucose solution, acacia mucilage, gelatin solution, sucrose and starch paste.
  • lubricants examples include, but are not limited to, talc, starch, magnesium or calcium stearate, lycopodium and stearic acid.
  • diluents examples include, but are not limited to, lactose, sucrose, starch, kaolin, salt, mannitol and dicalcium phosphate.
  • glidants examples include, but are not limited to, colloidal silicon dioxide.
  • disintegrating agents examples include, but are not limited to, crosscarmellose sodium, sodium starch glycolate, alginic acid, corn starch, potato starch, bentonite, methylcellulose, agar and carboxymethylcellulose.
  • coloring agents examples include, but are not limited to, any of the approved certified water soluble FD and C dyes, mixtures thereof; and water insoluble
  • sweetening agents examples include, but are not limited to, sucrose, lactose, mannitol and artificial sweetening agents such as sodium cyclamate and saccharin, and any number of spray-dried flavors.
  • flavoring agents examples include, but are not limited to, natural flavors extracted from plants such as fruits and synthetic blends of compounds that produce a pleasant sensation, such as, but not limited to peppermint and methyl salicylate.
  • wetting agents examples include, but are not limited to, propylene glycol monostearate, sorbitan monooleate, diethylene glycol monolaurate and polyoxyethylene lauryl ether.
  • anti-emetic coatings examples include, but are not limited to, fatty acids, fats, waxes, shellac, ammoniated shellac and cellulose acetate phthalates.
  • film coatings examples include, but are not limited to, hydroxyethylcellulose, sodium carboxymethylcellulose, polyethylene glycol 4000 and cellulose acetate phthalate.
  • the salt of the compound may optionally be provided in a composition that protects it from the acidic environment of the stomach.
  • the composition can be formulated in an enteric coating that maintains its integrity in the stomach and releases the active compound in the intestine.
  • the composition may also be formulated in combination with an antacid or other such ingredient.
  • dosage unit form When the dosage unit form is a capsule, it may optionally additionally comprise a liquid canier such as a fatty oil.
  • dosage unit forms may optionally additionally comprise various other materials that modify the physical form of the dosage unit, for example, coatings of sugar and other enteric agents.
  • Compounds according to the present invention may also be administered as a component of an elixir, suspension, syrup, wafer, sprinkle, chewing gum or the like.
  • a syrup may optionally comprise, in addition to the active compounds, sucrose as a sweetening agent and certain preservatives, dyes and colorings and flavors.
  • the DPP-IN inhibitors of the present invention may also be mixed with other active materials that do not impair the desired action, or with materials that supplement the desired action, such as antacids, H2 blockers, and diuretics.
  • active materials such as antacids, H2 blockers, and diuretics.
  • Examples of pharmaceutically acceptable carriers that may be included in tablets comprising DPP-IN inhibitors of the present invention include, but are not limited to binders, lubricants, diluents, disintegrating agents, coloring agents, flavoring agents, and wetting agents .
  • Enteric-coated tablets because of the enteric-coating, resist the action of stomach acid and dissolve or disintegrate in the neutral or alkaline intestines.
  • Sugar-coated tablets may be compressed tablets to which different layers of pharmaceutically acceptable substances are applied.
  • Film-coated tablets may be compressed tablets that have been coated with polymers or other suitable coating. Multiple compressed tablets may be compressed tablets made by more than one compression cycle utilizing the pharmaceutically acceptable substances previously mentioned.
  • Coloring agents may also be used in tablets..
  • Flavoring and sweetening agents may be used in tablets, and are especially useful in the formation of chewable tablets and lozenges.
  • liquid oral dosage forms examples include, but are not limited to, aqueous solutions, emulsions, suspensions, solutions and/or suspensions reconstituted from non-effervescent granules and effervescent preparations reconstituted from effervescent granules.
  • aqueous solutions examples include, but are not limited to, elixirs and syrups.
  • elixirs refer to clear, sweetened, hydroalcoholic preparations.
  • pharmaceutically acceptable carriers examples include, but are not limited to solvents.
  • solvents include glycerin, sorbitol, ethyl alcohol and syrup.
  • syrups refer to concentrated aqueous solutions of a sugar, for example, sucrose. Syrups may optionally further comprise a preservative.
  • Emulsions refer to two-phase systems in which one liquid is dispersed in the form of small globules throughout another liquid. Emulsions may optionally be oil-in-water or water-in-oil emulsions .
  • Examples of pharmaceutically acceptable carriers that may be used in emulsions include, but are not limited to non-aqueous liquids, emulsifying agents and preservatives.
  • Examples of pharmaceutically acceptable substances that may be used in non- effervescent granules, to be reconstituted into a liquid oral dosage form, include diluents, sweeteners and wetting agents.
  • Examples of pharmaceutically acceptable substances that may be used in effervescent granules, to be reconstituted into a liquid oral dosage form, include organic adds and a source of carbon dioxide.
  • Coloring and flavoring agents may optionally be used in all of the above dosage forms.
  • Particular examples of preservatives that may be used include glycerin, methyl and propylparaben, benzoic add, sodium benzoate and alcohol.
  • emulsifying agents include gelatin, acacia, tragacanth, bentonite, and surfactants such as polyoxyethylene sorbitan monooleate.
  • suspending agents include sodium carboxymethylcellulose, pectin, tragacanth, Neegum and acacia.
  • Diluents include lactose and sucrose.
  • Sweetening agents include sucrose, syrups, glycerin and artificial sweetening agents such as sodium cyclamate and saccharin.
  • wetting agents include propylene glycol monostearate, sorbitan monooleate, diethylene glycol monolaurate and polyoxyethylene lauryl ether.
  • organic acids that may be used include citric and tartaric acid.
  • S ources of carbon dioxide that may be used in effervescent compositions include sodium bicarbonate and sodium carbonate.
  • Coloring agents include any of the approved certified water soluble FD and C dyes, and mixtures thereof.
  • flavoring agents include natural flavors extracted from plants such fruits, and synthetic blends of compounds that produce a pleasant taste sensation.
  • the solution or suspension in for example propylene carbonate, vegetable oils or triglycerides, is preferably encapsulated in a gelatin capsule.
  • a gelatin capsule Such solutions, and the preparation and encapsulation thereof, are disclosed in U.S. Pat. ⁇ os.
  • the solution e.g. , for example, in a polyethylene glycol
  • a pharmaceutically acceptable liquid carrier e.g. water
  • liquid or semi-solid oral formulations may be prepared by dissolving or dispersing the active compound or salt in vegetable oils, glycols, triglycerides, propylene glycol esters (e.g. propylene carbonate) and other such carriers, and encapsulating these solutions or suspensions in hard or soft gelatin capsule shells.
  • Other useful formulations include those set forth in U.S. Pat. Nos. Re 28,819 and 4,358,603.
  • compositions designed to administer the DPP-TV inhibitors of the present invention by parenteral administration generally characterized by injection, either subcutaneously, intramuscularly or intravenously.
  • injectables may be prepared in any conventional form, for example as liquid solutions or suspensions, solid forms suitable for solution or suspension in liquid prior to injection, or as emulsions.
  • excipients examples include, but are not limited to water, saline, dextrose, glycerol or ethanol.
  • the injectable compositions may also optionally comprise minor amounts of non- toxic auxiliary substances such as wetting or emulsifying agents, pH buffering agents, stabilizers, solubility enhancers, and other such agents, such as for example, sodium acetate, sorbitan monolaurate, triethanolamine oleate and cyclodextrins.
  • Implantation of a slow- release or sustained-release system, such that a constant level of dosage is maintained see, e.g., U.S. Pat. No.
  • Parenteral adininistration of the formulations includes intravenous, subcutaneous and intramuscular administrations. Preparations for parenteral administration include sterile solutions ready for injection, sterile dry soluble products, such as the lyophilized powders described herein, ready to be combined with a solvent just prior to use, including hypodermic tablets, sterile suspensions ready for injection, sterile dry insoluble products ready to be combined with a vehicle just prior to use and sterile emulsions. The solutions may be either aqueous or nonaqueous.
  • suitable carriers include, but are not limited to physiological saline or phosphate buffered saline (PBS), and solutions containing thickening and solubilizing agents, such as glucose, polyethylene glycol, and polypropylene glycol and mixtures thereof.
  • pharmaceutically acceptable earners that may optionally be used in parenteral preparations include, but are not limited to aqueous vehicles, nonaqueous vehicles, antimicrobial agents, isotonic agents, buffers, antioxidants, local anesthetics, suspending and dispersing agents, emulsifying agents, sequestering or chelating agents and other pharmaceutically acceptable substances.
  • aqueous vehicles examples include Sodium
  • nonaqueous parenteral vehicles examples include fixed oils of vegetable origin, cottonseed oil, corn oil, sesame oil and peanut oil.
  • Antimicrobial agents in bacteriostatic or fungistatic concentrations may be added to parenteral preparations, particularly when the preparations are packaged in multiple-dose containers and thus designed to be stored and multiple aliquots to be removed.
  • antimicrobial agents include phenols or cresols, mercurials, benzyl alcohol, chlorobutanol, methyl and propyl p-hydroxybenzoic acid esters, thimerosal, benzalkonium chloride and benzethonium chloride.
  • Examples of isotonic agents that may be used include sodium chloride and dextrose.
  • Examples of buffers that may be used include phosphate and citrate.
  • antioxidants that may be used include sodium bisulfate.
  • Examples of local anesthetics that may be used include procaine hydrochloride.
  • Examples of suspending and dispersing agents that may be used include sodium carboxymethylcellulose, hydroxypropyl methylcellulose and polyvinylpynolidone.
  • Examples of emulsifying agents that may be used include Polysorbate
  • TWEEN 80 TWEEN 80
  • a sequestering or chelating agent of metal ions include EDTA.
  • Pharmaceutical carriers may also optionally include ethyl alcohol, polyethylene glycol and propylene glycol for water miscible vehicles and sodium hydroxide, hydrochloric acid, citric acid or lactic acid for pH adjustment.
  • the concentration of a DPP-IN inhibitor in the parenteral formulation may be adjusted so that an injection administers a pharmaceutically effective amount sufficient to produce the desired pharmacological effect.
  • concentration of a DPP-IN inhibitor and/or dosage to be used will ultimately depend on the age, weight and condition of the patient or animal as is known in the art.
  • Unit-dose parenteral preparations may be packaged in an ampoule, a vial or a syringe with a needle. All preparations for parenteral administration should be sterile, as is know and practiced in the art.
  • Injectables may be designed for local and systemic administration.
  • a therapeutically effective dosage is formulated to contain a concentration of at least about 0.1 % w/w up to about 90% w/w or more, preferably more than 1% w/w of the DPP-TV inhibitor to the treated tissue(s).
  • the DPP-TV inhibitor may be administered at once, or may be divided into a number of smaller doses to be administered at intervals of time. It is understood that the precise dosage and duration of treatment will be a function of the location of where the composition is parenterally administered, the carrier and other variables that may be determined empirically using known testing protocols or by extrapolation from in vivo or in vitro test data.
  • concentrations and dosage values may also vary with the age of the individual treated. It is to be further understood that for any particular subject, specific dosage regimens may need to be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the formulations. Hence, the concentration ranges set forth herein are intended to be exemplary and are not intended to limit the scope or practice of the claimed formulations.
  • the DPP-TV inhibitor may optionally be suspended in micronized or other suitable form or may be derivatized to produce a more soluble active product or to produce a prodrug. The form of the resulting mixture depends upon a number of factors, including the intended mode of administration and the solubility of the compound in the selected carrier or vehicle. The effective concentration is sufficient for ameliorating the symptoms of the disease state and may be empirically determined.
  • the DPP-IN inhibitors of the present invention may also be prepared as lyophilized powders, which can be reconstituted for administration as solutions, emulsions and other mixtures.
  • the lyophilized powders may also be formulated as solids or gels.
  • Sterile, lyophilized powder may be prepared by dissolving the compound in a sodium phosphate buffer solution containing dextrose or other suitable excipient. Subsequent sterile filtration of the solution followed by lyophilization under standard conditions known to those of skill in the art provides the desired formulation.
  • the lyophilized powder may optionally be prepared by dissolving dextrose, sorbitol, fructose, corn syrup, xylitol, glycerin, glucose, sucrose or other suitable agent, about 1-20%, preferably about 5 to 15%, in a suitable buffer, such as citrate, sodium or potassium phosphate or other such buffer known to those of skill in the art at, typically, about neutral pH.
  • a suitable buffer such as citrate, sodium or potassium phosphate or other such buffer known to those of skill in the art at, typically, about neutral pH.
  • a DPP-JN inhibitor is added to the resulting mixture, preferably above room temperature, more preferably at about 30-35 °C, and st red until it dissolves.
  • the resulting mixture is diluted by adding more buffer to a desired concentration.
  • the resulting mixture is sterile filtered or treated to remove particulates and to insure sterility, and apportioned into vials for lyophilization.
  • Each vial
  • the DPP-TV inhibitors of the present invention may also be administered as topical mixtures.
  • Topical mixtures may be used for local and systemic administration.
  • the resulting mixture may be a solution, suspension, emulsions or the like and are formulated as creams, gels, ointments, emulsions, solutions, elixirs, lotions, suspensions, tinctures, pastes, foams, aerosols, irrigations, sprays, suppositories, bandages, dermal patches or any other formulations suitable for topical admiriistration.
  • the DPP-IN inhibitors may be formulated as aerosols for topical application, such as by inhalation (see, U.S. Pat. ⁇ os. 4,044,126, 4,414,209, and 4,364,923, which describe aerosols for delivery of a steroid useful for treatment inflammatory diseases, particularly asthma).
  • These formulations for administration to the respiratory tract can be in the form of an aerosol or solution for a nebulizer, or as a microfine powder for insufflation, alone or in combination with an inert carrier such as lactose.
  • the particles of the formulation will typically have diameters of less than 50 microns, preferably less than 10 microns.
  • the DPP-IN inhibitors may also be formulated for local or topical application, such as for topical application to the skin and mucous membranes, such as in the eye, in the form of gels, creams, and lotions and for application to the eye or for intracisternal or intraspinal application.
  • Topical administration is contemplated for transdermal delivery and also for admiiiistration to the eyes or mucosa, or for inhalation therapies.
  • Nasal solutions of the DPP-TV inhibitor alone or in combination with other pharmaceutically acceptable excipients can also be administered.
  • Formulations for other routes of administration can also be administered.
  • rectal administration may also be used.
  • pharmaceutical dosage forms for rectal administration are rectal suppositories, capsules and tablets for systemic effect.
  • Rectal suppositories are used herein mean solid bodies for insertion into the rectum that melt or soften at body temperature releasing one or more pharmacologically or therapeutically active ingredients.
  • Pharmaceutically acceptable substances utilized in rectal suppositories are bases or vehicles and agents to raise the melting point.
  • bases examples include cocoa butter (theobroma oil), glycerin-gelatin, carbowax, (polyoxyethylene glycol) and appropriate mixtures of mono-, di- and triglycerides of fatty acids. Combinations of the various bases may be used.
  • Agents to raise the melting point of suppositories include spermaceti and wax. Rectal suppositories may be prepared either by the compressed method or by molding. The typical weight of a rectal suppository is about 2 to 3 gm. Tablets and capsules for rectal administration may be manufactured using the same pharmaceutically acceptable substance and by the same methods as for formulations for oral administration.
  • oral, intravenous and tablet formulations that may optionally be used with compounds of the present invention. It is noted that these formulations may be varied depending on the particular compound being used and the indication for which the formulation is going to be used.
  • the invention is also directed to kits and other articles of manufacture for treating diseases associated with DPP-JN. It is noted that diseases are intended to cover all conditions for which the DPP-IN possesses activity that contributes to the pathology and/or symptomology of the condition.
  • a kit comprising a composition comprising at least one DPP-IN inhibitor of the present invention in combination with instructions.
  • the instructions may indicate the disease state for which the composition is to be administered, storage information, dosing information and/or instructions regarding how to administer the composition.
  • the kit may also comprise packaging materials.
  • the packaging material may comprise a container for housing the composition.
  • the kit may also optionally comprise additional components, such as syringes for administration of the composition.
  • the kit may comprise the composition in single or multiple dose forms.
  • an article of manufacture comprises a composition comprising at least one DPP-JN inhibitor of the present invention in combination with packaging materials.
  • the packaging material may comprise a container for housing the composition.
  • the container may optionally comprise a label indicating the disease state for which the composition is to be administered, storage information, dosing information and/or instructions regarding how to administer the composition.
  • the kit may also optionally comprise additional components, such as syringes for administration of the composition.
  • the kit may comprise the composition in single or multiple dose forms.
  • the packaging material used in kits and articles of manufacture according to the present invention may form a plurality of divided containers such as a divided bottle or a divided foil packet.
  • the container can be in any conventional shape or form as known in the art which is made of a pharmaceutically acceptable material, for example a paper or cardboard box, a glass or plastic bottle or jar, a re-sealable bag (for example, to hold a "refill" of tablets for placement into a different container), or a blister pack with individual doses for pressing out of the pack according to a therapeutic schedule.
  • a pharmaceutically acceptable material for example a paper or cardboard box, a glass or plastic bottle or jar, a re-sealable bag (for example, to hold a "refill" of tablets for placement into a different container), or a blister pack with individual doses for pressing out of the pack according to a therapeutic schedule.
  • the container that is employed will depend on the exact dosage form involved, for example a conventional cardboard box would not generally be used to hold a liquid suspension. It is feasible that more than one container can be used together in a single package to market a single dosage form. For example, tablets may be contained in a bottle that is in
  • ⁇ ie kit form is particularly advantageous when the separate components are preferably administered in different dosage forms (e.g., oral, topical, transdermal and parenteral), are administered at different dosage intervals, or when titration of the individual components of the combination is desired by the prescribing physician.
  • different dosage forms e.g., oral, topical, transdermal and parenteral
  • Blister packs are well known in the packaging industry and are being widely used for the packaging of pharmaceutical unit dosage forms (tablets, capsules, and the like). Blister packs generally consist of a sheet of relatively stiff material covered with a foil of a preferably transparent plastic material. During the packaging process recesses are formed in the plastic foil. The recesses have the size and shape of individual tablets or capsules to be packed or may have the size and shape to accommodate multiple tablets and/or capsules to be packed. Next, the tablets or capsules are placed in the recesses accordingly and the sheet of relatively stiff material is sealed against the plastic foil at the face of the foil which is opposite from the direction in which the recesses were formed.
  • kits are a dispenser designed to dispense the daily doses one at a time in the order of their intended use.
  • the dispenser is equipped with a memory-aid, so as to further facilitate compliance with the regimen.
  • a memory-aid is a mechanical counter that indicates the number of daily doses that has been dispensed.
  • a memory-aid is a battery-powered micro-chip memory coupled with a liquid crystal readout, or audible reminder signal which, for example, reads out the date that the last daily dose has been taken and/or reminds one when the next dose is to be taken.
  • the diastereomers may then be separated in order to recover the optically pure enantiomers.
  • Dissociable complexes may also be used to resolve enantiomers (e.g., crystalline diastereoisomeric salts).
  • Diastereomers typically have sufficiently distinct physical properties (e.g., melting points, boiling points, solubilities, reactivity, etc.) that they can be readily separated by taking advantage of these dissimilarities.
  • diastereomers can typically be separated by chromatography or by separation/resolution techniques based upon differences in solubility.
  • a more detailed description of techniques that can be used to resolve stereoisomers of compounds from their racemic mixture can be found in Jean Jacques Andre Collet, Samuel H. Wilen, Enantiomers, Racemates and Resolutions, John Wiley & Sons, Inc. (1981).
  • Compounds according to the present invention can also be prepared as a pharmaceutically acceptable acid addition salt by reacting the free base form of the compound with a pharmaceutically acceptable inorganic or organic acid.
  • a pharmaceutically acceptable base addition salt of a compound can be prepared by reacting the free acid form of the compound with a pharmaceutically acceptable inorganic or organic base.
  • Inorganic and organic acids and bases suitable for the preparation of the pharmaceutically acceptable salts of compounds are set forth in the definitions section of this Application.
  • the salt forms of the compounds can be prepared using salts of the starting materials or intermediates.
  • the free acid or free base forms of the compounds can be prepared from the conesponding base addition salt or acid addition salt form.
  • a compound in an acid addition salt form can be converted to the conesponding free base by treating with a suitable base (e.g., ammonium hydroxide solution, sodium hydroxide, and the like).
  • a compound in a base addition salt form can be converted to the conesponding free acid by treating with a suitable acid (e.g., hydrochloric acid, etc).
  • N-oxides of compounds according to the present invention can be prepared by methods known to those of ordinary skill in the art.
  • N-oxides can be prepared by treating an unoxidized form of the compound with an oxidizing agent (e.g., trifluoroperacetic acid, permaleic acid, perbenzoic acid, peracetic acid, met ⁇ -chloroperoxybenzoic acid, or the like) in a suitable inert organic solvent (e.g., a halogenated hydrocarbon such as dichloromethane) at approximately 0 °C.
  • an oxidizing agent e.g., trifluoroperacetic acid, permaleic acid, perbenzoic acid, peracetic acid, met ⁇ -chloroperoxybenzoic acid, or the like
  • a suitable inert organic solvent e.g., a halogenated hydrocarbon such as dichloromethane
  • the N-oxides of the compounds can be prepared from the N-oxide of an appropriate starting material.
  • Compounds in an unoxidized form can be prepared from N-oxides of compounds by treating with a reducing agent (e.g., sulfur, sulfur dioxide, triphenyl phosphine, lithium borohydride, sodium borohydride, phosphorus trichloride, tribromide, or the like) in an suitable inert organic solvent (e.g., acetonitrile, ethanol, aqueous dioxane, or the like) at 0 to 80 °C.
  • a reducing agent e.g., sulfur, sulfur dioxide, triphenyl phosphine, lithium borohydride, sodium borohydride, phosphorus trichloride, tribromide, or the like
  • an inert organic solvent e.g., acetonitrile, ethanol, aqueous dioxane, or the like
  • Prodrug derivatives of the compounds can be prepared by methods known to those of ordinary skill in the art (e.g., for further details see Saulnier et ⁇ Z.(1994), Bioorganic and Medicinal Chemistry Letters, Vol. 4, p. 1985).
  • appropriate prodrugs can be prepared by reacting a non-derivatized compound with a suitable carbamylating agent (e.g., l,l-acyloxyalkylcarbonochloridate,/? ⁇ r ⁇ -nitrophenyl carbonate, or the like).
  • a suitable carbamylating agent e.g., l,l-acyloxyalkylcarbonochloridate,/? ⁇ r ⁇ -nitrophenyl carbonate, or the like.
  • Protected derivatives of the compounds can be made by methods known to those of ordinary skill in the art. A detailed description of the techniques applicable to the creation of protecting groups and their removal can be found in T.W. Greene, Protecting Groups in Organic Synthesis, 3 rd edition,
  • Hydrates of compounds of the present invention may be conveniently prepared by recrystallization from an aqueous/organic solvent mixture, using organic solvents such as dioxin, tetrahydrofuran or methanol.
  • Compounds according to the present invention can also be prepared as their individual stereoisomers by reacting a racemic mixture of the compound with an optically active resolving agent to form a pair of diastereoisomeric compounds, separating the diastereomers and recovering the optically pure enantiomer. While resolution of enantiomers can be carried out using covalent diastereomeric derivatives of compounds, dissociable complexes are prefened (e.g., crystalline diastereoisomeric salts). Diastereomers have distinct physical properties (e.g. , melting points, boiling points, solubilities, reactivity, etc.) and can be readily separated by taking advantage of these dissimilarities.
  • the diastereomers can be separated by chromatography or, preferably, by separation/resolution techniques based upon differences in solubility.
  • the optically pure enantiomer is then recovered, along with the resolving agent, by any practical means that would not result in racemization.
  • a more detailed description of the techniques applicable to the resolution of stereoisomers of compounds from their racemic mixture can be found in Jean Jacques Andre Collet, Samuel H. Wilen, Enantiomers, Racemates and Resolutions, John Wiley & Sons, Inc. (1981). [0379]
  • the symbols and conventions used in these processes, schemes and examples are consistent with those used in the contemporary scientific literature, for example, the Journal of the American Chemical Society or the Journal of Biological Chemistry.
  • M molar
  • mM millimolar
  • i.v. intravenous
  • Hz Hertz
  • Tr retention time
  • RP reverse phase
  • TEA triethylamine
  • TFA trifluoroacetic acid
  • TFAA trifluoroacetic anhydride
  • THF tetrahydrofuran
  • DME (1,2-dimethoxyethane); DCM (dichloromethane);
  • DCE dichloroethane
  • DMF N,N-dimethylformamide
  • DMPU N,N'-dimethylpropyleneurea
  • CDI 1,1-carbonyldiimidazole
  • IBCF isobutyl chloroformate
  • HO Ac acetic acid
  • HOSu N-hydroxysuccinimino
  • HOBT 1-hydroxybenzotriazole
  • Et 2 O diethyl ether
  • EDCI ethylcarbodnmino hydrochloride
  • TJPS triisopropylsilyl
  • TBS t-butyldimethylsilyl
  • DMAP 4-dimethylam opyridine
  • Me methyl
  • HPLC high pressure liquid chomatography
  • BOP bis(2-oxo-3-oxazolidinyl)phosphinic chloride
  • TBAF tetra-n-butylammonium fluoride
  • mCPB A metal-chloroperbenzoic acid.
  • MS mass spectra
  • compound purity data were acquired on a Waters ZQ LC/MS single quadrupole system equipped with electrospray ionization (EST) source, UN detector (220 and 254 nm), and evaporative light scattering detector (ELSD).
  • EST electrospray ionization
  • UN detector 220 and 254 nm
  • ELSD evaporative light scattering detector
  • Thin-layer chromatography was performed on 0.25 mm E. Merck silica gel plates (60F-254), visualized with UN light, 5% ethanolic phosphomolybdic acid, ⁇ inhydrin or p-anisaldehyde solution. Flash column chromatography was performed on silica gel (230-400 mesh, Merck).
  • DPP-IN inhibitors according to the present invention may be synthesized according to a variety of reaction schemes. Some illustrative schemes are provided herein in the examples. Other reaction schemes could be readily devised by those skilled in the art. [0384] In the reactions described hereinafter it may be necessary to protect reactive functional groups, for example hydroxy, amino, imino, thio or carboxy groups, where these are desired in the final product, to avoid their unwanted participation in the reactions. Conventional protecting groups may be used in accordance with standard practice, for examples see T.W. Greene and P. G. M. Wuts in "Protective Groups in Organic Chemistry” John Wiley and Sons, 1991.
  • Example 7 2-[2-(3-Amino-piperidin- l-yl)-7-methyl-6-oxo-6,7-dihydro-purin- 1- ylmethylj-benzonitrile
  • Example 8 2-[2-(3-Amino-piperidin- 1 -yl)-9-methyl-6-oxo-6,9-dihydro-purin- 1 - ylmethyl] -benzonitrile
  • Tne crude products 7A and 8A were converted to 7B and 8B as a mixture by the method used for the preparation of IC in 82% yield.
  • Example 7C 2-(2-CMoro-7-methyl-6-oxo-6,7-dmydro-purin-l-ylmethyl)- benzonitrile
  • a mixture of title compounds 7C and 8C was prepared by the method used in the preparation of ID.
  • Example 7 NMR (400 MHz CDQ 3 -CD 3 OD 10: 1): ⁇ 7.9 (br s, IH), 7.63 (d, IH, J
  • Example 9A A mixture of Example 9A and Example 9B was synthesized from 2,6-Dichloro- 7H-purine by the method described in syntheses of Example 7A and 8A.
  • Example 9B NMR (400 MHz CDC1 3 ): ⁇ 8.2 (s, IH), 7.3 (m, 5H), 5.70 (s, 2H), 4.63 (s, 2H).
  • MS (ES) [m+H] calculated for C ⁇ 3 H ⁇ 0 Cl 2 N 4 O+H, 309; found 309.

Abstract

Compounds, pharmaceuticals, kits and methods are provided for use with DPP-IV and other S9 proteases that comprise a compound comprising the formula (I): wherein Q is selected from the group consisting of CO, SO, SO2, or C=NR9; and R1, R2, R3 and R4 are as defined herein.

