CA2608672A1 - Pyridine [3,4-b] pyrazinones as pde-5 inhibitors - Google Patents

Pyridine [3,4-b] pyrazinones as pde-5 inhibitors Download PDF

Info

Publication number
CA2608672A1
CA2608672A1 CA002608672A CA2608672A CA2608672A1 CA 2608672 A1 CA2608672 A1 CA 2608672A1 CA 002608672 A CA002608672 A CA 002608672A CA 2608672 A CA2608672 A CA 2608672A CA 2608672 A1 CA2608672 A1 CA 2608672A1
Authority
CA
Canada
Prior art keywords
group
pyrazin
pyrido
formula
alkyl
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.)
Withdrawn
Application number
CA002608672A
Other languages
French (fr)
Inventor
Alan George Benson
Andrew Simon Bell
David Graham Brown
David Louis Brown
Yvette Marlene Fobian
John Nicholas Freskos
Steven Edward Heasley
Robert Owen Hughes
Eric Jon Jacobsen
Brent Virgil Mischke
John Major Molyneaux
Dafydd Rhys Owen
Michael John Palmer
Christopher Phillips
Donald Joseph Rogier, Jr.
John Keith Walker
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.)
Pharmacia and Upjohn Co LLC
Original Assignee
Pharmacia & Upjohn Company Llc
Alan George Benson
Andrew Simon Bell
David Graham Brown
David Louis Brown
Yvette Marlene Fobian
John Nicholas Freskos
Steven Edward Heasley
Robert Owen Hughes
Eric Jon Jacobsen
Brent Virgil Mischke
John Major Molyneaux
Dafydd Rhys Owen
Michael John Palmer
Christopher Phillips
Donald Joseph Rogier, Jr.
John Keith Walker
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 Pharmacia & Upjohn Company Llc, Alan George Benson, Andrew Simon Bell, David Graham Brown, David Louis Brown, Yvette Marlene Fobian, John Nicholas Freskos, Steven Edward Heasley, Robert Owen Hughes, Eric Jon Jacobsen, Brent Virgil Mischke, John Major Molyneaux, Dafydd Rhys Owen, Michael John Palmer, Christopher Phillips, Donald Joseph Rogier, Jr., John Keith Walker filed Critical Pharmacia & Upjohn Company Llc
Publication of CA2608672A1 publication Critical patent/CA2608672A1/en
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system

Abstract

Compounds, tautomers of the compounds, and pharmaceutically acceptable salts of the compounds or tautomers are disclosed, wherein the compounds have the structure of Formula I: wherein R2, X6, Y6, R6, and R8 are as defined in the specification. Corresponding pharmaceutical compositions, methods of treatment, synthetic methods, and intermediates are also disclosed.

Description

PYRIDINE [3,4-b] PYRAZINONES

Cross Reference to Related Applications This application claims priority from U. S. Provisional Application Serial Number 60/684,139 filed May 24, 2005, the disclosure of which is incorporated herein by reference in its entirety.
FIELD OF THE INVENTION
The present invention comprises a class Pyridine [3,4-b] Pyrazinone compounds having the structure of Formula I and pharmaceutical compositions comprising a compound of Formula I.
The present invention also comprises methods of treating a subject by administering a therapeutically effective amount of a compound of Formula I to the subject. In general, these compounds inhibit, in whole or in part, the enzyme: cyclic guanylate monophosphate-specific phosphodiesterase type 5 (PDE-5).

BACKGROUND OF THE INVENTION
The prevalence of hypertension in developed countries is about 20% of the adult population, rising to about 60-70% of those aged 60 or more. Hypertension is associated with an increased risk of stroke, myocardial infarction, atrial fibrillation, heart failure, peripheral vascular disease and renal impairment. Despite the large number of anti-hypertensive drugs available in various pharmacological categories, additional agents useful for the treatment of hypertension are stili needed.
Vascular endothelial cells secrete nitric oxide (NO). This acts on vascular smooth muscle cells and leads to the activation of guanylate cyclase and the accumulation of cyclic guanosine monophosphate (cGMP). The accumulation of cGMP causes the muscles to relax and the blood vessels to dilate, leading to a reduction in blood pressure. The cGMP is inactivated by hydrolysis to guanosine 5'-monophosphate (GMP) by a cGMP-specific phosphodiesterase. One important cGMP-phosphodiesterase has been identified as Phosphodiesterase type 5 (PDE5).
Inhibitors of PDE5 decrease the rate of hydrolysis of cGMP and so potentiate the actions of nitric oxide.
Improved drug therapies for the treatment of subjects suffering from or susceptible to a cardiovascular condition are desirable. In particular, there still is a need for a new class of PDE-5 inhibitors for treating cGMP-mediated conditions and corresponding drug therapies.

SUMMARY OF THE INVENTION
In one embodiment, the invention comprises compounds having the structure of Formula I:

N
~ I N

I

tI) wherein R2, X6, Y6, R6, and R8 are as defined in the detailed description of the invention.
In another embodiment, the invention comprises a pharmaceutical composition comprising a compound having the structure of Formula I.
In another embodiment, the invention comprises methods of treating a condition in a subject by administering a therapeutically effective amount of a compound having the Formula I to the subject. The conditions that can be treated in accordance with the present invention include cardiovascular diseases, metabolic diseases, central nervous system diseases, pulmonary diseases, sexual dysfunction, and renal dysfunction.
In another embodiment, the invention comprises a method for inhibiting PDE-5, and particularly methods for treating a condition (typically a pathological condition) mediated by PDE-5 activity by administering a compound having a structure of Formula I to the subject.
In another embodiment, the invention comprises intermediates useful in the synthesis of compounds having the structure of Formula I.
DETAILED DESCRIPTION OF THE INVENTION
This detailed description of embodiments is intended only to acquaint others skilled in the art with Applicants' inventions, its principles, and its practical application so that others skilled in the art may adapt and apply the inventions in their numerous forms, as they may be best suited to the requirements of a particular use. These inventions, therefore, are not limited to the embodiments described in this specification, and may be variously modified.

A. Abbreviations and Definitions As used in reference to'H NMR, the symbol "5"refers to a'H NMR chemical shift.
As used in reference to'H NMR, the abbreviation "br" refers to a broad'H NMR
signal.
As used in reference to'H NMR, the abbreviation "d" refers to a doublet'H NMR
peak.
As used in reference to'H NMR, the abbreviation "dd" refers to a doublet of doublets'H
NMR peak.
The abbreviation "HRMS" refers to High Resolution Mass Spectrocopy (electrospray ionisation positive scan).
The abbreviation "m/z" refers to a Mass spectrum peak.
As used in reference to iH NMR, the abbreviation "m" refers to a multiplet 1H
NMR peak.
As used in reference to'H NMR, the abbreviation "q" refers to a quartet'H NMR
peak.
As used in reference to'H NMR, the abbreviation "s" refers to a singlet'H NMR
peak.

As used in reference to'H NMR, the abbreviation 'Y' refers to a triplet iH NMR
peak.
The abbreviation 'TFA" refers to trifluoroacetic acid.
The term "alkyl" (alone or in combination with other term(s)) refers to a linear or branched-chain saturated hydrocarbyl substitutent (i.e., a substitutent containing only carbon and hydrogen) typically containing from about one to about twenty carbon atoms or;
in another embodiment from about one to about twelve carbon atoms; in another embodiment, from about one to about ten carbon atoms; in another embodiment, from about one to about six carbon atoms; and in another embodiment, from about one to about four carbon atoms.
Examples of such substituents include methyl, ethyl, propyl (including n-propyl and isopropyl), butyl (including n-butyl, isobutyl, sec-butyl and tert-butyl), pentyl, iso-amyl, hexyl and the like.
The term "alkenyl" (alone or in combination with other term(s)) refers to a linear or branched-chain hydrocarbyl substituent containing one or more double bonds and from about two to about twenty carbon atoms; in another embodiment, from about two to about twelve carbon atoms;in another embodiment, from about two to about six carbon atoms; and in another embodiment, from about two to about four carbon atoms. Examples of alkenyl radicals include ethenyl, allyi, propenyl, butenyl and 3-methylbutenyl.
The terms "alkenyl'", and "lower alkenyl", embrace radicals having "cis" and "trans"
- - ---or-ientations,-or alternatively,-"Z"and"EO' orientations-The term "alkynyl" (alone or in combination with other term(s)) refers to linear or branched-chain heterocarbyl substituents containing one or more triple bonds and from about two to about twenty carbon atoms; in another embodiment, from about two to about twelve carbon atoms; in another embodiment, from about two to about six carbon atoms; and in another embodiment, from about two to about four carbon atoms. Examples of alkynyl radicals include 1-propynyl, 2-propynyl, 1-butyne, 2-butynyl and 1-pentynyl.
The term "amino", alone or in combination with another term(s), refers to -NH2 when it is at a terminal position or to -NH-when it is used in combination with another term(s) and is not at a terminal position.
The term "aryl", alone or in combination with another term(s), refers to a carbocyclic aromatic system containing one, two or three rings wherein such rings may be attached together in a pendent manner or may be fused. Examples of aryl moieties include phenyl, naphthyl, tetrahydronaphthyl, indanyl and biphenyl.
The term "carboxy", alone or in combination with another term(s),. refers to a radical of the formula -C(O)OH.
The term "cyano", alone or in combination with another term(s), means -CN, which also may - -C N
be depicted:
The term "cycloalkyl", alone or in combination with another term(s), refers to saturated carbocyclic radicals having three to about twelve carbon atoms. In another embodiment, cycloalkyl radicals are "lower cycloalkyl" radicals having three to about eight carbon atoms.
Examples of such radicals include cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl.
The term "cycloalkylalkyl", alone or in combination with another term(s), refers to alkyl substituted with cycloalkyl. Examples of such substituents include cyclopropylmethyl, cyclobutylmethyl, cyclopentylmethyl, and cyclohexylmethyl.
The term "cycloalkenyl", alone or in combination with another term(s), refers to a partially unsaturated carbocyclyl substituent. Examples of such substituents include cyclobutenyl, cyclopentenyl, and cyclohexenyl.
The term "halogen" or "halo", alone or in combination with another term(s), refers to means a fluorine radical (which may be depicted as -F), chlorine radical (which may be depicted as -CI), bromine radical (which may be depicted as -Br), or iodine radical (which may be depicted as -I).
In another embodiment, the halogen is a fluorine or chlorine radical. In another embodiment, the halogen is a fluorine radical.
When used in combination with another term(s), the prefix "halo" indicates that the substituent to which the prefix is attached is substituted with one or more independently selected halogen radicals. For example, haloalkyl refers to an alkyl substituent wherein at least one hydrogen radical is replaced with a halogen radical. Where there are more than one hydrogens - ------r-eplaced-w-ith-halogens;-the-halogens-may be-the-same-or-different.-Exampies of-haloalkyls include chloromethyl, dichloromethyl, difluorochloromethyl, dichlorofluoromethyl, trichloromethyl, 1-bromoethyl, fluoromethyl, difluoromethyl, trifluoromethyl, 2,2,2-trifluoroethyl, difluoroethyl, pentafluoroethyl, difluoropropyl, dichloropropyl, and heptafluoropropyl.
Illustrating further, "haloalkoxy' means an alkoxy substituent wherein at least one hydrogen radical is replaced by a halogen radical. Examples of haloalkoxy substituents include chloromethoxy, 1 -bromoethoxy, fluoromethoxy, difluoromethoxy, trifluoromethoxy (also known as "perfluoromethyloxy"), and 2,2,2,-trifluoroethoxy. If a substituent is substituted by more than one halogen radical, those halogen radicals may be identical or different (unless otherwise stated).
The term "heterocyclyl" refers to a saturated, partially saturated, or completely unsaturated ring structure containing a total of 3 to 14 ring atoms. At least one of the ring atoms is a heteroatom (i.e., oxygen, nitrogen, or sulfur), with the remaining ring atoms being independently selected from the group consisting of carbon, oxygen, nitrogen, and sulfur.
A heterocyclyl may be a single ring, which typically contains from 3 to 10 ring atoms, more typically from 3 to 7 ring atoms, and even more typically 5 to 6 ring atoms.
Examples of single-ring heterocyclyls include furanyl, dihydrofurnayl, tetradydrofurnayl, thiophenyl (also known as "thiofuranyl"), dihydrothiophenyl, tetrahydrothiophenyl, pyrrolyl, isopyrrolyl, pyrrolinyl, pyrrolidinyl, imidazolyl, isoimidazolyl, imidazolinyl, imidazolidinyl, pyrazolyl, pyrazolinyl, pyrazolidinyl, triazolyl, tetrazolyl, dithiolyl, oxathiolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, thiazolinyl, isothiazolinyl, thiazolidinyl, isothiazolidinyl, thiodiazolyl, oxathiazolyl, oxadiazolyl (including oxadiazolyl, 1,2,4-oxadiazolyl (also known as "azoximyl"), 1,2,5-oxadiazolyl (also known as "furazanyl"), or 1,3,4-oxadiazolyl), oxatriazolyl (including 1,2,3,4-oxatriazolyl or 1,2,3,5-oxatriazolyl), dioxazolyl (including 1,2,3-dioxazoiyl, 1,2,4-dioxazolyl, 1,3,2-dioxazolyl, or 1,3,4-dioxazolyl), oxathiazolyl, oxathiolyl, oxathiolanyl, pyranyl (including 1,2-pyranyl or 1,4-pyranyl), dihydropyranyl, pyridinyl (also known as "azinyl"), piperidinyl, diazinyl (including pyridazinyl (also known as "1,2-diazinyP'), pyrimidinyl (also known as "1,3-diazinyP" or "pyrimidyl"), or pyrazinyl (also known as "1,4-diazinyP')), piperazinyl, triazinyl (including s-triazinyl (also known as "1,3,5-triazinyP'), as-triazinyl (also known 1,2,4-triazinyl), and v-triazinyl (also known as "1,2,3-triazinyP')), oxazinyl (including 1,2,3-oxazinyl, 1,3,2-oxazinyl, 1,3,6-oxazinyl (also known as "pentoxazolyP'), 1,2,6-oxazinyl, or 1,4-oxazinyl), isoxazinyl (including o-isoxazinyl or p-isoxazinyl), oxazolidinyl, isoxazolidinyl, oxathiazinyl (including 1,2,5-oxathiazinyl or 1,2,6-oxathiazinyl), oxadiazinyl (including 1,4,2-oxadiazinyl or 1,3,5,2-oxadiazinyl), morpholinyl, azepinyl, oxepinyl, thiepinyl, and diazepinyl. A heterocyclyl alternatively may comprise 2 or 3 rings fused together, wherein at least one such ring contains a heteroatom as a ring atom (e.g., nitrogen, oxygen, or sulfur). Examples of 2-fused-ring heterocyclyls include, indolizinyl, pyrindinyl, pyranopyrrolyl, 4H-quinolizinyl, purinyl, naphthyridinyl, pyridopyridinyl (including pyrido[3,4-b]-pyridinyl, pyrido[3,2-b]-pyridinyl, or pyrido[4,3-b]-pyridinyl), and pteridinyl, indolyl, isoindolyl, indoleninyl, isoindazolyl, benzazinyl, phthalazinyl, quinoxalinyl, quinazolinyl, benzodiazinyl, benzopyranyl, benzothiopyranyl, benzoxazolyl, indoxazinyl, anthranilyl, benzodioxolyl, benzodioxanyl, benzoxadiazolyl, benzofuranyl, isobenzofuranyl, benzothienyl, isobenzothienyl, benzothiazolyl, ------benzothiadiazolyl,-benzimidazolyl,--benzotriazolyl, benzoxazinyl, benzisoxazinyl, and tetrahydroisoquinolinyl. Other examples of fused-ring heterocyclyls include benzo-fused hetes-ccyclyls, such as indolyl, isoindolyl (also known as "isobenzazolyl" or "pseudoisoindolyl"), indoleninyl (also known as "pseudoindolyl"), isoindazolyl (also known as "benzpyrazolyl"), benzazinyl (including quinolinyl (also known as "1-benzazinyl") or isoquinolinyl (also known as "2-benzazinyl")), phthalazinyl, quinoxalinyl, quinazolinyl, benzodiazinyl (including cinnolinyl (also known as "1,2-benzodiazinyl") or quinazolinyl (also known as "1,3-benzodiazinyP')), benzopyranyl (including "chromanyl" or "isochromanyl"), benzothiopyranyl (also known as "thiochromanyl"), benzoxazolyl, indoxazinyl (also known as "benzisoxazolyl"), anthranilyl, benzodioxolyl, benzodioxanyl, benzoxadiazolyl, benzofuranyl (also known as "coumaronyP'), isobenzofuranyl, benzothienyl (also known as "benzothiophenyl," "thionaphthenyl," or "benzothiofuranyl"), isobenzothienyl (also known as "isobenzothiophenyl," "isothionaphthenyl," or "isobenzothiofuranyl"), benzothiazolyl, benzothiadiazolyl, benzimidazolyl, benzotriazolyl, benzoxazinyl (including 1,3,2-benzoxazinyl , 1,4,2-benzoxazinyl , 2,3,1-benzoxazinyl , or 3,1,4-benzoxazinyl ), benzisoxazinyl (including 1,2-benzisoxazinyl or 1,4-benzisoxazinyl), tetrahydroisoquinolinyl , carbazolyl, xanthenyl, and acridinyl.
The term "heteroaryl", alone or in combination with another term(s), refers to a completely unsaturated (i.e., aromatic) heterocyclyl containing from 5 to 14 ring atoms.
A heteroaryl may comprise a single ring or 2 or 3 fused rings. In one embodiment, heteroaryl radicals are 5- or 6-membered heteroaryl, containing one or two heteroatoms selected from sulphur, nitrogen and oxygen, selected from thienyl, furanyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, pyridyl and pyrazinyl. Examples of heteroaryl substituents include 6-membered ring substituents such as pyridyl, pyrazyl, pyrimidinyl, and pyridazinyl; 5-membered ring substituents such as 1,3,5-, 1,2,4- or 1,2,3-triazinyl, imidazyl, furanyl, thiophenyl, pyrazolyl, oxazolyl, isoxazolyl, and thiazolyl; 1,2,3-, 1,2,4-, 1,2,5-, or 1,3,4-oxadiazolyl and isothiazolyl; 6/5-membered fused ring substituents such as benzothiofuranyl, isobenzothiofuranyl, benzisoxazolyl, benzoxazolyl, purinyl, and anthranilyl; and 6/6-membered fused rings such as 1,2-, 1,4-, 2,3-and 2, 1-benzopyronyl, quinolinyl, isoquinolinyl, cinnolinyl, quinazolinyl, and 1,4-benzoxazinyl. Other heteroaryls include unsaturated 5 to 6 membered heteromonocyclyl groups containing 1 to 4 nitrogen atoms, for example, pyrrolyl, imidazolyl, pyrazolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, pyrimidyl, pyrazinyl, pyridazinyl, triazolyl [e.g., 4H-1,2,4-triazolyl, 1H-1,2,3-triazolyl, 2H-1,2,3-triazolyl]; unsaturated condensed heterocyclic groups containing 1 to 5 nitrogen atoms, for example, indolyl, isoindolyl, indolizinyl, benzimidazolyl, quinolyl, isoquinolyl, indazolyl, benzotriazolyl, tetrazolopyridazinyl [e.g., tetrazolo [1,5-b]pyridazinyl];
unsaturated 3 to 6-membered heteromonocyclic groups containing an oxygen atom, for example, pyranyl, 2-furyl, 3-furyl, etc.; unsaturated 5 to 6-membered heteromonocyclic groups containing a sulfur atom, for example, 2-thienyl, 3-thienyl, etc.; unsaturated 5- to 6-membered heteromonocyclic groups containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms, for example, isoxazolyi, oxadiazolyl [e.g., 1,2,4-oxadiazolyl, 1,3,4-oxadiazolyl, 1,2,5-oxadiazolyl]; unsaturated condensed heterocyclic groups containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms [e.g.
benzoxazolyl, ---------benzoxadiazoiyl]; unsaturated-5-to-6=membered-heteromonocyclic-groups containing- 1-to 2 sulfur atoms and 1 to 3 nitrogen atoms, for example, thiazolyl, thiadiazolyl [e.g., 1,2,4- thiadiazolyl, 1,3,4-thiadiazolyl, 1,2,5-thiadiazolyl]; unsaturated condensed heterocyclic groups containing 1 to 2 sulfur atoms and 1 to 3 nitrogen atoms [e.g., benzothiazolyl, benzothiadiazolyl] and the like.
The term also embraces radicals where heterocyclic radicals are fused with aryl radicals.
Examples of such fused bicyclic radicals include benzofuran, benzothiophene, and the like.
The term "heterocyclylalkyl", alone or in combination with another term(s), refers to alkyl substituted with a heterocyclyl.
The term "hydroxy", alone or in combination with another term(s), refers to -OH.
The term "mercapto" or "thiol" refers to a sulfhydryl substituent, which also may depicted as -SH.
The term "nitro", alone or in combination with another term(s), refers to -NOZ.
The term "sulfonyl", alone or in combination with another term(s), refers to -S(O)2-, which also may be depicted as:
OO
'2Z s ~.

Thus, for example, "alkyl-sulfonyl-alkyl" refers to alkyl-S(O)2-alkyl.
Examples of typically preferred alkylsulfonyl substituents include methylsulfonyl, ethylsulfonyl, and propylsulfonyl.
The term "sulfoxyl" , alone or in combination with another term(s), refers to -S(O) -, which also may be depicted as:
S
The term "thio" or "thia", alone or in combination with another term(s), refers to a thiaether substituent, i.e., an ether substituent wherein a divalent sulfur atom is in the place of the ether oxygen atom. Such a substituent may be depicted as -S-. This, for example, "alkyl-thio-alkyl"
means alkyl-S-alkyl.
If a substituent is described as being "optionally substituted", the substituent may be either (1) not substituted, or (2) substituted. If a carbon of a substituent is described as being optionally substituted with one or more of a list of substituents, one or more of the hydrogens on the carbon (to the extent there are any) may separately and%or together be replaced with an independently selected optional substituent. This specification uses the terms "substituent"
and "radical"
interchangeably.
The term "cGMP-mediated condition" refers to any condition mediated by cGMP, whether through direct regulation by cGMP, or through indirect regulation by cGMP as a component of a signaling pathway.
The term "composition" refers to an article of manufacture which results from the mixing or combining of more than one element or ingredient.
The term "hypertensive subject" refers to a subject having hypertension, suffering from the effects of hypertension or susceptible to a hypertensive condition if not treated to prevent or control such hypertension.
The term "pharmaceutically acceptable carrier" refers to a carrier that is compatible with the other ingredients of the composition and is not deleterious to the subject.
Such carriers may be pharmaceutically acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting a chemical agent. The preferred composition depends on the method of administration.
The terms "prevent," "prevention" or "preventing" refer to either preventing the onset of a preclinically evident condition altogether or preventing the onset of a preclinical evident stage of a condition in a subject. Prevention includes, but is not limited to, prophylactic treatment of a subject at risk of developing a condition.
The term "therapeutically effective amount" refers to that amount of drug or pharmaceutical agent that will elicit the biological or medical response of a tissue, system or animal that is being sought by a researcher or clinician.
The term "treatment" (and corresponding terms "treat" and "treating") includes palliative, restorative, and preventative treatment of a subject. The term "palliative treatment" refers to treatment that eases or reduces the effect or intensity of a condition in a subject without curing the condition. The term "preventative treatment" (and the corresponding term "prophylactic treatment") refers to treatment that prevents the occurrence of a condition in a subject. The term "restorative treatment" refers to treatment that halts the progression of, reduces the pathologic manifestations of, or entirely eliminates a condition in a subject.

B. Compounds The present invention comprises, in part, a novel class of pyridine [3,4-b]
pyrazinone compounds. These compounds are useful as inhibitors of PDE5.

Compounds of Formula (I) As used herein, compounds of the present invention include tautomers of the compounds and pharmaceutically acceptable salts of the compounds and tautomers.
The present invention is directed, in part, to a class of compounds having the structure of Formula I:

O N ~ R2 s_ ( ~ N
R YQN N
I

-(I) --------- ----- ---wherein R2 is selected from the group consistirig of aryl and 3 to 10 membered ring heterocycyl wherein R2 may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, cyano, nitro, oxo, alkyl, alkenyl, alkynyl, cycloalkyl, -OR100, -C(O)Ri o -OC(O)R100, -C(O)OR'00, -NRi R101 -N(R1 )C(O)R1 1-C(O)NRi R'01, -C(O NR10 C O R101, -SR10 -S O R10 and -S O R10 ) ( ) ( ) ( )2 , aziridinyl, azetidinyl, pyrrolidinyl, piperidinyl, morpholinyl, thiomorpholinyl, and piperazinyl wherein (a) the alkyl, alkenyl, alkylnyl and cycloalkyl substituents may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, oxo, -OR102, and -C(O)OR102; and (b) the aziridinyl, azetidinyl, pyrrolidinyl, piperidinyl, morpholinyl, thiomorpholinyl, and piperazinyl substituents may be optionally substituted with one or more substituents selected from the group consisting of alkyl, hydroxy and alkoxy;

R10 , R101 and R102 are independently selected from the group consisting of hydrogen, alkyl, alkenyl and alkynyl, wherein the alkyl may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, alkoxy, -C(O)OH and -C(O)NH2;

X6 is selected from the group consisting of hydrogen and alkyl wherein the X6 alkyl substituent may be optionally substituted with one or more substituents selected from the group consisting of chloro, fluoro, hydroxy and alkoxy;
Y6 represents a bond or is selected from the group consisting of alkyl, alkenyl and alkynyl, wherein Y6 may be optionally substituted with one ormore substituents independently selected from the group consisting of halogen, cyano, oxo, cycloalkyl, -OR103, -C(O)R103, -C(O)OR103, -OC O R'03' -NR1 03R104' -N R103 C O R104 1 3 1 4 ( ) ( ) ( ) , and -C(O)NR R ;

R1 3 and R'04 are independently selected from the group consisting of hydrogen and alkyl, wherein the alkyl may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, carboxy, cyano, oxo, alkynyl, haloalkynyl, hydroxyalkynyl, carboxyalkynyl, alkoxy, haloalkoxy, hydroxyalkoxy, and carboxyalkoxy;

R6 is selected from the group consisting of aryl, aryl-C(O)-, heterocyclyl, aryl-C(O)-NR105-heterocyclyl-C(O)-, and heterocyclyl-C(O)-NR105- wherein R 6 may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, cyano, oxo, alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heterocyclyl, -OR106, -C(O)R106, -C(O)OR'oe, _ OC(O)R106, -NR106Rim -N(R1 06)C(O)R107, -C(O)NR10sRio7, -C(O)NR'06 C(O)R1 07 --S(O)R106, -S(O)2R106, -N(R106)S(O)2R107, and -S(O)2NR106R107; wherein the alkyl, alkenyl, alkynyl, cycloalkyl, aryl and heterocyclyl susbstituents may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, carboxy, cyano, oxo, alkoxy, haloalkoxy, hydroxyalkoxy, and carboxyalkoxy;

R105 is independently selected from the group consisting of hydrogen and alkyl;

R106 and R107 are independently selected from the group consisting of hydrogen, alkyl, alkenyl, and alkynyl, wherein (a) the R106 and R107 alkyl and alkenyl substituents may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, carboxy, cyano, oxo, alkynyl, haloalkynyl, hydroxyalkynyl, carboxyalkynyl, alkoxy, haloalkoxy, hydroxyalkoxy, and carboxyalkoxy, and (b) the R106 and R107 alkynyl substituents may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, carboxy, cyano, oxo, alkoxy, haloalkoxy, hydroxyalkoxy, and carboxyalkoxy;

RB is alkyl; wherein RB may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, carboxy, cyano, alkynyl, -OR108, -C(O)R108, -C(O)OR108, -OC(O)Ri08, -NR108 R109, -N(R108)C(O)R109, -C(O)NR10eR1e9 , -SR108, -S(O)R108, -S(O)2R108, and -C(O)NR108C(O)R109, wherein the alkynyl substituents may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, carboxy, cyano, oxo, alkyl, and alkoxy; and R108 and R109 are independently selected from the group consisting of hydrogen, alkyl, alkenyl and alkynyl, wherein (a) when the alkyl is methyl, the methyl may be optionally substituted with 1, 2, or 3 fluoro substituents, (b) when the alkyl comprises at least two carbon atoms, the alkyl may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, carboxy, cyano, oxo, alkynyl, haloalkynyl, hydroxyalkynyl, carboxyalkynyi, alkoxy, haloalkoxy, hydroxyalkoxy, and carboxyalkoxy, and (c) the R10Band R109 alkenyl and alkynyl substituents may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, carboxy, cyano, oxo, alkoxy, haloalkoxy, hydroxyalkoxy, and carboxyalkoxy.
Selected subclasses of compounds of interest that fall within the scope of the compounds of Formula I are shown in Table A, wherein R 2, X6, Ys, Rs, and R8 can be as defined for compounds of Formula I and as defined in the various embodiments described thoughout this specification.
Illustrative embodiments of these subclasses of compounds are described later in the specification.
TABLE A
R10 R1o R8 R9 R11 R8 R9 Rii O N \\ N O N N
~
Rs_YsN ,N i N Ris R6_Y6 N ,N N R13 Xs (I-A) H (I-B) Rs-YO-N ~N N R13 s_ s 1 , N
Xs (I C) R Y XN s N (I D) O
O N \\ N O N N
\
Rs-YsN ~N ~ N ~ N
R YG-N N
Xs (I-E) ~6 (I-F) Rio Rio Re R9 N RS R9 / Rii O N
I R12 0 N \ R12 s_ 6~ N R13 ~ N R13 R Y N N Rs-Y~N N
X s (I-G) Xs (I-H) Rs R9 / R11 Rg R11 O N . \ R12 0 N

6_ 6 ~ I,N R13 6_ ~~ N
R Y H N R Y N N
I
(I I) X6 (I-J) R8 / R11 R$ R9 N' R11 O N \ \ O N Nr 6_ fi ~ N 6_ 6 ~ N R13 R Y N N R Y N N
X6 (I-K) X6 (I-L) 11 Rs cAh1 O N / ~ \ II
O N
\ \ N ~
s_ ~~ i N R13 R6_Y6 N ~N N R13 R Y H N (I-M) X6 (I-N) _ .. ------ ------- -- --..._ ---R9--- -- -- -- _.- -- - ------- - R9 8 , O N N p N8 \ p 6_ fi- /\ I N R13 ~ I ~ R13 j~s (1-0) j~s (I-P) R1o R1o Rs R9 O R8 R9 O

O N p /\N \ \ ~
s _ /' / N R13 I N R13 j(s (I-Q) j~s (I-R) Ra R6 /N
R6-Y N \N N R6-YG-N \N
H ~
(I-S) (I-T) O\N I R2 O N R2 Rs-Y N N R6_N \N

(I-U) x6 (I-V) ~ R8 O N R
I 0 N ~ fl2 R6N \N

Xs (I-W) x6 (I-X) \O~ O--) R6-Y~N \N N Rs-YsN \N
x6 (I-Y) X6 (I-Z) ~
O//>

O N R2 O R2 R6-Y6N ~N N R6-Y6 N ~N

j~6 (I-AA) X6 (I-BB) ~~o~ ~'/~O

Rs-YcH \N Rs-YsN \N N
(I-CC) H (I-DD) O N R

R6-Y6N H (I-FF) H (I-EE) N

R6-N \N /
H (I-GG) H N
(I-HH) Rii O N Rz O N N

s"\ I N
Rs/\N \N N R N N
(I H (I JJ) H
-II) Q~ R11 Ri i Q--) N \ \ N O N N
IN
R
s\~~H ~N Rs_ ~N ~ N

(I-KK) H (I-LL) Rii Rs-YaN \N I Rs-Ys-N N N

H (I-MM) H (INN) 2 ~
O N =~ R O N ~ Rz I \IY

Rs-N N R\~\ \ I~ N
H (I-00) H N
(I-PP) v \Q ~/ 'Q R11 O Rz O N \

H N
s~\ I N
s/\ N R N N
R N H (I RR) (I-QQ) O R11 ~~\Q R11 --) N I I N O N

R \~H CN N R6_N N / N
(I SS) H
(I-TT) O N \ N
~
R6-YGN \N ~ N
H (I-UU) Embodiments of Substituent In one embodiment of Formula I, R 2 is selected from the group consisting of aryl and 3 to 10 membered ring heterocycyl wherein R2 may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, cyano, nitro, oxo, alkyl, alkenyl, alkynyl, cycloalkyl, -OR100, -C(O)Ri00, -OC(O)Rioo, -C(O)OR10 , -NR10 R101, -N(R10 )C(O)R101, -C(O)NR100R101, -C(O)NR10 C(O)R101, and -S(O)mR10 wherein the alkyl, alkenyl, alkynyl and cycloalkyl substituents may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, oxo, -OR102, and -C(0)OR102; wherein R10 , R101 and R102 are independently selected from the group consisting of hydrogen, alkyl, alkenyl and alkynyl, wherein the alkyl may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, alkoxy, -C(O)OH and -C(O)NH2; and m is 0, 1, or 2.
In one embodiment of Formula I, R2 is selected from the group consisting of phenyl and a 3 to 10 membered ring heteroaryl, optionally substituted as described in Formula I. In another embodiment of Formula I, R2 is selected 'from the group consisting of phenyl and a 5 to 7 membered ring heterocyclyl, optionally substituted as described in Formula I.
In another embodiment of Formula I, R2 is selected from the group consisting of phenyl and a 5 to 7 membered ring heteroaryl, optionally substituted as described in Formula I. In another embodiment of Formula I, R2 is selected from the group consisting of phenyl and a 5-6 membered ring heteroaryl, optionally substituted as described in Formula I. In another embodiment of Formula I, R2 is a 5 to 6 membered ring heteroaryl that comprises 1, 2, or 3 ring heteroatoms selected from the group consisting of oxygen and nitrogen.
In one embodiment of Formula I, R2 is selected from the group consisting of phenyl, thienyl, furanyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, pyrazinyl pyridinyl, triazinyl, imidazyl, thiophenyl, pyrazolyl, oxazolyl, oxadiazolyl, pyridyl, pyrazyl, pyrimidinyl, pyridazinyl, benzofuran, and benzodioxolyl. In another embodiment of Formula I, R2 is selected from the group consisting of phenyl, pyridinyl, pyrimidinyl, isoxazolyl, pyrazolyl, benzofuran, and benzodioxolyl, optionally substituted as described in Formula I. In another embodiment of Formula I, R2 is selected from the group consisting of phenyl, N\NH \O ) \ N N \
N N

