CA2535619A1 - 4-pyrimidone derivatives and their use as peptidyl peptidase inhibitors - Google Patents

4-pyrimidone derivatives and their use as peptidyl peptidase inhibitors Download PDF

Info

Publication number
CA2535619A1
CA2535619A1 CA002535619A CA2535619A CA2535619A1 CA 2535619 A1 CA2535619 A1 CA 2535619A1 CA 002535619 A CA002535619 A CA 002535619A CA 2535619 A CA2535619 A CA 2535619A CA 2535619 A1 CA2535619 A1 CA 2535619A1
Authority
CA
Canada
Prior art keywords
alkyl
group
phenyl
amino
substituted
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA002535619A
Other languages
French (fr)
Inventor
Jun Feng
Stephen L. Gwaltney
Stephen W. Kaldor
Jeffrey A. Stafford
Michael B. Wallace
Zhiyuan Zhang
Bruce Elder
Paul Isbester
Grant Palmer
Luckner Ulysse
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Takeda Pharmaceutical Co Ltd
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Publication of CA2535619A1 publication Critical patent/CA2535619A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/32One oxygen, sulfur or nitrogen atom
    • C07D239/34One oxygen atom
    • C07D239/36One oxygen atom as doubly bound oxygen atom or as unsubstituted hydroxy radical
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • A61P3/08Drugs 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
    • A61P31/04Antibacterial agents
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/46Two or more oxygen, sulphur or nitrogen atoms
    • C07D239/47One nitrogen atom and one oxygen or sulfur atom, e.g. cytosine
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/46Two or more oxygen, sulphur or nitrogen atoms
    • C07D239/52Two oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic 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 three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/06Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/14Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond

Abstract

Compounds, pharmaceuticals, kits and methods are provided for use with DPP-IV
inhibitors comprising Formula (I), wherein the substituents are as described herein.

Description

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 IV (lUBMB Enzyme Nomenclature EC.3.4.14.5) is a type II
membrane protein that has been referred to in the literature by a wide a variety of names including DPP4, DP4, DAP-IV, FAP(3, 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 dipeptidylaminopeptidase; dipeptidyl-peptide hydrolase;
glycylprolyl aminopeptidase; dipeptidyl-aminopeptidase IV; DPP IV/CD26; amino acyl-prolyl dipeptidyl aminopeptidase; T cell triggering molecule Tp 103; X-PDAP.
Dipeptidyl Peptidase IV 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, VlP) in vitro.

S: ~:~-~ .. . ..._ ..... ...
[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 1I
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 (t1/2=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. Imrnunol., 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. Iminunol., 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. Immun.,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 HIV-1 and HIV-2 viruses in CD4+ T-cells (Wakselman, M., Nguyen, C., Mazaleyrat, J.-P., Callebaut, C., Krust, B., Hovanessian, A. G., Inhibition of HIV-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 24th European Peptide Symposium 1996).
Additionally, DPP-IV has been shown to associate with the enzyme adenosine deaminase (ADA) on the surface of T-cells (Kameoka, J., et al., Science, 193, 26 466).
ADA deficiency causes severe combined immunodeficiency disease (SCID) in humans. This ADA-interaction may provide clues to the pathophysiology of SLID. 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 DPP-IV 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-IV
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 /LTS93/017173 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 inactivation 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-IV can also cleave neuropeptides and has been shown to modulate the activity of neuroactive peptides substance P, neuropeptide Y and CLIP
(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-IV. Nonetheless, a need still exists for new DPP-IV inhibitors that have advantageous potency, stability, selectivity, toxicity and/or pharmacodynamics properties. In this regard, a novel class of DPP-IV
inhibitors are provided herein.
SUMMARY OF THE INVENTION
[0017] The present invention relates to compounds that have activity for inhibiting DPP-IV. 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-IV. 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-IV inhibitor according to the present invention as an active ingredient.
Pharmaceutical compositions according to the invention rnay optionally comprise 0.001 %-100%
of one or more DPP-IV 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 stmt), 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-IV.
[0020] In one embodiment, a kit is provided that comprises a composition comprising at least one DPP-IV 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.
[0021] Tn another embodiment, an article of manufacture is provided that comprises a composition comprising at least one DPP-IV 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-IV.
[0025] In another embodiment, the compounds, compositions, kits and articles of manufacture are used to treat a disease state for which DPP-IV possesses activity that contributes to the pathology andlor symptomology of the disease state.
[0026] In another embodiment, a compound is administered to a subject wherein DPP-IV
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-IV.
[0028] In another embodiment, a method of inhibiting DPP-IV is provided that comprises contacting DPP-IV 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-IV in vivo.
[0030] In another embodiment, a method of inhibiting DPP-IV 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-IV 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-1V, or which is known to be treated by DPP-IV
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-IV, or which is known to be treated by DPP-IV
inhibitors.
[0036] In another embodiment, a method is provided for treating a disease state for which DPP-IV 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-IV 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] In another embodiment, a method is provided for treating a disease state for which DPP-IV 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] In 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, andlor 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-IV, in particular diabetes, more 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, obesity, imrnunosuppressants 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-IV and thus may themselves be prodrugs for another compound. Such prodrugs of another compound may or may not themselves independently have DPP-IV inhibitory activity.
BRIEF DESCRIPTION OF THE FIGURE
[0043] Figure 1 illustrates a ribbon diagram overview of the structure of DPP-IV, highlighting the secondary structural elements of the protein.
DEFINITIONS
[0044] Unless otherwise stated, the following terms used in the specification and claims shall have the following meanings for the purposes of this Application.
[0045] "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.
[0046] "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.
[0047] "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 Cx_Y alkenyl are typically used where X
and Y
indicate the number of carbon atoms in the chain. For example, C2_6 alkenyl includes alkenyls that have a chain of between 2 and 6 carbons.
[0048] "Alkoxy" means an oxygen moiety having a further alkyl substituent. The alkoxy groups of the present invention can be optionally substituted.
[0049] "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 Cx_Y alkyl are typically used where X and Y indicate the number of carbon atoms in the chain.
For example, Cl_6 alkyl includes alkyls that have a chain of between l and 6 carbons (e.g., methyl, ethyl, propyl, isopropyl, butyl, sec-butyl, isobutyl, tert-butyl, vinyl, allyl,1-propenyl, isopropenyl, l-butenyl, 2-butenyl, 3-butenyl, 2-methylallyl, ethynyl, l-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_lo)aryl(Cl_3)alkyl includes, benzyl, phenethyl, 1-phenylethyl, 3-phenylpropyl, 2-thienylmethyl, 2-pyridinylmethyl and the like).
[0050] "Alkylene", unless indicated otherwise, means a straight or branched, saturated or unsaturated, aliphatic, divalent radical. Cx alkylene and Cx_Y alkylene are typically used where X and Y indicate the number of carbon atoms in the chain. For example, Cl_6 alkylene includes methylene (-CHZ-), ethylene (-CH2CH2-), trimethylene (-CHZCH2CH2-), tetramethylene (-CH2CH2CHZCH2-) 2-butenylene (-CHZCH=CHCH2-), 2-methyltetramethylene (-CHaCH(CH3)CHZCH2-), pentamethylene (-CH2CH2CH2CH2CH2-) and the like).
[0051] "Alkylidene" means a straight or branched saturated or unsaturated, aliphatic radical connected to the parent molecule by a double bond. Cx alkylidene and Cx_Y alkylidene are typically used where X and Y indicate the number of carbon atoms in the chain. For example, Cl_6 alkylidene includes methylene (=CH2), ethylidene (=CHCH3), isopropylidene (=C(CH3)2), propylidene (=CHCHZCH3), allylidene (=CH-CH=CHa), and the like).
[0052] "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 Cx_Y alkynyl are typically used where X
and Y
indicate the number of carbon atoms in the chain. For example, C2_6 alkynyl includes alk5myls that have a chain of between 2 and 6 carbons.
[0053] "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, -NHCI_3-alkyl, -N(Cl_3-alkyl)2 and the like.
Optionally, the two substituents together with the nitrogen may also form a ring. Unless indicated otherwise, the compounds of the invention containing amino moieties may include protected derivatives thereof. Suitable protecting groups for amino moieties include acetyl, tent-butoxycarbonyl, benzyloxycarbonyl, and the like.
[0054] "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.
[0055] "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).
[0056] "Aromatic" means a moiety wherein the constituent atoms make up an unsaturated ring system, all atoms in the ring system are sp2 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).
[0057] "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 Cx_Y aryl are typically used where X and Y indicate the number of atoms in the ring.
[0058] "Aryloxy" means an oxygen moiety having a further aryl substituent. The aryloxy groups of the present invention can be optionally substituted.
[0059] "Bicycloalkyl" means a saturated or partially unsaturated fused bicyclic or bridged polycyclic ring assembly.
[0060] "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 Cx_y 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.
[0061] "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 borneol, norbornane, 7-oxabicyclo[2.2.1]heptane, and the like.
One or both rings of the bicyclic system may also comprise heteroatoms.
[0062] "Carbamoyl" means the radical -OC(O)NRaRb where Ra and Rb are each independently two further substituents where a hydrogen or carbon atom is attached to the nitrogen.
[0063] "Carbocycle" means a ring consisting of carbon atoms.
[0064] "Carbocyclic ketone derivative" means a carbocyclic derivative wherein the ring contains a -CO- moiety.
[0065] "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.
[0066] "Carboxy" means the radical -COZ-. 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.
[0067] "Cyano" means the radical -CN.
[0068] "Cycloalkyl" means a non-aromatic, saturated or partially unsaturated, monocyclic, fused bicyclic or bridged polycyclic ring assembly. Cx cycloalkyl and Cx_y cycloalkyl are typically used where X and Y indicate the number of carbon atoms in the ring assembly. For example, C3_io cycloalkyl includes cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexenyl, 2,5-cyclohexadienyl, bicyclo[2.2.2]octyl, adamantan-1-yl, decahydronaphthyl, oxocyclohexyl, dioxocyclohexyl, thiocyclohexyl, 2-oxobicyclo[2.2.1]hept-1-yl, and the like.
[0069] "Cycloalkylene" means a divalent saturated or partially unsaturated, monocyclic or polycyclic ring assembly. Cx cycloalkylene and Cx_Y cycloalkylene are typically used where X and Y indicate the number of carbon atoms in the ring assembly.
[0070] "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, i.e., the "side effects" of such therapy.
[0071] "Fused ring" as used herein refers to a ring that is bonded to another ring to form a compound having 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.
[0072] "Halo" means fluoro, chloro, bromo or iodo.
[0073] "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 (Cl_3)alkyl includes chloromethyl, dichloromethyl, difluoromethyl, trifluoromethyl, 2,2,2-trifluoroethyl, perfluoroethyl, 2,2,2-trifluoro-1,1-dichloroethyl, and the like).
[0074] "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 from benzo[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[1,2a]pyridine, quinoline, isoquinoline, phthalazine, quinoxaline, naphthyridine, quinolizine, indole, isoindole, indazole, indoline, benzoxazole, benzopyrazole, benzothiazole, imidazo[1,5-a]pyridine, pyrazolo[1,5-a]pyridine, imidazo[1,2-a]pyrimidine, imidazo[1,2-c]pyrimidine, imidazo[1,5-a]pyrimidine, imidazo[1,5-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[1,5-a]pyridine, pyrrolo[1,2-b]pyridazine, pyrrolo[1,2-c]pyrimidine, pyrrolo[1,2-a]pyrimidine, pyrrolo[1,2-a]pyrazine, triazo[1,5-a]pyridine, pteridine, purine, carbazole, acridine, phenazine, phenothiazene, phenoxazine, 1,2-dihydropyrrolo[3,2,1-hi]indole, indolizine, pyrido[1,2-a]indole and 2(1H)-pyridinone. 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.
[0075] "Heteroaryloxy" means an oxygen moiety having a further heteroaryl substituent.
The heteroaryloxy groups of the present invention can be optionally substituted.
[0076] "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.
[0077] "Heteroatom moiety" includes a moiety where the atom by which the moiety is attached is not a carbon. Examples of heteroatom moieties include -N=, -NR~-, -N+(O-)=, -O-, -S- or -S(O)2-, wherein R~ is further substituent.
[0078] "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]hept-3-y1,2-aza-bicyclo[3.1.0]hex-2-y1,3-aza-bicyclo[3.1.0]hex-3-yl,andthe like.
[0079] "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(C4_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.
[0080] "Heterocycloalkyl" means cycloalkyl, as defined in this Application, 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.
[0081] "Heterocycloalkylene" means cycloalkylene, as defined in this Application, provided that one or more of the ring member carbon atoms is replaced by a heteroatom.
[0082] "Hydroxy" means the radical-OH.
[0083] "Iminoketone derivative" means a derivative comprising the moiety -C(NR)-, wherein R comprises a hydrogen or carbon atom attached to the nitrogen.
[0084] "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 minor 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 2'1-I
enantiomeric pairs, where h 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).
[0085] "Leaving group" means a moiety that may be displaced by nucleophilic substitution. Exemplary nucleophilically displaceable leaving groups, include, but are not limited to halo (fluoro, chloro, bromo, or iodo), thio, hydroxyl, nitro, azido, phenylsulfoxido, aryloxy, alkoxy, alkylsulfonate (particularly methylsulphonyloxy), and arylsulfonate (particularly p-tolylsulphonyloxy). A myriad of suitable leaving groups are known to those of skill in the art and may be used herein.
[0086] "Nitro" means the radical -NO2.
[0087] "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)oxaalkyl refers to a chain comprising between 2 and 6 carbons and one or more oxygen atoms positioned between the carbon atoms.
[0088] "Oxoalkyl" means an alkyl, further substituted with a carbonyl group.
The carbonyl group may be an aldehyde, ketone, ester, amide, acid, or acid chloride.
[0089] "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.
[0090] "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, malefic acid, fumaric acid, tartatic acid, citric acid, benzoic acid, o-(4-hydroxybenzoyl)benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid,1,2-ethanedisulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, p-chlorobenzenesulfonic acid, 2-naphthalenesulfonic acid, p-toluenesulfonic acid, camphorsulfonic acid, 4-methylbicyclo[2.2.2]oct-2-ene-1-carboxylic acid, glucoheptonic acid, 4,4'-methylenebis(3-hydroxy-2-ene-1-carboxylic acid), 3-phenylpropionic acid, trimethylacetic acid, tertiary butylacetic acid, lauryl sulfuric acid, gluconic acid, glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid, muconic acid and the like.
[0091] 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 ethanolamine, diethanolamine, triethanolamine, tromethamine, N methylglucamine and the like.
[0092] "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-IV 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 irz vivo to the amine compound.
[0093] "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.
[0094] "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.
[0095] "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.
[0096] "Sulfonyl" means the radical -S02-. 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.
[0097] "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.
[0098] "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.
[0099] "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).
[0100] 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 Cl alkyl indicates that there is one carbon atom but does not indicate what are the substituents on the carbon atom. Hence, a Cl alkyl comprises methyl (i.e., -CH3) as well as -CRaRbR~ where Ra, Rb, and R~ may each independently be hydrogen or any other substituent where the atom attached to the carbon is a heteroatom or cyano. Hence, CF3, CHaOH and CH2CN, for example, are all C1 alkyls.
DETAILED DESCRIPTION OF THE INVENTION
[0101] The present invention relates to compounds, compositions, kits and articles of manufacture that may be used to inhibit dipeptidyl peptidases IV (referred to herein as DPP-IV).
[0102] DPP-IV (EC.3.4.14.5 also known as DPP4, DP4, DAP-IV, 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-IV has a serine type mechanism of protease activity, cleaving off dipeptides from the amino-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-IV 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-IV is described in GenBank Accession Number NM_001935 ("Dipeptidyl peptidase IV (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 IV mRNA 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).
[0103] DPP-IV 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:
Subfamil,~: Dipeptidyl-peptidase; Oligopeptidase B (EC 3.4.21.83);
Oligopeptidase B; Prolyl oligopeptidase (EC 3.4.21.26);
Subfamil,~: Dipeptidyl aminopeptidase A; Dipeptidyl aminopeptidase B
Dipeptidyl-peptidase IV (EC 3.4.14.5); Dipeptidyl-peptidase V
Fibroblast activation protein alpha subunit; Seprase Subfamil,~: Acylaminoacyl-peptidase (EC 3.4.19.1) [0104] It is noted that the compounds of the present invention may also possess inhibitory activity for other S9 family members and thus may be used to address disease states associated with these other family members.
1. CRYSTAL STRUCTURE OF I)PP-IV
[0105] Syrrx, Inc. (San Diego, California) recently solved the crystal structure of DPP-IV.
Knowledge of the crystal structure was used to guide the design of the DPP-IV
inhibitors provided herein.
[0106] Figure 1 illustrates a ribbon diagram overview of the structure of DPP-IV, highlighting secondary structural elements of the protein. DPP-IV is a cylindrical shaped molecule with an approximate height of 70 ~ and a diameter of 60 ~. The catalytic triad of DPP-IV (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-IV. The catalytic domain is covalently linked to the (3-propeller domain. The catalytic domain of DPP-IV includes residues 1-67 and 511-778. The catalytic domain of DPP-IV adopts a characteristic a/(3 hydrolase fold. The core of this domain contains an 8-stranded [3-sheet with all strands being parallel except one. The a-sheet is significantly twisted and is flanked by three a-helices on one side and five a-helices on the other. The topology of the [3-strands is 1, 2, -1x, 2x and (lx) (J. S. Richardson: The anatomy and taxonomy of protein structure; (1981) Adv. Protein Claeyn. 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-IV
inhibitors of the present invention.
2. DPP-IV INHIBITORS
[0107] In one embodiment, DPP-IV inhibitors of the present invention comprise a compound of Formula I:
R2 Q~N/L-X
R3 ~N
I
wherein Q is selected from the group consisting of CO, SO, SOz, and C=NR4;
Z is selected from the group consisting of halo, perhalo(Cl-lo)alkyl, amino, cyano, thio, (C1-lo)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, carbonyl (Cl_3)alkyl, thiocarbonyl (Cl_3)alkyl, sulfonyl (Cl_3)alkyl, sulfinyl (Cl_3)alkyl, imino (Cl_3)alkyl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, 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;
Rz and R3 are each independently selected from the group consisting of hydrogen, halo, perhalo(Cl-io)alkyl, amino, cyano, nitro, thio, (Cl-lo)alkyl, alkene, alkyne, (C3_lz)cycloalkyl, hetero(C3-iz)cycloalkyl, aryl(Cl-lo)alkyl, heteroaryl (Cl-5)alkyl, (C9-iz)bicycloaryl, hetero(C8_lz)bicycloaryl, carbonyl (Cl_3)alkyl, thiocarbonyl (Cl_3)alkyl, sulfonyl (Cl_3)alkyl, sulfinyl (Cl_3)alkyl, imino (C1_3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;
R~ is selected from the group consisting of hydrogen, (Cl_lo)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, arylalkyl, heteroarylalkyl, bicycloaryl, heterobicycloaryl, each substituted or unsubstituted;
L is a linker providing 0-6 atom separation between X and the ring to which L
is attached; and X is selected from the group consisting of (Cl-lo)alkyl, (C3_12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(Cl-lo)alkyl, heteroaryl(Cl-5)alkyl, (C9_12)bicycloaryl, hetero(C~_12)bicycloaryl, carbonyl (Cl_3)alkyl, thiocarbonyl (Cl_3)alkyl, sulfonyl (Cl_3)alkyl, sulfinyl (Ci_3)alkyl, imino (C1_3)alkyl, amino, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, cyano, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
[0108] In another embodiment, DPP-IV inhibitors of the present invention comprise the compound of Formula II:
Rs Rs R2 Q~ ~
N~~~X
R3 ~N Z
a wherein nis0, 1,2,or3;
Q is selected from the group consisting of CO, SO, 502, and C=NR~;
Z is selected from the group consisting of halo, perhalo(Cl-lo)alkyl, amino, cyano, thin, (Cl-lo)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, carbonyl (Cl_3)alkyl, thiocarbonyl (Cl_3)alkyl, sulfonyl (Cl_3)alkyl, sulfinyl (Cl_3)alkyl, imino (Cl_3)alkyl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, 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;
R2 and R3 are each independently selected from the group consisting of hydrogen, halo, perhalo(Cl-lo)alkyl, amino, cyano, nitro, thio, (Cl-lo)alkyl, alkene, alkyne, (Cs-i2)cycloalkyl, hetero(C3-i2)cycloalkyl, aryl(Ci-io)alkyl, heteroaryl (Cl-5)alkyl, (C9-i2)bicycloaryl, hetero(C8_12)bicycloaryl, carbonyl (C1_3)alkyl, thiocarbonyl (Cl_3)alkyl, sulfonyl (Cl_3)alkyl, sulfinyl (Cl_3)alkyl, imino (Cl_3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;
R4 is selected from the group consisting of hydrogen, (C1_lo)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, arylalkyl, heteroarylalkyl, bicycloaryl, heterobicycloaryl, each substituted or unsubstituted;
RS and R6 are each independently selected from the group consisting of hydrogen, a substituted or unsubstituted (Cl_io)alkyl, a substituted or unsubstituted (C1_lo)alkoxy, cyano, and halo, or where RS and R6 are taken together to form a ring; and X is selected from the group consisting of (Cl-lo)alkYl, (C3_i2)cycloalkyl, hetero(C3-i2)cycloalkyl, aryl(Cl-lo)alkyl, heteroaryl(Cl-5)alkyl, (C9_12)bicycloaryl, hetero(C8_12)bicycloaryl, carbonyl (Cl_3)alkyl, thiocarbonyl (Cl_3)alkyl, sulfonyl (Cl_3)alkyl, sulfinyl (Cl_3)alkyl, imino (Ci_3)alkyl, amino, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, cyano, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
[0109] In another embodiment, DPP-IV inhibitors of the present invention comprise a compound of Formula 1B:

III
~m wherein m is 0, 1, 2, 3, 4, or 5;
n is 0, 1, 2, or 3;
Q is selected from the group consisting of CO, SO, 502, and C=NR4;
Z is selected from the group consisting of halo, perhalo(Cl-io)alkyl, amino, cyano, thio, (Cl-lo)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, carbonyl (Cl_3)alkyl, thiocarbonyl (Cl_3)alkyl, sulfonyl (Cl_3)alkyl, sulfinyl (Cl_3)alkyl, imino (Cl_3)alkyl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, 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;
R2 and R3 are each independently selected from the group consisting of hydrogen, halo, perhalo(Cl-lo)alkyl, amino, cyano, nitro, thio, (Cl-lo)alkYl, alkene, alkyne, (C3_12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(C1-lo)alkyl, heteroaryl (Cl-5)alkyl, (C9-iz)bicycloaryl, hetero(C8_12)bicycloaryl, carbonyl (Cl_3)alkyl, thiocarbonyl (Cl_3)alkyl, sulfonyl (Cl_3)alkyl, sulfinyl (C1_3)alkyl, imino (Cl_3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;
R~ is selected from the group consisting of hydrogen, (Cl_lo)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, arylalkyl, heteroarylalkyl, bicycloaryl, heterobicycloaryl, each substituted or unsubstituted;
RS and R6 are each independently selected from the group consisting of hydrogen, a substituted or unsubstituted (Cl_lo)alkyl, a substituted or unsubstituted (Cl_lo)alkoxy, cyano, and halo, or where RS and R6 are taken together to form a ring; and each R~ is independently selected from the group consisting of halo, perhalo(Cl-io)alkyl, CF3, (Cl_lo)alkyl, alkenyl, alkynyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, hydroxy, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
[0110] In another embodiment, DPP-1V inhibitors of the present invention comprise a compound of Formulae IVa, IVb and IVc:

IVa IVb IVc wherein nis0, 1,2,or3;
Q is selected from the group consisting of CO, SO, 502, and C=NR4;
each of T, U, V, W and Y is independently nitrogen or CR16, provided that no more than two of T, U, V, W and Y are nitrogen;
Z is selected from the group consisting of halo, perhalo(Cl-lo)alkyl, amino, cyano, thio, (C1-lo)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, carbonyl (Cl_3)alkyl, thiocarbonyl (Cl_3)alkyl, sulfonyl (Cl_3)alkyl, sulfinyl (C1_3)alkyl, imino (Cl_3)alkyl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, 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;
R2 and R3 are each independently selected from the group consisting of hydrogen, halo, perhalo(Cl-io)alkyl, amino, cyano, nitro, thio, (Cl-lo)alkyl, alkene, alkyne, (C3-i2)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(C1-lo)alkyl, heteroaryl (Cl-5)alkyl, (C9-i2)bicycloaryl, hetero(C$_12)bicycloaryl, carbonyl (C1_3)alkyl, thiocarbonyl (Cl_3)alkyl, sulfonyl (Cl_3)alkyl, sulfinyl (Cl_3)alkyl, imino (Cl_3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;
R4 is selected from the group consisting of hydrogen, (Cl_lo)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, arylalkyl, heteroarylalkyl, bicycloaryl, heterobicycloaryl, each substituted or unsubstituted;
RS and R6 are each independently selected from the group consisting of hydrogen, a substituted or unsubstituted (Cl_lo)alkyl, a substituted or unsubstituted (Cl_io)alkoxY, cyano, and halo, or where RS and R6 are taken together to form a ring; and each R16 is independently selected from the group consisting of halo, perhalo(Cl-lo)alkyl, CF3, (Cl_lo)alkyl, alkenyl, alkynyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, hydroxy, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
[0111] In another embodiment, DPP-IV inhibitors of the present invention comprise a compound of Formula V:
R2 QWN~Ri ,Rs Rio V
wherein Q is selected from the group consisting of CO, SO, 502, and C=NR4;
Rl is selected from the group consisting of (Cl-io)alkyl, (C3_i2)cycloalkyl, (C3_l2)cycloalkyl(Cl_5)alkyl, hetero(C3-12)cycloalkyl, hetero(C3-i2)cycloalkyl(Cl_5)alkyl, aryl(Cl-lo)alkyl, heteroaryl(C1-5)alkyl, (C9_12)bicycloaryl, (C9_12)bicycloaryl(Cl_5)alkyl, hetero(C8_12)bicycloaryl, hetero(C8_12)bicycloaryl(Cl-5)alkyl, carbonyl (Cl_3)alkyl, thiocarbonyl (C1_3)alkyl, sulfonyl (Cl_3)alkyl, sulfinyl (Cl_3)alkyl, imino (Cl_3)alkyl, amino, cyano, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;
R2 and R3 are each independently selected from the group consisting of hydrogen, halo, perhalo(C1-lo)alkyl, amino, cyano, nitro, thio, (Cl-lo)alkyl, alkene, alkyne, (C3_12)cycloalkyl, hetero(C3-i2)cycloalkyl, aryl(Cl-lo)alkyl, heteroaryl (Cl-5)alkyl, (C9-i2)bicycloaryl, hetero(C8_12)bicycloaryl, carbonyl (C1_3)alkyl, thiocarbonyl (Cl_3)alkyl, sulfonyl (Cl_3)alkyl, sulfinyl (Cl_3)alkyl, imino (Cl_3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;
R4 is selected from the group consisting of hydrogen, (Cl_lo)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, arylalkyl, heteroarylalkyl, bicycloaryl, heterobicycloaryl, each substituted or unsubstituted; and R9 and Rlo are each independently selected from the group consisting of hydrogen, perhalo(Cl-lo)alkyl, amino, cyano, (Cl-lo)alkyl, (C3_12)cycloalkyl, hetero(C3-ia)cycloalkyl, aryl(Cl-lo)alkyl, heteroaryl (Cl-5)alkyl, (C9_12)bicycloaryl, hetero(C8_12)bicycloaryl, carbonyl (Cl_3)alkyl, thiocarbonyl (Cl_3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, sulfonyl group, sulfinyl group, each substituted or unsubstituted, or R~ and Rlo are taken together to form a 5, 6, or 7 membered ring, each substituted or unsubstituted.
[0112] In another embodiment, DPP-IV inhibitors of the present invention comprise a compound of Formula VI:
R2 ~\N~L-X

Rio VI
wherein Q is selected from the group consisting of CO, SO, SO2, and C=NR4;
R2 and R3 are each independently selected from the group consisting of hydrogen, halo, perhalo(Cl-lo)alkyl, amino, cyano, nitro, thin, (Cl-lo)alkyl, alkene, alkyne, (C3_12)cycloalkyl, hetero(C3-iz)cycloalkyl, aryl(Cl-lo)alkyl, heteroaryl (Cl-5)alkyl, (C9_12)bicycloaryl, hetero(C8_12)bicycloaryl, carbonyl (Cl_3)alkyl, thiocarbonyl (C1_3)alkyl, sulfonyl (Cl_3)alkyl, sulfinyl (C1_3)alkyl, imino (Cl_3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;
R4 is selected from the group consisting of hydrogen, (Cl_lo)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, arylalkyl, heteroarylalkyl, bicycloaryl, heterobicycloaryl, each substituted or unsubstituted;
R9 and Rlo are each independently selected from the group consisting of hydrogen, perhalo(Cl-lo)alkyl, amino, cyano, (Cl-lo)alkyl, (C3_ia)cYcloalkyl, hetero(C3-ia)cYcloalkyl, aryl(Cl-lo)alkyl, heteroaryl (Cl-5)alkyl, (C9_12)bicycloaryl, hetero(C8_12)bicycloaryl, carbonyl (Cl_3)alkyl, thiocarbonyl (C1_3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, sulfonyl group, sulfinyl group, each substituted or unsubstituted, or R9 and Rlo are taken together to form a 5, 6, or 7 membered ring, each substituted or unsubstituted;
L is a linker providing 0-6 atom separation between X and the ring to which L
is attached; and X is selected from the group consisting of (Cl-lo)alkyl, (C3_12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(Cl-lo)alkYl, heteroaryl(C1-5)alkyl, (C9_12)bicycloaryl, hetero(C~_12)bicycloaryl, carbonyl (Cl_3)alkyl, thiocarbonyl (Cl_3)alkyl, sulfonyl (Cl_3)alkyl, sulfinyl (Cl_3)alkyl, imino (Cl_3)alkyl, amino, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, cyano, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
[0113] In yet another embodiment, DPP-IV inhibitors of the present invention comprise a compound of Formula VII:

~n R2 Qw I N
,Rs Rio VII
wherein n is 0, 1, 2, or 3;
Q is selected from the group consisting of CO, SO, SO2, and C=NR4;
R2 and R3 are each independently selected from the group consisting of hydrogen, halo, perhalo(Cl-lo)alkyl, amino, cyano, nitro, thio, (C1-lo)alkYl, alkene, alkyne, (Cs-ia)cycloalkyl, hetero(C3-ia)cycloalkyl, aryl(C1-lo)alkyl, heteroaryl (Cl-5)alkyl, (C9_12)bicycloaryl, hetero(C$_12)bicycloaryl, carbonyl (C1_3)alkyl, thiocarbonyl (Cl_3)alkyl, sulfonyl (Cl_~)alkyl, sulfinyl (C1_3)alkyl, imino (C1_3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;
R~. is selected from the group consisting of hydrogen, (Cl_lo)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, arylalkyl, heteroarylalkyl, bicycloaryl, heterobicycloaryl, each substituted or unsubstituted;

RS and R6 are each independently selected from the group consisting of hydrogen, a substituted or unsubstituted (Cl_lo)alkyl, a substituted or unsubstituted (Cl_lo)alkoxy, cyano, and halo, or where RS and R6 are taken together to form a ring;
R9 and Rio are each independently selected from the group consisting of hydrogen, perhalo(Cl-io)alkyl, amino, cyano, (Cl-lo)alkYl, (C3_12)cycloalkyl, hetero(C3-i2)cycloalkyl, aryl(Cl-lo)alkYl, heteroaryl (C1-5)alkyl, (C9_12)bicycloaryl, hetero(C8_12)bicycloaryl, carbonyl (Cl_3)alkyl, thiocarbonyl (C1_3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, sulfonyl group, sulfinyl group, each substituted or unsubstituted, or R9 and Rlo are taken together to form a 5, 6, or 7 membered ring, each substituted or unsubstituted; and X is selected from the group consisting of (Cl-lo)alkYl, (C3_12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(Cl-lo)alkYl, heteroaryl(Cl-5)alkyl, (C9_i2)bicycloaryl, hetero(C4_i2)bicycloaryl, carbonyl (C1_3)alkyl, thiocarbonyl (C1_3)alkyl, sulfonyl (Cl_3)alkyl, sulfinyl (Cl_3)alkyl, imino (C1_3)alkyl, amino, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, cyano, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
[0114] In another embodiment, DPP-IV inhibitors of the present invention comprise a compound of Formula VIII:
R

~R7)m R2 Q~
I N
~N/Rs Rio VIII
wherein n is 0, 1, 2, or 3;
m is 0, 1, 2, 3, 4, or 5;
Q is selected from the group consisting of CO, SO, 502, and C=NR~;
R2 and R3 are each independently selected from the group consisting of hydrogen, halo, perhalo(Cl-lo)alkyl, amino, cyano, vitro, thio, (Cl-lo)alkYl, alkene, alkyne, 2~

(C3_12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(Cl-lo)alkyl, heteroaryl (Cl-5)alkyl, (C9-ia)bicycloaryl, hetero(C$_12)bicycloaryl, carbonyl (Cl_3)alkyl, thiocarbonyl (Cl_3)alkyl, sulfonyl (Cl_3)alkyl, sulfinyl (C1_3)alkyl, imino (Cl_3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;
R4 is selected from the group consisting of hydrogen, (Cl_io)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, arylalkyl, heteroarylalkyl, bicycloaryl, heterobicycloaryl, each substituted or unsubstituted;
RS and R6 are each independently selected from the group consisting of hydrogen, a substituted or unsubstituted (Cl_lo)alkyl, a substituted or unsubstituted (Cl_lo)alkoxy, cyano, and halo, or where RS and R6 are taken together to form a ring;
each R~ is independently selected from the group consisting of halo, perhalo(Cl-lo)alkYl, CF3, (Cl_lo)alkyl, alkenyl, alkynyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thin, cyano, nitro, hydroxy, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted; and R9 and Rlo are each independently selected from the group consisting of hydrogen, perhalo(Cl-io)alkyl, amino, cyano, (Cl-lo)alkyl, (C3_12)cycloalkyl, hetero(C3-ia)cycloalkyl, aryl(Cl-lo)alkyl, heteroaryl (Cl-5)alkyl, (C9_l2)bicycloaryl, hetero(C8_12)bicycloaryl, carbonyl (Cl_3)alkyl, thiocarbonyl (Cl_3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, sulfonyl group, sulfinyl group, each substituted or unsubstituted, or R9 and Rlo are taken together to form a 5, 6, or 7 membered ring, each substituted or unsubstituted.
[0115] In yet another embodiment, DPP-IV inhibitors of the present invention comprise a compound of Formulae IXa, IXb, and IXc:

Rio . Rio IXa IXb IXc wherein n is 0, 1, 2, or 3;
Q is selected from the group consisting of CO, SO, SO2, and C=NRø;
each of T, U, V, W, and Y is independently nitrogen or CRl6, provided that no more than two of T, U, V, W, and Y are nitrogen;
RZ and R3 are each independently selected from the group consisting of hydrogen, halo, perhalo(Cl-lo)alkyl, amino, cyano, nitro, thio, (Cl-lo)alkyl, alkene, alkyne, (Cs-ia)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(Cl-lo)alkyl, heteroaryl (Ci-5)alkyl, (C9-ia)bicycloaryl, hetero(C$_12)bicycloaryl, carbonyl (Cl_3)alkyl, thiocarbonyl (Cl_3)alkyl, sulfonyl (Cl_3)alkyl, sulfinyl (Cl_3)alkyl, imino (Cl_3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;
R4 is selected from the group consisting of hydrogen, (Cl_lo)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, arylalkyl, heteroarylalkyl, bicycloaryl, heterobicycloaryl, each substituted or unsubstituted;
RS and R6 are each independently selected from the group consisting of hydrogen, a substituted or unsubstituted (Cl_lo)alkyl, a substituted or unsubstituted (Ci_lo)alkoxy, cyano, and halo, or where RS and R6 are taken together to form a ring;
each R16 is independently selected from the group consisting of halo, perhalo(Cl-lo)alkyl, CF3, (Cl_lo)alkyl, alkenyl, alkynyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, hydroxy, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted; and R9 and Rlo are each independently selected from the group consisting of hydrogen, perhalo(Cl-lo)alkyl, amino, cyano, (Cl-lo)alkyl, (C3_12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(Cl-lo)alkyl, heteroaryl (Cl-5)alkyl, (C9_12)bicycloaryl, hetero(C8_la)bicycloaryl, carbonyl (Cl_3)alkyl, thiocarbonyl (Cl_3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, sulfonyl group, sulfinyl group, each substituted or unsubstituted, or R9 and Rlo are taken together to form a 5, 6, or 7 membered ring, each substituted or unsubstituted.
[0116] In another embodiment, DPP-IV inhibitors of the present invention comprise a compound of Formula X:
R2 QwN~Ri (Ra)a X
wherein p is 0-10;
Q is selected from the group consisting of CO, SO, SO2, and C=NR4;
Rl is selected from the group consisting of (C1-lo)alkyl, (C3_12)cycloalkyl, (Cs-iz)cycloalkyl(C1_5)alkyl, hetero(C3-12)cycloalkyl, hetero(C3-ia)cycloalkyl(Cl_5)alkyl, aryl(Cl-lo)alkyl, heteroaryl(Cl-5)alkyl, (C9_12)bicycloaryl, (C9_la)bicycloaryl(Cl_5)alkyl, hetero(C8_12)bicycloaryl, hetero(C8_12)bicycloaryl(Cl-5)alkyl, carbonyl (Cl_3)alkyl, thiocarbonyl (Cl_3)alkyl, sulfonyl (C1_3)alkyl, sulfinyl (Cl_3)alkyl, imino (C1_3)alkyl, amino, cyano, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynYl, carbonyl group, inuno group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;
R2 and R3 are each independently selected from the group consisting of hydrogen, halo, perhalo(Cl-io)alkyl, amino, cyano, nitro, thio, (Cl-io)alkyl, alkene, alkyne, (C3-ia)cycloalkyl, hetero(C3-ia)cycloalkyl, aryl(Cl-lo)alkyl, heteroaryl (Cl-5)alkyl, (Cs-ia)bicycloaryl, hetero(C8_l~)bicycloaryl, carbonyl (C1_3)alkyl, thiocarbonyl (Cl_3)alkyl, sulfonyl (Cl_3)alkyl, sulfinyl (Cl_3)alkyl, imino (Cl_3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;

R4 is selected from the group consisting of hydrogen, (Cl_lo)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, arylalkyl, heteroarylalkyl, bicycloaryl, heterobicycloaryl, each substituted or unsubstituted; and each R8 is independently selected from the group consisting of halo, perhalo(Cl-lo)alkyl, CF3, (Cl_lo)alkyl, alkenyl, alkynyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, hydroxy, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
[0117] In another embodiment, DPP-IV inhibitors of the present invention comprise a compound of Formula XI:
Rs)P
XI
wherein p is 0-10;
Q is selected from the group consisting of CO, SO, 502, and C=NR~.;
R2 and R3 are each independently selected from the group consisting of hydrogen, halo, perhalo(Cl-lo)alkyl, amino, cyano, nitro, thio, (C1-lo)alkyl, alkene, alkyne, (C3_12)cycloalkyl, hetero(C3-ia)cycloalkyl, aryl(Cl-lo)alkYl, heteroaryl (Cl-5)alkyl, (C9-ia)bicycloaryl, hetero(C8_l2)bicycloaryl, carbonyl (Cl_3)alkyl, thiocarbonyl (Cl_3)alkyl, sulfonyl (Cl_3)alkyl, sulfinyl (Cl_3)alkyl, imino (C1_3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;
R4 is selected from the group consisting of hydrogen, (C1_lo)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, arylalkyl, heteroarylalkyl, bicycloaryl, heterobicycloaryl, each substituted or unsubstituted;
each Rg is independently selected from the group consisting of halo, perhalo(Cl-lo)alkyl, CF3, (Cl_lo)alkyl, alkenyl, alkynyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, hydroxy, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;
L is a linker providing 0-6 atom separation between X and the ring to which L
is attached; and X is selected from the group consisting of (Cl-lo)alkyl, (C3_12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(Cl-lo)alkyl, heteroaryl(Cl-5)alkyl, (C9_l2)bicycloaryl, hetero(C4_12)bicycloaryl, carbonyl (Cl_3)alkyl, thiocarbonyl (Cl_3)alkyl, sulfonyl (Cl_3)alkyl, sulfinyl (Cl_3)alkyl, imino (Cl_3)alkyl, amino, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, cyano, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
[0118] In another embodiment, DPP-IV inhibitors of the present invention comprise a compound of Formula XII:
Rs n ~N