Description

DIPEPTIDYL PEPTIDASE INHIBITORS
Field of the Invention
[0001] The invention relates to compounds that may be used to inhibit dipeptidyl peptidases as well as compositions of matter and kits comprising these compounds. The present invention also relates to methods for inhibiting dipeptidyl peptidases as well as treatment methods using compounds according to the present invention.
Description of Related Art [0002] Dipeptidyl Peptidase IN (IUBMB Enzyme Nomenclature EC.3.4.14.5) is a type H membrane protein that has been referred to in the literature by a wide a variety of names including DPP4, DP4, DAPTV, FAPβ, adenosine deaminase complexing protein 2, adenosine deaminase binding protein (ADAbp), dipeptidyl aminopeptidase IV; Xaa-Pro-dipeptidyl- aminopeptidase; Gly-Pro naphthylamidase; postproline dipeptidyl aminopeptidase IV; lymphocyte antigen CD26; glycoprotein GP110; dipeptidyl peptidase IV; glycylproline aminopeptidase; glycylproline aminopeptidase; X-prolyl dipeptidyl aminopeptidase; pep X; leukocyte antigen CD26; glycylprolyl dipepMylaminopeptidase; dipeptidyl-peptidehydrolase; glycylprolyl aminopeptidase; dipeptidyl-aminopeptidase IV; DPP IV/CD26; amino acyl-prolyl dipeptidyl aminopeptidase; T cell triggering molecule Tpl03; X-PDAP. Dipeptidyl Peptidase TV is referred to herein as "DPP-IV."
[0003] DPP-IV is a non-classical serine aminodipeptidase that removes Xaa-Pro dipeptides from the amino terminus (N-terminus) of polypeptides and proteins. DPP-IV dependent slow release of dipeptides of the type X-Gly or X-Ser has also been reported for some naturally occurring peptides.
[0004] DPP-IV is constitutively expressed on epithelial and endothelial cells of a variety of different tissues (intestine, liver, lung, kidney and placenta), and is also found in body fluids. DPP-IV is also expressed on circulating T-lymphocytes and has been shown to be synonymous with the cell-surface antigen, CD-26. DPP-IV has been implicated in a number of disease states, some of which are discussed below.
[0005] DPP-IV is responsible for the metabolic cleavage of certain endogenous peptides (GLP-1 (7-36), glucagon) in vivo and has demonstrated proteolytic activity against a variety of other peptides (GHRH, NPY, GLP-2, VIP) in vitro. [0006] GLP-1 (7-36) is a 29 amino-acid peptide derived by post-translational processing of proglucagon in the small intestine; GLP-1 (7-36) has multiple actions in vivo including the stimulation of insulin secretion, inhibition of glucagon secretion, the promotion of satiety, and the slowing of gastric emptying. Based on its physiological profile, the actions of GLP-1 (7- 36) are believed to be beneficial in the prevention and treatment of type II diabetes and potentially obesity. For example, exogenous administration of GLP-1 (7-36) (continuous infusion) in diabetic patients has been found to be efficacious in this patient population. Unfortunately, GLP-1 (7-36) is degraded rapidly in vivo and has been shown to have a short half-life in vivo (tl/2=1.5 minutes).
[0007] Based on a study of genetically bred DPP-IV knock out mice and on in vivo / in vitro studies with selective DPP-IV inhibitors, DPP-IV has been shown to be the primary degrading enzyme of GLP-1 (7-36) in vivo. GLP-1 (7-36) is degraded by DPP-IV efficiently to GLP-1 (9-36), which has been speculated to act as a physiological antagonist to GLP-1 (7-36). Inhibiting DPP-IV in vivo is therefore believed to be useful for potentiating endogenous levels of GLP-1 (7-36) and attenuating the formation of its antagonist GLP-1 (9-36). Thus, DPP-IV inhibitors are believed to be useful agents for the prevention, delay of progression, and/or treatment of conditions mediated by DPP-IV, in particular diabetes and more particularly, type 2 diabetes mellitus, diabetic dislipidemia, conditions of impaired glucose tolerance (IGT), conditions of impaired fasting plasma glucose (IFG), metabolic acidosis, ketosis, appetite regulation and obesity.
[0008] DPP-IV expression is increased in T-cells upon mitogenic or antigenic stimulation (Mattem, T„ et al., Scand. J. Immunol., 1991, 33, 737). It has been reported that inhibitors of DPP-IV and antibodies to DPP-IV suppress the proliferation of mitogen-stimulated and antigen-stimulated T-cells in a dose-dependant manner (Schon, E., et al., Biol. Chem., 1991, 372, 305). Various other functions of T-lymphocytes such as cytokine production, IL-2 mediated cell proliferation and B-cell helper activity have been shown to be dependent on DPP-IV activity (Schon, E., et al., Scand. J. Immunol., 1989, 29, 127). DPP-IV inhibitors, based on boroProline, (Flentke, G. R., et al., Proc. Nat. Acad. Sci. USA, 1991, 88, 1556) although unstable, were effective at inhibiting antigen-induced lymphocyte proliferation and IL-2 production in murine CD4+ T-helper cells. Such boronic acid inhibitors have been shown to have an effect in vivo in mice causing suppression of antibody production induced by immune challenge (Kubota, T. et al., Clin. Exp. ϊrnmun., 1992, 89, 192). The role of DPP- IV in regulating T lymphocyte activation may also be attributed, in part, to its cell-surface association with the transmembrane phosphatase, CD45. DPP-IV inhibitors or non-active site ligands may possibly disrupt the CD45-DPP-IV association. CD45 is known to be an integral component of the T-cell signaling apparatus. It has been reported that DPP-IV is essential for the penetration and infectivity of UPV-l and HTV-2 viruses in CD4+ T-cells (Wakselman, M., Nguyen, C, Mazaleyrat, J.-P., Callebaut, C, Krust, B., Hovanessian, A. G., Inhibition of HTV- 1 infection of CD 26+ but not CD 26-cells by a potent cyclopeptidic inhibitor of the DPP-IV activity of CD 26. Abstract P.44 of the 24.sup.th European Peptide Symposium 1996). Additionally, DPP-TV has been shown to associate with the enzyme adenosine deaminase (ADA) on the surface of T-cells (Kameoka, J., et al., Science, 193, 26466). ADA deficiency causes severe combined immunodeficiency disease (SCID) in humans. This ADA-CD26 interaction may provide clues to the pathophysiology of SCID. It follows that inhibitors of DPP-IV may be useful immunosuppressants (or cytokine release suppressant drugs) for the treatment of among other things: organ transplant rejection; autoimmune diseases such as inflammatory bowel disease, multiple sclerosis and rheumatoid arthritis; and the treatment of AIDS.
[0009] It has been shown that lung endothelial cell DPP-IV is an adhesion molecule for lung-metastatic rat breast and prostate carcinoma cells (Johnson, R. C, et al., J. Cell. Biol., 1993, 121, 1423). DPP-IV is known to bind to fibronectin and some metastatic tumor cells are known to carry large amounts of fibronectin on their surface. Potent DPP-IV inhibitors may be useful as drugs to prevent metastases of, for example, breast and prostrate tumors to the lungs.
[0010] High levels of DPPTV expression have also been found in human skin fibroblast cells from patients with psoriasis, rheumatoid arthritis (RA) and lichen planus (Raynaud, F., et al.,J. Cell. Physiol., 1992, 151, 378). Therefore, DPP-IV inhibitors may be useful as agents to treat dermatological diseases such as psoriasis and lichen planus.
[0011] High DPP-IV activity has been found in tissue homogenates from patients with benign prostate hypertrophy and in prostatosomes. These are prostate derived organelles important for the enhancement of sperm forward motility (Vanhoof, G., et al., Eur. J. Clin. Chem. Clin. Biochem., 1992, 30, 333). DPP-IV inhibitors may also act to suppress sperm motility and therefore act as a male contraceptive agent. Conversely, DPP-JN inhibitors have been implicated as novel for treatment of infertility, and particularly human female infertility due to Polycystic ovary syndrome (PCOS, Stein-Leventhal syndrome) which is a condition characterized by thickening of the ovarian capsule and formation of multiple follicular cysts. It results in infertility and amenorrhea.
[0012] DPP-IV is thought to play a role in the cleavage of various cytokines (stimulating hematopoietic cells), growth factors and neuropeptides.
[0013] Stimulated hematopoietic cells are useful for the treatment of disorders that are characterized by a reduced number of hematopoietic cells or their precursors in vivo. Such conditions occur frequently in patients who are immunosuppressed, for example, as a consequence of chemotherapy and/or radiation therapy for cancer. It was discovered that inhibitors of dipeptidyl peptidase type IV are useful for stimulating the growth and differentiation of hematopoietic cells in the absence of exogenously added cytokines or other growth factors or stromal cells. This discovery contradicts the dogma in the field of hematopoietic cell stimulation, which provides that the addition of cytokines or cells that produce cytokines (stromal cells) is an essential element for maintaining and stimulating the growth and differentiation of hematopoietic cells in culture. (See, e.g., PCT Intl. Application No. PCT /US93/017173 published as WO 94/03055).
[0014] DPP-IV in human plasma has been shown to cleave N-terminal Tyr- Ala from growth hormone-releasing factor and cause motivation of this hormone. Therefore, inhibitors of DPP-IV may be useful in the treatment of short stature due to growth hormone deficiency (Dwarfism) and for promoting GH-dependent tissue growth or re-growth. [0015] DPP-TV can also cleave neuropeptides and has been shown to modulate the activity of neuroactive peptides substance P, neuropeptide Y and CLP (Mentlein, R., Dahms, P., Grandt, D., Kruger, R., Proteolytic processing of neuropeptide Y and peptide YY by dipeptidyl peptidase IV, Regul. Pept, 49, 133, 1993; Wetzel, W., Wagner, T., Vogel, D., Demuth, H.-U., Balschun, D., Effects of the CLIP fragment ACTH 20-24 on the duration of REM sleep episodes, Neuropeptides, 31, 41, 1997). Thus DPP-IV inhibitors may also be useful agents for the regulation or normalization of neurological disorders. [0016] Several compounds have been shown to inhibit DPP-JN. Nonetheless, a need still exists for new DPP-JN inhibitors that have advantageous potency, stability, selectivity, toxicity and or pharmacodynamics properties. In this regard, a novel class of DPP-JN inhibitors are provided herein.
SUMMARY OF THE INVENTION
[0017] The present invention relates to compounds that have activity for inhibiting DPP- JN. It is noted that these compounds may also have activity for inhibiting other S9 proteases and thus may be used against these other S9 proteases as well as DPP-JN. The present invention also provides compositions, articles of manufacture and kits comprising these compounds.
[0018] In one embodiment, a pharmaceutical composition is provided that comprises a DPP-JN inhibitor according to the present invention as an active ingredient. Pharmaceutical compositions according to the invention may optionally comprise 0.001 %-100% of one or more DPP-JN inhibitors of this invention. These pharmaceutical compositions may be administered or coadministered by a wide variety of routes, including for example, orally, parenterally, intraperitoneally, intravenously, intraarterially, transdermally, sublingually, intramuscularly, rectally, transbuccally, intranasally, liposomally, via inhalation, vaginally, intraoccularly, via local delivery (for example by catheter or stent), subcutaneously, intraadiposally, intraarticularly, or intrathecally. The compositions may also be administered or coadministered in slow release dosage forms.
[0019] The invention is also directed to kits and other articles of manufacture for treating disease states associated with DPP-JN.
[0020] In one embodiment, a kit is provided that comprises a composition comprising at least one DPP-JN inhibitor of the present invention in combination with instructions. The instructions may indicate the disease state for which the composition is to be administered, storage information, dosing information and/or instructions regarding how to a ^ninister the composition. The kit may also comprise packaging materials. The packaging material may comprise a container for housing the composition. The kit may also optionally comprise additional components, such as syringes for administration of the composition. The kit may comprise the composition in single or multiple dose forms.
[0021] In another embodiment, an article of manufacture is provided that comprises a composition comprising at least one DPP-JN inhibitor of the present invention in combination with packaging materials. The packaging material may comprise a container for housing the composition. The container may optionally comprise a label indicating the disease state for which the composition is to be administered, storage information, dosing information and/or instructions regarding how to administer the composition. The kit may also optionally comprise additional components, such as syringes for administration of the composition. The kit may comprise the composition in single or multiple dose forms.
[0022] Also provided are methods for preparing compounds, compositions and kits according to the present invention. For example, several synthetic schemes are provided herein for synthesizing compounds according to the present invention.
[0023] Also provided are methods for using compounds, compositions, kits and articles of manufacture according to the present invention.
[0024] In one embodiment, the compounds, compositions, kits and articles of manufacture are used to inhibit DPP-JN.
[0025] In another embodiment, the compounds, compositions, kits and articles of manufacture are used to treat a disease state for which DPP-JN possesses activity that contributes to the pathology and/or symptomology of the disease state.
[0026] In another embodiment, a compound is administered to a subject wherein DPP-JN activity within the subject is altered, preferably reduced.
[0027] In another embodiment, a prodrug of a compound is administered to a subject that is converted to the compound in vivo where it inhibits DPP-JN.
[0028] In another embodiment, a method of inhibiting DPP-JN is provided that comprises contacting DPP-JN with a compound according to the present invention.
[0029] In another embodiment, a method of inhibiting DPP-IV is provided that comprises causing a compound according to the present invention to be present in a subject in order to inhibit DPP-JN in vivo.
[0030] In another embodiment, a method of inhibiting DPP-JN is provided that comprises administering a first compound to a subject that is converted in vivo to a second compound wherein the second compound inhibits DPP-JN in vivo. It is noted that the compounds of the present invention may be the first or second compounds.
[0031] In another embodiment, a therapeutic method is provided that comprises administering a compound according to the present invention. [0032] In another embodiment, a method of inhibiting cell proliferation is provided that comprises contacting a cell with an effective amount of a compound according to the present invention.
[0033] In another embodiment, a method of inhibiting cell proliferation in a patient is provided that comprises administering to the patient a therapeutically effective amount of a compound according to the present invention.
[0034] In another embodiment, a method of treating a condition in a patient which is known to be mediated by DPP-JN, or which is known to be treated by DPP-JN inhibitors, comprising administering to the patient a therapeutically effective amount of a compound according to the present invention.
[0035] In another embodiment, a method is provided for using a compound according to the present invention in order to manufacture a medicament for use in the treatment of disease state which is known to be mediated by DPP-JN, or which is known to be treated by DPP-JN inhibitors.
[0036] In another embodiment, a method is provided for treating a disease state for which
DPP-JN possesses activity that contributes. to the pathology and/or symptomology of the disease state, the method comprising: causing a compound according to the present invention to be present in a subject in a therapeutically effective amount for the disease state.
[0037] In another embodiment, a method is provided for treating a disease state for which
DPP-JN possesses activity that contributes to the pathology and/or symptomology of the disease state, the method comprising: administering a first compound to a subject that is converted in vivo to a second compound such that the second compound is present in the subject in a therapeutically effective amount for the disease state. It is noted that the compounds of the present invention may be the first or second compounds.
[0038] Tri another embodiment, a method is provided for treating a disease state for which
DPP-IN possesses activity that contributes to the pathology and/or symptomology of the disease state, the method comprising: administering a compound according to the present invention to a subject such that the compound is present in the subject in a therapeutically effective amount for the disease state.
[0039] Jrt another embodiment, a method is provided for treating a cell proliferative disease state comprising treating cells with a compound according to the present invention in combination with an anti-proliferative agent, wherein the cells are treated with the compound according to the present invention before, at the same time, and/or after the cells are treated with the anti-proliferative agent, referred to herein as combination therapy. It is noted that treatment of one agent before another is referred to herein as sequential therapy, even if the agents are also administered together. It is noted that combination therapy is intended to cover when agents are administered before or after each other (sequential therapy) as well as when the agents are administered at the same time.
[0040] Examples of diseases that may be treated by administration of compounds and compositions according to the present invention include, but are not limited to conditions mediated by DPP-TV, in particular diabetes, more particular type 2 diabetes mellitus, diabetic dislipidemia, conditions of impaired glucose tolerance (IGT), conditions of impaired fasting plasma glucose (TFG), metabolic acidosis, ketosis, appetite regulation, obesity, immunosuppressants or cytokine release regulation, autoimmune diseases such as inflammatory bowel disease, multiple sclerosis and rheumatoid arthritis, AIDS, cancers (prevention of metastases, for example, breast and prostrate tumors to the lungs), dermatological diseases such as psoriasis and lichen planus, treatment of female infertility, osteoporosis, male contraception and neurological disorders.
[0041] It is noted in regard to all of the above embodiments that the present invention is intended to encompass all pharmaceutically acceptable ionized forms (e.g., salts) and solvates (e.g., hydrates) of the compounds, regardless of whether such ionized forms and solvates are specified since it is well known in the art to administer pharmaceutical agents in an ionized or solvated form. It is also noted that unless a particular stereochemistry is specified, recitation of a compound is intended to encompass all possible stereoisomers (e.g., enantiomers or diastereomers depending on the number of chiral centers), independent of whether the compound is present as an individual isomer or a mixture of isomers. Further, unless otherwise specified, recitation of a compound is intended to encompass all possible resonance forms and tautomers. With regard to the claims, the language "compound comprising the formula" is intended to encompass the compound and all pharmaceutically acceptable ionized forms and solvates, all possible stereoisomers, and all possible resonance forms and tautomers unless otherwise specifically specified in the particular claim. [0042] It is further noted that prodrugs may also be administered which are altered in vivo and become a compound according to the present invention. The various methods of using the compounds of the present invention are intended, regardless of whether prodrug delivery is specified, to encompass the administration of a prodrug that is converted in vivo to a compound according to the present invention. It is also noted that certain compounds of the present invention may be altered in vivo prior to inhibiting DPP-JN and thus may themselves be prodrugs for another compound. Such prodrugs of another compound may or may not themselves independently have DPP-IN inhibitory activity.
BRIEF DESCRIPTION OF THE FIGURE
[0043] Figure 1 illustrates a ribbon diagram overview of the structure of DPP-JN, highlighting the secondary structural elements of the protein.
[0044] Figure 2 depicts different, representative 5 membered ring structures that may be formed when R3 and R4 are taken together to form a substituted or unsubstituted ring. [0045] Figure 3 depicts different, representative 6 membered ring structures that may be formed when R3 and R4 are taken together to form a substituted or unsubstituted ring. [0046] Figure 4 depict additional representative 5 or 6 membered ring structures that may be formed when R3 and R4 are taken together to form a substituted or unsubstituted ring. [0047] The rings shown in Figures 2, 3 and 4 are unsubstituted. It is noted that the rings may optionally be further substituted by one or more substituents.
DEFINITIONS
[0048] Unless otherwise stated, the following terms used in the specification and claims shall have the following meanings for the purposes of this Application. [0049] "Alicyclic" means a moiety comprising a non-aromatic ring structure. Alicyclic moieties may be saturated or partially unsaturated with one, two or more double or triple bonds. Alicyclic moieties may also optionally comprise heteroatoms such as nitrogen, oxygen and sulfur. The nitrogen atoms can be optionally quaternerized or oxidized and the sulfur atoms can be optionally oxidized. Examples of alicyclic moieties include, but are not limited to moieties with C3 - C8 rings such as cyclopropyl, cyclohexane, cyclopentane, cyclopentene, cyclopentadiene, cyclohexane, cyclohexene, cyclohexadiene, cycloheptane, cycloheptene, cycloheptadiene, cyclooctane, cyclooctene, and cyclooctadiene. [0050] "Aliphatic" means a moiety characterized by a straight or branched chain arrangement of constituent carbon atoms and may be saturated or partially unsaturated with one, two or more double or triple bonds.
[0051] "Alkenyl" represented by itself means a straight or branched, unsaturated, aliphatic radical having a chain of carbon atoms having at least one double bond between adjacent carbon atoms. Cx alkenyl and Cχ.γ alkenyl are typically used where X and Y indicate the number of carbon atoms in the chain. For example, C2- alkenyl includes alkenyls that have a chain of between 2 and 6 carbons.
[0052] "Alkoxy" means an oxygen moiety having a further alkyl substituent. The alkoxy groups of the present invention can be optionally substituted.
[0053] "Alkyl" represented by itself means a straight or branched, saturated or unsaturated, aliphatic radical having a chain of carbon atoms, optionally with oxygen (See "oxaalkyl") or nitrogen atoms (See "aminoalkyl") between the carbon atoms. Cx alkyl and Cχ.γ alkyl are typically used where X and Y indicate the number of carbon atoms in the chain. For example, Cι-6 alkyl includes alkyls that have a chain of between 1 and 6 carbons (e.g., methyl, ethyl, propyl, isopropyl, butyl, .fee-butyl, isobutyl, tert-butyl, vinyl, allyl, 1-propenyl, isopropenyl, 1 -butenyl, 2-butenyl, 3-butenyl, 2-methylalTyl, ethynyl, 1-propynyl, 2-propynyl, and the like). Alkyl represented along with another radical (e.g., as in arylalkyl, heteroarylalkyl) means a straight or branched, saturated or unsaturated aliphatic divalent radical having the number of atoms indicated or when no atoms are indicated means a bond (e.g., (C6-ιo)aryl(C1-3)alkyl includes, benzyl, phenethyl, 1-phenylethyl, 3-phenylpropyl, 2- thienylmethyl, 2-pyridinylmethyl and the like).
[0054] "Alkylene", unless indicated otherwise, means a straight or branched, saturated or unsaturated, aliphatic, divalent radical. Cx alkylene and Cχ.γ alkylene are typically used where X and Y indicate the number of carbon atoms in the chain. For example, Cι-6 alkylene includes methylene (-CH2-), ethylene (-CH2CH2-), trimethylene (-CH2CH2CH2-), tetramethylene (-CH2CH2CH2CH2-) 2-butenylene (-CH2CH=CHCH2-),
2-methyltetramethylene (-CH2CH(CH3)CH2CH2-), pentamethylene (-CH2CH2CH2CH2CH2-) and the like).
[0055] "Alkylidene" means a straight or branched saturated or unsaturated, aliphatic radical connected to the parent molecule by a double bond. Cx alkylidene and Cχ.γ alkylidene are typically used where X and Y indicate the number of carbon atoms in the chain. For example, Cι-6 alkylidene includes methylene (=CH2), ethylidene (=CHCH3), isopropylidene (=C(CH3)2), propylidene (=CHCH2CH3), allylidene (=CH-CH=CH2), and the like). [0056] "Alkynyl" represented by itself means a straight or branched, unsaturated, aliphatic radical having a chain of carbon atoms having at least one triple bond between adjacent carbon atoms. Cx alkynyl and Cχ.γ alkynyl are typically used where X and Y indicate the number of carbon atoms in the chain. For example, C2^ alkynyl includes alkynyls that have a chain of between 2 and 6 carbons.
[0057] "Amino" means a nitrogen moiety having two further substituents where a hydrogen or carbon atom is attached to the nitrogen. For example, representative amino groups include -NH2, -NHCH3, -N(CH3)2, -NHC1-3-alkyl, -N(Cι.3-alkyl)2 and the like. Unless indicated otherwise, the compounds of the invention containing amino moieties may include protected derivatives thereof. Suitable protecting groups for amino moieties include acetyl, tert-butoxycarbonyl, benzyloxycarbonyl, arid the like.
[0058] "Aminoalkyl" means an alkyl, as defined above, except where one or more substituted or unsubstituted nitrogen atoms (-N-) are positioned between carbon atoms of the alkyl. For example, an (C2-6) aminoalkyl refers to a chain comprising between 2 and 6 carbons and one or more nitrogen atoms positioned between the carbon atoms. [0059] "Animal" includes humans, non-human mammals (e.g. , dogs, cats, rabbits, cattle, horses, sheep, goats, swine, deer, and the like) and non-mammals (e.g., birds, and the like). [0060] "Aromatic" means a moiety wherein the constituent atoms make up an unsaturated ring system, all atoms in the ring system are sp hybridized and the total number of pi electrons is equal to 4n+2. An aromatic ring may be such that the ring atoms are only carbon atoms or may include carbon and non-carbon atoms (see Heteroaryl). [0061] "Aryl" means a monocyclic or polycyclic ring assembly wherein each ring is aromatic or when fused with one or more rings forms an aromatic ring assembly. If one or more ring atoms is not carbon (e.g., N, S), the aryl is a heteroaryl. Cx aryl and Cχ.γ aryl are typically used where X and Y indicate the number of atoms in the ring. [0062] "Aryloxy" means an oxygen moiety having a further aryl substituent. The aryloxy groups of the present invention can be optionally substituted. [0063] "Bicycloalkyl" means a saturated or partially unsaturated fused bicyclic or bridged polycyclic ring assembly.
[0064] "Bicycloaryl" means a bicyclic ring assembly wherein the rings are linked by a single bond or fused and at least one of the rings comprising the assembly is aromatic. Cx bicycloaryl and Cχ.γ bicycloaryl are typically used where X and Y indicate the number of carbon atoms in the bicyclic ring assembly and directly attached to the ring.
[0065] "Bridging ring" as used herein refers to a ring that is bonded to another ring to form a compound having a bicyclic structure where two ring atoms that are common to both rings are not directly bound to each other. Non-exclusive examples of common compounds having a bridging ring include borήeol, norbornane, 7-oxabicyclo[2.2. ljheptane, and the like.
One or both rings of the bicyclic system may also comprise heteroatoms.
[0066] "Carbamoyl" means the radical -OC(O)NRaRb where Ra and Rt, are each independently two further substituents where a hydrogen or carbon atom is attached to the nitrogen.
[0067] "Carbocycle" means a ring consisting of carbon atoms.
[0068] "Carbocyclic ketone derivative" means a carbocyclic derivative wherein the ring contains a -CO- moiety.
[0069] "Carbonyl" means the radical -CO-. It is noted that the carbonyl radical may be further substituted with a variety of substituents to form different carbonyl groups including acids, acid halides, aldehydes, amides, esters, and ketones.
[0070] "Carboxy" means the radical -CO2-. It is noted that compounds of the invention containing carboxy moieties may include protected derivatives thereof, i.e., where the oxygen is substituted with a protecting group. Suitable protecting groups for carboxy moieties include benzyl, tert-butyl, and the like.
[0071] "Cyano" means the radical -CN.
[0072] "Cycloalkyl" means a non-aromatic, saturated or partially unsaturated, monocyclic, fused bicyclic or bridged polycyclic ring assembly. Cx cycloalkyl and Cχ-Y cycloalkyl are typically used where X and Y indicate the number of carbon atoms in the ring assembly. For example, C3.10 cycloalkyl includes cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexenyl, 2,5-cyclohexadienyl, bicyclo[2.2.2]octyl, adamantan-1-yl, decahydronaphthyl, oxocyclohexyl, dioxocyclohexyl, thiocyclohexyl,
2-oxobicyclo [2.2. l]hept- 1-yl, and the like.
[0073] "Cycloalkylene" means a divalent saturated or partially unsaturated, monocyclic or polycyclic ring assembly. Cx cycloalkylene and Cχ.γ cycloalkylene are typically used where
X and Y indicate the number of carbon atoms in the ring assembly.
[0074] "Disease" specifically includes any unhealthy condition of an animal or part thereof and includes an unhealthy condition that may be caused by, or incident to, medical or veterinary therapy applied to that animal, Le., the "side effects" of such therapy.
[0075] "Fused ring" as used herein refers to a ring that is bonded to another ring to form a compound haying a bicyclic structure where the ring atoms that are common to both rings are directly bound to each other. Non-exclusive examples of common fused rings include decalin, naphthalene, anthracene, phenanthrene, indole, furan, benzofuran, quinoline, and the like.
Compounds having fused ring systems may be saturated, partially saturated, carbocyclics, heterocyclics, aromatics, heteroaromatics, and the like.
[0076] "Halo" means fluoro, chloro, bromo or iodo.
[0077] "Halo-substituted alkyl", as an isolated group or part of a larger group, means
"alkyl" substituted by one or more "halo" atoms, as such terms are defined in this Application.
Halo-substituted alkyl includes haloalkyl, dihaloalkyl, trihaloalkyl, perhaloalkyl and the like
(e.g. halo-substituted (C^alkyl includes chloromethyl, dichloromethyl, difluoromethyl, trifluoromethyl, 2,2,2-trifluoroethyl, perfluoroethyl, 2,2,2-trifluoro-l,l-dichloroethyl, and the like).
[0078] "Heteroaryl" means a cyclic aromatic group having five or six ring atoms, wherein at least one ring atom is a heteroatom and the remaining ring atoms are carbon. The nitrogen atoms can be optionally quaternerized and the sulfur atoms can be optionally oxidized.
Heteroaryl groups of this invention include, but are not limited to, those derived from furan, imidazole, isothiazole, isoxazole, oxadiazole, oxazole, 1,2,3-oxadiazole, pyrazine, pyrazole, pyridazine, pyridine, pyrimidine, pyrroline, thiazole, 1,3,4-thiadiazole, triazole and tetrazole.
"Heteroaryl" also includes, but is not limited to, bicyclic or tricyclic rings, wherein the heteroaryl ring is fused to one or two rings independently selected from the group consisting of an aryl ring, a cycloalkyl ring, a cycloalkenyl ring, and another monocyclic heteroaryl or heterocycloalkyl ring. These bicyclic or tricyclic heteroaryls include, but are not limited to, those derived frombenzo[b]furan, benzo[b]thiophene, benzimidazole, imidazo[4,5-c]pyridine, quinazoline, thieno[2,3-c]pyridine, thieno[3,2-b]pyridine, thieno[2,3-b]pyridine, indolizine, imidazo[l,2a]pyridine, quinoline, isoquinoline, phthalazine, quinoxaline, naphthyridine, quinolizine, indole, isoindole, indazole, indoline, benzoxazole, benzopyrazole, benzothiazole, imidazo[l,5-a]pyridine, pyrazolo[l,5-a]pyridine, imidazo[l,2-a]pyrirmdine, imidazo[l,2- c]pyrimidine, imidazo[l,5-a]pyrimidine, imidazo[l,5-c]pyrimidine, pyrrolo[2,3-b]pyridine, pyrrolo[2,3-c]pyridine, pyrrolo[3,2-c]pyridine, pyrrolo[3,2-b]pyridine, pyrrolo[2,3- d]pyrimidine, pyrrolo[3,2-d]pyrimidine, pyrrolo[2,3-b]pyrazine, pyrazolo[l,5-a]pyridine, pyrrolo[l,2-b]pyridazine, pyrrolo[l,2-c]pyrimidine, pyrrolo[l,2-a]pyrimidine, pyrrolo[l,2- a]pyrazine, triazo[l,5-a]ρyridine, pteridine, purine, carbazole, acridine, phenazine, phenothiazene, phenoxazine, l,2-dihydropyrrolo[3,2,l-/tt]indole, indolizine, pyrido[l,2- a]indole and 2(lH)-pvridinone. The bicyclic or tricyclic heteroaryl rings can be attached to the parent molecule through either the heteroaryl group itself or the aryl, cycloalkyl, cycloalkenyl or heterocycloalkyl group to which it is fused. The heteroaryl groups of this invention can be substituted or unsubstituted.
[0079] "Heteroaryloxy" means an oxygen moiety having a further heteroaryl substituent.
The heteroaryloxy groups of the present invention can be optionally substituted.
[0080] "Heteroatom" refers to an atom that is not a carbon atom. Particular examples of heteroatoms include, but are not limited to nitrogen, oxygen, and sulfur.
[0081] "Heteroatom moiety" includes a moiety where the atom by which the moiety is attached is not a carbon. Examples of heteroatom moieties include -N=, -NRc-, -^(0")=, -O-,
-S- or -S(O)2-, wherein Re is further substituent.
[0082] "Heterobicycloalkyl" means bicycloalkyl, as defined in this Application, provided that one or more of the atoms within the ring is a heteroatom. For example hetero(C9-12)bicycloalkyl as used in this application includes, but is not limited to, 3-aza- bicyclo[4.1.0]heρt-3-yl, 2-aza-bicyclo[3.1.0]hex-2-yl , 3-aza-bicyclo[3.1.0]hex-3-yl, and the like.
[0083] "Heterobicycloaryl" means bicycloaryl, as defined in this Application, provided that one or more of the atoms within the ring is a heteroatom. For example, hetero(C -1o)bicycloaryl as used in this Application includes, but is not limited to, 2-amino-
4-oxo-3,4-dihydropteridin-6-yl, tetrahydroisoquinolinyl, and the like. [0084] "Heterocycloalkyl" means cycloalkyl, as defined in this AppHcation, provided that one or more of the atoms forming the ring is a heteroatom selected, independently from N, O, or S. Non-exclusive examples of heterocycloalkyl include piperidyl, 4-morpholyl, 4- piperazinyl, pyrrolidinyl, perhydropyrrolizinyl, 1,4-diazaperhydroepinyl, 1,3-dioxanyl, 1,4- dioxanyl and the like.
[0085] "Heterocycloalkylene" means cycloalkylene, as defined in this Application, provided that one or more of the ring member carbon atoms is replaced by a heteroatom. [0086] "Hydroxy" means the radical -OH.
[0087] "Jminoketone derivative" means a derivative comprising the moiety -C(NR)-, wherein R comprises a hydrogen or carbon atom attached to the nitrogen. [0088] "Isomers" mean any compound having an identical molecular formulae but differing in the nature or sequence of bonding of their atoms or in the arrangement of their atoms in space. Isomers that differ in the arrangement of their atoms in space are termed "stereoisomers." Stereoisomers that are not mirror images of one another are termed "diastereomers" and stereoisomers that are nonsuperimposable mirror images are termed "enantiomers" or sometimes "optical isomers." A carbon atom bonded to four nonidentical substituents is termed a "chiral center." A compound with one chiral center has two enantiomeric forms of opposite chirality. A mixture of the two enantiomeric forms is termed a "racemic mixture." A compound that has more than one chiral center has 2 enantiomeric pairs, where n is the number of chiral centers. Compounds with more than one chiral center may exist as ether an individual diastereomer or as a mixture of diastereomers, termed a "diastereomeric mixture." When one chiral center is present a stereoisomer may be characterized by the absolute configuration of that chiral center. Absolute configuration refers to the arrangement in space of the substituents attached to the chiral center. Enantiomers are characterized by the absolute configuration of their chiral centers and described by the R- and S-sequencing rules of Cahn, Ingold and Prelog. Conventions for stereochemical nomenclature, methods for the determination of stereochemistry and the separation of stereoisomers are well known in the art (e.g., see "Advanced Organic Chemistry", 4th edition, March, Jerry, John Wiley & Sons, New York, 1992). [0089] "Nitro" means the radical -NO2. [0090] "Oxaalkyl" means an alkyl, as defined above, except where one or more oxygen atoms (-O-) are positioned between carbon atoms of the alkyl. For example, an (C2-6)oxaaikyl refers to a chain comprising between 2 and 6 carbons and one or more oxygen atoms positioned between the carbon atoms.
[0091] "Oxoalkyl" means an alkyl, further substituted with a carbonyl group. The carbonyl group may be an aldehyde, ketone, ester, amide, acid or acid chloride. [0092] "Pharmaceutically acceptable" means that which is useful in preparing a pharmaceutical composition that is generally safe, non-toxic and neither biologically nor otherwise undesirable and includes that which is acceptable for veterinary use as well as human pharmaceutical use.
[0093] "Pharmaceutically acceptable salts" means salts of inhibitors of the present invention which are pharmaceutically acceptable, as defined above, and which possess the desired pharmacological activity. Such salts include acid addition salts formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or with organic acids such as acetic acid, propionic acid, hexanoic acid, heptanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartatic acid, citric acid, benzoic acid, ø-(4-hydroxybenzoyl)benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1,2-ethanedisulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, /?-chlorobenzenesulfonic acid, 2-naphthalenesulfonic acid, p-toluenesulfonic acid, camphorsulfonic acid, 4-methylbicyclo[2.2.2]oct-2-ene-l-carboxyhc acid, glucoheptonic acid, 4,4'-methylenebis(3-hydroxy-2-ene-l-carboxyhc acid), 3-ρhenylpropionic acid, trimethylacetic acid, tertiary butylacetic acid, lauryl sulfuric acid, gluconic acid, glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid, muconic acid and the like. [0094] Pharmaceutically acceptable salts also include base addition salts which may be formed when acidic protons present are capable of reacting with inorganic or organic bases. Acceptable inorganic bases include sodium hydroxide, sodium carbonate, potassium hydroxide, aluminum hydroxide and calcium hydroxide. Acceptable organic bases include emanolamine, diethanolamine, triethanolamine, tromethamine, N-memylglucamine and the like. [0095] "Prodrug" means a compound that is convertible in vivo metabolically into an inhibitor according to the present invention. The prodrug itself may or may not also have DPP-JN inhibitory activity. For example, an inhibitor comprising a hydroxy group may be administered as an ester that is converted by hydrolysis in vivo to the hydroxy compound. Suitable esters that may be converted in vivo into hydroxy compounds include acetates, citrates, lactates, tartrates, malonates, oxalates, salicylates, propionates, succinates, fumarates, maleates, methylene-bis-b-hydroxynaphthoates, gentisates, isethionates, di-p-toluoyltartrates, methanesulfonates, ethanesulfonates, benzenesulfonates, p-toluenesulfonates, cyclohexylsulfamates, quinates, esters of amino acids, and the like. Similarly, an inhibitor comprising an amine group may be administered as an amide that is converted by hydrolysis in vivo to the amine compound.
[0096] "Protected derivatives" means derivatives of inhibitors in which a reactive site or sites are blocked with protecting groups. Protected derivatives are useful in the preparation of inhibitors or in themselves may be active as inhibitors. A comprehensive list of suitable protecting groups can be found in T.W. Greene, Protecting Groups in Organic Synthesis, 3rd edition, John Wiley & Sons, Inc. 1999.
[0097] "Substituted or unsubstituted" means that a given moiety may consist of only hydrogen substituents through available valencies (unsubstituted) or may further comprise one or more non-hydrogen substituents through available valencies (substituted) that are not otherwise specified by the name of the given moiety. For example, isopropyl is an example of an ethylene moiety that is substituted by -CH3. In general, a non-hydrogen substituent may be any substituent that may be bound to an atom of the given moiety that is specified to be substituted. Examples of substituents include, but are not limited to, aldehyde, alicyclic, aliphatic, alkyl, alkylene, alkylidene, amide, amino, aminoalkyl, aromatic, aryl, bicycloalkyl, bicycloaryl, carbamoyl, carbocyclyl, carboxyl, carbonyl group, cycloalkyl, cycloalkylene, ester, halo, heterobicycloalkyl, heterocycloalkylene, heteroaryl, heterobicycloaryl, heterocycloalkyl, oxo, hydroxy, iminoketone, ketone, nitro, oxaalkyl, and oxoalkyl moieties, each of which may optionally also be substituted or unsubstituted. [0098] "Sulfinyl" means the radical -SO-. It is noted that the sulfinyl radical may be further substituted with a variety of substituents to form different sulfinyl groups including sulfinic acids, sulfinamides, sulfinyl esters, and sulfoxides. [0100] "Sulfonyl" means the radical -SO2-. It is noted that the sulfonyl radical may be further substituted with a variety of substituents to form different sulfonyl groups including sulfonic acids, sulfonamides, sulfonate esters, and sulfones.
[0101] "Therapeutically effective amount" means that amount which, when administered to an animal for treating a disease, is sufficient to effect such treatment for the disease.
[0102] "Thiocarbonyl" means the radical -CS-. It is noted that the thiocarbonyl radical may be further substituted with a variety of substituents to form different thiocarbonyl groups including thioacids, thioamides, thioesters, and thioketones.
[0103] "Treatment" or "treating" means any administration of a compound of the present invention and includes:
(1) preventing the disease from occurring in an animal which may be predisposed to the disease but does not yet experience or display the pathology or symptomatology of the disease,
(2) inhibiting the disease in an animal that is experiencing or displaying the pathology or symptomatology of the diseased (i.e., arresting further development of the pathology and/or symptomatology), or
(3) ameliorating the disease in an animal that is experiencing or displaying the pathology or symptomatology of the disease (i.e., reversing the pathology and/or symptomatology).
[0104] It is noted in regard to all of the definitions provided herein that the definitions should be interpreted as being open ended in the sense that further substituents beyond those specified may be included. Hence, a alkyl indicates that there is one carbon atom but does not indicate what are the substituents on the carbon atom. Hence, a alkyl comprises methyl (i.e., -CH3) as well as -RaRbRc where Ra, Rb, and Rc may each independently be hydrogen or any other substituent where the atom attached to the carbon is a heteroatom or cyano. Hence, CF3, CH2OH and CH2CN, for example, are all alkyls.
DETAILED DESCRIPTION OF THE INVENTION
[0105] The present invention relates to compounds, compositions, kits and articles of manufacture that may be used to inhibit dipeptidyl peptidases JN (referred to herein as DPP- IV). [0106] DPP-JN (EC.3.4.14.5 also known as DPP4, DP4, DAP-TV, adenosine deaminase complexing protein 2, adenosine deaminase binding protein (ADAbp) or CD26) is a 766 residue, 240kDa protein that is a highly specific membrane bound non-classical serine aminodipeptidase. DPP-TV has a serine type mechanism of protease activity, cleaving off dipeptides from the ammo-terminus of peptides with proline or alanine at the penultimate position. In addition the slow release of dipeptides of the type X-Gly or X-Ser is reported for some naturally occurring peptides. DPP-IV is constitutively expressed on epithelial and endothelial cells of a variety of different tissues (intestine, liver, lung, kidney and placenta), and is also found in body fluids. DPP-JN is also expressed on circulating T-lymphocytes and has been shown to be synonymous with the cell-surface antigen, CD-26. The wild-type form of full length DPP-JN is described in GenBank Accession Number NM_001935 ("Dipeptidyl peptidase TV (CD 26) gene expression in enterocyte-like colon cancer cell lines HT-29 and Caco-2. Cloning of the complete human coding sequence and changes of dipeptidyl peptidase TN mRΝA levels during cell differentiation", Darmoul, D., Lacasa, M., Baricault, L., Marguet, D., Sapin, C, Trotot, P., Barbat, A. and Trugnan, G., J. Biol. Chem., 267 (7), 4824-4833, 1992).
[0107] DPP-JN is a member of the S9 family of serine proteases, more particularly the S9B family. Other members of the S9 family include, but are not limited to:
Subfamily S9A: Dipeptidyl-peptidase; Oligopeptidase B (EC 3.4.21.83): Oligopeptidase B; Prolyl oligopeptidase (EC 3.4.21.26): Subfamily S9B: Dipeptidyl aminopeptidase A; Dipeptidyl aminopeptidase B Dipeptidyl-peptidase TV (EC 3.4.14.5): Dipeptidyl-peptidase V Fibroblast activation protein alpha subunit; Seprase Subfamily S9C: Acvlaminoacvl-peptidase (EC 3.4.19.1) [0108] It is noted that the compounds of the present invention may also possess inhibitory activity for other S9 family members and thus maybe used to address disease states associated with these other family members.
1. CRYSTAL STRUCTURE OF DPP-IV
[0109] Syrrx, Inc. (San Diego, California) recently solved the crystal structure of DPP-JN. Knowledge of the crystal structure was used to guide the design of the DPP-JN inhibitors provided herein. [0110] Figure 1 illustrates a ribbon diagram overview of the structure of DPP-TV, highlighting secondary structural elements of the protein. DPP-TV is a cylindrical shaped molecule with an approximate height of 70 A and a diameter of 60 A. The catalytic triad of DPP-IN (Ser642, Asp720 and His752) is illustrated in the center of the figure by a "ball and stick" representation. This triad of amino acids is located in the peptidase domain or catalytic domain of DPP-TV. The catalytic domain is covalently linked to the β-propeller domain. The catalytic domain of DPP-TV includes residues 1-67 and 511-778. The catalytic domain of DPP-IN adopts a characteristic α/β hydrolase fold. The core of this domain contains an 8- stranded β-sheet with all strands being parallel except one. The α-sheet is significantly twisted and is flanked by three α-helices on one side and five α-helices on the other. The topology of the β-strands is 1, 2, -lx, 2x and (lx) (J. S. Richardson: The anatomy and taxonomy of protein structure; (1981) Adv. Protein Chem.269, 15076-15084.). A number of residues were identified that contribute to the shape and charge characteristics of the active site. Knowledge of these residues has been an important contribution to the design of DPP-JN inhibitors of the present invention.
2. DPP-IV INHIBITORS
[0111] In one embodiment, DPP-TV inhibitors of the present invention comprise Formula I:
Figure imgf000022_0001
I wherein
Q is selected from the group consisting of CO, SO, SO2, or C=NR ;
Ri is hydrogen or is selected from the group consisting of (Cι-1o)alkyl, (C3-i2)cycloalkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-i2)cycloalkyl, aryl(C1-10)alkyl, (C9-i2)bicycloaryl, hetero(C4-12)bicycloaryl, carbonyl (Cι-3)alkyl, thiocarbonyl (Cι-3)alkyl, sulfonyl (C1-3)alkyl, sulfinyl
Figure imgf000022_0002
amino, cyano, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;
R2 is selected from the group consisting of halo, perhalo(Cι-1o)alkyl, amino, cyano, thio, (Cι-io)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, carbonyl (Cι.3)alkyl, thiocarbonyl
Figure imgf000023_0001
sulfinyl (Cι.3)alkyl, imino
Figure imgf000023_0002
alkoxy, aryloxy, heteroaryloxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, and a substituted or unsubstituted 4, 5, 6 or 7 membered ring;
R3 and R4 are taken together to form a substituted or unsubstituted 5 or 6 membered ring; and
R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
[0112] In another embodiment, DPP-JN inhibitors of the present invention comprise Formula JJ:
6-Position 7-Position
Figure imgf000023_0003
II wherein
Q is selected from the group consisting of CO, SO, SO2, or C=ΝR9;
J, K, L, and M are each independently selected from the group of moieties where the ring atom is either C or N;
Ri is hydrogen or is selected from the group consisting of (C1-ιo)alkyl, (C3-i2)cycloalkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl, aryl(C1-1o)alkyl, (Cg-^bicycloaryl, hetero(C -i2)bicycloaryl, carbonyl (C1-3)alkyl, thiocarbonyl (C1-3)aιkyl, sulfonyl (C1-3)alkyl, sulfinyl (Ci^alkyl, imino (Cι.3)alkyl, amino, cyano, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted; R2 is selected from the group consisting of halo, perhalo(Cι-ιo)alkyl, amino, cyano, thio, (Cι-ιo)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, carbonyl (Cι-3)alkyl, thiocarbonyl (Ci^alkyl, sulfonyl (Cι-3)alkyl, sulfinyl (Cι.3)alkyl, imino ( ^alkyl, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, and a substituted or unsubstituted 4, 5, 6 or 7 membered ring; and
R is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
[0113] In one embodiment, DPP-JN inhibitors of the present invention having particular substitution patterns have significantly improved stability in biological assays. Improvement in the stabilities of the compounds in rat liver microsomes, for example, as demonstrated by the increase in the half life of the test compound in microsomal assays, has been demonstrated in particular substitutions of the DPP-IN inhibitors of the present invention. [0114] Jn one particular embodiment, DPP-IV inhibitors comprising the Formula JJ with substitution at the 6 and/or the 7-positions result in enhanced stability as demonstrated in biological assays. Jn one particular embodiment, compounds of the Formula JJ comprise quinazolinones that are substituted at the 6-position, the 7-position, or both the 6 and 7 positions of the compound of Formula TJ. Νon-limiting examples of the compounds of the Formula JI include quinazolinone derivatives, naphthyridine derivatives, 4H-pyrido[3,4- d]pyrimidine derivatives, etc.
[0115] In one embodiment, DPP-JN inhibitors comprise compounds of the Formula JJ wherein K is CR12, where R12 is independently selected from the group consisting of halo, perhalo(Cl-10)alkyl, CF3, cyano, nitro, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, a carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
[0116] In yet another variation, K is CR12 where R12 is independently selected from the group consisting of halo, perhalo(Cl-10)alkyl, CF3, cyano, nitro, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, a carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, and L is nitrogen.
[0117] In another variation,
K is CR12, where R12 is independently selected from the group consisting of halo, perhalo(C1-1o)alkyl, CF3, cyano, nitro, alkyl, aryloxy, heteroaryloxy, amino, and alkoxy, each substituted or unsubstituted;
Ri is selected from the group consisting of -(CH2)-(2-cyano)phenyl, -(CH2)-(3-cyano)phenyl, -(CH2)-(2-hydroxy)phenyl, -(CH2)-(3-hydroxy)phenyl, -(CH2)-(2- alkenyl)phenyl, -(CH2)-(3-alkenyl)phenyl, -(CH2)-(2-alkynyl)phenyl, -(CH2)-(3- alkynyl)phenyl, -(CH2)-(2-nitro)phenyl, -(CH2)-(3-nitro)phenyl, -(CH2)-(2-carboxy)phenyl, -(CH2)-(3-carboxy)phenyl, -(CH2)-(2-carboxamido)ρhenyl, -(CH2)-(3-carboxamido)phenyl, -(CH2)-(2-sulfonamido)phenyl, -(CH2)-(3-sulfonamido)phenyl, -(CH2)-(2-tetrazolyl)phenyl, -(CH2)-(3-tetrazolyl)phenyl,-(CH2)-(2-ammomemyl)phenyl, -(CH2)-(3-ammomemyl)phenyl^ -(CH2)-(2-amino)phenyl, -(CH2)-(3-amino)phenyl, -(CH2)-(2-hydroxymethyl)phenyl, -(CH2)-(3-hydroxymethyl)ρhenyl, -(CH2)-(2-ρhenyl)phenyl, -(CH2)-(3-phenyl)ρhenyl, -(CH2)-(2-CONH2)phenyl, -(CH2)-(3-CONH2)phenyl, -(CH2)-(2-CONH(Cr7)alkyl)phenyl, -(CH2)-(3-CONH(C1-7)alkyl)phenyl, -(CH2)-(2-CO2(Ci-7)aikyl)phenyl,
-(CH2)-(3-CO2(C1-7)alkyl)phenyl, -CH2-NH2, -CH2-OH, -CH2-(C3-7)alkyl, -CH2-alkene, -CH2-alkyne, -CH2-CCH, -CH2-(C3-7)cycloalkyl, and -CH2-aryl, each substituted or unsubstituted; and
R2 is selected from the group consisting of 3-amino-piperidin-l-yl, 3-aminomethyl- pyrrolidin-1-yl, azetidin-1-yl, 3-aminoazetidin-l-yl, pyrrolidin- 1-yl, 3-aminocyclopent-l-yl, 3- aminomethylcyclopent-1-yl, 3-aminomethylcyclohex-l-yl, hexahydroazepin-1-yl, 3- aminohexahydroazepin-1-yl, 3-amino-cyclohex-l-yl, piperazin- 1-yl, homopiperazin- 1-yl, 3- amino-pyrrolidin-1-yl, and R-3-aminopiperidin-l-yl, each substituted or unsubstituted. [0118] In yet another variation,
K is CRi2, where R12 is independently selected from the group consisting of halo, perhalo(C1-1o)alkyl, CF3, cyano, nitro, alkyl, aryloxy, heteroaryloxy, amino, and alkoxy, each substituted or unsubstituted;
Ri is selected from the group consisting of -(CH2)-(2-cyano)phenyl, -(CH2)-(3-cyano)ρhenyl, -(CH2)-(2-hydroxy)ρhenyl, -(CH2)-(3-hydroxy)phenyl, -(CH2)-(2- alkenyl)ρhenyl, -(CH2)-(3-alkenyl)ρhenyl, -(CH2)-(2-alkynyl)phenyl, -(CH2)-(3^ alkynyl)phenyl, -(CH2)-(2-nitro)phenyl, -(CH2)-(3-nitro)ρhenyl, -(CH2)-(2-carboxy)phenyl, -(CH2)-(3-carboxy)phenyl, -(CH2)-(2-carboxamido)phenyl, -(CH2)-(3-carboxamido)phenyl, -(CH2)-(2-sulfonarmdo)phenyl, -(CH2)-(3-sulfonamido)phenyl, -(CH2)-(2-tetrazolyl)phenyl, -(CH2)-(3-tetrazolyl)phenyl, -(CH2M2-arn^
-(CH2)-(2-amino)phenyl, -(CH2)-(3-amino)phenyl, -(CH2)-(2-hydroxymethyl)phenyl, -(CH2)-(3-hydroxymethyl)ρhenyl, -(CH2)-(2-phenyl)phenyl, -(CH )-(3-ρhenyl)phenyl, -(CH2)-(2-COiNuH2)phenyl, -(CH2)-(3-CONH2)phenyl, -(CH2)-(2-CONH(C1-7)alkyl)phenyl, -(CH2)-(3-CONH(C1-7)alkyl)phenyl, -(CH2)-(2-CO2(C1-7)alkyl)phenyl,
-(CH2)-(3-CO2(C1-7)alkyl)phenyl, -CH2-NH2, -CH2-OH, ,CH2-(C3-7)alkyl, -CH2-alkene, -CH2-alkyne, -CH2-CCH, -CH2-(C3- )cycloalkyl, and -CH2-aryl, each substituted or unsubstituted; and
R2 is selected from the group consisting of 3-amino-piperidin-l-yl, 3-aminomethyl- pyrrolidin-1-yl, azetidin-1-yl, 3-aminoazetidin-l-yl, pyrrolidin- 1-yl, 3-aminocyclopent-l-yl, 3- aminomethylcyclopent-1-yl, 3-aminomethylcyclohex-l-yl, hexahydroazepin-1-yl, 3- aminohexahydroazepin-1-yl, 3-amino-cyclohex-l-yl, piperazin- 1-yl, homopiperazin- 1-yl, 3- amino-pyrrolidin-1-yl, and R-3-aminopiperidin-l-yl, each substituted or unsubstituted. [0119] In another embodiment, DPP-IN inhibitors of the present invention comprise a member selected from the group consisting of Formulae HJa, JJIb, TJIc, Eld, UJe and IJJf:
