AU2016203312B2 - Pyridazine compounds, compositions and methods - Google Patents

Pyridazine compounds, compositions and methods Download PDF

Info

Publication number
AU2016203312B2
AU2016203312B2 AU2016203312A AU2016203312A AU2016203312B2 AU 2016203312 B2 AU2016203312 B2 AU 2016203312B2 AU 2016203312 A AU2016203312 A AU 2016203312A AU 2016203312 A AU2016203312 A AU 2016203312A AU 2016203312 B2 AU2016203312 B2 AU 2016203312B2
Authority
AU
Australia
Prior art keywords
compound
disease
substituted
formula
subject
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.)
Ceased
Application number
AU2016203312A
Other versions
AU2016203312A1 (en
Inventor
Jean-Jacques Bourguignon
Jacques Haiech
Marcel Hibert
Wenhui Hu
Linda Van Eldik
Anastasia Velentza
Martin D. Watterson
Magdalena 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
Priority claimed from AU2012216322A external-priority patent/AU2012216322A1/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 AU2016203312A priority Critical patent/AU2016203312B2/en
Publication of AU2016203312A1 publication Critical patent/AU2016203312A1/en
Application granted granted Critical
Publication of AU2016203312B2 publication Critical patent/AU2016203312B2/en
Ceased legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Landscapes

  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

dATRACT The invention relates to novei chemical compounds. compositions and methods of making and using the same in particular; the invention provides pyridazine compounds and/or related hete rocydlic derivatives. composlitions comprising the same, and methods of making ad using pyridazine compounds and/or related heterocyclic derivatives and composions composing the same, for modulation of cellular pathways (e~g signal transduction pathways), for treatment or prevention of inflammatory diseases (eg Alzhmer disease); for research, drg screening, and therpeud applations.

Description

The invention relates to nove; chemical compounds, compositions and methods of making and using the same. In particular, the invention provides pyddazine compounds· and/or related heterocyclic derivatives, compositions comprising the same, and methods of making and using pyhdazine compounds and/or related heterocyclic derivatives and compositions comprising the same, for modulation of cellular pathways (e.g, signal transduction pathways), for treatment <»· prevention of inflammatory diseases (eg Alzheimer's disease?, lor research, dmu soroening, and therapeutic applications.
2016203312 20 May 2016
Patents Act, 1990 ORIGINAL
COMPLETE SPECIFICATION
APPLICANT'S: Northwestern University,
Centre National De La Recherche Sdentlflque and
Umvershe de Strasbourg
INVENTORS: WATTERSON, Martin D
VAN ELDIK, Linda HAIECH, Jacques HIBERT, Marcel
BOURGUIGNON, Jean-Jacques VELENTZA, Anastasia HU, Wenhui ZASADZKI, Magdalena
ADDRESS FOR SERVICE : Peter Maxwell and Associates
Level 6 60 Pitt Street SYDNEY NSW 2000
INVENTION TITLE: PYRIDAZINE COMPOUNDS,
COMPOSITIONS AND METHODS
DIVISIONAL OF AU - 2012 216 322- 2 November 2005 which Is a divisional of AU - 2005 302 225 - 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\20051275\411751 .doc
2016203312 20 May 2016
PYR1DAZJNE CO WOUNDS, COMPOSITIONS AND METHODS
This invention was funded, in part, under ΝΪΗ grants POj AG21184 and R01NS47586.
The US government may have eestain rights in the invention.
BBIATED 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
60/723,124, filed October 3,2003, and 60/723.000, filed October 3,2005.
HELD OF INVENTION
The invention relates to novel chemical compounds, compositions and methods of making and using the same, fit particular, the invention provides pyridasine compounds and/or related heterocyclic derivatives, compositions comprising the same, and methods of using pyridazme 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.
BACKGROUND OF ΙΝΛΈΝΠΟΝ
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 them may be an overproduction of protisflammatory or other detrimental molecules drat mediate toxic cellular responses. Disreputation of signal transduction pathways involving protein kinases am often involved in the generation or progression of these diseases. For example, nemoinfliunmution is a process that results primarily from an abnormally high or chronic activation of glia (microglia and astrocytes). Ibis overactive state of glia results in increased levels of irfiammatory and oxidative stress molecules, which can lead to neuron damage or death. Neuronal damage/death can also induce glial activation, facilitating the propagation of a localized, detrimental cycle of neuroinfitmunation [7j.
The Inflammation (e.g,, nenroinflammatlon) cycle has been proposed as a potential therapeutic target in the development of new approaches to treat inflammatory disease (e.g,, Alzhehneris disease). However, the efficacy mid lexicological profile of compounds that focus only on classical non2016203312 20 May 2016 steroidal anti-inflamtnatoty' drug targets have been disappointing to date, for example, most antiinflammatory hierapeutics arc palliative, providing minimal, short-lived, symptomatic relief with limited effects on inflammatory disease (&g., netroiuflammafory diseases such as Alzheimer's disease) progression. Because the major societal impact from inflammatory diseases (e.g., mmomflammatery diseases such as Aizhehnea’s disease) is expected to increase gready 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 chemopreventive paraoigm as combinations of risk factors wife prognostic value are identified. In either therapeutic paradigm, now drugs must have a good thempettoc index, especially in regard to 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 neurohrflammation [1]. Thus, development of new classes of anti-inflartuuatery compounds that can modulate inflammatory disease-relevant pathways are urgently needed,
SUMMARY OP INVENTION
The present invention relates to novel chemical compounds, compositions and methods of making and using the same. In particular, the present invention provides pyridaxine compounds 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 substifoted heteroaryl, in particular piperazinyl substituted with pyiirmdinyl, or pyridinyk
In an 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 & compound of the Formula I, comprising I a and lb:
Formula 1
2016203312 20 May 2016
Figure AU2016203312B2_D0001
wherein Rl, R2, and R3 are independently hydrogen, hydroxyl, alkyl, alkenyl, alkynyl, alkylene, alfcenylene, alkoxy, alkenyloxy, cydoalkyl, cycloalkenyl, aryl, aryloxy, arylalkoxy, aroyl, heteroaryl, heterocyclic, acyl, acyloxy, sulfonyl, sulfinyl, sulfenyl, amino, imino, azido, thiol, thioalkyl, thioalkoxy, thioaryl, nitro, urn do, cyano, halo, silyl, silyloxy, silylalkyl, silylftio, ~O, -S, carboxyl, carbonyl, carbamoyl, or carboxamide; R7 is hydrogen, hydroxyl, alkyl, alkenyl, alkynyl, alkylene, alkenylene, alkoxy, alkenyloxy, cycloalkyl, cycloalkenyl, aryl, aryloxy, arylalkoxy, aroyl, heteroaryl, heterocyclic, acyl, acyloxv, sulfonyl, sulfinyl, sulfenyl, amino, imino, azido, thiol, thioalkyl, thioalkoxy, thioaryl, nitro, ureido, cyano, halo, silyl, silyloxy, silylalkyl, silyllhio, -O, ~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; R4, R5, and R* are independently hydrogen, alkyl, alkoxy, halo, or nitro; or R1 and R2, R1 and R7, or R3 and R3 may form a heteroaryl or heterocyclic ring; or an isomer or a pharmaceutically acceptable salt thereof.
In an embodiment, Rl is a piperazinyl or substituted piperazinyl, in particular a piperazinyl substituted with a pyrimidinyl of Formula A below.
vuwuvwtv
Ν'
Ν'
Figure AU2016203312B2_D0002
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 compound of the Formula II;
2016203312 20 May 2016
Figure AU2016203312B2_D0003
U wherein and Rn are independently hydrogen, hydroxyl, alkyl, alkenyl, alkynyl, alkylene, alfeenylenc, alkoxy, alkenyloxy, cycloalkyl, cycloalkenyl, aryl, aryloxy, arylalkoxy, aroyl, beteroaryl, heterocyclic, acyl, acyioxy, sulfonyl, sulfinyl, sulfenyl, amino, imino, addo, thiol, thioalkyl, thioalkoxy, thioaryl, nitro, uroido, cyano, halo, silyl, sllyloxy, 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 particular an inflammatory disease, in a subject comprising administering to the subject a compound of the Formula HI:
Figure AU2016203312B2_D0004
wherein R1S and K/0 are independently hydrogen, hydroxyl, alkyl, alkenyl, alkynyl, alkylene, alkenylene, alkoxy, alkenyloxy, cycloalkyl, cycloalkenyl, aryl, aryloxy, arylalkoxy, aroyl, faeteroaryl, heterocyclic, acyl, acyioxy, sulfonyl, sulfinyl, sulfenyl, amino, imino, azido, thiol, thioalkyl, thioalkoxy, thioaryl, nitro, ureido, cyano, halo, siivl, sllyloxy, silylalkyl, silylthio, ™O, ™S, carboxyl, carbonyl, carbamoyl, or carboxamide; or an isomer or a pharmaceutically acceptable salt thereof.
In another aspect, die 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;
2016203312 20 May 2016
Figure AU2016203312B2_D0005
wherein R'° is substituted or unsubstituted hydrogen, hydroxy], alkyl, alkenyl, alkynyl, alkylene, alkenyiene, alkoxy, alkenyloxy, cycloalkyl, cycioalkenyl, 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, ~O, ™S, carboxy], carbonyl, carbamoyl, or carboxamide, especially heterocyclic, heieroaryl, amino, and substituted amino and R'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 heating a disease disclosed herein, in particular an inflammatory disease, in a subject comprising administering to the subject a compound of the Formula V:
Figure AU2016203312B2_D0006
wherein Rs, RS1, and R52 are Independently hydrogen, hydroxyl, alkyl, alkenyl, alkynyl, alkylene, alkeuylene, alkoxy, alkenyloxy, eydoalkyl, cycloaJkenyl, 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, =G, ~S, carboxyl, carbonyl, carbamoyl, or carboxamide; or an isomer or a pharmaceutically acceptable salt thereof.
2016203312 20 May 2016 fe an aspect, a method is provided for treating s disease disclosed herein in a subject, comprising administering a compound of the Fonnola I, Π, ΠΙ, IV, or V as defined heroin, with the proviso that compounds depleted in Table 1 ars 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 die Formula I, II, DI,IV, or V, or a pharmaceutically acceptable salt thereof, or a composition comprising a compound of the Formula I, Π, 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 heating fe a subject a disease involving or characterized by inflammation, in particular neuroinfiaxnmation, comprising administering to the subject a therapeutically effective amount of a compound, of the Formula I,
II, DI, IV, or V, or a pharmaceutically acceptable salt thereof. fe a further aspect, a method is provided for treating in a subject a condition involving inflammation, in particular nouroflammation, comprising administering to the subject a therapeutically effective amount of a composition comprising a compound of the Formula I, Π, ID, IV, or V and a pharmaceutically acceptable carrier, excipient, or vehicle.
fe a further aspect, the invention provides a method Involving administering to a subject a therapeutic compound of the Formula I, H, 3Π, IV, or V, or a pharmaceutically acceptable salt thereof, or a composition comprising a compound of the Formula I, U, Iff. IV, or V, and a pharmaceutically acceptable carrier, excipient, or vehicle which inhibit, or reduce neuroflammatlon, activation of glia, proimflammatory cytokines, oxidative stress-related enzymes, acute phase proteins and/or components of the complement cascade.
fe another aspect, the invention provides a method for treating in a subject a disease associated with neuroinilannnation that can be decreased or inhibited with a compound disclosed herein comprising administering to fee subject a therapeutically effective amount of a compound of fee Formula I, Π, IH, fe7, or V, a pharmaceutically acceptable salt thereof, or a composition comprising a compound of the Formula I, Π, Iff, fe7, or V, n particular the compounds depicted in fee Figures and Tables, mote particularly Table 2, 3,4 or 5 and derivatives thereof and an isomer or pharmaceutically acceptable carrier, excipient, or vehicle.
Methods of fee 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
2016203312 20 May 2016 activation, neurons! cell damage, and/or neuronal cell death; inhibit ceil signaling molecule production (e.g., Π.,-Ιβ and TNF&), amelioriate progression of a disease or obtain a less severe stage of a disease in a subject suffering from such disease (e,g,, nemoinflammatory disease, in particular a neurodegenerative diseae, mom particularly Alzheimer’s disease); delay the progression of a disease (e.g, neuromflmnmajtory disease, in particular a neurodegenerative disease, more particularly Alzheimer’s disease), increase survival of a subject suffering from a disease (e.g. neuroinflam.ro story disease, in particular a neurodegenerative disease, more particularly Alzheimer’s disease; teat or prevent a neurodegenerative disease, in particular Alzheimer’s disease; treat mild cognitive impairment (MCI); reverse or inhibit nenroinflammation, activation of signaling pathways involved in inflammation (e,g., neuroinflammation), cell signaling molecule production, activation of glia or glial activation pathways and responses, proirfelmrun&tory cytokines or chemokrnes (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 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, wherein the mammal has oo 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 compound of the Formula I, II, ffl, IV, or V, or a pharmaceutically acceptable salt thereof, or a composition comprising a compound of the Formula 1, II, III, IV, or V and a pharmaceutically acceptable carrier, excipient, or vehicle.
The Invention relates to compounds of the Formula I. with the proviso that dm compounds depleted in Table 1 are excluded.
The invention relates to compounds of the Formula H with die proviso that the compounds depicted in Table 1 are excluded.
The invention also relates to compounds of the Formula ΙΠ with the proviso feat compounds depicted in Table 1 are excluded,
2016203312 20 May 2016
The invention also relates to compounds of the Formula IV with the proviso (fast compounds depleted 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,
A compound of the Formula I, H, SI, IV, or V may optionally comprise a carrier
Interacting with one or more radicals in the compound, for example R , R , R, R, R s R or R 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.
In accordance with aspects of the invention pyridazins 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, tee present invention provides new classes of chemical, compounds capable of modulating pro-mfiammatory and oxidative stress related, cellular signaling pathways (e.g., in activated glial cells), tit 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 those depicted in the Figures and Table 2, 3, 4, and/or 5 and derivatives thereof. In some embodiments, tee inveution provides MW01-3-5-183WH, MWO1-5-188WH, MWOI-2» O65UKH MWOI-2-.1.84 WH, MWGI-24S9WH and MW01-2451SRM and methods of synthesizing toe same.
In some embodiments, the invention provides MW01-3-5-183WH, MWO1-5-I88WH,
MWOT2T65IKM, MW01-2-184WH, MW01-2-151SRM, MW01-2-189WH, MWGl-i-Ol-LD07, and/or related heterocyclic derivatives of these compounds and methods of malting and using die same for modulating cellular pathways (e.g., signal transduction pathways) for use in research, drag screening, and therapeutic applications.
In an aspect, the invention provides compositions for prevention and/or treatment of a disease disclosed herein. Thus, the invention provides a pharmaceutical composition comprising a compound of the Formula I, Π, BI, Ft7, or V, in particular a therapeutically effective amount of a compound of the Formula I, U, Id, 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, die invention provides a pharmaceutical composition in a form adapted for administration to a
2016203312 20 May 2016 subject to provide therapeutic effects, in particular beneficial effects to treat a disease disclosed herein.
ht another aspect, the composition is is a fonn such that administration to a subject suffering from a disease results in a decrease or reversal of one or more of the following:
inflammation (e.g. neuromflammation), activation of signaling pathways involved in inflammation (e.g., neuroinflammation), cell signaling molecule oroduction, 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 (e.g,, neuronal cell death), A composition of fee invention can be in a form that results in one or more of a decrease or reversal of one or more of fee following: inflammation (e.g. neuroinflammation), activation of signaling pathways involved in inflammation (e.g,, neuroiaflammatisn), 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 stressrelated responses such as nitric oxide synthase production and uitric 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 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. neuroinflamrnation), activation of signaling pathways involved in inflammation (e,g„ neuroinflammation), cell signaling molecule production, activation of glia or glrel activation pathways and responses, proinflarn.mato.ty cytokines or chemokines (e.g., interleukin (IL) or tumor necrosis factor (INF), 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 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, III, IV, or V. Alter a composition is prepared, it can be placed in an appropriate container and labeled for treatment
2016203312 20 May 2016 w
of an indicated disease. For administration of a composition of fee 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, 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, II, III, 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 bound by any particular theory, the compounds and compositions may act to ameliorate die course of a disease using without limitation one or mom of fee following mechanisms: preventing, reducing and/or inhibiting inflammation (e.g, neuroinflammation), activation of signaling pathways involved in inflammation (e.g., nemoinflammarion), cell signaling molecule production, activation of glia or glial activation pathways and responses, proiufiammatory cytokines or chemokines (e.g., interleukin (IL) or tumor necrosis factor (TNF), oxidative stressrelated 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).