~Ra)P
XII
wherein nis0, 1,2,or3;
p is 0-10;
Q is selected from the group consisting of CO, SO, 502, and C=NR4;
R2 and R3 are each independently selected from the group consisting of hydrogen, halo, perhalo(Cl-lo)alkyl, amino, cyano, nitro, thio, (Cl-lo)alkyl, alkene, alkyne, (C3_12)cycloalkyl, hetero(C3-i2)cYcloalkyl, aryl(Cl-lo)alkYl, heteroaryl (Cl-5)alkyl, (C9-12)bicycloaryl, hetero(C8_12)bicycloaryl, carbonyl (C1_3)alkyl, thiocarbonyl (Cl_3)alkyl, sulfonyl (Cl_3)alkyl, sulfinyl (Cl_3)alkyl, imino (Cl_3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;

R4 is selected from the group consisting of hydrogen, (Cl_lo)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, arylalkyl, heteroarylalkyl, bicycloaryl, heterobicycloaryl, each substituted or unsubstituted;
R5 and R6 are each independently selected from the group consisting of hydrogen, a substituted or unsubstituted (Cl_lo)alkyl, a substituted or unsubstituted (Cl_lo)alkoxy, cyano, and halo, or where RS and R6 are taken together to form a ring;
each R$ is independently selected from the group consisting of halo, perhalo(Cl-lo)alkyl, CF3, (Cl_lo)alkyl, alkenyl, alkynyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thin, cyano, nitro, hydroxy, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted; and X is selected from the group consisting of (Cl-io)alkyl, (C3_12)cycloalkyl, hetero(C3-iz)cycloalkyl, aryl(Cl-lo)alkyl, heteroaryl(C1-5)alkyl, (C9_12)bicycloaryl, hetero(C4_12)bicycloaryl, carbonyl (Cl_3)alkyl, thiocarbonyl (Cl_3)alkyl, sulfonyl (Cl_3)alkyl, sulfinyl (Cl_3)alkyl, imino (Cl_3)alkyl, amino, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, cyano, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
[0119] In yet another embodiment, DPP-1V inhibitors of the present invention comprise a compound of Formula XITI:
~m Re)p XIB
wherein m is 0, l, 2, 3, 4, or 5;
nis0, 1,2,or3;
p is 0-10;
Q is selected from the group consisting of CO, SO, SO2, and C=NR4;

R2 and R3 are each independently selected from the group consisting of hydrogen, halo, perhalo(Cl-lo)alkyl, amino, cyano, nitro, thio, (Cl-lo)alkyl, alkene, alkyne, (Cs-ia)cycloalkyl, hetero(C3-iz)cycloalkyl, aryl(Cl-lo)alkyl, heteroaryl (Cl-5)alkyl, (C9-iz)bicycloaryl, hetero(C8_12)bicycloaryl, carbonyl (Cl_3)alkyl, thiocarbonyl (Ci_3)alkyl, sulfonyl (Cl_3)alkyl, sulfinyl (Cl_3)alkyl, imino (C1_3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;
R~ is selected from the group consisting of hydrogen, (Cl_lo)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, arylalkyl, heteroarylalkyl, bicycloaryl, heterobicycloaryl, each substituted or unsubstituted;
R5 and R6 are each independently selected from the group consisting of hydrogen, a substituted or unsubstituted (Cl_lo)alkyl, a substituted or unsubstituted (Cl_lo)alkoxy, cyano, and halo, or where RS and R6 are taken together to form a ring;
each R~ is independently selected from the group consisting of halo, perhalo(Cl-io)alkyl, CF3, (Cl_lo)alkyl, alkenyl, alkynyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, hydroxy, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted; and each R8 is independently selected from the group consisting of halo, perhalo(Cl-lo)alkyl, CF3, (Cl_lo)alkyl, alkenyl, alkynyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thin, cyano, nitro, hydroxy, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
[0120] In yet another embodiment, DPP-IV inhibitors of the present invention comprise a compound of Formulae XIVa, XIVb and XIVc:

XIV a ~V b XIV c wherein nis0, 1,2,or3;
p is 0-10;
Q is selected from the group consisting of CO, SO, SOz, and C=NR4;
each of T, U, V, W, and Y is independently nitrogen or CRl6, provided that no more than two of T, U, V, W, and Y are nitrogen;
Rz and R3 are each independently selected from the group consisting of hydrogen, halo, perhalo(Cl-lo)alkYl, amino, cyano, nitro, thin, (Cl-lo)alkyl, alkene, alkyne, (Cs-lz)cycloalkyl, hetero(C3-iz)cycloalkyl, aryl(Cl-lo)alkyl, heteroaryl (Cl-5)alkyl, (C9_lz)bicycloaryl, hetero(C$_lz)bicycloaryl, carbonyl (C1_3)alkyl, thiocarbonyl (Cl_3)alkyl, sulfonyl (Cl_3)alkyl, sulfinyl (Cl_3)alkyl, imino (Cl_3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;
R4 is selected from the group consisting of hydrogen, (Cl_lo)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, arylalkyl, heteroarylalkyl, bicycloaryl, heterobicycloaryl, each substituted or unsubstituted;
RS and R6 are each independently selected from the group consisting of hydrogen, a substituted or unsubstituted (Cl_lo)alkyl, a substituted or unsubstituted (Cl_lo)alkoxy, cyano, and halo, or where RS and R6 are taken together to form a ring;
each R8 is independently selected from the group consisting of halo, perhalo(C1-lo)alkyl, CF3, (Cl_lo)alkyl, alkenyl, alkynyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, hydroxy, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted; and ~p ~1~;; a .. I t ~'r t~;;:l~
P -SOt~l~,,.~'E~;~.,.,i~.".~~ 1't;,., i,;,.i~,i;.;. ~1;..~
f._t rt.,, ' s rode endently selected from the group consisting of halo, each Ri6 1 p CF Ci-lo)~kYh alkenyl, alkynYl, ~Yl, heteroaryl, aminosulfonyl, ~erhalo(Cl-lo)~kYl~ s' ( lsulfon 1, aryloxy, heteroaryloxy, arylalkyl, ~lkylsulfonyl, arylsulfonyl, heteroary y cloalkyl, heterocycloalkyl, a~no, thio, cyano, nitro, hydroxy, alkoxy, heteroarylalkyl, cy 'ne oup, sulfonyl group and sulfinyl group, each substituted or carbonyl group, 1~-unsubstituted.
other embodiment, DPP-1V inhibitors of the present invention comprise a [0121] In Yet an compound of Formula XV:
R2 Q\ ~L~X
N
M-wRis XV
wherein is selected from the group consisting of CO, SO, SOa' and C=NR4;
roviding 1-6 atom separation between R19 and the ring to which M is M is a moiety p attached;
are each independently selected from the group consisting of hydrogen, RZ and R3 thio, (Ci-io)alkyl, alkene, alkyne, h~o, perhalo(Crio)~kyh a~n°' cyano, nitro, tero C3-ia)cYcloalkyl, aryl(Cl-lo)~kYl~ heteroaryl (Cms)~kyl' (C3_12)cycloalkyl, he ( ro Cg_iz)bicycloaryl' carbonyl (Cl_3)alkyl, thiocarbonyl (Ci_3)~kyl, (C9_12)bicycloaryl, hete sulfin 1 (Cl_s)alkYl, imino (Ci-3)alkyl, aryl' heteroaryl, hydroxy, alkoxy, sulfonyl (Cl_3)alkYl, Y
alken 1, alkynyh carbonyl group, imine group' sulfonyl group and aryloxy, heteroaryloxy, Y
sulfinyl group, each substituted or unsubstituted;
from the group consisting of hydrogen, (Cl-io)~kyh cycloalkyl, Ra is selected heteroaryl, arylalkyl, heteroaryl~kyl, bicycloaryl, heterobicycloaryl, heterocycloalkyl, aryl' each substituted or unsubstituted;
a basic nitrogen atom that is capable of interacting with a carboxylic R19 comprises acid side chain of an active site residue of a protein;
roviding 0-6 atom separation between X and the ring to which L is L is a linker p attached; and X is selected from the group consisting of (Cl-io)alkyl, (C3_i2)cycloalkyl, hetero(C3-i2)cycloalkyl, aryl(C1-lo)alkyl, heteroaryl(Ci-5)alkyl, (C9_12)bicycloaryl, hetero(C4_i2)bicycloaryl, carbonyl (Cl_3)alkyl, thiocarbonyl (Cl_3)alkyl, sulfonyl (Cl_3)alkyl, sulfinyl (Cl_3)alkyl, imino (Cl_3)alkyl, amino, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, cyano, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
[0122] In one variation of the above embodiment, L provides 0,1, 2, or 3 atom separation between X and the ring. In another variation of the above embodiment, L
provides 0 atom separation between X and the ring. In yet another variation of the above embodiment, X is selected from the group consisting of a substituted or unsubstituted aryl or heteroaryl.
[0123] Synthetic intermediates that may be used to synthesize DPP-IV
inhibitors of the present invention are also provided.
[0124] In one embodiment, the synthetic intermediates comprise Formula XVI:
R2 Q~ /L-X
I N
R3 N Z' XVI
wherein Q is selected from the group consisting of CO, SO, 502, and C=NR~;
Z' is a leaving group;
R2 and R3 are each independently selected from the group consisting of hydrogen, halo, perhalo(Cl-io)alkyl, amino, cyano, nitro, thio, (Cl-io)alkyl, alkene, alkyne, (C3_12)cycloalkyl, hetero(C3-i2)cycloalkyl, aryl(Cl-io)alkyl, heteroaryl (C1-5)alkyl, (C9_12)bicycloaryl, hetero(C8_12)bicycloaryl, carbonyl (Cl_~)alkyl, thiocarbonyl (Cl_3)alkyl, sulfonyl (Cl_3)alkyl, sulfinyl (Cl_3)alkyl, imino (Cl_3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;
R4 is selected from the group consisting of hydrogen, (Cl_lo)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, arylalkyl, heteroarylalkyl, bicycloaryl, heterobicycloaryl, each substituted or unsubstituted;

L is a linker providing 0-6 atom separation between X and the ring to which L
is attached; and X is selected from the group consisting of cycloalkyl, aryl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
[0125] In one variation, Z' is not alkyl, alkylthio, NNH, NHa, H, HZN-C(O)-NH, cyano, alkoxy, or 2-trimethylsilyl-1-oxazolidinone (TMSO) when -LX is benzyl. In another variation, Z' is not halo when -LX is alkyl.
[0126] According to this embodiment, Z' may optionally be selected from the group consisting of halo, nitro, azido, phenylsulfoxido, aryloxy, alkoxy, alkylsulfonate, and arylsulfonate.
[0127] Also according to this embodiment, Z' may optionally be selected from the group consisting of halo, thio, hydroxyl, p-tolylsulphonyloxy and methylsulphonyloxy.
[0128] In another embodiment, the synthetic intermediates comprise Formula XVII:
R2 Qw NH

XVII
wherein Q is selected from the group consisting of CO, SO, SO~,, and C=NR4;
Z' is a leaving group;
R2 and R3 are each independently selected from the group consisting of hydrogen, halo, perhalo(Cl-io)alkyl, amino, cyano, nitro, thio, (Cl-lo)alkyl, alkene, alkyne, (Cs-ia)cycloalkyl, hetero(C3-1~)cycloalkyl, aryl(Cl-io)alkyl, heteroaryl (Ci-5)alkyl, (C9_la)bicycloaryl, hetero(C8_12)bicycloaryl, carbonyl (Cl_3)alkyl, thiocarbonyl (Cl_3)alkyl, sulfonyl (Cl_3)alkyl, sulfinyl (Ci_3)alkyl, imino (Cl_3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;
R4 is selected from the group consisting of hydrogen, (Cl_lo)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, arylalkyl, heteroarylalkyl, bicycloaryl, heterobicycloaryl, each substituted or unsubstituted;
L is a linker providing 0-6 atom separation between X and the ring to which L
is attached; and X is selected from the group consisting of (Cl-lo)alkyl, (C3_12)cycloalkyl, hetero(C3-iz)cycloalkyl, aryl(Cl-lo)alkyl, heteroaryl(Cl-5)alkyl, (C9_12)bicycloaryl, hetero(C4_12)bicycloaryl, carbonyl (Cl_3)alkyl, thiocarbonyl (Cl_3)alkyl, sulfonyl (Ci_3)alkyl, sulfinyl (C1_3)alkyl, imino (Cl_3)alkyl, amino, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, cyano, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
SUBSTITUENT L:
[0129] In one variation, the present invention comprises compounds where L is a linker providing 0, 1, 2, or 3 atom separation between X and the ring to which L is attached. In another variation, L provides 0 atom separation between X and the ring. In yet another variation, L provides 0, 1, 2, or 3 atom separation between X and the ring. In still another variation, the present invention comprises compounds where L is a linker providing 1, 2, or 3 atom separation between X and the ring to which L is attached. The atoms of the linker providing the separation may optionally be selected from the group consisting of carbon, oxygen, nitrogen, and sulfur. ~ptionally according to this variation, the 1, 2, or 3 atoms of L
providing the separation comprise and optionally consist of carbon atoms. The 1, 2, or 3 atoms of L providing the separation may also be selected from a group of linkers consisting of at least one oxygen, or at least one nitrogen atom.
[0130] In another variation, the present invention comprises compounds where L
provides 1 atom separation between X and the ring. The 1 atom separation may optionally be provided by a carbon, oxygen, nitrogen, or sulfur atom. In another variation, L is a carbonyl.
[0131] In another variation, the present invention comprises compounds where L
is a linker providing 0-6 atom separation between X and the ring to which L is attached, with the proviso that L is not -NH- or -N=CH-.
[0132] In yet another variation, the present invention comprises compounds where L is selected from the group consisting of (Cl-lo)alkyl, (C3_12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(Cl-lo)alkyl, heteroaryl(Cl-5)alkyl, (C9_1a)bicycloaryl, hetero(C~_12)bicycloaryl, carbonyl (Cl_3)alkyl, thiocarbonyl (Cl_3)alkyl, sulfonyl (Cl_3)alkyl, sul~nyl (Cl_3)alkyl, imino (Cl_3)alkyl, amino, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, cyano, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
[0133) Tn yet another variation, the present invention comprises compounds where L is selected from the group consisting of -CH2-, -CHRIa-, -C(R12)(R12>)-, -C(O)-, -C(S)-, -C(NH)-, -C(NR12)-, -O-, -N(H)-, -N(R12)-, and -S- where R12 and R12> are each independently selected from the group consisting of hydrogen, perhalo(Cl-lo)alkyl, amino, cyano, (Cl-io)~kyl, (C3_12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(Cl-lo)alkyl, heteroaryl (C1-5)alkyl, (C9_la)bicycloaryl, hetero(C8_12)bicycloaryl, carbonyl (Cl_3)alkyl, thiocarbonyl (Cl_3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alk5myl, carbonyl group, sulfonyl group, sulfinyl group, each substituted or unsubstituted or where or R12 and R12~ are taken together to form a 5, 6, or 7 membered ring, each substituted or unsubstituted.
[0134] In yet another variation, the present invention comprises compounds where L is selected from the group consisting of -CH2-, -CH2CH2-, -CH2CHZCH2-, -C(O)-, -CH2C(O)-, -C(O)CH2-, -CHZ-C(O)CH2-, -C(O)CH2CH2-, -CHZCH2C(O)-, -O-, -OCH2-, -CH20-, -CH20CH2-, -OCH2CH2-, -CH2CH20-, -N(CH3)-, -NHCHZ-, -CHZNH-, -CHZNHCH2-, -NHCHZCH2-, -CH2CHZNH-, -NH-C(O)-, -NCH3-C(O)-, -C(O)NH-, -C(O)NCHS-, -NHC(O)CHZ-, -C(O)NHCH2-, -C(O)CH2NH-, -CH2NHC(O)-, -CH2C(O)NH-, -NHCH2C(O)-, -S-, -SCH2-, -CH2S-, -SCH2CH2-, -CH~SCHZ-, -CH2CHaS-, -C(O)S-, -C(O)SCH2-, -CHZC(O)S-, -C(O)CH2S-, and -CH2SC(O)-. It is noted that one or more of the hydrogens shown may be substituted with other non-hydrogen substituents.
[0135] In another variation, the present invention comprises compounds where L
is selected from the group consisting of -CH2-, -C(O)-, -CHZC(O)-, -C(O)CH2-, -CHZ-C(O)CH2-, -C(O)CH2CH2-, and -CH2CHaC(O)-. In the described definitions of the linker L, the 0-6 atom separation refers to the direct linear linkage between the ring and X. As such, branching along the 0-6 atom separation is permitted as is clear from the variations described.
SUBSTITUENT X:
[0136] In one variation, the present invention comprises compounds where X is selected from the group consisting of (C1-lo)alkyl, (C3_12)cycloalkyl, hetero(C3-la)cycloalkyl, aryl(Cl-lo)alkyl, heteroaryl(Cl-5)alkyl, (C9_12)bicycloaryl, hetero(C4_12)bicycloaryl, carbonyl (Cl_3)alkyl, thiocarbonyl (Cl_3)alkyl, sulfonyl (Cl_3)alkyl, sulfinyl (C1_3)alkyl, imino (Ci_3)alkyl, amino, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, cyano, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
[0137] In yet another variation, the present invention comprises compounds where X is selected from the group consisting of cycloalkyl, aryl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
[0138] In another variation, the present invention comprises compounds where X
is a substituted or unsubstituted cycloalkyl or heterocycloalkyl. For example, X
may be a substituted or unsubstituted (C3_~)cycloalkyl or a substituted or unsubstituted (C3_~)heterocycloalkyl.
[0139) In another variation, the present invention comprises compounds where X
is a substituted or unsubstituted aryl or heteroaryl. For example, X may be a substituted or unsubstituted phenyl.
[0140] In yet another variation, the present invention comprises compounds where X is a ring having a non-hydrogen substituent at a 2 or 3 position of the ring.
[0141] In another variation, the present invention comprises compounds where X
has a non-hydrogen substituent at a 2 or 3 position of the ring where the non-hydrogen substituent is selected from the group consisting of (Cl-lo)alkyl, (C3_12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(Cl-lo)alkyl, heteroaryl(Cl-5)alkyl, (C9_12)bicycloaryl, hetero(C$_12)bicycloaryl, carbonyl (Cl_3)alkyl, thiocarbonyl (Cl_3)alkyl, sulfonyl (Cl_3)alkyl, sulfinyl (Cl_3)alkyl, imino (Cl_3)alkyl, amino, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, cyano, nitro, halo, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
[0142] In particular variations, X 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)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-CONHa)phenyl, (2-CONH(Cl-~)alkyl)phenyl, (3-CONH(Cl-~)alkyl)phenyl, (2-CO2(Cl-~)alkyl)phenyl, (3-CO~(Cl-~)alkyl)phenyl, -NH2, -OH, -(C3-)alkyl, -alkene, -alkyne, -CCH, -(C3-~)cycloalkyl, and -aryl, each substituted or unsubstituted.
[0143] In yet another variation, the present invention comprises compounds where X is selected from the group consisting of B ~ \A
B g B
wherein A is S, O, or NR21;
B is CRZ~, or N;
R2o is independently selected from the group consisting of hydrogen, halo, perhalo(Cl-1o)alkyl, amino, thin, cyano, CF3, vitro, (Cl-lo)alkyl, (C3_ia)cycloalkyl, hetero(C3-ia)cycloalkyl, aryl(Cl-lo)alkyl, heteroaryl (Cl-5)alkyl, (C9_t2)bicycloaryl, hetero(Cg_12)bicycloaryl, carbonyl (Cl_3)alkyl, thiocarbonyl (C1_3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy; heteroaryloxy, imino group, carbonyl group, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, sulfinyl group; and R21 is independently selected from the group consisting of hydrogen, perhalo(Cl-io)alkyl, amino, cyano, (Cl-lo)alkyl, (C3_l2)cycloalkyl, hetero(C3-ia)cycloalkyl, aryl(Ctu o)alkyl, heteroaryl (C1-S)alkyl, (C9_12)bicycloaryl, hetero(C8_12)bicycloaryl, carbonyl (Cl_3)alkyl, thiocarbonyl (Cl_3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, imino group, carbonyl group, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, sulfinyl group.
[0144] In yet another variation, the present invention comprises compounds where X is selected from the group consisting of N- -N
~ \ I/
\ I / \ I / \ I ~N
(R~)t (R~)t (R~)t (R~)t wherein t is 0, 1, 2, or 3; and each R~ is independently selected from the group consisting of halo, perhalo(Cl-lo)alkyl, CF3, (Cl_lo)alkyl, alkenyl, alkynyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, hydroxy, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
[0145] In yet another variation, the present invention comprises compounds where X is selected from the group consisting of - a s cRot S\
cRot wherein t is 0, 1, 2, or 3; and each R~ is independently selected from the group consisting of halo, perhalo(Cl-io)alkyl, CF3, (Cl_lo)alkyl, alkenyl, alkynyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, hydroxy, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
[0146] According to each of the above variations, R~ may independently be selected from the group consisting of -cyano, -methoxy, -nitro, -carboxy, -sulfonamido, -tetrazolyl, -aminomethyl, -hydroxymethyl, -phenyl, -halo, -CONH2, -CONH(C1-)alkyl, -COZ(Cl-)alkyl, -NHS, -OH, -(Cl-5)alkyl, -alkenyl, -alkynyl, -CCH, (Cl-5)cycloalkyl, aryl, heteroaryl, and heterocycloalkyl, each substituted or unsubstituted.
[0147] In yet another variation, the present invention comprises compounds where -LX is -ORl l, where Rll is selected from the group consisting of substituted or unsubstituted alkyl, cycloalkyl, aryl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl.
[0148] In yet another variation, the present invention comprises compounds where -LX is selected from the group consisting of -(CH2)-(2-cyano)phenyl, -(CHZ)-(3-cyano)phenyl, -(CHa)-(2-hydroxy)phenyl, -(CHZ)-(3-hydroxy)phenyl, -(CHZ)-(2-alkenyl)phenyl, -(CHZ)-(3-alkenyl)phenyl, -(CH2)-(2-alkynyl)phenyl, -(CH2)-(3-all~myl)phenyl, -(CH2)-(2-nitro)phenyl, -(CHZ)-(3-nitro)phenyl, -(CH2)-(2-carboxy)phenyl, -(CH2)-(3-carboxy)phenyl, -(CHZ)-(2-carboxamido)phenyl, -(CH2)-(3-carboxamido)phenyl, -(CH2)-(2-sulfonamido)phenyl, -(CHZ)-(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, -(CHZ)-(2-hydroxymethyl)phenyl, -(CH2)-(3-hydroxymethyl)phenyl, -(CHZ)-(2,-phenyl)phenyl, -(CH2)-(3-phenyl)phenyl, -(CH2)-(2-CONHZ)phenyl, -(CH2)-(3-CONH2)phenyl, -(CHZ)-(2-CONH(Cl-~)alkyl)phenyl, -(CHa)-(3-CONH(Cl-~)alkyl)phenyl, -(CH2)-(2-C02(Cl-~)alkyl)phenyl, -(CHI,)-(3-C02(C1-~)alkyl)phenyl, -CH2-NH2, -CHZ-OH, -CH2-(C3-)alkyl, -CH2-alkene, -CH2-alkyne, -CH2-CCH, -CH2-(C3-~)cycloalkyl, and -CHZ-aryl, each substituted or unsubstituted.
[0149] In yet another variation, the present invention comprises compounds where -LX is selected from the group consisting of -(Cl)alkyl-aryl, -(Cl)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, -S02-heterobicycloaryl, -SOZ-aryl, -S02-heteroaryl, -SOZ-bicycloaryl, -S02-heterobicycloaryl, -C(NR13)-aryl, -C(NRl3)-heteroaryl, -C(NRIS)-bicycloaryl, -C(NR13)-heterobicycloaryl, each substituted or unsubstituted where R13 is selected from the group consisting of hydrogen, perhalo(Cl-lo)alkyl, amino, cyano, (Cl-lo)alkyl, (C3_12)cycloalkyl, hetero(C3-ia)cycloalkyl, aryl(Cl-lo)alkyl, heteroaryl (Cl-5)alkyl, (C9_12)bicycloaryl, hetero(C8_12)bicycloaryl, carbonyl (Cl_3)alkyl, thiocarbonyl (C1_3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, sulfonyl group, sulfinyl group, each substituted or unsubstituted.
SUBSTITUENT Z:
[0150] In one variation, the present invention comprises compounds where Z is selected from the group consisting of halo, perhalo(Cl-lo)alkyl, amino, cyano, thin, (Cl-lo)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, carbonyl (Cl_3)alkyl, thiocarbonyl (Cl_3)alkyl, sulfonyl (Cl_3)alkyl, sulfinyl (Cl_3)alkyl, imino (Cl_3)alkyl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
[0151] In another variation, Z is selected from the group consisting of (C3_l2)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(Cl-io)alkyl, heteroaryl (Cl-5)alkyl, (C9_12)bicycloaryl, and hetero(C8_12)bicycloaryl, each substituted or unsubstituted.
[0152] In another variation, the present invention comprises compounds where Z
is selected from the group consisting of cycloalkyl, heterocycloalkyl, aryl and heteroaryl, each substituted or unsubstituted. For example, in a particular variation, Z is a substituted or unsubstituted 3, 4, 5, 6, or 7 membered ring. In one variation, Z is a substituted or unsubstituted 3, 4, 5, 6, or 7 membered cycloalkyl. In another variation, Z is a substituted or unsubstituted 4, 5, 6, or 7 membered heterocycloalkyl. In another variation, Z
is a substituted or unsubstituted aryl. In a particular variation, Z is a substituted or unsubstituted phenyl. In another variation, Z is a substituted or unsubstituted heteroaryl.
[0153] In another variation, Z comprises a substituted or unsubstituted (C3-~)cycloalkyl ring. Z may also optionally comprise O, N(O), N, S, SO, 502, or a carbonyl group in the ring.
[0154] In another variation, Z is -NR9Rlo where R9 and Rlo together are -(CH2)~6, with or without being interrupted by one O, S, SO, 502, carbonyl group, N(O), NH, N-(aryl), N-(aryl(Cl-5)alkyl), N-(carboxy(Cl-5)alkyl), or N-(Cl_2)alkyl) group, each substituted or unsubstituted.
[0155] In yet another variation, Z is -NR9Rlo, where R9 is selected from the group consisting of (C3_12)cycloalkyl, hetero(C3_12)cycloalkyl, (C6_I2)aryl, hetero(CS_12)aryl, (C9-i2)bicycloaryl, each substituted or unsubstituted and hetero(C8_12)bicycloaryl, and Rlo is selected from the group consisting of hydrogen and a substituted or unsubstituted (Cl_8)alkyl.
[0156] In another variation, Z is -NR9Rlo, where R9 and Rlo are taken together to form a substituted or unsubstituted 5, 6, or 7 membered heterocycloalkyl, heteroaryl, or heterobicycloaryl. In another particular variation, R9 and Rlo are taken together to form a pyrrole, pyrazole, triazole, imidazole, benzimidazole, indole, isoindole, piperidine, piperazine, pyrrolidine, homopiperazine, azetidine, and hexahydroazepine, each substituted or unsubstituted.
[0157] In another variation, Z is selected from the group consisting of -N~ -~_N -~_N~\ -~_N
~S R ~~ ~\(R8) ( 8)p (R8)p p (R8)p wherein p is 0-12 and each Rg is independently selected from the group consisting of halo, perhalo(Cl-lo)alkyl, CF3, cyano, nitro, hydroxy, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroat~yloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thin, alkoxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
[0158] In another variation, Z is selected from the group consisting of:
-~~-( R8)r (R8)r ~ (R8)r (R8)r (R8)r wherein r is 0-13 and each R$ is independently selected from the group consisting of halo, perhalo(Cl-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 substituted or unsubstituted.
[0159] In another variation, Z is selected from the group consisting of (C9_12)bicycloaryl and hetero(C4_12)bicycloaryl, each substituted or unsubstituted.
[0160] In another variation, Z 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.
[0161] In another variation, Z is selected from the group consisting of 3-amino-piperidinyl-1-yl, 3-aminomethyl-pyrrolidin-1-yl, 2-aminoazetidin-1-yl, aminoazetidin-1-yl, 3-amino-3-methylpiperidin-1-yl, 3-aminocyclopent-1-yl, 3-aminomethylcyclopent-1-yl, 3-aminomethylcyclohex-1-yl, 3-aminohexahydroazepin-1-yl, 3-amino-cyclohex-1-yl, piperazin-1-yl, homopiperazin-1-yl, 3-amino-pyrrolidin-1-yl, and R-3-aminopiperidin-1-yl, each substituted or unsubstituted.
[0162] In another variation, Z is selected from the group consisting of halo, thio, and hydroxyl.
SUBSTITUENT Rl:
[0163] In one variation Rl is selected from the group consisting of (C1-lo)alkyl, (C3-ia)cycloalkyl, (C3_l2)cycloalkyl(Cl_5)alkyl, hetero(C3-la)cycloalkyl, hetero(C3-12)cycloalkyl(Cl_5)alkyl, aryl(Cl-lo)alkyl, heteroaryl(Cl-5)alkyl, (C9_12)bicycloaryl, (C9_l~)bicycloaryl(Cl_5)alkyl, hetero(Cg_12)bicycloaryl, hetero(Cg_12)bicycloaryl(Cl-5)alkyl, carbonyl (Cl_3)alkyl, thiocarbonyl (Ci_3)alkyl, sulfonyl (Cl_3)alkyl, sulfinyl (Cl_3)alkyl, imino (Cl_3)alkyl, amino, cyano, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
[0164] In another variation, Rl is selected from the group consisting of:
A~ g g ~ \A
B B
wherein A is S, O, or NR21;
B is CRZO or N;
R2o is independently selected from the group consisting of hydrogen, halo, perhalo(Cl-io)alkyl, amino, thio, cyano, CF3, nitro, (C1-lo)alkYl, (C3_la)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(Cl-lo)alkyl, heteroaryl (Cl-5)alkyl, (C~_12)bicycloaryl, hetero(C8_12)bicycloaryl, carbonyl (C1_3)alkyl, thiocarbonyl (Cl_3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, imino group, carbonyl group, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, sulfinyl group; and R21 is independently selected from the group consisting of hydrogen, perhalo(C1-lo)alkyl, amino, cyano, (C1-lo)alkyl, (C3_12)cycloalkyl, hetero(C3-ia)cycloalkyl, aryl(C1-lo)alkYl, heteroaryl (Cl-5)alkyl, (C9_la)bicycloaryl, hetero(C8_12)bicycloaryl, carbonyl (Cl_3)alkyl, thiocarbonyl (Cl_3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, imino group, carbonyl group, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, sulfinyl group.
[0165] In another variation, Rl is selected from the group consisting of:
N- -N
I ~ ~ ~ I ~ ~ I ~ ''Lt," ~ I ~ N
(R~)t (R~)t (R~)t (R~)t wherein t is 0, l, 2, or 3; and each R~ is independently selected from the group consisting of halo, perhalo(Cl-lo)alkyl, CF3, (Cl_lo)alkyl, alkenyl, alkynyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, hydroxy, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
[0166] In another variation, Rl is selected from the group consisting of:
~ ~S ~R~)t S\
cR~)t wherein t is 0, 1, 2, or 3; and each R~ is independently selected from the group consisting of halo, perhalo(Cl-lo)alkyl, CF3, (Cl_lo)alkyl, alkenyl, alkynyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, hydroxy, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
SUBSTITUENTS R5 AND R6:
[0167] In one variation, RS and R6 are each independently selected from the group consisting of hydrogen, a substituted or unsubstituted (Cl_lo)alkyl, a substituted or unsubstituted (Cl_lo)alkoxy, cyano, and halo, or where R5 and R6 are taken together to form a ring.
[0168] In one variation, RS and R6 are taken together to form a ring.
[0169] In another variation, at least one of RS and R6 is a substituted or unsubstituted -(Cl_s)alkyleneRl~, wherein Rl~ is selected from the group consisting of (C3_12)cycloalkyl, hetero(C4_i2)cycloalkyl, (C6_12)aryl, heter0(CS_12)aryl, (C9-ia)bicycloalkyl, hetero(C9_ia)bicycloalkyl, (C9_12)bicycloaryl and hetero(Cs_12)bicycloaryl, each substituted or unsubstituted.
[0170] In another variation, at least one of RS and R6 is optionally substituted by one to three radicals selected from the group consisting of (Cl_6)alkyl, cyano, halo, vitro, halo-substituted(Cl_6)alkyl, -NRlsRls, -NRisC(O)ORIS, -NRISC(O)NRlsRls, -NRisC(NRis)NRisRis~ -ORIS~ -SRis~ -C(O)ORlBa -C(O)NRlsRis~ -S(O)aNRisRis~
-P(O)(ORis)ORis~ -OP(O)(ORis)ORis~ -NR18C(O)Risa -S(O)Ris~ -S(O)aRis~
-(Cl_8)alkyleneC(O)Ris, -(Cl_s)alkyleneNRlsRlsa -(Cl_s)alkyleneNRlsC(O)ORIS, -(Cl_s)alkyleneNRlBC(O)NRlBRIS, -(Cl_s)alkyleneNRlsC(NRIS)NRlsRls, -(Cl_s)alkYleneORls, -(Cl_s)alkyleneSRls, -(Cl_s)alkyleneC(O)ORlB, -(Cl_$)alkyleneC(O)NRlsRlsa -(C1_s)alkyleneS(O)2NR1sR18, -(Cl_8)alkyleneP(O)(ORIS)ORlB, -(C1_ s)alkyleneOP(O)(ORIS)ORlB, -(Cl_s)alkyleneNRlsC(O)Rls, -(Ci-s)alkyleneS(O)Rlg, -(Ci-s)alkyleneS(O)2Rls and -(Cl_s)alkyleneC(O)Rls where each Rls substituent is independently selected from the group consisting of hydrogen and a substituted or unsubstituted (Ci_s)alkyl.
[0171] According to each of the above variations, at least one of R5 and R6 is optionally substituted by one to three radicals selected from the group consisting of -NH2, -NHC(NH)NH2, -OH, -SH, -C(O)OH and -C(O)NH2.
[0172] In one variation, the present invention comprises compounds where RS
and R6 are hydrogen. In another variation, the present invention comprises compounds where RS and R6 are hydrogen and n is 1 or 2.
[0173] In still another variation, RS and R6 are hydrogen and each R~ is independently selected from the group consisting of halo, perhalo(Cl-lo)alkyl, CF3, (Cl_lo)alkyl, alkenyl, alkynyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, vitro, hydroxy, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted. In yet another variation, RS and R6 are hydrogen and two R~
are taken together to form a substituted or unsubstituted, fused or bridged ring.
[0174] In another variation, the present invention comprises compounds where RS and R6 are hydrogen and two R~ are taken together to form a substituted or unsubstituted fused or bridged ring. In yet another variation, two R~ are taken together to form a substituted or unsubstituted fused ring. In still another variation, two R~ are taken together to form a substituted or unsubstituted bridged ring.
[0175] In yet another variation, the present invention comprises compounds where RS and R6 are hydrogen, n is 1, and R~ is 2-cyano. In still another variation, the present invention comprises compounds where RS and R6 are hydrogen and R~ is 2-cyano.
[0176] In yet another variation, the present invention comprises compounds where n is 1, 2, or 3; RS and R6 are hydrogen; and each R~ is independently selected from the group consisting of halo, perhalo(Cl-lo)alkyl, CF3, heteroaryl, aryloxy, heteroaryloxy, cyano, nitro, hydroxy, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
[0177] In another variation, the present invention comprises compounds where n is 1 or 2; Z is selected from the group consisting of alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, imino group, sulfonyl group and sulfinyl group, and a substituted or unsubstituted 4, 5, 6, or 7 membered ring; and R5 and R6 are hydrogen.
SUBSTITITENT Q:
[0178] In one variation, Q is selected from the group consisting of CO, SO, S02 and C=NRø, where R~ is selected from the group consisting of hydrogen, (Cl_lo)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, arylalkyl, heteroarylalkyl, bicycloaryl, heterobicycloaryl, each substituted or unsubstituted. In another variation, Q is selected from the group consisting of CO, SO and 502. In yet another variation, Q is selected from the group consisting of CO
and 502. In still another variation, Q is C=NR4, where R4 is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted SUBSTITUENTS R~ AND R~:
[0179] In one variation, R2 and R3 are each independently selected from the group consisting of hydrogen, halo, perhalo(Cl-lo)alkyl, amino, cyano, nitro, thin, (Cl-lo)alkyl, alkene, alkyne, (C3_12)cycloalkyl, hetero(C3-i2)cycloalkyl, aryl(Cl-lo)alkyl, heteroaryl (Cl-5)alkyl, (C9_12)bicycloaryl, hetero(C8_12)bicycloaryl, carbonyl (Cl_3)alkyl, thiocarbonyl (C1_ 3)alkyl, sulfonyl (Cl_3)alkyl, sulfinyl (Cl_3)alkyl, imino (Cl_3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted. In another variation, R2 andlor R3 is hydrogen. In another variation, R2 and/or R3 is selected from the group consisting of -NH2, -NHCH3, -N(CH3)2, -NHCI_3-alkyl, and -N(Cl_3alkyl)2, each substituted or unsubstituted. In yet another variation, R2 and/or R3 is selected from the group consisting of hydrogen, halo, perhalo(Cl-lo)alkyl, amino, cyano, vitro, thio, (Cl-io)alkyl, (C3_iz)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(C1-lo)alkyl, heteroaryl (Cl-S)alkyl, (C9_12)bicycloaryl, hetero(C$_12)bicycloaryl, carbonyl (C1_3)alkyl, thiocarbonyl (Cl_3)alkyl, sulfonyl (Cl_3)alkyl, sulfinyl (Cl_3)alkyl, imino (Cl_3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
[0180] In another particular variation, R2 and/or R3 is selected from the group consisting of a (C2-lo)alkenyl; an amino, (Cl_lo)alkyl, or alkoxy carbonyl (Ci_3)alkenyl;
an anuno, (C1_ lo)alkY1 ,or alkoxy thiocarbonyl (Cl_3)alkenyl; an amino, (C1_lo)alkyl, or alkoxy sulfonyl (Cl_3)alkenyl; an amino, (Cl_lo)alkyl, or alkoxy sulfinyl (Cl_3)alkenyl; an amino, (Cl_lo)alkyl, or alkoxy imino (Cl_3)alkenyl; an aryl (Cl-5)alkenyl; and a heteroaryl (C1-5) alkenyl, each substituted or unsubstituted. In yet another particular variation, R2 and/or R3 is selected from the group consisting of a (C2-lo)alkynyl; an amino, (Cl_lo)alkyl, or alkoxy carbonyl (Cl_3)alkynyl; an amino, (Cl_lo)alkyl, or alkoxy thiocarbonyl (Cl_3)alkynyl;
an amino, (C1_ io)alkyl, or alkoxy sulfonyl (Cl_3)alkynyl; an amino, (Cl_lo)alkyl, or alkoxy sulfinyl (C1_ 3)alkynyl; an amino, (Cl_lo)alkyl, or alkoxy imino (Cl_3)alkynyl; an aryl (C1-5) alkynyl; and heteroaryl (Cl-5)alkynyl, each substituted or unsubstituted. In another variation, Ra and/or R3 is substituted or unsubstituted (C3_~)cycloalkyl. In yet another variation, RZ
and/or R3 is substituted or unsubstituted (C3_~)heterocycloalkyl.
[0181] In another particular variation, R2 and/or R3 is substituted or unsubstituted aryl. In yet another variation, R2 and/or R3 is substituted or unsubstituted phenyl. In another variation, R2 and/or R3 is selected from the group consisting of 2-fluorophenyl, 4-fluorostyryl, 2-methoxyphenyl, pyrrolidin-1-yl, imidazolyl, and 3-furanyl, each substituted or unsubstituted.
[0182] In another variation, R2 and/or R3 is substituted or unsubstituted heteroaryl. In yet another variation, R2 and/or R3 is selected from the group consisting of hydrogen, cyano, -CF3, or hydroxy. In another variation, R2 and/or R3 is selected from the group consisting of I, Br, Cl, and F. In yet another particular variation, R2 and/or R3 is -OR13 where R13 is selected from the group consisting of alkyl, cycloalkyl, aryl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
[0183] In one variation, R2 and/or R3 is a carbonyl group. In another variation, R2 andlor R3 is selected from the group consisting of an aldehyde, acid, amide, and ester. In another variation, RZ and/or R3 is selected from the group consisting of pyrrolidin-1-ylcarbonyl, piperidin-1-ylcarbonyl, and morpholin-4-ylcarbonyl, each substituted or unsubstituted. In yet another variation, RZ and/or R3 is selected from the group consisting of -SH, -SCH3, and -S(CI_3)alkyl, each substituted or unsubstituted. In yet another variation, RZ
and/or R3 is -NRl4Ris where R14 is selected from the group consisting of (C3_12)cycloalkyl, hetero(C3_iz)cycloalkyl, (C6_12)aryl, hetero(Cs_12)aryl, (C9_12)bicycloaryl and hetero(C8_12)bicycloaryl, each substituted or unsubstituted, and Rls is selected from the group consisting of hydrogen and a substituted or unsubstituted (Cl_8)alkyl, and where R14 and Rls together are -(CH2)a-s- optionally interrupted by one O, S, NH, or -N(C1_3)alkyl group, unsubstituted or substituted.
[0184] In another variation, R2 andlor R3 is selected from the group consisting of a (Cl_3)alkyl; an amino, (Cl_lo)alkyl, or alkoxy carbonyl (Cl_3)alkyl; an amino, (Cl_lo)alkyl, or alkoxy thiocarbonyl (Cl_3)alkyl; an amino, (C1_lo)alkyl, or alkoxy sulfonyl (Cl_3)alkyl; an amino, (Cl_lo)alkyl, or alkoxy sulfinyl (Cl_3)alkyl; an amino, (Cl_lo)alkyl, or alkoxy imino (Cl_3)alkyl; an aryl (Cl-s)alkyl; and a heteroaryl (C1-s)alkyl, each unsubstituted or substituted.
[0185] In another particular variation, R2 and/or R3 is selected from the group consisting of 1,2-dihydro-2-oxo-pyridinyl-, 1,4-dihydro-4-oxo-pyridinyl-, 2,3-dihydro-3-oxo-pyridazinyl-,1,2,3,6-tetrahydro-3,6-dioxo-pyridazinyl-,1,2-dihydro-2-oxo-pyrimidinyl-, 3,4-dihydro-4-oxo-pyrimidinyl-, 1,2,3,4-tetrahydro-2,4-dioxo-pyrimidinyl-, 1,2-dihydro-2-oxo-pyrazinyl, 1,2,3,4-tetrahydro-2,3-dioxo-pyrazinyl-, 2,3-dihydro-2-oxo-indolyl-, 2,3-dihydrobenzofuranyl-, 2,3-dihydro-2-oxo-1H-benzimidazolyl-, 2,3-dihydro-2-oxo-benzoxazolyl-,1,2-dihydro-2-oxo-quinolinyl-,1,4-dihydro-4-oxo-quinolinyl-,1,2-dihydro-1-oxo-iso-quinolinyl-, 1,4-dihydro-4-oxo-cinnolinyl-, 1,2-dihydro-2-oxo-quinazolinyl-, 1,4-dihydro-4-oxo-quinazolinyl-,1,2,3,4-tetrahydro-2,4-dioxo-quinazolinyl-,1,2-dihydro-2-oxo-quinoxalinyl-,1,2,3,4-tetrahydro-2,3-dioxo-quinoxalinyl-,1,2-dihydro-1-oxo-phthalazinyl-, 1,2,3,4-tetrahydro-1,4-dioxo-phthalazinyl-, chromanyl-, cumarinyl-, 2,3-dihydro-benzo[1,4]dioxinyl-, and 3,4-dihydro-3-oxo-2H-benzo[1,4]oxazinyl-, each unsubstituted or substituted.
[0186] In one variation, at least one of R2 and R3 is not hydrogen. In another variation, both R2 and R3 are not hydrogen.
[0187] In yet another variation, R2 is selected from the group consisting of 2-fluorophenyl, styryl, 4-fluorostyryl, 2-methoxyphenyl, pyrrolidin-1-yl, imidazolyl, and 3-furanyl each unsubstituted or substituted, and R3 is selected from the group consisting of hydrogen, (Cl_lo)alkyl, and aryl, each unsubstituted or substituted.
[0188] In one particular variation, R2 is selected from the group consisting of hydrogen, carboxyl, cyano, nitro, bromo, fluoro, chloro, iodo, -CF3, -CH3 and hydroxy, and R3 is selected from the group consisting of hydrogen, (C1_lo)alkyl, and aryl, each unsubstituted or substituted. In another particular variation, RZ is selected from the group consisting of hydrogen, carboxyl, cyano, nitro, bromo, fluoro, chloro, iodo, -CF3, -CH3 and hydroxy, and R3 is selected from the group consisting of hydrogen, (C3_~)cycloalkyl and aryl, each unsubstituted or substituted.
[0189] In one variation, the present invention comprises compounds where RZ or R3 is selected from the group consisting of -NH2, -NHCH3, -N(CH3)2, -NHCI_3-alkyl, and -N(Cl_3-alkyl)2 each unsubstituted or substituted; and Z is selected from the group consisting of 3-amino-piperidinyl-1-yl, 3-aminomethyl-pyrrolidin-1-yl, 2-aminoazetidin-1-yl, aminoazetidin-1-yl, 3-anuno-3-methylpiperidin-1-yl, 3-aminocyclopentyl, 3-aminomethylcyclopentyl, 3-aminomethylcyclohexyl, 3-aminohexahydroazepin-1-yl, 3-amino-cyclohexyl, piperazin-1-yl, homopiperazin-1-yl, 3-amino-pyrrolidin-1-yl, and R-aminopiperidin-1-yl, each unsubstituted or substituted.
[0190] In another variation, the present invention comprises compounds where Q
is CO;
and Rl is selected from the group consisting of -(CH2)-(2-cyano)phenyl, -(CH2)-(3-cyano)phenyl, -(CHZ)-(2-hydroxy)phenyl, -(CH2)-(3-hydroxy)phenyl, -(CH2)-(2-alkenyl)phenyl, -(CHZ)-(2-alkynyl)phenyl, -(CH2)-(2-methoxy)phenyl, -(CH2)-(3-methoxy)phenyl, -(CHZ)-(2-nitro)phenyl, -(CH2)-(3-nitro)phenyl, -(CHZ)-(3-carboxy)phenyl, -(CHZ)-(2-carboxarnido)phenyl, -(CH2)-(3-carboxamido)phenyl, -(CH2)-(2-sulfonamido)phenyl, -(CH2)-(3-sulfonamido)phenyl, -(CH2)-(2-tetrazolyl)phenyl, -(CHa)-(3-tetrazolyl)phenyl, -(CH2)-(2-aminomethyl)phenyl, -(CH2)-(2-amino)phenyl, -(CH2)-(3-amino)phenyl, -(CHZ)-(3-aminomethyl)phenyl, -(CH2)-(2-hydroxymethyl)phenyl, -(CHZ)-(3-hydroxymethyl)phenyl, -(CHZ)-(2-phenyl)phenyl, -(CH2)-(3-phenyl)phenyl, -(CHZ)-(2,-halo)phenyl, -(CHZ)-(3-halo)phenyl, -(CH2)-(2-CONH2)phenyl, -(CH2)-(2-CONH(Cl-~)alkyl)phenyl, -(CH2)-(2-C02(Ci-~)alkyl)phenyl, -CH2-NH2, -CH2-OH, -CHa-(C3-)alkyl, -CH2-alkene, -CH2-alkyne, -CH2-CCH, -CH2-(C3-~)cycloalkyl, -CH2-aryl, -CH2-heteroaryl, and -CH2-heterocycloalkyl, each substituted or unsubstituted.
[0191] In another variation, the present invention comprises compounds where Q
is CO;
and Z is selected from the group consisting of 3-amino-piperidinyl-1-yl, 3-aminomethyl-pyrrolidin-1-yl, 2-aminoazetidin-1-yl, 3-aminoazetidin-1-yl, 3-amino-3-methylpiperidin-1-yl, 3-aminocyclopentyl, 3-aminomethylcyclopentyl, 3-aminomethylcyclohexyl, 3-aminohexahydroazepin-1-yl, 3-amino-cyclohexyl, piperazin-1-yl, homopiperazin-1-yl, 3-amino-pyrrolidin-1-yl, and R-3-aminopiperidin-1-yl, each unsubstituted or substituted.
[0192] In another variation, the present invention comprises compounds where Q
is SO;
and Rl is selected from the group consisting of -(CHZ)-(2-cyano)phenyl, -(CH2)-(3-cyano)phenyl, -(CH2)-(2-hydroxy)phenyl, -(CH2)-(3-hydroxy)phenyl, -(CHZ)-(2-alkenyl)phenyl, -(CHZ)-(2-alkynyl)phenyl, -(CH2)-(2-methoxy)phenyl, -(CH2)-(3-methoxy)phenyl, -(CHZ)-(2-nitro)phenyl, -(CHZ)-(3-nitro)phenyl, -(CH2)-(2-carboxy)phenyl, -(CH2)-(3-carboxy)phenyl, -(CHZ)-(3-carboxamido)phenyl, -(CH2)-(2-sulfonamido)phenyl, -(CH2)-(3-sulfonamido)phenyl, -(CHZ)-(2-tetrazolyl)phenyl, -(CHZ)-(3-tetrazolyl)phenyl, -(CHZ)-(2-aminomethyl)phenyl, -(CH2)-(2-amino)phenyl, -(CH2)-(3-amino)phenyl, -(CH2)-(3-aminomethyl)phenyl, -(CHZ)-(2-hydroxymethyl)phenyl, -(CHZ)-(3-hydroxymethyl)phenyl, -(CHZ)-(2-phenyl)phenyl, -(CHZ)-(3-phenyl)phenyl, -(CH2)-(2-halo)phenyl, -(CH2)-(3-halo)phenyl, -(CH2)-(2.-CONH2)phenyl, -(CHZ)-(2-CONH(Cl-~)alkyl)phenyl, -(CHa)-(2-C02(Cl-~)alkyl)phenyl, -CH2-NH2, -CHZ-OH, -CH2-(C3-)alkyl, -CHa-alkene, -CH2-alkyne, -CH2-CCH, -CH2-(C3-~)cycloalkyl, -CH2-aryl, -CHZ-heteroaryl, and -CH2-heterocycloalkyl, each substituted or unsubstituted.
[0193] In another variation, the present invention comprises compounds where Q
is SO;
and Z is selected from the group consisting of 3-amino-piperidinyl-1-yl, 3-aminomethyl-pyrrolidin-1-yl, 2-aminoazetidin-1-yl, 3-aminoazetidin-1-yl, 3-amino-3-methylpiperidin-1-yl, 3-aminocyclopent-1-yl, 3-aminomethylcyclopent-1-yl, 3-aminomethylcyclohex-1-yl, 3-aminohexahydroazepin-1-yl, 3-amino-cyclohex-1-yl, piperazin-1-yl, homopiperazin-1-yl, 3-amino-pyrrolidin-1-yl, and R-3-aminopiperidin-1-yl, each unsubstituted or substituted.
[0194] In another variation, the present invention comprises compounds where Q
is 502;
and Rl 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)-(2-alkynyl)phenyl, -(CH2)-(2-methoxy)phenyl, -(CH2)-(3-methoxy)phenyl, -(CH2)-(2-nitro)phenyl, -(CH2)-(3-nitro)phenyl, -(CH2)-(2-carboxy)phenyl, -(CH2)-(3-carboxy)phenyl, -(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-hydroxymethyl)phenyl, -(CH2)-(3-hydroxymethyl)phenyl, -(CH2)-(2-phenyl)phenyl, -(CH2)-(3-phenyl)phenyl, -(CH2)-(2-halo)phenyl, -(CH2)-(3-halo)phenyl, -(CH2)-(2-CONH2)phenyl, -(CH2)-(2-CONH(Cl-~)alkyl)phenyl, -(CH2)-(2-C02(Cl-~)alkY1)phenyl, -CH2-NH2, -CH2-OH, -CH2-(C3-)alkyl, -CH2-alkene, -CH2-alkyne, -CH2-CCH, -CH2-(C3-~)cycloalkyl, -CH2-aryl, -CH2-heteroaryl, and -CH2-heterocycloalkyl, each substituted or unsubstituted.
[0195] In another variation, the present invention comprises compounds where Q
is 502;
and Z is selected from the group consisting of 3-amino-piperidinyl-1-yl, 3-aminomethyl-pyrrolidin-1-yl, 2-aminoazetidin-1-yl, 3-aminoazetidin-1-yl, 3-amino-3-methylpiperidin-1-yl, 3-aminocyclopent-1-yl, 3-aminomethylcyclopent-1-yl, 3-aminomethylcyclohex-1-yl, 3-aminohexahydroazepin-1-yl, 3-amino-cyclohex-1-yl, piperazin-1-yl, homopiperazin-1-yl, 3-amino-pyrrolidin-1-yl, and R-3-aminopiperidin-1-yl, each unsubstituted or substituted.
[0196] In another variation, the present invention comprises compounds where Q
is CO;
-L-X together are 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)-(2-alkynyl)phenyl, -(CH2)-(2-methoxy)phenyl, -(CH2)-(3-methoxy)phenyl, -(CH2)-(2-nitro)phenyl, -(CH2)-(3-nitro)phenyl, -(CH2)-(2-carboxy)phenyl, -(CH2)-(3-carboxy)phenyl, -(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-hydroxymethyl)phenyl, -(CH2)-(3-hydroxymethyl)phenyl, -(CH2)-(2.-phenyl)phenyl, -(CH2)-(3-phenyl)phenyl, -(CH2)-(2-halo)phenyl, -(CH2)-(3-halo)phenyl, -(CH2)-(2-CONH2)phenyl, -(CH2)-(2-CONH(Cl-~)alkyl)phenyl, -(CH2)-(2-C02(Cl-~)alkyl)phenyl, -CH2-NH2, -CH2-OH, -CH2-(C3-)alkyl, -CH2-alkene, -CH2-alkyne, -CH2-CCH, -CH2-(C3-~)cycloalkyl, -CH2-aryl, -CH2-heteroaryl, and -CH2-heterocycloalkyl, each substituted or unsubstituted; and Z is selected from the group consisting of 3-amino-piperidinyl-1-yl, 3-aminomethyl-pyrrolidin-1-yl, 2-aminoazetidin-1-yl, 3-aminoazetidin-1-yl, 3-amino-3-methylpiperidin-1-yl, 3-aminocyclopent-1-yl, 3-aminomethylcyclopent-1-yl, 3-aminomethylcyclohex-1-yl, 3-aminohexahydroazepin-1-yl, 3-axnino-cyclohex-1-yl, piperazin-1-yl, homopiperazin-1-yl, 3-amino-pyrrolidin-1-yl, and R-3-aminopiperidin-1-yl, each unsubstituted or substituted.
[0197] In another variation, the present invention comprises compounds where Q
is SO;
-L-X together are 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)-(2-alkynyl)phenyl, -(CH2)-(2-methoxy)phenyl, -(CH2)-(3-methoxy)phenyl, -(CH2)-(2-nitro)phenyl, -(CH2)-(3-nitro)phenyl, -(CH2)-(2-carboxy)phenyl, -(CH2)-(3-carboxy)phenyl, -(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, -(CHZ)-(2-amino)phenyl, -(CH2)-(3-amino)phenyl, -(CH2)-(2-hydroxymethyl)phenyl, -(CH2)-(3-hydroxymethyl)phenyl, -(CH2)-(2-phenyl)phenyl, -(CH2)-(3-phenyl)phenyl, -(CH2)-(2-halo)phenyl, -(CH2)-(3-halo)phenyl, -(CH2)-(2-CONH2)phenyl, -(CH2)-(2-CONH(Cl-~)alkyl)phenyl, -(CH2)-(2-C02(Cl-~)alkyl)phenyl, -CH2-NH2, -CH2-OH, -CH2-(C3-)alkyl, -CH2-alkene, -CH2-alkyne, -CH2-CCH, -CH2-(C3-~)cycloalkyl, -CH2-aryl, -CH2-heteroaryl, and -CH2-heterocycloalkyl, each substituted or unsubstituted; and Z is selected from the group consisting of 3-amino-piperidinyl-1-yl, 3-aminomethyl-pyrrolidin-1-yl, 2-aminoazetidin-1-yl, 3-aminoazetidin-1-yl, 3-amino-3-methylpiperidin-1-yl, 3-aminocyclopent-1-yl, 3-aminomethylcyclopent-1-yl, 3-aminomethylcyclohex-1-yl, 3-aminohexahydroazepin-1-yl, 3-amino-cyclohex-1-yl, piperazin-1-yl, homopiperazin-1-yl, 3-amino-pyrrolidin-1-yl, and R-3-aminopiperidin-1-yl, each unsubstituted or substituted.
[0198] In another variation, the present invention comprises compounds where Q
is 502;
-L-X together are selected from the group consisting of -(CH2)-(2-cyano)phenyl, -(CH2)-(3-cyano)phenyl, -(CH2)-(2-hydroxy)phenyl, -(CH2)-(3-hydroxy)phenyl, -(CHI)-(2-alkenyl)phenyl, -(CHZ)-(2-alkynyl)phenyl, -(CH2)-(2-methoxy)phenyl, -(CH2)-(3-methoxy)phenyl, -(CHZ)-(2,-nitro)phenyl, -(CH2)-(3-nitro)phenyl, -(CHZ)-(2-carboxy)phenyl, -(CH2)-(3-carboxy)phenyl, -(CH2)-(3-carboxamido)phenyl, -(CH2)-(2-sulfonamido)phenyl, -(CHa)-(3-sulfonamido)phenyl, -(CH2)-(2-tetrazolyl)phenyl, -(CH2)-(3-tetrazolyl)phenyl, -(CHa)-(2-aminomethyl)phenyl, -(CHZ)-(3-aminomethyl)phenyl, -(CH2)-(2-amino)phenyl, -(CH2)-(3-amino)phenyl, -(CH2)-(2-hydroxymethyl)phenyl, -(CH2)-(3-hydroxymethyl)phenyl, -(CH2)-(2-phenyl)phenyl, -(CH2)-(3-phenyl)phenyl, -(CH2)-(2-halo)phenyl, -(CH2)-(3-halo)phenyl, -(CHZ)-(2-CONHZ)phenyl, -(CHZ)-(2-CONH(Cl-~)alkyl)phenyl, -(CH2)-(2-CO2(Cl-~)alkyl)phenyl, -CH2-NH2, -CHI-OH, -CH2-(C3-)alkyl, -CH2-alkene, -CH2-alkyne, -CH2-CCH, -CHZ-(C3-~)cycloalkyl, -CHZ-aryl, -CH2-heteroaryl, and -CHa-heterocycloalkyl, each substituted or unsubstituted; and Z is selected from the group consisting of 3-amino-piperidinyl-1-yl, 3-aminomethyl-pyrrolidin-1-yl, 2-aminoazetidin-1-yl, 3-aminoazetidin-1-yl, 3-amino-3-methylpiperidin-1-yl, 3-aminocyclopent-1-yl, 3-aminomethylcyclopent-1-yl, 3-aminomethylcyclohex-1-yl, 3-aminohexahydroazepin-1-yl, 3-amino-cyclohex-1-yl, piperazin-1-yl, homopiperazin-1-yl, 3-amino-pyrrolidin-1-yl, and R-3-aminopiperidin-1-yl, each unsubstituted or substituted.
SUBSTITUENTS M AND R19:
[0199] In one embodiment, M is a moiety providing 1, 2, 3, 4, 5, or 6 atom separation between R19 and the ring to which M is attached. In one variation, M provides 1, 2, 3, or 4 atom separation between R19 and the ring. In another variation, M provides 1, 2, or 3 atom separation between R19 and the ring.
[0200] In yet another variation, M is selected from the group consisting of -CHZ-, -CHaCH2-, -CHZCHZCH2-, -C(O)-, -CH2C(O)-, -C(O)CH2-, -CHa-C(O)CHZ-, -C(O)CH2CH2-, -CH2CH2C(O)-, -O-, -OCHZ-, -CH20-, -CH20CH~-, -OCHZCH2-, -CHZCHZO-, -N(CH3)-, -NHCH2-, -CH2NH-, -CH2NHCHa-, -NHCHaCH2-, -CH2CHZNH-, -NH-C(O)-, -NCH3-C(O)-, -C(O)NH-, -C(O)NCHS-, -NHC(O)CHZ-, -C(O)NHCH2-, -C(O)CHZNH-, -CH2NHC(O)-, -CH2C(O)NH-, -NHCHZC(O)-, -S-, -SCHa-, -CH2S-, -SCH2CH2-, -CHZSCHZ-, -CH2CH2S-, -C(O)S-, -C(O)SCH2-, -CHZC(O)S-, -C(O)CHZS-, and -CH2SC(O)-.
[0201] In one embodiment, R19 comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein. In one variation, Rlg 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. In another variation, Rl9 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.
[0202] In still another variation, the basic nitrogen of R19 is separated from the ring atom to which M is attached by between 1-5 atoms. In another variation, the basic nitrogen of R19 forms part of a primary, secondary, or tertiary amine. In yet a further variation, R19 is selected from the group consisting of a heterocycloalkyl comprising a nitrogen ring atom or a heteroaryl comprising a nitrogen ring atom.
[0203] In one variation, -MR19 is selected from the group consisting of:
-N~ -~_N -~_N~ -~_N
~\R ~~ \R
8)p ~ ~ 8)p ~Re)p ~Rs)p wherein p is 0-12 and each R$ is independently selected from the group consisting of halo, perhalo(Cl-io)alkyl, CF3, cyano, vitro, 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 includes the basic nitrogen of R19.
[0204] In another variation, -MR19 is selected from the group consisting of:
-~~-'~R8)r -~--<~ - - -(R$)r ~ ~R8)r ~R8)r ~R8)r wherein r is 0-13 and each R8 is independently selected from the group consisting of halo, perhalo(Cl-io)alkyl, CF3, cyano, vitro, hydroxy, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, alkoxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, with the proviso that at least one R8 includes the basic nitrogen of R19.
[0205] In one particular variation, at least one R$ is a primary, secondary, or tertiary amine. In another particular variation, at least one R$ is a substituted or unsubstituted heterocycloalkyl comprising a nitrogen ring atom or a substituted or unsubstituted heteroaryl comprising a nitrogen ring atom. In still another variation, at least one Rg is selected from the group consisting of -NH2, -NH(Ci_5 alkyl), -N(Ci_5 alkyl) Z, piperazine, imidazole, and pyridine.
Particular examules of synthetic intermediates [0206] In one embodiment, particular examples of synthetic intermediates useful for the production of DPP-IV inhibitors according to the present invention include, but are not limited to:
5-Bromo-2-chloro-3H pyrimidin-4-one;
5-Bromo-2-chloro-6-methyl-3H-pyrimidin-4-one;
2-Chloro-5-ethyl-3H-pyrimidin-4-one;
2,5-Dichloro-3H-pyrimidin-4-one;
2-Chloro-5-methoxy-3H-pyrimidin-4-one; and 2-Chloro-5-fluoro-3H-pyrimidin-4-one.
[0207] In another embodiment, particular examples of synthetic intermediates useful for the production of DPP-IV inhibitors according to the present invention include, but are not limited to:
2-(5-Bromo-2-chloro-6-oxo-6H pyrimidin-1-ylmethyl)-benzonitrile;
2-[2-(3-Amino-piperidin-1-yl)-5-trimethylsilylethynyl-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile;
2-(5-Bromo-2-chloro-4-methyl-6-oxo-6H-pyrimidin-1-ylmethyl)-benzonitrile;
2-(2-Chloro-4,5-dimethyl-6-oxo-6H-pyrimidin-lylmethyl)-benzonitrile;
2-(2-Chloro-5-ethyl-6-oxo-6H-pyrimidin-1-ylmethyl)-benzonitrile;
2-(2,5-Dichloro-6-oxo-6H-pyrimidin-1-ylmethyl)-benzonitrile;
2-((5-fluoro-2-chloro-6-oxopyrimidin-1 (6H)-yl)methyl)-5-fluorobenzonitrile;
2-((5-fluoro-2-chloro-6-oxopyrimidin-1 (6H)-yl)methyl)-4-fluorobenzonitrile;
2-((5-bromo-2-chloro-6-oxopyrimidin-1 (6H)-yl)methyl)-4-fluorobenzonitrile;
2-(5-Bromo-2-chloro-6-oxo-6H pyrimidin-lylmethyl)-thiophene-3-carbonitrile;
2-(2-Chloro-5-methoxy-6-oxo-6H-pyrimidin-1-ylmethyl)-benzonitrile;