Figure imgf000027_0001
wherein
Q is selected from the group consisting of CO, SO, SO2, or C=NR ;
Ri is hydrogen or is selected from the group consisting of (Cι-io)alkyl, (C3-12)cycloalkyl, (C3.12)cycloalkyl(C1-5)alkyl, hetero(C3-i2)cycloalkyl, aryl(Cι-10)alkyl, (Cg-^bicycloaryl, hetero(C -12)bicycloaryl, carbonyl (C^alkyl, thiocarbonyl (C^alkyl, sulfonyl (C1-3)alkyl, sulfinyl (C^alkyl, imino (C1-3)alkyl, amino, cyano, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;
R2 is selected from the group consisting of halo, perhalo(Cι-1o)alkyl, amino, cyano, thio, (C1-1o)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, carbonyl ( ^alkyl, thiocarbonyl
Figure imgf000027_0002
sulfinyl (Cι-3)alkyl, imino (Cι-3)alkyl, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, and a substituted or unsubstituted 4, 5, 6 or 7 membered ring;
R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted; and each Rι9 is independently selected from the group consisting of hydrogen, halo, perhalo(Cι-ιo)alkyl, CF3, cyano, nitro, alkyl, alkene, alkyne, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, with the proviso that R1 is not alkylthio, arylthio, halo, cyano, nitro, and thio in the case where the ring atom to which R1 is bound is nitrogen.
[0120] Jn another embodiment, DPP-JN inhibitors of the present invention comprise Formula TN:
Figure imgf000028_0001
IV wherein
Q is selected from the group consisting of CO, SO, SO2, or C=ΝR ;
W, X, and Y are each independently selected from the group of moieties where the ring atom is either C, N, O or S;
Ri is hydrogen or is selected from the group consisting of (Cι-ιo)alkyl, (C3-i2)cycloalkyl, (C3-i2)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl, aryl(C1-io)alkyl, (C92)bicycloaryl, hetero(C ,12)bicycloaryl, carbonyl
Figure imgf000028_0002
sulfonyl (C1-3)alkyl, sulfinyl ( ^alkyl, imino (C^aHcyl, amino, cyano, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;
R2 is selected from the group consisting of halo, perhalo(C1-1o)alkyl, amino, cyano, thio, (C1-1o)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, carbonyl (Cι-3)alkyl, thiocarbonyl
Figure imgf000028_0003
hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, and a substituted or unsubstituted 4, 5, 6 or 7 membered ring; and R is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
[0121] In another embodiment, DPP-IV inhibitors of the present invention comprise Formulae Va, Vb or Vc:
Figure imgf000029_0001
wherein
Q is selected from the group consisting of CO, SO, SO2, or C=NR ;
Ri is hydrogen or is selected from the group consisting of (Ci-io)alkyl, (C3-12)cycloalkyl, (C3-i2)cycloalkyl(Cι-5)alkyl, hetero(C3-12)cycloalkyl, aryl(Ci-io)alkyl, (C9-12)bicycloaryl, hetero(C -12)bicycloaryl, carbonyl (C^alkyl, thiocarbonyl ( ^alkyl, sulfonyl (Cι.3)alkyl, sulfinyl (C1-3)alkyl, imino
Figure imgf000029_0002
amino, cyano, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;
R2 is selected from the group consisting of halo, perhalo(Ci-1o)alkyl, amino, cyano, thio, (Cι-1o)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, carbonyl ( ^alkyl, thiocarbonyl
Figure imgf000029_0003
sulfinyl (C1-3)alkyl, imino (C1-3)alkyl, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, and a substituted or unsubstituted 4, 5, 6 or 7 membered ring;
R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted; and each Rι9 is independently selected from the group consisting of hydrogen, halo, ρerhalo(C1-1o)alkyl, CF3, cyano, nitro, alkyl, alkene, alkyne, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, with the proviso that R19 is not alkylthio, arylthio, halo, cyano, nitro, and thio in the case where the ring atom to which Ri9 is bound is nitrogen.
[0122] In another embodiment, DPP-IV inhibitors of the present invention comprise Formula Via or Formula VJb:
Figure imgf000030_0001
wherein
Q is selected from the group consisting of CO, SO, SO2, or C=NR9;
W, X, and Y are each independently selected from the group of moieties where the ring atom is either C, N, O or S;
Ri is hydrogen or is selected from the group consisting of (Ci-io)alkyl, (C3-i2)cycloalkyl, (C3-i2)cycloalkyl(Ci-5)alkyl, hetero(C32)cycloalkyl, aryl(C1-1o)alkyl, (C9-ι2)bicycloaryl, hetero(C -i2)bicycloaryl, carbonyl ( ^alkyl, thiocarbonyl (Cι-3)alkyl, sulfonyl (Ci-3)alkyl, sulfinyl (Chalky!, imino ( ^alkyl, amino, cyano, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;
R2 is selected from the group consisting of halo, perhalo(Ci-io)alkyl, amino, cyano, thio, (Ci-1o)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, carbonyl (Ci^alkyl, thiocarbonyl
Figure imgf000030_0002
hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, and a substituted or unsubstituted 4, 5, 6 or 7 membered ring; and
R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
[0123] In another embodiment, DPP-IV inhibitors of the present invention comprise Formulae Vila, VHb, or VJJc:
Figure imgf000031_0001
wherein
Q is selected from the group consisting of CO, SO, SO2, or C=NR9;
X is selected from the group of moieties where the ring atom is either C, N, O or S in Formula VTJa, or X is selected from the group of moieties where the ring atom is either C or N in Formula VTJb or Formula VTJc;
Ri is hydrogen or is selected from the group consisting of (Ci-10)alkyl, (C3-i2)cycloalkyl, (C3.i2)cycloalkyl(Ci-5)alkyl, hetero(C3-12)cycloaIkyl, aryl(Cι-ιo)alkyl, C9-i2)bicycloaryl, hetero(C4.12)bicycloaryl, carbonyl ( ^alkyl, thiocarbonyl (Cι-3)alkyl, sulfonyl
Figure imgf000031_0002
imino (Ci-3)alkyl, amino, cyano, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;
R2 is selected from the group consisting of halo, ρerhalo(Ci-io)alkyl, amino, cyano, thio, (Crιo)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, carbonyl (C1-3)alkyl, thiocarbonyl (Ci-3)alkyl, sulfonyl
Figure imgf000031_0003
alkoxy, aryloxy, heteroaryloxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, and a substituted or unsubstituted 4, 5, 6 or 7 membered ring;
R is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted; and each R19 is independently selected from the group consisting of hydrogen, halo, perhalo(Ci-io)alkyl, CF3, cyano, nitro, alkyl, alkene, alkyne, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, with the proviso that Rι9 is not alkylthio, arylthio, halo, cyano, nitro, and thio in the case where the ring atom to which R19 is bound is nitrogen.
[0124] In another embodiment, DPP-TV inhibitors of the present invention comprise Formulae Villa VTUb and VTTIc:
Figure imgf000032_0001
vπia viπb vmc wherein
Q is selected from the group consisting of CO, SO, SO2, or C=NR9;
J, K, L, and M are each independently selected from the group of moieties where the ring atom is either C, N, O or S;
Ri is hydrogen or is selected from the group consisting of (Cι-io)alkyl, (C32)cycloalkyl, (C3-i2)cycloalkyl(Ci-5)alkyl, hetero(C3-12)cycloalkyl, aryl(Cι-i0)alkyl, (C9-ι2)bicycloaryl, hetero(C4.i2)bicycloaryl, carbonyl
Figure imgf000032_0002
sulfonyl
Figure imgf000032_0003
sulfinyl (Ci.3)alkyl, imino (Ci.3)alkyl, amino, cyano, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;
R2 is selected from the group consisting of halo, perhalo(Cι-ι0)alkyl, amino, cyano, thio, (Cι-ιo)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, carbonyl
Figure imgf000032_0004
thiocarbonyl
Figure imgf000032_0005
sulfinyl (Ci_3)alkyl, imino
Figure imgf000032_0006
hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, and a substituted or unsubstituted 4, 5, 6 or 7 membered ring; and
R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
[0125] In another embodiment, DPP-JV inhibitors of the present invention comprise Formula TX:
Figure imgf000033_0001
IX wherein
Q is selected from the group consisting of CO, SO, SO2, or C=NR9;
J, K, L, and M are each independently selected from the group of moieties where the ring atom is either C, N, O or S;
Ri is hydrogen or is selected from the group consisting of (Ci-io)alkyl, (C3-i2)cycloalkyl, (C3-i2)cycloalkyl(Ci-5)alkyl, hetero(C3-i2)cycloalkyl, aryl(Ci-io)alkyl, (C9.i2)bicycloaryl, hetero(C4-12)bicycloaryl, carbonyl (Ci^alkyl, thiocarbonyl (C1-3)alkyl, sulfonyl (Cι-3)alkyl, sulfinyl (Ci^alkyl, imino (Chalky!, amino, cyano, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;
R2 is selected from the group consisting of halo, perhalo(Cι-ιo)alkyl, amino, cyano, thio, (Cι-io)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, carbonyl (Ci.3)alkyl, thiocarbonyl
Figure imgf000033_0002
sulfonyl (C1-3)alkyl, sulfinyl (Ci_3)alkyl, imino
Figure imgf000033_0003
alkoxy, aryloxy, heteroaryloxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, and a substituted or unsubstituted 4, 5, 6 or 7 membered ring; and
R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
[0126] In another embodiment, DPP-TV inhibitors of the present invention comprise Formula X:
Figure imgf000033_0004
X wherein Q is selected from the group consisting of CO, CS, SO, SO2, or C=NR9;
Ri is -ZRm, where Z is a moiety providing 1-6 atom separation between Rm and the ring to which Ri is attached, and Rm is selected from the group consisting of a substituted or unsubstituted (C3- )cycloalkyl and aryl;
R2 is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring;
R3 and R4 are taken together to form a substituted or unsubstituted 5 or 6 membered ring; and
R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
[0127] In another embodiment, DPP-TV inhibitors of the present invention comprise Formula XI:
Figure imgf000034_0001
XI wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NR9;
J, K, L, and M are each independently selected from the group of CRι2 and N;
Ri is -ZRm, where Z is a moiety providing 1-6 atom separation between Rm and the ring to which Ri is attached, and Rm is selected from the group consisting of a substituted or unsubstituted (C3-7)cycloalkyl and aryl;
R2 is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring;
R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted; and each R12 is hydrogen or is independently selected from the group consisting of halo, perhalo(Ci-io)alkyl, CF3, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, alkoxy, a carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
[0128] In another embodiment, DPP-IV inhibitors of the present invention comprise
Formulae XJJa, Xllb, XJIc, XUd, XTJe, and Xϋf:
Figure imgf000035_0001
wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NR9;
Ri is -ZRm, where Z is a moiety providing 1-6 atom separation between Rra and the ring to which Ri is attached, and Rm is selected from the group consisting of a substituted or unsubstituted (C3-7)cycloalkyl and aryl;
R2 is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring; R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted; and each R19 is independently selected from the group consisting of hydrogen, halo, perhalo(Ci-io)alkyl, CF3, cyano, nitro, alkyl, alkene, alkyne, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, with the proviso that Rig is not alkylthio, arylthio, halo, cyano, nitro, and thio in the case where the ring atom to which Rι9 is bound is nitrogen.
[0129] Th another embodiment, DPP-IN inhibitors of the present invention comprise Formula XTJJ:
Figure imgf000036_0001
XIII wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=ΝR9;
W, X, and Y are each independently selected from the group of moieties where the ring atom is either C, N, O or S;
Ri is -ZRm, where Z is a moiety providing 1-6 atom separation between Rm and the ring to which Ri is attached, and Rm is selected from the group consisting of a substituted or unsubstituted (C3-7)cycloalkyl and aryl;
R2 is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring; and
R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
[0130] In another embodiment, DPP-TV inhibitors of the present invention comprise Formulae XTVa, XTVb, and XTVc:
Figure imgf000037_0001
wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NRg;
Ri is -ZRm, where Z is a moiety providing 1-6 atom separation between Rm and the ring to which Ri is attached, and Rm is selected from the group consisting of a substituted or unsubstituted (C3- )cycloalkyl and aryl;
R2 is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring;
R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted; and each Rι9 is independently selected from the group consisting of hydrogen, halo, perhalo(Ci-io)alkyl, CF3, cyano, nitro, alkyl, alkene, alkyne, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, with the proviso that R19 is not alkylthio, arylthio, halo, cyano, nitro, and thio in the case where the ring atom to which Rι9 is bound is nitrogen.
[0131] In another embodiment, DPP-JV inhibitors of the present invention comprise Formulae XVa and XVb:
Figure imgf000037_0002
wherein Q is selected from the group consisting of CO, CS, SO, SO2, or C=NR9;
W, X, and Y are each independently selected from the group of moieties where the ring atom is either C, N, O or S;
Ri is -ZRm, where Z is a moiety providing 1-6 atom separation between Rm and the ring to which Ri is attached, and Rm is selected from the group consisting of a substituted or unsubstituted (C3- )cycloalkyl and aryl;
R2 is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring; and
R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
[0132] In another embodiment, DPP-TV inhibitors of the present invention comprise Formulae XVIa, XVTb, and XVIc:
Figure imgf000038_0001
wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NR9;
X is selected from the group of moieties where the ring atom is either C, N, O or S in Formula XVIa, or X is selected from the group of moieties where the ring atom is either C or N in Formula XVTb or Formula XVTc;
Ri is -ZRm, where Z is a moiety providing 1-6 atom separation between Rm and the ring to which Ri is attached, and Rm is selected from the group consisting of a substituted or unsubstituted (C3- )cycloalkyl and aryl;
R2 is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring; Rg is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted; and each R19 is independently selected from the group consisting of hydrogen, halo, perhalo(Cι-ιo)alkyl, CF3, cyano, nitro, alkyl, alkene, alkyne, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, with the proviso that R19 is not alkylthio, arylthio, halo, cyano, nitro, and thio in the case where the ring atom to which R19 is bound is nitrogen.
[0133] In another embodiment, DPP-IN inhibitors of the present invention comprise Formulae XVJJa, XVJJb, and XVHc:
Figure imgf000039_0001
xviia xvπb xvπc
wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NRg;
J, K, L, and M are each independently selected from the group of moieties where the ring atom is either C, N, O or S;
Ri is -ZRm, where Z is a moiety providing 1-6 atom separation between Rm and the ring to which Ri is attached, and Rm is selected from the group consisting of a substituted or unsubstituted (C3-7)cycloalkyl and aryl; and
R2 is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring.
[0134] In another embodiment, DPP-I inhibitors of the present invention comprise Formula XV :
Figure imgf000040_0001
xvm wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NR9;
J, K, L, and M are each independently selected from the group of moieties where the ring atom is either C, N, O or S;
Ri is -ZRm, where Z is a moiety providing 1-6 atom separation between Rm and the ring to which.Ri is attached, and Rm is selected from the group consisting of a substituted or unsubstituted (C3- )cycloalkyl and aryl;
R2 is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring; and
R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
[0135] In another embodiment, DPP-TV inhibitors of the present invention comprise Formula XIX:
Figure imgf000040_0002
XIX wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NR9;
Ri is -ZRm, where Z is a moiety providing 1-6 atom separation between Rm and the ring to which Ri is attached, and -Rm is selected from the group consisting of a substituted or unsubstituted (C3- )cycloalkyl and aryl;
R2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein; R3 and R4 are taken together to form a substituted or unsubstituted 5 or 6 membered ring; and
R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
[0136] In another embodiment, DPP-IN inhibitors of the present invention comprise Formula XX:
Figure imgf000041_0001
XX wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NR9;
J, K, L, and M are each independently selected from the group of CR12 and N;
Ri is -ZRm, where Z is a moiety providing 1-6 atom separation between Rm and the ring to which Ri is attached, and -Rm is selected from the group consisting of a substituted or unsubstituted (C3- )cycloalkyl and aryl;
R2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein;
R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted; and each Rι2 is hydrogen or is independently selected from the group consisting of halo, perhalo(Cι-ιo)alkyl, CF3, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, alkoxy, a carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
[0137] In another embodiment, DPP-IV inhibitors of the present invention comprise Formulae XXIa, XXIb, XXIc, XXId, XXIe, and XXJf:
Figure imgf000042_0001
wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NR9;
Ri is -ZRm, where Z is a moiety providing 1-6 atom separation between Rm and the ring to which Ri is attached, and -Rm is selected from the group consisting of a substituted or unsubstituted (C3- )cycloalkyl and aryl;
R2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein;
R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted; and each R19 is independently selected from the group consisting of hydrogen, halo, perhalo(Ci-io)alkyl, CF3, cyano, nitro, alkyl, alkene, alkyne, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, with the proviso that Rig is not alkylthio, arylthio, halo, cyano, nitro, and thio in the case where the ring atom to which R19 is bound is nitrogen.
[0138] In another embodiment, DPP-TV inhibitors of the present invention comprise Formula XXII:
Figure imgf000043_0001
XXII wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NR9;
W, X, and Y are each independently selected from the group of moieties where the ring atom is either C, N, O or S;
Ri is -ZRm, where Z is a moiety providing 1-6 atom separation between Rm and the ring to which Ri is attached, and -Rm is selected from the group consisting of a substituted or unsubstituted (C3- )cycloalkyl and aryl;
R2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein; and
R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
[0139] In another embodiment, DPP-IV inhibitors of the present invention comprise Formulae XXTHa, XXJJIb, and XXTHc:
Figure imgf000044_0001
wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NRg;
Ri is -ZRm, where Z is a moiety providing 1-6 atom separation between Rm and the ring to which Ri is attached, and -Rm is selected from the group consisting of a substituted or unsubstituted (C3- )cycloalkyl and aryl;
R2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein;
R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted; and each R19 is independently selected from the group consisting of hydrogen, halo, perhalo(Cι-ιo)alkyl, CF3, cyano, nitro, alkyl, alkene, alkyne, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, with the proviso that Rι9 is not alkylthio, arylthio, halo, cyano, nitro, and thio in the case where the ring atom to which R19 is bound is nitrogen.
[0140] In another embodiment, DPP-IN inhibitors of the present invention comprise Formulae XXINa and XXIVb:
Figure imgf000045_0001
XXTVa XXTVb wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NRg;
W, X, and Y are each independently selected from the group of moieties where the ring atom is either C, N, O or S;
Ri is -ZRm, where Z is a moiety providing 1-6 atom separation between Rm and the ring to which Ri is attached, and -Rm is selected from the group consisting of a substituted or unsubstituted (C3-7)cycloalkyl and aryl;
R2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein; and
R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
[0141] In another embodiment, DPP-IV inhibitors of the present invention comprise Formulae XXVa, XXVb, and XXVc:
Figure imgf000045_0002
wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NR9;
X is selected from the group of moieties where the ring atom is either C, N, O or S in Formula XXVa, or X is selected from the group of moieties where the ring atom is either C or N in Formula XXVb or Formula XXVc; Ri is -ZRm, where Z is a moiety providing 1-6 atom separation between Rm and the ring to which Ri is attached, and -Rm is selected from the group consisting of a substituted or unsubstituted (C3-7)cycloalkyl and aryl;
R2 is -UN, where U is a moiety providing 1-6 atom separation between V and the ring to which R2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein;
Rg is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted; and each R19 is independently selected from the group consisting of hydrogen, halo, perhalo(Cι-ιo)alkyl, CF3, cyano, nitro, alkyl, alkene, alkyne, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, carbonyl group, irnine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, with the proviso that Ri9 is not alkylthio, arylthio, halo, cyano, nitro, and thio in the case where the ring atom to which R19 is bound is nitrogen.
[0142] In another embodiment, DPP-TV inhibitors of the present invention comprise Formulae XXVIa, XXVIb, and XXVIc:
Figure imgf000046_0001
XXVIa XXVIb XXVIc
wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=ΝR9;
J, K, L, and M are each independently selected from the group of moieties where the ring atom is either C, N, O or S;
Ri is -ZRm, where Z is a moiety providing 1-6 atom separation between Rm and the ring to which Ri is attached, and -Rm is selected from the group consisting of a substituted or unsubstituted (C3-7)cycloalkyl and aryl; and R2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein. [0143] In another embodiment, DPP-TV inhibitors of the present invention comprise Formula XXVTT:
Figure imgf000047_0001
xxvπ wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NR9;
J, K, L, and M are each independently selected from the group of moieties where the ring atom is either C, N, O or S;
Ri is -ZRm, where Z is a moiety providing 1-6 atom separation between Rm and the ring to which Ri is attached, and -Rm is selected from the group consisting of a substituted or unsubstituted (C3- )cycloalkyl and aryl;
R2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein; and
R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
[0144] In another embodiment, DPP-TV inhibitors of the present invention comprise Formula XXVTJJ:
Figure imgf000047_0002
wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NR9;
Ri is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring;
R2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein;
R3 and R4 are taken together to form a substituted or unsubstituted 5 or 6 membered ring; and
R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
[0145] In another embodiment, DPP-IV inhibitors of the present invention comprise Formula XXIX:
Figure imgf000048_0001
wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NR9;
J, K, L, and M are each independently selected from the group of CR12 and N;
Ri is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring;
R2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein;
R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted; and each R12 is hydrogen or is independently selected from the group consisting of halo, perhalo(Ci-io)alkyl, CF3, alkyl, aryl; heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, alkoxy, a carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
[0146] In another embodiment, DPP-IN inhibitors of the present invention comprise Formulae XXXa, XXXb, XXXc, XXXd, XXXe, and XXXf:
Figure imgf000049_0001
wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NR9;
Ri is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring; R2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein;
R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted; and each R19 is independently selected from the group consisting of hydrogen, halo, perhalo(Cι-ιo)alkyl, CF3, cyano, nitro, alkyl, alkene, alkyne, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, with the proviso that Rι9 is not alkylthio, arylthio, halo, cyano, nitro, and thio in the case where the ring atom to which Rig is bound is nitrogen.
[0147] In another embodiment, DPP-IN inhibitors of the present invention comprise Formula XXXI:
Figure imgf000050_0001
XXXI wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=ΝRg;
W, X, and Y are each independently selected from the group of moieties where the ring atom is either C, N, O or S;
Ri is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring;
R2 is -UN, where U is a moiety providing 1-6 atom separation between N and the ring to which R2 is attached and N comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein; and R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
[0148] In another embodiment, DPP-IN inhibitors of the present invention comprise Formulae XXXJJa, XXXTIb, and XXXIJc:
Figure imgf000051_0001
wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=ΝR9;
Ri is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring;
R2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein;
Rg is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted; and each Rig is independently selected from the group consisting of hydrogen, halo, perhalo(Ci-io)alkyl, CF3, cyano, nitro, alkyl, alkene, alkyne, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, with the proviso that Rig is not alkylthio, arylthio, halo, cyano, nitro, and thio in the case where the ring atom to which Rι9 is bound is nitrogen.
[0149] In another embodiment, DPP-IV inhibitors of the present invention comprise Formulae XXXJJJa and XXXITJb:
Figure imgf000052_0001
XXXIHa XXXHIb wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NR9;
W, X, and Y are each independently selected from the group of moieties where the ring atom is either C, N, O or S;
Ri is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring;
R2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein; and
Rg is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
[0150] In another embodiment, DPP-TV inhibitors of the present invention comprise Formulae XXXJNa, XXXIVb, and XXXTNc:
Figure imgf000052_0002
wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NR9; X is selected from the group of moieties where the ring atom is either C, N, O or S in Formula XXXTVa, or X is selected from the group of moieties where the ring atom is either C or N in Formula XXXIVb or Formula XXXTVc;
Ri is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring;
R2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein;
R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted; and each R19 is independently selected from the group consisting of hydrogen, halo, perhalo(Cι-ιo)alkyl, CF3, cyano, nitro, alkyl, alkene, alkyne, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, with the proviso that Ri is not alkylthio, arylthio, halo, cyano, nitro, and thio in the case where the ring atom to which Rι9 is bound is nitrogen.
[0151] In another embodiment, DPP-TV inhibitors of the present invention comprise Formulae XXXVa, XXXVb, and XXXVc:
Figure imgf000053_0001
XXXVa XXXVb XXXVc
wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NR9;
J, K, L, and M are each independently selected from the group of moieties where the ring atom is either C, N, O or S;
Ri is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring; and R2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein. [0152] Jn another embodiment, DPP-TV inhibitors of the present invention comprise Formula XXXVI:
Figure imgf000054_0001
XXXVI wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NR9;
J, K, L, and M are each independently selected from the group of moieties where the ring atom is either C, N, O or S;
Ri is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring;
R2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein; and
R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
[0153] In another embodiment, DPP-TV inhibitors of the present invention comprise Formula XXXVII:
Figure imgf000054_0002
XXXVII wherein Q is selected from the group consisting of CO, CS, SO, SO2, or C=NRg;
J, K, L, and M are each independently selected from the group of CR12 and N, provided that at least one of K and L is CR12 where Rι2 is not hydrogen;
Ri is -ZRm, where Z is a moiety providing 1-6 atom separation between Rm and the ring to which Ri is attached, and Rm is selected from the group consisting of a substituted or unsubstituted (C3- )cycloalkyl and aryl;
R2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein;
Rg is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted; and each R12 is hydrogen or is independently selected from the group consisting of halo, perhalo(Ci-io)alkyl, CF3, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, alkoxy, a carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
[0154] In another embodiment, DPP-TV inhibitors of the present invention comprise Formula XXXVHI:
Figure imgf000055_0001
XXXVΠI wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NR9;
J, K, L, and M are each independently selected from the group of CRι2 and N, provided that at least one of K and L is CR12 where R12 is not hydrogen;
Ri is -ZRm, where Z is a moiety providing 1-6 atom separation between Rm and the ring to which Ri is attached, and Rm is selected from the group consisting of a substituted or unsubstituted (C3- )cycloalkyl and aryl; R2 is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring;
R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted; and each R12 is hydrogen or is independently selected from the group consisting of halo, perhalo(Cι-ιo)alkyl, CF3, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, alkoxy, a carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
[0155] In another embodiment, DPP-IN inhibitors of the present invention comprise Formula XXXIX:
Figure imgf000056_0001
XXXIX wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NRg;
J, K, L, and M are each independently selected from the group of CR12 and N, provided that at least one of K and L is CR12 where R12 is not hydrogen;
Ri is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring;
R2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein; and
Rg is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted; and each R12 is hydrogen or is independently selected from the group consisting of halo, perhalo(Ci-io)alkyl, CF3, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, alkoxy, a carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
Substituents Ri and Rt:
[0156] In one variation of each of the above embodiments and variations comprising R3 and Rt, R3 and R4 are taken together to form an unsubstituted or substituted 5 or 6 membered cycloalkyl or heterocycloalkyl ring. In another variation, R3 and R4 are taken together to form a substituted or unsubstituted phenyl ring. In yet another variation, R3 and R4 are taken together to form a substituted or unsubstituted heteroaryl ring. In still another variation, R3 and R4 are taken together to form a substituted or unsubstituted heteroaryl selected from the group consisting of substituted or unsubstituted furan, thiophene, pyrrole, pyrazole, triazole, isoxazole, oxazole, thiazole, isothiazole, oxadiazole, pyridine, pyridazine, pyrimidine, pyrazine, triazine, benzofuran, isobenzofuran, benzothiophene, isobenzothiophene, imidazole, benzimidazole, indole, isoindole, quinoline, isoquinoline, cinnoline, quinazoline, naphthyridine, pyridopyridine, quinoxaline, phthalazine, and benzothiazole. [0157] In a further variation of each of the above embodiments and variations comprising R3 and R4, R3 and R are taken together to form a 5 or 6-membered ring where the ring comprises at least one CO group. In another variation of each of the above embodiments and variations comprising R3 and R4, R3 and R4 are taken together to form a 5 or 6-membered ring comprising of 1-3 nitrogen ring atoms. In yet another variation of each of the above embodiments and variations comprising R3 and R4, R3 and j are taken together to form a 5 or 6-membered ring where the ring comprises a sulfur atom. In one variation, the ring sulfur atom is in an oxidized form as SO or SO2.
[0158] In particular variations, R3 and R4 are taken together to form a ring system such that the compound formed is selected from the group consisting of substituted or unsubstituted 4-oxo-4H-quinazoline, 3H-pvrido[2,3-d]pyrir din-4-one, 3H-pyrido[3,2-d]pyrimidin-4-one, 3H-pyrido[3,4-d]pyrimidin-4-one and 3H-pyrido[4,3-d]pyrimidin-4-one. [0159] In still another variation of each of the above embodiments and variations wherein R3 and R4 are taken together to form a ring, the ring formed by R3 and R4 comprises substituents that form a ring fused to the ring formed by R3 and R4. The fused ring may be further substituted or unsubstituted, and may be a saturated or unsaturated ring. In one variation, the fused ring is a heterocyclic ring.
[0160] In another variation, the 5 or 6 membered ring is further substituted such that a 3 , 4, 5, 6 or 7 membered bridging ring is formed with the 5 or 6 membered ring. The 3, 4, 5, 6 or 7 membered ring may be further substituted or unsubstituted, and may be a saturated or unsaturated ring. In another variation, the 3, 4, 5, 6 or 7 membered bridged ring may be a heterocyclic ring.
Substituents J. K, L, M:
[0161] In one variation, J, K, L and M each comprises a carbon ring atom. In another variation, J comprises a nitrogen ring atom. In another variation, K comprises a nitrogen ring atom. In another variation, L comprises a nitrogen ring atom. In another variation, M comprises a nitrogen ring atom.
[0162] yet another variation, J and L each comprise a nitrogen ring atom or J and K each comprise a nitrogen ring atom. In another variation, K and L each comprise a nitrogen ring atom. In another variation, K and M each comprise a nitrogen ring atom. In yet another variation, J and M each comprise a nitrogen ring atom, or L and M each comprise a nitrogen ring atom. ;
[0163] In a further variation of each of the above embodiments and variations comprising J, K, L and M, at least one of J, K, L and M comprise a nitrogen ring atom. Jn yet another variation, at least two of J, K, L and M comprise a nitrogen ring atom. In another variation, at least three of J, K, L and M comprise a nitrogen ring atom. In still another variation of each of the above embodiments and variations comprising J, K, L and M, at least three of J, K, L and M comprise a nitrogen ring atom.
[0164] According to each of the above variations, the ring formed by J, K, L, and M may comprise substituents that form a ring fused or bridged (e.g., through available valencies) to the ring formed by J, K, L, and M. Jn particular with respect to the case of Formula VTJJib, J and M optionally form a fused or bridged ring relative to the ring formed by J, K, L, and M. [0165] In another variation of each of the above embodiments and variations comprising K, K is CR12, where R^ is independently selected from the group consisting of halo, perhalo(C1-io)alkyl, CF3, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, alkoxy, a carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
[0166] In still another variation of each of the above embodiments and variations comprising K, K is CRι2, where R12 is independently selected from the group consisting of halo, perhalo(Cι-ιo)alkyl, CF3, cyanό, nitro, alkyl, aryloxy, heteroaryloxy, amino, and alkoxy, each substituted or unsubstituted.
[0167] Jn yet another variation of each of the above embodiments and variations comprising K, K is CR12, where Rι2 is independently selected from the group consisting of heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryl, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, thio, a carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
[0168] In a further variation of each of the above embodiments and variations comprising
K, K is CR12, where R12 is independently selected from the group consisting of chloro, bromo, fluoro, iodo, methoxy, morpholin-4-yl, and pyrrolidin- 1-yl, each substituted or unsubstituted.
[0169] In one variation of each of the above embodiments and variations comprising L, L comprises a nitrogen ring atom.
[0170] In another variation of each of the above embodiments and variations comprising
L, L is CRu, where R12 is independently selected from the group consisting of halo, perhalo(C1-1o)alkyl, CF3, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, alkoxy, a carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
[0171] In still another variation of each of the above embodiments and variations comprising L, L is CR12, where R12 is independently selected from the group consisting of halo, perhalo(Cι-ιo)alkyl, CF3, cyano, nitro, alkyl, aryloxy, heteroaryloxy, amino, morpholin-
4-yl, and pyrrolidin- 1-yl, and alkoxy, each substituted or unsubstituted.
[0172] In one variation of each of the above embodiments and variations comprising J, K,
L and M, at least one of J, K, L and M is CO.
[0173] In another variation of each of the above embodiments and variations comprising
J, K, L and M, at least one of J, K, L and M is SO. [0174] In still another variation of each of the above embodiments and variations comprising J, K, L and M, at least one of J, K, L and M is SO2.
[0175] In one variation of each of the above embodiments and variations comprising J and M, J and M form a bridged ring relative to the ring formed by J, K, L, and M
[0176] In one variation of each of the above embodiments and variations comprising K and L, K and L are independently CR12, where R12 is independently selected from the group consisting of halo, perhalo(C1-1o)alkyl, CF3, cyano, nitro, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, a carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
[0177] In another variation of each of the above embodiments and variations comprising
K and L,
K is CR12, where R12 is independently selected from the group consisting of halo, perhalo(Ci-io)alkyl, CF3, cyano, nitro, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, a carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted; and
L is nitrogen. [0178] Jn one variation of each of the above embodiments and variations comprising R12, at least one R12 is halogen.
[0179] In another variation of each of the above embodiments and variations comprising R12, at least one Rι2 is fluorine.
Substituents W, X. and Y
[0180] In one variation, at least one of W, X, and Y is CO. In another variation, at least one of W, X, and Y is SO. In another variation, at least one of W, X, and Y is SO2. In one variation of each of the above embodiments and variations comprising W, W is selected from the group consisting of CO, SO or SO2. In one variation of each of the above embodiments and variations comprising Y, Y is selected from the group consisting of CO, SO or SO2. [0181] In another variation, at least one of W, X, and Y comprises a ring nitrogen atom. In yet another variation, at least two of W, X, and Y comprises a ring nitrogen atom. In yet another variation, W comprises a ring nitrogen atom. In still variation, X comprises a ring nitrogen atom. In a further variation, Y comprises a ring nitrogen atom.
[0182] In another variation, W and Y are taken together to form a substituted or unsubstituted bridged ring relative to the ring formed by W, X and Y. In yet another variation, at least two (t'.e., two or three) of W, X, and Y are taken together to form a substituted or unsubstituted ring fused or bridged (e.g., by substitution through available valencies) to the ring formed by W, X and Y.
[0183] In a further variation, W, X, and Y are selected such that the compound comprises a ring system selected from the group consisting of 4-oxo-4H-tMeno[3,2-d]pvrimidine, 7-oxo- l,2,3,7-tefrahydro-8-tMa-4,6-diaza-cyclopenta[a]mdene,7-memyl-6-oxo-6,7-dmydro-pιιrm and 6-oxo-6,9-dihydro-purine, each substituted or unsubstituted.
Substituent Ri:
[0184] In one variation, Ri is selected from the group consisting of -NH2, -NHCH3, -N(CH3)2, -NHCι-3-alkyl, and -N(Cι-3-alkyl)2, each unsubstituted or substituted through available valencies. In another variation, Ri is selected from the group consisting of a ( . 3)alkyl; an amino, alkyl, or alkoxy carbonyl (Cι-3)alkyl; an amino, alkyl, or alkoxy thiocarbonyl (Ci^alkyl; an amino, alkyl, or alkoxy sulfonyl (Cι-3)alkyl; an amino, alkyl, or alkoxy sulfinyl (Cι-3)alkyl; an amino, alkyl, or alkoxy imino (Ci^alkyl; and an aryl ( . 5)alkyl, each unsubstituted or substituted through available valencies. [0185] In another variation, Ri is selected from the group consisting of a (C2-ιo)alkenyl; an amino, alkyl, or alkoxy carbonyl (Ci^alkenyl; an amino, alkyl, or alkoxy thiocarbonyl ( . 3)alkenyl; an amino, alkyl, or alkoxy sulfonyl (Ci^alkenyl; an amino, alkyl, or alkoxy sulfinyl
Figure imgf000061_0001
and a heteroaryl ( .5) alkenyl, each unsubstituted or substituted through available valencies. In yet another variation, Ri is selected from the group consisting of a (C2-io)alkynyl; an amino, alkyl, or alkoxy carbonyl (Ci^alkynyl; an amino, alkyl, or alkoxy thiocarbonyl (Cι.3)alkynyl; an amino, alkyl, or alkoxy sulfonyl
Figure imgf000061_0002
an amino, alkyl, or alkoxy sulfinyl ( . 3)alkynyl; an amino, alkyl, or alkoxy imino
Figure imgf000061_0003
an aryl ( ..5) alkynyl; and heteroaryl (Cι-5)alkynyl, each unsubstituted or substituted through available valencies. [0186] In another variation, Ri is substituted or unsubstituted (C3.7)cycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted phenyl. [0187] In another variation of the invention, Ri is hydroxy. Jn yet another variation, Ri is
-ORn, where Rn is selected from the group consisting of substituted or unsubstituted alkyl, cycloalkyl, aryl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl.
[0188] In another variation, Ri is a carbonyl group. In yet another variation, Rl is selected from the group consisting of an aldehyde, acid, amide, and ester.
[0189] In yet another variation, Ri is selected from the group consisting of pyπolidin- 1 - ylcarbonyl, piperidin-1 -ylcarbonyl, and morpholin-4-ylcarbonyl, each unsubstituted or substituted through available valencies.
[0190] In another variation of the invention, Ri is -Z-Rm, where
Z is a moiety having a chain of 1-6 atoms selected from the group consisting of substituted or unsubstituted C, N, O, and S, attaching -Rm to the ring, or Z is a unsubstituted or substituted -(Ci-3)alkylene optionally interrupted by O, N(O), -NH, SO, SO2, or carbonyl; and
-Rm is selected from the group consisting of a substituted or unsubstituted (Ci- )alkyl, -(C2-7)alkene, -(C2-7)alkyne, -(C3-7)cycloalkyl, and aryl.
[0191] Jn another variation of each of the above embodiments and variations comprising Ri, Ri is -ZRm, where
Z is a moiety providing 1-6 atom separation between Rm and the ring to which Ri is attached, and
Rm is selected from the group consisting of a substituted or unsubstituted (C3-7)cycloalkyl and aryl.
[0192] In one variation, the 1-6 atoms of Z comprise of carbon atoms. In another variation, the 1-6 atoms of Z are selected from the group consisting of at least one oxygen, or at least one nitrogen atom. In yet another variation, Z comprises of 1 atom. [0193] In another variation, Z is selected from the group consisting of -CH2-, -CH2CH2-, -CH2CH2CH2-, -C(O)-, -CH2C(O)-, -C(O)CH2-, -CH2-C(O)CH2-, -C(O)CH2CH2-, -CH2CH2C(O)-, -O-, -OCH2-, -CH2O-, -CH2OCH2-, -OCH2CH2-, -CH2CH2O-, -N(CH3)-, -NHCH2-, -CH2NH-, -CH2NHCH2-, -NHCH2CH2-, -CH2CH2NH-, -NH-C(O)-, -NCH3-C(O)-, -C(O)NH-, -C(O)NCH3-, -NHC(O)CH2-, -C(O)NHCH2-, -C(O)CH2NH-, -CH2NHC(O)-, -CH2C(O)NH-, -NHCH2C(O)-, -S-, -SCH2-, -CH2S-, -SCH2CH2-, -CH2SCH2-, -CH2CH2S-, -C(O)S-, -C(O)SCH2-, -CH2C(O)S-, -C(O)CH2S-, and -CH2SC(O)-, each substituted or unsubstituted.
[0194] In yet another variation, Z is selected from the group consisting of -CH2-, -C(O)-, -CH2C(O)-, -C(O)CH2-, -CH2-C(O)CH2-, -C(O)CH2CH2-, and -CH2CH2C(O)-, each substituted or unsubstituted.
[0195] In one variation of each of the above embodiments and variations comprising Z, Z provides 1-3 atom separation between Rm and the ring.
[0196] In another variation of each of the above embodiments and variations comprising Z, Z provides 1 atom separation between Rm and the ring. In one variation, the 1 atom separation is provided by an atom selected from the group consisting of C, N, O, and S. In another variation, the 1 atom separation is provided by a carbon atom. In still another variation, the 1 atom separation is provided by an oxygen atom. In yet another variation, the 1 atom separation is provided by a nitrogen atom.
[0197] In a further variation of each of the above embodiments and variations comprising Z, Z is selected from the group consisting of -CH2-, -CHR9-, -C(R9)(R9)-, -C(O)-, -C(S)-, -C(NH)-, -C(NR9)-, -O-, -N(H)-, -N(R9)-, and -S-.
[0198] In yet another variation of each of the above embodiments and variations comprising Z, Z is a carbonyl.
[0199] In another variation according to the present invention, Rm is a substituted or unsubstituted (C3-7)cycloalkyl. In yet another variation, Rm is a substituted or unsubstituted aryl. In yet another variation, Rm is a substituted or unsubstituted phenyl. [0200] In another variation, Rm is a ring having a non-hydrogen substituent at a 2 or 3 position of the ring. In another variation according to the present invention, Rm has a non- hydrogen substituent at a 2 or 3 position of the ring selected from.the group consisting of ( - 10)alkyl, (C3-ι2)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(Cι-ιo)alkyl, heteroaryl(Ci-5)alkyl, (C9-i2)bicycloaryl, hetero(C -ι2)bicycloaryl, carbonyl (Ci^alkyl, thiocarbonyl (Ci^alkyl, sulfonyl (Cι-3)alkyl, sulfinyl
Figure imgf000063_0001
imino (Cι.3)alkyl, amino, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl group, cyano, nitro, halo, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
[0201] In another variation, Rm is selected from the group consisting of alkyl, cycloalkyl, aryl, arylalkyl, heteroarylalkyl, and bicycloaryl, each substituted or unsubstituted. [0202] In another variation of each of the above embodiments and variations comprising Rm, Rm is selected from the group consisting of (2-cyano)phenyl, (3-cyano)phenyl, (2- hydroxy)phenyl, (3-hydroxy)phenyl, (2-alkenyl)phenyl, (3-alkenyl)phenyl, (2-alkynyl)phenyl, (3-alkynyl)phenyl, (2-nitro)phenyl, (3-nitro)phenyl, (2-carboxy)phenyl, (3-carboxy)phenyl, (2-carboxamido)phenyl, (3-carboxamido)ρhenyl, (2-sulfonamido)phenyl,
(3-sulfonamido)phenylj (2-tetrazolyl)phenyl, (3-tetrazolyl)phenyl, (2-arninomethyl)phenyl, (3- aminomethyl)phenyl, (2-amino)phenyl, (3-amino)phenyl, (2-hydroxymethyl)phenyl, (3-hydroxymethyl)phenyl, (2-phenyl)phenyl, (3-phenyl)phenyl, (2-CONH2)phenyl, (3-CONH2)phenyl, (2-CONH(Cι-7)alkyl)phenyl, (3-CONH(Cι-7)alkyl)phenyl,
(2-CO2(Cι-7)alkyl)phenyl, (3-CO2(Cι-7)alkyl)phenyl, -NH2, -OH, -(C3-7)alkyl, -alkene, -alkyne, -CCH, -(C3-7)cycloalkyl, and -aryl, each substituted or unsubstituted. [0203] In another variation, Ri is -(CR5R6)o-2-(R7)0.2-R8, where
R5 and Rδ are each independently hydrogen, halo or substituted or unsubstituted (Cι-8)alkyl or R5 and R6 are taken together to form a (C3-6)cycloalkyl ring;
R7 is selected from the group consisting of CO; CS; (Cι-ιo)alkylene,
Figure imgf000064_0001
hetero(C3-ι2)cycloalkylene, amino, arylene, heteroarylene, imine, sulfonyl, and sulfinyl, each substituted or unsubstituted; and
R8 is hydrogen or is selected from the group consisting of (Cι-7)alkyl, (C2-7)alkenyl, (C2-7)alkynyl, (C3-7)cycloalkyl, (C3-7)heterocycloalkyl, aryl, aryl(Ci-3)alkyl, heteroaryl(Cι- 2)alkyl, heteroaryl, -SH, (Cι-7)alkyl-S-, (Ci-7)alkyl-SO-, (Cι-7)alkyl-SO2-, CO, CS, NHCONNH2, -NHCSNH2, -NHCONH2, -NHCO(Cι-7)alkyl, -NHS02(Cι-7)alkyl, -OCO-(Q- 5)alkyl, cyano, nitro, halo, hydroxy, perhalo(Cι-7)alkyl, perhalo(Ci-7)alkyloxy, -SO2NH2, ( - ιo)alkyloxy, (C2-ιo)alkenyloxy, (C20)alkynyloxy, aryloxy, or heteroaryloxy, each substituted or unsubstituted.
[0204] In another particular variation, Ri is selected from the group consisting of -(CH2)-(2-cyano)ρhenyl, -(CH2)-(3-cyano)phenyl, -(CH2)-(2-hydroxy)phenyl, -(CH2)-(3- hydroxy)ρhenyl, -(CH2)-(2-alkenyl)phenyl, -(CH2)-(3-alkenyι)ρhenyl, -(CH2)-(2- alkynyl)phenyl, -(CH2)-(3-alkynyl)ρhenyl, -(CH2)-(2-nitro)phenyl, -(CH2)-(3-nitro)phenyl, -(CH2)-(2-carboxy)phenyl, -(CH2)-(3-carboxy)phenyl, -(CH2)-(2-carboxamido)phenyl, -(CH2)-(3-carboxamido)phenyl, -(CH2)-(2-sulfonamido)phenyl,
-(CH2)-(3-sulfonamido)phenyl, -(CH2)-(2-tetrazolyl)ρhenyl, -(CH2)-(3-tetrazolyl)phenyl, -(CH2)-(2-aminomethyl)phenyl, -(CH2)-(3-aminomethyl)phenyl, -(CH2)-(2-amino)phenyl, -(CH2)-(3-am o)phenyl, -(CH2)-(2-hyα^oxyme l)ρhenyl, -(CH2)-(3-hydroxymethyl)phenyl, -(CH2)-(2-phenyl)phenyl, -(CH2)-(3-phenyl)phenyl, -(CH2)-(2-CONH2)ρhenyl,
-(CH )-(3-CONH2)ρhenyl, -(CH2)-(2-CONH(Ci-7)alkyl)phenyl,
-(CH2)-(3-CONH(Cι-7)alkyl)phenyl, -(CH2)-(2-CO2(Ci-7)alkyl)phenyl,
-(CH2)-(3-CO2(Cι-7)alkyl)phenyl, -CH2-NH2, -CH2-OH, -CH2-(C3-7)alkyl, -CH2-alkene, -CH2-alkyne, -CH2-CCH, -CH2-(C3-7)cycloalkyl, and -CH2-aryl, each substituted or unsubstituted.
[0205] In another variation of each of the above embodiments and variations comprising Ri, Ri is selected from the group consisting of -(Cι)alkyl-aryl, -(Cι)alkyl-bicycloaryl, -aminoaryl, -aminoheteroaryl, -aminobicycloaryl, -aminoheterobicycloaryl, -O-aryl, -O- heteroaryl, -O-bicycloaryl, -O-heterobicycloaryl, -(S)-aryl, -(S)-heteroaryl, -(S)-bicycloaryl, - S-heterobicycloaryl, -C(O)-aryl, -C(O)-heteroaryl, - ., C(O)-bicycloaryl, -C(O)- heterobicycloaryl, -C(S)-aryl, -C(S)-heteroaryl, -C(S)-bicycloaryl, -C(S)-heterobicycloaryl, -S(O)-aryl, -S(O)-heteroaryl, -S(O)-bicycloaryl, -SO2-heterobicycloaryl, -SO2-aryl, -SO2- heteroaryl, -SO2-bicycloaryl, -SO2-heterobicycloaryl, -C(NR9)-aryl, -C(NR9)-heteroaryl, - C(NR9)-bicycloaryl, -C(NR9)-heterobicycloaryl, each substituted or unsubstituted. [0206] In still another variation of each of the above embodiments and variations comprising Ri, Ri is a substituted or unsubstituted aryl. In a further variation of each of the above embodiments and variations comprising Ri, Ri is a substituted or unsubstituted phenyl. In yet another variation of each of the above embodiments and variations comprising Ri, Ri is a substituted or unsubstituted heteroaryl.
Substituent R2
[0207] In one variation of the invention, R2 is selected from the group consisting of halo, perhalo(Cι-ιo)alkyl, amino, cyano, thio, (Crιo)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, carbonyl
Figure imgf000065_0001
thiocarbonyl (Cι.3)alkyl, sulfonyl (Cι-3)alkyl, sulfinyl (Ci. 3)alkyl, imino (Ci^alkyl, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, and a substituted or unsubstituted 4, 5, 6 or 7 membered ring. [0208] In another variation, R2 is selected from the group consisting of (C3-i2)cycloalkyl(Cι.5)alkyl, hetero(C3-i2)cycloalkyl(Cι-5)alkyl, aryl(Crιo)alkyl, heteroaryl(Cι- 5)alkyl, and (Cg-i2)bicycloaryl(Cι-5)alkyl, each substituted or unsubstituted. [0209] In yet another variation, R2 is selected from the group consisting of I, Br, CI, and F. In another particular variation, R2 is selected from the group consisting of -SH, -SCH3, and -S(Cι.3)alkyl unsubstituted or substituted through available valencips. [0210] In another variation of the invention, R2 is selected from the group consisting of -NH2, -NHCH3, -N(CH3)2, -NH(Cι-3)alkyl, -N(Cι.3-alkyl)2, -NR14R15, where RM is selected from the group consisting of a substituted or unsubstituted (Cs-^cycloalkyl, hetero(C32)cycloalkyl, (C62)aryl, hetero(C52)aryl, (C9-ι2)bicycloaryl and hetero(C4-ι2)bicycloaryl, and R15 is selected from the group consisting of hydrogen and a substituted or unsubstituted (Cι-8)alkyl, and -NR10R11, where Rio and -Rn together are -(CEJ^s- optionally interrupted by one O, S, NH or -N(Cι-3)alkyl group; each unsubstituted or substituted through available valencies.
[0211] Jn another variation according to the present invention, R2 is selected from the group consisting of a (Ci^alkyl; an amino, alkyl, or alkoxy carbonyl (Ci^alkyl; an amino, alkyl, or alkoxy thiocarbonyl (Cι-3)alkyl; an amino, alkyl, or alkoxy sulfonyl (Cι.3)alkyl; an amino, alkyl, or alkoxy sulfinyl (Cι.3)alkyl; an amino, alkyl, or alkoxy imino
Figure imgf000066_0001
an aryl (Ci-5)alkyl; and a heteroaryl (Cι-5)alkyl, each unsubstituted or substituted through available valencies.
[0212] Jn another variation according to the present invention, R2 is selected from the group consisting of a (C2-ιo)alkenyl; an amino, alkyl, or alkoxy carbonyl (Cι-3)alkenyl; an amino, alkyl, or alkoxy thiocarbonyl (Cι-3)alkenyl; an amino, alkyl, or alkoxy sulfonyl (Q. 3)alkenyl; an amino, alkyl, or alkoxy sulfinyl (Ci^alkenyl; an amino, alkyl, or alkoxy imino (Cι-3)alkenyl; an aryl (Cι-5)alkenyl, and a heteroaryl (Cι-5)alkenyl, each unsubstituted or substituted through available valencies.
[0213] In yet another variation according to the present invention, R2 is selected from the group consisting of a (C2-ιo)alkynyl; an amino, alkyl, or alkoxy carbonyl (Cι_3)alkynyl; an amino, alkyl, or alkoxy thiocarbonyl (Cι.3)alkynyl; an amino, alkyl, or alkoxy sulfonyl ( . 3)alkynyl; an amino, alkyl, or alkoxy sulfinyl (Ci^alkynyl; an amino, alkyl, or alkoxy imino (Cι.3)alkynyl; an aryl (C1-5) alkynyl; and a heteroaryl (Cι-5)alkynyl, each unsubstituted or substituted through available valencies.
[0214] In one variation of each of the above embodiments and variations comprising R2,
R2 is a substituted or unsubstituted 3, 4, 5, 6, or 7 membered ring.
[0215] In another variation, R2 is substituted or unsubstituted (C3-7)cycloalkyl (i.e., a substituted or unsubstituted 3, 4, 5, 6, or 7 membered cycloalkyl). Jn another variation, R2 is substituted or unsubstituted (C3-7)heterocycloalkyl (e.g., a substituted or unsubstituted 4, 5,
6,or 7 membered heterocycloalkyl). In another variation, R2 is substituted or unsubstituted aryl. In another variation, R2 is substituted or unsubstituted phenyl. In yet another variation,
R2 is substituted or unsubstituted heteroaryl.
[0216] In another variation, R2 is hydroxy.
[0217] In yet another variation according to the present invention, R2 is -ORn, where Rn is selected from the group consisting of substituted or unsubstituted alkyl, cycloalkyl, aryl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl.
[0218] In yet another variation, R2 is a carbonyl group. In another variation according to the present invention, R2 is selected from the group consisting of an aldehyde, acid, amide, ketone and ester.
[0219] In another variation according to the present invention, R2 is selected from the group consisting of pyrrolidin- 1 -ylcarbonyl, piperidin-1 -ylcarbonyl, and morpholin-4- ylcarbonyl, each unsubstituted or substituted through available valencies.
