AU2012216322A1 - Pyridazine compounds, compositions and methods - Google Patents

Pyridazine compounds, compositions and methods Download PDF

Info

Publication number
AU2012216322A1
AU2012216322A1 AU2012216322A AU2012216322A AU2012216322A1 AU 2012216322 A1 AU2012216322 A1 AU 2012216322A1 AU 2012216322 A AU2012216322 A AU 2012216322A AU 2012216322 A AU2012216322 A AU 2012216322A AU 2012216322 A1 AU2012216322 A1 AU 2012216322A1
Authority
AU
Australia
Prior art keywords
compound
substituted
disease
alkyl
formula
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
AU2012216322A
Inventor
Jean-Jacques Bourguignon
Jacques Haiech
Marcel Hibert
Wenhui Hu
Linda Van Eldik
Anastasia Velentza
Martin D. Watterson
Magdelena Zasadzki
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.)
Centre National de la Recherche Scientifique CNRS
Universite de Strasbourg
Northwestern University
Original Assignee
Centre National de la Recherche Scientifique CNRS
Universite de Strasbourg
Northwestern University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from AU2005302225A external-priority patent/AU2005302225B2/en
Application filed by Centre National de la Recherche Scientifique CNRS, Universite de Strasbourg, Northwestern University filed Critical Centre National de la Recherche Scientifique CNRS
Priority to AU2012216322A priority Critical patent/AU2012216322A1/en
Publication of AU2012216322A1 publication Critical patent/AU2012216322A1/en
Priority to AU2016203312A priority patent/AU2016203312B2/en
Abandoned legal-status Critical Current

Links

Landscapes

  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention relates to novel chemical compounds, compositions and methods of making and using the same. In particular, the invention provides pyridazine compounds and/or related heterocyclic derivatives, compositions comprising the same, and methods of making and using pyridazine compounds and/or related heterocyclic derivatives and compositions comprising the same, for modulation of cellular pathways (e.g. signal transduction pathways), for treatment or prevention of inflammatory diseases (eg Alzheimer's disease), for research, drug screening, and therapeutic applications.