2-(2-Chloro-5-fluoro-6-oxo=6H pyrimidin-1-ylmethyl)-benzonitrile;
3-(2-Bromo-benzyl)-2-chloro-5-fluoro-3H pyrimidin-4-one; and 2-(2-Chloro-6-oxo-4-phenyl-6H pyrimidin-1-ylmethyl)-benzonitrile.
Particular examples of DPP-IV inhibitors [0208] Particular examples of DPP-IV inhibitors according to the present invention include, but are not limited to:
2-[2-(3-(R)-Amino-piperidin-1-yl)-6-oxo-5-phenyl-6H pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-furan-3-yl-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-(2-fluoro-phenyl)-6-oxo-6H pyrimidin-1-ylmethyl]-benzonitrile;
2-{ 2-(3-(R)-Amino-piperidin-1-yl)-5-[2-traps-(4-fluoro-phenyl)-vinyl]-6-oxo-pyrimidin-1 ylmethyl } -benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-(2-methoxy-phenyl)-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-bromo-4-methyl-6-oxo-6H pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-4-methyl-6-oxo-5-phenyl-6H pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-furan-3-yl-4-methyl-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-4-methyl-6-oxo-5-traps-styryl-6H pyrimidin-1-ylmethyl]-benzonitrile;
2-[2.-(3-(R)-Amino-piperidin-1-yl)-4,5-dimethyl-6-oxo-6H pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-ethyl-6-oxo-6H pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-iodo-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile;

2-[2-(3-(R)-Amino-piperidin-1-yl)-5-bromo-6-oxo-6H pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-ethynyl-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-chloro-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile;
(R)-2-((2-(3-axninopiperidin-1-yl)-6-oxo-5-(pyrrolidin-1-yl)pyrimidin-1 (6H)-yl)methyl)benzonitrile;
(R)-2-((2-(3-aminopiperidin-1-yl)-5-fluoro-6-oxopyrimidin-1 (6H)-yl)methyl)-5-fluorobenzonitrile;
(R)-2-((2-(3-aminopiperidin-1-yl)-5-fluoro-6-oxopyrimidin-1 (6H)-yl)methyl)-4-fluorobenzonitrile;
(R)-2-((2-(3-aminopiperidin-1-yl)-5-chloro-6-oxopyrimidin-1 (6H)-yl)methyl)-4-fluorobenzonitrile;
(R)-2-((2-(3-aminopiperidin-1-yl)-5-bromo-6-oxopyrimidin-1 (6H)-yl)methyl)-4-fluorobenzonitrile;
(R)-2-((2-(3-amino-3-methylpiperidin-1-yl)-5-fluoro-6-oxopyrimidin-1 (6H)-yl)methyl)-4-fluorobenzonitrile;
2-[2-(3-(R)-Amino-pyrimidin-1-yl)-5-bromo-6-oxo-6H pyrimidin-1-ylmethyl]-thiophene-3-carbonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-(3,3-dimethyl-but-1-ynyl)-6-oxo-6H
pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-prop-1-ynyl-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-(3-hydroxy-prop-1-ynyl)-6-oxo-6H pyrimidin-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-6-oxo-6H pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-6-oxo-5-vinyl-6H pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-methoxy-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile;

2-[2-(3-(R)-Amino-piperidin-1-yl)-6-oxo-5-(1H-pyrrol-3-yl)-6H-pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-fluoro-6-oxo-6H pyrimidin-1-ylmethyl]-benzonitrile;
2-(3-(R)-Amino-piperidin-1-yl)-3-(2-bromo-benzyl)-5-fluoro-3H pyrimidin-4-one;
2-[2-(3-(R)-Amino-piperidin-1-yl)-6-oxo-5-((E7-2-pyridin-3-yl-vinyl)-6H
pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-6-oxo-4-phenyl-6H-pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-3-methyl-piperidin-1-yl)-5-fluoro-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-3-methyl-piperidin-1-yl)-5-chloro-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile; and (R)-2-((2-(3-aminopiperidin-1-yl)-5-bromo-4-tent-butyl-6-oxopyrimidin-1 (6H)-yl)methyl)benzonitrile.
[0209] In one variation, particular examples of DPP-IV inhibitors according to the present invention include, but are not limited to:
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-bromo-6-oxo-6H pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-ethynyl-6-oxo-6H pyrimidin-1-ylmethyl]-benzonitrile;
2-[2.-(3-(R)-Amino-piperidin-1-yl)-4,5-dimethyl-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile;
2-[2,-(3-(R)-Amino-piperidin-1-yl)-5-chloro-6-oxo-6H pyrimidin-1-ylmethyl]-benzonitrile;
(R)-2-((2-(3-aminopiperidin-1-yl)-5-fluoro-6-oxopyrimidin-1 (6H)-yl)methyl)-4-fluorobenzonitrile;
(R)-2-((2-(3-aminopiperidin-1-yl)-5-chloro-6-oxopyrimidin-1 (6H)-yl)methyl)-4-fluorobenzonitrile;
(R)-2-((2-(3-aminopiperidin-1-yl)-5-bromo-6-oxopyrimidin-1 (6H)-yl)methyl)-4-fluorobenzonitrile;

2-[2-(3-(R)-Amino-piperidin-1-yl)-5-prop-1-ynyl-6-oxo-6H pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-fluoro-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile; and 2-[2-(3-(R)-Amino-3-methyl-piperidin-1-yl)-5-chloro-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile.
[0210] In another embodiment, the present invention provides the compounds in the form of a pharmaceutically acceptable salt. In one variation, the compound is in the form of a pharmaceutically acceptable salt selected from the group consisting of a TFA
salt, a tartaric acid salt, and an HCl salt. In another variation, DPP-IV inhibitors of the present invention include, but are not limited to:
2-[2-(3-(R)-Amino-piperidin-1-yl)-6-oxo-5-phenyl-6H pyrimidin-1-ylmethyl]-benzonitrile, TFA salt;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-furan-3-yl-6-oxo-6F1 pyrimidin-1-ylmethyl]-benzonitrile, TFA salt;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-(2-fluoro-phenyl)-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile, TFA salt;
2- { 2-(3-(R)-Amino-piperidin-1-yl)-5-[2-tracts-(4-fluoro-phenyl)-vinyl]-6-oxo-pyrimidin-lylmethyl}-benzonitrile, TFA salt;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-(2-methoxy-phenyl)-6-oxo-6H pyrimidin-1-ylmethyl]-benzonitrile, TFA salt;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-bromo-4-methyl-6-oxo-6H pyrimidin-1-ylmethyl]-benzonitrile, TFA salt;
2-[2-(3-(R)-Amino-piperidin-1-yl)-4-methyl-6-oxo-5-phenyl-6H pyrimidin-1-ylmethyl]-benzonitrile, TFA salt;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-furan-3-yl-4-methyl-6-oxo-6H pyrimidin-1-ylmethyl]-benzonitrile, TFA salt;
2-[2-(3-(R)-Amino-piperidin-1-yl)-4-methyl-6-oxo-5-tYans-styryl-6H-pyrimidin-1-ylmethyl]-benzonitrile, TFA salt;
2-[2-(3-(R)-Amino-piperidin-1-yl)-4,5-dimethyl-6-oxo-6H pyrimidin-1-ylmethyl]-benzonitrile, TFA salt;

2-[2-(3-(R)-Amino-piperidin-1-yl)-5-ethyl-6-oxo-6H pyrimidin-1-ylmethyl]-benzonitrile, TFA salt;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-iodo-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile, TFA salt;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-bromo-6-oxo-6H pyrimidin-1-ylmethyl]-benzonitrile, TFA salt;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-ethynyl-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-chloro-6-oxo-6H pyrimidin-1-ylmethyl]-benzonitrile, TFA salt;
(R)-2-((2-(3-aminopiperidin-1-yl)-6-oxo-5-(pyrrolidin-1-yl)pyrimidin-1 (6H)-yl)methyl)benzonitrile, TFA salt;
(R)-2-((2-(3-aminopiperidin-1-yl)-5-fluoro-6-oxopyrimidin-1 (6H)-yl)methyl)-5-fluorobenzonitrile, TFA salt;
(R)-2-((2-(3-aminopiperidin-1-yl)-5-fluoro-6-oxopyrimidin-1 (6H)-yl)methyl)-4-fluorobenzonitrile, TFA salt;
(R)-2-((2-(3-aminopiperidin-1-yl)-5-chloro-6-oxopyrimidin-1 (6H)-yl)methyl)-4-fluorobenzonitrile, TFA salt;
(R)-2-((2-(3-aminopiperidin-1-yl)-5-bromo-6-oxopyrimidin-1 (6H)-yl)methyl)-4-fluorobenzonitrile, TFA salt;
(R)-2-((2-(3-amino-3-methylpiperidin-1-yl)-5-fluoro-6-oxopyrimidin-1 (6H)-yl)methyl)-4-fluorobenzonitrile, TFA salt;
2-[2-(3-(R)-Amino-pyrimidin-1-yl)-5-bromo-6-oxo-6H pyrimidin-1-ylmethyl]-thiophene-3-carbonitrile, TFA salt;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-(3,3-dimethyl-but-1-ynyl)-6-oxo-6H
pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-prop-1-ynyl-6-oxo-6H pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-(3-hydroxy-prop-1-ynyl)-6-oxo-6H pyrimidin-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-6-oxo-6H pyrimidin-1-ylmethyl]-benzonitrile, TFA salt;
2-[2-(3-(R)-Amino-piperidin-1-yl)-6-oxo-5-vinyl-6H pyrimidin-1-ylmethyl]-benzonitrile, TFA salt;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-methoxy-6-oxo-6H pyrimidin-1-ylmethyl]-benzonitrile, TFA salt;
. 2-[2-(3-(R)-Amino-piperidin-1-yl)-6-oxo-5-(1H pyrrol-3-yl)-6H pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-fluoro-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile, TFA salt;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-fluoro-6-oxo-6H pyrimidin-1-ylmethyl]-benzonitrile, tartaric acid salt;
2,-(3-(R)-Amino-piperidin-1-yl)-3-(2-bromo-benzyl)-5-fluoro-3H-pyrimidin-4-one, HCl salt;
2-[2-(3-(R)-Amino-piperidin-1-yl)-6-oxo-5-((E~-2-pyridin-3-yl-vinyl)-6H
pyrimidin-1-ylmethyl]-benzonitrile, HCl salt;
2-[2-(3-(R)-Amino-piperidin-1-yl)-6-oxo-4-phenyl-6H-pyrimidin-1-ylmethyl]-benzonitrile, HCl salt;
2.-[2-(3-(R)-Amino-3-methyl-piperidin-1-yl)-5-fluoro-6-oxo-6H pyrimidin-1-ylmethyl]-benzonitrile, HCl salt;
2,-[2-(3-(R)-Amino-3-methyl-piperidin-1-yl)-5-chloro-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile, HCl salt; and (R)-2-((2-(3-aminopiperidin-1-yl)-5-bromo-4-tert-butyl-6-oxopyrimidin-1 (6H)-yl)methyl)benzonitrile, TFA salt.
[0211] 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.
[0212] In 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.
[0213] 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 stmt), subcutaneously, intraadiposally, intraarticularly, and intrathecally.
[0214] 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. In another embodiment, the present invention provides the kit that comprises the compound in a multiple dose form.
[0215] In another embodiment, the present invention provides an article of manufacture comprising a compound of the present invention, and packaging materials. In 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 andlor instructions regarding how to administer the composition.
[0216] In another variation, the present invention provides the article of manufacture wherein the article of manufacture comprises the compound in a multiple dose form.
[0217] In another embodiment, the present invention provides a method of inhibiting DPP-IV comprising contacting DPP-IV with a compound according to the present invention.
[0218] In another embodiment, the present invention provides a method of inhibiting DPP-IV comprising causing a compound according to the present invention to be present in a subject in order to inhibit DPP-IV ifz vivo.
[0219] In another embodiment, the present invention provides a method of inhibiting DPP-IV comprising: administering a first compound to a subject that is converted in vivo to a second compound wherein the second compound inhibits DPP-IV in vivo, the second compound being a compound of the present invention.
[0220] In another embodiment, the present invention provides therapeutic method comprising: administering a compound according to the present invention to a subject.
[0221] In another embodiment, the present invention provides a method of treating a disease state for which DPP-IV 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.
[0222] In another embodiment, the present invention provides a method of treating a disease where the disease is type I or type II diabetes.
[0223] 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.
[0224] 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.
[0225] 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.
[0226] In 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.
[0227] 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.
[0228] 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.
[0229] 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 a bone marrow disorder.
[0230] In another embodiment, the present invention provides a method of treating HIV
infection in a patient in need thereof, comprising administering to said patient a therapeutically effective amount of a compound according to the present invention.
[0231] In another embodiment, the present invention provides a method of treating a condition characterized by immunodeficiency symptoms in a patient in need thereof, comprising administering to said patient a therapeutically effective amount of a compound according to the present invention.
[0232] 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 [0233] 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.
[0234] 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;
othermineral acids and their corresponding salts such as sulfate, nitrate, phosphate, etc.; and alkyl and monoarylsulfonates such as ethanesulfonate, toluenesulfonate and benzenesulfonate; 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 (from mucic 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.
[0235] 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 limited 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), dicyclohexylamine, diethylamine, 2-diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethylmorpholine, N-ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, iso-propylamine, lidocaine, lysine, meglumine, N-methyl-D-glucamine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethanolamine, triethylamine, trimethylamine, tripropylamine and tris-(hydroxymethyl)-methylamine (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.
[0236] Compounds of the present invention that comprise basic nitrogen-containing groups may be quaternized with such agents as (Cl_4)alkyl halides, e.g., methyl, ethyl, iso-propyl and tert-butyl chlorides, bromides and iodides; di (Cl.~)alkyl sulfates, e.g., dimethyl, diethyl and diamyl sulfates; (Clo_18)alkyl halides, e.g., decyl, dodecyl, lauryl, myristyl and stearyl chlorides, bromides and iodides; and aryl (C1~)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.
[0237] 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, fneta-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.
[0238] 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 ij~ 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.,1,1-acyloxyalkylcarbonochloridate,para-nitrophenyl carbonate, or the like) or an acylating agent. Further examples of methods of making prodrugs are described in Saulnier et al.(1994), Bioorganic a~zd Medicinal Che~aistry Letters, Vol. 4, p.
1985.
[0239] 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.
[0240] 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 recrystallization from an aqueous/organic solvent mixture, using organic solvents such as dioxin, tetrahydrofuran, or methanol.
[0241] 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 administration 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
[0242] DPP-IV is believed to contribute to the pathology and/or symptomology of several different diseases such that reduction of the activity of DPP-IV in a subject through inhibition may be used to therapeutically address these disease states. Examples of various diseases that may be treated using the DPP-IV 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-IV plays in various pathways becomes more fully understood.
[0243] One set of indications that DPP-IV inhibitors of the present invention may be 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.
[0244] DPP-IV 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.
[0245] DPP-IV inhibitors of the present invention may also be used for treating various cancers including breast cancer, lung cancer and prostate cancer.
[0246] DPP-IV inhibitors of the present invention may also be used to treat dermatological diseases such as psoriasis, rheumatoid arthritis (RA) and lichen planus.
[0247] DPP-IV inhibitors of the present invention may also be used to treat infertility and amenorrhea.
[0248] DPP-IV 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.
[0249] DPP-IV inhibitors of the present invention may also be used prevent or reduce cleavage of N-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.
[0250] DPP-IV 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.
[0251] For oncology indications, DPP-IV 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-IV inhibitors of the present invention include, but are not limited to, retinoid acid and derivatives thereof, 2-methoxyestradiol, ANGIOSTATINTM protein, ENDOSTATINTM
protein, suramin, squalamine, tissue inhibitor of metalloproteinase-I, tissue inhibitor of metalloproteinase-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 ((1-azetidine-2-carboxylic acid (LACA)), cishydroxyproline, d,l-3,4-dehydroproline, thiaproline, beta.-aminopropionitrile fumarate, 4-propyl-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-a.ntiplasmin, bisantrene, lobenzarit disodium, n-2-carboxyphenyl-4-chloroanthronilic 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, VEGF-C, HGF/SF and Ang-1/Ang-2. Ferrara N.
and Alitalo, K. "Clinical application of angiogenic growth factors and their inhibitors" (1999) Nature Medicine 5:1359-1364.
4. COMPOSITIONS COMPRISING DPP-IV INHIBITORS
[0252] A wide variety of compositions and administration methods may be used in conjunction with the DPP-IV inhibitors of the present invention. Such compositions may include, in addition to the DPP-IV 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-IV 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.
[0253] 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 administration, capsules and tablets are typically used. For parenteral administration, reconstitution of a lyophilized powder, prepared as described herein, is typically used.
[0254] Compositions comprising DPP-IV 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 stmt), 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.
[0255] The DPP-IV 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-IV 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.
[0256] 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.
[0257] When DPP-IV 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 TWEEN, 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.
[0258] Upon mixing or adding DPP-1V inhibitors according to the present invention to a composition, a solution, suspension, emulsion, or the like may be formed. The form 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.
[0259] 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 animal subjects and packaged individually as is known in the art. Each unit-dose contains a predeternuned quantity of the therapeutically active compound sufficient to produce the desired therapeutic effect, in association with the required pharmaceutical calTier, vehicle or diluent. Examples of unit-dose forms include ampoules and syringes and individually packaged tablets or capsules. 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 pints or gallons. Hence, multiple dose form is a multiple of unit-doses that are not segregated in packaging.
[0260] In addition to one or more DPP-IV 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-1V inhibitor of the present invention to reduce DPP-IV activity irv vivo, thereby treating the disease state of the subject.
[0261] Dosage forms or compositions may optionally comprise one or more DPP-IV
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% (weightlweight) of one or more DPP-IV inhibitors, optionally 0.1-95%, and optionally 1-95%.
[0262] Salts, preferably sodium salts, of the DPP-IV 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 [0263] Oral pharmaceutical dosage forms 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.
[0264] In certain embodiments, DPP-IV 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.
[0265] 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.
[0266] Examples of lubricants that may be used include, but are not limited to, talc, starch, magnesium or calcium stearate, lycopodium and stearic acid.
[0267] Examples of diluents that may be used include, but are not limited to, lactose, sucrose, starch, kaolin, salt, mannitol and dicalcium phosphate.
[0268] Examples of glidants that may be used include, but are not limited to, colloidal silicon dioxide.
[0269] 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.
[0270] 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.
[0271] 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.
[0272] 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.
[0273] 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.
[0274] 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.
[0275] Examples of film coatings that may be used include, but are not limited to, hydroxyethylcellulose, sodium carboxymethylcellulose, polyethylene glycol 4000 and cellulose acetate phthalate.
[0276] 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.
[0277] When the dosage unit form is a capsule, it may optionally additionally comprise a liquid carrier 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.
[0278] 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.
[0279] The DPP-IV 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.
[0280] Examples of pharmaceutically acceptable carriers that may be included in tablets comprising DPP-1V 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.
[0281] Examples of liquid oral dosage forms that may be used include, but are not limited to, aqueous solutions, emulsions, suspensions, solutions andlor suspensions reconstituted from non-effervescent granules and effervescent preparations reconstituted from effervescent granules.
[0282] 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.
[0283] 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.
[0284] 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.
[0285] 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.
[0286] Coloring and flavoring agents may optionally be used in all of the above dosage forms.
[0287] Particular examples of preservatives that may be used include glycerin, methyl and propylparaben, benzoic add, sodium benzoate and alcohol.
[0288] Particular examples of non-aqueous liquids that may be used in emulsions include mineral oil and cottonseed oil.
[0289] Particular examples of emulsifying agents that may be used include gelatin, acacia, tragacanth, bentonite, and surfactants such as polyoxyethylene sorbitan monooleate.
[0290] Particular examples of suspending agents that may be used include sodium carboxymethylcellulose, pectin, tragacanth, Veegum and acacia. Diluents include lactose and sucrose. Sweetening agents include sucrose, syrups, glycerin and artificial sweetening agents such as sodium cyclamate and saccharin.
[0291] Particular examples of wetting agents that may be used include propylene glycol monostearate, sorbitan monooleate, diethylene glycol monolaurate and polyoxyethylene lauryl ether.
[0292] Particular examples of organic acids that may be used include citric and tartaric acid.
[0293] Sources 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.
[0294] 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.
[0295] 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. Nos.
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.
[0296] 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 [0297] The present invention is also directed to compositions designed to administer the DPP-IV 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.
[0298] 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.
[0299] Parenteral administration 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.
[0300] 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.
[0301] Examples of pharmaceutically acceptable carriers 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.
[0302] 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.
[0303] 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.
[0304] 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.
[0305] 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 polyvinylpyrrolidone. Examples of emulsifying agents that may be used include Polysorbate 80 (TWEEN 80). A sequestering or chelating agent of metal ions include EDTA.
[0306] 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.
[0307] The concentration of a DPP-IV 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-IV 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.
[0308) 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.
[0309] Injectables may be designed for local and systemic adnunistration.
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-IV
inhibitor to the treated tissue(s). The DPP-IV 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 adnunistered, the carrier and other variables that may be determined empirically using known testing protocols or by extrapolation from in vivo or ih 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.
[0310] The DPP-IV 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 [0311] The DPP-IV 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.
[0312] 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-IV inhibitor is added to the resulting mixture, preferably above room temperature, more preferably at about 30-35 °C, and stirred 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-IV inhibitor.
D. Topical administration [0313] The DPP-IV 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 administration.
[0314] The DPP-IV inhibitors may be formulated as aerosols for topical application, such as by inhalation (see, U.S. Pat. Nos. 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.
[0315] The DPP-IV 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 administration to the eyes or mucosa, or for inhalation therapies.
Nasal solutions of the DPP-IV inhibitor alone or in combination with other pharmaceutically acceptable excipients can also be administered.
E. Formulations for other routes of administration [0316] 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 [0317] 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 10-100 mg Invention Citric Acid Monohydrate 105 mg Sodium Hydroxide 18 mg Flavoring Water q.s. to 100 mL