The invention relates to the use of a composition comprising at least one compound of the Formula I, II, III. 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 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, Η, 111, IV, or V or a composition comprising a. compound of the Formula I, SL, DI, IV, or V. In an aspect, the invention provides a kit for preventing and/or heating a disease disclosed heroin comprising one or more compound of fee Formula I, fl, HI, 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.
2016203312 20 May 2016 π
The compounds of the Formula 1, II. Ill, IV. or V (in particular die 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. neuroinflammatiou), activation of signaling pathways involved in inflammation (e.g., neuroinflammatiou), ceil signaling molecule production, activation of glia or glial activation pathways and responses, proinfl&nnnstory cytokines or chemokines (e.g., interleukin (11,) 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 ceil death), wherein the compound comprise a structure of Formula I, H, HI, 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, Hs HI, IV, or V, in particular a compound depicted in Table 2, 3, 4, and/or 5 or derivatives thereof. In particular, the invention
IS contemplates a combinatorial library comprising compounds for decreasing or reversing one or more of the following; i.ufiamruahou (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 (INF), 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), wherein the compound comprise a structure of Formula I, Π, UI, IV, or V.
These and other aspects, features, and advantages of the present invention should be apparent to those skilled in the art from the followmg detailed description.
DESCRIPTION OF THE FIGURES
Figure I depicts a synthetic scheme for synthesis of MWG1-34S3WH.
Figure 2 A and B are graphs showing concentration dependent inhibition of proinflammatory cytokine production by MWQ1--3-183WH in BY-2 cells
Figure 3 A- H show graphs and micrographs of activity of MW01-5 188WH. A is a graph of )IL-lp and (B) TNFa levels by the BV2 microglial cell line. (€} Accumulation of the NO metabolite, nitrite, was not inhibited; Western blots of iNOS, CON-2 or apoE production in activated glia in (£>); iNOS, (E) COX-2 and (F) apoE from glia cultures. Micrographs of treatment with diluent and compound am shown in (G) and (H).
2016203312 20 May 2016
Figure- 4 A-H shows graphs and micrographs of activity of MW01~5~188WH after oral administetion.
Figure 5 A-C shows graphs of results of oral administration of MW01-5-188WH.
Figure 6 Ά-Β shows graphs and ixnmnnohfots illustrating the cell-based activity of
MW01 -2-151SRM in B V-2 microglial cells.
Figure. 7 A-G shows graphs illustrating in vivo activity of MW0I-2-151SRM in the Αβ infusion mouse model. Graphs are of MW01-2-151SSM suppression of Αβ-induced nenroinfiammation and. synaptic damage and activity in the Y-maze. Hippocampal sections or extracts from vehicle-infused mice (control), Αβ-mfused mice injected with solvent, and Αβ10 infused mice injected with MW01-2-151SRM were evaluated for neuroinilammutiou by measurement of the levels of the pro-inflammatory cytokines IL-Ιβ (A), TNFa (B), and S100B (C), and the number of GFAP-positive astrocytes, (D) and the presynaptic marker, synapfophysm, (E), and evaluated for synaptic damage by analysis of the levels of the post-synoptic density protein 95 (PSD-95) (F), and Y-maze. Data are from one of two independent experiments, and are the mean + SEM for 4-5 mice per experimental. group.
Figure 8 is a synthetic scheme for MW01-7-084WH.
Figure 9 is a synthetic scheme for MW01-7-085WH,
Figure .10 is a synthetic scheme for MW01-7-09lWK Figure 11 is a synthetic scheme forMW01-2-065LRM,
Figure 12 is a synthetic scheme for MW0I-2-069A-SRM.
Figure 13 is a synthetic scheme for MW01-2-1515RM.
Figure 14 is a synthetic scheme for MW01 -2-151SRM.
Figure 15 is a synthetic scheme for MW01-2-I51SRM.
Figure 16 is a synthetic scheme for MW01-5-188WH.
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- 189WB.
Figure 20 is a synthetic scheme for MWQ1-7 -029WH.
Figure 21 is a synthetic scheme forMW01-7'027B~WH.
Figure 22 is a synthetic scheme for MWO1-3-065SRM.
Figure 23 is a synthetic scheme for MW01-3-066SRM.
Figure 24 is a synthetic scheme for MW01-7-133 WFL Figure 25 is a synthetic scheme for MW01-7-107WH.
Figure 26 is a synthetic scheme for MWO1-7-057WH.
2016203312 20 May 2016
Figure 27 is a synthetic scheme: forhiW01-2-16'3MAS. figure 28 is a synthetic scheme for MWG1-7-084WH.
Figure 29 A-C shows graphical data of the assays used herein for MWGI-2--Q56WFI,
Figure 30 A-E shows graphical data of the assays used herein for MW01-2-056WI1
Figure 31 A-C shows graphical data of the assays used herein and immunobfots for biological activity of MW01-7-057WFL
Figure 32 A~E shows graphical data of the assays used herein for MW01-7-057WH,
Figure 33 A-C shows graphical data of the assays used herein and immunoblots for biological activity of MW01-2-065LKM.
Figure 34 A-E shows graphical data of the assays used herein for MW01-2-065LKM.
Figure 35 A-C shows graphical data of the assays used herein and immunoblots for biological activity of MW01-2-069A-SRM.
Figure 36 A-E shows graphical data of the assays used herein for MW01-2-069A-SRM. Figure 37 A-C show's graphical data of the assays used herein and immunoblots for biological activity of MW01-7-085WH,
Figure 38 A-E shows graphical data of the assays used herein for MWO1-7-O85WFL Figure 39 A-C shows graphical data of the assays used herein and inununoblots for biological activity of MW01-7-091WH.
Figure 40 A~E shows graphical data of the assays used herein for MW01-7-G91WH,
Figure 41 A-C shows graphical data of the assays used herein and inununoblots for biological activity of MW01-7-1O7WH.
Figure 42 A-E shows graphical data of the assays used herein for MW01-7-107WH.
Figure 43 A-C shows graphical data of the assays used herein and immunoblots for biological activity of MW01-7-127WH,
Figure 44 A-E show's graphical data of the assays used herein for MW01-7-127WH.
Figure 45 A-C shows graphical data of the assays used herein and inununoblots for biological activity of MW01-2-151SRM.
Figure 46 A-H shows graphical data of the assays used herein for MWG1-2-151SRM. Figure 47 A and B are graphs of response to MW01-2-151SRM, in a test for toxicity 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 <A„B) and rat (C,D) microsomes with MW01-2-151SRM in two different amounts, for two tune periods. B and F show' human
2016203312 20 May 2016 (F) and (F) rat microsomes with MW01-2-151SSM stability for different time periods compared to minaprine.
Pi gate 49 A-C shows graphical data of the assays used herein and immurufelots for biological activity of MW01-2-163MAS,
Figure 50 A-C shows graphical data of the assays used herein and imruunoblots for biological activity of MW01-6-189WH.
Figure 51 A-D shows graphical data of fee assays used herein for MWGlriS-WWH,
DETAILED DESCRIPTION
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 feat all numbers and fractions thereof ate presumed to be modified by the term about, The term “about” means plus or minus 0.1 to 50%, 5-50%, or 10-40%, preferably 1020%, 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.
As used herein fee 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 fee subject’s clinical condition, the site and method of administration, dosage, patient age, sex, body weight, and other factors known to physicians.
As used herein, the term ’’co-administration’’ refers to fee administration of at least two compounds or agent(s) (e.g., compound of fee Formula Ϊ, Π, ΠΙ, IV, or V or pyridazines) or therapies to a subject. In some embodiments, the co-administration of two or more agents/feerapies is concurrent In other embodiments, a first agent/therapy is administered prior to a second agentftherapy. Those of skill in fee ari understand that the Formulations and/or mutes of administration of fee various agenLVfeera.pi.es used may vary. Tire appropriate dosage for coadmifestration can be readily determined by one skilled in fee art. In some embodiments, when agents/therapies axe co-admimsteied, the respective agents/therapies are administered at lower dosages than appropriate for their administration alone, Thus, oo-adminisfration is especially desirable in
2016203312 20 May 2016 embodiments whew the C(>admim-shatxon 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 on© or more symptoms of such disease, to winch such term applies. Depending on 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 wife a disease, A treatment may be either perforated in an acute or chronic way, Tire term also refers to reducing fee 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 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 fee act of treating, as treating” is defined above.
Tbs terms “subject”, “individual”, or “patient” are used interchangeably herein and refer to an animal preferably a warm-blooded animal such as a mammal, Mammal includes without limitation any members of fee Mammalia. In general, fee terns refer to a human. The terms also include domestic animals bred for food or as pets, including equines, bovfees, sheep, poultry5, fish, porcines, canines, felines, and zoo animals, goats, apes (e.g. gorilla or chimpanzee), and rodents such as rats and mice,
In aspects of the invention, fee terms refer to organisms to be- treated by fee methods of fee present invention. Such organisms preferably include, but aw not limited to, mammals (e.g., murines, simians, equines, bovines, porcines, canines, felines, and the like), and most preferably includes humans. In fee context of particular aspects of fee invention, the term subject generally wife’s to an individual who will receive or who has received treatment (e,g., administration of a compound of the Formula I, li, IU, IV, or V or a pyridazine compouud(s), and optionally one or more other agents) for a condition characterized by inflammation, fee dvswgulation of protein Irinas© activity, and/or dysmguiation 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 feat have suffered a disease disclosed herein. A subject may or may uot 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 fee invention the subjects are suspectible to, or suffer from Alzheimer’s disease.
2016203312 20 May 2016
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, toe term modulate refers to the activity of a compound (e,g.f a compound of tire Formula I, Π, HI, TV, or V, or a pyridazane 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 fee invention, the beneficial effects include without limitation prevention, redaction, reversal, nr inhibition of one or more of the following: inflammation (e.g. necroinfiammation), 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 chemokises (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 some aspects, a beneficial effect is a favourable characteristic of a composition comprising a compound of the Formula I, fi, HI, IV. or V including without limitation enhanced stability, a longer half life, and/or enhanced uptake and transport across tire 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, H, DI, IV, or V versus fee effects without the compound or compound that is not within the scope of tire- invention. Statistically significant’ or significantly different” effects or levels may represent levels that are higher or lower than a standard, In aspects of tire invention, the difference may he 1.5, 2, 3,4, 5, or 6 times higher or lower comp .red with tire effect obtained without a compound of the Formula I, Π, 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
2016203312 20 May 2016 diluents, binders, adhesives, lubricants, disintegrates, bulking agents, wetting or emulsifying agents, pH buffering agents, and miscellaneous materials such as absorbants 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 of such media and agents for an active substance is well known in the art.
The compounds of the Formula I, 11., 111, IV, or V disclosed herein also include pharmaceutically acceptable salt(s)n. 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 benefit/risk ratio. Pharmaceutically acceptable salts are described for example, in S. M Berge, et at, J. Pharmaceutical Sciences, 1977, 66:1. Examples of salts include MWGi-1-0! -L-D10, MW0H-01-L-E02, MWOl-l-Ol-L-EOS, MW01-1-03-L-A05, MW01-1-16-L-D09, and MW011-17-L-G04.
A compound of the Formula I, Π, HI, IV, or V can contain one or more asymmetric
IS 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, Η, IE, 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 of the Formula I, II, III, IV, or V contains centers of geometric asymmetry, and unless specified otherwise, it is intended that the compounds inci ode both E and A geometric isomers. All tautomeric forms are also included within the scope of a compound of the Formula I, H, IH, IV, orV.
A compound of the Formula I, H, DI, IV, or V can exist in nnsolvated as well as solvated 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, Π, IS, IV, or V or composition comprising the same, that will lead to one or 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
2016203312 20 May 2016 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 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 heroin, refers to a chemically modified compound wherein tiro chemical modification takes place either at a functional group of the [0 compound or on tire aromatic ring. Non-limiting examples of derivatives of compounds of the Formula I, Π, ΙΠ, IV, or V (e.g,, pyridarine derivatives of the present invention) may include N-aeetyl, N-meihyl, N-hydroxy groups at atty of tire 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 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, aery!, styrene-, carbohydrate derived polymers, polyethylene glycol. (PEG), polyoxyethylene, polymethylerre glycol, poly-trime-thylene glycols, polyvinylpyrrolidone, polyoxyethylene-polyoxypropylene block polymers, and copolymers, salts, and derivatives
Z0 thereof. In aspects of tire invention, the polymer is poly(2-acrylamido-2-methyl-lpropanesulfonic acid); poly(2-acrylamido-2-methyl,-1 -propanesulfonk aeid-coacrylonitrile, poly(2-acryiamido~2-methyl-l-propanesulfonic acid-co-styrene). poly(vinylsulfenlc acid); poly(sodium 4-styrenesulfonic acid); and sulfates and sulfonates derived therefrom; poiy(acryiic acid), poly(methylaerylate), poly(methyl methacrylate), and poly(vmyl alcohol),
A “carbohydrate” as used herein refers to a polyhydroxyaldehyde, or polyhydroxyketorro aad 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 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 he substituted at one two, three or four positions, other than the position of linkage to a compound of tlie Formula I» Π, HI, TV, or V. For example, a carbohydrate may be substituted with one or more alkyl, amino, nitro, halo, thiol, carboxyl, or
2016203312 20 May 2016 hydroxyl groups, which are optionally substituted Blostrative substituted carbohydr'ates are glucosamine, or galaetosamine. In aspects of tire invention, the carbohydrate is a sugar, in particular a hexosc or pentose and may be an aldose or a fcetose. A sugar may be a member of the D or L series and can include amino sugars, deoxy sugars, and their uronic acid derivatives,
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, galaetosamine, glucosamine, in particular D-glucosamine (2~amino~2-doexy~Dglueose) or D-gaJactosamme (2-aasino-2-deoxy-D-galactose). Illustrative pentose sugars include arabinose, fucose, and ribose.
A sugar residue may be linked to a compound of the Formula I, Π, ΏΊ, TV, 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 1, Π, ΠΙ, IV, or V. An oxygen atom can be replaced one or more times by -Oij- or-S- groups,
The term “carbohydrate” also includes glycoproteins such as lectins (e.g. cancanavalin
A, wheat germ agglutinin, peanutaggiutimn, seromucoid, and orosomucoid) and glycolipids such as cerebrosids 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 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 carrier can be a polypeptide such as albumin, antitrypsin, macroglobulin, haptoglobin, caeruloplaxm, transferring, a- or β~ lipoprotein, 8- or γ- globulin or fibrinogen.
Approaches to designing peptide analogies, derivatives and mimetics are known in the art For example, see Farmer, F. S, in Drug Design (B. J. Ariens, ed.) Academic Press, New York, 1980. vol. 10, pp. 119-1.43; Bali. J. B. and Alewood, P. E (1990} J Moi. Recognition
3:55; Motg» , B. A, and Gainer, J. A, (1989) Ann. Rep. Med. Chem. 24:243; and Freidinger, R.
M. (1989) Trends Pharmacol. Sci. 10:270. Sec 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,
2016203312 20 May 2016
A. R. 3rd, et at (1995) J. Am. Chem. Soc. 117:11113-11123; Smith, A. B. 3rd, el al. ¢1994) 1 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, Π, Π1IV, or V through a functional group on the side chain of certain amino acids (e.g. serine) or other suitable functional groups. A earner may compose 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 cyateic add.
The term “alkyl, either alone or within other terms such as “thioalkyl and “uryialkyl”, 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~ butyl, n-pentyl, n-hexyl, isopropyl, isobutyl, isopentyl, amyl, sec-butyl, tert-butyl, tert-pentyl, nheptyl, n-octyl, n-nonyl, n-decyl, undecyl, n-dodecyl, n-tetradecyl, pentadecyl, n-hexadecyi, 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 CrCg lower alkyl comprising or selected fit u the group consisting of methyl, ethyl, n-propyl, n-butyl, n~ pentyl, n-hexyl, isopropyl, isooutyl, 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, 11, III, IV, or V and do not significantly reduce the- efficacy of the compounds, hr certain aspects of the invention, an alkyl radical is substituted with one to five substituents including halo, lower alkoxy, lower aliphatic, a substituted lower aliphatic, hydroxy, cyano, nitro, thio, annuo, keto, aldehyde, ester, amide, substituted amino, carboxyl, sulfonyl, sulfinyl, sulfenyl, sulfate, sulfoxide, substituted carboxyl, halogenated lower alkyl (e.g, Clfi), halogenated lower alkoxy, hydroxycarbonyl, lower alkoxycarbonyl, lower alkylcarbonyloxy, lower alkyicarbonylaroino, cycloaliphatic, substituted cycloaliphatic, or aryl (e.g., phenylmethyl (i.e.