Description

Regulation 3.2 Revised 2/98 AUSTRALIA Patents Act, 1990 ORIGINAL COMPLETE SPECIFICATION TO BE COMPLETED BY THE APPLICANT NAME OF APPLICANT: Northwestern University; Centre National de la Recherche Scientifique and University de Strasbourg ACTUAL INVENTORS: WATTERSON, Martin D VAN ELDIK, Linda HAIECH, Jacques HIBERT, Marcel BOURGUIGNON, Jean-Jacques VELENTZA, Anastasia HU, Wenhui ZASADZKI, Magdelena ADDRESS FOR SERVICE: Peter Maxwell and Associates Level 6 60 Pitt Street SYDNEY NSW 2000 INVENTION TITLE: PYRIDAZINE COMPOUNDS, COMPOSITIONS AND METHODS DETAILS OF ASSOCIATED APPLICATION NO(S): Divisional of Australia Patent Application No. 2005 302 225 filed on 2 November 2005 The following statement is a full description of this invention including the best method of performing it known to us. m:\docs\20051261 \259355.doc PYRIDAZINE COMPOUNDS, COMPOSITIONS AND METHODS This invention was funded, in part, under NIH grants P01AG21184 and R01NS47586. 5 The US government may have certain rights in the invention. RELATED APPLICATIONS This application claims priority benefit from prior U.S. Provisional Application Serial Number 60/624,346 filed November 2, 2004 and U.S. Provisional Applications Serial Number 10 60/723,124, filed October 3, 2005, and 601723,090, filed October 3, 2005. FMID OF INVENTION The invention relates to novel chemical compounds, compositions and methods of making and using the same. In particular, the invention provides pyridazine compounds and/or 15 related heterocyclic derivatives, compositions comprising the same, and methods of using pyridazine compounds and/or related heterocyclic derivatives and compositions comprising the same, for modulation of cellular pathways (e.g., signal transduction pathways), for treatment or prevention of inflammatory diseases (e.g., Alzheimer's disease), for research, drug screening, and therapeutic applications. 20 BACKGROUND OF INVENTION The majority of inflammatory conditions and diseases result from a disruption in the homeostatic balance between beneficial and detrimental responses of the organism. For example, there may be a decrease in the production of trophic molecules that mediate cell survival and other beneficial cellular processes, or there may be an overproduction of pro-inflammatory or other 25 detrimental molecules that mediate toxic cellular responses. Disregulation of signal transduction pathways involving protein kinases are often involved in the generation or progression of these diseases. For example, neuroinflammation is a process that results primarily from an abnormally high or chronic activation of glia (microglia and astrocytes). This overactive state of glia results in increased levels of inflammatory and oxidative stress molecules, which can lead to neuron damage or 30 death. Neuronal damage/death can also induce glial activation, facilitating the propagation of a localized, detrimental cycle of neuroinflammation [7]. The inflammation (e.g., neuroinflammation) cycle has been proposed as a potential therapeutic target in the development of new approaches to treat inflammatory disease (e.g., Alzheimer's disease). However, the efficacy and lexicological profile of compounds that focus only on classical non- 2 steroidal anti-inflammatory drug targets have been disappointing to date, for example, most anti inflammatory therapeutics are palliative, providing minimal, short-lived, symptomatic relief with limited effects on inflammatory disease (e.g., neuroinflammatory diseases such as Alzheimer's disease) progression. Because the major societal impact from inflammatory diseases (e.g., 5 neuroinflammatory diseases such as Alzheimer's disease) is expected to increase greatly in coming decades, there is an urgent need for anti-inflammatory therapeutics that impact disease progression when administered soon after diagnosis (e.g., diagnosis of cognitive decline), or in a chemo preventive paradigm as combinations of risk factors with prognostic value are identified. In either therapeutic paradigm, new drugs must have a good therapeutic index, especially in regard to 10 potential toxicology in the elderly. Despite an overwhelming need, and the presence of well-defined molecular targets, the current anti-inflammatory drug development pipeline is lacking chemically diverse compounds that work within the relevant therapeutic window and treatment paradigm needed for altering disease progression, An area of comparative neglect that fits this therapeutic window is 15 neuroinflammation [1]. Thus, development of new classes of anti-inflammatory compounds that can modulate inflammatory disease-relevant pathways are urgently needed. SUMMARY OF INVENTION The present invention relates to novel chemical compounds, compositions and methods of making and using the same. In particular, the present invention provides pyridazine compounds 0 and/or related heterocyclic derivatives, compositions comprising the same, and methods of using pyridazine compounds and/or related heterocyclic derivatives, and compositions comprising the same, for modulation of cellular pathways (e.g., signal transduction pathways), for treatment or prevention of inflammatory diseases (e.g., Alzheimer's disease), for research, drug screening, and therapeutic applications. :5 The invention provides a method for treating a disease disclosed herein, in particular an inflammatory disease in a subject comprising administering to the subject a pyridazinyl radical pendant with an aryl or substituted aryl, in particular phenyl or substituted phenyl, a heteroaryl or substituted heteroaryl, in particular piperazinyl substituted with pyrimidinyl, or pyridinyl. In an aspect, the invention provides a method for treating a disease disclosed herein, in 0 particular an inflammatory disease, in a subject comprising administering to the subject a compound of the Formula I, comprising I a and Ib: Formula I 3 R1 R o 0 R _R R4 R6 R 4 R0* R' R" Ia Th wherein R', R 2 , and R 3 are independently hydrogen, hydroxyl, alkyl, alkenyl, alkynyl, alkylene, 5 alkenylene, alkoxy, alkenyloxy, cycloalkyl, cycloalkenyl, aryl, aryloxy, arylalkoxy, aroyl, heteroaryl, heterocyclic, acyl, acyloxy, sulfonyl, sulfinyl, sulfenyl, amino, imino, azido, thiol, thioalkyl, thioalkoxy, thioaryl, nitro, ureido, cyano, halo, silyl, silyloxy, silylalkyl, silylthio, =0, =S, carboxyl, carbonyl, carbamoyl, or carboxamide; R 7 is hydrogen, hydroxyl, alkyl, alkenyl, alkynyl, alkylene, alkenylene, alkoxy, alkenyloxy, cycloalkyl, cycloalkenyl, aryl, aryloxy, 10 arylalkoxy, aroyl, heteroaryl, heterocyclic, acyl, acyloxy, sulfonyl, sulfinyl, sulfenyl, amino, imino, azido, thiol, thioalkyl, thioalkoxy, thioaryl, nitro, ureido, cyano, halo, silyl, silyloxy, silylalkyl, silylthio, =0, =S, carboxyl, carbonyl, carbamoyl, or carboxamide or R7 may be absent and there is a double bond between N at position 1 and C at position 6; R 4 , R 5 , and R 6 are independently hydrogen, alkyl, alkoxy, halo, or nitro; or R1 and R 2 , R' and R 7 , or R 2 and R 3 15 may form a heteroaryl or heterocyclic ring; or an isomer or a pharmaceutically acceptable salt thereof. In an embodiment, R 1 is a piperazinyl or substituted piperazinyl, in particular a piperazinyl substituted with a pyrimidinyl of Formula A below.
N
20 N A Therefore, the invention also provides a method for treating a disease disclosed herein, in particular an inflammatory disease, in a subject comprising administering to the subject a 25 compound of the Formula II: 4
R'
0 R (9 N N II wherein R 0 and R" are independently hydrogen, hydroxyl, alkyl, alkenyl, alkynyl, alkylene, alkenylene, alkoxy, alkenyloxy, cycloalkyl, cycloalkenyl, aryl, aryloxy, arylalkoxy, aroyl, 5 heteroaryl, heterocyclic, acyl, acyloxy, sulfonyl, sulfinyl, sulfenyl, amino, imino, azido, thiol, thioalkyl, thioalkoxy, thioaryl, nitro, ureido, cyano, halo, silyl, silyloxy, silylalkyl, silylthio, =0, =S, carboxyl, carbonyl, carbamoyl, or carboxamide; or an isomer or a pharmaceutically acceptable salt thereof In an aspect, the invention provides a method for treating a disease disclosed herein, in 10 particular an inflammatory disease, in a subject comprising administering to the subject a compound of the Formula III: Rs R16 YQN N wherein R 15 and R1 6 are independently hydrogen, hydroxyl, alkyl, alkenyl, alkynyl, alkylene, 15 alkenylene, alkoxy, alkenyloxy, cycloalkyl, cycloalkenyl, aryl, aryloxy, arylalkoxy, aroyl, heteroaryl, heterocyclic, acyl, acyloxy, sulfonyl, sulfinyl, sulfenyl, amino, imino, azido, thiol, thioalkyl, thioalkoxy, thioaryl, nitro, ureido, cyano, halo, silyl, silyloxy, silylalkyl, silylthio, =0, =S, carboxyl, carbonyl, carbamoyl, or carboxamide; or an isomer or a pharmaceutically acceptable salt thereof. 20 In another aspect, the invention provides a method for treating a disease disclosed herein, in particular an inflammatory disease, in a subject comprising administering to the subject a compound of the Formula IV: 5 R" R 71 0 N IV wherein R 7 is substituted or unsubstituted hydrogen, hydroxyl, alkyl, alkenyl, alkynyl, alkylene, alkenylene, alkoxy, alkenyloxy, cycloalkyl, cycloalkenyl, aryl, aryloxy, arylalkoxy, aroyl, heteroaryl, heterocyclic, acyl, acyloxy, sulfonyl, sulfinyl, sulfenyl, amino, imino, azido, thiol, 5 thioalkyl, thioalkoxy, thioaryl, nitro, ureido, cyano, halo, silyl, silyloxy, silylalkyl, silylthio, =0, =S, carboxyl, carbonyl, carbamoyl, or carboxamide, especially heterocyclic, heteroaryl, amino, and substituted amino and R 7 1 is aryl or substituted aryl; or an isomer or a pharmaceutically acceptable salt thereof. In a further aspect, the invention provides a method for treating a disease disclosed 10 herein, in particular an inflammatory disease, in a subject comprising administering to the subject a compound of the Formula V: rO R0 Rox R6 2 V wherein R 50 , R 51 , and Rs 2 are independently hydrogen, hydroxyl, alkyl, alkenyl, alkynyl, alkylene, alkenylene, alkoxy, alkenyloxy, cycloalkyl, cycloalkenyl, aryl, aryloxy, arylalkoxy, 15 aroyl, heteroaryl, heterocyclic, acyl, acyloxy, sulfonyl, sulfinyl, sulfenyl, amino, imino, azido, thiol, thioalkyl, thioalkoxy, thioaryl, nitro, ureido, cyano, halo, silyl, silyloxy, silylalkyl, silylthio, =0, =S, carboxyl, carbonyl, carbamoyl, or carboxamide; or an isomer or a pharmaceutically acceptable salt thereof.
6 In an aspect, a method is provided for treating a disease disclosed herein in a subject comprising administering a compound of the Formula I, H, Il, IV, or V as defined herein, with the proviso that compounds depicted in Table 1 are excluded. The invention relates to a method for treating diseases disclosed herein in a subject 5 comprising administering to the subject a therapeutically effective amount of one or more compound of the Formula I, I, M, IV, or V, or a pharmaceutically acceptable salt thereof, or a composition comprising a compound of the Formula I, , III, IV, or V and a pharmaceutically acceptable carrier, excipient, or vehicle. In an aspect the invention provides beneficial effects following treatment. The methods of the invention can be used therapeutically or .0 prophylactically in a subject susceptible to or having a genetic predisposition to a disease disclosed herein. In another aspect of the invention, a method is provided for treating in a subject a disease involving or characterized by inflammation, in particular neuroinflammation, comprising administering to the subject a therapeutically effective amount of a compound of the Formula I, 15 11, m, IV, or V, or a pharmaceutically acceptable salt thereof. In a further aspect, a method is provided for treating in a subject a condition involving inflammation, in particular neuroflammation, comprising administering to the subject a therapeutically effective amount of a composition comprising a compound of the Formula I, 1H, I1, IV, or V and a pharmaceutically acceptable carrier, excipient, or vehicle. 20 In a further aspect, the invention provides a method involving administering to a subject a therapeutic compound of the Formula I, 1, IM, IV, or V, or a pharmaceutically acceptable salt thereof, or a composition comprising a compound of the Formula I, II, I, IV, or V, and a pharmaceutically acceptable carrier, excipient, or vehicle which inhibit or reduce neuroflammation, activation of glia, proinflammatory cytokines, oxidative stress-related 25 enzymes, acute phase proteins and/or components of the complement cascade. In another aspect, the invention provides a method for treating in a subject a disease associated with neuroinflammation that can be decreased or inhibited with a compound disclosed herein comprising administering to the subject a therapeutically effective amount of a compound of the Formula I, If, III, IV, or V, a pharmaceutically acceptable salt thereof, or a 30 composition comprising a compound of the Formula I, HI, IH, IV, or V, n particular the compounds depicted in the Figures and Tables, more particularly Table 2, 3, 4 or 5 and derivatives thereof and an isomer or pharmaceutically acceptable carrier, excipient, or vehicle. Methods of the invention may be used to prevent or inhibit activation of protein kinases, in particular death associated protein kinase (DAPK); reduce or inhibit kinase activity, glial 7 activation, neuronal cell damage, and/or neuronal cell death; inhibit cell signaling molecule production (e.g., I-lp and TNFa), amelioriate progression of a disease or obtain a less severe stage of a disease in a subject suffering from such disease (e.g., neuroinflammatory disease, in particular a neurodegenerative disease, more particularly Alzheimer's disease); delay the 5 progression of a disease (e.g. neuroinflammatory disease, in particular a neurodegenerative disease, more particularly Alzheimer's disease), increase survival of a subject suffering from a disease (e.g. neuroinflammatory disease, in particular a neurodegenerative disease, more particularly Alzheimer's disease; treat or prevent a neurodegenerative disease, in particular Alzheimer's disease; treat mild cognitive impairment (MCI); reverse or inhibit 10 neuroinflammation, activation of signaling pathways involved in inflammation (e.g., neuroinflammation), cell signaling molecule production, activation of glia or glial activation pathways and responses, proinflammatory cytokines or chemokines (e.g., interleukin (IL) or tumor necrosis factor (TNF), oxidative stress-related responses such as nitric oxide synthase production and nitric oxide accumulation, acute phase proteins, components of the complement 15 cascade, protein kinase activity (e.g., death associated protein kinase activity), neuronal cell damage, and/or neuronal cell death, after the onset of cognitive deficits and Alzheimer's disease neuropathology in a subject; improve memory of a healthy subject or the memory of a subject with age impaired memory; improve memory, especially short-term memory and other mental dysfunction associated with the aging process; treat a mammal in need of improved memory, 20 wherein the mammal has no diagnosed disease, disorder, infirmity or ailment known to impair or otherwise diminish memory; and/or improve the lifespan of a subject suffering from Alzheimer's disease. The invention provides a method of preventing a disease disclosed herein in a subject with a genetic predisposition to such disease by administering an effective amount of a 25 compound of the Formula I H, IH, IV, or V, or a pharmaceutically acceptable salt thereof, or a composition comprising a compound of the Formula I, II, M, IV, or V and a pharmaceutically acceptable carrier, excipient, or vehicle. The invention relates to compounds of the Formula I with the proviso that the compounds depicted in Table 1 are excluded. 30 The invention relates to compounds of the Formula II with the proviso that the compounds depicted in Table 1 are excluded. The invention also relates to compounds of the Formula I with the proviso that compounds depicted in Table 1 are excluded.
8 The invention also relates to compounds of the Formula IV with the proviso that compounds depicted in Table 1 are excluded. The invention also relates to compounds of the Formula V with the proviso that compounds depicted in Table 1 are excluded. 5 A compound of the Formula I, H, III, IV, or V may optionally comprise a carrier interacting with one or more radicals in the compound, for example R1, R 2 , R 3 , R 4 , R 5 , R 6 or R 7 in Formula I. A carrier may be a polymer, carbohydrate, or peptide, or combinations thereof, and it may be optionally substituted, for example, with one or more alkyl, halo, hydroxyl, halo, or amino. 10 In accordance with aspects of the invention pyridazine compounds and/or related heterocyclic derivatives thereof (See, e.g., the Figures and Tables herein, in particular Table 2, 3, 4 or 5 or derivatives thereof) are provided for use in research, drug screening, for modulation of cellular pathways (e.g., signal transduction pathways), and for treatment or prevention of inflammatory diseases (e.g., Alzheimer's disease). In some embodiments, the present invention 15 provides new classes of chemical compounds capable of modulating pro-inflammatory and oxidative stress related, cellular signaling pathways (e.g., in activated glial cells). In some embodiments, one or more compounds of the Figures and Tables herein are used to modulate kinase activity alone or in combination with other compounds or therapies. In some embodiments, compounds, and methods of using the compounds, provided by the invention are 20 those depicted in the Figures and Table 2, 3, 4, and/or 5 and derivatives thereof. In some embodiments, the invention provides MWO1-3-5-183WH, MWO1-5-188WH, MWO1-2 065LKM, MWO1-2-184WH, MWO1-2-189WH and MWO1-2-151SRM and methods of synthesizing the same. In some embodiments, the invention provides MWO1-3-5-183WH, MWO1-5-188WH, 25 MWO1-2-065LKM, MWO1-2-184WH, MWO1-2-151SRM, MWO1-2-189WH, MWO1-1-01-L D07, and/or related heterocyclic derivatives of these compounds and methods of making and using the same for modulating cellular pathways (e.g., signal transduction pathways) for use in research, drug screening, and therapeutic applications. In an aspect, the invention provides compositions for prevention and/or treatment of a 30 disease disclosed herein. Thus, the invention provides a pharmaceutical composition comprising a compound of the Formula I,1H, MII, IV, or V, in particular a therapeutically effective amount of a compound of the Formula I, IT, III, IV, or V, more particularly a compound depicted in the Figures and Table 2, 3, 4, and/or 5 or derivatives thereof, for treating a disease. More particularly, the invention provides a pharmaceutical composition in a form adapted for administration to a 9 subject to provide therapeutic effects, in particular beneficial effects to treat a disease disclosed herein. In another aspect, the composition is in a form such that administration to a subject suffering from a disease results in a decrease or reversal of one or more of the following: 5 inflammation (e.g. neuroinflammation), activation of signaling pathways involved in inflammation (e.g., neuroinflammation), cell signaling molecule production, activation of glia or glial activation pathways and responses, proinflammatory cytokines or chemokines (e.g., interleukin (IL) or tumor necrosis factor (TNF), oxidative stress-related responses such as nitric oxide synthase production and nitric oxide accumulation, acute phase proteins, components of 10 the complement cascade, protein kinase activity (e.g., death associated protein kinase activity), cell damage (e.g., neuronal cell damage), and/or cell death (e.g., neuronal cell death). A composition of the invention can be in a form that results in one or more of a decrease or reversal of one or more of the following: inflammation (e.g. neuroinflammation), activation of signaling pathways involved in inflammation (e.g., neuroinflammation), cell signaling molecule 15 production, activation of glia or glial activation pathways and responses, proinflammatory cytokines or chemokines (e.g., interleukin (IL) or tumor necrosis factor (TNF), oxidative stress related responses such as nitric oxide synthase production and nitric oxide accumulation, acute phase proteins, components of the complement cascade, protein kinase activity (e.g., death associated protein kinase activity), cell damage (e.g., neuronal cell damage), and/or cell death 20 (e.g., neuronal cell death) in a subject. In an aspect, the invention features a composition comprising a compound of the invention in a therapeutically effective amount for decreasing or reversing of one or more of the following: inflammation (e.g. neuroinflammation), activation of signaling pathways involved in inflammation (e.g., neuroinflammation), cell signaling molecule production, activation of glia or 25 glial activation pathways and responses, proinflammatory cytokines or chemokines (e.g., interleukin (IL) or tumor necrosis factor (TNF), oxidative stress-related responses such as nitric oxide synthase production and nitric oxide accumulation, acute phase proteins, components of the complement cascade, protein kinase activity (e.g., death associated protein kinase activity), cell damage (e.g., neuronal cell damage), and/or cell death (e.g., neuronal cell death) in a 30 subject. The composition can be in a pharmaceutically acceptable carrier, excipient, or vehicle. Additionally the invention contemplates a method of preparing a stable pharmaceutical composition comprising one or more compound of the Formula I, II, II, IV, or V. After a composition is prepared, it can be placed in an appropriate container and labeled for treatment 10 of an indicated disease. For administration of a composition of the invention, such labeling would include amount, frequency, and method of administration. In some aspects the invention provides methods to make commercially available pills, tablets, caplets, soft and hard gelatin capsules, lozenges, sachets, cachets, vegicaps, liquid drops, 5 elixirs, suspensions, emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium) suppositories, sterile injectable solutions, and/or sterile packaged powders, which contain a compound of the Formula I,1H, lI, IV, or V of the invention. In an aspect, compounds and compositions of the invention may be administered therapeutically or prophylactically to treat a disease disclosed herein. While not wishing to be 10 bound by any particular theory, the compounds and compositions may act to ameliorate the course of a disease using without limitation one or more of the following mechanisms: preventing, reducing and/or inhibiting inflammation (e.g. neuroinflammation), activation of signaling pathways involved in inflammation (e.g., neuroinflammation), cell signaling molecule production, activation of glia or glial activation pathways and responses, proinflammatory 15 cytokines or chemokines (e.g., interleukin (IL) or tumor necrosis factor (TNF), oxidative stress related responses such as nitric oxide synthase production and nitric oxide accumulation, acute phase proteins, components of the complement cascade, protein kinase activity (e.g., death associated protein kinase activity), cell damage (e.g., neuronal cell damage), and/or cell death (e.g., neuronal cell death). 20 The invention relates to the use of a composition comprising at least one compound of the Formula I, 1, I, IV, or V for the preparation of a medicament for treating a disease disclosed herein. The invention additionally relates to uses of a pharmaceutical composition of the invention in the preparation of medicaments for the prevention and/or treatment of a disease disclosed herein. The medicament may be in a form suitable for consumption by a subject, for 25 example, a pill, tablet, caplet, soft and hard gelatin capsule, lozenge, sachet, cachet, vegicap, liquid drop, elixir, suspension, emulsion, solution, syrup, aerosol (as a solid or in a liquid medium) suppository, sterile injectable solution, and/or sterile packaged powder. The invention further relates to a kit comprising one or more compound of the Formula I, H, II, IV, or V or a composition comprising a compound of the Formula I, H, HI, IV, or V. In 30 an aspect, the invention provides a kit for preventing and/or treating a disease disclosed herein comprising one or more compound of the Formula I, H, M, IV, or V, a container, and instructions for use. The composition of a kit of the invention can further comprise a pharmaceutically acceptable carrier, excipient, or vehicle.
11 The compounds of the Formula I, II, I, IV, or V (in particular the compounds depicted in Table 2, 3, 4 and/or 5 or derivatives thereof) provide a structural scaffold on which to base compositions for decreasing or reversing one or more of the following: inflammation (e.g. neuroinflammation), activation of signaling pathways involved in inflammation (e.g., 5 neuroinflammation), cell signaling molecule production, activation of glia or glial activation pathways and responses, proinflammatory cytokines or chemokines (e.g., interleukin (IL) or tumor necrosis factor (TNF), oxidative stress-related responses such as nitric oxide synthase production and nitric oxide accumulation, acute phase proteins, components of the complement cascade, protein kinase activity (e.g., death associated protein kinase activity), cell damage (e.g., 10 neuronal cell damage), and/or cell death (e.g., neuronal cell death), wherein the compound comprise a structure of Formula I, IIM, IV, or V. Thus, the invention also contemplates libraries or collections of compounds all of which are represented by a compound of the Formula I, II, III, IV, or V, in particular a compound depicted in Table 2, 3, 4, and/or 5 or derivatives thereof. In particular, the invention 15 contemplates a combinatorial library comprising compounds for decreasing or reversing one or more of the following: inflammation (e.g. neuroinflammation), activation of signaling pathways involved in inflammation (e.g., neuroinflammation), cell signaling molecule production, activation of glia or glial activation pathways and responses, proinflammatory cytokines or chemokines (e.g., interleukin (IL) or tumor necrosis factor (TNF), oxidative stress-related 20 responses such as nitric oxide synthase production and nitric oxide accumulation, acute phase proteins, components of the complement cascade, protein kinase activity (e.g., death associated protein kinase activity), cell damage (e.g., neuronal cell damage), and/or cell death (e.g., neuronal cell death), wherein the compound comprise a structure of Formula I, II, I, IV, or V. These and other aspects, features, and advantages of the present invention should be 25 apparent to those skilled in the art from the following detailed description. DESCRIPTION OF THE FIGURES Figure 1 depicts a synthetic scheme for synthesis of MWO1-3-183WH. Figure 2 A and B are graphs showing concentration dependent inhibition of proinflammatory cytokine production by MWO1-3-183WH in BV-2 cells 30 Figure 3 A- H show graphs and micrographs of activity of MWO1-5-188WH. A is a graph of )IL-1p and (B) TNFa levels by the BV2 microglial cell line. (C) Accumulation of the NO metabolite, nitrite, was not inhibited; Western blots of iNOS, COX-2 or apoE production in activated glia in (D); iNOS, (E) COX-2 and (F) apoE from glia cultures. Micrographs of treatment with diluent and compound are shown in (G) and (H).
12 Figure 4 A-H shows graphs and micrographs of activity of MWO1-5-188WH after oral administration. Figure 5 A-C shows graphs of results of oral administration of MWO1-5-188WH. Figure 6 A-D shows graphs and immunoblots illustrating the cell-based activity of 5 MW01-2-151SRM in BV-2 microglial cells. Figure 7 A-G shows graphs illustrating in vivo activity of MWO1-2-151SRM in the As infusion mouse model. Graphs are of MW01-2-151SRM suppression of AP-induced neuroinflammation and synaptic damage and activity in the Y-maze. Hippocampal sections or extracts from vehicle-infused mice (control), Ap-infused mice injected with solvent, and Ap 10 infused mice injected with MW01-2-151SRM were evaluated for neuroinflammation by measurement of the levels of the pro-inflammatory cytokines IL-10 (A), TNFa (B), and SlOOB (C), and the number of GFAP-positive astrocytes, (D) and the presynaptic marker, synaptophysin, (E), and evaluated for synaptic damage by analysis of the levels of the post-synaptic density protein 95 (PSD-95) (F), and Y-maze. Data are from one of two independent experiments, and 15 are the mean + SEM for 4-5 mice per experimental group. Figure 8 is a synthetic scheme for MWO1-7-084WH. Figure 9 is a synthetic scheme for MW01-7-085WH. Figure 10 is a synthetic scheme for MWO1-7-091WH. Figure 11 is a synthetic scheme for MW01-2-065LKM. 20 Figure 12 is a synthetic scheme for MW01-2-069A-SRM. Figure 13 is a synthetic scheme for MWO1-2-151SRM. Figure 14 is a synthetic scheme for MW01-2-151SRM. Figure 15 is a synthetic scheme for MW01-2-151SRM. Figure 16 is a synthetic scheme for MWO1-5-188WH. 25 Figure 17 is a synthetic scheme for MW01-5-188WH. Figure 18 is a synthetic scheme for MW01-5-188WH. Figure 19 A and B are synthetic schemes for MW01-6-189WH. Figure 20 is a synthetic scheme for MWO1-7-029WH. Figure 21 is a synthetic scheme for MWO1-7-027B-WH. 30 Figure 22 is a synthetic scheme for MWO1-3-065SRM. Figure 23 is a synthetic scheme for MWO1-3-066SRM. Figure 24 is a synthetic scheme for MWO1-7-133WH. Figure 25 is a synthetic scheme for MW01-7-107WH. Figure 26 is a synthetic scheme for MWO1-7-057WH.
13 Figure 27 is a synthetic scheme for MW01-2-163MAS. Figure 28 is a synthetic scheme for MW01-7-084WH. Figure 29 A-C shows graphical data of the assays used herein for MW01-2-056WH. Figure 30 A-E shows graphical data of the assays used herein for MWO1-2-056WH. 5 Figure 31 A-C shows graphical data of the assays used herein and immunoblots for biological activity of MWO1-7-057WH. Figure 32 A-E shows graphical data of the assays used herein for MWO1-7-057WH. Figure 33 A-C shows graphical data of the assays used herein and immunoblots for biological activity of MW01-2-065LKM. 10 Figure 34 A-E shows graphical data of the assays used herein for MWO1-2-065LKM. Figure 35 A-C shows graphical data of the assays used herein and immunoblots for biological activity of MWO1-2-069A-SRM. Figure 36 A-E shows graphical data of the assays used herein for MWO1-2-069A-SRM. Figure 37 A-C shows graphical data of the assays used herein and immunoblots for 15 biological activity of MWO1-7-085WH. Figure 38 A-E shows graphical data of the assays used herein for MWO1-7-085WR Figure 39 A-C shows graphical data of the assays used herein and immunoblots for biological activity of MWO1-7-091WH. Figure 40 A-E shows graphical data of the assays used herein for MWO1-7-091WH. 20 Figure 41 A-C shows graphical data of the assays used herein and immunoblots for biological activity of MW01-7-107WH. Figure 42 A-E shows graphical data of the assays used herein for MWO1-7-107WH. Figure 43 A-C shows graphical data of the assays used herein and immunoblots for biological activity of MWO1-7-127WH. 25 Figure 44 A-E shows graphical data of the assays used herein for MWO1-7-127WH. Figure 45 A-C shows graphical data of the assays used herein and immunoblots for biological activity of MWO1-2-151SRM. Figure 46 A-H shows graphical data of the assays used herein for MW01-2-151SRM. Figure 47 A and B are graphs of response to MWO1-2-151SRM, in a test for toxicity 30 where DMSO in saline was the control, and QTc interval measurement. QT intervals were obtained at baseline and at 15 min, 30 min, 45 min, and 60 min after compound administration. Figure 48 A-F are graphs of stability data using human (AB) and rat (C,D) microsomes with MWO1-2-151SRM in two different amounts, for two time periods. E and F show human 14 (E) and (F) rat microsomes with MWO1-2-151SRM stability for different time periods compared to minaprine. Figure 49 A-C shows graphical data of the assays used herein and immunoblots for biological activity of MW01-2-163MAS. 5 Figure 50 A-C shows graphical data of the assays used herein and immunoblots for biological activity of MWO1-6-189WH. Figure 51 A-D shows graphical data of the assays used herein for MWO1-6-189WH. DETAILED DESCRIPTION 10 For convenience, certain terms employed in the specification, examples, and appended claims are collected here. Numerical ranges recited herein by endpoints include all numbers and fractions subsumed within that range (e.g. 1 to 5 includes 1, 1.5, 2, 2.75, 3, 3.90, 4, and 5). It is also to be understood that all numbers and fractions thereof are presumed to be modified by the term 15 "about." The term "about" means plus or minus 0.1 to 50%, 5-50%, or 10-40%, preferably 10 20%, more preferably 10% or 15%, of the number to which reference is being made. Further, it is .to be understood that "a," "an," and "the" include plural referents unless the content clearly dictates otherwise. Thus, for example, reference to a composition comprising "a compound" includes a mixture of two or more compounds. 20 As used herein the terms "administering" and "administration" refer to a process by which a therapeutically effective amount of a compound or composition contemplated herein is delivered to a subject for prevention and/or treatment purposes. Compositions are administered in accordance with good medical practices taking into account the subject's clinical condition, the site and method of administration, dosage, patient age, sex, body weight, and other factors 25 known to physicians. As used herein, the term "co-administration" refers to the administration of at least two compounds or agent(s) (e.g., compound of the Formula I, II, III, IV, or V or pyridazines) or therapies to a subject. In some embodiments, the co-administration of two or more agents/therapies is concurrent. In other embodiments, a first agent/therapy is administered prior to 30 a second agent/therapy. Those of skill in the art understand that the Formulations and/or routes of administration of the various agents/therapies used may vary. The appropriate dosage for co administration can be readily determined by one skilled in the art. In some embodiments, when agents/therapies are co-administered, the respective agents/therapies are administered at lower dosages than appropriate for their administration alone. Thus, co-administration is especially desirable in 15 embodiments where the co-administration of the agents/therapies lowers the requisite dosage of a known potentially harmful (e.g., toxic) agent(s). The term "treating" refers to reversing, alleviating, or inhibiting the progress of a disease, or one or more symptoms of such disease, to which such term applies. Depending on 5 the condition of the subject, the term also refers to preventing a disease, and includes preventing the onset of a disease, or preventing the symptoms associated with a disease. A treatment may be either performed in an acute or chronic way. The term also refers to reducing the severity of a disease or symptoms associated with such disease prior to affliction with the disease. Such prevention or reduction of the severity of a disease prior to affliction refers to administration of 10 a compound or composition of the present invention to a subject that is not at the time of administration afflicted with the disease. "Preventing" also refers to preventing the recurrence of a disease or of one or more symptoms associated with such disease. "Treatment" and "therapeutically," refer to the act of treating, as "treating" is defined above. The terms "subject", "individual", or "patient" are used interchangeably herein and refer 15 to an animal preferably a warm-blooded animal such as a mammal. Mammal includes without limitation any members of the Mammalia. In general, the terms refer to a human. The terms also include domestic animals bred for food or as pets, including equines, bovines, sheep, poultry, fish, porcines, canines, felines, and zoo animals, goats, apes (e.g. gorilla or chimpanzee), and rodents such as rats and mice. 20 In aspects of the invention, the terms refer to organisms to be treated by the methods of the present invention. Such organisms preferably include, but are not limited to, mammals (e.g., murines, simians, equines, bovines, porcines, canines, felines, and the like), and most preferably includes humans. In the context of particular aspects of the invention, the term "subject" generally refers to an individual who will receive or who has received treatment (e.g., administration of a 25 compound of the Formula I, II, III, IV, or V or a pyridazine compound(s), and optionally one or more other agents) for a condition characterized by inflammation, the dysregulation of protein kinase activity, and/or dysregulation of apototic processes. Typical subjects for treatment include persons afflicted with or suspected of having or being pre-disposed to a disease disclosed herein, or persons susceptible to, suffering from or that 30 have suffered a disease disclosed herein. A subject may or may not have a genetic predisposition for a disease disclosed herein such as Alzheimer's disease. In particular aspects, a subject shows signs of cognitive deficits and Alzheimer's disease neuropathology. In embodiments of the invention the subjects are suspectible to, or suffer from Alzheimer's disease.
16 As utilized herein, the term "healthy subject" means a subject, in particular a mammal, having no diagnosed disease, disorder, infirmity, or ailment, more particularly a disease, disorder, infirmity or ailment known to impair or otherwise diminish memory. The term "diagnosed," as used herein, refers to the recognition of a disease by its signs and 5 symptoms (e.g., resistance to conventional therapies), or genetic analysis, pathological analysis, histological analysis, and the like. As used herein, the term "modulate" refers to the activity of a compound (e.g., a compound of the Formula I H, III, 1V, or V, or a pyridazine compound) to affect (e.g., to promote or retard) an aspect of cellular function, including, but not limited to, cell growth, proliferation, apoptosis, and the .0 like. A "beneficial effect" refers to an effect of a compound of the invention or composition thereof in certain aspects of the invention, including favorable .pharmacological and/or therapeutic effects, and improved biological activity. In aspects of the invention, the beneficial effects include without limitation prevention, reduction, reversal, or inhibition of one or more of [5 the following: inflammation (e.g. neuroinflammation), activation of signaling pathways involved in inflammation (e.g., neuroinflammation), cell signaling molecule production, activation of glia or glial activation pathways and responses, proinflammatory cytokines or chemokines (e.g., interleukin (IL) or tumor necrosis factor (TNF), oxidative stress-related responses such as nitric oxide synthase production and nitric oxide accumulation, acute phase 20 proteins, components of the complement cascade, protein kinase activity (e.g., death associated protein kinase activity), cell damage (e.g., neuronal cell damage), and/or cell death (e.g., neuronal cell death). In some aspects, a beneficial effect is a favourable characteristic of a composition comprising a compound of the Formula I, II, Im, IV, or V including without limitation enhanced stability, a longer half life, and/or enhanced uptake and transport across the 25 blood brain barrier. The beneficial effect can be a statistically significant effect in terms of statistical analysis of an effect of a compound of the Formula I, II, I, IV, or V versus the effects without the compound or compound that is not within the scope of the invention. Statistically significant" or "significantly different" effects or levels may represent levels that are higher or lower than a 30 standard. In aspects of the invention, the difference may be 1.5, 2, 3, 4, 5, or 6 times higher or lower compared with the effect obtained without a compound of the Formula I, H, HI, IV, or V. The term "pharmaceutically acceptable carrier, excipient, or vehicle" refers to a medium which does not interfere with the effectiveness or activity of an active ingredient and which is not toxic to the hosts to which it is administered. A carrier, excipient, or vehicle includes 17 diluents, binders, adhesives, lubricants, disintegrates, bulking agents, wetting or emulsifying agents, pH buffering agents, and miscellaneous materials such as absorbents that may be needed in order to prepare a particular composition. Examples of carriers etc. include but are not limited to saline, buffered saline, dextrose, water, glycerol, ethanol, and combinations thereof. The use 5 of such media and agents for an active substance is well known in the art. The compounds of the Formula I, H, Il, IV, or V disclosed herein also include "pharmaceutically acceptable salt(s)". By pharmaceutically acceptable salts is meant those salts which are suitable for use in contact with the tissues of a subject or patient without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable 10 benefit/risk ratio. Pharmaceutically acceptable salts are described for example, in S. M. Berge, et al., J. Pharmaceutical Sciences, 1977, 66:1. Examples of salts include MW01-1-01-L-D10, MW01-1-01-L-E02, MW01-1-01-L-E08, MW01-1-03-L-A05, MWO1-1-16-L-D09, and MW01 1-17-L-G04. A compound of the Formula I, H, Il, IV, or V can contain one or more asymmetric 15 centers and may give rise to enantiomers, diasteriomers, and other stereoisomeric forms which may be defined in terms of absolute stereochemistry as (R)- or (S)-. Thus, compounds of the Formula I, II, III, IV, or V include all possible diasteriomers and enantiomers as well as their racemic and optically pure forms. Optically active (R)- and (S)-isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques. When a compound 20 of the Formula I, I II, IV, or V contains centers of geometric asymmetry, and unless specified otherwise, it is intended that the compounds include both E and A geometric isomers. All tautomeric forms are also included within the scope of a compound of the Formula I, I1, H, IV, or V. A compound of the Formula I, II, II, IV, or V can exist in unsolvated as well as solvated 25 forms with pharmaceutically acceptable solvents such as water, ethanol, and the like. The solvated forms may be considered equivalent to the unsolvated forms for the purposes of the present invention. "Therapeutically effective amount" relates to the amount or dose of an active compound of the Formula I, H, Ill, IV, or V or composition comprising the same, that will lead to one or 30 more desired effects, in particular, one or more therapeutic effects, more particularly beneficial effects. A therapeutically effective amount of a substance can vary according to factors such as the disease state, age, sex, and weight of the subject, and the ability of the substance to elicit a desired response in the subject. A dosage regimen may be adjusted to provide the optimum therapeutic response (e.g. sustained beneficial effects). For example, several divided doses may 18 be administered daily or the dose may be proportionally reduced as indicated by the exigencies of the therapeutic situation. As used herein, the term "pure" in general means better than 95% pure, and "substantially pure" means a compound synthesized such that the compound, as made or as 5 available for consideration into a composition or therapeutic dosage described herein, has only those impurities that can not readily nor reasonably be removed by conventional purification processes. The term "derivative" of a compound, as used herein, refers to a chemically modified compound wherein the chemical modification takes place either at a functional group of the [0 compound or on the aromatic ring. Non-limiting examples of derivatives of compounds of the Formula I, , , IV, or V (e.g., pyridazine derivatives of the present invention) may include N-acetyl, N-methyl, N-hydroxy groups at any of the available nitrogens in the compound. A "polymer" refers to molecules comprising two or more monomer subunits that may be identical repeating subunits or different repeating subunits. A monomer generally comprises a 15 simple structure, low-molecular weight molecule containing carbon. Polymers may optionally be substituted. Polymers that can be used in the present invention include without limitation vinyl, acryl, styrene, carbohydrate derived polymers, polyethylene glycol (PEG), polyoxyethylene, polymethylene glycol, poly-trimethylene glycols, polyvinylpyrrolidone, polyoxyethylene-polyoxypropylene block polymers, and copolymers, salts, and derivatives 20 thereof. In aspects of the invention, the polymer is poly(2-acrylamido-2-methyl-l propanesulfonic acid); poly(2-acrylamido-2-methyl,-1-propanesulfonic acid-coacrylonitrile, poly(2-acrylamido-2-methyl-1-propanesulfonic acid-co-styrene), poly(vinylsulfonic acid); poly(sodium 4-styrenesulfonic acid); and sulfates and sulfonates derived therefrom; poly(acrylic acid), poly(methylacrylate), poly(methyl methacrylate), and poly(vinyl alcohol). 25 A "carbohydrate" as used herein refers to a polyhydroxyaldehyde, or polyhydroxyketone and derivatives thereof. The term includes monosaccharides such as erythrose, arabinose, allose, altrose, glucose, mannose, threose, xylose, gulose, idose, galactose, talose, aldohexose, fructose, ketohexose, ribose, and aldopentose. The term also includes carbohydrates composed of monosaccharide units, including disaccharides, oligosaccharides, or polysaccharides. Examples 30 of disaccharides are sucrose, lactose, and maltose. Oligosaccharides generally contain between 3 and 9 monosaccharide units and polysaccharides contain greater than 10 monosaccharide units. A carbohydrate group may be substituted at one two, three or four positions, other than the position of linkage to a compound of the Formula I, II, II, IV, or V. For example, a carbohydrate may be substituted with one or more alkyl, amino, nitro, halo, thiol, carboxyl, or 19 hydroxyl groups, which are optionally substituted. Illustrative substituted carbohydrates are glucosamine, or galactosamine. In aspects of the invention, the carbohydrate is a sugar, in particular a hexose or pentose and may be an aldose or a ketose. A sugar may be a member of the D or L series and can include amino sugars, deoxy sugars, and their uronic acid derivatives. 5 In embodiments of the invention where the carbohydrate is a hexose, the hexose is glucose, galactose, or mannose, or substituted hexose sugar residues such as an amino sugar residue such as hexosamine, galactosamine, glucosamine, in particular D-glucosamine (2-amino-2-doexy-D glucose) or D-galactosamine (2-amino-2-deoxy-D-galactose). Illustrative pentose sugars include arabinose, fucose, and ribose. 10 A sugar residue may be linked to a compound of the Formula I, II, III, IV, or V from a 1,1 linkage, 1,2 linkage, 1,4 linkage, 1,5 linkage, or 1,6 linkage. A linkage may be via an oxygen atom of a compound of the Formula I, II, III, IV, or V. An oxygen atom can be replaced one or more times by -CH 2 - or -S- groups. The term "carbohydrate" also includes glycoproteins such as lectins (e.g. concanavalin 15 A, wheat germ agglutinin, peanutagglutinin, seromucoid, and orosomucoid) and glycolipids such as cerebroside and ganglioside. A "peptide" carrier for use in the practice of the present invention includes one, two, three, four, or five or more amino acids covalently linked through a peptide bond. A peptide can comprise one or more naturally occurring amino acids, and analogs, derivatives, and congeners 20 thereof. A peptide can be modified to increase its stability, bioavailability, solubility, etc. "Peptide analogue" and "peptide derivative" as used herein include molecules which mimic the chemical structure of a peptide and retain the functional properties of the peptide. A carrier for use in the present invention can be an amino acid such as alanine, glycine, proline, methionine, serine, threonine, histidine, asparagine, alanyl-alanyl, prolyl-methionyl, or glycyl-glycyl. A 25 carrier can be a polypeptide such as albumin, antitrypsin, macroglobulin, haptoglobin, caeruloplasm, transferring, a- or 0- lipoprotein, 0- or y- globulin or fibrinogen. Approaches to designing peptide analogues, derivatives and mimetics are known in the art. For example, see Farmer, P. S. in Drug Design (E. J. Ariens, ed.) Academic Press, New York, 1980, vol. 10, pp. 119-143; Ball. J. B. and Alewood, P. F. (1990) J Mol. Recognition 30 3:55; Morgan, B. A. and Gainor, I. A. (1989) Ann. Rep. Med. Chem. 24:243; and Freidinger, R. M. (1989) Trends Pharmacol. Sci. 10:270. See also Sawyer, T. K. (1995) "Peptidomimetic Design and Chemical Approaches to Peptide Metabolism" in Taylor, M. D. and Amidon, G. L. (eds.) Peptide-Based Drug Design: Controlling Transport and Metabolism, Chapter 17; Smith, 20 A. B. 3rd, et al. (1995) J. Am. Chem. Soc. 117:11113-11123; Smith, A. B. 3rd, et al. (1994) J. Am. Chem. Soc. 116:9947-9962; and Hirschman, R., et al. (1993) J. Am. Chem. Soc. 115:12550-12568. A peptide can be attached to a compound of the Formula I, II, III, IV, or V through a 5 functional group on the side chain of certain amino acids (e.g. serine) or other suitable functional groups. A carrier may comprise four or more amino acids with groups attached to three or more of the amino acids through functional groups on side chains. In an aspect, the carrier is one amino acid, in particular a sulfonate derivative of an amino acid, for example cysteic acid. 10 The term "alkyl", either alone or within other terms such as "thioalkyl" and "arylalkyl", means a monovalent, saturated hydrocarbon radical which may be a straight chain (i.e. linear) or a branched chain. An alkyl radical for use in the present invention generally comprises from about 1 to 20 carbon atoms, particularly from about 1 to 10, 1 to 8 or 1 to 7, more particularly about 1 to 6 carbon atoms, or 3 to 6. Illustrative alkyl radicals include methyl, ethyl, n-propyl, n 15 butyl, n-pentyl, n-hexyl, isopropyl, isobutyl, isopentyl, amyl, sec-butyl, tert-butyl, tert-pentyl, n heptyl, n-octyl, n-nonyl, n-decyl, undecyl, n-dodecyl, n-tetradecyl, pentadecyl, n-hexadecyl, heptadecyl, n-octadecyl, nonadecyl, eicosyl, dosyl, n-tetracosyl, and the like, along with branched variations thereof. In certain aspects of the invention an alkyl radical is a C-C 6 lower alkyl comprising or selected from the group consisting of methyl, ethyl, n-propyl, n-butyl, n 20 pentyl, n-hexyl, isopropyl, isobutyl, isopentyl, amyl, tributyl, sec-butyl, tert-butyl, tert-pentyl, and n-hexyl. An alkyl radical may be optionally substituted with substituents as defined herein at positions that do not significantly interfere with the preparation of compounds of the Formula I, II, III, IV, or V and do not significantly reduce the efficacy of the compounds. In certain aspects of the invention, an alkyl radical is substituted with one to five substituents including 25 halo, lower alkoxy, lower aliphatic, a substituted lower aliphatic, hydroxy, cyano, nitro, thio, amino, keto, aldehyde, ester, amide, substituted amino, carboxyl, sulfonyl, sulfinyl, sulfenyl, sulfate, sulfoxide, substituted carboxyl, halogenated lower alkyl (e.g. CF 3 ), halogenated lower alkoxy, hydroxycarbonyl, lower alkoxycarbonyl, lower alkylcarbonyloxy, lower alkylcarbonylamino, cycloaliphatic, substituted cycloaliphatic, or aryl (e.g., phenylmethyl (i.e. 30 benzyl)). Substituent on an alkyl group may themselves be substituted. As used herein in respect to certain aspects of the invention, the term "substituted aliphatic" refers to an alkyl or an alkane possessing less than 10 carbons where at least one of the aliphatic hydrogen atoms has been replaced by a halogen, an amino, a hydroxy, a nitro, a thio, a ketone, an aldehyde, an ester, an amide, a lower aliphatic, a substituted lower aliphatic, or a ring 21 (aryl, substituted aryl, cycloaliphatic, or substituted cycloaliphatic, etc.). Examples of such groups include, but are not limited to, 1-chloroethyl and the like. As used herein in respect to certain aspects of the invention, the term "lower-alkyl substituted-amino" refers to any alkyl unit containing up to and including eight carbon atoms 5 where one of the aliphatic hydrogen atoms is replaced by an amino group. Examples of such include, but are not limited to, ethylamino and the like. As used herein in respect to certain aspects of the invention, the term "lower-alkyl substituted-halogen" refers to any alkyl chain containing up to and including eight carbon atoms where one of the aliphatic hydrogen atoms is replaced by a halogen. Examples of such include, 10 but are not limited to, chlorethyl and the like. As used herein, the term "acetylamino" shall mean any primary or secondary amino that is acetylated. Examples of such include, but are not limited to, acetamide and the like. As used herein the term "alkenyl" refers to an unsaturated, acyclic branched or straight chain hydrocarbon radical comprising at least one double bond. An alkenyl radical may contain 15 from about 2 to 10 carbon atoms, in particular from about 3 to 8 carbon atoms and more particularly about 3 to 6 carbon atoms. Suitable alkenyl radicals include without limitation ethenyl, propenyl (e.g., prop-l-en-1-yl, prop-1-en-2-yl, prop-2-en-1-yl (allyl), prop-2-en-2-yl), buten-1-yl, but-1-en-2-yl, 2-methyl-prop-1-en-1-yl, but-2-en-1-yl, but-2-en-1-yl, but-2-en-2-yl, buta-1,3-dien-1-yl, buta-1,3-dien-2-yl, hexen-1-yl, 3-hydroxyhexen-1-yl, hepten-1-yl, and 20 octen-1-yl, and the like. An alkenyl radical may be optionally substituted similar to alkyl. As used herein, the term "alkynyl" refers to an unsaturated, branched or straight-chain hydrocarbon radical comprising one or more triple bonds. An alkynyl radical may contain about 1 to 20, 1 to 15, or 2-10 carbon atoms, particularly about 3 to 8 carbon atoms and more particularly about 3 to 6 carbon atoms. Suitable alkynyl radicals include without limitation 25 ethynyl, such as prop-1-yn-1-yl, prop-2-yn-1-yl, butynyls such as but-1-yn-1-yl, but-1-yn-3-yl, but-3-yn-1-yl, pentynyls such as pentyn-1-yl, pentyn-2-yl, 4-methoxypentyn-2-yl, 3 methylbutyn-1-yl, hexynyls such as hexyn-1-yl, hexyn-2-yl, hexyn-3-yl, and 3,3-dimethylbutyn 1-yl radicals and the like. An alkenyl may be optionally substituted similar to alkyl. The term "cycloalkynyl" refers to cyclic alkynyl groups. 30 As used herein the term "alkylene" refers to a linear or branched radical having from about 1 to 10 carbon atoms and having attachment points for two or more covalent bonds. Examples of such radicals are methylene, ethylene, propylene, butylene, pentylene, hexylene, ethylidene, methylethylene, and isopropylidene. When an alkenylene radical is present as a 22 substituent on another radical it is typically considered to be a single substituent rather than a radical formed by two substituents. As used herein the term "alkenylene" refers to a linear or branched radical having from about 2 to 10 carbon atoms, at least one double bond, and having attachment points for two or 5 more covalent bonds. Examples of alkenylene radicals include 1,1-vinylidene (-CH 2 =C-), 1,2 vinylidene (-CH=CH-), and 1,4-butadienyl (-CH=CH-CH=CH-). As used herein the term "halo" refers to a halogen such as fluorine, chlorine, bromine or iodine atoms. As used herein the term "hydroxyl" or "hydroxy" refers to an -OH group. 10 As used herein the term "cyano" refers to a carbon radical having three of four covalent bonds shared by a nitrogen atom, in particular -C=N. A cyano group may be substituted with substituents described herein. As used herein the term "alkoxy" refers to a linear or branched oxy-containing radical having an alkyl portion of one to about ten carbon atoms, such as a methoxy radical, which may 15 be substituted. In aspects of the invention an alkoxy radical may comprise about 1-10, 1-8 or 1-6 carbon atoms. In embodiments of the invention, an alkoxy radical comprises about 1-6 carbon atoms and includes a C-C 6 alkyl-O-radical wherein C-C 6 alkyl has the meaning set out herein. Examples of alkoxy radicals include without limitation methoxy, ethoxy, propoxy, butoxy, isopropoxy and tert-butoxy alkyls. An "alkoxy" radical may optionally be substituted with one 20 or more substitutents disclosed herein including alkyl atoms to provide "alkylalkoxy" radicals; halo atoms, such as fluoro, chloro or bromo, to provide "haloalkoxy" radicals (e.g. fluoromethoxy, chloromethoxy, trifluoromethoxy, difluoromethoxy, trifluoroethoxy, fluoroethoxy, tetrafluoroethoxy, pentafluoroethoxy, and fluoropropox) and "haloalkoxyalkyl" radicals (e.g. fluoromethoxymethyl, chloromethoxyethyl, trifluoromethoxymethyl, 25 difluoromethoxyethyl, and trifluoroethoxymethyl). As used herein the term "alkenyloxy" refers to linear or branched oxy-containing radicals having an alkenyl portion of about 2 to 10 ten carbon atoms, such as an ethenyloxy or propenyloxy radical. An alkenyloxy radical may be a "lower alkenyloxy" radical having about 2 to 6 carbon atoms. Examples of alkenyloxy radicals include without limitation ethenyloxy, 30 propenyloxy, butenyloxy, and isopropenyloxy alkyls. An "alkenyloxy" radical may be substituted with one or more substitutents disclosed herein including halo atoms, such as fluoro, chloro or bromo, to provide "haloalkenyloxy" radicals (e.g. trifluoroethenyloxy, fluoroethenyloxy, difluoroethenyhloxy, and fluoropropenyloxy).