INTRAVENOUS FORMULATION

Compound of the Present 0.1-10 mg Invention 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
[0318] The invention is also directed to kits and other articles of manufacture for treating diseases associated with DPP-IV. It is noted that diseases are intended to cover all conditions for which the DPP-IV possesses activity that contributes to the pathology and/or symptomology of the condition.
[0319] In one embodiment, a kit is provided that comprises a composition comprising at least one DPP-IV inhibitor of the present invention in combination with instructions. The instructions rnay 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.
[0320] In another embodiment, an article of manufacture is provided that comprises a composition comprising at least one DPP-IV 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.
[0321] 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. The 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.
[0322] 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.
[0323) 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 [0324] 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.
[0325] 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.
[0326) 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. (191).
[0327] 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.
[0328) The free acid or free base forms of the compounds can be prepared from the corresponding base addition salt or acid addition salt form. For example, a compound in an acid addition salt form can be converted to the corresponding 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 corresponding free acid by treating with a suitable acid (e.g., hydrochloric acid, etc).
[0329] 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, meta-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.
[0330] 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.
[0331] 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 al.
(1994), Bioorganic arid Medicinal Chemistry Letters, Vol. 4, p. 195). For example, appropriate prodrugs can be prepared by reacting a non-derivatized compound with a suitable carbamylating agent (e.g., 1,1-acyloxyalkylcarbonochloridate, para-nitrophenyl carbonate, or the like).
[0332] 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 ~z Sons, Inc. 1999.
[0333] 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.
[0334] 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 preferred (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).
[0335] 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);

~,L (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 THF (tetrahydrofuran);
anhydride);

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); HOAc (acetic acid);
HOSu (N-hydroxysuccinimino); HOBT (1-hydroxybenzotriazole);

Et2O (diethyl ether);
EDCI (ethylcarbodiimino hydrochloride);
BOC (tert-butyloxycarbonyl); FMOC (9-fluorenylmethoxycarbonyl);
DCC (dicyclohexylcarbodiimino); CBZ (benzyloxycarbonyl);
Ac (acetyl); atm (atmosphere);
TMSE (2-(trimethylsilyl)ethyl); TMS (trimethylsilyl);
TIPS (triisopropylsilyl); TBS (t-butyldimethylsilyl);
DMAP (4-dimethylaminopyridine); 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);
mCPBA (meta-chloroperbenzoic acid.
[0336] All references to ether or Et20 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.
[0337] 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).
[0338] Low-resolution mass spectra (MS) and compound purity data were acquired on a Waters ZQ LC/MS single quadrupole system equipped with electrospray ionization (ESI) source, UV 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 UV light, 5% ethanolic phosphomolybdic acid, Ninhydrin 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 [0339] DPP-IV 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.
[0340] 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.
[0341] 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:
O O CI O
R2 OR u~ R2 I NH POCK R2 I ~ N NaOH> R2 ~NH

R3 NH2 200 C R3 N~O R3 NCI R3 NCI
H
O O
R22X R2 I N. R22 Ras R~ I N. R22 R3 NCI R3 N~R23 [0342] By varying the R2, R3, 822X, and R23 groups shown above in this example, a wide variety of different DPP-IV inhibitors according to the present invention may be synthesized.

Representative Scheme for the Synthesis of Compounds According to Formula I where Q=502:
Scheme 2:
,R
R2 OSO I R2~ R2 OSO HR22 SnCl2 R2 OS NHR22 ~ R2 OS N 22 R' -NO R' -NO R' 'NH R ~N~S

O O O~ s0 O"O
~i R2 I S.N.R22 Oxidatio~ R2 I S~N.R22 R23 R2 I S~N.R22 R ~N~S~ R ~N~S~ R ~N~R
3 3 ~ i 3 23 O
[0343] By varying the R2, R3, R22NH2, and R23 groups shown above in this example, a wide variety of different DPP-IV inhibitors according to the present invention may be synthesized.
[0344] In each of the above reaction schemes, the various substituents may be selected from among the various substituents otherwise taught herein.
[0345] 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 [0346] The present invention is further exemplified, but not limited by, the following examples that describe the synthesis of particular compounds according to the invention.
Example 1: 2-[2-(3-(R)-Amino-piperidin-1-yl)-5-bromo-6-oxo-6H pyrimidin-1-ylmethyl]-benzonitrile, TFA salt O ~ CN
Br N
N~N ,,.NH2 Example 1A: 5-Bromo-2-chloro-3H pyrimidin-4-one O
Br NH
NCI
[0347] 5-Bromo-2,4-dichloro-pyrimidine (5.0 g, 22 mmol) was stirred in THF ( 10 mL) with 1N NaOH (30 mL) at r.t. for 3h. The solution was made slightly acidic with 1N HCl and was extracted with CHCl3. Organics were dried (MgS04) and concentrated in vacuo.
Precipitation from 20% CHC13/hexanes and collection by filtration gave 2.92 g (64%) of 5-bromo-2-chloro-3H-pyrimidin-4-one as a white solid. 1H NMR (400 MHz, DMSO-d6):

13.33 (br s, 1H), 8.35 (s, 1H). MS (ES) [m+H] calc'd for C4H2NaOBrCl, 209, 211, 213;
found 209, 211, 213.
Example 1B: 2-(5-Bromo-2-chloro-6-oxo-6H-pyrimidin-1-ylmethyl)-benzonitrile O ~ CN
Br N
N ~CI
[0348] 5-Bromo-2-chloro-3H pyrimidin-4-one (1.88 g, 9.0 mmol) was stirred in DME
(25 mL)/ DMF (5 mL) under nitrogen at 0 °C. Sodium hydride (95%, 238 mg, 9.4 mmol) was added in portions. After 10 min, lithium bromide ( 1.56 g, 17.9 mmol) was added and the reaction stirred for 15 min at r.t. a-Bromo-o-tolunitrile (3.5 g, 17.9 mmol) was added, and the reaction stirred at 65 °C for 8 h. The solution was diluted with EtOAc, washed with brine, dried (MgSO4) and concentrated in vacuo. Purification by silica gel chromatography (1:1:1 EtOAc/hexanes/CHC13) gave 997 mg (34%) of 2-(5-bromo-2-chloro-6-oxo-6H
pyrimidin-1-ylmethyl)-benzonitrile as a white solid. 1H NMR (400 MHz, CDCl3): 8 8.11 (s,1H), 7.73 (dd, 1H, J = 7.6, 1.2 Hz), 7.58 (dt, 1H, J = 7.6, 1.2 Hz), 7.45 (t, 1H, J = 7.6 Hz), 7.16 (d, 1H, J =
7.6 Hz), 5.69 (s, 2H). MS (ES) [m+H] calc'd for C12H~N30BrCl, 324, 326, 328;
found 324, 326, 328.
[0349] Also obtained from the reaction were impure fractions of the less polar O-alkylated isomer and 1.06 g (36%) of the more polar N3-alkylated isomer.

Example 1: 2-[2-(3-(R)-Amino-piperidin-1-yl)-5-bromo-6-oxo-6H pyrimidin-1-ylmethyl]-benzonitrile, TFA salt O ~ CN
Br N
N~N ,.NH2 [0350] 2-(5-Bromo-2-chloro-6-oxo-6H pyrimidin-1-ylmethyl)-benzonitrile (189 mg, 0.58 mmol), (R)-3-amino-piperidine, dihydrochloride ( 128 mg, 0.74 mmol) and sodium bicarbonate ( 195 mg, 2.32 mmol) were stirred in ethanol (5 mL) at 60 °C for 90 min. The reaction was diluted with EtOAc, washed with water and brine, dried (MgS04), and concentrated in vacuo. Purification by silica gel chromatography (5% MeOH/
CHC13) gave 139 mg (62°70) of the title compound as a clear oil. This was converted to the solid TFA salt by subjection to TFA in CH2C12 and concentration in vacuo. 1H NMR (400 MHz, DMSO-d6):
~ 8.18 (s, 1H), 7.98 (br s, 3H), 7.82 (d, 1H, J = 6.8 Hz), 7.64 (dt, 1H, J =
7.6, 1.2 Hz), 7.47 (t, 1H, J = 7.2 Hz), 7.27 (d, 1H, J = 7.6 Hz), 5.29 (AB q, 2H, J = 42.8, 15.2 Hz), 3.52-3.57 (m, 1H), 3.30-3.39 (m, 1H), 3.15-3.24 (m, 1H), 2.88-3.05 (m, 2H), 1.90-1.99 (m,1H), 1.75-1.83 (m, 1H), 1.49-1.63 (m, 2H). MS (ES) [m+H] calc'd for C1~H18NSOBr, 388, 390;
found 388, 390.
Example 2: 2-[2-(3-(R)-Amino-piperidin-1-yl)-6-oxo-5-phenyl-6H-pyrimidin-1-ylmethyl]-benzonitrile, TFA salt o ~ cN
N
N~N ,,NH2 [0351] 2-[2-(3-(R)-Amino-piperidin-1-yl)-5-bromo-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile (70 mg, 0.18 mmol), phenylboronic acid (33 mg, 0.27 mmol), and sodium carbonate (57 mg, 0.54 mmol) were stirred in DME (2 rnL)/ H20 (0.3 mL) in a flask purged with nitrogen. Tetrakis(triphenylphosphine)palladium (31 mg, 0.03 mmol) was added, and the reaction stirred at 88 °C for 2 h. The reaction was diluted with EtOAc, washed with brine, dried (MgSO4), and concentrated ifz vacuo. Purification by silica gel chromatography (5%
MeOH/CHCl3) followed by conversion to the TFA salt with TFA/CHZCl2 gave 76 mg (85%) of the title compound as a white solid. 1H NMR (400 MHz, DMSO-d6): b 8.08 (s, 1H), 7.93 (br s, 3H), 7.82 (d, 1H, J = 7.2 Hz), 7.63 (dt, 1H, J = 7.6, 1.2 Hz), 7.56 (d, 2H, J = 8.4 Hz), 7.45 (t, 1H, J = 7.6 Hz), 7.24-7.37 (m, 4H), 5.34 (AB q, 2H, J = 40.0, 15.2 Hz), 3.53-3.59 (m, 1H), 3.36-3.45 (m, 1H), 3.18-3.25 (m, 1H), 2.80-3.08 (m, 2H), 1.92-2.00 (m, 1H), 1.79-1.85 (m, 1H), 1.51-1.67 (m, 2H). MS (ES) [m+H] calc'd for C23H23N$~, 386; found 386.
Example 3: 2-[2-(3-(R)-Amino-piperidin-1-yl)-5-furan-3-yl-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile, TFA salt O I O ~ CN
~N~N ,.NH2 [0352] The title compound was prepared in 64% yield from 3-furanylboronic acid according to the general procedure outlined for Example 2. 1H NMR (400 MHz, DMSO-d6):
8 8.29 (s, 1H), 8.16 (s, 1H), 8.05 (br s, 3H), 7.82 (d, 1H, J = 7.2 Hz), 7.68 (s, 1H), 7.62 (t, 1H, J = 7.6 Hz), 7.44 (t, 1H, J = 7.6 Hz), 7.19 (d, 1H, J = 7.6 Hz), 7.01 (s, 1H), 5.36 (AB q, 2H, J
= 41.6, 15.2 Hz), 3.49-3.56 (m,1H), 3.36-3.45 (m,1H), 3.13-3.21 (m,1H), 3.01-3.09 (m,1H), 2.86-2.93 (m,1H),1.92-1.99 (m,1H),1.76-1.84 (m,1H),1.50-1.65 (m, 2H). MS (ES) [m+H]
calc'd for C21H21N5~2~ 376; found 376.
Example 4: 2-[2-(3-(R)-Amino-piperidin-1-yl)-5-(2-fluoro-phenyl)-6-oxo-6H
pyrimidin-1-ylmethyl]-benzonitrile, TFA salt ~ o ~ CN
N
N~N ,,.NH2 [0353] The title compound was prepared in 48% yield from 2-fluorophenylboronic acid according to the general procedure outlined for Example 2. 1H NMR (400 MHz, DMSO-d6):

b 7.92 (br s, 3H), 7.85 (s, 1H), 7.71 (d,1H, J = 7.2 Hz), 7.54 (t, 1H, J = 7.6 Hz), 7.35 (t, 1H, J
= 7.6 Hz), 7.06-7.27 (m, 5H), 5.23 (AB q, 2H, J = 40.4, 15.2 Hz), 3.47-3.54 (m, 1H), 3.36-3.45 (m, 1H), 3.12-3.20 (m, 1H), 2.93-3.02 (m, 1H), 2.83-2.90 (m, 1H), 1.22-1.90 (m, 1H), 1.70-1.78 (m, 1H), 1.42-1.58 (m, 2H). MS (ES) [m+H] calc'd for C23H22NSOF, 404; found 404.
Example 5: 2-{2-(3-(R)-Amino-piperidin-1-yl)-5-[2-traps-(4-fluoro-phenyl)-vinyl]-6-oxo-6H-pyrimidin-lylmethyl}-benzonitrile, TFA salt O ~ CN
/ ~ N
N~N ,,.NH2 [0354] The title compound was prepared in 62% yield from trafis-2-(4-fluorophenylvinylboronic acid according to the general procedure outlined for Example 2. 1H
NMR (400 MHz, DMSO-d6): S 8.09 (s, 1H), 8.01 (br s, 3H), 7.82 (d, 1H, J = 7.2 Hz), 7.63 (t, 1H, J = 7.6 Hz), 7.43-7.54 (m,~4H), 7.22 (d, 1H, J = 7.6 Hz), 7.14 (t, 2H, J =
8.8 Hz), 6.92 (d, 1H, J =16.4 Hz), 5.33 (AB q, 2H, J = 38.4, 15.2 Hz), 3.51-3.59 (m, 1H), 3.36-3.45 (m, 1H), 3.15-3 .23 (m, 1 H), 3 .O 1-3 .09 (m, 1 H), 2. 89-2. 96 (m, 1 H), 1.92-1.99 (m, 1 H), 1.77-1. 84 (m, 1H), 1.51-1.65 (m, 2H). MS (ES) [m+H] calc'd for CZSHaaNsOF, 430; found 430.
Example 6: 2-[2-(3-(R)-Amino-piperidin-1-yl)-5-(2-methoxy-phenyl)-6-oxo-6H
pyrimidin-1-ylmethyl]-benzonitrile, TFA salt O ~ CN_ N
O~ I N~N ,,.NH2 [0355] The title compound was prepared in 42% yield from 2-methoxyphenylboronic acid according to the general procedure outlined for Example 2. 1H NMR (400 MHz, DMSO-d6): 8 8.02 (br s, 3H), 7.80-7.84 (m, 2H), 7.66 (t, 1H, J = 7.6 Hz), 7.46 (t, 1H, J = 7.6 Hz), 7.12-7.31 (m, 3H), 6.99 (d, 1H, J = 8.4 Hz), 6.91 (t, 1H, J = 7.6 Hz), 5.33 (AB q, 2H, J =

43.2, 15.2 Hz), 3.58 (s, 3H), 3.49-3.56 (m, 1H), 3.36-3.45 (m, 1H), 3.15-3.21 (m, 1H), 3.01-3.09 (m, 1H), 2.89-2.96 (m, 1H), 1.92-1.99 (m, 1H), 1.75-1.84 (m, 1H), 1.50-1.65 (m, 2H).
MS (ES) [m+H] calc'd for C24H25NSO2, 416; found 416.
Example 7: 2-[2-(3-(R)-Amino-piperidin-1-yl)-5-ethynyl-6-oxo-6H pyrimidin-1-ylmethyl]-benzonitrile \ O ~ CN
~N
N~N ,,,NH2 Example 7A: 2-[2-(3-(R)-Amino-piperidin-1-yl)-5-trimethylsilylethynyl-6-oxo-6H pyrimidin-1-ylmethyl]-benzonitrile ~Si~\ O ~ CN
~N
N~N .~NH~
[0356] 2-[2-(3-(R)-Amino-piperidin-1-yl)-5-bromo-6-oxo-6H-pyrinudin-1-ylmethyl]-benzonitrile (189 mg, 0.49 mmol), trimethylsilylacetylene (103 ~,L, 0.73 mmol), triphenylphosphine (4 mg, 0.02 mmol), and triethylamine (102 ~,~L, 0.73 mmol) were stirred in THF (4 mL) in a flask purged with nitrogen.
Dichlorobis(triphenylphosphine)palladium(II]
( 17 mg, 0.024 mmol) was added, and after 10 min copper iodide (2 mg) was added. The reaction stirred for 18 h at r.t. The solution was diluted with EtOAc, washed with brine, dried (MgS04), and concentrated i~z vacuo. Purification by silica gel chromatography (5%
MeOHlCHCI3) gave 168 mg (85%) of the title compound as a clear oil. MS (ES) [m+H]
calc'd for C22H2~N50S1, 406; found 406.

Example 7: 2-[2-(3-(R)-Amino-piperidin-1-yl)-5-ethynyl-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile O ~ CN
~N
N~N ,,.NH2 [0357] Deprotection of 2-[2-(3-(R)-amino-piperidin-1-yl)-5-trimethylsilylethynyl-6-oxo-6H pyrimidin-1-ylmethyl]-benzonitrile (168 mg) was carried out by stirring the intermediate in THF (2 mL) with TBAF (1 N in THF, 0.8 mL, 0.8 mmol) for 1 h. The reaction was purified by silica gel chromatography (4 to 8% MeOH/CHC13) to give 98 mg (71 %) of the title compound as a faintly yellow oil/foam. 1H NMR (400 MHz, DMSO-dg): 8 8.01 (s,1H), 7.81 (dd,1H, J = 7.6,1.2 Hz), 7.63 (dt, 1H, J = 7.6, 1.2 Hz), 7.45 (t, 1H, J = 7.6 Hz), 7.19 (d, 1H, J
= 7.6 Hz), 5.22 (AB q, 2H, J =16.4, 15.2 Hz), 4.13 (s,1H), 3.25-3.46 (m, 2H), 2.82-2.89 (m, 1H), 2.70-2.77 (m, 1H), 2.58-2.63 (m, 1H), 1.74-1.82 (m, 1H), 1.63-1.71 (m, 1H), 1.45-1.57 (m, 1H), 1.11-1.19 (m, 1H). MS (ES) [m+H] calc'd for C19H19NSO, 334; found 334.
Example 8: 2-[2-(3-(R)-Amino-piperidin-1-yl)-5-bromo-4-methyl-6-oxo-6H
pyrimidin-1-ylmethyl]-benzonitrile, TFA salt O ~ CN
Br N
N~N ,.NH2 Example 8A: 5-Bromo-2-chloro-6-methyl-3H pyrimidin-4-one O
Br NH
NI 'CI
[0358] The title compound was prepared in 63% yield from 5-Bromo-6-methyl-1H-pyrimidine-2,4-dione utilizing a method analogous to the preparation of 2-chloro-5,6-dimethyl-3H-pyrimidin-4-one (see Lee, et al., WO 9605177). 1H NMR (400 MHz, DMSO-d6): 8 2.38 (s, 3H). MS (ES) [m+H] calc' d for CSH4N20BrCl, 223, 225, 227;
found 223, 225, 227.

Example 8B: 2-(5-Bromo-2-chloro-4-methyl-6-oxo-6H-pyrimidin-1-ylmethyl)-benzonitrile O ~ CN
Br N
NCI
[0359] The title compound was prepared in 52% yield from 5-bromo-2-chloro-6-methyl-3H pyrimidin-4-one according to the general procedure outlined for Example 1B.

(400 MHz, DMSO-d6): b 7.91 (dd, 1H, J = 7.6,1.2 Hz), 7.67 (dt,1H, J = 7.6, 1.2 Hz), 7.52 (t, 1H, J = 7.6 Hz), 7.26 (d, 1H, J = 7.6 Hz), 5.50 (s, 2H), 2.41 (s, 3H). MS (ES) [m+H] calc'd for C13H9N30BrCl, 338, 340, 342; found 338, 340, 342.
Example 8: 2-[2-(3-(R)-Amino-piperidin-1-yl)-5-bromo-4-methyl-6-oxo-6H
pyrimidin-1-ylmethyl]-benzonitrile, TFA salt O ~ CN
Br N
N~N ,,NH2 [0360] The title compound was prepared in 52% yield from 2-(5-bromo-2-chloro-4-methyl-6-oxo-6H-pyrimidin-1-ylmethyl)-benzonitrile according to the general procedure outlined for Example 1. 1H NMR (400 MHz, DMSO-d6): ~ 8.03 (br s, 3H), 7.81 (d, 1H, J =
7.2 Hz), 7.63 (t,1H, J = 7.6 Hz), 7.46 (t,1H, J = 7.2 Hz), 7.23 (d,1H, J = 7.6 Hz), 5.27 (AB q, 2H, J = 42.0, 15.2 Hz), 3.50-3.55 (m, 1H), 3.31-3.40 (m,1H), 3.13-3.20 (m,1H), 3.00-3.06 (s, 1H), 2.85-2.92 (m, 1H), 2.33 (s, 3H), 1.90-1.99 (m, 1H), 1.75-1.83 (m, 1H), 1.49-1.63 (m, 2H). MS (ES) [m+H] calc'd for C18H2oN50Br, 402, 404; found 402, 404.

Example 9: 2-[2-(3-(R)-Amino-piperidin-1-yl)-4-methyl-6-oxo-5-phenyl-6H
pyrimidin-1-ylmethyl]-benzonitrile, TFA salt o ~ cN
N
N~N ,,.NH2 [0361] The title compound was prepared in 64% yield from phenylboronic acid and 2-[2-(3-(R)-amino-piperidin-1-yl)-5-bromo-4-methyl-6-oxo-6H pyrimidin-1-ylmethyl]-benzonitrile according to the general procedure outlined for Example 2. 1H NMR (400 MHz, DMSO-d6):
8 8.09 (br s, 3H), 7.81 (d, 1H, J = 7.6 Hz), 7.64 (dt, 1H, J = 7.6, 1.2 Hz), 7.44 (t, 1H, J = 7.6 Hz), 7.14-7.37 (m, 6H), 5.30 (AB q, 2H, J = 43.2, 15.2 Hz), 3.51-3.57 (m,1H), 3.33-3.42 (m, 1H), 3.12-3.20 (m, 1H), 3.10-3.19 (m, 1H), 2.85-2.93 (m, 1H), 2.08 (s, 3H), 1.92-2.00 (m, 1H), 1.79-1.85 (m, 1H), 1.51-1.67 (m, 2H). MS (ES) [m+H] calc'd for C24HzsNsO, 400;
found 400.
Example 10: 2-[2-(3-(R)-Amino-piperidin-1-yl)-5-furan-3-yl-4-methyl-6-oxo-6H
pyrimidin-1-ylmethyl]-benzonitrile, TFA salt O ~ O ~ CN
~N
~N~N ,.NH2 [0362] The title compound was prepared in 64% yield from 3-furanylboronic acid and 2-[2-(3-(R)-amino-piperidin-1-yl)-5-bromo-4-methyl-6-oxo-6H pyrimidin-1-ylmethyl]-benzonitrile according to the general procedure outlined for Example 2. 1H NMR
(400 MHz, DMSO-d6): S 7.99 (br s, 3H), 7.79-7.84 (m, 2H), 7.59-7.67 (m, 2H), 7.44 (t,1H, J = 7.6 Hz), 7.17 (d, 1H, J = 7.6 Hz), 6.62 (d, 1H, J = 1.2 Hz), 5.29 (AB q, 2H, J = 40.4, 15.2 Hz), 3.49-3.56 (m, 1H), 3.33-3.41 (m, 1H), 3.01-3.17 (m, 2H), 2.82-2.89 (m, 1H), 2.30 (m, 3H), 1.92-1.99 (m,1H),1.76-1.84 (m, 1H),1.50-1.65 (m, 2H). MS (ES) [m+H] calc'd for C2aH23N5Oa, 390; found 390.

Example 11: 2-[2-(3-(R)-Amino-piperidin-1-yl)-4-methyl-6-oxo-5-traits-styryl-6H pyrimidin-1-ylmethyl]-benzonitrile, TFA salt O
~N
N~N
CN
,,.NH2 [0363] The title compound was prepared in 64% yield from traps-2-phenylvinylboronic acid and 2-[2-(3-(R)-amino-piperidin-1-yl)-5-bromo-4-methyl-6-oxo-6H-pyrimidin-ylmethyl]-benzonitrile according to the general procedure outlined for Example 2. 1H NMR
(400 MHz, DMSO-dg): ~ 8.08 (br s, 3H), 7.83 (d, 1H, J = 7.2 Hz), 7.61-7.66 (m, 2H), 7.42-7.50 (m, 3H), 7.31 (t, 2H, J = 7.6 Hz), 7.17-7.22 (m, 2H), 7.00 (d, 1H, J
=16.4 Hz), 5.30 (AB
q, 2H, J = 38.4,15.2 Hz), 3.51-3.58 (m,1H), 3.34-3.42 (m, 1H), 3.03-3.19 (m, 2H), 2.85-2.93 (m, 1H), 2.43 (m, 3H), 1.92-1.99 (m, 1H), 1.76-1.84 (m, 1H), 1.50-1.65 (m, 2H). MS (ES) [m+H] calc'd for C26H2~N50, 426; found 426.
Example 12: 2-[2-(3-(R)-Amino-piperidin-1-yl)-4,5-dimethyl-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile, TFA salt O ~ CN
~N
N~N .~NH2 Example 12A: 2-(2-Chloro-4,5-dimethyl-6-oxo-6H-pyrimidin-1-ylmethyl)-benzonitrile O ~ CN
~N
N ~CI
[0364] The title compound was prepared in 64% yield from 2-chloro-5,6-dimethyl-pyrimidin-4-one (see Lee, et al., WO 9605177) according to the procedure for Example 1B.
1H NMR (400 MHz, CDC13): 8 7.71 (d,1H, J = 7.6 Hz), 7.55 (dt,1H, J = 7.6,1.2 Hz), 7.41 (t, 1H, J = 7.6 Hz), 7.12 (d, 1H, J = 7.6 Hz), 5.63 (s, 2H), 2.32 (s, 3H), 2.10 (s, 3H). MS (ES) [m+H] calc'd for Cl4HiaNsOCI, 274, 276; found 274, 276.
[0365] Also obtained from the reaction was 30% yield of the less polar O-alkylated isomer.
Example 12: 2-[2-(3-(R)-Amino-piperidin-1-yl)-4,5-dimethyl-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile, TFA salt O ~ CN
~N
N~N ,,.NH2 [0366] The title compound was prepared in 74% yield from 2-(2-chloro-4,5-dimethyl-6-oxo-6H-pyrimidin-lylmethyl)-benzonitrile according to the general procedure outlined for Example 1. 1H NMR (400 MHz, DMSO-d6): b 7.95 (br s, 3H), 7.81 (d, 1H, J = 7.6 Hz), 7.61 (dt,1H, J = 7.6,1.2 Hz), 7.44 (t, 1H, J = 7.6 Hz), 7.09 (d, 1H, J = 7.6 Hz), 5.26 (AB q, 2H, J =
45.2, 15.2 Hz), 3.29-3.42 (m, 2H), 2.91-3.08 (m, 2H), 2.71-2.80 (m,1H), 2.19 (m, 3H), 1.89-1.96 (m, 1H),1.83 (s, 3H), 1.72-1.81 (m, 1H),1.46-1.61 (m, 2H). MS (ES) [m+H]
calc'd for C19H23N50~ 338; found 338.
Example 13: 2-[2-(3-(R)-Amino-piperidin-1-yl)-5-ethyl-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile, TFA salt O ~ CN
~N
~N~N ,,.NH2 Example 13A: 2-Chloro-5-ethyl-3H pyrimidin-4-one O
N CI
[0367] The title compound was prepared in 67% yield from 5-Ethyl-1H-pyrimidine-2,4-dione utilizing a method analogous to the preparation of 2-chloro-5,6-dimethyl-3H pyrimidin-4-one (see Lee, et al., WO 9605177). 1H NMR (400 MHz, DMSO-d6): 8 13.21 (br s, 1H), 7.90 (br s, 1H), 2.39 (q, 2H, J = 7.6 Hz), 1.08 (t, 3H, J = 7.6 Hz). MS (ES) [m+H] calc'd for C6H~N20C1, 159, 161; found 159, 161.
Example 13B: 2-(2-Chloro-5-ethyl-6-oxo-6H pyrimidin-1-ylmethyl)-benzonitrile O ~ CN
~N
~N~CI
[0368] The title compound was prepared in 20% yield from 2-chloro-5-ethyl-3H
pyrimidin-4-one according to the procedure for Example 1B. 1H NMR (400 MHz, CDC13): 8 7.70 (dd, 1H, J = 7.6, 1.2 Hz), 7.65 (d, 1H, J = 1.2 Hz), 7.55 (dt, 1H, J =
7.6, 1.2 Hz), 7.40 (t, 1H, J = 7.6 Hz), 7.10 (d,1H, J = 7.6 Hz), 5.63 (s, 2H), 2.51 (dq, 2H, J = 7.6, 1.2 Hz),1.20 (dt, 3H, J = 7.6, 1.2 Hz). MS (ES) [m+H] calc'd for C14H12N3OCl 274, 276; found 274, 276.
[0369] Also obtained from the reaction were impure fractions of the less polar O-alkylated isomer, and 38% yield of the more polar N3-alkylated isomer.
Example 13: 2-[2-(3-(R)-Amino-piperidin-1-yl)-5-ethyl-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile, TFA salt O ~ CN
_N
~N~N ,.NH2 [0370] The title compound was prepared in 54% yield from 2-(2-chloro-5-ethyl-6-oxo-6H pyrimidin-1-ylmethyl)-benzonitrile according to the general procedure outlined for Example 1. 1H NMR (400 MHz, DMSO-d6): ~ 8.05 (br s, 3H), 7.81 (dd,1H, J =
7.6,1.2 Hz), 7.69 (s, 1H), 7.61 (dt, 1H, J = 7.6, 1.2 Hz), 7.44 (t, 1H, J = 7.6 Hz), 7.10 (d, 1H, J = 7.6 Hz), 5.26 (AB q, 2H, J = 48.0,15.2 Hz), 3.29-3.42 (m, 2H), 2.91-3.08 (m, 2H), 2.75-2.84 (m,1H), 2.25 (q, 2H, J = 7.2 Hz),1.89-1.96 (m,1H),1.72-1.81 (m,1H),1.46-1.61 (m, 2H),1.00 (t, 3H, J = 7.2 Hz). MS (ES) [m+H] calc'd for C19H23Ns0, 338; found 338.

Example 14: 2-[2-(3-(R)-Amino-piperidin-1-yl)-5-iodo-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile O ~ CN
I N
N~N ,,.NH2 [0371] The title compound was prepared from 2,4-dichloro-5-iodopyrimidine [

0] according to the general procedure outlined for Example 1. 1H NMR (400 MHz, MeOD):
8 8.27 (s, 1H), 7.72 (d, J = 7.6 Hz, 1H), 7.63 (m, 1H), 7.46 (t, J = 7.6 Hz,1H), 7.38 (d, J = 8.0 Hz, 1H), 5.34-5.46 (AB q, J =15.2 Hz, 2H) 3.65-3.72 (m, 1H), 3.49-3.58 (m, 1H), 3.28-3.34 (m, 1H), 3.14-3.22 (m, 1H), 3.05-3.09 (m, 1H), 2.07-2.17 (m, 1H), 1.60-1.90 (m, 3H). MS
(ES) [M+H]: 436.
Example 15: 2-[2-(3-(R)-Amino-piperidin-1-yl)-5-chloro-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile, TFA salt O ~ CN
CI N
N~N ,,.NH2 Example 15A: 2,5-Dichloro-3H-pyrimidin-4-one O
CI NH
N ~CI
[0372] The title compound was prepared in 76% yield from 2,4,5-trichloropyrimidine according to the procedure for Example 1A. 1H NMR (400 MHz, DMSO-d6): 813.40 (br s, 1H), 8.24 (s, 1H). MS (ES) [m+H] calc'd for CøH2NZOC12, 165, 167; found 165, 167.

Example 15B: 2-(2,5-Dichloro-6-oxo-6H pyrimidin-1-ylmethyl)-benzonitrile O ~ CN
CI N
NCI
[0373] The title compound was prepared in 40% yield from 2,5-dichloro-3H
pyrimidin-4-one according to the procedure for Example 1B. 1H NMR (400 MHz, CDC13): ~ 8.00 (s,1H), 7.73 (dd, 1H, J = 7.6, 1.2 Hz), 7.59 (dt, 1H, J = 7.6, 1.2 Hz), 7.45 (t, 1H, J
= 7.6 Hz), 7.16 (d, 1H, J = 7.6 Hz), 5.69 (s, 2H). MS (ES) [m+H] calc'd for CI~H~N30C12, 280, 282;
found 280, 282.
[0374] Also obtained from the reaction were impure fractions of the less polar O-alkylated isomer, and 41 % yield of the more polar N3-alkylated isomer.
Example 15: 2-[2-(3-(R)-Amino-piperidin-1-yl)-5-chloro-6-oxo-6H pyrimidin-1-ylmethyl]-benzonitrile, TFA salt O ~ CN
CI N
N~N ,.NH2 [0375] The title compound was prepared in 55% yield from 2-(2,5-dichloro-6-oxo-pyrimidin-1-ylmethyl)-benzonitrile according to the general procedure outlined for Example 1. 1H NMR (400 MHz, DMSO-d6): S 8.30 (s, 1H), 8.09 (d, 1H, J = 6.8 Hz), 7.69 (br s, 3H), 7.64 (dt, 1H, J = 7.6, 1.2 Hz), 7.46 (t, 1H, J = 7.6 Hz), 7.27 (d, 1H, J = 7.6 Hz), 5.29 (AB q, 2H, J = 48.0, 15.2 Hz), 3.49-3.58 (m, 1H), 3.16-3.36 (m, 2H), 2.86-3.02 (m, 2H), 1.89-1.96 (m, 1H), 1.72-1.81 (m, 1H), 1.43-1.64 (m, 2H). MS (ES) [m+H] calc'd for C1~H18NSOCl, 344, 346; found 344, 346.