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
2016203312 20 May 2016 (aryl, substituted aryl, cycloaliphatic, or substituted cycloaliphatic, etc.). Examples of such groups include, but are not limited to, 1-chloroethyl and tire like.
As used heroin in respect to certain aspects of the invention, the term ’’lower-alkylsubstituted-amiuo refers to any alkyl unit containing up to mid including eight, carbon atoms 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-alkylsubstituted-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, but. are not limited to, chiorethyl 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 die like.
As used herein the tern alkenyl refers to an uns&turated, acyclic branched or straightchain hydrocarbon radical comprising at least one double bond. An alkenyl radical may contain 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 ethenvl, propenyl (e.g., prop-l-en-l-yl, prop-l-en-2-yk prop-2-en~l~yi (allyl), prop-2-en-2-yI)s buten-lvh bat-1 -en-2-yl, 2~.methy.l~prop~l~en-l-yl, bnt-2-en-l-yl, bm-2-en-l-yl, but-2-en-2-yl, huta-kS-dien-l-yl, feuta-l,3~dien-2-yl, hexea-l-yl, 3-hydrexyhexen-l-yl, hepten-l-yl, and oeten-l-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 ethynyl, such as prop-1-yn-l-yl, prop-2-yn-l-yl, batynyls such as hut-.l-yn-.l-yl, but-l-yn-3-yJ, but-3-yn-l-yl, penlynyls such as pentyn-l-yl, pentyn-2-yl, 4-methoxypentyn»2-yls 3methylbutyn-l-yi, hexynyls such as hexyn-l-yl, hexyn-2-yI, hexyn-3-yl, and 3,3-dimethylbutyn1-yl radicals and the like. An alkenyl may be optionally substituted similar to alkyl. The term cycloalkynyl refers to cyclic alkynyl groups,
As used herein the term alkylene refers to a linear or branched radical having from about 1 to 10 carbon atoms and having attarhment points for two or more covalent bonds. Examples of such radicals are methylene, ethylene, propylene, butylene, pentyleae, hexylene-, ethyli.de-ne, methylethylene, and isopropylidene. When an alkenylens radical is present as a
2016203312 20 May 2016 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 more covalent bonds. Examples of alkenylene radicals include 1,1-vinylidene (-CH2“C~), 1,2vinylidene (-CH€H-); and 1,4-butadienyI (-CH™CH-CH~CB~).
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.
As used herein the term eyano refers to a carbon radical having three of four covalent bonds shared by a nitrogen atom, in particular -CsN. 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 be substituted. In aspects of the invention an alkoxy radical may comprise about 1-10,1~S or 1-6 carbon atoms. In embodiments of the invention, an alkoxy radical comprises about I-6 carbon atoms and includes a Cj-Q, alkyl-O-radicai wherein Ci-Ce 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 or mote 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, triflnoromethoxy, dsfiuoromethoxy, trifinoroethoxy, fluoroethoxy, tetrafluoroethoxy, pentafluoroethoxy, and fluoropropox) and haloalkoxyalkyl radicals (e.g. fluoromethoxymethvl, chloromethoxyethyl, trifluorometlioxymethyl, 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, propenyloxy, buteuyloxy, and isopropeuyloxy 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 haloaBrnyloxy radicals (e.g. trifluoroethenyloxy, fluoroethenyloxy, difluoroeihenyhloxy, and flaoropropenytoxy).
2016203312 20 May 2016
A “csrbocylic” includes radicals derived from a saturated or unstatnratsd, 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, cycloalkenyi, aryl, in particular phenyl, naphthyl, norixsmanyl, bicycloheptadienyl, toMyl, xylenyl, indenyl, stubenyl, terphenylyl, diphsuylethylenyi, pheaylcycfofeexyl, acenapththylenyl, anthracenyl, biphenyl, bibenzylyl, and related bibenzylyl homologs, oetahydronaphthyl» tetrahydronaphthyl, oetahydroqninolinyk dime&oxyteirahydronaphthyl and the like.
As used herein, the term cycloalkyl refers to radicals having from about 3 to 15 carbon atoms turd containing one. two, three, or four- rings wherein such rings may be attached in a 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 die cycloalkyl radicals are lower eyeloalkyT’ radicals having from about 3 to 8 carbon atoms, in particular cyclopropyl, eyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl. In some aspects of the invention the term “cycloalkyl embraces radicals where cycloalkyl radicals are fused with 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 aw not limited to, 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 aldehyde, an ester, an amide, a lower aliphatic, a substituted lower aliphatic, or a ting (aryl, substituted aryl, cycloaliphatic, or substituted. cycloaliphatic). Examples of such include, but are not limited to, Uchloredecalyl and. the like.
A used herein, the term cycloalkenyi 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 rings may be attached in a pendant manner or may be fused. In certain aspects of the invention the cycloalkenyl radicals are lower cyeloalkenyF radicals having three to seven carbon atoms. Examples of cycloalkenyi radicals include without limitation cydobutenyl, cyclopentenyl, cyclohexenyl and cycloheptenyh A cycloalkenyi radical may be optionally substituted with
2016203312 20 May 2016 groups as disclosed herein, in particular 1, 2, or 3 substituents which may be the saxae 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 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 he 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.
Illustrative aryl radicals includes without limitation aromatic radicals such as phenyl, benzyl, naphthyl, indenyl, benzocyclooctenyl, beuzocycloheptenyl, peatalenyl, azulenyl, tetrahydronaphthyl, indanyl, biphenyl, acephthylenyl, fiuorenyl, phenalenyl, phenarsferenyl, and anferacenyh 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 substituted aryl includes without limitation arylamine and aryl&Ikyamine,
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, 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, isoqoinolizinyloxy, and the like.
As used herein the term arylaikoxy,” refers to an aryl group attached to an alkoxy group. Representative examples of aryl alkoxy include, hut are not limited to, 2-phenyleihoxy, 3naphth-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 he optionally substituted with groups as disclosed herein.
As used herein the term heteroayl refers to fully unsaturated heieroatom-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 tings and fee rings may
2016203312 20 May 2016 be attached in a pendant manner or may be fused. Examples of hetoroaryl” radicals, include without limitation, an unsaturated 5 to 6 membered heterumonocyclyl group containing 1 to 4 nitrogen atoms, in particular, pyrrolvl, pyrroHnyi, imidazolyl, pyrazolyl, 2-pyridyI, 3-pyridyl, 4pyridyl, pyridinyl, pyrirmdinyh pyrazinyl, pyridazinyl, iriazolyl, tetrazolyl and the like; an unsaturated condensed heterocyclic group containing 1 to 5 nitrogen atoms, in particular, indolyl, isoindolyl, indolirinyl, indazolyl, quinazolinyl, pteridinyl, quinolizidinyl, phthalazinyl, naphthyridinyl, quinoxalinyl, cinnolinyl, phenanthridinyl, acridinyl, phenanthrolinyl, phenazinyl, carbazolyl, purinyl, benzhnidazolyl, quinolyl, isoquinolyl, quinolinyl, isoquinolinyi, indazolyl, benzotriazolyl, tetrazolopyridazinyl and the like; an unsaturated 3 to 6~membered heteromonocyelic group containing aa oxygen atom, in particular, 2-furyI, 3-ftsryl, pyranyl, and the like; an unsaturated 5 to 6-membered heteromonocyelic group containing a sulfur atom, in particular, thienyl, 2~thi.cnyl, 3~thienyl, and die like; unsaturated 5 to 6~membered heteromonocyelic group containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms, in particular, fiirazanyl, benzofurazanyl, oxazolyl, isoxazolyh and oxadiazolyl; an unsaturated condensed heterocyclic group containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms, in particular benzoxazolyl, benzoxadiazolyl and tire like; an unsaturated 5 to 6-membered heteromonocyelic group containing 1 to 2 sulfur atoms and 1 to 3 nitrogen atoms, for example, feiazolyl, isothiazolyl, thiadiazolyl and the like; an nnsatorated condensed heterocyclic group containing 1 to 2 sulfur atoms and 1 to 3 nitrogen atoms such as benzofeiazolyl, benzothiadiazolyl and the like. The term also includes radicals where heterocyclic radicals are fused with aryl radicals, in particular bicyeiic radicals such as benzofuranyl, benzothiophenyl, phthalazinyl, chromenyl, x&nthenyl, and the like, A heteroaryl radical may be optionally substituted with groups as disclosed herein, for example wife an alkyl, amino, halogen, etc., in particular a heteroarylamine.
Ute 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 he attached in a pendant manner or may be fused, Examplary saturated heterocyclic radicals include without liroitiation a saturated 3 to d-membered hetercmonocylic group containing 1 to
4 nitrogen atoms [e.g. pyrrolidinyl, isaidazolidinyl, piperidinyl, and piperazinyl]; a saturated 3 to
6-membered hereromonocyclic group containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms [e.g, morpholinyl; sydnonyl]; and, a saturated 3 to 6-membered heteromonocyelic 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 dihydrotliiophene,
2016203312 20 May 2016 dihydropyrsnyl, dihydroftiranyl and dihydrothiazolyl. Eluatrative heterocyclic radicals include without limitation aziridinyl, azetidinyl, 2~pynolinyl, 3-pyrroEnyl, pyrrolidinyl, azepinyl, 1,3™ dioxolanyl, 2H-pyranyl, 4H-pyranyl, piperidinyl, 1,4-dioxaayl, morpholinyl, pyrazolinyi, 1,4dithianyl, thiomorpholinyl, 1,2,3,6-tetrahydropyridinyl, ©xiranyl, oxetanyl, teteahydrofuranyl, teirahydropyranyl, tetrahydropyridinyl, teteahydrothiopyranyi, thioxanyl, mdohnyh 2H-pyranyl, 4H~pyranyl, dioxanyl, 1,3-dioxolanyl, pyrazolinyi, dihydropyranyl, dihydrothieny'J, dihydrofuranyl, pyrazohdinyl, iiaidazolinyl, iutidazolidinyl, 3H-indolyI, qninndidinyl, quinolizinyl, and the like.
As used herein in respect to certain aspects of tire invention, the team 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- Eased rings, where at least one of the ring carbon atoms is replaced by oxygen, nitrogen or sulfur. Examples of such include, hut 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 ring (aryl, substituted aryl, cycloaliphatic, or substituted cycloaliphatic). Examples of such include, hut are not limited to 2-chloropyranyL
The foregoing hetcroaryl 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 alkylsulfenyl or arylsulfosyl, refers to the divalent radicals -SO2”. hi aspects of the Invention a sulfonyl group, the sulfonyl group may be attached to a substituted or unsuhstitnted hydroxyl, alkyl group, ether group, alkenyl group, alkynyl group, aryl group, cycloalkyl group, cycloalkenyl group, cycloalkynyl group, heterocyclic group, carbohydrate, peptide, or peptide derivative.
The term sulfmyl, used alone or linked to other terms such as alkylsulfinyi (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
2016203312 20 May 2016 chemical formula -NR^R22 where R2i and R23 can be any combination of hydrogen, hydroxyl, alkyl, cyclo alkyl, 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 (-OB) to provide an amine known as a hydroxylamine.
Illustrative examples of amino groups are amino (-NFfe), alkylaraino, acylamino, cydoamino, acyclodkylannno, arylamino, arylalkylamino, and lower aikylsilylamino, in particular methylantino, ethylamino, dimethylamino, 2-propytemino, buiylammo, isobutylamino, eyclopropylamino, benzylamine, allylamino, hydroxylamino, cyclohexyiamino, piperidinyl, hydraanyl, benzylamine, diphenylmethylamino, tritylamino, ttiroethylsilylarnino, and dimethyl10 tem-butylsilylamino, which may or may not be substituted.
As used herein the term “thiol” means -SR A thiol may be substituted with a substituent disclosed herein, in particular a.kyi (thioalkyl), aryl (thioaryl), alkoxy (thioaikoxy) or carboxyl. The term “sulfenyl” used alone or linked to other terms such as alkylsulfenyl, refers to the radical -SR** wherein R24 is not hydrogen. In aspects of the invention R24 is substituted or unsubstituted alkyl, eycloalkyl, 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 tbiopropyk A thioalkyl may be substituted with a substituted or unsubstitute carboxyl, aryl, heteroeylie, 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] 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 formula -SR25 where B? is aryl which may be substituted, Illustrative examples of thioaryl groups and substituted thioaryl groups are ihsophenyl, chlorotltiophenyl, para-chlorothiophenyl, tlnobenzyl, 4-methoxy-thiophenyl, 4~mtro-lhiophenyl, and para-nitrothiobenzyL
As used herein the term ’thioalkoxy, alone or in combination, refers to a chemical functional group where a suitor atom (S) is bonded to an alkoxy group with the general
2016203312 20 May 2016 s 28 chemical formula -SR30 where R30 is an alkoxy group which may be substituted, A “thioalkoxy group” may have 1-6 carbon atoms i.e. a -S-(O)-C>.-C6 alkyl group wherein €; -Q alkyl have the moaning as defined above, Biustrativc examples of a straight or branched thioalkoxy group or radical having from 1 to 6 carbon atoms, also known as a Ci -C$ thioalkoxy, include feiomethoxy 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 hereon, the term “carboxyl”, alone or in combination, refers to -C(Q)ORH~ or C<™O)ORi4 wherein Ru is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, amino, thiol, aryl, heteroaryl, thioalkyl, thioaryi, thioalkoxy, a heteroaryl, or a heterocyclic, which may optionally be substituted. Examples of carboxyl groups are methoxycarbonyl, butoxycarbonyl, tertmlkoxycarbonyl such as tefobufoxyc&rbonyl, 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, meihoxybenxyloxyearbonyl, diphenylmethoxycarbonyl, 2-bromoethoxycarbonyl, 2iodoethoxycarbonykertbufylearbonyl, 4-nitrobenzyloxycarbosyl, ' diphenylmethoxy-cafbonyl, benzhydroxycaibonyl, di-(4-mefeoxyphenyl-methoxycarbonyl, 2-bromoefhoxycarbonyh 2iodoethoxycatbonyl, 2-trimethylsilylethoxycarbonyl, or 2~triphenylsilyleihoxycarbonyl. Additional carboxyl groups in esterified form are silyloxycaibonyl groups including organic silyioxycarbonyh In aspects of the invention, the carboxyl group may be an alkoxy carbonyl, in particular methoxy carbonyl, ethoxy carbonyl, isopropoxy carbonyl, t-butoxvcarhonyl, tpentyloxycarbonyl, or heptyloxy carbonyl, especially methoxy carbonyl or ethoxy carbonyl.
As used herein, the term carbamoyl, alone or in combination, refers to amino, monoalkylamino, diaikylamino, monocycloalkylamino, alkylcycloalkylamino, and dicycloalkylandno 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, fee term “nitro” means
As used herein, the term acyl, alone or in combination, means a carbonyl or fhiocarhonyl group bonded to a radical selected from, for example, optionally substituted, hydrido, alkyl (e.g. haioalkyl), alkenyl, alkynyl, alkoxy (“acyloxy” including acetyloxy, hutyryloxy, iso-valeryloxy, phenylacetyloxy, benzoyloxy, p-mefeoxybenzoyloxy, and substituted acyloxy such as alkoxyalkyl and haloalkoxy), aryl, halo, heterocyclyl, heteroaryl, sulfinyl (e.g. aikylsulfinylalkyl), sulfonyl (e.g. alkylsulfonylalkyl), cycloalkyl, cycloalksnyl, tttioalkyl, thioaryl, amino (e.g alkylamiuo or diaikylamino), and aralkoxy, Illustrative examples
2016203312 20 May 2016 of «gcyl radicals are formyl, acetyl, 2-chloroacetyl, 2-bromacetyl, benzoyl, trilluoroaeetyl, phthaloyl, malonyl, nicotinyl, and the like.
As used herein, “ureido” refers to the group “-NKCONIT. A ureido radical includes an alkylurddo comprising a ureido substituted with an alkyl, in particular a lower alkyl attached to the terminal nitrogen of the ureido group. Examples of an alkylureido include without limitation N’-methylureido, N’-ethylureido, N’-n-propylureido, N'-i-propylumido and the like. A ureido radical also includes a NyN’-dialkylureido group containing a radical -NHCON where the terminal nitrogen is attached to two optionally substituted radicals including alkyl, aryl, heterocylic, and heteroaryl,
The terras used herein for radicals including “alkyl”, “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 nonual valency is not exceeded, and that the substitution results in a stable compound.