23 A "carbocylic" includes radicals derived from a saturated or unstaturated, substituted or unsubstituted 5 to 14 member organic nucleus whose ring forming atoms (other than hydrogen) are solely carbon. Examples of carbocyclic radicals are cycloalkyl, cycloalkenyl, aryl, in particular phenyl, naphthyl, norbornanyl, bicycloheptadienyl, tolulyl, xylenyl, indenyl, 5 stilbenyl, terphenylyl, diphenylethylenyl, phenylcyclohexyl, acenapththylenyl, anthracenyl, biphenyl, bibenzylyl, and related bibenzylyl homologs, octahydronaphthyl, tetrahydronaphthyl, octahydroquinolinyl, dimethoxytetrahydronaphthyl and the like. As used herein, the term "cycloalkyl" refers to radicals having from about 3 to 15 carbon atoms and containing one, two, three, or four rings wherein such rings may be attached in a 10 pendant manner or may be fused, in particular cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl, adamantyl, and the like. In certain aspects of the invention the cycloalkyl radicals are "lower cycloalkyl" radicals having from about 3 to 8 carbon atoms, in particular cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl. In some aspects of the invention the term "cycloalkyl" embraces radicals where cycloalkyl radicals are fused with 15 aryl radicals or heterocyclyl radicals. A cycloalkyl radical may be optionally substituted with groups as disclosed herein. As used herein in respect to certain aspects of the invention, the term "cycloaliphatic" refers to a cycloalkane possessing less than 8 carbons or a fused ring system consisting of no more than three fused cycloaliphatic rings. Examples of such include, but are not limited to, 20 decalin and the like. As used herein in respect to certain aspects of the invention, the term "substituted cycloaliphatic" refers to a cycloalkane possessing less than 8 carbons or a fused ring system consisting of no more than three fused rings, and where at least one of the aliphatic hydrogen atoms has been replaced by a halogen, a nitro, a thio, an amino, a hydroxy, a ketone, an 25 aldehyde, an ester, an amide, a lower aliphatic, a substituted lower aliphatic, or a ring (aryl, substituted aryl, cycloaliphatic, or substituted cycloaliphatic). Examples of such include, but are not limited to, 1-chlorodecalyl and the like. A used herein, the term "cycloalkenyl" refers to radicals comprising about 2 to 15 carbon atoms, one or more carbon-carbon double bonds, and one, two, three, or four rings wherein such 30 rings may be attached in a pendant manner or may be fused. In certain aspects of the invention the cycloalkenyl radicals are "lower cycloalkenyl" radicals having three to seven carbon atoms. Examples of cycloalkenyl radicals include without limitation cyclobutenyl, cyclopentenyl, cyclohexenyl and cycloheptenyl. A cycloalkenyl radical may be optionally substituted with 24 groups as disclosed herein, in particular 1, 2, or 3 substituents which may be the same or different. As used herein the term "cycloalkoxy" refers to cycloalkyl radicals attached to an oxy radical. Examples of cycloalkoxy radicals include cyclohexoxy and cyclopentoxy. A 5 cycloalkoxy radical may be optionally substituted with groups as disclosed herein. As used herein, the term "aryl", alone or in combination, refers to a carbocyclic aromatic system containing one, two or three rings wherein such rings may be attached together in a pendant manner or may be fused. The term "fused" means that a second ring is present (i.e, attached or formed) by having two adjacent atoms in common or shared with the first ring. 10 Illustrative "aryl" radicals includes without limitation aromatic radicals such as phenyl, benzyl, naphthyl, indenyl, benzocyclooctenyl, benzocycloheptenyl, pentalenyl, azulenyl, tetrahydronaphthyl, indanyl, biphenyl, acephthylenyl, fluorenyl, phenalenyl, phenanthrenyl, and anthracenyl. An aryl radical may be optionally subsitituted with groups as disclosed herein, in particular hydroxyl, alkyl, carbonyl, carboxyl, thiol, amino, and/or halo, in particular a 15 substituted aryl includes without limitation arylamine and arylalkyamine. As used herein in respect to certain aspects of the invention, the term "substituted aryl" refers to an aromatic ring, or fused aromatic ring system consisting of no more than three fused rings at least one of which is aromatic, and where at least one of the hydrogen atoms on a ring carbon has been replaced by a halogen, an amino, a hydroxy, a nitro, a thio, an alkyl, a ketone, 20 an aldehyde, an ester, an amide, a lower aliphatic, a substituted lower aliphatic, or a ring (aryl, substituted aryl, cycloaliphatic, or substituted cycloaliphatic). Examples of such include, but are not limited to, hydroxyphenyl, chlorophenyl and the like. As used herein, the term "aryloxy" refers to aryl radicals, as defined above, attached to an oxygen atom. Exemplary aryloxy groups include napthyloxy, quinolyloxy, 25 isoquinolizinyloxy, and the like. As used herein the term "arylalkoxy," refers to an aryl group attached to an alkoxy group. Representative examples of arylalkoxy include, but are not limited to, 2-phenylethoxy, 3 naphth-2-ylpropoxy, and 5-phenylpentyloxy. As used herein, the term "aroyl" refers to aryl radicals, as defined above, attached to a 30 carbonyl radical as defined herein, including without limitation benzoyl and toluoyl. An aroyl radical may be optionally substituted with groups as disclosed herein. As used herein the term "heteroaryl" refers to fully unsaturated heteroatom-containing ring-shaped aromatic radicals having at least one heteroatom selected from carbon, nitrogen, sulfur and oxygen. A heteroaryl radical may contain one, two or three rings and the rings may 25 be attached in a pendant manner or may be fused. Examples of "heteroaryl" radicals, include without limitation, an unsaturated 5 to 6 membered heteromonocyclyl group containing 1 to 4 nitrogen atoms, in particular, pyrrolyl, pyrrolinyl, imidazolyl, pyrazolyl, 2-pyridyl, 3-pyridyl, 4 pyridyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazolyl, tetrazolyl and the like; an 5 unsaturated condensed heterocyclic group containing 1 to 5 nitrogen atoms, in particular, indolyl, isoindolyl, indolizinyl, indazolyl, quinazolinyl, pteridinyl, quinolizidinyl, phthalazinyl, naphthyridinyl, quinoxalinyl, cinnolinyl, phenanthridinyl, acridinyl, phenanthrolinyl, phenazinyl, carbazolyl, purinyl, benzimidazolyl, quinolyl, isoquinolyl, quinolinyl, isoquinolinyl, indazolyl, benzotriazolyl, tetrazolopyridazinyl and the like; an unsaturated 3 to 6-membered 10 heteromonocyclic group containing an oxygen atom, in particular, 2-furyl, 3-furyl, pyranyl, and the like; an unsaturated 5 to 6-membered heteromonocyclic group containing a sulfur atom, in particular, thienyl, 2-thienyl, 3-thienyl, and the like; unsaturated 5 to 6-membered heteromonocyclic group containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms, in particular, furazanyl, benzofurazanyl, oxazolyl, isoxazolyl, and oxadiazolyl; an unsaturated condensed 15 heterocyclic group containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms, in particular benzoxazolyl, benzoxadiazolyl and the like; an unsaturated 5 to 6-membered heteromonocyclic group containing 1 to 2 sulfur atoms and 1 to 3 nitrogen atoms, for example, thiazolyl, isothiazolyl, thiadiazolyl and the like; an unsaturated condensed heterocyclic group containing 1 to 2 sulfur atoms and 1 to 3 nitrogen atoms such as benzothiazolyl, benzothiadiazolyl and the 20 like. The term also includes radicals where heterocyclic radicals are fused with aryl radicals, in particular bicyclic radicals such as benzofuranyl, benzothiophenyl, phthalazinyl, chromenyl, xanthenyl, and the like. A heteroaryl radical may be optionally substituted with groups as disclosed herein, for example with an alkyl, amino, halogen, etc., in particular a heteroarylamine. 25 The term "heterocyclic" refers to saturated and partially saturated heteroatom-containing ring-shaped radicals having at least one heteroatom selected from carbon, nitrogen, sulfur and oxygen. A heterocylic radical may contain one, two or three rings wherein such rings may be attached in a pendant manner or may be fused. Examplary saturated heterocyclic radicals include without limitiation a saturated 3 to 6-membered heteromonocylic group containing 1 to 30 4 nitrogen atoms [e.g. pyrrolidinyl, imidazolidinyl, piperidinyl, and piperazinyl]; a saturated 3 to 6-membered heteromonocyclic group containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms [e.g. morpholinyl; sydnonyl]; and, a saturated 3 to 6-membered heteromonocyclic group containing 1 to 2 sulfur atoms and 1 to 3 nitrogen atoms [e.g., thiazolidinyl] etc. Examples of partially saturated heterocyclyl radicals include without limitation dihydrothiophene, 26 dihydropyranyl, dihydrofuranyl and dihydrothiazolyl. Illustrative heterocyclic radicals include without limitation aziridinyl, azetidinyl, 2-pyrrolinyl, 3-pyrrolinyl, pyrrolidinyl, azepinyl, 1,3 dioxolanyl, 2H-pyranyl, 4H-pyranyl, piperidinyl, 1,4-dioxanyl, morpholinyl, pyrazolinyl, 1,4 dithianyl, thiomorpholinyl, 1, 2 ,3,6-tetrahydropyridinyl, oxiranyl, oxetanyl, tetrahydrofuranyl, 5 tetrahydropyranyl, tetrahydropyridinyl, tetrahydrothiopyranyl, thioxanyl, indolinyl, 2H-pyranyl, 4H-pyranyl, dioxanyl, 1,3-dioxolanyl, pyrazolinyl, dihydropyranyl, dihydrothienyl, dihydrofuranyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, 3H-indolyl, quinuclidinyl, quinolizinyl, and the like. As used herein in respect to certain aspects of the invention, the term "heterocyclic" 10 refers to a cycloalkane and/or an aryl ring system, possessing less than 8 carbons, or a fused ring system consisting of no more than three fused rings, where at least one of the ring carbon atoms is replaced by oxygen, nitrogen or sulfur. Examples of such include, but are not limited to, morpholino and the like. As used herein in respect to certain aspects of the invention, the term "substituted 15 heterocyclic" refers to a cycloalkane and/or an aryl ring system, possessing less than 8 carbons, or a fused ring system consisting of no more than three fused rings, where at least one of the ring carbon atoms is replaced by oxygen, nitrogen or sulfur, and where at least one of the aliphatic hydrogen atoms has been replaced by a halogen, hydroxy, a thio, nitro, an amino, a ketone, an aldehyde, an ester, an amide, a lower aliphatic, a substituted lower aliphatic, or a 20 ring (aryl, substituted aryl, cycloaliphatic, or substituted cycloaliphatic). Examples of such include, but are not limited to 2-chloropyranyl. The foregoing heteroaryl and heterocyclic groups may be C-attached or N-attached (where such is possible). As used herein the term "sulfonyl", used alone or linked to other terms such as 25 alkylsulfonyl or arylsulfonyl, refers to the divalent radicals -SO2~. In aspects of the invention a sulfonyl group, the sulfonyl group may be attached to a substituted or unsubstituted hydroxyl, alkyl group, ether group, alkenyl group, alkynyl group, aryl group, cycloalkyl group, cycloalkenyl group, cycloalkynyl group, heterocyclic group, carbohydrate, peptide, or peptide derivative. 30 The term "sulfinyl", used alone or linked to other terms such as alkylsulfinyl (i.e. -S(O) alkyl) or arylsulfinyl, refers to the divalent radicals -S(O)-. As used herein he term "amino", alone or in combination, refers to a radical where a nitrogen atom (N) is bonded to three substituents being any combination of hydrogen, hydroxyl, alkyl, cycloalkyl, alkenyl, alkynyl, aryl, silyl, heterocyclic, or heteroaryl with the general 27 chemical formula -NR 21 R where R 2 ' and R? 2 can be any combination of hydrogen, hydroxyl, alkyl, cycloalkyl, alkoxy, alkenyl, alkynyl, aryl, carbonyl carboxyl, amino, silyl, heteroaryl, or heterocyclic which may or may not be substituted. Optionally one substituent on the nitrogen atom may be a hydroxyl group (-OH) to provide an amine known as a hydroxylamine. 5 Illustrative examples of amino groups are amino (-NH 2 ), alkylamino, acylamino, cycloamino, acycloalkylamino, arylamino, arylalkylamino, and lower alkylsilylamino, in particular methylamino, ethylamino, dimethylamino, 2-propylamino, butylamino, isobutylamino, cyclopropylamino, benzylamino, allylamino, hydroxylamino, cyclohexylamino, piperidinyl, hydrazinyl, benzylamino, diphenylmethylamino, tritylamino, trimethylsilylamino, and dimethyl 10 tert.-butylsilylamino, which may or may not be substituted. As used herein the term "thiol" means -SH. A thiol may be substituted with a substituent disclosed herein, in particular alkyl (thioalkyl), aryl (thioaryl), alkoxy (thioalkoxy) or carboxyl. The term "sulfenyl" used alone or linked to other terms such as alkylsulfenyl, refers to the radical -SR wherein R 24 is not hydrogen. In aspects of the invention R 24 is substituted or 15 unsubstituted alkyl, cycloalkyl, alkenyl, alkynyl, aryl, silyl, silylalkyl, heterocyclic, heteroaryl, carbonyl, carbamoyl, alkoxy, or carboxyl. As used hererin, the term "thioalkyl", alone or in combination, refers to a chemical functional group where a sulfur atom (S) is bonded to an alkyl, which may be substituted. Examples of thioalkyl groups are thiomethyl, thioethyl, and thiopropyl. A thioalkyl may be 20 substituted with a substituted or unsubstitute carboxyl, aryl, heterocylic, carbonyl, or heterocyclic. A thiol may be substituted with a substituted or unsubstituted heteroaryl or heterocyclic, in particular a substituted or unsubstituted saturated 3 to 6-membered heteromonocylic group containing 1 to 4 nitrogen atoms [e.g. pyrrolidinyl, imidazolidinyl, piperidinyl, and piperazinyl] 25 or a saturated 3 to 6-membered heteromonocyclic group containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms [e.g. morpholinyl; sydnonyl], especially a substituted morpholinyl or piperidinyl. As used herein the term "thioaryl", alone or in combination, refers to a chemical functional group where a sulfur atom (S) is bonded to an aryl group with the general chemical 30 formula -SR where R 2 s is aryl which may be substituted. Illustrative examples of thioaryl groups and substituted thioaryl groups are thiophenyl, chlorothiophenyl, para-chlorothiophenyl, thiobenzyl, 4-methoxy-thiophenyl, 4-nitro-thiophenyl, and para-nitrothiobenzyl. As used herein the term "thioalkoxy", alone or in combination, refers to a chemical functional group where a sulfur atom (S) is bonded to an alkoxy group with the general 28 chemical formula -SR30 where R 30 is an alkoxy group which may be substituted. A "thioalkoxy group" may have 1-6 carbon atoms i.e. a -S-(O)-C 1
-C
6 alkyl group wherein C 1
-C
6 alkyl have the meaning as defined above. Illustrative examples of a straight or branched thioalkoxy group or radical having from 1 to 6 carbon atoms, also known as a C 1
-C
6 thioalkoxy, include 5 thiomethoxy and thioethoxy. As used herein, the term "carbonyl" refers to a carbon radical having two of the four covalent bonds shared with an oxygen atom. As used hererin, the term "carboxyl", alone or in combination, refers to -C(O)OR- 1 4 or C(=O)OR1 4 wherein R 1 4 is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, amino, 10 thiol, aryl, heteroaryl, thioalkyl, thioaryl, thioalkoxy, a heteroaryl, or a heterocyclic, which may optionally be substituted. Examples of carboxyl groups are methoxycarbonyl, butoxycarbonyl, tert.alkoxycarbonyl such as tert.butoxycarbonyl, arylmethyoxycarbonyl having one or two aryl radicals including without limitation phenyl optionally substituted by for example lower alkyl, lower alkoxy, hydroxyl, halo, and/or nitro, such as benzyloxycarbonyl, 15 methoxybenxyloxycarbonyl, diphenylmethoxycarbonyl, 2-bromoethoxycarbonyl, 2 iodoethoxycarbonyltert.butylcarbonyl, 4-nitrobenzyloxycarbonyl, diphenylmethoxy-carbonyl, benzhydroxycarbonyl, di-(4-methoxyphenyl-methoxycarbonyl, 2-bromoethoxycarbonyl, 2 iodoethoxycarbonyl, 2-trimethylsilylethoxycarbonyl, or 2-triphenylsilylethoxycarbonyl. Additional carboxyl groups in esterified form are silyloxycarbonyl groups including organic 20 silyloxycarbonyl. In aspects of the invention, the carboxyl group may be an alkoxy carbonyl, in particular methoxy carbonyl, ethoxy carbonyl, isopropoxy carbonyl, t-butoxycarbonyl, t pentyloxycarbonyl, or heptyloxy carbonyl, especially methoxy carbonyl or ethoxy carbonyl. As used herein, the term "carbamoyl", alone or in combination, refers to amino, monoalkylamino, dialkylamino, monocycloalkylamino, alkylcycloalkylamino, and 25 dicycloalkylamino radicals, attached to one of two unshared bonds in a carbonyl group. As used herein, the term "carboxamide" refers to the group -CONH-. As used herein, the term "nitro" means -NO 2 -. As used herein, the term "acyl", alone or in combination, means a carbonyl or thiocarbonyl group bonded to a radical selected from, for example, optionally substituted, 30 hydrido, alkyl (e.g. haloalkyl), alkenyl, alkynyl, alkoxy ("acyloxy" including acetyloxy, butyryloxy, iso-valeryloxy, phenylacetyloxy, benzoyloxy, p-methoxybenzoyloxy, and substituted acyloxy such as alkoxyalkyl and haloalkoxy), aryl, halo, heterocyclyl, heteroaryl, sulfinyl (e.g. alkylsulfinylalkyl), sulfonyl (e.g. alkylsulfonylalkyl), cycloalkyl, cycloalkenyl, thioalkyl, thioaryl, amino (e.g alkylamino or dialkylamino), and aralkoxy. Illustrative examples 29 of "acyl" radicals are formyl, acetyl, 2-chloroacetyl, 2-bromacetyl, benzoyl, trifluoroacetyl, phthaloyl, malonyl, nicotinyl, and the like. As used herein, "ureido" refers to the group "-NHCONH-". A ureido radical includes an alkylureido comprising a ureido substituted with an alkyl, in particular a lower alkyl attached to 5 the terminal nitrogen of the ureido group. Examples of an alkylureido include without limitation N'-methylureido, N'-ethylureido, N'-n-propylureido, N'-i-propylureido and the like. A ureido radical also includes a N',N'-dialkylureido group containing a radical -NHCON where the terminal nitrogen is attached to two optionally substituted radicals including alkyl, aryl, heterocylic, and heteroaryl. 10 The terms used herein for radicals including alkyll", "alkoxy", "alkenyl", "alkynyl", "hydroxyl" etc. refer to both unsubstituted and substituted radicals. The term "substituted," as used herein, means that any one or more moiety on a designated atom (e.g., hydrogen) is replaced with a selection from a group disclosed herein, provided that the designated atom's normal valency is not exceeded, and that the substitution results in a stable compound. 15 Combinations of substituents and/or radicals are permissible only if such combinations result in stable compounds. "Stable compound" refers to a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent. A radical in a compound of the Formula I, II, III, IV, or V may be substituted with one 20 or more substituents apparent to a person skilled in the art including without limitation alkyl, alkoxy, alkenyl, alkynyl, alkanoyl, alkylene, alkenylene, hydroxyalkyl, haloalkyl, haloalkylene, haloalkenyl, alkoxy, alkenyloxy, alkenyloxyalkyl, alkoxyalkyl, aryl, alkylaryl, haloalkoxy, haloalkenyloxy, heterocyclic, heteroaryl, sulfonyl, alkylsulfonyl, sulfinyl, sulfonyl, sulfenyl, alkylsulfinyl, aralkyl, heteroaralkyl, cycloalkyl, cycloalkenyl, cycloalkoxy, cycloalkenyloxy, 25 amino, oxy, halo, azido, thio, =0, =S, cyano, hydroxyl, phosphonato, phosphinato, thioalkyl, alkylamino, arylamino, arylsulfonyl, alkylcarbonyl, arylcarbonyl, heteroarylcarbonyl, heteroarylsulfinyl, heteroarylsulfony, heteroarylamino, heteroaryloxy, heteroaryloxylalkyl, arylacetamidoyl, aryloxy, aroyl, aralkanoyl, aralkoxy, aryloxyalkyl, haloaryloxyalkyl, heteroaroyl, heteroaralkanoyl, heteroaralkoxy, heteroaralkoxyalkyl, thioaryl, arylthioalkyl, 30 alkoxyalkyl, and acyl groups. These substitutents may themselves be substituted. A chemical substituent is "pendant" from a radical if it is bound to an atom of the radical. In this context, the substituent can be pending from a carbon atom of a radical, a carbon atom connected to a carbon atom of the radical by a chain extender, or a heteroatom of the radical.
30 A "disease" that can be treated and/or prevented using a compound, composition, or method of the invention includes a condition associated with or requiring modulation of one or more of inflammation (e.g. neuroinflammation), signaling pathways involved in inflammation (e.g., neuroinflammation), cell signaling molecule production, activation of glia or glial 5 activation pathways and responses, proinflammatory cytokines or chemokines (e.g., interleukin (IL) or tumor necrosis factor (TNF), oxidative stress-related responses such as nitric oxide synthase production and nitric oxide accumulation, acute phase proteins, components of the complement cascade, protein kinase activity (e.g., death associated protein kinase (DAPK) activity), cell damage (e.g., neuronal cell damage), and cell death (e.g., neuronal cell death). In 10 particular a disease is a dementing disorder, a neurodegenerative disorder, a CNS demyelinating disorder, an autoimmune disorder, or a peripheral inflammatory disease. A disease may be characterized by an inflammatory process due to the presence of macrophages activated by an amyloidogenic protein or peptide. Thus, a method of the invention may involve inhibiting macrophage activation and/or inhibiting an inflammatory process. A 15 method may comprise decreasing, slowing, ameliorating, or reversing the course or degree of macrophage invasion or inflammation in a patient. Examples of diseases that can be treated and/or prevented using the compounds, compositions and methods of the invention include Alzheimer's disease and related disorders, presenile and senile forms; amyloid angiopathy; mild cognitive impairment; Alzheimer's 20 disease-related dementia (e.g., vascular dementia or Alzheimer dementia); AIDS related dementia, tauopathies (e.g., argyrophilic grain dementia, corticobasal degeneration, dementia pugilistica, diffuse neurofibrillary tangles with calcification, frontotemporal dementia with parkinsonism, Prion-related disease, Hallervorden-Spatz disease, myotonic dystrophy, Niemann-Pick disease type C, non-Guamanian Motor Neuron disease with neurofibrillary 25 tangles, Pick's disease, postencephalitic parkinsonism, cerebral amyloid angiopathy, progressive subcortical gliosis, progressive supranuclear palsy, subacute sclerosing panencephalitis, and tangle only dementia), alpha-synucleinopathy (e.g., dementia with Lewy bodies, multiple system atrophy with glial cytoplasmic inclusions), multiple system atrophies, Shy-Drager syndrome, spinocerebellar ataxia (e.g., DRPLA or Machado-Joseph Disease); striatonigral 30 degeneration, olivopontocerebellar atrophy, neurodegeneration with brain iron accumulation type I, olfactory dysfunction, and amyotrophic lateral sclerosis); Parkinson's disease (e.g., familial or non-familial); Amyotrophic Lateral Sclerosis; Spastic paraplegia (e.g., associated with defective function of chaperones and/or triple A proteins); Huntington's Disease, spinocerebellar ataxia, Freidrich's Ataxia; cerebrovascular diseases including stroke, hypoxia, 31 ischemia, infarction, intracerebral hemorrhage; traumatic brain injury; Down's syndrome; head trauma with post-traumatic accumulation of amyloid beta peptide; Familial British Dementia; Familial Danish Dementia; Presenile Dementia with Spastic Ataxia; Cerebral Amyloid Angiopathy, British Type; Presenile Dementia With Spastic Ataxia Cerebral Amyloid 5 Angiopathy, Danish Type; Familial encephalopathy with neuroserpin inclusion bodies (FENIB); Amyloid Polyneuropathy (e.g., senile amyloid polyneuropathy or systemic Amyloidosis); Inclusion Body myositis due to amyloid beta peptide; Familial and Finnish Type Amyloidosis; Systemic amyloidosis associated with multiple myeloma; Familial Mediterranean Fever; multiple sclerosis, optic neuritis; Guillain-Barre Syndrome; chronic inflammatory demyelinating 10 polyneuropathy; chronic infections and inflammations; acute disseminated encephalomyelitis (ADEM); autoimmune inner ear disease (AIED); diabetes; myocardial ischemia and other cardiovascular disorders; pancreatitis; gout; inflammatory bowel disease; ulcerative colitis, Crohn's disease, rheumatoid arthritis, osteoarthritis; artheriosclerosis, inflammatory aortic aneurysm; asthma; adult respiratory distress syndrome; restenosis; ischemialreperfusion injury; 15 glomerulonephritis; sacoidosis cancer; restenosis; rheumatic fever; systemic lupus erythematosus; Reiter's syndrome; psoriatic arthritis; ankylosing spondylitis; coxarthritis; pelvic inflammatory disease; osteomyelitis; adhesive capsulitis; oligoarthritis; periarthritis; polyarthritis; psoriasis; Still's disease; synovitis; inflammatory dermatosis; and, wound healing. In aspects of the invention, a compound, composition, or method disclosed herein may be 20 utilized to prevent and/or treat a disease involving neuroinflammation (i.e., neuroinflammatory disease). Neuroinflammation is a characteristic feature of disease pathology and progression in a diverse array of neurodegenerative disorders that are increasing in their societal impact (for a recent review, see, e.g., Prusiner, S. B. (2001) New Engl. J. Med. 344,1516-1526). These neuroinflammation-related disorders include Alzheimer's disease (AD), amyotrophic lateral sclerosis, 25 autoimmune disorders, priori diseases, stroke and traumatic brain injury. Neuroinflammation is brought about by glial cell (e.g., astrocytes and microglia) activation, which normally serves a beneficial role as part of an organism's homeostatic response to injury or developmental change. However, disregulation of this process through chronic or excessive activation of glia contributes to the disease process through the increased production of proinflammatory cytokines and chemokines, 30 oxidative stress-related enzymes, acute phase proteins, and various components of the complement cascades. (See, e.g., Akiyama et al., (2000) Neurobiol. Aging 21,383-421). The direct linkage of glial activation to pathology that is a hallmark of disease underscores the importance of understanding the signal transduction pathways that mediate these critical glial cellular responses and the discovery of cell permeable ligands that can modulate these disease relevant pathways.
32 For Alzheimer's disease (AD) in particular, the deposition of [p-amyloid (AP) and neurofibrillary tangles are associated with glial activation, neuronal loss and cognitive decline. On a molecular level, Alzheimer's disease is characterized by; increased expression of nitric oxide synthase (NOS) in glial cells surrounding amyloid plaques; neuropathological evidence of peroxynitrite 5 mediated neuronal damage; and nitric oxide (NO) overproduction involved in Ap-induced brain dysfunction. NOSH (iNOS) is induced as part of the glial activation response and is an oxidative stress-related enzyme that generates NO. When NO is present in high levels along with superoxide, the highly reactive NO-derived molecule peroxynitrite is generated, leading to neuronal cell death. The pro-inflammatory cytokine IL-16 is also overexpressed in activated glia in AD brain and 10 polymorphisms in IL-10 genes are associated with an increased risk of early onset sporadic AD (See, e.g., Du et al., (2000) Neurology 55, 480-483). IL-1s0 can also influence amyloid plaque development and is involved in additional glial inflammatory and neuronal dysfunction responses (See, e.g., Griffin, et al., (1998) Brain Pathol. 8, 65-72; and Sheng, et al., (1996) Neurobiol. Aging 17, 761-766). Therefore, because glial activation and specific glial products are associated with neurodegenerative 15 disorders (e.g., Alzheimer's disease), the compounds and compositions disclosed herein that are capable of modulating cell signaling pathways (e.g., glial activation pathways) will have particular application in the treatment and prevention of inflammatory disease. In aspects of the invention, a compound, composition, or method disclosed herein may be utilized to prevent and/or treat a disease involving disregulation of protein kinase signaling. 20 Disregulation of protein kinase signaling often accompanies disregulation of cell signaling pathways (e.g., glial cell activation pathways). Protein kinases are a large family of proteins that play a central role in regulating a number of cellular functions including cell growth, differentiation and death. There are thought to be more than 500 protein kinases and 130 protein phosphatases exerting tight control on protein phosphorylation. Each protein kinase transfers the 25 y-phosphate of ATP to a specific residue(s) of a protein substrate. Protein kinases can be further categorized as tyrosine, serine/threonine or dual specific based on acceptor residue. Examples of serine/threonine kinases include MAP kinase, MAPK kinase (MEK), Akt/PKB, Jun kinase (INK), CDKs, protein kinase A (PRA), protein kinase C (PKC), and calmodulin (CaM) dependent kinases (CaMKs). Disregulated protein kinase activity (e.g., hyper- or hypo-active) 30 leads to abnormal protein phosphorylation, underlying a great number of diseases including diabetes, rheumatoid arthritis, inflammation, hypertension, and proliferative diseases such as cancer. Therefore, because aberrant kinase activity is associated with inflammatory disease (e.g., neurodegenerative disorders like Alzheimer's disease), the compounds and compositions that are 33 disclosed herein that are capable of modulating inases involved in cell signaling pathways will have particular application for treatment and prevention of inflammatory disease. Compounds The invention provides an isolated and pure, in particular, substantially pure, compound 5 of the Formula I wherein R', R 2 , and R 3 are independently hydrogen, hydroxyl, alkyl, alkenyl, alkynyl, alkylene, alkenylene, alkoxy, alkenyloxy, cycloalkyl, cycloalkenyl, aryl, aryloxy, arylalkoxy, aroyl, heteroaryl, heterocyclic, acyl, acyloxy, sulfonyl, sulfinyl, sulfenyl, amino, imino, azido, thiol, thioalkyl, thioalkoxy, thioaryl, nitro, ureido, cyano, halo, silyl, silyloxy, silylalkyl, silylthio, =0, =S, carboxyl, carbonyl, carbamoyl, or carboxamide;or R 7 is hydrogen, 10 hydroxyl, alkyl, alkenyl, alkynyl, alkylene, alkenylene, alkoxy, alkenyloxy, cycloalkyl, cycloalkenyl, aryl, aryloxy, arylalkoxy, aroyl, heteroaryl, heterocyclic, acyl, acyloxy, sulfonyl, sulfinyl, sulfenyl, amino, imino, azido, thiol, thioalkyl, thioalkoxy, thioaryl, nitro, ureido, cyano, halo, silyl, silyloxy, silylalkyl, silylthio, =0, =S, carboxyl, carbonyl, carbamoyl, or carboxamide; R 4 , R, and R' are independently hydrogen, alkyl, alkoxy, halo, or nitro or R7 15 may be absent with a double bond between N at position 1 and C at position 6; or R' and R 2 , R' and R 7 , or R 2 and R 3 may form a heteroaryl or heterocyclic ring; or an isomer or a pharmaceutically acceptable salt thereof. In some aspects, one or more of the following compounds do not fall within the scope of the present invention: 20 In some aspects, one or more of the following compounds do not fall within the scope of the present invention: a) a compound wherein when R1 is =0, R 3 is -COOCH 3 , CH=CHCOOCH 3 , CH=CHC(=O)-phenyl, -CH=CH(C(=0)OCH 3
)
2 , -S-phenyl, CH=CH(COCH 3
)(COOCH
3 ), CH=CH(COOCH 2
CH
3
)
2 , -phenyl-COOCH 3 , -CH=CHCO 25 phenyl, -CH 2 CH(Cl)(CH 2 OH), -methylphenyl, R 7 is hydrogen or -CH 2 0CH 3 , and R',
R
2 , R 4 , R' and R 6 are hydrogen; b) a compound wherein when R' is =0, R 2 is cyano, R 3 is -C(=O)OCH 3 , and R 3 , R 4 , Rs, and R are hydrogen; c) a compound wherein when R 1 is =0, R 2 is -methylthiophene or benzyl, R , R 4 , Rs, R 6 , 30 and R 7 are hydrogen; d) a compound wherein when R1 is =0, R 2 is methyl, R 5 is hydrogen, hydroxyl, chloro, or bromo, R 7 is hydrogen or ethylmorpholinyl, and R 3 , R 4 , and R 6 are hydrogen; e) a compound wherein when R 2 is methyl, R 5 is chloro, bromo, or hydrogen, R 7 is hydrogen or -CH 2
CH
2 -morpholinyl, and R 1 , R 3 , R 4 , and R 6 are hydrogen; 34 f) a compound wherein when R' is piperazinyl, piperazinyl substituted with pyridinyl, phenyl, or methyl, R 2 is hydrogen or methyl, and R 3 , R 4 , R, and R are hydrogen; g) a compound wherein when R 1 is chloro or bromo, R 2 is C 1
-C
3 alkyl, phenyl, amino, benzyl, morpholinyl, chloro, -C(=O)NH 2 , -NH 2 , C 1
-C
3 alkylphenyl, -CH(CH 3
)
2 , 5 CH 2
CH(CH
3
)
2 , -benzylchloro, and R 3 , R 4 , R 5 and R 6 are hydrogen; h) a compound wherein when R is chloro or bromo, R 3 is hydroxyl, chloro, bromo, C 1
-C
3 alkyl, phenyl, or -N(CH 3
)
2 , and R 2 , R 4 , R 5 and R 6 are hydrogen; i) a compound wherein when R' is chloro, R 2 is methyl, R 5 is hydroxyl, and R 3 , R 4 , and R 6 are hydrogen; 10 j) a compound wherein when R' is chloro, R 2 , R 3 , R 4 , R 5 and R 6 are hydrogen; k) a compound wherein when R' is hydroxyl, R 2 is C1-C4 alkyl, and R 3 , R 4 , R' and R 6 are hydrogen; 1) a compound wherein when R' is -C 1
-C
4 alkoxy, or C 1
-C
4 alkoxy substituted with N(CH 3
)
2 , morpholinyl, or piperidinyl substituted with benzyl, R 2 is hydrogen or methyl, 15 R 3 , R 4 , R and R 6 are hydrogen, R 7 is absent, hydrogen, or methyl; m) a compound wherein when R 1 is -SH, -SCH 3 , or -SCH 2
C(=O)CH
3 , R 2 is hydrogen or methyl, and R 3 , R 4 , R and R 6 are hydrogen; n) a compound wherein when R' is =S, R 2 is hydrogen or methyl, R 7 is methyl or benzyl, and R 3 , R 4 , and R 6 are hydrogen; 20 o) a compound wherein when R' is =S, R 2 is methyl and R 5 is chloro or R 7 is methyl, and
R
3 , R 4 , and R 6 are hydrogen; p) a compound wherein when R is hydroxyl, R 2 is hydrogen, methyl, or butyl, and R 3 , R 4 ,
R
5 and R 6 are hydrogen; q) a compound wherein when R is methoxy, R 2 , R 3 , , R R 5 and R 6 are hydrogen; 25 r) a compound wherein when R' is C 1
-C
2 alkoxy or C 1
-C
4 alkoxy substituted with morpholinyl,
-N(CH
3
)
2 , or piperidinyl substituted with benzyl, R 2 is methyl, and R 3 , R 4 , R' and R 6 are hydrogen; s) a compound wherein R 2 , R 3 , R 4 , R 5 and R 6 are hydrogen; t) a compound wherein R 1 is cyano or cyano substituted with -C(OCH 2
CH
3
)
2 , M0 CH(OH)(CH 3 ), -Si(CH 2
CH
3
)
2 , cyclohexol, -CH20-trimethyldiphenylsilyl or cyclohexyl substituted with hydroxyl, and R 3 , R 4 , R 5 and R 6 are hydrogen; u) a compound wherein R 1 is cyano substituted with -CH(OH)(CH 3
)
2 , -Si(CH 2
CH
3
)
2 , morpholinyl, trimethyldiphenylsilyl, or -CH(OCH 2
CH
3
)
2 , R 2 is methyl, and R 3 , R 4 , Rs and R 6 are hydrogen; 35 v) a compound wherein R 7 is oxy, and R 2 is hydrogen or methyl, and R 3 , R 4 , R 5 and R 6 are hydrogen; w) a compound wherein R' is methyl, and R 2 , R 3 , R 4 , R' and R 6 are hydrogen; x) a compound wherein R 2 is methyl, and R', R 3 , R 4 , R 5 and R 6 are hydrogen; 5 y) a compound wherein R' is methoxycarbonyl,
R
3 is hydrogen, and R 2 , R 4 , R 5 and R 6 are hydrogen; z) a compound wherein R' is -NH 2 , R 2 is methyl, chlorophenyl, methoxyphenyl, ethylphenyl, ethylmethoxyphenyl, propylphenyl, or -CH(CH 3
)
2 , R 4 , R 5 and R 6 are hydrogen, and R 7 is absent or -CH 2
CH
2
CH
2 COOH; 10 aa) a compound wherein R' is -OR 0 wherein R 80 is ethylmorpholinyl or -CH 2
CH
2
N(CH
3
)
2 and R 2 , R 3 , R 4 , Rs and R 6 are hydrogen; bb) a compound wherein R 1 is -NH 2 , R 3 is -NH 2 , and R 3 , R 4 , R' and R 6 are hydrogen; cc) a compound wherein R1 is -NH 2 , R 5 and R 6 are methoxy, and R 3 and R 4 are hydrogen; dd) a compound wherein R 1 is -NH 2 , R 3 is methyl and R 4 , R 5 and R 6 are hydrogen; 15 ee) a compound wherein R 1 is -NH 2 , R 5 is chloro, and R , R 4 and R 6 are hydrogen; ff) a compound wherein R' is -NH-chlorophenyl, and R 2 and R 3 form a phenyl group, and
R
4 , R 5 and R 6 are hydrogen; gg) a compound wherein R 1 is -NH 2 , R 4 and R 5 is methoxy, and R 2 , R 3 and R 6 are hydrogen; hh) a compound wherein R 1 is -NH 2 , R 2 is ethylmethoxyphenyl,
R
7 is carboxyethyl or 20 carboxypropyl, and R 3 , R 4 and R 6 are hydrogen; ii) a compound wherein R' is -NHR 22 wherein R 22 is ethylmorpholinyl, ethylmorpholinyl substituted with =0, -CH 2
CH
2 0CH 3 , -CH 2
CH
2
CH
2
CH
2
CH
2
CH
2
CH
2
CH
3 , CH 2
CH
2
CH
2
CH
2 OH, -CH 2
CH
2 OH, or -CH 2
CH
2 0CH 3 , R 2 is hydrogen, methyl, ethyl, CHO, -CH 2 OH, -COOH, chloro, -CH 2
CH
2
NH
2 , -NO 2 , -C=N, -C(=O)OCH 2
CH
3 , or 25 C(=O)NH 2 , and R 3 , R 4 , R 5 and R 6 are hydrogen; jj) a compound wherein R' is -NHR2 wherein R 22 is ethanol, methylpiperidinylbenzyl, ethylpiperidinyl, ethylpiperidinylbenzyl, or butylpiperidinylbenzyl,
R
2 is hydrogen, methyl, or -C(CH 3
)
2 , and R 3 , R 4 , R 5 and R 6 are hydrogen; kk) a compound wherein R 1 is -NHDR 2 wherein R 22 is hydrogen, and R 3 , R 4 , R' and R 6 are 30 hydrogen; 11) a compound wherein R 1 is -NHR 2 wherein R 22 is -CH 2
CH
2
N(CH
2
CH
3
)
2 or ethylmorpholinyl,
R
3 is ethyl, and R 4 , R' and R 6 are hydrogen; 36 mm) a compound wherein R' is -NHNH 2 , R 3 is hydrogen, alkyl, or phenyl, and R 3 ,
R
4 , R and R 6 are hydrogen; nn) a compound wherein R' is -NHR 2 wherein R 22 is NH 2 , -CH 2
CH
2 OH,
CH
2
CH(OH)(CH
3 ), ethylmorpholinyl, ethylmorpholinyl substituted with =0, 5 ethylphenyl, -CH 2
CH
2
NHCH
3 , -CH 2
CH
2
N(-CH
2
CH
2
CH
3
)
2 , ethylpiperidinyl, or ethylpiperidinylbenzyl, R 2 is methyl, and R 3 , R 4 , R 5 and R are hydrogen; oo) a compound wherein R' is morpholinyl, R 2 is -C(F) 3 , -C(=O), -CH 2 OH, -C(=O)H, COOH, chloro, -NO 2 , or cyano, and R 3 , R 4 , R 5 and R 6 are hydrogen; pp) a compound wherein R is -NHR 2 2 wherein R 22 is heptyl, phenyl, benzyl, or 10 ethylphenyl, R 2 is hydrogen, methyl, or chlorophenyl, R 4 , R 5 and R are hydrogen; qq) a compound wherein R 1 is -NR 2 1 wherein R 2 1 is phenyl and R 2 , R 3 , R 4 , R 5 and R 6 are hydrogen; rr) a compound wherein R is morpholinyl and R 2 , R 3 , R 4 , R 5 and R 6 are hydrogen; ss) a compound wherein R' is methylpiperazinyl and R 2 , R 3 , R 4 , R 5 and R 6 are hydrogen; 15 tt) a compound wherein R is -NHCH 2
CH
2 OH or NHCH 2
CH
2 0CH 3 , R 2 is phenyl and R 3 ,
R
4 , R 5 and R 6 are hydrogen; uu) a compound wherein R is -NHR 2 wherein R 22 is ethylamino, butylamino, ethylaminomethyl, and R 2 is hydrogen, methyl, or -C(=O)NH 2 , and R 3 , R 4 , RI and R 6 are hydrogen; 20 vv) a compound wherein R' is is -NHR 2 wherein R2 is ethylpiperidinyl, methylpiperidinylbenzyl, piperidinylbenzyl, ethylpiperidinylbenzyl, methylpyrrolidinylmethyl, ethylpiperazinylbenzyl, -CH2C(=O)-piperazinylbenzyl, C(=O)-methylnaphthyl,
-CH
2
CH
2
CH
2
CH
2
CH
2
N(CH
3
)(C
7 7 ), -CH2C(=O) piperidinylbenzyl, -C(=O)-methylpiperidinylbenzyl, or -CH(CH 3
)
2 , and R 3 , R 4 , R 5 and 25 R6 are hydrogen; ww) a compound wherein R' is -CHCH 2
CH
2 -isoquinolinyl, NHCH 2
CH
2
N(CH
2
CH
2
CH
3
)
2 , propyl substituted with piperidinyl fused to phenyl, NHCH 2
CH
2 , or -NHCH 2
CH
2
CH
2
CH
2
CH
2 substituted with a piperidinyl fused to two adjacent carbon atoms of a phenyl moiety; 30 xx) a compound wherein R' is -NH substituted with two pyrrolidinyl groups; R 3 is methyl, and R 2 , R 4 , R 5 and R 6 are hydrogen; yy) a compound wherein R' is -COOCH 3 , R 3 is methyl, and R 2 , R 4 , R 5 and R are hydrogen; zz) a compound wherein R 1 is hydrogen, R 2 is methyl, R 7 is oxygen; 37 aaa) a compound wherein R 7 is methyl or oxygen, and R', R 2 , R 4 , R 5 and R 6 are hydrogen; bbb) a compound wherein R 1 is -NHCH 2
CH
2
N(CH
2
CH
3
)
2 , R 3 is ethyl, and R 2 , R 4 , R 5 and R 6 are hydrogen; and 5 ccc) a compound wherein R'is -NHCH 2
CH(OH)(CH
3 ) or NHCH 2
CH
2
NHCH
2
CH
2 OH, R 2 is methyl, and R 3 , R 4 , R and R 6 are hydrogen. In an aspect, a compound of the Formula I is provided wherein: (a) R 1 is optionally substituted halo, hydroxyl, alkyl, alkenyl, alkoxy, cyano, amino, cycloalkyl, -sulfonyl, sulfinyl, sulfenyl, thioaryl, thioalkyl, carbonyl, silyl, piperazinyl, piperidinyl, pyrrolidinyl, morpholinyl, 10 SR24 wherein R 2 A is optionally substituted alkyl, carbonyl, carboxyl, carbamoyl, aryl, heterocylic, or heteroaryl; (b) R 2 is optionally substituted halo, hydroxyl, alkyl, alkenyl, alkoxy, carbonyl, carboxyl, phenyl, benzyl, amino, aryl, cyano, -COH, piperazinyl, alcohol, piperidinyl, morpholinyl, or naphthyl;(c) R 3 is hydrogen, halo, hydroxyl, alkyl, alkenyl, alkoxy, phenyl, piperazinyl, piperidinyl, pyrrolidinyl, morpholinyl, thiol, sulfenyl, sulfonyl, sulfinyl, or nitro; (d) 15 R 4 is hydrogen, halo, or nitro (e) Rs is hydrogen, halo, alkoxy, or amido;(f) R 7 is hydrogen halo, hydroxyl, alkyl, alkenyl, alkoxy, carboxy, morpholino; imidazolyl; piperazinyl, piperidinyl, pyrrolidinyl, morpholinyl or R 7 is absent and there is a double bond between N at position 1 and C at position 6; and/or (g) R' and R 2 , R' and R 7 or R 2 and R 3 may form a heteroaryl or heterocyclic ring. 20 In another aspect of the invention a compound of the Formula I is provided wherein R' is Cl or Br, -NH 2 , alkyl, -CN, =S, silyl, sulfonyl, thioalkyl, thioaryl, piperazinyl, piperazinyl, piperidinyl, piperidinyl, morpholinyl, pyrrolidinyl, pyrrolyl, or pyrrolidinyl, which may be optionally substituted with halo, =0, alkoxy, alkenyl, alkyl, substituted alkyl, -CN, -SR24 wherein R24 is optionally substituted methyl, ethyl, phenyl, heterocylic, or heteroaryl, or -CO 25 substituted with phenyl or substituted phenyl. In another aspect of the invention a compound of the Formula I is provided wherein R 2 is carbonyl, piperazinyl, morpholinyl, sulfonyl, sulfinyl, sulfenyl, or phenyl, -CN, -COH,
CH
2 OH, -OCH 2
CH
3 , or alkyl which may be optionally substituted with alkyl, alkoxy, amino, halo, phenyl, substituted phenyl, benzyl, hydroxyl, amino, piperidinyl, or morpholinyl. 30 In another aspect of the invention a compound of the Formula I is provided wherein R 3 is piperazinyl; substituted piperzinyl; alkyl which may optionally be substituted with amino; phenyl; substituted phenyl; amino which may be optionally substituted with alkyl or alkylamine (e.g., NHCOOC(CH 3
)
3 ), carboxyl, or substituted carboxyl; hydroxyl; or nitro.
38 In another aspect of the invention a compound of the Formula I is provided wherein R4 is nitro or hydrogen. In another aspect of the invention a compound of the Formula I is provided wherein R 5 is hydrogen, halo, -OCH 2
CH
2
CH
2
NHCOOC(CH
3
)
3 , or -OCH 3 . 5 In another aspect of the invention a compound of the Formula I is provided wherein R 7 is alkyl, morpholinyl, benzyl, imidazolyl, -CH 2
COOCH
2
CH
3 , CH 2
C=COOCH
2
CH
3 ,
CH
2
CH
2
CH
2
SO
2 OH, CH 2
CH
2
CH
2
SO
3 ~, CH 2
CH
2
CH
2
CH
2
PO(OH)
2 , or CH 2
CH
2
CH
2
PO(OH)
2 . In another aspect of the invention a compound of the Formula I is provided wherein R 7 is absent and there is a double bond between N at position 1 and C at position 6. 10 In a further aspect, a compound of the Formula I is provided wherein R', R 2 , R , and R 7 are independently substituted aliphatic, lower alkyl substituted amino, lower alkyl substituted halogen, cycloaliphatic, or substituted cycloaliphatic. In a still further aspect the invention a compound of the Formula I is provided wherein R' is a piperazinyl which may be substituted (e.g., with a pyrimidinyl moiety); halo; amino 15 which may be substituted; cyano; - SR 2 4 wherein R24 is alkyl or aryl (e.g. phenyl) which may be substituted (e.g., halo); substituted alkyl [e.g., alkyl substituted with halogen, such as CH(Br) 2 ]; morpholinyl; pyrrolyl which may be substituted; hydroxyl; - ORs wherein R 3 " is alkyl; -C=
CHR
3 6 wherein R 3 6 is alkyl; or pyrrolidinyl. In a still further aspect the invention a compound of the Formula I is provided wherein 20 R 2 is hydrogen; morpholinyl; piperazinyl which may be substituted (e.g., with a pyrimidinyl moiety); phenyl; alkyl; alkoxy (e.g. CH(OCH 3
)
2 ); substituted alkyl; substituted aryl (e.g., phenyl); cyano; or hydroxyl. In a still further aspect the invention a compound of the Formula I is provided wherein
R
3 is hydrogen; hydroxyl; alkyl which may be substituted (e.g., halo); amino which may be 25 substituted; -COR 3 7 wherein R 3 7 is hydrogen, hydroxyl, alkoxy (e.g. -OCH 3 ); or, aryl (e.g. phenyl) which may be substituted (e.g., alkyl). In a still further aspect the invention a compound of the Formula I is provided wherein R4 is hydrogen or halo; R5 is hydrogen or halo; R 6 is hydrogen or halo. In a still further aspect the invention a compound of the Formula I is provided wherein 30 R 7 is hydrogen; alkyl which may be substituted (e.g. with phenyl); -CH 2
CH
2 COOR' wherein
R
3 8 is alkyl, -CH 2 C=COOR' wherein R 38 is alkyl, CH 2
CH
2
CH
2
S(O)
2 0H, morpholinyl, benzyl, imidazolyl, or [CH 2 ]nPO(OH) 2 wherein n is 1 to 6, in particular 3 or 4. In a still further aspect the invention a compound of the Formula I is provided wherein R1 and R 2 form a piperidinyl ring which may optionally be substituted with a carboxyl.
39 In a still further aspect the invention a compound of the Formula I is provided wherein R' and R 7 form a pyrimidinyl ring which may optionally be substituted with alkyl, aryl, halo, or hydroxyl. In a particular aspect R' is -NR 2 1
R
22 wherein R 2 1 is hydrogen, and R 22 is hydrogen, 5 alkyl, carbonyl, aryl, amino, cycloalkane, heterocylic, or heteroaryl which may be substituted. In embodiments R 2 2 may comprise or be selected from the group consisting of hydrogen, C-C 6 alkyl (e.g. methyl or ethyl) which may be substituted with optionally substituted hydroxyl, alkyl, amino, carbonyl, carboxyl, morpholinyl, isoquinolinyl, or an amino which may be substituted with one or more of optionally substituted alkyl, benzyl, carboxyl, alcohol group, heteroaryl or 10 heterocyclic, a propanol group, phenyl which may be optionally substituted with halo, benzyl which may be substituted with alkoxy, cyclohexyl, piperidinyl which may be substituted with optionally substituted phenyl, pyrrolidinyl or pyrrolidinylalkyl which may be substituted with alkyl, -COOR40 wherein R'O is alkyl which may be substituted, or [CH 2 ]m-piperidinyl wherein m is 1 to 4, in particular 1 to 3 and the piperidinyl is optionally substituted with optionally 15 substituted alkyl, phenyl, or benzyl. In embodiments, R' is -R6 R6 wherein R60 is -NH[CH 2 ]wNH wherein w is 1 to 4, in particular 2 or 3, and R 5 1 is piperazinyl substituted with pyrimidinyl which may be substituted, in particular substituted with alkyl. In embodiments, R 2 ' is - 2
R
6 wherein R 62 is -[CH2]wN(CH 3 ) wherein w is 1 to 4, in 20 particular 2 or 3, and R 63 is piperazinyl substituted with pyrimidinyl which may be substituted, in particular substituted with alkyl. In an aspect of the invention, a compound of the Formula I is provided wherein R' is halo especially chloro or bromo, R 2 is alkyl which may be substituted, in particular substituted with alkoxy (e.g., methoxy, dimethoxy), substituted aryl which may be substituted with alkyl, 25 alkoxy, (e.g., benzyl, methoxy phenyl), halo (e.g. bromo or chIoro), or carbonyl, a substituted or unsubstituted saturated 3 to 6-membered heteromonocylic group containing 1 to 4 nitrogen atoms [e.g., piperidinyl, and piperazinyl] or a saturated 3 to 6-membered heteromonocyclic group containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms [e.g. morpholinyl; sydnonyl], in particular a substituted morpholinyl, piperazinyl,or piperazinyl substituted with a heteroaryl in 30 particular an unsaturated 5 to 6 membered heteromonocyclyl group containing 1 to 4 nitrogen atoms, in particular, pyrrolyl, pyrrolinyl, imidazolyl, pyrazolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, pyridinyl, pyrimidinyl, pyrazinyl, or pyridazinyl, especially pyrimidinyl, and optionally R 3 , R 4 , R', R 6 , and R 7 are hydrogen.