Example 16: (R)-2-((2-(3-aminopiperidin-1-yl)-6-oxo-5-(pyrrolidin-1-yl)pyrimidin-1 (6H)-yl)methyl)benzonitrile I
O / ~N
N N
I N~N ,,.NH2 [0376] The title compound was prepared by heating Example 1 with pyrrolidine at 150 °C using a microwave. The crude product was purified by HPLC and was isolated as the TFA salt. 1H NMR (400 MHz, MeOH-d4): ~ 7.71 (d, J = 7.6 Hz,1H), 7.60 (t, J =
7.6, 8.0 Hz, 1H), 7.43 (t, J = 8.0, 7.2 Hz, 1H), 7.29 (d, J = 7.6 Hz,1H), 5.30 (AB q, J
=15.6, 24.0 Hz, 2H), 3.71-2.90 (m, 8H), 2.28-1.57 (m, 9H). MS (ES) [m+H] calc'd for C~,1H26IV6O, 379, 22; found 379.22.
Example 17: (R)-2-((2-(3-aminopiperidin-1-yl)-5-fluoro-6-oxopyrimidin-1 (6H)-yl)methyl)-5-fluorobenzonitrile F
I
O / ~N
F
N
N~N ,,.NH2 Example 17A: 2-bromomethyl-5-fluoro-benzonitrile F F
NBS
( / --CN AIBN / CN
Br '17A
[0377] A solution of 1 (3.4 g, 25.2 mmol), NBS (4.63 g, 26 rnmol) and 100 mg AIBN
was refluxed for 2 hours under nitrogen. After cooling to room temperature, the solvent was removed and the residue was purified by column chromatography. 1H NMR (400 MHz, CDCl3): 7.51-7.59 (m, 1H), 7.26-7.39 (m, 2H), 4.61 (s, 2H).
Example 17B: 2-((5-fluoro-2-chloro-6-oxopyrimidin-1(6H)-yl)methyl)-5-fluorobenzonitrile F
O / ~N
F N
NCI
[0378] The title compound was prepared from 17A and 2-chloro-5-fluoropyrimidin-4(3H)-one according to the procedure outlined for Example 1B. 1H NMR (400 MHz, CDCl3): 7.96 (s, 1H), 7.64 (d, J = 8.0 Hz, 1H), 7.32-7.48 (m, 2H), 5.63 (s, 1H). MS (ES) [m+H] calc'd for C12H6C1FaN30, 282.02; found 282.02.
Example 17: (R)-2-((2-(3-aminopiperidin-1-yl)-5-fluoro-6-oxopyrimidin-1 (6H)-yl)methyl)-5-fluorobenzonitrile F
O / ~N
F N
N~N ,,.NH2 [0379] The title compound was prepared from 17B according to the general procedure outlined for Example 1 and was isolated as the TFA salt. 1H NMR (400 MHz, MeOH-d4): ~
7.85 (d, J = 4 Hz, 1H), 7.37-7.59 (m, 3H), 5.40 (AB q, J = 16.0, 28.0 Hz, 2H), 3.47-3.64 (m, 2H), 2.91-3.25 (m, 3H),1.57-1.91 (m, 4H). MS (ES) [m+H] calc'd for C1~H1~F2N50, 346.14;
found 346.14 Example 18: (R)-2-((2-(3-aminopiperidin-1-yl)-5-fluoro-6-oxopyrimidin-1 (6H)-yl)methyl)-4-fluorobenzonitrile F
I~
~N
F
N
N~N ,,.NH2 Example 18A: 4-fluoro-2-methyl-benzonitrile F F
I ~ Zn(~ F I ~ --~ I
gr Pd(PPh3)a CN AIBN CN
Br [0380] A mixture of ~ (3.5 g, 18.5 mmol) and CuCN (2 g, 22 mmol) in DMF was refluxed overnight. After cooling to room temperature, the reaction was diluted with water and extracted with hexane. The extract was dried over MgS04 and then the solvent was removed. The residue was purified by column chromatography. 1H NMR (400 MHz, CDC13): 7.60 (d, J = 5.6, 8.8 Hz, 1H), 6.96-7.06 (m, 2H), 2.55 (s, 3H).
Example 18B: 2-bromomethyl-4-fluoro-benzonitrile F
I
~CN
Br [0381] A solution of 18A (4.8 g, 25.4 mmol), NBS (4.5 g, 25.4 mmol) and 100 mg AIBN
was refluxed for 2 hours under nitrogen. After cooling to room temperature, the solvent was removed and the residue was purified by column chromatography. 1H NMR (400 MHz, CDCl3): 7.68 (d, J = 5.2, 8.4 Hz, 1H), 7.28 (d, J = 2.4, 8.8 Hz, 1H), 7.10-7.6 (m, 1H), 4.60 (s, 2H).

Example 18C: 2-((5-fluoro-2-chloro-6-oxopyrimidin-1(6H)-yl)methyl)-4-fluorobenzonitrile F
~N
F N
NCI
[0382] The title compound was prepared from 18B and 2-chloro-5-fluoropyrimidin-4(3H)-one according to the procedure outlined for Example 1B. 1H NMR (400 MHz, CDC13): 7.83 (d, J = 0.8 Hz, 1H), 7.73-7.78 (m, 1H), 7.13-7.20 (m,1H), 6.88 (dd, J = 2.4, 8.8 Hz, 1H), 5.66(s, 2H). MS (ES) [m+H] calc'd for C12H6C1FZN3O, 282.02; found 282.02.
Example 18: (R)-2-((2-(3-aminopiperidin-1-yl)-5-fluoro-6-oxopyrimidin-1(6H)-yl)methyl)-4-fluorobenzonitrile F
~N
F N
N~N ,.NH2 [0383] The title compound was prepared from 18C according to the general procedure outlined for Example 1 and was isolated as the TFA salt. 1H NMR (400 MHz, MeOH-d4): ~
7.75-7.88 (m, 2H), 7.18-7.27 (m, 2H), 5.42 (AB q, J =15.2, 31.6 Hz, 2H), 3.47-3.64 (m, 2H), 2.89-3.23 (m, 3H), 1.57-2.17 (m, 4H). MS (ES) [m+H] calc'd for CI~Hl~F2N50, 346.14;
found 346.14.
Example 19: (R)-2-((2-(3-aminopiperidin-1-yl)-5-chloro-6-oxopyrimidin-1 (6H)-yl)methyl)-4-fluorobenzonitrile F
~N
CI
~N
N~N ,,.NH2 [0384] The title compound was prepared from 15A according to the general procedure outlined for Example 1 and was isolated as the TFA salt. 1H NMR (400 MHz, MeOH-d4): S
8.01 (s, 1H), 7.76-7.82 (m, 1H), 7.20-7.27 (m, 2H), 5.39 (AB q, J =14.8, 24.0 Hz, 2H), 3.52-3.73 (m, 2H), 3.0-3.33 (m, 3H), 1.62-2.18 (m, 4H). MS (ES) [m+H] calc'd for CI~HmCIFNsO, 362.1; found 362.11 Example 20: (R)-2-((2-(3-aminopiperidin-1-yl)-5-bromo-6-oxopyrimidin-1 (6H)-yl)methyl)-4-fluorobenzonitrile F
I
~N
Br N
I N~N ,,.NH2 Example 20A: 2-((5-bromo-2-chloro-6-oxopyrimidin-1(6H)-yl)methyl)-4-fluorobenzonitrile F
I~
~N
Br N
NCI
[0385] The title compound was prepared from 1A according to the procedure outlined for Example 1S. 1H NMR (400 MHz, C1~C13): 8.15 (s, 1H), 7.71-7.77 (m, 1H), 7.12-7.19 (m, 1H), 6.88 (dd, J = 2.4, 4.8 Hz, 1H), 5.66 (s, 2H). MS (ES) [m+H] calc'd for C12H6BrC1FN3O, 341.94, 343.93; found 341.94, 343.93.
Example 20: (R)-2-((2-(3-aminopiperidin-1-yl)-5-bromo-6-oxopyrimidin-1 (6H)-yl)methyl)-4-fluorobenzonitrile F
I
~N
Br N
I N~N ,,.NH2 [0386] The title compound was prepared from 20A according to the general procedure outlined for Example 1 and was isolated as the TFA salt. 1H NMR (400 MHz, MeOH-d4): 8 8.13 (s, 1H), 7.76-7.82 (m, 1H), 7.20-7.27 (m, 2H), 5.39 (AB q, J =14.2, 26.6 Hz, 2H), 3.67-3.73 (m, 2H), 3.02-3.32 (m, 3H), 1.62-2.17 (m, 4H). MS (ES) [m+H] calc'd for C1~HI~BrFN50, 406.06, 408.06; found 406.06, 408.06 Example 21: (R)-2-((2-(3-amino-3-methylpiperidin-1-yl)-5-fluoro-6-oxopyrimidin-1 (6H)-yl)methyl)-4-fluorobenzonitrile F
I
O ~N
F
N
N~N ,,.NH2 [0387] The title compound was prepared from 18A and (R)-3-amino-3-methylpiperidine according to the general procedure outlined for Example 1 and was isolated as the TFA salt.
1H NMR (400 MHz, MeOH-dø): 8 7.86 (d, J = 2.0 Hz,1H), 7.77-7.82 (m, 2H), 7.17-7.27 (m, 2H), 5.46 (AB q, J =15.2, 98.4 Hz, 2H), 3.40-3.47 (m, l H), 3.14-3.22 (m, 2H), 2.92-3.01 (m, 1H), 1.71-1.94 (m, 4H), 1.57-2.17. MS (ES) [m+H] calc'd for C18H19F2N50, 360.16; found 360.16.
Example 22: 2-[2-(3-(R)-Amino-pyrimidin-1-yl)-5-bromo-6-oxo-6H pyrimidin-1-ylmethyl]-thiophene-3-carbonitrile CN
O
Br N
N~N ,,.NH2 Example 22A: 2-Bromomethyl-thiophene-3-carbonitrile CN
Br [0388] 2-Methyl-thiophene-3-carbonitrile [see Beaton, et al.; TetYahedrorc Lett. 39, 10, 1998, 1227-1230] (1.36 g, 11 mmol), N bromosuccinimide (2.56 g, 14.4 mmol), and a catalytic amount of benzoyl peroxide were stirred in benzene (30 mL) at 80 °C for 2h. The solution was diluted with EtOAc, washed with sat. NaHC03 and brine, dried (MgS04), and concentrated irc vacuo. Purification by silica gel chromatography (10% EtOAc /
hexanes) gave 1.03 g (46%) of the title compound as a white solid. 1H NMR (400 MHz, CDC13): 7.39 (d, 1H, J = 5.6 Hz), 7.17 (d, 1H, J = 5.6 Hz), 4.79 (s, 2H).
Example 22B: 2-(5-Bromo-2-chloro-6-oxo-6H pyrimidin-lylmethyl)-thiophene-3-carbonitrile CN
Br N
NCI
[0389] The title compound was prepared in 58% yield from 2-chloro-5-bromo-3H-pyrimidin-4-one (Example 1A) and 2-bromomethyl-thiophene-3-carbonitrile according to the general procedure outlined for Example 1B. 1H NMR (400 MHz, CDCl3): ~ 8.10 (s, 1H), 7.40 (d, 1H, J = 5.6 Hz), 7.21 (d, 1H, J = 5.6 Hz), 5.74 (s, 2H). MS (ES) [m+H] calc'd for CloH5N3OSBrCI, 330, 332; found 330, 332.
Example 22: 2-[2-(3-(R)-Amino-pyrimidin-1-yl)-5-bromo-6-oxo-6H pyrimidin-1-ylmethyl]-thiophene-3-carbonitrile CN
Br N
N~N .,.NH2 [0390] The title compound was prepared in 66% yield from 2-(5-bromo-2-chloro-6-oxo-6H pyrimidin-lylmethyl)-thiophene-3-carbonitrile according to the general procedure outlined for Example 1 and was isolated as the TFA salt. 1H NMR (400 MHz, DMSO-d6): 8 8.18 (s, 1H), 8.00 (br s, 3H), 7.69 (d, 1H, J = 5.6 Hz), 7.37 (d, 1H, J = 5.6 Hz), 5.45 (AB q, 2H, J =

28.0, 15.2 Hz), 3.26-3.52 (m, 3H), 2.80-2.95 (m, 2H), 1.92-2.02 (m, 1H), 1.62-1.81 (m, 2H), 1.41-1.51 (m, 1H). MS (ES) [m+H] calc'd for C1sH16IVsOSBr, 394, 396; found 394, 396.
Example 23: 2-[2-(3-(R)-Amino-piperidin-1-yl)-5-(3,3-dimethyl-but-1-ynyl)-6-oxo-6H pyrimidin-1-ylmethyl]-benzonitrile O ~ CN
~N
N~N ,.NH2 [0391] The title compound was prepared in 51 % yield using 3,3-dimethyl-1-butyne in the general procedure outlined for Example 7a. 1H NMR (400 MHz, DMSO-d6): 8 7.89 (s,1H), 7.82 (d, 1H, 7.2 Hz), 7.54 (t, 1H, J = 7.6 Hz), 7.45 (t, 1H, J = 7.2 Hz), 7.15 (d, 1H, J = 7.6 Hz), 5.22 (s, 2H), 3.20-3.40 (m, 2H), 2.71-2.76 (m, 2H), 2.53-2.59 (m, 1H), 1.46-1.82 (m, 3H),1.20 (s, 9H),1.10-1.19 (m,1H). MS (ES) [m+H] calc'd for C23H2~N50, 390;
found 390.
Example 24: 2-[2-(3-(R)-Amino-piperidin-1-yl)-5-prop-1-ynyl-6-oxo-6H
pyrimidin-1-ylmethyl]-benzonitrile O ~ CN
~N
N~N ,,.NH2 [0392] 2-[2-(3-(R)-Amino-piperidin-1-yl)-5-bromo-6-oxo-6H pyrimidin-1-ylmethyl]-benzonitrile (Example 1) (120 mg, 0.31 mmol) and tributyl(1-propynyl)tin (140 ~,I,, 0.46 mrnol) were stirred in dioxane (5 mL) in a flask purged with nitrogen.
Tetrakis(triphenylphosphine)palladium(0) (54 mg, 0.046 mmol) was added, and the solution stirred at 96 °C for 40 h. Concentration in vacuo and purification by silica gel chromatography ( 16% MeOH l CHC13) gave 82 mg (76%) of the title compound as a clear oil.
1H NMR (400 MHz, MeOD-d4): 8 7.82 (s,1H), 7.64 (d,1H, 7.6 Hz), 7.53 (t,1H, J =
7.6 Hz), 7.36 (t, 1H, J = 7.6 Hz), 7.16 (d, 1H, J = 7.6 Hz), 5.30 (s, 2H), 3.40-3.48 (m, 1H), 3.21-3.30 (m,1H), 2.78-2.89 (m, 2H), 2.58-2.66 (m,1H),1.91 (s, 3H),1.85-1.93 (m,1H),1.52-1.74 (m, 2H), 1.18-1.27 (m, 1H). MS (ES) [m+H] calc'd for CZOH21N5O, 348; found 348.
Example 25: 2-[2-(3-(R)-Amino-piperidin-1-yl)-5-(3-hydroxy-prop-1-ynyl)-6-oxo-6H pyrimidin-1-ylmethyl]-benzonitrile OH
O ~ CN
~N
N~N ,,.NH2 [0393] The title compound was prepared in 66°Io yield using propargyl alcohol in the general procedure outlined for Example 7a. 1H NMR (400 MHz, MeOD-d4): 8 7.90 (s, 1H), 7.63 (d, 1H, 7.6 Hz), 7.52 (t, 1H, J = 7.6 Hz), 7.36 (t, 1H, J = 7.6 Hz), 7.19 (d, 1H, J = 7.6 Hz), 5.28 (s, 2H), 4.24 (s, 2H), 3.48-3.54 (m, 1H), 3.31-3.38 (m,1H), 2.78-2.89 (m, 2H), 2.61-2.69 (m, 1H), 1.85-1.93 (m, 1H), 1.52-1.74 (m, 2H), 1.19-1.28 (m, 1H). MS (ES) [m+H]
calc'd for CZOHzINsOa, 364; found 364.
Example 26: 2-[2-(3-(R)-Amino-piperidin-1-yl)-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile O ~ CN
'N
N~N ,,.NH2 [0394] 2-[2-(3-(R)-Amino-piperidin-1-yl)-5-bromo-6-oxo-6H pyrimidin-1-ylmethyl]-benzonitrile (Example 1) (80 mg, 0.21 mmol) and tributyltin hydride (83 ~.I,, 0.31 mmol) were stirred in dry toluene (5 mL) under nitrogen.
Tetrakis(triphenylphosphine)palladium(0) (36 mg, 0.013 mmol) and a catalytic amount of AIBN were added, and the reaction stirred at 108 °C for 24 h. Concentration ire vacuo and purification by Prep HPLC
gave 51.8 mg (59%) of the title compound as the TFA salt. 1H NMR (400 MHz, CDCl3): ~ 8.40 (br s, 3H), 7.71 (d, 1H, J = 6.4 Hz), 7.63 (d, 1H, 7.6 Hz), 7.57 (t, 1H, J = 7.6 Hz), 7.40 (t, 1H, J = 7.6 Hz), 7.33 (d, 1H, J = 7.6 Hz), 6.25 (d, 1H, J = 6.4 Hz), 5.36 (AB q, 2H, J = 113.2, 15.2 Hz), 3.47-3.59 (m, 3H), 3.21-3.28 (m, 2H), 1.95-2.05 (m, 2H), 1.80-1.91 (m, 1H), 1.60-1.71 (m, 1H). MS
(ES) [m+H] calc'd for C1~H19N50, 310; found 310.
Example 27: 2-[2-(3-(R)-Amino-piperidin-1-yl)-6-oxo-5-vinyl-6H-pyrimidin-1-ylmethyl]-benzonitrile O ~ CN
,.NH2 N N
[0395] The title compound was prepared in 56% yield using tributylvinyltin in the general procedure outlined for Example 24 and was isolated as the TFA salt. 1H NMR
(400 MHz, CDCl3): ~ 8.37 (br s, 3H), 7.78 (s, 1H), 7.62 (d, 1H, 7.6 Hz), 7.56 (t, 1H, J
= 7.6 Hz), 7.39 (t, 1H, J = 7.6 Hz), 7.29 (d, 1H, J = 7.6 Hz), 6.49 (dd, 1H, J = 30.4, 11.6 Hz), 6.03 (d, 1H, J =
17.6 Hz), 5.37 (AB q, 2H, J = 100.8, 14.8 Hz), 5.33 (d, 1H, J = 11.6 Hz), 3.47-3.59 (m, 3H), 3.16-3.26 (m, 2H), 1.95-2.05 (m, 2H), 1.80-1.90 (m, 1H), 1.60-1.71 (m, 1H). MS
(ES) [m+H] calc'd for C19H21NSO, 336; found 336.
Example 28: 2-[2-(3-(R)-Amino-piperidin-1-yl)-5-methoxy-6-oxo-6H pyrimidin-1-ylmethyl]-benzonitrile O ~ CN
N
N~N ,.NH2 Example 28A: 2-Chloro-5-methoxy-3H-pyrimidin-4-one O
NH
NCI
[0396] The title compound was prepared in 8% yield from 2,4-dichloro-5-methoxy-pyrimidine [see Chesterfield et al., J. Chem. Soc., 1950, 4590-4594] according to the general procedure outlined for Example 1A. 1H NMR (400 MHz, DMSO-d6): b 7.58 (s,1H), 3.74 (s, 3H). MS (ES) [m+H] calc'd for C$HSN202C1, 161, 163; found 161, 163.
Example 28B: 2-(2-Chloro-5-methoxy-6-oxo-6H pyrimidin-1-ylmethyl)-benzonitrile O ~ CN
O N
NCI
[0397] The title compound was prepared in 33% yield from 2-chloro-5-methoxy-3H
pyrimidin-4-one according to the general procedure outlined for Example 1B. 1H
NMR (400 MHz, CDC13): 8 7.71 (d, 1H, J = 7.2 Hz), 7.55 (t, 1H, J = 7.2 Hz), 7.42 (t, 1H, J = 7.2 Hz), 7.34 (s, 1H), 7.12 (d, 1H, J = 7.2 Hz), 5.66 (s, 2H), 3.89 (s, 3H). MS (ES) [m+H] calc'd for ~13H10N3~2C1, 276, 278; found 276, 278.
Example 28: 2-[2-(3-(R)-Amino-piperidin-1-yl)-5-methoxy-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile [0398] The title compound was prepared in 49% yield from 2-(2-chloro-5-methoxy-oxo-6H pyrimidin-1-ylmethyl)-benzonitrile according to the general procedure outlined for Example 1 and was isolated as the TFA salt. 1H NMR (400 MHz, MeOD-d4): 8 7.71 (d, 1H, J = 7.2 Hz), 7.59 (t, 1H, J = 7.2 Hz), 7.47 (s, 1H), 7.42 (t, 1H, J = 7.2 Hz), 7.19 (d, 1H, J = 7.2 Hz), 5.46 (s, 2H), 3.79 (s, 3H), 3.31-3.40 (m,1H), 3.02-3.21 (m, 2H), 2.72-2.85 (m, 2H),1.92-2.02 (m,1H),1.61-1.82 (m, 2H), 1.38-1.48 (m,1H). MS (ES) [m+H] calc'd for ClgHa1N502, 340; found 340.

Example 29: 2-[2-(3-(R)-Amino-piperidin-1-yl)-6-oxo-5-(1H pyrrol-3-yl)-6H
pyrimidin-1-ylmethyl]-benzonitrile HN I O ~ CN
~N~N ,,.NH2 [0399] The title compound was prepared in 71 % yield using 1-(triisopropylsilyl)pyrrole-3-boronic acid in the general procedure outlined for Example 2, followed by TBAF
deprotection according to the procedure for Example 7. 1H NMR (400 MHz, MeOD-d4): ~
8.09 (s, 1H), 7.71 (d,1H, J = 7.2 Hz), 7.58 (t, 1H, J = 7.2 Hz), 7.36-7.47 (m, 2H), 7.14 (d,1H, J = 7.2 Hz), 6.74 (d, 1H, J = 4.4 Hz), 6.47 (d,1H, J = 4.4 Hz), 5.50 (s, 2H), 3.36-3.42 (m,1H), 3.00-3.18 (m, 2H), 2.72-2.88 (m, 2H), 1.92-2.02 (m, 1H), 1.61-1.82 (m, 2H), 1.29-1.39 (m, 1H). MS (ES) [m+H] calc'd for Ca1H22N60, 375; found 375 Example 30: 2-[2-(3-(R)-Amino-piperidin-1-yl)-5-fluoro-6-oxo-6H pyrimidin-1-ylmethyl]-benzonitrile O ~ CN
F N
N~N ,,.NH2 Example 30A: 2-Chloro-5-fluoro-3H pyrimidin-4-one O
F NH
N"CI
[0400] The title compound was prepared in 56% yield from 2,4-dichloro-5-fluoro-pyrimidine according to the procedure for Example 1A. 1H NMR (400 MHz, DMSO-d6): 8 13.98 (br s, 1H), 8.14 (d, 1H, J = 3.2 Hz).
[0401] Example 30A was also prepared as follows. Dimethylaniline ( 195 mL,1.54 mol) was added to a slurry of 5-fluorouracil (99.73 g, 0.77 mol) in phosphorus oxychloride (215 mL, 2.31 mol) at 95 °C under a nitrogen atmosphere. The reaction mixture was stirred at this temperature for 3.5 h, cooled to room temperature and then slowly added to a stirred mixture of ice (200 g) and 6 M HCl (200 mL). The resulting slurry was extracted with dichloromethane (2 x 400 mL) and the combined organic extracts were washed with DI water (4 x 275 mL), dried over MgSO4 and concentrated under reduced pressure to afford 111.77 g (87%, 98.1% AUC by HPLC) of 5-fluoro-2,4-dichloro-pyrimidine as an amber oil.
A molar solution of sodium hydroxide (1.34 L) was slowly added to a solution of the 5-fluoro-2,4-dichloro-pyrimidine ( 111.77 g, 0.67 mol) in THF (377 mL) at 0 °C.
After the reaction mixture was stirred for about 30 min at room temperature, the pH was adjusted to 6 by a slow addition of 1.0 M HCl. The aqueous solution was extracted with ethyl acetate (440 mL) to remove impurities following which the pH was adjusted to 1 with 1.0 M HCI. The acidic aqueous solution was extracted with ethyl acetate (4 x 555 mL) and the combined organic extracts were washed with brine (111 mL), dried over MgS04 and concentrated under reduced pressure to produce 88.35 g (89%, 99.2% AUC by HPLC) of 2-chloro-5-fluoro-3H
pyrimidin-4-one as an off-white powder.
Example 30B: 2-(2-Chloro-5-fluoro-6-oxo-6H pyrinudin-1-ylmethyl)-benzonitrile I~
O ~ CN
F
N
NCI
[0402] The title compound was prepared in 44% yield from 2-chloro-5-fluoro-3F1-pyrimidin-4-one according to the procedure for Example 1B. 1H NMR (400 MHz, CDC13): 8 7.81 (s, 1H), 7.74 (dd, 1H, J = 7.6, 1.2 Hz), 7.59 (td, 1H, J = 7.6, 1.2 Hz), 7.45 (t, 1H, J = 7.6 Hz), 7.15 (d, 1H, J = 7.6 Hz), 5.67 (s, 2H). MS (ES) [m+H] calc'd for C12H~N30FC1, 264, 266; found 264, 266.
[0403] Also obtained from the reaction were impure fractions of the less polar O-alkylated isomer, and the more polar N3-alkylated isomer.
[0404] Example 30B was also prepared from 5-fluoro-2-chloro-3H pyrimidin-4-one as follows. To a solution of 5-fluoro-2-chloro-3H pyrimidin-4-one (100.00 g, 0.67 mol) in 1:l DMF/DME (440 mL) under a nitrogen atmosphere was added sodium iodide ( 10 g, 67 rnmol) and the resulting slurry cooled to 0 °C. While maintaining the internal temperature to <_12 °C, 1,1,3,3-tetramethylguanidine (94 mL, 0.74 mol) was added dropwise over 45 minutes to form a homogeneous solution. The ice bath was removed and a solution of 2-(bromomethyl)benzonitrile (145 g, 0.74 mol) in 1:1 DMF/DME (600 mL) was added all at once and the reaction mixture heated to 70 °C for 18 h. In-process assay (by HPLC analysis);
showed complete consumption of the starting material. The reaction mixture was cooled to 30 °C and then MTBE (1.14 L) and DI water (2.29 L) were added to form a slurry which was stirred at ambient temperature for 1 h. The crude product was collected by filtration and washed with MTBE ( 1.14 L). The phases of the filtrate were separated and the organic layer was combined with the filter-cake and concentrated to a thick slurry which was subsequently reslurried with DME (1.00 L). Insoluble impurities were removed by vacuum filtration and washed with DME (500 rnL). The filtrate was concentrated to a wet residue which was reslurried with 2-propanol (365 mL) and the solid product collected by vacuum filtration.
After removal of residual IPA, the product was reslurried in ethyl acetate (6 vol) at 70 °C and heptane (6 vol) was added slowly while maintaining the internal temperature at 68-70 °C. At the end of the addition of heptane, insoluble materials were still visible.
The mixture was stirred for an additional 10 min after which insoluble impurities were removed by hot vacuum filtration. The filtrate was slowly cooled to room temperature and then cooled further to about 0 °C by means of a salt-ice bath. The crystallized product was collected by vacuum filtration and then reslurried in ethanol (6 vol) at 50 °C, cooled to 40 °C, and the product collected by vacuum filtration to give 80.62 g (45%, 95.6% AUC by HPLC) of 2-(2-chloro-5-fluoro-6-oxo-6H-pyrimidin-1-ylmethyl)-benzonitrile as an off white solid.
Example 30: 2-[2-(3-(R)-Amino-piperidin-1-yl)-5-fluoro-6-oxo-6H pyrimidin-1-ylmethyl]-benzonitrile O ~ CN
F
N
N~N ,,.NH2 [0405] The title compound was prepared in 68% yield from 2-(2-chloro-5-fluoro-6-oxo-6H pyrimidin-1-ylmethyl)-benzonitrile according to the general procedure outlined for Example 1 and was isolated as the TFA salt. 1H NMR (400 MHz, MeOD-d4): 8 7.99 (d, 1H, J = 0.8 Hz), 7.85 (d, 1H, J = 7.2 Hz), 7.64 (t,1H, J = 7.6 Hz), 7.47 (t,1H, J
= 7.6 Hz), 7.20 (d, 1H, J = 7.6 Hz), 5.33 (s, 2H), 3.49-3.58 (m,1H), 3.10-3~ 19 (m, 1H), 2.68-2.76 (m, 2H), 2.48-2.58 (m, 1H), 1.60-1.80 (m, 2H), 1.41-1.51 (m, 1H), 1.10-1.19 (m, 1H). MS (ES) [m+H]
calc'd for C1~H18NSOF, 328; found 328.
[0406] Example 30 was also prepared from 2-(2-chloro-5-fluoro-6-oxo-6H-pyrimidin-1-ylmethyl)-benzonitrile as follows. A mixture of 2-(2-chloro-5-fluoro-6-oxo-6H-pyrimidin-1-ylmethyl)-benzonitrile (80.62 g, 0.31 mol), (R)-3-aminopiperidine dihydrochloride (58.00 g, 0.34 mol) and potassium carbonate (186 g, 1.35 mol) in 10% water in IPA (807 mL) was heated at 45 °C for 1 h. After cooling to room temperature, isopropyl acetate (807 mL) and 2 M HCl (807 mL) were added. Following phase separation, the organic layer was extracted with 2.0 M HCl (2 x 807 mL). The aqueous extracts were combined, washed with isopropyl acetate (807 mL), cooled to 10 °C and the pH adjusted to 13 with caustic soda. The alkaline slurry was extracted with isopropyl acetate (2 x 807 mL), and the combined organic extracts concentrated to afford 93.50 g (93%, 98.4% AUC by HPLC) of Example 30 as a viscous oil (93.49 g, 93%).
[0407] The tartrate salt was prepared by adding a solution of L-tartaric acid in 5% water in IPA (3.00 L) to a solution of Example 30 (93.00 g, 284 mmol) in methanol (982 mL) at 65 °C. The mixture was stirred for 20 min and then cooled to room temperature. The resulting precipitate was collected by vacuum filtration, washed with 5% water in IPA (2 x 560 mL), and dried in a vacuum oven at 75 °C to give 112.78 g (77%, 100% AUC by HPLC) of the salt as a white solid.
Example 31: 2-(3-(R)-Amino-piperidin-1-yl)-3-(2-bromo-benzyl)-5-fluoro-3H
pyrimidin-4-one O ~ Br F N
N~N ,,.NH2 Example 31A: 3-(2-Bromo-benzyl)-2-chloro-5-fluoro-3H-pyrimidin-4-one O ~ Br F
N
NCI
[0408] The title compound was prepared in 11 % yield from 2-chloro-5-fluoro-3H
pyrimidin-4-one and 2-bromobenzyl bromide according to the procedure for Example 1B. 1H
NMR (400 MHz, CDC13): 8 7.82 (d, 1H, J =1.2 Hz), 7.62 (dd,1H, J = 7.6, 1.2 Hz), 7.17-7.30 (m, 2H), 6.78 (d, 1H, J = 7.6 Hz), 5.52 (s, 2H). MS (ES) [m+H] calc'd for C11H~N~OFCIBr, 317, 319; found 317, 319.
[0409] Also obtained from the reaction were impure fractions of the less polar O-alkylated isomer, and the more polar N3-alkylated isomer.
Example 31: 2-(3-(R)-Amino-piperidin-1-yl)-3-(2-bromo-benzyl)-5-fluoro-3H
pyrimidin-4-one O ~ Br F
N
N~N ,,.NH2 [0410] The title compound was prepared in 75% yield from 3-(2-bromo-benzyl)-2-chloro-5-fluoro-3H pyrirnidin-4-one according to the general procedure outlined for Example 1 and was isolated as the HCl salt. 1H NMR (400 MHz, DMSO-d6): 8 8.48 (br s, 3H), 8.00 (d, 1H, J = 1.2 Hz), 7.61 (d, 1H, J = 7.6 Hz), 7.31 (t, 1H, J = 7.6 Hz), 7.21 (t, 1H, J = 7.6 Hz), 7.00 (d, 1H, J = 7.6 Hz), 5.14 (AB q, 2H, J = 35.6, 15.6 Hz), 3.42-3.53 (m, 1H), 3.16-3.26 (m, 1H), 2.95-3.06 (m, 2H), 2.62-2.72 (m, 1H), 1.90-1.99 (m, 1H), 1.67-1.76 (m, 1H), 1.39-1.59 (m, 2H). MS (ES) [m+H] calc'd for C16H18N~OFBr, 381, 383; found 381, 383.

.WO 2005/016911.-.- - . PCT/US2004/026265 Example 32: 2-[2-(3-(R)-Amino-piperidin-1-yl)-6-oxo-5-((E~-2-pyridin-3-yl-vinyl)-6H-pyrimidin-1-ylmethyl]-benzonitrile \
O ~ CN
\ / ~ N
N~N ,.NH2 Example 32A: 3-((L~-2-Tributylstannanyl-vinyl)-pyridine [0411] 3-Ethynyl-pyridine ( 1.03 g, 10 mmol) and tributyltin hydride (3.2 mL, 12 mmol) were stirred in dry THF (10 mL) with a catalytic amount of AIBN at 50 °C for 18 h. The solution was concentrated ih vacuo and purified by silica gel chromatography (10% EOAc /
hexanes) to give 2.09 g (53%) of the title compound as a clear oil. 1H NMR
(400 MHz, DMSO-d6): 8 8.61 (s, 1H), 8.45 (d, 1H, J = 3.2 Hz), 7.75 (d, 1H, J = 7.6 Hz), 7.25-7.29 (m, 1H), 6.93 (AB q, 2 H, J = 57.2,19.6 Hz),1.49-1.59 (m, 6H),1.28-1.39 (m, 6H), 0.87-1.00 (m, 15H).
[0412] Also obtained from the reaction was 0.91 g (23%) of the less polar 3-((~-2-tributylstannanyl-vinyl)-pyridine. 1H NMR (400 MHz, DMSO-d6): 8 8.48-8.52 (m, 2H), 7.53-7.58 (m, 2H), 7.24-7.27 (m, 1H), 6.38 (d, 1H, J =14.0 Hz), 1.35-1.45 (m, 6H),1.20-1.30 (m, 6H), 0.78-0.90 (m, 15H).
Example 32: 2-[2-(3-(R)-Amino-piperidin-1-yl)-6-oxo-5-((E~-2-pyridin-3-yl-vinyl)-6H pyrimidin-1-ylmethyl]-benzonitrile \
O ~ CN
\ / ~ N
N~N ,,.NH2 " WO 2005/016911,. ,. . ....... "". ....... ,.._ .".., PCT/US2004/026265 [0413] The title compound was prepared in 55% yield using 3-((~-2-tributylstannanyl-vinyl)-pyridine the general procedure outlined for Example 24. 1H NMR (400 MHz, MeOD-d4): 8 8.53 (d, 1H, J =1.6 Hz), 8.32 (d, 1H, J = 4.8 Hz), 8.04 (s, 1H), 7.92 (d, 1H, J = 7.6 Hz), 7.70 (d, 1H, J = 7.6 Hz), 7.50-7.61 (m, 2H), 7.31-7.47 (m, 2H), 7.24 (d, 1H, J
= 7.6 Hz), 7.00 (d, 1H, J = 16.4 Hz), 5.41 (s, 2H), 3.51-3.58 (m, 1H), 3.29-3.37 (m, 1H), 2.81-2.99 (m, 2H), 2.70-2.78 (m, 1H), 1.90-1.99 (m, 1H), 1.62-1.79 (m, 2H), 1.26-1.36 (m, 1H). MS
(ES) [m+H] calc'd for C2qH2qN6O, 413; found 413.
Example 33: 2-[2-(3-(R)-Amino-piperidin-1-yl)-6-oxo-4-phenyl-6H pyrimidin-1-ylmethyl]-benzonitrile O ~ CN
~N
N~N .,.NH2 Example 33A: 2-(2-Chloro-6-oxo-4-phenyl-6F1-pyrimidin-1-ylmethyl)-benzonitrile O ~ CN
~N
NCI
[0414] The title compound was prepared in 49% yield from 2-chloro-4-phenyl-3H
pyrimidin-4-one [see Elmoghayar, et al.; Acta Clae~n. Scared. Ser. B, 37, 2, 1983, 109-114]
according to the procedure for Example 1B. 1H NMR (400 MHz, CDC13): 8 7.96 (d, 2H, J =
7.6 Hz), 7.73 (d, 1H, J = 7.6 Hz), 7.41-7.60 (m, 5H), 7.21 (d, 1H, J = 7.6 Hz), 6.92 (s, 1H), 5.69 (s, 2H). MS (ES) [m+H] calc'd for C1gH12N30C1, 322, 324; found 322, 324.
[0415] Also obtained from the reaction were impure fractions of the less polar O-alkylated isomer.

.-WO 2005/016911". :, , ""." ..". .~.,. "". ".", PCT/US2004/026265 Example 33: 2-[2-(3-(R)-Amino-piperidin-1-yl)-6-oxo-4-phenyl-6H pyrimidin-1-ylmethyl]-benzonitrile O ~ CN
~N
N~N ,,.Nf-f2 [0416] The title compound was prepared in 72% yield from 2-(2-chloro-6-oxo-4-phenyl-6H pyrimidin-1-ylmethyl)-benzonitrile according to the general procedure outlined for Example 1 and was isolated as the HCl salt. 1H NMR (400 MHz, DMSO-d6): 8 8.51 (s, 3H), 8.06-8.14 (m, 2H), 7.81 (d, 1H, J = 7.6 Hz), 7.62 (t, 1H, J = 7.6 Hz), 7.39-7.51 (m, 4H), 7.23 (d,1H, J = 7.6 Hz), 6.69 (s,1H), 5.33 (AB q, 2H, J = 36.8, 15.2 Hz), 3.67-3.76 (m,1H), 3.35-3.45 (m, 1H), 3.15-3.26 (m, 2H), 2.90-3.00 (m, 1H), 1.95-2.05 (m, 1H), 1.78-1.88 (m, 1H), 1.53-1.70 (m, 2H). MS (ES) [m+H] calc'd for C23H23NSO, 386; found 386.
Example 34: 2-[2-(3-(R)-Amino-3-methyl-piperidin-1-yl)-5-fluoro-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile O ~ CN
F N
N~N ,,.NH2 [0417] The title compound was prepared in 87% yield from 2-(2-chloro-5-fluoro-6-oxo-6H pyrimidin-1-ylmethyl)-benzonitrile (Example 30B) and (R)-3-methyl-piperidin-3-ylamine according to the general procedure outlined for Example 1 and was isolated as the HCl salt.
1H NMR (400 MHz, DMSO-d6): 8 8.36 (br s, 3H), 8.01 (d, 1H, J =1.6 Hz), 7.83 (d, 1H, J =
7.6 Hz), 7.64 (t, 1H, J = 7.6 Hz), 7.46 (t,1H, J = 7.6 Hz), 7.21 (d,1H, J =
7.6 Hz), 5.40 (AB q, 2H, J =107.2, 15.2 Hz), 3.29-3.37 (m, 1H), 2.99-3.10 (m, 2H), 2.76-2.82 (m, 1H), 1.80-2.00 (m, 2H), 1.55-1.66 (m, 2H), 1.28 (m, 3H). MS (ES) [m+H] calc'd for ClgH2oN50F, 342;
found 342.