'L N I I I N
_Z and each optionally substituted as provided in Formula I. In another embodiment of Formula I, R2 is selected from the group consisting of phenyl and pyridinyl, optionally substituted as described in Formula I.
In one embodiment of Formula I, R2 is optionally substituted with one or more substituents selected from the group consisting of halogen, oxo, alkyl, -OR10 , -C(O)R10 , -OC(O)R'oo, -C(O)OR10 , -NR10 R101 and -C(O)NR10 R101, wherein the alkyl substitutent may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, oxo, -OR102, and -C(O)OR102; wherein R'oo R'o' and R102 are independently selected from the group consisting of hydrogen and C, to C4 alkyl.
In another embodiment of Formula I, R 2 is optionally substituted with one or more substituents independently selected from the group consisting of halogen, cyano, oxo, C, to C4 alkyl, -OR100, -C(O)OR10 , and -C(O)NR10 R'01, wherein (a) when the alkyl is methyl, the methyl may be optionally substituted with 1, 2, or 3 halogen substituents, (b) when the alkyl comprises at least two carbon atoms, the alkyl may be optionally substituted with one or more substituents selected from the group consisting of halogen, C, to C4 alkoxy and hydroxy;
and wherein R'oo and R101 are independently selected from the group consisting of hydrogen and Ci to C2 alkyl.
In another embodiment of Formula I, R2 is optionally substituted with one or more substituents selected from the group consisting of halogen, C, to C4 alkyl, -OR100, and -NR100R101; wherein (a) when the alkyl is methyl, the methyl may be optionally substituted with 1, 2, or 3 halogen substituents, (b) when the alkyl comprises at least two carbon atoms, the alkyl may be optionally substituted with one or more substituents selected from the group consisting of halogen, oxo, C1 to C2 alkoxy and hydroxy; and wherein R10 and R101 are independently selected from the group consisting of hydrogen and Ci to C2 alkyl.
In one embodiment of Formula I, R2 is optionally substituted with one or more substituents selected from the group consisting of chloro, fluoro, methyl, ethyl, propyl, butyl, pentyl, hexyl, trifluoromethyl, hydroxy, methoxy, ethoxy, propoxy, butoxy, amino, methylamino, dimethylamino, ethylamino, and diethylamino. In another embodiment of Formula I, R2 is optionally substituted with one or more substituents selected from the group consisting of fluoro, methyl, trifluoromethyl, methoxy, trifluoromethoxy, amino, methylamino, and dimethylamino.
In one embodiment of Formula I, R2 is substituted with one or more fluoro substituents. In another embodiment of Formula I, R2 is substituted=with one fluoro substituent. In another embodiment of Formula I, R2 is substituted with two fluoro substituents.
In one embodiment of Formula I, R2 is substituted with methoxy.
In one embodiment of Formula I, R2 is substituted at the para position with a substituent selected from the group consisting of fluoro, methyl, trifluoromethyl, methoxy, trifluoromethoxy, amino, methylamino, and dimethylamino. In another embodiment of Formula I, R2 is substituted at the para position with a substituent selected from the group consisting of fluoro, methyl, trifluoromethyl, methoxy, and trifluoromethoxy.
In one embodiment of Formula I, R2 is selected from the group in Table A
consisting of Formula I-A, Formula I-C, Formula I-G, Formula I-H, Formula I-I, Formula I-J, Formula I-K, Formula I-L, Formula I-M, Formula I-N, Formula 1-0, Formula I-P, Formula I-Q
and Formula I-R, wherein R9, R10, R", R'2 and R'3 are independently selected from the group consisting of hydrogen, halogen, oxo, alkyl, -OR10 , -C(O)R100, -OC(O)R10 , -C(O)OR10 , -NR10 R101 and -C(O)NR100R101, wherein the alkyl substitutent may be optionally substituted with one or more substituents independentiy selected from the group consisting of halogen, oxo, -OR102, and -C(O)OR102; wherein R10 , R101, and R102 are independently selected from the group consisting of hydrogen and Ci toC4 alkyl. In another embodiment of Formula I, R2 is selected from the group in Table A consisting of Formula I-H, Formula I-A and Formula I-L, wherein R9, R'o, R", R'Z and R13 are independently selected from the group consisting of hydrogen, halogen, oxo, alkyl, -OR100, -C(O)R100, -OC(0)R100, -C(O)OR10 , -NR10 R101 and -C(O)NR'ooR10' wherein the alkyl substitutent may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, oxo, -OR102, and -C(O)OR102; wherein R'oo R101, and R102 are independently selected from the group consisting of hydrogen and Ci to C4 alkyl. In another embodiment of Formula I, R 2 is selected from the group in Table A consisting of Formula I-A, Formula I-D, Formula I-H and Formula I-L, wherein R9, R'o, R", R'2 and R13 are independently selected from the group consisting of hydrogen, halogen, alkyl, haloalkyl, oxy, alkoxy, hydroxy, and carboxy. In another embodiment of Formula I, R 2 is selected from the group in Table A
consistingof_Formula, -A,_ Formula I-C, Formula I-D and Formula I-H, wherein R9, R10, R", R'z and R13 are independently selected from the group consisting of hydrogen, fluoro, methyl, trifluoromethyl, and methoxy. In another embodiment of Formula I, 'r~2 is selected from the group in Table A consisting of Formula I-D and Formula I-H, wherein R9, R'O. R", R12 and R13 are independently selected from the group consisting of hydrogen, fluoro, methyl, trifluoromethyl, and methoxy.
In one embodiment of Formula I, R2 is as described by the structures in Table A consisting of Formula I-D and Formula I-H wherein R9, R'o, R", R12and R13 are independently selected from the group consisting of hydrogen, fluoro, methyl, trifluoromethyl, and methoxy. In another embodiment of Formula I, R2 is as described in Formula I-D in Table A, wherein R" is selected from the group consisting of hydrogen, fluoro, methyl, trifluoromethyl, and methoxy. In another embodiment of Formula I, the R2 substituent is as described in Formula I-E in Table A.
Embodiments of X6 Substituent In one embodiment of Formula I, X6 is selected from the group consisting of hydrogen and Ci to C4 alkyl wherein the X6 Cy to C4 alkyl substituent may be optionally substituted with one or more substituents selected from the group consisting of chloro, fluoro, hydroxy and alkoxy.
In one embodiment of Formula I, X6 is alkyl, optionally substituted with 1 to 3 substituents independently selected from the group consisting of fluoro and chloro. In another embodiment of Formula I, X6 alkyl is optionally substituted with 1 to 3 fluoro substituents.
In one embodiment of Formula I, X6 is selected from the group consisting of hydrogen and Ci to C6 alkyl. In another embodiment of Formula I, X6 is selected from the group consisting of hydrogen, m,ethyl, and ethyl. In another embodiment, X6 is hydrogen.

Embodiments of Ys Substituent In one embodiment of Formula I, Y6 represents a bond or is selected from the group consisting of alkyl, alkenyl and alkynyl, wherein the Y6 alkyl, alkenyl and alkynyl substituents may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, cyano, oxo, cycloalkyl, -OR103, -C(O)R103, -C(O)OR103, _OC(O)R'03_ NR103R104, -N R103 C O R104, and -C O NR'03R10a 103 and R10a ( )() () ; and wherein R are independently selected from the group consisting of hydrogen and alkyl, wherein the alkyl may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, carboxy, cyano, oxo, alkynyl, haloalkynyl, hydroxyalkynyl, carboxyalkynyl, alkoxy, haloalkoxy, hydroxyalkoxy, and carboxyalkoxy.
In one embodiment of Formula I, Ys represents a bond or is alkyl, optionally substituted as described in Formula I. In another embodiment of Formula I, Y6 represents a bond or is Ci to C6 alkyl, optionally substituted as described in Formula I. In another embodiment of Formula I, Y6 represents a bond or is selected from the group consisting of Ci to C4 alkyl and hydroxyCi to C6.
alkyi. In another embodiment of Formula I, Y6 represents a bond or is selected from the group consisting of methyl, ethyl, propyl, butyl, hydroxyethyl, hydroxypropyl, hydroxybutyl, _dihydroxyeth~rl,.and dihy_droxybutyl,_ In another embodiment of Formula I, Y6 represents a bond or is selected from the group consisting of methyl, ethyl, propyl, butyl, hydroxyethyl, hydroxypropyl, hydroxybutyl, dihydroxyethyl, HO HO HO T HO,,__HO~ HO--,,,_ HO

~ (R6) (R6) (Rs) (Rs) (R6) (R6) \,(R6), and HO
In one embodiment of Formula I, Ys represents a bond or is selected from the group consisting of methyl, ethyl, propyl and butyl. In another embodiment of Formula I, Y6 represents a bond.
In one embodiment of Formula I, Y6 is alkyl optionally substituted with one or more substituents independently selected from the group consisting of halogen, cyano, oxo, cycloalkyl, -OR103, -C(O)R103, _C(O)OR103, _OC(O)R103, _NR103R104, -N(R103)C(O)R104 , and -C(O)NR103 104 R ;
wherein R103 and R104 are independently selected from the group consisting of hydrogen and Ci to C4 alkyl. In another embodiment of Formula I, Y6 is alkyl substituted with one or more substituents independently selected from the group consisting of fluoro, chloro, oxo, cycloalkyl, hydroxy, and carboxy. In another embodiment of Formula I, Y6 is alkyl substituted with one to three substituents independently selected from the group consisting of hydroxy and cyclohexyl.
In another embodiment of Formula I, Y6 is unsubstituted alkyl. In another embodiment of Formula I, Ys is selected from the group consisting of methyl, ethyl and propyl. In another embodiment, Y6 is selected from the group consisting of -CH2-, -CH2CH2-, -CH2CH2CH2-, -CH2(CH3)-, -CH2CH2(CH3)-, -CH2(CH3)CH2-, -CH2C(CH3) 2CH-, -C(CH3) 2CH-, -CH2C(CH3)-, and -CH2CH2CH2CH2-. In another embodiment of Formula I, Y6 is methyl.

Embodiments of 146 Substituent In one embodiment of Formula I, R 6 is selected from the group consisting of phenyl, phenyl-C(O)-, 3 to 10 membered ring heterocyclyl, phenyl-C(O)-NR105-, 3 to 10 membered ring heterocyclyl-C(O)-, and 3 to 10 membered ring heterocyclyl-C(O)-NR105-, wherein R6 is optionally substituted as described in Formula I, wherein R105 is selected from the group consisting of hydrogen and Ci to C6 alkyl. In another embodiment of Formula I, R6 is selected from the group consisting of phenyl, phenyl-C(O)-, 5 to 7 membered ring heterocyclyl, phenyl-C(O)-NR105- 5 to 7 membered ring heterocyclyl-C(O)-, and 5 to 7 membered ring heterocyclyl-C(O)-NR1o5-wherein R6 is optionally substituted as described in Formula I, wherein R105 is selected from the group consisting of hydrogen and Ci to C4 alkyl. In another embodiment of Formula I, R6 is selected from the group consisting of phenyl, phenyl-C(O)-, 5 to 6 membered ring heterocyclyl, phenyl-C(O)-NR105-, 5 to 6 membered ring heterocyclyl-C(O)-, and 5 to 6 membered ring heterocyclyl-C(O)-NR105-, wherein R6 is optionally substituted as described in Formula I wherein R105 is hydrogen. In another embodiment, R6 is selected from the group consisting of phenyl, pyridinyl, piperidinyl, piperizinyl, morpholino, pyrazinyl, tetrahydropyran, tetrahydrofuran, isoxazole, imidazole, pyrrolidine, wherein R6 is optionally substituted as described in Formula I.
In another embodiment, R 6 is selected from the group consisting of morpholinyl, tetrahydropyranyl, tetrahydrofuranyl, piperidinyl, and pyrrolidinyl, wherein R6 is optionally substituted as provided iR? Formula I.
In one embodiment of Formula I, R6 is selected from the group consisting of phenyl, phenyl C(O)NH-, and phenyl C(O)-.
In one embodiment of Formula I, R6 is selected from the group consisting of 5 to 7 membered ring heteroaryl, 5 to 7 membered ring heteroaryl-C(O)-, and 5 to 7 membered ring heteroaryl-C(O)-NR105-, wherein R6 is optionally substituted as described in Formula I, wherein R105 is selected from the group consisting of hydrogen and Ci to C4 alkyl. In another embodiment of Formula I, R6 is a 3 to 10 membered ring heteroaryl, optionally substituted as described in Formula I. In another embodiment of Formula I, R6 is a 5 to 7 membered ring heteroaryl, optionally substituted as described in Formula I. In another embodiment of Formula I, R6 is a 5 to 6 membered ring heteroaryl, optionally substituted as described in Formula I. In another embodiment of Formula I, R6 is a 5 to 6 membered ring heteroaryl containing one to three heteroatoms selected from the group consisting of 0 and N, optionally substituted as described in Formula I. In another embodiment of Formula I, R6 is selected from the group consisting of imidazole, isoxazole, pyridinyl and pyrazynyl, optionally substituted as described in Formula I. In another embodiment of Formula I, R6 is selected from the group consisting of imidazole, isoxazole, pyridinyl and pyrazynyl, optionally substituted with one or more substituents independently selected from the group consisting of alkyl, -OR105, and -C(O)R105, wherein R105 is selected from the group consisting of hydrogen, methyl, and ethyl. In another embodimeht, R6 is ~ffi.
N N ~ n(Zr"?.
N~ O-N NJ
selected from the group consisting of , , , and In one embodiment of Formula I, R6 is selected from the group consisting of 3 to 10 membered ring fully or partially saturated heterocylyl, 3 to 10 membered ring fully or partially saturated heterocyclyl-C(O)-, 3 to 10 membered ring fully or partially saturated heterocyclyl-C(O)NH-, optionally substituted as described in Formula I. In another embodiment of Formula I, R6 is a 5 to 7 membered ring fully or partially saturated heterocylyl, 5 to 7 membered ring fully or partially saturated heterocyclyl-C(O)-, 5 to 7 membered ring fully or partially saturated heterocyclyl-C(O)NH-, wherein R6 is optionally substituted as described in Formula I. In another embodiment of Formula I, R6 is a 5 to 6 membered ring fully saturated heterocylyl, 5 to 6 membered ring fully saturated heterocyclyl-C(O)-, 5 to 6 membered ring fully saturated heterocyclyl-C(O)NH-, wherein R6 is optionally substituted as described in Formula I. In another embodiment of Formula I, R6 is a 5 to 6 membered ring fully saturated heterocylyl, wherein R6 is optionally substituted as described in Formula I. In another embodiment of Formula I, Rs is selected from the group consisting of tetrahydrofuran, tetrahydropyran, pyrrolidinyl, piperidinyl, -- --- piper-azinyl;-and-morpholino; wher-ein-R6 is-optionallysubstituted as-described-in Formula I. In another embodiment of Formula I, R6 is selected from the group consisting of tetrahydrofuran, tetrahydropyran, pyrrolidinyl, piperidinyl, piperazinyl, and morpholino, wherein R6 is optionally substituted with one or more substituents independently selected from the group consisting of alkyl, -OR105, -C(O)R'05 and -C(O)OR105 wherein R1 5 is selected from the group consisting of hydrogen, methyl, and ethyl. In another embodiment, R6 is selected from the group consisting O
~-Z-n1'~ N
of > > O > > , O

~. N
~~
HN
N
> > , , O N
olf r J~
HNJ NJ

> > a O

N ~ rN
\/NJ NJ
O o O

_JN~' JN
~ rN
/ \O N O~/ O~/ O

N JL, H
OJ
and In another embodiment of Formula I, R6 is optionally substituted with one or more substituents independently selected from the group consisting of -OR'06, -C(O)R106, -C(O)OR106, -NR106Ri07 -N(Ry06)C(O)Ri07, -N(R1 06)C(O)ORi07, -C(O)NR106R107, -NHC(O)NR106 Ri07, -N(R106)S(O)2R107, wherein R106 and R107 are independently selected from the group consisting of hydrogen, methyl, ethyl, methylethyl, tert-butyl, cyclopentyl, cyclohexyl, optionally substituted with one or more substituent selected from the group consisting of halogen, oxo, hydroxy, and methyl. In another embodiment of Formula I, R6 is optionally substituted with one or more substituents independently selected from the group consisting of -OH, -OCH3, -OCH2CH3, -OCH2(CH3)CH3, -C(O)CH2CH2OH, -C(O)OH, -C(O)OCH2(CH3)(CH3)(CH3), -NH2, -NH(CH2CH3), -N(CH3)(CH3), -N(CH2CH3)CH2CH3, -N(H)CH2(CH3)CH3, -N(H)CH2CH2CH2OH, -N(H)CH2C(O)CH3, -NH(CH2CH2OH), -N(H)C(O)OCH3, -N(H)C(O)CH2(CH3)(CH3)(CH3), -NHC(O)OCH2(CH3)(CH3)(CH3), -NHC(O)COCH3, -NHC(O)NHCOCH3, -NCH3C(O)CH3, -NCH3(O)C(O)CH3i N(CH(O)CH3)C(O)CH3, -C(O)NH2, -C(O)NH(CH3), -C(O)NCH3(CH3), -C(O)NHCH2(CH3)(CH3)(CH3), -C(O)NHR106, -N(H)S(O)2CH3i -N(H)S(O)2CHF3, wherein R106 is independently selected from the group consisting of cyclopentyl and cyclohexyl, and wherein the R106 cyclohexyl substituent is optionally substituted with hydroxy; and wherein (b) the cyclohexyl R6 substituent is optionally substituted with hydroxy. In another embodiment of Formula I, R6 is optionally substituted with one or more substituents independ-ently selected from the group consisting of alkyl, --C(O)R'osand -C(O)OR106, wherein R106 is selected from the group consisting of hydrogen, methyl, and ethyl. In another embodiment of Formula I, R6 is optionally substituted with one or more substituents independently selected from the group consisting of -OH, -CH3, -CH2CH3i -C(O)CH3, -C(O)CH2CH3, -C(O)OCH2CH3.
Embodiments of A8 Substituent In one embodiment of Formula I, RB is alkyl; wherein the R8 substituent may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, carboxy, cyano, alkynyl, -OR10S, -SR108, -C(O)R108, -C(O)OR108, -OC(O)R108, -NR'08R10s, -N(R108)C(O)R109, -C(O)NR108R109, and -C(O)NR108C(O)R109, wherein the alkynyl substituents may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, carboxy, cyano, oxo, alkyl, and alkoxy; and wherein R10S and R109 are independently selected from the group consisting of hydrogen, alkyl, and alkynyl, wherein (a) when the alkyl is methyl, the methyl may be optionally substituted with 1, 2, or 3 fluoro substituents, (b) when the alkyl comprises at least two carbon atoms, the alkyl may be optionally substituted with one or more substituents independently selected from the group _- -consisting_of-halogen,h_y_dx_oxy, carh.oxy,_.cyano,_oxo,-alkyny_i,h.aloalkynayl,hydr.oxyalkynyl, carboxyalkynyl, alkoxy, haloalkoxy, hydroxyalkoxy, and carboxyalkoxy, and (c) the R108and R109 alkynyl substituents may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, carboxy, cyano, oxo, alkoxy, haloalkoxy, hydroxyalkoxy, and carboxyalkoxy.
In one embodiment of Formula I, R8 is Ci to C10 alkyl, optionally substituted as described in Formula I. In another embodiment of Formula I, R8 is C. to C$ alkyl, optionally substituted as described in Formula I. In another embodiment of Formula I, R8 is Ci to C6 alkyl, optionally substituted as described in Formula I. In another embodiment of Formula I, R8 is Ci to C4 alkyl, optionally substituted as described in Formula I. In another embodiment of Formula I, R8 is ethyl, optionally substituted as described in Formula I.
In one embodiment of Formula I, R8 is optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, carboxy, cyano, -OR'08-C(O)R108, -C(O)OR108, -OC(O)R108, -NR108R109-N(R108)C(O)R709 -C(O)NR108R109, and -C(O)NR108C(O)R109, wherein R108 and R109 are independently selected from the group consisting of hydrogen and Ci to C6 alkyl, wherein (a) when the C, to C6 alkyl is methyl, the methyl may be optionally substituted with 1, 2, or 3 fluoro substituents, (b) when the Ci to C6 alkyl comprises at least two carbon atoms, the alkyl may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, carboxy, cyano, oxo, alkynyl, haloalkynyl, hydroxyalkynyl, carboxyalkynyl, alkoxy, haloalkoxy, hydroxyalkoxy, and carboxyalkoxy. In another embodiment of Formula I, R8 is optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, carboxy, cyano, alkoxy, haloalkyl, hydroxyalkyl, carboxyalkyl, haloalkoxy, hydroxyalkoxy, and carboxyalkoxy. In another embodiment of Formula I, Ra is optionally substituted with one or more substituents independently selected from the group consisting of halogen, haloalkoxy, hydroxy, carboxy, cyano, oxo, and alkoxy. In another embodiment of Formula I, R8 is optionally substituted with one or more substituents independently selected from the group consisting of halogen, haloalkoxy, hydroxy, oxo, and alkoxy. In another embodiment of Formula I, R8 is optionally substituted with one or more substituents independently selected from the group consisting of haloalkoxy and alkoxy. In another embodiment of Formula I, R 8 is ethyl optionally substituted with one or more substituents independently selected from the group consisting of halogen, haloalkoxy, hydroxy, carboxy, cyano, oxo, and alkoxy. In another embodiment of Formula I, R8 is ethyl optionally substituted with one or more substituents independently selected from the group consisting of haloalkoxy and alkoxy.
In another embodiment of Formula I, R8 is alkoxyalkyl, optionally substituted as described in Formula I. In another embodiment of Formula I, R8 is a(Cl to C4)alkoxy(C, to C4)alkyl, optionally substituted as described in Formula I. In another embodiment of Formula I, R 8 is methoxyethyl, as described in Formula 1-Y in Table A. In another embodiment of Formula I, R8 is ethoxyethyl, as described in Formula I-Z in Table A. In another embodiment of Formula I, R$
is propoxyethyl, as described in Formulae I-AA and I-BB in Table A. In another embodiment of Formula I, R8 is trifluoroethoxyethyl. In another embodiment of Formula I, R8 is selected from the group consisting of propoxyethyl and ethoxyethyl as described in Formulae I-Z, I-AA
and I-BB in Table A, respectively.

Embodiments of Multiple Substituents The following are additional embodiments of the compounds of Formula I. Unless otherwise specified, substituents are as described in Formula I. Further embodiments of Formula I described when R2, X6, Y6, R6 and R8 are selected from the various embodiments described above.

Embodiments where X6 is Hydrogen, R2 is phenyl or 3-10 membered heterocyclyl In one embodiment of Formula I, X6 is hydrogen and R2 is selected from the group consisting of phenyl, thienyl, furanyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, pyrazinyl pyridinyl, triazinyl, imidazyl, thiophenyl, pyrazolyl, oxazolyl, oxadiazolyl, pyridyl, pyrazyl, pyrimidinyl, pyridazinyl, benzofuran, and benzodioxolyl. In another embodiment of Formula I, X6 is hydrogen and RZ is selected from the group consisting of phenyl, pyridinyl, pyrimidinyl, isoxazolyl, pyrazolyl, benzofuran, and benzodioxolyl. In another embodiment of Formula I, X6 is hydrogen and R2 is selected from the group consisting of phenyl, pyridinyl and pyrimidinyl, as described in Formulae I-I, I-B and I-M of Table A. In another embodiment of Formula I, X6 is hydrogen and R2 is selected from the group consisting of phenyl and pyridinyl, as described in Formulae I-I and I-B of Table A. In another embodiment of Formula I, X6 is hydrogen and R2 is phenyl, as described in Formulae I-B of Table A, wherein R9, R1o R11, R12and R13are independently selected from the group consisting of hydrogen, hydroxy, and fluoro. In another embodiment of Formula I, X6 is hydrogen and R 2 is pyridinyl, as described in Formulae I-B of Table A, wherein R9, R10 and R13 are each hydrogen, and R" is methoxy.

Embodiments where X6 is Hydrogen, R8 is alkoxyalkyl In one embodiment of Formula I, X6 is hydrogen and R8 is alkoxyalkyl, optionally substituted as described in Formula I. In another embodiment of Formula I, X6 is hydrogen and Ra is a(C1 to C4)alkoxy(C1 to C4)alkyl, optionally substituted as described in Formula I.
In another embodiment of Formula I, X6 is hydrogen and R8 is selected from the group consisting of methoxyethyl, ethoxyethyl, and propoxyethyl, optionally substituted as described in Formula I.
In another embodiment, X6 is hydrogen and R8 is selected from the group consisting of methoxyethyl, ethoxyethyl, propoxyethyl, and trifluoroethylethoxy. In another embodiment of Formula I, X6 is hydrogen and RB is selected from the group consisting of propoxyethyl and ethoxyethyl, as described in Formulae I-Z, I-AA and I-BB of Table A, respectively.

Embodiments where X6 is Hydrogen, R8 is alkoxyalkyl, and Ys represents a bond or is alkyl In one embodiment of Formula I, X6 is hydrogen, R8 is alkoxyalkyl, optionally substituted as --- - described-in-Formula-I-and Y6 r-epresents-a-bond-or-is-alkyl,-optionally-substituted as described in Formula I. In another embodiment of Formula I, X6 is hydrogen and R8 is a(Ci to C4)alkoxy(Ci to C4)alkyl, optionally substituted as described in Formula I, and Y6 represents a bond or is C, to C6 alkyl, optionally substituted as described in Formula I. In another embodiment of Formula I, X6 is hydrogen, R8 is selected from the group consisting of methoxyethyl, ethoxyethyl, and propoxyethyl, wherein R8 is optionally substituted as described in Formula I, and Y6 represents a bond or is selected from the group consisting of methyl, ethyl, propyl, butyl, methylethyl, tert-butyl, hydroxyethyl, hydroxypropyl, hydroxybutyl, dihydroxyethyl, HO HO HO HO~HO~ HO~_ HO
s (R6 ) (Rs) (R6) (R6) (R6) (R6), and R ( ) HO

In another embodiment of Formula I, X6 is hydrogen, Ra is selected from the group consisting of methoxyethyl, ethoxyethyl, propoxyethyl, and trifluoroethoxyethyl, and Ys represents a bond or is selected from the group consisting of inethyl, ethyl, propyl and butyl. In another embodiment of Formula I, X6 is hydrogen, R8 is selected from the group consisting of propoxyethyl and ethoxyethyl, and Y6 represents a bond or is selected from the group consisting of methyl and ethyl. In another embodiment of Formula I, X6 is hydrogen, R8 is ethoxyethyl, and Y6 represents a bond or is selected from the group consisting of methyl and ethyl, as described in Formulae I-GG, I-HH and I-II in Table A, respectively. In another embodiment of Formula I, X6 is hydrogen, RB is propoxyethyl, and Y6 represents a bond or is selected from the group consisting of methyl and ethyl, as described in Formulae 1-00, I-QQ and I-PP
in Table A, respectively.

Embodiments where Ra is alkoxyalkyl, and R2 is phenyl or 3-10 membered heterocyclyl In one embodiment of Formula I, R8 is alkoxyalkyl, optionally substituted as described in Formula I and R2 is selected from the group consisting of phenyl, thienyl, furanyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, pyrazinyl pyridinyl, triazinyl, imidazyl, thiophenyl, pyrazolyl, oxazolyl, oxadiazolyl, pyridyl, pyrazyl, pyrimidinyl, pyridazinyl, benzofuran, and benzodioxolyl, wherein R2 is optionally substituted as described in Formula I.
In another embodiment of Formula I, R8 is a(C, to C4)alkoxy(Cy to C4)alkyl, optionally substituted as described in Formula I, and R2 is selected from the group consisting of phenyl, pyridinyl, pyrimidinyl, isoxazolyl, pyrazolyl, benzofuran, and benzodioxolyl, wherein R2 is optionally substituted as described in Formula I. In another embodiment of Formula I, R8 is selected from the group consisting of trifluroethoxyethyl, ethoxyethyl, and propoxyethyl, and R2 is selected from the group consisting of phenyl, pyridinyl and pyrimidinyl, wherein R2 is optionally substituted as described in Formula I. In another embodiment of Formula I, R8 is selected from the group consisting of ethoxyethyl and propoxyethyl, and R2 is selected from the group consisting of phenyl and pyridinyl, wherein R2 is optionally substituted as described in Formula I. In another embodiment of Formula I, R8 is ethoxyethyl, and R2 is pyridinyl, optionally substituted as described in Formula I. In another embodiment of Formula I, R8 is ethoxyethyl and R2 is a --- methoxy-pyr-idinyl,-as-descr-ibed-in-For-mula-I-F-of Table A. In-another-embodiment of Formula I, R8 is propoxyethyl and R2 is a methoxy pyridinyl.
Embodiments where X6 is hydrogen, R8 is alkoxyalkyl, and R2 is phenyl or 3-10 membered heterocyclyl In one embodiment of Formula I, X6 is hydrogen, R8 is alkoxyalkyl, optionally substituted as described in Formula I and R2 is selected from the group consisting of phenyl, thienyl, furanyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, pyrazinyl pyridinyl, triazinyl, imidazyl, thiophenyl, pyrazolyl, oxazolyl, oxadiazolyl, pyridyl, pyrazyl, pyrimidinyl, pyridazinyl, benzofuran, and benzodioxolyl, wherein R2 is optionally substituted as described in Formula I. In another embodiment of Formula I, X6 is hydrogen, R$ is a(Cl to C4)alkoxy(Ci to C4)alkyl, optionally substituted as described in Formula I, and R2 is selected from the group consisting of phenyl, pyridinyl, pyrimidinyl, isoxazolyl, pyrazolyl, benzofuran, and benzodioxolyl, wherein R2 is optionally substituted as described in Formula I. In another embodiment of Formula I, X6 is hydrogen, R8 is selected from the group consisting of trifluoroethoxyethyl, ethoxyethyl, and propoxyethyl, wherein R8 is optionally substituted as described in Formula I
and R2 is selected from the group consisting of phenyl, pyridinyl and pyrimidinyl, wherein R2 is optionally substituted as described in Formula I. In another embodiment of Formula I, X6 is hydrogen, R 8 is selected from the group consisting of ethoxyethyl and propoxyethyl, and R2 is selected from the group consisting of phenyl and pyridinyl wherein R2 is optionally substituted as described in Formula I.
In another embodiment of Formula I, X6 is hydrogen, R8 is ethoxyethyl, and R2 is pyridinyl, optionally substituted as described in Formula I. In another embodiment of Formula I, X6 is hydrogen, R$ is ethoxyethyl and R2 is a para-substitued pyridinyl, as described in Formula I-FF of Table A. In another embodiment of Formula I, X6 is hydrogen, R8 is propoxyethyl and R2 is a para-substitued pyridinyl, as described in Formula I-NN of Table A. In another embodiment of Formula I, X6 is hydrogen, R8 is propoxyethyl and R2 is a pyridinyl, substituted at the para position with a methoxy as described in Formula I-UU of Table A.
Embodiments where X6 is hydrogen, R8 is alkoxyalkyl, Y6 represents a bond or is alkyl and R2 is phenyl or 3-10 membered heterocyclyl In one embodiment of Formula I, X6 is hydrogen, Y6 is a bond or is alkyl, optionally substituted as described in Formula I, R8 is alkoxyalkyl, optionally substituted as described in Formula I, and R2 is selected from the group consisting of phenyl, thienyl, furanyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, pyrazinyl pyridinyl, triazinyl, imidazyl, thiophenyl, pyrazolyi, oxazolyl, oxadiazolyl, pyridyl, pyrazyl, pyrimidinyl, pyridazinyl, benzofuran, and benzodioxolyl, wherein R2 is optionally substituted as described in Formula I.
In another embodiment of Formula I, X6 is hydrogen, Y6 represents a bond or is Ci to C6 alkyl, optionally substituted as described in Formula I, R8 is a(C1 to C4)alkoxy(Ci to C4)alkyl, optionally substituted as described in Formula I, and R2 is selected from the group consisting of phenyl, pyridinyl, pyrimidinyl, isoxazolyl, pyrazolyl, benzofuran, and benzodioxolyl wherein R2 is optionally substituted as described in Formula I. In another embodiment of Formula I, X6 is - -_- hydrogen,_R$_is.selected from_the_group_consisting of_triflur.oethoxyethyl,_ethoxyethyl,_and propoxyethyl, optionally substituted as described in Formula I and R 2 is selected from the group consisting of phenyl, pyridinyl and pyrimidinyl wherein R2 is optionally substituted as described in Formula I. In another embodiment of Formula I, X6 is hydrogen, Y6 represents a bond or is selected from the group consisting of methyl, ethyl, propyl, butyl, hydroxyethyl, hydroxypropyl, hydroxybutyl, dihydroxyethyl, and dihydroxybutyl, R8 is selected from the group consisting of ethoxyethyl and propoxyethyl, and R2 is selected from the group consisting of phenyl and pyridinyl wherein R 2 is optionally substituted as described in Formula I. In another embodiment of Formula I, X6 is hydrogen, Y6 represents a bond or is selected from the group consisting of methyl, ethyl, methylethyl, propyl and butyl, R8 is selected from the group consisting of ethoxyethyl and propoxyethyl, and R2 is pyridinyl, optionally substituted as described in Formula 1. In another embodiment of Formula I, X6 is hydrogen, Ys represents a bond, R
8 is ethoxyethyl and R2 is a para-substitued pyridinyl, as described in Formula I-LL of Table A. In another embodiment of Formula I, X6 is hydrogen, Y6 is methyl, R8 is ethoxyethyl and R2 is a para-substitued pyridinyl, as described in Formula I-JJ of Table A. In another embodiment of Formula I, X6 is hydrogen, Y6 is ethyl, R 8 is ethoxyethyl and R2 is a para-substitued pyridinyl, as described in Formula I-KK of Table A. In another embodiment of Formula I, X6 is hydrogen, Y6 represents a bond, R8 is propoxyethyl and R2 is a para-substitued pyridinyl, as described in Formula I-TT of Table A. In another embodiment of Formula I, X6 is hydrogen, Y6 is methyl, Ra is propoxyethyl and R2 is a para-substitued pyridinyl, as described in Formula I-RR of Table A. In another embodiment of Formula I, X6 is hydrogen, Y6 is ethyl, Re is propoxyethyl and R2 is a para-substitued pyridinyl, as described in Formula I-SS of Table A.