40 In another aspect of the invention, a compound of the Formula I is provided wherein R' is halo especially chloro or bromo, and R 3 is a substituted or unsubstituted saturated 3 to 6 membered heteromonocylic group containing 1 to 4 nitrogen atoms [e.g., piperidinyl, and piperazinyl] or a saturated 3 to 6-membered heteromonocyclic group containing 1 to 2 oxygen 5 atoms and 1 to 3 nitrogen atoms [e.g. morpholinyl; sydnonyl], in particular a substituted morpholinyl, piperazinyl,or piperazinyl substituted with alkyl or a heteroaryl in particular an unsaturated 5 to 6 membered heteromonocyclyl group containing 1 to 4 nitrogen atoms, in particular, pyrrolyl, pyrrolinyl, imidazolyl, pyrazolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, pyridinyl, pyrimidinyl, pyrazinyl, or pyridazinyl, especially pyrimidinyl, or R 2 is a substituted amino, in .0 particular amino substituted with alkyl or substituted alkyl, in particular alkyl substituted with alkoxy carbony, and optionally R 2 , R 4 , R 5 , R 6 , and R 7 are hydrogen. In further aspect R' is halo, especially bromo or chloro, and R 2 and R 3 form an unsaturated ring, in particular phenyl, R 5 is a heteroaryl, in particular a substituted or unsubstituted unsaturated 5 to 6 membered heteromonocyclyl group containing 1 to 4 nitrogen 15 atoms, in particular, pyrrolyl, pyrrolinyl, imidazolyl, pyrazolyl, especially imidazolyl, and optionally R 4 , R 6 and R 7 are hydrogen. In a further aspect, R' is halo, especially bromo or chloro, and R' is nitro, and optionally R2, R 3 , R 5 , R, and R 7 are hydrogen. In a further aspect, the invention provides a compound of the Formula I wherein R1 is a 20 thiol substituted with alkyl (thioalkyl); substituted alkyl, in particular alkyl substituted with a substituted or unsubstituted saturated 3 to 6-membered heteromonocylic group containing 1 to 4 nitrogen atoms [e.g. pyrrolidinyl, imidazolidinyl, piperidinyl, and piperazinyl] or a saturated 3 to 6-membered heteromonocyclic group containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms [e.g. morpholinyl; sydnonyl], especially a substituted morpholinyl or piperidinyl; aryl; 25 substituted aryl; carboxyl which may be substituted with substituted or unsubstituted aryl; optionally R 2 is alkyl, in particular lower alkyl; optionally R 3 is alkyl, in particular lower alkyl or nitro; optionally R 5 is alkoxy; optionally R 7 is alkyl; and optionally R 4 , R 5 , and R 6 , are hydrogen. In a further aspect of the invention, a compound of the Formula I is provided wherein R 1 30 is =S, and optionally R 2 is alkyl, in particular lower alkyl, R' is alkoxy, and R 3 , R 4 , R 6 and R 7 are hydrogen. In a further aspect of the invention, a compound of the Formula I is provided wherein R1 is sulfonyl which may be substituted with substituted or unsubstituted aryl, in particular substituted phenyl, and optionally R 2 is alkyl and R 3 , R 4 , R 5 , R 6 , and R 7 are hydrogen.
41 In a further aspect of the invention, a compound of the Formula I is provided wherein R 1 is substituted or unsubstituted alkyl or ailcynyl, in particular alkyl substituted with aryl, substituted aryl, halo, cyano, or alkynyl substituted with alkyl; and optionally R 2 is alkyl, R is alkyl, and R 3 , R 4 , R-, and R' are hydrogen. 5 In a further aspect of the invention, a compound of the Formula I is provided wherein R 1 is cyano and R2 is aryl or alkyl, and optionally R 3 , R 4 , R 5 , R 6 , and R 7 are hydrogen. In a further aspect of the invention, a compound of the Formula I is provided wherein one or both of R 1 and R 2 are a saturated 3 to 6-membered heteromonocyclic group containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms [e.g. morpholinyl; sydnonyl], especially a 10 substituted morpholinyl, and optionally R 3 , R 4 , R 5 , R 6 , and R 7 are hydrogen. In a further aspect of the invention, a compound of the Formula I is provided wherein R' is a saturated 3 to 6-membered heteromonocylic group containing 1 to 4 nitrogen atoms [e.g. pyrrolidinyl], which may be substituted with substituted or unsubstituted carboxyl; R 2 is alkyl or halo, and optionally
R
3 , R 4 , R , R 6 , and R 7 are hydrogen. L5 In a further aspect of the invention, a compound of the Formula I is provided wherein R" is hydroxyl;
R
2 is alkyl or substituted alkyl or R 3 is a saturated 3 to 6-membered heteromonocylic group containing 1 to 4 nitrogen atoms [e.g. piperidinyl, and piperazinyl] which may optionally be substituted with a heteroaryl [e.g., pyrimidinyl], and the other of R 2 or
R
3 is hydrogen, and optionally R 4 , Rs, R, and R 7 are hydrogen. 0 In a further aspect of the invention, a compound of the Formula I is provided wherein R' is a saturated 3 to 6-membered heteromonocylic group containing 1 to 4 nitrogen atoms [e.g., piperidinyl and piperazinyl] which may be substituted with carboxyl or carboxyl substituted with alkyl or alkoxy or with purinyl or substituted purinyl; R 2 is alkyl or substituted alkyl, in particular alkylaryl, and optionally R , R 4 , R', R', and R 7 are hydrogen. 5 In a further aspect of the invention, a compound of the Formula I is provided wherein R1 is =0, and R 2 is alkyl, alkylaryl, cyano, alkoxy, or substituted alkoxy, and optionally
R
3 , R 4 , R 5 ,
R
6 , and R 7 are hydrogen. In a further aspect of the invention, a compound of the Formula I is provided wherein R1 is alkoxy, R 2 is alkyl, substituted alkyl, or alkoxy, and optionally
R
3 , R 4 , R 5 , R 6 , and R 7 are 0 hydrogen. In a further aspect of the invention, a compound of the Formula I is provided wherein R' and R 2 form a heterocyclic, in particular a saturated 3 to 6-membered heteromonocylic group containing 1 to 4 nitrogen atoms, in particular a 6-membered ring comprising 1 or 2 nitrogen 42 atoms [e.g., piperidinyl and piperazinyl] which may be substituted for example with alkyl, halo, carboxyl, or alkoxy carbonyl, and optionally R 3 , R 4 , R 5 , R 6 , and R 7 are hydrogen. In a further aspect of the invention, a compound of the Formula I is provided wherein R, and R 7 form a heteroaryl, in particular an unsaturated 5 to 6 membered heteromonocyclyl group 5 containing 1 to 4 nitrogen atoms, in particular, pyrrolyl, pyrrolinyl, imidazolyl, pyrazolyl, 2 pyridyl, 3-pyridyl, 4-pyridyl, pyridinyl, pyrimidinyl, pyrazinyl, or pyridazinyl, R 2 is hydrogen or alkyl, and R 3 , R 4 , R 5 , R , and R 7 are hydrogen. In a further aspect of the invention, a compound of the Formula I is provided wherein R, is silyl which may be substituted, in particular substituted with alkyl, R 2 is alkyl, and R', R 4 , R 5 , 10 R 6 , and R 7 are hydrogen. In an embodiment,
R
1 is a piperazinyl or substituted piperazinyl, in particular a piperazinyl substituted with a pyrimidinyl of the Formula II below. 0 RP \/N N N--\N N-N N 15 Thus, the invention also provides an isolated and pure, in particular, substantially pure, compound of the Formula II wherein one or both of R1 0 and R" are independently substituted or unsubstituted hydrogen, hydroxyl, alkyl, alkenyl, alkynyl, alkylene, alkenylene, alkoxy, alkenyloxy, cycloalkyl, cycloalkenyl, aryl, aryloxy, arylalkoxy, aroyl, heteroaryl, heterocyclic, 20 acyl, acyloxy, sulfonyl, sulfinyl, sulfenyl, amino, imino, azido, thiol, thioalkyl, thioalkoxy, thioaryl, nitro, ureido, cyano, halo, silyl, silyalkyl, silyloxy, silylthio, =0, =S, carboxyl, carbonyl, or carbamoyl, or an isomer or a pharmaceutically acceptable salt thereof. In an aspect of the compound of the Formula II is provided wherein R1 0 is hydrogen; hydroxyl; alkyl; aryl [e.g. phenyl which is optionally substituted (e.g., halide)]; piperazinyl 25 which may be substituted (e.g. substituted with a pyrimidinyl);
-NR
55 Rs 6 wherein R 5 is hydrogen or alkyl, and R 5 6 is phenyl which may be substituted or alkyl which may be substituted (e.g. amino, in particular - CH 2
CH
2
NH
2 ; CH 2
CH
2
NHCOOC(CH
3
)
3 ); morpholinyl which may be substituted; or -SR 25 wherein R 2is phenyl which may be substituted; and Ru is hydrogen, or aryl (e.g. phenyl) which may be substituted. 30 In a particular aspect of the invention a compound of the Formula II is provided wherein
R
10 is hydrogen, halo, optionally substituted hydroxyl, alkyl, pyridinyl, phenyl, benzyl, 43 piperazinyl, amino, morpholinyl, or -SR2" wherein R2 is alkyl or aryl. In an embodiment, R1 0 is
-NH[CH
2
],NR
60
R
61 wherein m is 1 to 6, in particular 2 to 4, R 60 is hydrogen, R 6 1 is a carboxyl, in particular
-COOC(CH
3
)
3 . In an aspect of the invention a compound of the Formula II is provided wherein R" is 5 hydrogen, halo, optionally substituted alkyl, pyridinyl, piperidinyl, morpholinyl, piperazinyl, or phenyl. In another aspect of the invention a compound of the Formula II is provided wherein both of R'O and R" are not hydrogen. In particular embodiments of the invention one or more of R 10 and R" are alkyl, in 10 particular C 1
-C
6 alkyl and the other of R'O and R" is hydrogen. In particular embodiments of the invention one or more of R 10 and R" are aryl in particular phenyl or benzyl and the other of R1 0 and R" is hydrogen. In particular embodiments of the invention a compound of the Formula II is a compound in Table 3, more particularly MWO1-2-065LKM, MWO1-2-069SRM, MWO1-2-151SRM, 15 MWO1-5-188WH, MW01-6-127WH, MWO1-6-189WH, MW01-7-107WH, and derivatives thereof. The invention also provides an isolated and pure, in particular, substantially pure, compound of the Formula III wherein R's and R1 6 are independently substituted or unsubstituted hydrogen, hydroxyl, alkyl, alkenyl, alkynyl, alkylene, alkenylene, alkoxy, alkenyloxy, 20 cycloalkyl, cycloalkenyl, aryl, aryloxy, arylalkoxy, aroyl, heteroaryl, heterocyclic, acyl, acyloxy, sulfonyl, sulfinyl, sulfenyl, amino, imino, azido, thiol, thioalkyl, thioalkoxy, thioaryl, nitro, ureido, cyano, halo, silyl, silyloxy, silylthio, =0, =S, carboxyl, carbonyl, or carbamoyl, or an isomer or pharmaceutically acceptable salt thereof. The invention also provides an isolated and pure, in particular, substantially pure, 25 compound of the Formula IV wherein R 70 and R 7 1 are independently substituted or unsubstituted hydrogen, hydroxyl, alkyl, alkenyl, alkynyl, alkylene, alkenylene, alkoxy, alkenyloxy, cycloalkyl, cycloalkenyl, aryl, aryloxy, arylalkoxy, aroyl, heteroaryl, heterocyclic, acyl, acyloxy, sulfonyl, sulfinyl, sulfenyl, amino, imino, azido, thiol, thioalkyl, thioalkoxy, thioaryl, nitro, ureido, cyano, halo, silyl, silyloxy, silylthio, =0, =S, carboxyl, carbonyl, or carbamoyl, or 0 an isomer or pharmaceutically acceptable salt thereof.. In an aspect, the invention relates to a compound of the Formula IV wherein R 7 0 is a heterocylic, in particular a saturated 3 to 6-membered heteromonocylic group containing 1 to 4 nitrogen atoms more particularly, pyrrolidinyl, imidazolidinyl, piperidinyl, and piperazinyl, especially piperazinyl or piperidinyl, which may be substituted with alkyl especially methyl, 44 dimethyl, cycloalkyl especially cyclohexyl, aryl especially phenyl, a substituted or unsubstituted unsaturated condensed heterocyclic group containing 1 to 5 nitrogen atoms, in particular, indolyl, isoindolyl, indolizinyl, indazolyl, quinazolinyl, pteridinyl, quinolizidinyl, phthalazinyl, naphthyridinyl, quinoxalinyl, cinnolinyl, phenanthridinyl, acridinyl, phenanthrolinyl, 5 phenazinyl, carbazolyl, purinyl, benzimidazolyl, quinolyl, isoquinolyl, quinolinyl, isoquinolinyl,or indazolyl, especially benzimidazolyl substituted with oxy. The invention also relates to a compound of the Formula IV wherein R 70 is amino or substituted amino, and optionally R 71 is aryl, in particular phenyl. In an aspect R 70 is -N-R 2 ' wherein R 2 1 is hydrogen or alkyl, in particular C 1
-C
6 alkyl, more particularly methyl or 10 dimethyl, or -N-R 2 1
R
22 wherein R 2 1 is hydrogen or alkyl, in particular C-C 6 alkyl, more particularly methyl and R 22 is alkyl substituted with amino or substituted amino, heterocyclic, substituted heterocylic, or cycloalkyl. In an embodiment, R 7 is -N-R 21 R wherein R 2 ' is hydrogen or alkyl, in particular C 1
-C
6 alkyl, more particularly methyl and R is C-C 6 alkyl, especially methyl or ethyl substituted with a cycloalkyl especially cyclopropyl, a heterocyclic 15 especially piperidinyl, pyrrolidinyl, or morpholinyl which may be substituted in particular substituted with aryl, especially benzyl. A compound of the Formula IV may comprise structure 124, 125, 126, 127, 128, 129, 130, 131, 132, 133, 134, 135, 136, 137, 138, or 139 in Table 5 or derivatives thereof. The invention also provides an isolated and pure, in particular, substantially pure, 20 compound of the Formula V wherein R 50 and R 5 1 are independently substituted or unsubstituted hydrogen, hydroxyl, alkyl, alkenyl, alkynyl, alkylene, alkenylene, alkoxy, alkenyloxy, cycloalkyl, cycloalkenyl, aryl, aryloxy, arylalkoxy, aroyl, heteroaryl, heterocyclic, acyl, acyloxy, sulfonyl, sulfinyl, sulfenyl, amino, imino, azido, thiol, thioalkyl, thioalkoxy, thioaryl, nitro, ureido, cyano, halo, silyl, silyloxy, silylthio, =0, =S, carboxyl, carbonyl, or carbamoyl, or 25 an isomer or pharmaceutically acceptable salt thereof. The invention relates to a compound of the Formula V wherein Rso is substituted or unsubstituted hydrogen, alkyl, aryl, or heterocyclic; R 5 1 is substituted or unsubstituted hydrogen or alkyl, and R 5 2 is substituted or unsubstituted hydrogen, alkyl, cycloalkyl, heteroaryl or halo. In an aspect, the invention relates to a compound of the Formula V wherein R 50 is hydrogen, C 30 C 6 alkyl which may be substituted with alkyl, especially methyl or trimethyl, phenyl, or a 3 to 6 membered heteromonocylic group containing 1 to 4 nitrogen atoms more particularly, piperidinyl or morpholinyl,
R
51 is hydrogen or alkyl especially methyl, and R 2 is hydrogen, alkyl especially methyl, dimethyl, ethyl, or propyl, cyclohexyl, chloro, or an unsaturated 5 to 6 membered heteromonocyclyl group containing 1 to 4 nitrogen atoms, in particular, pyrrolyl, 45 pyrrolinyl, imidazolyl, pyrazolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, pyridinyl, pyrimidinyl, pyrazinyl, or pyridazinyl, especially pyridinyl. In an embodiment, Re 5 is aryl, R 5 1 is hydrogen, and Rs 2 is C 1 -C alkyl. A compound of the Formula V may comprise compound MWO1-7-057WH, or structure 5 32, 34, 36, 38, 39, 40, 41, 42, 43, 44, 46, 47, 48, 49, 63, 69, 70, 71, 75, 76, 77, 78, 79, 80, 81, and 82 in Table 5 or derivatives thereof In some embodiments, the present invention provides novel organic compounds, and/or heterocyclic derivatives thereof, depicted in Tables 2, 3, 4 or 5. Derivative groups that may be used to modify the compounds of the Formula I, T, H[, IV, or V 10 can be found in U.S. Patent Application No. 20030176437 (herein incorporated by reference in its entirety for all purposes). A compound of the Formula I, H, II, IV, or V may be in the form of a prodrug that is converted in vivo to an active compound. For example, in a compound of the Formula I one or more of R', R 2 , R3' R 4 , R', R 6 , and R 7 may comprise a cleavable group that is cleaved after 15 administration to a subject to provide an active (e.g., therapeutically active) compound, or an intermediate compound that subsequently yields the active compound. A cleavable group can be an ester that is removed either enzymatically or non-enzymatically. A compound of the Formula I, H, II, IV, or V may comprise a carrier, such as one or more of a polymer, carbohydrate, peptide or derivative thereof, which may be directly or 20 indirectly covalently attached to the compound. A carrier may be substituted with substituents described herein including without limitation one or more alkyl, amino, nitro, halogen, thiol, thioalkyl, sulfate, sulfonyl, sulfinyl, sulfoxide, hydroxyl groups. In aspects of the invention the carrier is an amino acid including alanine, glycine, praline, methionine, serine, threonine, asparagine, alanyl-alanyl, prolyl-methionyl, or glycyl-glycyl. A carrier can also include a 25 molecule that targets a compound of the Formula I, H, IM, IV, or V to a particular tissue or organ. Thus, a carrier may facilitate or enhance transport of a compound of the Formula 1, H, M, IV or V to the brain. PROCESS Compounds of the Formula I, H, IM, IV, or V can be prepared using reactions and 30 methods generally known to the person of ordinary skill in the art, having regard to that knowledge and the disclosure of this application including the Examples. The reactions are performed in a solvent appropriate to the reagents and materials used and suitable for the reactions being effected. It will be understood by those skilled in the art of organic synthesis that the functionality present on the compounds should be consistent with the proposed reaction 46 steps. This will sometimes require modification of the order of the synthetic steps or selection of one particular process scheme over another in order to obtain a desired compound of the invention. It will also be recognized that another major consideration in the development of a synthetic route is the selection of the protecting group used for protection of the reactive 5 functional groups present in the compounds described in this invention. An authoritative account describing the many alternatives to the skilled artisan is Greene and Wuts (Protective Groups In Organic Synthesis, Wiley and Sons, 1991). The starting materials and reagents used in preparing compounds or the invention are either available from commercial suppliers or are prepared by methods well known to a person 10 of ordinary skill in the art, following procedures described in such references as Fieser and Fieser's Reagents for Organic Synthesis, vols. 1-17, John Wiley and Sons, New York, N.Y., 1991; Rodd's Chemistry of Carbon Compounds, vols. 1-5 and supps., Elsevier Science Publishers, 1989; Organic Reactions, vols. 1-40, John Wiley and Sons, New York, N.Y., 1991; March J.: Advanced Organic Chemistry, 4th ed., John Wiley and Sons, New York, N.Y.; and 15 Larock: Comprehensive Organic Transformations, VCH Publishers, New York, 1989. The starting materials, intermediates, and compounds of the Formula I, H, M, IV, or V may be isolated and purified using conventional techniques, such as precipitation, filtration, distillation, crystallization, chromatography, and the like. The compounds of the Formula I, H, 1I, IV, or V may be characterized using conventional methods, including physical constants and 20 spectroscopic methods, in particular HPLC. The compounds of the Formula I , IH, IV, or V which are basic in nature can form a wide variety of different salts with various inorganic and organic acids. In practice is it desirable to first isolate a compound of the Formula I, II, I, IV, or V from the reaction mixture as a pharmaceutically unacceptable salt and then convert the latter to the free base compound by !5 treatment with an alkaline reagent and subsequently convert the free base to a pharmaceutically acceptable acid addition salt. The acid addition salts of the base compounds of the Formula I, HI, I, IV, or V are readily prepared by treating the base compound with a substantially equivalent amount of the chosen mineral or organic acid in an aqueous solvent medium or in a suitable organic solvent such as methanol or ethanol. Upon careful evaporation of the solvent, the 0 desired solid salt is obtained. Compounds of the Formula I, II, I, IV, or V which are acidic in nature are capable of forming base salts with various pharmacologically acceptable cations. These salts may be prepared by conventional techniques by treating the corresponding acidic compounds with an aqueous solution containing the desired pharmacologically acceptable cations and then 47 evaporating the resulting solution to dryness, preferably under reduced pressure. Alternatively, they may be prepared by mixing lower alkanolic solutions of the acidic compounds and the desired alkali metal alkoxide together and then evaporating the resulting solution to dryness in the same manner as before. In either case, stoichiometric quantities of reagents are typically 5 employed to ensure completeness of reaction and maximum product yields. In particular aspects, the present invention provides methods of making the compounds disclosed herein, comprising the steps provided (See, e.g., the Figures and Materials and Methods). Broad Process Description for Pyr PCT Application In an aspect, the invention provides a process for preparing a compound of the formula II 10 wherein R11 is hydrogen and R10 is an unsaturated 5 to 6 membered heteromonocyclyl group containing 1 to 4 nitrogen atoms, in particular, pyrrolyl, pyrrolinyl, imidazolyl, pyrazolyl, 2 pyridyl, 3-pyridyl, 4-pyridyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazolyl, or tetrazolyl, more particularly pyridinyl, which comprises reacting a compound of the formula H wherein R10 is halo, in particular chloro, and R 11 is hydrogen, with boronic acid substituted with an 15 unsaturated 5 to 6 membered heteromonocyclyl group containing 1 to 4 nitrogen atoms, in particular, pyrrolyl, pyrrolinyl, imidazolyl, pyrazolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazolyl, or tetrazolyl, more particularly pyridinyl, under suitable conditions to prepare a compound of the formula H wherein R 11 is hydrogen and R10 is an unsaturated 5 to 6 membered heteromonocyclyl group containing 1 to 4 nitrogen atoms, in 2O particular, pyrrolyl, pyrrolinyl, imidazolyl, pyrazolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazolyl, or tetrazolyl, more particularly pyridinyl. In an embodiment, R10 is phenyl substituted with halo. In another aspect, the invention provides a process for preparing a compound of the formula H wherein R 11 is hydrogen and RIO is a substituted aryl which comprises reacting a !5 compound of the formula EH wherein R10 is halo, in particular chloro, and R1 1 is hydrogen, with a substituted aryl boronic acid under suitable conditions to prepare a compound of the formula H wherein R11 is hydrogen and R10 is a substituted aryl. In an embodiment, RIO is phenyl substituted with halo. In another aspect, the invention provides a process for preparing a compound of the 0 formula IH wherein RIO is hydrogen and RI 1 is alkyl which comprises reacting a compound of the formula H wherein R1 1 is halo, in particular chloro, and RIO is hydrogen, with an alkyl boronic acid under suitable conditions to prepare a compound of the formula H wherein RIO is hydrogen and R11 is alkyl. In an embodiment, R11 is lower alkyl, in particular methyl or ethyl, and a 48 compound of the formula IH wherein RI 1 is chloro is reacted with lower alkyl boronic acid, in particular methyl or ethyl boronic acid under suitable conditions. In another aspect, the invention provides a process for preparing a compound of the formula 11 wherein RIO is hydrogen and R11 is an alkyl which comprises reacting a pyridazine 5 substituted at the C3 position with halo (e.g., chloro), at the C4 position with alkyl, and at the 6 position with phenyl, with 2 -(piperidin-4-yloxy)pyrimidine under suitable conditions to prepare a compound of the formula 1H wherein R10 is hydrogen and R11 is an alkyl. In an embodiment, R 11 is methyl or ethyl. In another aspect, the invention provides a process for preparing a compound of the 10 formula H1 wherein R10 is hydrogen and R11 is aryl which comprises reacting a compound of the formula H wherein R10 is hydrogen and RI 1 is halo (e.g., chloro), with pyridazine substituted at the C3 position with halo (e.g., chloro), at the C4 position with aryl, and at the 6 position with phenyl, with 2 -(piperidin-4-yloxy)pyrimidine under suitable conditions to prepare a compound of the formula H wherein R10 is hydrogen and R11 is aryl. In an embodiment, R11 is phenyl. [5 In another aspect, the invention provides a process for preparing a compound of the formula H wherein RIO is hydrogen and R 11 is an unsaturated 5 to 6 membered heteromonocyclyl group containing 1 to 4 nitrogen atoms, in particular, pyrrolyl, pyrrolinyl, imidazolyl, pyrazolyl, 2 pyridyl, 3-pyridyl, 4-pyridyl, pyridinyl, pyriniidinyl, pyrazinyl, pyridazinyl, triazolyl, or tetrazolyl, more particularly pyridinyl which comprises reacting a compound of the formula II wherein R 11 !0 is halo, in particular chloro, and R10 is hydrogen, with a boronic acid substituted with an unsaturated 5 to 6 membered heteromonocyclyl group containing 1 to 4 nitrogen atoms, in particular, pyrrolyl, pyrrolinyl, imidazolyl, pyrazolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazolyl, or tetrazolyl, more particularly pyridinyl, under suitable conditions to prepare a compound of the formula H wherein RIO is hydrogen and R 11 is :5 an unsaturated 5 to 6 membered heteromonocyclyl group containing 1 to 4 nitrogen atoms, in particular, pyrrolyl, pyrrolinyl, imidazolyl, pyrazolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazolyl, or tetrazolyl, more particularly pyridinyl. In an embodiment, the invention provides a process for preparing a compound of the formula H wherein RIO is hydrogen and R 11 is pyridinyl which comprises reacting a compound 0 of the formula H wherein R11 is halo, in particular chloro, and RIO is hydrogen, with a pyridinyl boronic acid under suitable conditions to prepare a compound of the formula H wherein RIO is hydrogen and Ri 1 is pyridinyl. In another aspect, the invention provides a process for preparing a compound of the formula H wherein RIO is hydrogen and R 11 is an unsaturated 5 to 6 membered heteromonocyclyl 49 group containing 1 to 4 nitrogen atoms, in particular, pyrrolyl, pyrrolinyl, imidazolyl, pyrazolyl, 2 pyridyl, 3-pyridyl, 4-pyridyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazolyl, or tetrazolyl, more particularly pyridinyl which comprises reacting a pyridazine substituted at the C3 position with halo, at the C4 position with an unsaturated 5 to 6 membered heteromonocyclyl group 5 containing 1 to 4 nitrogen atoms, in particular, pyrrolyl, pyrrolinyl, imidazolyl, pyrazolyl, 2 pyridyl, 3-pyridyl, 4-pyridyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazolyl, or tetrazolyl, more particularly pyridinyl, and at the 6 position with phenyl, with 2-(piperidin-4 yloxy)pyrimidine under suitable conditions to prepare a compound of the formula H wherein RIO is hydrogen and R11 is an unsaturated 5 to 6 membered heteromonocyclyl group containing I to 4 10 nitrogen atoms, in particular, pyrrolyl, pyrrolinyl, imidazolyl, pyrazolyl, 2-pyridyl, 3-pyridyl, 4 pyridyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazolyl, or tetrazolyl, more particularly pyridinyl. In an embodiment, the invention provides a process for preparing a compound of the formula IH wherein RIO is hydrogen and R11 is pyridinyl which comprises reacting a pyridazine 15 substituted at the C3 position with halo, at the C4 position with pyridinyl, and at the 6 position with phenyl, with 2 -(piperidin-4-yloxy)pyrimidine under suitable conditions to prepare a compound of the formula II wherein R10 is hydrogen and R 11 is pyridinyl. In another aspect, the invention provides a process for preparing a compound of the formula H1 wherein RIO is hydrogen and Ri1 is piperidinyl or substituted piperidinyl which 20 comprises reacting a compound of the formula II wherein R1I is halo, in particular chloro, and R10 is hydrogen with piperazinyl or substituted piperazinyl under suitable conditions to prepare a compound of the formula II wherein RIO is hydrogen and R11 is piperidinyl or substituted piperidinyl. In another aspect, the invention provides a process for preparing a compound of the 25 formula I wherein R1 is piperazinyl or piperazinyl substituted with alkyl, aryl, or cycloalkyl, R2 is aryl, R3, R4, R5 and R6 are hydrogen and R7 is absent, which comprises reacting a pyridazine substituted at the C3 position with halo, at the C4 position with aryl with a piperazinyl or piperazinyl substituted with alkyl, aryl, or cycloalkyl, under suitable conditions to prepare a compound of the formula I wherein R1 is piperazinyl or piperazinyl substituted with alkyl, aryl, or 30 cycloalkyl, R2 is aryl, R3, R4, R5 and R6 are hydrogen and R7 is absent. In another aspect, the invention provides a process for preparing a compound of the formula I wherein R1 is piperazinyl or piperazinyl substituted with alkyl, aryl, or cycloalkyl, R2 is an unsaturated 5 to 6 membered heteromonocyclyl group containing 1 to 4 nitrogen atoms, in particular, pyrrolyl, pyrrolinyl, imidazolyl, pyrazolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, pyridinyl, 50 pyrimidinyl, pyrazinyl, pyridazinyl, triazolyl, or tetrazolyl, more particularly pyridinyl, R3, R4, R5 and R6 are hydrogen and R7 is absent, which comprises reacting a pyridazine substituted at the C3 position with halo, at the C4 position with an unsaturated 5 to 6 membered heteromonocyclyl group containing 1 to 4 nitrogen atoms, in particular, pyrrolyl, pyrrolinyl, 5 imidazolyl, pyrazolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazolyl, or tetrazolyl, more particularly pyridinyl, with piperazinyl or piperazinyl substituted with alkyl, aryl, or cycloalkyl under suitable conditions to prepare a compound of the formula I wherein Ri is piperazinyl or piperazinyl substituted with alkyl, aryl, or cycloalkyl, R2 is an unsaturated 5 to 6 membered heteromonocyclyl group containing 1 to 4 nitrogen atoms, in 10 particular, pyrrolyl, pyrrolinyl, imidazolyl, pyrazolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazolyl, or tetrazolyl, more particularly pyridinyl, R3, R4, R5 and R6 are hydrogen and R7 is absent. In another aspect, the invention provides a process for preparing a compound of the formula I wherein RI is substituted amino in particular amino substituted with substituted 15 morpholinyl, in particular morpholinoethyl, R2 is aryl or an unsaturated 5 to 6 membered heteromonocyclyl group containing 1 to 4 nitrogen atoms, in particular, pyrrolyl, pyrrolinyl, imidazolyl, pyrazolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazolyl, or tetrazolyl, in particular pyridinyl, R3, R4, R5 and R6 are hydrogen and R7 is absent, which comprises reacting a pyridazine substituted at the C3 position with halo, at the 0 C4 position with aryl or an unsaturated 5 to 6 membered heteromonocyclyl group containing 1 to 4 nitrogen atoms, in particular, pyrrolyl, pyrrolinyl, imidazolyl, pyrazolyl, 2-pyridyl, 3-pyridyl, 4 pyridyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazolyl, or tetrazolyl, more particularly pyridinyl, with substituted amino in particular amino substituted with substituted morpholinyl, in particular morpholinoethyl, under suitable conditions to prepare a compound of the formula I '5 wherein RI is substituted amino in particular amino substituted with substituted morpholinyl, R2 is aryl, R3, R4, R5 and R6 are hydrogen and R7 is absent. In another aspect, the invention provides a process for preparing a compound of the formula V wherein R50 is aryl, R51 is hydrogen, and R52 is alkyl comprising reacting a pyridazine substituted at position C3 with halo, at position C4 with aryl and position 6 with alkyl 0 with 1-( 2 -pyrimidyl)piperazine under suitable conditions to prepare a compound of the formula V wherein R50 is aryl, R51 is hydrogen, and R52 is alkyl. In another aspect, the invention provides a process for preparing a compound of the formula I wherein R1 is substituted amino, R2 is an unsaturated 5 to 6 membered heteromonocyclyl group containing 1 to 4 nitrogen atoms, in particular, pyrrolyl, pyrrolinyl, 51 imidazolyl, pyrazolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazolyl, or tetrazolyl, in particular pyridinyl, R3, R4, R5 and R6 are hydrogen and R7 is absent comprising reacting a pyridazine substituted at the C3 position with halo, at the C4 position with an unsaturated 5 to 6 membered heteromonocyclyl group containing 1 to 4 nitrogen 5 atoms, in particular, pyrrolyl, pyrrolinyl, imidazolyl, pyrazolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazolyl, or tetrazolyl, in particular pyridinyl, and at the C6 position phenyl, and a substituted amino under suitable conditions to prepare a compound of the formula I wherein R1 is substituted amino, R2 is an unsaturated 5 to 6 membered heteromonocyclyl group containing 1 to 4 nitrogen atoms, in particular, pyrrolyl, 10 pyrrolinyl, imidazolyl, pyrazolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazolyl, or tetrazolyl, in particular pyridinyl, R3, R4, R5 and R6 are hydrogen and R7 is absent. In the preparation of compounds of the Formula R, a key precursor (See, e.g., Figure 1) that may be utilized was obtained commercially and used directly for the synthesis of the illustrated 15 compound MWO1-3-183WH without further purification. Compounds may be synthesized with yields of 81 - 96%. All purified compounds may be characterized by HPLC, mass spectrometry and NMR in order to confirm syntheses. In Figure 1, a synthetic scheme is shown, for synthesis of MWO1-3 183WH with unconstrained aromatic ring at position 6 and no modification at position 5. Reagent and condition: (a) 1-BuOH, NH 4 Ci, 2-(piperazin-1-yl) pyrimidine. 20 Thus, in an aspect, the invention provides a method for preparing a compound of the Formula If wherein a substituted 6-phenylpyridazine is reacted with 2-(piperazin-lyl)pyridmidine to produce a compound of the Formula H wherein R' 0 and R" are hydrogen. A compound of the formula H1 wherein RIO and R11 are hydrogen can be reacted under suitable conditions and with suitable reagents to introduce the radicals R' 0 and R" which are independently hydrogen, hydroxyl, alkyl, alkenyl, 25 alkynyl, alkylene, alkenylene, alkoxy, alkenyloxy, cycloalkyl, cycloalkenyl, aryl, aryloxy, arylalkoxy, aroyl, heteroaryl, heterocyclic, acyl, acyloxy, sulfonyl, sulfinyl, sulfenyl, amino, imino, azido, thiol, thioalkyl, thioalkoxy, thioaryl, nitro, ureido, cyano, halo, silyl, silyloxy, silylalkyl, silylthio, =0, =S, carboxyl, carbonyl, carbamoyl, or carboxamide Therapeutic efficacy and toxicity of compounds, compositions and methods of the 30 invention may be determined by standard pharmaceutical procedures in cell cultures or with experimental animals such as by calculating a statistical parameter such as the ED 50 ( the dose that is therapeutically effective in 50% of the population) or LID 5 0 (the dose lethal to 50% of the population) statistics. The therapeutic index is the dose ratio of therapeutic to toxic effects and it can be expressed as the ED 5 0
/LD
50 ratio. Pharmaceutical compositions which exhibit large 52 therapeutic indices are preferred. By way of example, one or more of the therapeutic effects, in particular beneficial effects disclosed herein, can be demonstrated in a subject or disease model, for example, a TgCRND8 mouse with symptoms of Alzheimer's disease. Biological investigations were done with compounds disclosed herein that were >95% 5 homogenous as determined by HPLC/MS analysis. As part of a hierarchal, cell-based screening protocol, the compounds were screened for their ability to block IL1p and TNFa production by BV-2 mouse microglial cells stimulated with LPS. The data for MWO1-3-183WH is shown in Figure 2. Derivative groups that may be used to modify the compounds of the present invention can be found in U.S. Patent Application No. 20030176437 (herein incorporated by reference in its entirety for all 10 purposes). The compounds disclosed herein can be tested for liver toxicity which is an important initial consideration for orally administered compounds since the liver is the major site of initial drug metabolism and is critical to overall metabolism and homeostasis of an animal. An example of an in vivo liver toxicity test in animals is illustrated in Example 2. Compounds 15 disclosed herein may also be tested for cardiac safety by testing for hERG channel inhibition, for example using the method illustrated in Example 3. COMPOSITIONS AND KITS A compound of the Formula I, II, III, IV, or V of the invention may be formulated into a pharmaceutical composition for administration to a subject. Pharmaceutical compositions of the 20 present invention or fractions thereof comprise suitable pharmaceutically acceptable carriers, excipients, and vehicles selected based on the intended form of administration, and consistent with conventional pharmaceutical practices. Suitable pharmaceutical carriers, excipients, and vehicles are described in the standard text, Remington's Pharmaceutical Sciences, Mack Publishing Company (Remington's Pharmaceutical Sciences, Mack Publishing Company, 25 Easton, Pa., USA 1985). By way of example for oral administration in the form of a capsule or tablet, the active components can be combined with an oral, non-toxic pharmaceutically acceptable inert carrier such as lactose, starch, sucrose, methyl cellulose, magnesium stearate, glucose, calcium sulfate, dicalcium phosphate, mannitol, sorbital, and the like. For oral administration in a liquid form, the drug components may be combined with any oral, non-toxic, 10 pharmaceutically acceptable inert carrier such as ethanol, glycerol, water, and the like. Suitable binders (e.g. gelatin, starch, corn sweeteners, natural sugars including glucose; natural and synthetic gums, and waxes), lubricants (e.g. sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, and sodium chloride), disintegrating agents (e.g. starch, methyl cellulose, agar, bentonite, and xanthan gum), flavoring agents, and coloring 53 agents may also be combined in the compositions or components thereof. Compositions as described herein can further comprise wetting or emulsifying agents, or pH buffering agents. A composition of the invention can be a liquid solution, suspension, emulsion, tablet, pill, capsule, sustained release formulation, or powder. The compositions can be formulated as a 5 suppository, with traditional binders and carriers such as triglycerides. Oral formulations can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc. Various delivery systems are known and can be used to administer a composition of the invention, e.g. encapsulation in liposomes, microparticles, microcapsules, and the like. 10 Formulations for parenteral administration may include aqueous solutions, syrups, aqueous or oil suspensions and emulsions with edible oil such as cottonseed oil, coconut oil or peanut oil. Dispersing or suspending agents that can be used for aqueous suspensions include synthetic or natural gums, such as tragacanth, alginate, acacia, dextran, sodium carboxymethylcellulose, gelatin, methylcellulose, and polyvinylpyrrolidone. 15 Compositions for parenteral administration may include sterile aqueous or non-aqueous solvents, such as water, isotonic saline, isotonic glucose solution, buffer solution, or other solvents conveniently used for parenteral administration of therapeutically active agents. A composition intended for parenteral administration may also include conventional additives such as stabilizers, buffers, or preservatives, e.g. antioxidants such as methylhydroxybenzoate or 0 similar additives. Compositions of the invention can be formulated as pharmaceutically acceptable salts as described herein. A composition of the invention may be sterilized by, for example, filtration through a bacteria retaining filter, addition of sterilizing agents to the composition, irradiation of the !5 composition, or heating the composition. Alternatively, the compounds or compositions of the present invention may be provided as sterile solid preparations e.g. lyophilized powder, which are readily dissolved in sterile solvent immediately prior to use. After pharmaceutical compositions have been prepared, they can be placed in an appropriate container and labeled for treatment of an indicated condition. For administration of 0 a composition of the invention, such labeling would include amount, frequency, and method of administration. The invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of a pharmaceutical composition of the invention to provide a beneficial effect, in particular a sustained beneficial effect. Associated 54 with such container(s) can be various written materials such as instructions for use, or a notice in the form prescribed by a governmental agency regulating the labeling, manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use, or sale for human administration. 5 The invention also provides a kit comprising a compound or a pharmaceutical composition of the invention. The kit can be a package which houses a container which contains a composition of the invention and also houses instructions for administering the composition to a subject. APPLICATIONS 10 The invention contemplates the use of compounds of the Formula I, II, 111, IV, or V and compositions comprising the same for treating a disease disclosed herein, in particular preventing, and/or ameliorating disease severity, disease symptoms, and/or periodicity of recurrence of a disease disclosed herein. The invention also contemplates treating in mammals, diseases using the compounds, compositions or treatments of the invention. The present 15 invention in embodiments may provide a composition comprising a compound that provides beneficial effects including greater solubility, stability, efficacy, potency, and/or utility, in particular greater solubility and stability. Novel compounds and methods for new therapeutic interventions are needed for many areas of medicine and disease treatment. For example, chronic and acute inflammatory conditions form the 10 basis for diseases affecting all organ systems including, but not limited to, asthma, acute inflammatory diseases, vascular inflammatory disease, chronic inflammation, atherosclerosis, angiopathy, myocarditis, nephritis, Crohn's disease, arthritis, type I and II diabetes and associated vascular pathologies. The incidence of these inflammatory conditions is on the rise and the expense is large. For example, for just one form of inflammatory disease, Alzheimer's disease, the direct costs !5 (such as medications, doctors' fees, and nursing home care) and indirect costs (loss of productivity of those suffering Alzheimer's disease and loss of productivity of those caring for these individuals) are estimated to exceed one-hundred billion dollars per year. With reference to the following examples and related discussions, the present invention provides various methods relating to the modulation of inflammation, glial activation or 0 phosphorylation pathways and/or new therapeutic routes relating thereto. As illustrated more fully elsewhere herein, such methods include but are not limited to use of the compounds and compositions of this invention, preferably in a dose dependent fashion, to selectively inhibit protein kinase activity, glial activation response, oxidative stress-related responses such as nitric oxide synthase production and nitric oxide accumulation, cellular apoptosis and/or death 55 associated protein kinase activity, and/or proinflammatory cytokine responses such as interleukin or tumor necrosis factor production. Such methods can include the preparation and/or formulation of a composition with subsequent administration and/or delivery to activated glial cells, tissue, culture or a related physiological system or medium, such 5 administration/delivery in a dose or at a compositional concentration sufficient to effect the desired regulation and/or inhibition, without substantially inhibiting other desired endogenous anti-inflammatory responses. In an aspect, the present invention relates to the inhibition of neuronal cell death. Selective neuronal cell death is a characteristic feature of the pathology of a number of 10 neurodegenerative diseases, including Alzheimer's disease (AD), and traumatic brain injury, and stroke. Selected compounds and compositions of the present invention may be used to reduce or inhibit A3-induced neuronal cell death and in particular to reduce or inhibit calmodulin regulated protein kineses, such as death associated protein kinase (DAPK). In some embodiments, the present invention provides methods of inhibiting cell signaling 15 molecule production (e.g., IL-l and TNFa), comprising administering compositions comprising one or more of the compounds of the Formula I , III V, or V, in particular one or more compounds depicted in the Figures and Tables herein, in particular the compounds depicted in Tables 2, 3, 4, or 5, or derivatives of these compounds. The present invention also provides compounds (e.g., compounds listed in the Figures and Z0 Tables for use in 1) lowering amounts of pro-inflammatory cytokines (e.g., TNFa or IL-lp) and/or 2) maintaining normal levels of postsynaptic proteins (e.g., PSD-95). In some embodiments, the reduction of pro-inflammatory cytokines reduces cytokines to levels found in a normal, healthy individual. In some embodiments, the compounds are provided to an individual displaying characteristics of an inflammatory disease (e.g., Alzheimer's disease), such that treatment with the !5 compounds reduces aberrantly high pro-inflammatory cytokine production caused by the disease (e.g., Ap- induced increase in pro-inflammatory cytokines). In another aspect, selected compounds and compositions of the invention may be used to modulate cytokine-mediated neuronal cell death, in particular modulate cytokine-induced generation of NO, TNFa signaling through the Fas/TNFR family of death receptors, and/or 0 DAPK, in Alzheimer's disease and other neurodegenerative disorders, and brain injury, and stroke. The evidence for the involvement of pro-inflammatory cytokines and NO in neuronal cell death has been reviewed in Akiyama, H., et al., (2000) Neurobiol. Agir g 21, 383-421; 56 Prusiner, S.B. (2001) New Engl. J. Med. 344, 15 16-1526). cytokine-induced neuronal death could involve DAPK. In part, the present invention also relates to the inhibition of cell death or tissue loss and cell activation in addition to brain glia and neurons. For example, cell activation and tissue 5 damage is a characteristic of other diseases such as acute lung injury (ALI). ALI due to sepsis, trauma or mechanical ventilation is associated with high mortality and morbidity, yet there are few effective therapies for the treatment of ALI. ALI is common during sepsis, which itself has an annual mortality equal to acute myocardial infarction. Endothelial cell (EC) dysfunction and activation has been implicated in the in vivo responses linked to ALI, and EC protein kineses, 10 such as myosin light chain kinase (MLCK), have been shown to be critical to EC barrier function and activation. Similarly, the response of the heart to stress and acute injury results in acute and chronic injuries in which protein phosphorylation regulated pathways and cell activation has been linked to cell death and tissue damage. MLCK and related enzymes such as Rho kinase have been implicated in these processes and may be targets for new therapeutics. 15 Accordingly, compounds of the Formula I, , Ml, IV, or V can be used to reduce injury from hypoxia-ischemia, acute lung injury and/or endothelial cell dysfunction in lung or vascular tissue. In another aspect of the invention, a method is provided for treating in a subject a disease involving or characterized by inflammation, in particular neuroinflammation, comprising 0 administering to the subject a therapeutically effective amount of a compound of the Formula I, II, I, IV, or V, or a pharmaceutically acceptable salt thereof. In a further aspect, a method is provided for treating in a subject a condition involving inflammation, in particular neuroflammation, comprising administering to the subject a therapeutically effective amount of a composition comprising a compound of the Formula I, II, IH, IV, or V and a pharmaceutically 5 acceptable carrier, excipient, or vehicle. In a further aspect, the invention provides a method involving administering to a subject a therapeutic compound of the Formula I, II, IM, IV, or V, or a pharmaceutically acceptable salt thereof, or a composition comprising a compound of the Formula I 1I, IH, IV, or V, and a pharmaceutically acceptable carrier, excipient, or vehicle which inhibit or reduce 0 neuroflammation, activation of glia, proimflammatory cytokines, oxidative stress-related enzymes, acute phase proteins and/or components of the complement cascade. In another aspect, the invention provides a method for treating in a subject a disease associated with neuroinflammation that can be decreased or inhibited with a compound disclosed herein comprising administering to the subject a therapeutically effective amount of a 57 compound of the Formula I, II, IH, IV, or V, a pharmaceutically acceptable salt thereof, or a composition comprising a compound of the Formula I, II, I, IV, or V and a pharmaceutically acceptable carrier, excipient, or vehicle. In another aspect, the invention provides a method for preventing or inhibiting activation 5 of protein kinases, in particular DAPK, in a subject comprising administering a therapeutically effective amount of a compound of the Formula I, II, El, IV, or V a pharmaceutically acceptable salt thereof, or a composition comprising a compound of the Formula I, I, I, IV, or V and a pharmaceutically acceptable carrier, excipient, or vehicle. In a further aspect, the invention provides a method for reducing or inhibiting kinase 10 activity, glial activation, neuronal cell damage, and/or neuronal cell death in a subject comprising administering to the subject a therapeutically effective amount of a compound of the Formula I, H, II, IV, or V a pharmaceutically acceptable salt thereof, or a composition comprising a compound of the Formula I, II, m, IV, or V and a pharmaceutically acceptable carrier, excipient, or vehicle. 15 In some embodiments, the invention provides methods of inhibiting cell signaling molecule production (e.g., IL-1P and TNFa), comprising administering compositions comprising one or more compounds of the Formula I, II, IV, or V, in particular the compounds depicted in the Figures and Tables, more particularly Table 2, 3, 4 or 5, or derivatives of these compounds. In some embodiments, one or more of the compounds, in particular the compounds depicted in the Figures and 20 Tables, more particularly Table 2, 3, 4 or 5 or derivatives of these compounds, are co-administered with other recognized therapeutics to treat inflammatory disease (e.g., neuroinflammatory disease, in particular Alzheimer's disease). In some embodiments, the invention provides compounds (e.g., compounds listed in the Figures and Tables) for use in 1) lowering amounts of pro-inflammatory cytokines (e.g., TNFa or IL-10) and/or 2) maintaining normal levels of postsynaptic proteins (e.g., 25 PSD-95) for research, drug screening, or therapeutic purposes. In some embodiments, the reduction of pro-inflammatory cytokines reduces cytokines to levels found in a normal, healthy individual. In some embodiments, the compounds are provided to an individual displaying characteristics of an inflammatory disease (e.g., neuroinflammatory disease, in particular Alzheimer's disease), such that treatment with the compounds reduces aberrantly high pro-inflammatory cytokine production caused 30 by the disease (e.g., A-induced increase in pro-inflammatory cytokines). In an aspect, the invention provides a method for amelioriating progression of a disease or obtaining a less severe stage of a disease in a subject suffering from such disease (e.g., neuroinflammatory disease, in particular a neurodegenerative diseae, more particularly Alzheimer's disease) comprising administering a therapeutically effective amount of a 58 compound of the Formula I, II, III, IV, or V, a pharmaceutically acceptable salt thereof, or a composition comprising a compound of the Formula I, H, III, IV, or V and a pharmaceutically acceptable carrier, excipient, or vehicle. The invention relates to a method of delaying the progression of a disease (e.g. 5 neuroinflammatory disease, in particular a neurodegenerative disease, more particularly Alzheimer's disease) comprising administering a therapeutically effective amount of a compound of the Formula , IT, E[, IV, or V, a pharmaceutically acceptable salt thereof, or a composition comprising a compound of the Formula I, H, I, IV, or V and a pharmaceutically acceptable carrier, excipient, or vehicle. 10 The invention also relates to a method of increasing survival of a subject suffering from a disease (e.g. neuroinflammatory disease, in particular a neurodegenerative disease, more particularly Alzheimer's disease) comprising administering a therapeutically effective amount of a compound of the Formula I, H, III, IV, or V, a pharmaceutically acceptable salt thereof, or a composition comprising a compound of the Formula I, II, 111, IV, or V and a pharmaceutically 15 acceptable carrier, excipient, or vehicle. The invention has particular applications in treating or preventing a neurodegenerative disease, in particular Alzheimer's disease. In an aspect of the invention a compound of the Formula I H, III, IV, or V is utilized in the treatment of Alzheimer's disease. Alzheimer's disease may be treated by administering a therapeutically effective amount of a compound of 20 the Formula I, II, H, IV, or V. Such treatment may be effective for retarding the degenerative effects of Alzheimer's disease, including specifically, but not exclusively, neuroinflammation, deterioration of the central nervous system, loss of mental facilities, loss of short term memory, and disorientation. In another aspect, the invention provides a method for treating Alzheimer's disease by 25 providing a composition comprising a compound of the invention in an amount sufficient to reverse or inhibit neuroinflammation, activation of signaling pathways involved in inflammation (e.g., neuroinflammation), cell signaling molecule production, activation of glia or glial activation pathways and responses, proinflammatory cytokines or chemokines (e.g., interleukin (IL) or tumor necrosis factor (TNF), oxidative stress-related responses such as nitric oxide 0 synthase production and nitric oxide accumulation, acute phase proteins, components of the complement cascade, protein kinase activity (e.g., death associated protein kinase activity), neuronal cell damage, and/or neuronal cell death for a prolonged period following administration.