,~WO 2005/016911._, .. . ..,..,. ~._.. ",.,., "..~ ",.., PCT/US2004/026265 Example 35: 2-[2-(3-(R)-Amino-3-methyl-piperidin-1-yl)-5-chloro-6-oxo-6H
pyrimidin-1-ylmethyl]-benzonitrile I~
O ~ CN
CI
NI
N~N ,,.NH2 [0418] The title compound was prepared in 65% yield from 2-(2,5-dichloro-6-oxo-pyrimidin-1-ylmethyl)-benzonitrile (Example 15B) and (R)-3-methyl-piperidin-3-ylamine according to the general procedure outlined for Example 1 and was isolated as the HCl salt.
1H NMR (400 MHz, DMSO-d~): ~ 8.43 (br s, 3H), 8.09 (s, 1H), 7.80 (d,1H, J =
7.6 Hz), 7.64 (t,1H, J = 7.6 Hz), 7.45 (t, 1H, J = 7.6 Hz), 7.22 (d,1H, J = 7.6 Hz), 5.39 (AB q, 2H, J = 98.0, 14.8 Hz), 3.39-3.47 (m, 1H), 3.01-3.15 (m, 2H), 2.80-2.90 (m, 1H), 1.81-2.00 (m, 2H),1.56-1.66 (m, 2H), 1.30 (m, 3H). MS (ES) [m+H] calc'd for C18H2oN50C1, 358, 360;
found 358, 360.
Example 36: (R)-2-((2-(3-aminopiperidin-1-yl)-5-bromo-4-tert-butyl-6-oxopyrimidin-1 (6H)-yl)methyl)benzonitrile O ~ CN
Br N
I N~N ,,NH2 O O I ~ CN O I ~ CN amine O I ~ CN
Br2, t-BuONO Br I ~ Br NaH Br I ~ NaHC03 Br I N
.. ' ' ~ ~.~.NHz N NH2 CHCI3 N Br pME-DMF N Br MeOH N N
[0419] To a suspended mixture of Compound 36A ( 1.0 g, 5.9 mmol), bromine ( 1.0 g) and CuBr (1.01 g, 7.1 rnmol) in CHC13 (50 mL), were slowly added Br2 (1.0 g) and 97%
isoamyl nitrite (2.9 g mL) in CHC13 (5 mL) at the same time over 10 minutes.
The mixture ~WO 2005/016911,. a . _~ ..,.. A..,.~ .~.. 3..,. PCT/US2004/026265 then was stirred under reflux for 2 h, diluted with DCM, washed with saturated Na2SO3, dried over Na2S04, concentrated and purified by chromatography to give Compound 36B.
[0420] Compound 36B (200 mg) in DME-DMF (4:1) was converted to Example 36 in two steps according to the procedures outlined for Example 1B and Example 1 and was isolated as the TFA salt. 1H NMR (400 MHz, CDCl3-CD3OD 10:1) 8 7.68 - 7.74 (d, J=7.5 Hz, 1H), 7.62 (t, J=8.5 Hz, 2H), 7.39 (t, J=7.6 Hz, 1H), 5.57 (q, J=13.6 Hz, 2H), 4.80 (d, J=11.4 Hz, 1H), 4.40 (d, J=12.6 Hz, 1H), 3.29 (m, 1H), 3.15 - 3.26 (m, 1H), 3.02 (t, J=11.4 Hz, 1H), 2.16 (d, J=8.1 Hz, 1H), 1.80 (d, J=12.4 Hz, 1H), 1.53 (d, J=13.4 Hz, 2H), 1.41 (s, 9H). MS (ES) [m+H] calc'd for C21HZ~BrN50, 444; found 444.
3. Examples of Ih vitro Assays [0421] The protease inhibitory activities of DPP-IV inhibitors can be readily determined by methods known to those of ordinary skill in the art since suitable ifa 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-IV Assay [0422] 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 0.lmg/mL BSA. Human DPP-IV
(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 ~,M final concentration). The total volume of the reaction mixture was 10-100~.L depending on assay formats used (384 or 96 well plates). The reaction was followed kinetically (excitation X400 nm; emission ~,=505 nm) for minutes or an end-point was measured after 10 minutes. Inhibition constants (ICSO) were calculated from the enzyme progress curves using standard mathematical models.
FAPa Assay [0423] 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 FAPa (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 ~.tM final concentration). The total volume of the reaction mixture was 10-100~.I. depending on assay formats used (384 or 96 well plates). The reaction was followed kinetically (excitation ~-400 nm; emission ~,=505 nm) for 5-10 minutes or an end-point was measured after 10 minutes. Inhibition constants (ICSO) were calculated from the enzyme progress curves using standard mathematical models.
PREP Assay [0424] Solutions of test compounds in varying concentrations (<_lOmM final concentration) were prepared in Dimethyl Sulfoxide (DMSO) and then diluted into assay buffer comprising: 20rnM Sodium Phosphate, pH 7.4; 0.5mM EDTA; 0.5mM DTT; and 0.1mg/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 minutes at ambient temperature before the reaction was initiated with Z-G-P-AMC ( 10 ~1M
final concentration). The total volume of the reaction mixture was 10-100~,I, depending on assay formats used (384 or 96 well plates). The reaction was followed kinetically (excitation ?i,=375 nm; emission ar-460 nm) for 10 minutes or an end-point was measured after 10 minutes. Inhibition constants (ICso) were calculated from the enzyme progress curves using standard mathematical models.
Tryptase Assay [0425] Solutions of test compounds in varying concentrations (__<lOmM final concentration) were prepared in Dimethyl Sulfoxide (DMSO) and then diluted into assay buffer comprising: 100mM 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 temper ature. The enzymatic reaction was initiated with 25 ~,M Z-lys-SBzl and 400~,M DTNB. The total volume of the reaction mixture was 100~.L, in Costar A/2 96 well plates. The reaction was followed colorimetrically 0405 nm) for 10 minutes. Inhibition constants (ICso) were calculated from the enzyme progress curves using standard mathematical models.
[0426] Compounds of the invention were tested according to the above-described assays for protease inhibition and observed to exhibit selective DPP-IV inhibitory activity. For example, compounds of the invention were found to inhibit DPP-IV activity at concentrations ::WO 2005/016911.- PCT/US2004/026265 that are at least 50 fold less than those concentrations required to produce an equiactive inhibition of protease activity for FAPa. The apparent inhibition constants (K;) for compounds of the invention, against DPP-IV, were in the range from about 10-9M
to about 10-5M.
[0427] It will be apparent to those skilled in the art that various modifications and variations can be made in 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 (184)

1. A compound comprising Formula I:
wherein Q is selected from the group consisting of CO, SO, SO2, and C=NR4;
Z is selected from the group consisting of halo, perhalo(C1-10)alkyl, amino, cyano, thio, (C1-10)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, carbonyl (C1-3)alkyl, thiocarbonyl (C1-3)alkyl, sulfonyl (C1-3)alkyl, sulfinyl (C1-3)alkyl, imino (C1-3)alkyl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, 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;
R2 and R3 are each independently selected from the group consisting of hydrogen, halo, perhalo(C1-10)alkyl, amino, cyano, nitro, thio, (C1-10)alkyl, alkene, alkyne, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(C1-10)alkyl, heteroaryl (C1-5)alkyl, (C9-12)bicycloaryl, hetero(C8-12)bicycloaryl, carbonyl (C1-3)alkyl, thiocarbonyl (C1-3)alkyl, sulfonyl (C1-3)alkyl, sulfinyl (C1-3)alkyl, imino (C1-3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;
R4 is selected from the group consisting of hydrogen, (C1-10)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, arylalkyl, heteroarylalkyl, bicycloaryl, heterobicycloaryl, each substituted or unsubstituted;
L is a linker providing 0-6 atom separation between X and the ring to which L
is attached; and X is selected from the group consisting of (C1-10)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl, hetero(C4-12)bicycloaryl, carbonyl (C1-3)alkyl, thiocarbonyl (C1-3)alkyl, sulfonyl (C1-3)alkyl, sulfinyl (C1-3)alkyl, imino (C1-3)alkyl, amino, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, cyano, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
2. ~A compound comprising Formula II:
wherein n is 0, 1, 2, or 3;
Q is selected from the group consisting of CO, SO, SO2, and C=NR4;
Z is selected from the group consisting of halo, perhalo(C1-10)alkyl, amino, cyano, thio, (C1-10)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, carbonyl (C1-
3)alkyl, thiocarbonyl (C1-3)alkyl, sulfonyl (C1-3)alkyl, sulfinyl (C1-3)alkyl, imino (C1-3)alkyl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, 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;
R2 and R3 are each independently selected from the group consisting of hydrogen, halo, perhalo(C1-10)alkyl, amino, cyano, nitro, thio, (C1-10)alkyl, alkene, alkyne, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(C1-10)alkyl, heteroaryl (C1-5)alkyl, (C9-12)bicycloaryl, hetero(C8-12)bicycloaryl, carbonyl (C1-3)alkyl, thiocarbonyl (C1-3)alkyl, sulfonyl (C1-3)alkyl, sulfinyl (C1-3)alkyl, imino (C1-3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;
R4 is selected from the group consisting of hydrogen, (C1-10)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, arylalkyl, heteroarylalkyl, bicycloaryl, heterobicycloaryl, each substituted or unsubstituted;
R5 and R6 are each independently selected from the group consisting of hydrogen, a substituted or unsubstituted (C1-10)alkyl, a substituted or unsubstituted (C1-10)alkoxy, cyano, and halo, or where R5 and R6 are taken together to form a ring; and X is selected from the group consisting of (C1-10)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl, hetero(C4-12)bicycloaryl, carbonyl (C1-3)alkyl, thiocarbonyl (C1-3)alkyl, sulfonyl (C1-3)alkyl, sulfinyl (C1-3)alkyl, imino (C1-3)alkyl, amino, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, cyano, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
3. A compound comprising Formula III:
wherein m is 0, 1, 2, 3, 4, or 5;
n is 0, 1, 2, or 3;
Q is selected from the group consisting of CO, SO, SO2, and C=NR4;
Z is selected from the group consisting of halo, perhalo(C1-10)alkyl, amino, cyano, thio, (C1-10)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, carbonyl (C1-3)alkyl, thiocarbonyl (C1-3)alkyl, sulfonyl (C1-3)alkyl, sulfinyl (C1-3)alkyl, imino (C1-3)alkyl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, 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;
R2 and R3 are each independently selected from the group consisting of hydrogen, halo, perhalo(C1-10)alkyl, amino, cyano, nitro, thio, (C1-10)alkyl, alkene, alkyne, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(C1-10)alkyl, heteroaryl (C1-5)alkyl, (C9-12)bicycloaryl, hetero(C8-12)bicycloaryl, carbonyl (C1-3)alkyl, thiocarbonyl (C1-3)alkyl, sulfonyl (C1-3)alkyl, sulfinyl (C1-3)alkyl, imino (C1-3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;

R4 is selected from the group consisting of hydrogen, (C1-10)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, arylalkyl, heteroarylalkyl, bicycloaryl, heterobicycloaryl, each substituted or unsubstituted;
R5 and R6 are each independently selected from the group consisting of hydrogen, a substituted or unsubstituted (C1-10)alkyl, a substituted or unsubstituted (C1-10)alkoxy, cyano, and halo, or where R5 and R6 are taken together to form a ring; and each R7 is independently selected from the group consisting of halo, perhalo(C1-10)alkyl, CF3, (C1-10)alkyl, alkenyl, alkynyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, hydroxy, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
4. A compound comprising one of Formulae IVa, IVb and IVc:
wherein n is 0, 1, 2, or 3;
Q is selected from the group consisting of CO, SO, SO2, and C=NR4;
each of T, U, V, W and Y is independently nitrogen or CR16, provided that no more than two of T, U, V, W and Y are nitrogen;
Z is selected from the group consisting of halo, perhalo(C1-10)alkyl, amino, cyano, thio, (C1-10)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, carbonyl (C1-3)alkyl, thiocarbonyl (C1-3)alkyl, sulfonyl (C1-3)alkyl, sulfinyl (C1-3)alkyl, imino (C1-3)alkyl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, 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;
R2 and R3 are each independently selected from the group consisting of hydrogen, halo, perhalo(C1-10)alkyl, amino, cyano, nitro, thio, (C1-10)alkyl, alkene, alkyne, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(C1-10)alkyl, heteroaryl (C1-
5)alkyl, (C9-12)bicycloaryl, hetero(C8-12)bicycloaryl, carbonyl (C1-3)alkyl, thiocarbonyl (C1-3)alkyl, sulfonyl (C1-3)alkyl, sulfinyl (C1-3)alkyl, imino (C1-3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;
R4 is selected from the group consisting of hydrogen, (C1-10)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, arylalkyl, heteroarylalkyl, bicycloaryl, heterobicycloaryl, each substituted or unsubstituted;
R5 and R6 are each independently selected from the group consisting of hydrogen, a substituted or unsubstituted (C1-10)alkyl, a substituted or unsubstituted (C1-10)alkoxy, cyano, and halo, or where R5 and R6 are taken together to form a ring; and each R16 is independently selected from the group consisting of halo, perhalo(C1-10)alkyl, CF3, (C1-10)alkyl, alkenyl, alkynyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, hydroxy, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
5. A compound comprising Formula V:
wherein Q is selected from the group consisting of CO, SO, SO2, and C=NR4;
R1 is selected from the group consisting of (C1-10)alkyl, (C3-12)cycloalkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl, hetero(C8-12)bicycloaryl(C1-5)alkyl, carbonyl (C1-3)alkyl, thiocarbonyl (C1-3)alkyl, sulfonyl (C1-3)alkyl, sulfinyl (C1-3)alkyl, imino (C1-3)alkyl, amino, cyano, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;
R2 and R3 are each independently selected from the group consisting of hydrogen, halo, perhalo(C1-10)alkyl, amino, cyano, nitro, thio, (C1-10)alkyl, alkene, alkyne, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(C1-10)alkyl, heteroaryl (C1-5)alkyl, (C9-12)bicycloaryl, hetero(C8-12)bicycloaryl, carbonyl (C1-3)alkyl, thiocarbonyl (C1-3)alkyl, sulfonyl (C1-3)alkyl, sulfinyl (C1-3)alkyl, imino (C1-3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;
R4 is selected from the group consisting of hydrogen, (C1-10)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, arylalkyl, heteroarylalkyl, bicycloaryl, heterobicycloaryl, each substituted or unsubstituted; and R9 and R10 are each independently selected from the group consisting of hydrogen, perhalo(C1-10)alkyl, amino, cyano, (C1-10)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(C1-10)alkyl, heteroaryl (C1-5)alkyl, (C9-12)bicycloaryl, hetero(C8-12)bicycloaryl, carbonyl (C1-3)alkyl, thiocarbonyl (C1-3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, sulfonyl group, sulfinyl group, each substituted or unsubstituted, or R9 and R10 are taken together to form a 5, 6, or 7 membered ring, each substituted or unsubstituted.
6. A compound comprising Formula VI:
wherein Q is selected from the group consisting of CO, SO, SO2, and C=NR4;
R2 and R3 are each independently selected from the group consisting of hydrogen, halo, perhalo(C1-10)alkyl, amino, cyano, nitro, thio, (C1-10)alkyl, alkene, alkyne, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(C1-10)alkyl, heteroaryl (C1-5)alkyl, (C9-12)bicycloaryl, hetero(C8-12)bicycloaryl, carbonyl (C1-3)alkyl, thiocarbonyl (C1-3)alkyl, sulfonyl (C1-3)alkyl, sulfinyl (C1-3)alkyl, imino (C1-3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;
R4 is selected from the group consisting of hydrogen, (C1-10)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, arylalkyl, heteroarylalkyl, bicycloaryl, heterobicycloaryl, each substituted or unsubstituted;
R9 and R10 are each independently hydrogen, perhalo(C1-10)alkyl, amino, cyano, (C1-10)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(C1-10)alkyl, heteroaryl (C1-5)alkyl, (C9-12)bicycloaryl, hetero(C8-12)bicycloaryl, carbonyl (C1-3)alkyl, thiocarbonyl (C1-3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, sulfonyl group, sulfinyl group, each substituted or unsubstituted, or R9 and R10 are taken together to form a 5, 6, or 7 membered ring, each substituted or unsubstituted;
L is a linker providing 0-6 atom separation between X and the ring to which L
is attached; and X is selected from the group consisting of (C1-10)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl, hetero(C4-12)bicycloaryl, carbonyl (C1-3)alkyl, thiocarbonyl (C1-3)alkyl, sulfonyl (C1-3)alkyl, sulfinyl (C1-3)alkyl, imino (C1-3)alkyl, amino, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, cyano, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
7. A compound comprising Formula VII:
wherein n is 0,1, 2, or 3;
Q is selected from the group consisting of CO, SO, SO2, and C=NR4;
R2 and R3 are each independently selected from the group consisting of hydrogen, halo, perhalo(C1-10)alkyl, amino, cyano, nitro, thio, (C1-10)alkyl, alkene, alkyne, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(C1-10)alkyl, heteroaryl (C1-5)alkyl, (C9-12)bicycloaryl, hetero(C8-12)bicycloaryl, carbonyl (C1-3)alkyl, thiocarbonyl (C1-3)alkyl, sulfonyl (C1-3)alkyl, sulfinyl (C1-3)alkyl, imino (C1-3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;
R4 is selected from the group consisting of hydrogen, (C1-10)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, arylalkyl, heteroarylalkyl, bicycloaryl, heterobicycloaryl, each substituted or unsubstituted;
R5 and R6 are each independently selected from the group consisting of hydrogen, a substituted or unsubstituted (C1-10)alkyl, a substituted or unsubstituted (C1-10)alkoxy, cyano, and halo, or where R5 and R6 are taken together to form a ring;
R9 and R10 are each independently hydrogen, perhalo(C1-10)alkyl, amino, cyano, (C1-10)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(C1-10)alkyl, heteroaryl (C1-5)alkyl, (C9-12)bicycloaryl, hetero(C8-12)bicycloaryl, carbonyl (C1-3)alkyl, thiocarbonyl (C1-3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, sulfonyl group, sulfinyl group, each substituted or unsubstituted, or R9 and R10 are taken together to form a 5, 6, or 7 membered ring, each substituted or unsubstituted; and X is selected from the group consisting of (C1-10)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl, hetero(C4-12)bicycloaryl, carbonyl (C1-3)alkyl, thiocarbonyl (C1-3)alkyl, sulfonyl (C1-3)alkyl, sulfinyl (C1-3)alkyl, imino (C1-3)alkyl, amino, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, cyano, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
8. ~A compound comprising Formula VIII:

wherein n is 0, 1, 2, or 3;
m is 0, 1, 2, 3, 4, or 5;
Q is selected from the group consisting of CO, SO, SO2, and C=NR4;
R2 and R3 are each independently selected from the group consisting of hydrogen, halo, perhalo(C1-10)alkyl, amino, cyano, nitro, thio, (C1-10)alkyl, alkene, alkyne, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(C1-10)alkyl, heteroaryl (C1-5)alkyl, (C9-12)bicycloaryl, hetero(C8-12)bicycloaryl, carbonyl (C1-3)alkyl, thiocarbonyl (C1-3)alkyl, sulfonyl (C1-3)alkyl, sulfinyl (C1-3)alkyl, imino (C1-3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;
R4 is selected from the group consisting of hydrogen, (C1-10)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, arylalkyl, heteroarylalkyl, bicycloaryl, heterobicycloaryl, each substituted or unsubstituted;
R5 and R6 are each independently selected from the group consisting of hydrogen, a substituted or unsubstituted (C1-10)alkyl, a substituted or unsubstituted (C1-10)alkoxy, cyano, and halo, or where R5 and R6 are taken together to form a ring;
each R7 is independently selected from the group consisting of halo, perhalo(C1-10)alkyl, CF3, (C1-10)alkyl, alkenyl, alkynyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, hydroxy, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted; and R9 and R10 are each independently hydrogen, perhalo(C1-10)alkyl, amino, cyano, (C1-10)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(C1-10)alkyl, heteroaryl (C1-5)alkyl, (C9-12)bicycloaryl, hetero(C8-12)bicycloaryl, carbonyl (C1-3)alkyl, thiocarbonyl (C1-3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, sulfonyl group, sulfinyl group, each substituted or unsubstituted, or R9 and R10 are taken together to form a 5, 6, or 7 membered ring, each substituted or unsubstituted.
9. ~A compound comprising one of Formulae IXa, IXb, and IXc:
wherein n is 0, 1, 2, or 3;
Q is selected from the group consisting of CO, SO, SO2, and C=NR4;
each of T, U, V, W, and Y is independently nitrogen or CR16, provided that no more than two of T, U, V, W, and Y are nitrogen;
R2 and R3 are each independently selected from the group consisting of hydrogen, halo, perhalo(C1-10)alkyl, amino, cyano, nitro, thio, (C1-10)alkyl, alkene, alkyne, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(C1-10)alkyl, heteroaryl (C1-5)alkyl, (C9-12)bicycloaryl, hetero(C8-12)bicycloaryl, carbonyl (C1-3)alkyl, thiocarbonyl (C1-3)alkyl, sulfonyl (C1-3)alkyl, sulfinyl (C1-3)alkyl, imino (C1-3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;
R4 is selected from the group consisting of hydrogen, (C1-10)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, arylalkyl, heteroarylalkyl, bicycloaryl, heterobicycloaryl, each substituted or unsubstituted;
R5 and R6 are each independently selected from the group consisting of hydrogen, a substituted or unsubstituted (C1-10)alkyl, a substituted or unsubstituted (C1-
10)alkoxy, cyano, and halo, or where R5 and R6 are taken together to form a ring;

each R16 is independently selected from the group consisting of halo, perhalo(C1-10)alkyl, CF3, (C1-10)alkyl, alkenyl, alkynyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, hydroxy, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted; and R9 and R10 are each independently hydrogen, perhalo(C1-10)alkyl, amino, cyano, (C1-10)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(C1-10)alkyl, heteroaryl (C1-5)alkyl, (C9-12)bicycloaryl, hetero(C8-12)bicycloaryl, carbonyl (C1-3)alkyl, thiocarbonyl (C1-3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, sulfonyl group, sulfinyl group, each substituted or unsubstituted, or R9 and R10 are taken together to form a 5, 6, or 7 membered ring, each substituted or unsubstituted.

10. ~A compound comprising Formula X:
wherein p is 0-10;
Q is selected from the group consisting of CO, SO, SO2, and C=NR4;
R1 is selected from the group consisting of (C1-10)alkyl, (C3-12)cycloalkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl, hetero(C8-12)bicycloaryl(C1-5)alkyl, carbonyl (C1-3)alkyl, thiocarbonyl (C1-3)alkyl, sulfonyl (C1-3)alkyl, sulfinyl (C1-3)alkyl, imino (C1-3)alkyl, amino, cyano, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;
R2 and R3 are each independently selected from the group consisting of hydrogen, halo, perhalo(C1-10)alkyl, amino, cyano, nitro, thio, (C1-10)alkyl, alkene, alkyne, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(C1-10)alkyl, heteroaryl (C1-5)alkyl, (C9-12)bicycloaryl, hetero(C8-12)bicycloaryl, carbonyl (C1-3)alkyl, thiocarbonyl (C1-3)alkyl, sulfonyl (C1-3)alkyl, sulfinyl (C1-3)alkyl, imino (C1-3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;
R4 is selected from the group consisting of hydrogen, (C1-10)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, arylalkyl, heteroarylalkyl, bicycloaryl, heterobicycloaryl, each substituted or unsubstituted; and each R8 is independently selected from the group consisting of halo, perhalo(C1-10)alkyl, CF3, (C1-10)alkyl, alkenyl, alkynyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, hydroxy, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
11. ~A compound comprising Formula XI:
wherein p is 0-10;
Q is selected from the group consisting of CO, SO, SO2, and C=NR4;
R2 and R3 are each independently selected from the group consisting of hydrogen, halo, perhalo(C1-10)alkyl, amino, cyano, nitro, thio, (C1-10)alkyl, alkene, alkyne, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(C1-10)alkyl, heteroaryl (C1-5)alkyl, (C9-12)bicycloaryl, hetero(C8-12)bicycloaryl, carbonyl (C1-3)alkyl, thiocarbonyl (C1-3)alkyl, sulfonyl (C1-3)alkyl, sulfinyl (C1-3)alkyl, imino (C1-3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;
R4 is selected from the group consisting of hydrogen, (C1-10)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, arylalkyl, heteroarylalkyl, bicycloaryl, heterobicycloaryl, each substituted or unsubstituted;
each R8 is independently selected from the group consisting of halo, perhalo(C1-10)alkyl, CF3, (C1-10)alkyl, alkenyl, alkynyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, hydroxy, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;
L is a linker providing 0-6 atom separation between X and the ring to which L
is attached; and X is selected from the group consisting of (C1-10)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-
12)bicycloaryl, hetero(C4-12)bicycloaryl, carbonyl (C1-3)alkyl, thiocarbonyl (C1-3)alkyl, sulfonyl (C1-3)alkyl, sulfinyl (C1-3)alkyl, imino (C1-3)alkyl, amino, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, cyano, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.

12. ~A compound comprising Formula XII:
wherein n is 0, 1, 2, or 3;
p is 0-10;
Q is selected from the group consisting of CO, SO, SO2, and C=NR4;
R2 and R3 are each independently selected from the group consisting of hydrogen, halo, perhalo(C1-10)alkyl, amino, cyano, nitro, thin, (C1-10)alkyl, alkene, alkyne, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(C1-10)alkyl, heteroaryl (C1-5)alkyl, (C9-12)bicycloaryl, hetero(C8-12)bicycloaryl, carbonyl (C1-3)alkyl, thiocarbonyl (C1-3)alkyl, sulfonyl (C1-3)alkyl, sulfinyl (C1-3)alkyl, imino (C1-3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;
R4 is selected from the group consisting of hydrogen, (C1-10)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, arylalkyl, heteroarylalkyl, bicycloaryl, heterobicycloaryl, each substituted or unsubstituted;
R5 and R6 are each independently selected from the group consisting of hydrogen, a substituted or unsubstituted (C1-10)alkyl, a substituted or unsubstituted (C1-10)alkoxy, cyano, and halo, or where R5 and R6 are taken together to form a ring;
each R8 is independently selected from the group consisting of halo, perhalo(C1-10)alkyl, CF3, (C1-10)alkyl, alkenyl, alkynyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, hydroxy, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted; and X is selected from the group consisting of (C1-10)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl, hetero(C4-12)bicycloaryl, carbonyl (C1-3)alkyl, thiocarbonyl (C1-3)alkyl, sulfonyl (C1-3)alkyl, sulfinyl (C1-3)alkyl, imino (C1-3)alkyl, amino, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, cyano, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
13. A compound comprising Formula XIII:
wherein m is 0, 1, 2, 3, 4, or 5;
n is 0, 1, 2, or 3;

p is 0-10;
Q is selected from the group consisting of CO, SO, SO2, and C=NR4;
R2 and R3 are each independently selected from the group consisting of hydrogen, halo, perhalo(C1-10)alkyl, amino, cyano, nitro, thio, (C1-10)alkyl, alkene, alkyne, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(C1-10)alkyl, heteroaryl (C1-5)alkyl, (C9-12)bicycloaryl, hetero(C8-12)bicycloaryl, carbonyl (C1-3)alkyl, thiocarbonyl (C1-3)alkyl, sulfonyl (C1-3)alkyl, sulfinyl (C1-3)alkyl, imino (C1-3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;
R4 is selected from the group consisting of hydrogen, (C1-10)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, arylalkyl, heteroarylalkyl, bicycloaryl, heterobicycloaryl, each substituted or unsubstituted;
R5 and R6 are each independently selected from the group consisting of hydrogen, a substituted or unsubstituted (C1-10)alkyl, a substituted or unsubstituted (C1-10)alkoxy, cyano, and halo, or where R5 and R6 are taken together to form a ring;
each R7 is independently selected from the group consisting of halo, perhalo(C1-10)alkyl, CF3, (C1-10)alkyl, alkenyl, alkynyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, hydroxy, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted; and each R8 is independently selected from the group consisting of halo, perhalo(C1-10)alkyl, CF3, (C1-10)alkyl, alkenyl, alkynyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thin, cyano, nitro, hydroxy, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
14. ~A compound comprising one of Formulae XIVa, XIVb and XIVc:

wherein n is 0, 1, 2, or 3;
p is 0-10;
Q is selected from the group consisting of CO, SO, SO2, and C=NR4;
each of T, U, V, W, and Y is independently nitrogen or CR16, provided that no more than two of T, U, V, W, and Y are nitrogen;
R2 and R3 are each independently selected from the group consisting of hydrogen, halo, perhalo(C1-10)alkyl, amino, cyano, nitro, thio, (C1-10)alkyl, alkene, alkyne, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(C1-10)alkyl, heteroaryl (C1-5)alkyl, (C9-12)bicycloaryl, hetero(C8-12)bicycloaryl, carbonyl (C1-3)alkyl, thiocarbonyl (C1-3)alkyl, sulfonyl (C1-3)alkyl, sulfinyl (C1-3)alkyl, imino (C1-3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;
R4 is selected from the group consisting of hydrogen, (C1-10)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, arylalkyl, heteroarylalkyl, bicycloaryl, heterobicycloaryl, each substituted or unsubstituted;
R5 and R6 are each independently selected from the group consisting of hydrogen, a substituted or unsubstituted (C1-10)alkyl, a substituted or unsubstituted (C1-10)alkoxy, cyano, and halo, or where R5 and R6 are taken together to form a ring;
each R8 is independently selected from the group consisting of halo, perhalo(C1-10)alkyl, CF3, (C1-10)alkyl, alkenyl, alkynyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, hydroxy, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted; and each R16 is independently selected from the group consisting of halo, perhalo(C1-10)alkyl, CF3, (C1-10)alkyl, alkenyl, alkynyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, hydroxy, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.~
15. A compound comprising Formula XV:
wherein Q is selected from the group consisting of CO, SO, SO2, and C=NR4;
M is a moiety providing 1-6 atom separation between R19 and the ring to which M is attached;
R2 and R3 are each independently selected from the group consisting of hydrogen, halo, perhalo(C1-10)alkyl, amino, cyano, nitro, thio, (C1-10)alkyl, alkene, alkyne, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(C1-10)alkyl, heteroaryl (C1-5)alkyl, (C9-12)bicycloaryl, hetero(C8-12)bicycloaryl, carbonyl (C1-3)alkyl, thiocarbonyl (C1-3)alkyl, sulfonyl (C1-3)alkyl, sulfinyl (C1-3)alkyl, imino (C1-3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;
R4 is selected from the group consisting of hydrogen, (C1-10)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, arylalkyl, heteroarylalkyl, bicycloaryl, heterobicycloaryl, each substituted or unsubstituted;
R19 comprises a basic nitrogen atom that is capable of interacting with a carboxylic acid side chain of an active site residue of a protein;

L is a linker providing 0-6 atom separation between X and the ring to which L
is attached; and X is selected from the group consisting of (C1-10)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl, hetero(C4-12)bicycloaryl, carbonyl (C1-3)alkyl, thiocarbonyl (C1-3)alkyl, sulfonyl (C1-3)alkyl, sulfinyl (C1-3)alkyl, imino (C1-3)alkyl, amino, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, cyano, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
16. A compound according to claim 15, wherein M provides 1-4 atom separation between R19 and the ring.
17. A compound according to claim 15, wherein M provides 1-3 atom separation between R19 and the ring.
18. A compound according to claim 15, wherein M 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)-.
19. A compound according to any one of claims 15-18, wherein R19 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.
20. A compound according to any one of claims 15-18, wherein R19 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.
21. A compound according to any one of claims 15-18, wherein the basic nitrogen of R19 is separated from the ring atom to which M is attached by between 1-5 atoms.
22. A compound according to any one of claims 15-18, wherein the basic nitrogen of R19 forms part of a primary, secondary, or tertiary amine.
23. A compound according to any one of claims 15-18, wherein R19 is selected from the group consisting of a heterocycloalkyl comprising a nitrogen ring atom or a heteroaryl comprising a nitrogen ring atom.
24. A compound according to claim 15, wherein -MR19 is selected from the group consisting of:

wherein p is 0-12 and each R8 is independently selected from the group consisting of halo, perhalo(C1-10)alkyl, CF3, cyano, nitro, hydroxy, alkyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thin, alkoxy, carbonyl group, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted, with the proviso that at least one R8 includes the basic nitrogen of R19.
25. A compound according to claim 24, wherein at least one R8 is a primary, secondary, or tertiary amine.
26. A compound according to claim 24, 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.
27. A compound according to claim 24, wherein at least one R8 is selected from the group consisting of -NH2, -NH(C1-5 alkyl), -N(C1-5 alkyl) 2, piperazine, imidazole, and pyridine.
28. A compound according to claim 15, wherein -MR19 is selected from the group consisting of:

wherein r is 0-13 and each R8 is independently selected from the group consisting of halo, perhalo(C1-10)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 includes the basic nitrogen of R19.
29. A compound according to claim 28, wherein at least one R8 is a primary, secondary, or tertiary amine.
30. A compound according to claim 28, 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.
31. A compound according to claim 28, wherein at least one R8 is selected from the group consisting of -NH2, -NH(C1-5 alkyl), -N(C1-5 alkyl)2, piperazine, imidazole, and pyridine.
32. A compound according to any one of claims 15-31, wherein L provides 0-3 atom separation between X and the ring.
33. A compound according to any one of claims 15-31, wherein L provides 0 atom separation between X and the ring.
34. A compound according to any one of claims 15-31, wherein X is selected from the group consisting of a substituted or unsubstituted aryl or heteroaryl.
35. A compound comprising Formula XVI:
wherein Q is selected from the group consisting of CO, SO, SO2, and C=NR4;
Z' is a leaving group;
R2 and R3 are each independently selected from the group consisting of hydrogen, halo, perhalo(C1-10)alkyl, amino, cyano, nitro, thio, (C1-10)alkyl, alkene, alkyne, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(C1-10)alkyl, heteroaryl (C1-5)alkyl, (C9-12)bicycloaryl, hetero(C8-12)bicycloaryl, carbonyl (C1-3)alkyl, thiocarbonyl (C1-3)alkyl, sulfonyl (C1-3)alkyl, sulfinyl (C1-3)alkyl, imino (C1-3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;
R4 is selected from the group consisting of hydrogen, (C1-10)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, arylalkyl, heteroarylalkyl, bicycloaryl, heterobicycloaryl, each substituted or unsubstituted;
L is a linker providing 0-6 atom separation between X and the ring to which L
is attached; and X is selected from the group consisting of cycloalkyl, aryl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted, with the provisos that (a) Z' is not alkyl, alkylthio, NNH, NH2, H, H2N-C(O)-NH, cyano, alkoxy, or TMSO when -LX is benzyl and (b) Z' is not halo when -LX is alkyl.
36. A compound comprising Formula XVII:
wherein Q is selected from the group consisting of CO, SO, SO2, and C=NR4;
Z' is a leaving group;
R2 and R3 are each independently selected from the group consisting of hydrogen, halo, perhalo(C1-10)alkyl, amino, cyano, nitro, thio, (C1-10)alkyl, alkene, alkyne, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(C1-10)alkyl, heteroaryl (C1-5)alkyl, (C9-12)bicycloaryl, hetero(C8-12)bicycloaryl, carbonyl (C1-3)alkyl, thiocarbonyl (C1-3)alkyl, sulfonyl (C1-3)alkyl, sulfinyl (C1-3)alkyl, imino (C1-3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted;
R4 is selected from the group consisting of hydrogen, (C1-10)alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, arylalkyl, heteroarylalkyl, bicycloaryl, heterobicycloaryl, each substituted or unsubstituted;
L is a linker providing 0-6 atom separation between X and the ring to which L
is attached; and X is selected from the group consisting of (C1-10)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl, hetero(C4-12)bicycloaryl, carbonyl (C1-3)alkyl, thiocarbonyl (C1-3)alkyl, sulfonyl (C1-3)alkyl, sulfinyl (C1-3)alkyl, imino (C1-3)alkyl, amino, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, cyano, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
37. A compound according to any one of claims 35 and 36, wherein Z' is selected from the group consisting of halo, nitro, azido, phenylsulfoxido, aryloxy, alkoxy, alkylsulfonate, and arylsulfonate.
38. A compound according to any one of claims 35 and 36, wherein Z' is selected from the group consisting of halo, thio, hydroxyl, p-tolylsulphonyloxy and methylsulphonyloxy.
39. A compound according to any one of claims 1, 6,11, and 35-38, wherein L
provides 0-3 atom separation between X and the ring.
40. A compound according to any one of claims 1, 6,11, and 35-38, wherein L
provides 1-3 atom separation between X and the ring.
41. A compound according to any one of claims 1, 6, 11, and 35-38, wherein L
is a linker providing 1, 2, or 3 atom separation between X and the ring to which L is attached, wherein the atoms of the linker providing the separation are selected from the group consisting of carbon, oxygen, nitrogen, and sulfur
42. A compound according to any one of claims 1, 6, 11, and 35-38, wherein L
is a linker providing 1, 2, or 3 atom separation between X and the ring to which L is attached and the 1, 2, or 3 atoms of L providing the separation comprise carbon atoms.
43. A compound according to any one of claims 1, 6, 11, and 35-38, wherein L
is a linker providing 1, 2, or 3 atom separation between X and the ring to which L is attached and the 1, 2, or 3 atoms of L providing the separation are selected from the group of linkers consisting of at least one oxygen or at least one nitrogen atom.
44. A compound according to any one of claims l, 6, 11, and 35-38, wherein L
provides 1 atom separation between X and the ring.
45. A compound according to claim 44, wherein the 1 atom separation is provided by an atom selected from the group consisting of C, N, O, and S.
46. A compound according to claim 44, wherein the 1 atom separation is provided by a carbon atom.
47. A compound according to claim 44, wherein the 1 atom separation is provided by an oxygen atom.
48. A compound according to claim 44, wherein the 1 atom separation is provided by a nitrogen atom.
49. A compound according to any one of claims 1, 6, 11, and 35-38, wherein L
is a carbonyl.
50. A compound according to any one of claims 1, 6, 11, and 35-38, wherein L
is a linker providing 0-6 atom separation between X and the ring to which L is attached, with the proviso that L is not -NH- or -N=CH-.
51. A compound according to any one of claims 1, 6, 11, and 35-38, wherein L
is selected from the group consisting of (C1-10)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl, hetero(C4-12)bicycloaryl, carbonyl (C1-3)alkyl, thiocarbonyl (C1-3)alkyl, sulfonyl (C1-3)alkyl, sulfinyl (Cl_S)alkyl, imino (C1-3)alkyl, amino, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, cyano, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
52. A compound according to any one of claims 1, 6, 11, and 35-38, wherein L
is selected from the group consisting of -, -CHR12-, -C(R12)(R12')-, -C(O)-, -C(S)-, -C(NH)-, -C(NR12)-, -O-, -N(H)-, -N(R12)-, and -S- where R12 and R12' are each independently selected from the group consisting of hydrogen, perhalo(C1-10)alkyl, amino, cyano, (C1-10)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(C1-10)alkyl, heteroaryl (C1-5)alkyl, (C9-12)bicycloaryl, hetero(C8-12)bicycloaryl, carbonyl (C1-3)alkyl, thiocarbonyl (C1-3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, sulfonyl group, sulfinyl group, each substituted or unsubstituted or where or R12 and R12' are taken together to form a 5, 6, or 7 membered ring, each substituted or unsubstituted.
53. A compound according to any one of claims 1, 6, 11, and 35-38, wherein L
is selected from the group consisting of -, CH2-, CH2CH2-, -C(O)-, C(O)-, -C(O)CH2-, -CH2-C(O)CH2-, -C(O)CH2CH2-, CH2C(O)-, -O-, -OCH2-, O-, -CH2OCH2-,, -OCH2CH2-, CH2CH2O-, -N(CH3)-, -NHCH2-, NH2NH-, 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 SC(O)-.
54. A compound according to any one of claims 1, 6, 11, and 35-38, wherein L
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)-.
55. A compound according to any one of claims 1, 2, 6, 7, 11, 12, and 35-53, wherein X is selected from the group consisting of (C1-12)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl, hetero(C4-12)bicycloaryl, carbonyl (C1-3)alkyl, thiocarbonyl (C1-3)alkyl, sulfonyl (C1-3)alkyl, sulfinyl (C1-3)alkyl, imino (C1-3)alkyl, amino, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, cyano, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
56. A compound according to any one of claims 1, 2, 6, 7, 11, 12, and 35-53, wherein X is selected from the group consisting of cycloalkyl, aryl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
57. A compound according to any one of claims 1, 2, 6, 7, 11, 12, and 35-53,wherein X is a substituted or unsubstituted cycloalkyl or heterocycloalkyl.
58. A compound according to any one of claims 1, 2, 6, 7, 11, 12, and 35-53,wherein X is a substituted or unsubstituted (C3-7)cycloalkyl.
59. A compound according to any one of claims 1, 2, 6, 7, 11, 12, and 35-53,wherein X is a substituted or unsubstituted (C3-7)heterocycloalkyl.
60. A compound according to any one of claims 1, 2, 6, 7, 11, 12, and 35-53,wherein X is a substituted or unsubstituted aryl or heteroaryl.
61. A compound according to any one of claims 1, 2, 6, 7, 11, 12, and 35-53,wherein X is a substituted or unsubstituted phenyl.
62. A compound according to any one of claims 1, 2, 6, 7, 11, 12, and 35-53,wherein X is a substituted or unsubstituted heteroaryl.
63. A compound according to any one of claims 1, 2, 6, 7, 11, 12, and 35-53,wherein X is a ring having a non-hydrogen substituent at a 2 or 3 position of the ring.
64. A compound according to any one of claims 1, 2, 6, 7, 11, 12, and 35-53,wherein X
has a non-hydrogen substituent at a 2 or 3 position of the ring wherein the non-hydrogen substituent is selected from the group consisting of (C1-10)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl, hetero(C8-12)bicycloaryl, carbonyl (C1-3)alkyl, thiocarbonyl (C1-3)alkyl, sulfonyl (C1-3)alkyl, sulfinyl (C1-3)alkyl, imino (C1-3)alkyl, amino, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, cyano, nitro, halo, imino group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
65. A compound according to any one of claims 1, 2, 6, 7, 11, 12, and 35-53, wherein X 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)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(C1-7)alkyl)phenyl, (3-CONH(C1-7)alkyl)phenyl, (2-CO2(C1-7)alkyl)phenyl, (3-CO2(C1-7)alkyl)phenyl, -NH2, -OH, -(C3-7)alkyl, -alkene, -alkyne, -CCH, -(C3-7)cycloalkyl, and -aryl, each substituted or unsubstituted.
66. A compound according to any one of claims 1, 2, 6, 7, 11, 12, and 35-53,wherein X is selected from the group consisting of:
wherein A is S, O, or NR21;
B is CR20 or N;
R20 is independently selected from the group consisting of hydrogen, halo, perhalo(C2-10)alkyl, amino, thin, cyano, CF3, nitro, (C1-10)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(C1-10)alkyl, heteroaryl (C1-5)alkyl, (C9-12)bicycloaryl, hetero(C8-12)bicycloaryl, carbonyl (C1-3)alkyl, thiocarbonyl (C1-3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, imino group, carbonyl group, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, sulfinyl group; and R21 is independently selected from the group consisting of hydrogen, perhalo(C1-10)alkyl, amino, cyano, (C1-10)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(C1-10)alkyl, heteroaryl (C1-5)alkyl, (C9-12)bicycloaryl, hetero(C8_12)bicycloaryl, carbonyl (C1-3)alkyl, thiocarbonyl (C1-3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, imino group, carbonyl group, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, sulfinyl group.
67. A compound according to any one of claims 1, 2, 6, 7, 11, 12, and 35-53,wherein X
is selected from the group consisting of:
wherein t is 0, 1, 2, or 3; and each R7 is independently selected from the group consisting of halo, perhalo(C1-10)alkyl, CF3, (C1-10)alkyl, alkenyl, alkynyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thin, cyano, nitro, hydroxy, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
68. A compound according to any one of claims 1, 2, 6, 7, 11, 12, and 35-53, wherein X is selected from the group consisting of:
wherein t is 0, 1, 2, or 3; and each R7 is independently selected from the group consisting of halo, perhalo(C1-10)alkyl, CF3, (C1-10)alkyl, alkenyl, alkynyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thin, cyano, nitro, hydroxy, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
69. A compound according to claim 68, wherein R7 is independently selected from the group consisting of -cyano, -methoxy, -nitro, -carboxy, -sulfonamido, -tetrazolyl, -aminomethyl, -hydroxymethyl, -phenyl, -halo, -CONH2, -CONH(C1-7)alkyl, -CO2(C1-7)alkyl, -NH2, -OH, -(C1-5)alkyl, -alkenyl, -alkynyl, -CCH, (C1-5)cycloalkyl, aryl, heteroaryl, and heterocycloalkyl, each substituted or unsubstituted.
70. A compound according to any one of claims 1, 6, 11, and 35-69, wherein -LX
is -OR11, where R11 is selected from the group consisting of substituted or unsubstituted alkyl, cycloalkyl, aryl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl.
71. A compound according to any one of claims 1, 6, 11, and 35-69, wherein -LX
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)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-hydroxymethyl)phenyl, -(CH2)-(3-hydroxymethyl)phenyl, -(CH2)-(2-phenyl)phenyl, -(CH2)-(3-phenyl)phenyl, -(CH2)-(2-CONH2)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-)alkyl, -CH2-alkene, -CH2-alkyne, -CH2-CCH, -CH2-(C3-7)cycloalkyl, and -CH2-aryl, each substituted or unsubstituted.
72. A compound according to any one of claims 1, 6, 11, and 35-71, wherein -LX
is selected from the group consisting of -(C1)alkyl-aryl, -(C1)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(NR13)-aryl, -C(NR13)-heteroaryl, -C(NR13)-bicycloaryl, -C(NR13)-heterobicycloaryl, each substituted or unsubstituted where R13 is selected from the group consisting of hydrogen, perhalo(C1-10)alkyl, amino, cyano, (C1-10)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(C1-10)alkyl, heteroaryl (C1-5)alkyl, (C9-12)bicycloaryl, hetero(C8-12)bicycloaryl, carbonyl (C1-3)alkyl, thiocarbonyl (C1-3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, sulfonyl group, sulfinyl group, each substituted or unsubstituted.
73. A compound according to any one of claims 1, 2, 3, 4, and 39-72, wherein Z
is selected from the group consisting of (C3_12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(C1-10)alkyl, heteroaryl (C1-5)alkyl, (C9-12)bicycloaryl, and hetero(C8-12)bicycloaryl, each substituted or unsubstituted.
74. A compound according to any one of claims 1, 2, 3, 4, and 39-72, wherein Z
is a substituted or unsubstituted 3, 4, 5, 6, or 7 membered ring.
75. A compound according to any one of claims 1, 2, 3, 4, and 39-72, wherein Z
is a substituted or unsubstituted 3, 4, 5, 6, or 7 membered cycloalkyl.
76. A compound according to any one of claims 1, 2, 3, 4, and 39-72, wherein Z
is a substituted or unsubstituted 4, 5, 6, or 7 membered heterocycloalkyl.
77. A compound according to claim 76, wherein Z comprises O, N(O), N, S, SO, SO2, or a carbonyl group in the ring.
78. A compound according to any one of claims 1, 2, 3, 4, and 39-72, wherein Z
is a substituted or unsubstituted aryl.
79. A compound according to any one of claims 1, 2, 3, 4, and 39-72, wherein Z
is substituted or unsubstituted phenyl.
80. A compound according to any one of claims 1, 2, 3, 4, and 39-72, wherein Z
is a substituted or unsubstituted heteroaryl.
81. A compound according to any one of claims 1, 2, 3, 4, and 39-72, wherein Z
is -NR9R10 where R9 and R10 together are -(CH2)4-6 with or without being interrupted by one O, S, SO, SO2, carbonyl group, N(O), NH, N-(aryl), N-(aryl(C1-5)alkyl), N-(carboxy(C1-5)alkyl), or N-(C1-2)alkyl) group, each substituted or unsubstituted.
82. A compound according to any one of claims 1, 2, 3, 4, and 39-72, wherein Z
is -NR9R10, where R9 is selected from the group consisting of (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C6-12)aryl, hetero(C5-12)aryl, (C9-12)bicycloaryl, each substituted or unsubstituted and hetero(C8-12)bicycloaryl, and R10 is selected from the group consisting of hydrogen and a substituted or unsubstituted (C1-8)alkyl.
83. A compound according to any one of claims 1, 2, 3, 4, and 39-72, wherein Z
is -NR9R10 where R9 and R10 are taken together to form a substituted or unsubstituted 5, 6, or 7 membered heterocycloalkyl, heteroaryl, or heterobicycloaryl.
84. A compound according to claim 83, wherein R9 and R10 are taken together to form a pyrrole, pyrazole, triazole, imidazole, benzimidazole, indole, isoindole, piperidine, piperazine, pyrrolidine, homopiperazine, azetidine, and hexahydroazepine, each substituted or unsubstituted.
85. A compound according to any one of claims 1, 2, 3, 4, and 39-72, wherein Z
is selected from the group consisting of:
wherein p is 0-12 and each R8 is independently selected from the group consisting of halo, perhalo(C1-10)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.
86. A compound according to any one of claims 1, 2, 3, 4, and 39-72, wherein Z
is selected from the group consisting of wherein r is 0-13 and each R8 is independently selected from the group consisting of halo, perhalo(C1-10)alkyl, CF3, cyano, nitro, hydroxy, alkyl, aryl, heteroaryl, anunosulfonyl, 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.
87. A compound according to any one of claims 1, 2, 3, 4, and 39-72, wherein Z
is selected from the group consisting of (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted.
88. A compound according to any one of claims 1, 2, 3, 4, and 39-72, wherein Z
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.
89. A compound according to any one of claims 1, 2, 3, 4, and 39-72, wherein Z
is selected from the group consisting of 3-amino-piperidinyl-1-yl, 3-aminomethyl-pyrrolidin-1-yl, 2-aminoazetidin-1-yl, 3-aminoazetidin-1-yl, 3-amino-3-methylpiperidin-1-yl, 3-aminocyclopent-1-yl, 3-aminomethylcyclopent-1-yl, 3-aminomethylcyclohex-1-yl, aminohexahydroazepin-1-yl, 3-amino-cyclohex-1-yl, piperazin-1-yl, homopiperazin-1-yl, 3-amino-pyrrolidin-1-yl, and R-3-aminopiperidin-1-yl, each substituted or unsubstituted.
90. A compound according to any one of claims 1, 2, 3, 4, and 39-72, wherein Z
is selected from the group consisting of halo, thio, hydroxyl.
91. A compound according to any one of claims 5 and 10, wherein R1 is selected from the group consisting of.