Embodiments where X6 is hydrogen, R8 is alkoxyalkyl, Y6 represents a bond or is alkyl and R2 is phenyl or 3-10 membered heterocyclyl, and R6 is selected from various embodiments In one embodiment of Formula I, X6 is hydrogen, Y6 represents a bond, R8 is selected from the group consisting of propoxyethyl and ethoxyethyl and R2 is a para-substitued pyridinyl, R6 is selected from the group consisting of selected from the group consisting of phenyl, phenyl C(O)NH-, phenyl C(O)-, 3 to 10 membered ring heterocyclyl, phenyl-C(O)-NH-, 3 to 10 membered ring heterocyclyl-C(O)-, and 3 to 10 membered ring heterocyclyl-C(O)-NH, optionally substituted with one or more substituents independently selected from the group consisting of -OR105, -C(O)R105, -C(O)OR105, -NR. 105R106, -N(R105)C(O)R106, -N(R105)C(O)OR106, -C(O)NR105 R106 , -NHC(O)NR'05R106, -N(R105)S(O)2R106, wherein R105 and R106 are independently selected from the group consisting of hydrogen, methyl, ethyl, methylethyl, tert-butyl, cyclopentyl, cyclohexyl, optionally substituted with one or more substituent selected from the group consisting of halogen, oxo, hydroxy, and methyl. In another embodiment of Formula I, X6 is hydrogen, Y6 is methyl, R8 is selected from the group consisting of propoxyethyl and ethoxyethyl, and R2 is a para-substitued pyridinyl, optionally substituted as described in Formula I, and R6 is selected from the group consisting of phenyl, phenyl C(O)NH-, phenyl C(O)-, 5 to 7 membered ring heterocyclyl, phenyl-C(O)-NH-, 5 to 7 membered ring heterocyclyl-C(O)-, and 5 to 7 membered ring ----heterocyel-yl-C(O)-NH,-optionally-s-ubstituted-with-one-or-more-substituents-independently selected from the group consisting of of -OR106, -C(O)R106, -C(O)OR'06 -NR106R107, -N(R106)C(O)R107 -N(R106)C(O)OR107, -C(O)NR106R107 -NHC(O)NR106R107, -N(R106)S(O)2R107, wherein R106 and R107 are independently selected from the group consisting of hydrogen, methyl, ethyl, methylethyl, tert-butyl, cyclopentyl, cyclohexyl, optionally substituted with one or more substituent selected from the group consisting of halogen, oxo, hydroxy, and methyl. In another embodiment of Formula I, X6 is hydrogen, Y6 is ethyl, R8 is selected from the group consisting of propoxyethyl and ethoxyethyl, R2 is a para-substitued pyridinyl, optionally substituted as described in Formula I, and R6 is selected from the group consisting of 5 to 6 membered ring heteroaryl, 5 to 6 membered ring heteroaryl-C(O)-, and 5 to 6 membered ring heteroaryl-C(O)-NH, optionally substituted with one or more substituents independently selected from the group consisting of alkyl, -OR106, and -C(O)R106, wherein R106 is selected from the group consisting of hydrogen, methyl, and ethyl. In another embodiment of Formula I, X6 is hydrogen, Y6 is ethyl, R8 is selected from the group consisting of propoxyethyl and ethoxyethyl, R2 is a para-substitued pyridinyl optionally substituted as described in Formula I, and R6 is selected from the group consisting of 5 to 6 membered ring fully saturated heterocyclyl, 5 to 6 membered ring fully saturated heterocyclyl-C(O)-, and 5 to 6 membered ring fully saturated heterocyclyl-C(O)-NH, optionally substituted with one or more substituents independently selected from the group consisting of alkyl, -OR106, and -C(O)R106, wherein R106 is selected from the group consisting of hydrogen, methyl, and ethyl.

Compounds of Formula (I-B) The present invention is directed, in part, to a class of compounds having the structure of Formula I-B:
Rio R$ R9 Rii O N
\ \ N
( R6-Yrl'N ~N ~ N R13 H (I-B) wherein R9, R10, R" and R13 are independently selected from the group consisting of hydrogen, halogen, oxo, alkyl, -OR100, -C(O)R100, -OC(O)R100, -C(O)OR'o -NR100R101 and -C(O)NR100R101, wherein the alkyl substitutent may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, oxo, -OR102, and -C(O)OR102;
wherein R10 , R101, and R102 are independently selected from the group consisting of hydrogen and Ci to C4 alkyl;

y6 represents a bond or is selected from the group consisting of alkyl, alkenyl and alkynyl, wherein (a) the Y6 alkyl, alkenyl and alkynyl substituents may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, cyano, oxo, cycloaiky,, -OR103, -C(O)R103, -C(O)OR103, -OC(O)R103, -NR103R104, -N(R103)C(O)R104, and -C(O)NR103R104;

R103 and R104 are independently selected from the group consisting of hydrogen and alkyl, wherein the alkyl may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, carboxy, cyano, oxo, alkynyl, haloalkynyl, hydroxyalkynyl, carboxyalkynyl, alkoxy, haloalkoxy, hydroxyalkoxy, and carboxyalkoxy;

R6 is selected from the group consisting of aryl, aryl-C(O)-, heterocyclyl, aryl-C(O)-NR105-, heterocyclyl-C(O)-, and heterocyclyl-C(O)-NR105- wherein R6 may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, cyano, oxo, alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heterocyclyi, -OR106, -C(O)R106, -C(O)OR106, -OC(O)R 106, -NR106 R107, -N(R106)C(O)R107, -C(O)NR10sR107, -C(O)NR10sC(O)R107, , -S(O)R106, -S(O)2R106, -N(R106)S(O)2R107, and -S(O)2NR106R107; wherein the alkyl, alkenyl, alkynyl, cycloalkyl, aryl and heterocyclyl susbstituents may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, carboxy, cyano, oxo, alkoxy, haloalkoxy, hydroxyalkoxy, and carboxyalkoxy;

R105 is independently selected from the group consisting of hydrogen and alkyl;

R106 and R107 are independently selected from the group consisting of hydrogen, alkyl, alkenyl, and alkynyl, wherein (a) the R106 and R107 alkyl and alkenyl substituents may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, carboxy, cyano, oxo, alkynyl, haloalkynyl, hydroxyalkynyl, carboxyalkynyl, alkoxy, haloalkoxy, hydroxyalkoxy, and carboxyalkoxy, and (b) the R106 and R107 alkynyl substituents may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, carboxy, cyano, oxo, alkoxy, haloalkoxy, hydroxyalkoxy, and carboxyalkoxy;
RB is alkoxyalkyl, optionally substituted with halogen.
Embodiments of R9. RYO, R1 i and R13 In one embodiment of the compounds of Formula I-B, R9, R10, R" and R'3are selected from the group consisting of hydrogen, chloro, fluoro, methyl, ethyl, propyl, butyl, pentyl, hexyl, trif luorom ethyl, hydroxy, methoxy, ethoxy, propoxy, butoxy, amino, methylamino, dimethylamino, ethylamino, and diethylamino. In another embodiment of Formula I-B, R9, R10, R" and R'3are - ---selected-fr-om-the gr-oup-consisting-of-hydr-ogen,-fluor-o,-rnethyl,-trifluor-omethyl; methoxy, trifluoromethoxy, amino, methylamino, and dimethylamino. In another embodiment of Formula I-B, R" and R13 are hydrogen, and R9 and R1 are selected from the group consisting ot hydrogen, chloro, fluoro, methyl, ethyl, propyl, butyl, pentyl, hexyl, trifluoromethyl, hydroxy, methoxy, ethoxy, propoxy, butoxy, amino, methylamino, dimethylamino, ethylamino, and diethylamino. In another embodiment of Formula I-B, R9 and R10 are hydrogen, and R" and R'3are selected from the group consisting of hydrogen, methoxy, and fluoro. In another embodiment of Formula I-B, R9 R10 and R13 are hydrogen, and R" is methoxy.
Embodiments of 1 6 In one embodiment of the compounds of Formula I-B, Ys represents a bond or is selected from the group consisting of alkyl, alkenyl and alkynyl, wherein the Y6 alkyl, alkenyl and alkynyl substituents may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, cyano, oxo, cycloalkyl, -OR103, -C(O)R103, -C(O)OR103, -OC(O)R'03, -NR103R104, -N(R103)C(O)R'04 , and -C(O)NR 103 R104 ; and wherein R103 and R104 are independently selected from the group consisting of hydrogen and alkyl, wherein the alkyl may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, carboxy, cyano, oxo, alkynyl, haloalkynyl, hydroxyalkynyl, carboxyalkynyl, alkoxy, haloalkoxy, hydroxyalkoxy, and carboxyalkoxy.
In one embodiment of Formula I-B, Y6 represents a bond or is alkyl, optionally substituted as described in Formula I-B. In another embodiment of Formula I-B, Y6represents a bond or is Ci to C6 alkyl, optionally substituted as described in Formula I-B. In another embodiment of Formula I-B, Y6 represents a bond or is selected from the group consisting of Ci to C4 alkyl and hydroxyCi to C4-alkyl. In another embodiment of Formula I-B, Y6 represents a bond or is selected from the group consisting of methyl, ethyl, propyl, butyl, hydroxyethyl, hydroxypropyl, hydroxybutyl, dihydroxyethyl, and dihydroxybutyl. In one embodiment of Formula I-B, Y6 represents a bond or is selected from the group consisting of methyl, ethyl, propyl and butyl.
In one embodiment of Formula I-B, Y6 is alkyl optionally substituted with one or more substituents independently selected from the group consisting of halogen, cyano, oxo, cycloalkyl, -OR'03, -C(O)R103, -C(O)OR103, -OC(O)R 103, -NR 103 R104, -N(R103)C(O)R 104, and -C(O)NR103 R104 ;
wherein R103 and R104 are independently selected from the group consisting of hydrogen and Ci to C4 alkyl. In another embodiment of Formula I-B, Y6 is alkyl substituted with one or more substituents independently selected from the group consisting of fluoro, chloro, oxo, cycloalkyl, hydroxy, and carboxy. In another embodiment of Formula I-B, Y6 is alkyl substituted with one to three substituents independently selected from the group consisting of hydroxy and cyclohexyl.
In another embodiment of Formula I-B, Y6 is unsubstituted alkyl.
Embodiments of R6 Substituent In one embodiment of Formula I-B, R6 is selected from the group consisting of phenyl, phenyl-C(O)-, 3 to 10 membered ring heterocyclyl, phenyl-C(O)-NR105-, 3 to 10 membered ring heterocyclyl-C(O)-, and 3 to 10 membered ring heterocyclyl-C(O)-NR105-, wherein R6 is optionally substituted as described in Formula I-B, wherein R105 is selected from the group consisting of _..by_dr_o.geriansLC1-to_C6-aIkylJn_an_olher-embodiment_oiEormu.la1,J36.is ~elecfed_fr_om_the_group consisting of phenyl, phenyl-C(O)-, 5 to 7 membered ring heterocyclyl, phenyl-C(O)-NR105-, 5 to 7 membered ring heterocyclyl-C(O)-, and 5t07 membered ring I,Eterocyclyl-C(O)-NR105-, wherein R6 is optionally substituted as described in Formula I-B, wherein R105 is selected from the group consisting of hydrogen and C7 to C4 alkyl. In another embodiment of Formula I-B, R6 is selected from the group consisting of phenyl, phenyl-C(O)-, 5 to 6 membered ring heterocyclyl, phenyl-C(O)-NR105-, 5 to 6 membered ring heterocyclyl-C(O)-, and 5 to 6 membered ring heterocyclyl-C(O)-NR105-, wherein R6 is optionally substituted as described in Formula I-B
wherein R105 is hydrogen. In another embodiment, R6 is selected from the group consisting of phenyl, pyridinyl, piperidinyl, piperizinyl, morpholino, pyrazinyl, tetrahydropyran, tetrahydrofuran, isoxazole, imidazole, pyrrolidine, wherein R 6 is optionally substituted as described in Formula I.
In another embodiment, R6 is selected from the group consisting of morpholinyl, tetrahydropyranyl, tetrahydrofuranyl, piperidinyl, and pyrrolidinyl, wherein R6 is optionally substituted as provided in Formula I.
In one embodiment of Formula I, R6 is selected from the group consisting of phenyl, phenyl C(O)NH-, and phenyl C(O)-.
In one embodiment of Formula I, R6 is selected from the group consisting of 5 to 7 membered ring heteroaryl, 5 to 7 membered ring heteroaryl-C(O)-, and 5 to 7 membered ring heteroaryl-C(O)-NR105-, wherein R 6 is optionally substituted as described in Formula I, wherein R105 is selected from the group consisting of hydrogen and Ci to C4 alkyl. In another embodiment of Formula I, R6 is a 3 to 10 membered ring heteroaryl, optionally substituted as described in Formula I. In another embodiment of Formula I, R6 is a 5 to 7 membered ring heteroaryl, optionally substituted as described in Formula I. In another embodiment of Formula I, R6 is a 5 to 6 membered ring heteroaryl, optionally substituted as described in Formula 1. In another embodiment of Formula I, R 6 is a 5 to 6 membered ring heteroaryl containing one to three heteroatoms selected from the group consisting of 0 and N, wherein R6 is optionally substituted as described in Formula I. In another embodiment of Formula I, R6 is selected from the group consisting of imidazole, isoxazole, pyridinyl and pyrazynyl, wherein R6 is optionally substituted as described in Formula I. In another embodiment of Formula I, R6 is selected from the group consisting of imidazole, isoxazole, pyridinyl and pyrazynyl, wherein R 6 is optionally substituted with one or more substituents independently selected from the group consisting of alkyl, -OR105, and -C(O)R105, wherein R105 is selected from the group consisting of hydrogen, N\
~
methyl, and ethyl. In another embodiment, R6 is selected from the group consisting of N tq~
N O-N NJ' and In one embodiment of Formula I, R 6 is selected from the group consisting of 3 to 10 membered ring fully or partially saturated heterocylyl, 3 to 10 membered ring fully or partially - -saturated-heterocYclYI-C()- O-,-to-1-0-membered-rin g-fullY-orpartiallY
saturated heterocYclYI-C(O)NH-, wherein R6 is optionally substituted as described in Formula I. In another embodiment of Formula I, R6 is a 5 to 7 membered ring fully or partially saturated heterocylyl, 5 to 7 membered ring fully or partially saturated heterocyclyl-C(O)-, 5 to 7 membered ring fully or partially saturated heterocyclyi-C(0)NH-, wherein R 6 is optionally substituted as described in Formula 1. In another embodiment of Formula I, R6 is a 5 to 6 membered ring fully saturated heterocylyl, 5 to 6 membered ring fully saturated heterocyclyl-C(O)-, 5 to 6 membered ring fully saturated heterocyclyl-C(O)NH-, wherein R6 is optionally substituted as described in Formula I.
In another embodiment of Formula I, R6 is a 5 to 6 membered ring fully saturated heterocylyl, optionally substituted as described in Formula I. In another embodiment of Formula I, R6 is selected from the group consisting of tetrahydrofuran, tetrahydropyran, pyrrolidinyl, piperidinyl, piperazinyl, and morpholino, wherein R 6 is optionally substituted as described in Formula I. In another embodiment of Formula I, R6 is selected from the group consisting of tetrahydrofuran, tetrahydropyran, pyrrolidinyl, piperidinyl, piperazinyl, and morpholino, wherein R6 is optionally substituted with one or more substituents independently selected from the group consisting of alkyl, -OR105, -C(O)R105 and -C(O)OR105 wherein R105 is selected from the group consisting of hydrogen, methyl, and ethyl. In another embodiment, R6 is selected from the group consisting O
JL
N

of ~ O

~
HN
N

O
O N "N' N J~*-' HNJ N
~O N

O
N N''' N J~-~ ~
J N

O O
O
N-Zi' rN
rN OJ Oj O N
j and O

rN'jL'N-~-HOJ
.
In another embodiment of Formula I, Rs is optionally substituted with one or more substituents independently selected from the group consisting of -OR105, -C(O)R'05 -C(O)OR'05 -NR'0eRi06s -N(Ri05)C(O)R106-N(R10e)C(O)ORi06, -C(O)NR105Ri06, -NHC(O)NRioa.RiOS' -N(R105)S(O)2R106, wherein R105 and R'06 are independently selected from the group consisting of hydrogen, methyl, ethyl, methylethyl, tert-butyl, cyclopentyl, cyclohexyl, optionally substituted with one or more substituent selected from the group consisting of halogen, oxo, hydroxy, and methyl.. In another embodiment of Formula I, R6 is optionally substituted with one or more substituents independently selected from the group consisting of alkyl, --C(O)R105 and -C(O)OR105, wherein R105 is selected from the group consisting of hydrogen, methyl, and ethyl. In another embodiment of Formula I, R6 is optionally substituted with one or more substituents independently selected from the group consisting of -OH, -CH3, -CH2CH3, -C(O)CH3, -C(O)CH2CH3, -C(O)OCH2CH3.
Embodiments of R8 Substituent In one embodiment of Formula I-B, R8 is a(C, to C4)alkoxy(Cl to C4)alkyl, optionally substituted with one to three substituents selected from fluoro and chloro. In another embodiment of Formula I, R8 is methoxyethyl. In another embodiment of Formula I, R8 is ethoxyethyl. In another embodiment of Formula I, R8 is propoxyethyl. In another embodiment of Formula I, R8 is trifluoroethoxyethyl. In another embodiment of Formula I, R8 is selected from the group consisting of propoxyethyl and ethoxyethyl.
Embodiments of Multiple Substituents The following are additional embodiments of the compounds of Formula I-B.
Unless otherwise specified, substituents are as described in Formula I-B. Further embodiments of Formula I-B described when R9, R10, R", R'3, Y6, R6 and R8 are selected from the various embodiments described above.
Embodiments where R9, R10 are hydrogen, Ys represents a bond or is alkyl In one embodiment of Formula I-B, Y6 is a bond or is alkyl, optionally substituted as -- --descr-ibed-in-For-r-nula-I-B, and-R9 and-R'- are-hydr-ogen-In-another-embodiment-of Formula I-B, y6 is a bond or is alkyl, optionally substituted as described in Formula I-B, R9 and R10 are hydrogen and R" and R13 are selected from the group consisting of hydrogen, methoxy, and fiuoro. In another embodiment of Formula I-B, Y6 is a bond or is alkyl, optionally substituted as described in Formula I-B, R9 R10 and R'3 are hydrogen, and R" is methoxy.
Embodiments where R9, R10, R13 are hydrogen, Y6 represents a bond or isaikyl In one embodiment of Formula I-B, Y6 is a bond or is alkyl, optionally substituted as described in Formula I-B, and R9, R10 and R13 are hydrogen. In another embodiment of Formula I-B, Y6 is a bond or is alkyl, optionally substituted as described in Formula I-B, R9, R10 and R'3 are hydrogen and R" is selected from the group consisting of hydrogen, methoxy, and fluoro. In another embodiment of Formula I-B, Y6 is a bond or is alkyl, optionally substituted as described in Formula I-B, R9 R10 and R'3 are hydrogen, and R" is methoxy.
Embodiments where R9, R10, R'3 are hydrogen and R8 is ethoxyethyl or propoxyethyl In one embodiment of Formula I-B, R9, R10 and R'3 are hydrogen, and R8 is selected from the group consisting of ethoxyethyl and propoxyethyl. In another embodiment of Formula I-B, R9, R10 and R'3 are hydrogen and R" is selected from the group consisting of hydrogen, methoxy, and fluoro, and R8 is selected from the group consisting of ethoxyethyl and propoxyethyl. In another embodiment of Formula I-B, R9 R10 and R'3 are hydrogen, R" is methoxy, and R8 is ethoxyethyl, as described in Formula I-FF in Table A. In another embodiment of Formula I-B, R9 R10 and R'3 are hydrogen, R" is methoxy, and R8 is propoxyethyl, as described in Formula 1-UU in Table A.
Embodiments of Formula I-FF and 1-UU

In one embodiment of Formulae I-FF and I-UU, Y6 represents a bond or is alkyl optionally substituted as described in Formula I-B, and R6 is selected from the group consisting of hydrogen, halogen, oxo, hydroxy, amino, aminoalkyl, alkyl, alkynyl, alkoxy, alkylamino, and cycloalkyl, optionally substituted as described in Formula I-B. In another embodiment of Formulae I-FF and I-UU, Y6 represents a bond or is selected from the group consisting of unsubstituted Cl to C4 alkyl and unsubstituted hydroxyC, to C4-alkyl, and R6 is selected from the group consisting of phenyl, phenyl-C(O)-, 5 to 6 membered ring heterocyclyl, phenyl-C(O)-NR105-, 5 to 6 membered ring heterocyclyl-C(O)-, and 5 to 6 membered ring heterocyclyl-C(O)-NR105-, wherein R6 is optionally substituted as described in Formula I-B
wherein R105 is hydrogen. In another embodiment of Formulae I-FF and I-UU, Y6 represents a bond or is selected from the group consisting of methyl, ethyl, propyl, butyl, hydroxyethyl, hydroxypropyl, hydroxybutyl, dihydroxyethyl, and dihydroxybutyl, and R 6 is selected from the group consisting of phenyl, phenyl-C(O)-, 5 to 6 membered ring heterocyclyi, phenyl-C(O)-NH-, 5 to 6 membered ring heterocyclyl-C(O)-, and 5 to 6 membered ring heterocyclyl-C(O)-NH-, wherein R6 is optionally substituted with one or more substituents independently selected from the group consisting of -OH, -CH3, -CH2CH3, -C(O)CH3, -C(O)CH2CH3, -C(O)OCH2CH3.
Embodiments where R9, R10, R'3 are hydrogen, R8 is ethoxyethyl or propoxyethyl and R6 is selected from various embodiments In one embodiment of Formula I-B, R9, R10 and R'3 are hydrogen, R8 is selected from the group consisting of ethoxyethyl and propoxyethyl, and R 6 is selected from the group consisting of phenyl, phenyl C(O)NH-, phenyl C(O)-, 5 to 7 membered ring heterocyclyl, pher,yl-C(O)-NH-, 5 to 7 membered ring heterocyclyl-C(O)-, and 5 to 7 membered ring heterocyclyl-C(O)-NH, optionally substituted with one or more substituents independently selected from the group consisting of of -OR105, -C(O)R105, -C(O)OR'05-NR105R106, -N(R105)C(O)R1 6, -N(R105)C(O)OR106, -C(O)NR1 5R'06, -NHC(O)NR'0eRt06-N(R108)S(O)2R,1)6,0s 106 , wherein R and R
are independently selected from the group consisting of hydrogen, methyl, ethyl, methylethyl, tert-butyl, cyclopentyl, cyclohexyl, optionally substituted with one or more substituent selected from the group consisting of halogen, oxo, hydroxy, and methyl. In another embodiment of Formula I-B, R9, R10 and R'3 are hydrogen, R8 is selected from the group consisting of ethoxyethyl and propoxyethyl, and R6 is selected from the group consisting of 5 to 6 membered ring heteroaryl, 5 to 6 membered ring heteroaryl-C(O)-, and 5 to 6 membered ring heteroaryl-C(O)-NH, optionally substituted with one or more substituents independently selected from the group consisting of alkyl, -OR105, and -C(O)R105, wherein R105 is selected from the group consisting of hydrogen, methyl, and ethyl. In another embodiment of Formula I-B, R9, R10 and R13 are hydrogen, R8 is selected from the group consisting of ethoxyethyl and propoxyethyl, and R6 is selected from the group consisting of 5 to 6 membered ring fully saturated heterocyclyl, 5 to 6 membered ring fully saturated heterocyclyl-C(O)-, and 5 to 6 membered ring fully saturated heterocyclyl-C(O)-NH, optionally substituted with one or more substituents independently selected from the group consisting of alkyl, -OR105, and -C(O)R105, wherein R105 is selected from the group consisting of hydrogen, methyl, and ethyl.

Compounds of Formula (I-I) The present invention is directed, in part, to a class of compounds having the structure of Formula I-I:
R1o 8 R9 / R"
R

O ~ N \ ~ I R12 H
(I-I) wherein Ro, R10, R", R'2 and R'3 are independently selected from the group consisting of hydrogen, halogen, oxo, alkyl, -OR100, -C(O)R100, -OC(O)R100, -C(O)OR100, NR100R101 and -C(O)NR100R101 - , wherein the alkyl substitutent may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, oxo, -OR102, and -C(O)OR102;
wherein R10 , R101, and R102 are independently selected from the group consisting of hydrogen - - - and C1 to C4 alkyl; - - - - ---- --- - - -- - - -y6 represents a bond or is selected from the group consisting of alkyl, alkenyl and alkynyl, wherein (a) the Y6 alkyl, alkenyl and alkynyl substituents may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, cyano, oxo, cycloalkyl, -OR'03, -C(O)R103, -C(O)OR103, -OC(O)R103, -NR103R104, -N(R103)C(O)R104' and -C(O)NR103R104, , R103 and R104 are independently selected from the group consisting of hydrogen and alkyl, wherein the alkyl may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, carboxy, cyano, oxo, alkynyl, haloalkynyl, hydroxyalkynyl, carboxyalkynyl, alkoxy, haloalkoxy, hydroxyalkoxy, and carboxyalkoxy;

R6 is selected from the group consisting of aryl, aryl-C(O)-, heterocyclyl, aryl-C(O)-NR105-, and heterocyclyl-C(O)-NR105- wherein R6 may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, cyano, ~ -oxo, alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heterocyclyl, -OR106, -C(O)R106, -C(O)OR'06 OC(O)R106, -NR106R107, -N(R106)C(O)R107, -C(O)NR106 R107, -C(O)NR10sC(O)R107, -S(O)R106, -S(O)2R106, -N(R106)S(O)2R'07, and -S(O)2NR106R107; wherein the alkyl, alkenyl, alkynyl, cycloalkyl, aryl and heterocyclyl susbstituents may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, carboxy, cyano, oxo, alkoxy, haloalkoxy, hydroxyalkoxy, and carboxyalkoxy;