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, Π, Hl, IV, or V may be substituted with one or more substituents apparent to a person skilled in the art including without limitation alkyl, alkoxy, alkenyl, alkynyl, alkanoyl, alkylene, alkenytene, hydroxyalkyl, haloalkyl, haloalkyiene, haloalkeny], alkoxy, alkenyloxy, alkenyioxyalkyl, alkoxyalkyl, aryl, alkylaryl, haloalkoxy, haloalkenyloxy, heterocyclic, heteroaryl, sulfonyl, alkylsulfonyl, sulfinyl, sulfonyl, sulfenyl, alkylsulfinyl, aralkyl, heteroaralkyl, cycloalkyi. cycloalkenyl, cycloalkoxy, cycloalkenyloxy, amino, oxy, halo, azido, thio, =0, ~S, oyano, hydroxyl, phosphonato, phosphinato, thioalkyl, alkylanri.no, arylammo, arylsulfonyl, alkylcarbonyl, arylcarbonyl, heterosrylcarhonyl, heteroaryteulfinyl, heteroarylsulfony, beteroarylamino, heteroaryloxy, heteroaryloxyk-tikyl, arylacetamidoyl, aryioxy, aroyl, arfdkanoyl, amlkoxy, aryloxyalkyl, haloaryloxyalkyl, hetcroaroyl, heteroaralkanoyl, heteroaralfcoxy, hetcroaralkoxyalkyl, thiosryl, aryithioalkyl, 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.
2016203312 20 May 2016
A “disease” tfiat 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,s neuroinflannnation), cell signaling molecule production, activation of glia or glial activation pathways and responses, proinflammatory cytokines or chemokines (e.g., interleukin (31-,) or tumor necrosis factor (TNF), oxidative stress-related responses such as nitne oxide synthase production and nitric oxide accumulation, acme phase proteins, components of the complement cascade, protein kinase activity (e.g,, death associated protein kinase (DAPK) activity), sell damage (e.g., neuronal cell damage), and sell death <e-.g„ neuronal cell death). In 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 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, pro-senile and senile forms; amyloid angiopathy; mild cognitive impairment; Alzheimer's disease-related dementia (e.g,, vascular dementia or Alzheimer dementia); AIDS related dementia, tauopatiues (e.g., argyrophilic grain dementia, cortieobasal 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, oon-Guamanian Motor Neuron disease with neurofibrillary tangles, .feck's disease, postencephalitic parkinsonism, cerebral amyloid angiopathy, progressive subcortical gliosis, progressive supranuclear palsy, subacute sclerosing paneneephalitis, and tangle only dementia), alpha-synucleinopathy (e.g., dementia wife Lewy bodies, multiple system atrophy with glial cytoplasmic inclusions), multiple system atrophies, Shy-Drag® syndrome, spinocerebellar ataxia (e.g., DRPLA or Machado-Joseph Disease); striatouigral 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,
2016203312 20 May 2016 ischemia, infarction, mhacerebral 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
Angiopathy, Danish Type; Familial encephalopathy with nenroserpin inclusion bodies (EENIB); 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 polyneuropathy; chronic infections and inflammations; acute disseminated encephalomyelitis (ADEM); autoimmune inner ear disease (A1ED); diabetes; myocardial ischemia and other cardiovascular disorders; pancreatitis; gout inflammatory bowel disease; ulcerative colitis, Crohn’s disease, rheumatoid arthritis, osteoarthritis; arthsriosclerosis, inflammatory aortic aneurysm; asthma; adult respiratory distress syndrome; restenosis; ischemia/reperfusion injury;
glomerulonephritis; sscoidosis cancer; restenosis; rheumatic fever; systemic lupus erythematosus; Reiter's syndrome; psoriatic arthritis; ankylosing spondylitis; eoxarthritis; pelvic inflammatory disease; osteomyelitis; adhesive capsulitis; oligoarthritis; periarthritis; polyarthritis; psoriasis; Still’s disease; synovitis; inflammatory dermatosis; and, wound healing.
hi aspects of the invention, a compound, composition, or method disclosed herein may be utilized to prevent and/or treat a disease involving neuroinfiammation (i.e., neuroinflammatory disease), Nenminflammatiou is a characteristic feature of disease pathology and progression in a diverse array of neurodegenerative disorders that am increasing in their societal impact (for a recent review, see, e.g,, Frusiner, S. B. (2001) New Engl. J. Med, 344,1516-1526), These neuroinflammation-related disorders include Alzheimer's disease (AD), am>tetrophie lateral sclerosis, autoimmune disorders, priori diseases, stoke and traumatic brain injury, Neuminflammatson is brought about by glial cell (e.g., astrocytes and microglia) activation, which normally serves & 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 oxidative stress-related enzymes, acute phase protons, and various components of the complement cascades. (See, e.g., Akiyama et ah, (2000) Neuroblol. Aging 21,383-421). The direct linkage of glial activation to pathology that is a hallmark of disease underscores the importance of uurtetending 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,
2016203312 20 May 2016
For Afetenefs disease (AD) in particular, the deposition of β-amyloid (Αβ) and neurofibrillary tangles are associated with glial activation, neuronal loss and cognitive decline. On a molecular level, Alzheimer's disease is characterised by; increased expression of nitric oxide synthase (NOS) in glial cells surrounding amyloid plaques; neuropathological evidence of peroxynitrite5 mediated neuronal damage; and nitric oxide (NO) overproduction involved in Αβ-induced brain dysfunction. NOSH (iNOS) is induced as pat of die 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-sterived molecule peroxynitrite is generated, leading to neuronal cell death. The pro-inflammatory cytoldne ΙΕ-Ιβ is also overexpressed in activated glia in AD brain and polymorphisms in IL-Ιβ genes are associated with an increased risk of early onset sporadic AD (See, e.g,, Du et al., (2000) Neurology 55,480-483), IL-Ιβ 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 Shag, et al., (1996) Neurobiol. Aging 17, 761-766). Tlrerefore, because glial activation and specific glial products are associated with neurodegenerative 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 inflammatoty 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
Bisregulation of protein kinase signaling often accompanies disre-gulafion of cell signaling pathways (e.g,s 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 titan 500 protein kinases and 130 protein phosphatases exerting tight control on protein phosphorylation. Each protein kinase transfers the y-phosphate of ATP to a specific residue^) 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, MARK kinase (MKK), AlxTKM fun kinase (INK), CDKr, protein kinase A (PRA), protein kinase C (PKC), and calmodulin (CaM)~ dependent kinases (CsMKs). Bisregulated protein kinase activity (e,g.s hyper- or hypo-active) leads to abnormal protein phosphorylation, underlying a groat number of diseases including diabetes, rheumatoid arthritis, inSfrmmation, hypertension, and proliferative diseases such as cancer. Therefore, because aberrant kinase activity is associated with inflammatory disease (e.g., neurodegenera Live disorders like Alzheimer's disease), tire compounds and compositions that are
2016203312 20 May 2016 disclosed herein that are capable of modulating kinases 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 R1, R2, and R3 are independently hydrogen, hydroxyl, alkyl, alkenyl, alkynyl, alkylene, alkenylene, alkoxy, alkenyloxy, cycloalkyl, cyoloalkenyi, aryl, aryloxy, arylalkoxy, aroyl, heteroaryh heterocyclic, acyl, acyloxy, sulfonyl, sulfinyl, sulfenyl, amino, imino, azido, thiol, thioalkyl, thioalkoxy, thioaryl, nitro, ureido, cyano, halo, silyl, silyloxy, silylalkyl, silylthio, -O, =S, carboxyl, carbonyl, carbamoyl, or carboxamide;or R7 is hydrogen, hydroxyl., alkyl, alkenyl, alkynyl, alkylene, alkenylene, alkoxy, alkenyloxy, eyeloalkyl, cycloalkenyl, aryl, aryloxy, arylalkoxy, aroyl, heteroaryl, heterocyclic, acyl, acyloxy, sulfonyl, sulfinyl, sulfenyl, amino, imino, azido, thiol, thioalkyl, thioalkoxy, tbioaryl, nitre, ureido, cyano, halo, silyl, silyloxy, silylalkyl, silylthio, -O, -S, carboxyl, carbonyl, carbamoyl, or carboxamide: R4, R5, and R5 are independently hydrogen, alkyl, alkoxy, halo, or nitro or R' may be absent with a double bond between N at position 1 and C at position 6; or R1 and R2, R* and R7, or R2 and RJ 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:
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? is -COOCH3, CH^CHCOOCHs, ~
CHsCHC(=O>phenyl, -CH-CH(C(^0)OCH3)2, -S-phenyl,
CH=€H(C0CH3)(CO0CH3), CH^CH(COOCH2CH3)3, ~pfaenyl~C0OCH3, -CH=CHCO25 phenyl, -CHaCIlfClXCl-bOH), -methylphenyl, R7 is hydrogen or -CHjOCHs, and R1,
R2, R4, Rs and R6 are hydrogen;
b) a compound wherein when R* is -0, R2 is cyano, Ra is ~C(ss0)OCH3, and R3, R4, R\ and B? are hydrogen;
c) a compound wherein when Rl is =0, R* is -methylthiophene or benzyl, R\ R4, Rs, Re, and R7 are hydrogen;
d) a compound wherein when R1 is -0, R2 is methyl, R3 is hydrogen, hydroxyl, chloro, or bromo, R'' is hydrogen or ethylmorpholinyl, and R2, R4, and Rs are hydrogen;
e) a compound wherein when R~ is methyl, R5 is chloro, bromo, or hydrogen, R7 is hydrogen or>~CH2CH?-morpholinyl, and R5 , RJ, R4, andR6 are hydrogen;
2016203312 20 May 2016
f) a compound wherein when R* is piperazlnyl., piperaxinyi substituted with pyddinyl, phenyl, or methyl, IV’ is hydrogen or methyl, tmdR3, R\ R\ and R* are hydrogen;
g) a compound wherein when R? is chloro or bromo, R2 is C1-C3 alkyl, phenyl, amino, benzyl, morpholinyl, chloro, -C(ssO)NH2, -NH2, C-.-C3 alkylphenyl, ~ΟΗ<£Ή3) 5 CHjCHCCi^h, benzylcbloro, and R3, R4, R3 and R6 are hydrogen;
h) a compound wherein when R1 is chloro or bromo, R3 is hydroxyl, chloro, bromo, CpCb alkyl, phenyl, or -NCCHsh, and RF R4, R5 and R* are hydrogen;
i) a compound wherein when R5 is chloro, R2 is methyl, R5 is hydroxyl, and RV R4, and R6 are hydrogen;
j) a compound wherein when R1 is chloro, R2, R'\ R\ R5 and Ks are hydrogen;
k) a compound wherein when R1 is hydroxyl, R2 is C1-C4 alkyl, and R3, R4, R5 and Re are hydrogen;
l) a compound wherein when R1 is -C1-C4 alkoxy, or Cj-th alkoxy substituted with ™ N(CH3)2, morphohnyl, or piperidinyl substituted with benzyl, R2 is hydrogen or methyl,
5, R4, R5 and R” are hydrogen, R7 is absent, hydrogen, or methyl;
m) a compound wherein when R1 is -SH, -SCH3, or -SCH2C(-O)CHi, R2 is hydrogen or methyl, and R3, R4, R and R*5 are hydrogen;
n) a compound wherein when R1 is ~3, R2 is hydrogen or methyl, R7 is methyl or benzyl, and R3, R\ and Rw are hydrogen;
0) a compound wherein when R1 is -S, R* is methyl and R5 is chloro or R? is methyl, and
R3, RR and R& are hydrogen:
p) a compound wherein when R; is hydroxyl, R2 is hydrogen, methyl, or butyl, and R\ R4, R5 and R6 ate hydrogen;
q) a compound wherein when R* is methoxy; R2, R\ R\ R5 and R° are hydrogen;
r) a compound wherein when R? is C1-C2 alkoxy or Cj-Q alkoxy substituted with inorpholinyl, -XC'CHsjfe, or piperidinyl substituted with benzyl, R2 is methyl, and R3, R4, Rs and R6 are hydrogen;
s) a compound wherein R2, R3, R4, Rs and R6 ate hydrogen;
t) a compound wherein R3 is cyano or evano substituted with -C(OCH;.CIi3)2, 10 CHCOHXCBs), -SitCHsCH;!);:, cyclohexol, -CBsO-ttimethyldiphenylsilyl or cyclobexyl substituted with hydroxyl, and R3, R'\ R5 and R® ate hydrogen;
u) a compound wherein R1 is cyano substituted with -CH(OB)(CH3}2, -Si(CH>CBj)2.; morphollnyl, trimethyldiphesylsilyk or -CH(OCH2CH352, R2 is methyl, and R3, R4, R5 and R6 are hydrogen;
2016203312 20 May 2016
v) a compound wherein R7 is oxy, and R2 is hydrogen or methyl, and R3, R4, R5 and R6 are hydrogen;
w) a compound wherein R? is methyl, and R2, R3, R4, R5 and R6 are hydrogen;
x) a compound wherein R2 is methyl, and R5, S3, R4, R5 and Rs are hydrogen;
y) a compound wherein R1 is methcxycarbenyl, R3 is hydrogen, and R2, R y Rs and R6 are hydrogen;
z) a compound wherein R1 is -NTU, P? is methyl, ehlorophenyl, methoxyphenyl, ethyiphenyl, ethylmethoxyphenyl, propylphenyl, or -CH(CH3)2, R4, Rs and 86 are hydrogen, andR7 is absent or --CH2CH2CH2COOH;
aa) a compound wherein R5 is -OR50 wherein R80 is ethylmorpholinyl or -CHsCIfeNCCHah and Rz, RJ, R\ Rs and R6 are hydrogen;
bb) a compound wherein R'1 is ~NH2, R-’ is -NH2, and R\ R4, R'“ and P? are hydrogen; cc) a compound wherein R* is --NHa, R5 and R~ are methoxy, arid R3 and R4 are hydrogen; dd) a compound wherein R.3 is ~NH2, R3 is methyl and R4, R5 and R& an hydrogen;
ee) a compound wherein R1 is -NIU, R' is chloro, and R3, R4 and R4 are hydrogen;
£Q a compound wherein R3 is ™NH-o.hIorophenyl, and R2 and R3 form a phenyl group, and R4 R5 and R6 are hydrogen;
gg) a compound wherein E? is ~NH2, R4 and R5 is methoxy, and R2, R3 and Rfi are hydrogen; hip a compound wherein R1 is -NH2, R2 is ethylmethoxyphenyl, R7 is carboxyethyl or carboxypropyl, and R3, R4 and R6 are hydrogen;
ii) a compound wherein R1 is -NHR22 wherein R22 is ethylmorpholinyl, ethylmorpholinyl substituted with «Ο, -CiiaCHsOCBb, CH2CH2CH3, CH2CH2CH2CH2GH, -CH2CH2OH, or ~CH2CH3OCH3, R2 is hydrogen, methyl, ethyl, CHO, -CH2OH, -COOH, chloro, -CH2.CH2NH2, NO2, -ON, -C(~G)OCH2CH3j or25 C(=O)NH2, and R3, R4, R5 and R6 are hydrogen;
jj) a compound wherein R? is -NHR22 wherein R22 is ethanol, methyl piped dinyibenzyl, etirylpiperidinyL ethyjpipeddmylbenzyl, or buiylpiperidinylbenzyl, Rz is hydrogen, methyl, or -C(CHS)2, and R3, S4, R5 and R6 are hydrogen;
kk) a compound wherein RA is NHR22 wherein R22 is hydrogen, and RJ, R4, Rs and R* are 30 hydrogen;
II) a compound wherein R* is -NHR22 wherein R22 is -OfcCHsNCCHaCHah or ethylmorpholinyl, RJ is ethyl, and R4, Rs and R* are hydrogen;
2016203312 20 May 2016 36 min) a compound wherein R3 is -NHKHfe, R3 is hydrogen, alkyl, car phenyl, and R3, R \ Rs aud R& are hydrogen;
nn) a compound wherein R1 is -NHR22 wherein R22 is NBj, -CH^CTBOB, CH2CH(OH)(GH3)s etliylmotpholinyl, ethylmorpholinyl substituted with ~O, efhylphen.y.1, -CHsCEfeNHCHs, 'CHsCBj.Nf'-CBjCHaCH.ifi, ethylpiperidinyl, or ethylpiperidinylbenzyl, R2 is methyl, and R3, R4, R5 and Rs are hydrogen;
<x>) a compound wherein R1 is morpholinyl, R1' is -COP)», --0(-0), “CEfcOH, -C(™O)H, « COOH, chloro, -NO;., or eyano, and R3, R4, R5 and IV are hydrogen;
pp)a compound wherein R1 is -NBRz2 wherein R22 Is heptyl, phenyl, benzyl, or 10 ethylphenyl, R2 is hydrogen, methyl, or chlorophenyl, R4, R5 and Rs are hydrogen;
<jq)a compound wherein Rl is -NR2i wherein R23 is phenyl and R2, R\ R.\ Rs and R6 are hydrogen;
ir) a compound wherein R3 is morpholinyl and R2, R3, R4, R5 and R6 are hydrogen; as) a compound wherein R1 is methylpiperazinyl and R2, R3, R4, R and P? are hydrogen;
tt) a compound wherein R3 is -NHCH2CH2OB or NHCH2CH2OCH3, R~ Is phenyl and R3, R4, R5 sod R6 are hydrogen;
uu)a compound wherein R5 is -NBR22 wherein R22 is ethylamino, butylamiuo, ethylaminomethyl, and R2 is hydrogen, methyl, or ~C(”O)N%, and R3, R4, R5 and R6 are hydrogen;
vv) a compound wherein R1 is is -NHR22 wherein R22 is ethylpiperidinyl, methylpiperidinylbenzyl, piperidinylbenzyl, ethyipiperidinylhenzyl, methylpyrrolidinylmethyl, e&ylpiperaziny Ibenzyl, ~CH2C(~O)»piperazinylbenzyl? C(=O)-methylnaphthyl, -CWCHsCHs^CBaNOfeXCdf?), -CH2C^O)· piperidinylbenzyl, ~C(™O)> methylpiperidinylbenzyl, or -CHCCHjh, and R3, R4, R5 and
R6 are hydrogen;
ww) a compound wherein R1 is -CHCHsCHa-ssoquinolinyl,
NliCBsCHsNCCIiiCHiCI-R'h, propyl substituted with piperidinyl fused to phenyl, ~ NBC.H2.CH7., or “NHCHsCBsCH^CHiCH^ substituted with a piperidinyl fused to two adjacent carbon atoms of a phenyl moiety;
xx) a compound wherein R3 is -NH substituted with two pyrrehdinyl groups; R3 is methyl, and. R2, R4, R5 and Rs are hydrogen;
yy) a compound wherein R3 is -COOCH3, R3 is methyl, and R2, R4, R5 and Re fire hydrogen; zz) a compound wherein R* is hydrogen, R'1 is methyl, R7 is oxygen;
2016203312 20 May 2016
.. 37 aaa) a compound wherein R' is methyl or oxygen, and R3, R2, R4, R5 and R6 are hydrogen;
bbb) a compound wherein R1 is -NHCHjCHsNCCHaCHsis, R5 is ethyl, and R2, R4, R5 and R'5 are- hydrogen; and ccc) a compound wherein R3is -NHCH:jCH(OH)(CHj) or NHCH2CH2WCH2CH20iL R:? is methyl, and R3, R4, R5 and R« are hydrogen.