59 In a further aspect, the invention provides a method for treating Alzheimer's disease in a patient in need thereof which includes administering to the individual a composition that provides a compound of the invention in a dose sufficient to reverse or inhibit neuroinflammation, activation of signaling pathways involved in inflammation (e.g., 5 neuroinflammation), cell signaling molecule production, activation of glia or glial activation pathways and responses, proinflammatory cytokines or chemokines (e.g., interleukin (IL) or tumor necrosis factor (TNF), oxidative stress-related responses such as nitric oxide synthase production and nitric oxide accumulation, acute phase proteins, components of the complement cascade, protein kinase activity (e.g., death associated protein kinase activity), neuronal cell 10 damage, and/or neuronal cell death for a prolonged period following administration. The invention in an embodiment provides a method for treating Alzheimer's disease, the method comprising administering to a mammal in need thereof a composition comprising a compound of the invention in an amount sufficient to reduce cognitive decline for a prolonged period following administration, thereby treating the Alzheimer's disease. 15 In as aspect, the invention relates to a method of treatment comprising administering a therapeutically effective amount of one or more compound of the Formula I, II, III, IV, or V, a pharmaceutically acceptable salt thereof, or a composition comprising a compound of the Formula I, II, 111, IV, or V and a pharmaceutically acceptable carrier, excipient, or vehicle, which upon administration to a subject with symptoms of a neurodegenerative disease, in 20 particular Alzheimer's disease, produces one or more therapeutic effect, in particular a beneficial effect, more particularly a sustained beneficial effect. In an embodiment, a beneficial effect is evidenced by a decrease or inhibition of one or more of the following: inflammation (e.g. neuroinflammation), activation of signaling pathways involved in inflammation (e.g., neuroinflammation), cell signaling molecule production, activation of glia or glial activation 25 pathways and responses, proinflammatory cytokines or chemokines (e.g., interleukin (IL) or tumor necrosis factor (TNF), oxidative stress-related responses such as nitric oxide synthase production and nitric oxide accumulation, acute phase proteins, components of the complement cascade, protein kinase activity (e.g., death associated protein kinase activity), cell damage (e.g., neuronal cell damage), and/or cell death (e.g., neuronal cell death). 50 In an embodiment, where the disease is Alzheimer's disease, beneficial effects of a compound or composition or treatment of the invention can manifest as one, two, three, four, five, six, seven, eight, or all of the following, in particular five or more, more particularly 7 or more of the following: 60 a) A reduction in protein kinase activity (e.g. DAPK), in particular at least about a 0.05%, 0.1%, 0.5%, 1%, 2%, 5%, 10%, 15%, 20%, 30%, 33%, 35%, 40%, 45%, 50%, 60%, 70%, 80%, 90%, 95%, or 99% decrease in protein kinase activity. b) A reduction in glial activation response, in particular, at least about a 0.05%, 0.1%, 0.5%, 5 1%, 2%, 5%, 10%, 15%, 20%, 30%, 33%, 35%, 40%, 45%, 50%, 60%, 70%, 80%, 90%, 95%, or 99% reduction in glial activation. c) A reduction in glial activity in the brain, relative to the levels determined in the absence of a compound of the Formula I, II, III, IV, or V in subjects with symptoms of Alzheimer's disease. In particular, the compounds induce at least about a 2%, 5%, 10%, 15%, 20%, 10 30%, 40%, 50%, 60%, 70%, 80%, or 90% decrease in glial activity d) A reduction in oxidative stress-related responses (e.g., nitric oxide synthase production and/or nitric oxide accumulation), in particular at least about a 0.05%, 0.1%, 0.5%, 1%, 2%, 5%, 10%, 15%, 20%, 30%, 33%, 35%, 40%, 45%, 50%, 60%, 70%, 80%, 90%, 95%, or 99% reduction in oxidative stress-related responses such as nitric oxide synthase 15 production and nitric oxide accumulation. e) A reduction in cellular apoptosis and/or death associated protein kinase activity, in particular a 0.05%, 0.1%, 0.5%, 1%, 2%, 5%, 10%, 15%, 20%, 30%, 33%, 35%, 40%, 45%, 50%, 60%, 70%, 80%, 90%, 95%, or 99% reduction in cellular apoptosis and/or death associated protein kinase activity. 20 f) A reduction in proinflammatory cytokine responses in particular a 0.05%, 0.1%, 0.5%, 1%, 2%, 5%, 10%, 15%, 20%, 30%, 33%, 35%, 40%, 45%, 50%, 60%, 70%, 80%, 90%, 95%, or 99% reduction in proinflammatory cytokine responses. g) A reduction in interleukin-1 and/or tumor necrosis factor production in particular a 0.05%, 0.1%, 0.5%, 1%, 2%, 5%, 10%, 15%, 20%, 30%, 33%, 35%, 40%, 45%, 50%, !5 60%, 70%, 80%, 90%, 95%, or 99% reduction in interleukin-10 and/or tumor necrosis factor production. h) A slowing of the rate of disease progression in a subject with Alzheimer's disease. i) Increase in survival in a subject with symptoms of Alzheimer's disease. In particular aspects of the invention beneficial effects of compounds, compositions or 10 treatments of the invention can manifest as (a) and (b); (a), (b) and (c); (a) through (d); (a) through (e); (a) through (f); (a) through (g); (a) through (h); or (a) through (i). Compounds, pharmaceutical compositions and methods of the invention can be selected that have sustained beneficial effects. In an embodiment, a pharmaceutical composition with 61 statistically significant sustained beneficial effects is provided comprising a therapeutically effective amount of a compound of the invention. The invention provides a method for treating mild cognitive impairment (MCI) comprising administering a therapeutically effective amount of a compound of the Formula I, H, 5 III, IV, or V, a pharmaceutically acceptable salt thereof, or a composition comprising a compound of the Formula I, II, III, IV, or V and a pharmaceutically acceptable carrier, excipient, or vehicle. In an embodiment, the invention provides a method of reversing or inhibiting neuroinflammation, activation of signaling pathways involved in inflammation (e.g., 10 neuroinflammation), cell signaling molecule production, activation of glia or glial activation pathways and responses, proinflammatory cytokines or chemokines (e.g., interleukin (IL) or tumor necrosis factor (TNF), oxidative stress-related responses such as nitric oxide synthase production and nitric oxide accumulation, acute phase proteins, components of the complement cascade, protein kinase activity (e.g., death associated protein kinase activity), neuronal cell 15 damage, and/or neuronal cell death, after the onset of cognitive deficits and Alzheimer's disease neuropathology in a subject comprising administering to the subject a therapeutically effective amount of a compound of the Formula I, II, I, IV, or V, a pharmaceutically acceptable salt thereof, or a composition comprising a compound of the Formula I, II, IH, IV, or V and a pharmaceutically acceptable carrier, excipient, or vehicle. 20 The invention provides a method of preventing a disease disclosed herein in a subject with a genetic predisposition to such disease by administering an effective amount of a compound of the Formula I, H, I, IV, or V, or a pharmaceutically acceptable salt thereof, or a composition comprising a compound of the Formula I, II, I, IV, or V and a pharmaceutically acceptable carrier, excipient, or vehicle. 25 The invention provides a method of improving memory of a healthy subject or the memory of a subject with age impaired memory by administering an effective amount of a compound of the Formula I H, III IV, or V, or a pharmaceutically acceptable salt thereof, or a composition comprising a compound of the Formula I, II, IH, IV, or V and a pharmaceutically acceptable carrier, excipient, or vehicle. 30 The further provides a method for improving memory, especially short-term memory and other mental dysfunction associated with the aging process comprising administering an effective amount of a compound of the Formula I, II, I, IV, or V, or a pharmaceutically acceptable salt thereof, or a composition comprising a compound of the Formula LI, I, IV, or V and a pharmaceutically acceptable carrier, excipient, or vehicle.
62 In an embodiment, a method is provided for treating a mammal in need of improved memory, wherein the mammal has no diagnosed disease, disorder, infirmity or ailment known to impair or otherwise diminish memory, comprising the step of administering to the mammal an effective memory-improving amount of a compound of the Formula I, HI, I11, IV, or V, a 5 pharmaceutically acceptable salt thereof, or a composition comprising a compound of the Formula I, II, III, IV, or V and a pharmaceutically acceptable carrier, excipient, or vehicle. In an aspect, the invention relates to a method of improving the lifespan of a subject suffering from Alzheimer's disease comprising administering a therapeutically effective amount of a compound of the Formula I, II, I, IV, or V, a pharmaceutically acceptable salt thereof, or a 10 composition comprising a compound of the Formula I, II, III, IV, or V and a pharmaceutically acceptable carrier, excipient, or vehicle. . In some aspects, greater efficacy and potency of a treatment of the invention may improve the therapeutic ratio of treatment, reducing untoward side effects and toxicity. Selected methods of the invention may also improve long-standing disease even when treatment is begun 15 long after the appearance of symptoms. The compositions and methods described herein are indicated as therapeutic agents or methods either alone or in conjunction with other therapeutic agents or other forms of treatment. They may be combined or formulated with one or more therapies or agents used to treat a condition described herein. Compositions of the invention may be administered concurrently, 20 separately, or sequentially with other therapeutic agents or therapies.Therefore, the compounds of the Formula I, II, II, IV, and/or V may be co-administered with one or more additional therapeutic agents including without limitation beta-secretase inhibitors, alpha-secretase inhibitors, and epsilon-secretase inhibitors, agents that are used for the treatment of complications resulting from or associated with a disease, or general medications that treat or Z5 prevent side effects. The invention also contemplates the use of a composition comprising at least one compound of the invention for the preparation of a medicament in treating a disease disclosed herein. In an embodiment, the invention relates to the use of a therapeutically effective amount of at least one compound of the invention for preparation of a medicament for providing 0 therapeutic effects, in particular beneficial effects, more particularly sustained beneficial effects, in treating a disorder or disease. In a still further embodiment the invention provides the use of a compound of the invention for the preparation of a medicament for prolonged or sustained treatment of a disease.
63 ADMINISTRATION Compounds and compositions of the present invention can be administered by any means that produce contact of the active agent(s) with the agent's sites of action in the body of a subject or patient to produce a therapeutic effect, in particular a beneficial effect, in particular a 5 sustained beneficial effect. The active ingredients can be administered simultaneously or sequentially and in any order at different points in time to provide the desired beneficial effects. A compound and composition of the invention can be formulated for sustained release, for delivery locally or systemically. It lies within the capability of a skilled physician or veterinarian to select a form and route of administration that optimizes the effects of the 10 compositions and treatments of the present invention to provide therapeutic effects, in particular beneficial effects, more particularly sustained beneficial effects. The compositions may be administered in oral dosage forms such as tablets, capsules (each of which includes sustained release or timed release formulations), pills, powders, granules, elixirs, tinctures, suspensions, syrups, and emulsions. They may also be administered [5 in intravenous (bolus or infusion), intraperitoneal, subcutaneous, or intramuscular forms, all utilizing dosage forms well known to those of ordinary skill in the pharmaceutical arts. The compositions of the invention may be administered by intranasal route via topical use of suitable intranasal vehicles, or via a transdermal route, for example using conventional transdermal skin patches. A dosage protocol for administration using a transdermal delivery system may be !0 continuous rather than intermittent throughout the dosage regimen. A sustained release formulation can also be used for the therapeutic agents. An amount of a therapeutic of the invention which will be effective in the treatment of a particular disorder or disease to provide effects, in particular beneficial effects, more particularly sustained beneficial effects, will depend on the nature of the condition or disorder, :5 and can be determined by standard clinical techniques. The precise dose to be employed in the formulation will also depend on the route of administration, and the seriousness of the disease, and should be decided according to the judgement of the practitioner and each patient's circumstances. Thus, the dosage regimen of the invention will vary depending upon known factors such 0 as the pharmacodynamic characteristics of the agents and their mode and route of administration; the species, age, sex, health, medical condition, and weight of the patient, the nature and extent of the symptoms, the kind of concurrent treatment, the frequency of treatment, the route of administration, the renal and hepatic function of the patient, and the desired effect.
64 Suitable dosage ranges for administration are particularly selected to provide therapeutic effects, in particular beneficial effects, more particularly sustained beneficial effects. A dosage range is generally effective for triggering the desired biological responses. The dosage ranges are generally about .5 mg to about 2 g per kg, about 1 mg to about 1 g per kg, about 1 mg to 5 about 200 mg per kg, about 1 mg to about 100 mg per kg, about 1 mg to about 50 mg per kg, about 10 mg to about 100 mg per kg, or about 30 mg to 70 mg per kg of the weight of a subject. A composition or treatment of the invention may comprise a unit dosage of at least one compound of the invention to provide beneficial effects. A "unit dosage" or "dosage unit" refers to a unitary i.e., a single dose which is capable of being administered to a patient, and 10 which may be readily handled and packed, remaining as a physically and chemically stable unit dose comprising either the active agents as such or a mixture with one or more solid or liquid pharmaceutical excipients, carriers, or vehicles. A subject may be treated with a compound of the Formula I, II, III, IV, or V or composition or formulation thereof on substantially any desired schedule. A composition of the 15 invention may be administered one or more times per day, in particular 1 or 2 times per day, once per week, once a month or continuously. However, a subject may be treated less frequently, such as every other day or once a week, or more frequently. A compound, composition or formulation of the invention may be administered to a subject for about or at least about 1 week, 2 weeks to 4 weeks, 2 weeks to 6 weeks, 2 weeks to 8 weeks, 2 weeks to 10 20 weeks, 2 weeks to 12 weeks, 2 weeks to 14 weeks, 2 weeks to 16 weeks, 2 weeks to 6 months, 2 weeks to 12 months, 2 weeks to 18 months, or 2 weeks to 24 months, periodically or continuously. The invention will be described in greater detail by way of specific examples. The following examples are offered for illustrative purposes, and are not intended to limit the 25 invention in any manner. EXAMPLES Example 1. General Materials and Methods Synthetic Chemistry. All reagents and solvents were used as purchased without further purification. 30 Syntheses were done using variations of established methods and in-parallel synthetic schemes. Briefly, diversification of position 3 of the pyridazine ring was done by reaction of a common halogenated pyridazine precursor. For the compounds used in this report, a mixture of 0.01 mol of substituted chloropyridazine, 0.05 mol of substituted piperazine and 0.01 mol of ammonium hydrochloride in 30 mL of 1-BuOH was stirred at 130*C for 48 h. The solvent was removed under 65 reduced pressure. The residue was then extracted with ethyl acetate, washed with water and brine, and dried over anhydrous NaiSO-i. Removal of solvent was followed by recrystallization from 95% ethanol. Amination of 3-chloro-6-phenylpyridazine by 2-(piperazin-l- yl)pyrimidine (Figure 1 2) easily 5 led to 2-(4-(6-phenylpyridazin-3-yl)piperazin-1-yl)pyrimidine (MW01-3-183WH): Light yellow crystals, yield 96.4%; HPLC: 97.4% purity; HRMS calculated 318.1587, found 318.1579; 1HNMR (CDC13): 5 8.356 (d, J=4.5, 2H), 8.011(d, J=7.5,11 2H), 7.692 (d, 3=9.5,1H), 7.468 (t, 5 J=6.0,2H), 7.417 (d, J=7.5, 111), 7.047 (d, J=9.5, 1H), 6.546 (t, J=4.5,1H), 4.013 (t, J=5.0,41H), 3.826 (tj=5.0,41). 0 Reactions were monitored by analytical HPLC (Rainin Instruments System Woburn, MA), done on a reverse phase column CIS (25 cm x 4.6 mm, 5 um, Supelco, Bellefbnte, PA) with two different UV wavelengths (X=260 nm and X=220 nm or 300 nm). Eluents were (A): 0.1% (v/v) TFA in water and (B) 80% (v/v) acetonitrile/water containing 0.08% TFA. A linear gradient of 10010 to 0/100 A/B over 34 min at ImIJmin was used. 1H-NMR spectra were obtained using Varian INOVA 15 (500 MHz) spectrometer. High resolution mass spectra were obtained on a VG70-250SE mass spectrometer. Cell Culture Assays. BV-2 mouse microglial cells (5x103 cells/well in a 48-well plate) were cultured and treated for 16 hrs with the standard glial activating stimulus lipopolysaccharide (LPS, from Salmonella typhimuriun; 100 ng/ml final concentration) in the presence or absence of 20 aminopyridazine compounds, as described previously [4,17]. EL-1/3 and TNFa levels in cell lysates were determined by electro-chemiluminescent detection in a Meso-Scale Discovery (MSD) kit, as per the manufacturer's instructions. In vivo Assays. AP-42 infusions and treatment of C57B1/6 mice with MWO1-2-151WH were performed as previously described [5]. Briefly, oligomeric A/31-42 was infused ICV for 28 25 days with a micro-osmotic pump. At post-operative day 21 and continuing for 14 days thereafter, mice were injected intraperitoneally once daily with either a test compound (2.5 mg/kg per day) or solvent control (10% DMSO in saline). At post-operative day 60, mice were perfused and sacrificed, and hippocampal endpoints measured as previously described [5]. Endpoint assays included immunohistochemical detection of activated astrocytes and microglia by glial fibrillary acidic protein 30 (GFAP) and F4/80 staining, measurement of the levels of the pro-inflammatory cytokines IL-1|S, TNFa, and S100B by ELISA, and determination of synaptic damage by analysis of the levels of postsynaptic density protein-95 (PSD-95). Brain Uptake Assays. MWO1-2-151WH was administered to mice (25-30g) by oral gavage using 2.5 mg/kg compound in 0.5% carboxymethylcellulose vehicle. At various times (0-60 min) after 66 administration, mice were sacrificed, blood removed by cardiac puncture, and brains immediately harvested, weighed, quick-frozen in liquid nitrogen, and stored at -80C until assayed. Brain tissue was homogenized in 1.5 ml of 0.1 M perchloric acid. After centrifugation (12,000 x g for 10 min), the supernatant was neutralized with 1 M NaOH and then extracted three times with 2 ml of 5 dichloromethane by centrifugation at 3,000 x g for 5 min. The organic phases from the three successive extractions were pooled and then vaporated to dryness under reduced pressure. The dried sample was reconstituted in 100 ul of HPLC mobile phase (80% acetonitrile, 0.08% formic acid, 20%
H
2 0), and 20 Al of the reconstituted material was injected into the HPLC system. The HPLC system for detection of MWO1-2-151WH was a Luna 5 pmCIS, 250 mm x 2 mm internal diameter column 10 together with a guard column (Phenomenex, Tonance, CA, USA), with HPLC solvent delivered at 0.2 mi/min (Dionex, model P680 pump) and absorbance monitored at 282 run (Dionex, model UVD 170U detector). Under these experimental conditions, the retention time of the test compound was 15.3 min. A standard curve of the test compound was prepared by adding increasing concentrations of the compound to brain tissue from untreated mice, then extracting the brains and performing HPLC 15 analysis as described above. The area under the curve increased linearly with the concentration of the compound over the range of concentrations investigated, with a correlation coefficient of 0.99. Under our experimental conditions, the compound was extracted reproducibly, with mean recoveries of 29 +/-2%. Graded dose, acute toxicity assays Vehicle (30% DMSO) or test compound (3.1, 12.5 or 50 20 mg/kg) in 0.5% carboxymethylcellulose was administered by oral gavage once daily for 3 days. On the 4th day, mice were anesthetized with pentobarbital, intubated and the lungs were inflated with an aircontaining syringe. The mice were perfused through the right ventricle and the lungs, liver and kidneys were then harvested and fixed in 4% paraformaldehyde for histology. Paraffinembedded hematoxylin & eosin stained sections of each organ were prepared by standard techniques. A 25 pathologist blinded to the treatment groups performed microscopic assessment of the tissue for injury. Results MWO1-3-183WH suppressed both IL-p and TNFa production in a concentration dependent manner (Figure 2). As shown in Figure 2 A and B, concentration dependent inhibition of proinflammatory cytokine production by MWO1-3-183WH1 in BV-2 cells were treated with LPS 30 (100 ng/ml) in the absence or presence of increasing concentrations of MWO1-3-183WH for 16 hrs with levels of IL-1f and TNFa in cell lysates measured by the Meso-Scale Discovery electrochemiluminescentdetection assay (See, Example 1., Materials and Methods). Data are the mean +/- SEM of triplicate determinations.
67 Figure 3 illustrates that MWO1-5-188WH is a concentration-dependent and selective inhibitor of proinflammatory cytokine production by activated glia and does not cause liver injury after chronic oral administration. As shown in Figure 3, concentration-dependent inhibition by MWO1-5-188WIH of LPS-induced increases in (A)IL-1p and (B) TNFa levels by 5 the BV2 microglial cell line. (C) Accumulation of the NO metabolite, nitrite, was not inhibited at concentrations up to 33 pM. MWO1-5-188WH (188) does not suppress iNOS, COX-2 or apoE production in activated glia, as evidenced by the representative western blots for (D) iNOS, (E) COX-2 and (F) apoE from glia cultures. Cultures were treated with control buffer, C, or activated in the absence, A, or presence, A+188, of 7.5 pM MW01-5-188WH. Daily oral 10 administration of MWO1-5-188WH does not bring about liver injury (G). Mice were administered either diluent or MWO1-5-188WH (2.5 mg/kg/day) for 2 weeks, then liver sections were stained with haematoxylin and eosin. Bar = 125 pim. Figure 4 illustrates that oral administration of MWOl-5-188WH suppresses human AP-induced neuroinflammation in mouse hippocampus in the absence of a detectable effect on the number of nitrotyrosine-labeled 15 neurons, or on amyloid plaque deposition. Oral administration of MW01-5-188WH suppreses human AP-induced neuroinflammation in mouse hippocampus in the absence of a detectable effect on the number of nitrotyrosine-labeled neurons, or on amyloid plaque deposition. A schematic of the experimental paradigm is shown. Daily oral administration (2.5 mg/kg) of MWO1-5-188WH (188-treated) for 2 weeks results in significant suppression of the human AP 0 induced increase in (A) IL- 10, (B) TNFt and (C) S100B levels in hippocampal extracts (n= 10 mice/group). MWO1-5-188WH treatment also decreased the number of (D) GFAP-positive activated astrocytes and (E) F4/80 positive microglia in the hippocampus. (F) MWO1-5-188WH treatment (188 treated) does not alter the profile of nitrotyrosine stained neurons, an indicator of oxidative stress linked injury. Representative micrographs are shown for hippocampus sections !5 stained for nitrotyrosine-positive neurons, from vehicle-infused mice, AP-infused mice (no treatment), and AP-infused mice orally administered MW0I-5-188WH (188-treated). Bars= 25 pm. The number (G) of amyloid plaques or the area occupied by amyloid plaques (H) is not altered by MW01-5-188WH therapy. Quantification of amyloid burden from all mice (n=5/group) was done by determination of the amyloid load. Data are mean & SEM. 0 Significantly different from AP-infused: *p<0.05, **p<0.01. Figure 5 illustrates that oral administration of MWO1-5-188WH attenuates hippocampal synaptic dysfunction and hippocampus-linked behavioral deficits. MWO1-5-188WH administration significantly attenuated the loss of synaptophysin and PSD-95, and the behavioral deficit in the Ymaze. Oral administration of MWO1-5-188WH attenuates hippocampal synaptic dysfunction and 68 hippocampus-linked behavioral deficits. A schematic of the experimental paradigm is shown in Figure 5. MWO1-5-188WH administration significantly attenuated the loss of synaptophysin and PSD-95, and the behavioral deficit in the Ymaze. Levels of (A) the pre-synaptic protein synaptophysin and (B) the post-synaptic protein PSD-95 were measured in hippocampal extracts 5 from vehicle-infused mice (control), Ap-infused mice (no treatment), and Ap-infused mice administered MWOl-5-188WH (188-treated) at 2.5 mg/kg by oral gavage once daily for 2 weeks. (C) Spontaneous alternation of mice in the Y-maze, a hippocampus-dependent spatial learning task, was measured for 10 days during the 7th and 8th week after the start of Ap infusion. Data are mean ยฑ SEM of n= 5 or 10 per group. Significantly different from Ap-infused 10 (*p<0.05, **p<0.01, ***p<0.001). MWO1-2-151SRM was found to suppress IL-10 in a concentration dependent manner. Immunoreactive levels of glial fibrillary acidic protein (GFAP), a marker of activated astrocytes were increased in Af.-infused mice (No treatment) compared to vehicle-infused mice (Control). The A0 infused increase in GFAP levels was suppressed by administration of MW01-2 15 151RSM. MWO1-2-151RSM also blocked the Ao-induced increase in the pro-inflammatory cytokines IL-10, TNF-ax, and S100B and the A$-induced loss of the pre-synaptic protein, synaptophysin and post-synaptic density protein PSD-95. These results are consistent with the results of the cell based assay. (See Figures 6 and 7.) Example 2. Acute and Chronic Toxicity Assays 20 Liver toxicity is an especially important initial consideration for orally administered compounds, as the liver is the major site of initial drug metabolism and is critical to overall metabolism and homeostasis of an animal. Liver injury is also a component of idiopathic tissue injury seen in certain chronically administered drugs. Therefore, it is important to do initial assessments of liver toxicity after oral administration of compounds to mice. 15 Methods: A standard approach is to test compounds in two initial in vivo toxicity assays: an acute, escalating-dose paradigm and a chronic, therapeutic dose regimen. For the escalating-dose, acute toxicity assays, mice (5 per experimental group) are administered either compound or vehicle in 0.5% carboxymethylcellulose (alternatively, castor oil or sesame oil can be used) by 10 oral gavage once daily for 3 days. Standard compound doses are 3.1, 12.5, and 50 mg/kg; the highest dose is 20X a therapeutic dose. On the 4f day, mice are sacrificed and the liver harvested and fixed for histology. Paraffin-embedded, hematoxylin & eosin (H&E)-stained sections of liver tissue are analyzed microscopically for injury by two individuals blinded to the 69 treatment groups. A semi-quantitative histological scoring system from 0 (best) to 9 (worst) is applied that considers architecture features (normal to extensive fibrosis), cellular features (normal to extensive edema and widespread necrosis), and degree of inflammatory infiltrate (normal to extensive infiltrate). For each acute toxicity assay, 15 mg of compound is required. 5 For the therapeutic dose, chronic toxicity assays, mice (5 per experimental group) are administered either compound or vehicle in 0.5% carboxymethylcellulose by oral gavage once daily for 2 weeks at a therapeutic dose of 2.5 mg/kg/day. After two weeks of treatment, mice are sacrificed and liver toxicity analyzed as described above. For each chronic toxicity assay, 5 mg of compound is required. 10 Results: MWO1-5-188WH has been tested in the acute, escalating-dose assay and the chronic, therapeutic dose assay. There was no histological evidence of tissue toxicity at the lower doses but some vacuolisation was observed at the 50 mg/kg dose. MWO1-2-151SRM has been tested in the chronic, therapeutic dose assay. There was no 15 histological evidence of tissue toxicity; no differences were seen by histology in livers from mice treated with vehicle or with compound. This compound is currently being tested in the acute, escalating dose assay. MW01-6-189WH has been tested in the chronic, therapeutic dose assay. There was no histological evidence of tissue toxicity; no differences were seen by histology in livers from 20 mice treated with vehicle or with compound. This compound is being tested in the acute, escalating dose assay. Example 3. hERG channel inhibition assays and cardiac OT interval assays Compounds have been screened for hERG (human ether-a-go-go) potassium ion channel binding and inhibition in order to eliminate early in the process any compounds with high 25 potential to induce prolongation of cardiac QT interval in later studies due to off-target toxicities. The hERG channel conducts rapidly activating delayed rectifier potassium currents that critically contribute to cardiac repolarization. Mutations in the hERG channel gene and drug-induced blockade of the currents have been linked to delayed repolarization of action potentials resulting in prolonged QT interval (Finlayson et al., 2004; Recanatini et al., 2005; 30 Roden, 2004). QT prolongation is considered a significant risk factor against cardiac safety of new drugs. Therefore, consideration of cardiac safety early in the development process by testing for hERG channel inhibition provides an efficient and predictive means to assess potential compound cardiac safety liabilities. In addition, the FDA (USA) is considering this as 70 an approval criteria in the future and has specific recommendations at this time. The assays done to date have been by a commercial service (MDS PharmaService). The initial assay is a radioligand binding assay that tests the ability of the test compound to compete with 3 H-astemizole (a reference standard that binds to hERG channels with nM 5 affinity) for binding to recombinant hERG channels stably expressed on human HBEK-293 cells. This cell line was chosen because it is of human origin, has been fully characterized with regard to hERG electrophysiology and pharmacology and displays the expected characteristics of IKr current as well as expected pharmacological sensitivities, and is easy to maintain in culture (Zhou et al., 1998). A single concentration (10 M) of test compound is assayed, and % 10 inhibition of 3 H-astemizole binding is calculated. Generally, any compounds that show >50% inhibition are tested further in the hERG channel activity assay. This is usual for medium throughout screens but is not recommended in the FDA document and tends to give false positives, as evidenced by the results reported below. The hERG channel activity inhibition assay provides whole cell electrophysiological 15 data about compound effects on the hERG K' channel function. Whole cell patch clamp methodology is generally considered to be the gold-standard determination of ion channel activity, rather than simply measuring channel binding. The standard testing procedure is to use 3 to 5 concentrations of compound at log dilutions with each concentration tested in triplicate (three cells). This allows a balance between achieving a reasonably accurate IC50 measurement 20 against a broad concentration range, and reducing cell attrition that would occur during more protracted experiment durations. After completion of compound dose-response procedures, a known hERG channel inhibitor, such as astemizole, is applied as a positive control. Compounds which exhibit inhibition of hERG channel activity are verified as positives (the hERG channel activity assay can give false positives and false negatives) by testing in vivo 5 for prolongation of cardiac QT interval. The QT interval studies are performed by evaluating compounds for effects on QT interval in Lead II electrocardiograms measured in anesthetized guinea pigs (Hirohashi et al., 1991), one of the species recommended in the FDA white paper. Vehicle or compound is administered orally at 15 mg/kg (dosing volume of 10 ml/kg) to groups of male guinea pigs (weighing 3 3 0-350g), with 5 animals per group. This dose corresponds ;0 approximately to 20-fold the therapeutic dose by taking into account the body surface area of the animals. Heart rate, arterial blood pressure, and QT intervals are measured at baseline, and at 15, 30, 45, and 60 min after compound administration. Sotalol administered iv at 0.3 mg/kg serves as the positive control compound. The QT intervals are corrected for changes in heart 71 rate using both Bazett's and Fridericia's formulae. Any increase in QT interval values over baseline values exceeding the upper 95% confidence limit of the mean changes at the corresponding time point in the vehicle-treated control group for two consecutive observation times indicates significant QT interval prolongation in the individually treated animals. This 5 functional testing in early discovery provides a rapid and cost-effective method to better anticipate and eliminate compounds that may have adverse QT prolongation potential in humans. Calculations of Amount of Compound Needed: Competition binding assay: 1-2 mg 10 Patch clamp assay: 1-2 mg QT interval assay: 5 mg/animal/dose= 25 mg per assay at 15 mg/kg dose Because the ex vivo activity assays are subject to false positives and negatives, it is considered better to complete studies of in vivo QT interval assay following the guidelines of the FDA position paper. 15 Results: Competition inhibition assay: MWOI-5-188WH, MWO1-2-151SRM, and MWO1-6-127WH were tested at 10pM concentration. MWO1-5-188WH showed 91% inhibition at 10 pM. MWO1-2-151SRM and MW01-6 20 127WH were negative, showing only 8% and 19% inhibition, respectively. Patch clamp inhibition assay: MWO1-2-15iSRM and MW01-6-189WH were tested at three concentrations (0.1, 1, 10 PM). These compounds showed minimal inhibition, with IC 50 values of 4.81 PM for MWO1-6 189WH and 9.21 pM for MWO1-2-151SRM. 25 Cardiac OT interval prolongation assay: MWO1-5-188WH and MWOl-2-151SRM were administered PO at 15 mg/kg to 5 guinea pigs
(
3 30-350g weight). QT intervals were obtained at baseline and at 15 min, 30 min, 45 min, and 60 min after compound administration. Neither compound increased cardiac QT interval above the mean + 2SD of corresponding values in the vehicle control group. There were also no ;0 significant effects on mean blood pressure or heart rate after compound administration. Example 4. Preparation of 2
-(
4
-(
6 -henvlinvridazin-3-vl)pinerazin-1-vl)nvrimidine (MWO1-3-183WH).
72 Figure 1 depicts a synthetic scheme for the preparation of 2
-(
4 -(6-phenylpyridazin-3-yl)piperazin-1 yl)pyrimidine (MWO1-3-183WH). Reagent and condition: (a) 1-BuOH, NH 4 C, and 2-( piperazin-1 yl)pyrimidine. A typical reaction mixture of comprised about 0.01 mol of 3-chloro-6 phenylpyridazine by 2 -(piperazin-1- yl)pyrimidine, about 0.05 mol of 2 -(piperazin-1-yl)pyrimidine 5 and about 0.01 mol of ammonium hydrochloride was prepared in about 15ml of 1-BuOH. The mixture was stirred at about 130"C for about 48h, and then the solvent was removed under reduced pressure. The remaining residue was then extracted with ethyl acetate, washed with water and brine, dried over anhydrous Na 2
SO
4 . Removal of solvent followed by recrystallization from 95% ethanol yielded light yellow crystals, yield 96.4%; HPLC: 97.4% purity; HRMS calculated 318.1587, 10 found 318.1579; 1H NMR (CDC13): 8 8.356 (d, J=4.5, 2H), 8.011(d, J=7.5, 11 2H), 7.692 (d, J=9.5, 1H), 7.468 (t, J=6.0, 2H), 7.417 (d, J=7.5, 1H), 7.047 (d, J=9.5, 1H), 6.546 (t, J=4.5, I1), 4.013 (t, J=5.0, 4H), 3.826 (t, J=5.0, 4H). Example 5. Biological Activity, Metabolic Stability and Toxicity of 4-methyl-6-phenyl-3-(4 15 Dyrimidin- 2 -vlinierazin-1-vI)Dyridazine (WO1-2-151SRM) As shown in Figures 6A-D and 7 A-G, the biological activity of MWO1-2-151SRM was investigated using the assays described herein for biological activity of the compounds. The compound was orally available, and targeted to glia reponses to protect against neurodeneration, but did not suppress the same inflammatory response endpoints outside the brain. The compound was a 20 selective suppressor of activated glia responses, especially key proinflammatory responses that have been linked to AD pathology. It also showed efficacy in the mouse model of human AP-induced neuroinflammation and neuronal injury as shown by the assays of Figure 7 A-G. The stability of MWO1-2-151SRM in microsomes, as shown in Figures 47 and 48, was demonstrated for MW01-2-151SRM. The stability of MWO1-5-188WH (1 yM) in a standard 5 incubation with rat liver microsomes (BD Biosciences) and an NADPH-regenerating system was done at 37*C for the times shown. Reactions were stopped by acetonitrile, and the reaction mixture was centrifuged at 16 000xg for 10 min. 10l of the supernatant was analyzed by calibrated HPLC to quantify the percentage of the initial amount of MWO1-2-151SRM remaining after the incubation. The HPLC system (Dionex Corp., Sunnyvale, CA) includes a Dionex P480 pump, a Phenomenex 0 Luna C18 column (250 x 2.0 mm, 50m) with a guard column (Phenomenex, Torrance, CA) and a Dionex UVD34OU Ultraviolet (UV) detector. The mobile phase consisted of 0.1% formic acid as reagent A and 0.08% formic acid/water in 80% acetonitrile as reagent B, at a flow rate of 0.2 ml per minute. The gradient consisted of the following linear and isocratic gradient elution changes in reagent B: isocratic at 60% from 0 to 5 min, 60% to 90% from 5 to 39 min, isocratic at 90% until 44 min. Peak 73 quantification was done based on absorption measured at 260 nm relative to a standard curve obtained by using serial dilutions of MWO1-2-151SRM. Liver toxicity after chronic in vivo administration of MW01-2-151SRM was investigated, see Figure 47 A. Mice were administered by oral gavage either MWO1-2-151SRM (2.5 mg/kg/day) or 5 diluent (10% DMSO) in a 0.5% (w/v) carboxymethylcellulose suspension once daily for two weeks. Mice were anesthetized and sacrificed. Livers were removed, fixed in 4% (v/v) paraformaldehyde and paraffin-embedded for histology. To assess histological toxicity, 4 Om liver sections were stained with haematoxylin and eosin. Two independent observers blinded to the treatment groups performed microscopic assessment of the tissue for injury. 10 Example 6. Preparation of N-(cvclopropVlmethVl)-6-phenvl4-(pyridin-4-vI)pyridazin-3 amine (MWO1-7-084WH). A synthetic scheme for the prepration of N-(cyclopropylmethyl)-6-phenyl-4-(pyridin-4 yl)pyridazin-3-amine (MW01-7-084WH) is depicted in Figure 8, and synthesis was carried out 15 as described herein. 4 -chloro-6-phenylpyridazin-3(2H)-one (MWO1-6-093WH) 4 -chloro-6-phenylpyridazin-3(2H-one was synthesized according to the procedure described by Coudert, P. [18]. 4-chloro- 2 -(methoxymethyl)-6-henylpydazin-3(2H)-one (MW01-7-053WH) 20 A mixture of chloropyridazinone 1 (25.5 g, 0.12mol), 4-NN-dimethylaninopyridine (0.20 g) and i Pr2NEt (26.7g, 0.21mol) in anhydrous CH 2 Cl 2 (300mL) was stirred at 0"C (ice bath) for 30 mi. Methoxymethyl chloride (25g, 0.31mol) was added and the mixture was stirred at 0 iiC for 1h and then allowed to warm to room termperature. The reaction was stirred at room temperature till complete. The solvent was then removed in vacuo, the residue was treated with water, washed with 25 dilute Na 2
CO
3 solution and extracted with EtOAc. The organic layer was dried over anhydrous Na 2
SO
4 , filtered and evaporated. The residue was then purified by recrystallization from 95% ethanol to give 20.1 light yellow solid. Yield 66.9%. 6 -phenyl- 4 -(pyridin-4-vl)pridazin-3(2H)-one (MW01-7-069 WH) The protected pyridazinone MWO1-7-053WH (l.Oequiv.) was mixed with arylboronic acid 30 (1.37equiv.), Pd(PPh3)4 (0.05 equiv.) and K2C03 (3.1equiv) and 200 mL of DME in a 350ml of pressure vessel, flushed with argon for 3 min, and the mixture was then stirred and refluxed (oil bath, 120"C) until the starting material had disappeared. After cooling, the solution was concentrated to dryness under reduced pressure, the residue was treated with water and filtered off. The filter cake was washed with water over filter funnel and then used for next step directly. The residue obtained above 74 was dissolved in 200ml of EtOH, 6 N HCl (200 mL) was added and the reaction mixture was refluxed (oil bath, 120 i9C) for 6 h, then it was allowed to cool to room termperature, and concentrated to dryness under reduced pressure. The residue was neutralized with dilute NaOH solution. The suspension was then filter off, washed with water and dried over filter funnel. Recrystallization from 5 90% ethanol provided brown yellow solid. Yield 80.4%. ESI-MS: m/z 294.3 (M+H+) 3-chloro6 (MW1-7-076WHg) 3 -chloro-6-phenyl-4-(pyridin-4-yl)pyridazine (MW01-7-076WH) (66mmol) was suspended in 75ml phosphorus oxychloride and heated with stirring at 100"C for 3h. After cooling to room temperature the mixture was poured onto crushed ice. The mixture was then neutralized with 10 NaOH solution to give white suspension. The precipitation was filtered off, washed with water, dried over filter funnel to yielding a light yellow solid. ESI-MS: m/z 268.4 (M+H+). yridin-4-v1)pyridazin-3-an (MW1-7-084WH) A mixture of N-(cyclopropylethyl)-6-phenyl-4-(pyridin-4-yl)pyridazin-3-amine (MW01-7 084WH) ( 0.5mmol), C-Cyclopropyl-methylamine (2.Ommol) in 3ml of 1-BuOH was heated 15 with stirring at 1300* for 7days. The solvent was removed by evaporation in vacuo, the residue was treated with water to give a suspension. The solid was then filtered off, washed with water, then 1:3, Ethyl Acetate: Petroleum ether, dried over filter funnel in vacuo yielding gray solid. ESI-MS: m/z 330.4 (M+H+). 20 Example 7. Preparation of 3
-(
4 -methylpierazin-1-vl)-6-henvI-4-(avridin-4-vl)pvridazine (MWO1-7-085WH). A mixture of 3-chloro-6-phenyl-4-(pyridin-4-yl)pyridazine (MW01-7-076WH) (0.5mmol), 1 methyl-piperazine (2.Ommol) in 3ml of 1-BuOH was heated with stirring at 130"C for about 7 days. The solvent was removed by evaporation in vacuo, the residue was treated with water to 25 give a suspension. The solid was then filtered off, washed with water, then 1:3, Ethyl Acetate: Petroleum ether, dried over filter funnel in vacuo to yield a brown solid. ESI-MS: m/z 332.2 (M+H). A synthetic reaction scheme for the preparation of 3 -(4-methylpiperazin-1-yl)-6 phenyl-4-(pyridin-4-yl)pyridazine (MWO1-7-085WH) is depicted in Figure 9. 30 Example 8. Prearation of N-( 2 -mornholinoethvl)-6-phenvl-4-(Pvridin-4-vl)pvyridazin-3 amine (MWO1-7-091WH). A mixture of 3-chloro-6-phenyl-4-(pyridin-4-yl)pyridazine (MWO1-7-076WH) (0.5mmol), 2 Morpholin-4-yl-ethylamine (2.0 mmol) in 3ml of 1-BuOH was heated with stirring at 130"C for about 7 days. The solvent was removed by evaporation in vacuo, the residue was treated with 75 water to give a suspension. The solid was then filtered off, washed with water, then 1:3, Ethyl Acetate: Petroleum ether, dried over filter funnel in vacuo to yield a gray solid. ESI-MS: m/z 362.2 (M+W). A synthetic reaction scheme for the preparation of 3-(4-methylpiperazin-1-yl)
N-(
2 -morpholinoethyl)-6-phenyl-4-(pyridin-4-yl)pyridazin-3-amine (MWO1-7-091WH) is 5 depicted in Figure 10. Example 9. Preparation of 5-(4-Fluorophenvl)-3-phenyl-6-(4-pvrimidin-2-vlpiperazin-1 Vl)pyridazine (MWO1-2-065LKM). A synthetic reaction scheme for the preparation of 5-(4-Florophenyl)-3-phenyl-6-(4-pyrimidin 10 2 -ylpiperazin-1-yl)pyridazine (MW01-2-065LKMv) is depicted in Figure 11, and synthesis was carried out as described herein. 3 -phenl- 6 -(4-pyimidin-2-lpiperazin--ylpridazin-5-o1 (MWO1-6-006WH) This compound was prepared from 3-chloro-5-hydroxy-6-phenylpyridazine (1.4g, 6.8mmol) in the same manner as described for MWO1-6-121WH, yielding white solid (2.12g, 6.15mmol, 90.4%). 15 MALDI-TOF: m/z 335.7 (M+H+). 1H NMR (DMSO): d 8.433 (t, J=2.0, J=2.4, 211), 7.773 (d, J=3.2, 2), 7.497 (t, J=2.0, J=3.6, 3H), 7.182 (s, 1H), 6.727 (s, 1H), 3.949 (s, 411). 5-chloro-3-phenyl-6-(4-pyrim-2-vlpiperazin-1-vl)prdazine (MW01-6-015WH) 3 -chloro-5-hydroxy-6-phenylpyridazine (66mmol) was suspended in 75ml phosphorus oxychloride and heated with stirring at 100"C for 3h. After cooling to room temperature the 20 mixture was poured onto crushed ice. The mixture was then neutralized with NaOH solution to give white suspension. The precipitation was filtered off, washed with water, dried over filter funnel to yield white solid (98.8%). ESI-MS: m/z 353.3 (M+H+). 1H NMR (CDCl3): d 8.375 (d, J=5.0, 2H), 7.776 (d, J=7.0, 2H), 7.487 (m, 3H), 7.073 (s, 111), 6.588 (t, J=4.5, 1H), 4.046 (t, J=4.5, J=5.5, 4H), 3.849 (t, J=5.5, J=5.0, 41). 25 5-(4-Horophenyll-3-phenyl-6-(4-pyrimidin-2-vlpiperazin-l-vlpyridazine (MW1-2-065LKM) This compound was prepared in the same manner as described for MWO1-7-069WHWH, yielding a white solid (60.4%). MAIDI-TOF: m/z 413.4 (M+H+). Example 10. Preparation of 5-(4-vridyl)-3-henvl-6-(4-yrimidin-2.ylpiperazin- 30 vl)pyridazine (MW1-2-069A-SRM). A synthetic reaction scheme for the preparation of 5-(4-pyridyl)-3-phenyl-6-(4-pyrimidin-2 ylpiperazin-1-yl)pyridazine (MWOl-2-069A-SRM) is depicted in Figure 12, and synthesis was carried out as described herein. This compound was prepared in the same manner as described for MWO1-2-065LKM, yielding white solid (65.4%). MALDI-TOF: m/z 396.2 (M+H+).
76 Example 11. Preparation of 4 -methyl-6-phenyl-3-(4-pyrimidin-2-ylpiperazin-1 yl)pyridazine (MWO1-2-151SRM). 4-methyl-6-phenyl-3-(4-pyrmidin-2-ylpiperazin-1-yl)pyridazine (MWO1 -2-151SRM) was 5 prepared by several synthetic schemes as depicted in Figure 13 (Scheme 1), Figure 14 (Scheme 2), and Figure 15 (Scheme 3), which were carried out as described in detail herein. The various reaction schemes (Schemes 1, 2, and 3) are generally applicable to the compounds of the present invention and are not restricted in utility only to the preparation of MW01-2-151SRM. Scheme 1 10 3 -chloro-6-phenylpyridazin-4-ol was synthesized according to the procedure described by Coudert, P., et al. [18]. 6 -:phenyl-3-44-(p3midin-2-yl)p*erazin-1-vl)pyridazin-4-ol (MW01-7-121WH) This compound was prepared from 3 -chloro-4-hydroxy-6-phenylpyridazine (14g, 68mmol) in the same manner as described below, yielding white solid (22.1g, 66mmol, 97.3%). ESI-MS: 15 m/z 335.2 (M+H+). 1H NMR (DMSO): 1H NMR (DMSO): d 8.406 (d, J=6.5, 2H), 7.740 (d, J=4.0, 2H), 7.558 (s, 3H), 6.686 (t, J=4.8, J=4.4, 1H), 6.841 (s, 1H), 3.881 (s, 4H), 3.620 (s, 4H), 3.776 (s, 411). yn-2-ylpiperazin-1-vl)pyridazine (MWO1-6-127WH) 6-phenyl-3-(4-pyrimidin-2-ylpiperazin-1-yl)pyridazin-4-ol (22.0g, 66mmol) was suspended in 20 75ml phosphorus oxychloride and heated with stirring at 100"C for 3h. After cooling to room temperature the mixture was poured onto crushed ice. The mixture was then neutralized with NaOH solution to give white suspension. The precipitation was filtered off, washed with water, dried over filter funnel to provide white solid (21.3g, 60.3mmol, 91.4%). ESI-MS: m/z 353.4 (M+H+). 1H NMR (CDCI3): d 8.377 (d, J=4.5, 2H), 8.036 (d, J=7.5, 2H), 7.833 (s, 1H), 7.508 25 (m, 3H), 6.564 (t, J=4.5, 111), 4.073 (t, J=4.0, J=4.5, 4H), 3.672 (t, J=4.0, J=4.5, 411). 4-methyl-6-phenyl-3-(4-pyrimidin-2-vlpiperazin-1-vllpyrdazine (MW01-2-151SRM) Into a reaction tube were added MW01-6-127WH (1.4g, 4.Ommol), K2C03 powder (1.7g, 12.4mmol), Pd(dppf)C12 (326mg, 0.4mmol), silver oxide (2.3g, 10mmol), methylboronic acid (324mg, 5.4mmol) and 20ml of THF. Argon was then flushed through the tube for 3min. The 30 tube was then sealed tightly and heated with stirring at 80 degree for 12h. After cooled down, the mixture was quenched with 10% NaOH solution and extracted with ethyl acetate. The organic phase was concentrated in vacuo and the residue was purified by column chromatography eluting with 1:4, Ethyl Acetate: Petroleum ether. White powder solid was obtained (0.60g, 1.8mmol, yield 45.2%). ESI-MS: m/z 333.4 (M+H+). 1H NMR (CDCl3): d 77 8.380 (d, J=5.0, 2H), 7.065 (d, J=7.0, 2H), 7.626 (s, 1H), 7.473 (m, 3H), 6.567 (t, J=4.5, J=5.0, 111), 4.056 (t, J=5.0, 4H), 3.475 (t, J=5.0, 41), 2.456 (s, 3H). Scheme 2 Into a reaction tube were added MWO1-6-127WH (1.4g, 4.0mmol), K2C03 powder (1.7g, 5 12.4mmol), Pd(PPh 3
)
4 (240mg, 0.2mmol), silver oxide (2.3g, 10mmol), methylboronic acid
(