R105 is independently selected from the group consisting of hydrogen and alkyl;
R106 and R107 are independently selected from.the group consisting of hydrogen, alkyl, alkenyl, and alkynyl, wherein (a) the R106 and R107 alkyl and alkenyl substituents may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, carboxy, cyano, oxo, alkynyl, haloalkynyl, hydroxyalkynyl, carboxyalkynyl, alkoxy, haloalkoxy, hydroxyalkoxy, and carboxyalkoxy, and (b) the R106and R107 alkynyl substituents may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, carboxy, cyano, oxo, alkoxy, haloalkoxy, hydroxyalkoxy, and carboxyalkoxy;
R8 is alkoxyalkyl, optionally substituted with halogen.
Embodiments of R9, R10. Ri1, R'2 and R13 In one embodiment of the compounds of Formula I-I, R9, R10, R" R12, and R13 are selected from the group consisting of hydrogen, chloro, fluoro, methyl, ethyl, propyl, butyl, pentyl, hexyl, ___ tr-if-luoromethylr.-hydr-oxy,-methoxy,-ethoxy, propoxy, butoxy, amino, methylamino, dimethylamino, ethylamino, and diethylamino. In another embodiment of Formula I-I, R9, R10, R" R'2, and R13 are selected from the group consisting of hydrogen, fluoro, methyl, trifluoromethyl, methoxy, trifluoromethoxy, amino, methylamino, and dimethylamino. In another embodiment of Formula I-I, R9, R12 and R13 are hydrogen, and R10 and R'1 are selected from the group consisting of hydrogen, chloro, fluoro, methyl, ethyl, propyl, butyl, pentyl, hexyl, trifluoromethyl, hydroxy, methoxy, ethoxy, propoxy, butoxy, amino, methylamino, dimethylamino, ethylamino, and diethylamino. In another embodiment of Formula I-I, R9, R12 and R13 are hydrogen, and R" and R'3are selected from the group consisting of hydrogen, methoxy, trifluoromethyl, and fluoro. In another embodiment of Formula I-I, R9, R10, R'2 and R'3 are hydrogen, and R"
is fluoro.
Embodiments of Y6 In one embodiment of the compounds of Formula I-I, Ys represents a bond or is selected from the group consisting of alkyl, alkenyl and alkynyl, wherein the Y6 alkyl, alkenyl and alkynyl substituents may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, cyano, oxo, cycloalkyl, -OR103, -C(O)R103, -C(O)OR103, -OC(O)R103, -NR'03R104, -N(R103)C(O)R104, and -C(O)NR1 3R'04; and wherein R103 and R104 are independently selected from the group consisting of hydrogen and alkyl, wherein the alkyl may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, carboxy, cyano, oxo, alkynyl, haloalkynyl, hydroxyalkynyl, carboxyalkynyl, alkoxy, haloalkoxy, hydroxyalkoxy, and carboxyalkoxy.
In one embodiment of Formula I-I, Y6 represents a bond or is alkyl, optionally substituted as described in Formula I-I. In another embodiment of Formula I-I, Y6 represents a bond or is C1 to C6 alkyl, optionally substituted as described in Formula I-I. In another embodiment of Formula I-I, Y6 represents a bond or is selected from the group consisting of C, to C4 alkyl and hydr6xyC1 to C4_alkyl. In another embodiment of Formula I-I, Y6 represents a bond or is selected from the group consisting of methyl, ethyl, propyl, butyl, hydroxyethyl, hydroxypropyl, hydroxybutyl, dihydroxyethyl, and dihydroxybutyl. In one embodiment of Formula I-I, Y6 represents a bond or is selected from the group consisting of methyl, ethyl, propyl and butyl.
In one embodiment of Formula I-I, Ys is alkyl optionally substituted with one or more substituents independently selected from the group consisting of halogen, cyano, oxo, cycloalkyl, -OR103, -C(O)R1 031 -C(O)ORi03, -OC(O)Rioa, -NR703R' oa, -N(R10a)C(O)Rioa, and -C(O)NR,oaR,oa;
wherein R103 and R104 are independently selected from the group consisting of hydrogen and C, to C4 alkyl. In another embodiment of Formula I-I, Y6 is alkyl substituted with one or more substituents independently selected from the group consisting of fluoro, chloro, oxo, cycloalkyl, hydroxy, and carboxy. In another embodiment of Formula I-I, Y6 is alkyl substituted with one to three substituents independently selected from the group consisting of hydroxy and cyclohexyl.
In another embodiment of Formula I-I, Y6 is unsubstituted alkyl.
Embodiments of R6 Substituent In one embodiment of Formula I-I, R6 is selected from the group consisting of phenyl, phenyl-C(O)-, 3 to 10 membered ring heterocyclyi, phenyl-C(O)-NR105-, 3 to 10 membered ring heterocyclyl-C(O)-, and 3 to 10 membered ring heterocyclyl-C(O)-NR105- wherein R6 is optionally substituted as described in Formula I-I, wherein R105 is selected from the group consisting of hydrogen and C1 to C6 alkyl. In another embodiment of Formula I-I, R6 is selected from the group consisting of phenyl, phenyl-C(O)-, 5 to 7 membered ring heterocyclyi, phenyl-C(O)-NR105-, 5 to 7 membered ring heterocyclyl-C(O)-, and 5 to 7 membered ring heterocyclyl-C(O)-NR105-, wherein R6 is optionally substituted as described in Formula I-I, wherein R105 is selected from the group consisting of hydrogen and C1 to C4 alkyl. In another embodiment of Formula I-I, R6 is selected from the group consisting of phenyl, phenyl-C(O)-, 5 to 6 membered ring heterocyclyl, phenyl-C(O)-NR105-, 5 to 6 membered ring heterocyclyl-C(O)-, and 5 to 6 membered ring heterocyclyl-C(O)-NR105-, wherein R 6 is optionally substituted as described in Formula I-I wherein R105 is hydrogen. In another embodiment, R6 is selected from the group consisting of phenyl, pyridinyl, piperidinyl, piperizinyl, morpholino, pyrazinyl, tetrahydropyran, tetrahydrofuran, isoxazole, imidazole, pyrrolidine, wherein R6 is optionally substituted as described in Formula I-I.
In another embodiment, R6 is selected from the group consisting of morpholinyl, tetrahydropyranyl, tetrahydrofuranyl, piperidinyl, and pyrrolidinyl, wherein R6 is optionally substituted as provided in Formula I-I.
In one embodiment of Formula I-I, R6 is selected from the group consisting of phenyl, phenyl C(O)NH-, and phenyl C(O)-.
In one embodiment of Formula I-I, R6 is selected from the group consisting of 5 to 7 membered ring heteroaryl, 5 to 7 membered ring heteroaryl-C(O)-, and 5 to 7 membered ring heteroaryl-C(O)-NR105- wherein R6 is optionally substituted as described in Formula I-I, wherein R105 is selected from the group consisting of hydrogen and C, to C4 alkyl. In another embodiment of Formula I-I, R6 is a 3 to 10 membered ring heteroaryl, optionally substituted as described in Formula I-I. In another embodiment of Formula I-I, R 6 is a 5 to 7 membered ring heteroaryl, optionally substituted as described in Formula I-I. In another embodiment of Formula I-I, R 6 is a 5 to 6 membered ring heteroaryl, optionally substituted as described in Formula I-I. In another embodiment of Formula I-I, R6 is a 5 to 6 membered ring heteroaryl containing one to ' three heteroatoms selected from the group consisting of 0 and N, optionally substituted as described in Formula I-I. In another embodiment of Formula I-I, R6 is selected from the group consisting of imidazole, isoxazole, pyridinyl and pyrazynyl, wherein R6 is optionally substituted as described in Formula I-I. In another embodiment of Formula I-I, R6 is selected from the group consisting of imidazole, isoxazole, pyridinyl and pyrazynyl, wherein R6 is optionally substituted with one or more substituents independently selected from the group consisting of alkyl, -OR'05and -C(O)R105, wherein R105 is selected from the group consisting of hydrogen, methyl, and ethyl.
N~'2. N
~ NJ
In another embodiment, R6 is selected from the group consisting of ~ ~~ ~p.
O-N NJ
,and - - ---- ---In one enibodirnenf of Formula I-I,-R is selec~d from~he group consisting of-3 to 10-membered ring fully or partially saturated heterocylyi, 3 to 10 membered ring fully or partially saturated heterocyclyl-C(O)-, 3 to 10 membered ring fully or partially saturated heterocyclyl-C(O)NH-, wherein R6 is optionally substituted as described in Formula I-I. In another embodiment of Formula I-I-I, R6 is a 5 to 7 membered ring fully or partially saturated heterocylyl, 5 to 7 membered ring fully or partially saturated heterocyclyl-C(O)-, 5 to 7 membered ring fully or partially saturated heterocyclyl-C(O)NH-, wherein R6 is optionally substituted as described in Formula I-I. In another embodiment of Formula I-I, R6 is a 5 to 6 membered ring fully saturated heterocylyl, 5 to 6 membered ring fully saturated heterocyclyl-C(O)-, 5 to 6 membered ring fully saturated heterocyclyl-C(O)NH-, wherein R 6 is optionally substituted as described in Formula I-I.
In another embodiment of Formula I-I, R 6 is a 5 to 6 membered ring fully saturated heterocylyl, optionally substituted as described in Formula I-I. In another embodiment of Formula I-I, R6 is selected from the group consisting of tetrahydrofuran, tetrahydropyran, pyrrolidinyl, piperidinyl, piperazinyl, and morpholino, wherein R 6 is optionally substituted as described in Formula I-I. In another embodiment of Formula I-I, R 6 is selected from the group consisting of tetrahydrofuran, tetrahydropyran, pyrrolidinyl, piperidinyl, piperazinyl, and morpholino, wherein R6 is optionally substituted with one or more substituents independently selected from the group consisting of alkyl, -OR105, -C(O)R105 and -C(O)OR'05 wherein R105 is selected from the group consisting of hydrogen, methyl, and ethyl. In another embodiment, R6 is selected from the group consisting O
of , , O , , N. , O
O
~.
HN
N

O
O N N
r~
HNJ N
~O N

O
rN rN
N " NJ

O O
O

~ N rNNI rN
N J OJ OJ
and O

N H
OJ

In another embodiment of Formula I-I, R6 is optionally substituted with one or more substituents independently selected from the group consisting of -OR105, -C(O)R105, -C(O)OR105, -NR105R106, -N(R105)~=i(O)R106, -N(R105)C(0)OR106, -C(O)NR105 R106, -NHC(O)NR105R 106 ,-N(R1 5)S(O)2R106, wherein R105 and R106 are independently selected from the group consisting of hydrogen, methyl, ethyl, methylethyl, tert-butyl, cyclopentyl, cyclohexyl, optionally substituted with one or more substituent selected from the group consisting of halogen, oxo, hydroxy, and methyl.. In another embodiment of Formula I-I, R6 is optionally substituted with one or more substituents independently selected from the group consisting of alkyi, --C(O)R105 and -C(O)OR105, wherein R105 is selected from the group consisting of hydrogen, methyl, and ethyl. In another embodiment of Formula I-I, R6 is optionally substituted with one or more substituents independently selected from the group consisting of -OH, -CH3, -CH2CH3, -C(O)CH3, -C(O)CH2CH3, -C(O)OCH2CH3.
Embodiments of R( Substituent In one embodiment of Formula I-I, R8 is a(C1 to C4)alkoxy(C1 to C4)alkyl, optionally substituted with one to three substituents selected from fluoro and chloro. In another embodiment of Formula I-I, R 8 is trifluroethoxyethyl. In another embodiment of Formula I-I, R8 is ethoxyethyl. In another embodiment of Formula I, R 8 is propoxyethyl. In another embodiment of Formula I-I, R8 is trifluoroethoxyethyl. In another embodiment of Formula I-I, R8 is selected from the group consisting of propoxyethyl and ethoxyethyl.

Embodiments of Multiple Substituents The following are additional embodiments of the compounds of Formula I-I.
Unless otherwise specified, substituents are as described in Formula I-I. Further embodiments of ----For-mula-l--I -descr-ibed when-R9-, R10; Rii-R-'3;-Y6, Rs and-R8 are selected from the various embodiments described above.
Embodiments where R9, R12 and R13 are hydrogen, Ys represents a bond or is alkyl In one embodiment of Formula I-I, Y6 is a bond or is alkyl, optionally substituted as described in Formula I-I, and R9, R12 and R13 are hydrogen. In another embodiment of Formula I-(, y6 is a bond or is alkyl, optionally substituted as described in Formula I-I, R9, R'Z and R13 are hydrogen and R10 and R" are selected from the group consisting of hydrogen, methyl, methoxy, trifluoromethyl, and fluoro. In another embodiment of Formula I-I, Y6 is a bond or is alkyl, optionally substituted as described in Formula I-I, R9, R10, R'2 and R'3 are hydrogen, and R" is fluoro.
Embodiments where R9, R10, R13 are hydrogen and Re is ethoxyethyl or propoxyethyl In one embodiment of Formula I-I, R9, R10 and R'3 are hydrogen, and R8 is selected from the group consisting of ethoxyethyl and propoxyethyl. In another embodiment of Formula I-I, R9, R10 and R'3 are hydrogen and R" and R'2 are independently selected from the group consisting of hydrogen, methyl, methoxy, fluoro and trifluoromethyl, and R8 is selected from the group consisting of ethoxyethyl and propoxyethyl. In another embodiment of Formula I-I, R9 R1 and R13 are hydrogen, R" and R12 are independently selected from the group consisting of hydrogen and fluoro, and R8 is selected from the group consisting of ethoxyethyl and propoxyethyl.
Embodiments where R9, R70, R13 are hydrogen, R8 is ethoxyethyl or propoxyethyl and R 6 is selected from various embodiments In one embodiment of Formula I-I, R9, R10 and R'3 are hydrogen, R8 is selected from the group consisting of ethoxyethyl and propoxyethyl, and R6 is selected from the group consisting of phenyl, phenyl C(O)NH-, phenyl C(O)-, 5 to 7 membered ring heterocyclyl, phenyl-C(O)-NH-, 5 to 7 membered ring heterocyclyl-C(O)-, and 5 to 7 membered ring heterocyclyi-C(O)-NH, wherein R 6 is optionally substituted with one or more substituents independently selected from the group consisting of of -OR105, -C(O)R105, -C(O)OR105,-NR'05R'0s, -N(R105)C(O)R'0s, -N(R105)C(O)OR106, -C(O)NR105R106, -NHC(O)NR105Ri06, -N(R1e5)S(O)2R106, wherein R105 and R106 are independently selected from the group consisting of hydrogen, methyl, ethyl, methylethyl, tert-butyl, cyclopentyl, cyclohexyl, optionally substituted with one or more substituent selected from the group consisting of halogen, oxo, hydroxy, and methyl. In another embodiment of Formula I-I, R9, R10 and R'3 are hydrogen, R8 is selected from the group consisting of ethoxyethyl and propoxyethyl, and R 6 is selected from the group consisting of 5 to 6 membered ring heteroaryl, 5 to 6 membered ring heteroaryl-C(O)-, and 5 to 6 membered ring heteroaryl-C(O)-NH, wherein R 6 is optionally substituted with one or more substituents independently selected from the group consisting of alkyl, -OR105, and -C(O)R105, wherein R105 is selected from the group consisting of hydrogen, methyl, and ethyl. In another embodiment of Formula I-I, R9, R10 and R13 are hydrogen, R8 is selected from the group consisting of ethoxyethyl and propoxyethyl, and R 6 is selected from the group consisting of 5 to 6 membered ring fully saturated heterocyclyl, 5 to 6 membered ring fully saturated heterocyclyl-C(O)-, and 5 to 6 membered ring fully saturated heterocyclyl-C(O)-NH, wherein R 6 is optionally substituted with one or more substituents independently selected from the group consisting of alkyl, -OR105, and -C(O)R105, wherein R105 is selected from the group consisting of hydrogen, methyl, and ethyl.
C. Isomers When an asymmetric center is present in a compound of Formulae (I) through (I-UU) the compound will exist in the form of enantiomers. In one embodiment, the present invention comprises optical isomers and mixtures, including racemic mixtures of the compounds of Formulae (I) through (I-UU). In another embodiment, the present invention comprises diastereomeric forms (individual diastereomers and mixtures thereof) of compounds of Formulae (I) through (I-UU). When a compound of Formulae (I) through (1-UU) contains an alkenyl group or moiety, geometric isomers may arise.

D. Tautomeric Forms The present invention comprises the tautomeric forms of compounds of Formulae (I) through (1-UU). For instance, a tautomeric form of the following compound:

OH
O N
0 N ~

ON, /-N \N N
H

may be represented by:

NH
O
N
ON ---HN I i The various ratios of the tautomers in solid and liquid form is dependent on the various substituents on the molecule as well as the particular crystallization technique used to isolate a compound.
E. Salts The compounds of this invention may be used in the form of salts derived from inorganic or organic acids. Depending on the particular compound, a sait of the compound may be advantageous due to one or more of the salt's physical properties, such as enhanced pharmaceutical stability in differing temperatures and humidities, or a desirable solubility in water or oil. In some instances, a salt of a compound also may be used as an aid in the isolation, purification, and/or resolution of the compound.
Wher-e-a-salt is-intended to be administer-ed-to-a-patient (as opposed-to, for-example, being used in an in vitro context), the salt preferably is pharmaceutically acceptable. The term "pharmaceutically acceptable salt" refers to a salt prepared by combining a compound of Formuiae (I) - (I-CC) with an acid whose anion, or a base whose cation, is generally considered suitable for human consumption. Pharmaceutically acceptable salts are particularly useful as products of the methods of the present invention because of their greater aqueous solubility relative to the parent compound. For use in medicine, the salts of the compounds of this invention are non-toxic "pharmaceutically acceptable salts." Salts encompassed within the term "pharmaceutically acceptable salts" refer to non-toxic salts of the compounds of this invention which are generally prepared by reacting the free base with a suitable organic or inorganic acid.
Suitable pharmaceutically acceptable acid addition salts of the compounds of the present invention when possible include those derived from inorganic acids, such as hydrochloric, hydrobromic, hydrofluoric, boric, fluoroboric, phosphoric, metaphosphoric, nitric, carbonic, sulfonic, and sulfuric acids, and organic acids such as acetic, benzenesulfonic, benzoic, citric, ethanesulfonic, fumaric, gluconic, glycolic, isothionic, lactic, lactobionic, maleic, malic, methanesulfonic, trifluoromethanesulfonic, succinic, toluenesulfonic, tartaric, and trifluoroacetic acids. Suitable organic acids generally include, for example, aliphatic, cycloaliphatic, aromatic, araliphatic, heterocyclyl, carboxyic, and sulfonic classes of organic acids.
Specific examples of suitable organic acids include acetate, trifluoroacetate, formate, propionate, succinate, glycolate, gluconate, digluconate, lactate, malate, tartaric acid, citrate, ascorbate, glucuronate, maleate, fumarate, pyruvate, aspartate, glutamate, benzoate, anthranilic acid, mesylate, stearate, salicylate, p-hydroxybenzoate, phenylacetate, mandelate, embonate (pamoate), methanesulfonate, ethanesulfonate, benzenesulfonate, pantothenate, toluenesulfonate, 2-hydroxyethanesulfonate, sufanilate, cyclohexylaminosulfonate, algenic acid, f3-hydroxybutyric acid, galactarate, galacturonate, adipate, alginate, butyrate, camphorate, camphorsulfonate, cyclopentanepropionate, dodecylsulfate, glycoheptanoate, glycerophosphate, heptanoate, hexanoate, nicotinate, 2-naphthalesulfonate, oxalate, palmoate, pectinate, 3-phenylpropionate, picrate, pivalate, thiocyanate, and undecanoate.
In another embodiment, examples of suitable addition salts formed include the acetate, aspartate, benzoate, besylate, bicarbonate/carbonate, bisulphate/sulphate, borate, camsyate, citrate, edisylate, esylate, formate, fumarate, gluceptate, gluconate, glucuronate, hexafluorophosphate, hibenzate, hydrochloride/chloride, hydrobromide/bromide, hydroiodide/iodide, isethionate, lactate, malate, maleate, nitrate, orotate, oxalate, palmitate, pamoate, phosphate/hydrogen phosphate/dihidrogen phosphate, saccharate, stearate, succinate, tartrate, tosylate and trifluoroacetate salts.
In another embodiment, representative salts include benzenesulfonate, hydrobromide and hydrochloride.
Furthermore, where the compounds of the invention carry an acidic moiety, suitable pharmaceutically acceptable salts thereof may include alkali metal salts, e.g., sodium or potassium salts; alkaline earth metal salts, e.g., calcium or magnesium salts;
and salts formed ____with suitable organic ligands, e.g.., quaternary amrnonium_salts.._._.
In another embodiment, base salts are formed from bases which form non-toxic salts, including aluminum, arginine, benzathine, choline, diethylamine, diolamine, glycine, lysine, meglumine, olamine, tromethamine and zinc salts.
Organic salts may be made from secondary, tertiary or quaternary amine salts, such as tromethamine, diethylamine, N,N'-dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, meglumine (N-methylglucamine), and procaine.
Basic nitrogen-containing groups may be quaternized with agents such as lower alkyl (Ci to C6) halides (e.g., methyl, ethyl, propyl, and butyl chlorides, bromides, and iodides), dialkyl sulfates (e.g., dimethyl, diethyl, dibuytl, and diamyl sulfates), long chain halides (e.g., decyl, lauryl, myristyl, and stearyl chlorides, bromides, and iodides), arylalkyl halides (e.g., benzyl and phenethyl bromides), and others.
In one embodiment, salts of the compounds of this invention include hydrochloric acid (HCI) salts, trifluoroacetate (CF3COOH or 'TFA") salts, mesylate salts, and tosylate salts.
Pharmaceutically acceptable salts of compounds of Formulae (I) to (I-UU) may be prepared by one or more of three methods:
(i) by reacting the compound of any one of Formulae (I)- (I-UU) with the desired acid or base;
(ii) by removing an acid- or base-labile protecting group from a suitable precursor of the compound of any one of Formulae (I)- (I-UU) or by ring-opening a suitable cyclic precursor, for example, a lactone or lactam, using the desired acid or base;
and (iii) by converting one salt of the a compound of Formulae (I) through (I-UU) to another by reaction with an appropriate acid or base or by means of a suitable ion exchange column.
All three reactions are typically carried out in solution. The resulting salt may precipitate out and be collected by filtration or may be recovered by evaporation of the solvent. The degree of ionization in the resulting salt may vary from completely ionized to almost non-lonised.

F. Prodrups Also within the scope of the present invention are so-called "prodrugs" of the compounds of Formulae (I) through (1-UU). Thus, certain derivatives of compounds of any of Formulae (I) through (1-UU) which may have little or no pharmacological activity themselves can, when administered into or onto the body, be converted into compounds of any of Formulae (I) through (1-UU) having the desired activity, for example, by hydrolytic cleavage. Such derivatives are referred to as "prodrugs". Further information on the use of prodrugs may be found in "Pro-drugs as Novel Delivery Systems, Vol. 14, ACS Symposium Series (T Higuchi and W
Stella) and "Bioreversible Carriers in Drug Design", Pergamon Press, 1987 (ed. E B Roche, American -Pharm_aceutical Association)._ P_r_odrugs .in.acoordance with_the ir_wention can, for_example,._be produced by replacing appropriate functionalities present in the compounds of any of Formulae (I) through (1-UU) with certain moieties known to those skilled iri the art as "pro-moieties" as described, for example, in "Design of Prodrugs" by H Bundgaard New York, NY
(Elseview, 1985).
G. Methods of Treatment The present invention further comprises methods for treating a condition in a subject having or susceptible to having such a condition, by administering to the subject a therapeutically-effective amount of one or more compounds of Formulae (I) through (1-UU) as described above.
In one embodiment, the treatment is preventative treatment.
In another embodiment, the treatment is palliative treatment.
In another embodiment, the treatment is restorative treatment.
The conditions that can be treated in accordance with the present invention include, but are not limited to, cardiovascular diseases, metabolic diseases, central nervous system diseases, pulmonary diseases, sexual dysfunction, and renal dysfunction.

Conditions The present invention further comprises methods for treating a condition in a subject having or susceptible to having such a condition, by administering to the subject a therapeutically-effective amount of one or more compounds of Formula (I) through (I-UU). In another embodiment, the condition is a cGMP-mediated condition.
The conditions that can be treated in accordance with the present invention are PDE-5 mediated conditions. Such conditions include cardiovascular diseases, metabolic diseases, central nervous system diseases, pulmonary diseases, sexual dysfunction, and renal dysfunction.
In one embodiment, the condition is a cardiovascular disease, particularly a cardiovascular disease selected from the group consisting of hypertension (such as essential hypertension, pulmonary hypertension, secondary hypertension, isolated systolic hypertension, hypertension associated with diabetes, hypertension associated with atherosclerosis, and renovascular hypertension) ; complications associated with hypertension (such as vascular organ damage, congestive heart failure, angina, stroke, glaucoma and impaired renal function); valvular insufficiency; stable, unstable and variant (Prinzmetal) angina; peripheral vascular disease;
myocardial infarct; stroke; thromboembolic disease; restenosis;
arteriosclerosis; atherosclerosis;
pulmonary arterial hypertension; angiostenosis after bypass; angioplasty (such as percutaneous transluminal angioplasty, or percutaneous transluminal coronary angioplasty);
hyperlipidemia;
hypoxic vasoconstriction; vasculitis, such as Kawasaki's syndrome; heart failure (such as congestive, decompensated, systolic, diastolic, left ventricular heart failure, right ventricular heart failure, left ventricular hypertrophy); Raynaud's disease; preeclampsia;
pregnancy-induced high blood pressure; cardiomyopathy; and arterial occlusive disorders.
In another embodiment, the condition is hypertension. In another embodiment, the condition _______is_pulm.onary_aderial_h_yp.er_tension-IrLarLother-embodiment, the condition is heart failure. In another embodiment, the condition is diastolic heart failure. In another embodiment, the condition is systolic heart failure. In another embodiment, the condition is angina. In another embodiment, the condition is thrombosis. In another embodiment, the condition is stroke.
In another embodiment, the condition is a metabolic disease, particularly a metabolic disease selected from the group consisting of Syndrome X; insulin resistance or impaired glucose tolerance; diabetes (such as type I and type II diabetes); syndromes of insulin resistance (such as insulin receptor disorders, Rabson-Mendenhall syndrome, leprechaunism, Kobberling-Dunnigan syndrome, Seip syndrome, Lawrence syndrome, Cushing syndrome, acromegaly, pheochomocytoma, glucagonoma, pri'mary aldosteronism, somatostatinoma, Lipoatrophic diabetes, R-cell toxin induced diabetes, Grave's disease, Hashimoto's thyroiditis and idiopathic Addison's disease); diabetic complications (such as diabetic gangrene, diabetic arthropathy, diabetic nephropathy, diabetic glomerulosclerosis, diabetic deramatopathy, diabetic neuropathy, peripheral diabetic neuropathy, diabetic cataract, and diabetic retinopathy);
hyperglycemia; and obesity.
In another embodiment, the condition is insulin resistance. In another embodiment, the condition is nephropathy.
In another embodiment, the condition is a disease of the central nervous system, particularly a disease of the central nervous system selected from the group consisting of vascular dementia;
craniocerebral trauma; cerebral infarcts; dementia; concentration disorders;
Alzheimer's disease;
Parkinson's disease; amyolateral sclerosis (ALS); Huntington's disease;
multiple sclerosis;
Creutzfeld-Jacob; anxiety; depression; sleep disorders; and migraine. In one embodiment, the condition is Alzheimer's disease. In another embodiment, the condition is Parkinson's disease.

In one embodiment, the condition is ALS. In another embodiment, the condition is a concentration disorder.
In one embodiment, the condition is a pulmonary disease, particularly a pulmonary disease, selected from the group consiting of asthma; acute respiratory distress;
cystic fibrosis; chronic obstructive pulmonary disease (COPD); bronchitis; and chronic reversible pulmonary obstruction.
In one embodiment, the condition is sexual dysfunction, particularly sexual dysfunction selected from the group consiting of impotence (organic or psychic); male erectile dysfunction;
clitoral dysfunction; sexual dysfunction after spinal cord injury; female sexual arousal disorder;
female sexual orgasmic dysfunction; female sexual pain disorder; and female hypoactive sexual desire disorder. In another embodiment, the condition is erectile dysfunction.
In another embodiment, the condition is renal dysfunction, particularly a renal dysfunction selected from the group consisting of acute or chronic renal failure;
nephropathy (such as diabetic nephropathy); glomerulopathy; and nephritis.
In another embodiment, the condition is pain. In another embodiment, the condition is acute pain. Examples of acute pain include acute pain associated with injury or surgery. In another embodiment, the condition is chronic pain. Examples of chronic pain include neuropathic pain (including postherpetic neuralgia and pain associated with peripheral, cancer or diabetic -neur-opathy),-carpal't-unnel-syndrorne,-back-pain-(including-pain-associated-with-her.niated or ruptured intervertabral discs or abnormalities of the lumber facet joints, sacroiliac joints, paraspinal muscles or the posterior longitudinal ligament), headache, cancer pain (including tumour related pain such as bone pain, headache, facial pain or visceral pain) or pain associated with cancer therapy (including postchemotherapy syndrome, chronic postsurgical pain syndrome, post radiation syndrome, pain associated with immunotherapy, or pain associated with hormonal therapy), arthritic pain (including osteoarthritis and rheumatoid arthritis pain), chronic post-surgical pain, post herpetic neuralgia, trigeminal neuralgia, HIV neuropathy, phantom limb pain, central post-stroke pain and pain associated with chronic alcoholism, hypothyroidism, uremia, multiple sclerosis, spinal cord injury, Parkinson's disease, epilepsy and vitamin deficiency. In another embodiment, the condition is nociceptive pain (including pain from central nervous system trauma, strains/sprains, burns, myocardial infarction and acute pancreatitis, post-operative pain (pain following any type of surgical procedure), posttraumatic pain, renal colic, cancer pain and back pain). In another embodiment, the condition is pain associated with inflammation (including arthritic pain (such as osteoarthritis and rheumatoid disease pain), ankylosing spondylitis, visceral pain (including inflammatory bowel disease, functional bowel disorder, gastro-esophageal reflux, dyspepsia, irritable bowel syndrome, functional abdominal pain syndrome, Crohn's disease, ileitis, ulcerative colitis, dysmenorrheal, cystitis, pancreatitis and pelvic pain). In another embodiment, the condition is pain resulting from musculo-skeletal disorders (including myalgia, fibromyalgia, spondylitis, sero-negative (non-rheumatoid) arthropathies, non-articular rheumatism, dystrophinopathy, glycogenolysis, polymyositis and pyomyositis). In another embodiment, the condition is selected from the group consisting of heart and vascular pain (including pain caused by angina, myocardical infarction, mitral stenosis, pericarditis, Raynaud's phenomenon, scieredoma and skeletal muscle ischemia).
In another embodiment, the condition is selected from the group consisting of head pain (including migraine such as migraine with aura and migraine without aura), cluster headache, tension-type headache mixed headache and headache associated with vascular disorders; orofacial pain, including dental pain, otic pain, burning mouth syndrome and temporomandibular myofascial pain).
In another embodiment, the condition is a urologic condition selected from the group consisting of bladder outlet obstruction; incontinence and benign prostatic hyperplasia.
In another embodiment, the condition is an ophthalmic condition selected from retinal disease; macular degeneration and glaucoma.
In another embodiment, the condition is selected from the group consisting of tubulointerstitial disorders; anal fissure; baldness; cancerous cachexia;
cerebral apoplexy;
disorders of gut motility; enteromotility disorders; dysmenorrhoea (primary and secondary);
glaucoma; macular degeneration; antiplatelet; haemorrhoids; incontinence;
irritable bowel syndrome (IBS); tumor metastasis; multiple sclerosis; neoplasia; nitrate intolerance; nutcracker oesophagus; osteoporosis; infertility; premature labor; psoriasis; retinal disease; skin necrosis;
and urticaria. In another embodiment, the condition is osteoporosis.
In another embodiment, the condition is associated with endothelial dysfunction, particularly ----- c-onditions-selec#ed from-the-group-consisting-of--atherosclerotic-lesions, myocardial ischaemia, peripheral ischaemia, valvular insufficiency, pulmonary arterial hypertension, angina, vascular complications after vascular bypass, vascular dilation, vascular repermeabilisation, and heart transplantation.
The methods and compositions of the present invention are suitable for use with, for example, mammalian subjects such as humans, other primates (e.g., monkeys, chimpanzees), companion animals (e.g., dogs, cats, horses), farm animals (e.g., goats, sheep, pigs, cattle), laboratory animals (e.g., mice, rats), and wild and zoo animals (e.g., wolves, bears, deer). In another embodiment, the subject is a human.

Hypothesized Mechanism Without being held to a particular theory, it is hypothesized that compounds of Formulae (I) through (I-UU) inhibit PDE-5 and increase intracellular cGMP levels. This increase in intracellular cGMP reduces intracellular calcium signaling, resulting in vascular smooth muscle relaxation, and a reduction in hypertension.
Selected embodiments of the invention, therefore, comprise methods for treating a cGMP-mediated condition. via PDE-5 inhibition. A condition in which, for instance, insufficient cGMP is a major component, and whose production or action is modulated in response to PDE-5, would therefore be considered a disorder mediated by cGMP. Thus, compounds of Formulae (I) through (I-UU) would be therapeutically useful in methods for treating hypertension by administering to a hypertensive subject a therapeutically-effective amount of a compound of Forumulae (I) through (I-UU). Other examples of circulatory-related disorders which can be treated by compounds of the invention include congestive heart failure, renal failure, angina, and glaucoma.

Co-administration One or more compounds of the present invention can be used, alone or in combination with other therapeutic agents, in the treatment of various conditions or disease states. The compound(s) of the present invention and other therapeutic agent(s) may be may be administered simultaneously (either in the same dosage form or in separate dosage forms) or sequentially.
For instance, in one embodiment, one or more compounds of Formulae (I) through (I-UU) may be administered with aspirin.
In one embodiment, one or more compounds of Formulae (I) through (I-UU) may be co-administered with one or more angiotensin converting enzyme (ACE) inhibitors.
Examples of the one or more ACE inhibitors for use with the one or morecompound of Formulae (I) -(I-UU) include quinapril (such as ACCUPRILTM), perindopril (such as ACEONTM), captopril (such as CAPOTENTM), enalapril (such as VASOTECTM), ENALAPRILATTM, ramipril (such as ALTACETM), cilazapril, delapril, fosenopril (such as MONOPRILTM), zofenopril, indolapril, benazepril (such as L-O-T-E-NSINT); lisinopr-il-(such-as-PRINIVILTM or-ZESTRILT-"'); spirapril;
trandolapr-il-(such as MAVIKTM), perindep, pentopril, moexipril (such as UNIVASCTM) or pivopril.
In another embodiment, one or more compounds of Formulae (I) through (I-UU) may be co-administered with one or more alpha blockers such as dozazosin (such as CA;DURAT"~), phenoxybenzamine (such as DIBENZYLINETM), or terazosin (such as HYTRINTM), and CR-2991.
In another embodiment, one or more compounds of Formulae (I) through (1-UU) may be co-administered with one or more alpha-beta blockers such as Iabetalol (such as NORMODYNETM
or TRANDATETM).
In another embodiment, one or more compounds of Formulae (I) through (1-UU) may be co-administered with one or more angiotensin II receptor blockers such as candesartan (such as ATACANDTM), eprosartan (such as TEVETENTM), irbesartan (such as AVEPROTM), losartan (such as COZAARTM), olmesartan, olmesartan medoxomil (such as BENICARTM), tasosartan, telmisartan (such as MICARDISTM), valsartan (such as DIOVANTM) or zolasartan, FI-6828K, RNH-6270, UR-7198, Way-126227, KRH-594, TAK-536, BRA-657, and TA-606.
In another embodiment, one or more compounds of Formulae (I) through (I-UU) may be co-administered with one or more alpha-2-delta ligands such as gabapentin, pregabalin (such as LYRICATM), [(1R,5R,6S)-6-(aminomethyl)bicyclo[3.2.0]hept-6-yI]acetic acid, 3-(1-aminomethyl-cyclohexyimethyl)-4H-[1,2,4]oxadiazol-5-one, C-[1-(1 H-tetrazol-5-ylmethyl)-cycloheptyl]-methylamine, (3S,4S)-(1-aminomethyl-3,4-dimethyl-cyclopentyl)-acetic acid, (1a,3a,5(x)-(3-amino-methyl-bicyclo[3.2.0]hept-3-yl)-acetic acid, (3S,5R)-3-aminomethyl-5-methyl-octanoic acid, (3S,5R)-3-amino-5-methyl-heptanoic acid, (3S,5R)-3-amino-5-methyl-nonanoic acid and (3S,5R)-3-amino-5-methyl-octanoic acid), (2S,4S)-4-(3-Chlorophenoxy)praline, or (2S,4S)-4-(3-Fluorobenzyl)praline.
In another embodiment, one or more compounds of Formulae (I) through (I-UU) may be co-administered with one or more beta blockers such as timolol (such as BLOCARDENTM), carteolol (such as CARTROLTM), carvedilol (such as COREGTM), nadolol (such as CORGARDTM), propranolol (such as INNOPRAN XLTM), betaxolol (such as KERLONETM), penbutolol (such as LEVATOLTM), metoprolol (such as LOPRESSORTM or TOPROL-XLTM), atenolol (such as TENORMINTM), or pindolol (such as VISKENTM), and bisoprolol.
In another embodiment, one or more compounds of Formulae (I) through (I-UU) may be co-administered with one or more calcium channel blockers such as nifedipine (such as ADALATTM, ADALAT CCTM or PROCARDIATM), verapamil (such as CALANTM, COVERA-HST"', ISOPTIN
SRTM or VERELANTM), diltiazem (such as CARDIZEMTM CARDIZEM CDTM, CARDIZEM
LATM, CARDIZEM SRTM, DILACORTM, TIAMATETM or TIAZACTM), isradipine (such as DYNACIRCTM or DYNACIRC CRTM), amlodipine (such as NORVASCT'"), felodipine (such as PLENDILTM), nisoldipine (such as SULARTM), or bepridil (such as VASCORTM), vatanidipine, clevidipine, lercanidipine, dilitiazem, and NNC-55-0396.
In another embodiment, one or more compounds Formulae (I) through (1-UU) may be co-ister-ed with_one_or-mor_ecentral_antiadr.energics-such_as_methyldopa-(such-as-_- .-.
ALDOMETTM), clonidine (such as CATAPRESTM or CATAPRES-TTSTM), guanfacine (such as TENEXTM), or guanabenz (such as WYTENSINTM).
In another embodiment, one or more compounds oF Formulae (I) through (1-UU) may be co-administered with one or more diruretics such as hydroclorothiazide (such as MICROZIDETM or ORETICTM), hydroflumethiazide (such as SALURONTM), bemetanide (such as BUMEXTM), torsemide (such as DEMADEXTM), metolazone (such as ZAROXOLYNTM), chlorothiazide (such as DIURILTM, ESIDRIXTM or HYDRODIURILTM), triamterene (such as DYRENIUMTM), ethacrynic acid (such as EDECRINTM), chlorthalidone (such as HYGROTONTM), furosemide (such as LASIXTM), indapamide (such as LOZOLTM), or amiloride (such as MIDAMORTM or MODURETICTM).
In another embodiment, one or more compounds of Formulae (I) through (1-UU) may be co-administered with one or more glycosides / inotropic agents such as digoxin (such as LANOXINTM).
In another embodiment, one or more compounds of Formulae (I) through (1-UU) may be co-administered with one or more organic nitrates or an NO donors. "Nitric oxide donor" or "NO
donor" refers to a compound that donates, releases and/or directly or indirectly transfers a nitrogen monoxide species, and/or stimulate the endogenous production of nitric oxide or endothelium-derived relaxing factor (EDRF) in vivo and/or elevate endogenous levels of nitric oxide or EDRF in vivo. "NO donor" also includes compounds that are substrates for nitric oxide synthase. Examples of the one or more NO donors for use with one or more compounds of Formulae (I) through (I-UU) include S-nitrosothiols, nitrites, nitrates, N-oxo-N-nitrosamines, SPM
3672, SPM 5185, SPM 5186 and analogues thereof, sodium nitroprusside, nitroglycerin, isosorbide dinitrate, isosorbide mononitrate, molsidomine, SIN-1 or substrates of the various isozymes of nitric oxide synthase.
In another embodiment, one or more compounds of Formulae (I) through (I-UU) may be co-administered with one or more human B-type natriuretic peptides (hBNP) such as nesiritide (such as NATRECORTM).
In another embodiment, one or more compounds of Formulae (I) through (I-UU) may be co-administered with one or more renin inhibitors such as Aliskiren (SPP 100), SPP-500/600 and YS-004-39.
In another embodiment, one or more compounds of Formulae (I) through (I-UU) may be co-administered with one or more soluble guanylate cyclase activator ("sGCa").
An example of a suitable soluble guanylate cyclase activator is BAY-41-8543.
In another embodiment, one or more compounds of Formulae (I) through (I-UU) may be co-administered with one or more neutral endopeptidase (NEP) inhibitors, such as, for example, ( \
O
NH N

omapatrilat, fasidotril, mixanpril, sampatrilat, Z13752A , BMS-189921 , or H

N

CYS'-AN O O OH
O = H

~ MDL-100240 In another embodiment, one or more compounds of Formulae (I) through (I-UU) may be co-administered with one or more aldosterone receptor antagonists such as eplerenone (such as INSPRATM) or spironolactone (such as ALDACTONETM).
In another embodiment, one or more compounds of Formulae (I) through (I-UU) may be co-administered with one or more bradykinin agonists.
In another embodiment, one or more compounds of Formulae (I) through (I-UU) may be co-administered with one or more endothelian antagonists. Examples of suitable endothelin antagonists include ambrisentan, darusentan, J-1 04132, SPP-301, TBC-371 1, YM-62899, YM-91746, and BMS-193884.
In another embodiment, one or more compounds of Formulae (I) through (I-UU) may be co-administered with niacin or one or more nicotinic acid derivatives, such as NIACORTM, NIASPANTM, NICOLARTM, or SLO-NIACINr"'.