In an aspect, a compound of the Formula I is provided wherein: (a) R3 is optionally substituted halo, hydroxyl, alkyl, alkenyl, alkoxy, cyano, amino, cycioalkyl, -sulfonyl, sulfinyl, suifenyl, thioaryl, thioalkyl, carbonyl, silyl, piperazinyl, piperidinyl, pyrrohdinyl, morpholinyl, 10 SR24 wherein S?4 is optionally substituted alkyl, carbonyl, carboxyl, carbamoyl, aryl, heterocylic, or heteroaryl; (b) R2 is optionally substituted halo, hydroxyl, alkyl, alkenyl, alkoxy, carbonyl, carboxyl, phenyl, benzyl, amino, aryl, cyano, -COH, piperazinyl, alcohol, piperidinyl» morpholinyl, or naphthyl:(c) R3 is hydrogen, halo, hydroxyl, alkyl, alkenyl, alkoxy, phenyl, piperazinyl, piperidinyl, pyrrohdinyl, morpholinyl, thiol, suifenyl, sulfonyl, sulfinyl, or nitro; (d)
R4 is hydrogen, halo, or nitro (e) R5 is hydrogen, halo, alkoxy, or amido;(f) R7 Is hydrogen halo, hydroxyl, alkyl, alkenyl, alkoxy, carboxy, morpholino; imidazolyl; piperazinyl, piperidinyl, pyiTolidinyl, morpholinyl or R7 is absent and there is a double bond between N at position 1 and C at position 6; and/or (g) R3 and R‘, R1 and Rz or R2 and R3 may fonn a heteroaryl or heterocyclic ring,
Jn another aspect of the invention a compound of the Formula I is provided wherein R1 is Cl or Rr, -NFF, alkyl, -CN, -S, silyl, sulfonyl, thioalkyl, thioaryl, piperazinyl, piperazinyl, piperidinyl, piperidinyl, mmphoiinyl, pyrrohdinyl, pyrrolyl, or pyrrohdinyl, which may be optionally substituted with halo, ~O, alkoxy, alkenyl, alkyl, substituted alkyl, -CN, -SR24 wherein R24 is optionally substituted methyl, ethyl, phenyl, heterocylic, or heteroaryl, or -CO substituted with phenyl or substituted phenyl,
In another aspect of the invention a compound of the Formula I is provided wherein Rz is carbonyl, piperazinyl, morpholinyl, sulfonyl, sulfinyl, suifenyl, or phenyl, -CN, -COH, CH’OH, -OCH^CHii, or alkyl which may be optionally substituted with alkyl, alkoxy, amino, halo, phenyl, substituted phenyl, benzyl, hydroxyl, amino, piperidinyl, or morpholinyl.
In another aspect of the invention a compound of the Formula I is provided wherein 1C is piperazinyl; substituted piperzinyl; alkyl which may optionally be substituted with amino; phenyl; substituted phenyl; amino which may be optionally substituted with alkyd or alkylamine (e.g., NHCOOCfCHsh), carboxyl, or substituted carboxyl; hydroxyl; or nitro.
2016203312 20 May 2016
In another aspect of the invention a compound of the Formula I is provided wherein 1Z4 is nitro or hydrogen.
In another aspect of the invention a compound of the Formula I is provided wherein R5 is hydrogen, halo, -OCHjCFIsCH^NHCOOCfCHsls, or -OCHs5 In another aspect of the Invention a compound of the Formula I b provided wherein K;
is alkyl, morpholinyl, benzyl, imidaxolyl, -ChbCOOCHsCHa, CH2C=COOCH2CH3s CH2CH2CH2SO2OH, CHjCHjCHsSOf, CH3CH2CH2CH3PO(OH)2> or CH2CH2CH3pO(OH)2.
In another aspect of the invention a compound of the Formula 1 is provided wherein R7 is absent and there is a double bond, between N at. position 1 anti C at position 6.
In a further aspect, a compound of the Formula I is provided wherein R1, R2, R3, and R7 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 pipemmyl which may be substituted (e.g., with a pyrimidlnyl moiety); halo; amino which may be substituted; cvano; - SR24 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 he substituted; hydroxyl; - OR3* wherein R35 is alkyl; -C“ CHR*'6 wherein R** is alkyl; or pyrrohdinyh
In a still further aspect the invention a compound of the Formula I is provided wherein
R2 is hydrogen; morpholinyl; piperazinyl which may be substituted (e.g,, with a pvrimidinyl moiety); phenyl; alkyl; alkoxy (e.g. CH(OCH3}2); substituted alkyl; substituted aryl (e.g., phenyl); cvano; or hydroxyl.
In a still further aspect the invention a compound of tire Formula 1 Is provided wherein RJ is hydrogen; hydroxyl; alkyl which may be substituted (e.g., halo); annuo which may be substituted; -COR37 wherein R37 is hydrogen, hydroxyl, alkoxy (e.g. -OCHs); 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; Rc is hydrogen or halo.
In a still further aspect the invention a compound of the Formula. I is provided wherein
R7 is hydrogen; alkyl which may be substituted (e.g, with phenyl); -CH^CHsCOGR?8 wherein R38 is alkyl, -CUzC-CGOR38 wherein R38 is alkyl, CH2CHzCH2S(O)2OH. morpholinyl, benzyl, imidazolyl, or [CH2]aPO(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 R2 form a piperidinyl ring which may optionally be substituted walls a carboxyl.
2016203312 20 May 2016
In a still further aspect the invention a compound of the Formula I. is provided wherein S? and R' form a pyrimidinyl ring which may optionally be substituted with alkyl, aryl, halo, or hydroxyl, in a particular aspect. R‘ is -NR^R22 wherein R2i is hydrogen, and R22 Is hydrogen, 5 alkyl, carbonyl, aryl, annuo, cycloalkane, heterocylic, or heterosryl which may be substituted. In embodiments R22 may comprise or be selected from the group consisting of hydrogen, Cy Gs alkyl (e.g, methyl or ethyl! which may be substituted with optionally substituted hydroxyl, alkyl, amino, carbonyl, carboxyl, morpholinyl· isoquinolinvl, or an amino which may be substituted with one or more of optionally substituted alkyl, benzyl, carboxyl, alcohol group, heteroaryl or heterocyclic, a propanol group, phenyl which may be optionally substituted with halo, benzyl which may be substituted with alkoxy, cyclohexyl, piperidinyl which may he substituted with optionally substituted phenyl· pyrrolidiuyl or pyrrolidinylafkyl which may be substituted with alkyl, -COOK40 wherein R43 is alkyl which may be substituted, or [CH?Jlr/pipcridinyl wherein m is 1 to 4, in particular 1 to 3 and the pjperidinyl is optionally substituted with optionally substituted alkyl, phenyl, or benzyl.
In embodiments, R'2 is -R63R63 wherein R61' is -NHfCHajwNH wherein w is 1 to 4, in particular 2 or 3, and R5i is piper&rinyl substituted with pyrimidinyl which may be substituted, in particular substituted with alkyl·
In embodiments, R22 is ~R62R wherein R’“ is -[CH2.]WN(CH3) wherein w is .1 to 4, in 20 particular 2 or 3, and R*J 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 R1 is halo especially chloro or bromo, R2 is alkyl which may be substituted, in particular substituted with alkoxy (e.g., methoxy, diroeihoxy}, substituted aryl which may be substituted with alkyl, alkoxy, (e.g., benzyl, methoxy phenyl), halo (e.g. bromo or chloro), 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 heteromonocyclie group containing 1 to 2 oxygen atoms and I to 3 nitrogen atoms [e.g, morpholinyl; sydoonyl], in particular a substituted morpholinyl, piperazinykor piperazinyl substituted with a heteroaryl in particular an unsaturated 5 to 6 membered heteromonoeyclyl group containing 1 to 4 nitrogen atoms, in particular, pyrrolyl, pyrrohnyl, imidazolyi, pyrazolyl, 2-pyridyl, 3-pyridyl, 4-pytidyl, pyridinyl, pyrimidinyl, pyrazhtyl, or pyridazinyl, especially pyrimidinyl, and optionally ΚΛ R5, Rs, and R7 are hydrogen.
2016203312 20 May 2016
In another aspect of the invention., a compound of the Formula I is provided wherein R5 is halo especially chloro or bromo, and R3 is a substituted or unsnhstituted saturated 3 to ti~ membered heteromonocylic group containing 1 to 4 nitrogen atoms [e.g.s piperidinyl, and piperaxinyl] or a saturated 3 to ό-mernbered heteromonocyelie group containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms [e,g. morpholinyl; sydnonyl], in particular a substituted morpholinyl, ptperazinyhor pipersxwyl substituted with alkyl or a heteroaryl In particular an unsaturated 5 to 6 membered heleromonocydyl group containing 1 to 4 nitrogen atoms, in particular, pyrrolyl, pyrrolinyl. hnidaxolyl, pyrazolyi, 2-pyridyl, 3-pyridyl, 4-pyridyI, pyridinyl, pyrimidinyl, pyrazinyl, or pyridazinyl, especially pyrimidinyl, or R2 Is a substituted amino, in .0 particular amino substituted with alkyl or substituted alkyl, in particular alky] substituted with alkoxy carhony, and optionally R2, R4, Rs, R6, and R7 are hydrogen.
In further aspect R3 is halo, especially bromo or chloro, and R2 and R3 form as unsaturated ring, in particular phenyl, R5 is a heteroaryl, in particular a substituted or ussubstituted unsaturated 5 to 6 membered heteromonocyelyl group containing 1 to 4 nitrogen atoms, in particular, pymolyl, pyrrohuyl, itnidazolyl, pyrazoiyl, especially inaidazolyi, and optionally R4, Re and R7 are hydrogen.
In a further aspect, R1 is halo, especially bromo or chloro, and R* is nitro, and optionally R2, R3, R5, R0. and R7 are hydrogen.
In a further aspect, the invention provides a compound of the Formula 1 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. pyrroiidinyl, imidazoiidmyl, piperidinyl, and piperaanyl] or a saturated 3 to 6~membered heteromonocyelie group containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms [e.g. morpholinyl; sydnonyl], especially a substituted morpholinyl or piperidinyl; aryl;
substituted sty]; carboxyl which may be substituted with substituted or unsubstituted aryl; optionally R2 is alkyl, in particular lower alkyl; optionally R3 is alkyl, In particular lower alkyl or nitro; optionally R5 is alkoxy; optionally R7 is alkyl; and optionally R4, Rss and R6, am hydrogen.
In a further aspect of the invention, a compound of the Formula I Is provided wherein R1 is »S, and optionally R2 is alkyl, in particular lower alkyl, R5 is alkoxy, and R'\ R4, R* and R7 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 R2 is alkyl and R3, R4, R5, R/\ and R7 are hydrogen.
2016203312 20 May 2016
In a further aspect of the invention, a compound of the Formula I is provided wherein K! is substituted or onsubsthnted alkyl or alkynyl, in particular alkyl substituted with aryl, substituted aryl, halo, cyano, or alkynyl substituted with alkyl; and optionally R2 is alkyl, R7 is alkyl, and R3, R4, R5, and Rs are hydrogen.
In a further aspect of tire invention, a compound of the Formula I is provided wherein R5 is cyano and RA is aryl or alkyl, and optionally R3, R4, R5, R6, and R7 are hydrogen.
In a further aspect of the invention, a compound of the Formula I is provided wherein one or both of R5 and R2 are a saturated 3 to 6-membered heteromonocyctic group containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms (e.g. morphoiinyh sydnonyl], especially a substituted rnorpholinyi, and optionally R'\ R'\ Rs, R6, and R7 are hydrogen.
In a further aspect of the invention, a compound of the .Formula I is provided wherein R1 is a saturated 3 to 6-membered heteromonoeylic group containing 1 to 4 nitrogen atoms [e.g, pyrrolidinyl], which may be substituted with substituted or unsubstituted carboxyl; R2 is alkyl or halo, and optionally R3, R.t Rs, Rs, and R7 are hydrogen.
In a further aspect of the invention, a compound of the Formula I is provided wherein Rr is hydroxyl; R2 is alkyl or substituted alkyl or R3 is a saturated 3 to 6-membered heteromonocylic group containing 1 to 4 nitrogen atoms [e.g. piperidiuyi, and piperazinyl] which may optionally be substituted with a heteroaryl [e.g.. pyrimidinyl], and the other of R2 or R3 is hydrogen, and optionally R\ R5, Rfl, and R: are hydrogen,
In a further aspect of the invention, a compound of the Formula I is provided wherein R5 is a saturated 3 to 6-memhered heteromonocylic group containing 1 to 4 nitrogen atoms [e.g,, piperidiuyi and piperazinyl] which may be substituted with carboxyl or carboxyl substituted with alkyl or alkoxy or with purinyl or substituted purinyl; R2 is alkyl or substituted alkyl, in particular alkyiaryl, and optionally R3, R\ Rs, R®, and R7 are hydrogen.
.5 In a further aspect of the invention, & compound of fee Formula I is provided wherein R5 is 'O, and R2 is alkyl, alkyiaryl, cyano, alkoxy, or substituted alkoxy, and optionally R\ R4, R5, Rs, and R? are hydrogen.
In a further aspect of the invention, a compound of fee Formula 1 is provided wherein R1 is alkoxy, RA is alkyl, substituted alkyl, or alkoxy, and optionally R3, R4, R3, R®, and R7 are hydrogen.
In a further aspect of the invention, a compound of the Formula I is provided wherein Rx and R2 form a heterocyclic, in particular a saturated 3 to 6-membered heteromonocylic group containing I to 4 nitrogen atoms, in particular a 6-membered ring comprising 1 or 2 nitrogen
2016203312 20 May 2016 atoms [e.g„ japeridinyl and piperazinyl] which may be substituted for example wife alkyl, halo, carboxyl, or alkoxy carbonyl, and optionally R'\ R4, R5, Iff, and R7 are hydrogen.