3 2 4mg, 5.4mmol) and 20ml of DME. Argon was then flushed through the tube for 3min. The tube was then sealed tightly and heated with stirring at 120*C for 24h. After cooled down, the mixture was filter through acelite earth, the filtrate was then concentrated and the residue was purified by column chromatography eluting with 1:4, Ethyl Acetate: Petroleum ether. White 10 powder solid was obtained (0.64g, 1.93mmol, yield 48.1%). ESI-MS: m/z 333.4 (M+H+). 1H NMR (CDCl3): d 8.380 (d, J=5.0, 211), 7.065 (d, J=7.0, 2H), 7.626 (s, 1H), 7.473 (m, 3H), 6.567 (t, J=4.5, J=5.0, 1H), 4.056 (t, J=5.0, 4H), 3.475 (t, J=5.0, 411), 2.456 (s, 3H). Scheme 3 yrin (MW01-8-004WH 15 7.7 g (40 mmole) of 2 -methyl-4-oxo-4-phenylbutanoic acid was added to a 100 ml single necked round bottom flask followed by 3.0 ml (60 mmole) of hydrazine monohydrate and then 20 ml of reagent grade ethanol (100%, 95% of ethanol should be fine also). The flask was fitted with a reflux condenser and the reaction mixture was heated to reflux in an oil bath at 110 C (temperature of oil bath) and stirred for 2 h. The flask was then removed from the oil bath and 20 the reaction mixture cooled to ambient temperature. The stir bar was removed and the solvent was evaporated in vacuo in a water bath at 45"C. The residue was then treated with 50 ml of Milli-Q water and stirred for 10 minutes to give a suspension. The precipitate was collected by filtering, washed with 100 ml of 2N NaHCO 3 , then washed with 60 ml Milli-Q water three times, and dried over a medium frit sintered glass funnel in vacuo to give 7.15 g of white 25 crystals (Syn. ID, WH-8-004). Yield, 95%, confirmed by ESI-MS. ESI-MS: m/z 189.2 (M+H+). 4 -methyl-6-phenylpyridazin-3(2)-one (MWO1-8-008WH) 7.0 g (35 mmole) of MWO1-8-004WH was placed in a 100 ml single-necked round bottom flask followed by 9.4 g (70 mmole) of anhydrous copper (II) chloride and then 30 ml of acetonitrile to give a brown yellow suspension. A reflux condenser was connected to the flask and a dry tube 30 filled with CaCl2 was fitted to the top of the condenser. The reaction mixture was heated to reflux in an oil bath (1 10*C) for 3 h. The color of the reaction suspension changed to dark yellow once the reflux started. After the completion of the reaction (monitored by HPLC), the flask was removed from the oil bath and cooled to ambient temperature. The mixture was poured on to 300g of crushed ice and stirred vigorously for 10 minutes to give a gray precipitate 78 and blue liquid. The precipitate was then collected by filtering (pH of the filtrate was 1.5-2.0), and washed with 100 ml of a IN HC1 solution to rid the solid of any remaining copper byproducts. This is followed by washing with 100 ml of Milli-Q water to get rid of the acid in the solid, and is monitored by checking the pH value of the filtrate. The solid was washed until 5 the filtrate shows a pH of 7, after approximately 5 washes. The solid was dried over a medium frit sintered glass funnel in vacuo to give 6.3 g of a blue gray solid. Yield was 96.7% and confirmed by ESI-MS. ESI-MS: m/z 187.3 (M+H+). 3-chloro-4-methyl-6-pnvlpyridazine(MW 1-8-012WH) 6.0 g (32 mmole) of MWO1-8-008WH and 30 ml (320 mmole) of phosphorus oxychloride were 10 placed in a 100 ml single-necked round bottom flask. The flask was connected with a reflux condenser and a dry tube filled with anhydrous CaC 2 was fitted to the top of the condenser. (HC1 gas is formed in the reaction so a basic solution such as NaOH may be needed to absorb HC1 in a large-scale synthesis). The reaction mixture was stirred in an oil bath (90"C) for 2 h, then cooled to ambient temperature and poured onto crushed ice. (phosphorus oxychloride can 15 be decomposed by water to give HC1 and H 3 P0 4 ). The mixture was then stirred vigorously for 10 minutes to give a white suspension. The suspension was neutralized with a 2N NaOH solution until the pH of the suspension was pH = 7. The precipitate was filtered, washed three times with 100 ml of Milli-Q water and dried over a medium frit sintered glass funnel in vacuo to provide 5.9 g of a light pink powder (Syn. ID, WH-8-012). Yield was 89.4% and confirmed 20 by ESI-MS. ESI-MS: m/z 205.4 (M+H+). 2
-(
4
-(
4 -methyl-6-phenlpydazin-3-l)piperazin-1-l)pyrimidine (MWO1-2-151SRM) 0.82 g (4.Ommole) of WH-8-012 was placed in a 30 ml pressure vessel followed by addition of 2.6 g (16.Ommole) of 1-(2-pyrimidyl) piperazine and then 15 ml of 1-BuOH. The vessel was sealed tightly and placed into an oil bath and stirred at 130 C (temperature of oil bath) for 2.5 25 days. The reaction mixture was then cooled to ambient temperature and transferred to a single necked flask for evaporation under reduced pressure. Removal of solvent gave rise to a brown red residue that was treated with 30 ml of water to give a brown sticky oil. The mixture was kept at ambient temperature overnight while the oil solidified gradually. The formed solid was then broken into small pieces with a steel spatula. The solid was collected by filtering and 0 washed with 50 ml of Milli-Q water three times and dried over a filter funnel in vacuo to provide 1.25 g of light yellow solid (Syn. ID, W.H-8-020). Yield was 94%. (Alternative separation is to use precipitation procedure instead of solidification process. Solidification is a simple and cheap operation, yet time-consuming. Precipitation is time efficient, yet more costly than the former one. So it is up to the process chemist to decide which procedure to pick for the 79 manufacture. The precipitation process is below: The oil product was dissolved completely in 10 ml of reagent grade ethanol or acetone to form a solution. The solution was then added dropwise to 150 ml of ice water under vigorous stirring. Light yellow suspension was then formed gradually. The solid was collected by filtering, washed with Milli-Q water, dried over 5 filter funnel in vacuo to give the desired product.) The final compound was confirmed by ESI MS and NMR. ESI-MS: m/z 333.8 (M+H+). 1H NIR (CDCl3): d 8.380 (d, J=5.0, 21), 7.065 (d, J=7.0, 2H), 7.626 (s, 1H), 7.473 (m, 311), 6.567 (t, J=4.5, J=5.0, 111), 4.056 (t, J=5.0, 4H), 3.475 (t, J=5.0, 4H), 2.456 (s, 3H). 10 Example 12. Prepartion of 4
,
6 -diphenvl-3-(4-pyrimidin-2-vlpiperazin-1-vI)pyridazine (MWO1-5-188WH). 4,6-dphenyl-3-(4-pyrimidin-2-ylpiperazin-1-yl)pyridazine (MW01-5-188WH).was prepared by several synthetic schemes as depicted in Figure 16 (Scheme 1), Figure 17 (Scheme 2), and Figure 18 (Scheme 3), which were carried out as described in detail herein. The various 15 reaction schemes (Schemes 1, 2, and 3) are generally applicable to the compounds of the present invention and are not restricted in utility only to the preparation of MW01-2-188WH. Scheme 1 3 -chloro-6-phenylpyridazin-4-ol was synthesized according to the procedure described by Coudert, P., et al. [18]. 20 6 -phenyl- 3 -(4-(pyrimidin-2-yl)piperazin-l-vl)pyridazin-4-ol (MW01-7-121WH) The compound was prepared from 3 -chloro-4-hydroxy-6-phenylpyridazine (14g, 68mmol). A mixture of 3 -chloro-4,6-diphenylpyridazine (267mg, 1.0mmol), 1-(2-pyrimidyl)piperazine (656mg, 4.Ommol) in 3ml of 1-BuOH was heated with stirring at 130*C for 3days. The solvent was removed by evaporation in vacuo, the residue was treated with water to give a suspension. 25 The solid was then filtered off, washed with water, dried over filter funnel in vacuo to give light pink solid. yielding white solid (22.1g, 66mmol, 97.3%). ESI-MS: m/z 335.2 (M+H+). 1H NMR (DMSO): 1H NMR (DMSO): d 8.406 (d, J=6.5, 2H), 7.740 (d, J=4.0, 2H), 7.558 (s, 3H), 6.686 (t, J=4.8, J=4.4, 111), 6.841 (s, 1H), 3.881 (s, 4H), 3.620 (s, 4H), 3.776 (s, 4H). 4 -chloro- 6 -phenl-34-vriidin-2ylpiperazin-1-vl)pyridazine (MWO1-6-127WH) 30 6-phenyl-3-(4-pyrimidin-2-ylpiperazin-1-yl)pyridazin-4-ol (22.0g, 66mmol) was suspended in 75ml phosphorus oxychloride and heated with stirring at 100*C for 3h. After cooling to room temperature the mixture was poured onto crushed ice. The mixture was then neutralized with NaOH solution to give white suspension. The precipitation was filtered off, washed with water, dried over filter funnel to provide white solid (21.3g, 60.3mmol, 91.4%). ESI-MS: m/z 353.4 80 (M+H+). 1HI NMR (CDCl3): d 8.377 (d, 1=4.5, 2H), 8.036 (d, J=7.5, 2H), 7.833 (s, 11), 7.508 (n, 311), 6.564 (t, J=4.5, 1H), 4.073 (t, J=4.0, J=4.5, 411), 3.672 (t, J=4.0, J=4.5, 41). 4,6-diphenyI-3-(4-pyrimidin-2-vlpiperazin-1-vIpyrdazine (M 01-5-188WH m (MWl-1 8WH) A mixture of 3 -chloro- 4 ,6-diphenylpyridazine (267mg, 1.Ommol), 1-( 2 -pyrimidyl)piperazine 5 ( 6 56mg, 4.Ommol) in 3ml of 1-BuOH was heated with stirring at 130*C for 3days. The solvent was removed by evaporation in vacuo, the residue was treated with water to give a suspension. The solid was then filtered off, washed with water, dried over filter funnel in vacuo to give light pink solid. ( 3 20mg, 0.81mmol, yield 81.1%). ESI-MS: m/z 395.5 (M+H+). HRMS calcd 395.1979, found 395.1973; 1H NMR (CDCl3): d 8.329 (d, J=5.0, 211), 8.101 (d, J=7.5, 211), 10 7.734 (d, J=7.5, 211), 7.655 (s, 1H), 7.509 (m, 611), 6.530 (t, J=4.5, 111), 3.836 (t, J=4.5, J=5.0, 4H), 3.394 (t, J=5.0, J=4.5, 411). Scheme 2 4,5-dihydro-6-phenvl-4-phenvyridazin-3(2H)-one 135 ml (135 mmole) of a solution of phenylmagnesium bromide (IM) in THF was added to a 15 hot suspension of 6 -phenylpyridazinone compound 7.8g (45 mole) in dry toluene (50 ml). The mixture was refluxed for 8h, left overnight at ambient temperature, then decomposed with a saturated solution of ammonium chloride. The organic layer was separated, and the aqueous layer was extracted with 100ml of ethyl acetate. The solvent was removed and the residue was crystallized from ethanol. The crystals were collected by filtering and dried over a medium fit 20 sintered glass funnel in vacuo to give 5.6 g of white crystals. Yield was 50%, confirmed by ESI MS. ESI-MS: m/z 250.1 (M+H+). 6 -phenyl-4-phenylpyridazin-3(2H)-one 4.4 g (17.5 mmole) of 6 -pyridazinone obtained above was placed in a 50 ml single-necked round bottom flask followed by 4.7 g (35 mmole) of anhydrous copper (II) chloride and then 20 25 ml of acetonitrile to give a brown yellow suspension. A reflux condenser was connected to the flask and a dry tube filled with CaCl2 was fitted to the top of the condenser. The reaction mixture was heated to reflux in an oil bath (110 C) for 3 h. The color of the reaction suspension changed to dark yellow once the reflux started. After the completion of the reaction (monitored by HPLC), the flask was removed from the oil bath and cooled to ambient 30 temperature. The mixture was poured on to 200 g of crushed ice and stirred vigorously for 10 minutes to give a gray precipitate and blue liquid. The precipitate was then collected by filtering (pH of the filtrate was 1.5-2.0), and washed with 50 ml of a 1N HCl solution to rid the solid of any remaining copper byproducts. This is followed by washing with 100 ml of Milli-Q water to get rid of the acid in the solid, and is monitored by checking the pH value of the filtrate.
81 The solid was washed until the filtrate shows a pH of 7, after approximately 5 washes. The solid was dried over a medium frit sintered glass funnel in vacuo to give 3.9 g of a blue gray solid. Yield was 90%, confirmed by ESI-MS. ESI-MS: m/z 248.1 (M+H+). 3-chloro-6-phenyl-4-phenylpyridazine 5 2.0 g (8 mmole) of 6-phenylpyridazinone obtained above and 10 ml (54 mmole) of phosphorus oxychloride (reagent grade, Aldrich) were placed in a 50 ml single-necked round bottom flask. The flask was connected with a reflux condenser and a dry tube filled with CaCl2 was fitted to the top of the condenser. (HC1 gas is formed in the reaction so a basic solution such as NaOH may be needed to absorb HCl in a large-scale synthesis). The reaction mixture was stirred in an 10 oil bath (90 C) for 2 h, then cooled to ambient temperature and poured onto crushed ice. (phosphorus oxychloride can be decomposed by water to give HCI and H3P04). The mixture was then stirred vigorously for 10 minutes to give a white suspension. The suspension was neutralized with a 2N NaOH solution until the pH of the suspension was pH = 7. The precipitate was filtered, washed three times with 100 ml of water and dried over a medium frit 15 sintered glass funnel in vacuo to provide 1.8 g of a light pink powder. Yield was 85%, confirmed by ESI-MS. ESI-MS: m/z 266.4 (M+H+). 2-(4-(6-phenyl-4-phenylpyridazin-3-ylpiperazin-1-yl)pyriidine 1.1 g (4.Ommole) of 3-chloropyridazine obtained above was placed in a 30 ml pressure vessel followed by addition of 2.6 g (16.0mmole) of 1-(2-pyrimidyl) piperazine and then 15 ml of 1 20 BuOH (reagent grade). The vessel was sealed tightly and placed into an oil bath and stirred at 130 0 C (temperature of oil bath) for 3 days. The reaction mixture was then cooled to ambient temperature and transferred to a single-necked flask for evaporation under reduced pressure. Removal of solvent gave rise to a brown-red residue that was treated with 30 ml of water to give a brown suspension. The solid was collected by filtering and washed with 50 mL of water three 25 times and dried over a filter funnel in vacuo to provide 0.96 g of light yellow solid. Yield was 90%, ESI-MS: m/z 395.5 (M+H+). BRMS calcd 395.1979, found 395.1973; 1H NMR (CDC3): d 8.329 (d, J=5.0, 2H), 8.101 (d, J=7.5, 2H), 7.734 (d, J=7.5, 2H), 7.655 (s, 111), 7.509 (m, 6H), 6.530 (t, J=4.5, 11), 3.836 (t, 1=4.5, J=5.0, 4H), 3.394 (t, J=5.0, J=4.5, 41). Scheme 3 30 3-chloro-6-phenylpyridazin-4-ol was synthesized according to the procedure described by Coudert, P., et al. [18]. 4,6-diphenyl-3-(4-pyrimidin-2-ylpiperazin-1-yl)pyridazine (MW01-5-188WH) A mixture of 3-chloro-4,6-diphenylpyridazine (267mg, 1.0mmol), 1-(2-pyrimidyl)piperazine (656mg, 4.0mmol) in 3ml of 1-BuOH was heated with stirring at 130 C for 3days. The solvent 82 was removed by evaporation in vacuo, the residue was treated with water to give a suspension. The solid was then filtered off, washed with water, dried over filter funnel in vacuo to give light pink solid. (320mg, 0.81mmol, yield 81.1%). ESI-MS: m/z 395.5 (M+H+). HRMS calcd 395.1979, found 395.1973; 1H NMR (CDCl3): d 8.329 (d, J=5.0, 2H), 8.101 (d, J=7.5, 211), 5 7.734 (d, J=7.5, 2H), 7.655 (s, 1H), 7.509 (m, 61), 6.530 (t, 1=4.5, 1H), 3.836 (t, 1=4.5, J=5.0, 4H), 3.394 (t, J=5.0, J=4.5, 4H). Example 13. Prepartion of 4 -vyridvl-6-phenv-3-(4-pvrimidin-2-ylvperazin1.
VA)pyridazine (MWO1-6-189WH). 10 4-pyridyl-6-phenyl-3-(4-pyrmidin-2-ylpiperazin-1-yl)pyridazine (MWO1-6-189WH) was prepared by two synthetic schemes as depicted in Figure 19a and 19b, which were carried out as described in detail herein. The various reaction schemes (Schemes 1 and 2) are generally applicable to the compounds of the present invention and are not restricted in utility only to the preparation of MWO1-2-189WH. 15 ' Scheme 1 3 -chloro-6-phenylpyridazin-4-ol was synthesized according to the procedure described by Coudert, P., et al. [18]. 6Ypiprzn---pyiain4o (MW0-7-121WH) This compound was prepared from 3 -chloro-4-hydroxy-6-phenylpyridazine (14g, 68mmol) A 20 mixture of 3 -chloro-4,6-diphenylpyridazine (267mg, 1.Ommol), 1-( 2 -pyrimidyl)piperazine (656mg, 4.Ommol) in 3ml of 1-BuOH was heated with stirring at 130*C for 3days. The solvent was removed by evaporation in vacuo, the residue was treated with water to give a suspension. The solid was then filtered off, washed with water, dried over filter funnel in vacuo to give light pink solid. yielding white solid ( 2 2.lg, 66mmol, 97.3%). ESI-MS: m/z 335.2 (M+H+). 1H 25 NMR (DMSO): 1H NMR (DMSO): d 8.406 (d, J=6.5, 2H), 7.740 (d, J=4.0, 211), 7.558 (s, 3H), 6.686 (t, J=4.8, J=4.4, 1H), 6.841 (s, 1H), 3.881 (s, 4H), 3.620 (s, 4H), 3.776 (s, 41). 4 -eyL~3-(4 nvlpiperazin-1-vl)pyridazine (MW1-6-127WH) 6-phenyl-3-(4-pyrimidin-2-ylpiperazin-1-yl)pyridazin-4-ol 1h (22.0g, 66mmol) was suspended in 75ml phosphorus oxychloride and heated with stirring at 100 for 3h. After cooling to room 30 temperature the mixture was poured onto crushed ice. The mixture was then neutralized with NaOH solution to give white suspension. The precipitation was filtered off, washed with water, dried over filter funnel to provide white solid (21.3g, 60.3mmol, 91.4%). ESI-MS: m/z 353.4 (M+H+). 1H NMR (CDCl3): d 8.377 (d, 1=4.5, 2H), 8.036 (d, J=7.5, 2H), 7.833 (s, 1H), 7.508 (m, 31), 6.564 (t, J=4.5, 111), 4.073 (t, J=4.0, J=4.5, 4H), 3.672 (t, J=4.0, J=4.5, 4H).
83 4-pyrdyl-6-pMhenyl-3-(4-pyrimidin-2-vlpiperazin-1-vlpyaine W01-6-189WH) Into a reaction tube were added WH-6-127 (1.4g, 4.0mmol), K2C03 powder (1.7g, 12.4mmol), Pd(PPh3)4 (240mg, 0.2mmol), 4 -pyridineboronic acid (664mg, 5.4mmol) and 20ml of DME. Argon was then flushed through the tube for 3min. The tube was then sealed tightly and heated 5 with stirring at 120 degree for 24h. After cooled down, the mixture was filter through a celite earth, the filtrate was then concentrated and the residue was purified by column chromatography eluting with 1:4, Ethyl Acetate: Petroleum ether. Light yellow needle crystals were obtained (0.
6 5g, 1.65mmol, yield 41.2%). Confirmed by ESI-MS and NMR. ESI-MS: m/z 396.2 (M+H+). 1H NMR (CDCl3): d 8.809 (d, J=6.0, 2H), 8.335 (d, J=5.0, 211), 8.090 (d, J=7.5, 2H), 10 7.750 (m, 6H), 6.543 (t, J=4.5, 1H), 3.868 (t, J=5.0, 4H), 3.404 (t, J=5.0, 4H). Scheme 2 4yridin-4-vl)pyrdazin-3(2H)-one To a 200 ml, three-necked, round-bottomed flask equipped with a magnetic stir bar, 150 ml pressure-equalizing addition funnel, reflux condenser and a glass stopper, was added 21 g (135 15 mole) of 4 -bromopyridine and 70 of anhydrous THF. The system was oven-dried and flushed with argon before use. 135 ml (135 mmole) of THF solution of phenylmagnesiumn bromide (1M) was placed in the pressure-equalizing addition funnel. Then, the grignard solution was added dropwise over a period of 10 minutes. After the addition, the reaction was stirred for 15 minutes for completion. The solution of Grignard reagent was then obtained. A solution of 4 10 pyridylmagnesium bromide obtained above was added to a hot suspension of 6 phenylpyridazinone compound 7.8g (45 mole) in dry toluene (50 ml). The mixture was refluxed for 8h, left overnight at ambient temperature, then decomposed with a saturated solution of ammonium chloride. The organic layer was separated, and the aqueous layer was extracted with 100ml of ethyl acetate. The solvent was removed and the residue was crystallized !5 from ethanol. The crystals were collected by filtering and dried over a medium fruit sintered glass funnel in vacuo to give 5.6 g of white crystals. Yield was 50%, confirmed by ESI-MS. ESI-MS: m/z 252.1 (M+H+). 6 -phenyl-4( vridin-4-y)yridazin-3(2H) -one 4.4 g (17.5 mmole) of 6 -pyridazinone obtained above was placed in a 50 ml single-necked 0 round bottom flask followed by 4.7 g (35 mmole) of anhydrous copper (II) chloride and then 20 ml of acetonitrile to give a brown yellow suspension. A reflux condenser was connected to the flask and a dry tube filled with CaCI2 was fitted to the top of the condenser. The reaction mixture was heated to reflux in an oil bath (110 C) for 3 h. The color of the reaction suspension changed to dark yellow once the reflux started. After the completion of the reaction 84 (monitored by HPLC), the flask was removed from the oil bath and cooled to ambient temperature. The mixture was poured on to 200 g of crushed ice and stirred vigorously for 10 minutes to give a gray precipitate and blue liquid. The precipitate was then collected by filtering (pH of the filtrate was 1.5-2.0), and washed with 50 ml of a 1N HCl solution to rid the 5 solid of any remaining copper byproducts. This is followed by washing with 100 ml of Milli-Q water to get rid of the acid in the solid, and is monitored by checking the pH value of the filtrate. The solid was washed until the filtrate shows a pH of 7, after approximately 5 washes. The solid was dried over a medium frit sintered glass funnel in vacuo to give 3.9 g of a blue gray solid. Yield was 90%, confirmed by ESI-MS. ESI-MS: m/z 250.1 (M+H+). 10 3 -chloro- 6 -henyl-4-(yridin-4-yl)pyridazine 2.0 g (8 mmole) of 6 -phenylpyridazinone obtained above and 10 ml (54 mmole) of phosphorus oxychloride (reagent grade, Aldrich) were placed in a 50 ml single-necked round bottom flask. The flask was connected with a reflux condenser and a dry tube filled with CaCl2 was fitted to the top of the condenser. (HCl gas is formed in the reaction so a basic solution such as NaOH 15 may be needed to absorb HCl in a large-scale synthesis). The reaction mixture was stirred in an oil bath (90 C) for 2 h, then cooled to ambient temperature and poured onto crushed ice. (phosphorus oxychloride can be decomposed by water to give HCl and H3PO4). The mixture was then stirred vigorously for 10 minutes to give a white suspension. The suspension was neutralized with a 2N NaOH solution until the pH of the suspension was pH = 7. The 20 precipitate was filtered, washed three times with 100 ml of water and dried over a medium frit sintered glass funnel in vacuo to provide 1.8 g of a light pink powder. Yield was 85%, confirmed by ESI-MS. ESI-MS: m/z 268.4 (M+H+). 2vii (MW-1-6-189WH) 1.1 g ( 4 .0mmole) of 3 -chloropyridazine obtained above was placed in a 30 ml pressure vessel 25 followed by addition of 2.6 g (16.Ommole) of 1-(2-pyrimidyl) piperazine and then 15 ml of 1 BuOH (reagent grade). The vessel was sealed tightly and placed into an oil bath and stirred at 130"C (temperature of oil bath) for 3 days. The reaction mixture was then cooled to ambient temperature and transferred to a single-necked flask for evaporation under reduced pressure. Removal of solvent gave rise to a brown-red residue that was treated with 30 ml of water to give 30 a brown suspension. The solid was collected by filtering and washed with 50 mL of water three times and dried over a filter funnel in vacuo to provide 0.96 g of light yellow solid. Yield was 90%, confirmed by ESI-MS and NMR. ESI-MS: m/z 396.2 (M+H+). 1H NMR (CDCl3): d 8.809 (d, 1=6.0, 2H), 8.335 (d, J=5.0, 2H), 8.090 (d, J=7.5, 211), 7.750 (m, 6H), 6.543 (t, J=4.5, 1H), 3.868 (t, J=5.0, 411), 3.404 (t, J=5.0,44H).
85 Example 14. Preparation of 4
,
6 -diphenvl-3-(4henvlpiperazin-1-VI)yridazine (MW1-7 029WH). A synthetic reaction scheme for the preparation of 4 ,6-diphenyl-3-(4-phenylpiperazin-1 5 yl)pyridazine (MWO1-7-029WH) is depicted in Figure 20, and synthesis was carried out as described herein. The compound was prepared from 3-chloro-4,6-diphenylpyridazine (100mg, 0.37mmol) in the same manner as described for MWO1-7-057WH, yielding white solid (123mg, 0.31mmol, 83.1%). ESI-MS: m/z 393.2 (M+H+). NMR (CDCl3): d 8.107 (d, J=8.0, 2H), 7.736 (d, J=7.5, 2H), 7.651 (s, 111), 7.500 (m, 5H), 7.290 (t, J=8.5, J=6.5, 311), 6.958 (d, J=7.5, 211), 10 6.899 (t, J=7.0, 11), 3.487 (s, 4H), 3.214 (s, 4H). Example 15. Preparation of 4,6-dipheny-3-(4-methylpiperazin-1-yllpyridazine (MWO1-7 027B-WH). A synthetic reaction scheme for the preparation of 4
,
6 -diphenyl-3-(4-methylpiperazin-1 15 yl)pyridazine (MW01-7-027B-WH) is depicted in Figure 21, and synthesis was carried out as described herein. The compound was prepared from 3 -chloro-4,6-diphenylpyridazine (100mg, 0.37mmol) in the same manner as described for MWO1-7-057WH, yielding white solid (1 19mg, 0.35mmol, 94.5%). ESI-MS: m/z 331.1 (M+H+). NMR (CDC13): d 8.089 (d, J=7.5, 2H), 7.643 (d, J=7.5, 211), 7.611 (s, 111), 7.510 (m, 6H), 3.365 (s, 311), 2.472 (s, 4H), 2.337 (s, 411). 20 Example 16. Preparation of 4
,
6 -dinhenl-3-(4-cvclohexvlpiperazin-1-vl)Dyridazine (MWO1-3-065SRM) A synthetic reaction scheme for the preparation of 4
,
6 -diphenyl-3-(4-cyclohexylpiperazin-1 yl)pyridazine (MWO1-3-065SRM) is depicted in Figure 22, and synthesis was carried out as 25 described herein. The compound was prepared from 3 -chloro-4,6-diphenylpyridazine
(
3 0 0mg,1.1mmol) in the same manner as described for MWO1-7-057WH, yielding white solid (350mg, 0.87mmol, 87%). ESI-MS: m/z 399.2 (M+H+). 1H NMNR (CDCI3): d 8.09 (d, J=7.5, 2H), 7.68 (d, J=7.5, 2H), 7.59 (s, 1H), 7.56-7.42 (m, 6H), 3.39 (s, 41), 2.62 (s, 411), 2.273(s, 1H), 2.01-1.78 (m, 4H), 1.63 (d, J=12.5, 111), 1.33-1.08 (m, 5H). 30 Example 17. Preparation of 4,6-diphenvi-3-4-isopropyloinerazin-1-vl~byridazine (MW01 3-066SRM). A synthetic reaction scheme for the preparation of 4
,
6 -diphenyl-3-(4-isopropylpiperazin-1 yl)pyridazine (MWO1-3-066SRM) is depicted in Figure 23, and synthesis was carried out as 86 described herein. The compound was prepared from 3 -chloro-4,6-diphenylpyridazine (300mg, 1.1mmol) in the same manner as described for MW01-7-057WH, yielding white solid (290mg, 0.8lmmol, 72%). m/z 359.2 (M+H+). 1H NMR (CDCI3): d 8.09 (d, J= 7.5, 2H), 7.69 (d, J=7.5, 2H), 7.61 (s, 111), 7.54-7.46 (m, 6H), 3.40 (s, 4H), 2 .72(m, 1H), 2.59 (s, 4H), 1.10 (d, J=6, 611). 5 Example 18. Preparation of 4
,
6 -dinhenvl-3-piyerazinvlpvridazine (MWO1-7-133WH) A synthetic reaction scheme for the preparation of 4
,
6 -diphenyl-3-piperazinylpyridazine (MWO1-7-133WH) is depicted in Figure 24, and synthesis was carried out as described herein. The compound was prepared from 3 -chloro-4,6-diphenylpyridazine (533mg, 20mmole) in the 10 same manner as described for MWO1-7-057WH, yielding light yellow solid (550mg, 17 .4mmole, yield 86.9%). ESI-MS: m/z 317.3 (M+H+). 1H NMR (CDCl3): d 8.086 (d, J=7.5, 2H), 7.705 (d, J=7.5, 211), 7.619 (s, 1H), 7.498 (m, 6H), 3.318 (d, J=4.0, 4H), 2.932 (d, J=4.0, 4H) 1.896 (s, 1H). 15 Example 19. Preparation of 2
-(
4
(
6 -Dhenvl-4-(piperidin-1-vl)pyridazin-3-vl)piperazin-1 Vl)pyrimidine (MW1-7-107WH) A synthetic reaction scheme for the preparation of 2
-(
4
-(
6 -phenyl-4-(piperidin-1-yl)pyridazin-3 yl)piperazin-1-yl)pyimidine (MW01-7-107WH) is depicted in Figure 25, and synthesis was carried out as described herein. The compound was prepared from MWO1-6-127WH (200mg, 20 0.57mmole) in the same manner as described for MWO1-7-057WH, yielding light yellow solid (220mg, 0.55mmole, yield 96.3%). ESI-MS: m/z 402.5 (M+H+). Example 20. Preparation of 6 -methl-v-4-phenvl-3-(4-vrimidin-2-vlpiperazin-1 Vl)pyridazine (MWO1-7-057) 25 A synthetic reaction scheme for the preparation of 6 -methyl-4-phenyl-3-(4-pyrimdin-2 ylpiperazin-1-y1)pyridazine (MWOl-7-057) is depicted in Figure 26, and synthesis was carried out as described herein. A mixture of 3 -chloro- 6 -methyl-4-phenylpyridazine (100mg, 0.5mmol), 1-( 2 -pyrimidyl)piperazine (400mg, 2.Ommol) in 3ml of 1-BuOH was heated with stirring at 130 C for 7days. The solvent was removed by evaporation in vacuo, the residue was 30 treated with water to give a suspension. The solid was then filtered off, washed with water, then 1:3, Ethyl Acetate: Petroleum ether, dried over filter funnel in vacuo to give light yellow solid (68mg, 0.20mmol, yield 41.7%). Purity >95%; ESI-MS: m/z 333.1 (M+H+). 1H NMR (CDCl3): d 8.310 (d, J=5.0, 2H), 7.678 (d, J=7.5, 211), 7.476 (m, 3H), 7.119 (s, 11), 6.509 (t, J=4.5, 1H), 3.785 (t, J=4.5, J=5.0, 4H), 3.277 (t, 1=4.5, J=5.0, 4H), 2.669 (s, 311).
87 Example 21. Preparation of 2-(4-(5-phenvl-6-vrdin-4-vl)pyridazin-3-vl)piperazin-1 v)pyrimidine (MWO1-2-163MAS) A synthetic reaction scheme for the preparation of 2
-(
4
-(
5 -phenyl-6-(pyridin-4-yl)pyridazin-3 5 yl)piperazin-1-yl)pyrirmidine (MWO1-2-163MAS) is depicted in Figure 27, and synthesis was carried out as described herein. 1, 2 -dihvdro-4-phenvlpyridazine-3,6-dione (MWn1-2-077A-MAS) 4.0 g (23 mole) of 3 -phenylfuran-2,5-dione was added to a 100 ml single-necked round bottom flask followed by 2.9g (27.6 mmole) of hydrazine monohydrate and then 20 ml of 10 reagent grade ethanol (95%). The flask was fitted with a reflux condenser and the reaction mixture was heated to reflux in an oil bath at 110 degree (temperature of oil bath) and stirred for 2 h. The flask was then removed from the oil bath and the reaction mixture cooled to ambient temperature. The stir bar was removed and the solvent was evaporated in vacuo in a water bath at 45 degree. The residue was then treated with 50 ml of Milli-Q water and stirred for 10 15 minutes to give a suspension. The precipitate was collected by filtering, washed with 100 ml of Milli-Q water, and dried over a medium frit sintered glass funnel in vacuo to give 3.9g of white solid. Yield, 91%, confirmed by ESI-MS. ESI-MS: m/z 189.2 (M+H*). 3
.
6 -dichloro-4-phenvlpyridazine (MW01-2-082A-MAS) 1.5 g (8 mmole) of 6 -phenylpyridazinone obtained above and 10 ml (54 mmole) of phosphorus 20 oxychloride (reagent grade, Aldrich) were placed in a 50 ml single-necked round bottom flask. The flask was connected with a reflux condenser and a dry tube filled with CaCl2 was fitted to the top of the condenser. (HCl gas is formed in the reaction so a basic solution such as NaOH may be needed to absorb HCI in a large-scale synthesis). The reaction mixture was stirred in an oil bath (90'C) for 2 h, then cooled to ambient temperature and poured onto crushed ice. 25 (phosphorus oxychloride can be decomposed by water to give HCl and H3PO4). The mixture was then stirred vigorously for 10 minutes to give a white suspension. The suspension was neutralized with a 2N NaOH solution until the pH of the suspension was pH = 7. The precipitate was filtered, washed three times with 100 ml of water and dried over a medium frit sintered glass funnel in vacuo to provide 1.5 g of a white solid. Yield was 85%, confirmed by W0 ESI-MS. ESI-MS: m/z 226.1 (M+H). 2-(4-(6-chloro-5-phenylpyridazin-3-vlp iperazin-1-vpvimidine (MW01-2-114B-MAS) A mixture of 3
,
6 -dichloro-4-phenylpyridazine (1.35g, 6mmol), 1-( 2 -pyrimidyl)piperazine (1.2g, 6.Ommol) in 10ml of 1-BuOH was heated with stirring at 80 0 C for 12h. The solvent was removed by evaporation in vacuo, the residue was treated with water to give a suspension. The 88 solid was then filtered off, washed with water, dried over filter funnel in vacuo to givewhite solid (1.8g, 5.2mmol, yield 86.0%). ESI-MS: m/z 353.9. 2Yl)piperazin-1-yl)pyrimidine (MW01-2-163MAS) Into a reaction tube were added MWO1-2-114B-MAS (1.4g, 4.0mmol), K 2 C0 3 powder (1.7g, 5 12.4mmol), Pd(PPh3)4 (240mg, 0.2mmol), 4 -pyridineboronic acid (664mg, 5.4mmol) and 20ml of DME. Argon was then flushed through the tube for 3min. The tube was then sealed tightly and heated with stirring at 120*C for 24h. After cooled down, the mixture was filter through a celite earth, the filtrate was then concentrated and the residue was purified by column chromatography eluting with 1:4, Ethyl Acetate: Petroleum ether. Light yellow needle crystals 10 were obtained (0.69g, 1.74mmol, yield 43.5%). Confirmed by ESI-MS and NMR. ESI-MS: m/z 396.2 (M+H*). Examle 22. Preparation of N.(ccloropylmethyl)-6-phenyl-4-(ridin-4-vI)Dridazin-3 amine (MWO1-7-084WH). 15 A synthetic reaction scheme for the preparation of N-(cyclopropylmethyl)-6-phenyl-4-(pyridin 4-yl)pyridazin-3-aine (MWO1-7-084WH) is depicted in Figure 28, and synthesis was carried out as described herein. 4 -chloro- 6 -phenylpvridazin-3(2 H-one (MW1-6-093WH) was synthesized according to the procedure described by Coudert, P., et al. [18]. 20 4 -chl ro- 2 -(methoxvm-thvl)6phenylpridazin-3(2H)-one (MW1-7-053WH) A mixture of chloropyridazinone 1 (25.5 g, 0.12mol), 4 -N,N-dimethylaminopyridine (0.20 g) and i-Pr2NEt (26.7g, 0.21mol) in anhydrous CH2Cl2 (300mL) was stirred at 0 j5C (ice bath) for 30 min. Methoxymethyl chloride (25g, 0.31mol) was added and the mixture was stirred at 0"C for lh and then allowed to warm to r. t. The reaction was stirred at r.t. till it complete. The 25 solvent was then removed in vacuo, the residue was treated with water, washed with dilute Na 2
CO
3 solution and extracted with EtOAc. The organic layer was dried over anhydrous Na 2
SO
4 , filtered and evaporated. The residue was then purified by recrystallization from 95% ethanol to give 20.1 light yellow solid. Yield 66.9%. 6 -phenyl-(pyidin4.vl)pyridazin-3(2H)-one (MW1-7-069WH) 30 The protected pyridazinone MWO1-7-053WH (1.Oequiv.) was mixed with arylboronic cid (1.37equiv.), Pd(PPh3)4 (0.05 equiv.) and K2C03 (3.1equiv) and 200 mL of DME in a 350mI of pressure vessel, flushed with argon for 3 min, and the mixture was then stirred and refluxed (oil bath, 120*C) until the starting material had disappeared. After cooling, the solution was concentrated to dryness under reduced pressure, the residue was treated with water and filtered 89 off. The filter cake was washed with water over filter f.unnel and then used for next step directly. The residue obtained above was dissolved in 200ml of EtOH, 6 N HC1 (200 mL) was added and the reaction mixture was refluxed (oil bath, 120*C) for 6 h, then it was allowed to cool to room temperature, and concentrated to dryness under reduced pressure. The residue was 5 neutralized with dilute NaOH solution. The suspension was then filter off, washed with water and dried over filter funnel. Recrystallization from 90% ethanol provided brown yellow solid. Yield 80.4%. ESI-MS: m/z 294.3 (M+H*) 3 -chloro 6 hen14 4 pyridinee (MW1-7-076W ) This compound was prepared from MW01-7-069WH in the same manner as described for 10 MW01-6-127WH, yielding light yellow solid. ESI-MS: m/z 268.4 (M+H+). N 4-(pyridin-4-vl~prdazin-3-amine (MW1-7-084WH) This compound was prepared from MWO1-7-076WH in the same manner as described for MW01-7-057WH, yielding gray solid. ESI-MS: m/z 330.4 (M+H*). 15 The present invention is not to be limited in scope by the specific embodiments described herein, since such embodiments are intended as but single illustrations of one aspect of the invention and any functionally equivalent embodiments are within the scope of this invention. Indeed, various modifications of the invention in addition to those shown and 20 described herein will become apparent to those skilled in the art from the foregoing description and accompanying drawings. Such modifications are intended to fall within the scope of the appended claims. All publications, patents and patent applications referred to herein are incorporated by reference in their entirety to the same extent as if each individual publication, patent or patent 25 application was specifically and individually indicated to be incorporated by reference in its entirety. All publications, patents and patent applications mentioned herein are incorporated herein by reference for the purpose of describing and disclosing the methods etc. which are reported therein which might be used in connection with the invention. Nothing herein is to be construed as an admission that the invention is not entitled to antedate such disclosure by virtue 10 of prior invention.
90 Table 1 Compound Compound Synthetic Number Structure Code 1 H MW01-ES1 N N 0 0 4 0 MW01-ES112 H' N N O 0 10 0 MWO1-ES159 N IN O 11 0 MW1-ES21 H'N O - 0 N 12 OMWO1-ES3 1 13 H, MWO1-ES60 N N 0 O0 91 14 0 )MWOI-ES61 H, N 00 N 0" - 0 16 H,0 AM01-ES75 H.N 17
H,
0 0MWO1-ES8 1 I i J 0 18 0MW-E1 H, N ci wlE9 N-O 20 C H 3 MW01-1-04-L-D04 N N CI 23 r \N MWO1-1-15-L-H07 24 '. MWO1-1-16--L-FO5 N 0 .. H 3 25 ' MWO1-1-18-L-B09 NN N--
N)
31 92 MWO1-1-O35LIXL Br 40 0-CH 3 MW110--O \/ \
NH
2 N-N HO -~0 41
H
3 0 MWO1-2-03-L-C02 0)> 0 \/NH N-\ "'/ N-N 43 MWO1-1-15-L-EO9 - N-N
NH
2 44
H
3 C MWO1-1-16-L-B1 1 \ / \ /NH N-N NH 2 _________ H4N7 MWO1-4-198B-Z 48 MWO1-5-144A-Z 0
H
2 N \/ 0 49 N MWO1-4-198C-Z
N-N
50 cI 9 AMO-5-144C-Z
H
2 N\/ 51 -o N MWOl-5-144D-Z
H
2 N / /\ N-N 52 MWOI-5-145A-Z
H
2 N / /\ N-N 54 NM1-5-189Z
H
2 N / /\ N-N 55N MW1-5-202B-Z 61 OH MW01-1.-01-L-D06 CNH "N N 65 H2N, NH IvWO1-1-O 1-L-E1O
KN
66 H3C. s 94MWO1-1-02-L-E08 N 71 SH NMO1-03-L-F03 N 73 OH 3 MWO1-1-03-L-G1O
K
0 N N 75 MWO1- 1-04-L-C03 N
N
95 76
CH
3 MWO1- 1-07-L-H04 lN N 88 '-.MWO1-1-100-L-A04 N N 89 MWO1-1-100-L-A05 HN N' CIH N. 90 HNfMWO1-1-100-L-AO8 HN I
CIH
96 ___________ 91 fNCXH3 MWO1-1-100-L-A09 HN CIH N'CIH Nz 92 MWO1-1- 11-L-E08 HN 0 N 94 N MWO1-1-15-L-G09 974W - 1--0 N HN 0 106 - ~ -MWOI-9-039Z N-N 107 (-N\ / N MWOI-9-040Z &N
N-N
97 108 -- N N MWO1-9-041Z 109N 109 H 2 N MWO1-9-104A-Z N-N 110 C MWO1-9-105A-Z N-N 111 OH MW01-9-11OA-Z CI \ /
N-N
112
NH
2 MWO1-9-133A-Z
H
2 N / N-N 113 MWO1-9-149A-Z Cl N-N 114 C MW01-9-159A-Z CI \ -115 rqNMWO1-9-171Z N-N 116 MW1-9-172Z N-N 118 H MW1-9-204Z N Cl -N 120 MWO1-1-16-L-G08 HN
N
98 122 AMO-1-17-L-G05 N HN "N NH 125 MW1-1-17-L-H03 NN HN N
HN
99 _ _ _ _ _ _ _ _ _ _ _ 127 MWO1l-1-17-L.H1 1 N' N HN "N -N
I
130 MWO 1-1-1 8-L-A08 N) HN N -N N'
N
13749 ~' MWOI1--2OSR N ND' NN,) H3C-H _ _ _ _ _ ~100 _ _ _ _ _ 150 MWO1-1-18-L-B03 NH 151
-
/ MvW01-1-18-L-B09 158 H 2 MWO1-3-033WH _________ N-N 159 Br MWOl-3-009VVH N 17 / /N MWO1-2-03-L-D02 NN 175 MWO1-2-06-L-F04 175A H??9 N N N 101 179 H3C, s MWO1-2--33-L-B02 180 MWO1-3-O1-L-G07 OH N 184N MWOl-5--161WH N N 189 MWOI1-6-041WH HN N -N 19 0 H 2 N MWOI-6-044WI <N 192 H 2 N~jN MWOI1-6-050WI-H ~N
~N
102 197 OH MWO1-1-01-L-A1O ~ NH NN -N 198
OH
3 )W1-1-01-L-B03 0 NH N 199 OH NffOl-1-01-L-B09 (NH CNH
H
3 0 AN N 201
OH
3 Cl IMO1-1-01-E3 'N N 202 0
.X.H
3 MWO1-1-01-L-E04
H-
3 0
AN
103 205 MfWO1-1-18-L-B07 N
H
3 C OH 3 H N5 N N 208 NMO-1-03-L-G03 cl Nr2 I N-~ /' 0 2100 MW01-1-04-L-C03 o,' 217
OH
3 MWO1-1 -02-L-E03 0 H)
H
3 C N HN HN
H
3 0 " --- --- --- ___________ ~~104 _ _ _ _ _ _ _ _ _ _ 221 MfWO1-1-02-L-F02 N N 223 OH MWO1-1-02-L-F08
H
3 C "N 225
YH
3 AM1-1-02-L-G05
H
3 N N 226OH MWol-1-02-L-GO6 N ,CH3 0
H
3 C "N N 227
YH
3 MWO-1-1-03-L-A02 0 NH 'N "'N
N
229 1 7CI- 0 MWO1-1-03-L-B09
H
3 C N 230-O MO1-03-L-B 10
H
3 C N N 231 Br MWO1-1-03-L-C03
H
3 C N N 23 3- C I MW1-1-03-L-C08 I -i -N 235 H2N, NH MWO1-1-03-L-E08
H
3 C N N 23 N 2 2C MWO1-1-03-L-EO9 106 240 HO.... CH 3 AMo-1-o4-L-A06
H
3 C N -N 242
H
3 C ~ oMWO1-1-04-L-DIO N 250 ("' MWO1-1-05-L-B 11
H
3 C "N 251 0MWO1-1-05-LC02 KN N 254 0MW0l1-05-L-G1 1 KN H N
KS
107 255 S.C3MWO1-1-05-L-H05
H
3 C ~NCH3 266 MW01-1-08-L-D09 HN l
H
3 C N 2-6-8 MWO1-1-09-L-C06 CHsHN O NH 270 271 OH MWO1-1-09-L-G07 NHOH N N 0 T7- c3MW11-9K-O 108 272 OH 3
NH
2 MWO1-1O09-L-3O9
H
3 0 OH 274 ci MWO1-1-09-L-H07 'NN 275 OH MWO1-1-15-L-A04
H
3 0 N N 27 6 cI MWO1-1-15-L-B02 'NN 278 H 3 0 CH8 1 NfWO1-1-15-L-B 10 N 280 H 3 C OH MWOI1-15-L-C04 -N N 282 OH 3 CI MWO1-1-15-L-D03
H
3 CN
N
109__ _ _ _ _ _ _ _ _ _ 284 H 3 C N MO-1-15-L-GIO 292 MOI-1-17-L-A09 OON
H
3 C 293 0 NMO-1-17-L-A1l CHs3H N N'- N ) 294 CH 3 H MO-1-17-L-B02 I ,,-,OH N'- N 295 OH 3 H MWO1-1-17-L-B1O N,,, H NH3 ~- N' H 296 CH 3 H MWO1-1-17-L-E11 N N K
OH
3 297 CH 3 H MWO1-1-17-L-F03 N' N NK 298 (0 MWO1-1-17-L-HO5
OH
8 H N' 0 1 N,,) N N 110__ _ _ _ _ _ _ _ _ 299 -MWO1-1-18-L-A09 N
CH
3 H N3 N NA 308 F0 O MWO1-2-03-L-B08 F FH CN KW1 -N%), W" NN 310 0MWOI-2-03-L-C05 HN N NN 31 3 OH()MWO1-2-03-L-G07 HN N' N) 318 H0 MW01-2-101-L-H08 H N N "N 319 M0)WOI-2-1'0-L.E05 COOH N N-,
NN)
320 0 MWO1-2-10-L-E06 CI H (N 'N 32 H2 MWOI-2-20-L-B02
NH
2 '2 323
NO
2 H MWOI-2-20-L-D05 I N,_,-.. 324 N MWOI-2-20-L-E09 H S N'~ ~ 326 cI MWOI-2-25-L-.H06 -N H' N 328
C
1 H AMO-3-01-L-G03
OH
3 H C H 3 'N N' No 112 331
CH
3 MWOI-3-O1-L--G08
CH
3 0 "CH 3 "" N' 32
H
3 C~ MWOI-3-O1-L-G09
OH
3 Si ,CH 3
~
1
CH
3 335 MIWO1-3-06-L-E09
CH
3 Q ~kCH3
OH
3 0 6C 337 MWOl-1-07-L-G07 N. CN N N 339
OH
3 MWO1-1-15-L-Cl 1 I 340 HMWO1-1-15-L-E09 N , NH 2 ANH
NN
113 346
CH
3 NMO-1-17-L-FIO CHS H N CH 3 I N,~ N N 347 0MWOl-1-17-L-F1 1
OH
3 H N "N 350 MWO1-2-20-L-B 11 Hp H3CA N CN 352 0 MWO1-3-O1-L-F09 H3C 0CH N'N 39 N~ cI MWO1-1-03-L-E05 "N 360 S AMO1-1-03-L-A08
N
361 H 3 C 11 MWO1-1-03-L-H08 0 ".0 362 0 MIWO1-1-O1-L-H04 S NH IN 363 MW0T1-1-01-L-HO6 INH 366 S MWO1-1-03-L-E07
H
3 C NH 367 NO 2 MWO1- 1-05-L-E05 HN4 368 0 NMO-1-03-L-B03 HsC INH
IN-
_____ _____115 371 H 2 N MWO1-1-05-L-E07 N N0 2 372 0 MWO1-1-03-L-A03
H
3 C NH N 373 cl MWO1-1 -03-L-E03
H
3 C N N 374 OH A4~l11-L-H1O
H
3 0 Ne CH 3 ("N 375 NIMO1-1-04-L-1H08 N t N 376 CH 3 MWO1-1-O1-L-GIO 0 NN NH HO 377 Cl MWO1-1-03-L-G1 1 +0e 116 380 MWO1-1-04-L-B07 CIH N 0 381 MW01-1-04-L-C09 0 CIH N- NN N 0 Br 382 MW01-1-10-L-G05 0 N-N\ HOOCCOOH
N
117 Table 2 Compound Compound Synthetic Number Structure Code 22 MWO1-1-15-L-E08 CH3 & /N CH, 26 MW01-2-02-L-H09 \ N N4 \ OH 29 MWO1-1-030A-LKM N N N)N Y N ( N N N N N 30 Br MWO1l-1-030B-LKM ('N -N _____ C_ N 32 Br MW01-1-048AB N LKM O H 0
H
33 118 MWO-2-65LKM N ..N N ' N F 34I 34 MWO1-2-127LKM{ Nr (N) N N N- NfN H 35NMOI-2-134LKM N-rN N <"N -N 36 A MWO1-2-146LKM N ~N
N
119 37 MWO1-2-147LK:M N
H
2 N N N -N H 38
OH
3 MWO1-1-02-L-B11 \/ /N NH N-N ' 39
H
3 0-O ICH 3 NMO-1-04-L-F1O 0 ____ ____N-N 42 H 3 C OH 3 0MWO1-2-33-L-A11 X\ / - N-N 45 MWO 1-1- 17-L-E06 \/NH
N-N
120 46 MWOL-1-O45MAS N NH 56 MWOI-7-127AB-Z 00H NH N N 6 OH MWOl-7-17AB-Z NHH N N
-N
_____ ____121 62 roMWO1-1-O 1-L-D 10 HN 0 1 "N H ci N 63 (No IVWO-1-01-L-E02 H ,H HN IN c 64 IMO1-1-01-L-E08 Br ,C~H 3 -N Br N 67 -N MWO1-1-02-L-HIO N KN Br 122 69 OH MWO1-1-03-L-B08 <H N N 72 Br Br MVW1-1-03-L-G09 KN N 93 HO MWO1-1-13-L-E06 S,0 N NN NH HO' NN
N
123 95 NH 2 0 MWfO1-1-16-L-D09 96 MWO1-1-16-L-E02 CIH HN CIHj N N / -N k- -N 121 roMWO1-1 -17-L-G04 N HN N CIH -N
CIH
__________ ~~124____ ______ 124 1Mwofl-1-17-L-H02 %NO HNX N
H
3 C.-- N H) 128 MWOL-1-18-L-A02 QN _ _ _ _ N 125 129 AM01-1-18-L-A03
H
3C -N N N N 136 (0 MWOI-2-018SRM N ro N NV N 1N N), N,,) N 147 MWOI-2-177A-WH N)N N N N ON
N
126 148 N MWO1-2-177B-WH N N- CI N N N N 153 O H C MW01-2-184WH ON N N H II -N 155 MWO1-2-191A-WH N -N N O N U - , H O N _N N H 156 MWO1-2-193B-WH 00 HN NH N 157 HO MWO1-3-003WH \/N N-{,) N-NN N 160 -N MW1-3-019A-WH -N N 127__ _ _ _ _ _ _ _ _ 161 CI MWOI-3-060A-W*H -NN 162
NH
2 MWOI-3-072WH NN N~ NN 163 CI MWO1-3-1 17WH N 164 ci MWOI-3-1 18VH N H 166 CN ~ WOI-3-183WH N N '- N ~ N-N 171 MWI20--O \ / & NMW-20L03 N-N OH 172 0 MWOI-2-03-L-C04 & NH N ___N-N 0 174 Q -- MWOI-2-03-L-G03 N-N 0 - \_ \176/N r-H MWO1-2-102-L-C11
N-N
128 177 NH 2 MWO1-2-21-L-F04 N 178 OH MWOI-2-24-L-G09 N 0 N 181Al N 186 / MWO1-5-188W11 ND - N-N N\-' N 18 8 -OH MWO1-6-OO3W~H ~N ~N __ __~ N0 191 H 2 NIN12 MWO1-6-046WH 'N N N "NN
-
i N 'NN 203 MWOI-2-03-L-D09 OH H N NN KNH 204 'NMWO1-1-01-L-B02 N N NH OH N C0) 206 OH Kro MWOI-2-03-L-D09 NZ N
%HN-
130 207 MWO1-2-03-L-G04 NH
H
3 C 'N N)N H 'N ',N N 211 MWO1-1-04-L-C03 NI-I 'NH 212N MWO1-1-01-L-Ell HN, NH
H
8 C N
AN
131 213
CH
3 O MWO1-1-01-L-F02 KO HN N
H
3 C~'0 "N N 214 MWO1-1-01-L-F03 NNa
H
3 C N 215 L MW01-1-01-L-G08
H
3 C N N 216 OH MWO1-1-02-L-D11 N "rOH 0 HN
H
3 C N N 219 rCH3 MW01-1-02-L-E04 NHO Of
H
3 C N
N
_____ ____132 220 N MWO1-1-02-L-E11 HNN
H
3 C -24 H'N2MWO1-1-02-L-F09 228 MW01-1-03-L-AO4
H
3 C N -N 23 2
H
3 C_O 0 MWO1-1-03-L-CO4
NH
2
H
3 0
-N
234 13MWO1-1-03-L-E04
H
3 0 cI "N 237
OH
3 MWiT1-1-03-L-E1O 0 N
H
3 C N "N 'N 239 H C3a MO1- 1-03-L-G04
H
3 C"N
NZ
134__ _ _ _ _ _ _ _ _ 241 OH MWO1-1-04-L-D08 (N) N H3C N NN 2441 MWOl-1-04-L-E03 N
H
3 0 N 245
N~
5 ~ MWO1-1-04-L-E09
H
3 C
N
135 246
CH
3 MWOl-1-04-L-E06
K
0 HN
H
3 C N 247 Br Cl MWO1-1-04-L-G06 BrN 248
CH
3 MWO1-1-04-L-HO6 (I N 249CH MOI--0-LH- 136__ _ _ _ _ _ _ _ _ 252 MWO1-1-05-L-F05
H
3 N HN H)
H
3 C N 253 COOH MWO1-1-05-L.GIO HOOC KN 256 OHF MWO1-1-05-L-H07 O=6=0 NH
H
3 C 257 N HN'NH2 Mwol-1-05-L-H09 -N 16K _____ ____137 259 07 MWO1l-1-07-L-E07
H
3 C " N N 261 -I --- lWO1-1-07-L-H03
H
3 0 NN 262 ~ ~ c ~ 7 ~OHWO1-1-07-L-HO3 H3C N" N
A-
138 263 NMO -1-07-L-H06 "N HON HN
H
3 C N HN 264 HC MW'1'NL-O 'N -N 265 -MWO1-1-08-L-C09 0 N N 267 H 3 CN 0 roMWO1-1-08-L-E04 HN -N
%HNN
139 269 MWO1- 1-09-L-GT04 N 0,OCH 3 0 273 IINH MWO1-1-09-L-G1 1 '0 N) N 279 CHS MWOl-1-15-L-B 11
H
3 C OH Nr~ N 281 CI AM01-1-15-L-D02 NN H3% N 282 OH 3 CI MWO1-1-15-L-D03
H
3 0 140__ _ _ _ _ _ _ _ _ 283
H
3 C,. N CH 3 N'WO1-.1-15-L-EIO N
H
3 C N 285 0 C MWO1-1-15-L-H09 HC 286 MWfO1-1-16-L-EO5 HC N - HN 287 HMW -- O-F1 0 NN Clll-1L CI 28 8 Nl MWO1-1-17-L-B05 H N 290
CH
3 CH 3 MWO1-1-16-L-E08 ~.
NH
2 0 N' 0
N'
141 291 H 3 0 OH 3 AM1-1-16-L-G07
H
3 C CH. Os..H N N 297
OH
3 H MWO1-1-17-L-F03 300 CH3H9 MWO1-1-18-L-B04 CH3 N, , N' 301 OH 3 MWO1-1-18-L-BlO H N' N H30) 30-2 MWO1-1-18-L-Bll
H
3 O OH 3 9T HN N N 303 01 MWO I-1-18-L-C05 0N
WHO
142 304
CH
3 MWO1-1-18-L-C06 H N N N' N 3C) 305
CH
3 r MW1-1-18-L-C08 N NN 306 0 MWO1-1-18-L-C1O
OH
3 N "OH N N 307
H
3 C NMO-1--18-L-D04 H",N N N N' N 3 ) 309 1MRWO1-2 -03-L- 03 CH N 311 NH2O(0 MWO1-2-03-L-DO7 H N
'N
143 312 0 NMO-2-03-L-D08
CH
3 0 IN< N'i NN 0 314
CH
3 MWO1-2-03-L-G1O , NH 2 00 N+ 0 315
OH
3 H MWOI-2-06-L-F06 316 O ~N'No MWO1-2-09-L-BO8 - cl 317
OH
3 _NMWO1-2-09-L-E1O N OH 322 N MW 12- 0BI H N N 325
OH
3 MWO 1-2-24-L-A05 .0--CH 3 327 CH MO1-3-01-L-G02 330 CH 14 MWO1-3-01-L-G05
OH
3 Si,-H <- H 3 333 CH 3 MWO1-3-06-L-B07
~-NH
2 -~ OH 334 OH 3 MW~01-3-06-L-B08 y. NH 2 HO a '-'N'N+OH 336 MWol-1-07-L-G07
H
3 C -~ Nr ' N 0N 338 H NMOT-1-08-L-D03
H
3 C ~. N OH 3 N'N 342 MWO1-1-16-L-E09 N H NY N N I-I
N"'N
__________ 145 M 017L 0
-------
H
3 C N" W I11--O H N 345 ~ " Iw y-7L-o N'N
OH
3 N H NY N N 348 y MO-1-1 8-L-A04 H
H
3 C ~.N N 349 MWO1-1-18-L-B05 (N
H
3 C N N 3511 MWOI-2-33-L-A1O HO NO,
H
3 N
N
146 356
C
1 H AM01-1-01-L-E06 O , H 3 HN 0 NH 0 357 CI 0 MWO1-1-01-L-H109 N NH cl N 358 CI MWO1-1-O5-L-D07 N N N 0 365
OH
3 MWO1-1-03-L-D04 OH IBr N : N 369 S MWOI-1-04-L-G02
'H
3 0 NH
K"
379I 147Mw O -2-24-L-EO7 H N NN 0 N
H
3 C% MWO1-O1-O1-L-B07 rNH2 NH N N N N NN
IN
148 0 MWOl-7-09 1WH H) NN N N
CH
3 MfWO1-10-12-L-G05
H
3 gC KN Nj~~ MWOI-7-057WH
N
149 Table 3 Compounds of the Formula II 2
-(
4
-(
6 -phenylpyridazin-3-yl)piperazine- 1-yl) pyrimidine and Derivatives Compound Compound Synthetic Number Structure Code MWo1-2-5 069A-SRM N N N N) 10 MWO1-6 0 N127WH N N CE MWo1-6 189WH N -N N N~ N' N N MW01-7 107WH NN ONN
-N
150 NYN DMWO1-2 151SRM N
N)
N C N N NMO-2 N~r N069A-SRM N ~ N
N
151MW1 [IN 030A-LKM (N) N N' 33MW-2 N N 065LKM (N N N F 34 MWOI -2 N~f N127LKM (N) N 35 NMO-2 NAfN 134LKM N ('N
N
362 MWO1-2 Ny N 146LKM N 37 MWOI-2 N YN 147LKM YN N N)
H
2 N tN ~~N' N H MWO 1-i 045MAS (N) ยง/)- N \/ / \MWOI-9-038Z N N/ N 153 138 - MWOI-2 N N 023SRM (N) NN I I 147MW0T1-2 177A-WH N NN -I 155 MWO1 N N 2-191A 0 N H (Z O NN N \ N N
OW
/ 154 1 -N /MWO1 - N= 3-019A \/ \/N N( WI' 5 1 88WH 25 N-N ~-N K-ol c. F05 N
H
3 C HN
H
3 C "N -N 263 CMWO1 1-07-L H106 KN N HO N HN
H
30 :' N
N
155 Table 4 Compound Final Code Concentration Dependent Activity in Cell Culture Assays
CH
3 MWOI-01-02-L-G05 N N
H
3 C N MWOl-'i-03-L-EI N N N
N
156 OH MWOI-Oi-04-L-DO8 N
H
3 C MWOl-01-18-L-AD2 N MWOI-01-18-L-002 jN
NH
157 MWOI-02-03-L-GO4 N' N Nk N 0 N
N&
158 '~I MWOI-2-141SRM N) N '~ 'iN N k MWOI -2-163MAS N .N N N MWOl-2-177A-WII N N y 0 159 K Y NMWOI-2-191A-WH N H o N~~N 'N MWOI-3-024SRM Ny tN) N KN N MWOI-3-027SRM Nir N
N
I MWOI-3-057SRM
N
160 MWOI-3-065SRM )-N JN N-N MWOI-3-066SRM MWOI-3-183WH~ I ~MWCI-4-179LKM N N' N4 MWO1-7-4J27B-WH ~ C) 161 MWO-7-029WH N N IN MWO1-7-031WH HN N MW1-7-IIWH N N H3CO N N N MWOI-7-102WH NyN N HaCO N N 162 H MWOI-7-133W1-( N N MWOI-9-039MZ MWOI-9-040MZ NNN MWOI-2102LPI (N) NN
N
163 MWOI-21M3LPI N -N N) NH
N
164 Table 5
N~A
-
N- N N~ N 1 N 7- - N N N N N IN - - HN NN .Structure14 Istructue15 Joe, N N N NX N IShrucurei19 Structure2l 165 N Structure Stulre8 N Sir 2 Structure13 N N Structure17 Structure18 N N IStructure22 Structure3 N N p N N (N1(' NN Shxwctre212 Structure23 166 N LStructure24 Stnrcure25 N N , N): N stuctureSo -Structu 2 N N Structre~J6Structure38 StructUre~l'tutr4 167 NN N-" N Structre26 Strucke27 N -N y TN NN Z N Steue9 Structure0) NN N nd Structure4 re44 168 N )N N c N N NN N -N NN Structue63 Structure3 Istructure6oStuuel 169 N N N IN N )~ I~~ ~ ~ Structure48 tu gd, N2 Har fStrucure7e Structure59 IN~ Structure 'Sructures3 170 ONV HN H HN' HNH N K~ I IStrcture84 iStvucture65 N -N N y N) N N (N N N NN N- N T' I(N NNN Structucel9 > lstrucwrSttuctureO 171
N
NN Ql N Structure Structureff N N** NN NN GOutw7 Struclure7l O.N YN N y (NN IsN N 8 JSncre7i ISbucure72 172 YI ~N NN N NN J N NIN lStructure87 -futue4 N N a% N I Structures lStructure143 173 FNN N ac g N N .St'cireeO Structurregg HN f NH Nl N Sutureg s"rCturew~
N
174 ,N HN HNN (N N Structure101 iStruure102 N NH N N Structure1r5 Strutre106 N Structurelog Structurell S NH .-. NN IStructure113 Srcuel1 175 NN IstructtjrelO3f StructurelO8 H) IStructurell11 IStrcturell2 HN NH N N [S~curelS Structurel 16 176 NQ~ (4 N7 I N HN N ISrcure 121 Structurel218 HNN N- N Stmture1 C lStructrel22 N N -N
-
N-N N Structure 12
N
177 HN N HN1 NQ_ Structure 119 StructureI2O NII N IS~ftrI23Strctrel24 N N NN NN Structure127 >JI StructurelI28 H- NO N NH N. .~ Strucburel3'1 Structur9132 178 I~I N - - -N Strutrel133N lStlucturel 34 HNNO N N
N
Stiucturel38 NHN Strucurel4l iStructurel44 179 NN N N N N N Strucure136 lsuctwe135 N N IStructurel39 e14 N ,Structurel45 180 References [1] Akiyama H, Barger S, Bamum S, Bradt B, Bauer J, Cole GM, et al. Inflammation and Alzheimer's disease. Neurobiol Aging 21: 383-421 (2000). 5 [2] Mirzoeva S, Sawkar A, Zasadzki M, Quo L, Velentza AV, Dunlap V, et al. discovery of a 3 -amino-6-phenyl-pyridazine derivative as a new synthetic anti-neuroinflammatory compound. J MedChem45: 563-566(2002). [3] Watterson DM, Haiech J and Van Eldik U. Discovery of new chemical classes of synthetic ligands that suppress neuroinflammatory responses. J Mol Neurosci 19: 89-94 (2002). 10 [4] Watterson DM, Velentza AV, Zasadzki M, Craft JM, Haiech J and Van Eldik U. Discovery of a new class of synthetic protein kinase inhibitors that, suppress selective aspects of glial activation and protect against [J-amyloid induced injury. A foundation for future medicinal chemistry efforts focused on targeting Alzheimer's disease progression. J Mol Neurosci 20: 411 424(2003). 15 [5] Craft JM, Watterson DM, Frautschy SA and Van Eldikl U. Aminopyridazines inhibit p amyloid induced glial activation and neuronal damage in vivo. Neurobiol. Aging 25: 1283-1292 (2004). [6] Craft JM, Van Eldik U, Zasadzki M, Hu W, Watterson DM. Aminopyridazines attenuate hippocampus dependent behavioral deficits induced by human (J-amyloid in a urine model of 10 neuroinflammation. J Mol Neurosci 24: 115-122(2004). [7] Griffin WST, Sheng JG, Royston MC, Gentleman SM, McKenzie IE, Graham DI, et al. Glial-neuronal interactions in Alzheimers disease: the potential role of a "cytokine cycle" in disease progression. Brain Pathol 8: 65-72 (1998). [8] WermuthCG. Search for new lead compounds: The exampleof thechemical and 15 pharmacological dissection of aminopyridazines. J Heterocyclic Chem 35: 1091-1100 (1998). [9] Frautschy SA, Yang F, Calderon L and Cole GM Rodent models of Alzheimer's disease: rat A p infusion approaches to amyloid deposits. Neurobiol Aging 17: 311-21 1996). [10] Veber DF, Johnson SR, Cheng HY, Smith BR, Ward KW and Kopple KD Molecular 30 properties that influence the oral bioavailability of drug candidates. J Med Chem 45: 2615-2623 0 (2002). [11] Vieth M, Siegel MG, Higgs RE, Watson IA, Robertson DH, Savin KA, et al (2004) Characteristic physical properties and structural fragments of marketed oral drugs. J Med Chem 47: 224-232 (2004).
181 [12] Cignarella G, Barlocco D, Pinna G, Loriga M, Curzu MI, Tofanetti 0, et al. Synthesis and biological evaluation of substituted benzo[A]cinnolinones and 3H-benzo[6,7]cyclohepta[l,2 c]pyridazinones: higher homologues of the antihypertensive and antithrombotic 5H-indeno[l,2 c]pyridazinones. J Med Chem 32: 2277-2282 (1989). [13] Costantino L, Rastelli G, Vescovini K, Cignarella G, Vianello P, Corso AD, et al. Synthesis,activity, and molecular modeling of a new series of tricyclic pyridazinones as selective aldose reductase inhibitors. J Med Chem 39: 4396-4405 (1996). [14] Sotelo E and Ravina B. Efficient aromatization of 4 ,5-dihydro-3-(2H)-pyridazinones substituted at 5 position by using anhydrous copper (II) chloride. Synthetic Communications 30: 1-7 (2000). [15] Wermuth CO, Bourguignon JJ, Schlewer G, Gies JP, Schoenfelder A, Melikian A, et al. Synthesis and structure-activity relationships of a series of aminopyridazine derivatives of y aminobutyric acid acting as selective GABAj, antagonists. J Med Chem 30: 239-249 (1987). [16] Wermuth CG, Schlewer G, Bourguignon JJ, Maghioros G, Bouchet MJ, Moire C, et al. (1989) 3-amiaopyridazine derivatives with atypical antidepressant, serotonergic, and dopaminergic activities. J Med Chem 32: 528-537 (1989). [17] Mirzoeva S, Koppal T, Petrova TV, Lukas TJ, Watterson DM and Van Eldik LJ Screening in a cell-based assay for inhibitors of microglial nitric oxide production reveals calmodulin regulated protein kinases as potential drug discovery targets. Brain Res 844: 126-134 (1999). [18] Coudert, P.; Couquelet, J.; Tronche, P. A new synthetic route to 4,6-diarylpyridazinones and some of their derivatives. Joumal of Heterocyclic Chemistry. 1988, 25(3), 799-802.