wherein A is S, O, or NR21;

B is CR20 or N;

R20 is independently selected from the group consisting of hydrogen, halo, perhalo(C1-10)alkyl, amino, thin, cyano, CF3, nitro, (C1-10)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(C1-10)alkyl, heteroaryl (C1-5)alkyl, (C9-12)bicycloaryl, hetero(C8-12)bicycloaryl, carbonyl (C1-3)alkyl, thiocarbonyl (C1-3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, imino group, carbonyl group, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, sulfinyl group; and R21 is independently selected from the group consisting of hydrogen, perhalo(C1-10)alkyl, amino, cyano, (C1-10)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(C1-10)alkyl, heteroaryl (C1-5)alkyl, (C9-12)bicycloaryl, hetero(C8-12)bicycloaryl, carbonyl (C1-3)alkyl, thiocarbonyl (C1-3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, imino group, carbonyl group, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, sulfinyl group.
92. A compound according to any one of claims 5 and 10, wherein R1 is selected from the group consisting of:

wherein t is 0, 1, 2, or 3; and each R7 is independently selected from the group consisting of halo, perhalo(C1-10)alkyl, CF3, (C1-10)alkyl, alkenyl, alkynyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, hydroxy, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
93. A compound according to any one of claims 5 and 10, wherein R1 is selected from the group consisting of:

wherein t is 0, 1, 2, or 3; and each R7 is independently selected from the group consisting of halo, perhalo(C1-10)alkyl, CF3, (C1-10)alkyl, alkenyl, alkynyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, hydroxy, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
94. A compound according to any one of claims 2-4, 7-9, 12-14, and 39-93, wherein R5 and R6 are taken together to form a ring.
95. A compound according to any one of claims 2-4, 7-9, 12-14, and 39-93, wherein at least one of R5 and R6 is a substituted or unsubstituted -(C1-8)alkyleneR17, wherein R17 is selected from the group consisting of (C3-12)cycloalkyl, hetero(C4-12)cycloalkyl, (C6-12)aryl, hetero(C5-12)aryl, (C9-12)bicycloalkyl, hetero(C9-12)bicycloalkyl, (C9-12)bicycloaryl and hetero(C8-12)bicycloaryl, each substituted or unsubstituted.
96. A compound according to any one of claims 2-4, 7-9, 12-14, and 39-93, wherein at least one of R5 and R6 is optionally substituted by one to three radicals selected from the group consisting of (C1-6)alkyl, cyano, halo, nitro, halo-substituted(C1-6)alkyl, -NR18R18, -NR18C(O)OR18 -NR18C(O)NR18R18 -NR18C(NR18)NR18R18 -OR18 -SR18 -C(O)OR18, -C(O)NR18R18 -S(O)2NR18R18, -P(O)(OR18)OR18, -OP(O)(OR18)OR18, -NR18C(O)R18, -S(O)R18, -S(O)2R18, -(C1-8)alkyleneC(O)R18, -(C1-8)alkyleneNR18R18, -(C1-8)alkyleneNR18C(O)OR18, -(C1-8)alkyleneNR18C(O)NR18R18, -(C1-8)alkyleneNR18C(NR18)NR18R18, -(C1-8)alkyleneOR18, -(C1-8)alkyleneSR18, -(C1-8)alkyleneC(O)OR18, -(C1-8)alkyleneC(O)NR18R18, -(C1-8)alkyleneS(O)2NR18R18, -(C1-8)alkyleneP(O)(OR18)OR18, -(C1-8)alkyleneOP(O)(OR18)OR18, -(C1-8)alkyleneNR18C(O)R18, -(C1-8)alkyleneS(O)R18, -(C1-8)alkyleneS(O)2R18 and -(C1-8)alkyleneC(O)R18 where each R18 substituent is independently selected from the group consisting of hydrogen and a substituted or unsubstituted (C1-8)alkyl.
97. A compound according to any one of claims 2-4, 7-9, 12-14, and 39-93, wherein at least one of R5 and R6 is optionally substituted by one to three radicals selected from the group consisting of -NH2, -NHC(NH)NH2, -OH, -SH, -C(O)OH and -C(O)NH2.
98. A compound according to any one of claims 2-4, 7-9, 12-14, and 39-93, wherein R5 and R6 are hydrogen.
99. A compound according to claim 98, wherein n is 1 or 2.
100. A compound according to any one of claims 3, 8, 13, and 39-99, wherein R5 and R6 are hydrogen and each R7 is independently selected from the group consisting of halo, perhalo(C1-10)alkyl, CF3, (C1-10)alkyl, alkenyl, alkynyl, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, cycloalkyl, heterocycloalkyl, amino, thio, cyano, nitro, hydroxy, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
101. A compound according to any one of claims 3, 8,13, and 39-99, wherein R5 and R6 are hydrogen and two R7 are taken together to form a substituted or unsubstituted fused or bridged ring.
102. A compound according to any one of claims 3, 8,13, and 39-99, wherein R5 and R6 are hydrogen, n is 1, and R7 is 2-cyano.
103. A compound according to any one of claims 3, 8, 13, and 39-99, wherein:

n is 1, 2, or 3;

R5 and R6 are hydrogen; and each R7 is independently selected from the group consisting of halo, perhalo(C1-10)alkyl, CF3, heteroaryl, aryloxy, heteroaryloxy, cyano, nitro, hydroxy, alkoxy, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
104. A compound according to any one of claims 3, 8, 13, and 39-99, wherein R5 and R6 are hydrogen and R7 is 2-cyano.
105. A compound according to any one of claims 3, 8, 13, and 39-99, wherein two R7 are taken together to form a substituted or unsubstituted fused ring.
106. A compound according to any one of claims 3, 8, 13, and 39-99, wherein two R7 are taken together to form a substituted or unsubstituted bridged ring.
107. A compound according to any one of claims 2, 3, 4, and 39-106, wherein:

n is 1 or 2;

Z is selected from the group consisting of alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, imino group, sulfonyl group and sulfinyl group, and a substituted or unsubstituted 4, 5, 6, or 7 membered ring; and R5 and R6 are hydrogen.
108. A compound according to any one of claims 1-107, wherein Q is selected from the group consisting of CO, SO and SO2.
109. A compound according to any one of claims 1-107, wherein Q is selected from the group consisting of CO and SO2.
110. A compound according to any one of claims 1-107, wherein Q is C=NR4 and R4 is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
111. A compound according to any one of claims 1-110, wherein at least one of R2 and R3 is hydrogen.
112. A compound according to any one of claims 1-110, wherein at least one of R2 and R3 is selected from the group consisting of -NH2, -NHCH3, -N(CH3)2, -NHC1-3-alkyl, and -N(C1-3-alkyl)2, each substituted or unsubstituted.
113. A compound according to any one of claims 1-110, wherein at least one of R2 and R3 is selected from the group consisting of hydrogen, halo, perhalo(C1-10)alkyl, amino, cyano, nitro, thio, (C1-10)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, aryl(C1-10)alkyl, heteroaryl (C1-5)alkyl, (C9-12)bicycloaryl, hetero(C8-12)bicycloaryl, carbonyl (C1-3)alkyl, thiocarbonyl (C1-3)alkyl, sulfonyl (C1-3)alkyl, sulfinyl (C1-3)alkyl, imino (C1-3)alkyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, heteroaryloxy, alkenyl, alkynyl, carbonyl group, imine group, sulfonyl group and sulfinyl group, each substituted or unsubstituted.
114. A compound according to any one of claims 1-110, wherein at least one of R2 and R3 is selected from the group consisting of a (C2-10)alkenyl; an amino, (C1-10)alkyl,or alkoxy carbonyl (C1-3)alkenyl; an amino, (C1-10)alkyl,or alkoxy thiocarbonyl (C1-3)alkenyl; an amino, (C1-10)alkyl,or alkoxy sulfonyl (C1-3)alkenyl; an amino, (C1-10)alkyl,or alkoxy sulfinyl (C1-3)alkenyl; an amino, (C1-10)alkyl,or alkoxy imino (C1-3)alkenyl; an aryl (C1-5)alkenyl; and a heteroaryl (C1-5) alkenyl, each substituted or unsubstituted.
115. A compound according to any one of claims 1-110, wherein at least one of R2 and R3 is selected from the group consisting of a (C2-10)alkynyl; an amino, (C1-10)alkyl,or alkoxy carbonyl (C1-3)alkynyl; an amino, (C1-10)alkyl,or alkoxy thiocarbonyl (C1-3)alkynyl; an amino, (C1-10)alkyl,or alkoxy sulfonyl (C1-3)alkynyl; an amino, (C1-10)alkyl,or alkoxy sulfinyl (C1-3)alkynyl; an amino, (C1-10)alkyl,or alkoxy imino (C1-3)alkynyl; an aryl (C1-5) alkynyl; and heteroaryl (C1-5)alkynyl, each substituted or unsubstituted.
116. A compound according to any one of claims 1-110, wherein at least one of R2 and R3 is substituted or unsubstituted (C3-7)cycloalkyl.
117. A compound according to any one of claims 1-110, wherein at least one of R2 and R3 is substituted or unsubstituted (C3-7)heterocycloalkyl.
118. A compound according to any one of claims 1-110, wherein at least one of R2 and R3 is substituted or unsubstituted aryl.
119. A compound according to any one of claims 1-110, wherein at least one of R2 and R3 is substituted or unsubstituted phenyl.
120. A compound according to any one of claims 1-110, wherein at least one of R2 and R3 is selected from the group consisting of 2-fluorophenyl, 4-fluorostyryl, 2-methoxyphenyl, pyrrolidin-1-yl, imidazolyl, and 3-furanyl, each substituted or unsubstituted.
121. A compound according to any one of claims 1-110, wherein at least one of R2 and R3 is substituted or unsubstituted heteroaryl.
122. A compound according to any one of claims 1-110, wherein at least one of R2 and R3 is selected from the group consisting of hydrogen, cyano, -CF3, or hydroxy.
123. A compound according to any one of claims 1-110, wherein at least one of R2 and R3 is selected from the group consisting of I, Br, Cl, and F.
124. A compound according to any one of claims 1-110, wherein at least one of R2 and R3 is -OR13 where R13 is selected from the group consisting of alkyl, cycloalkyl, aryl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, bicycloaryl, and heterobicycloaryl, each substituted or unsubstituted.
125. A compound according to any one of claims 1-110, wherein at least one of R2 and R3 is a carbonyl group.
126. A compound according to any one of claims 1-110, wherein at least one of R2 and R3 is selected from the group consisting of an aldehyde, acid, amide, and ester.
127. A compound according to any one of claims 1-110, wherein at least one of R2 and R3 is selected from the group consisting of pyrrolidin-1-ylcarbonyl, piperidin-1-ylcarbonyl, and morpholin-4-ylcarbonyl, each substituted or unsubstituted.
128. A compound according to any one of claims 1-110, wherein at least one of R2 and R3 is selected from the group consisting of -SH, -SCH3, and -S(C1-3)alkyl, each substituted or unsubstituted.
129. A compound according to any one of claims 1-110, wherein at least one of R2 and R3 is -NR14R15 where R14 is selected from the group consisting of (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C6-12)aryl, hetero(C5-12)aryl, (C9-12)bicycloaryl and hetero(C8-12)bicycloaryl, each substituted or unsubstituted, and R15 is selected from the group consisting of hydrogen and a substituted or unsubstituted (C1-8)alkyl, and where R14 and R15 together are -(CH2)4-5- optionally interrupted by one O, S, NH, or -N(C1-3)alkyl group, unsubstituted or substituted.
130. A compound according to any one of claims 1-110, wherein at least one of R2 and R3 is selected from the group consisting of a (C1-3)alkyl; an amino, (C1-10)alkyl,or alkoxy carbonyl (C1-3)alkyl; an amino, (C1-10)alkyl,or alkoxy thiocarbonyl (C1-3)alkyl; an amino, (C1-10)alkyl,or alkoxy sulfonyl (C1-3)alkyl; an amino, (C1-10)alkyl,or alkoxy sulfinyl (C1-3)alkyl; an amino, (C1-10)alkyl,or alkoxy imino (C1-3)alkyl; an aryl (C1-5)alkyl; and a heteroaryl (C1-5)alkyl, each unsubstituted or substituted.
131. A compound according to any one of claims 1-110, wherein at least one of R2 and R3 is selected from the group consisting of 1,2-dihydro-2-oxo-pyridinyl-, 1,4-dihydro-4-oxo-pyridinyl-, 2,3-dihydro-3-oxo-pyridazinyl-, 1,2,3,6-tetrahydro-3,6-dioxo-pyridazinyl-, 1,2-dihydro-2-oxo-pyrimidinyl-, 3,4-dihydro-4-oxo-pyrimidinyl-, 1,2,3,4-tetrahydro-2,4-dioxo-pyrimidinyl-, 1,2-dihydro-2-oxo-pyrazinyl, 1,2,3,4-tetrahydro-2,3-dioxo-pyrazinyl-, 2,3-dihydro-2-oxo-indolyl-, 2,3-dihydrobenzofuranyl-, 2,3-dihydro-2-oxo-1H-benzimidazolyl-, 2,3-dihydro-2-oxo-benzoxazolyl-, 1,2-dihydro-2-oxo-quinolinyl-, 1,4-dihydro-4-oxo-quinolinyl-,1,2-dihydro-1-oxo-iso-quinolinyl-,1,4-dihydro-4-oxo-cinnolinyl-,1,2-dihydro-2-oxo-quinazolinyl-, 1,4-dihydro-4-oxo-quinazolinyl-, 1,2,3,4-tetrahydro-2,4-dioxo-quinazolinyl-,1,2-dihydro-2-oxo-quinoxalinyl-,1,2,3,4-tetrahydro-2,3-dioxo-quinoxalinyl-, 1,2-dihydro-1-oxo-phthalazinyl-, 1,2,3,4-tetrahydro-1,4-dioxo-phthalazinyl-, chromanyl-, cumarinyl-, 2,3-dihydro-benzo[1,4]dioxinyl-, and 3,4-dihydro-3-oxo-2H-benzo[1,4]oxazinyl-, each unsubstituted or substituted.
132. A compound according to any one of claims 1-110, wherein at least one of R2 and R3 is not hydrogen.
133. A compound according to any one of claims 1-110, wherein both R2 and R3 are not hydrogen.
134. A compound according to any one of claims 1-110, wherein R2 is selected from the group consisting of 2-fluorophenyl, styryl, 4-fluorostyryl, 2-methoxyphenyl, pyrrolidin-1-yl, imidazolyl, and 3-furanyl each unsubstituted or substituted, and R3 is selected from the group consisting of hydrogen, (C1-10)alkyl, and aryl, each unsubstituted or substituted.
135. A compound according to any one of claims 1-110, wherein R2 is selected from the group consisting of hydrogen, carboxyl, cyano, nitro, bromo, fluoro, chloro, iodo, -CF3, -CH3 and hydroxy, and R3 is selected from the group consisting of hydrogen, (C1-10)alkyl, and aryl, each unsubstituted or substituted.
136. A compound according to any one of claims 1-110, wherein R2 is selected from the group consisting of hydrogen, carboxyl, cyano, nitro, bromo, fluoro, chloro, iodo, -CF3, -CH3 and hydroxy, and R3 is selected from the group consisting of hydrogen, (C3-7)cycloalkyl and aryl, each unsubstituted or substituted.
137. A compound according to any one of claims 1, 2, 3, 4, and 39-136, wherein:

R2 or R3 is selected from the group consisting of -NH2, -NHCH3, -N(CH3)2, -NHC1-3-alkyl, and -N(C1-3-alkyl)2, each unsubstituted or substituted; and Z is selected from the group consisting of 3-amino-piperidinyl-1-yl, 3-aminomethyl-pyrrolidin-1-yl, 2-aminoazetidin-1-yl, 3-aminoazetidin-1-yl, 3-amino-3-methylpiperidin-1-yl, 3-aminocyclopentyl, 3-aminomethylcyclopentyl, 3-aminomethylcyclohexyl, 3-aminohexahydroazepin-1-yl, 3-amino-cyclohexyl, piperazin-1-yl, homopiperazin-1-yl, 3-amino-pyrrolidin-1-yl, and R-3-aminopiperidin-1-yl, each unsubstituted or substituted.
138. A compound according to any one of claims 5, 10, and 108-136, wherein:

Q is CO; and R1 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)-(2-alkynyl)phenyl, -(CH2)-(2-methoxy)phenyl, -(CH2)-(3-methoxy)phenyl, -(CH2)-(2-nitro)phenyl, -(CH2)-(3-nitro)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-aminomethyl)phenyl, -(CH2)-(2-amino)phenyl, -(CH2)-(3-amino)phenyl, -(CH2)-(3-aminomethyl)phenyl, -(CH2)-(2-hydroxymethyl)phenyl, -(CH2)-(3-hydroxymethyl)phenyl, -(CH2)-(2-phenyl)phenyl, -(CH2)-(3-phenyl)phenyl, -(CH2)-(2-halo)phenyl, -(CH2)-(3-halo)phenyl, -(CH2)-(2-CONH2)phenyl, -(CH2)-(2,-CONH(C1-7)alkyl)phenyl, -(CH2)-(2-CO2(C1-7)alkyl)phenyl, -CH2-NH2, -CH2-OH, -CH2-(C3-7)alkyl, -CH2-alkene, -CH2-alkyne, -CH2-CCH, -CH2-(C3-7)cycloalkyl, -CH2-aryl, -CH2-heteroaryl, and -CH2-heterocycloalkyl, each substituted or unsubstituted.
139. A compound according to any one of claims 1, 2, 3, 4, and 39-136, wherein:
Q is CO; and Z is selected from the group consisting of 3-amino-piperidinyl-1-yl, 3-aminomethyl-pyrrolidin-1-yl, 2-aminoazetidin-1-yl, 3-aminoazetidin-1-yl, 3-amino-3-methylpiperidin-1-yl, 3-aminocyclopentyl, 3-aminomethylcyclopentyl, 3-aminomethylcyclohexyl, 3-aminohexahydroazepin-1-yl, 3-amino-cyclohexyl, piperazin-1-yl, homopiperazin-1-yl, 3-amino-pyrrolidin-1-yl, and R-3-aminopiperidin-1-yl, each unsubstituted or substituted.
140. A compound according to any one of claims 5, 10, and 108-136, wherein:
Q is SO; and R1 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)-(2-alkynyl)phenyl, -(CH2)-(2-methoxy)phenyl, -(CH2)-(3-methoxy)phenyl, -(CH2)-(2-nitro)phenyl, -(CH2)-(3-nitro)phenyl, -(CH2)-(2-carboxy)phenyl, -(CH2)-(3-carboxy)phenyl, -(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)-(2-amino)phenyl, -(CH2)-(3-amino)phenyl, -(CH2)-(3-aminomethyl)phenyl, -(CH2)-(2-hydroxymethyl)phenyl, -(CH2)-(3-hydroxymethyl)phenyl, -(CH2)-(2-phenyl)phenyl, -(CH2)-(3-phenyl)phenyl, -(CH2)-(2-halo)phenyl, -(CH2)-(3-halo)phenyl, -(CH2)-(2-CONH2)phenyl, -(CH2)-(2-CONH(C1-7)alkyl)phenyl, -(CH2)-(2-CO2(C1-7)alkyl)phenyl, -CH2-NH2, -CH2-OH, -CH2-(C3-)alkyl, -CH2-alkene, -CH2-alkyne, -CH2-CCH, -CH2-(C3-7)cycloalkyl, -CH2-aryl, -CH2-heteroaryl, and -CH2-heterocycloalkyl, each substituted or unsubstituted.
141. A compound according to any one of claims 1, 2, 3, 4, and 39-136, wherein:
Q is SO; and Z is selected from the group consisting of 3-amino-piperidinyl-1-yl, 3-aminomethyl-pyrrolidin-1-yl, 2-aminoazetidin-1-yl, 3-aminoazetidin-1-yl, 3-amino-3-methylpiperidin-1-yl, 3-aminocyclopent-1-yl, 3-aminomethylcyclopent-1-yl, 3-aminomethylcyclohex-1-yl, 3-aminohexahydroazepin-1-yl, 3-amino-cyclohex-1-yl, piperazin-1-yl, homopiperazin-1-yl, 3-amino-pyrrolidin-1-yl, and R-3-aminopiperidin-1-yl, each unsubstituted or substituted.
142. A compound according to any one of claims 5, 10, and 108-136, wherein:
Q is SO2; and R1 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)-(2-alkynyl)phenyl, -(CH2)-(2-methoxy)phenyl, -(CH2)-(3-methoxy)phenyl, -(CH2)-(2-nitro)phenyl, -(CH2)-(3-nitro)phenyl, -(CH2)-(2-carboxy)phenyl, -(CH2)-(3-carboxy)phenyl, -(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-hydroxymethyl)phenyl, -(CH2)-(3-hydroxymethyl)phenyl, -(CH2)-(2-phenyl)phenyl, -(CH2)-(3-phenyl)phenyl, -(CH2)-(2-halo)phenyl, -(CH2)-(3-halo)phenyl, -(CH2)-(2-CONH2)phenyl, -(CH2)-(2-CONH(C1-7)alkyl)phenyl, -(CH2)-(2-CO2(C1-7)alkyl)phenyl, -CH2-NH2, -CH2-OH, -CH2-(C3-)alkyl, -CH2-alkene, -CH2-alkyne, -CH2-CCH, -CH2-(C3-7)cycloalkyl, -CH2-aryl, -CH2-heteroaryl, and -CH2-heterocycloalkyl, each substituted or unsubstituted.
143. A compound according to any one of claims 1, 2, 3, 4, and 39-136, wherein:
Q is SO2; and Z is selected from the group consisting of 3-amino-piperidinyl-1-yl, 3-aminomethyl-pyrrolidin-1-yl, 2-aminoazetidin-1-yl, 3-aminoazetidin-1-yl, 3-amino-3-methylpiperidin-1-yl, 3-aminocyclopent-1-yl, 3-aminomethylcyclopent-1-y1, 3-aminomethylcyclohex-1-yl, 3-aminohexahydroazepin-1-yl, 3-amino-cyclohex-1-yl, piperazin-1-yl, homopiperazin-1-yl, 3-amino-pyrrolidin-1-yl, and R-3-aminopiperidin-1-yl, each unsubstituted or substituted.
144. A compound according to claim 1, wherein:
Q is CO;
-L-X together are 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)-(2-alkynyl)phenyl, -(CH2)-(2-methoxy)phenyl, -(CH2)-(3-methoxy)phenyl, -(CH2)-(2.-nitro)phenyl, -(CH2)-(3-nitro)phenyl, -(CH2)-(2-carboxy)phenyl, -(CH2)-(3-carboxy)phenyl, -(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-hydroxymethyl)phenyl, -(CH2)-(3-hydroxymethyl)phenyl, -(CH2)-(2-phenyl)phenyl, -(CH2)-(3-phenyl)phenyl, -(CH2)-(2-halo)phenyl, -(CH2)-(3-halo)phenyl, -(CH2)-(2-CONH2)phenyl, -(CH2)-(2-CONH(C1-7)alkyl)phenyl, -(CH2)-(2-CO2(C1-7)alkyl)phenyl, -CH2-NH2, -CH2-OH, -CH2-(C3-7)alkyl, -CH2-alkene, -CH2-alkyne, -CH2-CCH, -CH2-(C3-7)cycloalkyl, -CH2-aryl, -CH2-heteroaryl, and -CH2-heterocycloalkyl, each substituted or unsubstituted; and Z is selected from the group consisting of 3-amino-piperidinyl-1-yl, 3-aminomethyl-pyrrolidin-1-yl, 2-aminoazetidin-1-yl, 3-aminoazetidin-1-yl, 3-amino-3-methylpiperidin-1-yl, 3-aminocyclopent-1-yl, 3-aminomethylcyclopent-1-yl, 3-aminomethylcyclohex-1-yl, 3-aminohexahydroazepin-1-yl, 3-amino-cyclohex-1-yl, piperazin-1-yl, homopiperazin-1-yl, 3-amino-pyrrolidin-1-yl, and R-3-aminopiperidin-1-yl, each unsubstituted or substituted.
145. A compound according to claim 1, wherein:
Q is SO;
-L-X together are 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)-(2-alkynyl)phenyl, -(CH2)-(2-methoxy)phenyl, -(CH2)-(3-methoxy)phenyl, -(CH2)-(2-nitro)phenyl, -(CH2)-(3-nitro)phenyl, -(CH2)-(2-carboxy)phenyl, -(CH2)-(3-carboxy)phenyl, -(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-hydroxymethyl)phenyl, -(CH2)-(3-hydroxymethyl)phenyl, -(CH2)-(2-phenyl)phenyl, -(CH2)-(3-phenyl)phenyl, -(CH2)-(2,-halo)phenyl, -(CH2)-(3-halo)phenyl, -(CH2)-(2-CONH2)phenyl, -(CH2)-(2-CONH(C1-7)alkyl)phenyl, -(CH2)-(2-CO2(C1-7)alkyl)phenyl, -CH2-NH2, -CH2-OH, -CH2-(C3-)alkyl, -CH2-alkene, -CH2-alkyne, -CH2-CCH, -CH2-(C3-7)cycloalkyl, -CH2-aryl, -CH2-heteroaryl, and -CH2-heterocycloalkyl, each substituted or unsubstituted; and Z is selected from the group consisting of 3-amino-piperidinyl-1-yl, 3-aminomethyl-pyrrolidin-1-yl, 2-aminoazetidin-1-yl, 3-aminoazetidin-1-yl, 3-amino-3-methylpiperidin-1-yl, 3-aminocyclopent-1-yl, 3-aminomethylcyclopent-1-yl, 3-aminomethylcyclohex-1-yl, 3-aminohexahydroazepin-1-yl, 3-amino-cyclohex-1-yl, piperazin-1-yl, homopiperazin-1-yl, 3-amino-pyrrolidin-1-yl, and R-3-aminopiperidin-1-yl, each unsubstituted or substituted.
146. A compound according to claim 1, wherein:
Q is 502;
-L-X together are 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)-(2-alkynyl)phenyl, -(CH2)-(2-methoxy)phenyl, -(CH2)-(3-methoxy)phenyl, -(CH2)-(2-nitro)phenyl, -(CH2)-(3-nitro)phenyl, -(CH2)-(2-carboxy)phenyl, -(CH2)-(3-carboxy)phenyl, -(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-hydroxymethyl)phenyl, -(CH2)-(3-hydroxymethyl)phenyl, -(CH2)-(2-phenyl)phenyl, -(CH2)-(3-phenyl)phenyl, -(CH2)-(2-halo)phenyl, -(CH2)-(3-halo)phenyl, -(CH2)-(2-CONH2)phenyl, -(CH2)-(2-CONH(C1-7)alkyl)phenyl, -(CH2)-(2-CO2(C1-7)alkyl)phenyl, -CH2-NH2, -CH2-OH, -CH2-(C3-7)alkyl, -CH2-alkene, -CH2-alkyne, -CH2-CCH, -CH2-(C3-7)cycloalkyl, -CH2-aryl, -CH2-heteroaryl, and -CH2-heterocycloalkyl, each substituted or unsubstituted; and Z is selected from the group consisting of 3-amino-piperidinyl-1-yl, 3-aminomethyl-pyrrolidin-1-yl, 2-aminoazetidin-1-yl, 3-aminoazetidin-1-yl, 3-amino-3-methylpiperidin-1-yl, 3-aminocyclopent-1-yl, 3-aminomethylcyclopent-1-yl, 3-aminomethylcyclohex-1-yl, 3-aminohexahydroazepin-1-yl, 3-amino-cyclohex-1-yl, piperazin-1-yl, homopiperazin-1-yl, 3-amino-pyrrolidin-1-yl, and R-3-aminopiperidin-1-yl, each unsubstituted or substituted.
147. A compound selected from the group consisting of:
5-Bromo-2-chloro-3H pyrimidin-4-one;
5-Bromo-2-chloro-6-methyl-3H pyrimidin-4-one;
2-Chloro-5-ethyl-3H-pyrimidin-4-one;
2,5-Dichloro-3H-pyrimidin-4-one;
2-Chloro-5-methoxy-3H-pyrimidin-4-one; and 2-Chloro-5-fluoro-3H pyrimidin-4-one.
148. A compound selected from the group consisting of:
2-(5-Bromo-2-chloro-6-oxo-6H pyrimidin-1-ylmethyl)-benzonitrile;
2-[2-(3-Amino-piperidin-1-yl)-5-trimethylsilylethynyl-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile;
2-(5-Bromo-2-chloro-4-methyl-6-oxo-6H pyrimidin-1-ylmethyl)-benzonitrile;
2-(2-Chloro-4,5-dimethyl-6-oxo-6H-pyrimidin-1ylmethyl)-benzonitrile;
2-(2-Chloro-5-ethyl-6-oxo-6H-pyrimidin-1-ylmethyl)-benzonitrile;
2-(2,5-Dichloro-6-oxo-6H pyrimidin-1-ylmethyl)-benzonitrile;
2-((5-fluoro-2-chloro-6-oxopyrimidin-1-(6H)-yl)methyl)-5-fluorobenzonitrile;
2-((5-fluoro-2-chloro-6-oxopyrimidin-1-(6H)-yl)methyl)-4-fluorobenzonitrile;
2-((5-bromo-2-chloro-6-oxopyrimidin-1-(6H)-yl)methyl)-4-fluorobenzonitrile;
2-(5-Bromo-2-chloro-6-oxo-6H pyrimidin-1ylmethyl)-thiophene-3-carbonitrile;
2-(2-Chloro-5-methoxy-6-oxo-6H pyrimidin-1-ylmethyl)-benzonitrile;
2.-(2-Chloro-5-fluoro-6-oxo-6H pyrimidin-1-ylmethyl)-benzonitrile;
3-(2-Bromo-benzyl)-2-chloro-5-fluoro-3H-pyrimidin-4-one; and 2-(2-Chloro-6-oxo-4-phenyl-6H-pyrimidin-1-ylmethyl)-benzonitrile.
149. A compound selected from the group consisting of:
2-[2-(3-(R)-Amino-piperidin-1-yl)-6-oxo-5-phenyl-6H-pyrimidin-1-ylmethyl]-benzonitrile;

2-[2-(3-(R)-Amino-piperidin-1-yl)-5-furan-3-yl-6-oxo-6H pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-(2-fluoro-phenyl)-6-oxo-6H pyrimidin-1-ylmethyl]-benzonitrile;
2-{2-(3-(R)-Amino-piperidin-1-yl)-5-[2-trans-(4-fluoro-phenyl)-vinyl]-6-oxo-6H
pyrimidin-1ylmethyl}-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-(2-methoxy-phenyl)-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-bromo-4-methyl-6-oxo-6H pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-4-methyl-6-oxo-5-phenyl-6H pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-furan-3-yl-4-methyl-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-4-methyl-6-oxo-5-trans-styryl-6H-pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-4,5-dimethyl-6-oxo-6H pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-ethyl-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-iodo-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-bromo-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-ethynyl-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-chloro-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile;
(R)-2-((2-(3-aminopiperidin-1-yl)-6-oxo-5-(pyrrolidin-1-yl)pyrimidin-1(6H)-yl)methyl)benzonitrile;

(R)-2-((2-(3-aminopiperidin-1-yl)-5-fluoro-6-oxopyrimidin-1(6H)-yl)methyl)-5-fluorobenzonitrile;
(R)-2-((2-(3-aminopiperidin-1-yl)-5-fluoro-6-oxopyrimidin-1(6H)-yl)methyl)-4-fluorobenzonitrile;
(R)-2-((2-(3-aminopiperidin-1-yl)-5-chloro-6-oxopyrimidin-1(6H)-yl)methyl)-4-fluorobenzonitrile;
(R)-2-((2-(3-aminopiperidin-1-yl)-5-bromo-6-oxopyrimidin-1(6H)-yl)methyl)-4-fluorobenzonitrile;
(R)-2-((2-(3-amino-3-methylpiperidin-1-yl)-5-fluoro-6-oxopyrimidin-1(6H)-yl)methyl)-4-fluorobenzonitrile;
2-[2-(3-(R)-Amino-pyrimidin-1-yl)-5-bromo-6-oxo-6H-pyrimidin-1-ylmethyl]-thiophene-3-carbonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-(3,3-dimethyl-but-1-ynyl)-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-prop-1-ynyl-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-(3-hydroxy-prop-1-ynyl)-6-oxo-6H-pyrimidin-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-6-oxo-5-vinyl-6H-pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-methoxy-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-6-oxo-5-(1H-pyrrol-3-yl)-6H-pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-fluoro-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile;
2-(3-(R)-Amino-piperidin-1-yl)-3-(2-bromo-benzyl)-5-fluoro-3H pyrimidin-4-one;
2-[2-(3-(R)-Amino-piperidin-1-yl)-6-oxo-5-((E)-2-pyridin-3-yl-vinyl)-6H-pyrimidin-1-ylmethyl]-benzonitrile;