In another embodiment, one or more compounds of Formulae (I) through (I-UU) may be co-administered with one or more fibric acid derivatives, such as clofibrate (such as ATROMID-STM), gemfibrozil (such as LOPIDTM), or fenofibrate (such as TRICORTM).
In another embodiment, one or more compounds of Formulae (1) through (I-UU) may be co-administered with one or more cholesteryl ester transport protein inhibitors (CETPi), such as torcetrapib.
In another embodiment, one or more compounds of Formulae (I) through (1-UU) may be co-administered with one or more bile acid sequestants, such as colestipol (such as COLESTIDTM), cholestyramine (such as LOCHOLESTTM, PREVALITETM, QUESTRANTM, or QUESTRAN
LIGHTTM), colesevelam (such as WELCHOLTM).
In another embodiment, one or more compounds of Formulae (I) through (I-UU) may be co-administered with an apical sodium-dependent bile acid cotransporter inhibitors, such as SD-5613, AZD7806 or 264W94.
In another embodiment, one or more compounds of Formulae (I) through (1-UU) may be co-administered with one or more cholesterol absorbtion inhibitors, such as ezetimibe (such as ZETIATM).
In another embodiment, one or more compounds of Formulae (I) through (I-UU) may be co-_ -administexedLwith one_or_rnoce_3=b-ydr_oxy--3--meth_yJglutar_yJ
coQnzyme_A_(HMG-CoA) reductase inhibitors (statins) such as fluvastatin (such as LESCOLTM), atorvastatin (such as LIPITORTM), lovastatin (such as ALTOCORTM or MEVACORTM), pravastatin (such as PRAVACHOLTM), rosuvastatn (such as CRESTORTM), or simvastatin (such as ZOCORTM).
In another embodiment, one or more compounds of Formulae (I) through (I-UU) may be co-administered with one or more alpha glucosidase inhibitors, such as miglitol (such as GLYSETT"'), or acarbose (such as PRECOSETM).
In another embodiment, one or more compounds of Formulae (I) through (1-UU) may be co-administered with one or more biguanides, such as roseiglitazone (such as AVANDAMETT"'), or metformin (such as GLUCOPHAGETM or GLUCOPHAGE XRTM).
In another embodiment, one or more compounds of Formulae (I) through (I-UU) may be co-administered with one or more insulins, such as HUMALOGTM, HUMALOG 50/50T"', HUMALOG
75/25TM, HUMULIN 50/50T"', HUMALIN 75/25TM, HUMALIN LTM, HUMALIN NTM, HUMALIN
RrM, HUMALIN R U-500T"', HUMALIN UTM, ILETIN II LENTETM, ILETIN II NPHTM, ILETIN II
REGULART"~, LANTUSTM, NOVOLIN 70/30T~~, NOVILIN NTM, NOVILIN RT~", NOVOLOGTM, or VELOSULIN BRTM, and EXUBERATM.
In another embodiment, one or more compounds of Formulae (I) through (I-UU) may be co-administered with one or more meglitnides, such as repaglinide (such as PRANDINTM) or nateglinide (such as STARLIXTM).
In another embodiment, one or more compounds of Formulae (I) through (I-UU) may be co-administered with one or more sulfonylureas, such as glimepiride (such as AMARYLTM), glyburide (such as DIABETAT"~, GLYNASE PRESTABTM or MICRONASET"'), or glipizide (such as GLUCOTROLTM, or GLUCOTROL XLTM).

In another embodiment, one or more compounds of Formulae (I) through (I-UU) may be co-administered with one or more thiazolidinediones, such as pioglitazone (such as ACTOSTM) or rosiglitazone (such as AVANDIATM).

Administration and Dosing Typically, a compound described in this specification is administered in an amount effective to inhibit PDE-5. The compounds of the present invention are administered by any suitable route in the form of a pharmaceutical composition adapted to such a route, and in a dose effective for the treatment intended. Therapeutically effective doses of the compounds required to prevent or arrest the progress of or to treat the medical condition are readily ascertained by one of ordinary skill in the art using preclinical and clinical approaches familiar to the medicinal arts.
The dosage regimen for the compounds and/or compositions containing the compounds is based on a variety of factors, including the type, age, weight, sex and medical condition of the patient; the severity of the condition; the route of administration; and the activity of the particular compound employed. Thus the dosage regimen may vary widely. Dosage levels of the order from about 0.01 mg to about 100 mg per kilogram of body weight per day are useful in the treatment of the above-indicated conditions. In one embodiment, the total daily dose of a _ compound of Formulae_(I)_throu-gh (I_U_lJ)_(administered in singleor_divided doses) is typically from about 0. 01 to about 100 mg/kg. In another embodiment, total daily dose of the compound of Formulae (I) through (IX) is from about 0.1 to about 50 mg/kg, aizcf in another embodiment, from about 0.5 to about 30 mg/kg (i.e., mg compound of Formulae (I).through (1-UU) per kg body weight). In one embodiment, dosing is from 0.01 to 10 mg/kg/day. In another embodiment, dosing is from 0.1 to 1.0 mg/kg/day. Dosage unit compositions may contain such amounts or submultiples thereof to make up the daily dose. In many instances, the administration of the compound will be repeated a plurality of times in a day (typically no greater than 4 times).
Multiple doses per day typically may be used to increase the total daily dose, if desired.
For oral adminisiration, the compositions may be provided in the form of tablets containing 0.01, 0.05, 0.1, 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 25.0, 50.0, 75.0, 100, 125, 150, 175, 200, 250 and 500 milligrams of the active ingredient for the symptomatic adjustment of the dosage to the patient to be treated. A medicament typically contains from about 0.01 mg to about 500 mg of the active ingredient, or in another embodiment, from about 1 mg to about 100 mg of active ingredient. Intravenously, doses may range from about 0.1 to about 10 mg/kg/minute during a constant rate infusion.

H. Use in the Preparation of a Medicament In one embodiment, the present invention comprises methods for the preparation of a pharmaceutical composition (or "medicament) comprising the compounds of Formulae (I) through (I-UU) in combination with one or more pharmaceutically-acceptable carriers and/or other active ingredients for use in treating a cGMP-mediated condition.

In another embodiment, the invention comprises the use of one or more compounds of Formulae (I) through (1-UU) in the preparation of a medicament for the treatment of hypertension.
In another embodiment, the invention comprises the use of one or more compounds of Formulae (I) through (I-UU) in the preparation of a medicament for the treatment of angina.
In another embodiment, the invention comprises the use of one or more compounds of Formulae (I) through (1-UU) in the preparation of a medicament for the treatment of congestive heart failure.
In another embodiment, the invention comprises the use of one or more compounds of Formulae (I) through (1-UU) in the preparation of a medicament for the treatment of thrombosis.
In another embodiment, the invention comprises the use of one or more compounds of Formulae (I) through (I-UU) in the preparation of a medicament for the treatment of erectile dysfunction.

1. Pharmaceutical Compositions For the treatment of the conditions referred to above, the compounds of Formulae (I) through (I-UU) can be administered as compound per se. Alternatively, pharmaceutically acceptable salts are suitable for medical applications because of their greater aqueous solubility relative to __ the parent compound.___ In another embodiment, the present invention comprises pharmaceutical compositions.
Such pharmaceutical compositions comprise compounds of Formulae (I) through (I-UU) presented with a pharmaceutically-acceptable carrier. The carrier can be a solid, a liquid, or both, and may be formulated with the compound as a unit-dose composition, for example, a tablet, which can contain from 0.05% to 95% by weight of the active compounds.
Compounds of Formulae (I) through (I-UU) may be coupled with suitable polymers as targetable drug carriers.
Other pharmacologically active substances can also be present.
The active compounds of the present invention may be administered by any suitable route, preferably in the form of a pharmaceutical composition adapted to such a route, and in a dose effective for the treatment intended. The active compounds and compositions, for example, may be administered orally, rectally, parenterally, or topically.
Oral administration of a solid dose form may be, for example, presented in discrete units, such as hard or soft capsules, pills, cachets, lozenges, or tablets, each containing a predetermined amount of at least one compound of the present invention. In another embodiment, the oral administration may be in a powder or granule form. In another embodiment, the oral dose form is sub-lingual, such as, for example, a lozenge. In such solid dosage forms, the compounds of Formulae (I) through (I-UU) are ordinarily combined with one or more adjuvants. In the case of capsules, tablets, and pills, the dosage forms also may comprise buffering agentsor may be prepared with enteric coatings.
In another embodiment, oral administration may be in a liquid dose form.
Liquid dosage forms for oral administration include, for example, pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs containing inert diluents commonly used in the art (e.g., water). Such compositions also may comprise adjuvants, such as wetting, emulsifying, suspending, flavoring (e.g., sweetening), and/or perfuming agents.
In another embodiment, the present invention comprises a parenteral dose form.
"Parenteral administration" includes, for example, subcutaneous injections, intravenous injections, intraperitoneally, intramuscular injections, intrasternal injections, and infusion. Injectable preparations (e.g., sterile injectable aqueous or oleaginous suspensions) may be formulated according to the known art using suitable dispersing, wetting agents, and/or suspending agents.
In another embodiment, the present invention comprises a topical dose form.
"Topical administration" includes, for example, transdermal administration, such as via transdermal patches or iontophoresis devices, intraocular administration, or intranasal or inhalation administration. Compositions for topical administration also include, for example, topical gels, sprays, ointments, and creams. A topical formulation may include a compound which enhances absorption or penetration of the active ingredient through the skin or other affected areas. When the compounds of this invention are administered by a transdermal device, administration will be accomplished using a patch either of the reservoir and porous membrane type or of a solid matrix variety. Formulations suitable for topical administration to the eye include, for example, eye drops wherein the compound of this invention is dissolved or suspended in suitable carrier.
For intranasal administration or administration by inhalation, the active compounds of the invention are conveniently delivered in the form of a solution or suspension from a pump spray container that is squeezed or pumped by the patient or as an aerosol spray presentation from a pressurized container or a nebulizer, with the use of a suitable propellant.
In another embodiment, the present invention comprises a rectal dose form.
Such rectal dose form may be in the form of, for example, a suppository.
Other carrier materials and modes of administration known in the pharmaceutical art may also be used. Pharmaceutical compositions of the invention may be prepared by any of the well-known techniques of pharmacy, such as effective formulation and administration procedures.
The above considerations in regard to effective formulations and administration procedures are well known in the art and are described in standard textbooks. Formulation of drugs is discussed in, for example, Hoover, John E., Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, Pennsylvania, 1975; Liberman, et al., Eds., Pharmaceutical Dosage Forms, Marcel Decker, New York, N.Y., 1980; and Kibbe, et al., Eds., Handbook of Pharmaceutical Excipients (3rd Ed.), American Pharmaceutical Association, Washington, 1999.

J. Kits The present invention further comprises kits that are suitable for use in performing the methods of treatment or prevention described above. In one embodiment, the kit contains a first dosage form comprising one or more of the compounds of the present invention and a container for the dosage, in quantities sufficient to carry out the methods of the present invention.
n another embodiment, the kit of the present invention comprises one or more compounds of Formulae (I) through (I-UU) and an ACE inhibitor.

In another embodiment, the kit of the present invention comprises one or more compounds of Formulae (I) through (I-UU) and an angiotensin II receptor antagonist.
In another embodiment, the kit of the present invention comprises one or more compounds of Formulae (I) through (I-UU) and an aidosterone receptor antagonist.
In another embodiment, the kit of the present invention comprises one or more compounds of Formulae (I) through (I-UU) and a NO donor.

L. Compound Preparations Schemes The starting materials used herein are commercially available or may prepared by routine methods well known to those of ordinary skill in the art (such as those methods disclosed in standard reference books such as the COMPENDIUM OF ORGANIC SYNTHETIC METHODS, Vol. I-VI (published by Wiley-Interscience)).
The compounds of the present invention may be prepared using the methods illustrated in the general synthetic schemes and experimental procedures detailed below. The' general synthetic schemes are presented for purposes of illustration and are not intended to be limiting.
Scheme 1 CI Boc20 CI nBuLi F CI
I\ I\ then F+ I\
H2N ~ N BocHN ~ N. BocHN N
II III IV
Ra Ra i i NH2R8 HN \ CI Acid HN CI
I~N I~
BocHN H2N
V V VI

CIC(O)CO2Et R$ POCI3 R8 NR3, heat O N Cl nPrCN, reflux O N CI
OR ' I OR I
Oxalic acid, O H (COCI)2, DMF, CI N
H3O + VII CH2CI2, rt VIII
R8 R$
NHXs(Ys-Rs) s O N ~ CI ArB(OH)2 O N Ar R.Ys o R2.
N N ~ Pd N N
, N i Xs IX R2 I
Scheme 1 outlines a general procedure for the preparation of 7-aryl, 1, 3-disubstituted pyrido[3,4-b]pyrazin-2(1 H)-one of formula I. The starting material is the commercially available 6-chloropyridin-3-amine II. 6-chloropyridin-3-amine II is converted to tert-butyl 6-chloropyridin-3-ylcarbamate III by treatment with reagents such as di-tert-butyl dicarbonate, (2E)-{[(tert-butoxycarbonyl)oxy]imino}(phenyl)acetonitrile and tert-butyl phenyl carbonate.
This reaction is carried out in solvents such as dioxane, tetrahydrofuran, water, ethyl acetate or dichloromethane, in the presence or absence of inorganic bases such as potassium carbonate or sodium bicarbonate or organic bases such as triethylamine, 4-methylmorpholine, pyridine or N,N-diisopropylethylamine at temperatures ranging from room temperature to 110 C. tert-butyl 6-chloropyridin-3-ylcarbamate III is converted to tert-butyl 6-chloro-4-fluoropyridin-3-ylcarbamate IV by metallation followed by quenching with an electrophilic fluorine source.
Lithation could be achieved be treating tert-butyl 6-chloropyridin-3-ylcarbamate III with an organolithium such as n-butyl lithium or t-butyl lithium in the presence or absence of additives such as N,N,N;N'-tetramethylethylenediamine in solvents such as diethyl ether or tetrahydrofuran at temperatures ranging from -80 C to 0 C. Suitable electrophilic fluorine sources include N-flurobenzenesulfonimide. Addition of primary and benzylic amines to tert-butyl 6-chloro-4-fluoropyridin-3-ylcarbamate IV afforded amines of the formula V. This conversion could be achieved by treatment of IV with amines in solvents such as ethyl alcohol, isopropyl alcohol, dimethylformamide, dimethylactemide, toluene, dioxane and dichloroethane in the presence or absence of inorganic bases such as potassium carbonate or sodium bicarbonate or organic bases such as triethylamine, 4-methylmorpholine, pyridine or N,N-diisopropylethylamine at temperatures ranging from room temperature to 110 C. Amines of the formula V
could be converted to diamines of the formula VI by removing the carbamate protecting group under standard conditions, as described in Green, T., Wuts, P. Protecting Groups in Organic Synthesis, John Wiley & Sons, INC, Second edition, 1991, pp 309-405. The diamines of formula VI could be converted to the diones of formula VII using various reaction procedures.
In one procedure, this conversion is achieved by refluxing an aqueous solution of VI in the presence of oxalic acid and a catalytic amount of a mineral acid such as HCI. Alternatively, this conversion to structures of formula VII could be achieved by addition of either methyl chlorooxoacetate or oxalyl chloride to a solution of VI in the presence of an organic base such as triethylamine, 4-methylmorpholine, or N,N-diisopropylethylamine, at 0 C, followed by warming to either room temp or the reflux temperature of the solvent. Suitable solvents include toluene, dichloromethane, dicholroethane, dioxane, or tetrahydrofuran. The chloroimidate of formula VIII could be prepared by a number of methods. In one procedure, a dione of formula VII could be heated to reflux in the presence of phosphorous oxychloride and a phase transfer catalyst such as tetraethylammonium chloride.
Suitable solvents for this reaction include propionitrile or acetonitrile. In an alternate procedure, the formation of chloroimidate VII is achieved by dissolving VII in a suitable solvent such as dichloromethane, tetrahydrofuran, or dioxane and treating it with oxalyl chloride in the presence of a catalytic amount of dimethylformamide between 0 C and rt. The 6-aminopyrazinones of formula IX could be prepared by the addition of various primary and secondary amines to chloroimidate VIII in the presence of an organic base such as triethylamine, 4-methylmorpholine, or N,N-diisopropylethylamine at temperatures ranging from 0 C to rt. Suitable solvents include dich lorom ethane, tetrahydrofuran, and dioxane. Formation of the desired pteridinone of formula I
could be prepared through a standard palladium catalyzed Suzuki coupling between chloride IX
and suitable boronic acids, as described in Miyaura, N., Suzuki, A; Chem Rev.
1995, 95, 2457-2483. A solution of the chloride, IX, in a suitable solvent such as tetrahydrofuran or dioxane is heated to reflux in the presence of the desired boronic, an inorganic base such as sodium carbonate or cesium carbonate, and a palladium(0) source such palladium(II) acetate or tetrakis(triphenylphosphine)palladium to give compounds of formula I.

Scheme 2 CI nBuLi, I+ ' CI NH2Rg RBHN z CI
BocHN N BocHN ~N BocHN I ~N
III x V

Scheme 2 outlines an alternate conversion of tert-butyl 6-chloropyridin-3-ylcarbamate III to amines of formula V. tert-butyl 6-chloropyridin-3-ylcarbamate III is converted to tert-butyl 6-chloro-4-iodopyridin-3-ylcarbamate X by metallation followed by quenching with an electrophilic iodine source. Lithation could be achieved be treating tert-butyl 6-chloropyridin-3-ylcarbamate III
with an organolithium such as n-butyl lithium or t-butyl lithium in the presence or absence of additives such as N,N,N;N'tetramethylethylenediamine in solvents such as diethyl ether or tetrahydrofuran at temperatures ranging from -80 C to 0 C. Suitable electrophilic iodine sources include molecular iodine and 1-iodopyrrolidine-2,5-dione. Addition of primary and benzylic amines to tert-butyl 6-chloro-4-iodopyridin-3-ylcarbam ate IV
afforded amines of the formula V. Amines of the formula X could be converted to diamines of the formula V by standard coupling techniques as described in Ley, S., Thomas, A.; Angew. Chem. Int. Ed.
2003, 42, 5400-5449. A solution of iodide, X, in a suitable solvent such as tetrahydrofuran, dioxane, toluene, benzene, N,N dimethylformamide, isopropanol, ethanol or propionitrile is stirred at temperatures ranging from room temperature to reflux in the presence of the desired amine, a base such as sodium carbonate, cesium carbonate, potassium phosphate, or sodium tert-butoxide and a palladium with ligand and/or a copper source. Suitable sources of palladium include palladium(II) acetate, tetrakis(triphenylphosphine)palladium, dichloro[1,1'-bis(diphenylphosphino)ferrocene]palladium (II) dichloromethane adduct and tris(dibenzylideneacetone)dipalladium(0). Suitable ligands include triphenylphosphine, tri-2-furylphosphine, 4,5-bis(diphenylphosphine)-9-9-dimethylxathene, tricyclohexylphospine, tert-butylphospine and 2,2'-bis(diphenylphosphino)-1,1'-binapthyl. Suitable sources of copper include copper(II) acetate, copper(l) iodide and copper(l) chloride.

Scheme 3 outlines a one-pot procedure for the conversion of diaminopyridine of formula VI
to amino substituted pyrazinone of formula IX. The pyridine VI is dissolved in a solvent such as dichloromethane, tetrahydrofuran, or dioxane and cooled to 0 C. The mixture is treated with oxalyl chloride and allowed to slowly warm to room temperature. The reaction is typically mixed for 4-24 hours. The reaction mixture is then recooled to 0 C, treated with an organic base such as triethylamine, 4-methylmorpholine, or N,IV diisopropylethylamine, followed by addition of the requisite primary or secondary amine leading to isolation of the desired amine of formula IX.

Scheme 3 1) (COCI)2 R
R1 CH2CI2, 0 C_ rt O Ni I ~ CI
HN CI 2) Cool to 0 C R21 N~ ~ N
add organic base i N
H2N H Rs ~N, VI Rs R2 IX

The preparation of substituted amine analogs in the C(6) position similar to compounds of formula XIII or XIV are shown in Scheme 5. The starting amines XI can be prepared by utilizing mono-protected diamines in the final step of Scheme 1. The protecting groups used are typically carbamates which are removed using standard conditions to afford free amines of formula XII.2 Derivatives of formula XIII can be prepared by adding the desired aldehyde to a solution of XII in the presence of a catalytic amount of acids such as hydrochloric or acetic acid. A reducing agent such as sodium borohydride, sodium cyanoborohydride, or sodium triacetoxyborohydride is then added to the mixture leading to amines of formula XIII. Suitable solvents include tetrahydrofuran, dioxane, or dichloromethane. Amide derivatives of formula XIV could be prepared by treatment of the amine XII with activated esters such a acid chlorides, or acid derivatives prepared from acids utilizing_peptide coupling reagents such as 1-[3_(Dimethylaminopropy]-3-ethylcarbodiimide methiodide , 1,3-Dicyclohexylcarbodiimide, or 0-(7-Azabenzotriazol-1-yl)-1,1,3,3-tetramethyluromium hexafluorophosphate in the presence of an organic base such as triethylamine, N, N-diisopropylethylamine, 4-methyimorpholine. Suitable solvents include dichloromethane, tetrahydrofuran, or dioxane.

Scheme 5 O N "z Ar R$
Acid O N Ar N
~N~H TIN H2N ~N
I ~ N
~pp H

XI XII

p 0 N Ar.
RAH \I-H N
N
n H
H+, Reducing agent s O N Ar XIII
N

1n H O

R$
Base, 0 C I
- O N Ar - --- -- ~ . ~ p - -------OR R ' NH~~,- ~N_ ~ N
O 11n H
RsA-pH xiv Coupling agent, base, rt Compound Examples The following illustrate the synthesis of various compounds of the Formulae (I)-(I-EE). Other compounds of this invention may be prepared using the methods illustrated in these Examples, either alone or in combination with techniques generally known in the art.

Example 1 0f ~~ N
O O )CCra N N
H
7-(6-methoxypyridin-3-yl)-3-[(2-morpholin-4-ylethyl)amino]-1-(2-propoxyethyl)pyrido[3,4-b]pyrazin-2(1 H)-one.
Step 1: Preparation of tert-butyl 6-chloropyridin-3-yicarbamate.
A solution of 5-amino-2-chloropyridine (30.94 g, 236 mmol) and di-tert-butyldicarbonate (65.36 g, 299 mmol) in 1,4-dioxane (300 mL) was stirred at reflux for 20 hours. Additional di-tert-butyidicarbonate (8.30 g, 38 mmol) was added and the reaction was stirred at reflux for 7 hours.
The reaction was cooled to room temperature and poured into water. The layers were separated and the aqueous layer was extracted with ethyl acetate. The combined organic layers were washed with brine, dried over magnesium sulfate, and solvent was removed at reduced pressure to give a brown oil. The oil was triturated with diethyl ether and filtered to give tert-butyl 6-chloropyridin-3-ylcarbamate as a tan solid. (49.84 g, 92% yield). 'H NMR
(CDCI3) b 8.24 (m, 1 H), 7.96 (1 H), 7.27 (1 H), 6.65 (1 H), 1.51 (9H).
Step 2: Preparation of tert-butyl 6-chloro-4-fluoropyridin-3-ylcarbamate.
F
H
OyN ~
~ 0 ~ ~
N CI
To a-63 C solution of tert-butyl 6-chloropyridin-3-ylcarbamate (24.99 g, 109.3 mmol) and TMEDA (39 mL, 260.0 mmol) in diethyl ether (700 mL) was added a 1.6M n-butyl lithium solution in hexane (193 mL, 308.8 mmol) over a period of 30 minutes while maintaining the temperature of the reaction at -60 to -50 C. The reaction was stirred at -60 C for an additional 10 minutes after the addition was complete then warmed to -10 C and stirred at -25 to -10 C for 2.0 hours.
The reaction was cooled to -60 C and a solution of N-fluorobenzenesulfonimide (53.49 g, 169.6 15-mmel)-in tetrahydrofu"rari (155-mQ-Was ad"ded'while keeping-the temperature below -50 C. It precipitated on addition and stirring became difficult. The reaction was then allowed to slowly warm to 0 C over 1 hour. The reaction was quenched with saturated ammonium chloride solution (400 mL). The layers were separated and the aqueous layer was extracted with ethyl acetate (2 x 250 mL). The combined organic layers were washed with brine, dried over magnesium sulfate, and solvent was removed at reduced pressure to give an oily brown solid. The material was passed through a column of silica gel with 20% ethyl acetate/ hexane. The 6-chloro-4-fluoropyridin-3-ylcarbamate was obtained as a yellow solid. (15.88 g, 59%
yield). 'H NMR
(CDCI3) b 9.09 (1 H), 7.12 (1 H), 6.55 (1 H), 1.54 (s, 9H).
Step 3: Preparation of tert-butyl 6-chloro-4-f(2-propoxyethyl)aminolpyridin-3-ylcarbamate.
O
INH
H
>rOYN
0 ( N CI
A solution of tert-butyl 6-chloro-4-fluoropyridin-3-ylcarbamate (11.96 g, 48.5 mmol) and 2-n-propoxyethylamine (11.8 mL, 97.2 mmol) in ethanol (120 mL) was stirred at reflux for 22 hours.
The reaction was cooled to room temperature and solvent was removed at reduced pressure to give a yellow solid which was triturated with diethyl ether and filtered to give 6-chloro-4-[(2-propoxyethyl)amino]pyridin-3-ylcarbamate as a white solid. (13.08 g, 82%
yield). 'H NMR

(CDCI3) S 7.92 (1 H), 6.54 (1 H), 5.77 (1 H), 5.11 (1 H), 3.65 (2H), 3.44 (2H), 3.34-3.29 (2H), 1.65-1.56 (2H), 1.49 (9H), 0.94 (3H).
Step 4: Preparation of 6-ch!oro-N4-(2-propoxyethyl)pyridine-3,4-diamine.
O
HN ~ CI

~ /N

A solution of tert-butyl 6-ch!oro-4-[(2-propoxyethyl)amino]pyridin-3-y!carbamate (7.08 g, 21.4 mmol) in 1,4-dioxane (20 mL) was treated with 4N HCI in 1,4-dioxane (100 mL) and stirred at room temperature for one hour. The reaction was partitioned between ethyl acetate and saturated sodium bicarbonate solution. The layers were separated and the aqueous layer was extracted with ethyl acetate. The combined organic layers were washed with brine, dried over magnesium sulfate, and solvent was removed at reduced pressure to give 6-ch!oro-N4-(2-propoxyethyl)pyridine-3,4-diamine as a brown oil. (4.93 g, 100% yield). 1H NMR
(CDCI3) 8 7.63 (1 H), 6.45 (1 H), 4.67 (1 H), 3.67 (2H), 3.43 (2H), 3.32-3.27 (2H), 2.92 (2H), 1.64-1.55 (2H), 0.93 (3H).
Step 5: Preparation of 3,7-dich!oro-1-(2-propoxyethy!)pyr!do[3,4-blpyrazin-2(1H)-one.
0J"' ~
O N CI
CI~N N
A 0 C solution of 6-ch!oro-N4-(2-propoxyethyl)pyridine-3,4-diamine (2.80 g, 12.2 mmol) and diisopropylethylamine (4.6 mL, 25.7 mmol) in dichloromethane (100 mL) was treated with methyl ch!orooxoacetate(1.1 mL, 11.7 mmol), allowed to warm to room temperature and stirred for four hours. The reaction was diluted with dichloromethane and washed with saturated sodium bicarbonate solution, dried over magnesium sulfate, and solvent was removed at reduced pressure. The residue was dissolved in toluene (30 mL) and heated at 105 C for four hours. The solvent was removed at reduced pressure and the resulting solid taken up in dichloromethane (100 mL) and treated with oxalyl chloride (2.1 mL, 24.1 mmol) and DMF (3 drops). The reaction was stirred at room temperature for 6 hours. The solvent was removed at reduced pressure to give a brown solid. This was passed through a column of silica gel with 70%
ethyl acetate/
hexane to give 3,7-dich!oro-1-(2-propoxyethyl)pyrido[3,4-b]pyrazin-2(1 H)-one as a white solid.
(2.44 g, 66% yield). 1 H NMR (CDCI3) b 8.78 (1 H), 7.59 (1 H), 4.40 (2H), 3.80 (2H), 3.35 (2H), 1.52-1.46 (2H), 0.82 (3H).

Step 6: Preparation of 7-chloro-3-f(2-morpholin-4-ylethyl)aminol-l-(2-propoxyethyl)pyridof3,4-blpyrazin-2(1 H)-one.