In a further aspect of the invention, a compound of the Formula I is provided wherein B? and R' form a heteroaryl. in particular an unsaturated 5 to 6 membered hetcromonocyclyl group containing 1 to 4 nitrogen atoms, In particular, pyrrolyl pvrrolinyh imidazolyl, pyrazoivl, 2pyndyl, 3-pyridyl, 4-pvridyl, pyridinyl, pyrimidinyl, pyrazinyl. or pyridazinyl, R2 is hydrogen or alkyl, and R3, R4, Rl R® and R7 are hydrogen.
In a further aspect of the invention, a compound of the Formula I is provided wherein Rj is silyl which may be substituted, in particular substituted wife alkyl, R is alkyl, and R3, R4, R5,
R6, and R' arc hydrogen.
Is an embodiment, Rl is a piperazinyl or substituted piperazinyl, in particular a piperazinyl substituted with a pyrimidinyl of the Formula H below.
Figure AU2016203312B2_D0007
Thus, fee invention also provides an isolated and pure, in particular, substantially pure, compound of the Formula H wherein one or both of Rw and R;i are independently substituted or tmsubstitnted hydrogen, hydroxyl, alkyl, alkenyl, alkynyl, alkylene, alkenyleae, alkoxy, alkenyloxy, cycloalkyl, cycloalkenyl, aryl, aryloxy, arylalkoxy, aroyl, heteroaryl, heterocyclic, acyl, acyloxy, sulfonyl, sulfinyl, suifenyl, amino, imino, azido, thiol, thioalkyl, thioalkoxy, thioaryl, nitro, ureido, cyano, halo, silyl, silyalkyl, silyloxy, silylthio, ~O, ~5, carboxyl, carbonyl, or carbamoyl, or an isomer or a pharmaceutically acceptable salt thereof.
In an aspect of the compound of the Formula H is provided wherein RiS is hydrogen; hydroxyl; alkyl; aryl [e.g, phenyl which is optionally substituted (e.g., halide)]; piperazinyl which may be substituted (e.g, substituted with a pyrimidinyl); -NR5SR5S wherein R55 is hydrogen or alkyl, and Rss is phenyl which may be substituted or alkyl winch may be substituted (e.g. amino, in particular - CHsCHjNHr CBaCHiNHCOOCCCH^b); morpholinyl which may be substituted; or -SR25 wherein Rz5 is phenyl which may be substituted; and RiS is hydrogen, or aryl (e.g. phenyl) which may be substituted.
In a particular aspect of the invention a compound of fee Formula Π is provided wherein Ri0 is hydrogen, halo, optionally substituted hydroxyl, alkyl, pyridinyl, phenyl, benzyl,
2016203312 20 May 2016 piperazinyl, amino, moroholinyl, or -SR24 wherein R24 is alkyl or aryl· In an embodiment, Rw is “NH[CIl2]mNR*iSRs·' wherein m. is 1 to 6, in particular 2 to 4. Re<i is hydrogen, RM is a carboxyl, in particular ~COOC(CHa)3.
In an aspect of the invention a compound of the Formula H is provided wherein Rn is hydrogen, halo, optionally substituted alkyl, pyridinyl, piperidinyl, morpholinvl, piperazaiyl, or phenyl.
In another aspect of the invention a compound of the Formula Π is provided wherein both of Ri0 and Rn are not hydrogen.
In particular embodiments of the invention one or more of Rie and Rn are alkyl, in particular C3 -C6 alkyl and the other of R*~ and R'1 is hydrogen.
hi particular embodiments of the invention one or more of Rw and RV: are aryl in particular phenyl or benzyl and the other of Rw and Rn is hydrogen.
In particular embodiments of the invention a compound of the Formula II is a compound in Table 3, more particularly hW01-2-065LKM, MW01-2-O69SRM, MW01-2-151SRM,
MWO2-5-188WH, MW01-6427WH, MW01-6-189WH, MW01-7407WH, and derivatives thereof.
The invention also provides an isolated and pure, in particular, substantially pure, compound of the Formula 111 wherein R1S and R16 are independently substituted or unsubstituted hydrogen, hydroxyl, alkyl, alkenyl, alkynyl, alkylene, alkenylene, alkoxy, alkenyloxy, cycloalkyl, cycloalkenyl, aryl, aryloxy, arylalkoxy, aroyl, heteroaryl, heterocyclic, acyl, acyioxy, sulfenyl, sulfinyl, sulfenyl, amino, imino, azido, thiol, thioalkyl, thioalkoxy, thioaryl, nitro, ureido, cyano, halo, silyl, sllyloxy, silylthio, ~O, ™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, compound of the Formula IV wherein R7e and Rn are independently substituted or unsubstituted hydrogen, hydroxyl, alkyl, alkenyl, alkynyl, alkylene, alkenylene, alkoxy, alkenyloxy, cycloalkyl, cycloalkenyl, aryl, aryloxy, arylalkoxy, aroyl, heteroaryl, heterocyclic, acyl, acyioxy, sulfonyl, sulfinyl, sulfenyl, amino, imino, azido, thiol, thioalkyl, thioalkoxy, thioaryl, nitro, ureido, cyano, halo, silyl, sllyloxy, silylthio, ~O. ~S, carboxyl, carbonyl, or carbamoyl, or an isomer nr pharmaceutically acceptable salt thereof.
In an aspect, the invention relates to a compound of the Formula IV wherein R70 is a heteroeylic, In particular a saturated 3 to 6-membered heteramonocylie group containing 1 to 4 nitrogen atoms more particularly, pyrrolidinyl, imidazolidinyl, piperidinyl, and piperazlnyl, especially piperaziny! or piperidinyl, which may be substituted with alkyl especially methyl,
2016203312 20 May 2016 dimethyl, cycloalkyl especially cyclohexyl, aryl especially phenyl, a substituted or u&substituted unsaturated condensed heterocyclic group containing 1 to 5 nitrogen atoms, in particular, indolyl, isoindolyl, indolizinyl, indazolyl, quinazolinyl, pteridinyl, quinolizidinyl, phthalazinyl, naphthyridiuyl, quinoxalinyl, cinnolinyl, phenanthridinyl, acridinyl, phenanthrolnyl, phenazinyl, carbazolyl, putinyl, benzimidazolyl, quinolyl, isoquinolyl, quinolinyl, isoquinolinykor indazolyl, especially benzimidazolyl substituted with oxy.
The invention also relates to a compound of the Formula IV wherein R7S is amino or substituted amino, and optionally R7! is aryl, is particular phenyl. In an aspect R70 is -N-R21 wherein R21 is hydrogen or alkyl, in particular Cj-Cg alkyl, more particularly methyl or dimethyl, or -N-R^R22 wherein R21 is hydrogen or alkyl, in particular CVCy alkyl, more particularly methyl and R'4i is alkyl substituted with amino or substituted amino, heterocyclic, substituted hcterocylic, or cycloalkyl. In an embodiment, R78 is ~-N~R21R22 wherein R25 is hydrogen or alkyl, in particular CrC« alkyl, more particularly methyl and R22 is Cj-Cg alkyl, especially methyl or ethyl substituted with a cycloalkyl especially cyclopropyl, a heterocyclic especially piperidinyl, pyrrolidinyl, or morphollnyl 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, 130,13.1, .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, compound of the Formula V wherein Ry and R5i are independently substituted or unsubstituted hydrogen, hydroxyl, alkyl, alkenyl, alkynyl, alkylene, alkenylene, alkoxy, allenyloxy, cycloalkyl, eycloalkenyl, aryl, aryloxy, arylalkoxy, aroyl, heteroaryl, heterocyclic, acyl, acyloxy, sulfonyl, sulfinyl, sulfenyl, amino, imino, azido, thiol, thioalkyl, thioalkoxy, thioaryi, nitro, ureido, cyano, halo, silyl, silyloxy, sllyithio, ~«Q, ™S, carboxyl, carbonyl, or carbamoyl, or an isomer or pharmaceutically acceptable salt thereof.
The Invention relates io a compound of the Formula V wherein R5f! is substituted or unsnbstituted hydrogen, alky], aryl, or heterocyclic; R53 is substituted or unsnbstitated hydrogen or alkyl, and R,52 is substituted or unsubstituted hydrogen, alkyl, cycloalkyl, heteroaryl or halo. In as aspect, fee invention relates to a compound of the Formula V wherein R58 is hydrogen, Cj30 Cg alkyl which may be substituted with alkyl, especially methyl, or tthnethyl, phenyl, or a 3 to 6~ merobered heteromonocylic group containing 1 to 4 nitrogen atoms more particularly, piperidinyl or morphollnyl, RS1 is hydrogen or alkyl especially methyl, and R52 is hydrogen, alkyl especially methyl, dimethyl, ethyl, or propyl, cyclohexyl, chloro, or an unsaturated 5 to 6 membered heteromonoeyclyl group containing I to 4 nitrogen atoms, in particular, pyrrolyi,
2016203312 20 May 2016 pyrrolinyl, iraidazolyl, pyrazolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, pyridinyl, pyrimidinyl, pyraztnyl, or pyridazinyl, especially pyridinyl. In an embodiment, R5C is aryl, RSI is hydrogen, andR52 is C^-Cg 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 ihe Formula I, K, SI, IV, or V 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, Π, 553, IV, or V may be in the form of a prodrag that is converted in vivo to an active compound. For example, in a compound of the Formula I one or more of R\ Rs, R3’ R4, R5, R°, and R7 may comprise a cleavable group that is cleaved after 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-enzyroalically,
A compound of the Formula I, H, HI, 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 indirectly covalently attached to the compound. A carrier may be substituted with substituents described heroin including without limitation one or more alkyl, amino, nitro, halogen, thiol, thioalkyi, sulfate, sulfonyl, sulfinyl, sulfoxide, hydroxyl groups, hr 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 molecule that targets a compound of tire Formula I, II, III, IV, or V to a particular tissue or organ. Thus, a carrier may facilitate or enhance transport of a compound of the Formula I. Π, ΪΠ, IV or V to the brain.
PROCESS
Compounds of the Formula I, II, III, IV, or V can be prepared using reactions and 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 die functionality present on the compounds should be consistent with, the proposed reaction
2016203312 20 May 2016 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 he recognised teat another major consideration in the development of a synthetic route is the selection of the protecting group used for protection of the reactive 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 to preparing compounds or the invention are either available mom commercial suppliers or are prepared by methods well known to a person of ordinary skill in the ad., following procedures described in such references as Fieser and Fieser’s Rengente /or Organic Sw/rerir, vols, 1-17, John Wiley and Sons, New York, N.Y.,
Eodd’k CAemfrrty of Carbon Compounds, vols. 1-5 and supps., Elsevier Science Publishers, 1989; Organic Reactions, vols, 1-40, John Whey and Sons, New York, N.Y., 1991; March J.: Advanced Organic Chemistry, 4th ed., John Wiley and Sons, New York, N.Y.; and
Larock: Conmrehenrivc Orgnnfo Transformations, VCH Publishers, New York. 1989.
The starting materials, intermediates, and compounds of the Formula I, II, III, 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 1, H, Hl, IV, or V may be characterized using conventional methods, including physical constants and spectroscopic methods, in particular HPLC.
The compounds of the Formula I, Π, ΙΠ, 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, H, HI, IV, or V from tee reaction mixture as a pharmaceutically unacceptable salt and then convert the latter to the free base compound by treatment with an alkaline reagent and subsequently convert the free base to a pharmaceutically acceptable acid addition salt. The add addition salts of the base compounds of the Formula I, Π, ΙΠ, IV, or V are readily prepared by treating tee 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
Ό desired solid sal t is obtained.
Compounds of tee Formula I, If, ΙΠ, 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 testing tee corresponding acidic compounds with an aqueous solution containing the desired pharmacologically acceptable cations and. then
2016203312 20 May 2016 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 toe 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 employed to ensure- completeness of reaction and maximum product yields.
In particular aspects, the present invention provides methods of making toe compounds disclosed herein, comprising the steps provided (See, e.g., toe 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 toe formula. Π wherein Rli is hydrogen aud R10 is an unsaturated 5 to 6 membered hetercanonocyclyl group containing 1 to 4 nitrogen atoms, in particular, pyrrolyl, pyrrolinyl, itmdazolyl, pyrazolyl, 2pyridyl, 3-pyridyl, 4-pyridyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazolyl, or tetrazolyl, more particularly pyridinyl, which comprises reacting a compound of the formula Π wherein R10 is halo, in particular chloro, and Rll is hydrogen, with boronic acid substituted with an unsaturated 5 to 6 membered heteromonocyclyl group containing 1 to 4 nitrogen atoms, in particular, pyrrolyl, pyrrolinyl, imidazolyf, 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 toe formula II wherein Rll is hydrogen and R10 is an unsaturated 5 to 6 membered heteromonoeyclyl group containing I to 4 nitrogen atoms, in particular, pyrrolyl, pyrrolinyl, imidazolyl, pyrazolyl, 2-pyndyJ, 3-pyridyi, 4-pyridyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridnzinyi, triazolyl, or tetrazolyl, more particularly pyridinyl. In an embodiment, R10 is phenyl substituted with halo.
fa another aspect, tire invention provides a precess for preparing a compound of toe formula II wherein Rll is hydrogen and R10 is a substituted aryl which comprises reacting a compound of the formula II wherein R10 is halo, in particular chloro, anti Ri 1 is hydrogen, with a substituted aryl boronic acid under suitable conditions to prepare a compound of the formula H wherein RU is hydrogen and R10 is a substituted aryl, hr an embodiment, R10 is phenyl substituted with halo.
In another aspect, toe invention provides a process for preparing a compound of the :0 formula Π wherein R10 is hydrogen and Rl1 is alkyl which comprises reacting a compound of tire formula Π wherein Rll is halo, in particular chloro, and R10 is hydrogen, with an alkyl boronic acid under suitable conditions to prepare a compound of the formula II wherein RIO is hydrogen and RU is alkyl, In an embodiment, Rll. is lower alkyl, in particular methyl or ethyl, and a
2016203312 20 May 2016 compound of the formula Π wherein R11 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 H wherein RIO is hydrogen and Rll is an alkyl which comprises reacting a pyrfdazme . 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)pyriutidine under suitable conditions to prepare a compound of the formula H wherein RIO is hydrogen and R.1.1 is an alkyl. In an embodiment, RU is methyl or ethyl.
In another aspect, the invention provides a process for preparing a compound of fee formula If wherein £10 is hydrogen and. R11 is aryl which comprises reacting a compound of the formula II wherein R10 is hydrogen and Rll 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-(pipe,ridin-4-yloxy)pyrimidine under suitable conditions to prepare a compound of the formula H wherein R10 is hydrogen and Rll is aryl. In an embodiment, Rll is phenyl.
In another aspect, the invention provides a process for preparing a compound of fee formula R wherein R10 is hydrogen and R11 is an unsaturated 5 to 6 membered heteromonocyclyi group containing 1 to 4 nitrogen atoms, in particular, pyrrolyl, pyrrolinyl, rraidazolyl. pyrazolyl, 2pyridyk 3-pyridyl, 4-pyridyl, pyridlnyk pyrimidinyl, pyrazinyk pyridaanyl, triazolyl, or tetrazolyl, more particularly pyridinyi which comprises reacting a compound of fee formula U wherein Rll is halo. In particular chloro, and R10 is hydrogen, with a boronic acid substituted with an unsatnrated 5 to 6 membered heteromonocyclyi group containing 1 to 4 nitrogen atoms, in particular, pyrrolyl, pyrrollnyl, iroidazolyl, pyrazolyl, 2-pyridyi, 3-pyridyl, 4-pyridyl, pyridinyi, pyriroidinyk pyrazinyk pyridarinyl, triazolyl, or tefmzolyk more particularly pyridinyi, under suitable conditions to prepare a compound of the formula H wherein R10 is hydrogen and Rll is
'.5 an onsaturated 5 to 6 membered heteromonocyclyi group containing 1 io 4 nitrogen atoms, in particular, pyrrolyl, pyrrolinyk iroidazolyl, pyrazolyl, 2-pyridyi, 3-pyridyl, 4-pyridyl, pyridinyi, pyrimidinyl, pyrazinyk pyridazinyh triazolyl, or tetrazolyl, more particularly pyridinyi.