[0220] In another variation according to the present invention, R2 is selected from the group consisting of 3-amino-piperidin-l-yl, 3-aminomethyl-pyrrolidin-l-yl, azetidin-1-yl, 3- aminoazetidin-1-yl, pyrrolidin- 1-yl, 3-aminocyclopent-l-yl, 3-aminomethylcyclopent-l-yl, 3- aminomethylcyclohex-1-yl, hexahydroazepin-1-yl, 3-aminohexahydroazepin-l-yl, 3-amino- cyclohex-1-yl, piperazin- 1-yl, homopiperazin- 1-yl, 3-amino-pyrrolidin-l-yl, and R-3- aminopiperidin-1-yl, each substituted or unsubstituted.
[0221] In another variation of the present invention, R2 is MRaRb, where
M is CH or N, Ra is H or (Cι-6)alkyl, and R is amino(Co4)alkyl; or
M together with Ra and Rb form a ring of 4-7 atoms selected from the group consisting of C, N, O, and S each is independently substituted or unsubstituted and where C, N and S are optionally oxidized. [0222] In another variation, R2 comprises a primary or secondary amine where the nitrogen of the arnine is 1-6 atoms from the nearest ring atom. In another particular variation, R2 comprises a primary or secondary amine where the nitrogen of the amine is 2-5 atoms from the nearest ring atom. In yet another variation, R2 comprises a primary or secondary amine where the nitrogen of the amine is 3-4 atoms from the nearest ring atom. [0223] In a further variation of each of the above embodiments and variations comprising R2, R2 is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring wherein at least one substituent is selected from the group consisting of a primary, secondary or tertiary amine, a heterocycloalkyl comprising a nitrogen ring atom, and a heteroaryl comprising a nitrogen ring atom.
[0224] In a further variation of each of the above embodiments and variations comprising R2, R2 is selected from the group consisting of
Figure imgf000068_0001
wherein p is 0-12 and each R8 is independently selected from the group consisting of halo, perhalo(Cι-ιo)alkyl, CF3, cyano, nitro, hydroxy, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted. [0225] In yet another variation of each of the above embodiments and variations comprising R2, R2 is selected from the group consisting of
Figure imgf000068_0002
wherein r is 0-13 and each R8 is independently selected from the group consisting of halo, perhalo(Cι-ιo)alkyl, CF3, cyano, nitro, hydroxy, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
[0226] In another variation of each of the above embodiments and variations comprising
R2, R2 is a substituted or unsubstituted heteroaryl selected from the group consisting of furan, thiophene, pyrrole, pyrazole, triazole, isoxazole, oxazole, thiazole, isothiazole, oxadiazole, pyridine, pyridazine, pyrimidine, pyrazine, triazine, benzofuran, isobenzofuran, benzothiophene, isobenzothiophene, imidazole, benzimidazole, indole, isoindole, quinoline, isoquinoline, cinnoline, quinazoline, naphthyridine, pyridopyridine, quinoxaline, phthalazine, and benzothiazole, each substituted or unsubstituted.
[0227] In still another variation of each of the above embodiments and variations comprising R2, R2 is selected from the group consisting of (C3..ι2)cycloalkyl, hetero(C3-i2)cycloalkyl, aryl(Cι-ιo)alkyl, heteroaryl (Cι-5)alkyl, (Cg-^bicycloaryl, and hetero(C4.i2)bicycloaryl, each substituted or unsubstituted.
[0228] In a further variation of each of the above embodiments and variations comprising
R2, R2 is a substituted or unsubstituted (C3- )cycloalkyl ring, optionally comprising O, N(O),
N, S, SO, SO2 or a carbonyl group in the ring.
Substituent Rio
[0229] In one variation of each of the above embodiments and variations comprising R19, two R19 are taken together to form a substituted or unsubstituted ring.
[0230] In another variation of each of the above embodiments and variations comprising
R19, two Rig are taken together to form a substituted or unsubstituted fused or bridged ring.
[0231] In still another variation of each of the above embodiments and variations comprising Rig, two R19 are taken together to form a substituted or unsubstituted bridged or spiro ring.
Substituents U and V
[0232] In one variation of each of the above embodiments and variations comprising U,
U provides 1-4 atom separation between V and the ring. In another variation, U provides 1-3 atom separation between V and the ring.
[0233] In still another variation, U is selected from the group consisting of -CH2-,
-CH2CH2-, -CH2CH2CH2-, -C(O)-, -CH2C(O)-, -C(O)CH2-, -CH2-C(O)CH2-, -C(O)CH2CH2-,
-CH2CH2C(O)-, -O-, -OCH2-, -CH2O-, -CH2OCH2-, -OCH2CH2-, -CH2CH2O-, -N(CH3)-, -NHCH2-, -CH2NH-, -CH2NHCH2-, -NHCH2CH2-, -CH2CH2NH-, -NH-C(O)-, -NCH3-C(O)-,
-C(O)NH-, -C(O)NCH3-, -NHC(O)CH2-, -C(O)NHCH2-, -C(O)CH2NH-, -CH2NHC(O)-,
-CH2C(O)NH-, -NHCH2C(O)-, -S-, -SCH2-, -CH2S-, -SCH2CH2-, -CH2SCH2-, -CH2CH2S-,
-C(O)S-, -C(O)SCH2-, -CH2C(O)S-, -C(O)CH2S-, and -CH2SC(O)-, each substituted or unsubstituted.
[0234] In a further variation of each of the above embodiments and variations comprising
U, U is selected from the group consisting of -CH2-, -CHR9-, -C(R9)(R9)-, -O-, -N(H)-,
-N(R9)-, and -S-.
[0235] In one variation of each of the above embodiments and variations comprising V,
V is selected from the group consisting of a primary, secondary or tertiary amine, a heterocycloalkyl comprising a nitrogen ring atom, and a heteroaryl comprising a nitrogen ring atom
[0236] In another variation of each of the above embodiments and variations comprising
V, V is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring wherein at least one substituent is selected from the group consisting of a primary, secondary or tertiary amine, a heterocycloalkyl comprising a nitrogen ring atom, and a heteroaryl comprising a nitrogen ring atom.
[0237] In still another variation of each of the above embodiments and variations comprising V, the basic nitrogen of V is separated from the ring atom to which R2 is attached by between 1-5 atoms.
[0238] In a further variation of each of the above embodiments and variations comprising
V, the basic nitrogen of V forms part of a primary, secondary or tertiary amine.
[0239] In yet another variation of each of the above embodiments and variations comprising V, the basic nitrogen of V is a nitrogen ring atom of a heterocycloalkyl comprising a nitrogen ring atom or a heteroaryl comprising a nitrogen ring atom.
[0240] In one variation of each of the above embodiments and variations comprising
-UV, -UV is selected from the group consisting of
Figure imgf000070_0001
wherein p is 0-12 and each R8 is independently selected from the group consisting of halo, perhalo(Cι-ιo)alkyl, CF3, cyano, nitro, hydroxy, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, with the proviso that at least one R8 serves as V.
[0241] In another variation of each of the above embodiments and variations comprising -UV, -UV is selected from the group consisting of
Figure imgf000071_0001
wherein r is 0-13 and each R8 is independently selected from the group consisting of halo, perhalo(Cι-ιo)alkyl, CF3, cyano, nitro, hydroxy, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, with the proviso that at least one R8 serves as V.
[0242] In one variation of each of the above embodiments and variations comprising R8, at least one R8 is a primary, secondary or tertiary amine.
[0243] In another variation of each of the above embodiments and variations comprising
R8, at least one R8 is a substituted or unsubstituted heterocycloalkyl comprising a nitrogen ring atom or a substituted or unsubstituted heteroaryl comprising a nitrogen ring atom.
[0244] In still another variation of each of the above embodiments and variations comprising R8, at least one R8 is selected from the group consisting of -NH2, -NH(Cι_s alkyl), -
N(Cι-5 alkyl) 2, piperazine, imidazole, and pyridine.
[0245] In another variation according to the present invention,
Ri is selected from the group consisting of -(CH2)-(2-cyano)phenyl,
-(CH2)-(3-cyano)ρhenyl, -(CH )-(2-hydroxy)ρhenyl, -(CH2)-(3-hydroxy)ρhenyl, -(CH2)-(2- alkenyl)phenyl, -(CH2)-(3-alkenyl)phenyl, -(CH2)-(2-alkynyl)phenyl, -(CH2)-(3- alkynyl)phenyl, -(CH2)-(2-nitro)phenyl, -(CH2)-(3-nitro)phenyl, -(CH2)-(2-carboxy)ρhenyl, -(CH2)-(3-carboxy)phenyl, -(CH2)-(2-carboxamido)phenyl, -(CH2)-(3-carboxamido)phenyl, -(CH2)-(2-sulfonarmdo)phenyl, -(CH2)-(3-sulfonamido)phenyl, -(CH2)-(2-tetrazolyl)phenyl, -(CH2)-(3-tefrazolyl)phenyl, -(CH2)-(2-ammomemyl)phenyl, -(CH2)-(3-aminomethyl)phenyl, -(CH2)-(2-amino)phenyl, -(CH2)-(3-amino)phenyl, -(CH2)-(2-hydroxymethyl)phenyl, -(CH2)-(3-hydroxymethyl)ρhenyl, -(CH2)-(2-ρhenyl)phenyl, -(CH2)-(3-ρhenyl)ρhenyl, -(CH2)-(2-halo)phenyl, -(CH2)-(3-halo)phenyl, -(CH2)-(2-CONH2)phenyl,
-(CH2)-(3-CONH2)phenyl, -(CH2)-(2-CONH(Cι-7)alkyl)phenyl,
-(CH2)-(3-CONH(Cι-7)alkyl)phenyl, -(CH2)-(2-CO2(Ci-7)alkyl)phenyl,
-(CH2)-(3-CO2(Ci-7)alkyl)phenyl, -CH2-NH2, -CH2-OH, -CH2-(C3-7)alkyl, -CH2-alkene, -CH2-alkyne, -CH2-CCH, -CH2-(C3-7)cycloalkyl, and -CH2-aryl, each substituted or unsubstituted, and
R2 is selected from the group consisting of 3-amino-piperidin-l-yl, 3-aminomethyl- pyrrolidin-1-yl, azetidin-1-yl, 3-aminoazetidin-l-yl, pyrrohdin-1-yl, 3-aminocyclopent-l-yl, 3- aminomethylcyclopent-1-yl, 3-aminomethylcyclohex-l-yl, hexahydroazepin-1-yl, 3- aminohexahydroazepin-1-yl, 3-amino-cyclohex-l-yl, piperazin- 1-yl, homopiperazin- 1-yl, 3- amino-pyrrolidin-1-yl, and R-3-aminopiperidin-l-yl, each substituted or unsubstituted. [0246] In another particular variation according to the present invention,
Ri is selected from the group consisting of -NH2, -NHCH3, -N(CH3)2, -NHCi-3-alkyl, and -N(Cι-3-alkyl)2 each unsubstituted or substituted through available valencies, and
R2 is selected from the group consisting of 3-amino-piperidin-l-yl, 3-aminomethyl- pyrrolidin-1-yl, azetidin-1-yl, 3-aminoazetidin-l-yl, pyrrolidin- 1-yl, 3-aminocyclopent-l-yl, 3- aminomethylcyclopent-1-yl, 3-aminomethylcyclohex-l-yl, hexahydroazepin-1-yl, 3- aminohexahydroazepin-1-yl, 3-amino-cyclohex-l-yl, piperazin- 1-yl, homopiperazin- 1-yl, 3- amino-pyrrolidin-1-yl, and R-3-aminopiperidin-l-yl, each substituted or unsubstituted. [0247] In another variation according to the present invention,
Q is CO,
Ri is selected from the group consisting of -(CH2)-(2-cyano)phenyl, -(CH2)-(3-cyano)phenyl, -(CH )-(2-hydroxy)phenyl, -(CH2)-(3-hydroxy)phenyl, -(CH2)-(2- alkenyl)phenyl, -(CH2)-(3-alkenyl)phenyl, -(CH2)-(2-alkynyl)phenyl, -(CH2)-(3- alkynyl)ρhenyl, -(CH2)-(2-nitro)phenyl, -(CH2)-(3-nitro)ρhenyl, -(CH2)-(2-carboxy)ρhenyl, -(CH2)-(3-carboxy)phenyl, -(CH2)-(2-carboxamido)phenyl, -(CH2)-(3-carboxamido)phenyl, -(CH2)-(2-sulfonamido)phenyl, -(CH2)-(3-sulfonamido)phenyl, -(CH2)-(2-tetrazolyl)phenyl, -(CH2)-(3-tetrazolyl)phenyl, -(CH2)-(2-aπι ^
-(CH2)-(2-amino)phenyl, -(CH2)-(3-amino)phenyl, -(CH2)-(2-hydroxymethyi)phenyl, -(CH2)-(3-hydroxymethyl)phenyl, -(CH2)-(2-phenyl)phenyl, -(CH2)-(3-ρhenyl)phenyl, -(CH2)-(2-halo)phenyl, -(CH2)-(3-halo)phenyl, -(CH2)-(2-CONH2)phenyl,
-(CH2)-(3-CONH2)phenyl, -(CH2)-(2-CONH(Cι-7)alkyl)phenyl,
-(CH2)-(3-CONH(Cι-7)alkyl)phehyl, -(CH2)-(2-CO2(Cι-7)alkyl)phenyl,
-(CH2)-(3-CO2(Cι-7)alkyl)phenyl, -CH2-NH2, -CH2-OH, -CH2-(C3-7)alkyl, -CH2-alkene, -CH2-alkyne, -CH2-CCH, -CH2-(C3-7)cycloalkyl, and -CH2-aryl, each substituted or unsubstituted, and
R2 is selected from the group consisting of 3-amino-piperidin-l-yl, 3-aminomethyl- pyrrolidin-1-yl, azetidin-1-yl, 3-aminoazetidin-l-yl, pyrrolidin- 1-yl, 3-aminocyclopent-l-yl, 3- aminomethylcyclopent-1-yl, 3-aminomethylcyclohex-l-yl, hexahydroazepin-1-yl, 3- aminohexahydroazepin-1-yl, 3-amino-cyclohex-l-yl, piperazin- 1-yl, homopiperazin- 1-yl, 3- amino-pyπOlidin-1-yl, and R-3-aminopiperidin-l-yl, each substituted or unsubstituted. [0248] In another variation according to the present invention,
Q is SO,
Ri is selected from the group consisting of -(CH2)-(2-cyano)phenyl, -(CH2)-(3-cyano)ρhenyl, -(CH2)-(2-hydroxy)ρhenyl, -(CH2)-(3-hydroxy)phenyl, -(CH2)-(2- alkenyl)phenyl, -(CH2)-(3-alkenyl)phenyl, -(CH2)-(2-alkynyl)phenyl, -(CH2)-(3- alkynyl)phenyl, -(CH2)-(2-nitro)phenyl, -(CH2)-(3-nitro)phenyl, -(CH2)-(2-carboxy)phenyl, -(CH2)-(3-carboxy)phenyl, -(CH2)-(2-carboxamido)phenyl, -(CH2)-(3-carboxamido)phenyl, -(CH2)-(2-sulfonamido)phenyl, -(CH2)-(3-sulfonamido)phenyl, -(CH2)-(2-tetrazolyl)phenyl, -(CH2)-(3-tefrazolyl)phenyl, -(CH2)-(2-aminomemyl)phenyl, -(CH2)-(3-aminomethyl)phenyl, -(CH2)-(2-amino)phenyl, -(CH2)-(3-amino)phenyl, -(CH2)-(2-hycfroxymethyl)phenyl, -(CH2)-(3-hydroxymethyl)ρhenyl, -(CH2)-(2-ρhenyl)ρhenyl, -(CH2)-(3-ρhenyl)phenyl, -(CH2)-(2-halo)phenyl, -(CH2)-(3-halo)phenyl, -(CH2)-(2-CONH2)phenyl,
-(CH2)-(3-CONH2)phenyl, -(CH2)-(2-CONH(Ci-7)alkyl)phenyl,
-(CH2)-(3-CONH(Cι-7)alkyl)phenyl, -(CH2)-(2-CO2(Cι-7)alkyl)phenyl,
-(CH2)-(3-CO2(Cι-7)alkyl)phenyl, -CH2-NH2, -CH2-OH, -CH2-(C3-7)alkyl, -CH2-alkene, -CH2-alkyne, -CH2-CCH, -CH2-(C3- )cycloalkyl, and -CH2-aryl, each substituted or unsubstituted, and
R2 is selected from the group consisting of 3-amino-piperidin-l-yl, 3-aminomethyl- pyrcolidin-1-yl, azetidin-1-yl, 3-ammoazetidin-l-yl, pyrrohdin-1-yl, 3-aminocyclopent-l-yl, 3- aminomethylcyclopent-1-yl, 3-aminomethylcyclohex-l-yl, hexahydroazepin-1-yl, 3- aminohexahydroazepin-1-yl, 3-amino-cyclohex-l-yl, piperazin- 1-yl, homopiperazin- 1-yl, 3- ammo-pyrrolidin-1-yl, and R-3-aminopiperidin-l-yl, each substituted or unsubstituted. [0249] In yet another variation according to the present invention,
Q is SO2,
Ri is selected from the group consisting of-(CH2)-(2-cyano)phenyl, -(CH2)-(3-cyano)phenyl, -(CH2)-(2-hydroxy)phenyl, -(CH2)-(3-hydroxy)phenyl, -(CH2)-(2- alkenyl)ρhenyl, -(CH2)-(3-alkenyl)phenyl, -(CH2)-(2-alkynyl)phenyl, -(CH2)-(3- alkynyl)phenyl, -(CH2)-(2-nitro)phenyl, -(CH2)-(3-nitro)phenyl, -(CH2)-(2-carboxy)phenyl, -(CH2)-(3-carboxy)phenyl, -(CH2)-(2-carboxarmdo)phenyl, -(CH2)-(3-carboxamido)phenyl, -(CH2)-(2-sulfonamido)phenyl, -(CH2)-(3-sulfonamido)phenyl, -(CH2)-(2-tetrazolyl)phenyl, -(CH2)-(3-tefrazolyl)phenyl, -(CH2)-(2-ammomemyl)phenyl, -(CH2)-(3-am omethyl)phenyl, -(CH2)-(2-amino)phenyl, -(CH2)-(3-amino)phenyl, -(CH2)-(2-hydroxymethyl)phenyl, -(CH2)-(3-hydroxymethyl)phenyl, -(CH2)-(2-ρhenyl)phenyl, -(CH2)-(3-phenyl)phenyl, -(CH2)-(2-halo)phenyl, -(CH2)-(3-halo)phenyl, -(CH2)-(2-CONH2)ρhenyl,
-(CH2)-(3-CONH2)phenyl, -(CH2)-(2-CONH(Cι-7)alkyl)phenyl,
-(CH2)-(3-CONH(Ci-7)alkyl)phenyl, -(CH2)-(2-CO2(Cι-7)alkyl)phenyl,
-(CH2)-(3-CO2(Ci-7)alkyl)phenyl, -CH2-NH2, -CH2-OH, -CH2-(C3-7)alkyl, -CH2-alkene, -CH2-alkyne, -CH2-CCH, -CH2-(C3- )cycloalkyl, and -CH2-aryl, each substituted or unsubstituted, and
R2 is selected from the group consisting of 3-amino-piperidin-l-yl, 3-aminomethyl- pyrrohdin-1-yl, azetidin-1-yl, 3-aminoazetidin-l-yl, pyrroUdin-1-yl, 3-aminocyclopent-l-yl, 3- aminomethylcyclopent-1-yl, 3-aminomethylcyclohex-l-yl, hexahydroazepin-1-yl, 3- aminohexahydroazepin-1-yl, 3-amino-cyclohex-l-yl, piperazin- 1-yl, homopiperazin- 1-yl, 3- amino-pyπolidin-1-yl, and R-3-aminopiperidin-l-yl, each substituted or unsubstituted. [0250] In yet another variation according to the present invention,
Q is CO, Ri is selected from the group consisting of -NH2, -NHCH3, -N(CH3)2, -NHCι.3-alkyl, and -N(Cι-3-alkyl)2 each unsubstituted or substituted through available valencies, and
R2 is selected from the group consisting of 3-amino-piperidin-l-yl, 3-aminomethyl- pyrrohdin-1-yl, azetidin-1-yl, 3-aminoazetidin-l-yl, pyrrohdin-1-yl, 3-aminocyclopent-l-yl, 3- aminomethylcyclopent-1-yl, 3-aminomethylcyclohex-l-yl, hexahydroazepin-1-yl, 3- aminohexahydroazepin-1-yl, 3-amino-cyclohex-l-yl, piperazin- 1-yl, homopiperazin- 1-yl, 3- amino-pyrrolidin-1-yl, and R-3-aminopiperidin-l-yl, each substituted or unsubstituted. [0251] In another variation according to the present invention,
Q is SO,
Ri is selected from the group consisting of -NH2, -NHCH3, -N(CH3)2, -NHC1.3-a.kyl, and -N(Ci-3-alkyl)2 each unsubstituted or substituted through available valencies, and
R2 is selected from the group consisting of 3-amino-piperidin-l-yl, 3-aminomethyl- pyrcolidin-1-yl, azetidin-1-yl, 3-aminoazetidin-l-yl, pyrrolidin- 1-yl, 3-aminocyclopent-l-yl, 3- aminomethylcyclopent-1-yl, 3-aminomethylcyclohex-l-yl, hexahydroazepin-1-yl, 3- aminohexahydroazepin-1-yl, 3-amino-cyclohex-l-yl, piperazin- 1-yl, homopiperazin- 1-yl, 3- amino-pyrrolidin-1-yl, and R-3-aminopiperidin-l-yl, each substituted or unsubstituted. [0252] In another variation according to the present invention,
Q is SO2,
Ri is selected from the group consisting of -NH2, -NHCH3, -N(CH3)2, -NHCι.3-alkyl, and -N(Cι-3-alkyl)2 each unsubstituted or substituted through available valencies, and
R2 is selected from the group consisting of 3-amino-piperidin-l-yl, 3-aminomethyl- pyrcolidin-1-yl, azetidin-1-yl, 3-aminoazetidin-l-yl, pyrrolidin- 1-yl, 3-aminocyclopent-l-yl, 3- aminomethylcyclopent-1-yl, 3-aminomethylcyclohex-l-yl, hexahydroazepin-1-yl, 3- aminohexahydroazepin-1-yl, 3-amino-cyclohex-l-yl, piperazin- 1-yl, homopiperazin- 1-yl, 3- amino-pyrrolidin-1-yl, and R-3-aminopiperidin-l-yl, each substituted or unsubstituted. [0253] Particular examples of DPP-TV inhibitors according to the present invention include:
2-[2-(3-Anιino-piperidin-l-yl)-4-oxo-4H-quinazolin-3-ylmemyl]-benzonitrile;
2,4-Dichloro-quinazoline;
2-Chloro-3H-quinazolin-4-one;
2-(2-CUoro-4-oxo-4H-quinazolm-3-ylmethyl)-benzonitrile; 2-[2-(3-Amino-piperidm-l-yl)-6,7-dimethoxy-4-oxo-4H-quinazolin-3-ylmethyl]- benzonitrile;
2-Chloro-6,7-dimethoxy-3H-quinazolin-4-one;
2-(2-CMoro-6,7-άιmemoxy-4-oxo-4H-quinazolm-3-ylmemyl)-benzoniuile;
2-[2-(3-Ammo-piperidin-l-yl)-8-methoxy-4-oxo-4H-quinazolin-3-ylmethyl]- benzonitrile;
8-Methoxy-lH-quinazoline-2,4-dione;
2,4-Dichloro-8-methoxy-quinazoline;
2-Chloro-8-methoxy-3H-quinazolin-4-one;
2-(2-CUoro-8-memoxy-4-oxo-4H-quinazolin-3-yhnemyl)-benzonitrile;
2-[2-(3-Amino-piperidin-l-yl)-7-chloro-4-oxo-4H-quinazolin-3-ylmethyl]- benzonitrile, TFA salt;
2,7-Dichloro-5H-quinazolin-4-one;
2-(2,7-Dichloro-4^oxo-4H-qumazolin-3-ylmethlyl)-benzonitrile;
2-[2-(3-Amino-piperidin-l-yl)-8-chloro-4-oxo-4H-quinazolin-3-ylmethyl]- benzonitrile, TFA salt;
2,8-Dichloro-3Η-quinazolin-4-one;
2-(2,8-Dichloro-4-oxo-4H-quinazolin-3-ylmethlyl)-benzonitrile;
6-Fluoro- lH-quinazoline-2,4-dione;
2,4-Dichloro-6-fluoro-quinazoline;
2-Chloro-6-fluoro-3H-quinazolin-4-one;
2-(2-CUoro-6-fluoro-4-oxo-4H-quinazolin-3-ylmethyl)-benzonitrile;
(R) 2-[2-(3-An3ino-piperi(hn-l-yl)-6-fluoro-4-oxo-4H-qumazolin-3-ylmethyl]- benzonitrile TFA salt;
2-[2-(3-Armno-piperidin-l-yl)-7-memyl-6-oxo-6,7-dihydro-purin-l-ylmethyl]- benzonitrile;
2-[2-(3-Arnino-piperidin-l-yl)-9-memyl-6-oxo-6,9-dihydro-purin-l-ylmethyl]- benzonitrile;
2,6-Dichloro-7-methyl-7H-purine;
2,6-Dichloro-9-methyl-9H-purine;
2-Chloro-7-methyl- 1 ,7-dihydro-purin-6-one; 2-Chloro-9-methyl- 1 ,9-άιhydro-purin-6-one;
2-(2-Chloro-7-memyl-6-oxo-6,7-dihydro-purin- 1 -ylmethyl)-benzonitrile;
2-(2-Chloro-9-methyl-6-oxo-6,9-dihydro-purin- 1 -ylmethyl)-benzonitrile;
2-{2-[(R)-3-Amino-piperidin-l-yl]-6-oxo-6,7-dmydro-purin-l-ylmethyl}- benzonitrile;
7-Benzyloxymethyl-2,6-dichloro-7H-purine;
9-Benzyloxymethyl-2,6-dichloro-9H-purine;
7-Benzyloxymethyl-2-chloro- 1 ,7-dihydro-purin-6-one;
9-Benzyloxymethyl-2-chloro- 1 ,9-dihydro-purin-6-one;
2-(7-Berιzyloxymemyl-2-chloro-6-oxo-6,7-α hydro-purm-l-ylmemyl)-benzomt^^
2-(9-Benzyloxymemyl-2-c oro-6-oxo-6,9-dΗιydro-purm-l-ylmemyl)-benzonitrile;
2-(2-Chloro-6-oxo-6,9-dihydro-purin- 1 -ylmethyl)-benzonitrile;
2-[2-(3-(R)-Amino-piperidm-l-yl)-6-cUoro-4-oxo-4H-quinazoUn-3-ylmethyl]- benzonitrile, TFA salt;
2,6-Dichloro-3H-quinazolin-4-one;
2-(2,6-DicWoro-4-oxo-4H-quinazolm-3-ylmemyl)-benzonitrile;
2-[2-(3-(R)-Ammo-piperidin-lyl)-7-fluoro-6-methoxy-4-oxo-4H-quinazolin-3- ylmethylj-benzonitrile, TFA salt;
7-Fluoro-6-methoxy-lH-quinazoline-2,4-dione;
2-Chloro-7-fluoro-6-methoxy-3H-quinazolin-4-one;
2-(2-Chloro-7-fluoro-6-memoxy-4-oxo-4H-qumazolin-3-ylmemyl)-benzonitrile;
2-[2-(3-(R)-Anιino-piperiά n-lyl)-6-methoxy-4-oxo-4H-pyrido[3,4--flpyrirnidin-3- ylmethyl]-benzonitrile, TFA salt;
6-Memoxy-lH-pyrido[3,4- Ipyrimidine-2,4-dione;
2-Chloro-6-methoxy-3H-pyrido [3 ,4-<i]pyrimidin-4-one;
2-(2-CWoro-6-memoxy-4-oxo-4H-pyrido[3,4--i]pyrimidin-3-ylmethyl)- benzonitrile;
2-[6-(3-(R)-Ammo-piperichn-l-yl)-l-memyl-4-oxo-l,4-dmydro-pyrazolo[3,4- <^pyrirnidin-5-ylmethyl]-benzonitrile, TFA salt;
6-Chloro-l-memyl-l,5-o hydro-pyrazolo[3,4-<i]pyriπήdin-4-one; 2-(6-CUoro-l-memyl-4-oxo-l,4-dmydro-pyrazoto^ benzonitrile;
2-[2-(3-(R)-Amino-piperid -lyl)-5-fluoro-4-oxo-4H-quinazolin-3-ylmethyl]- benzonitrile, TFA salt;
2-Chloro-5-fluoro-3H-quinazolin-4-one;
2-(2-CWoro-5-fluoro-4-oxo-4H-quinazoUn-3-ylmethyl)-benzonitrile;
2-[5-(3-(R)-Amino-piperidin- 1 -yl)- 1 -methyl-7-oxo- 1 ,7-dihydro-[ 1 ,2,3]triazolo[4,5- ^pyrimidin-6-ylmethyl]-benzonitrile, TFA salt;
5-Chloro- 1 -methyl- 1 ,6-dihydro-[ 1 ,2,3]triazolo[4,5--i]pvrirnidin-7-one;
2-(5-Chloro- 1 -methyl-7-oxo- 1 ,7-dihydro-[ 1 ,2,3]triazolo[4,5-^pyrirnidin-6- ylmethyl)-benzonitrile;
2-[5-(3-(R)-Amino-ρiperiώn-l-yl)-2-me l-7-oxo-2,7-dihydro[l,2,3]triazolo[4,5- cflpyrimidin-6-ylmethyl] -benzonitrile, TFA salt;
5-CMoro-2-memyl-2,6-dihydro-[ 1 ,2,3]triazolo[4,5-d]pyrimidin-7-one;
2-(5-Chloro-2-me l-7-oxo-2,7-mhydro-[l,2,3]triazolo[4,5-^ρvrimidin-6- ylmefhyl)-benzonitrile;
2-[2-(3-(R)-Ammo-piperidin-lyl)-4-oxo-5,6,7,8-tetrahydro-4H-quinazolin-3- ylmethyl]-benzonitrile, TFA salt;
2-Chloro-5,6,7,8-tetrahydro-3H-quinazolin-4-one;
2-(2-Chloro-4-oxo-5,6,7,8-tefrahydro-4H-quinazolin-3-ylmemyl)-benzomtrile;
2-[2-(3-(R)-Ammo-piperidin-lyl)-6-cUoro-4-oxo-4H-pyrido[3,4-cflpyrimidin-3- ylmethyl] -benzonitrile, TFA salt;
1 ,7-Dihydro-pyrido [3 ,4--i]pyrimidine-2,4,6-trione;
2,6-Dichloro-3H-pyrido[3,4- ]pyrimidine-4-one;
2-(2,6-DicUoro-4-oxo-4H-pyrido[3,4-^pyrimidin-3-ylmethyl)-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-lyl)-4-oxo-6-pyrrolidin-lyl-4H-pyrido[3,4- <i]pyrimidin-3-ylmethyl]-benzonitrile;
2-[(R)-3-Amino-piperidin-l-yl]-6-fluoro-3-(2-trifluoromethyl-benzyl)-3Η- quinazolin-4-one;
2-CWoro-6-fluoro-3-(2-trifluoromemyl-benzyl)-3H-quinazolin-4-one; 2- { 2- [(R)-3-Amino-piperidin- 1 -yl]-7risopropyl-6-oxo-6,7-dmydro-purin- 1 - ylmethyl } -benzonitrile;
2-[2-(3-Amino-azepan-l-yl)-6-oxo-6,7-dmydro-purin-l-ylmemyl]-benzonitrile;
2- { 2- [(R)-3-Amino-piperidin- 1 -yl] -7-benzyl-6-oxo-6-hydro-purin- 1 -ylmethyl } - benzonitrile;
2- { 2- [(R)-3-Amino-piperidin- 1 -yl] -9-(2-cyano-benzyl)-6-oxo-6-hydro-purin- 1 - ylmethyl } -benzonitrile;
2- { 2- [(R)-3- Amino-piperidin- 1 -yl]-6-oxo-9-propyl-6,9-dihydro-purin- 1 -ylmethyl } - benzonitrile;
2-{2-[(R)-3-Arnmo-piperidin-l-yl]-6-oxo-7-propyl-6,7-d ydro-purin-l-ylmethyl}- benzonitrile;
2-Chloro-9-propyl- 1 ,9-dihydiO-purin-6-one;
2-Chloro-7-propyl- 1 ,7-dihydro-purin-6-one;
2-(2-CUoro-6-oxo-9-propyl-6,9-dihydro-purin- 1 -ylmethyl)-benzonitrile;
2-(2-Chloro-6-oxo-7-propyl-6,7-dihydro-purin- 1 -ylmethyl)-benzonitrile;
2-{2-[(R)-(3-Amino-piperidin-l-yl)]-9-isopropyl-6-oxo-8-trifluoromethyl-6,9- dihydro-purin- 1 -ylmethyl } -benzonitrile ;
6-Chloro-N4-isopropyl-pyrimidine-2,4,5-triamine;
6-Chloro-9-isopropyl-8-trifluoromethyl-9H-purm-2-ylamine;
2-(2-Aπιmo-9-isopropyl-6-oxo-8-frifluoromethyl-6,9-dmydro-purin-l-ylmethyl)- benzonitrile;
2-(2-Bromo-9-isopropyl-6-oxo-8-trifluoromethyl-6,9-dihydro-purin-l-ylmethyl)- benzonitrile;
2-[2-(3-(R)-Amino-piperidin-l-yl)-6-bromo-4-oxo-4H-quinazolin-3-ylmethyl]- benzonitrile;
6-Bromo- lH-quinazoline-2,4-dione;
6-Bromo-2-chloro-3H-quinazolin-4-one;
2-(6-Bromo-2-c oro-4-oxo-4H-quinazolin-3-ylmemyl)-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-l-yl)-6-bromo-4-oxo-4H-quinazoUn-3-ylmethyl]- benzonitrile, TFA salt; 2-[2-(3-(R)-Aιmno-pyrrohdin-l-yl)-6-bromo-4-oxo-4H-quinazolin-3-ylmethyl]- benzonitrile, TFA salt;
2-[2-(3-(R)-Amino-piperidin-l-yl)-6,8-dichloro-4-oxo-4H-quinazolin-3-ylmethyl]- benzonitrile;
6,8-Dichloro- 1 H-quinazoline-2,4-dione;
2,6,8-Trichloro-3H-quinazolin-4-one;
2-(2,6,8-Trichloro-4-oxo-4H-quinazolin-3-ylmethyl)-benzonitrile;
2-[2-(3-(R)-Ammo-piperiojn-l-yl)-6-memoxy-4-oxo-4H-quinazolin-3-ylmethyl]- benzonitrile;
6-Methoxy- lH-quinazoline-2,4-dione;
2,4-Dichloro-6-methoxy-quinazoline;
2-Chloro-6-methoxy-3H-quinazolin-4-one;
2-(2-C oro-6-memoxy-4-oxo-4H-quinazolin-3-ylmemyl)-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-l-yl)-6-fluoro-4-oxo-4H-quinazolin-3-ylmethyl]- benzamide;
2-[2-(3-(R)-Amino-piperidin-l-yl)-6-fluoro-7-morpholin-4-yl-4-oxo-4H- quinazolin-3-ylmethyl]-benzonitrile;
6,7-Difluoro-lH-quinazoline-2,4-dione;
6-Ruoro-7-morpholin-4-yl- 1 H-quinazoline-2,4-dione;
2,4-Dichloro-6-fluofo-7-morphoUn-4-yl-quinazoline;
2-Crdoro-6-fluoro-7-morpholin-4-yl-3H-quinazolin-4-one;
2-(2-CUoro-6-fluoro-7-morpholin-4-yl-4-oxo-4H-quinazolin-3-ylmethyl)- benzonitrile;
2-[2-(3-Amino-piperidin-l-yl)-6-fluoro-4-oxo-4H-quinazolin-3-ylmethyl]- benzamide;
2-[3-(R)-Anιino-piperiό n-l-yl]-6-fluoro-3-(2-trifluoromethyl-benzyl)-3H- quinazolin-4-one;
2-Aminomethyl-3-phenyl-3H-quinazolin-4-one;
2-Emylaminomethyl-3-phenyl-3H-quinazolin-4-one;
[(4-Oxo-3-phenyl-3,4-dihydro-quinazoUn-2-ylmethyl)-amino]-acetic acid methyl ester; [(4-Oxo-3-phenyl-3 ,4-dmydro-quinazol -2-ylmemyl)-amino]-acetic acid;
2-Aminomemyl-3-(2,4-ά^cMoro-phenyl)-3H-quinazolin-4-one;
2-Aminomethyl-3-(2-chloro-phenyl)-3H-quinazolin-4-one;
2-Aminomemyl-3-(4-cWoro-phenyl)-3H-quinazolin-4-one;
2-(3-Amino-piperiό^n-l-yl)-6,7-dimethoxy-3-(2-nitro-benzyl)-3H-quinazolin-4- one;
2-[2-(3-Amino-piperidin-l-yl)-6,7-dimemoxy-4-oxo-4H-qumazoUn-3-ylmethyl]- benzoic acid ethyl ester;
2-[2-(3-Amino-piperidin-l-yl)-6-fluoro-4-oxo-4H-quinazolin-3-ylmethyl]-berizoic acid ethyl ester;
2-[2-(3-Amino-piperidin-l-yl)-6,7-d memoxy-4-oxo-4H-quinazθiin-3-ylmethyl]- benzoic acid;
2-[2-(3-Anωno-piperidm-l-yl)-6-fluoro-4-oxo-4H-quinazolin-3-ylmem^ acid; and
2-(6,7-Dimemoxy-4-oxo-2-piperiά^n-l-yl-4H-quinazolin-3-ylmethyl)-benzonitrile. [0254] In another embodiment, the present invention provides the compounds in the form of a pharmaceutically acceptable salt.
[0255] In yet another embodiment, the present invention provides the compounds present in a mixture of stereoisomers. In yet another embodiment, the present invention provides the compounds as a single stereoisomer.
[0256] Jn yet another embodiment, the present invention provides pharmaceutical compositions comprising the compound as an active ingredient. In yet another variation, the present invention provides pharmaceutical compositions wherein the composition is a solid formulation adapted for oral administration. In yet another particular variation, the present invention provides pharmaceutical composition wherein the composition is a tablet. In another particular variation, the present invention provides the pharmaceutical composition wherein the composition is a liquid formulation adapted for oral administration. In yet another particular variation, the present invention provides pharmaceutical composition wherein the composition is a liquid formulation adapted for parenteral administration. [0257] In yet another particular variation, the present invention provides the pharmaceutical composition comprising the compound of the invention wherein the composition is adapted for administration by a route selected from the group consisting of orally, parenterally, intraperitoneally, intravenously, intraarterially, transdermally, sublingually, intramuscularly, rectally, transbuccally, intranasally, liposomally, via inhalation, vaginally, intraoccularly, via local delivery (for example by catheter or stent), subcutaneously, intraadiposally, intraarticularly, and intrathecally.
[0258] In another embodiment, the present invention provides a kit comprising a compound of the present invention and instructions which comprise one or more forms of information selected from the group consisting of indicating a disease state for which the compound is to be administered, storage information for the compound, dosing information and instructions regarding how to administer the compound. Jn another embodiment, the present invention provides the kit that comprises the compound in a multiple dose form. [0259] In another embodiment, the present invention provides an article of manufacture comprising a compound of the present invention, and packaging materials. Jn another variation, the packaging material comprises a container for housing the compound. In yet another variation, the invention provides the article of manufacture wherein the container comprises a label indicating one or more members of the group consisting of a disease state for which the compound is to be administered, storage information, dosing information and/or instructions regarding how to administer the composition.
[0260] In another variation, the present invention provides the article of manufacture wherein the article of manufacture comprises the compound in a multiple dose form. [0261] In another embodiment, the present invention provides a method of inhibiting DPP-IN comprising contacting DPP-IN with a compound according to the present invention. [0262] In another embodiment, the present invention provides a method of inhibiting DPP-IN comprising causing a compound according to the present invention to be present in a subject in order to inhibit DPP-IN in vivo.
[0263] In another embodiment, the present invention provides a method of inhibiting DPP-IN comprising: administering a first compound to a subject that is converted in vivo to a second compound wherein the second compound inhibits DPP-IN in vivo, the second compound being a compound of the present invention.
[0264] Jn another embodiment, the present invention provides therapeutic method comprising: administering a compound according to the present invention to a subject. [0265] In another embodiment, the present invention provides a method of treating a disease state for which DPP-IN possesses activity that contributes to the pathology and/or symptomology of the disease state, the method comprising causing a compound of the present invention to be present in a subject in a therapeutically effective amount for the disease state.
[0266] In another embodiment, the present invention provides a method of treating cancer in a patient in need thereof, comprising administering to said patient a therapeutically effective amount of a compound according to the present invention.
[0267] In another embodiment, the present invention provides a method of treating a disease where the disease is type I or type U diabetes.
[0268] In another embodiment, the present invention provides a method of treating autoimmune disorders such as, but not limited to, rheumatoid arthritis, psoriasis, and multiple sclerosis in a patient in need thereof, comprising administering to said patient a therapeutically effective amount of a compound according to the present invention.
[0269] In yet another embodiment, the present invention provides a method of treating cancer where the cancer treated is colorectal, prostate, breast, thyroid, skin, lung, or head and neck.
[0270] I another embodiment, the present invention provides a method of treating a condition characterized by inadequate lymphocyte or hemapoietic cell activation or concentration in a patient in need thereof, comprising administering to said patient a therapeutically effective amount of a compound according to the present invention.
[0271] In another embodiment, the present invention provides a method of treating HTV infection in a patient in need thereof, comprising admimstering to said patient a therapeutically effective amount of a compound according to the present invention.
[0272] In yet another embodiment, the present invention provides a method of treating a condition characterized by inadequate lymphocyte or hemapoietic cell activation or concentration in a patient in need thereof, wherein the condition is a side effect of chemotherapy or radiation therapy.
[0273] In yet another embodiment, the present invention provides a method of treating a condition characterized by inadequate lymphocyte or hemapoietic cell activation or concentration in a patient in need thereof, wherein the condition is a result of kidney failure. [0274] Jn yet another embodiment, the present invention provides a method of treating a condition characterized by inadequate lymphocyte or hemapoietic cell activation or concentration in a patient in need thereof, wherein the condition is a result of a bone marrow disorder.
[0275] In another embodiment, the present invention provides a method of treating a condition characterized by immunodeficiency symptoms in a patient in need thereof, comprising adrninistering to said patient a therapeutically effective amount of a compound according to the present invention.
[0276] It is noted in regard to all of the embodiments, and any further embodiments, variations, or individual compounds described or claimed herein that all such embodiments, variations, and/or individual compounds are intended to encompass all pharmaceutical acceptable salt forms whether in the form of a single stereoisomer or mixture of stereoisomers unless it is specifically specified otherwise. Similarly, when one or more potentially chiral centers are present in any of the embodiments, variations, and/or individual compounds specified or claimed herein, both possible chiral centers are intended to be encompassed unless it is specifically specified otherwise.
A. Salts, Hydrates, and Prodrugs of DPP-IV Inhibitors
[0277] It should be recognized that the compounds of the present invention may be present and optionally administered in the form of salts, hydrates and prodrugs that are converted in vivo into the compounds of the present invention. For example, it is within the scope of the present invention to convert the compounds of the present invention into and use them in the form of their pharmaceutically acceptable salts derived from various organic and inorganic acids and bases in accordance with procedures well known in the art. [0278] When the compounds of the present invention possess a free base form, the compounds can be prepared as a pharmaceutically acceptable acid addition salt by reacting the free base form of the compound with a pharmaceutically acceptable inorganic or organic acid, e.g. , hydrohalides such as hydrochloride, hydrobromide, hydroiodide; other mineral acids and their conesponding salts such as sulfate, nitrate, phosphate, etc.; and alkyl and monoarylsulfonates such as ethanesulfonate, toluenesulfonate andbenzenesulfonate; and other organic acids and their corresponding salts such as acetate, tartrate, maleate, succinate, citrate, benzoate, salicylate and ascorbate. Further acid addition salts of the present invention include, but are not limited to: adipate, alginate, arginate, aspartate, bisulfate, bisulfite, bromide, butyrate, camphorate, camphorsulfonate, caprylate, chloride, chlorobenzoate, cyclopentanepropionate, digluconate, dihydrogenphosphate, dinitrobenzoate, dodecylsulfate, fumarate, galacterate (frommucic acid), galacturonate, glucoheptaoate, gluconate, glutamate, glycerophosphate, hemisuccinate, hemisulfate, heptanoate, hexanoate, hippurate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethanesulfonate, iodide, isethionate, iso- butyrate, lactate, lactobionate, malate, malonate, mandelate, metaphosphate, methanesulfonate, methylbenzoate, monohydrogenphosphate, 2-naphthalenesulfonate, nicotinate, nitrate, oxalate, oleate, pamoate, pectinate, persulfate, phenylacetate, 3- phenylpropionate, phosphate, phosphonate and phthalate. It should be recognized that the free base forms will typically differ from their respective salt forms somewhat in physical properties such as solubility in polar solvents, but otherwise the salts are equivalent to their respective free base forms for the purposes of the present invention. [0279] When the compounds of the present invention possess a free acid form, a pharmaceutically acceptable base addition salt can be prepared by reacting the free acid form of the compound with a pharmaceutically acceptable inorganic or organic base. Examples of such bases are alkali metal hydroxides including potassium, sodium and lithium hydroxides; alkaline earth metal hydroxides such as barium and calcium hydroxides; alkali metal alkoxides, e.g. potassium ethanolate and sodium propanolate; and various organic bases such as ammonium hydroxide, piperidine, diethanolamine and N-methylglutamine. Also included are the aluminum salts of the compounds of the present invention. Further base salts of the present invention include, but are not limited to: copper, ferric, ferrous, lithium, magnesium, manganic, manganous, potassium, sodium and zinc salts. Organic base salts include, but are not l mited to, salts of primary, secondary and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, e.g., arginine, betaine, caffeine, chloroprocaine, choline, N,N'-dibenzylethylenediamine (benzathine), chcyclohexylamine, diemylamine, 2-diethylaminoethanol, 2- dimethylaminoethanol, ethanolamine, ethyleneάiamine, N-emylmo holine, N- ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, iso-propylamine, lidocaine, lysine, meglumine, N-methyl-D-glucamine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethanolamine, triemylamine, trimethylamine, tripropylamine and tris-(hydroxymemyl)-memylamine (tromethamine). It should be recognized that the free acid forms will typically differ from their respective salt forms somewhat in physical properties such as solubility in polar solvents, but otherwise the salts are equivalent to their respective free acid forms for the purposes of the present invention. [0280] Compounds of the present invention that comprise basic nitrogen-containing groups may be quaternized with such agents as (Ci^alkyl halides, e.g., methyl, ethyl, isopropyl and tert-butyl chlorides, bromides and iodides; di
Figure imgf000086_0001
sulfates, e.g., dimethyl, diethyl and diamyl sulfates; (Cιo-i8)alkyl halides, e.g., decyl, dodecyl, lauryl, myristyl and stearyl chlorides, bromides and iodides; and aryl (Ci^alkyl halides, e.g., benzyl chloride and phenethyl bromide. Such salts permit the preparation of both water-soluble and oil-soluble compounds of the present invention.
[0281] N-oxides of compounds according to the present invention can be prepared by methods known to those of ordinary skill in the art. For example, N-oxides can be prepared by treating an unoxidized form of the compound with an oxidizing agent (e.g., trifluoroperacetic acid, permaleic acid, perbenzoic acid, peracetic acid, metα-chloroperoxybenzoic acid, or the like) in a suitable inert organic solvent (e.g., a halogenated hydrocarbon such as dichloromethane) at approximately 0 °C. Alternatively, the N-oxides of the compounds can be prepared from the N-oxide of an appropriate starting material.
[0282] Prodrug derivatives of compounds according to the present invention can be prepared by modifying substituents of compounds of the present invention that are then converted in vivo to a different substituent. It is noted that in many instances, the prodrugs themselves also fall within the scope of the range of compounds according to the present invention. For example, prodrugs can be prepared by reacting a compound with a carbamylating agent (e.g., l -acyloxyalkylcarbonochloridate.pαrα-nitrophenyl carbonate, or the like) or an acylating agent. Further examples of methods of making prodrugs are described in Saulnier et al.(1994), Bioorganic and Medicinal Chemistry Letters, Vol. 4, p. 1985.
[0283] Protected derivatives of compounds of the present invention can also be made. Examples of techniques applicable to the creation of protecting groups and their removal can be found in T.W. Greene, Protecting Groups in Organic Synthesis, 3rd edition, John Wiley & Sons, Inc. 1999.
[0284] Compounds of the present invention may also be conveniently prepared, or formed during the process of the invention, as solvates (e.g. hydrates). Hydrates of compounds of the present invention may be conveniently prepared by reciystallization from an aqueous/organic solvent mixture, using organic solvents such as dioxin, tetrahydrofuran or methanol.
[0285] A "pharmaceutically acceptable salt", as used herein, is intended to encompass any compound according to the present invention that is utilized in the form of a salt thereof, especially where the salt confers on the compound improved pharmacokinetic properties as compared to the free form of compound or a different salt form of the compound. The pharmaceutically acceptable salt form may also initially confer desirable pharmacokinetic properties on the compound that it did not previously possess, and may even positively affect the pharmacodynamics of the compound with respect to its therapeutic activity in the body. An example of a pharmacokinetic property that may be favorably affected is the manner in which the compound is transported across cell membranes, which in turn may directly and positively affect the absorption, distribution, biotransformation and excretion of the compound. While the route of adrmmstration of the pharmaceutical composition is important, and various anatomical, physiological and pathological factors can critically affect bioavailability, the solubility of the compound is usually dependent upon the character of the particular salt form thereof, which it utilized. One of skill in the art will appreciate that an aqueous solution of the compound will provide the most rapid absorption of the compound into the body of a subject being treated, while lipid solutions and suspensions, as well as solid dosage forms, will result in less rapid adsorption of the compound.
3. INDICATIONS FOR USE OF DPP-IV INHIBITORS
[0286] DPP-TV is believed to contribute to the pathology and/or symptomology of several different diseases such that reduction of the activity of DPP-TV in a subject through inhibition maybe used to therapeutically address these disease states. Examples of various diseases that may be treated using the DPP-TV inhibitors of the present invention are described herein. It is noted that additional diseases beyond those disclosed herein may be later identified as the biological roles that DPP-TV plays in various pathways becomes more fully understood. [0287] One set of indications that DPP-TV inhibitors of the present invention maybe used to treat are those involving the prevention and treatment of diabetes and obesity, in particular type 2 diabetes mellitus, diabetic dislipidemia, conditions of impaired glucose tolerance (IGT), conditions of impaired fasting plasma glucose (IFG), metabolic acidosis, ketosis, appetite regulation and obesity.
[0288] DPP-IN inhibitors of the present invention may also be used as immunosuppressants (or cytokine release suppressant drugs) for the treatment of among other things: organ transplant rejection; autoimmune diseases such as inflammatory bowel disease, multiple sclerosis and rheumatoid arthritis; and the treatment of AIDS. [0289] DPP-TV inhibitors of the present invention may also be used for treating various cancers including breast cancer, lung cancer and prostate cancer.
[0290] DPP-IN inhibitors of the present invention may also be used to treat dermatological diseases such as psoriasis, rheumatoid arthritis (RA) and lichen planus. [0291] DPP-IN inhibitors of the present invention may also be used to treat infertility and amenorrhea.
[0292] DPP-IN inhibitors of the present invention may also be used to modulate cleavage of various cytokines (stimulating hematopoietic cells), growth factors and neuropeptides. For example, such conditions occur frequently in patients who are immunosuppressed, for example, as a consequence of chemotherapy and/or radiation therapy for cancer. [0293] DPP-IN inhibitors of the present invention may also be used prevent or reduce cleavage of Ν-terminal Tyr- Ala from growth hormone-releasing factor. Accordingly, these inhibitors may be used in the treatment of short stature due to growth hormone deficiency (Dwarfism) and for promoting GH-dependent tissue growth or re-growth. [0294] DPP-TV inhibitors of the present invention may also be used to address disease states associated with cleavage of neuropeptides and thus may be useful for the regulation or normalization of neurological disorders.
[0295] For oncology indications, DPP-TV inhibitors of the present invention may be used in conjunction with other agents to inhibit undesirable and uncontrolled cell proliferation. Examples of other anti-cell proliferation agents that may be used in conjunction with the DPP- IN inhibitors of the present invention include, but are not limited to, retinoid acid and derivatives thereof, 2-methoxyestradiol, AΝGIOSTATTΝ™ protein, EΝDOSTATIΝ™ protein, suramin, squalamine, tissue inhibitor of metalloproteinase-I, tissue inhibitor of metalloprote ase-2, plasminogen activator inhibitor-1, plasminogen activator inhibitor-2, cartilage-derived inhibitor, paclitaxel, platelet factor 4, protamine sulfate (clupeine), sulfated chitin derivatives (prepared from queen crab shells), sulfated polysaccharide peptidoglycan complex (sp-pg), staurosporine, modulators of matrix metabolism, including for example, proline analogs ((l-azetidine-2-carboxylic acid (LACA)), cishydroxyproline, d,l-3,4- dehydroproline, thiaproline, beta.-aminopropionitrile fumarate, 4-proρyl-5-(4-pyridinyl)- 2(3H)-oxazolone, methotrexate, mitoxantrone, heparin, interferons, 2 macroglobulin-serum, chimp-3, chymostatin, beta. -cyclodextrin tetradecasulfate, eponemycin; fumagillin, gold sodium thiomalate, d-penicillamine (CDPT), beta.-1-anticollagenase-serum, alpha.2- antiplasmin, bisantrene, lobenzarit disodium, n-2-carboxyphenyl-4-c oroanthromlic acid disodium or "CCA", thalidomide; angostatic steroid, carboxyaminoimidazole; metalloproteinase inhibitors such as BB94. Other anti-angiogenesis agents that may be used include antibodies, preferably monoclonal antibodies against these angiogenic growth factors: bFGF, aFGF, FGF-5, VEGF isoforms, NEGF-C, HGF/SF and Ang- l/Ang-2. Ferrara Ν. and Alitalo, K. "Clinical application of angiogenic growth factors and their inhibitors" (1999) Nature Medicine 5:1359-1364.
4. COMPOSITIONS COMPRISING DPP-IV INHIBITORS
[0296] A wide variety of compositions and administration methods may be used in conjunction with the DPP-JN inhibitors of the present invention. Such compositions may include, in addition to the DPP-IN inhibitors of the present invention, conventional pharmaceutical excipients, and other conventional, pharmaceutically inactive agents. Additionally, the compositions may include active agents in addition to the DPP-IN inhibitors of the present invention. These additional active agents may include additional compounds according to the invention, and/or one or more other pharmaceutically active agents. [0297] The compositions may be in gaseous, liquid, semi-liquid or solid form, formulated in a manner suitable for the route of administration to be used. For oral aciministration, capsules and tablets are typically used. For parenteral adirώiistration, reconstitution of a lyophilized powder, prepared as described herein, is typically used. [0298] Compositions comprising DPP-IN inhibitors of the present invention may be administered or coadministered orally, parenterally, intraperitoneally, intravenously, intraarterially, transdermally, sublingually, intramuscularly, rectally, transbuccally, intranasally, liposomally, via inhalation, vaginally, intraoccularly, via local delivery (for example by catheter or stent), subcutaneously, intraadiposally, intraarticularly, or intrathecally. The compounds and/or compositions according to the invention may also be administered or coadministered in slow release dosage forms.
[0299] The DPP-JN inhibitors and compositions comprising them may be administered or coadministered in any conventional dosage form. Co-administration in the context of this invention is intended to mean the administration of more than one therapeutic agent, one of which includes a DPP-IN inhibitor, in the course of a coordinated treatment to achieve an improved clinical outcome. Such co-administration may also be coextensive, that is, occurring during overlapping periods of time.
[0300] Solutions or suspensions used for parenteral, intradermal, subcutaneous, or topical application may optionally include one or more of the following components: a sterile diluent, such as water for injection, saline solution, fixed oil, polyethylene glycol, glycerine, propylene glycol or other synthetic solvent; antimicrobial agents, such as benzyl alcohol and methyl parabens; antioxidants, such as ascorbic acid and sodium bisulfite; chelating agents, such as ethylenediaminetetraacetic acid (EDTA); buffers, such as acetates, citrates and phosphates; agents for the adjustment of tonicity such as sodium chloride or dextrose, and agents for adjusting the acidity or alkalinity of the composition, such as alkaline or acidifying agents or buffers like carbonates, bicarbonates, phosphates, hydrochloric acid, and organic acids like acetic and citric acid. Parenteral preparations may optionally be enclosed in ampules, disposable syringes or single or multiple dose vials made of glass, plastic or other suitable material.
[0301] When DPP-TV inhibitors according to the present invention exhibit insufficient solubility, methods for solubilizing the compounds may be used. Such methods are known to those of skill in this art, and include, but are not limited to, using cosolvents, such as dimethylsulfoxide (DMSO), using surfactants, such as TWEEΝ, or dissolution in aqueous sodium bicarbonate. Derivatives of the compounds, such as prodrugs of the compounds may also be used in formulating effective pharmaceutical compositions. [0302] Upon mixing or adding DPP-IN inhibitors according to the present invention to a composition, a solution, suspension, emulsion or the like may be formed. The fonn of the resulting composition will depend upon a number of factors, including the intended mode of administration, and the solubility of the compound in the selected carrier or vehicle. The effective concentration needed to ameliorate the disease being treated may be empirically determined.
[0303] Compositions according to the present invention are optionally provided for administration to humans and animals in unit dosage forms, such as tablets, capsules, pills, powders, dry powders for inhalers, granules, sterile parenteral solutions or suspensions, and oral solutions or suspensions, and oil-water emulsions containing suitable quantities of the compounds, particularly the pharmaceutically acceptable salts, preferably the sodium salts, thereof. The pharmaceutically therapeutically active compounds and derivatives thereof are typically formulated and administered in unit-dosage forms or multiple-dosage forms. Unit- dose forms, as used herein, refers to physically discrete units suitable for human and aiiimal subjects and packaged individually as is known in the art. Each unit-dose contains a predetermined quantity of the therapeutically active compound sufficient to produce the desired therapeutic effect, in association with the required pharmaceutical carrier, vehicle or diluent. Examples of unit-dose forms include ampoules and syringes individually packaged tablet or capsule. Unit-dose forms may be administered in fractions or multiples thereof. A multiple-dose form is a plurality of identical unit-dosage forms packaged in a single container to be administered in segregated unit-dose form. Examples of multiple-dose forms include vials, bottles of tablets or capsules or bottles of pint or gallons. Hence, multiple dose form is a multiple of unit-doses that are not segregated in packaging.
[0304] In addition to one or more DPP-TV inhibitors according to the present invention, the composition may comprise: a diluent such as lactose, sucrose, dicalcium phosphate, or carboxymethylcellulose; a lubricant, such as magnesium stearate, calcium stearate and talc; and a binder such as starch, natural gums, such as gum acaciagelatin, glucose, molasses, polvinylpyrrolidine, celluloses and derivatives thereof, povidone, crospovidones and other such binders known to those of skill in the art. Liquid pharmaceutically administrable compositions can, for example, be prepared by dissolving, dispersing, or otherwise mixing an active compound as defined above and optional pharmaceutical adjuvants in a carrier, such as, for example, water, saline, aqueous dextrose, glycerol, glycols, ethanol, and the like, to form a solution or suspension. If desired, the pharmaceutical composition to be administered may also contain minor amounts of auxiliary substances such as wetting agents, emulsifying agents, or solubilizing agents, pH buffering agents and the like, for example, acetate, sodium citrate, cyclodextrine derivatives, sorbitan monolaurate, triethanolamine sodium acetate, triethanolamine oleate, and other such agents. Actual methods of preparing such dosage forms are known in the art, or will be apparent, to those skilled in this art; for example, see Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa., 15th Edition, 1975. The composition or formulation to be administered will, in any event, contain a sufficient quantity of a DPP-JN inhibitor of the present invention to reduce DPP-JN activity in vivo, thereby treating the disease state of the subject.
[0305] Dosage forms or compositions may optionally comprise one or more DPP-TV inhibitors according to the present invention in the range of 0.005% to 100% (weight/weight) with the balance comprising additional substances such as those described herein. For oral administration, a pharmaceutically acceptable composition may optionally comprise any one or more commonly employed excipients, such as, for example pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, talcum, cellulose derivatives, sodium crosscarmellose, glucose, sucrose, magnesium carbonate, sodium saccharin, talcum. Such compositions include solutions, suspensions, tablets, capsules, powders, dry powders for inhalers and sustained release formulations, such as, but not limited to, implants and microencapsulated delivery systems, and biodegradable, biocompatible polymers, such as collagen, ethylene vinyl acetate, polyanhydrides, polyglycolic acid, polyorthoesters, polylactic acid and others. Methods for preparing these formulations are known to those skilled in the art. The compositions may optionally contain 0.01%- 100% (weight/weight) of one or more DPP-IN inhibitors, optionally 0.1-95%, and optionally 1-95%.
[0306] Salts, preferably sodium salts, of the DPP-IN inhibitors may be prepared with carriers that protect the compound against rapid elimination from the body, such as time release formulations or coatings. The formulations may further include other active compounds to obtain desired combinations of properties.
A. Formulations for oral administration
[0307] Oral pharmaceutical dosage fonns may be as a solid, gel or liquid. Examples of solid dosage forms include, but are not limited to tablets, capsules, granules, and bulk powders. More specific examples of oral tablets include compressed, chewable lozenges and tablets that may be enteric-coated, sugar-coated or film-coated. Examples of capsules include hard or soft gelatin capsules. Granules and powders may be provided in non-effervescent or effervescent forms. Each may be combined with other ingredients known to those skilled in the art.