)-10 ~
O N CI
N,,_~ N
N N
H
A solution of 3,7-dichloro-1 -(2-propoxyethyl)pyrido[3,4-b]pyrazin-2(1 H)-one (193 mg, 0.64 mmol), 4-(2-aminoethyl)morpholine (118 mg, 0.91 mmol) and triethylamine (0.15 mL, 1.07 mmol) in THF (3 mL) was stirred at room temperature for one hour. The reaction was partitioned between ethyl acetate and water. The layers were separated and the aqueous layer was extracted with ethyl acetate. The combined organic layers were washed with brine, dried over magnesium sulfate, and solvent was removed at reduced pressure to give a brown oil. This was passed through a column of silica gel with 80-100% ethyl acetate/ hexane to give 7-chloro-3-[(2-morpholin-4-ylethyl)amino]-1-(2-propoxyethyl)pyrido[3,4-b]pyrazin-2(1H)-one as a pink oil. (183 mg, 72% yield). 1H NMR (CDCI3) b 8.49 (1 H), 7.36 (1 H), 6.93 (1 H), 4.36 (2H), 3.77-3.72 (6H), - - - ----3:64 3.-58-(2H), 3.36 (2H),--2:66-(2H), 2.-53-2.50 (-H)-, 1:54-1.47 (2H); 0.83 (3H).
Step 7: Preparation of 7-(6-methoxypyridin-3-yl)-3-f(2-morpholin-4-ylethyl)aminol-l-(2-propoxyethyl)pyridof3,4-blpyrazin-2(1 H)-one.

~ oll, cx N N
H
A solution of 7-chloro-3-[(2-morpholin-4-ylethyl)amino]-1-(2-propoxyethyl)pyrido[3,4-b]pyrazin-2(1 H)-one (72 mg, 0.18 mmol) in 1,4-dioxane (2.5 mL) was treated with"
tetrakis(triphenylphosphine) palladium(0) (19 mg, 0.016 mmol) and stirred at room temperature for five minutes. A warm solution of 2-methoxy-5-pyridineboronic acid (41 mg, 0.27 mmol) in ethanol (0.5 mL) and 2.0 M aqueous sodium carbonate 1.5 mL) were added. The mixture was refluxed for 2.0 hours, filtered hot through celite and the filtrate was concentrated under reduced pressure. The residue was partitioned between ethyl acetate and water, and the Cayers were separated. The aqueous layer was extracted with ethyl acetate. The combined organic layers were washed with brine, dried over magnesium sulfate, concentrated under reduced pressure, and passed through a column of silica gel with 2% methanol/ dichloromethane.
Fractions were concentrated at reduced pressure and triturated with diethyl ether. The 7-(6-methoxypyridin-3-yl)-3-[(2-morpholin-4-ylethyl)amino]-1-(2-propoxyethyl)pyrido[3,4-b]pyrazin-2(1 H)-one was obtained as a pink powder. (47 mg, 47% yield).

'H NMR (CDCI3) b 8.78 (1 H), 8.74 (1 H), 8.23 (1 H), 7.66 (1 H), 6.92 (1 H), 6.86 (1 H), 4.46 (2H), 4.00 (3H), 3.80 (2H), 3.76-3.73 (4H), 3.69-3.63 (2H), 3.37 (2H), 2.68 (2H), 2.55-2.52 (4H), 1.53-1.46 (2H), 0.77 (3H); HRMS m/z469.2559 (calcd for M+H, 469.2558).
Example 2 0~1 O N N

N~N \ N
N
~H
/
7-(6-methoxypyridin-3-yl)-1-(2-propoxyethyl)-3-[(pyridin-2-ylmethyl)amino]pyrido[3,4-b]pyrazin-2(1 H)-one.
Prepared as described in example 1 using 2-(aminomethyl)pyridine in step 6.
' H NMR (CDCI3) b 8.83 (1 H), 8.76 (1 H), 8.62 (1 H), 8.25 (1 H), 7.73-7.67 (2H), 7.61-7.59 (1 H), 7.40 (1 H), 7.24-7.22 (1 H), 6.87 (1 H), 4.88 (2H), 4.49 (2H), 4.01 (3H), 3.82 (2H), 3.37 (2H), 1.53-1.46 (2H), 0.77 (3H); HRMS m/z447.2129 (calcd for M+H, 447.2134).
- -- ---Example 3- _ . _ .__. - ---- -_,_ .-- - -oJI"

I - oNI
xx ~
7-(6-methoxypyridin-3-y1)-1-(2-propoxyethyl)-3-[(tetrahydrofuran-2-ylmethyl)amino]pyrido[3,4-b]pyrazin-2(1 H)-one.
Prepared as described in example 1 using tetrahydrofurfurylamine in step 6.
'H NMR (CDCI3) b 8.77 (1H), 8.74 (1H), 8.23 (1H), 7.66 (1H), 6.85 (1H), 6.76-6.72 (1H), 4.45 (2H), 4.22-4.14 (1 H), 4.00 (3H), 3.97-3.90 (1 H), 3.84-3.76 (4H), 3.59-3.50 (1 H), 3.36 (2H), 2.10-2.02 (1 H), 1.99-1.89 (2H), 1.73-1.63 (1 H), 1.55-1.43 (2H), 0.77 (3H); HRMS
m/z 440.2272 (calcd for M+H, 440.2292).
Example 4 O1~, ~;c / i O\O N \ N

~N\ NN N
II H
ri N
1-(2-isopropoxyethyl)-7-(6-methoxypyridin-3-yl)-3-{[(5-methy/pyrazin-2-yl)methyl]amino)pyrido[3,4-b]pyrazin-2(1 H)-one.
Prepared as described in example 1 using 2-isopropoxyethylamine in step 3 and (5-m ethyl pyrazin-2-yl)m ethylam ine in step 6.
'H NMR (CDCI3) 8 8.80 (1 H), 8.75 (1 H), 8.60 (1 H), 8.44 (1 H), 8.25 (1 H), 7.71 (1 H), 7.33-7.29 (1 H), 6.86 (1 H), 4.88 (2H), 4.44 (2H), 4.00 (3H), 3.79 (2H), 3.56-3.50 (1 H), 2.57 (3H), 1.77 (3H);
HRMS m/z462.2253 (calcd for M+H, 462.2248).
Example 5 O~
0~1 O N N ---NN N

H
N
7-(6-methoxypyridin-3-yl)-3-{[(5-methylpyrazin-2-yl)methyl]amino)-1-(2-propoxyethyl)pyrido[3,4-b]pyrazin-2(1 H)-one.
Prepared as described in example 1 using (5-methylpyrazin-2-yl)methylamine in step 6.
'H NMR (CDCI3) b 8.79 (1 H), 8.74 (1 H), 8.60 (1 H), 8.43 (1 H), 8.22 (1 H), 7.67 (1 H), 7.33-7.29 (1 H), 6.84 (1 H), 4.88 (2H), 4.46 (2H), 3.99 (3H), 3.80 (2H), 3.36 (2H), 2.56 (3H), 1.52-1.46 (2H), 0.76 (3H); HRMS m/z462.2218 (calcd for M+H, 462.2248).
Example 6 O' \ I
I u O N N
N N \ N
OH

7 -(2-isopropoxyethyl)-7-(6-methoxypyridin-3-yl)-3-[(tetrahydro-2H-pyran-4-ylmethyl)amino]pyrido[3,4-b]pyrazin-2(1 H)-one.
Prepared as described in example 1 using 2-isopropoxyethylamine in step 3 and tetrahydro-2H-pyran-4-ylmethylamine in step 6.

' H NMR (CDCI3) S 8.80 (1 H), 8.76 (1 H), 8.26 (1 H), 7.70 (1 H), 6.87 (1 H), 6.53-6.49 (1 H), 4.44 (2H), 4.04-3.99 (5H), 3.80 (2H), 3.60-3.46 (3H), 3.42-3.38 (2H), 2.01-1.95 (1H), 1.76-1.72 (2H), 1.52-1.38 (2H), 1.09 (6H); HRMS m/z454.2451 (calcd for M+H, 454.2449).
Example 7 ), O

~ O"
O N ,' .N

HN1-N ~N
r:~
Oly N
tert-butyl 4-({[7-(6-meth oxypyridin-3-yl)-2-oxo-1-(2-propoxyethyl)-1,2-dihydropyrido[3, 4-b]pyrazin-3-yl]amino]methyl)piperidine-1-carboxylate.
Prepared as described in example 1 using tert-butyl 4-(aminomethyl)piperidine-l-carboxylate in step 6.
' H NMR (CDCI3) b 8.80 (1 H), 8.75 (1 H), 8.24 (1 H), 7.68 (1 H), 6.87 (1 H), 6.53-6.49 (1 H); 4.47 (2H), 4.16-4.12 (2H), 4.01 (3H), 3.81 (2H), 3.50 (2H), 3.38 (2H), 2.74-2.69 (2H), 1.90-1.78 (3H), 1.54-1.49 (2H), 1.47 (9H), 1.29-1.24 (2H), 1.09 (6H); HRMS m/z 553.3145 (calcd for M+H, 553.3133).
Example 8 o~

O\
I
ON N
y r N \N
HNI~H
7-(6-methoxypyridin-3-yl)-3-[(piperidin-4-ylmethyl)amino]-1-(2-propoxyethyl)pyrido(3,4-b]pyrazin-2(1 H)-one.
A solution of tert-butyl 4-({[7-(6-methoxypyridin-3-yl)-2-oxo-1-(2-propoxyethyl)-1,2-dihydropyrido[3,4-b]pyrazin-3-yl]amino}methyl)piperidine-1-carboxylate from example 10 (60 mg, 0.11 mmol) in 4N HCI in 1,4-dioxane (10 mL) was stirred at room temperature for two hours. The solvent was removed at reduced pressure and the residue was recrystallized from methanol/ethyl acetate to give 7-(6-methoxypyridin-3-yl)-3-[(piperidin-4-ylmethyl)amino]-1-(2-propoxyethyl)pyrido[3,4-b]pyrazin-2(1 H)-one as a white solid. (18 mg, 34%
yield). 'H NMR
(CD3OD) 5 8.73 (1 H), 8.72 (1 H), 8.24 (1 H), 8.22 (1 H), 7.13 (1 H), 4.68 (2H), 4.06 (3H), 3.85 (2H), 3.54 (2H), 3.46-3.36 (4H), 3.05-2.96 (2H), 2.18 (1H), 2.08-2.03 (2H),1.61-1.53~2H), 1.49-1.40 (2H), 0.72 (3H); HRMS m/z453.2638 (calcd for M+H, 453.2609).

Example 9 o 'f~ I
I
I ~ o O N N
N" N N
\ ~l H
p-N
7-(6-methoxypyridin-3-yl)-3-{((5-methylisoxazol-3-yl)methyl]amino]-1-(2-propoxyethyl)pyridol3,4-6]pyrazin-2(1 H)-one.
Prepared as described in example 1 using (5-methylisoxazol-3-yl)methylamine in step 6.
'H NMR (CDCI3) b 8.83 (1 H), 8.76 (1 H), 8.25 (1 H), 7.70 (1 H), 6.87-6.84 (1 H), 6.07 (1 H), 4.82 (2H), 4.46 (2H), 4.01 (3H), 3.81 (2H), 3.37 (2H), 2.42 (3H), 1.53-1.46 (2H), 0.78 (3H); HRMS m/z 451.2057 (calcd for M+H, 451.2088).
Example 10 I
o O N N

N~\H N N
NJ
3-{(3-(1 H-imidazol-1-yl)propyl]amin o]-7-(6-methoxypyridin-3-yl)-1-(2-propoxyethyl)pyrido(3, 4-b]pyrazin-2(1 H)-one.
Prepared as described in example 1 using 3-(1H-imidazol-1-yl)propylamine in step 6.
'H NMR (CDCI3) 8 8.80 (1 H), 8.75 (1 H), 8.25 (1 H), 7.68 (s, 1 H), 7.58 (1 H), 7.11(1 H), 7.01 (1 H), 6.87 (1 H), 6.50-6.46 (1 H), 4.47 (2H), 4.11(2H), 4.01 (3H), 3.81 (2H), 3.65-3.58 (2H), 3.38 (2H), 2.25-2.21 (2H), 1.53-1.46 (2H), 0.78 (3H); HRMS m/z464.2384 (calcd for M+H, 464.2405).
Example 11 o'r N' O~1 O Nr / I \ 1N
N
H
3-[(2-morpholin-4-ylethyl)amino]-1-(2-propoxyethyl)-7-pyrimidin-5-ylpyrido[3,4-b]pyrazin-2(1 H)-one.
Prepared as described in example 1 using pyrimidine-5-boronic acid in step 7.
' H NMR (CDCI3) b 9.29 (2H), 9.18 (1 H), 8.78 (1 H), 7.78 (1 H), 7.02-7.00 (1 H), 4.44 (2H), 3.79 (2H), 3.72-3.69 (4H), 3.66-3.60 (2H), 3.32 (2H), 2.64 (2H), 2.51-2.48 (4H), 1.47-1.40 (2H), 0.71 (3H); HRMS m/z440.2427 (calcd for M+H, 440.2405).

Example 12 of~
I r I
\ N
ON Xll~N /
r N N
OI~H
7-(6-methoxypyridin-3-y1)-1-(2-propoxyethyl)-3-((tetrahydro-2H-pyran-4-ylmethyl)amino]pyrido(3,4-b]pyrazin-2(1 H)-one.
Prepared as described in example 1 using tetrahydro-2H-pyran-4-ylmethylamine in step 6.
' H NMR (CDCI3) b 8.80 (1 H), 8.74 (1 H), 8.24 (1 H), 7.67 (1 H), 6.86 (1 H), 6.53-6.49 (1 H), 4.46 (2H), 4.04-4.00 (5H), 3.80 (2H), 3.52-3.45 (2H), 3.42-3.35 (4H), 2.00-1.94 (1 H), 1.76-1.72 (2H), 1.53-1.39 (4H), 0.78 (3H); HRMS m/z454.2401 (calcd for M+H, 454.2449).
Example 13 of~
I
f i I O~
O~ O IN I N
~1N N " _N N
Y"'-H
O
3-(2-morpholino-2-oxoethylamino)-7-(6-m ethoxypyridin-3-yl)-1-(2-propoxyethyl)pyrido[3, 4-b]pyrazin-2(1 H)-one Prepared as described in example 1 using 2-morpholin-4-yl-2-oxoethylamine in step 6.
'H NMR (CDCI3) b 8.77 (1 H), 8.75 (1 H), 8.22 (1 H), 7.68 (1 H), 7.38 (1 H), 6.85 (1 H), 4.46 (2H), 4.33 (2H), 4.00 (3H), 3.81-3.71 (8H), 3.58-3.56 (2H), 3.35 (2H), 1.50-1.45 (2H), 0.78 (3H);
HRMS m/z483.2348 (calcd for M+H, 483.2350).
Example 14 o-1-o O N I N

HN" 'N IN
O

7-(6-mefhoxypyridin-3-yl)-1-(2-propoxyethyl)-3-(((2S)-tetrahydrofuran-2-ylmethyl]amino)pyrido[3,4-b]pyrazin-2(1 H)-one Prepared as described in example 1 using (S)-(+)-tetrahydrofurfurylamine in step 6.
' H NMR (CDCI3) b 8.75 (1 H), 8.72 (1 H), 8.20 (1 H), 7.64 (1 H), 6.83 (1 H), 6.75-6.71 (1 H), 4.44 (2H), 4.18-4.15 (1 H), 3.98 (3H), 3.96-3.89 (1 H), 3.83-3.75 (4H), 3.58-3.50 (1 H), 3.35 (2H), 2.07-2.01 (1 H), 1.95-1.91 (2H), 1.69-1.62 (1 H), 1.51-1.44 (2H), 0.76 (3H); HRMS
m/z440.2299 (calcd for M+H, 440.2292).
Example 15 I "" 0~1 O N / \ N

O N~ N \ N
~H
7-(6-methoxypyridin-3-yl)-1-(2-propoxyethyl)-3-{((2R)-tetrahydrofuran-2-ylmethyl]amino)pyrido[3, 4-b]pyrazin-2(1 H)-one.
Prepared as described in example 1 using (R)-(-)-tetrahydrofurfurylamine in step 6.
' H NMR (CDCI3) b 8.78 (1 H), 8.74 (1 H), 8.24 (1 H), 7.67 (1 H), 6.86 (1 H), 6.76-6.72 (1 H), 4.46 (2H), 4.20-4.16 (1 H), 4.00 (3H), 3.98-3.90 (1 H), 3.87-3.77 (4H), 3.60-3.51 (1 H), 3.36 (2H), 2.11-2.03 (1 H), 2.00-1.90 (2H), 1.73-1.62 (1 H), 1.55-1.43 (2H), 0.77 (3H); HRMS
m/z 440.2306 (calcd for M+H, 440.2292).
Example 16 of'*' 0 ? ~ i O~1 N O N N
~N" 'N N
H
ethyl 4-{[7-(6-methoxypyridin-3-yl)-2-oxo-1-(2-propoxyethyl)-1,2-dihydropyrido[3,4-b]pyrazin-3-yl]amino}piperidine-l-carboxylate.
Prepared as described in example 1 using ethyl 4-am inopiperidine-1 -carboxylate in step 6.
'H NMR (CDCI3) b 8.79 (1 H), 8.74 (1 H), 8.24 (1 H), 7.68 (1 H), 6.87 (1 H), 6.35 (1 H), 4.46 2H), 4.20-4.12 (5H), 4.01 (3H), 3.80 (2H), 3.37 (2H), 3.11-3.07 (2H), 2.15-2.11 (2H), 1.53-1.46 (4H), 1.28 (3H), 0.78 (3H); HRMS m/z511.2686 (calcd for M+H, 511.2663).
Example 17 O~
O~1 I I
HNO N N
N" 'N \ N
H
7-(6-methoxypyridin-3-yl)-3-(piperidin-4-ylamino)-1-(2-propoxyethyl)pyrido[3,4-b]pyrazin-2(1 H)-one.
Prepared as described in example 1 using 4-aminopiperidine in step 6.
iH NMR (CDCI3) b 8.74 (2H), 8.22 (1H), 7.60 (1H), 6.84 (1H), 4.84-4.80 (2H), 4.40 (2H), 3.98 (3H), 3.77 (2H), 3.35 (2H), 3.13-3.04 (2H), 2.99 (1 H), 1.98-1.94 (2H), 1.54-1.45 (4H), 0.77 (3H);
HRMS m/z439.2455 (calcd for M+H, 439.2452).
Example 18 0 ~
~N O N N
~N" 'N \ N
H
3-((1-acetylpiperidin-4-yl)amino]-7-(6-methoxypyridin-3-yl)-1-(2-propoxyethyl)pyrido[3, 4-b]pyrazin-2(1 H)-one.
A solution of 7-(6-methoxypyridin-3-yl)-3-(piperidin-4-ylamino)-1-(2-propoxyethyl)pyrido[3,4-b]pyrazin-2(1 H)-one from example 28 (72 mg, 0.16 mmol), acetyl chloride (27 mg, 0.34 mmol) and triethylamine (0.05 mL, 0.36 mmol) in dichloromethane (2 mL) was stirred at room temperature for 15 hours. The reaction was partitioned between ethyl acetate and water. The layers were separated and the aqueous layer was extracted with ethyl acetate.
The combined organic layers were washed with brine, dried over magnesium sulfate, solvent was removed at reduced pressure to give and passed through a column of silica gel with 2.5%
methanol/
dichloromethane to give 3-[(1-acetylpiperidin-4-yl)amino]-7-(6-methoxypyridin-3-yl)-1-(2-propoxyethyl)pyrido[3,4-b]pyrazin-2(1H)-one as a yellow solid. (15 mg, 18%
yield).
'H NMR (CDCI3) b 8.77-8.76 (2H), 8.29 (1 H), 7.65 (1 H), 6.87 (1 H), 5.38-5.35 (1 H), 4.87-4.82 (2H), 4.43 (2H), 4.11-4.07 (1 H), 4.01 (3H), 3.79 (2H), 3.37 (2H), 3.18-3.10 (2H), 2.12-2.09 (2H), 2.04 (3H), 1.53-1.44 (4H), 0.78 (3H); HRMS m/z481.2538 (calcd for M+H, 481.2558).
Example 19 ~
O

O\
O N N

NXN N
H
7-(6-methoxypyridin-3-yl)-3-[(2-piperidin-1-ylethyl)amino]-1-(2-propoxyethyl)pyrido[3,4-b]pyrazin-2(1 H)-one.
Prepared as described in example 1 using 2-piperidin-1-ylethylamine in step 6.
iH NMR (CDCI3) b 8.77 (1 H), 8.73 (1 H), 8.22 (1 H), 7.64 (1 H), 7.00-6.97 (1 H), 6.84 (1 H), 4.44 (2H), 3.99 (3H), 3.79 (2H), 3.64-3.59 (2H), 3.36 (2H), 2.61 (2H), 2.44 (4H), 1.62-1.56 (4H), 1.52-1.42 (4H), 0.76 (3H); HRMS m/z467.2762 (calcd for M+H, 467.2765).
Example 20 0f \ O\
I N
O N /N
NHN \N

7-(6-methoxypyridin-3-y1)-1-(2-prc; yoxyethyl)-3-[(2-pyridin-2-ylethyl)amino]pyrido[3,4-b]pyrazin-2(1H)-one.
Prepared as described in example 1 using 2-(2-aminoethyl)pyridine in step 6.
'H NMR (CDCI3) b 8.80 (s, 1 H), 8.74 (1 H), 8.62-8.60 (1 H), 8.26 (1 H), 7.69-7.63 (2H), 7.24-7.15 (3H), 6.86 (1 H), 4.45 (2H), 4.04-3.98 (5H), 3.79 (2H), 3.36 (2H), 3.22 (2H), 1.52-1.45 (2H), 0.77 (3H); HRMS m/z461.2346 (calcd for M+H, 461.2296).
Example 21 o~~
OMe N~ O N N

\~lN' N1 N N

3-(2-(4-ethylpiperazin-1-yl)-2-oxoethylamino)-7-(6-methoxypyridin-3-yl)-1-(2-propoxyethyl)pyrido[3,4-b]pyrazin-2(1 H)-one Prepared as described in example 1 using 2-amino-l-(4-ethylpiperazin-1-yl)ethanone in step 6. The Suzuki reaction of step 7 in example 1 was performed using the following modified procedure. A solution of 3-(2-(4-ethylpiperazin-1-yl)-2-oxoethylamino)-7-chloro-l-(2-propoxyethyl)pyrido[3,4-b]pyrazin-2(1H)-one (306 mg, 0.70 mmol) in 1,4-dioxane (5.0 mL) was treated with dichloro[1,1'-bis(diphenylphosphino)ferrocene]palladium (II) dichloromethane adduct (54 mg, 0.066 mmol), 6-methoxypyridin-3-yl-3-boronic acid (250 mg, 1.6 mmol) and 2.5 mL of water containing Na2CO3 (264 mg, 2.5 mmol). The mixture was refluxed for 4.0 hours. The mixture was partitioned between ethyl acetate and water, and the layers were separated. The aqueous layer was extracted with ethyl acetate. The combined organic layer were dried over sodium sulfate, concentrated under reduced pressure, and purified by column chromatography.
3-(2-(4-ethylpiperazin-1 -yl)-2-oxoethylamino)-7-(6-methoxypyridin-3-yl)-1 -(2-propoxyethyl)pyrido[3,4-b]pyrazin-2(1 H)-one was obtained. (177 mg, 48%
yield).Prepared as described in example 1 using in step 6 and the Suzuki conditions of example 47 step 7.

1H NMR (CDCI3) 58.79 (1 H), 8.75 (1 H), 8.24 (1 H), 7.68 (1 H), 7.42 (1 H), 6.84 (1 H), 4.47 (t, 2H, J
= 5.3 Hz), 4.33 (2H), 4.00 (3H), 3.80 (2H), 3.75 (2H), 3.58 (2H), 3.36 (2H), 2.54 - 2.43 (6H), 1.52 -1.45 (2H), 1.47 (3H), 0.77 (3H); HRMS m/z 510.2837 (calcd for M+H, 510.2823).
Example 22 O OMe N~ O N N 11 ~IN N11N N
~H
ethyl4-(2-(1,2-dihydro-7-(6-methoxypyridin-3-yl)-2-oxo-1-(2-propoxyethyl)pyrido[3,4-b]pyrazin-3-y/amino)acetyl)piperazine-1-carboxylate Prepared as described in example 1 using ethyl 4-(2-aminoacetyl)piperazine-l-carboxylate in step 6 and the Suzuki conditions of example 21 step 7.

yH NMR (CDCI3) 08.78 (1 H), 8.75 (1 H), 8.24 (1 H), 7.68 (1 H), 7.37 (1 H), 6.84 (1 H), 4.47 (2H), 4.35 (2H), 4.22 - 4.15 (2H), 4.00 (3H), 3.80 (2H), 3.72 - 3.68 (2H), 3.58 -3.51 (7H), 3.36 (2H), 1.52 - 1.45 (2H), 1.31 - 1.25 (5H), 0.77 (3H); HRMS m/z 554.2738 (calcd for M+H, 554.2722).
Example 23 OMe r I
O N N
ON N~N N
~H

3-(2-oxo-2-(pyrrolidin-1-yl)ethylamino)-7-(6-methoxypyridin-3-yl)-1-(2-propoxyethyl)pyrido[3, 4-b]pyrazin-2(1 H)-one Prepared as described in example 1 using 2-amino-l-(pyrrolidin-1-yl)ethanone in step 6 and the Suzuki conditions of example 21 step 7.

iH NMR (CDCI3) 08.78 (1 H), 8.75 (1 H), 8.24 (1 H), 7.68 (1 H), 7.37 (1 H), 6.84 (1 H), 4.46 (2H), 4.25 (2H), 4.00 (3H), 3.80 (2H), 3.60 - 3.53 (4H), 3.36 (2H), 2.08 - 2.01 (2H), 1.97 - 1.90 (2H), 1.52 -1.45 (2H), 0.77 (3H); HRMS m/z467.2394 (calcd for M+H, 467.2401).
Example 24 o~~
O
O~ O N ~ I o N
'~ NJ=N N

3-(2-morpholino-2-oxoethylamino)-7-(3,5-dimethylisoxazol-4-yl)-1-(2-propoxyethyl)pyrido[3,4-b]pyrazin-2(1H)-one Prepared as described in example 1 using 2-amino-l-morpholinoethanone in step 6 and and the Suzuki conditions of example 21 step 7 with 3,5-dimethylisoxazol-4-yl-4-boronic acid.
'H NMR (CDCI3) 08.78 (1 H), 7.41 (1 H), 7.34 (1 H), 4.41 (2H), 4.33 (2H), 3.99 - 3.97 (1 H), 3.81 -3.73 (10H), 3.59 - 3.56 (2H), 3.43 (1 H), 3.34 (2H), 2.59 (3H), 2.44 (3H), 0.75 (3H); HRMS m/z 471.2374 (calcd for M+H, 471.2350).
Example 25 J / OMe ( I
O N N
HN N N
~/ ~H N

3-(2-oxo-2-(piperazin-1-yl)ethylamino)-7-(6-methoxypyridin-3-yl)-1-(2-propoxyethyl)pyrido[3,4-b]pyrazin-2(1 H)-one Prepared as described in example 1 using 2-amino-l-(piperazin-1-yl)ethanone in step 6,and the Suzuki conditions of example 21 step 7.
' H NMR (CDCI3) 08.78 - 8.77 (2H), 8.28 - 8.26 (1 H), 7.67 (1 H), 6.85 (1 H), 4.44 (2H), 4.01 - 3.99 (6H), 3.81 - 3.78 (4H), 3.54 (2H), 3.37 (2H), 1.70 (4H), 1.53 - 1.46 (2H), 1.27 - 1.25 (2H), 0.76 (3H); HRMS m/z 482.2541 (calcd for M+H, 482.2510).
Example 26 OMe r I
O N ~ ~ N
Me ~ ~N
-~'H N
O
3-(2-(4-methylpiperazin-1-yl)-2-oxoethylamino)-7-(6-methoxypyridin-3-yl)-1-(2-propoxyethyl)pyrido[3,4-b]pyrazin-2(1 H)-one Prepared as described in example 1 using 2-amino-l-(4-methylpiperazin-1-yl)ethanone in step 6 and the Suzuki conditions of example 21 step 7.
'H NMR (CDCI3) 08.78 (1 H), 8.75 (1 H), 8.26 - 8.22 (1 H), 7.68 (1 H), 7.43 -7.40 (1 H), 6.85 (1 H), 4.47 (2H), 4.33 (2H), 4.00 (3H), 3.80 (2H), 3.73 - 3.71 (2H), 3.58 (2H), 3.36 (2H), 2.51 - 2.43 (4H), 2.34 (3H), 1.52 - 1.45 (2H), 0.77 (3H); HRMS m/z496.2632 (calcd for M+H, 496.2667).
Example 27 O~/
OMe ( I
OI N N
HO~
N ~H NN
O
3-(2-(4-hydroxypiperidin-1-yl)-2-oxoethylamino)-7-(6-methoxypyridin-3-yl)-1-(2-propoxyethyl)pyrido[3,4-b]pyrazin-2(1 H)-one Prepared as described in example 1 using 2-amino-l-(4-hydroxypiperidin-1-yl)ethanone in step 6 and the Suzuki conditions of example 21 step 7.
iH NMR (CDCI3) b8.79 (1 H), 8.75 (1 H), 8.26 - 8.22 (1 H), 7.69 (1 H), 7.44 (1 H), 6.84 (1 H), 4.47 (2H), 4.33 (2H), 4.09 - 4.05 (1 H), 4.00 (3H), 3.82 - 3.78 (2H), 3.43 - 3.31 (3H), 3.17 -1.96 (1 H), 1.67 (6H), 1.55 - 1.42 (2H), 0.77 (3H); HRMS m/z 497.2543 (calcd for M+H, 497.2507).
Example 28 o~~
OMe r I
ON N
C N' ~H ~N 1 N
o 3-(2-oxo-2-(piperidin-1-yl)ethylamino)-7-(6-methoxypyridin-3-yl)-1-(2-propoxyethyl)pyrido[3,4-b]pyrazin-2(1 H)-one Prepared as described in example 1 using 2-amino-l-(piperidin-1-yi)P.thanone in step 6 and the Suzuki conditions of example 21 step 7.
' H NMR (CDCI3) 08.79 (1 H), 8.75 (1 H), 8.26 - 8.22 (1 H), 7.68 (1 H), 7.48 (1 H), 6.85 (1 H), 4.46 (2H), 4.31 (2H), 4.00 (3H), 3.80 (2H), 3.66 - 3.63 (2H), 3.50 - 3.46 (2H), 3.38 - 3.34 (2H), 1.68 -1.43 (8H), 0.77 (3H); HRMS m/z481.2574 (calcd for M+H, 481.2558).
Example 29 o/\
N O~
O Nf \ \ ~

~, N'-/-'N1 N I N
H
3-(2-morpholinoethylamino)-1-(2-isopropoxyethyl)-7-(6-methoxypyridin-3-yl)pyrido[3,4-b]pyrazin-2(1 H)-one Prepared as described in example 1 using 2-isopropoxyethanamine in step 3.
iH NMR (CDCI3) b8.78 (1 H), 8.75 (1 H), 8.27 - 8.24 (1 H), 7.69 (1 H), 6.93 (1 H), 6.84 (1 H), 4.43 (2H), 3.99 (3H), 3.80 - 3.67 (8H), 3.57 - 3.51 (1 H), 2.70 (2H), 2.62 (4H), 1.06 (6H); HRMS m/z 469.2524 (calcd for M+H, 469.2558).
Example 30 O-t--I
N O~
O Nr \ \ I
N N (NN
l- H
3-((pyridin-2-y1)methylamino)-1-(2-isopropoxyethyl)-7-(6-methoxypyridin-3-yl)pyrido[3,4-b]pyrazin-2(1 H)-one Prepared as described in example 1 using 2-isopropoxyethanamine in step 3 and (pyridin-2-yl)methanamine in step 6.
iH NMR (CDCI3) b8.82 (1 H), 8.76 (1 H), 8.62 - 8.61 (1 H), 8.30 - 8.27 (1 H), 7.72 - 7.67 (2H), 7.60 - 7.58 (1 H), 7.38 (1 H), 7.26 - 7.22 (1 H), 6.85 (1 H), 4.87 (2H), 4.45 (2H), 4.00 (3H), 3.80 (2H), 3.57 - 3.51 (1 H), 1.05 (6H); HRMS m/z 447.2120 (calcd for M+H, 447.2139).
Example 31 o' \
N O~
O IN I
0~,. O.
N N
H
3-(((S)-tetrahydrofuran-2-yl)methylamino)-1-(2-isopropoxyethyl)-7-(6-methoxypyridin-3-yl)pyrido[3,4-b]p_~, razin-2(1 H)-one Prepared as described in example 1 using 2-isopropoxyethanamine in step 3 and ((S)-tetrahydrofuran-2-yl)methanamine in step 6 and the Suzuki conditions of example 21 step 7.
iH NMR (CDCI3) b8.84 (1 H), 8.76 (1 H), 8.26 - 8.22 (1 H), 7.69 (1 H), 6.84 (1 H), 6.74 (1 H), 4.42 (2H), 4.28 - 4.16 (1 H), 4.00 (3H), 3.97 - 3.90 (1 H), 3.85 - 3.72 (4H), 3.59 -3.50 (2H), 2.10 -1.94 (3H), 1.72 - 1.63 (1 H), 1.06 (6H); HRMS m/z 440.2270 (calcd for M+H, 440.2292).
Example 32 r O~
~ U
0 Nf N N (NN
H
1-(3-ethoxypropyl)-7-(6-methoxypyridin-3-yl)-3-[(pyridin-2-ylmethyl)amino]pyrido[3, 4-b]pyrazin-2(1 H)-one Prepared as described in example 1 using 3-ethoxypropan-l-amine in step 3 and (pyridin-2-yl)methanamine in step 6 and the Suzuki conditions of example 21 step 7.
' H NMR (CDCI3) 08.82 (1 H), 8.79 (1 H), 8.62 - 8.61 (1 H), 8.27 - 8.25 (1 H), 7.70 - 7.56 (3 H), 7.37 (1 H), 7.24 - 7.21 (1 H), 6.84 - 6.82 (1 H), 4.87 (2H), 4.40 (2H), 3.99 (3H), 3:52 - 3.45 (4H), 2.09 - 2.04 (2H), 1.24 - 1.19 (3H); HRMS m/z 447.2139 (calcd for M+H, 447.2139).