In an embodiment, tire invention provides a process for preparing a compound of the formula II wherein RIO is hydrogen and Rll Is pyridinyi which comprises reacting a compound of the formula II wherein R11 is halo, in particular chloro, and R10 is hydrogen, with a pyridinyi boronic acid under suitable conditions to prepare a compound of tire formula ΙΪ wherein R10 is hydrogen and R11 is pyridinyi, hi another aspect, the invention provides a process for preparing a compound of the formula ΙΊ wherein RIO is hydrogen and Rll is an unsatumted 5 to 6 membered heteromonocyclyi
2016203312 20 May 2016 group containing 1 to 4 nitrogen atoms, in particular, pyrrolyl, pyrrolinyl, imidazolyi, pyrazolyl, 2pyridyl, 3-pyridyl, 4-pyridyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazolyl, or tetrazolyl, more particularly pyridinyl which comprises reacting a pyridazme 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, imidazolyi, pyrazolyl, 2pyridyl, 3-pyridyl, 4-pyridyi, pyridinyl, pyrumdinyi, pyrazinyl, pyridazinyl, triazolyl, or tetrazolyl, more particularly pyridinyl, and at the 6 position with phenyl, with 2-(pi.peridm~4~ yloxyjpyritnidine under suitable conditions to prepare a compound of the formula II 'wherein RIO is hydrogen and Rll is an unsaturated 5 to 6 membered heteromonocyclyl group containing 1 to 4 nitrogen atoms, in particular, pyrrolyl, pyrrolinyl, imidazolyi, pyrazolyl, 2-pyridyl, 3-pyridyl, 4pyridyl, 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 II wherein RIO is hydrogen and Rll is pyridinyl which comprises reacting a pyridazine substituted at tbs C3 position with halo, at the C4 position with pyridinyl, and at the 6 position with phenyl, with 2-(piperidin-4-yIoxy)pyrimidine under suitable conditions to prepare a compound of tire formula 31 wherein R 1.0 is hydrogen and R11 is pyridinyl.
In another aspect, the invention provides a process for preparing a compound of the formula II wherein RIO is hydrogen and Rll is piperidinyl or substituted pipsridinyl which comprises reacting a compound of the formula Ii wherein Rll is halo, in particular chloro, and RIO is hydrogen with piperazinyl or substituted piperazinyl under suitable conditions to prepare a compound of the formula Π wherein R10 is hydrogen and Rll is pipsridinyl or substituted piperidinyl.
In another aspect, the invention provides a process for preparing a compound of the formula I wherein R1 is pipsrazinyl or pipsrazinyl substituted with alkyl, aryl, or cycioalkyl, R2 is aryl, R3, R4, R5 and Rd 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 cycioalkyl, under suitable- conditions to prepare a compound of the formula I wherein R1 is piperazinyl or piperazinyl substituted with alkyl, aryl, or cycioalkyl, R2 Is aryl, S3, R4, S3 and R6 are hydrogen and R7 is absent.
In another aspect, the invention provides & process for preparing a compound of the formula I wherein R1 is piperazinyl or piperazinyl substituted with alkyl, aryl, or cycioalkyl, R2 is an unsaturated 5 to 6 membered heteromonocyclyl group containing 1 to 4 nitrogen atoms, in particular, pyrrolyl, pyrrolinyl, imidazolyi, pyrazolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, pyridinyl,
2016203312 20 May 2016 pyrimidinyi, pyrazinyl, pyridazinyl, triazolyl, or tetrazolyl, more· particularly pyridinyi, R3, R.4, R5 and R6 are hydrogen and R7 is absent, which comprises reacting a pyridaziue substituted at the C3 position with halo, at the C4 position with an unsataated 5 to 6 membered heieromonocyelyl group containing 1 to 4 nitrogen atoms, in particular, pyrrolyl, pyirolinyl, intidazolyh pyrazolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, pyridinyi, pyrimidinyi, pyrazinyl, pyridazinyl, triazolyl, or tetrazolyl, more particularly pyridinyi. wife piperazinyl or piperazinyl substituted wife alkyl, aryl, or cycloalkyl under suitable conditions to prepare a compound of the fonnulal wherein R1 is piperazinyl or piperazinyl substituted wife alkyl, aryl, or cycloalkyl, R2 is an unsaturated 5 to 6 membered heieromonocyelyl group containing 1 to 4 nitrogen atoms, in particular, pyirolyi, pyrrolinyl, imidazolyl, pyrazolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, pyridinyi, pyrimidinyi, pyrazinyl, pyridazinyl, triazolyl, or tetrazolyl, more particularly pyridinyi, R3, R4, R5 andRti 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 substituted ainino in particular amino substituted with substituted morpholinyl, in particular morpholinoethyi, R2 is aryl or an unsaturated 5 to 6 membered heteromouocydy] group containing 1 to 4 n?tiofen atoms, In particular, pyrrolyl, pyrrolinyl, imidazolyl, pyrazolyl, 2-pyridyl, 3-pyridyl, 4-pyridyi, pyridinyi, pyrimidinyi, pyrazinyl, pyridazinyl, triazolyl, or tetrazolyl, in particular pyridinyi, R3, R4, R5 and R6 are hydrogen and R7 Is absent, which comprises reacting a pyridszine substituted at the C3 position with halo, at the
C4 position wife aryl or an unsaturated 5 to δ membered heteromonocyclyl group containing 1 to 4 nitrogen atoms, in particular, pyrrolyl, pyrrolinyl, imidazolyl, pyrazolyl, 2-pyridyl, 3-pyridyl, 4pyridyl, pyridinyi, pyrimidinyi, pyrazinyl, pyridazinyl, triazolyl, or tetrazolyl, more particularly pyridinyi, with substituted amino in particular amino substituted with substituted morpholinyl, is particular morpholinoethyi, under suitable conditions to prepare a compound of the formula I wherein R.1 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, R5.1 is hydrogen, and R52 is alkyl comprising reacting a pyridszine substituted at position C3 wife halo, at position €4 with aryl and position 6 with alkyl wife. l--(2-pyrimidyi)pipsrazine under suitable conditions to prepare a compound of fee 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 fee formula I wherein R1 is substituted amino, 112 is an unsaturated 5 to 6 membered heteromonocyclyl group containing .1 to 4 nitrogen atoms, in particular, pyrrolyl, pyrrolinyl,
2016203312 20 May 2016 imidazolyl, pwazoiyL 2-pyridyl, 3-pyridyl, 4-pyridyl, pyridinyl, pyrirnidinyk 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 tire C4 position with an unsaturated 5 to 6 membered heteromonocyclyl group containing 1 to 4 nitrogen atoms, in particular, pyrrolyl, pyrrolinyl, hnidazolyi, pyrazoiyl, 2-pyridyb 3-pyridyl, 4-pyridyl, pyridinyl, pyrirnidinyk pyrazinyl, pyridazinyl, triazolyl, or tetrazolyl, in particolar 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, pyoroiyl, pyrrolinyl, imidazolyl pyrazoiyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, pyridinyl, pyrirnidinyk 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 Π, a key precursor (See, e.g., Figure 1) that may be- utilized was obtained commercially and used directly for the synthesis of the illustrated compound MW01-3-183WH 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. hr Figure 1, a synthetic scheme is shown, for synthesis of .MWO1-3183WH with unconstrained aromatic ring at position 6 and no modification at position. 5. Reagent and condition: (a) i-BuOH, NfhCI, 2-(piperazm-!-yI) pyrimidine.
Thus, in an aspect, Coe invention provides a method for preparing a compound of die Formula
Π wherein a substituted ό-phenylpyridazme is reacted with 2-(pipefarin-lyl)pyridmidine to produce a compound of the Formula II wherein RKi and. Ru are hydrogen. A compound of the formula. II wherein. R10 and Ri 1 are hydrogen can be reacted under suitable conditions and with suitable reagents to introduce fee radicals R10 and Ru which are independently hydrogen, hydroxyl, alkyl, alkenyl, alkynyk alkylene, alfcenylene, alkoxy, alkenyioxy, cycloalkyl, cycloalkenyl, aryl, aryloxy, arylalkoxy. aroyl, heteroaryl, heterocyclic, acyl, acyloxy, sulfonyl, sulfinyl, sulfenyk amino, imino, azido, thiol, thioalkyl, thioalkoxy, thioaryl, nitro, ureido, cyano, halo, silyl, silyloxy, siiylalkyl, silyl thio, =0, ~S, carboxyl, carbonyl, carbamoyl, or caihoxamide
Therapeutic efficacy and toxicity of compounds, compositions and methods of the invention may be determined by standard pharmaceutical procedures is cell cultures or with experimental animals such as by calculating a statistical parameter such as the ED^q (the dose that is therapeutically effective in 50% of the population) or LEh;· (the dose lethal to 50% of the population) statistics. The therapeutic index is the dose ratio of therapeutic to toxic effects and it can he expressed as the FIWLD50 ratio. Pharmaceutical compositions which exhibit large
2016203312 20 May 2016 therapeutic indices are preferred. By way of example, one or more of fee therapeutic effects, in particular beneficial effects disclosed herein, can be demonstrated in a subject or disease model, for example, a TgCRNDS mouse with symptoms of Alzheimer’s disease.
Biological investigations were done wife compounds disclosed herein feat were >95% homogenous as determined by HPLOMS analysis. As part of a hierarchy, cell-based screening protocol, fee compounds were screened for their ability to block IL-Ιβ and TNFct production by B V-2 mouse microglial cells stimulated with EPS. The data for MW01-3483WH is shown in Figure 2. Derivative groups that may be used to modify the compounds of the present invention can be found in US, Patent Application No. 20030176437 (heroin incorporated by reference in its entirety for ail purposes).
The compounds disclosed herein can be tested for hvsr toxicity which is an important initial consideration for orally administered compounds since the liver is tire 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 disclosed herein may also be tested for cardiac safety by testing for hERG channel inhibition, for example using fee method illustrated in Example 3.
COMPOSITIONS AND KITS
A compound of the Formula ί, Π, HI, IV, or V of the invention may be formulated into a pharmaceutical composition for administration to a subject. Pharmaceutical compositions of the present invention or fractions thereof comprise suitable phaKnaceutically acceptable carriers, excipients, and vehicles selected based on the intended form of administration, and consistent with conventional pharmaceutical practices. Suitable pharmaeeutical carriers, excipients, and vehicles are described in fee standard text, Remington’s Pharmaceutical Sciences, Mack Publishing Company (Remington's Pharmaceutical Sciences, Mack Publishing Company,
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 wife 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 wife any oral, non-toxic, 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
2016203312 20 May 2016 agents may also be combined in the compositions c-r 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 suppository, wife traditional binders and carriers such as triglycerides. Oral formulations can in» hide standard earners 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, mierocapsuies, and the like,
Formulations for parenteral attain istration may include- aqueous solutions, syrups, aqueous or oil suspensions aud emulsions with edible oil such as cottonseed oil, coconut oil or peanut oik Dispersing or suspending agents that can be used for aqueous suspensions include synthetic or natural gums, such as tragacanth, alginate, acacia, dextran, sodium carboxvmethyicellnlose, gelatin, methylceliulose» and polyvinylpyrrolidone.
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, buffets, or preservatives, e.g. antioxidants such as mcthylhydroxybenzoate or ti) similar additives.
Compositions of the Invention can be formulated as pharmaceutically acceptable salts as described herein.
A composition of the invention may be sterilized bv, 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 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
2016203312 20 May 2016 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 bv the agency of manufacture, use, or sale for human administration.
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
The invention contemplates the use of compounds of the Formula I, 1Ϊ, ΠΙ, 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 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 basis for diseases affecting all organ systems including, but not limited to, asthma, acute inflammatory diseases, vascular inflammatory disease, chronic inflammation, afoerosderosis, angiopathy, myocarditis, nephritis, Crohn’s disease, arthritis, type I and S diabetes aad associated vascular pathologies. The incidence of these inflammatory conditions is on the rise aid the expense is large. For example, for just one form of inflammatory disease, Alzheimer's disease, the direct costs (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 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
2016203312 20 May 2016 associated protein kinase activity, and/or proinfiammatory 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 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 neurodegenerative diseases, including Alzheimer's disease (AD), and traumatic brain injury, and stroke. Selected compounds and compositions of the present invention may he used, to reduce or inhibit Αβ-induced neuronal ceil death and in particular to reduce or inhibit calmodulin regulated protein .kineses, such as death associated protein kinase (DARK).
In some embodiments, the present invention provides methods of inhibiting cell signaling molecule production (e.g., IL-Ιβ and INFo), comprising administering compositions comprising one or more of the compounds of the Formula I, H, III, IV, 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
Tables for use in 1) lowering amounts of prourTunumatary cytokines (e.g., TNFa or IL~lp) and/or 2) maintaining normal levels of postsynaptic proteins (e.g., PSD-95), fc some embodiments, the reduction of pm-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 tire compounds reduces aberrantly high pto-inflammator/ cytokine production caused by the disease (e.g,,
Αβ- induced increase in pro-inflammatory cytokines).
fir another aspect, selected compounds and compositions of the invention may be used to modulate cytokine-mediated neuronal cell death, in particular modulate cytoHnednduced generation of NO. TNFa signaling through the Fas/TNFR family of death receptors, and/or
DAPK, in Alzheimer’s disease and other neurodegencrative disorders, and brain injury, and stroke. The evidence for tire involvement of pro-inflammatory cytokines and NO in neuronal cell death has been reviewed in Akiyama, IL, et al„, (2000) NeurobioL Agir g 21, 383-421;
2016203312 20 May 2016
L5 »0 :5
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 cel] activation in addition to brain glia and neurons. For example, ceil activation and tissue damage is a characteristic of other diseases such as acute lung injury (All). AU duo to sepsis, trauma or mechanical ventilation is associated with high mortality and morbidity, yet there are few effective therapies for the treatment of ALL AU is common during sepsis, which Itself has as annual mortality equal to acute myocardial infarction. Endothelial cell (EC) dysfunction and activation has been implicated in the m w responses linked to ALI, and EC protein kineses, such as myosin light chain kinase (MLCK), have beer: 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 ceil 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. Accordingly, compounds of the Formula 1, ΪΪ, IE, IV, or V cart 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 administering to the subject a therapeutically effective amount of a compound of the Formula I, II, DI, 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 neuroilanunatiou. comprising administering to the subject a therapeutically effective amount of a composition comprising a compound of the Formula I, Π, ill, IV, or V and a pharmaceutically 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,11, IE, IV, or V, or a pharmaceutically acceptable salt thereof, or a composition comprising a compound of the Formula I, Π. ΙΠ, IV, or Vf and a pharmaceutically acceptable carrier, excipient, or vehicle which inhibit or reduce nsuroflammatiors, activation of glia, pmimflammatory 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 neuroinflammatiou that can be decreased or inhibited with a compound disclosed herein comprising administering to the- subject a therapeutically effective amount of a
2016203312 20 May 2016 compound of the Formula I, JI, ΪΙΪ, IV, or V, a pharmaceutically acceptable salt thereof, or a composition comprising a compound of the Formula ϊ, Π, ΠΙ, IV, or V and a pharmaceutically acceptable carrier, excipient, or vehicle.
In another aspect, the invention provides a method for preventing or inhibiting activation of protein kinases, in particular DAPK, in a subject conrprising administering a therapeutically effective amount of a compound of the Formula I, K, Hl, IV, or V a pharmaceutically acceptable salt thereof, or a composition comprising a compound of the Formula ϊ, II, ΙΠ, IV, ox V and a pharmaceutically acceptable carrier, excipient, or vehicle.
In a further aspect, the invention provides 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 therapeutically effective amount of a compound of the Formula I, Π, ΠΙ, IV, or V a pharmaceutically acceptable salt thereof, or a composition comprising a compound, of the Formula I, Π, Πϊ, IV, or V and a pharmaceutically acceptable carrier, excipient, or vehicle.
In some embodiments, the invention provides methods of inhibiting cell, signaling molecule production (e.g., IL-Ιβ and TNFa), comprising administering compositions comprising one or more compounds of the Formula ϊ, ϊϊ, Πϊ, 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 fee Figures and
Tables, more particularly Table 2, 3, 4 or 5 or derivatives of these compounds, are co-administered with oilier recognized therapeutics to treat inflammatory disease (e.g., neuroinflammatory disease, in particular zAIzheiirter's disease), hi 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-Ιβ) and/or 2) maintaining normal levels of postsynaptic proteins (e.g.,
PSD-95) for research, drag screening, or therapeutic purposes. In some embodiments, fee reduction of pro-infiammatcay 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 feat treatment with the compounds reduces aberrantly high pro-inflammatory cytokine production caused by fee disease (e.g., Αβ-induced increase in pro-iafiamnsatory cytokines),
In an aspect, fee invention provides a method for amsiioriating progression of a disease or obtaining a less severe stags 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 feerapeutically effective amount of a
2016203312 20 May 2016 compound of the Formula I, II, III, IV, or V, a pharmaceutically acceptable salt thereof, or a composition comprising a compound of tire Formula I, R, 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.
neuroinflammatory disease, in particular a neurodegenerative disease, more particularly Alzheimer’s disease) comprising administering a therapsutically effective amount of a compound of the Formula I, Π, 01, IV, or V, a pharmaceutically acceptable salt thereof, or a composition comprising a compound of tire Formula Ϊ, H, HI, IV, or V and a pharmaceutically acceptable carrier, excipient, or vehicle.