2-[2-(3-(R)-Amino-piperidin-1-yl)-6-oxo-4-phenyl-6H-pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-3-methyl-piperidin-1-yl)-5-fluoro-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-3-methyl-piperidin-1-yl)-5-chloro-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile; and (R)-2-((2-(3-aminopiperidin-1-yl)-5-bromo-4-tert-butyl-6-oxopyrimidin-1-(6H)-yl)methyl)benzonitrile.
150. A compound selected from the group consisting of:
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-bromo-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-ethynyl-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile;
2.-[2-(3-(R)-Amino-piperidin-1-yl)-4,5-dimethyl-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-chloro-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile;
(R)-2-((2-(3-aminopiperidin-1-yl)-5-fluoro-6-oxopyrimidin-1-(6H)-yl)methyl)-4-fluorobenzonitrile;
(R)-2-((2-(3-aminopiperidin-1-yl)-5-chloro-6-oxopyrimidin-1-(6H)-yl)methyl)-4-fluorobenzonitrile;
(R)-2.-((2-(3-aminopiperidin-1-yl)-5-bromo-6-oxopyrimidin-1-(6H)-yl)methyl)-4-fluorobenzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-prop-1-ynyl-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-fluoro-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile; and 2-[2-(3-(R)-Amino-3-methyl-piperidin-1-yl)-5-chloro-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile.
151. A compound according to any one of claims 1-150, wherein the compound is in the form of a pharmaceutically acceptable salt.
152. A compound according to claim 151, wherein the compound is in the form of a pharmaceutically acceptable salt selected from the group consisting of a TFA
salt, a tartaric acid salt, and an HC1 salt.
153. A compound selected from the group consisting of:
2-[2-(3-(R)-Amino-piperidin-1-yl)-6-oxo-5-phenyl-6H-pyrimidin-1-ylmethyl]-benzonitrile, TFA salt;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-furan-3-yl-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile, TFA salt;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-(2-fluoro-phenyl)-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile, TFA salt;
2-{2-(3-(R)-Amino-piperidin-1-yl)-5-[2-trans-(4-fluoro-phenyl)-vinyl]-6-oxo-6H-pyrimidin-1ylmethyl}-benzonitrile, TFA salt;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-(2-methoxy-phenyl)-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile, TFA salt;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-bromo-4-methyl-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile, TFA salt;
2-[2-(3-(R)-Amino-piperidin-1-yl)-4-methyl-6-oxo-5-phenyl-6H-pyrimidin-1-ylmethyl]-benzonitrile, TFA salt;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-furan-3-yl-4-methyl-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile, TFA salt;
2-[2-(3-(R)-Amino-piperidin-1-yl)-4-methyl-6-oxo-5-trans-styryl-6H-pyrimidin-1-ylmethyl]-benzonitrile, TFA salt;
2-[2-(3-(R)-Amino-piperidin-1-yl)-4,5-dimethyl-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile, TFA salt;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-ethyl-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile, TFA salt;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-iodo-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile, TFA salt;

2-[2-(3-(R)-Amino-piperidin-1-yl)-5-bromo-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile, TFA salt;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-ethynyl-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-chloro-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile, TFA salt;
(R)-2-((2-(3-aminopiperidin-1-yl)-6-oxo-5-(pyrrolidin-1-yl)pyrimidin-1 (6H)-yl)methyl)benzonitrile, TFA salt;
(R)-2-((2-(3-aminopiperidin-1-yl)-5-fluoro-6-oxopyrimidin-1-(6H)-yl)methyl)-5-fluorobenzonitrile, TFA salt;
(R)-2-((2-(3-aminopiperidin-1-yl)-5-fluoro-6-oxopyrimidin-1-(6H)-yl)methyl)-4-fluorobenzonitrile, TFA salt;
(R)-2-((2-(3-aminopiperidin-1-yl)-5-chloro-6-oxopyrimidin-1-(6H)-yl)methyl)-4-fluorobenzonitrile, TFA salt;
(R)-2-((2-(3-aminopiperidin-1-yl)-5-bromo-6-oxopyrimidin-1-(6H)-yl)methyl)-4-fluorobenzonitrile, TFA salt;
(R)-2-((2-(3-amino-3-methylpiperidin-1-yl)-5-fluoro-6-oxopyrimidin-1-(6H)-yl)methyl)-4-fluorobenzonitrile, TFA salt;
2-[2-(3-(R)-Amino-pyrimidin-1-yl)-5-bromo-6-oxo-6H-pyrimidin-1-ylmethyl]-thiophene-3-carbonitrile, TFA salt;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-(3,3-dimethyl-but-1-ynyl)-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-prop-1-ynyl-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-(3-hydroxy-prop-1-ynyl)-6-oxo-6H-pyrimidin-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile, TFA
salt;
2-[2-(3-(R)-Amino-piperidin-1-yl)-6-oxo-5-vinyl-6H-pyrimidin-1-ylmethyl]-benzonitrile, TFA salt;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-methoxy-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile, TFA salt;
2-[2-(3-(R)-Amino-piperidin-1-yl)-6-oxo-5-(1H-pyrrol-3-yl)-6H-pyrimidin-1-ylmethyl]-benzonitrile;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-fluoro-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile, TFA salt;
2-[2-(3-(R)-Amino-piperidin-1-yl)-5-fluoro-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile, tartaric acid salt;
2-(3-(R)-Amino-piperidin-1-yl)-3-(2-bromo-benzyl)-5-fluoro-3H-pyrimidin-4-one, HCl salt;
2-[2-(3-(R)-Amino-piperidin-1-yl)-6-oxo-5-((E)-2-pyridin-3-yl-vinyl)-6H-pyrimidin-1-ylmethyl]-benzonitrile, HCl salt;
2-[2-(3-(R)-Amino-piperidin-1-yl)-6-oxo-4-phenyl-6H-pyrimidin-1-ylmethyl]-benzonitrile, HCl salt;
2-[2-(3-(R)-Amino-3-methyl-piperidin-1-yl)-5-fluoro-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile, HCl salt;
2-[2-(3-(R)-Amino-3-methyl-piperidin-1-yl)-5-chloro-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile, HCl salt; and (R)-2-((2-(3-aminopiperidin-1-yl)-5-bromo-4-tert-butyl-6-oxopyrimidin-1(6H)-yl)methyl)benzonitrile, TFA salt.
154. A compound according to any one of claims 1-153, wherein the compound is present in a mixture of stereoisomers.
155. A compound according to any one of claims 1-153, wherein the compound comprises a single stereoisomer.
156. A pharmaceutical composition comprising, as an active ingredient, a compound according to any one of claims 1-155.
157. A pharmaceutical composition according to claim 156, wherein the composition is a solid formulation adapted for oral administration.
158. A pharmaceutical composition according to claim 156, wherein the composition is a tablet.
159. A pharmaceutical composition according to claim 156, wherein the composition is a liquid formulation adapted for oral administration.
160. A pharmaceutical composition according to claim 156, wherein the composition is a liquid formulation adapted for parenteral administration.
161. A pharmaceutical composition comprising a compound according to any one of claims 1-155, 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, subcutaneously, intraadiposally, intraarticularly, and intrathecally.
162. A kit comprising:
a compound according to any one of claims 1-155; 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.
163. A kit according to claim 162, wherein the kit comprises the compound in a multiple dose form.
164. An article of manufacture comprising:
a compound according to any one of claims 1-155; and packaging materials.
165. An article of manufacture according to claim 164, wherein the packaging material comprises a container for housing the compound.
166. An article of manufacture according to claim 165, 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.
167. An article of manufacture according to claim 164, wherein the article of manufacture comprises the compound in a multiple dose form.
168. A method of inhibiting DPP-IV comprising:
contacting DPP-IV with a compound according to any one of claims 1-155.
169. A method of inhibiting DPP-IV comprising:
causing a compound according to any one of claims 1-155 to be present in a subject in order to inhibit DPP-IV in vivo.
170. A method of inhibiting DPP-IV comprising:
administering a first compound to a subject that is converted in vivo to a second compound wherein the second compound inhibits DPP-IV in vivo, the second compound being a compound according to any one of claims 1-155.
171. A therapeutic method comprising:
administering a compound according to any one of claims 1-155 to a subject.
172. A method of treating a disease state for which DPP-IV possesses activity that contributes to the pathology and/or symptomology of the disease state, the method comprising causing a compound according to any one of claims 1-155 to be present in a subject in a therapeutically effective amount for the disease state.
173. A method according to claim 172, wherein the disease is type I or type II
diabetes.
174. 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 any one of claims 1-155.
175. The method according to claim 174, wherein the cancer treated is colorectal, prostate, breast, thyroid, skin, lung, or head and neck.
176. A method of treating autoimmune disorders in a patient in need thereof, comprising administering to said patient a therapeutically effective amount of a compound according to any one of claims 1-155.
177. A method according to claim 176, wherein the autoimmune disorder is selected from the group consisting of rheumatoid arthritis, psoriasis, and multiple sclerosis.
178. 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 any one of claims 1-155.
179. A method according to claim 178, wherein said condition is a side effect of chemotherapy or radiation therapy.
180. A method according to claim 178, wherein said condition is a result of kidney failure.
181. A method according to claim 178, wherein said condition is a result of a bone marrow disorder.
182. A method of treating HIV infection in a patient in need thereof, comprising administering to said patient a therapeutically effective amount of a compound according to any one of claims 1-155.
183. A method of treating a condition characterized by immunodeficiency symptoms in a patient in need thereof, comprising administering to said patient a therapeutically effective amount of a compound according to any one of claims 1-155.~~
184
CA002535619A 2003-08-13 2004-08-12 4-pyrimidone derivatives and their use as peptidyl peptidase inhibitors Abandoned CA2535619A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US49523803P 2003-08-13 2003-08-13
US60/495,238 2003-08-13
PCT/US2004/026265 WO2005016911A1 (en) 2003-08-13 2004-08-12 4-pyrimidone derivatives and their use as peptidyl peptidase inhibitors

Publications (1)

Publication Number Publication Date
CA2535619A1 true CA2535619A1 (en) 2005-02-24

Family

ID=33564066

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002535619A Abandoned CA2535619A1 (en) 2003-08-13 2004-08-12 4-pyrimidone derivatives and their use as peptidyl peptidase inhibitors

Country Status (17)

Country Link
US (6) US7790736B2 (en)
EP (1) EP1506967B1 (en)
JP (1) JP4854943B2 (en)
KR (1) KR20060041309A (en)
CN (1) CN1867560A (en)
AR (1) AR045455A1 (en)
AU (1) AU2004265341A1 (en)
BR (1) BRPI0413452A (en)
CA (1) CA2535619A1 (en)
DE (1) DE602004010206T2 (en)
IL (1) IL173490A0 (en)
MX (1) MXPA06001601A (en)
NO (1) NO20061157L (en)
RU (1) RU2006107553A (en)
TW (1) TW200524895A (en)
WO (1) WO2005016911A1 (en)
ZA (1) ZA200602051B (en)

Families Citing this family (57)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8025566B2 (en) * 2003-04-16 2011-09-27 Igt Gaming device methods and apparatus employing audio/video programming outcome presentation
WO2003002531A2 (en) 2001-06-27 2003-01-09 Smithkline Beecham Corporation Fluoropyrrolidines as dipeptidyl peptidase inhibitors
WO2004087053A2 (en) 2003-03-25 2004-10-14 Syrrx, Inc. Dipeptidyl peptidase inhibitors
US7790736B2 (en) 2003-08-13 2010-09-07 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
US7790734B2 (en) 2003-09-08 2010-09-07 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
CN102127057A (en) * 2004-03-15 2011-07-20 武田药品工业株式会社 Dipeptidyl peptidase inhibitors
WO2005113538A1 (en) * 2004-05-19 2005-12-01 Basf Aktiengesellschaft 2-substituted pyrimidines and their use as pesticides
WO2006068978A2 (en) 2004-12-21 2006-06-29 Takeda Pharmaceutial Company Limited Dipeptidyl peptidase inhibitors
JP2008539235A (en) 2005-04-29 2008-11-13 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 Peptides and peptidomimetics for treating disease states characterized by inflammatory responses
US8017624B2 (en) * 2005-08-26 2011-09-13 Merck Sharp & Dohme Corp. Fused aminopiperidines as dipeptidyi peptidase-IV inhibitors for the treatment or prevention of diabetes
ATE532518T1 (en) 2005-09-14 2011-11-15 Takeda Pharmaceutical DIPEPTIDYL PEPTIDASE INHIBITORS FOR THE TREATMENT OF DIABETES
PT1931350E (en) * 2005-09-14 2014-02-12 Takeda Pharmaceutical Administration of dipeptidyl peptidase inhibitors
WO2007033265A1 (en) * 2005-09-14 2007-03-22 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors for treating diabetis
TW200745080A (en) * 2005-09-16 2007-12-16 Takeda Pharmaceuticals Co Polymorphs of tartrate salt of 2-[2-(3-(R)-amino-piperidin-1-yl)-5-fluoro-6-oxo-6H-pyrimidin-1-ylmethyl]-benzonitrile and methods of use therefor
WO2007035629A2 (en) 2005-09-16 2007-03-29 Takeda Pharmaceutical Company Limited Process for the preparation of pyrimidinedione derivatives
TW200745079A (en) * 2005-09-16 2007-12-16 Takeda Pharmaceuticals Co 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 and methods of use therefor
GB0526291D0 (en) 2005-12-23 2006-02-01 Prosidion Ltd Therapeutic method
RU2438671C2 (en) 2005-12-28 2012-01-10 Такеда Фармасьютикал Компани Лимитед Therapeutic antidiabetic agent
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
BRPI0709984A2 (en) 2006-04-12 2011-08-02 Probiodrug Ag enzyme inhibitors
TW200811133A (en) * 2006-07-04 2008-03-01 Astrazeneca Ab New pyridine analogues III 334
BRPI0713401A2 (en) * 2006-07-04 2012-04-17 Astrazeneca Ab compound, pharmaceutical composition, use of a compound, and method of treating a platelet aggregation disorder
AU2007284801A1 (en) 2006-08-08 2008-02-21 The Regents Of The University Of Californina Salicylanilides enhance oral delivery of therapeutic peptides
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
ES2370873T3 (en) * 2006-09-13 2011-12-23 Takeda Pharmaceutical Company Limited USE OF 2-6- (3-AMINO-PIPERIDIN-1-IL) -3-METIL-2,4-DIOXO-3,4-DIHIDRO-2H-PIRIMIDIN-1-ILMETIL-4-FLUORO-BENZONITRILE FOR TREATMENT OF DIABETES, CANCER, AUTOIMMUNITY DISORDERS AND HIV INFECTION.
EP2089383B1 (en) 2006-11-09 2015-09-16 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
JP5523107B2 (en) 2006-11-30 2014-06-18 プロビオドルグ エージー Novel inhibitors of glutaminyl cyclase
US8093236B2 (en) 2007-03-13 2012-01-10 Takeda Pharmaceuticals Company Limited Weekly administration of dipeptidyl peptidase inhibitors
DK2142514T3 (en) 2007-04-18 2015-03-23 Probiodrug Ag Thiourea derivatives as glutaminyl cyclase inhibitors
CA2694620C (en) 2007-07-19 2016-05-31 Takeda Pharmaceutical Company Limited Solid preparation comprising alogliptin and metformin hydrochloride
TW201014850A (en) * 2008-09-25 2010-04-16 Takeda Pharmaceutical Solid pharmaceutical composition
US20100144140A1 (en) * 2008-12-10 2010-06-10 Novellus Systems, Inc. Methods for depositing tungsten films having low resistivity for gapfill applications
ES2548913T3 (en) 2009-09-11 2015-10-21 Probiodrug Ag Heterocyclic derivatives such as glutaminyl cyclase inhibitors
WO2011079778A1 (en) 2009-12-30 2011-07-07 China Shanghai Fochon Pharmaceutical Co Ltd 3-(3-aminopiperidin-1-yl)-5-oxo-1,2,4-triazine derivates as dipeptidyl peptidase iv(dpp-iv) inhibitors
WO2011107530A2 (en) 2010-03-03 2011-09-09 Probiodrug Ag Novel inhibitors
US8269019B2 (en) 2010-03-10 2012-09-18 Probiodrug Ag Inhibitors
WO2011131748A2 (en) 2010-04-21 2011-10-27 Probiodrug Ag Novel inhibitors
JP2014515345A (en) * 2011-01-10 2014-06-30 グラクソスミスクライン・リミテッド・ライアビリティ・カンパニー Pyrimidinone derivatives as fatty acid synthase inhibitors
EP2686313B1 (en) 2011-03-16 2016-02-03 Probiodrug AG Benzimidazole derivatives as inhibitors of glutaminyl cyclase
WO2012149563A1 (en) 2011-04-29 2012-11-01 Kinemed, Inc. Chemical modification of apolipoprotein mimetic peptides for the production of therapeutic agents
CN103044392B (en) * 2011-10-13 2015-10-14 连云港润众制药有限公司 A kind of preparation method of efficient DPP-IV inhibitor
CN103044391B (en) * 2011-10-13 2015-11-25 中国科学院广州生物医药与健康研究院 A kind of DPP-IV inhibitor efficiently
PT2794597T (en) 2011-12-21 2018-01-16 Ono Pharmaceutical Co Pyridinone and pyrimidinone derivatives as factor xia inhibitors
KR102057877B1 (en) 2012-07-18 2019-12-20 노스 앤드 사우스 브라더 파마시 인베스트먼트 컴파니 리미티드 Nitrogenous heterocyclic derivatives and their application in drugs
EP3083584B1 (en) 2013-12-19 2018-02-21 Sunshine Lake Pharma Co., Ltd. Nitrogenous heterocyclic derivatives and their application in the treatment of tissue fibrosis
WO2016019333A1 (en) 2014-07-31 2016-02-04 Kinemed, Inc. The effect of phospholipid composition of reconstituted hdl on its cholesterol efflux and anti-inflammatory properties
EP3702357B1 (en) 2014-12-10 2022-08-24 ONO Pharmaceutical Co., Ltd. Dihydroindolizinone derivative
SG11201707276PA (en) 2015-03-09 2017-10-30 Intekrin Therapeutics Inc Methods for the treatment of nonalcoholic fatty liver disease and/or lipodystrophy
CN107250126B (en) * 2015-11-05 2019-10-25 四川科伦博泰生物医药股份有限公司 Long-acting dipeptidyl peptidase-iv inhibitor, its purposes and preparation method thereof
CA3026149A1 (en) 2016-06-02 2017-12-07 Cadent Therapeutics, Inc. Potassium channel modulators
MX2019008682A (en) 2017-01-23 2019-09-18 Cadent Therapeutics Inc Potassium channel modulators.
JP2020515639A (en) 2017-04-03 2020-05-28 コヒラス・バイオサイエンシズ・インコーポレイテッド PPARγ agonists for the treatment of progressive supranuclear palsy
PL3461819T3 (en) 2017-09-29 2020-11-30 Probiodrug Ag Inhibitors of glutaminyl cyclase
CN109942556A (en) * 2017-12-21 2019-06-28 上海青煜医药科技有限公司 Pyrimidinone compound and its application
CN112567468A (en) 2018-06-14 2021-03-26 阿斯利康(英国)有限公司 Method for lowering blood glucose using dipeptidyl peptidase-4 inhibitor pharmaceutical composition

Family Cites Families (290)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB699812A (en) * 1950-11-29 1953-11-18 British Ind Solvents Ltd Manufacture of substituted pyrimidones
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
BE792206A (en) * 1971-12-02 1973-06-01 Byk Gulden Lomberg Chem Fab
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
EP0260621A3 (en) * 1986-09-18 1989-03-15 F. HOFFMANN-LA ROCHE & CO. Aktiengesellschaft 3-aryl uracil-enol ethers and their use as herbicides
EP0306228B1 (en) 1987-09-04 1999-11-17 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
US5814460A (en) 1990-02-14 1998-09-29 Diatide, Inc. Method for generating and screening useful peptides
US5366862A (en) 1990-02-14 1994-11-22 Receptor Laboratories, Inc. Method for generating and screening useful peptides
CA2036304C (en) 1990-02-15 2001-04-17 Takehiko Naka Pyrimidinedione derivatives, their production 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
US6825169B1 (en) 1991-10-22 2004-11-30 Trustees Of Tufts College Inhibitors of dipeptidyl-aminopeptidase type IV
ES2153831T3 (en) 1991-10-22 2001-03-16 New England Medical Center Inc TYPE IV DIPEPTIDIL-AMINOPEPTIDASE INHIBITORS.
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
DE4305602A1 (en) 1992-06-17 1993-12-23 Merck Patent Gmbh imidazopyridines
CZ181493A3 (en) 1992-09-10 1994-03-16 Lilly Co Eli The use of rhodanine derivative for the preparation of a pharmaceutical composition reducing level of sugar in blood 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
US5798344A (en) 1994-03-08 1998-08-25 Otsuka Pharmaceutical Factory, Inc. Phosphonic ester derivatives of quinazolinones
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
EP0764151A2 (en) 1994-06-10 1997-03-26 Universitaire Instelling Antwerpen Purification of serine protease and synthetic inhibitors thereof
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
WO1996018681A1 (en) * 1994-12-14 1996-06-20 Du Pont-Mitsui Polychemicals Co., Ltd. Thermoplastic resin composition and its use
US5614379A (en) 1995-04-26 1997-03-25 Eli Lilly And Company Process for preparing anti-obesity protein
US6325989B1 (en) 1995-06-01 2001-12-04 Dana-Farber Cancer Institute, Inc. Form of dipeptidylpeptidase IV (CD26) found in human serum
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
PT748800E (en) 1995-06-09 2001-10-30 Hoffmann La Roche DERIVATIVES OF PYRIMIDINADIONA PYRIMIDINATRIONA TRIAZINADIONA AS ANTAGONISTS OF THE ALPHA-1-ADRENERGIC RECEPTOR
JPH0928376A (en) 1995-07-21 1997-02-04 Ajinomoto Co Inc New dipeptidyl peptidase iv and its production
US20020006899A1 (en) * 1998-10-06 2002-01-17 Pospisilik Andrew J. Use of dipeptidyl peptidase IV effectors for lowering blood pressure in mammals
DE19616486C5 (en) 1996-04-25 2016-06-30 Royalty Pharma Collection Trust Method for lowering the blood glucose level 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
BR9711098B1 (en) 1996-07-01 2011-10-04 heterocyclic compounds, processes for their preparation and pharmaceutical compositions containing them and their use in the treatment of diabetes and related diseases.
US7235627B2 (en) 1996-08-30 2007-06-26 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
US6458924B2 (en) 1996-08-30 2002-10-01 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
AU735901C (en) * 1996-12-05 2004-02-12 Amgen, Inc. Substituted pyrimidinone and pyridone compounds and methods of use
GB9702701D0 (en) 1997-02-01 1997-04-02 Univ Newcastle Ventures Ltd Quinazolinone compounds
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
EP0981630B1 (en) 1997-05-16 2008-11-19 Novozymes, Inc. Polypeptides having prolyl pipeptidyl aminopeptidase activity and nucleic acids encoding same
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
TR200000815T2 (en) 1997-09-29 2000-12-21 Point Therapeutics Inc. 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
EP1043328B1 (en) 1997-11-18 2008-03-19 Zaidan Hojin Biseibutsu Kagaku Kenkyu Kai Novel physiologically active substance sulphostin, process for producing the same, and use thereof
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
US6184020B1 (en) 1997-12-16 2001-02-06 Novo Nordisk Biotech, Inc. Polypeptides having aminopeptidase activity and nucleic acids encoding same
US6380357B2 (en) * 1997-12-16 2002-04-30 Eli Lilly And Company Glucagon-like peptide-1 crystals
JP2002501889A (en) 1998-02-02 2002-01-22 トラスティーズ オブ タフツ カレッジ Methods for regulating glucose metabolism and reagents related thereto
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
IL139885A (en) 1998-06-05 2004-05-12 Point Therapeutics Inc Cyclic boroproline compounds
DE19828113A1 (en) 1998-06-24 2000-01-05 Probiodrug Ges Fuer Arzneim Prodrugs of Dipeptidyl Peptidase IV Inhibitors
DE19828114A1 (en) 1998-06-24 2000-01-27 Probiodrug Ges Fuer Arzneim Produgs of unstable inhibitors of dipeptidyl peptidase IV
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
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
BR0010750A (en) * 1999-05-17 2002-02-26 Conjuchem Inc Long-acting 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
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
US6172081B1 (en) 1999-06-24 2001-01-09 Novartis Ag Tetrahydroisoquinoline 3-carboxamide derivatives
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
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
EP1216233A1 (en) 1999-09-28 2002-06-26 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
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
CA2390231A1 (en) 1999-11-12 2001-05-17 Paul Jackson Dipeptidyl peptidase iv inhibitors and methods of making and using dipeptidyl peptidase iv inhibitors
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
GB9928330D0 (en) * 1999-11-30 2000-01-26 Ferring Bv Novel antidiabetic agents
US20030040478A1 (en) * 1999-12-08 2003-02-27 Drucker Daniel J 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
ES2301142T3 (en) 2000-01-21 2008-06-16 Novartis Ag COMBINATIONS THAT INCLUDE INHIBITORS OF DIPEPTIDILPEPTIDASA-IV AND ANTIDIABETIC AGENTS.
TR200401142T4 (en) 2000-01-27 2004-07-21 Eli Lilly And Company Process for dissolving glucagon-like peptide 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
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
US6395767B2 (en) 2000-03-10 2002-05-28 Bristol-Myers Squibb Company Cyclopropyl-fused pyrrolidine-based inhibitors of dipeptidyl peptidase IV and method
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
US7022702B2 (en) 2000-04-07 2006-04-04 Qlt Inc. Antiproliferative 1,2,3-thiadiazole compounds
US6545170B2 (en) 2000-04-13 2003-04-08 Pharmacia Corporation 2-amino-5, 6 heptenoic acid derivatives useful as nitric oxide synthase inhibitors
AU2001255667C1 (en) 2000-04-25 2016-06-09 Icos Corporation Inhibitors of human phosphatidyl-inositol 3-kinase delta
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
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
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
US6620821B2 (en) 2000-06-15 2003-09-16 Bristol-Myers Squibb Company HMG-CoA reductase inhibitors and method
US6627636B2 (en) 2000-06-15 2003-09-30 Bristol-Myers Squibb Company HMG-CoA reductase inhibitors and method
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
GB0014969D0 (en) 2000-06-19 2000-08-09 Smithkline Beecham Plc Novel method of treatment
AU6895801A (en) * 2000-07-04 2002-01-14 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
US6495544B2 (en) 2000-08-01 2002-12-17 Pharmacia Corporation Homoiminopiperidinyl hexanoic acid inhibitors of inducible nitric oxide synthase
JP2004519422A (en) 2000-08-04 2004-07-02 ワーナー−ランバート・カンパニー、リミテッド、ライアビリティ、カンパニー 2- (4-Pyridyl) amino-6-dialkoxyphenylpyrido [2,3-d] pyrimidin-7-one
EP1950199B1 (en) 2000-08-10 2009-12-02 Mitsubishi Tanabe Pharma Corporation Proline derivatives and use thereof as drugs
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
CA2423141A1 (en) 2000-09-27 2002-04-04 Merck & Co., Inc. Benzopyrancarboxylic acid derivatives for the treatment of diabetes and lipid disorders
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
CN1257891C (en) * 2000-10-06 2006-05-31 田边制药株式会社 Nitrogenous five-membered ring compounds
IL155245A0 (en) 2000-10-12 2003-11-23 Ferring Bv Novel serine protease genes related to dppiv
AUPR107800A0 (en) * 2000-10-27 2000-11-23 University Of Sydney, The Peptide and nucleic acid molecule ii
CA2424475A1 (en) 2000-10-27 2002-05-02 Probiodrug Ag Method for the treatment of neurological and neuropsychological disorders
NZ537490A (en) 2000-10-30 2006-11-30 Ortho Mcneil Pharm Inc Treatment of diabetes and syndrome X using a sulfamate derivate such as topiramate and metformin
WO2002060434A2 (en) 2000-10-31 2002-08-08 Merck & Co., Inc. Benzopyrancarboxylic acid derivatives for the treatment of diabetes and lipid disorders
AU2002246568A1 (en) 2000-10-31 2002-08-06 Vanderbilt University Biological markers and diagnostic tests for angiotensin converting enzyme inhibitor- and vasopeptidase inhibitor-associated angioedema
WO2002038742A2 (en) 2000-11-08 2002-05-16 The 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
EP1343763A1 (en) 2000-11-20 2003-09-17 Bristol-Myers Squibb Company Pyrodone derivatives as ap2 inhibitors
CZ20031910A3 (en) 2000-12-11 2003-12-17 Tularik Inc. Compound exhibiting CXCR3 antagonistic activity and pharmaceutical preparation
EP1354882A1 (en) 2000-12-27 2003-10-22 Kyowa Hakko Kogyo Co., Ltd. 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
ATE366724T1 (en) 2001-02-02 2007-08-15 Takeda Pharmaceutical CONDENSED HETEROCYCLIC DERIVATIVES
TWI255817B (en) 2001-02-14 2006-06-01 Kissei Pharmaceutical Glucopyranosyloxybenzylbenzene derivatives and medicinal use thereof
EP2298769B1 (en) * 2001-02-24 2013-10-30 Boehringer Ingelheim Pharma GmbH & Co. KG Xanthin derivatives, their production and utilisation as medicine
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
JP4178816B2 (en) 2001-03-15 2008-11-12 田辺三菱製薬株式会社 Pharmaceutical composition
NZ541703A (en) 2001-03-19 2007-04-27 Novartis Ag Combinations comprising an antidiarrheal agent and an epothilone or an epothilone derivative for treating a proliferative disease
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
US20030095958A1 (en) * 2001-04-27 2003-05-22 Bhisetti Govinda R. Inhibitors of bace
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
EP1406872B1 (en) 2001-06-20 2007-12-19 Merck & Co., Inc. Dipeptidyl peptidase inhibitors for the treatment of diabetes
ES2257555T3 (en) 2001-06-20 2006-08-01 MERCK &amp; CO., INC. DIPEPTIDILPEPTIDASE INHIBITORS FOR THE TREATMENT OF DIABETES.
DE10154689A1 (en) 2001-11-09 2003-05-22 Probiodrug Ag Substituted amino ketone compounds
ATE385238T1 (en) 2001-06-27 2008-02-15 Probiodrug Ag USE OF DPIV INHIBITORS AS THERAPEUTICS FOR MEDICAL NEUROLOGICAL CONDITIONS
US20030130199A1 (en) 2001-06-27 2003-07-10 Von Hoersten Stephan Dipeptidyl peptidase IV inhibitors and their uses as anti-cancer agents
US20030135023A1 (en) 2001-06-27 2003-07-17 Hans-Ulrich Demuth Peptide structures useful for competitive modulation of dipeptidyl peptidase IV catalysis
DE10150203A1 (en) 2001-10-12 2003-04-17 Probiodrug Ag Use of dipeptidyl peptidase IV inhibitor in treatment of cancer
JP4357293B2 (en) 2001-06-27 2009-11-04 スミスクライン ビーチャム コーポレーション Fluoropyrrolidines as dipeptidyl peptidase inhibitors
RU2003105463A (en) 2001-06-27 2004-11-27 Пробиодруг Аг (De) PEPTIDE STRUCTURES SUITABLE FOR COMPETITIVE MODULATION OF CATALYSIS BY DIPEPTIDYLPEPTIDASE IV
WO2003002531A2 (en) 2001-06-27 2003-01-09 Smithkline Beecham Corporation Fluoropyrrolidines as dipeptidyl peptidase inhibitors
DE60223920T2 (en) 2001-06-27 2008-11-13 Smithkline Beecham Corp. PYRROLIDINES AS DIPEPTIDYL-PEPTIDASE INHIBITORS
US7368421B2 (en) 2001-06-27 2008-05-06 Probiodrug Ag Use of dipeptidyl peptidase IV inhibitors in the treatment of multiple sclerosis
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
AU2002322585A1 (en) 2001-07-20 2003-03-03 Adipogenix, Inc. Fat accumulation-modulating 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
JP2003128551A (en) 2001-08-15 2003-05-08 Sankyo Co Ltd New antidiabetic pharmaceutical composition
DE10143840A1 (en) 2001-09-06 2003-03-27 Probiodrug Ag New acylated hydroxamates useful for the treatment of e.g. wound healing
US6844316B2 (en) * 2001-09-06 2005-01-18 Probiodrug Ag Inhibitors of dipeptidyl peptidase I
EP1426366A4 (en) 2001-09-14 2004-12-08 Mitsubishi Pharma Corp Thiazolidine derivative and medicinal use thereof
US6673829B2 (en) 2001-09-14 2004-01-06 Novo Nordisk A/S Aminoazetidine,-pyrrolidine and -piperidine derivatives
US20030186963A1 (en) 2001-09-14 2003-10-02 Dorwald Florencio Zaragoza Substituted piperidines
WO2003024965A2 (en) 2001-09-19 2003-03-27 Novo Nordisk A/S Heterocyclic compounds that are inhibitors of the enzyme dpp-iv
ATE312827T1 (en) 2001-09-21 2005-12-15 Mitsubishi Pharma Corp 3-SUBSTITUTED-4-PYRIMIDONE DERIVATIVES
DE60233335D1 (en) 2001-09-21 2009-09-24 Bristol Myers Squibb Co LACTOMATIC COMPOUNDS AND THEIR DERIVATIVES AS FACTOR XA HEMMER
EA007576B1 (en) * 2001-09-21 2006-12-29 Мицубиси Фарма Корпорейшн 3-substituted-4-pyrimidone derivatives
US7019010B2 (en) 2001-09-27 2006-03-28 Novertis Ag Combinations
AR036663A1 (en) * 2001-10-01 2004-09-22 Bristol Myers Squibb Co ESPIRO-HYDANTOINE COMPOUNDS USED AS ANTI-INFLAMMATORY AGENTS, PHARMACEUTICAL COMPOSITIONS THAT CONTAIN THEM AND THE INTERMEDIARIES TO PREPARE SUCH COMPOUNDS
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
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
WO2003045977A2 (en) 2001-11-26 2003-06-05 Trustees Of Tufts College Peptidomimetic Inhibitors of Post-Proline Cleaving Enzymes
JP2005511636A (en) 2001-11-26 2005-04-28 トラスティーズ オブ タフツ カレッジ Method for treating autoimmune disease and reagent related thereto
US6664273B2 (en) 2001-11-26 2003-12-16 Schering Corporation Piperidine based MCH antagonists for treatment of obesity and CNS disorders
AU2002350217A1 (en) 2001-12-04 2003-06-17 Bristol-Myers Squibb Company Glycinamides as factor xa inhibitors
JP2005518376A (en) 2001-12-14 2005-06-23 ノボ ノルディスク アクティーゼルスカブ Compounds and their use to reduce the activity of hormone sensitive lipases
EP1458382A1 (en) 2001-12-21 2004-09-22 Novo Nordisk A/S 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
WO2003059372A2 (en) 2001-12-29 2003-07-24 Novo Nordisk A/S Combined use of a glp-1 compound and a modulator of diabetic late complications
WO2003057200A2 (en) 2002-01-11 2003-07-17 Novo Nordisk A/S Compositions comprising inhibitors of dpp-iv and nep enzymes for the treatment of diabetes
AU2003218969A1 (en) 2002-02-01 2003-09-02 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
CN1308311C (en) 2002-02-13 2007-04-04 霍夫曼-拉罗奇有限公司 Novel pyridine- and quinoline-derivatives
WO2003072197A1 (en) 2002-02-27 2003-09-04 Pfizer Products Inc. Acc inhibitors
JP2005527504A (en) 2002-02-28 2005-09-15 プロシディオン・リミテッド DPIV inhibitors based on glutaminyl
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
MXPA04009784A (en) 2002-04-08 2004-12-13 Torrent Pharmaceuticlas Ltd Thiazolidine-4-carbonitriles and analogues and their use as dipeptidyl-peptidas 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
WO2003092605A2 (en) 2002-04-30 2003-11-13 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
US7034033B2 (en) 2002-05-23 2006-04-25 Chiron Corporation 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
WO2003101449A2 (en) * 2002-06-04 2003-12-11 Pfizer Products Inc. Process for the preparation of 3,3,4,4-tetrafluoropyrrolidine and derivatives thereof
CN100348599C (en) * 2002-06-06 2007-11-14 卫材R&D管理有限公司 Novel fused imidazole derivative
US20040009972A1 (en) * 2002-06-17 2004-01-15 Ding Charles Z. Benzodiazepine inhibitors of mitochondial F1F0 ATP hydrolase and methods of inhibiting F1F0 ATP hydrolase
US20060252943A1 (en) 2002-06-17 2006-11-09 Amogh Boloor 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
WO2004017989A1 (en) 2002-08-09 2004-03-04 Prosidion Ltd. Methods for improving islet signaling in diabetes mellitus and for its prevention
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
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
JP2004123738A (en) 2002-09-11 2004-04-22 Takeda Chem Ind Ltd Sustained-release preparation
ATE461212T1 (en) 2002-09-18 2010-04-15 Prosidion Ltd SECONDARY BINDING SITE OF DIPEPTIDYLPEPTIDASE 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
KR20050067418A (en) 2002-10-18 2005-07-01 머크 앤드 캄파니 인코포레이티드 Beta-amino heterocyclic dipeptidyl peptidase inhibitors for the 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
US6995180B2 (en) 2002-10-23 2006-02-07 Bristol Myers Squibb Company Glycinenitrile-based inhibitors of dipeptidyl peptidase IV and methods
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
MXPA05005260A (en) 2002-11-18 2005-07-25 Pfizer Prod 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
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
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
US7135467B2 (en) * 2003-01-13 2006-11-14 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
US20050014732A1 (en) 2003-03-14 2005-01-20 Pharmacia Corporation Combination of an aldosterone receptor antagonist and an anti-diabetic agent
WO2004087053A2 (en) 2003-03-25 2004-10-14 Syrrx, Inc. Dipeptidyl peptidase inhibitors
TWI357408B (en) * 2003-03-26 2012-02-01 Mitsubishi Tanabe Pharma Corp 3-substituted-4-pyrimidone derivatives
US7371871B2 (en) 2003-05-05 2008-05-13 Probiodrug Ag Inhibitors of glutaminyl cyclase
WO2004099134A2 (en) 2003-05-05 2004-11-18 Prosidion Ltd. Glutaminyl based dp iv-inhibitors
KR20110059664A (en) 2003-05-05 2011-06-02 프로비오드룩 아게 Use of effectors of glutaminyl and glutamate cyclases
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
SI1638970T1 (en) 2003-06-20 2011-03-31 Hoffmann La Roche Pyrid (2, 1-a) - isoquinoline derivatives as dpp-iv inhibitors
NZ543863A (en) 2003-06-20 2009-05-31 Hoffmann La Roche Hexahydropyridoisoquinolines 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
US7790736B2 (en) 2003-08-13 2010-09-07 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
WO2005019168A2 (en) * 2003-08-20 2005-03-03 Pfizer Products Inc. Fluorinated lysine derivatives as dipeptidyl peptidase iv inhibitors
US7790734B2 (en) 2003-09-08 2010-09-07 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
EP1697342A2 (en) 2003-09-08 2006-09-06 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
AU2005210004B2 (en) 2004-02-05 2010-10-28 Probiodrug Ag Novel inhibitors of glutaminyl cyclase
CN102127057A (en) 2004-03-15 2011-07-20 武田药品工业株式会社 Dipeptidyl peptidase inhibitors
WO2006068978A2 (en) 2004-12-21 2006-06-29 Takeda Pharmaceutial Company Limited Dipeptidyl peptidase inhibitors
ATE532518T1 (en) 2005-09-14 2011-11-15 Takeda Pharmaceutical DIPEPTIDYL PEPTIDASE INHIBITORS FOR THE TREATMENT OF DIABETES
PT1931350E (en) 2005-09-14 2014-02-12 Takeda Pharmaceutical Administration of dipeptidyl peptidase inhibitors
TW200745079A (en) 2005-09-16 2007-12-16 Takeda Pharmaceuticals Co 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 and methods of use therefor

Also Published As

Publication number Publication date
US20050075330A1 (en) 2005-04-07
WO2005016911A1 (en) 2005-02-24
EP1506967B1 (en) 2007-11-21
ZA200602051B (en) 2007-10-31
MXPA06001601A (en) 2006-08-25
DE602004010206T2 (en) 2008-10-09
EP1506967A1 (en) 2005-02-16
US7790736B2 (en) 2010-09-07
US7579357B2 (en) 2009-08-25
JP2005060401A (en) 2005-03-10
RU2006107553A (en) 2007-09-20
US20050070535A1 (en) 2005-03-31
US20050070706A1 (en) 2005-03-31
TW200524895A (en) 2005-08-01
US7470700B2 (en) 2008-12-30
US20050065148A1 (en) 2005-03-24
AU2004265341A1 (en) 2005-02-24
IL173490A0 (en) 2006-06-11
KR20060041309A (en) 2006-05-11
AR045455A1 (en) 2005-10-26
US20050070531A1 (en) 2005-03-31
CN1867560A (en) 2006-11-22
NO20061157L (en) 2006-05-11
DE602004010206D1 (en) 2008-01-03
US7723344B2 (en) 2010-05-25
US20050070530A1 (en) 2005-03-31
JP4854943B2 (en) 2012-01-18
BRPI0413452A (en) 2006-10-17

Similar Documents

Publication Publication Date Title
US7723344B2 (en) Dipeptidyl peptidase inhibitors
CA2559302C (en) 6-amino-1h-pyrimidine-2,4-dione derivatives as dipeptidyl peptidase inhibitors
US7825242B2 (en) Dipeptidyl peptidase inhibitors
US7790734B2 (en) Dipeptidyl peptidase inhibitors
US7687638B2 (en) Dipeptidyl peptidase inhibitors
US20050065144A1 (en) Dipeptidyl peptidase inhibitors
EP1911754B1 (en) Dipeptidyl peptidase inhibitors
AU2011203217B2 (en) Dipeptidyl peptidase inhibitors

Legal Events

Date Code Title Description
EEER Examination request
FZDE Discontinued