Example 33 r O
N O~
O N \ \ I

JI NH
1-(3-ethoxypropyl)-7-(6-methoxypyridin-3-yl)-3-{[(5-methylpyrazin-2-yl)methyl]amino)pyrido[3,4-b]pyrazin-2(1 H)-one Prepared as described in example 1 using 3-ethoxypropan-l-amine in step 3 and (5-methylpyrazin-2-yl)methanamine in step 6 and the Suzuki conditions of example 21 step 7.
' H NMR (CDCI3) b8.81 (1 H), 8.78 (1 H), 8.59 (1 H), 8.43 (1 H), 8.26 - 8.23 (1 H), 7.63 (1 H), 7.30 (1 H), 6.82 (1 H), 4.86 (2H), 4.39 (2H), 3.99 (3H), 3.52 - 3.45 (4H), 2.56 (3H), 2.16 - 2.02 (2H), 1.24 - 1.19 (3H); HRMS m/z 462.2270 (calcd for M+H, 462.2248).
Example 34 r N o~
O N \ \ I
0,. N N I N
H

1-(3-ethoxypropyl)-7-(6-methoxypyridin-3-yl)-3-{[(2S)-tetrahydrofuran-2-ylmethyl]amino)pyrido[3, 4-b]pyrazin-2(1 H)-one Prepared as described in example 1 using 3-ethoxypropan-l -am ine in step 3 and ((S)-tetrahydrofuran-2-yl)methanamine in step 6 and the Suzuki conditions of example 21 step 7.
'H NMR (CDCI3) 08.77 - 8.76 (2H), 8.25 - 8.22 (1 H), 7.61 (1 H), 6.82 (1 H), 6.76 - 6.74 (1 H), 4.37 (2H), 4.18 - 4.15 (1 H), 3.98 (3H), 3.95 - 3.89 (1 H), 3.84 - 3.76 (2H), 3.56 -3.44 (4H), 2.08 -2.02 (3H), 1.97 -1.91 (2H), 1.68 -1.65 (1 H), 1.23 -1.19 (4H); HRMS m/z 440.2282 (calcd for M+H, 440.2292).
Example 35 I
O
OMe O-') O IN N
~,N NX N ~N
~H
3-(2-morpholino-2-oxoethylamino)-1-(3-ethoxypropyl)-7-(6-methoxypyridin-3-yl)pyrido[3, 4-b]pyrazin-2(1 H)-one Prepared as described in example 1 using 2-amino-l-morpholinoethanone in step 3 and 3-methoxypropan-l-amine in step 6 and the Suzuki conditions of example 21 step 7.

iH NMR (CDCI3) 08.78 - 8.77 (2H), 8.23 (1 H), 7.62 (1 H), 7.39 (1 H), 6.82 (1 H), 4.38 (2H), 4.31 (2H), 3.98 (3H), 3.84 - 3.71 (6H), 3.57 - 3.55 (2H), 3.51 - 3.44 (4H), 2.16 -2.01 (2H), 1.21 (3H);
HRMS m/z483.2376 (calcd for M+H, 483.2350).
Example 36 Oi N
O
O ON
N~~N ~N I ~ N
H
7-(3,5-dimethylisoxazol-4-y1)-1-(2-ethoxyethyl)-3-[(2-morpholin-4-ylethyl)amino]pyrido[3,4-b]pyrazin-2(1 H)-one Prepared as described in example 1 using 2-ethoxyethanamine in step 3 and and the Suzuki conditions of example 21 step 7 with 3,5-dimethylisoxazol-4-yl-4-boronic acid.
' H NMR (CDCI3) 08.79 (1 H), 7.31 (1 H), 6.96 (1 H), 4.40 (2H), 3.81 - 3.68 (8H), 3.50 - 3.43 (3H), 2.64 (2H), 2.59 - 2.57 (6H), 2.44 (3H), 1.09 (3H); HRMS m/z443.2418 (calcd for M+H, 443.2401).
Example 37 OJ
N O1~
O N( 0. NN N
H
3-(((S)-tetrahydrofuran-2-yl)methylamino)- 1-(2-ethoxyethyl)-7-(6-methoxypyridin-3-yl)pyrido[3,4-b]pyrazin-2(1H)-one Prepared as described in example 1 using 2-ethoxyethanamine in step 3 and ((S)-tetrahydrofuran-2-yi)methanamine in step 6 and the Suzuki conditions of example 21 step 7.
iH NMR (CDCI3) b8.77 (1 H), 8.73 (1 H), 8.26 - 8.22 (1 H), 7.66 (1 H), 7.15 (1 H), 6.84 (1 H), 6.75 (1 H), 4.45 (2H), 4.21 - 4.14 (1 H), 3.99 (3H), 3.97 - 3.90 (1 H), 3.86 - 3.74 (4H), 3.59 - 3.39 (2H), 2.11 -1.89 (3H), 1.72 -1.64 (1 H), 1.14 (3H); HRMS m/z 426.2121 (calcd for M+H, 426.2136).
Example 38 OJ
N O~
O Nr ~

N'~-NXN N
H
3-(2-(pyrrolidin-1-yl)ethylamino)-1-(2-ethoxyethyl)-7-(6-methoxypyridin-3-yl)pyrido[3,4-b]pyrazin-2(1 H)-one Prepared as described in example 1 using 2-ethoxyethanamine in step 3 and 2-(pyrrolidin-l-yl)ethanamine in step 6 and the Suzuki conditions of example 21 step 7.

' H NMR (CDCI3) 08.78 (1 H), 8.74 (1 H), 8.25 (1 H), 7.66 (1 H), 7.00 (1 H), 6.84 (1 H), 4.45 (2H), 4.00 (3H), 3.82 - 3.76 (4H), 3.51 - 3.44 (2H), 2.93 (2H), 2.76 (4H), 1.89 (4H), 1.08 (3H); HRMS
m/z 439.2424 (calcd for M+H, 439.2452).
Example 39 OJ

0 Nf ON"--- N11 N N
H
3-(2-(piperidin- y-yl)ethylamino)-1-(2-ethoxyethyl)-7-(6-methoxypyridin-3-yl)pyrido[3,4-b]pyrazin-2(1 H)-one Prepared as described in example 1 using 2-ethoxyethanamine in step 3 and 2-(piperidin-l-yl)ethanamine in step 6 and the Suzuki conditions of example 21 step 7.
'H NMR (CDCI3) 08.78 (1 H), 8.75 (1 H), 8.25 (1 H), 7.66 (1 H), 7.26 (1 H), 6.85 (1 H), 4.45 (2H), 4.00 (3H), 3.80 (2H), 3.66 (2H), 3.48 (2H), 2.68 (2H), 2.51 (4H), 1.65 (4H), 1.48 (2H), 1.12 (3H);
HRMS m/z453.2642 (calcd for M+H, 453.2609).
Example 40 O~
-i O~1 O N
~N'/~N~N N
H
3-(2-morpholinoethylamino)-1-(2-ethoxyethyl)-7-(pyridin-3-yl)pyrido[3,4-b]pyrazin-2(1H)-one Prepared as described in example 1 using 2-ethoxyethanamine in step 3 and the Suzuki conditions of example 21 step 7 with pyridin-3-yl-3-boronic acid.
'H NMR (CDCI3) 09.20 - 9.19 (1 H), 8.83 (1 H), 8.64 - 8.62 (1 H), 8.37 - 8.33 (1 H), 7.79 (1 H), 7.43 - 7.38 (1 H), 7.00 (1 H), 4.49 (2H), 3.84 - 3.66 (8H), 3.48 (2H), 2.71 (2H), 2.56 (4H), 1.12 (3H);
HRMS m/z425.2303 (calcd for M+H, 425.2296).
Example 41 OJ
OMe r I
O~\ O N N
N N~N iN
~H

3-(2-morpholino-2-oxoethylamino)- i -(2-ethoxyethyl)-7-(6-methoxypyridin-3-yl)pyrido[3, 4-b]pyrazin-2(1 H)-one Prepared as described in example 1 using 2-ethoxyethanamine in step 3 and 2-amino-l-morpholinoethanone in step 6 and the Suzuki conditions of example 21 step 7.

' H NMR (CDCI3) 08.78 - 8.74 (2H), 8.28 - 8.25 (1 H), 7.69 (1 H), 7.40 (1 H), 6.85 (1 H), 4.46 (2H), 4.32 (2H), 4.00 (3H), 3.82 - 3.72 (8H), 3.58 (2H), 3.47 (2H), 1.11 (3H); HRMS
m/z469.2183 (calcd for M+H, 469.2194).
Example 42 oJ
OMe f I
O N N
N~ N~N N

J NH
3-((5-methylpyrazin-2-yl)methylamino)- 1-(2-ethoxyethyl)-7-(6-methoxypyridin-3-yl)pyrido[3,4-b]pyrazin-2(1H)-one Prepared as described in example 1 using 2-ethoxyethanamine in step 3 and (5-methylpyrazin-2-yl)methanamine in step 6 and the Suzuki conditions of example 21 step 7.
'H NMR (CDCI3) 08.82 (1 H), 8.75 - 8.74 (1 H), 8.60 (1 H), 8.44 (1 H), 8.30 -8.27 (1 H), 7.70 (1 H), 7.32 (1 H), 6.86 (1 H), 4.88 (2H), 4.47 (2H), 4.00 (3H), 3.81 (2H), 3.48 (2H), 2.58 (3H), 1.11 (3H);
HRMS m/z448.2050 (calcd for M+H, 448.2092).
Example 43 oJ
OMe f I ,N
N'~-, H N
/
3-((pyridin-2-yl)methylamino)-1-(2-ethoxyethyl)-7-(6-methoxypyridin-3-yl)pyrido[3,4-b]pyrazin-2(1 H)-one Prepared as described in example 1 using 2-ethoxyethanamine in step 3 and (pyridin-2-yl)methanamine in step 6 and the Suzuki conditions of example 21 step 7.
'H NMR (CDCI3) 08.83 (1 H), 8.75 (1 H), 8.61 (1 H), 8.29 (1 H), 7.74 - 7.68 {2H), 7.61 (1 H), 7.39 (1 H), 7.25 - 7.22 (1 H), 6.86 (1 H), 4.88 (2H), 4.48 (2H), 4.00 (3H), 3.82 (2H), 3.48 (2H), 1.12 (3H); HRMS m/z433.2018 (calcd for M+H, 433.1983).
Example 44 OJ
OMe f O O N N
N~'N~N I N
H
3-(2-morpholinoethylamino)-1-(2-ethoxyethyl)-7-(6-m ethoxypyridin-3-yl)pyrido[3, 4-b]pyrazin-2(1H)-one Prepared as described in example 1 using 2-ethoxyethanamine in step 3 and the Suzuki conditions of example 21 step 7.
' H NMR (CDCI3) 08.78 (1 H), 8.74 (1 H), 8.26 (1 H), 7.67 (1 H), 6.97 (1 H), 6.85 (1 H), 4.46 (2H), 4.00 (3H), 3.82 - 3.72 (8H), 3.48 (2H), 2.75 (2H), 2.60 (4H), 1.12 (3H); HRMS
m/z455.2378 (calcd for M+H, 455.2401).
Example 45 JI", ~ / F
o:xxr 3-(2-morpholinoethylamino)-7-(4-fluorophenyl)- 1-(2-propoxyethyl)pyrido[3,4-b]pyrazin-2(1 H)-one Prepared as described in example 1 and the Suzuki conditions of example 21 step 7 with 4-fluorophenylboronic acid.

1 H NMR (CDCI3) b 8.78 (1 H), 7.99-7.94 (2H), 7.68 (1 H), 7.17-7.11 (2H), 6.93-6.90 (1 H), 4.46 (2H), 3.80 (2H), 3.76-3.72 (4H), 3.68-3.62 (2H), 3.37 (2H), 2.67 (2H), 2.54-2.51 (4H), 1.524.45 (2H), 0.78 (3H); HRMS m/z456.2407 (calcd for M+H, 456.2405).
Example 46 Jr o 0 N O >
o I

N'-~NN \ N
H
7-(1, 3-benzodioxol-5-yl)-3-[(2-morpholin-4-ylethyl)amino]-1-(2-propoxyethyl)pyrido[3, 4-b]pyrazin-2(1 H)-one.
Prepared as described in example 1 and the Suzuki conditions of example 21 step 7using 3,4-(methylenedioxy)phenylboronic acid.
'H NMR (CDCI3) b 8.75 (1H), 7.62 (1H), 7.51-7.47 (2H), 6.90-6.87 (2H), 6.01 (2H), 4.46 (2H), 3.80 (2H), 3.76-3.72 (4H), 3.68-3.62 (2H), 3.37 (2H), 2.67 (2H), 2.54-2.51 (4H), 1.54-1.47 (2H), 0.79 (3H); HRMS m/z482.2354 (calcd for M+H, 482.2398).
Example 47 oJ

ON, oXcc H

3-(2-morpholinoethylamino)-1-(2-ethoxyethyl)-7-(4-methoxyphenyl)pyrido[3,4-b]pyrazin-2(1 H)-one Prepared as described in example 1 using 2-ethoxyethanamine in step 3 and the Suzuki conditions of example 21 step 7 with 4-methoxyphenylboronic acid.
'H NMR (CDCI3) b8.78 (1 H), 7.95 (2H), 7.66 (1 H), 7.01 (2H), 6.91 (1 H), 4.47 (2H), 3.87 (3H), 3.83 - 3.67 (8H), 3.49 (2H), 2.71 (2H), 2.57 (4H), 1.13 (3H); HRMS m/z 454.2425 (calcd for M+H, 454.2449).
Example 48 OJ
?;C~ O
O , NN1 N N
H
7-(2,3-dihydro-1-benzofuran-5-yl)-1-(2-ethoxyethyl)-3-[(2-morpholin-4-ylethyl)amino]pyrido(3,4-b]pyrazin-2(1 H)-one Prepared as described in example 1 using 2-ethoxyethanamine in step 3 and the Suzuki conditions of example 21 step 7 with 2,3-dihydrobenzofuran-5-yl-5-boronic acid.
'H NMR (CDCI3) b 8.77 (1 H), 7.91 (1 H), 7.75 - 7.71 (1 H), 7.63 (1 H), 6.89 -6.85 (2H), 4.64 (2H), 4.47 (2H), 3.83 - 3.70 (6 H), 3.68 - 3.64 (2H), 3.51 (2H), 3.28 (2H), 2.72 -2.70 (2H), 2.57 - 2.53 (4H), 1.14 (3H); HRMS m/z466.2420 (calcd for M+H, 466.2449).
Example 49 oJ

? CF3 O~1 O N ' I
NNIN I N
H
1-(2-ethoxyethyl)-3-[(2-morpholin-4-ylethyl)amino]-7-[4-(trifluoromethyl)phenyl]pyrido[3, 4-b]pyrazin-2(1 H)-one Prepared as described in example 1 using 2-ethoxyethanamine in step 3 and the Suzuki conditions of example 21 step 7 with 4-(trifluoromethyl)phenylboronic acid.
1 H NMR (CDCI3) 08.83 (1 H), 8.13 (2H), 7.82 (1 H), 7.73 (2H), 7.01 (1 H), 4.49 (2H), 3.85 - 3.70 (8H), 3.46 (2H), 2.73 (2H), 2.58 (4H), 1.12 (3H); HRMS m/z492.2181 (calcd for M+H, 492.2217).
Example 50 O~I O N 11 , N---~-NX, N . ~ N
H

1-(2-ethoxyethyl)-3-[(2-morpholin-4-ylethyl)amino]-7-(3-(trifluoromethyl)phenyl]pyrido[3, 4-b]pyrazin-2(1 H)-one Prepared as described in example 1 using 2-ethoxyethanamine in step 3 and the Suzuki conditions of example 21 step 7 with 3-(trifluoromethyl)phenylboronic acid.
' H NMR (CDCI3) 08.82 (s), 8.28 (s), 8.21 (1 H), 7.82 (1 H), 7.66 - 7.59 (2H), 7.0 (1 H), 4.49 (2H), 3.85 - 3.69 (8H), 3.51 (2H), 2.72 (2H), 2.57 (4H), 1.13 (3H); HRMS m/z 492.2194 (calcd for M+H, 492.2217).
Example 51 oJ
ON O N
~~ N~N N
H
3-(2-morpholinoethylamino)-1-(2-ethoxyethyl)-7-phenylpyrido[3,4-b]pyrazin-2(1H)-one Prepared as described in example 1 using 2-ethoxyethanamine in step 3 and the Suzuki conditions of example 21 step 7 with phenylboronic acid.
' H NMR (CDCI3) 08.82 (1 H), 8.00 (2H), 7.74 (1 H), 7.50 - 7.37 (3H), 6.94 (1 H), 4.48 (2H), 3.83 -3.68 (8H), 3.49 (2H), 2.71 (2H), 2.56 (4H), 1.14 (3H); HRMS m/z 424.2306 (calcd for M+H, 424.2343).
Example 52 oJ
J / F
f I
co H
3-(2-morpholinoethylamino)-1-(2-ethoxyethyl)-7-(4-fluorophenyl)pyrido[3, 4-b]pyrazin-2(1 H)-one Prepared as described in example 1 using 2-ethoxyethanamine in step 3 and the Suzuki conditions of example 21 step 7 with 4-fluorophenylboronic acid.
1H NMR (CDCI3) b8.79 (1 H), 8.01 - 7.96 (2H), 7.69 (1 H), 7.15 (2H), 6.95 (1 H), 4.47 (2H), 3.83 -3.68 (8H), 3.48 (2H), 2.27 (2H), 2.57 (4H), 1.13 (3H); HRMS m/z442.2222 (calcd for M+H, 442.2249).
Example 53 oJ
O N F

0 ~'N~N N
H
3-(2-morpholinoethylamino)-1-(2-ethoxyethyl)-7-(3-fluorophenyl)pyrido[3, 4-b]pyrazin-2(1 H)-one Prepared as described in example 1 using 2-ethoxyethanamine in step 3 and the Suzuki conditions of example 21 step 7 with 3-fluorophenylboronic acid.
'H NMR (CDCI3) 08.80 (1 H), 7.79 - 7.71 (3H), 7.47 - 7.39 (1 H), 7.08 - 7.05 (1 H), 6.97 (1 H), 4.48 (2H), 3.84 - 3.65 (8H), 3.49 (2H), 2.72 (2H), 2.56 (4H), 1.13 (3H); HRMS m/z 442.2262 (calcd for M+H, 442.2249).
Example 54 oJ
F
O O Nf I F
NNX N N
H
3-(2-morpholinoethylamino)-1-(2-ethoxyethyl)-7-(3,4-difluorophenyl)pyrido[3,4-b]pyrazin-2(1 H)-one Prepared as described in example 1 using 2-ethoxyethanamine in step 3 and the Suzuki conditions of example 21 step 7 with 3,4-difluorophenylboronic acid.
' H NMR (CDCI3) ~8.78 (1 H), 7.90 - 7.84 (1 H), 7.75 - 7.71 (2H), 7.23 - 7.20 (1 H), 7.00 (1 H), 4.48 (2H), 3.84 - 3.79 (8H), 3.48 (2H), 2.74 (2H), 2.60 (4H), 1.13 (3H); HRMS m/z 460.2122 (calcd for _--M+H; 460:2155).-----Example 55 o.,/
OMe o;I::Jcc*N
3-(2-morpholinoethylam ino)-1-(3-ethoxypropyl)-7-(6-methoxypyridin-3-yl)pyrido(3, 4-b]pyrazin-2(1 H)-one Example 56 OMe O Nf N

X, N N
H
O
1-(3-(1,2-dihydro-7-(6-methoxypyridin-3-yl)-2-oxo-1-(2-propoxyethyl)pyrido(3,4-b]pyrazin-3-ylamino)propyl)pyrrolidine-2, 5-dione Example 57 HO OMe O N N

tN--"~,-N X N N
H
3-(2-(3-hydroxypyrrolidin-1-yl)ethylam6io)-7-(6-methoxypyridin-3-yl)-1-(2-propoxyethyl)pyrido[3, 4-b]pyrazin-2(1 H)-one Example 58 OMe HO O fN \ \ N
N,_~ N11 N I ~ N
H
3-(2-(4-hydroxypiperidin-1-yl)ethy/amino)-7-(6-methoxypyridin-3-yl)-1-(2-propoxyethyl)pyrido[3,4-b]pyrazin-2(1 H)-one Example 59 - -- --- ---- - -- - -- - - -- _ _-- -- ~/- - -- -------O OMe H2N I O Nf N
N~~N I N N
H
1-(2-(1,2-dihydro-7-(6-methoxypyridin-3-yl)-2-oxo-1-(2-propoxyethyl)pyrido(3, 4-bJpyrazin-3-ylamino)ethyl)piperidine-4-carboxamide Example 60 o--~
O NH2 ? OMe I
O N
N-,---N cx:xr H

(2-(1,2-dihydro-7-(6-methoxypyridin-3-yl)-2-oxo-1-(2-propoxyethyl)pyrido[3,4-b]pyrazin-3-ylamino)ethyl)piperidine-3-carboxamide Example 61 o~~
J / OMe ON O Nf N
I N

3-(N-methyl-N-(2-morpholinoethyl)amino)-7-(6-methoxypyridin-3-yl)-1-(2-propoxyethyl)pyrido[3, 4-b]pyrazin-2(1 H)-one Example 62 o~\J
OMe r I
O N \ \ N
NNX, N I ~ N
~
3-(N-methyl-N-(2-(piperidin-1-y1)ethyl)amino)-7-(6-methoxypyridin-3-y1)-1-(2-propoxyethyl)pyrido(3,4-b]pyrazin-2(yH)-one Example 63 OMe f I
o ON N
NJk H~H ~N N
o1-,) N-(3-(1,2-dihydro-7-(6-methoxypyridin-3-yl)-2-oxo-1-(2-propoxyethyl)pyrido[3,4-b]pyrazin-3-ylamino)propyl)morpholine-4-carboxamide Example 64 J / OMe r I
O O N N
N-/~N~N N H
0 "
N-(2-(1,2-dihydro-7-(6-methoxypyridin-3-yl)-2-oxo-1-(2-propoxyethyl)pyrido(3, 4-b]pyrazin-3-ylamino)ethyl)morpholine-4-carboxamide Example 65 o~~
OMe r I
O N N
X, a_N'--"N ~N 20 H N

3-(3-(pyridin-2-ylamino)propylamino)-7-(6-methoxypyridin-3 yl)-1-(2-propoxyethyl)pyrido[3,4-b]pyrazin-2(1 H)-one Additional compounds of Formula I that can be prepared in accordance with the synthetic methods of the present invention include those compounds described in Table B:

Table B

O'CF3 OCF3 N OMe N OMe O N \ \ ~ ~N~ O N \ \ ~

ON N~N I N ~N ~N~N N
~H H O H

O~CF3 OCF3 O N OMe N ~ ~N ~ N
oXO HO N I
H 'O' H

o~ O~CF3 O~CF3 O O
O \ I > O N >
N O HN O
~ H IN
I N N I N
/ N
' N
N N ' O H

O a O
O N > ~ O N >
HN O N \ O
N~N N ~N NX N I N
~H ~H

0 OMe O N \ I ~ HO O N \ N
/-N
N N1N N NN ~N ~N
H
~H

M. In Vitro Assays Method 1: Human Platelet PDE5 Enzyme Inhibition Scintillation Proximity Assay The IC50 of test compounds can be measured using an in vitro assay using PDE5 enzyme isolated from human platelets. The IC50 is the concentration of test compound required to inhibit the hydrolysis of cGMP to GMP by the PDE5 enzyme by 50% relative to the activity of uninhibited controls. The PDE5 enzyme for use in the assay can be obtained from human platelets by appropriate modification of the method of Thompson, WJ et al.;
Biochemistry 18(23), 5228-5237, 1979, as described by Ballard SA et al.; J. Urology 159(6), 2164-2171, 1998. The PDE5 enzyme so obtained can be used to catalyze the hydrolysis of [3H]cGMP
(Amersham Biosciences) to 5' nucleotide [3H]GMP. The [3H]GMP binds to yttrium silicate SPA beads (Amersham Biosciences) and is detected by scintillation counting. More specifically, the effect of the test compound at different concentrations can be evaluated in the assay by contacting the compound with a fixed amount of PDE5 enzyme in the presence of substrate (cGMP
or cAMP in a 3:1 ratio unlabelled to [3H]-labeled). Scintillation counting can be used as described above to determine relative PDE5 enzyme activity. The inhibition of PDE5 enzyme activity is then calculated relative to.total PDE5 enzyme activity of uninhibited controls.

PDE5 IC50 Assay: 96-well microtiter plate format Reagents Buffer A: 20 mM Tris-HCI, 5 mM MgCI2, pH 7.4 Buffer B: 2 mg/mi BSA in Buffer A (enzyme buffer) cGMP substrate: Final concentration of 500 nM in assay The amount of 3H-labeled substrate added depends upon the specific activity of [3H]cGMP, and the cGMP substrate is diluted with a 10 mM stock of cold cGMP in Buffer A for a final substrate concentration of 500 nM in the assay.
PDE enzyme: Prepared in Buffer B. The dilution factor is determined by enzyme activity.
SPA beads: 20 mg/mi suspension prepared in dH2O.
Positive Control Negative Control Standard/Test compound 2 pl 100% DMSO 2 pl 100% DMSO 2 pl Standard/Test compound pl Buffer A 25 pi Buffer A 25 pl Buffer A
25 NI Enzyme 25 pl Buffer B 25 pl Enzyme 20 50 pl Substrate 50 pl Substrate 50 pl Substrate 50 pl SPA to stop 50 pl SPA to stop 50 pl SPA to stop Stocks of standard and test compounds are prepared at 5 mM in 100% DMSO. The compound is serially diluted in a dilution plate using a 10-point 1/2 log dilution format. 2 pl of the 25 compound dilution is added in duplicate to the wells of the assay plate. 2 pl of 100% DMSO are added to designated control wells. 25 NI of Buffer A are added to all wells.
25 pl of Buffer B are added to the negative control wells. 25 pl of enzyme are added to the remaining wells. 50 pl of substrate are added to each well. Plates are sealed and incubated for 60 minutes on a plate shaker at 30 C. 50 pl of SPA beads are added to stop the reaction. The plates are again sealed and shaken for 15 minutes to allow the beads to bind the GMP product. The beads are allowed to settle for 30 minutes and then read on a NXT TopCount scintillation counter. Data are analyzed with a curve fitting application for plate-based screening. Percent inhibition in this assay is calculated as follows:

Inhibition (%) = [(mean maximum - compound value/ (mean maximum - mean minimum)] x 100.
The IC50value is determined from sigmoid dose-response curves of enzyme activity versus compound concentration.

Method 2: Alternative Human Platelet PDE5 Enzyme Inhibition Scintillation Proximity Ass~

The IC50 of test compounds also can be measured in an alternative in vitro assay that varies from Method 1 as described below:
PDE5 IC50 Assay: 96-well microtiter plate format Reagents Buffer A: 20 mM Tris-HCI, 5 mM MgCI2, pH 7.4 Buffer B: 2 mg/mI BSA in Buffer A (enzyme buffer) cGMP substrate: Final concentration of 50 nM in assay The amount of 3H-labeled substrate added depends upon the specific activity of [3H]cGMP, and it is diluted in Buffer A.
PDE enzyme: Prepared in Buffer B. The dilution factor is determined by enzyme activity.
SPA beads: 4 mg/mi suspension prepared in dH2O.

Positive Control Negative Control Standard/Test compound 3 pl 100% DMSO 3 pl 100% DMSO 3 pl Standard/Test compound 27 pl Buffer A 27 pl Buffer A 27 pl Buffer A
30 pl Enzyme 30 pl Buffer B 30 pl Enzyme 30 pl Substrate 30 pl Substrate 30 pl Substrate 30 pl SPA to stop 30 pi SPA to stop 30 pl SPA to stop Stocks of standard and test compound are prepared at 2 mM in 100% DMSO. The test compound is serially diluted in a dilution plate using an 8-point 1/5 log dilution format such that the starting concentration in the assay is 2 pM for an initial IC50 screen. 27 ial of.Buffer A are added to the wells of the assay plates. From the dilution plate, 3 pl of diluted compound is delivered in duplicate or 3 pl of 100 % DMSO (for positive and negative controls) are added. 30 pl of enzyme are added. For the negative control wells, Buffer B is substituted in place of the enzyme. 30 pl of labeled substrate are added to all wells.

After incubating for 60 minutes at room temperature, the reaction is stopped with the addition of 30 pl of the yttrium silicate beads. These beads are dense and require constant agitation while being added to the plate. The plates are sealed and shaken on a plate shaker for fifteen minutes to allow the beads to bind the GMP product.

After allowing the beads to settle for 30 minutes, plates are read on a NXT
TopCount scintillation counter and the data are analyzed as follows. Percent inhibition values are calculated using the means of the 0% and 100% controls on each plate. The estimates of the 4-parameters of the logistic, sigmoid dose-response model are then calculated using the well-level percent inhibition value for the compound. The formula for the four-parameter logistic model may be expressed as Y=((a - d) / (1 + ( X / c) A b) ) + d, where Y is the response, X
is the concentration, a is the lower asymptote (minimum response), d is the upper asymptote (maximum response), c is the model IC50 (in the same units as X),.and b is the slope (as described in De Lean, A., P. J. Munson, and D. Rodbard ("Simultaneous analysis of families of sigmoidal curves: application to bioassay, radioligand assay, and physiological dose-response curves." Am. J. Physiol. 235(2): E97-E102, 1978). These estimates are used to calculate the concentration that corresponds to 50% inhibition.

IC50 values are shown in Table C for compounds tested in accordance with Method 2 above.

Table C Example IC50 Enzyme (nM) Example IC50 Enzyme (nM) #
# (Method B) (Method B) 33 1.38 1 0.0710 34 1.35 2 0.0440 35 4.25 3 0.0450 36 1.93 4 0.121 37 0.116 0.0600 38 6.22 6 0.0890 39 1.66 7 0.038 40 36.9 8 0.0660 41 0.812 9 0.169 42 0.168 20.0 43 0.141 11 0.0550 44 0.273 12 0.150 45 0.356 13 0.0450 46 0.0220 14 0.0400 47 3.61 2.96 48 14.5 16 0.284 49 132 17 0.466 50 3.48 18 0.0740 51 4.97 19 1.01 52 2.00 0.127 53 1.80 21 0.0860 54 0.925 22 -- 0.0820 55 2.21 23 2.42 56 0.093 24 3.07 57 0.337 1.65 58 0.353 26 0.0720 59 0.139 27 0.134 60 0.115 28 0.0690 61 0.506 29 0.333 62 1.51 0.249 63 0.117 31 0.261 64 0.047 32 1.02 65 0.034 N. Biological Protocols--/n Vivo Assays Method 5: CulexTM Assay The effect of the test compound on systemic arterial blood pressure can be evaluated in a 5 conscious pre-cannulated spontaneously hypertensive rat ("SHR") model. This assay is conducted using an automated blood sampler ("ABS") system. The CulexT~" ABS
system (Bioanalytical System, Inc., West Lafayette, IN) comprises a laptop computer, four control units and metabolic cages. This ABS system allows for the collection of multiple blood samples from a single rat without causing undue stress to the animal. In addition, the ABS
system allows for the 10 collection of urine samples that can be potentially used for biomarker identifications. Through this approach, efficacy and standard pharmacokinetic studies are conducted in the conscious unrestrained SHR rats simultaneously to define the relationship between plasma free drug concentration or potential biomarker(s) and pharmacological effect (reduction of mean arterial blood pressure).

SHR rats at 12 to 16 weeks of age, weighing about 300g, undergo surgerical cannulation of both jugular veins and the right carotid artery. After surgical recovery, animals are placed in the CulexTM cages and tethered to a movement-responsive arm with a sensor that controls cage movement when the animal moves to prevent the catheters from being twisted.
Connections are made between the right jugular catheter and the CulexTM sterile tubing set for blood sampling, and the left jugular catheter for compound administration, and the catheter in the right carotid artery is connected to a pressure transducer for monitoring blood pressure. To keep the patency of the catheters, the right jugular cannula is maintained by the "tend"
function of the CulexTM that flushes the catheter with 20 pL heparin saline (10 units/mL) every 12 minutes or between sampling events, and the left jugular cannula is filled with heparin saline (20 units/mL). The patency of the right carotid cannula is maintained by slow infusion of heparin saline either directly into the extend tubing when blood pressure is not recorded or through the pressure transducer during the blood pressure monitoring. Animals are allowed to acclimate for at least two hours before compound evaluation. The test compound may be administered intravenously or by oral gavage. Blood sampling protocols (sampling time and volume) are programmed using the CulexTM software. The total amount of blood withdrawn from each animal will not exceed 750 pU24 hrs and 10 mUkg within two weeks. Heart rate, blood pressure, and drug concentration are monitored. Systemic arterial blood pressure and heart rate are recorded by PONEMAH
(Gould Instrument System, Valley View, OH), a pressure transducer through a data acquisition system for recording blood pressure and heart rate, for 6 to 24 hours based on experimental protocol. Mean arterial blood pressure (primary endpoint) is analyzed for assessing the efficacy of the compound.