Claims (29)

  1. 2. A method according to claim 1 comprising administering to the subject a compound of the Formula Ia or Ib 10 R2' R 7R 2 2 Qt R4 F R 4 tR 6 Re Re or wherein R', R 2 , and R 3 are independently hydrogen, hydroxyl, alkyl, alkenyl, alkynyl, 15 alkylene, alkenylene, alkoxy, alkenyloxy, cycloalkyl, cycloalkenyl, aryl, aryloxy, arylalkoxy, aroyl, heteroaryl, heterocyclic, acyl, acyloxy, sulfonyl, sulfinyl, sulfenyl, amino, imino, azido, thiol, thioalkyl, thioalkoxy, thioaryl, nitro, ureido, cyano, halo, silyl, silyloxy, silylalkyl, silylthio, =0, =S, carboxyl, carbonyl, carbamoyl, or carboxamide; R 7 is hydrogen, hydroxyl, alkyl, alkenyl, alkynyl, alkylene, alkenylene, 20 alkoxy, alkenyloxy, cycloalkyl, cycloalkenyl, aryl, aryloxy, arylalkoxy, aroyl, heteroaryl, heterocyclic, acyl, acyloxy, sulfonyl, sulfinyl, sulfenyl, amino, imino, azido, thiol, thioalkyl, thioalkoxy, thioaryl, nitro, ureido, cyano, halo, silyl, silyloxy, silylalkyl, silylthio, =0, =S, carboxyl, carbonyl, carbamoyl, or carboxamide or R7 may be absent and there is a double bond between N at position 1 and C at position 6; R 4 , R 5 , and R 6 25 are independently hydrogen, alkyl, alkoxy, halo, or nitro; or R' and R 2 , R 1 and R 7 , or R2 and R3 may form a heteroaryl or heterocyclic ring; or an isomer or a pharmaceutically acceptable salt thereof.
  2. 3. A method according to claim 1 wherein the heteroaryl is piperazinyl substituted with pyrimidinyl, or pyridinyl. 183
  3. 4. A method according to claim 3 comprising administering to the subject a compound of the Formula H1: R 1 R' NU 5 N-N wherein R1 0 and Ru are independently hydrogen, hydroxyl, alkyl, alkenyl, alkynyl, alkylene, alkenylene, alkoxy, alkenyloxy, cycloalkyl, cycloalkenyl, aryl, aryloxy, arylalkoxy, aroyl, heteroaryl, heterocyclic, acyl, acyloxy, sulfonyl, sulfinyl, sulfenyl, 10 amino, imino, azido, thiol, thioalkyl, thioalkoxy, thioaryl, nitro, ureido, cyano, halo, silyl, silyloxy, silylalkyl, silylthio, =0, =S, carboxyl, carbonyl, carbamoyl, or carboxamide; or an isomer or a pharmaceutically acceptable salt thereof.
  4. 5. A method according to claim 1 comprising administering to the subject a compound of the Formula III: 15 R' 5 R'1 N wherein R" and R" are independently hydrogen, hydroxyl, alkyl, alkenyl, alkynyl, alkylene, alkenylene, alkoxy, alkenyloxy, cycloalkyl, cycloalkenyl, aryl, aryloxy, 20 arylalkoxy, aroyl, heteroaryl, heterocyclic, acyl, acyloxy, sulfonyl, sulfinyl, sulfenyl, amino, imino, azido, thiol, thioalkyl, thioalkoxy, thioaryl, nitro, ureido, cyano, halo, silyl, silyloxy, silylalkyl, silylthio, =0, =S, carboxyl, carbonyl, carbamoyl, or carboxamide; or an isomer or a pharmaceutically acceptable salt thereof. 184
  5. 6. A method according to claim 1 comprising administering to the subject a compound of the Formula IV: R 70 ON R 7 1 0 N IV 5 wherein R 70 is substituted or unsubstituted hydrogen, hydroxyl, alkyl, alkenyl, alkynyl, alkylene, alkenylene, alkoxy, alkenyloxy, cycloalkyl, cycloalkenyl, aryl, aryloxy, arylalkoxy, aroyl, heteroaryl, heterocyclic, acyl, acyloxy, sulfonyl, sulfinyl, sulfenyl, amino, imino, azido, thiol, thioalkyl, thioalkoxy, thioaryl, nitro, ureido, cyano, halo, silyl, 10 silyloxy, silylalkyl, silylthio, =0, =S, carboxyl, carbonyl, carbamoyl, or carboxamide, especially heterocyclic, heteroaryl, amino, and substituted amino and R 7 1 is aryl or substituted aryl; or an isomer or a pharmaceutically acceptable salt thereof.
  6. 7. A method according to claim I comprising administering to the subject a compound of the Formula V: N N N -'N R51 5 v wherein R 50 , R 51 , and Rs 2 are independently hydrogen, hydroxyl, alkyl, alkenyl, alkynyl, alkylene, alkenylene, alkoxy, alkenyloxy, cycloalkyl, cycloalkenyl, aryl, aryloxy, 185 arylalkoxy, aroyl, heteroaryl, heterocyclic, acyl, acyloxy, sulfonyl, sulfinyl, sulfenyl, amino, imino, azido, thiol, thioalkyl, thioalkoxy, thioaryl, nitro, ureido, cyano, halo, silyl, silyloxy, silylalkyl, silylthio, =0, =S, carboxyl, carbonyl, carbamoyl, or carboxamide; or an isomer or a pharmaceutically acceptable salt thereof. 5 8. A method according to any preceding claim wherein the compounds depicted in Table 1 are excluded.
  7. 9. An isolated and substantially pure compound as defined in any preceding claim.
  8. 10. A compound according to claim 9 further comprising a carrier.
  9. 11. An isolated and substantially pure compound of Formula la or Ib RI R' R 2 R R 2 R5 RR R 4 R 6 10 or wherein R1, R 2 , and R 3 are independently hydrogen, hydroxyl, alkyl, alkenyl, alkynyl, alkylene, alkenylene, alkoxy, alkenyloxy, cycloalkyl, cycloalkenyl, aryl, aryloxy, 15 arylalkoxy, aroyl, heteroaryl, heterocyclic, acyl, acyloxy, sulfonyl, sulfinyl, sulfenyl, amino, imino, azido, thiol, thioalkyl, thioalkoxy, thioaryl, nitro, ureido, cyano, halo, silyl, silyloxy, silylalkyl, silylthio, =0, =S, carboxyl, carbonyl, carbamoyl, or carboxamide; R 7 is hydrogen, hydroxyl, alkyl, alkenyl, alkynyl, alkylene, alkenylene, alkoxy, alkenyloxy, cycloalkyl, cycloalkenyl, aryl, aryloxy, arylalkoxy, aroyl, heteroaryl, 20 heterocyclic, acyl, acyloxy, sulfonyl, sulfinyl, sulfenyl, amino, imino, azido, thiol, thioalkyl, thioalkoxy, thioaryl, nitro, ureido, cyano, halo, silyl, silyloxy, silylalkyl, silylthio, =0, =S, carboxyl, carbonyl, carbamoyl, or carboxamide or R7 may be absent and there is a double bond between N at position 1 and C at position 6; R 4 , R 5 , and R are independently hydrogen, alkyl, alkoxy, halo, or nitro; or R 1 and R 2 , R' and R 7 , or R 2 25 and R 3 may form a heteroaryl or heterocyclic ring; or an isomer or a pharmaceutically acceptable salt thereof, with the proviso that compounds of Table 1 are excluded. 186
  10. 12. An isolated and substantially pure compound of compound according to claim 11 wherein the heteroaryl is piperazinyl substituted with pyrimidinyl, or pyridinyl.
  11. 13. An isolated and substantially pure compound of Formula II: R 1 0 R 1 1 N N 5 II wherein RO and R" are independently hydrogen, hydroxyl, alkyl, alkenyl, alkynyl, alkylene, alkenylene, alkoxy, alkenyloxy, cycloalkyl, cycloalkenyl, aryl, aryloxy, arylalkoxy, aroyl, heteroaryl, heterocyclic, acyl, acyloxy, sulfonyl, sulfinyl, sulfenyl, amino, imino, azido, thiol, thioalkyl, thioalkoxy, thioaryl, nitro, ureido, cyano, halo, silyl, [0 silyloxy, silylalkyl, silylthio, =0, =S, carboxyl, carbonyl, carbamoyl, or carboxamide; or an isomer or a pharmaceutically acceptable salt thereof, with the with the proviso that compounds of Table 1 are excluded.
  12. 14. An isolated and substantially pure compound of of Formula III: R 1 6 R' 6 N Iny 5 wherein Ri 5 and R" are independently hydrogen, hydroxyl, alkyl, alkenyl, alkynyl, alkylene, alkenylene, alkoxy, alkenyloxy, cycloalkyl, cycloalkenyl, aryl, aryloxy, arylalkoxy, aroyl, heteroaryl, heterocyclic, acyl, acyloxy, sulfonyl, sulfinyl, sulfenyl, amino, imino, azido, thiol, thioalkyl, thioalkoxy, thioaryl, nitro, ureido, cyano, halo, silyl, 0 silyloxy, silylalkyl, silylthio, =0, =S, carboxyl, carbonyl, carbamoyl, or carboxamide; or an isomer or a pharmaceutically acceptable salt thereof; with the with the proviso that compounds of Table 1 are excluded. 187
  13. 15. An isolated and substantially pure compound of Formula IV: R74 N R 7 N IV wherein R 70 is substituted or unsubstituted hydrogen, hydroxyl, alkyl, alkenyl, alkynyl, 5 alkylene, alkenylene, alkoxy, alkenyloxy, cycloalkyl, cycloalkenyl, aryl, aryloxy, arylalkoxy, aroyl, heteroaryl, heterocyclic, acyl, acyloxy, sulfonyl, sulfinyl, sulfenyl, amino, imino, azido, thiol, thioalkyl, thioalkoxy, thioaryl, nitro, ureido, cyano, halo, silyl, silyloxy, silylalkyl, silylthio, =0, =S, carboxyl, carbonyl, carbamoyl, or carboxamide, especially heterocyclic, heteroaryl, amino, and substituted amino and R 7 1 is aryl or 10 substituted aryl; or an isomer or a pharmaceutically acceptable salt thereof; with the with the proviso that compounds of Table I are excluded.
  14. 16. An isolated and substantially pure compound of Formula V: N N N N R 5 * R 5 ' R 62 V wherein R 50 , Rsi, and R 2 are independently hydrogen, hydroxyl, alkyl, alkenyl, alkynyl, 15 alkylene, alkenylene, alkoxy, alkenyloxy, cycloalkyl, cycloalkenyl, aryl, aryloxy, arylalkoxy, aroyl, heteroaryl, heterocyclic, acyl, acyloxy, sulfonyl, sulfinyl, sulfenyl, amino, imino, azido, thiol, thioalkyl, thioalkoxy, thioaryl, nitro, ureido, cyano, halo, silyl, 188 silyloxy, silylalkyl, silylthio, =0, =S, carboxyl, carbonyl, carbamoyl, or carboxamide; or an isomer or a pharmaceutically acceptable salt thereof; with the with the proviso that compounds of Table 1 are excluded.
  15. 17. A prodrug comprising a compound as defined in any preceding claim wherein one or 5 more of radicals of the compound comprise a cleavable group that is cleaved after administration to a subject to provide a therapeutically effective amount of the compound.
  16. 18. A pharmaceutical composition comprising a compound as defined in any preceding claim, and a pharmaceutically acceptable carrier, excipient, or vehicle.
  17. 19. A method for treating a disease requiring modulation of one or more of inflammation, 10 signaling pathways involved in inflammation, cell signaling molecule production, activation of glia or glial activation pathways and responses, proinflammatory cytokines or chemokines, oxidative stress-related responses, acute phase proteins, components of the complement cascade, protein kinase activity, cell damage, and cell death signal transduction pathways in a subject comprising administering to the subject a 15 therapeutically effective amount of a compound according to claim 9 or 10.
  18. 20. A method for treating in a subject a disease involving or characterized by inflammation comprising administering a therapeutically effective amount of a compound according to claim 9 or 10.
  19. 21. A method for treating in a subject a condition associated with neuroinflammation that 20 can be decreased or inhibited with a compound of the Formula I according to any preceding claim comprising administering to the subject a therapeutically effective amount of a compound according to claim 9 or 10.
  20. 22. A method for reducing or inhibiting kinase activity, glial activation, neuronal cell damage, and/or neuronal cell death in a subject comprising administering to the subject a 25 therapeutically effective amount of a compound according to claim 9 or 10, and a pharmaceutically acceptable carrier, excipient, or vehicle.
  21. 23. A method of inhibiting cell signaling molecule production comprising administering compositions comprising one or more compounds according to claim 9 or 10.
  22. 24. A method of claim 17 wherein the signaling molecule is IL10 and/or TNFa. 30 25. A method of delaying the progression of a neuroinflammatory disease in a subject comprising administering to the subject a therapeutically effective amount of a compound according to claim 9 or 10 and a pharmaceutically acceptable carrier, excipient, or vehicle. 189
  23. 26. A method for amelioriating progression of a disease or obtaining a less severe stage of a disease in a subject suffering from such disease comprising administering a therapeutically effective amount of a compound according to claim 9 or 10.
  24. 27. A method of any preceding claim wherein the disease is a dementing disorder, a 5 neurodegenerative disorder, a CNS demyelinating disorder, an autoimmune disorder, or a peripheral inflammatory disease.
  25. 28. A method according to any preceding claim wherein the disease is Alzheimer's disease.
  26. 29. A method of increasing survival of a subject suffering from Alzheimer's disease 10 comprising administering a therapeutically effective amount of a compound of the Formula I or a composition comprising a compound of the Formula I according to any preceding claim.
  27. 30. A method for treating mild cognitive impairment (MCI) in a subject comprising administering to the subject a therapeutically effective amount of a compound of 15 according to claim 9 or 10.
  28. 31. Use of a compound according to claim 9 or 10 for the preparation of a medicament for treating a disease.
  29. 32. A kit comprising a compound according to claim 9 or 10 for preventing and/or treating a disease, a container, and instructions for use. 20 Dated the 21st day of August 2012 Northwestern University; Centre National de la Recherche Scientifique and Universit4 de Strasbourg Patent Attorneys for the Applicant PETER MAXWELL AND ASSOCIATES
AU2012216322A 2004-11-02 2012-08-22 Pyridazine compounds, compositions and methods Abandoned AU2012216322A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
AU2012216322A AU2012216322A1 (en) 2004-11-02 2012-08-22 Pyridazine compounds, compositions and methods
AU2016203312A AU2016203312B2 (en) 2004-11-02 2016-05-20 Pyridazine compounds, compositions and methods