[0308] In certain embodiments, DPP-IN inhibitors according to the present invention are provided as solid dosage forms, preferably capsules or tablets. The tablets, pills, capsules, troches and the like may optionally contain one or more of the following ingredients, or compounds of a similar nature: a binder; a diluent; a disintegrating agent; a lubricant; a glidant; a sweetening agent; and a flavoring agent.
[0309] Examples of binders that may be used include, but are not limited to, microcrystalline cellulose, gum tragacanth, glucose solution, acacia mucilage, gelatin solution, sucrose and starch paste.
[0310] Examples of lubricants that may be used include, but are not limited to, talc, starch, magnesium or calcium stearate, lycopodium and stearic acid.
[0311] Examples of diluents that may be used include, but are not limited to, lactose, sucrose, starch, kaolin, salt, mannitol and dicalcium phosphate.
[0312] Examples of glidants that may be used include, but are not limited to, colloidal silicon dioxide.
[0313] Examples of disintegrating agents that may be used include, but are not limited to, crosscarmellose sodium, sodium starch glycolate, alginic acid, corn starch, potato starch, bentonite, methylcellulose, agar and carboxymethylcellulose.
[0314] Examples of coloring agents that may be used include, but are not limited to, any of the approved certified water soluble FD and C dyes, mixtures thereof; and water insoluble
FD and C dyes suspended on alumina hydrate.
[0315] Examples of sweetening agents that may be used include, but are not limited to, sucrose, lactose, mannitol and artificial sweetening agents such as sodium cyclamate and saccharin, and any number of spray-dried flavors.
[0316] Examples of flavoring agents that may be used include, but are not limited to, natural flavors extracted from plants such as fruits and synthetic blends of compounds that produce a pleasant sensation, such as, but not limited to peppermint and methyl salicylate. [0317] Examples of wetting agents that may be used include, but are not limited to, propylene glycol monostearate, sorbitan monooleate, diethylene glycol monolaurate and polyoxyethylene lauryl ether.
[0318] Examples of anti-emetic coatings that may be used include, but are not limited to, fatty acids, fats, waxes, shellac, ammoniated shellac and cellulose acetate phthalates.
[0319] Examples of film coatings that may be used include, but are not limited to, hydroxyethylcellulose, sodium carboxymethylcellulose, polyethylene glycol 4000 and cellulose acetate phthalate.
[0320] If oral administration is desired, the salt of the compound may optionally be provided in a composition that protects it from the acidic environment of the stomach. For example, the composition can be formulated in an enteric coating that maintains its integrity in the stomach and releases the active compound in the intestine. The composition may also be formulated in combination with an antacid or other such ingredient.
[0321] When the dosage unit form is a capsule, it may optionally additionally comprise a liquid canier such as a fatty oil. In addition, dosage unit forms may optionally additionally comprise various other materials that modify the physical form of the dosage unit, for example, coatings of sugar and other enteric agents.
[0322] Compounds according to the present invention may also be administered as a component of an elixir, suspension, syrup, wafer, sprinkle, chewing gum or the like. A syrup may optionally comprise, in addition to the active compounds, sucrose as a sweetening agent and certain preservatives, dyes and colorings and flavors.
[0323] The DPP-IN inhibitors of the present invention may also be mixed with other active materials that do not impair the desired action, or with materials that supplement the desired action, such as antacids, H2 blockers, and diuretics. For example, if a compound is used for treating asthma or hypertension, it may be used with other bronchodilators and antihypertensive agents, respectively.
[0324] Examples of pharmaceutically acceptable carriers that may be included in tablets comprising DPP-IN inhibitors of the present invention include, but are not limited to binders, lubricants, diluents, disintegrating agents, coloring agents, flavoring agents, and wetting agents . Enteric-coated tablets, because of the enteric-coating, resist the action of stomach acid and dissolve or disintegrate in the neutral or alkaline intestines. Sugar-coated tablets may be compressed tablets to which different layers of pharmaceutically acceptable substances are applied. Film-coated tablets may be compressed tablets that have been coated with polymers or other suitable coating. Multiple compressed tablets may be compressed tablets made by more than one compression cycle utilizing the pharmaceutically acceptable substances previously mentioned. Coloring agents may also be used in tablets.. Flavoring and sweetening agents may be used in tablets, and are especially useful in the formation of chewable tablets and lozenges.
[0325] Examples of liquid oral dosage forms that may be used include, but are not limited to, aqueous solutions, emulsions, suspensions, solutions and/or suspensions reconstituted from non-effervescent granules and effervescent preparations reconstituted from effervescent granules.
[0326] Examples of aqueous solutions that may be used include, but are not limited to, elixirs and syrups. As used herein, elixirs refer to clear, sweetened, hydroalcoholic preparations. Examples of pharmaceutically acceptable carriers that may be used in elixirs include, but are not limited to solvents. Particular examples of solvents that may be used include glycerin, sorbitol, ethyl alcohol and syrup. As used herein, syrups refer to concentrated aqueous solutions of a sugar, for example, sucrose. Syrups may optionally further comprise a preservative.
[0327] Emulsions refer to two-phase systems in which one liquid is dispersed in the form of small globules throughout another liquid. Emulsions may optionally be oil-in-water or water-in-oil emulsions . Examples of pharmaceutically acceptable carriers that may be used in emulsions include, but are not limited to non-aqueous liquids, emulsifying agents and preservatives.
[0328] Examples of pharmaceutically acceptable substances that may be used in non- effervescent granules, to be reconstituted into a liquid oral dosage form, include diluents, sweeteners and wetting agents.
[0329] Examples of pharmaceutically acceptable substances that may be used in effervescent granules, to be reconstituted into a liquid oral dosage form, include organic adds and a source of carbon dioxide.
[0330] Coloring and flavoring agents may optionally be used in all of the above dosage forms. [0331] Particular examples of preservatives that may be used include glycerin, methyl and propylparaben, benzoic add, sodium benzoate and alcohol.
[0332] Particular examples of non-aqueous liquids that may be used in emulsions include mineral oil and cottonseed oil.
[0333] Particular examples of emulsifying agents that may be used include gelatin, acacia, tragacanth, bentonite, and surfactants such as polyoxyethylene sorbitan monooleate.
[0334] Particular examples of suspending agents that may be used include sodium carboxymethylcellulose, pectin, tragacanth, Neegum and acacia. Diluents include lactose and sucrose. Sweetening agents include sucrose, syrups, glycerin and artificial sweetening agents such as sodium cyclamate and saccharin.
[0335] Particular examples of wetting agents that may be used include propylene glycol monostearate, sorbitan monooleate, diethylene glycol monolaurate and polyoxyethylene lauryl ether.
[0336] Particular examples of organic acids that may be used include citric and tartaric acid.
[0337] S ources of carbon dioxide that may be used in effervescent compositions include sodium bicarbonate and sodium carbonate. Coloring agents include any of the approved certified water soluble FD and C dyes, and mixtures thereof.
[0338] Particular examples of flavoring agents that may be used include natural flavors extracted from plants such fruits, and synthetic blends of compounds that produce a pleasant taste sensation.
[0339] For a solid dosage form, the solution or suspension, in for example propylene carbonate, vegetable oils or triglycerides, is preferably encapsulated in a gelatin capsule. Such solutions, and the preparation and encapsulation thereof, are disclosed in U.S. Pat. Νos.
4,328,245; 4,409,239; and 4,410,545. For a liquid dosage form, the solution, e.g. , for example, in a polyethylene glycol, may be diluted with a sufficient quantity of a pharmaceutically acceptable liquid carrier, e.g. water, to be easily measured for administration.
[0340] Alternatively, liquid or semi-solid oral formulations may be prepared by dissolving or dispersing the active compound or salt in vegetable oils, glycols, triglycerides, propylene glycol esters (e.g. propylene carbonate) and other such carriers, and encapsulating these solutions or suspensions in hard or soft gelatin capsule shells. Other useful formulations include those set forth in U.S. Pat. Nos. Re 28,819 and 4,358,603.
B. Injectables, solutions and emulsions [0341] The present invention is also directed to compositions designed to administer the DPP-TV inhibitors of the present invention by parenteral administration, generally characterized by injection, either subcutaneously, intramuscularly or intravenously. Injectables may be prepared in any conventional form, for example as liquid solutions or suspensions, solid forms suitable for solution or suspension in liquid prior to injection, or as emulsions.
[0342] Examples of excipients that may be used in conjunction with injectables according to the present invention include, but are not limited to water, saline, dextrose, glycerol or ethanol. The injectable compositions may also optionally comprise minor amounts of non- toxic auxiliary substances such as wetting or emulsifying agents, pH buffering agents, stabilizers, solubility enhancers, and other such agents, such as for example, sodium acetate, sorbitan monolaurate, triethanolamine oleate and cyclodextrins. Implantation of a slow- release or sustained-release system, such that a constant level of dosage is maintained (see, e.g., U.S. Pat. No. 3,710,795) is also contemplated herein. The percentage of active compound contained in such parenteral compositions is highly dependent on the specific nature thereof, as well as the activity of the compound and the needs of the subject. [0343] Parenteral adininistration of the formulations includes intravenous, subcutaneous and intramuscular administrations. Preparations for parenteral administration include sterile solutions ready for injection, sterile dry soluble products, such as the lyophilized powders described herein, ready to be combined with a solvent just prior to use, including hypodermic tablets, sterile suspensions ready for injection, sterile dry insoluble products ready to be combined with a vehicle just prior to use and sterile emulsions. The solutions may be either aqueous or nonaqueous.
[0344] When administered intravenously, examples of suitable carriers include, but are not limited to physiological saline or phosphate buffered saline (PBS), and solutions containing thickening and solubilizing agents, such as glucose, polyethylene glycol, and polypropylene glycol and mixtures thereof. [0345] Examples of pharmaceutically acceptable earners that may optionally be used in parenteral preparations include, but are not limited to aqueous vehicles, nonaqueous vehicles, antimicrobial agents, isotonic agents, buffers, antioxidants, local anesthetics, suspending and dispersing agents, emulsifying agents, sequestering or chelating agents and other pharmaceutically acceptable substances.
[0346] Examples of aqueous vehicles that may optionally be used include Sodium
Chloride Injection, Ringers Injection, Isotonic Dextrose Injection, Sterile Water Injection,
Dextrose and Lactated Ringers Injection.
[0347] Examples of nonaqueous parenteral vehicles that may optionally be used include fixed oils of vegetable origin, cottonseed oil, corn oil, sesame oil and peanut oil.
[0348] Antimicrobial agents in bacteriostatic or fungistatic concentrations may be added to parenteral preparations, particularly when the preparations are packaged in multiple-dose containers and thus designed to be stored and multiple aliquots to be removed. Examples of antimicrobial agents that may used include phenols or cresols, mercurials, benzyl alcohol, chlorobutanol, methyl and propyl p-hydroxybenzoic acid esters, thimerosal, benzalkonium chloride and benzethonium chloride.
[0349] Examples of isotonic agents that may be used include sodium chloride and dextrose. Examples of buffers that may be used include phosphate and citrate. Examples of antioxidants that may be used include sodium bisulfate. Examples of local anesthetics that may be used include procaine hydrochloride. Examples of suspending and dispersing agents that may be used include sodium carboxymethylcellulose, hydroxypropyl methylcellulose and polyvinylpynolidone. Examples of emulsifying agents that may be used include Polysorbate
80 (TWEEN 80). A sequestering or chelating agent of metal ions include EDTA.
[0350] Pharmaceutical carriers may also optionally include ethyl alcohol, polyethylene glycol and propylene glycol for water miscible vehicles and sodium hydroxide, hydrochloric acid, citric acid or lactic acid for pH adjustment.
[0351] The concentration of a DPP-IN inhibitor in the parenteral formulation may be adjusted so that an injection administers a pharmaceutically effective amount sufficient to produce the desired pharmacological effect. The exact concentration of a DPP-IN inhibitor and/or dosage to be used will ultimately depend on the age, weight and condition of the patient or animal as is known in the art. [0352] Unit-dose parenteral preparations may be packaged in an ampoule, a vial or a syringe with a needle. All preparations for parenteral administration should be sterile, as is know and practiced in the art.
[0353] Injectables may be designed for local and systemic administration. Typically a therapeutically effective dosage is formulated to contain a concentration of at least about 0.1 % w/w up to about 90% w/w or more, preferably more than 1% w/w of the DPP-TV inhibitor to the treated tissue(s). The DPP-TV inhibitor may be administered at once, or may be divided into a number of smaller doses to be administered at intervals of time. It is understood that the precise dosage and duration of treatment will be a function of the location of where the composition is parenterally administered, the carrier and other variables that may be determined empirically using known testing protocols or by extrapolation from in vivo or in vitro test data. It is to be noted that concentrations and dosage values may also vary with the age of the individual treated. It is to be further understood that for any particular subject, specific dosage regimens may need to be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the formulations. Hence, the concentration ranges set forth herein are intended to be exemplary and are not intended to limit the scope or practice of the claimed formulations. [0354] The DPP-TV inhibitor may optionally be suspended in micronized or other suitable form or may be derivatized to produce a more soluble active product or to produce a prodrug. The form of the resulting mixture depends upon a number of factors, including the intended mode of administration and the solubility of the compound in the selected carrier or vehicle. The effective concentration is sufficient for ameliorating the symptoms of the disease state and may be empirically determined.
C. Lyophilized powders
[0355] The DPP-IN inhibitors of the present invention may also be prepared as lyophilized powders, which can be reconstituted for administration as solutions, emulsions and other mixtures. The lyophilized powders may also be formulated as solids or gels. [0356] Sterile, lyophilized powder may be prepared by dissolving the compound in a sodium phosphate buffer solution containing dextrose or other suitable excipient. Subsequent sterile filtration of the solution followed by lyophilization under standard conditions known to those of skill in the art provides the desired formulation. Briefly, the lyophilized powder may optionally be prepared by dissolving dextrose, sorbitol, fructose, corn syrup, xylitol, glycerin, glucose, sucrose or other suitable agent, about 1-20%, preferably about 5 to 15%, in a suitable buffer, such as citrate, sodium or potassium phosphate or other such buffer known to those of skill in the art at, typically, about neutral pH. Then, a DPP-JN inhibitor is added to the resulting mixture, preferably above room temperature, more preferably at about 30-35 °C, and st red until it dissolves. The resulting mixture is diluted by adding more buffer to a desired concentration. The resulting mixture is sterile filtered or treated to remove particulates and to insure sterility, and apportioned into vials for lyophilization. Each vial may contain a single dosage or multiple dosages of the DPP-IN inhibitor.
D. Topical administration
[0357] The DPP-TV inhibitors of the present invention may also be administered as topical mixtures. Topical mixtures may be used for local and systemic administration. The resulting mixture may be a solution, suspension, emulsions or the like and are formulated as creams, gels, ointments, emulsions, solutions, elixirs, lotions, suspensions, tinctures, pastes, foams, aerosols, irrigations, sprays, suppositories, bandages, dermal patches or any other formulations suitable for topical admiriistration.
[0358] The DPP-IN inhibitors may be formulated as aerosols for topical application, such as by inhalation (see, U.S. Pat. Νos. 4,044,126, 4,414,209, and 4,364,923, which describe aerosols for delivery of a steroid useful for treatment inflammatory diseases, particularly asthma). These formulations for administration to the respiratory tract can be in the form of an aerosol or solution for a nebulizer, or as a microfine powder for insufflation, alone or in combination with an inert carrier such as lactose. In such a case, the particles of the formulation will typically have diameters of less than 50 microns, preferably less than 10 microns.
[0359] The DPP-IN inhibitors may also be formulated for local or topical application, such as for topical application to the skin and mucous membranes, such as in the eye, in the form of gels, creams, and lotions and for application to the eye or for intracisternal or intraspinal application. Topical administration is contemplated for transdermal delivery and also for admiiiistration to the eyes or mucosa, or for inhalation therapies. Nasal solutions of the DPP-TV inhibitor alone or in combination with other pharmaceutically acceptable excipients can also be administered. E. Formulations for other routes of administration
[0360] Depending upon the disease state being treated, other routes of administration, such as topical application, transdermal patches, and rectal administration, may also be used. For example, pharmaceutical dosage forms for rectal administration are rectal suppositories, capsules and tablets for systemic effect. Rectal suppositories are used herein mean solid bodies for insertion into the rectum that melt or soften at body temperature releasing one or more pharmacologically or therapeutically active ingredients. Pharmaceutically acceptable substances utilized in rectal suppositories are bases or vehicles and agents to raise the melting point. Examples of bases include cocoa butter (theobroma oil), glycerin-gelatin, carbowax, (polyoxyethylene glycol) and appropriate mixtures of mono-, di- and triglycerides of fatty acids. Combinations of the various bases may be used. Agents to raise the melting point of suppositories include spermaceti and wax. Rectal suppositories may be prepared either by the compressed method or by molding. The typical weight of a rectal suppository is about 2 to 3 gm. Tablets and capsules for rectal administration may be manufactured using the same pharmaceutically acceptable substance and by the same methods as for formulations for oral administration.
F. Examples of Formulations
[0361] The following are particular examples of oral, intravenous and tablet formulations that may optionally be used with compounds of the present invention. It is noted that these formulations may be varied depending on the particular compound being used and the indication for which the formulation is going to be used.
ORAL FORMULATION
Compound of the Present Invention 10-100 mg
Citric Acid Monohydrate 105 mg
Sodium Hydroxide 18 mg
Flavoring
Water q.s. to 100 mL
INTRAVENOUS FORMULATION
Compound of the Present Invention 0.1-10 mg
Dextrose Monohydrate q.s. to make isotonic Citric Acid Monohydrate 1.05 mg
Sodium Hydroxide 0.18 mg
Water for Injection q.s. to 1.0 mL
TABLET FORMULATION
Compound of the Present Invention 1 %
Microcrystalline Cellulose 73%
Stearic Acid 25%
Colloidal Silica 1%.
5. KITS COMPRISING DPP-IV INHIBITORS
[0362] The invention is also directed to kits and other articles of manufacture for treating diseases associated with DPP-JN. It is noted that diseases are intended to cover all conditions for which the DPP-IN possesses activity that contributes to the pathology and/or symptomology of the condition.
[0363] In one embodiment, a kit is provided that comprises a composition comprising at least one DPP-IN inhibitor of the present invention in combination with instructions. The instructions may indicate the disease state for which the composition is to be administered, storage information, dosing information and/or instructions regarding how to administer the composition. The kit may also comprise packaging materials. The packaging material may comprise a container for housing the composition. The kit may also optionally comprise additional components, such as syringes for administration of the composition. The kit may comprise the composition in single or multiple dose forms.
[0364] In another embodiment, an article of manufacture is provided that comprises a composition comprising at least one DPP-JN inhibitor of the present invention in combination with packaging materials. The packaging material may comprise a container for housing the composition. The container may optionally comprise a label indicating the disease state for which the composition is to be administered, storage information, dosing information and/or instructions regarding how to administer the composition. The kit may also optionally comprise additional components, such as syringes for administration of the composition. The kit may comprise the composition in single or multiple dose forms. [0365] It is noted that the packaging material used in kits and articles of manufacture according to the present invention may form a plurality of divided containers such as a divided bottle or a divided foil packet. The container can be in any conventional shape or form as known in the art which is made of a pharmaceutically acceptable material, for example a paper or cardboard box, a glass or plastic bottle or jar, a re-sealable bag (for example, to hold a "refill" of tablets for placement into a different container), or a blister pack with individual doses for pressing out of the pack according to a therapeutic schedule. The container that is employed will depend on the exact dosage form involved, for example a conventional cardboard box would not generally be used to hold a liquid suspension. It is feasible that more than one container can be used together in a single package to market a single dosage form. For example, tablets may be contained in a bottle that is in turn contained within a box. Typically the kit includes directions for the administration of the separate components. Ηie kit form is particularly advantageous when the separate components are preferably administered in different dosage forms (e.g., oral, topical, transdermal and parenteral), are administered at different dosage intervals, or when titration of the individual components of the combination is desired by the prescribing physician.
[0366] One particular example of a kit according to the present invention is a so-called blister pack. Blister packs are well known in the packaging industry and are being widely used for the packaging of pharmaceutical unit dosage forms (tablets, capsules, and the like). Blister packs generally consist of a sheet of relatively stiff material covered with a foil of a preferably transparent plastic material. During the packaging process recesses are formed in the plastic foil. The recesses have the size and shape of individual tablets or capsules to be packed or may have the size and shape to accommodate multiple tablets and/or capsules to be packed. Next, the tablets or capsules are placed in the recesses accordingly and the sheet of relatively stiff material is sealed against the plastic foil at the face of the foil which is opposite from the direction in which the recesses were formed. As a result, the tablets or capsules are individually sealed or collectively sealed, as desired, in the recesses between the plastic foil and the sheet. Preferably the strength of the sheet is such that the tablets or capsules can be removed from the blister pack by manually applying pressure on the recesses whereby an opening is formed in the sheet at the place of the recess. The tablet or capsule can then be removed via said opening. [0367] Another specific embodiment of a kit is a dispenser designed to dispense the daily doses one at a time in the order of their intended use. Preferably, the dispenser is equipped with a memory-aid, so as to further facilitate compliance with the regimen. An example of such a memory-aid is a mechanical counter that indicates the number of daily doses that has been dispensed. Another example of such a memory-aid is a battery-powered micro-chip memory coupled with a liquid crystal readout, or audible reminder signal which, for example, reads out the date that the last daily dose has been taken and/or reminds one when the next dose is to be taken.
EXAMPLES
1. Preparation Of DPP-IV Inhibitors
[0368] Various methods may be developed for synthesizing compounds according to the present invention. Representative methods for synthesizing these compounds are provided in the Examples. It is noted, however, that the compounds of the present invention may also be synthesized by other synthetic routes that others may devise.
[0369] It will be readily recognized that certain compounds according to the present invention have atoms with linkages to other atoms that confer a particular stereochemistry to the compound (e.g. , chiral centers). It is recognized that synthesis of compounds according to the present invention may result in the creation of mixtures of different stereoisomers (enantiomers, diastereomers). Unless a particular stereochemistry is specified, recitation of a compound is intended to encompass all of the different possible stereoisomers. [0370] Various methods for separating mixtures of different stereoisomers are known in the art. For example, a racemic mixture of a compound may be reacted with an optically active resolving agent to form a pair of diastereoisomeric compounds. The diastereomers may then be separated in order to recover the optically pure enantiomers. Dissociable complexes may also be used to resolve enantiomers (e.g., crystalline diastereoisomeric salts). Diastereomers typically have sufficiently distinct physical properties (e.g., melting points, boiling points, solubilities, reactivity, etc.) that they can be readily separated by taking advantage of these dissimilarities. For example, diastereomers can typically be separated by chromatography or by separation/resolution techniques based upon differences in solubility. A more detailed description of techniques that can be used to resolve stereoisomers of compounds from their racemic mixture can be found in Jean Jacques Andre Collet, Samuel H. Wilen, Enantiomers, Racemates and Resolutions, John Wiley & Sons, Inc. (1981).
[0371] Compounds according to the present invention can also be prepared as a pharmaceutically acceptable acid addition salt by reacting the free base form of the compound with a pharmaceutically acceptable inorganic or organic acid. Alternatively, a pharmaceutically acceptable base addition salt of a compound can be prepared by reacting the free acid form of the compound with a pharmaceutically acceptable inorganic or organic base. Inorganic and organic acids and bases suitable for the preparation of the pharmaceutically acceptable salts of compounds are set forth in the definitions section of this Application. Alternatively, the salt forms of the compounds can be prepared using salts of the starting materials or intermediates.
[0372] The free acid or free base forms of the compounds can be prepared from the conesponding base addition salt or acid addition salt form. For example, a compound in an acid addition salt form can be converted to the conesponding free base by treating with a suitable base (e.g., ammonium hydroxide solution, sodium hydroxide, and the like). A compound in a base addition salt form can be converted to the conesponding free acid by treating with a suitable acid (e.g., hydrochloric acid, etc).
[0373] The N-oxides of compounds according to the present invention can be prepared by methods known to those of ordinary skill in the art. For example, N-oxides can be prepared by treating an unoxidized form of the compound with an oxidizing agent (e.g., trifluoroperacetic acid, permaleic acid, perbenzoic acid, peracetic acid, metα-chloroperoxybenzoic acid, or the like) in a suitable inert organic solvent (e.g., a halogenated hydrocarbon such as dichloromethane) at approximately 0 °C. Alternatively, the N-oxides of the compounds can be prepared from the N-oxide of an appropriate starting material.
[0374] Compounds in an unoxidized form can be prepared from N-oxides of compounds by treating with a reducing agent (e.g., sulfur, sulfur dioxide, triphenyl phosphine, lithium borohydride, sodium borohydride, phosphorus trichloride, tribromide, or the like) in an suitable inert organic solvent (e.g., acetonitrile, ethanol, aqueous dioxane, or the like) at 0 to 80 °C. [0375] Prodrug derivatives of the compounds can be prepared by methods known to those of ordinary skill in the art (e.g., for further details see Saulnier et αZ.(1994), Bioorganic and Medicinal Chemistry Letters, Vol. 4, p. 1985). For example, appropriate prodrugs can be prepared by reacting a non-derivatized compound with a suitable carbamylating agent (e.g., l,l-acyloxyalkylcarbonochloridate,/?αrα-nitrophenyl carbonate, or the like). [0376] Protected derivatives of the compounds can be made by methods known to those of ordinary skill in the art. A detailed description of the techniques applicable to the creation of protecting groups and their removal can be found in T.W. Greene, Protecting Groups in Organic Synthesis, 3rd edition, John Wiley & Sons, Inc. 1999.
[0377] Compounds according to the present invention may be conveniently prepared, or formed during the process of the invention, as solvates (e.g. hydrates). Hydrates of compounds of the present invention may be conveniently prepared by recrystallization from an aqueous/organic solvent mixture, using organic solvents such as dioxin, tetrahydrofuran or methanol.
[0378] Compounds according to the present invention can also be prepared as their individual stereoisomers by reacting a racemic mixture of the compound with an optically active resolving agent to form a pair of diastereoisomeric compounds, separating the diastereomers and recovering the optically pure enantiomer. While resolution of enantiomers can be carried out using covalent diastereomeric derivatives of compounds, dissociable complexes are prefened (e.g., crystalline diastereoisomeric salts). Diastereomers have distinct physical properties (e.g. , melting points, boiling points, solubilities, reactivity, etc.) and can be readily separated by taking advantage of these dissimilarities. The diastereomers can be separated by chromatography or, preferably, by separation/resolution techniques based upon differences in solubility. The optically pure enantiomer is then recovered, along with the resolving agent, by any practical means that would not result in racemization. A more detailed description of the techniques applicable to the resolution of stereoisomers of compounds from their racemic mixture can be found in Jean Jacques Andre Collet, Samuel H. Wilen, Enantiomers, Racemates and Resolutions, John Wiley & Sons, Inc. (1981). [0379] As used herein the symbols and conventions used in these processes, schemes and examples are consistent with those used in the contemporary scientific literature, for example, the Journal of the American Chemical Society or the Journal of Biological Chemistry. Standard single-letter or thee-letter abbreviations are generally used to designate amino acid residues, which are assumed to be in the L-configuration unless otherwise noted. Unless otherwise noted, all starting materials were obtained from commercial suppliers and used without further purification. Specifically, the following abbreviations may be used in the examples and throughout the specification: g (grams); mg (milligrams);
L (liters); mL (milliliters); μL (microliters); psi (pounds per square inch);
M (molar); mM (millimolar); i.v. (intravenous); Hz (Hertz);
MHz (megahertz); mol (moles); mmol (millimoles); RT (ambient temperature); min (minutes) ;h (hours); mp (melting point); TLC (thin layer chromatography);
Tr (retention time); RP (reverse phase);
MeOH (methanol); i-PrOH (isopropanol);
TEA (triethylamine); TFA (trifluoroacetic acid);
TFAA (trifluoroacetic anhydride); THF (tetrahydrofuran);
DMSO (dimethylsulfoxide); EtOAc (ethyl acetate);
DME (1,2-dimethoxyethane); DCM (dichloromethane);
DCE (dichloroethane); DMF (N,N-dimethylformamide);
DMPU (N,N'-dimethylpropyleneurea); CDI (1,1-carbonyldiimidazole); IBCF (isobutyl chloroformate); HO Ac (acetic acid);
HOSu (N-hydroxysuccinimino); HOBT (1-hydroxybenzotriazole); Et2O (diethyl ether); EDCI (ethylcarbodnmino hydrochloride);
BOC (tert-butyloxycarbonyl); FMOC (9-fluorenylmethoxycarbonyl);
DCC (dicyclohexylcarbodiimino); CBZ (benzyloxycarbonyl);
Ac (acetyl); atm (atmosphere);
TMSE (2-(trimethylsilyl)ethyl); TMS (trimethylsilyl);
TJPS (triisopropylsilyl); TBS (t-butyldimethylsilyl); DMAP (4-dimethylam opyridine); Me (methyl);
OMe (methoxy); Et (ethyl);
Et (ethyl); tBu (tert-butyl);
HPLC (high pressure liquid chomatography);
BOP (bis(2-oxo-3-oxazolidinyl)phosphinic chloride);
TBAF (tetra-n-butylammonium fluoride); mCPB A (meta-chloroperbenzoic acid. [0380] All references to ether or Et2O are to diethyl ether; brine refers to a saturated aqueous solution of NaCl. Unless otherwise indicated, all temperatures are expressed in °C (degrees Centigrade). All reactions conducted under an inert atmosphere at RT unless otherwise noted.
[0381] 1H NMR spectra were recorded on a Bruker Avance 400. Chemical shifts are expressed in parts per million (ppm). Coupling constants are in units of hertz (Hz). Splitting patterns describe apparent multiplicities and are designated as s (singlet), d (doublet), t (triplet), q (quartet), m (multiplet), br (broad).
[0382] Low-resolution mass spectra (MS) and compound purity data were acquired on a Waters ZQ LC/MS single quadrupole system equipped with electrospray ionization (EST) source, UN detector (220 and 254 nm), and evaporative light scattering detector (ELSD). Thin-layer chromatography was performed on 0.25 mm E. Merck silica gel plates (60F-254), visualized with UN light, 5% ethanolic phosphomolybdic acid, Νinhydrin or p-anisaldehyde solution. Flash column chromatography was performed on silica gel (230-400 mesh, Merck).
2. Synthetic Schemes For DPP-IV Inhibitors Of The Present Invention
[0383] DPP-IN inhibitors according to the present invention may be synthesized according to a variety of reaction schemes. Some illustrative schemes are provided herein in the examples. Other reaction schemes could be readily devised by those skilled in the art. [0384] In the reactions described hereinafter it may be necessary to protect reactive functional groups, for example hydroxy, amino, imino, thio or carboxy groups, where these are desired in the final product, to avoid their unwanted participation in the reactions. Conventional protecting groups may be used in accordance with standard practice, for examples see T.W. Greene and P. G. M. Wuts in "Protective Groups in Organic Chemistry" John Wiley and Sons, 1991.
[0385] Compounds according to the present invention may optionally be synthesized according to the following reaction schemes:
Representative Scheme for the Synthesis of Compounds According to Formula I where Q=CO:
Scheme 1:
Figure imgf000109_0001
Figure imgf000109_0002
[0386] By varying the RiX and R2 groups shown above in this example, a wide variety of different DPP-IN inhibitors according to the present invention may be synthesized.
Representative Scheme for the Synthesis of Compounds According to Formula I where Q=SO2:
Scheme 2:
Figure imgf000109_0003
Mel
Figure imgf000109_0004
[0387] By varying the Ri, R2, R3 and R4 groups shown above in this example, a wide variety of different DPP-TV inhibitors according to the present invention may be synthesized. [0388] In each of the above reaction schemes, the various substituents may be selected from among the various substituents otherwise taught herein.
[0389] Descriptions of the syntheses of particular compounds according to the present invention based on the above reaction schemes are set forth herein.
3. Examples Of DPP-IV Inhibitors
[0390] The present invention is further exemplified, but not limited by, the following examples that describe the synthesis of particular compounds according to the invention. Synthesis of Ex. 1:
Figure imgf000110_0001
1A 1B 1C
Figure imgf000110_0002
Example 1: 2-[2-(3-Amino-piperidin-l-yl)-4-oxo-4H-quinazolin-3-ylmethyl]- benzonitrile
Figure imgf000110_0003
Example IB: 2,4-Dichloro-quinazoline
Figure imgf000111_0001
[0391] To 3.2 g of lH-quinazoline-2,4-dione (IA) in 20 mL POCl3 was added 0.8 mL N,N-dimethylaniline. The mixture was then heated at reflux for 16 hours. Excess POCI3 was removed in vacuo, providing crude product IB.
Example IC: 2-Chloro-3H-quinazolin-4-one
Figure imgf000111_0002
[0392] A mixture of 20 mL of IN NaOH, 20 mL of THF, and 2 g of IB was stiπed at room temperature under N2 for 4 hours. The solution was chilled and adjusted to pH 5 with AcOH. The solids that precipitated were filtered to give 1.62 g (90%) of product IC.
Example ID: 2-(2-Chloro-4-oxo-4H-quinazolin-3-ylmethyl)-benzonitrile
Figure imgf000111_0003
[0393] A mixture of 0.36 g of IC, 0.47 g of 2-cyanobenzylbromide and 0.35 g of K2CO3 in 10 mL of DMF was stined over night. The reaction mixture was diluted with water, extracted with ethyl acetate, and dried over MgSO4. Removal of the solvent gave crude product ID (containing O-alkylated product).
[0394] A mixture of 200 mg of crude ID, 3 eq. of 3-aminopiperidne dihydrochloride, 5 eq. of NaHCO3, and 3 mL of ethanol in a sealed tube was heated to 150 °C for 6 hours. After cooling to room temperature and filtering out the inorganic salts, purification via LC MS afforded 108 mg (45% yield) of product of Example 1. 1H NMR (400 MHz, CDCI3); δ 8.05 (d, J = 7.60 Hz, IH), 7.69-7.79 (m, 2H), 7.56-7.62 (m, 2H), 7.37-7.46 (m, 2H), 7.30 (d, J = 8.0 Hz, IH), 5.52 (AB q, J = 15.2 Hz, 2H), 3.64-3.71 (m, IH), 3.55 (br s, IH), 3.19-3.32 (m, 2H), 2.98-3.08 (m, IH), 2.10-2.18 (m, IH), 1.62-1.94 (m, 3H). MS: (ES) M+H calc'd for C2ιH2ιN50, 360; found 360.
Example 2: 2-[2-(3-Amino-piperidin-l-yl)-6,7-chmethoxy-4-oxo-4H-quinazolin- 3-ylmethyl]-benzonitrile
Figure imgf000112_0001
Example 2A: 2-Chloro-6,7-dimethoxy-3H-quinazolin-4-one
Figure imgf000112_0002
[0395] 2,4-Dichloro-6,7-dimethoxy-quinazoline (1.02 g, 3.95 mmol) was converted to the title compound (664 mg, 70%) by the method used for example IC. !H NMR (400 MHz, DMSO): δ 13.1 (s, IH), 7.41 (s, IH), 7.13 (s, IH), 3.90 (s, 3H), 3.87 (s, 3H). MS: (ES)M+H calc'd for C10H9C1N2O3, 241; found 241.
Example 2B: 2-(2-Chloro-6,7-dimethoxy-4-oxo-4H-quinazolin-3-ylmethyl)- benzonitrile
Figure imgf000112_0003
no [0396] To a stined solution of 2A (280 mg, 1.17 mmol) in DME (2 mL) and DMF (0.5 mL) at 0 °C was added NaH (30 mg, 1.23 mmol). After ten minutes, LiBr (203 mg, 2.33 mmol) was added and the mixture was allowed to warm to RT. After 15 minutes, α-bromo-o- tolunitrile (457 mg, 2.33 mmol) was added and the mixture was heated at 65 C overnight. After cooling, water (10 mL) was added. A precipitate formed. This precipitate was filtered and dried to give 2B which was not further purified.
[0397] 2B (215 mg, 0.6 mmol) was converted to the title compound by the method used for Example 1. The product was recrystallized to give purified 2, 2-[2-(3-Amino-piperidin-l- yl)-6,7-dimethoxy-4-oxo-4H-quinazoUn-3-ylmethyl]-benzonitrile, (95 mg). 1H NMR (400 MHz, DMSO): δ 7.84 (dd, J = 0.89, 7.7 Hz, IH), 7.60 (ddd, J = 1.0, 1.1, 7.7 Hz, IH), 7.43 (t, J = 7.6 Hz, IH), 7.32 (s, IH), 7.05 (d, J = 7.9 Hz), 7.01 (s, IH), 5.41 (s, 2H), 3.90 (s, 3H), 3.81 (s, 3H), 3.25 (m, IH), 3.17 (m, IH), 2.72 (m, 2H), 1.80 (m, IH), 1.67 (m, 2H), 1.52 (m, IH), 1.11 (m, IH). MS: (ES) M+H calc'd for C23H25N5O3, 420; found 420.
Example 3: 2-[2-(3-Amino-piρeridin-l-yl)-8-methoxy-4-oxo-4H-quinazohn-3- ylmethyl] -benzonitrile
Figure imgf000113_0001
Example 3A: 8-Methoxy-lH-quinazoline-2,4-dione
Figure imgf000113_0002
[0398] 2-amino-3-methoxybenzoic acid (842 mg, 5 mmol) and urea (1.5 g, 25 mmol) were heated together at 200 °C for 1.5 hours. The mixture was cooled and triturated with water. The solid was filtered and dried to give 3A (843 mg, yellow solid, 88%). MS: (ES) M+H calc'd for C9H8N2O3, 193; found 193.
Example 3B: 2,4-Dichloro-8-methoxy-quinazoline
Figure imgf000114_0001
[0399] 3A (843 mg, 4.39 mmol) was converted to crude 3B by the method used for Example IB.
Example 3C: 2-Chloro-8-methoxy-3H-quinazolin-4-one
Figure imgf000114_0002
[0400] Crude 3B was converted to 3C (388 mg) by the method used for Example IC. MS: (ES) M+H calc'd for C9H7ClN2O2, 211; found 211.
Example 3D: 2-(2-Chloro-8-methoxy-4-oxo-4H-quinazolin-3-ylmethyl)- benzonitrile
Figure imgf000114_0003
[0401] 3C (210 mg, 1 mmol) was converted to 3D by the procedure used for Example 2B. MS: (ES) M+H calc'd for C17H12ClN3O2, 326; found 326.
[0402] 3D (230 mg, 0.7 mmol) was converted to Example 3, 2-[2-(3-Amino-ρiρeridin-l- yl)-8-me1hoxy-4-oxo-4H-qumazolin-3-ylmethyl]-benzonitrile (100 mg, 37%) by the method used for Example 1. 1H NMR (400 MHz, DMSO): δ 7.79 (dd, J = 1.2, 7.9 Hz, IH), 7.68 (dd, J = 0.98, 7.6 Hz, IH), 7.47 (ddd, J = 1.2, 1.3, 7.8 Hz, IH), 7.31 (m, 2H), 7.18 (dd, J = 1.1, 8.0 Hz), 7.03 (d, J = 7.8 Hz, IH), 5.57 (s, 2H), 4.01 (s, 3H), 3.35 (m, IH), 3.22 (m, IH), 2.96 (m, 2H), 2.76 (dd, J = 9.1, 11.9 Hz, IH), 1.96 (m, IH), 1.80 (m, IH), 1.67 (m, IH), 1.25 (m, IH). MS: (ES) M+H calc'd for C22H23N5O2, 390; found 390.
Example 4: 2- [2-(3- Amino-piperidin- l-yl)-7-chloro-4-oxo-4H-quinazohn-3- ylmethyl] -benzonitrile, TFA salt
Figure imgf000115_0001
Example 4A: 2,7-Dichloro-5H-quinazolin-4-one
Figure imgf000115_0002
[0403] The title compound was prepared from 7-chloro- lΗ-quinazoline-2,4-dione (see Schneller et. al., J. Org. Chem., 46, 8, 1981, 1699-1702) in 58% yield according to the procedures of examples IB and IC. 1H NMR (400 MHz, DMSO-αV); δ 13.41 (br s, IH), 8.07 (d, IH, J = 6.3 Hz), 7.70 (d, IH, J = 1.5 Hz), 7.57 (dd, IH, J = 6.3, 1.5 Hz). MS (ES) [m+H] calculated for H^OCl i-H 214.98, 216.97; found 215.07, 217.08. Example 4B: 2-(2,7-Dichloro-4-oxo-^H-quinazoUn-3-ylmethlyl)-benzonitrile
Figure imgf000116_0001
[0404] The title compound was prepared from 2,7-dichloro-3H-quinazoUn-4-one in 70% yield according to the procedure for 2B. 1H NMR (400 MHz, CDC13); δ 8.22 (d, IH, J = 6.3 Hz), 7.74 (dd, IH, J = 5.7, 0.9 Hz), 7.68 (d, IH, J = 0.3 Hz), 7.49-7.57 (m, 2H), 7.43 (t, IH, J = 5.7 Hz), 7.15 (d, IH, J = 5.7 Hz), 5.73 (s, 2H). MS (ES) [m+H] calculated for C16H9N3OCl2+H 330.02, 332.02; found 329.99, 332.00.
[0405] The title compound, Example 4, 2-[2-(3-Amino-piperidin-l-yl)-7-chloro-4-oxo- 4H-quinazolin-3-ylmethyl]-benzonitrile, TFA salt, was prepared from 2-(2,7-dichloro-4-oxo- 4H-qumazolin-3-ylmethlyl)-benzonitrile in 80% yield according to the procedure for compound 1. 1H NMR (400 MHz, DMSO- ); δ 7.90-8.01 (m, 4H), 7.81 (d, IH, J = 5.7 Hz), 7.56-7.64 (m, 2H), 7.38-7.48 (m, 2H), 7.26 (d, IH, J = 5.7 Hz), 5.36 (dd, 2H, J = 34.8, 11.4 Hz), 3.52-3.58 (m, IH), 3.36-3.46 (m, IH), 3.03-3.24 (m, 2H), 2.87-2.94 (m, IH), 1.92-1.99 (m, IH), 1.78-1.85 (m, IH), 1.50-1.69 (m, 2H). MS (ES) [m+H] calculated for C2ιH20N5OCl+H 394.14, 396.14; found 394.02, 395.96.
Example 5: 2-[2-(3-Amino-piperidin- l-yl)-8-chloro-4-oxo-4H-quinazolin-3- ylmethyl] -benzonitrile, TFA salt
Figure imgf000116_0002
Example 5A: 2,8-Dichloro-3H-quinazolin-4-one
Figure imgf000117_0001
[0406] The title compound was prepared from 8-chloro- lH-quinazoline-2,4-dione (see Bindra, US4085213) in 37% yield according to the procedures of examples IB and IC. 1H NMR (400 MHz, DMSO- ); δ 13.50 (br s, IH), 8.04 (dd, IH, J = 6.0, 1.2 Hz), 7.98 (dd, IH, J = 6.0, 1.2 Hz), 7.51 (t, IH, J = 6.0 Hz). MS (ES) [m+H] calculated for C8H4N2OCl2+H 214.98, 216.97; found 215.07, 217.08.
Example 5B: 2-(2,8-Dichloro-4-oxo-4H-quinazolin-3-ylmethlyl)-benzonitrile
Figure imgf000117_0002
[0407] The title compound was prepared from 2,8-dicUoro-5H-quinazolin-4-one in 72% yield according to the procedure for 2B. 1H NMR (400 MHz, CDC13); δ 8.22 (dd, IH, J = 6.0, 1.2 Hz), 7.89 (dd, IH, J = 6.0, 1.2 Hz), 7.74 (dd, IH, J = 6.0, 0.9 Hz), 7.42-7.76 (m, 3H), 7.14 (d, IH, J = 6.0 Hz), 5.75 (s, 2H). MS (ES) [m+H] calculated for Cι6H9N3OCl2+H 330.02, 332.02; found 329.93, 331.94.
[0408] The title compound, Example 5, 2-[2-(3-Amino-piperidin-l-yl)-8-chloro-4-oxo- 4H-quinazolin-3-ylmethyl]-benzonitrile, TFA salt, was prepared from 2-(2,8-dichloro-4-oxo- ^H-quinazolin-3-ylmethlyl)-benzonitrile in 76% yield according to the procedure for compound 1.. 1H NMR (400 MHz, DMSO-αV); δ 7.88-8.02 (m, 5H), 7.81 (dd, IH, J = 5.7, 0.6 Hz), 7.60 (dt, IH, J = 5.7, 0.9 Hz), 7.44 (t, IH, J = 5.7 Hz), 7.27-7.36 (m, 2H), 5.37 (dd, 2H, J = 33.3, 11.4 Hz), 3.60-3.66 (m, IH), 3.41-3.50 (m, IH), 3.15-3.25 (m, 2H), 2.90-2.99 (m, IH), 1.92-1.99 (m, IH), 1.79-1.87 (m, IH), 1.51-1.69 (m, 2H). MS (ES) [m+H] calculated for C2ιH2oN5OCl+H 394.14, 396.14; found 394.04, 396.06.
Example 6: 2-[2-(3-Amino-piperidin- l-yl)-6-fluoro-4-oxo-4H-quinazolin-3- ylmethyl]-benzonitrile
Figure imgf000118_0001
Example 6A: 6-Fluoro-lH-quinazoline-2,4-dione
Figure imgf000118_0002
[0409] 2-Amino-6-fluoro-benzoic acid was converted to the title compound by the method used for Example 3 A.
Example 6B: 2,4-Dichloro-6-fluoro-quinazoline
Figure imgf000118_0003
[0410] 6A was converted to the title compound by the method used for Example IB.
Example 6C: 2-Chloro-6-fluoro-3H-quinazolin-4-one
Figure imgf000119_0001
[0411] 6B was converted to the title compound by the method used for Example IC.
Example 6D: 2-(2-CUoro-6-fluoro-4-oxo-4H-qumazolin-3-ylmethyl)-benzoxιitrile
Figure imgf000119_0002
[0412] 6C was converted to the title compound by the method used for Example ID. 1H NMR (400 MHz CDC13): 87.93 (dd, J = 2.8, 8.0 Hz, IH) 7.68-7.75 (m, 2H), 7.50-7.60 (m, 2H), 7.42 (dd, J = 7.2, 7.6 Hz, IH), 7.15 (d, J = 8.0 Hz, IH), 5.74 (s, 2H). MS (ES) [m+H] calculated for Cι6H9ClFN30, 314; found 314.
[0413] 6D was converted to the title compound by the method used for Example 1. 1H NMR (400 MHz. CD3OD): 87.52-7.9 (m, 5H), 7.41-7.51 (m, IH), 7.35 (d, J = 8.0 Hz, IH), 5.44-5.66 (AB q, J = 16.0 Hz, 2H), 3.62-3.71 (m, IH), 3.55-3.60 (m, IH), 3.19-3.33 (m, 2H), 2.94-3.05 (m, IH), 2.11-2.20 (m, IH), 1.60-1.95 (m, 3H). MS (ES) [m+H] calculated for C2ιH20FN5O, 378; found 378.
Example 6E: (R) 2-[2-(3-Aιruno-piperidin-l-yl)-6-fluoro-4-oxo-4H-quinazolin-3- ylmethyl]-benzonitrile TFA salt
Figure imgf000119_0003
[0414] The (R) enantiomer form of 6D where was converted to 6E by the method used for Example 1. 1H NMR (400 MHz CD3OD) δ 7.52-7.9 (m, 5H), 7.41-7.51 (m, IH), 7.35 (d, J = 8.0 Hz, IH), 5.44-5.66 (AB q, J = 16.0 Hz, 2H), 3.62-3.71 (m, IH), 3.55-3.60 (m, IH), 3.19-3.33 (m, 2H), 2.94-3.05 (m, IH), 2.11-2.20 (m, IH), 1.60-1.95 (m, 3H). MS (ES) [m+H] calculated for C21H20FN50, 378; found 378.
Example 7: 2-[2-(3-Amino-piperidin- l-yl)-7-methyl-6-oxo-6,7-dihydro-purin- 1- ylmethylj-benzonitrile
Figure imgf000120_0001
Example 8: 2-[2-(3-Amino-piperidin- 1 -yl)-9-methyl-6-oxo-6,9-dihydro-purin- 1 - ylmethyl] -benzonitrile
Figure imgf000120_0002
Example 7A: 2,6-Dichloro-7-methyl-7H-purine
Figure imgf000120_0003
Example 8A: 2,6-Dichloro-9-methyl-9H-purine
Figure imgf000121_0001
[0415] To a cold solution of 2,6-Dichloro-7H-purine (lg, 5.29 mmol) in DMF (5 mL) was added 60% NaH (233 mg, 5.8 mmol) under N2. After stirring for 10 min, Mel (910 μL) was added, the reaction mixture was stined for 30 min at this temperature and then overnight at room temperature, concentrated and co-evaporated with H2O to dryness. The residue was suspended in 2 mL cold water and filtered to the give crude mixture of 7A and 8A in about 1 : 1 ratio.
[0416] Selected data for 7A: NMR (400 MHz DMSO): β 8.79 (s, IH), 4.06 (s, 2H). [0417] Selected data for 8A: NMR (400 MHz DMSO): δ 8.66 (s, IH), 3.82 (s, 3H). MS (ES) [m+H] calculated for C6H C12N4+H, 203; found 203.
Example 7B: 2-Chloro-7-methyl-l,7-dihydro-purin-6-one
Figure imgf000121_0002
Example 8B: 2-Chloro-9-methyl-l,9-dihydro-ρurin-6-one
Figure imgf000121_0003
[0418] Tne crude products 7A and 8A were converted to 7B and 8B as a mixture by the method used for the preparation of IC in 82% yield.
[0419] Selected data for 7B: NMR (400 MHz DMSO): β 8.79 (s, IH), 4.06 (s, 2H). [0420] Selected data for 8B: NMR (400 MHz DMSO): 88.66 (s, IH), 3.82 (s, 3H). MS (ES) [m+H] calculated for C6H5ClN4O+H, 185; found 185.
Example 7C: 2-(2-CMoro-7-methyl-6-oxo-6,7-dmydro-purin-l-ylmethyl)- benzonitrile
Figure imgf000122_0001
Example 8C: 2-(2-Chloro-9-methyl-6-oxo-6,9-dihydro-purin-l-ylmethyl)- benzonitrile
Figure imgf000122_0002
[0421] A mixture of title compounds 7C and 8C was prepared by the method used in the preparation of ID.
[0422] Selected data for 7C: NMR (400 MHz CDC13): β 8.04 (Bs, IH), 5.74 (s, 2H), 4.11
(s, 3H).
[0423] Selected data for 8C: NMR (400 MHz CDC13): β 7.91 (Bs, IH), 5.77 (s, 2H), 3.85
(s, 3H). MS (ES) [m+H] calculated for 014*1100^0+ 1, 300; found 300.
[0424] A mixture of compound 7C and 8C was converted by the method used in the preparation of Example 1 to the title compounds 7 and 8, which were purified by LC-MS.
[0425] Example 7: NMR (400 MHz CDQ3-CD3OD 10: 1): β 7.9 (br s, IH), 7.63 (d, IH, J
= 7.2 Hz), 7.53 (t, IH, J = 7.3 Hz), 7.37 (t, IH, 7.3Hz), 7.20 (d, IH, J = 8.0 Hz), 5.50-5.38
(AB q, 2H, J = 15.2 Hz), 3.78 (s, 3H), 3.60 (br d, IH, J = 12.4 Hz), 3.53 (m, IH), 3.22 (dd, IH, J = 8.4 and 12 Hz), 3.1 (m, IH), 2.96 (m, IH), 2.19 (m, IH), 1.87 (m, IH), 1.72 (m, 2H). MS (ES) [m+H] calculated for Cι9H2ιN7O+H, 364.18; found 364.17. [0426] Example δ: NMR (400 MHz CDCI3-CD3OD 10:l): β7.91 (brs, lH), 7.69 (d, IH, J = 7.2 Hz), 7.58 (t, IH, J = 7.3 Hz), 7.42 (t, IH, 7.3 Hz), 7.20 (d, IH, J = 8.0 Hz), 5.57-5.43 (AB q, 2H, J = 15.2 Hz), 4.02 (s, 3H), 3.56 (br d, IH, J = 12.4 Hz), 3.50 (m, IH), 3.15 (m, 2H), 2.96 (m, IH), 2.11 (m, IH), 1.84 (m, IH), 1.70 (m, 2H). MS (ES) [m+H] calculated for Cι9H2ιN7O+H, 364.18; found 364.17.
Example 9: 2-{2-[(R)-3-Amino-piperidin-l-yl]-6-oxo-6,7-dihydro-purin-l- ylmethyl } -benzonitrile
Figure imgf000123_0001
Example 9A: 7-Benzyloxymethyl-2,6-dichloro-7H-purine
Figure imgf000123_0002
Example 9B: 9-Benzyloxymethyl-2,6-dichloro-9H-purine
Figure imgf000123_0003
[0427] A mixture of Example 9A and Example 9B was synthesized from 2,6-Dichloro- 7H-purine by the method described in syntheses of Example 7A and 8A. [0428] Example 9B: NMR (400 MHz CDC13): β 8.2 (s, IH), 7.3 (m, 5H), 5.70 (s, 2H), 4.63 (s, 2H). MS (ES) [m+H] calculated for Cι30Cl2N4O+H, 309; found 309.
Example 9C: 7-Benzyloxymemyl-2-cMoro-l,7-d ydro-purin-6-one
Figure imgf000124_0001
Example 9D: 9-Benzyloxymethyl-2-chloro-l,9-dihydro-purin-6-one
Figure imgf000124_0002
[0429] The mixture of Example 9A and Example 9B was converted to Example 9C and Example 9D by the method used in the preparation of Example IC.
Example 9E: 2-(7-Benzyloxymethyl-2-chloro-6-oxo-6,7-dihydro-purin-l- ylmethyl)-benzonitrile
Figure imgf000124_0003
Example 9F: 2-(9-Benzyloxymethyl-2-chloro-6-oxo-6,9-dihydro-purin-l- ylmethyl)-benzonitrile
Figure imgf000125_0001
Example 9G: 2-(2-Clιloro-6-oxo-6,9-dihydro-purin-l-ylmethyl)-benzonitrile
Figure imgf000125_0002
[0430] Alkylation of the mixture of Example 9C and Example 9D by the method used for Example IC gave a mixture of Example 9E and Example 9F, which was treated with 4N
HCl in dioxane at 40 °C overnight to give Example 9G.
[0431] Example 9G: NMR (400 MHz CDC13): β 8.02 (br s, IH), 7.70 (d, IH, J = 7.6
Hz), 7.53 (t, IH, J = 7.6 Hz), 7.40 (t, IH, 7.6Hz), 7.06 (d, IH, J = 8.0 Hz), 5.72 (s, 2H). MS
(ES) [m+H] calculated for Cι3H8ClN5O+H, 286.04; found 286.03.
[0432] The title compound of Example 9, 2-{2-[(R)-3-Amino-piperidin-l-yl]-6-oxo-6,7- dihydro-purin- 1 -ylmethyl } -benzonitrile, was prepared from Example 9G by the method used in the preparation of Example 1.
[0433] Example 9: NMR (400 MHz CDCI3-CD3OD 10: 1): β 8.05 (s, IH), 7.59 (d, IH, J
= 7.2 Hz), 7.46 (t, IH, J = 7.6 Hz), 7.31 (t, IH, 7.6 Hz), 7.09 (d, IH, J = 7.2 Hz), 5.50-5.33
(AB q, 2H, J= 15.6 Hz), 3.47 (m, IH), 3.24 (m, IH), 3.07 (m, IH), 2.92 (m, IH), 2.01 (m,
IH), 1.74 (m, 2H), 1.60(m, IH). MS (ES) [m+H] calculated for Cι89N7O+H, 350.2 found
350.2.
Example 10: 2-[2-(3-(R)-Amino-piperidin-l-yl)-6-chloro-4-oxo-4H-quinazolin-3- ylmethyl] -benzonitrile, TFA salt
Figure imgf000126_0001
Example 10A: 2,6-Dichloro-3H-quinazolin-4-one
Figure imgf000126_0002
[0434] The title compound was prepared from 6-chloro-lH-quinazohne-2,4-dione (see McKee et al., J. Amer. Chem. Soc, 69, 1947, 940) in 59% yield according to the procedures of examples IB and IC. 1H NMR (400 MHz, DMSO- ):. δ 13.44 (br s, IH), 8.01 (d, IH, J = 2.4 Hz), 7.85 (dd, IH, J = 8.4, 2.4 Hz), 7.63 (d, IH, J = 8.4 Hz). MS (ES) [m+H] calc'd for C8H N2OCl2215, 217; found 215, 217.
Example 10B: 2-(2,6-Dichloro-4-oxo-4H-quinazolin-3-ylmethyl)-benzonitrile
Figure imgf000126_0003
[0435] The title compound was prepared from 2,6-dic oro-5H-qumazolin-4-one in 63 % yield according to the procedure for example 2B. 1H NMR (400 MHz, CDC13); δ 8.26 (s, IH), 7.72-7.77 (m, 2H), 7.63 (d, IH, J = 8.8 Hz), 7.54 (dt, IH, J = 7.6, 1.2 Hz), 7.43 (t, IH, J = 7.6 Hz), 7.15 (d, IH, J = 7.6 Hz), 5.74 (s, 2H). MS (ES) [m+H] calc'd for Cι6H9N3OCl2330, 332; found 330, 332.