Blood samples are analyzed for measuring plasma drug concentration, using the LC/MS/MS
method described below, and for evaluating potential biomarkers.

LC/MS/MS Method Sample Preparation: Plasma samples (50 pL unknown, control or blank) are mixed with 10 pL acetonitrile:water or a standard solution of the test compound and 150 pL
of internal standard solution (100 ng/mL of the test compound in acetonitrile). The mixture is centrifuged at 3000 rpm for 5 min, and 125 pL of the supernatant transferred to a 96 well plate. The solvent is evaporated under a stream of nitrogen and the residue is reconstituted with 80 pL
acetonitrile/0.1% aqueous formic acid (20:80 v/v).

A 20 pL volume of each prepared sample is injected onto a Phenomenex Synergi 4 pm MAX-RP 2.0 x 75 mm column and eluted at 0.4 mUmin using gradient elution from 0.1%
aqueous formic acid (mobile phase A) to acetonitrile (mobile phase B). The gradient program consists of initial application of 90% mobile phase A, followed by a linear gradient to 75% mobile phase B from 0.2 to 1.15 min after injection and held at 75% mobile phase B
until 2.0 min. The mobile phase was linearly changed back to 90% mobile phase A from 2.00 to 2.10 minutes, and the next injection took place at 3.00 min. Detection was performed by mass spectrometry using positive ion electrospray (ESI) with multiple reaction monitoring of the transitions m/z 454.00 (MH+ the Carboxypiperidine Compound) - m/z 408.00, m/z 466.24 (MH+ the Carboxypiperidine Compound) -> 409.33 . The ion spray voltagea is set at 5000. A calibration curve is constructed by using peak area ratios of the analyte relative to the internal standard.
Subject concentrations are determined by inverse prediction from their peak area ratios against the calibration curve.

Method 6: Implantation of Radio Transmitters and Subsequent Blood Pressure Screening by Telemetry in Spontaneously Hypertensive Rats SHR Rats are anesthetized with isoflurane gas via an isoflurane anesthesia machine that is calibrated to deliver isoflurane over a range of percentages as oxygen passes through the machine's inner chambers. The animals are placed in an induction chamber and administered isoflurane at 4-5% to reach a surgical plane of anesthesia. They are then maintained at 1-2%
during the surgical procedure via a nose cone, with isoflurane delivered via a smaller isoflurane anesthesia device on the surgical table.

Following administration of anesthesia, the rats are implanted with transmitters using aseptic procedures with commercially available sterile radio-telemetry units (Data Sciences, International, Roseville, MN 551 1 3-1 1 36). Prior to surgery the surgical field is shaved, scrubbed with DialT"' brand antimicrobial solution (containing 4% chlorhexidine gluconate and 4% isopropyl alcohol) followed by an application of iodine (10%) spray solution. A 2.5 to 3.0 cm laparotomy is preformed and the radio-telemetry units implanted into the abdomen, with the catheter tip inserted into the abdominal aorta. Baby Weitlaner retractors are used to retain soft tissue. A 1 cm section of the abdominal aorta is partially dissected and. that section cross-clamped briefly, punctured with a 21-gauge needle and the transmitter catheter tip introduced into the vessel and secured by a single 4.0 silk suture anchored to the adjacent psoas muscle. The transmitter body is then inserted into the abdominal cavity and simultaneously secured to the abdominal muscle wall while closing with running 4.0 silk suture. The skin layer is closed with subdermal continuous 4.0 absorbable suture. A subcutaneous (s.c.) administration of marcaine followed by a topical application of iodine is administered into and around the suture line, respectively, upon closing.
All rats receive a postoperative injection of buprenorphine C 0.05mg/kg, s.c.
before regaining consciousness. A typical dose volume for a 0.300kg rat will be 0.050m1. The rats must be fully recovered from their operative anesthesia before the administration of buprenorphine. They then receive the same dose once daily for 2 consecutive days, unless the animal demonstrates that it is in compromising postoperative pain.

Following surgery, the rats are returned to their cages and housed individually on solid bottom caging with paper bedding. A period of no less than 7 days is allowed for recovery before experimental procedures are initiated. It has been observed that the rats are typically hypertensive for several days following surgery and return to "normotensive"
levels by approximately the 7th day post-surgery. They are fed standard rat chow and water ad libitum throughout the experimental time line.

Test compounds are administered intragastrically (i.g.) via gavage, using of a stainless steel, 21h inch, 18 gauge gavage needle with a balled end. For single daily dosing, the target volume is 3.33 ml/kg, i.g. The dose volume for the test compound is approximately 1 ml/ rat.
The vehicles in which the test compound is administered is methylcellulose (0.5%) + Tween 80 (0.1%) in 50mM citrate buffer pH=5Ø

Blood pressure data will be obtained using Data Sciences International's data acquisition program (Version 3.0). Blood pressure samples are recorded at 1.5-3 minute intervals for a 5 second duration 24 hours per day for the entire study. This data is processed by Data Science's data analysis software into averages of a desired time inervals. All other data reduction is performed in Microsoft ExcelTM spreadsheets.

As various changes could be made in the above invention(s) without departing from the scope of the invention(s), it is intended that all matter contained in the above description be interpreted as illustrative and not in a limiting sense. All documents mentioned in this application are expressly incorporated by reference as if fully set forth at length, with the definitions of the present application controlling. When introducing elements of the present invention or the various embodiment(s) thereof, the articles "a", "an", "the" and "said" are intended to mean that -- --there are one or-more of-the-elements: -The terms "comprising", "including" and "having" are intended to be inclusive and mean that there may be additional elements other than the listed elements.

Claims (15)

1. A compound having the structure of Formula I:
wherein R2 is selected from the group consisting of aryl and 3 to 10 membered ring heterocycyl wherein R2 may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, cyano, nitro, oxo, alkyl, alkenyl, alkynyl, cycloalkyl, -OR100, -C(O)R100, -OC(O)R100, -C(O)OR100, -NR100R101 -N(R100)C(O)R101, -C(O)NR100R101, -C(O)NR100C(O)R101, and -S(O)m R100, aziridinyl, azetidinyl, pyrrolidinyl, piperidinyl, morpholinyl, thiomorpholinyl, and piperazinyl wherein (a) the alkyl, alkenyl, alkylnyl and cycloalkyl substituents may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, oxo, -OR102, and -C(O)OR102; and (b) the aziridinyl, azetidinyl, pyrrolidinyl, piperidinyl, morpholinyl, thiomorpholinyl, and piperazinyl substituents may be optionally substituted with one or more substituents selected from the group consisting of alkyl, hydroxy and alkoxy;

R100, R101 and R102 are independently selected from the group consisting of hydrogen, alkyl, alkenyl and alkynyl, wherein the alkyl may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, alkoxy, -C(O)OH and -C(O)NH2;

X6 is selected from the group consisting of hydrogen and alkyl wherein the X6 alkyl substituent may be optionally substituted with one or more substituents selected from the group consisting of chloro, fluoro, alkoxy and hydroxy;

Y6 represents a bond or is selected from the group consisting of alkyl, alkenyl and alkynyl, wherein (a) the Y6 alkyl, alkenyl and alkynyl substituents may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, cyano, oxo, cycloalkyl, -OR103, -C(O)R103, -C(O)OR103, -OC(O)R103, -NR103R104-N(R103)C(O)R104 and -C(O)NR103R104;

R103 and R104 are independently selected from the group consisting of hydrogen and alkyl, wherein the alkyl may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, carboxy, cyano, oxo, alkynyl, haloalkynyl, hydroxyalkynyl, carboxyalkynyl, alkoxy, haloalkoxy, hydroxyalkoxy, and carboxyalkoxy;

R6 is selected from the group consisting of aryl, aryl-C(O)-, heterocyclyl, aryl-C(O)-NR105-heterocyclyl-C(O)-, and heterocyclyl-C(O)-NR105- wherein R6 may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, cyano, oxo, alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heterocyclyl, -OR106, -C(O)R106-C(O)OR106, -OC(O)R106 -NR106R107, -N(R107)C(O)R107, -C(O)NR106R107-C(O)NR106C(O)R107, -SR106, -S(O)R106, -S(O)2R106, -N(R106)S(O)2R107 , and -S(O)2NR106 R107; wherein the alkyl, alkenyl, alkynyl, cycloalkyl, aryl and heterocyclyl susbstituents may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, carboxy, cyano, oxo, alkoxy, haloalkoxy, hydroxyalkoxy, and carboxyalkoxy;

R105 is independently selected from the group consisting of hydrogen and alkyl;

R106 and R107 are independently selected from the group consisting of hydrogen, alkyl, alkenyl, and alkynyl, wherein (a) the R106 and R107 alkyl and alkenyl substituents may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, carboxy, cyano, oxo, alkynyl, haloalkynyl, hydroxyalkynyl, carboxyalkynyl, alkoxy, haloalkoxy, hydroxyalkoxy, and carboxyalkoxy, and (b) the R106 and R107 alkynyl substituents may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, carboxy, cyano, oxo, alkoxy, haloalkoxy, hydroxyalkoxy, and carboxyalkoxy;

R8 is alkyl; wherein R8 may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, carboxy, cyano, alkynyl, -OR108, -C(O)R108, -C(O)OR108, -OC(O)R108, -NR108R109, -N(R108)C(O)R109, -C(O)NR108R109, -SR108, -S(O)R108, -S(O)2R108,and -C(O)NR108C(O)R109, wherein the alkynyl substituents may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, carboxy, cyano, oxo, alkyl, and alkoxy; and R108 and R109 are independently selected from the group consisting of hydrogen, alkyl, alkenyl and alkynyl, wherein (a) when the alkyl is methyl, the methyl may be optionally substituted with 1, 2, or 3 fluoro substituents, (b) when the alkyl comprises at least two carbon atoms, the alkyl may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, carboxy, cyano, oxo, alkynyl, haloalkynyl, hydroxyalkynyl, carboxyalkynyl, alkoxy, haloalkoxy, hydroxyalkoxy, and carboxyalkoxy, and (c) the R108 and R109 alkynyl substituents may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, carboxy, cyano, oxo, alkoxy, haloalkoxy, hydroxyalkoxy, and carboxyalkoxy.
2. A compound of claim 1 wherein X6 is selected from the group consisting of hydrogen, C1 to C4 alkyl, wherein said C1 to C4 alkyl is optionally substituted with one or more substituents selected from the group consisting of C1 to C4 alkoxy and hydroxy.
3. A compound of claim 2 wherein R2 is selected from the group consisting of R9, R10, R11, R12 and R13 are independently selected from the group consisting of hydrogen, halogen, oxo, alkyl, -OR100, -C(O)R100, -OC(O)R100, -C(O)OR100, -NR100R101 and -C(O)NR100R101, wherein the alkyl substitutent may be optionally substituted with one or more substituents independently selected from the group consisting of halogen, oxo, -OR102, and -C(O)OR102; and R100, R101, and R 102 are independently selected from the group consisting of hydrogen and C1 to C4 alkyl.
4. A compound of claim 2 wherein R2 is selected from the group consisting of R9, R10, R11, R12 and R13 are independently selected from the group consisting of hydrogen, fluoro, methyl, trifluoromethyl, and methoxy.
5. A compound of claim 2 wherein Y6 represents a bond or is selected from the group consisting of alkyl and hydroxyalkyl.
6. A compound of claim 2 wherein Y6 represents a bond or is selected from the group consisting of methyl, ethyl, propyl, butyl, tert-butyl, hydroxyethyl, hydroxypropyl, hydroxybutyl, and dihydroxyethyl .
7. A compound of claim 2 wherein R6 is selected from the group consisting of phenyl C(O)NH-,
8. A compound of claim 2 wherein R8 is alkoxyalkyl optionally substituted as described in claim 1.
9. A compound of claim 8 wherein R2 is selected from the group consisting of phenyl and pyridinyl, optionally substituted as described in claim 1;
y6 represents a bond or is selected from the group consisting of methyl, ethyl, and propyl; and R8 is selected from the group consisting of ethoxyethyl and propoxyethyl.
10. A compound of claim 8 wherein:

R2 is selected from the group consisting of R9, R10, R11, R12 and R13 are independently selected from the group consisting of hydrogen, fluoro, methyl, trifluoromethyl, and methoxy;
Y6 represents a bond or is selected from the group_consisting of methyl, ethyl, and propyl;
R8 is selected from the group consisting of propoxyethyl and ethoxyethyl; and R6 is selected from the group consisting of is selected from the group consisting of phenyl, phenyl C(O)NH-, phenyl C(O)-, 5 to 6 membered ring fully saturated heterocyclyl, 5 to 6 membered ring fully saturated heterocyclyl-C(O)-, and 5 to 6 membered ring fully saturated heterocyclyl-C(O)-NH, optionally substituted with one or more substituents independently selected from the group consisting of alkyl, -OR106, and -C(O)R106, wherein R106 is selected from the group consisting of hydrogen, methyl, and ethyl.
11. The compound according to claim 1 selected from the group consisting of:
7-(6-methoxypyridin-3-yl)-3-[(2-morpholin-4-ylethyl)amino]-1-(2-propoxyethyl)pyrido[3,4-b]pyrazin-2(1H)-one;
7-(6-methoxypyridin-3-yl)-1-(2-propoxyethyl)-3-[(pyridin-2-ylmethyl)amino]pyrido[3,4-b]pyrazin-2(1H)-one;
7-(6-methoxypyridin-3-yl)-1-(2-propoxyethyl)-3-[(tetrahydrofuran-2-ylmethyl)amino]pyrido[3,4-b]pyrazin-2(1H)-one;
1-(2-isopropoxyethyl)-7-(6-methoxypyridin-3-yl)-3-{{(5-methylpyrazin-2-yl)methyl]amino}pyrido[3,4-b]pyrazin-2(1H)-one;
7-(6-methoxypyridin-3-yl)-3-{[(5-methylpyrazin-2-yl)methyl]amino}-1-(2-propoxyethyl)pyrido[3,4-b]pyrazin-2(1H)-one;
1-(2-isopropoxyethyl)-7-(6-methoxypyridin-3-yl)-3-[(tetrahydro-2H-pyran-4-ylmethyl)amino]pyrido[3,4-b]pyrazin-2(1H)-one;

tert-butyl 4-({[7-(6-methoxypyridin-3-yl)-2-oxo-1-(2-propoxyethyl)-1,2-dihydropyrido[3,4-b]pyrazin-3-yl]amino}methyl)piperidine-1-carboxylate;
7-(6-methoxypyridin-3-yl)-3-[(piperidin-4-ylmethyl)amino]-1-(2-propoxyethyl)pyrido[3,4-b]pyrazin-2(1H)-one;
7-(6-methoxypyridin-3-yl)-3-{[(5-methylisoxazol-3-yl)methyl]amino}-1-(2-propoxyethyl)pyrido[3,4-b]pyrazin-2(1H)-one;
3-{[3-(1H-imidazol-1-yl)propyl]amino}-7-(6-methoxypyridin-3-yl)-1-(2-propoxyethyl)pyrido[3,4-b]pyrazin-2(1H)-one;
3-[(2-morpholin-4-ylethyl)amino]-1-(2-propoxyethyl)-7-pyrimidin-5-ylpyrido[3,4-b]pyrazin-2(1H)-one;
7-(6-methoxypyridin-3-yl)-1-(2-propoxyethyl)-3-[(tetrahydro-2H-pyran-4-ylmethyl)amino]pyrido[3,4-b]pyrazin-2(1H)-one;
3-(2-morpholino-2-oxoethylamino)-7-(6-methoxypyridin-3-yl)-1-(2-propoxyethyl)pyrido[3,4-b]pyrazin-2(1H)-one;
7-(6-methoxypyridin-3-yl)-1-(2-propoxyethyl)-3-{[(2S)-tetrahydrofuran-2-ylmethyl]am ino}pyrido[3,4-b]pyrazin-2(1H)-one;
7-(6-methoxypyridin-3-yl)-1-(2-propoxyethyl)-3-{[(2R)-tetrahydrofuran-2-ylm ethyl]am ino}pyrido[3,4-b]pyrazin-2(1H)-one;

ethyl 4-{[7-(6-m ethoxypyridin-3-yl)-2-oxo-1-(2-propoxyethyl)-1,2-d ihydropyrido[3,4-b]pyrazin-3-yl]am ino}piperidine-1-carboxylate;
7-(6-methoxypyridin-3-yl)-3-(piperidin-4-ylamino)-1-(2-propoxyethyl)pyrido[3,4-b]pyrazin-2(1H)-one;

3-[(1-acetylpiperidin-4-yl)am ino]-7-(6-methoxypyridin-3-yl)-1-(2-propoxyethyl)pyrido[3,4-b]pyrazin-2(1H)-one; and 7-(6-methoxypyridin-3-yl)-3-[(2-piperidin-1-ylethyl)amino]-1-(2-propoxyethyl)pyrido[3,4-b]pyrazin-2(1H)-one.
12. The compound according to claim 1 selected from the group consisting of:
7-(6-methoxypyridin-3-yl)-1-(2-propoxyethyl)-3-[(2-pyridin-2-ylethyl)amino]pyrido[3,4-b]pyrazin-2(1H)-one;

3-(2-(4-ethylpiperazin-1-yl)-2-oxoethylamino)-7-(6-methoxypyridin-3-yl)-1 -(2-propoxyethyl)pyrido[3,4-b]pyrazin-2(1H)-one;
ethyl 4-(2-(1,2-dihydro-7-(6-methoxypyridin-3-yl)-2-oxo-1-(2-propoxyethyl)pyrido[3,4-b]pyrazin-3-ylamino)acetyl)piperazine-1-carboxylate;
3-(2-oxo-2-(pyrrolidin-1-yl)ethylamino)-7-(6-methoxypyridin-3-yl)-1-(2-propoxyethyl)pyrido[3,4-b]pyrazin-2(1H)-one;

3-(2-morpholino-2-oxoethylamino)-7-(3,5-dimethylisoxazol-4-yl)-1-(2-propoxyethyl)pyrido[3,4-b]pyrazin-2(1H)-one;
3-(2-oxo-2-(piperazin-1-yl)ethylam ino)-7-(6-m ethoxypyridin-3-yl)-1-(2-propoxyethyl)pyrido[3,4-b]pyrazin-2(1H)-one;
3-(2-(4-methylpiperazin-1-yl)-2-oxoethylamino)-7-(6-methoxypyridin-3-yi)-1-(2-propoxyethyl)pyrido[3,4-b]pyrazin-2(1H)-one;
3-(2-(4-hydroxypiperidin-1-yl)-2-oxoethylamino)-7-(6-methoxypyridin-3-yl)-1-(2-propoxyethyl)pyrido[3,4-b]pyrazin-2(1H)-one;
3-(2-oxo-2-(piperidin-1-yl)ethylam ino)-7-(6-m ethoxypyridin-3-yl)-1-(2-propoxyethyl)pyrido[3,4-b]pyrazin-2(1H)-one;
3-[(1S,4S)-2,5-diazabicyclo[2.2.1]hept-2-yl]-7-(6-methoxypyridin-3-yl)-1-(2-propoxyethyl)pyrido[3,4-b]pyrazin-2(1H)-one;
3-(2-morpholinoethylamino)-1-(2-isopropoxyethyl)-7-(6-methoxypyridin-3-yl)pyrido[3,4-b]pyrazin-2(1H)-one;
3-((pyridin-2-yl)methylamino)-1-(2-isopropoxyethyl)-7-(6-methoxypyridin-3-yl)pyrido[3,4-b]pyrazin-2(1H)-one;
3-(((S)-tetrahydrofuran-2-yl)methylamino)-1-(2-isopropoxyethyl)-7-(6-methoxypyridin-3-yl)pyrido[3,4-b]pyrazin-2(1H)-one;
1-(3-ethoxypropyl)-7-(6-methoxypyridin-3-yl)-3-[(pyridin-2-ylmethyl)amino]pyrido[3,4-b]pyrazin-2(1H)-one;
1-(3-ethoxypropyl)-7-(6-methoxypyridin-3-yl)-3-{[(5-methylpyrazin-2-yl)methyl]amino}pyrido[3,4-b]pyrazin-2(1H)-one;
1-(3-ethoxypropyl)-7-(6-methoxypyridin-3-yl)-3-{[(2S)-tetrahydrofuran-2-ylmethyl]amino}pyrido[3,4-b]pyrazin-2(1H)-one;
3-(2-morpholino-2-oxoethylamino)-1-(3-ethoxypropyl)-7-(6-methoxypyridin-3-yl)pyrido[3,4-b]pyrazin-2(1H)-one; and 7-(3,5-dimethylisoxazol-4-yl)-1 (2-ethoxyethyl)-3-[(2-morpholin-4-ylethyl)amino]pyrido[3,4-b]pyrazin-2(1H)-one.
13. The compound according to claim 1 selected from the group consisting of:
3-(((S)-tetrahydrofuran-2-yl)methylamino)-1-(2-ethoxyethyl)-7-(6-methoxypyridin-3-yl)pyrido[3,4-b]pyrazin-2(1H)-one;
3-(2-(pyrrolidin-1-yl)ethylamino)-1-(2-ethoxyethyl)-7-(6-methoxypyridin-3-yl)pyrido[3,4-b]pyrazin-2(1H)-one;
3-(2-(piperidin-1-yl)ethylamino)-1-(2-ethoxyethyl)-7-(6-methoxypyridin-3-yi)pyrido[3,4-b]pyrazin-2(1H)-one;
3-(2-morpholinoethylamino)-1-(2-ethoxyethyl)-7-(pyridin-3-yl)pyrido[3,4-b]pyrazin-2(1H)-one;
3-(2-morpholino-2-oxoethylamino)-1-(2-ethoxyethyl)-7-(6-methoxypyridin-3-yl)pyrido[3,4-b]pyrazin-2(1H)-one;

3-((5-methylpyrazin-2-yl)methylamino)-1-(2-ethoxyethyl)-7-(6-methoxypyridin-3-yI)pyrido[3,4-b]pyrazin-2(1H)-one;
3-((pyridin-2-yl)methylamino)-1-(2-ethoxyethyl)-7-(6-methoxypyridin-3-yl)pyrido[3,4-b]pyrazin-2(1H)-one;
3-(2-morpholinoethylamino)-1-(2-ethoxyethyl)-7-(6-methoxypyridin-3-yl)pyrido[3,4-b]pyrazin-2(1H)-one;
3-(2-morpholinoethylamino)-7-(4-fluorophenyl)-1-(2-propoxyethyl)pyrido[3,4-b]pyrazin-2(1H)-one;

7-(1,3-benzodioxol-5-yl)-3-[(2-morpholin-4-ylethyl)amino]-1-(2-propoxyethyl)pyrido[3,4-b]pyrazin-2(1H)-one;

3-(2-morpholinoethylamino)-1-(2-ethoxyethyl)-7-(4-methoxyphenyl)pyrido[3,4-b]pyrazin-2(1H)-one;
7-(2,3-dihydro-1-benzofuran-5-yl)-1-(2-ethoxyethyl)-3-[(2-morpholin-4-ylethyl)amino]pyrido[3,4-b]pyrazin-2(1 H)-one;
1-(2-ethoxyethyl)-3-[(2-morpholin-4-ylethyl)amino]-7-[4-(trifluoromethyl)phenyl]pyrido[3,4-b]pyrazin-2(1H)-one;
1-(2-ethoxyethyl)-3-[(2-morpholin-4-ylethyl)amino]-7-[3-(trifluoromethyl)phenyl]pyrido[3,4-b]pyrazin-2(1H)-one;
3-(2-morpholinoethylamino)-1-(2-ethoxyethyl)-7-phenylpyrido[3,4-b]pyrazin-2(1H)-one;
3-(2-morpholinoethylamino)-1-(2-ethoxyethyl)-7-(4-fluorophenyl)pyrido[3,4-b]pyrazin-2(1H)-one;
3-(2-morpholinoethylamino)-1-(2-ethoxyethyl)-7-(3-fluorophenyl)pyrido[3,4-b]pyrazin-2(1H)-one; and 3-(2-morpholinoethylamino)-1-(2-ethoxyethyl)-7-(3,4-difluorophenyl)pyrido[3,4-b]pyrazin-2(1H)-one.
14. A pharmaceutical composition comprising a therapeutically-effective amount of a compound of claim 1.
15. A method of treating a condition selected from the group consisting of cardiovascular disease, metabolic disease, central nervous system disease, pulmonary disease, sexual dysfunction, and renal dysfunction in a subject, comprising administering to the subject a therapeutically-effective amount of a compound of claim 1.
CA002608672A 2005-05-24 2006-05-17 Pyridine [3,4-b] pyrazinones as pde-5 inhibitors Withdrawn CA2608672A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US68413905P 2005-05-24 2005-05-24
US60/684,139 2005-05-24
PCT/IB2006/001387 WO2006126082A2 (en) 2005-05-24 2006-05-17 Pyridine [3,4-b] pyrazinones as pde-5 inhibitors

Publications (1)

Publication Number Publication Date
CA2608672A1 true CA2608672A1 (en) 2006-11-30

Family

ID=37056557

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002608672A Withdrawn CA2608672A1 (en) 2005-05-24 2006-05-17 Pyridine [3,4-b] pyrazinones as pde-5 inhibitors

Country Status (2)

Country Link
CA (1) CA2608672A1 (en)
WO (1) WO2006126082A2 (en)

Families Citing this family (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2007242555B2 (en) * 2006-04-21 2010-12-16 Pfizer Products Inc. Pyridine[3,4-b]pyrazinones
FR2902009B1 (en) * 2006-06-13 2012-12-07 Bioprojet Soc Civ USE OF A VASOPEPTIDASE INHIBITOR FOR THE TREATMENT OF PULMONARY ARTERIAL HYPERTENSION
EP2247292B1 (en) * 2008-03-05 2017-04-12 Merck Patent GmbH Pyridopyrazinones derivatives insulin secretion stimulators, methods for obtaining them and use thereof for the treatment of diabetes
WO2012131539A1 (en) 2011-03-31 2012-10-04 Pfizer Inc. Novel bicyclic pyridinones
WO2012172449A1 (en) 2011-06-13 2012-12-20 Pfizer Inc. Lactams as beta secretase inhibitors
ES2605565T3 (en) 2011-08-31 2017-03-15 Pfizer Inc Hexahydropyran [3,4-D] [1,3] thiazin-2-amine compounds
WO2013164730A1 (en) 2012-05-04 2013-11-07 Pfizer Inc. Heterocyclic substituted hexahydropyrano [3,4-d] [1,3] thiazin- 2 -amine compounds as inhibitors of app, bace1 and bace 2.
JP2015529239A (en) 2012-09-20 2015-10-05 ファイザー・インク Alkyl-substituted hexahydropyrano [3,4-d] [1,3] thiazin-2-amine compounds
UA110688C2 (en) 2012-09-21 2016-01-25 Пфайзер Інк. Bicyclic pirydynony
CA2893256A1 (en) 2012-12-11 2014-06-19 Pfizer Inc. Hexahydropyrano [3,4-d][1,3]thiazin-2-amine compounds as inhibitors of bace1
CA2893333C (en) 2012-12-19 2017-10-24 Pfizer Inc. Carbocyclic-and heterocyclic-substituted hexahydropyrano[3,4-d][1,3]thiazin-2-amine compounds
EP2956458B1 (en) 2013-02-13 2017-08-09 Pfizer Inc Heteroaryl-substituted hexahydropyrano[3,4-d][1,3]thiazin-2-amine compounds
US9233981B1 (en) 2013-02-15 2016-01-12 Pfizer Inc. Substituted phenyl hexahydropyrano[3,4-d][1,3]thiazin-2-amine compounds
KR20150119370A (en) 2013-02-19 2015-10-23 화이자 인코포레이티드 Azabenzimidazole compounds as inhibitors of pde4 isozymes for the treatment of cns and other disorders
CA2925743C (en) 2013-10-04 2018-03-06 Pfizer Inc. Novel bicyclic pyridinones as gamma-secretase modulators
AP2016009465A0 (en) 2014-04-01 2016-09-30 Pfizer Chromene and 1,1 a,2,7b-tetrahydrocyclopropa[c]chromene pyridopyrazinediones as gamma-secretase modulators
WO2015155626A1 (en) 2014-04-10 2015-10-15 Pfizer Inc. 2-AMINO-6-METHYL-4,4a,5,6-TETRAHYDROPYRANO[3,4-d][1,3]THIAZIN-8a(8H)-YL-1,3-THIAZOL-4-YL AMIDES
JP6713982B2 (en) 2014-07-24 2020-06-24 ファイザー・インク Pyrazolopyrimidine compounds
PE20170295A1 (en) 2014-08-06 2017-03-30 Pfizer IMIDAZOPYRIDAZINE COMPOUNDS
AU2016214102B2 (en) 2015-02-03 2018-09-27 Pfizer Inc. Novel cyclopropabenzofuranyl pyridopyrazinediones
CR20170572A (en) 2015-06-17 2018-03-20 Pfizer TRICYCLIC COMPOUNDS AND THEIR USE AS INHIBITORS OF PHOSPHODIESTERASE
JP2018531924A (en) 2015-09-24 2018-11-01 ファイザー・インク Tetrahydropyrano [3,4-D] [1,3] oxazine derivatives and their use as BACE inhibitors
AU2016325665A1 (en) 2015-09-24 2018-03-08 Pfizer Inc. N-[2-(2-amino-6,6-disubstituted-4, 4a, 5, 6-tetrahydropyrano [3,4-d][1,3] thiazin-8a (8h)-yl) -1, 3-thiazol-4-yl] amides
JP2018534251A (en) 2015-09-24 2018-11-22 ファイザー・インク N- [2- (3-Amino-2,5-dimethyl-1,1-dioxide-5,6-dihydro-2H-1,2,4-thiadiazin-5-yl) -1 useful as a BACE inhibitor , 3-Thiazol-4-yl] amide
ES2775674T3 (en) 2016-02-23 2020-07-27 Pfizer 6,7-Dihydro-5H-Pyrazolo [5,1-b] [1,3] Oxazine-2-Carboxamide Compounds
EP3872078A1 (en) 2016-07-01 2021-09-01 Pfizer Inc. 5,7-dihydro-pyrrolo-pyridine derivatives for use in the treament of depression, anxiety or panic disorders
DK3642202T3 (en) 2017-06-22 2023-01-30 Pfizer Dihydro-pyrrolo-pyridine derivatives
ES2942837T3 (en) 2018-03-23 2023-06-07 Pfizer Azaspiro piperazine derivatives

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6465465B1 (en) * 2000-03-14 2002-10-15 Arzneimittelwerk Dresden Gmbh Process for the treatment of erectile dysfunction and product therefor
EP1364950A4 (en) * 2001-02-26 2005-03-09 Tanabe Seiyaku Co Pyridopyrimidine or naphthyridine derivative
WO2002076954A1 (en) * 2001-03-23 2002-10-03 Smithkline Beecham Corporation Compounds useful as kinase inhibitors for the treatment of hyperproliferative diseases
US7067658B2 (en) * 2002-09-30 2006-06-27 Bristol-Myers Squibb Company Pyridino and pyrimidino pyrazinones

Also Published As

Publication number Publication date
WO2006126082A3 (en) 2007-01-25
WO2006126082A2 (en) 2006-11-30

Similar Documents

Publication Publication Date Title
AU2007242555B2 (en) Pyridine[3,4-b]pyrazinones
CA2608672A1 (en) Pyridine [3,4-b] pyrazinones as pde-5 inhibitors
WO2006126081A2 (en) Pyridino [2 , 3-b] pyrazinones as pde-5 inhibitors
WO2006126083A1 (en) Pyridine [3 , 4-b] pyrazinone compounds as pde-5 inhibitors
ES2717680T3 (en) Zeste homolog 2 inhibitor potentiator
US7951801B2 (en) Beta-carbolines useful for treating inflammatory disease
US20100093713A1 (en) Beta-carbolines useful for treating inflammatory disease
WO2007020521A1 (en) Pyridoyrazinones as pde-5 inhibitors
AU2009318855A1 (en) Lactams as beta secretase inhibitors
TW202122382A (en) Hydantoin derivative
JP7464526B2 (en) 4-HYDROXYPIPERIDINE DERIVATIVES AND THEIR USE AS INHIBITORS OF UBIQUITIN-SPECIFIC PROTEASE 19 (USP19) - Patent application
CA3121376A1 (en) Pharmaceutical compounds and their use as inhibitors of ubiquitin specific protease 19 (usp19)
WO2024052692A1 (en) Novel compounds as ck2 inhibitors

Legal Events

Date Code Title Description
EEER Examination request
AZWI Withdrawn application