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US60/624,346 2004-11-02
US60/723,124 2005-10-03
US60/723,090 2005-10-03
AU2005302225A AU2005302225B2 (en) 2004-11-02 2005-11-02 Pyridazine compounds, compositions and methods
AU2012216322A AU2012216322A1 (en) 2004-11-02 2012-08-22 Pyridazine compounds, compositions and methods

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU2005302225A Division AU2005302225B2 (en) 2004-11-02 2005-11-02 Pyridazine compounds, compositions and methods

Related Child Applications (1)

Application Number Title Priority Date Filing Date
AU2016203312A Division AU2016203312B2 (en) 2004-11-02 2016-05-20 Pyridazine compounds, compositions and methods

Publications (1)

Publication Number Publication Date
AU2012216322A1 true AU2012216322A1 (en) 2012-09-06

Family

ID=46785944

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2012216322A Abandoned AU2012216322A1 (en) 2004-11-02 2012-08-22 Pyridazine compounds, compositions and methods

Country Status (1)

Country Link
AU (1) AU2012216322A1 (en)

Similar Documents

Publication Publication Date Title
AU2005302225B2 (en) Pyridazine compounds, compositions and methods
EP2063894B1 (en) Formulations containing pyridazine compounds for treating neuroinflammatory diseases
US8158627B2 (en) Compositions and treatments using pyridazine compounds and cholinesterase inhibitors
WO2007130383A2 (en) Compositions and treatments using pyridazine compounds and secretases
US8063047B2 (en) Pyridazine compounds and methods
AU2012216322A1 (en) Pyridazine compounds, compositions and methods
AU2016203312B2 (en) Pyridazine compounds, compositions and methods

Legal Events

Date Code Title Description
MK5 Application lapsed section 142(2)(e) - patent request and compl. specification not accepted