[0436] The title compound, Example 10, 2-[2-(3-(R)-Amino-ρiρeridin-l-yl)-6-chloro-4- oxo-4H-quinazolm-3-ylmethyl]-benzonitrile, TFA salt, was prepared from 2-(2,6-dichloro-4- oxo-4H-quinazolin-3-ylmethyl)-benzonitrile in 70% yield according to the procedure for example 1. 1H NMR (400 MHz, DMSO-d*): δ 7.99 (br s, 3H), 7.88 (d, IH, J = 1.2 Hz), 7.76- 7.83 ( , 2H), 7.54-7.63 (m, 2H), 7.44 (t, IH, J = 7.6 Hz), 7.25 (d, IH, J = 7.6 Hz), 5.38 (AB q, 2H, J = 48.0, 15.2 Hz), 3.51-3.59 (m, IH), 3.38-3.45 (m, IH), 3.02-3.21 (m, 2H), 2.84-2.93 (m, IH), 1.91-2.00 (m, IH), 1.79-1.88 (m, IH), 1.50-1.69 (m, 2H). MS (ES) [m+H] calc'd for C2ιH20N5OCl 394, 396; found 394, 396.
Example 11: 2-[2-(3-(R)-Amino-piperidin-lyl)-7-fluoro-6-methoxy-4-oxo-4H- quinazolin-3-ylmethyl]-benzonitrile, TFA salt
Figure imgf000127_0001
Example 11 A: 7-Fluoro-6-methoxy-lH-quinazoline-2,4-dione
Figure imgf000127_0002
[0437] The title compound was prepared from 2-amino-4-fluoro-5-methoxy-benzoic acid methyl ester (see EP602851) in 90% yield according to the procedure for example 3A. 1H NMR (400 MHz, DMSO-α ):. δ 11.05 (br s, 2H), 7.50 (d, IH, J = 9.2 Hz), 6.98 (d, IH, J = 12.0 Hz), 3.88 (s, 3H). MS (ES) [m+H] calc'd for C9H7N2O3F 211; found 211.
Example 11B: 2-CUoro-7-fluoro-6-memoxy-3H-quinazolin-4-one
Figure imgf000128_0001
[0438] The title compound was prepared from 7-fluoro-6-methoxy- lH-quinazoline-2,4- dione in 80% yield according to the procedures of examples IB and IC. 1H NMR (400 MHz, DMSO-αV): δ 13.29 (br s, IH), 7.62 (d, IH, J = 9.2 Hz), 7.51 (d, IH, J = 12.0 Hz), 3.95 (s, 3H). MS (ES) [m+H] calc'd for C9H6N2O2FCl 229, 231; found 229, 231.
Example IIC: 2-(2-CMoro^7-fluoro-6-methoxy-4-oxo-4H-quinazolin-3- ylmethyl)-benzonitrile
Figure imgf000128_0002
[0439] The title compound was prepared from 2-chloro-7-fluoro-6-methoxy-3H- quinazolin-4-one in 67% yield according to the procedure for example 2B. 1H NMR (400 MHz, CDC13); δ 7.71-7.74 (m, 2H), 7.54 (dt, IH, J = 7.6, 1.2 Hz), 7.36-7.44 (m, 2H), 7.14 (d, IH, J = 7.6 Hz), 5.74 (s, 2H), 4.01 (s, 3H). MS (ES) [m+H] calc'd for C17HπN3O2FCl 344, 346; found 344, 346.
[0440] The title compound, Example 11, 2-[2-(3-(R)-Amino-piperidin-lyl)-7-fluoro-6- methoxy-4-oxo-4H-quinazolin-3-ylmethyl]-benzonitrile, TFA salt, was prepared from 2-(2- chloro-7-fluoro-6-methoxy-4-oxo-4H-quinazolin-3-ylmethyl)-benzonitrile in 85% yield according to the procedure for compound 1. 1H NMR (400 MHz, DMSO-α* <j): δ 7.93 (br s, 3H), 7.82 (d, IH, J = 7.6 Hz), 7.60 (dt, IH, J = 7.6, 1.2 Hz), 7.52 (d, IH, J = 9.2 Hz), 7.38- 7.46 (m, 2H), 7.21 (d, IH, J = 7.6 Hz), 5.39 (AB q, 2H, J = 51.2, 15.2 Hz), 3.89 (s, 3H), 3.46- 3.53 (m, IH), 3.34-3.42 (m, IH), 3.01-3.18 (m, 2H), 2.81-2.89 (m, IH), 1.91-1.99 (m, IH), 1.78-1.86 (m, IH), 1.49-1.70 (m, 2H). MS (ES) [m+H] calc'd for C22H22N5O2F 408; found 408. Example 12: 2-[2-(3-(R)-Arnino-piperidin-lyl)-6-methoxy-4-oxo-4H-ρyrido[3,4- αlpyrimidin-S-ylmethyy-benzonitrile, TFA salt
Figure imgf000129_0001
Example 12A: 6-Methoxy-lH-pyrido[3,4-αr|pyrinιidiήe-2,4-dione
Figure imgf000129_0002
[0441] Ihe title compound was prepared from 5-amino-2-methoxypyridine-4-carboxylic acid (see Rewcastle et al., J. Chem. Soc. Perkin Trans. 1, 1996, 2221-2226) in 77% yield according to the procedure for example 3A. 1H NMR (400 MHz, DMSO-αV); δ 11.29 (br s, 2H), 8.16 (s, IH), 7.12 (s, IH), 3.85 (s, 3H). MS (ES) [m+H] calc'd for C8H7N3O3 194; found 194.
Example 12B: 2-Chloro-6-methoxy-3H-pyrido[3,4-αlpyrimidin-4-one
Figure imgf000129_0003
[0442] The title compound was prepared from 6-methoxy-lH-pyrido[3,4-αlpyrimidine- 2,4-dione in 62% yield according to the procedures of examples IB and IC. 1H NMR (400 MHz, DMSO-αV):. δ 13.32 (br s, IH), 8.66 (s, IH), 7.25 (s, IH), 3.93 (s, 3H). MS (ES) [m+H] calc'd for HeNgOaCl 212, 214; found 212, 214.
Example 12C: 2-(2-Chloro-6-memoxy-4-oxo-4H-pyrido[3,4-^pyrimidin-3- ylmethyl)-benzonitrile
Figure imgf000130_0001
[0443] The title compound was prepared from 2-chloro-6-methoxy-3H-pyrido[3,4- αlpyrimidin-4-one in 63% yield according to the procedure for example 2B. 1H NMR (400 MHz, CDC13): δ 8.75 (s, IH), 7.74 (dd, IH, J = 7.6, 1.2 Hz), 7.55 (dt, IH, J = 7.6, 1.2 Hz), 7.42-7.47 (m, 2H), 7.14 (d, IH, J = 7.6 Hz), 5.72 (s, 2H), 4.04 (s, 3H). MS (ES) [m+H] calc'd for Cι6HnN4O2Cl 327, 329; found 327, 329.
[0444] The title compound, Example 12, 2-[2-(3-(R)-Am o-piperidin-lyl)-6-methoxy-4- oxo-4H-pyrido [3, 4-ατ]pyrimidin-3-ylmethyl] -benzonitrile, TTA salt, was prepared from 2-(2- chloro-6-memoxy-4-oxo-4H-pyrido[3,4-αlpyrimidin-3-ylmethyl)-benzonitrile in 51% yield according to the procedure for example 1. 1H NMR (400 MHz, DMSO-αV): δ 8.62 (s, IH), 7.94 (br s, 3H), 7.81 (d, IH, J = 7.6 Hz), 7.60 (dt, IH, J = 7.6, 1.2 Hz), 7.44 (t, IH, J = 7.6 Hz), 7.26 (d, IH, J = 7.6 Hz), 7.16 (s, IH), 5.39 (AB q, 2H, J = 50.8, 15.2 Hz), 3.90 (s, 3H), 3.50-3.57 (m, IH), 3.32-3.41 (m, IH), 3.00-3.18 (m, 2H), 2.80-2.88 (m, IH), 1.91-2.00 (m, IH), 1.78-1.86 (m, IH), 1.48-Ϊ.66 (m, 2H). MS (ES) [m+H] calc'd for C2ιH22N6O2 391; found 391.
Example 13: 2-[6-(3-(R)-Amino-ρiρeridin-l-yl)-l-methyl-4-oxo-l,4-dihydro- pyrazolo[3,4-αlpyrirnidin-5-ylmethyl]-benzonitrile, TFA salt
Figure imgf000131_0001
Example 13A: 6-Cliloro-l-memyl-l,5-dmydro-pyrazolo[3,4-αlpyrirnidin-4-one
Figure imgf000131_0002
[0445] The title compound was prepared from 1 -methyl- 1,7-dihydro-pyrazolo [3,4- α^pyrimidine-4,6-dione (see Cheng et al., J. Org. Chem., 23, 1958, 852-855) in 11% yield according to the procedures of examples IB and IC. 1H NMR (400 MHz, MeOD-d ): δ 8.00 (s, IH), 3.92 (s, 3H). MS (ES) [m+H] calc'd for C6H5N4OCl 185, 187; found 185, 187.
Example 13B: 2-(6-Chloro-l-methyl-4-oxo-l,4-dihydro-pyrazolo[3,4- α'lpyrimidin-S-ylmethy^-benzonitrile
Figure imgf000131_0003
[0446] The title compound was prepared from 6-chloro-l -methyl- 1,5-dihydro- pyrazolo[3,4--i]pyrirnidin-4-one in 78% yield according to the procedure for example 2B. 1H NMR (400 MHz, CDC13): δ 8.09 (s, IH), 7.72 (dd, IH, J = 7.6, 1.2 Hz), 7.53 (dd, IH, J = 7.6, 1.2 Hz), 7.41 (t, IH, J = 7.6 Hz), 7.10 (d, IH, J = 7.6 Hz), 5.71 (s, 2H), 3.99 (s, 3H). MS (ES) [m+H] calc'd for C14H10N5OCI 300, 302; found 300, 302. [0447] The title compound, Example 13, 2-[6-(3-(R)-Ammo-piperidin-l-yl)-l-methyl-4- oxo-l,4-ά hydro-pyrazolo[3,4-α^pyrimidin-5-ylmemyl]-benzo trile, TFA salt, was prepared from 2-(6-chloro- 1 -methyl-4-oxo- 1 ,4-dihydro-pyrazolo [3 ,4-α^pyrimidin-5-ylmethyl)- benzonitrile in 72% yield according to the procedure for example 1. 1H NMR (400 MHz, DMSO-αV): δ 8.00 (br s, 3H), 7.92 (s, IH), 7.81 (dd, IH, J = 7.6, 1.2 Hz), 7.59 (dt, IH, J = 7.6, 1.2 Hz), 7.43 (t, IH, J = 7.6 Hz), 7.15 (d, IH, J = 7.6 Hz), 5.33 (AB q, 2H, J = 44.8, 15.2 Hz), 3.85 (s, 3H), 3.51-3.59 (m, IH), 3.35-3.44 (m, IH), 3.15-3.23 (m, IH), 3.02-3.11 (m, IH), 2.88-2.96 (m, IH), 1.91-2.00 (m, IH), 1.77-1.85 (m, IH), 1.50-1.67 (m, 2H). MS (ES) [m+H] calc'd for d9H2ιN7O 364; found 364.
Example 14: 2-[2-(3-(R)-Amino-piperidin-lyl)-5-fluoro-4-oxo-4H-quinazolin-3- ylmethyl] -benzonitrile, TTA salt
Figure imgf000132_0001
Example 14A: 2-Chloro-5-fluoro-3H-quinazolin-4-one
Figure imgf000132_0002
[0448] The title compound was prepared from 5-fluoro-lH-quinazoline-2,4-dione (see Michel et al., Tetrahedron, 53 (25), 1997, 8457-8478) in 11% yield according to the procedures of examples IB and IC. 1H NMR (400 MHz, OMSO-dδ): δ 13.31 (br s, IH), 7.77- 7.83 (m, IH), 7.41 (d, IH, J = 7.6 Hz), 7.26-7.32 (m, IH). MS (ES) [m+H] calc'd for H N2OFCI 199, 201; found 199, 201. Example 14B: 2-(2-CMoro-5-fluoro-4-oxo-4H-qumazolin-3-ylmethyl)- benzonitrile
Figure imgf000133_0001
[0449] The title compound was prepared from 2-chloro-5-fluoro-3H-quinazolin-4-one in 70% yield according to the procedure for example 2B. 1H NMR (400 MHz, CDC13): δ 7.70- 7.79 (m, 2H), 7.40-7.58 (m, 3H), 7.17-7.24 (m, 2H), 5.72 (s, 2H). MS (ES) [m+H] calc'd for Cι6H9N3OFCl 314, 316; found 314, 316.
[0450] The title compound, Example 14, 2-[2-(3-(R)-Amino-piρeridin- lyl)-5-fluoro-4- oxo-4H-quinazolin-3-ylmethyl]-benzonitrile, TFA salt, was prepared from 2-(2-chloro-5- fluoro-4-oxo-4H-qumazolm-3-ylmemyl)-benzonitrile in 53% yield according to the procedure for example 1. 1H NMR (400 MHz, DMSO-α ): δ 7.81 (dd, IH, J = 7.6, 1.2 Hz), 7.68-7.73 (m, IH), 7.61 (dt, IH, J = 7.6, 1.2 Hz), 7.44 (t, IH, J = 6.8 Hz), 7.32 (d, IH, J = 7.6 Hz), 7.26 (d, IH, J = 7.6 Hz), 7.08-7.13 (m, IH), 6.97 (br s, 2H), 5.33 (AB q, 2H, J = 35.6, 15.2 Hz), 3.49-3.55 (m, IH), 3.17-3.36 (m, 2H), 2.81-2.99 (m, 2H), 1.90-1.99 (m, IH), 1.78-1.86 (m, IH), 1.41-1.66 (m, 2H). MS (ES) [m+H] calc'd for C2ιH20N5OF 378; found 378.
Example 15: 2- [5-(3-(R)-Amino-piperidin- 1 -yl)- 1 -methyl-7-oxo- 1 ,7-dihydro- [l,2,3]triazolo[4,5-α1pyrin3idin-6-ylmethyl]-benzonitrile, TFA salt
Figure imgf000133_0002
Example 15A: 5-CUoro-l-methyl-l,6-d ydro-[l,2,3]triazolo[4,5-α^pyrirrridin-7- one
Figure imgf000134_0001
[0451] The title compound was prepared from l-memyl-l,4-dihydro-[l,2,3]triazolo[4,5- απpyrimidine-5,7-dione (see Smirnova et al., J. Org. Chem. USSR Eng. Trans., 14, 1978, 1617) in 64% yield according to the procedures of examples IB and IC. 1H NMR (400 MHz, DMSO-α ): δ 13.39 (br s, IH), 4.36 (s, 3H). MS (ES) [m+H] calc'd for C5H N5OCI 186, 188; found 186, 188.
Example 15B: 2-(5-Chloro-l-methyl-7-oxo-l,7-dihydro-[l,2,3]triazolo[4,5- α^pyrimidin-6-ylmethyl)-benzonitrile
Figure imgf000134_0002
[0452] The title compound was prepared from 5-chloro-l -methyl- 1,6-dihydro- [l,2,3]triazolo[4,5-αlρyrimidin-7-one in 70% yield according to the procedure for example 2B. 1H NMR (400 MHz, CDCI3): δ 7.73 (dd, IH, J = 7.6, 1.2 Hz), 7.55 (dt, IH, J = 7.6, 1.2 Hz), 7.43 (t, IH, J = 7.6 Hz), 7.10 (d, IH, J = 7.6 Hz), 5.76 (s, 2H), 4.45 (s, 3H). MS (ES) [m+H] calc'd for d3H9N6OCl 301, 303; found 301, 303.
[0453] The title compound, Example 15, 2-[5-(3-(R)-Amino-piperidin-l-yl)-l-methyl-7- oxo-l,7-dmydro-[l,2,3]triazolo[4,5-α^pyrimidin-6-ylmethyl]-benzonitrile, TFA salt, was prepared from 2-(5-cUoro-l-memyl-7-oxo-l,7-dmydro-[l,2,3]triazolo[4,5-α1pyrimidin-6- ylmethyl)-benzonitrile in 46% yield according to the procedure for example 1. 1H NMR (400 MHz, DMSO-α ): δ 7.96 (br s, 3H), 7.81 (dd, IH, J = 7.6, 1.2 Hz), 7.61 (dt, IH, J = 7.6, 1.2 Hz), 7.44 (t, IH, J = 7.6 Hz), 7.24 (d, IH, J = 7.6 Hz), 5.38 (AB q, 2H, J = 48.0, 15.2 Hz), 4.31 (s, 3H), 3.49-3.56 (m, IH), 3.31-4.41 (m, IH), 3.22-3.30 (m, IH), 2.98-3.06 (m, IH), 2.82-2.90 (m, IH), 1.91-1.99 (m, IH), 1.77-1.85 (m, IH), 1.48-1.65 (m, 2H). MS (ES) [m+H] calc'd for Cι8H20N8O 365; found 365.
Example 16: 2-[5-(3-(R)-Amino-piρeridin-l-yl)-2-methyl-7-oxo-2,7- dmydro[l,2,3]triazolo[4,5-αlpyrirnidin-6-ylmethyl]-benzonitrile, TFA salt
Figure imgf000135_0001
Example 16A: 5-CUoro-2-methyl-2,6-dmydro-[l,2,3]1riazolo[4,5-α^pyrimidin-7- one
Figure imgf000135_0002
[0454] The title compound was prepared from 2-methyl-2,4-dihydro- [ 1 ,2,3]triazolo [4,5- αlpyrimidine-5,7-dione (see Smimova et al., J. Org. Chem. USSR Eng. Trans., 14, 1978, 1617) in 36% yield according to the procedures of examples IB and IC. 1H NMR (400 MHz, DMSO-α* <j): δ 13.65 (br s, IH), 4.30 (s, 3H). MS (ES) [m+H] calc'd for C5H4N5OCI 186, 188; found 186, 188.
Example 16B: 2-(5-Chloro-2-methyl-7-oxo-2,7-dihydro-[l,2,3]triazolo[4,5- απpyrimidin-6-ylmethyl)-benzonitrile
Figure imgf000136_0001
[0455] The title compound was prepared from 5-chloro-2-methyl-2,6-dihydro- [l,2,3]triazolo[4,5-α^pyrimidin-7-one in 58% yield according to the procedure for example 2B. 1H NMR (400 MHz, CDC13): δ 7.75 (dd, IH, J = 7.6, 1.2 Hz), 7.57 (dt, IH, J = 7.6, 1.2 Hz), 7.45 (t, IH, J = 7.6 Hz), 7.09 (d, IH, J = 7.6 Hz), 5.74 (s, 2H), 4.47 (s, 3H). MS (ES) [m+H] calc'd for Cι3H9N6OCl 301, 303; found 301, 303.
[0456] The title compound, Example 16, 2-[5-(3-(R)-Ammo-ρiperidin-l-yl)-2-methyl-7- oxo-2,7-dmydro[l,2,3]1riazolo[4,5-αlpyrinιidin-6-ylmemyl]-benzonitrile, TFA salt, was prepared from 2-(5-c oro-2-methyl-7-oxo-2,7-dihydro- [ 1 ,2,3]triazolo [4,5-α1pyrimidin-6- ylmethyl)-benzonitrile in 74% yield according to the procedure for example 1. 1H NMR (400 MHz, OMSO-d6): δ 7.97 (br s, 3H), 7.83 (dd, IH, J = 7.6, 1.2 Hz), 7.61 (dt, IH, J = 7.6, 1.2 Hz), 7.46 (t, IH, J = 7.6 Hz), 7.25 (d, IH, J = 7.6 Hz), 5.39 (AB q, 2H, J = 48.0, 15.2 Hz), 4.24 (s, 3H), 3.49-3.56 (m, IH), 3.31-4.41 (m, IH), 3.22-3.30 (m, IH), 2.98-3.06 (m, IH), 2.82-2.90 (m, IH), 1.91-1.99 (m, IH), 1.77-1.85 (m, IH), 1.48-1.65 (m, 2H). MS (ES) [m+H] calc'd for C18H20N8O 365; found 365.
[0457] Example 17: 2-[2-(3-(R)-Amino-piperidin-lyl)-4-oxo-5,6,7,8-tetrahydro-4H- quinazolm-3-ylme1hyl]-benzonitrile, TTA salt
Figure imgf000136_0002
Example 17A: 2-Chloro-5,6,7,8-tetrahydro-3H-quinazolin-4-one
Figure imgf000137_0001
[0458] The title compound was prepared from 5,6,7,8-tefrahydro-lH-quinazoline-2,4- dione (see EP604920) in 47% yield according to the procedures of examples IB and IC. 1H NMR (400 MHz, DMSO-αV): δ 13.20 (br s, IH), 2.58-2.72 (m, 4H), 1.75-1.92 (m, 4H). MS (ES) [m+H] calc'd for C8H9N2OCl 185, 187; found 185, 187.
Example 17B: 2-(2-Chloro-4-oxo-5,6,7,8-tetrahydro-4H-quinazolin-3-ylmethyl)- benzonitrile
Figure imgf000137_0002
[0459] The title compound was prepared from 2-chloro-5,6,7,8-tetrahydro-3H- quinazolin-4-one in 59% yield according to the procedure for example 2B. 1H NMR (400 MHz, CDC13): δ 7.70 (dd, IH, J = 7.6, 1.2 Hz), 7.55 (dt, IH, J = 7.6, 1.2 Hz), 7.41 (t, IH, J = 7.6 Hz), 7.14 (d, IH, J = 7.6 Hz), 5.62 (s, 2H), 2.59-2.65 (m, 2H), 2.50-2.58 (m, 2H), 1.71- 1.87 (m, 4H). MS (ES) [m+H] calc'd for QeHi^OCl 300, 302; found 300, 302. [0460] 2-(2-CUoro-4-oxo-5,6,7,8-tetiahyάro-4H-quinazolm-3-ylmethyl)-benzonitrile (150 mg, 0.5 mmol), 3-(R)-amino-piperidine dihydrochloride (104 mg, 0.6 mmol) and sodium bicarbonate (168 mg, 2.0 mmol) were stined in ethanol (5 mL) at 60 °C for 2h. The reaction was diluted with EtOAc, washed with brine, dried (MgSO4), and concentrated in vacuo. Purification by preparative ΗPLC gave 163 mg (68%) of the title compound, Example 17, 2- [2-(3-(R)-Amino-piperidin-lyl)-4-oxo-5,6,7,8-tetiahydro-4H-qumazolm-3-ylmethyl]- benzonitrile, TFA salt, as a white solid. 1H NMR (400 MHz, DMSO-αV): δ 8.01 (br s, 3H), 7.82 (d, IH, J = 7.6 Hz), 7.61 (t, IH, J = 7.6 Hz), 7.44 (t, IH, J = 7.6 Hz), 7.09 (d, IH, J = 7.6 Hz), 5.26 (AB q, 2H, J = 44.8, 15.2 Hz), 3.25-3.40 (m, 2H), 2.90-3.08 (m, 2H), 2.70-2.80 (m, IH), 2.48 (br s, 2H), 2.23 (br s, 2H), 1.89-1.98 (m, IH), 1.42-1.80 (m, 7H). MS (ES) [m+H] calc'd for C21H25N5O 364; found 364.
Example 18: 2-[2-(3-(R)-Amino-piperidin-lyl)-6-chloro-4-oxo-4H-pyrido[3,4- α^pyrimidin-3-ylmethyl]-benzonitrile, TFA salt
Figure imgf000138_0001
Example 18A: l,7-Dmydro-pyrido[3,4-αlpyrimidine-2,4,6-trione
Figure imgf000138_0002
[0461] The title compound was prepared from 5-amino-2-hydroxypyridine-4-carboxyUc acid (see Rewcastle et al., /. Chem. Soc Perkin Trans. 1, 1996, 2221-2226) in 73% yield according to the procedure for example 3A. 1H NMR (400 MHz, DMSO-αV): δ 11.02 (br s, 3H), 7.92 (s, IH), 6.92 (s, IH).
Example 18B: 2,6-Dichloro-3H-pyrido[3,4-αlpyrimidine-4-one
Figure imgf000138_0003
[0462] The title compound was prepared from l,7-dmydro-pyrido[3,4-α^pyrimidine- 2,4,6-trione in 16% yield according to the procedures of examples IB and IC. 1H NMR (400 MHz, DMSO-αV): δ 13.70 (br s, IH), 8.84 (s, IH), 7.96 (s, IH).
Example 18C: 2-(2,6-Dichioro-4-oxo-4H-pyrido[3,4-αlpyrimidin-3-ylmethyl)- benzonitrile
Figure imgf000139_0001
[0463] The title compound was prepared from 2,6-dicWoro-3H-pyrido[3,4-αlpyrimidine- 4-one in 63% yield according to the procedure for example 2B. 1H NMR (400 MHz, CDC13): δ 8.91 (s, IH), 8.10 (s, IH), 7.75 (dd, IH, J = 7.6, 1.2 Hz), 7.56 (dt, IH, J = 7.6, 1.2 Hz), 7.45 (t, IH, J = 7.6 Hz), 7.14 (d, IH, J = 7.6 Hz), 5.74 (s, 2H). MS (ES) [m+H] calc'd for Cι5H8N4OCl2331, 333; found 331, 333.
[0464] The title compound, Example 18, 2-[2-(3-(R)-Amino-piperidin- lyl)-6-chloro-4- oxo-4H-pyrido[3,4-αlpyrin idm-3-ylmemyl]-berLzonitrile, TFA salt, was prepared from 2-(2,6- dichloro-4-oxo-4H-pyrido[3,4-α^pyrimidm-3-ylmethyl)-benzonitrile in 63% yield according to the procedure for example 17. 1H NMR (400 MHz, DMSO-αV): δ 8.77 (s, IH), 8.03 (br s, 3H), 7.80-7.84 (m, 2H), 7.61 (t, IH, J = 7.6 Hz), 7.45 (t, IH, J = 7.6 Hz), 7.34 (d, IH, J = 7.6 Hz), 5.37 (AB q, 2H, J = 44.4, 15.2 Hz), 3.41-3.60 (m, 2H), 3.20-3.27 (m, IH), 3.06-3.14 (m, IH), 2.89-2.97 (m, IH), 1.94-2.02 (m, IH), 1.79-1.87 (m, IH), 1.51-1.69 (m, 2H). MS (ES) [m+H] calc'd for C209N6OCl 395, 397; found 395, 397.
[0465] Example 19: 2-[2-(3-(R)-Amino-piρeridin-lyl)-4-oxo-6-ρynolidin-lyl-4H- pyrido [3 ,4-αlpyrimldin-3-ylmethyI] -benzonitrile
Figure imgf000140_0001
[0466] 2-[2-(3-(R)-Amino-piperidin-lyl)-6-cUoro-4-oxo-4H-pyrido[3,4-olpyrirmdin-3- ylmethylj-benzonitrile (Example 18) (120 mg, 0.3 mmol), pynoUdine (75 μL, 0.91 mmol) and sodium bicarbonate (76 mg, 0.91 mmol) were stined in EtOH in a sealed tube at 150 C for 16 h. After cooling, the reaction was diluted with EtOAc, washed with brine, dried (MgSO4), and concentrated in vacuo. Purification by preparative HPLC yielded 54 mg (32%) of the title compound as a bright orange solid. 1H NMR (400 MHz, MeOD-α ): δ 8.40 (s, IH), 7.64 (dd, IH, J = 7.6, 1.2 Hz), 7.48 (dt, IH, J = 7.6, 1.2 Hz), 7.33 (t, IH, J = 7.6 Hz), 7.07 (d, IH, J = 7.6 Hz), 6.67 (s, IH), 5.43 (s, 2H), 3.20-3.32 (m, 5H), 3.02-3.10 (m, IH), 2.81-2.90 (m, IH), 2.69-2.78 (m, IH), 2.59-2.67 (m, IH), 1.87-1.99 (m, 5H), 1.51-1.73 (m, 2H), 1.18-1.28 (m, IH). MS (ES) [m+H] calc'd for H27N7O 430; found 430.
Example 20: 2-[(R)-3-Amino-piperidin- l-yl]-6-fluoro-3-(2-trifluoromethyl- benzyl)-3H-quinazolin-4-one
Figure imgf000140_0002
Example 20A: 2-Chloro-6-fluoro-3-(2-trifluoromethyl-benzyl)-3H-quinazolin-4- one
Figure imgf000140_0003
[0467] The title compound Example 20A was prepared from 6C based on the procedures described in the synthesis of ID. Without further characterization, 20A was converted to Example 20 by the method used for Example 1.
[0468] Example 20: NMR (400 MHz CDCI3-CD3OD 10:1): β 8.56 (br s, 3H), 7.96 (s, IH), 7.78 (d, IH, J = 7.6 Hz), 7.52-7.7 (m, 5H), 5.88-5.76 (AB q, 2H, J = 12.8 Hz), 4.51 (m, IH), 4.18 (m, 2H), 3.85 (m, IH), 3.66 (m, IH), 2.21 (m, IH), 2.03 (m, 2H), 1.70 (m, IH). MS (ES) [m+H] calculated for C2ιH20FN4O+H, 421.2, found 421.1.
Example 21: 2-{2-[(R)-3-Amino-ρiperidin-l-yl]-7-isopropyl-6-oxo-6,7-dihydro- purin- 1 -ylmethyl } -benzonitrile
Figure imgf000141_0001
[0469] The title compound was prepared from Example 9G by treatment with NaH and isobutyl bromide, followed by the method used for Example 1.
[0470] Example 21: NMR (400 MHz CDCI3-CD3OD 10: 1): δ 7.88 (s, IH), 7.72 (d, IH, J = 8 Hz), 7.62 (d, IH, J = 7.6 Hz), 7.57 (t, IH, J = 7.6 Hz), 7.09 (t, IH, J = 7.6 Hz), 5.73-5.52 (AB q, 2H, J = 13.4 Hz), 4.66 (m, IH), 4.56 (d, IH, J = 13.2 Hz), 4.21 (m, IH), 3.4-3.2 (m, 3H), 2.01 (m, IH), 1.77 (m, IH), 1.66 (m, IH), 1.55 (m, IH), 1.49 (s, 3H), 1.48 (s, 3H). MS (ES) [m+H] calculated for C2ιH25N7O+H, 392.2, found 392.2.
Example 22: 2-[2-(3-Amino-azepan-l-yl)-6-oxo-6,7-dihydro-purin-l-ylmethyl]- benzonitrile
Figure imgf000141_0002
[0471] The title compound was prepared from Example 9G using the method for Example 1. [0472] Example 22: NMR (400 MHz CDCI3-CD3OD 10: 1): δ 8.47 (s, IH), 7.74 (m, 2H), 7.68 (t, IH, J = 7.6 Hz), 7.52 (t, IH, J = 7.6 Hz), 5.84-5.72 (AB q, 2H, J = 13.2 Hz), 4.17 (m, IH), 3.81 (m, IH), 3.59 (m, IH), 3.29 (m, IH), 2.21 (m, IH), 1.88 (m, 2H), 1.62 (m, IH), 1.44 (m, 2H). MS (ES) [m+H] calculated for Cι9H21N7O+H, 363.2, found 363.3.
Example 23: 2-{2-[(R)-3-Aniino-piperidin-l-yl]-7-benzyl-6-oxo-6-hydro-purin-l- ylmethyl } -benzonitrile
Figure imgf000142_0001
[0473] The title compound was prepared from Example 9G by treatment with NaH and benzyl bromide, followed by the method used for Example 1.
[0474] Example 23: NMR (400 MHz CDCI3-CD3OD 10: 1): β 7.88 (d, IH, J = 7.6 Hz), 7.80-7.70 (m, 2H), 7.67-7.55 (m, 2H), 7.45 (t, IH, J = 7.6 Hz), 7.22 (d, IH, J = 8 Hz), 5.60- 5.42 (m,4H), 3.6-3.45 (m, 2H), 3.23-3.10 (m, 2H), 2.94 (m, lH), 2.12 (m, IH), 1.90-1.70 (m, 2H), 1.6 (m, IH), 1.62 (m, IH), 1.44 (m, 2H). MS (ES) [m+H] calculated for C25H25N7O+H, 440.2, found 440.1.
Example 24: 2- { 2-[(R)-3-Amino-piperidin- 1 -yl]-9-(2-cyano-benzyl)-6-oxo-6- hydro-purin- 1 -ylmethyl }
Figure imgf000142_0002
[0475] The title compound was prepared from Example 9G by treatment with NaH and δ-cyano-benzyl bromide and the procedure used for Example 1.
[0476] Example 24: NMR (400 MHz CDCI3-CD3OD 10: 1): β 7.86 (s, IH), 7.74 (d, IH,
J = 8 Hz), 7.68-7.60 (m, 2H), 7.55-7.45 (m, 3H), 7.39-7.20 (m, 2H), 5.40-5.50 (m, 4H), 3.58 (d, IH, J = 12.8 Hz), 3.52, (m, IH), 3.21 (dd, IH, J = 8.8 and 12 Hz), 3.06 (m, 2H), 2.10 (m, IH), 1.87 (m, IH), 1.70 (m, 2H). MS (ES) [m+H] calculated for C26H24N8O+H, 464.2, found 464.1.
Example 25 : 2- { 2- [(R)-3- Amino-piperidin- 1 -yl] -6-oxo-r9-propyl-6,9-dihydro- purin- 1 -ylmethyl } -benzonitrile
Figure imgf000143_0001
Example 26: 2-{2-[(R)-3-Amino-piperidin-l-yl]-6-oxo-7-propyl-6,7-dihydro- purin- 1 -ylmethyl } -benzonitrile
Figure imgf000143_0002
Example 25A: 2-CUoro-9-propyl-l,9-dmycu-o-purin-6-one
Figure imgf000143_0003
Example 26A: 2-Chloro-7-propyl-l,7-dihydro-purin-6-one
Figure imgf000143_0004
[0477] Compounds 25A and 26A were prepared from 2,6-dichloro-7H-purine by the methods used for Example 7B and Example 8B.
[0478] Example 25A: NMR (400 MHz CDC13): β 7.96 (s, IH), 4.14 (t, 2H, J = 7.2 Hz),
1.91 (m, 2H), 0.97 (t, 3H, J = 7.6 Hz). MS (ES) [m+H] calculated for C8H9ClN O+H, 213.1, found 213.1.
[0479] Example 26A: NMR (400 MHz CDC13): 88.33 (s, IH), 4.37 (t, 2H, J = 6.8 Hz),
1.95 (m, 2H), 0.96 (t, 3H, J = 7.6 Hz). MS (ES) [m+H] calculated for C8H9ClN4O+H, 213.1, found 213.1.
Example 25B: 2-(2-Chloro-6-oxo-9-propyl-6,9-dihydro-purin-l-ylmethyl)- benzonitrile
Figure imgf000144_0001
[0480] The title compound Example 25B was prepared from 25A by the method used for
ID.
[0481] Example 25B: NMR (400 MHz CDCI3-CD3OD 10: 1): β 7.98 (s, IH), 7.73 (d,
IH, J = 8 Hz), 7.55 (t, IH, J = 7.6 Hz), 7.43 (t, IH, J = 7.6 Hz), 7.14 (d, IH, J = 8 Hz), 4.17 (t,
2H, J = 7.2 Hz), 1.94 (m, 2H), 1.00 (t, 3H, J = 7.2 Hz). MS (ES) [m+H] calculated for
C164ClN5O+H, 328.1, found 328.1.
Example 26B: 2-(2-Chloro-6-oxo-7-propyl-6,7-dihydro-purin- 1 -ylmethyl)- benzonitrile
Figure imgf000144_0002
[0482] The title compound Example 26B was prepared from 26A by the method used for
ID. [0483] Example 26B: NMR (400 MHz CDCI3-CD3OD 10: 1): § 8.1 (s, IH), 7.76 (d, IH,
J = 8 Hz), 7.58 (t, IH, J = 7.6 Hz), 7.45 (t, IH, J = 7.6 Hz), 7.10(d, IH, J = 8 Hz), 4.37 (t, 2H,
J = 6.4 Hz), 1.94 (m, 2H), 0.97 (t, 3H, J = 6.8Hz). MS (ES) [m+H] calculated for
64ClN5O+H, 328.1, found 328.1.
[0484] The title compound, Example 25, was prepared from Example 25B by the method used for Example 1.
[0485]
[0486] Example 25: NMR (400 MHz CDCI3-CD3OD 10: 1): 88.06 (s, IH), 7.64 (d, IH,
J = 8 Hz), 7.55 (t, IH, J = 7.6 Hz), 7.37 (t, IH, J = 8 Hz), 7.22 (t, IH, J = 7.6 Hz), 5.53-5.37
(AB q, 2H, J = 15.2 Hz), 4.14 (t, IH, J = 7.2 Hz), 3.62 (d, IH, J = 12.4Hz), 3.51 (m, IH), 3.19
(dd, IH, J = 9.2 and 12.4 Hz), 3.12 (d, IH, J = 12.4 Hz), 2.94 (m, IH), 2.12 (m, IH), 1.90 (m,
3H), 1.72 (m, IH), 0.97 (t, 6H, J =7.6 Hz). MS (ES) [m+H] calculated for C2ιH25N7O+H,
392.2, found 392.2.
[0487] The title compound of Example 26 was prepared from Example 26B by the method used for Example 1.
[0488] Example 26: NMR (400 MHz CDCI3-CD3OD 10: 1): 87.92 (s, IH), 7.69 (d, IH,
J = 8 Hz), 7.55 (t, IH, J = 7.6 Hz), 7.41 (t, IH, J = 8 Hz), 7.14 (t, IH, J = 7.6 Hz), 5.60-5.37
(AB q, 2H, J = 15.6 Hz), 4.29 (t, IH, J = 6.4 Hz), 3.54 (m, 2H), 3.19 (m, 2H), 2.95 (m, IH),
2.11 (m, IH), 1.88 (m, 3H), 1.70 (m, IH), 0.92 (t, 6H, J = 7.2 Hz). MS (ES) [m+H] calculated for C2ιH25N7O+H, 392.2, found 392.2.
Scheme for the Preparation of Example 27
Figure imgf000146_0001
Figure imgf000146_0002
27A 27B
Figure imgf000146_0003
Example 27: 2-{2-[(R)-(3-Amino-piperidin-l-yl)]-9-isopropyl-6-oxo-8- ttifluoromemyl-6,9-dihydro-ρurin- 1 -ylmethyl } -benzonitrile
Figure imgf000146_0004
Example 27A: 6-Chloro-N4-isoρropyl-pyrimidine-2,4,5-triarnine
Figure imgf000147_0001
[0489] Example 27A was prepared from 4,6-Dichloro-pyrirnidin-2-ylamine by the procedures described in J. Het. Chem., 1990, 27, 1409.
Example 27B: 6-CMoro-9-isopropyl-8-tiifluoromethyl-9H-purin-2-ylamine
Figure imgf000147_0002
[0490] To a mixture of 28A (lg, 5.0 mmol) and Et3N in DMF (5 mL) was added a solution of trifluoroacetic anhydride in DMF (ImL) dropwise. After stirring at room temperature for 30 min, the mixture was heated at 120 °C overnight, then further heated at 160
C for 5h. After cooling, the residue was suspended in hot water, and then cooled and filtered.
The crude product was suspended in hot MeOH-water, cooled and filtered to give the title compound (1.2 g). MS (ES) [m+H] calculated for C9H10F3N5O+H, 262.1, found 262.2.
Example 27C : 2-(2-Amino-9-isopropyl-6-oxo-8-trifluoromethyl-6,9-dihydro- purin- 1 -ylmethyl)-benzonitrile
[0491] The title compound was prepared by converting Example 27B under the condition used for ID.
[0492] Example 27C: NMR (400 MHz CDCI3-CD3OD 10:1): β 7.71 (d, IH, J = 8.0 Hz), 7.57 (t, IH, J = 8 Hz), 7.42 (t, IH, J = 7.2 Hz), 7.20 (d, IH, J = 8 Hz), 5.53 (s, 2H), 4.70 (m, IH), 1.67 (d, 6H, J = 6.8 Hz). MS (ES) [m+H] calculated for Cι75F3N6O+H, 377.1, found 377.1. Example 27D: 2-(2-BromoL9-isopropyl-6-oxo-8-trMuoromemyl-6,9-dihydro- purin- 1 -ylmethyl)-benzonitrile
Figure imgf000148_0001
[0493] To a hot (60 C) mixture of Example 27C (100 mg), Br2 (50 μL) and CuBr (60 mg) in CHCI3 (4 mL), was added dropwise a solution of Br2 (150 μL) in CHCI3 (200 μL) and tert-butyl nitrite (300 μL) at the same time. The mixture was refluxed for 1 h, and then diluted with CH2C12 and washed with 10% Na2S2O3. The organic layer was dried and concentrated to give crude product (90 mg).
[0494] Example 27E: NMR (400 MHz CDCI3-CD3OD 10: 1): β 7.73 (d, IH, J = 7.6 Hz), 7.56 (t, IH, J = 7.2 Hz), 7.44 (t, IH, J = 7.2 Hz), 7.11 (d, IH, J = 7.6 Hz), 5.81 (s, 2H), 4.84 (m, IH), 1.73 (d, 6H, J = 6.8 Hz). MS (ES) [m+H] calculated for Cι73BrF3N5O+H, 440.0, found 440.2.
[0495] The title compound Example 27, 2-{2-[(R)-(3-Amino-ρiρeridin-l-yl)]-9- isopropyl-6-oxo-8-trMuoromethyl-6,9-dihydro-purin- 1 -ylmethyl } -benzonitrile, was synthesized from Example 27E by the method used for Example 1. [0496] Example 27: NMR (400 MHz CDCI3-CD3OD 10: 1): β 7.64-7.50 (m, 2H), 7.40- 7.26 (m, 2H), 7.44 (t, IH, J = 7.2 Hz), 7.11 (d, IH, J = 7.6 Hz), 5.52-5.30 (AB q, 2H, J = 15.8 Hz), 4.76 (m, IH), 3.7-3.50 (m, 2H), 3.18, (m, 2H), 2.94 (t, IH, J = 10.0 Hz), 2.13 (m, IH), 1.90 (m, IH), 1.80-1.62 (m, 8H). MS (ES) [m+H] calculated for C^H^^O+H, 460.2, found 460.3.
Example 28: 2-[2-(3-(R)-Amino-piperidin- l-yl)-6-bromo-4-oxo-4H-quinazolin-3- ylmethyl] -benzonitrile
Figure imgf000149_0001
Example 28A: 6-Bromo-lH-quinazoline-2,4-dione
Figure imgf000149_0002
[0497] The title compound was prepared from Methyl 2-amino-5-bromo-benzoate in 90% yield according to the procedure for example 3A. MS: (ES) M+H calc'd for C8HsBrN2O2, 240, 242; found 240, 242.
Example 28B: 6-Bromo-2-chloro-3H-quinazolin-4-one
Figure imgf000149_0003
[0498] The title compound was prepared from 28A according to the procedures of examples IB and IC. MS: (ES) M+H calc'd for C8H4BrClN20, 260; found 260.
Example 28C: 2-(6-Bromo-2-chloro-4-oxo-4H-quinazolin-3-ylmethyl)- benzonitrile
Figure imgf000149_0004
[0499] The title compound was prepared from 28C as a mixture of N- and O- alkylation products according to the procedure for ID. MS: (ES) M+H calc'd for Cι6H9BrClN30, 375; found 375. [0500] Example 28 was prepared from 28C according to the procedure for compound 1. 1H NMR (400 MHz, CDC13): δ 8.23 (d, J = 1.77 Hz, IH), 7.77 (d, J = 6.82 Hz, IH), 7.61 (d, J = 7.33 Hz, IH), 7.55 (dd, J = 7.58, 7.07 Hz, IH), 7.43 - 7.49 (m, IH), 7.31 - 7.41 (m, 2H), 5.44 (AB q, J = 137.18, 14.91 Hz, 2H), 3.48 - 3.81 (m, 3H), 3.18 - 3.34 (m, 2H), 1.83 - 2.14 (m, 3H), 1.64 - 1.76 (m, IH). MS: (ES) M+H calc'd for C2ιH20BrCN50, 438; found 438.
Example 29: 2-[2-(3-(R)-Amino-pynolidin-l-yl)-6-bromo-4-oxo-4H-quinazolin- 3-ylmethyl]-benzonitrile, TFA salt
Figure imgf000150_0001
[0501] The title compound was prepared from 28C according to the procedure for compound 1. 1H NMR (400 MHz, DMSO-D6): δ 7.97 (d, J = 2.27 Hz, IH), 7.85 (d, J = 7.58 Hz, IH), 7.78 (dd, J = 8.59, 2.53 Hz, IH), 7.62 (dd, J = 6.82, 6.82 Hz, IH), 7.46 (dd, J = 7.58, 7.07 Hz, IH), 7.33 (d, J = 8.84 Hz, IH), 7.27 (d, J = 7.83 Hz, IH), 5.33 (s, 2H), 3.56 (m, 2H), 3.43 (m, 2H), 3.06 (m, IH), 1.88 (m, 2H), 1.57 (m, IH). MS: (ES) [m+H] calculated for C2oHι8BrCN50, 424, 426; found 424, 426.
Example 30: 2-[2-(3-(R)-Amino-piperidin-l-yl)-6,8-dichloro-4-oxo-4H- quinazolin-3-ylmethyl]-benzonitrile
Figure imgf000150_0002
Example 30A: 6,8-Dichloro-lH-quinazoline-2,4-dione
Figure imgf000150_0003
[0502] 2-Amino-3,5-dichlorobenzoic acid (lg, 4.85 mmol) and urea (1 g, 16.7 mmol) were heated together at 200 °C for 1 hour. The mixture was cooled and triturated with water. The solid was filtered and dried to give 30A (0.9 mg, green solid, 80%). This material was used in the next step without further purification. MS: (ES) M+H calc'd for C8H4Cl2N2O2, 230; found 230.
Example 30B: 2,6,8-Trichloro-3H-quinazolin-4-one
Figure imgf000151_0001
[0503] The title compound was prepared from 6,8-άicMoro-lH-qumazoline-2,4-dione in 69% yield according to the procedures of examples IB and IC. MS: (ES) M+H calc'd for C8H3Cl3N20, 250; found 250.
Example 30C: 2-(2,6,8-Trichloro-4-oxo-4H-quinazolin-3-ylmethyl)-benzonitrile
Figure imgf000151_0002
[0504] To a stined solution of 30B (400 mg, 1.6 mmol) in DME (4 mL) and DMF (1 mL) at 0 °C was added NaH (43 mg, 1.8 mmol, 95%). After ten minutes, LiBr (280 mg, 3.2 mmol) was added and the mixture was allowed to warm to RT. After 15 minutes, α- bromo-o- tolunitrile (350 mg, 1.8 mmol) was added and the mixture was heated at 65 °C overnight.
After cooling, water (10 mL) was added. A precipitate formed. This precipitate was filtered and dried to give 30C which was not further purified. MS (ES) [m+H] calculated for
6H8Cl3N3O, 363; found 363.
[0505] A mixture of 30C (92 mg, 0.25 mmol), 3-aminopiperidine dihydrochloride (66 mg, 0.38 mmol), NaHCO3 (63 mg, 0.75 mmol) and 2 mL of ethanol in a sealed tube was heated to 150 °C for 6 hours. After cooling to room temperature and filtering the inorganic salts, purification via LC MS afforded 55 mg (51 % yield) of product 30. 1H NMR (400 MHz, MeOD): δ 7.93 (d, J = 2.53 Hz, IH), 7.88 (d, J = 2.53 Hz, IH), 7.71 (dd, J = 7.58, 1.01 Hz, IH), 7.61 (ddd, J = 7.58, 7.58, 1.26 Hz, IH), 7.44 (dd, J = 7.58, 2.4 Hz, IH), 7.39 (d, J = 7.83 Hz, IH), 5.47 (AB q, J = 34.86, 15.16 Hz, 2H), 3.61 - 3.80 (m, 2H), 3.34 - 3.42 (m, IH), 3.24 - 3.27 (m, IH), 3.10 - 3.19 (m, IH), 2.10 - 2.20 (m, IH), 1.64 - 1.90 (m, 3H). MS: (ES) [m+H] calculated for C2ιH19Cl2N50, 428; found 428.
Example 31: 2-[2-(3-(R)-Amino-piperidin-l-yl)-6-methoxy-4-oxo-4H-quinazolin- 3-ylmethyl] -benzonitrile
Figure imgf000152_0001
Example 31A: 6-Methoxy-lH-quinazoline-2,4-dione
Figure imgf000152_0002
[0506] 2-Amino-5-methoxybenzoic acid (2g, 12 mmol) and urea (2.2 g, 36 mmol) were heated together at 200 °C for 1 hour. The mixture was cooled and triturated with water. The solid was filtered and dried to give 31A (2.1 g, green solid, 90%). This material was used in the next step without further purification. MS: (ES) [m+H] calculated for C9H8N2θ3, 193; found 193.
Example 31B: 2,4-Dichloro-6-methoxy-quinazoline
Figure imgf000152_0003
[0507] To 2.1 g of 31A in 10 mL POCl3 was added 0.5 mL N,N-dimethylaniline. The mixture was then heated at reflux for 16 hours. Excess POCI3 was removed in vacuo and the residue was purified by column chromatography (Hexane: Ethyl acetate = 4:1), providing crude 1.8 g of product 31B. MS (ES) [m+H] calculated for C9H6Cl2N20, 230; found 230.
Example 31C: 2-Chloro-6-methoxy-3H-quinazolin-4-one
Figure imgf000153_0001
[0508] The title compound was prepared from 31C in 80% yield according to the procedure for example IC. MS: (ES) [m+H] calculated for C9H7ClN2O2, 211; found 211.
Example 31D: 2-(2-Chloro-6-memoxy-4-oxo-4H-quinazoιin-3-ylmethyl)- benzonitrile
Figure imgf000153_0002
[0509] The title compound was prepared from 31C in 91% yield according to the procedure for example 2B. MS (ES) [m+H] calculated for C172ClN3O2, 326; found 326. [0510] A mixture of 31 (99 mg, 0.3 mmol), 3-aminopiperidine dihydrochloride (80 mg, 0.46 mmol), NaHCO3 (76 mg, 0.9 mmol) and 2 mL of ethanol in a sealed tube was heated to 120 °C for 6 hours. After cooling to room temperature and filtering the inorganic salts, purification via LC/MS afforded 38 mg (44% yield) of product 31. 1H NMR (400 MHz, CDCI3): δ 7.53 - 7.68 (m, 3H), 7.32 - 7.47 (m, 3H), 7.25 - 7.30 (m, IH), 5.42 (AB q, J = 72.76, 14.65 Hz, 2H), 3.84 - 3.94 (m, IH), 3.65 - 3.79 (m, 2H), 3.33 - 3.50 (m, 2H), 2.10 - 2.23 (m, IH), 1.91 - 2.05 (m, 2H), 1.70 - 1.82 (m, IH). MS: (ES) [m+H] calculated for C22H23N5O2, 390; found 390. Example 32: 2-[2-(3-(R)-Ajnino-piρeridin- 1 -yl)-6-fluoro-4-oxo-4H-quinazolin-3- ylmethyl]-benzamide
Figure imgf000154_0001
[0511] To a solution of 2-[2-(3-amino-piperidin- 1 -yl)-6-fluoro-4-oxo-4H-quinazolin-3- ylmethyl]-benzonitrile (Example 6, 50 mg) in 5 mL of THF was added 2 mL of IN NaOH, followed by 2 mL H2O2. After stirring at room temperature overnight, the mixture was acidified with concentrated HCl. Removal of the solvent and purification by Preparative LC MS provided the title compound. 1H NMR (400 MHz, MeOD): δ 7.68 - 7.74 (m, IH), 7. 59 - 7.66 (m, IH), 7.48 - 7.58 (m, 2H), 7.29 - 7.40 (m, 2H), 7.02 (d, J = 6.57 Hz, IH), 5.58 (AB q, J = 22.74, 15.66 Hz, 2H), 3.49 - 3.59 (m, 2H), 3.17 - 3.26 (m, IH), 3.01 - 3.10 (m, 2H), 1.64 - 2.09 (m, 4H). MS: (ES) [m+H] calculated for C2ιH22FN5O2, 396; found 396.
Example 33: 2-[2-(3-(R)-Amino-piperidin-l-yl)-6-fluoro-7-morpholin-4-yl-4- oxo-4H-quinazolin-3-ylmethyl]-benzonitrile
Figure imgf000154_0002
Example 33A: 6,7-Difluoro-lH-quinazoline-2,4-dione
Figure imgf000154_0003
[0512] 2-Amino-4,5-difluorobenzoic acid (4g, 23 mmol) and urea (4.2 g, 69 mmol) were heated together at 200 °C for 1 hour. The mixture was cooled and triturated with water. The solid was filtered and dried to give 33A (4.1 g, green solid, 90%). This material was used in the next step without further purification.
Example 33B: 6-Fluoro-7-morpholin-4-yl-lH-quinazohne-2,4-dione
Figure imgf000155_0001
[0513] A mixture of 33A ( 1 g, 5.1 mmol) and 2mL of morpholine in 5 mL of DMS O was stined at 90 C for 2 hours. The mixture was diluted with water and acidified with concentrated HCl. The solid product was filtered out and dried under vacuum to give lg (yield 74%) of product 33B. MS: (ES) [m+H] calculated for C12H12FN3O3, 266; found 266.
Example 33C: 2,4-Dichloro-6-fluoro-7-morphohn-4-yl-quinazoline
Figure imgf000155_0002
[0514] To 1 g of 33B in 10 mL POCl3 was added 0.5 mL N,N-dimethylaniline. The mixture was then heated at reflux for 16 hours. Excess POCI3 was removed in vacuo, and the residue was purified by column chromatography (Hexane: Ethyl acetate = 4: 1), providing 0.38 g of product 33C. MS: (ES) [m+H] calculated for C12H10CI2FN30, 302; found 302.
Example 33D: 2-CUoro-6-fluoro-7-morpholin-4-yl-3H-quinazolin-4-one
Figure imgf000155_0003
[0515] A mixture of 5 mL of IN NaOH, 10 mL of THF, and 0.38 g of 33C was stined at room temperature under N2 overnight. The solution was acidified with HCl. The solids that precipitated were filtered to give 0.1 g (27%) of product 33D. MS: (ES) [m+H] calculated for Ci2HnClFN3O2, 384; found 384.
Example 33E: 2-(2-Chloro-6-fluoro-7-morpholin-4-yl-4-oxo-4H-quinazolin-3- ylmethyl)-benzonitrile
Figure imgf000156_0001
[0516] To a stined solution of 33D (100 mg, 0.35 mmol) in DME (2 mL) and DMF (0.5 mL) at 0 °C was added NaH (9.6 mg, 0.4 mmol). After ten minutes, LiBr (61 mg, 0.7 mmol) was added and the mixture was allowed to warm to RT. After 15 minutes, α- bromo-o- tolunitrile (76.4 mg, 0.39 mmol) was added and the mixture was heated at 65 °C overnight. After cooling, water (10 mL) was added. A precipitate formed. This precipitate was filtered and dried to give 33E which was not further purified. MS: (ES) [m+H] calculated for C20H16C1FN4O2, 399; found 399.
[0517] A mixture of 50 mg of crude 33E, 2 eq. of 3-aminopiperidine dihydrochloride, 5 eq. of NaHCO3, and 2 mL of ethanol in a sealed tube was heated to 150 °C for 6 hours. After cooling to room temperature and filtering the inorganic salts, purification viaLC MS afforded 28 mg (47% yield) of product 33. 1H NMR (400 MHz, MeOD): δ 7.71 (d, J = 8.0 Hz, IH), 7.55-7.62 (m, 2H), 7.43 (dd, J = 8.0, 7.2 Hz, IH), 7.29 (d, J = 8.0 Hz, IH), 7.07 (d. J = 8.0Hz, IH), 5.49 (AB q, J = 15.2, 34.8 Hz, 2H), 3.82-3.90 (m, 4H), 3.49-3.65 (m, 2H), 3.15-3.27 (m, 6H), 2.92-3.02 (m, IH), 2.09-2.17 (m, IH), 1.60-1.90 (m, 3H). MS: (ES) [m+H] calculated for C257FN6O2, 463; found 463.
Example 34: 2-[2-(3-Amino-piperidin- 1 -yl)-6-fluoro-4-oxo-4H-quinazolin-3- ylmethyl]-benzamide
Figure imgf000157_0001
[0518] To a solution of 6E in THF was added H2O2 (30%, 5mL) and IN NaOH (5mL). The mixture was stined at room temperature for 4 hours, neutralized by HCl and extracted with DCM. The organics were dried over MgSO and the solvent removed in vacuo. The residue was purified by HPLC. 1H NMR (400 MHz CD3OD): β7.74-7.48 (m, 4H), 7.28-7.39 (m, 2H), 7.02 (d, J=6.8Hz, IH), 5.52-5.64 (AB q, J = 15.6, 22.8 Hz, 2H), 3.49-3.59 (m, 2H), 3.18-3.26 (m, IH), 2.99-3.09 (m, 2H), 1.65-2.09 (m, 4H). MS (ES) [m+H] calculated for C2ιH22FN5O2, 395; found 395.
Example 35: 2-[3-(R)-Arnino-piperidin-l-yl]-6-fluoro-3-(2-trifluoromethyl- benzyl)-3H-quinazolin-4-one
Figure imgf000157_0002
[0519] Example 35 was synthesized from 2-Chloro-6-fluoro-3H-quinazolin-4-one (example 6C) by two steps, benzylation and amine replacement. The procedure for benzylation was described in the synthesis of example 2B, and the procedure for amine replacement was described in the synthesis of example 1. 1H-NMR (400 MHz, CDCI3- CD3OD, 10:1), δ 8.56 (s, 2 H), 7.96 (d, 7=5.3 Hz, 1 H), 7.78 (d, 7=7.8 Hz, 1 H), 7.56 - 7.67 (m, 5 H), 5.77 - 5.88 (ABq, 7= 10.6 and 23.5 Hz, 2 H), 4.53 (brs, 1 H), 4.18 (s, 2 H), 3.86 (s, 1 H), 3.66 (s, 1 H), 2.21 (s, 1 H), 2.03 (s, 2 H), 1.71 (s, 1 H). MS (ES) [m+H] calc'd for C21H.1F4N40, 421.2; found, 421.2.
Figure imgf000158_0001
Example 36: 2-Aminomethyl-3-phenyl-3H-quinazolin-4-one
Figure imgf000158_0002
[0520] To a solution of 2-Amino-N-ρhenyl-benzamide, 36A, (500 mg, 2.5 mmol) in THF (10 mL), was added bromo-acetyl bromide (2mL). The reaction was carried out at rt for 0.5 h, and then 50 °C for 2h, then concentrated in vacuo. The residue was dissolved in CH2C12, washed with aq NaHCO3, dried over Na2SO4, and concentrated to give the crude product 36B that was used for the next step without further purification.
[0521] A mixture of compound 36B (100 mg, 0.32 mmol) and potassium phthalimide (120 mg, 0.64 mmol) in dry DMF (3 mL) was heated at 100 °C for 5 h, then concentrated in vacuo. The residue was dissolved in CH2C12 washed with water, dried and concentrated to give crude product 36C. Compound 36C in MeOH (5 mL), was treated with NH2NH2 at rt for 2h, 50 °C for 5 min, and then dried in vacuo, co-evaporated with water, and purified by LC- MS to give compound 36. 1H-NMR (400 MHz, CDC13) δ 8.30 (d, 7=7.8 Hz, 1 H), 7.73 - 7.82 (m, 2 H), 7.48 - 7.59 (m, 4 H), 7.22 - 7.28 (m, 2 H), 3.49 (s, 2H). MS (ES) [m+H] calc'd for
Figure imgf000159_0001
252; found, 252.
Example 37: 2-Emylaminomethyl-3-phenyl-3H-quinazolin-4-one
Figure imgf000159_0002
[0522] A solution of compound 36B (lOOmg) in THF (0.5 mL) was added to a stirring solution of 2 M EtNH2 in THF (2 mL). The mixture was stined at rt for 2h, and then purified by LC-MS to give compound example 37. 1H-NMR (400 MHz, CD3C1) δ ppm 8.21 (dd, 7=8.1 and 1.5 Hz, 1 H) 7.80 (dt, 7=7.1 and 1.2 Hz, 1 H), 7.72 (d, 7= 7.8 Hz, IH), 7.57-7.49 (m, 4 H) 7.28 (m, 2 H) 3.80 (s, 2 H) 3.08 (q, 7=7.3 Hz, 2 H) 1.30 (t, 7=7.2 Hz, 3 H). MS (ES) [m+H] calc'd for d78N30, 280; found, 280.
Example 38: [(4-Oxo-3-phenyl-3 ,4-dihydro-quinazolin-2-ylmethyl)-amino]-acetic acid methyl ester
Figure imgf000159_0003
[0523] To a pre-stined mixture of glycine methyl ester hydrochloride (300 mg, 2.4 mmol) and NaHCO3 (250 mg, 2.6 mmol) in DMF (3 mL) was added a solution of compound 36B (80 mg, 0.25 mmol) in DMF (ImL). The mixture was stined at rt for 2h, and purified by LC-MS to give compound example 38. 1H-NMR (400 MHz, CD3OD) δ 8.22 (dd, 7=8.0, 1.4 Hz, 1 H) 7.77 (dt, 7=7.6 and 1.1 Hz, 1 H) 7.72 (d, J= 7.6 Hz, 1 H) 7.46 - 7.56 (m, 4 H) 7.25 (m, 2 H) 3.61 (s, 3 H) 3.44 (s, 2 H) 3.41 (s, 2 H). MS (ES) [m+H] calc'd for Cι88N3O3, 324; found, 324. Example 39: [(4-Oxo-3-phenyl-3,4-dmydro-qu azolin-2-ylmethyl)-amino]-acetic acid
Figure imgf000160_0001
[0524] Compound 38 (45 mg, 0.14 mmol) in THF (3 ml) was treated with 2M aq LiOH (350 μL) at rt overnight. The residue was purified by LC-MS to give example 39. 1H-NMR (400 MHz, MeOD) δ 8.24 (d, 7=7.8 Hz, 1 H), 7.80 (t, 7= 8.0 Hz, 1 H), 7.74 (d, 7= 7.9 Hz, IH), 7.50 - 7.60 (m, 4 H), 7.26 - 7.34 (m, 3 H), 3.87-3.90 (4H). MS (ES) [m+H] calc'd for Cι7Hi6N3O3, 310; found, 310.
Example 40:
Figure imgf000160_0002
[0525] 2,4-dichloro-aniline (322 mg, 2.0 mmol) in DMF was treated with 95% NaH (53 mg, 2.1 mmol) at 0°C, and then 2-nitro-benzoyl chloride was added (390 mg, 2.1 mmol). After stirring for 2 h at rt, the mixture was concentrated and co-evaporated with water, diluted with CH2C12, washed with aq. NaHCO3, dried and concentrated. The crude residue was dissolved in EtOH, and subjected to hydrogenation at 1 atm overnight, filtered through Celite and concentrated to give the crude product 2- amino-benzamide.
[0526] The crude product was converted to the title compound using the procedures described in the synthesis of example 36 to give example 40. 1H-NMR (400 MHz, CDCI3) δ ppm 8.21 (d, 7=7.8 Hz, 1 H) 7.69 (t, 7=7.5Hz, 1 H) 7.59 (d, 7=7.6 Hz, 1 H) 7.55 (s, 1 H) 7.48 (t, 7=7.5 Hz, 1 H) 7.38 (d, 7=8.1 Hz, 1 H) 7.22 (d, 7=8.6 Hz, 1 H) 3.74-3.58 (ABq, 7=15.2 and 63.4 Hz, 2 H). MS (ES) [m+H] calc'd for C152Cl2N30, 320; found, 320.
Example 41: 2-Anιinomethyl-3-(2-chloro-phenyl)-3H-quinazolin-4-one
Figure imgf000161_0001
[0527] Example 41 was synthesized through the procedures described in the synthesis of example 40. 1H-NMR (400 MHz, DMSO-D6) δ 8.52 (s, 2 H) 8.21 (d, 7=7.3 Hz, 1 H) 7.98 (t, 7=7.3 Hz, 1 H) 7.81 (t, 7=7.1 Hz, 2 H) 7.76 (d, 7=6.3 Hz, 1 H) 7.66 (m, 3 H) 3.84 (d, 7=17.2 Hz, 1 H), 3.43 (d 7=17.2 Hz, 1 H). MS (ES) [m+H] calc'd for C15H13CIN30, 286; found, 286.
Example 42: 2-Aminomethyl-3-(4-chloro-ρhenyl)-3H-quinazohn-4-one
Figure imgf000161_0002
[0528] Example 42 was synthesized through the procedures described in the synthesis of example 40. 1H-NMR (400 MHz, DMSO-D6) δ ppm 8.45 (s, 2 H) 8.18 (d, 7=7.8 Hz, 1 H) 7.95 (t, 7=7.6 Hz, 1 H) 7.76 (d, 7=8.1 Hz, 1 H) 7.71 (d, 7=8.1 Hz, 2 H) 7.63 (t, 7=7.3 Hz, 1 H) 7.57 (d, 7=8.3 Hz, 2 H) 3.69 (s, 2 H). MS (ES) [m+H] calc'd for C15H13CIN30, 286; found, 286.
Example 43: 2-(3-Amino-piperidin- 1 -yl)-6,7-dimethoxy-3-(2-nitro-benzyl)-3H- quinazolin-4-one
Figure imgf000161_0003
[0529] 2A (242 mg, 1.01 mmol) was converted to 2-Chloro-6,7-dimethoxy-3-(2-niuO- benzyl)-3H-quinazolin-4-one by the method used for Example 2B using 2-nitrobenzylchloride in place of α-bromo-o-tolunitrile. The crude product was not further purified, but was converted to example 43 by the method used for example ID. 1HNMR (400MHzCDCi3): δ , 8.12 (dd, J=1.2, 8.1 Hz, IH), 7.55 (s, IH), 7.52 (m, IH), 7.44 (t, J = 8.1 Hz, IH), 7.03 (s, IH), 7.01 (m, IH), 5.73 (s, 2H), 4.03 (s, 3H), 3.98 (s, 3H), 3.24 (m, IH), 3.12 (m, IH), 2.89-2.79 (m, 2H), 2.64 (dd, J=9, 11.7 Hz, IH), 1.91 (m, IH), 1.73 (m, IH), 1.29-1.21 (m, 2H). MS (ES) [m+H] calculated for C22H26N5O5, 440; found 440.
Example 44: 2-[2-(3-Amino-piperidin- 1 -yl)-6,7-dimethoxy-4-oxo-4H-quinazolin- 3-ylmethyι]~benzoic acid ethyl ester
Figure imgf000162_0001
[0530] Example 2A was converted to 2-Chloro-6,7-dimethoxy-4-oxo-4H-quinazolin-3- ylmethyl)-benzoic acid ethyl ester using the procedure described for example 2B, except that ethyl 2-bromomethylbenzoate [7115-91-5] was used in place of α-bromo-o-tolunitrile. The crude product was not further purified but was converted to example 44 using the method described for example 1. 1H NMR (400 MHz DMSO): β 8.31 (m, 3H), 7.89 (d, J=7.0 Hz, IH), 7.47 (m, IH), 7.35 (m, 2H), 7.06 (s, IH), 6.82 (d, J=6.7 Hz, IH), 5.47-5.66 (AB q, J = 16.4, 59.5 Hz, 2H), 4.35 (m, 2H), 3.92 (s, 3H), 3.83 (s, 3H), 3.50 (m, IH), 2.99-3.11 (m, 3H), 2.63 (m, IH), 1.97 (m, IH), 1.69 (m, IH), 1.45 (m, 2H), 1.35 (t, J=6.7 Hz, 3H). MS (ES) [m+H] calculated for C25H3ιN4O5, 467; found 467.
Example 45: 2-[2-(3-Amino-piperidin-l-yl)-6-fluoro-4-oxo-4H-quinazolin-3- ylmethyTJ-benzoic acid ethyl ester
Figure imgf000163_0001
[0531] Example 6C was converted to 2-(2-Chloro-6-fluoro-4-oxo-4H-quinazolin-3- ylmethyl)-benzoic acid ethyl ester using the procedure described for example 2B, except that ethyl 2-bromomethylbenzoate [7115-91-5] was used in place of α-bromo-o-tolunitrile. The crude product was not further purified but was converted to example 45 using the method described for example 1. 1H NMR (400 MHz CDC13): β 8.03 (dd, J=1.2, 7.7 Hz, IH), 7.84 (dd, J=3.0, 8.4 Hz, IH), 7.61 (dd, J=4.7, 9.0 Hz, IH), 7.44 (m, 2H), 7.34 (m, IH), 6.91 (d, J=7.7 Hz, IH), 5.77 (AB q, J = 17.1, 24.0 Hz, 2H), 4.41 (q, J=7.2 Hz, 2H), 3.34 (m, IH), 3.23 (m, IH), 3.09 (m, 2H), 2.98 (m, IH), 1.89 (m, IH), 1.72-1.78 (m, 2H), 1.57 (m, IH), 1.43 (t, J=7.1 Hz, 3H). MS (ES) [m+H] calculated for C23H26FN4O3, 425; found 425.
Example 46: 2-[2-(3-Amino-piperidin-l-yl)-6,7-dimethoxy-4-oxo-4H-quinazolin- 3 -ylmethyl] -benzoic acid
Figure imgf000163_0002
[0532] To a stined solution of example 44 (HCl salt, 87 mg, 0.19 mmol) in THF (5 mL) was added LiOH (IM, 2 mL). The reaction was stined overnight at RT. The mixture was concentrated and purified by HPLC to give example 46 (TFA salt, 86 mg, 82%). 1H NMR (400 MHz DMSO): β 7.90-7.95 (m, 4H), 7.42 (m, IH), 7.35 (m, 2H), 7.00 (s, IH), 6.73 (d, J=7.6 Hz, IH), 5.49-5.68 (AB q, J = 16.5, 59.0 Hz, 2H), 3.90 (s, 3H), 3.82 (s, 3H), 3.11 (m, 2H), 2.92 (m, 2H), 2.90 (m, IH), 2.61 (m, IH), 1.94 (m, IH), 1.67 (m, IH), 1.41 (m, 2H). MS (ES) [m+H] calculated for C25H26N4O5, 439; found 439. Example 47: 2-[2-(3-Anιino-piperidin-l-yl)-6-fluoro-4-oxo-4H-quinazoιin-3- ylmethyl]-benzoic acid
Figure imgf000164_0001
[0533] Example 45 (70 mg, 0.17 mmol) was converted to the title compound (TFA salt, 61 mg, 70%) using the procedure described for Example 46. 1H NMR (400 MHz DMSO): β 13.2 (br s, IH), 7.99 (m, 3H), 7.91 (d, J=7.3 Hz, IH), 7.56-7.69 (m, 3H), 7.41 (m, IH), 7.34 (m, IH), 6.85 (d, J=7.5 Hz, IH), 5.58 (AB q, J = 16.9, 53.4 Hz, 2H), 3.52 (d, J=10.8 Hz, IH), 3.14 (m, 2H), 2.95 (m, IH), 2.65 (m, IH), 1.95 (s, IH), 1.67 (s, IH), 1.42 (s, 2H). MS (ES) [m+H] calculated for C2ιH22FN O3, 397; found 397.
Example 48: 2-(6,7-Dimethoxy-4-oxo-2-piperidm-l-yl-4H-qmnazolin-3-yιmethyl)- benzonitrile
Figure imgf000164_0002
[0534] Example 2B (125 mg, 0.35 mmol) was converted to the title compound (50 mg) using the procedure described for Example 1, except that piperidine was used in the place of 3-aminopiperidine. 1H NMR (400 MHz DMSO): β 7.83 (d, J=7.5 Hz, IH), 7.59 (t, J=7.5 Hz, IH), 7.42 (t, J=7.4 Hz, IH), 7.32 (s, IH), 7.04 (d, J=7.9 Hz, IH), 6.99 (s, IH), 5.40 (s, 2H), 3.89 (s, 3H), 3.80 (s, 3H), 2.98 (br s, 4H), 1.53 (br s, 6H). MS (ES) [m+H] calculated for C23H25N4O3, 405; found 405.
4. Examples Of In vitro Assays
[0535] The protease inhibitory activities of DPP-IN inhibitors can be readily determined by methods known to those of ordinary skill in the art since suitable in vitro assays for measuring protease activity and the inhibition thereof by test compounds are known. Examples of assays that may be used for measuring protease inhibitory activity and selectivity are set forth below.
DPP-IN Assay
[0536] Solutions of test compounds in varying concentrations (≤lOmM final concentration) were prepared in Dimethyl Sulfoxide (DMSO) and then diluted into assay buffer comprising: 20mM Tris, pH 7.4; 20mM KCl; and O.lmg mL BSA. Human DPP-IN (0.1 nM final concentration) was added to the dilutions and pre-incubated for 10 minutes at ambient temperature before the reaction was initiated with A-P-7-amido-4- trifluoromethylcoumarin (AP-AFC; 10 μM final concentration). The total volume of the reaction mixture was 10-10QμL depending on assay formats used (384 or 96 well plates). The reaction was followed kinetically (excitation λ=400 nm; emission ?ι=505 nm) for 5-10 minutes or an end-point was measured after 10 minutes. Inhibition constants (IC50) were calculated from the enzyme progress curves using standard mathematical models.
FAPα Assay
[0537] Solutions of test compounds in varying concentrations (≤lOmM final concentration) were prepared in Dimethyl Sulfoxide (DMSO) and then diluted into assay buffer comprising: 20mM Tris, pH 7.4; 20mM KCl; and O.lmg/mL BSA. Human FAPα (2 nM final concentration) was added to the dilutions and pre-incubated for 10 minutes at ambient temperature before the reaction was initiated with A-P-7-amido-4- trifluoromethylcoumarin (AP-AFC; 40 μM final concentration). The total volume of the reaction mixture was 10- lOOμL depending on assay formats used (384 or 96 well plates). The reaction was followed kinetically (excitation λ=400 nm; emission λ=505 nm) for 5-10 minutes or an end-point was measured after 10 minutes. Inhibition constants (IC50) were calculated from the enzyme progress curves using standard mathematical models.
PREP Assay
[0538] Solutions of test compounds in varying concentrations (≤lOmM final concentration) were prepared in Dimethyl Sulfoxide (DMSO) and then diluted into assay buffer comprising: 20mM Sodium Phosphate, pH 7.4; 0.5mM EDTA; 0.5mM DTT; and 0. lmg/mL BSA. PREP (EC3.4.21.26 from Flavobacterium meningosepticum; 0.2 nM final concentration) was added to the dilutions. The PREP and compound were pre-incubated for 10 minutes at ambient temperature before the reaction was initiated with Z-G-P-AMC (10 μM final concentration). The total volume of the reaction mixture was 10-lOOμL depending on assay formats used (384 or 96 well plates). The reaction was followed kinetically (excitation λ=375 nm; emission λ=460 nm) for 10 minutes or an end-point was measured after 10 minutes. Inhibition constants (IC50) were calculated from the enzyme progress curves using standard mathematical models.
Tryptase Assay
[0539] Solutions of test compounds in varying concentrations (≤lOmM final concentration) were prepared in Dimethyl Sulfoxide (DMSO) and then diluted into assay buffer comprising: lOOmM Hepes, pH 7.4; 0.01% Brij35; and 10% glycerol. Tryptase (rhLung beta; 0.1 nM final concentration) was added to the dilutions and pre-incubated with compound for 10 minutes at ambient temperature. Ihe enzymatic reaction was initiated with 25 μM Z-lys-SBzl and 400μM DT^NB. The total volume of the reaction mixture was lOOμL in Costar A/2 96 well plates. The reaction was followed colorimetrically (λ=405 nm) for 10 minutes. Inhibition constants (IC50) were calculated from the enzyme progress curves using standard mathematical models.
[0540] Compounds of the invention were tested according to the above-described assays for protease inhibition and observed to exhibit selective DPP-IN inhibitory activity. For example, compounds of the invention were found to inhibit DPP-TV activity at concentrations that are at least 50 fold less than those concentrations required to produce an equiactive inhibition of protease activity for FAPα. The apparent inhibition constants (Ki) for compounds of the invention, against DPP-TV, were in the range from about 10"9M to about 10" 5M.
Microsomal Stability: [0541] Several compounds were evaluated for stability in rat liver microsomes. [0542] The test compounds (1 μM) were incubated at 37 °C in phosphate buffer (50 mM, pH 7.4) containing rat liver microsomes (1 mg/mL protein) and ΝADPH (Νicotinamide Adenine Dinucleotide Phosphate, reduced form) (4 mM). The incubation mixtures were quenched with trichloroacetic acid (0.3 M) over 0, 5, 15, 30 min time-course. Quenched solutions were centrifuged and supernatants were transfened for LC MS quantitation. The half life of a test compounds was derived from the compound stability curve over the time course.
[0543] As can be seen from the test results in Table 1, substitution at the 6 and/or 7 position of the compounds of Formula JJ appears to significantly improve the compound metabolic stability relative to the 6 and/or 7 unsubstituted analogues. Accordingly, one aspect of the present invention relates to the DPP1N inhibitors as described herein where the compounds of Formula IT are substituted at the 6 and/or 7 position.
[0544] In one variation, substituents at the 6 and/or 7 position are independently selected from the group consisting of halo, perhalo(Cι-ιo)alkyl, CF3, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, amino, thio, cyano, nitro, alkoxy, aryloxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
[0545] In another particular variation, substituents at the 6 and/or 7 position are independently selected from the group consisting of alkyl, aryl, heteroaryl, arylalkyl, heteroarylalkyl, cycloalkyl, and heterocycloalkyl, each substituted or unsubstituted.
[0546] In yet another particular variation, substituents at the 6 position are selected from the group consisting of halo, substituted or unsubstituted (Cι-ιo)alkoxy, cyano, nitro, and substituted or unsubstituted heteroaryloxy.
[0547] In yet another particular variation, DPP-JN inhibitors comprising the Formula JJ are substituted as the 6-chloro, 6-bromo, 6-fluoro, 6-iodo, 6-methoxy, 7-fluoro, or 6,7- dimethoxy quinazolinone derivatives. In another particular embodiment, DPP-JN inhibitors comprising the Formula JJ wherein L is nitrogen and the 6-position substituents are selected from the group consisting of halo, (Cι-ι0)alkoxy, cyano, nitro, morpholin-4-yl, pynolidin-1-yl, and heteroaryloxy, each substituted or unsubstituted.
[0548] In yet another variation, DPP-IN inhibitors comprising the Formula IT are substituted as the 6,7-disubstituted derivatives, where substituents at the 6 and 7 positions are independently selected from the group consisting of bromo, chloro, fluoro, iodo,
(Cι-ιo)alkoxy, cyano, nitro, morpholin-4-yl, pyπolidin-1-yl and heteroaryloxy, each substituted or unsubstituted. In a particular variation, DPP-JN inhibitors comprising the Formula TJ are substituted as the 6-fluoro,7-morpholin-l-yl quinazolinone derivatives. [0549] The improvement in stability in rat liver microsomes for specific compounds that are substituted at the 6 and/or 7 position of the compound of Formula IT are shown in Table 1.
TABLE 1
Figure imgf000168_0001
[0550] It will be apparent to those skilled in the art that various modifications and variations can be made to the compounds, compositions, kits, and methods of the present invention without departing from the spirit or scope of the invention. Thus, it is intended that the present invention cover the modifications and variations of this invention provided they come within the scope of the appended claims and their equivalents.

Claims

What is claimed is:
1. A compound comprising Formula X:
Figure imgf000169_0001
X wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NR9;
Ri is -ZRm, where Z is a moiety providing 1-6 atom separation between Rm and the ring to which Ri is attached, and Rm is selected from the group consisting of a substituted or unsubstituted (C3-7)cycloalkyl and aryl;
R2 is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring;
R3 and R are taken together to form a substituted or unsubstituted 5 or 6 membered ring; and
R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
2. A compound according to claim 1, wherein Z provides 1-3 atom separation between Rm and the ring.
3. A compound according to claim 1, wherein Z provides 1 atom separation between Rm and the ring.
4. A compound according to claim 3, wherein the 1 atom separation is provided by an atom selected from the group consisting of C, N, O, and S.
5. A compound according to claim 3, wherein the 1 atom separation is provided by a carbon atom.
6. A compound according to claim 3, wherein the 1 atom separation is provided by an oxygen atom.
7. A compound according to claim 3, wherein the 1 atom separation is provided by a nitrogen atom.
8. A compound according to claim 1, wherein Z is selected from the group consisting of -CH2-, -CH2CH2-, -CH2CH2CH2-, -C(O)-, -CH2C(O)-, -C(O)CH2-, -CH2-C(O)CH2-, -C(O)CH2CH2-, -CH2CH2C(O)-, -O-, -OCH2-, -CH2O-, -CH2OCH2-, -OCH2CH2-, -CH2CH2O-, -N(CH3)-, -NHCH2-, -CH2NH-, -CH2NHCH2-, -NHCH2CH2-, -CH2CH2NH-, -NH-C(O)-, -NCH3-C(O)-, -C(O)NH-, -C(O)NCH3-, -NHC(O)CH2-, -C(O)NHCH2-, -C(O)CH2NH-, -CH2NHC(O)-, -CH2C(O)NH-, -NHCH2C(O)-, -S-, -SCH2-, -CH2S-, -SCH2CH2-, -CH2SCH2-, -CH2CH2S-, -C(O)S-, -C(O)SCH2-, -CH2C(O)S-, -C(O)CH2S-, and -CH2SC(O)-, each substituted or unsubstituted.
9. A compound according to claim 1, wherein Z is selected from the group consisting of -CH2-, -CHR9-, -C(R9)(R9)-, -C(O)-, -C(S)-, -C(NH)-, -C(NR9)-, -O-, -N(H)-, -N(R9)-, and -S-.
10. A compound according to claim 1, wherein Rm is a substituted or unsubstituted (C3-7)cycloalkyl.
11. A compound according to claim 1, wherein Rm is a substituted or unsubstituted aryl.
12. A compound according to claim 1, wherein Rm is a substituted or unsubstituted phenyl.
13. A compound according to claim 1, wherein Rm is selected from the group consisting of (2-cyano)phenyl, (3-cyano)phenyl, (2-hydroxy)phenyl, (3-hydroxy)phenyl, (2- alkenyl)phenyl, (3-alkenyl)phenyl, (2-alkynyl)phenyl, (3-alkynyl)phenyl, (2-nitro)phenyl, (3-nitro)phenyl, (2-carboxy)phenyl, (3-carboxy)phenyl, (2-carboxamido)phenyl, (3- carboxamido)phenyl, (2-sulfonamido)phenyl, (3-sulfonamido)phenyl, (2-tetrazolyl)phenyl, (3-tetrazolyl)ρhenyl, (2-aminomethyl)phenyl, (3-aminomethyl)phenyl, (2-amino)phenyl, (3-amino)phenyl, (2-hydroxymethyl)ρhenyl, (3-hydroxymethyl)phenyl, (2-ρhenyl)phenyl, (3-phenyl)phenyl, (2-CONH2)phenyl, (3-CONH2)phenyl, (2-CONH(Cι-7)alkyl)phenyl, (3-CONH(Cι-7)alkyl)phenyl, (2-CO2(Cι-7)alkyl)phenyl, (3-CO2(Cι-7)alkyl)phenyl, -NH2, -OH, -(C3- )alkyl, -alkene, -alkyne, -CCH, -(C3-7)cycloalkyl, and -aryl, each substituted or unsubstituted.
14. A compound according to claim 1, wherein Ri is -ORn, where Rn is selected from the group consisting of substituted or unsubstituted alkyl, cycloalkyl, aryl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl.
15. A compound according to claim 1, wherein Z is a carbonyl.
16. A compound according to claim 1, wherein Ri is selected from the group consisting of -(CH2)-(2-cyano)phenyl, -(CH2)-(3-cyano)ρhenyl, -(CH2)-(2-hydroxy)phenyl, -(CH2)-(3- hydroxy)phenyl, -(CH2)-(2-alkenyl)ρhenyl, -(CH2)-(3-alkenyl)ρhenyl, -(CH2)-(2- alkynyl)phenyl, -(CH2)-(3-alkynyl)ρhenyl, -(CH2)-(2-nitro)phenyl, -(CH2)-(3-nitro)ρhenyl, -(CH2)-(2-carboxy)phenyl, -(CH2)-(3-carboxy)phenyl, -(CH2)-(2-carboxamido)phenyl, -(CH2)-(3-carboxamido)phenyl, -(CH2)-(2-sulfonamido)phenyl, -(CH2)-(3-sulfonamido)phenyl, -(CH2)-(2-tetrazolyl)phenyl, -(CH2)-(3-tetrazolyl)phenyl, -(CH2)-(2-an inome l)phenyl, -(CH2)-(3-aminomeιhyl)phenyl, -(CH2)-(2-amino)phenyl, -(CH2)-(3-amino)phenyl, -(CH2)-(2-hydroxymethyl)phenyl,
-(CH2)-(3-hydroxymethyl)ρhenyl, -(CH2)-(2-phenyl)phenyl, -(CH2)-(3-phenyl)phenyl, -(CH2)-(2-CONH2)phenyl, -(CH2)-(3-CONH2)phenyl, -(CH2)-(2-CONH(Cι-7)alkyl)phenyl, -(CH2)-(3-CONH(Cι-7)alkyl)phenyl, -(CH2)-(2-CO2(Cι-7)alkyl)phenyl, -(CH2)-(3-CO2(Cι-7)alkyl)phenyl, -CH2-NH2, -CH2-OH, -CH2-(C3-7)alkyl, -CH2-alkene, -CH2-alkyne, -CH2-CCH, -CH2-(C3-7)cycloalkyl, and -CH2-aryl, each substituted or unsubstituted.
17. A compound according to claim 1, wherein Ri is selected from the group consisting of -(Cι)alkyl-aryl, -(Cι)alkyl-bicycloaryl, -aminoaryl, -aminoheteroaryl, -aminobicycloaryl, - aminoheterobicycloaryl, -O-aryl, -O-heteroaryl, -O-bicycloaryl, -O-heterobicycloaryl, -(S)- aryl, -(S)-heteroaryl, -(S)-bicycloaryl, -S-heterobicycloaryl, -C(O)-aryl, -C(O)-heteroaryl, - C(O)-bicycloaryl, - C(O)-heterobicycloaryl, -C(S)-aryl, -C(S)-heteroaryl, -C(S)- bicycloaryl, -C(S)-heterobicycloaryl, -S(O)-aryl, -S(O)-heteroaryl, -S(O)-bicycloaryl, - SO2-heterobicycloaryl, -SO2-aryl, -SO2-heteroaryl, -SO2-bicycloaryl, -SO2- heterobicycloaryl, -C(NR9)-aryl, -C(NR9)-heteroaryl, -C(NR9)-bicycloaryl, -C(NR9)- heterobicycloaryl, each substituted or unsubstituted.
18. A compound according to claim l,wherein R2 is a substituted or unsubstituted 3, 4, 5, 6, or 7 membered cycloalkyl.
19. A compound according to claim 1, wherein R2 is a substituted or unsubstituted 4, 5, 6, or 7 membered heterocycloalkyl.
20. A compound according to claim 1, wherein R2 is a substituted or unsubstituted aryl.
21. A compound according to claim 1, wherein R2 is substituted or unsubstituted phenyl.
22. A compound according to claim 1, wherein R2 is a substituted or unsubstituted heteroaryl.
23. A compound according to claim 1, wherein R2 is selected from the group consisting of
Figure imgf000172_0001
wherein p is 0-12 and each R8 is independently selected from the group consisting of halo, perhalo(Cι-ιo)alkyl, CF3, cyano, nitro, hydroxy, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
24. A compound according to claim 1, wherein R2 is selected from the group consisting of
Figure imgf000173_0001
wherein r is 0-13 and each R8 is independently selected from the group consisting of halo, perhalo(Cι-ιo)alkyl, CF3, cyano, nitro, hydroxy, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
25. A compound according to claim 1, wherein R2 is a substituted or unsubstituted heteroaryl selected from the group consisting of furan, thiophene, pynole, pyrazole, triazole, isoxazole, oxazole, thiazole, isothiazole, oxadiazole, pyridine, pyridazine, pyrimidine, pyrazine, triazine, benzofuran, isobenzofuran, benzothiophene, isobenzothiophene, imidazole, benzimidazole, indole, isoindole, quinoline, isoquinoline, cinnoline, quinazoline, naphthyridine, pyridopyridine, quinoxaline, phthalazine, and benzothiazole, each substituted or unsubstituted.
26. A compound according to claim 1, wherein R2 is selected from the group consisting of (C3-i2)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(Cι-ι0)alkyl, heteroaryl (Cι-5)alkyl, (C -i2)bicycloaryl, and hetero(C42)bicycloaryl, each substituted or unsubstituted.
27. A compound according to claim 1, wherein R2 is a substituted or unsubstituted (C3-7)cycloalkyl ring, optionally comprising O, N(O), N, S, SO, SO2 or a carbonyl group in the ring.
28. A compound according to claim 1, where R3 and R4 are taken together to form an unsubstituted or substituted 5 or 6 membered cycloalkyl or heterocycloalkyl ring.
29. A compound according to claim 1 , where R3 and Rt are taken together to form a substituted or unsubstituted phenyl ring.
30. A compound according to claim 1, where R3 and t are taken together to form a substituted or unsubstituted heteroaryl ring.
31. A compound according to claim 1, where R3 and t are taken together to form a substituted or unsubstituted heteroaryl selected from the group consisting of substituted or unsubstituted furan, thiophene, pynole, pyrazole, triazole, isoxazole, oxazole, thiazole, isothiazole, oxadiazole, pyridine, pyridazine, pyrimidine, pyrazine, triazine, benzofuran, isobenzofuran, benzothiophene, isobenzothiophene, imidazole, benzimidazole, indole, isoindole, quinoline, isoquinoline, cinnoline, quinazoline, naphthyridine, pyridopyridine, quinoxaline, phthalazine, and benzothiazole.
32. A compound according to claim 1, where R3 and Rt are taken together to form a 5 or 6-membered ring where the ring comprises at least one CO group.
33. A compound according to claim 1, where R3 and R are taken together to form a 5 or . 6-membered ring comprising of 1-3 nitrogen ring atoms.
34. A compound according to claim 1, where R3 and t are taken together to form a 5 or 6-membered ring where the ring comprises a sulfur atom.
35. A compound according to claim 34, wherein the ring sulfur atom is in an oxidized form as SO or S02.
36. A compound according to claim 1, wherein the ring formed by Rj and R* comprises substituents that form a ring fused to the ring fonned by Ra and R*.
37. A compound according to claim 1, wherein R3 and R4 are taken together to form a ring system such that the compound of Formula X formed is selected from the group consisting of substituted or unsubstituted 4~oxo-4H-quϊnazoline, 3H-pyrido[2,3-d]pyrirnidin-4-one, 3H-pyrido[3,2-d]pyri idiπ-4-one, 3H-pyridQ[3,4-d]pyrimidin- -one and 3H-pyrido[4,3- d]pyrimidin-4-one.
38. A compound comprising Formula XI:
Figure imgf000175_0001
XI wherein
Q is selected from, the group consisting of CO, CS, SO, S02, or C=NR9;
J, K, L, and M are each independently selected from the group of CR12 and N;
Ri is -ZRm, where Z is a moiety providing 1-6 atom separation between Rm and the ring to which Rj is attached, and Rm is selected from the group consisting of a substituted or unsubstituted (C3-7)cycloalkyl and aryl;
R2 is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring;
Rs> is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted; and each R12 is hydrogen or is independently selected from the group consisting of halo, perhalo(C1-1o)alkyl, CF3, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, alkoxy, a carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
39. A compound according to claim 38, wherein the compound is a compound where J, K, L and M each comprise a carbon ring atom.
40. A compound according to claim 38, wherein the compound is a compound where J comprises a nitrogen ring atom.
41. A compound according to claim 38, wherein the compound is a compound where K comprises a nitrogen ring atom.
42. A compound according to claim 38, wherein the compound is a compound where L comprises a nitrogen ring atom.
43. A compound according to claim 38, wherein the compound is a compound where M comprises a nitrogen ring atom.
44. A compound according to claim 38, wherein the compound is a compound where J and L each comprise a nitrogen ring atom or J and K each comprise a nitrogen ring atom.
45. A compound according to claim 38, wherein the compound is a compound where K and L each comprise a nitrogen ring atom.
46. A compound according to claim 38, wherein the compound is a compound where K and M each comprise a nitrogen ring atom.
47. A compound according to claim 38, wherein the compound is a compound where J and M each comprise a nitrogen ring atom or L and M each comprise a nitrogen ring atom.
48. A compound according to claim 38, wherein at least two of J, K, L and M comprise a nitrogen ring atom.
49. A compound according to claim 38, wherein at least three of J, K, L and M comprise a nitrogen ring atom.
50. A compound according to claim 38, wherein the ring formed by J, K, L, and M comprises substituents that form a ring fused to or bridged to the ring formed by J, K, L, and M.
51. A compound according to claim 38, wherein K is CR12, where R12 is independently selected from the group consisting of halo, perhalo(C1-1o)alkyl, CF3, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, alkoxy, a carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
52. A compound according to claim 38, wherein K is CR12, where R12 is independently selected from the group consisting of halo, perhalo(C1-1o)alkyl, CF3, cyano, nitro, alkyl, aryloxy, heteroaryloxy, amino, and alkoxy, each substituted or unsubstituted.
53. A compound according to claim 38, wherein K is CR12, where R12 is independently selected from the group consisting of heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryl, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, thio, a carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
54. A compound according to claim 38, wherein K is CRι2, where R12 is independently selected from the group consisting of chloro, bromo, fluoro, iodo, methoxy, morpholin-4- yl, and pyrrolidin- 1-yl, each substituted or unsubstituted.
55. A compound according to claim 38, wherein L is CR12, where R12 is independently selected from the group consisting of halo, perhalo(C1-1o)alkyl, CF3, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, alkoxy, a carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
56. A compound according to claim 38, wherein L is CR12, where R12 is independently selected from the group consisting of halo, perhalo(C1-1o)alkyl, CF3, cyano, nitro, alkyl, aryloxy, heteroaryloxy, amino, morpholin-4-yl, and pyrrolidin- 1-yl, and alkoxy, each substituted or unsubstituted.
57. A compound according to claim 38, wherein K and L are independently CR12, where R12 is independently selected from the group consisting of halo, perhalo(C1-10)alkyl, CF3, cyano, nitro, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, a carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
58. A compound according to claim 38, wherein:
K is CR12, where R12 is independently selected from the group consisting of halo, perhalo(C1-1o)alkyl, CF3, cyano, nitro, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, a carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted; and
L is nitrogen.
59. A compound comprising a member selected from the group consisting of Formulae XHa, XHb, Xπc, XHd, πe and XHf :
Figure imgf000179_0001
wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NR ;
Ri is -ZRm, where Z is a moiety providing 1-6 atom separation between Rm and the ring to which Ri is attached, and Rm is selected from the group consisting of a substituted or unsubstituted (C3- )cycloalkyl and aryl;
R2 is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring;
R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted; and each Rig is independently selected from the group consisting of hydrogen, halo, ρerhalo(Cι-ιo)alkyl, CF3, cyano, nitro, alkyl, alkene, alkyne, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, carbonyl group, irnine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, with the proviso that R19 is not alkylthio, arylthio, halo, cyano, nitro, and thio in the case where the ring atom to which R19 is bound is nitrogen.
60. A compound according to claim 59, wherein two R19 are taken together to form a substituted or unsubstituted fused or bridged ring.
61. A compound comprising Formula XHI:
Figure imgf000180_0001
wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NR9;
W, X, and Y are each independently selected from the group of moieties where the ring atom is either C, N, O or S;
Ri is -ZRm, where Z is a moiety providing 1-6 atom separation between Rm and the ring to which Ri is attached, and Rm is selected from the group consisting of a substituted or unsubstituted (C3- )cycloalkyl and aryl;
R2 is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring;
R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
62. A compound according to claim 61, wherein at least one of W, X, and Y is CO.
63. A compound according to claim 61, wherein at least one of W, X, and Y is SO.
64. A compound according to claim 61, wherein at least one of W, X, and Y is SO2.
65. A compound according to claim 61, wherein at least one of W, X, and Y comprises a ring nitrogen atom.
66. A compound according to claim 61, wherein at least two of W, X, and Y comprises a ring nitrogen atom.
67. A compound according to claim 61, wherein W and Y are taken together to form a substituted or unsubstituted bridged ring relative to the ring formed by , X and Y.
68. A compound according to claim 61, wherein two of W, X, and Y are taken together to form a substituted or unsubstituted ring fused to the ring formed by W, X and Y.
69. A compound comprising a member selected from the group consisting of Formulae XTVa, XlNb or XlNc:
Figure imgf000181_0001
wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NR9;
Rt is -ZRm, where Z is a moiety providing 1-6 atom separation between Rm and the ring to which Ri is attached, and Rm is selected from the group consisting of a substituted or unsubstituted (C3-7)cycloalkyl and aryl; R2 is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring;
R is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted; and each R1 is independently selected from the group consisting of hydrogen, halo, perhalo(C1-1o)alkyl, CF3, cyano, nitro, alkyl, alkene, alkyne, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, with the proviso that R19 is not alkylthio, arylthio, halo, cyano, nitro, and thio in the case where the ring atom to which R1 is bound is nitrogen.
70. A compound according to claim 69, wherein two R19 are taken together to form a substituted or unsubstituted bridged or spiro ring.
71. A compound comprising Formula XVa or Formula XNb:
Figure imgf000182_0001
wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=ΝR9;
W, X, and Y are each independently selected from the group of moieties where the ring atom is either C, N, O or S;
Ri is -ZRm, where Z is a moiety providing 1-6 atom separation between Rm and the ring to which Ri is attached, and Rm is selected from the group consisting of a substituted or unsubstituted (C3- )cycloalkyl and aryl;
R2 is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring; and R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
72. A compound according to claim 71, wherein the compound is a compound of Formula XNa wherein Y is selected from the group consisting of CO, SO or SO2.
73. A compound according to claim 71, wherein the compound is a compound of Formula XNb wherein W is selected from the group consisting of CO, SO or SO2.
74. A compound according to claim 71, wherein W comprise a ring nitrogen atom.
75. A compound according to claim 71, wherein X comprise a ring nitrogen atom.
76. A compound according to claim 71, wherein Y comprise a ring nitrogen atom.
77. A compound according to claim 71, wherein at least two of W, X, and Y comprises a ring nitrogen atom.
78. A compound according to claim 71, wherein two of W, X, and Y are taken together and substituted through available valencies to form a substituted or unsubstituted ring fused or bridged to the ring formed by W, X and Y.
79. A compound according to claim 71, wherein W, X, and Y are selected such that the compound comprises a ring system selected from the group consisting of 4-oxo-4H- thieno [3 ,2-d]pyrimidine, 7-oxo- 1 ,2,3 ,7-tetrahydro-8-thia-4,6-diaza-cyclopenta[a]indene, 7- memyl-6-oxo-6,7-dihydro-purine, and 6-oxo-6,9-dihydro-purine, each substituted or unsubstituted.
80. A compound comprising Formulae XNIa, XNIb, or XVIc:
Figure imgf000184_0001
wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NR ;
X is selected from the group of moieties where the ring atom is either C, N, O or S in Formula XVIa, or X is selected from the group of moieties where the ring atom is either C or N in Formula XVIb or Formula XVIc;
Ri is -ZRm, where Z is a moiety providing 1-6 atom separation between Rm and the ring to which Ri is attached, and Rm is selected from the group consisting of a substituted or unsubstituted (C3-7)cycloalkyl and aryl;
R2 is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring;
R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted; and each Ri9 is independently selected from the group consisting of hydrogen, halo, perhalo(Ci-io)alkyl, CF3, cyano, nitro, alkyl, alkene, alkyne, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, with the proviso that R19 is not alkylthio, arylthio, halo, cyano, nitro, and thio in the case where the ring atom to which R19 is bound is nitrogen.
81. A compound according to claim 80, wherein two R19 are taken together to form a substituted or unsubstituted ring.
82. A compound according to claim 80, wherein the compound comprises Formula XVIa and the two Rι9 are taken together to form a substituted or unsubstituted fused or bridged ring.
83. A compound comprising a member selected from the group of Formulae XVIIa, XVTIb and XVHc:
Figure imgf000185_0001
XVHa xvπb xvπc
wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NR9;
J, K, L, and M are each independently selected from the group of moieties where the ring atom is either C, N, O or S;
Ri is -ZRm, where Z is a moiety providing 1-6 atom separation between Rm and the ring to which Ri is attached, and Rm is selected from the group consisting of a substituted or unsubstituted (C3- )cycloalkyl and aryl; and
R2 is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring.
84. A compound according to claim 83, wherein the compound is a compound where J, K, L and M each comprise a carbon ring atom.
85. A compound according to claim 83, wherein at least one of J, K, L and M comprise a nitrogen ring atom.
86. A compound according to claim 83, wherein the compound is a compound where J and K each comprise a nitrogen ring atom or J and L each comprise a nitrogen ring atom.
87. A compound according to claim 83, wherein the compound is a compound where K and L each comprise a nitrogen ring atom or K and M each comprise a nitrogen atom.
88. A compound according to claim 83, wherein the compound is a compound where J and M each comprise a nitrogen ring atom or L and M each comprise a nitrogen ring atom.
89. A compound according to claim 83, wherein at least two of J, K, L and M comprise a nitrogen ring atom.
90. A compound according to claim 83, wherein at least three of J, K, L and M comprise a nitrogen ring atom.
91. A compound according to claim 83, wherein at least one of J, K, L and M is CO.
92. A compound according to claim 83, wherein at least one of J, K, L and M is SO.
93. A compound according to claim 83, wherein at least one of J, K, L and M is SO2.
94. A compound according to claim 83, wherein the ring formed by J, K, L, and M comprises substituents, through available valencies, that form a ring fused to the ring formed by J, K, L, and M or, in the case of Formula XVHb, J and M form a bridged ring relative to the ring formed by J, K, L, and M.
95. A compound comprising Formula XVHI:
Figure imgf000186_0001
xvm wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NR ; J, K, L, and M are each independently selected from the group of moieties where the ring atom is either C, N, O or S;
Ri is -ZRm, where Z is a moiety providing 1-6 atom separation between Rm and the ring to which Ri is attached, and Rm is selected from the group consisting of a substituted or unsubstituted (C3-7)cycloalkyl and aryl;
R2 is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring; and
R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
96. A compound according to claim 95, wherein at least one of J, K, L and M is CO.
97. A compound according to claim 95, wherein at least one of J, K, L and M is SO.
98. A compound according to claim 95, wherein at least one of J, K, L and M is SO2.
99. A compound according to claim 95, wherein the compound is a compound where J, K, L and M each comprise a carbon ring atom.
100. A compound according to claim 95, wherein the compound is a compound where J comprises a nitrogen ring atom.
101. A compound according to claim 95, wherein the compound is a compound where K comprises a nitrogen ring atom.
102. A compound according to claim 95, wherein the compound is a compound where L comprises a nitrogen ring atom.
103. A compound according to claim 95, wherein the compound is a compound where M comprises a nitrogen ring atom.
104. A compound according to claim 95, wherein the compound is a compound where J and K each comprise a nitrogen ring atom or J and L each comprise a nitrogen ring atom.
105. A compound according to claim 95, wherein the compound is a compound where K and L each comprise a nitrogen ring atom or K and M each comprise a nitrogen atom.
106. A compound according to claim 95, wherein the compound is a compound where J and M each comprise a nitrogen ring atom or L and M each comprise a nitrogen ring atom.
107. A compound according to claim 95, wherein at least two of J, K, L and M comprise a nitrogen ring atom.
108. A compound according to claim 95, wherein at least three of J, K, L and M comprise a nitrogen ring atom.
109. A compound according to claim 95, wherein the ring formed by J, K, L, and M comprises substituents that form a ring fused to the ring formed by J, K, L, and M.
110. A compound according to claim 95, wherein the ring formed by J, K, L, and M comprises substituents that form a bridged ring relative to the ring formed by J, K, L, and M.
111. A compound selected from the group consisting of: 2-[2-(3-Amino-piperidin-l-yl)-4-oxo-4H-quinazohn-3-ylmemyl]-benzomtrile; 2,4-Dichloro-quinazoline;
2-Chloro-3H-quinazolin-4-one;
2-(2-Chloro-4-oxo-4H-quinazolin-3-ylmethyl)-benzonitrile; 2-[2-(3-Amino-piperidin-l-yl)-6,7-dimemoxy-4-oxo-4H-quinazolin-3-ylmethyl]- benzonitrile;
2-Chloro-6,7-dimethoxy-3H-quinazoUn-4-one; 2-(2-Cωoro-6,7-dimemoxy-4-oxo-4H-quinazolin-3-ylmethyl)-benzonitrile;
2-[2-(3-Amino-piperid^n-l-yl)-8-memoxy-4-oxo-4H-qumazolin-3-ylmethyl]- benzonitrile;
8-Methoxy- lH-quinazoline-2,4-dione;
2,4-Dichloro-8-methoxy-quinazoline;
2-Chloro-8-methoxy-3H-quinazoUn-4-one;
2-(2-CWoro-8-memoxy-4-oxo-4H-qu azolin-3-ylmethyl)-benzonitrile;
2-[2-(3-Amino-piperidin-l-yl)-7-chloro-4-oxo- H-quinazolin-3-ylmethyl]- benzonitrile, TFA salt;
2,7-Dichloro-5H-quinazolin-4-one;
2-(2,7-Dicmoro-4-oxo-4H-quinazolin-3-ylmethlyl)-benzonitrile;
2-[2-(3-Ammo-piperidin-l-yl)-8-cUoro-4-oxo-^H-qumazolm-3-ylmethyl]- benzonitrile, TFA salt;
2,8-Dichloro-3Η-quinazolin-4-one;
2-(2,8-Dichloro-4-oxo- H-quinazolin-3-ylmethlyl)-benzonitrile;
6-Fluoro-lΗ-quinazoline-2,4-dione;
2,4-Dichloro-6-fluoro-quinazoline;
2-Chloro-6-fluoro-3H-quinazolin-4-one;
2-(2-Chloro-6-fluoro-4-oxo-4H-quinazolin-3-ylmethyl)-benzonitrile;
(R) 2-[2-(3-Amino-piperidin-l-yl)-6-fluoro-4-oxo-4H-quinazolin-3-ylmethyl]- benzonitrile TFA salt;
2-[2-(3-Ammo-ρiperidin-l-yl)-7-methyl-6-oxo-6,7-(imydro-purin-l-ylmethyl]- benzonitrile;
2-[2-(3-Amino-piperidin-l-yl)-9-methyl-6-oxo-6,9-dihydro-purin-l-ylmethyl]- benzonitrile;
2,6-Dichloro-7-methyl-7H-purine;
2,6-Dichloro-9-methyl-9H-purine;
2-CUoro-7-memyl-l,7-dihydro-purin-6-one;
2-Chloro-9-methyl- 1 ,9-dihydro-purin-6-one;
2-(2-CWoro-7-memyl-6-oxo-6,7-ά^hydro-purm-l-ylmemyl)-benzonitrile;
2-(2-CMoro-9-memyl-6-oxo-6,9-ά^ydro-purin-l-ylmethyl)-benzonitrile; 2-{2-[(R)-3-Ammo-piperidm-l-yl]-6-oxo-6,7-dihydro-purin-l-ylmethyl}- benzonitrile;
7-Benzyloxymethyl-2,6-dichloro-7H-purine;
9-Benzyloxymethyl-2,6-dichloro-9H-ρurine;
7-Benzyloxymethyl-2-chloro- 1 ,7-dihydro-purin-6-one;
9-Benzyloxymethyl-2-chloro- 1 ,9-dihydro-purin-6-one;
2-(7-Benzyloxymethyl-2-cWoro-6-oxo-6,7-dmydro-purm-l-ylmethyl)-benzonitrile;
2-(9-Benzyloxymemyl-2-chloro-6-oxo-6,9-ά hydro-purin-l-ylmemyl)-benzonitrile;
2-(2-Chloro-6-oxo-6,9-dihydro-purin- 1 -ylmethyl)-benzonitrile;
2-[2-(3-(R)-Amino-piperiά n-l-yl)-6-chloro-4-oxo-4H-quinazolin-3-ylmethyl]- benzonitrile, TFA salt;
2,6-Dichloro-3H-quinazolin-4-one;
2-(2,6-Dichloro-4-oxo-4H-quinazolin-3-ylmethyl)-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-lyl)-7-fluoro-6-methoxy-4-oxo-4H-quinazolin-3- ylmethyl]-benzonitrile, TFA salt;
7-Fluoro-6-methoxy-lH-quinazoline-2,4-dione;
2-Chloro-7-fluoro-6-methoxy-3H-quinazolin-4-one;
2-(2-CUoro-7-fluoro-6-memoxy-4-oxo-4H-quinazolin-3-ylmemyl)-benzonitrile;
2-[2-(3-(R)-Am o-phperidm-lyl)-6-memoxy-4^ ylmethyl] -benzonitrile, TFA salt;
6-Methoxy-lH-pyrido[3,4-(i]pyrimidine-2,4-dione;
2-Chloro-6-methoxy-3H-pyrido [3 ,4- ]pyrimidin-4-one;
2-(2-Chloro-6-methoxy-4-oxo-4H-pyrido [3 ,4--i]pyrimidin-3-ylmethyl)- benzonitrile;
2-[6-(3-(R)-Amino-piperidin-l-yl)-l-methyl-4-oxo-l,4-dihydro-pyrazolo[3,4- <flpyrimidin-5-ylmethyl]-benzonitrile, TFA salt;
6-Chloro-l-memyl-l,5-dmydro-pyrazolo[3,4-^pyriroidin-4-one;
2-(6-CUoro-l-memyl-4-oxo-l,4-alhydro-pyrazolo[3,4-^pyrimidin-5-ylmethyl)- benzonitrile;
2-[2-(3-(R)-Ammo-ρiperiώn-lyl)-5-fluoro-4-oxo-4H-quinazolin-3-ylmethyl]- benzonitrile, TFA salt; 2-Chloro-5-fluorb-3H-quinazolin-4-one;
2-(2-Chloro-5-fluoro-4-oxo-4H-quinazolin-3-ylmethyl)-benzonitrile;
2-[5-(3-(R)-Amino-piperidin- 1 -yl)- 1 -methyl-7-oxo- 1 ,7-dihydro-[ 1 ,2,3]triazolo[4,5- -i]pyrimidin-6-ylmethyl]-benzonitrile, TFA salt;
5-Chloro- 1 -methyl- 1 ,6-dihydro-[ 1 ,2,3]txiazolo[4,5-^pyrirmdin-7-one;
2-(5-Chloro-l-methyl-7-oxo- 1 ,7-dihydro-[l ,2,3]triazolo[4,5-d]ρyrimidin-6- ylmethyl)-benzonitrile;
2-[5-(3-(R)-Amino-ρiρeridin- 1 -yl)-2-methyl-7-oxo-2,7-dihydro[ 1 ,2,3]triazolo[4,5- ]pyrirnidin-6-ylmethyl] -benzonitrile, TFA salt;
5-Chloro-2-memyl-2,6-dmydro-[l,2,3]triazolo[4,5--i]pyrimidin-7-one;
2-(5-CWoro-2-me l-7-oxo-2,7-dihydro-[l,2,3]triazolo[4,5-d]pyrinndin-6- ylmethyl)-benzonitrile;
2-[2-(3-(R)-Ammo-piperidin-lyl)-4-oxo-5,6,7,8-tetrahydro-4H-qumazolin-3- ylmethyl] -benzonitrile, TFA salt;
2-Chloro-5,6,7,8-tetrahydro-3H-quinazolin-4-one;
2-(2-CUoro-4-oxo-5,6,7,8-te1xahyα^o-4H-quinazolin-3-ylmethyl)-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-lyl)-6-chloro-4-oxo-4H-pyrido[3,4-<i]pyrimidin-3- ylmethyl]-benzonitrile, TFA salt; l,7-Dmydro-pyrido[3,4-rf]pyrimidine-2,4,6-trione;
2,6-DicWoro-3H-pyrido[3,4--^ρyrimidine-4-one;
2-(2,6-Dichloro-4-oxo-4H-pyrido [3 ,4-d]pyrimidin-3-ylmethyl)-benzonitrile;
2-[2-(3-(R)-Arnino-piperidin- lyl)-4-oxo-6-pyrrohdin- lyl-4H-pyrido[3,4- ]pyrimidin-3-ylmethyl]-benzonitrile;
2-[(R)-3-Ammo-piperichn-l-yl]-6-fluoro-3-(2-trifluoromethyl-benzyl)-3Η- quinazohn-4-one;
2-Chloro-6-fluoro-3-(2-trifluoromethyl-benzyl)-3H-quinazolin-4-one;
2-{ 2-[(R)-3-Amino-piperidin- 1 -yl]-7-isopropyl-6-oxo-6,7-dihydro-purin- 1 - ylmethyl}-benzonitrile;
2-[2-(3-Amino-azepan-l-yl)-6-oxo-6,7-dihydro-purin-l-ylmethyl]-benzonitrile;
2-{2-[(R)-3-Amino-piperidin-l-yl]-7-benzyl-6-oxo-6-hydro-purin-l-ylmethyl}- benzonitrile; 2- { 2-[(R)-3-Amino-piperidin- l-yl]-9-(2-cyano-benzyl)-6-oxo-6-hydro-purin- 1- ylmethyl } -benzonitrile;
2-{2-[(R)-3-Amino-piperidin-l-yl]-6-oxo-9-propyl-6,9-dmydro-purin-l-ylmethyl}- benzonitrile;
2-{2-[(R)-3-Amino-piperidin-l-yl]-6-oxo-7-proρyl-6,7-dmydro-purin-l-ylmethyl}- benzonitrile;
2-Chloro-9-propyl- 1 ,9-dihydro-purin-6-one;
2-Chloro-7-propyl- 1 ,7-dihydro-purin-6-one;
2-(2-CUoro-6-oxo-9-propyl-6,9-dmydro-purin-l-ylmemyl)-benzonitrile;
2-(2-Crdoro-6-oxo-7-propyl-6,7-dihydro-purin-l-ylmethyl)-benzonitrile;
2-{2-[(R)-(3-Aιrrmo-piperimn-l-yl)]-9-isopropyl-6-oxo-8-trifluoromethyl-6,9- dihydro-purin- 1 -ylmethyl } -benzonitrile;
6-CMoro-N4-isopropyl-pyrimidine-2,4,5-triamine;
6-CWoro-9-isopropyl-8-trifluoromethyl-9H-purin-2-ylamine;
2-(2-Amino-9-isopropyl-6-oxo-8- fluoromemyl-6,9-dmydro-purin-l-ylmethyl)- benzonitrile;
2-(2-Bromo-9-isopropyl-6-oxo-8-trifluoromemyl-6,9-dmydro-purin-l-ylmethyl)- benzonitrile;
2-[2-(3-(R)-Amino-piperidin-l-yl)-6-bromo-4-oxo-4H-quinazolin-3-ylmethyl]- benzonitrile;
6-Bromo- lH-quinazoline-2,4-dione;
6-Bromo-2-chloro-3H-quinazolin-4-one;
2-(6-Bromo-2-chloro-4-oxo-4H-quinazolin-3-ylmethyl)-benzonitrile;
2-[2-(3-(R)-Armno-piperidin-l-yl)-6-bromo-4-oxo-4H-quinazohn-3-ylmethyl]- benzonitrile, TFA salt;
2-[2-(3-(R)-Amino-py τoliά^n-l-yl)-6-bromo-4-oxo-4H-quinazohn-3-ylmethyl]- benzonitrile, TFA salt;
2-[2-(3-(R)-Ammo-piperid1n-l-yl)-6,8-dichloro-4-oxo-4H-quinazolin-3-ylmethyl]- benzonitrile;
6,8-Dichloro-lH-quinazoline-2,4-dione;
2,6,8-Trichloro-3H-quinazolin-4-one; 2-(2,6,8-Trichloro-4-oxo-4H-quinazolin-3-ylmethyl)-benzonitrile;
2-[2-(3-(R)-Arn o-piperidin-l-yl)-6-memoxy-4-oxo-4H-quinazolin-3-ylmethyl]- benzonitrile;
6-Methoxy- 1 H-quinazoline-2,4-dione;
2,4-Dichloro-6-methoxy-quinazoline;
2-Chloro-6-methoxy-3H-quinazolin-4-one;
2-(2-Chloro-6-methoxy-4-oxo-4H-quinazolin-3-ylmethyl)-benzonitrile;
2-[2-(3-(R)-Anήno-piperidin-l-yl)-6-fluoro-4-oxo-4H-quinazolin-3-ylmethyl]- benzamide;
2-[2-(3-(R)-Amino-piperidin-l-yl)-6-fluoro-7-morpholin-4-yl-4-oxo-4H- quinazolin-3-ylmethyl]-benzonitrile;
6,7-Difluoro-lH-quinazoline-2,4-dione;
6-Huoro-7-moφholin-4-yl-lH-quinazoline-2,4-dione;
2,4-Dichloro-6-fluoro-7-morpholin-4-yl-quinazoline;
2-CWoro-6-fluoro-7-morpholin-4-yl-3H-quinazolin-4-one;
2-(2-Chloro-6-fluoro-7-morpholin-4-yl-4-oxo-4H-quinazolin-3-ylmethyl)- benzonitrile;
2-[2-(3-Amino-piperidin-l-yl)-6-fluoro-4-oxo-4H-quinazolin-3-ylmethyl]- benzamide;
2-[3-(R)-Ammo-piperidm-l-yl]-6-fluoro-3-(2-trifluoromethyl-benzyl)-3H- quinazolin-4-one;
2-(3-An ino-piperidin-l-yl)-6,7-dimemoxy-3-(2-nitro-benzyl)-3H-quinazolin-4- one;
2-[2-(3-Ammo-piperidin-l-yl)-6,7-dimethoxy-4-oxo-4H-quinazolin-3-ylmethyl]- benzoic acid ethyl ester;
2-[2-(3-Amino-piperidin-l-yl)-6-fluoro-4-oxo-4H-quinazolin-3-ylmethyl]-benzoic acid ethyl ester;
2-[2-(3-Arnmo-piperiώn-l-yl)-6,7-dimethoxy-4-oxo-4H-quinazolin-3-ylmethyl]- benzoic acid;
2-[2-(3-Amino-piperidin-l-yl)-6-fluoro-4-oxo-4H-quinazolin-3-ylmethyl]-benzoic acid; and 2-(6,7-Dimemoxy-4-oxo-2-piperidin-l-yl-4H-quinazolin-3-ylmemyl)-benzonitrile.
112. A compound comprising Formula XIX:
Figure imgf000194_0001
XIX wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NR9;
Ri is -ZRm, where Z is a moiety providing 1-6 atom separation between Rm and the ring to which Ri is attached, and -Rm is selected from the group consisting of a substituted or unsubstituted (C3-7)cycloalkyl and aryl;
R2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein;
R3 and R). are taken together to form a substituted or unsubstituted 5 or 6 membered ring; and
R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
113. A compound according to claim 112, wherein V is selected from the group consisting of a primary, secondary or tertiary amine, a heterocycloalkyl comprising a nitrogen ring atom, and a heteroaryl comprising a nitrogen ring atom
114. A compound according to claim 112, wherein R2 is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring wherein at least one substituent is selected from the group consisting of a primary, secondary or tertiary amine, a heterocycloalkyl comprising a nitrogen ring atom, and a heteroaryl comprising a nitrogen ring atom.
115. A compound according to claim 112, wherein the basic nitrogen of V is separated from the ring atom to which R2 is attached by between 1-5 atoms.
116. A compound according to claim 112, wherein the basic nitrogen of V forms part of a primary, secondary or tertiary amine.
117. A compound according to claim 112, wherein the basic nitrogen of V is a nitrogen ring atom of a heterocycloalkyl comprising a nitrogen ring atom or a heteroaryl comprising a nitrogen ring atom.
118. A compound according to claim 112, wherein -UN is selected from the group consisting of
Figure imgf000195_0001
wherein p is 0-12 and each R8 is independently selected from the group consisting of halo, perhalo(Cι-ιo)alkyl, CF3, cyano, nitro, hydroxy, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, with the proviso that at least one R8 serves as V.
119. A compound according to claim 118, wherein at least one R8 is a primary, secondary or tertiary amine.
120. A compound according to claim 118, wherein at least one R8 is a substituted or unsubstituted heterocycloalkyl comprising a nitrogen ring atom or a substituted or unsubstituted heteroaryl comprising a nitrogen ring atom.
121. A compound according to claim 118, wherein at least one R8 is selected from the group consisting of -NH2, -NH(Cι.5 alkyl), -N(Cι.s alkyl) 2, piperazine, imidazole, and pyridine.
122. A compound according to claim 112, wherein -UN is selected from the group consisting of
Figure imgf000196_0001
wherein r is 0-13 and each R8 is independently selected from the group consisting of halo, ρerhalo(Cι-ιo)alkyl, CF3, cyano, nitro, hydroxy, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, with the proviso that at least one R8 serves as V.
123. A compound according to claim 122, wherein at least one R8 is a primary, secondary or tertiary arnine.
124. A compound according to claim 122, wherein at least one R8 is a substituted or unsubstituted heterocycloalkyl comprising a nitrogen ring atom or a substituted or unsubstituted heteroaryl comprising a nitrogen ring atom.
125. A compound according to claim 122, wherein at least one R8 is selected from the group consisting of - H2, -NH(Cι.5 alkyl), -N(Cι-5 alkyl) 2, piperazine, imidazole, and pyridine.
126. A compound according to claim 112, wherein R2 is selected from the group consisting of 3-amino-piperidin-l-yl, 3-aminomethyl-pyrrolidin-l-yl, azetidin-1-yl, 3- aminoazetidin-1-yl, pyrrolidin- 1-yl, 3-aminocyclopent-l-yl, 3-aminomethylcyclopent-l-yl, 3-aminomethylcyclohex-l-yl, hexahydroazepin-1-yl, 3-aminohexahydroazepin-l-yl, 3- amino-cyclohex-1-yl, piperazin- 1-yl, homopiperazin- 1-yl, 3-amino-pyrrolidin-l-yl, and R- 3-aminopiperidin-l-yl, each substituted or unsubstituted.
127. A compound according to claim 112, wherein Z provides 1-3 atom separation between Rm and the ring.
128. A compound according to claim 112, wherein Z provides 1 atom separation between Rm and the ring.
129. A compound according to claim 128, wherein the 1 atom separation is provided by an atom selected from the group consisting of C, N, O, and S.
130. A compound according to claim 128, wherein the 1 atom separation is provided by a carbon atom.
131. A compound according to claim 128, wherein the 1 atom separation is provided by an oxygen atom.
132. A compound according to claim 128, wherein the 1 atom separation is provided by a nitrogen atom.
133. A compound according to claim 112, wherein Z is selected from the group consisting of -CH2-, -CH2CH2-, -CH2CH2CH2-, -C(O)-, -CH2C(O)-, -C(O)CH , -CH2-C(O)CH2-, -C(O)CH2CH2-, -CH2CH2C(O)-, -O-, -OCH2-, -CH2O-, -CH2OCH2-, -OCH2CH2-, -CH2CH2O-, -N(CH3)-, -NHCH2-, -CH2NH-, -CH2NHCH2-, -NHCH2CH2-, -CH2CH2NH-, -NH-C(O)-, -NCH3-C(O)-, -C(O)NH-, -C(O)NCH3-, -NHC(O)CH2-, -C(O)NHCH2-, -C(O)CH2NH-, -CH2NHC(O)-, -CH2C(O)NH-, -NHCH2C(O)-, -S-, -SCH2-, -CH2S-, -SCH2CH2-, -CH2SCH2-, -CH2CH2S-, -C(O)S-, -C(O)SCH2-, -CH2C(O)S-, -C(O)CH2S-, and -CH2SC(O)-, each substituted or unsubstituted.
134. A compound according to claim 112, wherein Z is selected from the group consisting of -CH2-, -C(O)-, -C(S)-, -C(NH)-, -C(NR9)-, -O-, -N(H)-, -N(R9)-, and -S-.
135. A compound according to claim 112, wherein Rm is a substituted or unsubstituted (C3-7)cycloalkyl.
136. A compound according to claim 112, wherein Rm is a substituted or unsubstituted aryl.
137. A compound according to claim 112, wherein Rm is a substituted or unsubstituted phenyl.
138. A compound according to claim 112, wherein Rm is selected from the group consisting of (2-cyano)phenyl, (3-cyano)phenyl, (2-hydroxy)phenyl, (3-hydroxy)phenyl, (2-alkenyl)phenyl, (3-alkenyl)phenyl, (2-alkynyl)phenyl, (3-alkynyl)phenyl, (2-nitro)phenyl, (3-nitro)phenyl, (2-carboxy)phenyl, (3-carboxy)phenyl, (2-carboxamido)phenyl, (3-carboxamido)phenyl, (2-suifonamido)phenyl, (3-sulfonamido)phenyl, (2-tetrazolyl)ρhenyl, (3-tetrazolyl)phenyl, (2-aminomethyl)ρhenyl, (3-aminomethyl)phenyl, (2-amino)phenyl, (3-amino)phenyl, (2-hydroxymethyl)phenyl, (3-hydroxymethyl)phenyl, (2-phenyl)phenyl, (3-phenyl)phenyl, (2-CONH2)phenyl, (3-CONH2)phenyl, (2-CONH(Ci-7)alkyl)phenyl, (3-CONH(Ci-7)alkyl)phenyl, (2-Cθ2(Cι-7)alkyl)phenyl, (3-CO2(Cι-7)alkyl)phenyl, -NH2, -OH, -(C3-7)alkyl, -alkene, -alkyne, -CCH, -(C3- )cycloalkyl, and -aryl, each substituted or unsubstituted.
139. A compound according to claim 112, wherein Ri is -ORn, where Rn is selected from the group consisting of substituted or unsubstituted alkyl, cycloalkyl, aryl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl.
140. A compound according to claim 112, wherein Z is a carbonyl.
141. A compound according to claim 112, wherein Ri is selected from the group consisting of -(CH2)-(2-cyano)phenyl, -(CH2)-(3-cyano)phenyl, -(CH2)-(2-hydroxy)phenyl, -(CH2)-(3-hydroxy)phenyl, -(CH2)-(2-alkenyl)phenyl, -(CH2)-(3-alkenyl)phenyl, -(CH2)-(2-alkynyl)phenyl, -(CH2)-(3-alkynyl)phenyl, -(CH2)-(2-nitro)phenyl, -(CH2)-(3- nitro)phenyl, -(CH2)-(2-carboxy)phenyl, -(CH2)-(3-carboxy)phenyl, -(CH2)-(2-carboxamido)phenyl, -(CH2)-(3-carboxamido)phenyl, -(CH2)-(2-sulfonamido)ρhenyl, -(CH2)-(3-sulfonamido)ρhenyl, -(CH2)-(2- tetrazolyl)phenyl, -(CH2)-(3-tetrazolyl)ρhenyl, -(CH2)-(2-aminomemyι)ρhenyl, -(CH2)~(3- aminomethyl)phenyl, -(CH2)-(2-amino)phenyl, -(CH2)-(3-amino)phenyl, -(CH2)-(2-hydroxymethyl)phenyl, -(CH2)-(3-hydroxymethyl)phenyl, -(CH2)-(2- ρhenyl)ρhenyl, -(CH2)-(3-ρhenyl)phenyl, -(CH2)-(2-CONH2)phenyl, -(CH2)-(3-CONH2)phenyl, -(CH2)-(2-CONH(Ci-7)alkyl)phenyl, -(CH2)-(3-CONH(Cι-7)alkyl)phenyl, -(CH2)-(2-Cθ2(Cι-7)alkyl)phenyl, -(CH2)-(3-CO2(Ci-7)alkyl)phenyl, -CH2-NH2, -CH2-OH, -CH2-(C3-7)alkyl, -CH2-alkene, -CH2-alkyne, -CH2-CCH, -CH2-(C3-7)cycloalkyl, and -CH2-aryl, each substituted or unsubstituted.
142. A compound according to claim 112, wherein Ri is selected from the group consisting of -(Ci)alkyl-aryl, -(Cι)alkyl-bicycloaryl, -aminoaryl, -aminoheteroaryl, - aminobicycloaryl, -aminoheterobicycloaryl, -O-aryl, -O-heteroaryl, -O-bicycloaryl, -O- heterobicycloaryl, -(S)-aryl, -(S)-heteroaryl, -(S)-bicycloaryl, -S-heterobicycloaryl, -C(O)- aryl, -C(O)-heteroaryl, - C(O)-bicycloaryl, - C(O)-heterobicycloaryl, -C(S)-aryl, -C(S)- heteroaryl, -C(S)-bicycloaryl, -C(S)-heterobicycloaryl, -S(O)-aryl, -S(O)-heteroaryl, -S(O)- bicycloaryl, -Sθ2-heterobicycloaryl, -SO2-aryl, -Sθ2-heteroaryl, -Sθ2-bicycloaryl, -SO2- heterobicycloaryl, -C(NR9)-aryl, -C(NR9)-heteroaryl, -C(NR9)-bicycloaryl, -C(NR9)- heterobicycloaryl, each substituted or unsubstituted.
' 143. A compound according to claim 112, where R3 and t are taken together to form an unsubstituted or substituted 5 or 6 membered cycloalkyl or heterocycloalkyl ring.
144. A compound according to claim 112, where R3 and Rt are taken together to form a substituted or unsubstituted phenyl ring.
145. A compound according to claim 112, where R3 and R4 are taken together to form a substituted or unsubstituted heteroaryl ring.
146. A compound according to claim 112, where R3 and t are taken together to form a substituted or unsubstituted heteroaryl selected from the group consisting of substituted or unsubstituted furan, thiophene, pyrrole, pyrazole, triazole, isoxazole, oxazole, thiazole, isothiazole, oxadiazole, pyridine, pyridazine, pyrimidine, pyrazine, triazine, benzofuran, isobenzofuran, benzothiophene, isobenzothiophene, imidazole, benzimidazole, indole, isoindole, quinoline, isoquinoline, cinnoline, quinazoline, naphthyridine, pyridopyridine, quinoxaline, phthalazine, and benzothiazole.
147. A compound according to claim 112, where R3 and t are taken together to form a 5 or 6-membered ring where the ring comprises at least one CO group.
148. A compound according to claim 112, where R3 and R4 are taken together to form a 5 or 6-membered ring comprising of 1-3 nitrogen ring atoms.
149. A compound according to claim 112, where R and R are taken together to form a 5 or 6-membered ring where the ring comprises a sulfur atom.
150. A compound according to claim 149, wherein the ring sulfur atom is in an oxidized form as SO or SO2.
151. A compound according to claim 112, wherein the ring formed by R3 and φ comprises substituents that form a ring fused to the ring formed by R3 and Rt.
152. A compound according to claim 112, wherein R3 and t are taken together to form a ring system such that the compound of Formula XIX formed is selected from the group consisting of substituted or unsubstituted 4-oxo-4H-quinazoline, 3H-pyrido[2,3- d]pyrimidin-4-one, 3H-pyrido [3 ,2-d]pyrimidin-4-one, 3H-pyrido[3 ,4-d]pyrimidin-4-one and 3H-pyrido[4,3-d]pyrimidin-4-one.
153. A compound comprising Formula XX:
Figure imgf000201_0001
XX wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NR9;
J, K, L, and M are each independently selected from the group of CR12 and N;
Ri is -ZRm, where Z is a moiety providing 1-6 atom separation between Rm and the ring to which Ri is attached, and -Rm is selected from the group consisting of a substituted or unsubstituted (C3-7)cycloalkyl and aryl;
R2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein;
R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted; and each R12 is hydrogen or is independently selected from the group consisting of halo, ρerhalo(Cι-ιo)alkyl, CF3, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, alkoxy, a carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
154. A compound according to claim 153, wherein the compound is a compound where J, K, L and M each comprise a carbon ring atom.
155. A compound according to claim 153, wherein the compound is a compound where J comprises a nitrogen ring atom.
156. A compound according to claim 153, wherein the compound is a compound where K comprises a nitrogen ring atom.
157. A compound according to claim 153, wherein the compound is a compound where L comprises a nitrogen ring atom.
158. A compound according to claim 153, wherein the compound is a compound where M comprises a nitrogen ring atom.
159. A compound according to claim 153, wherein the compound is a compound where J and L each comprise a nitrogen ring atom or J and K each comprise a nitrogen ring atom.
160. A compound according to claim 153, wherein the compound is a compound where K and L each comprise a nitrogen ring atom.
161. A compound according to claim 153, wherein the compound is a compound where K and M each comprise a nitrogen ring atom.
162. A compound according to claim 153, wherein the compound is a compound where J and M each comprise a nitrogen ring atom or L and M each comprise a nitrogen ring atom.
163. A compound according to claim 153, wherein at least two of J, K, L and M comprise a nitrogen ring atom.
164. A compound according to claim 153, wherein at least three of J, K, L and M comprise a nitrogen ring atom.
165. A compound according to claim 153, wherein the ring formed by J, K, L, and M comprises substituents that form a ring fused to or bridged to the ring formed by J, K, L, and M.
166. A compound according to claim 153, wherein K is CR12, where Ri2 is independently selected from the group consisting of halo, perhalo(Ci-io)alkyl, CF3, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, aikoxy, a carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
167. A compound according to claim 153, wherein K is CR12, where Rι2 is independently selected from the group consisting of halo, perhalo(Ci-io)alkyl, CF3, cyano, nitro, alkyl, aryloxy, heteroaryloxy, amino, and alkoxy, each substituted or unsubstituted.
168. A compound according to claim 153, wherein K is CR12, where R12 is independently selected from the group consisting of heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryl, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, thio, a carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
169. A compound according to claim 153, wherein K is CR12, where Rι2 is independently selected from the group consisting of chloro, bromo, fluoro, iodo, methoxy, morpholin-4-yl, and pyrrolidin- 1-yl, each substituted or unsubstituted.
170. A compound according to claim 153, wherein L is CR12, where R12 is independently selected from the group consisting of halo, perhalo(Cι-ιo)alkyl, CF3, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, alkoxy, a carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
171. A compound according to claim 153, wherein L is CR12, where R12 is independently selected from the group consisting of halo, perhalo(Cι-ιo)alkyl, CF3, cyano, nitro, alkyl, aryloxy, heteroaryloxy, amino, morpholin-4-yl, and pyrrolidin- 1-yl, and alkoxy, each substituted or unsubstituted.
172. A compound according to claim 153, wherein and L are independently CR12, where Rι2 is independently selected from the group consisting of halo, perhalo(Cι-iQ)alkyl, CF3, cyano, nitro, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, a carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
173. A compound according to claim 153, wherein:
K is CR12, where Rι2 is independently selected from the group consisting of halo, perhalo(Cι-ιo)alkyl, CF3, cyano, nitro, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, a carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted; and
L is nitrogen.
174. A compound comprising a member selected from the group consisting of Formulae XXIa, XXIb, XXIc, XXId, XXIe and XXIf :
Figure imgf000205_0001
wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NR9;
Ri is -ZRm, where Z is a moiety providing 1-6 atom separation between Rm and the ring to which Ri is attached, and -Rm is selected from the group consisting of a substituted or unsubstituted (C3- )cycloalkyl and aryl;
R2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein;
R is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted; and each Rι9 is independently selected from the group consisting of hydrogen, halo, perhalo(Cι-ιo)alkyl, CF3, cyano, nitro, alkyl, alkene, alkyne, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, with the proviso that Rι9 is not alkylthio, arylthio, halo, cyano, nitro, and thio in the case where the ring atom to which Rι9 is bound is nitrogen.
175. A compound according to claim 174, wherein two Rι are taken together to form a substituted or unsubstituted fused or bridged ring.
176. A compound comprising Formula XXII:
Figure imgf000206_0001
wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NR ;
W, X, and Y are each independently selected from the group of moieties where the ring atom is either C, N, O or S;
Ri is -ZRm, where Z is a moiety providing 1-6 atom separation between Rm and the ring to which Ri is attached, and -Rm is selected from the group consisting of a substituted or unsubstituted (C3-7)cycloalkyl and aryl;
R2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein;
R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
177. A compound according to claim 176, wherein at least one of W, X, and Y is CO.
178. A compound according to claim 176, wherein at least one of W, X, and Y is SO.
179. A compound according to claim 176, wherein at least one of W, X, and Y is SO2.
180. A compound according to claim 176, wherein at least one of W, X, and Y comprises a ring nitrogen atom.
181. A compound according to claim 176, wherein at least two of W, X, and Y comprises a ring nitrogen atom.
182. A compound according to claim 176, wherein W and Y are taken together to form a substituted or unsubstituted bridged ring relative to the ring formed by W, X and Y.
183. A compound according to claim 176, wherein two of W, X, and Y are taken together to form a substituted or unsubstituted ring fused to the ring formed by W, X and Y.
184. A compound comprising a member selected from the group consisting of Formulae XXHIa, XXmb or XXHIc:
Figure imgf000207_0001
wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NR9;
Ri is -ZRm, where Z is a moiety providing 1-6 atom separation between Rm and the ring to which Ri is attached, and -Rm is selected from the group consisting of a substituted or unsubstituted (C3-7)cyclόalkyl and aryl; R2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein;
R is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted; and each Rι9 is independently selected from the group consisting of hydrogen, halo, perhalo(Cι-ιo)alkyl, CF3, cyano, nitro, alkyl, alkene, alkyne, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, with the proviso that Rι9 is not alkylthio, arylthio, halo, cyano, nitro, and thio in the case where the ring atom to which Rι9 is bound is nitrogen.
185. A compound according to claim 184, wherein two Rι9 are taken together to form a substituted or unsubstituted bridged or spiro ring.
186. A compound comprising Formula XXIVa or Formula XXIVb:
Figure imgf000208_0001
XXTVa wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NR ;
W, X, and Y are each independently selected from the group of moieties where the ring atom is either C, N, O or S;
Ri is -ZRm, where Z is a moiety providing 1-6 atom separation between Rm and the ring to which Ri is attached, and -Rm is selected from the group consisting of a substituted or unsubstituted (C3- )cycloalkyl and aryl; R2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein; and
R is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
187. A compound according to claim 186, wherein the compound is a compound of Formula XXTVa wherein Y is selected from the group consisting of CO, SO or SO2.
188. A compound according to claim 186, wherein the compound is a compound of Formula XXT b wherein W is selected from the group consisting of CO, SO or SO2.
189. A compound according to claim 186, wherein W comprise a ring nitrogen atom.
190. A compound according to claim 186, wherein X comprise a ring nitrogen atom.
191. A compound according to claim 186, wherein Y comprise a ring nitrogen atom.
192. A compound according to claim 186, wherein at least two of W, X, and Y comprises a ring nitrogen atom.
193. A compound according to claim 186, wherein two of W, X, and Y are taken together and substituted through available valencies to form a substituted or unsubstituted ring fused or bridged to the ring formed by , X and Y.
194. A compound according to claim 186, wherein , X, and Y are selected such that the compound comprises a ring system selected from the group consisting of 4-oxo-4H- thieno [3 ,2-d]pyrimidine, 7-oxo- 1 ,2,3 ,7-tetrahydro-8-thia-4,6-diaza-cyclopenta[a]indene, 7- methyl-6-oxo-6,7-dihydro-purine, and 6-oxo-6,9-dihydro-purine, each substituted or unsubstituted.
195. A compound comprising Formulae XXVa, XXVb, or XXVc:
Figure imgf000210_0001
wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NR9;
X is selected from the group of moieties where the ring atom is either C, N, O or S in Formula XXVa, or X is selected from the group of moieties where the ring atom is either C or N in Formula XXVb or Formula XXVc;
Ri is -ZRm, where Z is a moiety providing 1-6 atom separation between Rm and the ring to which Ri is attached, and -Rm is selected from the group consisting of a substituted or unsubstituted (C3-7)cycloalkyl and aryl;
R2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein;
R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted; and each Rι9 is independently selected from the group consisting of hydrogen, halo, perhalo(Cι-ιo)alkyl, CF3, cyano, nitro, alkyl, alkene, alkyne, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, with the proviso that R19 is not alkylthio, arylthio, halo, cyano, nitro, and thio in the case where the ring atom to which Rι9 is bound is nitrogen.
196. A compound according to claim 195, wherein two Rι are taken together to form a substituted or unsubstituted ring.
197. A compound according to claim 195, wherein the compound comprises Formula XXVa and the two Rι9 are taken together to form a substituted or unsubstituted fused or bridged ring.
198. A compound comprising a member selected from the group of Formulae XXVIa, XXVIb and XXVIc:
Figure imgf000211_0001
XXVIa XXVIb XXVIc
wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NR9;
J, K, L, and M are each independently selected from the group of moieties where the ring atom is either C, N, O or S;
Ri is -ZRm, where Z is a moiety providing 1-6 atom separation between Rra and the ring to which Ri is attached, and -Rm is selected from the group consisting of a substituted or unsubstituted (C3- )cycloalkyl and aryl; and
R2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein.
199. A compound according to claim 198, wherein the compound is a compound where J, K, L and M each comprise a carbon ring atom.
200. A compound according to claim 198, wherein at least one of J, K, L and M comprise a nitrogen ring atom.
201. A compound according to claim 198, wherein the compound is a compound where J and K each comprise a nitrogen ring atom or J and L each comprise a nitrogen ring atom.
202. A compound according to claim 198, wherein the compound is a compound where K and L each comprise a nitrogen ring atom or K and M each comprise a nitrogen atom.
203. A compound according to claim 198, wherein the compound is a compound where J and M each comprise a nitrogen ring atom or L and M each comprise a nitrogen ring atom.
204. A compound according to claim 198, wherein at least two of J, K, L and M comprise a nitrogen ring atom.
205. A compound according to claim 198, wherein at least three of J, K, L and M comprise a nitrogen ring atom.
206. A compound according to claim 198, wherein at least one of J, K, L and M is CO.
207. A compound according to claim 198, wherein at least one of J, K, L and M is SO.
208. A compound according to claim 198, wherein at least one of J, K, L and M is SO2.
209. A compound according to claim 198, wherein the ring formed by J, K, L, and M comprises substituents, through available valencies, that form a ring fused to the ring formed by J, K, L, and M or, in the case of Formula XXVIb, J and M form a bridged ring relative to the ring formed by J, K, L, and M.
210. A compound comprising Formula XXVII:
Figure imgf000213_0001
xxvπ wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NR9;
J, K, L, and M are each independently selected from the group of moieties where the ring atom is either C, N, O or S;
Ri is -ZRm, where Z is a moiety providing 1-6 atom separation between Rm and the ring to which Ri is attached, and -Rm is selected from the group consisting of a substituted or unsubstituted (C3- )cycloalkyl and aryl;
R2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein; and
R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
211. A compound according to claim 210, wherein at least one of J, K, L and M is CO.
212. A compound according to claim 210, wherein at least one of J, K, L and M is SO.
213. A compound according to claim 210, wherein at least one of J, K, L and M is SO2.
214. A compound according to claim 210, wherein the compound is a compound where J, K, L and M each comprise a carbon ring atom.
215. A compound according to claim 210, wherein the compound is a compound where J comprises a nitrogen ring atom.
216. A compound according to claim 210, wherein the compound is a compound where K comprises a nitrogen ring atom.
217. A compound according to claim 210, wherein the compound is a compound where L comprises a nitrogen ring atom.
218. A compound according to claim 210, wherein the compound is a compound where M comprises a nitrogen ring atom.
219. A compound according to claim 210, wherein the compound is a compound where J and K each comprise a nitrogen ring atom or J and L each comprise a nitrogen ring atom.
220. A compound according to claim 210, wherein the compound is a compound where K and L each comprise a nitrogen ring atom or K and M each comprise a nitrogen atom.
221. A compound according to claim 210, wherein the compound is a compound where J and M each comprise a nitrogen ring atom or L and M each comprise a nitrogen ring atom.
222. A compound according to claim 210, wherein at least two of J, K, L and M comprise a nitrogen ring atom.
223. A compound according to claim 210, wherein at least three of J, K, L and M comprise a nitrogen ring atom.
224. A compound according to claim 210, wherein the ring formed by J, K, L, and M comprises substituents that form a ring fused to the ring formed by J, K, L, and M.
225. A compound according to claim 210, wherein the ring formed by J, K, L, and M comprises substituents that form a bridged ring relative to the ring formed by J, K, L, and M.
226. A compound comprising Formula XXVHI:
Figure imgf000215_0001
XXVIII wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NR9;
Ri is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring;
R2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein;
R3 and R4 are taken together to form a substituted or unsubstituted 5 or 6 membered ring; and
R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
227. A compound according to claim 226, wherein U provides 1-4 atom separation between V and the ring.
228. A compound according to claim 226, wherein U provides 1-3 atom separation between V and the ring.
229. A compound according to claim 226, wherein U is selected from the group consisting of -CH2-, -CH2CH2-, -CH2CH2CH2-, -C(O)-, -CH2C(O)-, -C(O)CH2-, -CH2-C(O)CH2-, -C(O)CH2CH2-, -CH2CH2C(O)-, -O-, -OCH2-, -CH2O-, -CH2OCH2-, -OCH2CH2-, -CH2CH2O-, -N(CH3)-, -NHCH2-, -CH2NH-, -CH2NHCH2-, -NHCH2CH2-, -CH2CH2NH-, -NH-C(O)-, -NCH3-C(O)-, -C(O)NH-, -C(O)NCH3-, -NHC(O)CH2-, -C(O)NHCH2-, -C(O)CH2NH-, -CH2NHC(O)-, -CH2C(O)NH-, -NHCH2C(O)-, -S-, -SCH2-, -CH2S-, -SCH2CH2-, -CH2SCH2-, -CH2CH2S-, -C(O)S-, -C(O)SCH2-, -CH2C(O)S-, -C(O)CH2S-, and -CH2SC(O)-, each substituted or unsubstituted.
230. A compound according to claim 226, wherein U is selected from the group consisting of -CH2-, -CHR9-, -C(R9)(R9)-, -O-, -N(H)-, -N(R9)-, and -S-.
231. A compound according to claim 226, wherein V is selected from the group consisting of a primary, secondary or tertiary amine, a heterocycloalkyl comprising a nitrogen ring atom, and a heteroaryl comprising a nitrogen ring atom.
232. A compound according to claim 226, wherein the basic nitrogen of V is separated from the ring atom to which R2 is attached by between 1-5 atoms.
233. A compound according to claim 226, wherein the basic mtrogen of V forms part of a primary, secondary or tertiary amine.
234. A compound according to claim 226, wherein the basic nitrogen of V is a nitrogen ring atom of a heterocycloalkyl comprising a nitrogen ring atom or a heteroaryl comprising a nitrogen ring atom.
235. A compound according to claim 226, wherein Ri is a substituted or unsubstituted aryl.
236. A compound according to claim 226, wherein Ri is a substituted or unsubstituted phenyl.
237. A compound according to claim 226, wherein Ri is a substituted or unsubstituted heteroaryl.
238. A compound comprising Formula XXIX:
Figure imgf000217_0001
XXIX wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NR ;
J, K, L, and M are each independently selected from the group of CRι2 and N;
Ri is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring;
R2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein;
R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted; and each Ri2 is hydrogen or is independently selected from the group consisting of halo, perhalo(Ci-io)alkyl, CF3, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, alkoxy, a carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
239. A compound according to claim 238, wherein the compound is a compound where J, K, L and M each comprise a carbon ring atom.
240. A compound according to claim 238, wherein the compound is a compound where J comprises a nitrogen ring atom.
241. A compound according to claim 238, wherein the compound is a compound where K comprises a nitrogen ring atom.
242. A compound according to claim 238, wherein the compound is a compound where L comprises a nitrogen ring atom.
243. A compound according to claim 238, wherein the compound is a compound where M comprises a nitrogen ring atom.
244. A compound according to claim 238, wherein the compound is a compound where J and L each comprise a nitrogen ring atom or J and K each comprise a nitrogen ring atom.
245. A compound according to claim 238, wherein the compound is a compound where K and L each comprise a nitrogen ring atom.
246. A compound according to claim 238, wherein the compound is a compound where K and M each comprise a nitrogen ring atom.
247. A compound according to claim 238, wherein the compound is a compound where J and M each comprise a nitrogen ring atom or L and M each comprise a nitrogen ring atom.
248. A compound according to claim 238, wherein at least two of J, K, L and M comprise a nitrogen ring atom.
249. A compound according to claim 238, wherein at least three of J, K, L and M comprise a nitrogen ring atom.
250. A compound according to claim 238, wherein the ring formed by J, K, L, and M comprises substituents that form a ring fused to or bridged to the ring formed by J, K, L, and M.
251. A compound according to claim 238, wherein K is CRι2, where Rι2 is independently selected from the group consisting of halo, perhalo(Cι-ι0)alkyl, CF3, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, alkoxy, a carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
252. A compound according to claim 238, wherein K is CRι2, where R12 is independently selected from the group consisting of halo, perhalo(Cι-ιo)alkyl, CF3, cyano, nitro, alkyl, aryloxy, heteroaryloxy, amino, and alkoxy, each substituted or unsubstituted.
253. A compound according to claim 238, wherein K is CRι2, where Rι2 is independently selected from the group consisting of heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryl, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, thio, a carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
254. A compound according to claim 238, wherein K is CR12, where R12 is independently selected from the group consisting of chloro, bromo, fluoro, iodo, methoxy, morpholin-4-yl, and pyrrolidin- 1-yl, each substituted or unsubstituted.
255. A compound according to claim 238, wherein L is CR12, where R12 is independently selected from the group consisting of halo, perhalo(Ci-i0)alkyl, CF3, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, alkoxy, a carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
256. A compound according to claim 238, wherein L is CR12, where Rι2 is independently selected from the group consisting of halo, perhalo(Cι-ιo)alkyl, CF3, cyano, nitro, alkyl, aryloxy, heteroaryloxy, amino, morpholin-4-yl, and pyrrolidin- 1-yl, and alkoxy, each substituted or unsubstituted.
257. A compound according to claim 238, wherein K and L are independently CR12, where Rι2 is independently selected from the group consisting of halo, perhalo(Ci-i0)alkyl, CF3, cyano, nitro, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, a carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
258. A compound according to claim 238, wherein:
K is CRι2, where Rι2 is independently selected from the group consisting of halo, perhalo(Ci-io)alkyl, CF3, cyano, nitro, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, a carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted; and
L is nitrogen.
259. A compound comprising a member selected from the group consisting of Formulae XXXa, XXXb, XXXc, XXXd, XXXe and XXXf:
Figure imgf000221_0001
wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NR ;
Ri is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring;
R2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein;
R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted; and each Rι9 is independently selected from the group consisting of hydrogen, halo, perhalo(Ci-io)alkyl, CF3, cyano, nitro, alkyl, alkene, alkyne, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, with the proviso that Rι9 is not alkylthio, arylthio, halo, cyano, nitro, and thio in the case where the ring atom to which Rι9 is bound is nitrogen.
260. A compound according to claim 259, wherein two Rι9 are taken together to form a substituted or unsubstituted fused or bridged ring.
261. A compound comprising Formula XXXI:
Figure imgf000222_0001
wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NR9;
W, X, and Y are each independently selected from the group of moieties where the ring atom is either C, N, O or S;
Ri is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring;
R2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein; and
R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
262. A compound according to claim 261, wherein at least one of W, X, and Y is CO.
263. A compound according to claim 261, wherein at least one of W, X, and Y is SO.
264. A compound according to claim 261 , wherein at least one of W, X, and Y is SO2.
265. A compound according to claim 261, wherein at least one of W, X, and Y comprises a ring nitrogen atom.
266. A compound according to claim 261, wherein at least two of W, X, and Y comprises a ring nitrogen atom.
267. A compound according to claim 261, wherein W and Y are taken together to form a substituted or unsubstituted bridged ring relative to the ring formed by W, X and Y.
268. A compound according to claim 261 , wherein two of W, X, and Y are taken together to form a substituted or unsubstituted ring fused to the ring formed by W, X and Y.
269. A compound comprising a member selected from the group consisting of Formulae XXXTIa, XXXHb or XXXIIc:
Figure imgf000223_0001
wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NR9;
Ri is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring;
R2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein; R is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted; and each Ri9 is independently selected from the group consisting of hydrogen, halo, perhalo(Ci-io)alkyl, CF3, cyano, nitro, alkyl, alkene, alkyne, aryl,.heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, with the proviso that Rι9 is not alkylthio, arylthio, halo, cyano, nitro, and thio in the case where the ring atom to which Rι9 is bound is nitrogen.
270. A compound according to claim 269, wherein two Rι9 are taken together to form a substituted or unsubstituted bridged or spiro ring.
271. A compound comprising Formula XXXπia or Formula XXXmb :
Figure imgf000224_0001
wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NR9;
W, X, and Y are each independently selected from the group of moieties where the ring atom is either C, N, O or S;
Ri is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring;
R2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein; and R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
272. A compound according to claim 271 , wherein the compound is a compound of Formula XXXπia wherein Y is selected from the group consisting of CO, SO or SO2.
273. A compound according to claim 271 , wherein the compound is a compound of Formula XXXIIIb wherein W is selected from the group consisting of CO, SO or SO2.
274. A compound according to claim 271 , wherein W comprise a ring nitrogen atom.
275. A compound according to claim 271 , wherein X comprise a ring nitrogen atom.
276. A compound according to claim 271 , wherein Y comprise a ring nitrogen atom.
277. A compound according to claim 271 , wherein at least two of W, X, and Y comprises a ring nitrogen atom.
278. A compound according to claim 271 , wherein two of W, X, and Y are taken together and substituted through available valencies to form a substituted or unsubstituted ring fused or bridged to the ring formed by W, X and Y.
279. A compound according to claim 271 , wherein W, X, and Y are selected such that the compound comprises a ring system selected from the group consisting of 4-oxo-4H- thieno [3 ,2-d]pyrimidine, 7-oxo- 1 ,2,3 ,7-tetrahydro-8-thia-4,6-diaza-cyclopenta[a]indene, 7- methyl-6-oxo-6,7-dihydro-purine, and 6-oxo-6,9-dihydro-purine, each substituted or unsubstituted.
280. A compound comprising Formulae XXXIVa, XXXIVb, or XXXIVc:
Figure imgf000226_0001
wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NR9;
X is selected from the group of moieties where the ring atom is either C, N, O or S in Formula XXTVa, or X is selected from the group of moieties where the ring atom is either C or N in Formula XXXTVb or Formula XXXTVc;
Ri is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring;
R2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein;
R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted; and each Rι9 is independently selected from the group consisting of hydrogen, halo, perhalo(Cι-ιo)alkyl, CF3, cyano, nitro, alkyl, alkene, alkyne, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, with the proviso that Rι9 is not alkylthio, arylthio, halo, cyano, nitro, and thio in the case where the ring atom to which Rι9 is bound is nitrogen.
281. A compound according to claim 280, wherein two Rι9 are taken together to form a substituted or unsubstituted ring.
282. A compound according to claim 280, wherein the compound comprises Formula XXXIVa and the two Ri9 are taken together to form a substitated or unsubstituted fused or bridged ring.
283. A compound comprising a member selected from the group of Formulae XXXVa, XXXVb and XXXVc:
Figure imgf000227_0001
XXXVa XXXVb XXXVc wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NR9;
J, K, L, and M are each independently selected from the group of moieties where the ring atom is either C, N, O or S;
Ri is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring; and
R2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein.
284. A compound according to claim 283, wherein the compound is a compound where J, K, L and M each comprise a carbon ring atom.
285. A compound according to claim 283, wherein at least one of J, K, L and M comprise a nitrogen ring atom.
286. A compound according to claim 283, wherein the compound is a compound where J and K each comprise a nitrogen ring atom or J and L each comprise a nitrogen ring atom.
287. A compound according to claim 283, wherein the compound is a compound where K and L each comprise a nitrogen ring atom or K and M each comprise a nitrogen atom.
288. A compound according to claim 283, wherein the compound is a compound where J and M each comprise a nitrogen ring atom or L and M each comprise a nitrogen ring atom.
289. A compound according to claim 283, wherein at least two of J, K, L and M comprise a nitrogen ring atom.
290. A compound according to claim 283, wherein at least three of J, K, L and M comprise a nitrogen ring atom.
291. A compound according to claim 283, wherein at least one of J, K, L and M is CO.
292. A compound according to claim 283, wherein at least one of J, K, L and M is SO.
293. A compound according to claim 283, wherein at least one of J, K, L and M is SO2.
294. A compound according to claim 283, wherein the ring formed by J, K, L, and M comprises substituents, through available valencies, that form a ring fused to the ring formed by J, K, L, and M or, in the case of Formula XXXVb, J and M form a bridged ring relative to the ring formed by J, K, L, and M.
295. A compound comprising Fonnula XXXVI:
Figure imgf000228_0001
XXXVI wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NR9; J, K, L, and M are each independently selected from the group of moieties where the ring atom is either C, N, O or S;
Ri is selected from the group consisting of a substitated or unsubstituted 3, 4, 5, 6 or 7 membered ring;
R2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein; and
R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substitated or unsubstituted.
296. A compound according to claim 295, wherein at least one of J, K, L and M is CO.
297. A compound according to claim 295, wherein at least one of J, K, L and M is SO.
298. A compound according to claim 295, wherein at least one of J, K, L and M is SO2.
299. A compound according to claim 295, wherein the compound is a compound where J, K, L and M each comprise a carbon ring atom.
300. A compound according to claim 295, wherein the compound is a compound where J comprises a nitrogen ring atom.
301. A compound according to claim 295, wherein the compound is a compound where K comprises a nitrogen ring atom.
302. A compound according to claim 295, wherein the compound is a compound where L comprises a nitrogen ring atom.
303. A compound according to claim 295, wherein the compound is a compound where M comprises a nitrogen ring atom.
304. A compound according to claim 295, wherein the compound is a compound where J and K each comprise a nitrogen ring atom or J and L each comprise a nitrogen ring atom.
305. A compound according to claim 295, wherein the compound is a compound where K and L each comprise a nitrogen ring atom or K and M each comprise a nitrogen atom.
306. A compound according to claim 295, wherein the compound is a compound where J and M each comprise a nitrogen ring atom or L and M each comprise a nitrogen ring atom.
307. A compound according to claim 295, wherein at least two of J, K, L and M comprise a nitrogen ring atom.
308. A compound according to claim 295, wherein at least three of J, K, L and M comprise a nitrogen ring atom.
309. A compound according to claim 295, wherein the ring formed by J, K, L, and M comprises substituents that form a ring fused to the ring formed by J, K, L, and M.
310. A compound according to claim 295, wherein the ring formed by J, K, L, and M comprises substituents that form a bridged ring relative to the ring formed by J, K, L, and M.
311. A compound selected from the group consisting of: 2-Aminomethyl-3-phenyl-3H-quinazolin-4-one; 2-Etaylaminomethyl-3-phenyl-3H-quinazolin-4-one; [(4-Oxo-3-phenyl-3,4-dihydro-quinazolin-2-ylmethyl)-amino]-acetic acid methyl ester;
[(4-Oxo-3-phenyl-3,4-dihydro-quinazolin-2-ylmethyl)-amino]-acetic acid; 2-Aminomethyl-3-(2,4-dichloro-phenyl)-3H-quinazolin-4-one; 2-An nomemyl-3-(2-cUoro-phenyl)-3H-quinazolin-4-one; and 2-Aminomemyl-3-(4-chloro-ρhenyl)-3H-quinazolin-4-one.
312. A compound comprising Formula XXXVII:
Figure imgf000231_0001
xxxvπ wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NR9;
J, K, L, and M are each independently selected from the group of CR12 and N, provided that at least one of K and L is CRι2 where Rι2 is not hydrogen;
Ri is -ZRm, where Z is a moiety providing 1-6 atom separation between Rm and the ring to which Ri is attached, and Rm is selected from the group consisting of a substitated or unsubstituted (C3-7)cycloalkyl and aryl;
R2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein;
R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted; and each Rι2 is hydrogen or is independently selected from the group consisting of halo, perhalo(Cι-ιo)alkyl, CF3, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, alkoxy, a carbonyl group, imine group, sulfonyl group and sulfinyl group, each substitated or unsubstituted.
313. A compound according to claim 312, wherein V is selected from the group consisting of a primary, secondary or tertiary amine, a heterocycloalkyl comprising a nitrogen ring atom, and a heteroaryl comprising a nitrogen ring atom
314. A compound according to claim 312, wherein V is selected from the group consisting of a substituted or unsubstituted 3, 4, 5, 6 or 7 membered ring wherein at least one substituent is selected from the group consisting of a primary, secondary or tertiary amine, a heterocycloalkyl comprising a nitrogen ring atom, and a heteroaryl comprising a nitrogen ring atom.
315. A compound according to claim 312, wherein the basic nitrogen of V is separated from the ring atom to which R2 is attached by between 1-5 atoms.
316. A compound according to claim 312, wherein the basic nitrogen of V forms part of a primary, secondary or tertiary amine.
317. A compound according to claim 312, wherein the basic nitrogen of V is a nitrogen ring atom of a heterocycloalkyl comprising a nitrogen ring atom or a heteroaryl comprising a nitrogen ring atom.
318. A compound according to claim 312, wherein R2 is selected from the group consisting of
Figure imgf000232_0001
wherein p is 0-12 and each R8 is independently selected from the group consisting of halo, ρerhalo(Cι-ιo)alkyl, CF3, cyano, nitro, hydroxy, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, with the proviso that at least one R8 serves as V.
319. A compound according to claim 318, wherein at least one R8 is a primary, secondary or tertiary amine.
320. A compound according to claim 318, wherein at least one R8 is a substituted or unsubstituted heterocycloalkyl comprising a nitrogen ring atom or a substituted or unsubstituted heteroaryl comprising a nitrogen ring atom.
321. A compound according to claim 318, wherein at least one R8 is selected from the group consisting of -NH2, -NH(Cι-5 alkyl), -N(Cι-5 alkyl) 2, piperazine, imidazole, and pyridine.
322. A compound according to claim 312, wherein R2 is selected from the group consisting of
Figure imgf000233_0001
wherein r is 0-13 and each R8 is independently selected from the group consisting of halo, perhalo(Ci-io)alkyl, CF3, cyano, nitro, hydroxy, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substitated or unsubstitated, with the proviso that at least one R8 serves as V.
323. A compound according to claim 322, wherein at least one R8 is a primary, secondary or tertiary amine.
324. A compound according to claim 322, wherein at least one R8 is a substitated or unsubstituted heterocycloalkyl comprising a nitrogen ring atom or a substitated or unsubstitated heteroaryl comprising a nitrogen ring atom.
325. A compound according to claim 322, wherein at least one R8 is selected from the group consisting of -NH2, -NH(Cι-5 alkyl), -N(Cι-5 alkyl) 2, piperazine, imidazole, and pyridine.
326. A compound according to claim 312, wherein R2 is selected from the group consisting of 3-amino-piperidin-l-yl, 3-aminomethyl-pyπolidin-l-yl, azetidin-1-yl, 3- aminoazetidin-1-yl, pyrrolidin- 1-yl, 3-aminocyclopent-l-yl, 3-aminomethylcyclopent-l-yl, 3-aminomethylcyclohex-l-yl, hexahydroazepin-1-yl, 3-aminohexahydroazepin-l-yl, 3- amino-cyclohex-1-yl, piperazin- 1-yl, homopiperazin- 1-yl, 3-amino-pyrrolidin-l-yl, and R- 3-aminopiperidin-l-yl, each substitated or unsubstitated.
327. A compound according to claim 312, wherein R2 is selected from the group consisting of a substitated or unsubstitated 3, 4, 5, 6 or 7 membered ring.
328. A compound according to claim 327, wherein Ri is -ZRm, where Z is a moiety providing 1-6 atom separation between Rm and the ring to which Ri is attached, and Rm is selected from the group consisting of a substituted or unsubstituted (C3- )cycloalkyl and aryl.
329. A compound according to claim 312, wherein Ri is -ZRm, where Z is a moiety providing 1-6 atom separation between Rm and the ring to which Ri is attached, and Rm is selected from the group consisting of a substituted or unsubstituted (C3- )cycloalkyl and aryl.
330. A compound according to claim 312, wherein at least one Rι2 is halogen.
331. A compound according to claim 312, wherein at least one Rι2 is fluorine.
332. A compound comprising Formula XXXVHI:
Figure imgf000235_0001
xxxvπi wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or G=NR9;
J, K, L, and M are each independently selected from the group of CR12 and N, provided that at least one of K and L is CR12 where Rι2 is not hydrogen;
Ri is -ZRm, where Z is a moiety providing 1-6 atom separation between Rm and the ring to which Ri is attached, and Rm is selected from the group consisting of a substitated or unsubstituted (C3- )cycloalkyl and aryl;
R2 is selected from the group consisting of a substitated or unsubstitated 3, 4, 5, 6 or 7 membered ring;
R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstitated; and each Rι2 is hydrogen or is independently selected from the group consisting of halo, perhalo(Cπo)alkyl, CF3, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, alkoxy, a carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstitated.
333. A compound according to claim 332, wherein Z provides 1-3 atom separation between Rm and the ring.
334. A compound according to claim 332, wherein Z provides 1 atom separation between Rm and the ring.
335. A compound according to claim 334, wherein the 1 atom separation is provided by an atom selected from the group consisting of C, N, O, and S.
336. A compound according to claim 334, wherein the 1 atom separation is provided by a carbon atom.
337. A compound according to claim 334, wherein the 1 atom separation is provided by an oxygen atom.
338. A compound according to claim 334, wherein the 1 atom separation is provided by a nitrogen atom.
339. A compound according to claim 332, wherein Z is selected from the group consisting of -CH2-, -CH2CH2-, -CH2CH2CH2-, -C(O)-, -CH2C(O)-, -C(O)CH2-, -CH2-C(O)CH2-, -C(O)CH2CH2-, -CH2CH2C(O)-, -O-, -OCH2-, -CH2O-, -CH2OCH2-, -OCH2CH2-, -CH2CH2O-, -N(CH3)-, -NHCH2-, -CH2NH-, -CH2NHCH2-, -NHCH2CH2-, -CH2CH2NH-, -NH-C(O)-, -NCH3-C(O)-, -C(O)NH-, -C(O)NCH3-, -NHC(O)CH2-, -C(O)NHCH2-, -C(O)CH2NH-, -CH2NHC(O)-, -CH2C(O)NH-, -NHCH2C(O)-, -S-, -SCH2-, -CH2S-, -SCH2CH2-, -CH2SCH2-, -CH2CH2S-, -C(O)S-, -C(O)SCH2-, -CH2C(O)S-, -C(O)CH2S-, and -CH2SC(O)-, each substituted or unsubstitated.
340. A compound according to claim 332, wherein Z is selected from the group consisting of -CH2-, -C(O)-, -C(S)-, -C(NH)-, -C(NR9)-, -O-, -N(H)-, -N(R9)-, and -S-.
341. A compound according to claim 332, wherein Rm is a substituted or unsubstituted -(C3.7)cycloalkyl.
342. A compound according to claim 332, wherein Rm is a substituted or unsubstituted aryl.
343. A compound according to claim 332, wherein Rm is a substituted or unsubstituted phenyl.
344. A compound according to claim 332, wherein Rm is selected from the group consisting of (2-cyano)phenyl, (3-cyano)phenyl, (2-hydroxy)phenyl, (3-hydroxy)phenyl, (2-alkenyl)phenyl, (3-alkenyl)phenyl, (2-alkynyl)phenyl, (3-alkynyl)phenyl, (2-nitro)phenyl, (3-nitro)phenyl, (2-carboxy)phenyl, (3-carboxy)phenyl, (2-carboxamido)phenyl, (3-carboxamido)ρhenyl, (2-sulfonamido)phenyl, (3-sulfonamido)phenyl, (2-tetrazolyl)phenyl, (3-tetrazolyl)phenyl, (2-aminomethyl)phenyl, (3-aminomethyl)phenyl, (2-amino)phenyl, (3-amino)phenyl, (2-hydroxymethyl)phenyl, (3-hydroxymethyl)phenyl, (2-phenyl)phenyl, (3-phenyl phenyl, (2-CONH2)phenyl, (3-CONH2)phenyl, (2-CONH(Cι-7)alkyl)phenyl, (3-CONH(Cι-7)alkyl)phenyl, (2-C02(C1-7)alkyl)phenyl, (3-CO2(Cι-7)alkyl)phenyl, -NH2, -OH, -(C3-7)alkyl, -alkene, -alkyne, -CCH, -(C3- )cycloalkyl, and -aryl, each substitated or unsubstituted.
345. A compound according to claim 332, wherein Ri is -ORn, where Rn is selected from the group consisting of substituted or unsubstituted alkyl, cycloalkyl, aryl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl.
346. A compound according to claim 332, wherein Z is a carbonyl.
347. A compound according to claim 332, wherein Ri is selected from the group consisting of -(CH2)-(2-cyano)phenyl, -(CH2)-(3-cyano)phenyl, -(CH2)-(2-hydroxy)phenyl, -(CH2)-(3-hychoxy)phenyl, -(CH2)-(2-alkenyl)ρhenyl, -(CH2)-(3-alkenyl)phenyl, -(CH2)-(2-alkynyl)phenyl, -(CH2)-(3-alkynyl)phenyl, -(CH2)-(2-nitro)phenyl, -(CH2)-(3- nitro)phenyl, -(CH2)-(2-carboxy)phenyl, -(CH2)-(3-carboxy)phenyl, -(CH2)-(2-carboxamido)ρhenyl, -(CH2)-(3-carboxamido)phenyl, -(CH2)-(2-sulfonamido)phenyl, -(CH2)-(3-sulfonamido)phenyl, -(CH2)-(2- tetrazolyl)phenyl, -(CH2)-(3-tetrazolyl)phenyl, -(CH2)-(2-aminomethyl)phenyl, -(CH2)-(3- aminomethyl)phenyl, -(CH2)-(2-amino)phenyl, -(CH2)-(3-amino)phenyl, -(CH2)-(2-hyα^oxymethyl)ρhenyl, -(CH2)-(3-hydroxymethyl)ρhenyl, -(CH2)-(2- ρhenyl)phenyl, -(CH2)-(3-phenyl)phenyl, (CH2)-(2-CONH2)ρhenyl, -(CH2)-(3-CONH2)phenyl, -(CH2)-(2-CONH(Cι-7)alkyl)phenyl, -(CH2)-(3-CONH(Cι-7)alkyl)phenyl, -(CH2)-(2-CO2(Cι-7)alkyl)phenyl, -(CH2)-(3-CO2(Cι-7)alkyl)phenyl, -CH2-NH2, -CH2-OH, -CH2-(C3-7)alkyl, -CH2-alkene, -CH2-alkyne, -CH2-CCH, -CH2-(C3-7)cycloalkyl, and -CH2-aryl, each substitated or unsubstitated.
348. A compound according to claim 332, wherein Ri is selected from the group consisting of -(Ci)alkyl-aryl, -(Cι)alkyl-bicycloaryl, -aminoaryl, -aminoheteroaryl, - aminobicycloaryl, -aminoheterobicycloaryl, -O-aryl, -O-heteroaryl, -O-bicycloaryl, -O- heterobicycloaryl, -(S)-aryl, -(S)-heteroaryl, -(S)-bicycloaryl, -S-heterobicycloaryl, -C(O)- aryl, -C(O)-heteroaryl, - C(O)-bicycloaryl, - C(O)-heterobicycloaryl, -C(S)-aryl, -C(S)- heteroaryl, -C(S)-bicycloaryl, -C(S)-heterobicycloaryl, -S(O)-aryl, -S(O)-heteroaryl, -S(O)- bicycloaryl, -SO2-heterobicycloaryl, -SO2-aryl, -SO2-heteroaryl, -SO2-bicycloaryl, -SO2- heterobicycloaryl, -C(NR9)-aryl, -C(NR9)-heteroaryl, -C(NR9)-bicycloaryl, -C(NR9)- heterobicycloaryl, each substituted or unsubstituted.
349. A compound comprising Formula XXXIX:
Figure imgf000238_0001
XXXIX wherein
Q is selected from the group consisting of CO, CS, SO, SO2, or C=NR9;
J, K, L, and M are each independently selected from the group of CRι2 and N, provided that at least one of K and L is CRι2 where Rι2 is not hydrogen;
Ri is selected from the group consisting of a substitated or unsubstituted 3, 4, 5, 6 or 7 membered ring;
R2 is -UV, where U is a moiety providing 1-6 atom separation between V and the ring to which R2 is attached and V comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein; R9 is hydrogen or is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substitated or unsubstitated; and each Rι2 is hydrogen or is independently selected from the group consisting of halo, perhalo(Cι-ιo)alkyl, CF3, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, alkoxy, a carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
350. A compound according to claim 349, wherein U provides 1-4 atom separation between V and the ring.
351. A compound according to claim 349, wherein U provides 1-3 atom separation between V and the ring.
352. A compound according to claim 349, wherein U is selected from the group consisting of -CH2-, -CH2CH2-, -CH2CH2CH2-, -C(O)-, -CH2C(O)-, -C(O)CH2-, -CH2-C(O)CH2-, -C(O)CH2CH2-, -CH2CH2C(O)-, -O-, -OCH2-, -CH2O-, -CH2OCH2-, -OCH2CH2-, -CH2CH2O-, -N(CH3)-, -NHCH2-, -CH2NH-, -CH2NHCH2-, -NHCH2CH2-, -CH2CH2NH-, -NH-C(O)-, -NCH3-C(O)-, -C(O)NH-, -C(O)NCH3-, -NHC(O)CH2-, -C(O)NHCH2-, -C(O)CH2NH-, -CH2NHC(O)-, -CH2C(O)NH-, -NHCH2C(O)-, -S-, -SCH2-, -CH2S-, -SCH2CH2-, -CH2SCH2-, -CH2CH2S-, -C(O)S-, -C(O)SCH2-, -CH2C(O)S-, -C(O)CH2S-, and -CH2SC(O)-, each substituted or unsubstitated.
353. A compound according to claim 349, wherein U is selected from the group consisting of -CH2-, -CHR9-, -C(R9)(R9)-, -O-, -N(H)-, -N(R9)-, and -S-.
354. A compound according to claim 349, wherein V is selected from the group consisting of a primary, secondary or tertiary amine, a heterocycloalkyl comprising a nitrogen ring atom, and a heteroaryl comprising a nitrogen ring atom
355. A compound according to claim 349, wherein the basic nitrogen of V is separated from the ring atom to which R2 is attached by between 1-5 atoms.
356. A compound according to claim 349, wherein the basic nitrogen of V forms part of a primary, secondary or tertiary amine.
357. A compound according to claim 349, wherein the basic nitrogen of V is a nitrogen ring atom of a heterocycloalkyl comprising a nitrogen ring atom or a heteroaryl comprising a nitrogen ring atom.
358. A compound according to claim 349, wherein Ri is a substitated or unsubstituted aryl.
359. A compound according to claim 349, wherein Ri is a substitated or unsubstitated phenyl.
360. A compound according to claim 349, wherein Ri is a substituted or unsubstituted heteroaryl.
PCT/US2004/009217 2003-03-25 2004-03-24 Dipeptidyl peptidase inhibitors WO2004087053A2 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
EP04758366A EP1608317B1 (en) 2003-03-25 2004-03-24 Dipeptidyl peptidase inhibitors
CA002518465A CA2518465A1 (en) 2003-03-25 2004-03-24 Dipeptidyl peptidase inhibitors
JP2006509315A JP4887139B2 (en) 2003-03-25 2004-03-24 Dipeptidyl peptidase inhibitor

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US45778503P 2003-03-25 2003-03-25
US60/457,785 2003-03-25

Publications (3)

Publication Number Publication Date
WO2004087053A2 WO2004087053A2 (en) 2004-10-14
WO2004087053A9 true WO2004087053A9 (en) 2004-11-11
WO2004087053A3 WO2004087053A3 (en) 2006-08-31

Family

ID=33131718

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2004/009217 WO2004087053A2 (en) 2003-03-25 2004-03-24 Dipeptidyl peptidase inhibitors

Country Status (7)

Country Link
US (4) US20040242566A1 (en)
EP (1) EP1608317B1 (en)
JP (1) JP4887139B2 (en)
KR (1) KR20050122220A (en)
CN (1) CN1894234A (en)
CA (1) CA2518465A1 (en)
WO (1) WO2004087053A2 (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8883714B2 (en) 2008-04-07 2014-11-11 Arena Pharmaceuticals, Inc. Pharmaceutical compositions comprising GPR119 agonists which act as peptide YY (PYY) secretagogues
US8906901B2 (en) 2005-09-14 2014-12-09 Takeda Pharmaceutical Company Limited Administration of dipeptidyl peptidase inhibitors
US9266892B2 (en) 2012-12-19 2016-02-23 Incyte Holdings Corporation Fused pyrazoles as FGFR inhibitors
US9388185B2 (en) 2012-08-10 2016-07-12 Incyte Holdings Corporation Substituted pyrrolo[2,3-b]pyrazines as FGFR inhibitors
US9533954B2 (en) 2010-12-22 2017-01-03 Incyte Corporation Substituted imidazopyridazines and benzimidazoles as inhibitors of FGFR3
US9533984B2 (en) 2013-04-19 2017-01-03 Incyte Holdings Corporation Bicyclic heterocycles as FGFR inhibitors
US9580423B2 (en) 2015-02-20 2017-02-28 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US11566028B2 (en) 2019-10-16 2023-01-31 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US11591329B2 (en) 2019-07-09 2023-02-28 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors

Families Citing this family (122)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2518465A1 (en) 2003-03-25 2004-10-14 Takeda San Diego, Inc. Dipeptidyl peptidase inhibitors
EP1911754B1 (en) * 2003-08-13 2013-10-09 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
KR20060041309A (en) 2003-08-13 2006-05-11 다케다 야쿠힌 고교 가부시키가이샤 4-pyrimidone derivatives and their use as peptidyl peptidase inhibitors
US7169926B1 (en) 2003-08-13 2007-01-30 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
US7678909B1 (en) 2003-08-13 2010-03-16 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
WO2005026148A1 (en) * 2003-09-08 2005-03-24 Takeda San Diego, Inc. Dipeptidyl peptidase inhibitors
US7732446B1 (en) 2004-03-11 2010-06-08 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
CN102134231B (en) 2004-03-15 2020-08-04 武田药品工业株式会社 Dipeptidyl peptidase inhibitors
JP2008501714A (en) 2004-06-04 2008-01-24 武田薬品工業株式会社 Dipeptidyl peptidase inhibitor
WO2006019965A2 (en) 2004-07-16 2006-02-23 Takeda San Diego, Inc. Dipeptidyl peptidase inhibitors
AR051596A1 (en) * 2004-10-26 2007-01-24 Irm Llc CONDENSED HETEROCICLIC COMPOUNDS NITROGENATED AS INHIBITORS OF THE ACTIVITY OF THE CANABINOID RECEIVER 1; PHARMACEUTICAL COMPOSITIONS THAT CONTAIN THEM AND THEIR EMPLOYMENT IN THE PREPARATION OF MEDICINES FOR THE TREATMENT OF FOOD DISORDERS
JP2008520744A (en) 2004-11-19 2008-06-19 ザ・レジェンツ・オブ・ザ・ユニバーシティ・オブ・カリフォルニア Anti-inflammatory pyrazolopyrimidine
AU2011201488B2 (en) * 2004-11-29 2011-11-17 Merck Sharp & Dohme Corp. Fused aminopiperidines as dipeptidyl peptidase-IV inhibitors for the treatment or prevention of diabetes
US7872124B2 (en) 2004-12-21 2011-01-18 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
DOP2006000008A (en) 2005-01-10 2006-08-31 Arena Pharm Inc COMBINED THERAPY FOR THE TREATMENT OF DIABETES AND RELATED AFFECTIONS AND FOR THE TREATMENT OF AFFECTIONS THAT IMPROVE THROUGH AN INCREASE IN THE BLOOD CONCENTRATION OF GLP-1
EP2390264A1 (en) 2005-02-11 2011-11-30 Amylin Pharmaceuticals Inc. GIP analog and hybrid polypeptides with selectable propperties
WO2007022946A1 (en) 2005-08-21 2007-03-01 Abbott Gmbh & Co. Kg Heterocyclic compounds and their use as binding partners for 5-ht5 receptors
ATE549926T1 (en) 2005-08-26 2012-04-15 Merck Sharp & Dohme CONDENSED AMINOPIPERIDINES AS DIPEPTIDYLPEPTIDASE IV INHIBITORS FOR THE TREATMENT OR PREVENTION OF DIABETES
BRPI0616077B8 (en) 2005-09-14 2021-05-25 Takeda Pharmaceuticals Co pharmaceutical composition formulated in a single dose, kit, article of manufacture, use of the pharmaceutical composition and use of one or more antidiabetic compounds
CA2622642C (en) 2005-09-16 2013-12-31 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
AU2006327069A1 (en) * 2005-12-23 2007-06-28 Novartis Ag Condensed heterocyclic compounds useful as DPP-IV inhibitors
WO2007112347A1 (en) 2006-03-28 2007-10-04 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
EP1999108A1 (en) * 2006-03-28 2008-12-10 Takeda Pharmaceutical Company Limited Preparation of (r)-3-aminopiperidine dihydrochloride
GB2453058A (en) 2006-04-04 2009-03-25 Univ California Kinase antagonists
PE20071221A1 (en) 2006-04-11 2007-12-14 Arena Pharm Inc GPR119 RECEPTOR AGONISTS IN METHODS TO INCREASE BONE MASS AND TO TREAT OSTEOPOROSIS AND OTHER CONDITIONS CHARACTERIZED BY LOW BONE MASS, AND COMBINED THERAPY RELATED TO THESE AGONISTS
KR20090004950A (en) 2006-04-12 2009-01-12 프로비오드룩 아게 Enzyme inhibitors
JP5421108B2 (en) * 2006-08-23 2014-02-19 ニューロジェン・コーポレーション 2-phenoxypyrimidinone analogs
WO2008024433A2 (en) * 2006-08-23 2008-02-28 Neurogen Corporation Haloalkyl-substituted pyrimidinone derivatives
US8324383B2 (en) 2006-09-13 2012-12-04 Takeda Pharmaceutical Company Limited Methods of making polymorphs of benzoate salt of 2-[[6-[(3R)-3-amino-1-piperidinyl]-3,4-dihydro-3-methyl-2,4-dioxo-1(2H)-pyrimidinyl]methyl]-benzonitrile
KR101129868B1 (en) 2006-10-04 2012-04-12 화이자 프로덕츠 인코포레이티드 Pyrido[4,3-d]pyrimidin-43h-one derivatives as calcium receptor antagonists
WO2008055945A1 (en) 2006-11-09 2008-05-15 Probiodrug Ag 3-hydr0xy-1,5-dihydr0-pyrr0l-2-one derivatives as inhibitors of glutaminyl cyclase for the treatment of ulcer, cancer and other diseases
TW200838536A (en) * 2006-11-29 2008-10-01 Takeda Pharmaceutical Polymorphs of succinate salt of 2-[6-(3-amino-piperidin-1-yl)-3-methyl-2,4-dioxo-3,4-dihydro-2H-pyrimidin-1-ylmethy]-4-fluor-benzonitrile and methods of use therefor
US9126987B2 (en) 2006-11-30 2015-09-08 Probiodrug Ag Inhibitors of glutaminyl cyclase
US8093236B2 (en) 2007-03-13 2012-01-10 Takeda Pharmaceuticals Company Limited Weekly administration of dipeptidyl peptidase inhibitors
KR20120030570A (en) 2007-04-03 2012-03-28 미쓰비시 타나베 파마 코퍼레이션 Combined use of dipeptidyl peptidase iv inhibitor compound and sweetener
JP5667440B2 (en) 2007-04-18 2015-02-12 プロビオドルグ エージー Thiourea derivatives as glutaminyl cyclase inhibitors
SI2152712T1 (en) 2007-05-11 2012-03-30 Pfizer Amino-heterocyclic compounds
WO2009046448A1 (en) 2007-10-04 2009-04-09 Intellikine, Inc. Chemical entities and therapeutic uses thereof
US8193182B2 (en) 2008-01-04 2012-06-05 Intellikine, Inc. Substituted isoquinolin-1(2H)-ones, and methods of use thereof
CN101965336B (en) 2008-01-04 2015-06-17 英特利凯恩有限责任公司 Certain chemical entities, compositions and methods
US8993580B2 (en) 2008-03-14 2015-03-31 Intellikine Llc Benzothiazole kinase inhibitors and methods of use
WO2009114870A2 (en) 2008-03-14 2009-09-17 Intellikine, Inc. Kinase inhibitors and methods of use
US20090257979A1 (en) * 2008-04-15 2009-10-15 Intermune, Inc. Novel Inhibitors of Hepatitis C Virus Replication
WO2010006072A2 (en) 2008-07-08 2010-01-14 The Regents Of The University Of California Mtor modulators and uses thereof
MX2011000216A (en) 2008-07-08 2011-03-29 Intellikine Inc Kinase inhibitors and methods of use.
US20100029371A1 (en) * 2008-08-01 2010-02-04 Gennady Medvinsky Personal Game Services Commerce System (PGSCS)
CA2738429C (en) 2008-09-26 2016-10-25 Intellikine, Inc. Heterocyclic kinase inhibitors
DK2358720T3 (en) 2008-10-16 2016-06-06 Univ California Heteroarylkinaseinhibitorer fused-ring
US8476431B2 (en) 2008-11-03 2013-07-02 Itellikine LLC Benzoxazole kinase inhibitors and methods of use
CN101817833B (en) * 2009-02-26 2013-03-06 中国科学院广州生物医药与健康研究院 DPP-IV inhibitor
EP2427195B1 (en) 2009-05-07 2019-05-01 Intellikine, LLC Heterocyclic compounds and uses thereof
AR077642A1 (en) 2009-07-09 2011-09-14 Arena Pharm Inc METABOLISM MODULATORS AND THE TREATMENT OF DISORDERS RELATED TO THE SAME
EP2475428B1 (en) 2009-09-11 2015-07-01 Probiodrug AG Heterocylcic derivatives as inhibitors of glutaminyl cyclase
US8980899B2 (en) 2009-10-16 2015-03-17 The Regents Of The University Of California Methods of inhibiting Ire1
TW201121968A (en) * 2009-11-09 2011-07-01 Intermune Inc Novel inhibitors of hepatitis C virus replication
ES2567168T3 (en) 2009-11-12 2016-04-20 F. Hoffmann-La Roche Ag N7-substituted purine and pyrazolopyrimidine compounds, compositions and methods of use
EA021568B1 (en) 2009-12-23 2015-07-30 Такеда Фармасьютикал Компани Лимитед Fused heteroaromatic pyrrolidinones as syk inhibitors
US8648073B2 (en) 2009-12-30 2014-02-11 Fochon Pharma, Inc. Certain dipeptidyl peptidase inhibitors
WO2011107530A2 (en) 2010-03-03 2011-09-09 Probiodrug Ag Novel inhibitors
CA2789440C (en) 2010-03-10 2020-03-24 Probiodrug Ag Heterocyclic inhibitors of glutaminyl cyclase (qc, ec 2.3.2.5)
WO2011127051A1 (en) 2010-04-06 2011-10-13 Arena Pharmaceuticals, Inc. Modulators of the gpr119 receptor and the treatment of disorders related thereto
JP5945532B2 (en) 2010-04-21 2016-07-05 プロビオドルグ エージー Benzimidazole derivatives as inhibitors of glutaminyl cyclase
AU2011255218B2 (en) 2010-05-21 2015-03-12 Infinity Pharmaceuticals, Inc. Chemical compounds, compositions and methods for kinase modulation
CN102311448B (en) * 2010-07-07 2014-02-19 中国科学院广州生物医药与健康研究院 Thieno-pyrimidone DPP-IV (dipeptidyl peptidase) inhibitor
CN102311447B (en) * 2010-07-07 2013-11-27 中国科学院广州生物医药与健康研究院 Heterocyclo pyrimidone dipeptidyl peptidase-IV (DPP-IV) inhibitor
EA201390421A1 (en) 2010-09-22 2013-09-30 Арена Фармасьютикалз, Инк. GPR119 RECEPTOR MODULATORS AND TREATMENT OF RELATED DISORDERS
WO2012064973A2 (en) 2010-11-10 2012-05-18 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
AR084824A1 (en) 2011-01-10 2013-06-26 Intellikine Inc PROCESSES TO PREPARE ISOQUINOLINONES AND SOLID FORMS OF ISOQUINOLINONAS
JP6130305B2 (en) 2011-02-23 2017-05-17 インテリカイン, エルエルシー Combinations of kinase inhibitors and their use
US8907086B2 (en) 2011-03-03 2014-12-09 Merck Sharp & Dohme Corp. Fused bicyclic heterocycles useful as dipeptidyl peptidase-IV inhibitors
ES2570167T3 (en) 2011-03-16 2016-05-17 Probiodrug Ag Benzimidazole derivatives as glutaminyl cyclase inhibitors
US20140018371A1 (en) 2011-04-01 2014-01-16 Arena Pharmaceuticals, Inc. Modulators Of The GPR119 Receptor And The Treatment Of Disorders Related Thereto
US20140066369A1 (en) 2011-04-19 2014-03-06 Arena Pharmaceuticals, Inc. Modulators Of The GPR119 Receptor And The Treatment Of Disorders Related Thereto
US20140051714A1 (en) 2011-04-22 2014-02-20 Arena Pharmaceuticals, Inc. Modulators Of The GPR119 Receptor And The Treatment Of Disorders Related Thereto
US20140038889A1 (en) 2011-04-22 2014-02-06 Arena Pharmaceuticals, Inc. Modulators Of The GPR119 Receptor And The Treatment Of Disorders Related Thereto
WO2012170702A1 (en) 2011-06-08 2012-12-13 Arena Pharmaceuticals, Inc. Modulators of the gpr119 receptor and the treatment of disorders related thereto
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
EP2734520B1 (en) 2011-07-19 2016-09-14 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
JP6027611B2 (en) 2011-07-19 2016-11-16 インフィニティー ファーマシューティカルズ, インコーポレイテッド Heterocyclic compounds and uses thereof
BR112014003981A2 (en) 2011-08-23 2017-03-07 Endo Pharmaceuticals Inc pyrimido-pyridazinone compounds and their use
US8785470B2 (en) 2011-08-29 2014-07-22 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
JP6342805B2 (en) 2011-09-02 2018-06-13 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア Substituted pyrazolo [3,4-D] pyrimidine and uses thereof
WO2013055910A1 (en) 2011-10-12 2013-04-18 Arena Pharmaceuticals, Inc. Modulators of the gpr119 receptor and the treatment of disorders related thereto
CN103130819B (en) * 2011-12-01 2016-01-20 中国科学院上海药物研究所 Thiophene [3,2-d] pyrimidin-4-one compounds, its preparation method, pharmaceutical composition and purposes
US8940742B2 (en) 2012-04-10 2015-01-27 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
NZ730134A (en) 2012-06-13 2018-07-27 Incyte Holdings Corp Substituted tricyclic compounds as fgfr inhibitors
US8828998B2 (en) 2012-06-25 2014-09-09 Infinity Pharmaceuticals, Inc. Treatment of lupus, fibrotic conditions, and inflammatory myopathies and other disorders using PI3 kinase inhibitors
AU2013323426A1 (en) 2012-09-26 2015-04-23 The Regents Of The University Of California Modulation of ire1
WO2014074668A1 (en) 2012-11-08 2014-05-15 Arena Pharmaceuticals, Inc. Modulators of gpr119 and the treatment of disorders related thereto
US9481667B2 (en) 2013-03-15 2016-11-01 Infinity Pharmaceuticals, Inc. Salts and solid forms of isoquinolinones and composition comprising and methods of using the same
UY35675A (en) 2013-07-24 2015-02-27 Novartis Ag SUBSTITUTED DERIVATIVES OF QUINAZOLIN-4-ONA
PL3052485T3 (en) 2013-10-04 2022-02-28 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
WO2015051241A1 (en) 2013-10-04 2015-04-09 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US20160187724A1 (en) * 2013-10-08 2016-06-30 Sharp Kabushiki Kaisha Image display device
CA2943075C (en) 2014-03-19 2023-02-28 Infinity Pharmaceuticals, Inc. Heterocyclic compounds for use in the treatment of pi3k-gamma mediated disorders
US20150320755A1 (en) 2014-04-16 2015-11-12 Infinity Pharmaceuticals, Inc. Combination therapies
KR101739003B1 (en) * 2014-07-11 2017-05-23 에스티팜 주식회사 Novel triazolopyrimidinone or triazolopyridone derivatives, and use thereof
GB201415598D0 (en) 2014-09-03 2014-10-15 Univ Birmingham Elavated Itercranial Pressure Treatment
US9708348B2 (en) 2014-10-03 2017-07-18 Infinity Pharmaceuticals, Inc. Trisubstituted bicyclic heterocyclic compounds with kinase activities and uses thereof
US10851105B2 (en) 2014-10-22 2020-12-01 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
MA41551A (en) 2015-02-20 2017-12-26 Incyte Corp BICYCLIC HETEROCYCLES USED AS FGFR4 INHIBITORS
TWI712601B (en) 2015-02-20 2020-12-11 美商英塞特公司 Bicyclic heterocycles as fgfr inhibitors
KR20220070057A (en) 2015-03-09 2022-05-27 인테크린 테라퓨틱스, 아이엔씨. Methods for the treatment of nonalcoholic fatty liver disease and/or lipodystrophy
MX2018003058A (en) 2015-09-14 2018-08-01 Infinity Pharmaceuticals Inc Solid forms of isoquinolinone derivatives, process of making, compositions comprising, and methods of using the same.
KR101730790B1 (en) 2015-12-24 2017-04-28 한국과학기술연구원 5,6,7,8-Tetrahydroquinazolin-2-amine compounds having inhibitory activity of serotonine 5-HT6
WO2017161116A1 (en) 2016-03-17 2017-09-21 Infinity Pharmaceuticals, Inc. Isotopologues of isoquinolinone and quinazolinone compounds and uses thereof as pi3k kinase inhibitors
US10919914B2 (en) 2016-06-08 2021-02-16 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
WO2017223422A1 (en) 2016-06-24 2017-12-28 Infinity Pharmaceuticals, Inc. Combination therapies
TW201825465A (en) 2016-09-23 2018-07-16 美商基利科學股份有限公司 Phosphatidylinositol 3-kinase inhibitors
TW201815787A (en) 2016-09-23 2018-05-01 美商基利科學股份有限公司 Phosphatidylinositol 3-kinase inhibitors
TW201813963A (en) 2016-09-23 2018-04-16 美商基利科學股份有限公司 Phosphatidylinositol 3-kinase inhibitors
MX2019011867A (en) 2017-04-03 2020-01-09 Coherus Biosciences Inc Pparî³ agonist for treatment of progressive supranuclear palsy.
AR111960A1 (en) 2017-05-26 2019-09-04 Incyte Corp CRYSTALLINE FORMS OF A FGFR INHIBITOR AND PROCESSES FOR ITS PREPARATION
ES2812698T3 (en) 2017-09-29 2021-03-18 Probiodrug Ag Glutaminyl cyclase inhibitors
MA52494A (en) 2018-05-04 2021-03-10 Incyte Corp SOLID FORMS OF FGFR INHIBITOR AND THEIR PREPARATION PROCEDURES
CR20200591A (en) 2018-05-04 2021-03-31 Incyte Corp Salts of an fgfr inhibitor
WO2020185532A1 (en) 2019-03-08 2020-09-17 Incyte Corporation Methods of treating cancer with an fgfr inhibitor
BR112022007163A2 (en) 2019-10-14 2022-08-23 Incyte Corp BICYCLIC HETEROCYCLES AS FGFR INHIBITORS
CA3162010A1 (en) 2019-12-04 2021-06-10 Incyte Corporation Derivatives of an fgfr inhibitor
WO2021113479A1 (en) 2019-12-04 2021-06-10 Incyte Corporation Tricyclic heterocycles as fgfr inhibitors
WO2021127071A1 (en) * 2019-12-18 2021-06-24 Merck Sharp & Dohme Corp. Bicyclic heterocycle compounds methods of use thereof for the treatment of herpes viruses
TW202313611A (en) 2021-06-09 2023-04-01 美商英塞特公司 Tricyclic heterocycles as fgfr inhibitors

Family Cites Families (308)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US150343A (en) * 1874-04-28 Ezra milleb
US114141A (en) * 1871-04-25 Improvement in steam-traps
US150344A (en) * 1874-04-28 Improvement in fluid-meters
US150353A (en) * 1874-04-28 Improvement in bearing-bars for furnace-grates
CH317122A (en) * 1952-09-03 1956-11-15 Basf Ag Process for the production of vat dyes
DE1249281B (en) * 1963-05-18
DE1670912C3 (en) 1967-08-18 1981-06-11 Bayer Ag, 5090 Leverkusen Herbicidal agents based on 1,2,4-triazin-5-ones
GB1377642A (en) * 1971-01-14 1974-12-18 Koninklijke Gist Spiritus Penicillanic and cephalosporanic acid derivatives
US3960949A (en) * 1971-04-02 1976-06-01 Schering Aktiengesellschaft 1,2-Biguanides
DE2150686A1 (en) 1971-10-12 1973-04-19 Basf Ag 6-amino-uracil-5-carboxylic acid thioamides - inters for dyestuffs and pharmaceuticals
AU5996573A (en) 1972-09-11 1975-03-06 Commonwealth Scientific And Industrial Research Organisation Pyridinium salts
US3823135A (en) 1972-12-26 1974-07-09 Shell Oil Co Pyrimidone herbicides
GB1464248A (en) 1973-11-01 1977-02-09 Ici Ltd Substituted triazinediones their preparation and use as herbicides
DE2361551A1 (en) 1973-12-11 1975-06-19 Basf Ag Watersol. azo dyes derived from substd. pyridinium chlorides - dyeing natural and synthetic polyamides yellow-violet shades
DE2500024A1 (en) 1975-01-02 1976-07-08 Basf Ag Soluble azo dyes for dying polyamides - prepd. by coupling of 4,6-diaminopyridone derivs
JPS535180A (en) 1976-07-01 1978-01-18 Sumitomo Chem Co Ltd Preparation of 3,4-dihydro-2 (1h) quinazoline derivatives
DE2720085A1 (en) 1977-05-05 1978-11-16 Hoechst Ag PYRIMIDO (6.1-A) ISOCHINOLIN-2-ON DERIVATIVES
US4494978A (en) * 1976-12-30 1985-01-22 Chevron Research Company Herbicidal N-(N'-hydrocarbyloxycarbamylalkyl)-2,6-dialkyl-alpha-haloacetanilides
CH657851A5 (en) * 1983-06-28 1986-09-30 Ciba Geigy Ag CHROMOGENEOUS CHINAZOLONE COMPOUNDS.
AR240698A1 (en) 1985-01-19 1990-09-28 Takeda Chemical Industries Ltd Process for the preparation of 5-(4-(2-(5-ethyl-2-pyridil)-ethoxy)benzyl)-2,4-thiazolodinedione and their salts
US5614492A (en) * 1986-05-05 1997-03-25 The General Hospital Corporation Insulinotropic hormone GLP-1 (7-36) and uses thereof
EP0842925A1 (en) 1987-09-04 1998-05-20 Beecham Group Plc Substituted thiazolidinedione derivatives
US4935493A (en) * 1987-10-06 1990-06-19 E. I. Du Pont De Nemours And Company Protease inhibitors
GB8900382D0 (en) 1989-01-09 1989-03-08 Janssen Pharmaceutica Nv 2-aminopyrimidinone derivatives
US5433955A (en) * 1989-01-23 1995-07-18 Akzo N.V. Site specific in vivo activation of therapeutic drugs
IE63502B1 (en) 1989-04-21 1995-05-03 Zeneca Ltd Aminopyrimidine derivatives useful for treating cardiovascular disorders
US5366862A (en) 1990-02-14 1994-11-22 Receptor Laboratories, Inc. Method for generating and screening useful peptides
US5814460A (en) 1990-02-14 1998-09-29 Diatide, Inc. Method for generating and screening useful peptides
ATE169915T1 (en) 1990-02-15 1998-09-15 Takeda Chemical Industries Ltd PYRIMIDINEDIONE DERIVATIVES, THEIR PREPARATION AND USE
US5462928A (en) 1990-04-14 1995-10-31 New England Medical Center Hospitals, Inc. Inhibitors of dipeptidyl-aminopeptidase type IV
DE4110019C2 (en) 1991-03-27 2000-04-13 Merck Patent Gmbh Imidazopyridines, processes for their production and pharmaceutical preparations containing them
US5387512A (en) * 1991-06-07 1995-02-07 Merck & Co. Inc. Preparation of 3-[z-benzoxazol-2-yl)ethyl]-5-(1-hydroxyethyl)-6-methyl-2-(1H)-pyridinone by biotransformation
EP0610317B1 (en) 1991-10-22 2001-01-31 New England Medical Center Hospitals, Inc. Inhibitors of dipeptidyl-aminopeptidase type iv
US6825169B1 (en) 1991-10-22 2004-11-30 Trustees Of Tufts College Inhibitors of dipeptidyl-aminopeptidase type IV
US5350752A (en) 1991-12-16 1994-09-27 E. R. Squibb & Sons, Inc. Dihydropyrimidine derivatives
DE4141788A1 (en) 1991-12-18 1993-06-24 Merck Patent Gmbh imidazopyridines
EP0626964A1 (en) * 1992-02-19 1994-12-07 Pfizer Inc. Heterocyclic compounds for enhancing antitumor activity
DE4305602A1 (en) 1992-06-17 1993-12-23 Merck Patent Gmbh imidazopyridines
EP0915090A1 (en) 1992-09-10 1999-05-12 Eli Lilly And Company Compounds useful as hypoglycemic agents and for treating Alzheimer's disease
IL106998A0 (en) 1992-09-17 1993-12-28 Univ Florida Brain-enhanced delivery of neuroactive peptides by sequential metabolism
US5811281A (en) 1993-07-12 1998-09-22 Cornell Research Foundation, Inc. Immortalized intestinal epithelial cell lines
IL111785A0 (en) * 1993-12-03 1995-01-24 Ferring Bv Dp-iv inhibitors and pharmaceutical compositions containing them
DE4341453A1 (en) 1993-12-06 1995-06-08 Merck Patent Gmbh imidazopyridines
CN1066739C (en) 1994-03-08 2001-06-06 株式会社大塚制药工场 Phosphonic diester deriv
US5580979A (en) 1994-03-15 1996-12-03 Trustees Of Tufts University Phosphotyrosine peptidomimetics for inhibiting SH2 domain interactions
US5543396A (en) 1994-04-28 1996-08-06 Georgia Tech Research Corp. Proline phosphonate derivatives
US6090786A (en) 1994-06-10 2000-07-18 Fondatech Benelux N.V. Serine proteases, their activity and their synthetic inhibitors
US5601986A (en) * 1994-07-14 1997-02-11 Amgen Inc. Assays and devices for the detection of extrahepatic biliary atresia
DE4432860A1 (en) 1994-09-15 1996-03-21 Merck Patent Gmbh imidazopyridines
US5512549A (en) * 1994-10-18 1996-04-30 Eli Lilly And Company Glucagon-like insulinotropic peptide analogs, compositions, and methods of use
US5864468A (en) * 1995-01-13 1999-01-26 Methode Electronics, Inc. Removable optoelectronic module with grounding means
US5879173A (en) * 1995-01-13 1999-03-09 Methode Electronics, Inc. Removable transceiver module and receptacle
US5717533A (en) * 1995-01-13 1998-02-10 Methode Electronics Inc. Removable optoelectronic module
US5734558A (en) * 1995-01-13 1998-03-31 Poplawski; Daniel S. Removable optoelectronic module
US5614379A (en) 1995-04-26 1997-03-25 Eli Lilly And Company Process for preparing anti-obesity protein
US6265551B1 (en) 1995-06-01 2001-07-24 Dana-Farber Cancer Institute, Inc. Form of dipeptidylpeptidase IV (CD26) found in human serum, antibodies thereto, and uses thereof
US6325989B1 (en) 1995-06-01 2001-12-04 Dana-Farber Cancer Institute, Inc. Form of dipeptidylpeptidase IV (CD26) found in human serum
ATE201016T1 (en) 1995-06-09 2001-05-15 Hoffmann La Roche PYRIMIDINDIONE, PYRIMIDINDRION, TRIAZINDIONE DERIVATIVES AS ALPHA-1 ADRENERGIC RECEPTOR ANTAGONISTS
JPH0928376A (en) 1995-07-21 1997-02-04 Ajinomoto Co Inc New dipeptidyl peptidase iv and its production
TW445263B (en) * 1996-02-29 2001-07-11 Janssen Pharmaceutica Nv Novel esters of 1,4-disubstituted piperidine derivatives
DE122010000020I1 (en) 1996-04-25 2010-07-08 Prosidion Ltd Method for lowering the blood glucose level in mammals
US20020006899A1 (en) * 1998-10-06 2002-01-17 Pospisilik Andrew J. Use of dipeptidyl peptidase IV effectors for lowering blood pressure in mammals
US5965532A (en) 1996-06-28 1999-10-12 Trustees Of Tufts College Multivalent compounds for crosslinking receptors and uses thereof
US5885997A (en) * 1996-07-01 1999-03-23 Dr. Reddy's Research Foundation Heterocyclic compounds, process for their preparation and pharmaceutical compositions containing them and their use in the treatment of diabetes and related diseases
CA2258949C (en) 1996-07-01 2008-05-06 Dr. Reddy's Research Foundation Novel heterocyclic compounds process for their preparation and pharmaceutical compositions containing them and their use in the treatment of diabetes and related diseases
US6458924B2 (en) 1996-08-30 2002-10-01 Novo Nordisk A/S Derivatives of GLP-1 analogs
US6006753A (en) 1996-08-30 1999-12-28 Eli Lilly And Company Use of GLP-1 or analogs to abolish catabolic changes after surgery
US7235627B2 (en) 1996-08-30 2007-06-26 Novo Nordisk A/S Derivatives of GLP-1 analogs
US6011155A (en) 1996-11-07 2000-01-04 Novartis Ag N-(substituted glycyl)-2-cyanopyrrolidines, pharmaceutical compositions containing them and their use in inhibiting dipeptidyl peptidase-IV
GB9702701D0 (en) 1997-02-01 1997-04-02 Univ Newcastle Ventures Ltd Quinazolinone compounds
JP3468007B2 (en) * 1997-02-05 2003-11-17 住友電装株式会社 Lever connector
US20030060434A1 (en) * 1997-02-18 2003-03-27 Loretta Nielsen Combined tumor suppressor gene therapy and chemotherapy in the treatment of neoplasms
US6100234A (en) 1997-05-07 2000-08-08 Tufts University Treatment of HIV
DE69840230D1 (en) 1997-05-16 2009-01-02 Novozymes Inc POLYPEPTIDES WITH PROLYLDIPEPTIDYLAMINOPEPTIDASE ACTIVITY AND NUCLEIC ACIDS THAT CODE FOR IT
EP0897012A1 (en) 1997-07-05 1999-02-17 Societe Des Produits Nestle S.A. Cloning of the prolyl-dipeptidyl-peptidase from aspergillus oryzae
US6235493B1 (en) 1997-08-06 2001-05-22 The Regents Of The University Of California Amino acid substituted-cresyl violet, synthetic fluorogenic substrates for the analysis of agents in individual in vivo cells or tissue
IL125950A0 (en) * 1997-09-05 1999-04-11 Pfizer Prod Inc Methods of administering ampa receptor antagonists to treat dyskinesias associated with dopamine agonist therapy
JP2001518290A (en) 1997-09-29 2001-10-16 ポイント セラピューティクス, インコーポレイテッド Stimulation of hematopoietic cells in vitro
US6485955B1 (en) 1997-10-06 2002-11-26 The Trustees Of Tufts University Quiescent cell dipeptidyl peptidase: a novel cytoplasmic serine protease
US6342611B1 (en) * 1997-10-10 2002-01-29 Cytovia, Inc. Fluorogenic or fluorescent reporter molecules and their applications for whole-cell fluorescence screening assays for capsases and other enzymes and the use thereof
JP4095772B2 (en) * 1997-11-18 2008-06-04 財団法人微生物化学研究会 Novel bioactive substance sulfostine, its production method and its use
FR2771004B1 (en) 1997-11-19 2000-02-18 Inst Curie USE OF BENZHYDRYL SULFINYL DERIVATIVES FOR THE MANUFACTURE OF MEDICINAL PRODUCTS HAVING A WAKING EFFECT IN SITUATIONS OF DRUG-BASED VIGILANCE DISORDERS
US6380357B2 (en) 1997-12-16 2002-04-30 Eli Lilly And Company Glucagon-like peptide-1 crystals
KR100556071B1 (en) * 1997-12-16 2006-03-10 노보자임스 에이/에스 Polypeptides having signal peptide activity and nucleic acids encoding same
EP1052994A2 (en) 1998-02-02 2000-11-22 Trustees Of Tufts College Use of dipeptidylpetidase inhibitors to regulate glucose metabolism
US20020061839A1 (en) * 1998-03-09 2002-05-23 Scharpe Simon Lodewijk Serine peptidase modulators
FR2777283B1 (en) 1998-04-10 2000-11-24 Adir NOVEL GLUCAGON-PEPTIDE- 1 (7-37) ANALOGUE PEPTIDE COMPOUNDS, PROCESS FOR THEIR PREPARATION AND THE PHARMACEUTICAL COMPOSITIONS CONTAINING THE SAME
DE19823831A1 (en) * 1998-05-28 1999-12-02 Probiodrug Ges Fuer Arzneim New pharmaceutical use of isoleucyl thiazolidide and its salts
JP2002517401A (en) * 1998-06-05 2002-06-18 ポイント セラピューティクス, インコーポレイテッド Cyclic boroproline compound
DE19828114A1 (en) 1998-06-24 2000-01-27 Probiodrug Ges Fuer Arzneim Produgs of unstable inhibitors of dipeptidyl peptidase IV
DE19828113A1 (en) * 1998-06-24 2000-01-05 Probiodrug Ges Fuer Arzneim Prodrugs of Dipeptidyl Peptidase IV Inhibitors
US6129911A (en) 1998-07-10 2000-10-10 Rhode Island Hospital, A Lifespan Partner Liver stem cell
DE19834591A1 (en) * 1998-07-31 2000-02-03 Probiodrug Ges Fuer Arzneim Use of substances that decrease the activity of dipeptidyl peptidase IV to increase blood sugar levels, e.g. for treating hypoglycemia
DE19844693A1 (en) * 1998-09-29 2000-03-30 Delphi Automotive Systems Gmbh Two part electrical connector
US20030176357A1 (en) 1998-10-06 2003-09-18 Pospisilik Andrew J. Dipeptidyl peptidase IV inhibitors and their uses for lowering blood pressure levels
CO5150173A1 (en) 1998-12-10 2002-04-29 Novartis Ag COMPOUNDS N- (REPLACED GLYCLE) -2-DIPEPTIDYL-IV PEPTIDASE INHIBITING CYANOPIRROLIDINS (DPP-IV) WHICH ARE EFFECTIVE IN THE TREATMENT OF CONDITIONS MEDIATED BY DPP-IV INHIBITION
GB9906715D0 (en) * 1999-03-23 1999-05-19 Ferring Bv Compositions for promoting growth
US6548529B1 (en) 1999-04-05 2003-04-15 Bristol-Myers Squibb Company Heterocyclic containing biphenyl aP2 inhibitors and method
AU3958100A (en) 1999-04-20 2000-11-02 Novo Nordisk A/S New compounds, their preparation and use
CN1191273C (en) 1999-05-17 2005-03-02 康久化学公司 Long lasting insulinotropic peptides
DE19926233C1 (en) * 1999-06-10 2000-10-19 Probiodrug Ges Fuer Arzneim Production of thiazolidine, useful as pharmaceutical intermediate, comprises reacting hexamethylenetetramine with cysteamine
US6172081B1 (en) * 1999-06-24 2001-01-09 Novartis Ag Tetrahydroisoquinoline 3-carboxamide derivatives
US6107317A (en) 1999-06-24 2000-08-22 Novartis Ag N-(substituted glycyl)-thiazolidines, pharmaceutical compositions containing them and their use in inhibiting dipeptidyl peptidase-IV
US6110949A (en) 1999-06-24 2000-08-29 Novartis Ag N-(substituted glycyl)-4-cyanothiazolidines, pharmaceutical compositions containing them and their use in inhibiting dipeptidyl peptidase-IV
US6528486B1 (en) * 1999-07-12 2003-03-04 Zealand Pharma A/S Peptide agonists of GLP-1 activity
US6617340B1 (en) 1999-07-29 2003-09-09 Novartis Ag N-(substituted glycyl)-pyrrolidines, pharmaceutical compositions containing them and their use in inhibiting dipeptidyl peptidase-IV
DE19940130A1 (en) 1999-08-24 2001-03-01 Probiodrug Ges Fuer Arzneim New effectors of Dipeptidyl Peptidase IV for topical use
WO2001016114A2 (en) * 1999-08-27 2001-03-08 Chemocentryx, Inc. Heterocyclic compounds and methods for modulating cxcr3 function
ES2234662T3 (en) * 1999-09-16 2005-07-01 Curis, Inc. HEDGEHOG SIGNALING ROUTE MEDIATORS, COMPOSITIONS AND USES RELATED TO THEMSELVES.
US6559188B1 (en) * 1999-09-17 2003-05-06 Novartis Ag Method of treating metabolic disorders especially diabetes, or a disease or condition associated with diabetes
US6414002B1 (en) * 1999-09-22 2002-07-02 Bristol-Myers Squibb Company Substituted acid derivatives useful as antidiabetic and antiobesity agents and method
WO2001023364A1 (en) 1999-09-28 2001-04-05 Merck Patent Gmbh Quinazolinones
US6447772B1 (en) 1999-10-01 2002-09-10 Klaire Laboratories, Inc. Compositions and methods relating to reduction of symptoms of autism
US6251391B1 (en) * 1999-10-01 2001-06-26 Klaire Laboratories, Inc. Compositions containing dipepitidyl peptidase IV and tyrosinase or phenylalaninase for reducing opioid-related symptons
JP2001172257A (en) * 1999-10-05 2001-06-26 Fujisawa Pharmaceut Co Ltd Organic sulfonamide compound
WO2001027090A1 (en) 1999-10-08 2001-04-19 Meiji Seika Kaisha, Ltd. m-SUBSTITUTED BENZOIC ACID DERIVATIVES EXHIBITING INTEGRINαvβ3 ANTAGONISM
US6261794B1 (en) 1999-10-14 2001-07-17 Saint Louis University Methods for identifying inhibitors of methionine aminopeptidases
US7230000B1 (en) * 1999-10-27 2007-06-12 Cytokinetics, Incorporated Methods and compositions utilizing quinazolinones
KR20050049562A (en) * 1999-10-27 2005-05-25 싸이토키네틱스, 인코포레이티드 Methods and compositions utilizing quinazolinones
US20040152745A1 (en) 1999-11-12 2004-08-05 Guilford Pharmaceuticals, Inc. Dipeptidyl peptidase IV inhibitors and methods of making and using dipeptidyl peptidase IV inhibitors
CA2390231A1 (en) 1999-11-12 2001-05-17 Paul Jackson Dipeptidyl peptidase iv inhibitors and methods of making and using dipeptidyl peptidase iv inhibitors
GB9928330D0 (en) * 1999-11-30 2000-01-26 Ferring Bv Novel antidiabetic agents
US6517382B2 (en) * 1999-12-01 2003-02-11 Tyco Electronics Corporation Pluggable module and receptacle
AU2020501A (en) * 1999-12-08 2001-06-18 1149336 Ontario Inc. Chemotherapy treatment
US6380398B2 (en) 2000-01-04 2002-04-30 Novo Nordisk A/S Therapeutically active and selective heterocyclic compounds that are inhibitors of the enzyme DPP-IV
ATE385421T1 (en) 2000-01-21 2008-02-15 Novartis Pharma Gmbh COMPOSITIONS CONSISTING OF DIPEPTIDYLPEPTIDASE-IV INHIBITORS AND ANTIDIABETICS
CA2396920A1 (en) * 2000-01-27 2001-08-02 Eli Lilly And Company Process for solubilizing glucagon-like peptide 1 (glp-1) compounds
US6569901B2 (en) 2000-01-28 2003-05-27 Novo Nordisk A/S Alkynyl-substituted propionic acid derivatives, their preparation and use
US7217722B2 (en) 2000-02-01 2007-05-15 Kirin Beer Kabushiki Kaisha Nitrogen-containing compounds having kinase inhibitory activity and drugs containing the same
US6395767B2 (en) * 2000-03-10 2002-05-28 Bristol-Myers Squibb Company Cyclopropyl-fused pyrrolidine-based inhibitors of dipeptidyl peptidase IV and method
US6448045B1 (en) 2000-03-10 2002-09-10 The Regents Of The University Of California Inducing insulin gene expression in pancreas cells expressing recombinant PDX-1
US6608038B2 (en) 2000-03-15 2003-08-19 Novartis Ag Methods and compositions for treatment of diabetes and related conditions via gene therapy
EP1136482A1 (en) 2000-03-23 2001-09-26 Sanofi-Synthelabo 2-Amino-3-(alkyl)-pyrimidone derivatives as GSK3beta inhibitors
US6555519B2 (en) * 2000-03-30 2003-04-29 Bristol-Myers Squibb Company O-glucosylated benzamide SGLT2 inhibitors and method
CZ20023234A3 (en) * 2000-03-31 2003-01-15 Probiodrug Ag Medicament against diabetes mellitus
US6573096B1 (en) 2000-04-01 2003-06-03 The Research Foundation At State University Of New York Compositions and methods for inhibition of cancer invasion and angiogenesis
US6545170B2 (en) * 2000-04-13 2003-04-08 Pharmacia Corporation 2-amino-5, 6 heptenoic acid derivatives useful as nitric oxide synthase inhibitors
ES2567260T3 (en) * 2000-04-25 2016-04-21 Icos Corporation Human phosphatidylinositol 3-kinase delta inhibitors
GB0010183D0 (en) 2000-04-26 2000-06-14 Ferring Bv Inhibitors of dipeptidyl peptidase IV
GB0010188D0 (en) * 2000-04-26 2000-06-14 Ferring Bv Inhibitors of dipeptidyl peptidase IV
US6783757B2 (en) * 2000-06-01 2004-08-31 Kirkman Group, Inc. Composition and method for increasing exorphin catabolism to treat autism
TW583185B (en) 2000-06-13 2004-04-11 Novartis Ag N-(substituted glycyl)-2-cyanopyrrolidines and pharmaceutical composition for inhibiting dipeptidyl peptidase-IV (DPP-IV) or for the prevention or treatment of diseases or conditions associated with elevated levels of DPP-IV comprising the same
US6432969B1 (en) 2000-06-13 2002-08-13 Novartis Ag N-(substituted glycyl)-2 cyanopyrrolidines, pharmaceutical compositions containing them and their use in inhibiting dipeptidyl peptidase-IV
US6627636B2 (en) 2000-06-15 2003-09-30 Bristol-Myers Squibb Company HMG-CoA reductase inhibitors and method
US6620821B2 (en) 2000-06-15 2003-09-16 Bristol-Myers Squibb Company HMG-CoA reductase inhibitors and method
GB0014969D0 (en) 2000-06-19 2000-08-09 Smithkline Beecham Plc Novel method of treatment
US7078397B2 (en) 2000-06-19 2006-07-18 Smithkline Beecham Corporation Combinations of dipeptidyl peptidase IV inhibitors and other antidiabetic agents for the treatment of diabetes mellitus
MXPA02012272A (en) * 2000-07-04 2003-04-25 Novo Nordisk As Heterocyclic compounds, which are inhibitors of the enzyme dpp-iv.
JP2002042960A (en) * 2000-07-25 2002-02-08 Yazaki Corp Connector support mechanism
AU2001281083A1 (en) 2000-08-01 2002-02-13 Pharmacia Corporation Hexahydro-7-1h-azepin-2-yl-haxanoic acid derivatives as inhibitors of inducible nitric oxide synthase
US7022711B2 (en) 2000-08-04 2006-04-04 Warner-Lambert Company 2-(4-Pyridyl)amino-6-dialkoxyphenyl-pyrido[2,3-d]pyrimidin-7-ones
JP4101053B2 (en) 2000-08-10 2008-06-11 田辺三菱製薬株式会社 Proline derivative and its pharmaceutical use
US20020165237A1 (en) 2000-08-11 2002-11-07 Fryburg David Albert Treatment of the insulin resistance syndrome
US20020037829A1 (en) * 2000-08-23 2002-03-28 Aronson Peter S. Use of DPPIV inhibitors as diuretic and anti-hypertensive agents
US6900226B2 (en) * 2000-09-06 2005-05-31 Hoffman-La Roche Inc. Neuropeptide Y antagonists
US20020064736A1 (en) * 2000-09-27 2002-05-30 Fuji Photo Film Co., Ltd. Dye-forming coupler, silver halide photographic light-sensitive material, and method for producing an azomethine dye
EP1324995A2 (en) * 2000-09-27 2003-07-09 Merck & Co., Inc. Benzopyrancarboxylic acid derivatives for the treatment of diabetes and lipid disorders
IL154893A0 (en) * 2000-10-06 2003-10-31 Tanabe Seiyaku Co Aliphatic nitrogen-containing cyclic compounds, methods for the preparation thereof and pharmaceutical compositions containing the same
RU2305133C2 (en) 2000-10-12 2007-08-27 Ферринг Бв Dppiv-cognated new serine protease gene
AUPR107800A0 (en) * 2000-10-27 2000-11-23 University Of Sydney, The Peptide and nucleic acid molecule ii
JP2004512300A (en) 2000-10-27 2004-04-22 プロバイオドラッグ アーゲー Methods of treating neurological and neuropsychological disorders
EP1333887B1 (en) * 2000-10-30 2006-08-02 Ortho-McNeil Pharmaceutical, Inc. Combination therapy comprising anti-diabetic and anticonvulsant agents
CA2427610A1 (en) 2000-10-31 2002-08-08 Merck & Co., Inc. Benzopyrancarboxylic acid derivatives for the treatment of diabetes and lipid disorders
WO2002059343A2 (en) 2000-10-31 2002-08-01 Vanderbilt University Biological markers and diagnostic tests for angiotensin converting enzyme inhibitor- and vasopeptidase inhibitor-associated angioedema
US6864363B2 (en) 2000-11-08 2005-03-08 University Of Georgia Research Foundation, Inc. Dipeptidylpeptidases and methods of use
US20020155565A1 (en) 2000-11-10 2002-10-24 Pilar Garin-Chesa FAP-activated anti-tumor compounds
TWI243162B (en) * 2000-11-10 2005-11-11 Taisho Pharmaceutical Co Ltd Cyanopyrrolidine derivatives
US20030055052A1 (en) * 2000-11-10 2003-03-20 Stefan Peters FAP-activated anti-tumor compounds
US20030203946A1 (en) 2000-11-17 2003-10-30 Carsten Behrens Glucagon antagonists/inverse agonists
WO2002040448A1 (en) 2000-11-20 2002-05-23 Bristol-Myers Squibb Company Pyridone derivatives as ap2 inhibitors
NZ526622A (en) 2000-12-11 2006-07-28 Amgen Sf Llc CXCR3 antagonists
JPWO2002051836A1 (en) 2000-12-27 2004-04-22 協和醗酵工業株式会社 Dipeptidyl peptidase-IV inhibitor
DE10100053A1 (en) 2001-01-02 2002-08-22 Keyneurotek Ag I G Use of enzyme inhibitors of dipeptidyl peptidase IV and aminopeptidase N and pharmaceutical preparations therefrom for the prevention and / or therapy of ischemia-related acute and chronic neurodegenerative processes and diseases
BR0206831A (en) * 2001-02-02 2004-07-06 Takeda Chemical Industries Ltd Compound, crystal, pharmaceutical agent, agents for the prophylaxis or treatment of diabetes, diabetic complications, impaired glucose tolerance and obesity, peptidase inhibitor, use of a compound, and method of producing a compound
WO2002062798A2 (en) * 2001-02-05 2002-08-15 Dr. Reddy's Laboratories Ltd. Salts of pyrimidine derivatives for use against coronary heart disease and atherosclerose
TWI255817B (en) 2001-02-14 2006-06-01 Kissei Pharmaceutical Glucopyranosyloxybenzylbenzene derivatives and medicinal use thereof
CZ301487B6 (en) * 2001-02-24 2010-03-17 Boehringer Ingelheim Pharma Gmbh & Co. Kg Xanthine derivatives, process of their preparation and use as a medicament
US6337069B1 (en) 2001-02-28 2002-01-08 B.M.R.A. Corporation B.V. Method of treating rhinitis or sinusitis by intranasally administering a peptidase
US6371787B1 (en) * 2001-03-07 2002-04-16 International Business Machines Corporation Pull-to-release type latch mechanism for removable small form factor electronic modules
PT1372650E (en) * 2001-03-19 2009-02-25 Novartis Ag Combinations comprising an antidiarrheal agent and an epothilone or an epothilone derivative
US7026316B2 (en) 2001-03-27 2006-04-11 Merck & Co., Inc. Dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
FR2822826B1 (en) 2001-03-28 2003-05-09 Servier Lab NOVEL ALPHA-AMINO ACID SULPHONYL DERIVATIVES, PROCESS FOR PREPARING THEM AND PHARMACEUTICAL COMPOSITIONS CONTAINING SAME
DE10115921A1 (en) 2001-03-30 2002-10-02 Boehringer Ingelheim Pharma Process for the preparation of 4,6-diaminopyrimido [5,4-d] pyrimidines
US6890905B2 (en) 2001-04-02 2005-05-10 Prosidion Limited Methods for improving islet signaling in diabetes mellitus and for its prevention
GB0109146D0 (en) 2001-04-11 2001-05-30 Ferring Bv Treatment of type 2 diabetes
PE20021080A1 (en) * 2001-04-12 2003-02-12 Boehringer Ingelheim Int A SPECIFIC ANTIBODY FAPO BIBH1 IN THE TREATMENT OF CANCER
US6573287B2 (en) 2001-04-12 2003-06-03 Bristo-Myers Squibb Company 2,1-oxazoline and 1,2-pyrazoline-based inhibitors of dipeptidyl peptidase IV and method
FR2824825B1 (en) * 2001-05-15 2005-05-06 Servier Lab NOVEL ALPHA-AMINOACID DERIVATIVES, PROCESS FOR THEIR PREPARATION AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
US20030060494A1 (en) 2001-05-18 2003-03-27 Nobuyuki Yasuda Pharmaceutical use of N-carbamoylazole derivatives
US7105556B2 (en) * 2001-05-30 2006-09-12 Bristol-Myers Squibb Company Conformationally constrained analogs useful as antidiabetic and antiobesity agents and method
US6794379B2 (en) * 2001-06-06 2004-09-21 Tularik Inc. CXCR3 antagonists
KR100490048B1 (en) * 2001-06-19 2005-05-17 삼성전자주식회사 In-line system for manufacturing liquid crystal display and method of manufacturing liquid crystal display using the same
JP2004535433A (en) 2001-06-20 2004-11-25 メルク エンド カムパニー インコーポレーテッド Dipeptidyl peptidase inhibitors for the treatment of diabetes
ATE318139T1 (en) 2001-06-20 2006-03-15 Merck & Co Inc DIPEPTIDYLPEPTIDASE INHIBITORS FOR THE TREATMENT OF DIABETES
ES2296962T3 (en) 2001-06-27 2008-05-01 Smithkline Beecham Corporation PIRROLIDINS AS INHIBITORS OF DIPEPTIDIL PEPTIDASA.
US7183290B2 (en) 2001-06-27 2007-02-27 Smithkline Beecham Corporation Fluoropyrrolidines as dipeptidyl peptidase inhibitors
DE10150203A1 (en) 2001-10-12 2003-04-17 Probiodrug Ag Use of dipeptidyl peptidase IV inhibitor in treatment of cancer
US20030135023A1 (en) 2001-06-27 2003-07-17 Hans-Ulrich Demuth Peptide structures useful for competitive modulation of dipeptidyl peptidase IV catalysis
WO2003002593A2 (en) 2001-06-27 2003-01-09 Probiodrug Ag Peptide structures useful for competitive modulation of dipeptidyl peptidase iv catalysis
US7368421B2 (en) * 2001-06-27 2008-05-06 Probiodrug Ag Use of dipeptidyl peptidase IV inhibitors in the treatment of multiple sclerosis
EP1406873B1 (en) 2001-06-27 2007-09-26 Smithkline Beecham Corporation Fluoropyrrolidines as dipeptidyl peptidase inhibitors
US20030130199A1 (en) * 2001-06-27 2003-07-10 Von Hoersten Stephan Dipeptidyl peptidase IV inhibitors and their uses as anti-cancer agents
DE10154689A1 (en) 2001-11-09 2003-05-22 Probiodrug Ag Substituted amino ketone compounds
CA2418543A1 (en) 2001-06-27 2003-01-09 Probiodrug Ag New dipeptidyl peptidase iv inhibitors and their uses as anti-cancer agents
US6869947B2 (en) * 2001-07-03 2005-03-22 Novo Nordisk A/S Heterocyclic compounds that are inhibitors of the enzyme DPP-IV
UA74912C2 (en) 2001-07-06 2006-02-15 Merck & Co Inc Beta-aminotetrahydroimidazo-(1,2-a)-pyrazines and tetratriazolo-(4,3-a)-pyrazines as inhibitors of dipeptylpeptidase for the treatment or prevention of diabetes
US20030144350A1 (en) 2001-07-20 2003-07-31 Adipogenix, Inc. Fat accumulation-modulation compounds
EP1285922A1 (en) 2001-08-13 2003-02-26 Warner-Lambert Company 1-Alkyl or 1-cycloalkyltriazolo[4,3-a]quinazolin-5-ones as phosphodiesterase inhibitors
WO2003016335A2 (en) 2001-08-13 2003-02-27 Probiodrug Ag Irreversible cysteine protease inhibitors of legumain
US6844316B2 (en) * 2001-09-06 2005-01-18 Probiodrug Ag Inhibitors of dipeptidyl peptidase I
DE10143840A1 (en) 2001-09-06 2003-03-27 Probiodrug Ag New acylated hydroxamates useful for the treatment of e.g. wound healing
US6673829B2 (en) * 2001-09-14 2004-01-06 Novo Nordisk A/S Aminoazetidine,-pyrrolidine and -piperidine derivatives
CA2460512A1 (en) 2001-09-14 2003-03-27 Mitsubishi Pharma Corporation Thiazolidine derivatives and medicinal use thereof
US20030186963A1 (en) 2001-09-14 2003-10-02 Dorwald Florencio Zaragoza Substituted piperidines
AU2002331311A1 (en) 2001-09-19 2003-04-01 Novo Nordisk A/S Heterocyclic compounds that are inhibitors of the enzyme dpp-iv
EA007224B1 (en) 2001-09-21 2006-08-25 Мицубиси Фарма Корпорейшн 3-substituted-4-pyrimidone derivatives
NZ531636A (en) 2001-09-21 2005-11-25 Mitsubishi Pharma Corp 3-substituted-4-pyrimidone derivatives as tau protein kinase 1 inhibitor
SI1427415T1 (en) 2001-09-21 2009-12-31 Bristol Myers Squibb Co Lactam-containing compounds and derivatives thereof as factor xa inhibitors
US7019010B2 (en) * 2001-09-27 2006-03-28 Novertis Ag Combinations
MXPA04002993A (en) * 2001-10-01 2004-07-15 Bristol Myers Squibb Co Spiro-hydantoin compounds useful as anti-inflammatory agents.
WO2003030946A1 (en) 2001-10-09 2003-04-17 Novartis Ag Regulation of insulin production
US7064135B2 (en) 2001-10-12 2006-06-20 Novo Nordisk Inc. Substituted piperidines
TWI301834B (en) 2001-10-22 2008-10-11 Eisai R&D Man Co Ltd Pyrimidone compound and pharmaceutical composition including the same
GB0125445D0 (en) * 2001-10-23 2001-12-12 Ferring Bv Protease Inhibitors
US6861440B2 (en) 2001-10-26 2005-03-01 Hoffmann-La Roche Inc. DPP IV inhibitors
EP1442028A4 (en) * 2001-11-06 2009-11-04 Bristol Myers Squibb Co Substituted acid derivatives useful as antidiabetic and antiobesity agents and method
WO2003039460A2 (en) * 2001-11-07 2003-05-15 Merck & Co., Inc. Mitotic kinesin inhibitors
US20030125304A1 (en) * 2001-11-09 2003-07-03 Hans-Ulrich Demuth Substituted amino ketone compounds
US20030089935A1 (en) * 2001-11-13 2003-05-15 Macronix International Co., Ltd. Non-volatile semiconductor memory device with multi-layer gate insulating structure
IL161717A0 (en) 2001-11-26 2004-09-27 Schering Corp Piperidine-based mch antagonists for treatment of obesity and cns disorders
EP2769715A3 (en) 2001-11-26 2014-09-17 Trustees Of Tufts College Methods for treating autoimmune disorders, and reagents related thereto
EP1469873A4 (en) 2001-11-26 2007-10-03 Tufts College Peptidomimetic inhibitors of post-proline cleaving enzymes
AU2002350217A1 (en) 2001-12-04 2003-06-17 Bristol-Myers Squibb Company Glycinamides as factor xa inhibitors
DE60222302T2 (en) * 2001-12-06 2008-05-29 Merck & Co., Inc. INHIBITORS OF MITOTIC KINESINE
CZ2004714A3 (en) 2001-12-14 2004-10-13 Novoánordiskáa@S Compounds and uses thereof for decreasing activity of hormone-sensitive lipase
JP2005518391A (en) 2001-12-21 2005-06-23 ノボ ノルディスク アクティーゼルスカブ Amide derivatives as GK activators
TW200301698A (en) 2001-12-21 2003-07-16 Bristol Myers Squibb Co Acridone inhibitors of IMPDH enzyme
US6727261B2 (en) * 2001-12-27 2004-04-27 Hoffman-La Roche Inc. Pyrido[2,1-A]Isoquinoline derivatives
DE60217918T2 (en) 2001-12-29 2007-11-15 Novo Nordisk A/S COMBINED USE OF A GLP-1 COMPOUND AND AN ALDOSE REDUCTASE INHIBITOR
ATE409466T1 (en) 2002-01-11 2008-10-15 Novo Nordisk As METHOD AND COMPOSITION FOR TREATING DIABETES, HYPERTENSION, CHRONIC HEART FAILURE AND CONDITIONS ASSOCIATED WITH FLUID RETENTION
WO2003063903A2 (en) 2002-02-01 2003-08-07 Probiodrug Ag Modulation of t lymphocytes using dp iv inhibitors
US7101898B2 (en) 2002-02-01 2006-09-05 Novo Nordisk A/S Amides of aminoalkyl-substituted azetidines, pyrrolidines, piperidines and azepanes
BR0307665A (en) * 2002-02-13 2005-01-04 Hoffmann La Roche Compounds, process for their manufacture, pharmaceutical compositions comprising them, method for treating and / or prophylaxis of diseases associated with dpp iv and use of the compounds
MXPA04007743A (en) 2002-02-13 2004-10-15 Hoffmann La Roche Novel pyridine- and quinoline-derivatives.
EA200400980A1 (en) 2002-02-27 2005-02-24 Пфайзер Продактс Инк. ACC INHIBITORS
ATE349435T1 (en) 2002-02-28 2007-01-15 Prosidion Ltd GLUTAMINE-BASED DPIV INHIBITORS
US20080070864A1 (en) 2002-03-07 2008-03-20 X-Ceptor Therapeutics, Inc. Quinazolinone Modulators Of Nuclear Receptors
US20030232761A1 (en) 2002-03-28 2003-12-18 Hinke Simon A. Novel analogues of glucose-dependent insulinotropic polypeptide
EP1492777A1 (en) 2002-04-08 2005-01-05 Torrent Pharmaceuticals Ltd Thiazolidine-4-carbonitriles and analogues and their use as dipeptidyl-peptidase inhibitors
US20040106802A1 (en) * 2002-04-08 2004-06-03 Torrent Pharmaceuticals Ltd. Novel compounds and therapeutic uses thereof
JP2003300977A (en) 2002-04-10 2003-10-21 Sumitomo Pharmaceut Co Ltd Xanthine derivative
EP2204181A3 (en) 2002-04-30 2010-09-22 Trustees Of Tufts College Protease inhibitors
TW200307667A (en) * 2002-05-06 2003-12-16 Bristol Myers Squibb Co Sulfonylaminovalerolactams and derivatives thereof as factor Xa inhibitors
US7057046B2 (en) * 2002-05-20 2006-06-06 Bristol-Myers Squibb Company Lactam glycogen phosphorylase inhibitors and method of use
MXPA04011557A (en) 2002-05-23 2005-07-05 Chiron Corp Substituted quinazolinone compounds.
GB0212412D0 (en) 2002-05-29 2002-07-10 Novartis Ag Combination of organic compounds
US6710040B1 (en) 2002-06-04 2004-03-23 Pfizer Inc. Fluorinated cyclic amides as dipeptidyl peptidase IV inhibitors
AU2003233010A1 (en) 2002-06-04 2003-12-19 Pfizer Products Inc. Process for the preparation of 3,3,4,4-tetrafluoropyrrolidine and derivatives thereof
CN100469778C (en) * 2002-06-06 2009-03-18 卫材R&D管理有限公司 New fused imidazole derivative
WO2003106628A2 (en) * 2002-06-17 2003-12-24 Bristol-Myers Squibb Company Benzodiazepine inhibitors of mitochondial f1f0 atp hydrolase and methods of inhibiting f1f0 atp hydrolase
CA2489648A1 (en) 2002-06-17 2003-12-24 Smithkline Beecham Corporation Chemical process
SE0201976D0 (en) 2002-06-24 2002-06-24 Astrazeneca Ab Novel compounds
US20040054171A1 (en) * 2002-07-04 2004-03-18 Jensen Anette Frost Polymorphic forms of a 4H-thieno[3,2-E]-1,2,4-thiadiazine 1,1-dioxide derivative
AU2002331226A1 (en) 2002-08-09 2004-03-11 Prosidion Ltd. Methods for improving islet signaling in diabetes mellitus and for its prevention
AU2003262747A1 (en) * 2002-08-21 2004-03-11 Cytokinetics, Inc. Compounds, compositions, and methods
US7407955B2 (en) * 2002-08-21 2008-08-05 Boehringer Ingelheim Pharma Gmbh & Co., Kg 8-[3-amino-piperidin-1-yl]-xanthines, the preparation thereof and their use as pharmaceutical compositions
DE10238477A1 (en) * 2002-08-22 2004-03-04 Boehringer Ingelheim Pharma Gmbh & Co. Kg New purine derivatives, their production and their use as medicines
US7495005B2 (en) 2002-08-22 2009-02-24 Boehringer Ingelheim Pharma Gmbh & Co. Kg Xanthine derivatives, their preparation and their use in pharmaceutical compositions
US6998502B1 (en) 2002-09-05 2006-02-14 Sabinsa Corporation Convenient process of manufacture for difluoromethylornithine and related compounds
EP1398032A1 (en) 2002-09-10 2004-03-17 PheneX Pharmaceuticals AG 4-Oxo-quinazolines as LXR nuclear receptor binding compounds
EP1407774A1 (en) * 2002-09-10 2004-04-14 LION Bioscience AG 2-Amino-4-quinazolinones as LXR nuclear receptor binding compounds
JP2004123738A (en) 2002-09-11 2004-04-22 Takeda Chem Ind Ltd Sustained-release preparation
EP1543023B1 (en) 2002-09-18 2010-03-17 Prosidion Limited Secondary binding site of dipeptidyl peptidase iv (dp iv)
US20040058876A1 (en) * 2002-09-18 2004-03-25 Torsten Hoffmann Secondary binding site of dipeptidyl peptidase IV (DP IV)
US7262207B2 (en) 2002-09-19 2007-08-28 Abbott Laboratories Pharmaceutical compositions as inhibitors of dipeptidyl peptidase-IV (DPP-IV)
US6869966B2 (en) * 2002-09-30 2005-03-22 Banyu Pharmaceutical Co., Ltd. N-substituted-2-oxodihydropyridine derivatives
DE60320008T2 (en) 2002-10-18 2009-06-18 Merck & Co., Inc. HETEROCYCLIC BETA-AMINO COMPOUNDS AS INHIBITORS OF DIPEPTIDYLPEPTIDASE FOR TREATMENT OR PREVENTION OF DIABETES
AU2003301662A1 (en) 2002-10-21 2004-05-13 Bristol-Myers Squibb Company Quinazolinones and derivatives thereof as factor xa inhibitors
DE60332856D1 (en) 2002-10-23 2010-07-15 Bristol Myers Squibb Co GLYCINNITRIL BASED DIPEPTIDYLPEPTIDASE IV INHIBITORS
US7482337B2 (en) 2002-11-08 2009-01-27 Boehringer Ingelheim Pharma Gmbh & Co. Kg Xanthine derivatives, the preparation thereof and their use as pharmaceutical compositions
CA2502068A1 (en) * 2002-11-18 2004-06-03 Pfizer Products Inc. Dipeptidyl peptidase iv inhibiting fluorinated cyclic amides
DE10254304A1 (en) 2002-11-21 2004-06-03 Boehringer Ingelheim Pharma Gmbh & Co. Kg New xanthine derivatives, their production and their use as medicines
DE10256264A1 (en) 2002-12-03 2004-06-24 Boehringer Ingelheim Pharma Gmbh & Co. Kg New trisubstituted dihydro-imidazo-pyridazinone or imidazo-pyridinone derivatives, useful as dipeptidylpeptidase-IV inhibitors for e.g. treating diabetes mellitus type I or II, arthritis or obesity
US7109192B2 (en) * 2002-12-03 2006-09-19 Boehringer Ingelheim Pharma Gmbh & Co Kg Substituted imidazo-pyridinones and imidazo-pyridazinones, the preparation thereof and their use as pharmaceutical compositions
UY28103A1 (en) * 2002-12-03 2004-06-30 Boehringer Ingelheim Pharma NEW IMIDAZO-PIRIDINONAS REPLACED, ITS PREPARATION AND ITS EMPLOYMENT AS MEDICATIONS
WO2004062613A2 (en) 2003-01-13 2004-07-29 Bristol-Myers Squibb Company Hiv integrase inhibitors
US7148246B2 (en) 2003-02-27 2006-12-12 Sanofi-Aventis Deutschland Gmbh Cycloalkyl derivatives having bioisosteric carboxylic acid groups, processes for their preparation and their use as pharmaceuticals
WO2004078758A1 (en) * 2003-03-07 2004-09-16 Astrazeneca Ab Novel fused heterocycles and uses thereof
US20050014732A1 (en) * 2003-03-14 2005-01-20 Pharmacia Corporation Combination of an aldosterone receptor antagonist and an anti-diabetic agent
CA2518465A1 (en) 2003-03-25 2004-10-14 Takeda San Diego, Inc. Dipeptidyl peptidase inhibitors
TWI357408B (en) 2003-03-26 2012-02-01 Mitsubishi Tanabe Pharma Corp 3-substituted-4-pyrimidone derivatives
EP1961416B1 (en) * 2003-05-05 2013-01-23 Probiodrug AG Use of inhibitors of glutaminyl cyclase for treating psoriasis, rheumatoid arthritis or atherosclerosis.
WO2004098591A2 (en) 2003-05-05 2004-11-18 Probiodrug Ag Inhibitors of glutaminyl cyclase and their use in the treatment of neurological diseases
US20040229848A1 (en) 2003-05-05 2004-11-18 Hans-Ulrich Demuth Glutaminyl based DP IV-inhibitors
WO2004103993A1 (en) 2003-05-14 2004-12-02 Syrrx, Inc. Dipeptidyl peptidase inhibitors
US7566707B2 (en) * 2003-06-18 2009-07-28 Boehringer Ingelheim International Gmbh Imidazopyridazinone and imidazopyridone derivatives, the preparation thereof and their use as pharmaceutical compositions
NZ544221A (en) 2003-06-20 2009-05-31 Hoffmann La Roche Pyrido[2,1-a]-isoquinoline derivatives as DPP-IV inhibitors
MXPA05013734A (en) 2003-06-20 2006-03-08 Hoffmann La Roche Hexahydropyridoisoqinolines as dpp-iv inhibitors.
JO2625B1 (en) * 2003-06-24 2011-11-01 ميرك شارب اند دوم كوربوريشن Phosphoric acid salt of a dipeptidyl peptidase-IV inhibitor
US6995183B2 (en) * 2003-08-01 2006-02-07 Bristol Myers Squibb Company Adamantylglycine-based inhibitors of dipeptidyl peptidase IV and methods
KR20060041309A (en) 2003-08-13 2006-05-11 다케다 야쿠힌 고교 가부시키가이샤 4-pyrimidone derivatives and their use as peptidyl peptidase inhibitors
WO2005019168A2 (en) * 2003-08-20 2005-03-03 Pfizer Products Inc. Fluorinated lysine derivatives as dipeptidyl peptidase iv inhibitors
WO2005026148A1 (en) 2003-09-08 2005-03-24 Takeda San Diego, Inc. Dipeptidyl peptidase inhibitors
JP2007513058A (en) 2003-09-08 2007-05-24 武田薬品工業株式会社 Dipeptidyl peptidase inhibitor
AU2004293012A1 (en) * 2003-11-19 2005-06-09 Rustum S. Boyce Quinazolinone compounds with reduced bioaccumulation
US7304086B2 (en) 2004-02-05 2007-12-04 Probiodrug Ag Inhibitors of glutaminyl cyclase
CN102134231B (en) 2004-03-15 2020-08-04 武田药品工业株式会社 Dipeptidyl peptidase inhibitors
US7872124B2 (en) 2004-12-21 2011-01-18 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8906901B2 (en) 2005-09-14 2014-12-09 Takeda Pharmaceutical Company Limited Administration of dipeptidyl peptidase inhibitors
US8883714B2 (en) 2008-04-07 2014-11-11 Arena Pharmaceuticals, Inc. Pharmaceutical compositions comprising GPR119 agonists which act as peptide YY (PYY) secretagogues
US9533954B2 (en) 2010-12-22 2017-01-03 Incyte Corporation Substituted imidazopyridazines and benzimidazoles as inhibitors of FGFR3
US9388185B2 (en) 2012-08-10 2016-07-12 Incyte Holdings Corporation Substituted pyrrolo[2,3-b]pyrazines as FGFR inhibitors
US9266892B2 (en) 2012-12-19 2016-02-23 Incyte Holdings Corporation Fused pyrazoles as FGFR inhibitors
US9533984B2 (en) 2013-04-19 2017-01-03 Incyte Holdings Corporation Bicyclic heterocycles as FGFR inhibitors
US9580423B2 (en) 2015-02-20 2017-02-28 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US11591329B2 (en) 2019-07-09 2023-02-28 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US11566028B2 (en) 2019-10-16 2023-01-31 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors

Also Published As

Publication number Publication date
US20040259870A1 (en) 2004-12-23
CN1894234A (en) 2007-01-10
CA2518465A1 (en) 2004-10-14
KR20050122220A (en) 2005-12-28
US20040242566A1 (en) 2004-12-02
US20040242568A1 (en) 2004-12-02
EP1608317A2 (en) 2005-12-28
JP4887139B2 (en) 2012-02-29
WO2004087053A3 (en) 2006-08-31
WO2004087053A2 (en) 2004-10-14
US7550590B2 (en) 2009-06-23
EP1608317B1 (en) 2012-09-26
US20050004117A1 (en) 2005-01-06
US7687625B2 (en) 2010-03-30
JP2007524600A (en) 2007-08-30
EP1608317A4 (en) 2006-12-27

Similar Documents

Publication Publication Date Title
US7550590B2 (en) Dipeptidyl peptidase inhibitors
US7872124B2 (en) Dipeptidyl peptidase inhibitors
EP1506967B1 (en) Dipeptidyl peptidase inhibitors
US7825242B2 (en) Dipeptidyl peptidase inhibitors
EP1697342A2 (en) Dipeptidyl peptidase inhibitors
WO2005095381A1 (en) Dipeptidyl peptidase inhibitors
US7550598B2 (en) Kinase inhibitors
EP1911754B1 (en) Dipeptidyl peptidase inhibitors

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

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

AL Designated countries for regional patents

Kind code of ref document: A2

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

COP Corrected version of pamphlet

Free format text: PAGE 173, CLAIMS, REPLACED BY A NEW PAGE 173

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2518465

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2006509315

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 1020057018103

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 2004758366

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 20048119000

Country of ref document: CN

WWP Wipo information: published in national office

Ref document number: 2004758366

Country of ref document: EP

Ref document number: 1020057018103

Country of ref document: KR

DPEN Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed from 20040101)