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, Jl, III, IV, or V, a pharmaceutically acceptable salt thereof, or a composition comprising a compound of the Formula I, Π, ΠΙ, IV, or V and a pharmaceutically 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, II, HI, 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 the Formula I, H, HI, IV, or V. Such treatment may be effective for retarding tire degenerative effects of Alzheimer’s disease, including specifically, but not exclusively, neuromflanunation, 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 providing a composition comprising a compound of the- invention in an amount sufficient to reverse or inhibit neuroinfiammatlon, activation of signaling pathways involved in inflammation (e.g., neuroinflammation), cell signaling molecule production, activation of glia or glial activation pathways and responses, proinfiammatory cytokines or ehemokines (e.g., interleukin (IL) or tumor necrosis factor (TNF), oxidative st.ress-relat.eii 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 damage, and/or neuronal cell dead's for a prolonged period following administration.
2016203312 20 May 2016
In afordhsr 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 fee invention in a dose sufficient to reverse or inhibit neuroinflammation, activation of signaling pathways Involved in inflammation (e.g., neuroinfianwaation), cell signaling molecule production, activation of glia or glial activation pathways and responses, proinflammatay cytokines or chemokincs (e.g., interleukin (IL) or tumor necrosis factor (IKE), 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 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 &d=feu Is nation, thereby treating fee Alzheimer’s disease.
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, Π, ΙΠ, IV, or V, a pharmaceutically acceptable salt thereof, or a composition comprising a compound of the Formula I, U, Ill, IV, or V and a pharmaceutically acceptable carrier, excipient, or vehicle, which upon administration to a subject with symptoms of a neurodegenerative disease, in 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 affect is evidenced by a decrease or inhibition of one or more of the following: inflammation (e.g. nenroinfiarnmation), activation of signaling pathways involved in inflammation (e.g,, neutOlnflammation), cell signaling molecule production, activation of glia or glial activation
IS pathways and responses, proinfiammatory cytokines or chemokincs (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).
H) 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 mom, more- particularly 7 or more of the following:
2016203312 20 May 2016
a) A reduction m protein. kinase activity (e.g. DAPK), in particular at least about a 0.05%, 0.1%, 05%, 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%,
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 m glial activity in the brain, relative to the levels determined in the absence of a compound of the Formula I, H, ΪΗ, 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%,
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 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,
Ϊ0 f) A reduction in proinfiammatory 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-lB and/or tumor necrosis factors production 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 interleukin-ΐβ and/or humor necrosis factora 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 t0 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
2016203312 20 May 2016 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,
ΙΕί, IV, or V, a pharmaceutically acceptable salt thereof, or a composition comprising a compound of the Formula I, Π, III, IV, or V and a pharmaceutically acceptable carrier, excipient, or vehicle.
In an embodiment, the invention provides a method of reversing or inhibiting uemoinflannnation, activation of signaling pathways involved in inflammation (e.g., neuroin.fiamination), 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 fee complement 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 comprising administering to the subject a therapeutically effective amount of a compound of the Formula I, Π, ΙΠ, IV, or V, a pharmaceutically acceptable salt thereof, or a composition comprising a compound of the Formula I, II, ill, IV, or V and a pharmaceutically acceptable carrier, excipient, or vehicle.
2Q 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, Π, III, TV, or V, or a pharmaceutically acceptable salt thereof, or a composition comprising a compound of the Formula I, H, HI, IV, or V and a pharmaceutically acceptable carrier, excipient, or vehicle.
The invention provides a method of improving memory of a healthy subject or fee memory of a subject wife age impaired memory by administering an effective amount of a compound of fee Formula I, Π, III. IV, or V. or a pharmaceutically acceptable salt thereof, or a composition comprising a compound of the Formula I, II, ΪΠ, IV, or V and a pharmaceutically acceptable carrier, excipient, or vehicle.
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, H, HI, IV, or V, or a pharmaceutically acceptable salt thereof, or a composition comprising a compound of the Formula I, H, ΠΙ, IV, or V and a pharmaceutically acceptable carrier, excipient, or vehicle.
2016203312 20 May 2016 & 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 foe mammal an effective memory-improving amount of a compound of the Formula I, H, HL IV, or V, a pharmaceutically acceptable salt thereof, or a composition comprising a compound of die Formula I, Π, 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, H, Hl, IV, or V, a pharmaceutically acceptable salt thereof, or a composition comprising a compound of foe Formula I, Ii, HI, 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 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, separately, or sequentially with other therapeutic agents or therapies.Therefore, the compounds of the Formula I, ΪΪ, HI, IV, and/or V may be co-admirtistered with one or more additional therapeutic agents including without limitation beta-secretas© inhibitors, alpha--secretase inhibitors, .and eprilon-secretas© inhibitors, agents that are used for the treatment of complications resulting from or associated with a disease, or general medications that treat or prevent side effects.
The invention also contemplates the use of a composition comprising at least one compound of foe 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 foe invention for preparation of a medicament for providing 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 foe invention for foe preparation of a medicament for prolonged or sustained treatment of a disease,
2016203312 20 May 2016 ϋ3
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 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 fee invention can be formulated for sustained release, for delivery locally or systemicallv. 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 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 fonnulations), pills, powders, granules, elixirs, tinctures, suspensions, syrups, and emulsions. They may also be administered
L5 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 intonasal 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 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, 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 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, fee frequency of treatment, the route of administration, the renal and hepatic function of the patient, and the desired effect.
2016203312 20 May 2016
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 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 onecompound 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 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 he treated with a compound of the Formula ϊ, Π, 111, IV, or V or composition or formulation thereof on substantially any desired schedule. A composition of the 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 weeks, 2 weeks to 12 weeks, 2- weeks io 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 he described in greater detail by way of specific examples. The following examples are offered for illustrative purposes, and are not intended to limit the invention in any manner,
EXAMPLES
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 ehlorepyridaxine, 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. Trie solvent was removed under·
2016203312 20 May 2016 reduced pressure. The residue was then extracted with ethyl acetate, washed with water and brine, and dried ever anhydrous NaiSO-i. Removal of solvent was followed by reay'staiiization from 95% ethanol
Amination of 3-chloio-6-phenylpyridarfne by 2-(pipcrazm~l- yi)pyrimidine (Figure 12) easily fed to 2“(4”(frpheuylpyridarin3“yi)pipernrin-l-yi)pyrisudme (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, 1=4.5, 2H), 8.011(d, 1=7.5,11 2H), 7.692 (d, 3=9,5,1¾ 7.468 (t, 5 .1=6.0,2H), 7.417 (d, 1=7.5,1H), 7.047 (d, 1=9.5,1H), 6-546 (t, 3=4.5,1H), 4.013 (t, 3=5.0,4H), 3.826 (U=5.0,4B).
.0 Reactions were monitored by analytical liFLC (Rairnn Instruments System. Woburn, MA), done on a reverse phase column CIS (25 cm x 4.6 nan, 5 an, Supelco, Bellefonte, PA) with two different TJV wavelengths (X=260 nm and X=22Q nm or 300 nm)- Eluents were (A): 0,1% (v/v) TFA in water and (B) 80% (v/v) acetonittfoe/water containing 0.08% TFA. A linear gradient of 100/0 to 0/100 A/B over 34 min at hnL/min was used. 1H-NMR spectra were obtained using Vatian INOVA (500 MHz) spectiomcter. 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 acti vating stimulus Epopolysaccharide (LPS, from Salmemtta typkimurimt; 100 ng/ml final concentration) in the presence or absence of amxnopyridazice compounds, as described previously [4,17], Et-1/3 and TNFa levels in cell lysates were determined by electee-chemiluminescent detection in a Meso-Scsle Discovery (M.SD) Idt, as per the manufacturer's instructions.
fs Wra Assays, Αβ-42 infusions and treatment of C57B1/6 mice with MTV01-2--15IWH were performed as previously described [5], Briefly, oligomeric A/31-42 was infused ICV for 28 days with a micro-osmotic pump. At post-operative day 21 and continuing for 14 days thereafter, mice were injected intraperitoneatiy 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 gtial fibrillary acidic protein (GFAP) and F4/80 staining, measurement of the levels of the pro-inflammatory cytokines IL-1]S, TNFa, and SiOOB by ELISA, and determination of synaptic damage by analysis of the levels of postsynaptic density protein-95 (PSD-95).
Brata Uptake Assays, MW01-2-15IWH was administered to mice (25~30g) by oral gavage using 2.5 mg/kg compound In Q.5% carboxymethyleedluiose vehicle. At various times (0-60 min) after
2016203312 20 May 2016 administration, mice were sacrificed, Wood removed by cardiac puncture, and brains iromediatdy harvested, weighed, quick-frozen in liquid nitrogen, and stored at -SOT until assayed. Stain tissue was homogenized in 1 .5 tai of 0.1 M perchloric acid. After centrifugation (12,000 x g for 10 min), foe supernatant was neutralized with 1 M NaOH and then extracted tee times with 2 ml of dichloromcthane 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% H2O), and 20 gl of the reconstituted material was injected into the HPLC system. The HPLC system for detection of MWO1-2-151WH was a Luna 5 /tmCTS, 250 mm x 2 ram internal diameter column together with a guard column (Phenomeaex, Torrance, CA, USA), with HPLC solvent delivered at 0.2 ml/min (Dionsx, 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 foe compound to brain tissue from untreated mice, then extracting the brains and performing HPLC analysis as described above. The area under foe curve increased linearly with the concentration of foe 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 mg/kg) in 0.5% carboxyrnethylcelluiose was administered by oral gavage once daily for 3 days. On the 4th day, mice were anesthetized with peatobaibital, intubated and tire lung» were Inflated with. an airccatiainiug 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 pafeologist blinded to the treatment groups performed microscopic assessment of the tissue for injury.
Results
MW01-3-183WH suppressed both IL-Ιβ and TNFa production in a concentration dependent manner (Figure 2). As shown in Figure 2 A and B, concentration dependent inhibition of proinfiammatory cytokine production by MWO1-3-183WH in BV-2 cells were treated with LPS (100 ng/ml) in the absence or presence of increasing concentrations of MW01-3-183WH for 16 hrs with levels of IL-Ιβ and TNFa in cell lysates measured by the Mesa-Scale Discovery elecrfoehemilumineseeotdetection assay (See, Example L, Materials and Methods). Data are the mean +/- SEM of triplicate determinations.
2016203312 20 May 2016
Figure 3 illustrates that MWG1-5-1SSWH is a concentration-dependent and selective inhibitor of preinfLauanatory cytokine production by activated glia and docs not cause liver injury after chronic oral adminlshation. As shown in Figure 3, concentration-dependent inhibition by MW01-5-18SWH of EPS-induced Increases in (Α)ΪΕ-Ιβ and (B) TNFa levels by 5 the BV2 microglial cell fine. (C) Accumulation, of the NO metabolite, nitrite, was not inhibited at concentrations up to 33 pM. MW01-5-188WH (188) does not suppress iNOS, COX-2 or apoE production in activated glia, as evidenced by the representative western blots for (D) iNOS, (B) 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 μΜ MW01-5-I88WH. Daily oral 10 administration of MW01-5-188WH does not bring about liver injury (G). Mice were administered either diluent or MW01-5-188WH (2.5 mg/kg/day) for 2 weeks, then liver sections were stained with haematoxylin and eosin. Bar = 125 pm. Figure 4 illustrates that oral administration of MW01-5-188WIi sundresses human AB-induced neurointlammation in mouse hippocampus in toe absence of a detectable effect on the number of nitrotyroshie-labeled 15 neurons, or on amyloid plaque deposition. Oral administration of MW01-5-188WH suppreses human Αβ-induced neuroinflammation in mouse hippocampus in the absence of a delectable effect on the number of nitrotyresine-labeled neurons, or on amyloid plaque schematic of toe experimental paradigm is shown. Daily oral
MWO1-5-188WH (188-treated) for 2 weeks .results in significant suppression of the human Αβ ti) induced increase in (A) Π,-Ίβ, (B) TNFa and (C) SWOB levels in hippocampal extracts (n~ 10 mice/group). MW01-5-188WH treatment also decreased the number of (D) GFAP-positive activated astrocytes and (E) F4/80 positive microglia in the hippocampus. (F) MW01-5-188WH treatment (1.88 treated) does not alter the profile of nitrotyrosine stained neurons, an indicator of oxidative stress linked injury. Representative micrographs are shown for hippocampus sections 15 stained for nitrotyresine-positive neurons, from vehicle-infused mice, Αβ-infused mice (no treatment), and Αβ-infused mice orally administered MW01 -5-188WH (188-treated). Bars- 25 pm. The number (G) of amyloid plaques or tire area oceuoied by amyloid plaques (H) is not tinistration (2,5 mg/fcg) of (n=5/group) was done by determination of the amyloid load. Data are mean ± SEM, 0 Significantly different from Αβ-infused: *p<G.05, **p<O,0X, Figure 5 illustrates that oral administration of M'W01-5-l 88WH attenuates hippocampal synaptic dysfunction and hippocampus-linked behavioral deficits. MW01-5-I88WH administration significantly attenuated the loss of synaptophysin and PSD-95, and the behavioral deficit in the Yrnaze. Oral administration of MW01-5-188WH attenuates hippocampal synaptic dysfunction and
2016203312 20 May 2016 hinpocampus-iinked behavioral deficits. A schematic of the experimental paradigm is shown in Figure 5, MW01-5-188WH administration significantly attenuated the loss of synaptophysin and PSD-95, and the behavioral deficit in the Ymase. Levels of (A) the pre-synaptic protein synaptophysin and (B) the nost-synaptic protein PSD-95 were measured In hippocampal extracts from vehicle-infused mice (control), Αβ-infused mice (no treatment), and Αβ-infhsed mice administered MW01-5-188WH ('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 Αβ infusion. Data are mean + SEM of 5 or 10 per group. Significantly different from Αβ-infused (*p<0.05, **p<0.01, ***p<0,00I),
MW01-2-151SRM was .found to suppress IL-Ιβ in a conoermation dependent manner, hnmunoreaetive levels of glial fibrillary' acidic protein (GFAP), a marker of activated astrocytes were increased in Αβ-infused mice (No treatment) compared to vehicle-infused mice (Control). The Αβ infused increase in GFAP levels was suppressed by administration of MW01-2151RSM. MW01-2-15IRSM also blocked the Αβ-induced increase in the pro-inflammatory cytokines IL-lp, TNF-α» and SI GOB and the Αβ-induced loss of the pre-synaptic protein, synaptophysin and post-synaptic density protein PSD-95. These results are consistent with the results of die cell based assay. (See Figures 6 and 7.)
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.
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% carhoxymethylcellulosc (alternatively, castor· oil or st.same oil can be used) by oral gavage once daily for 3 days. Standard compound doses are 3.1, 12.5, and 50 rag/kg; the highest dose is 20X a therapeutic dose. On die 4ih 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
2016203312 20 May 2016 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.
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.
Ashis;
MW01-5-i88WH 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 rng/kg dose.
MW01-2-151SRM 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 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 mice treated with vehicle or with compound. This compound, is being tested In the acute, escalating dose assay.
have been screened for hERG (human ether-a-go-go) potassium ion channel binding arid inhibition in order to eliminate early in the process any compounds with high potential to induce prolongation of cardiac QT interval in later studies due to off-target toxicides. 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 foe currents have been linked to delayed repolarization of action potentials resulting in prolonged QT interval (Finlayson et al., 2004; Recanatini et ah, 2005: Roden, 2004), QT prolongation is considered a significant risk factor against cardiac safety of new thugs. 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
2016203312 20 May 2016 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 FharmaSendee).
The initial assay is a radioligand binding assay that tests the ability of the test compound to compete with H-astemizoie (a reference standard that binds to hERG channels with nM affinity) for binding to recombinant hERG channels stably expressed on human HEK-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 Ik, current as well as expected pharmacological sensitivities, and is easy to maintain in culture (Zhou et al, 1998). A single concentration (10 μΜ) of test compound is assayed, and % inhibition of ’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 PDA document and tends to give false positi ves, as evidenced by the results reported below.
The hERG channel activity inhibition assay provides whole cell electrophysiological 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, rattier 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 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 astemizoie, 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 jn vivo for prolongation of cardiac QT interval, The QT interval studies are performed by evaluating compounds for effects on QT interval in Lead H 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 mgdeg (dosing volume of 10 ml/kg) to groups of male guinea pigs (weighing 33O~35Og), 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 rog/kg serves as the positive control compound. The QT intervals are corrected for changes in heart
2016203312 20 May 2016 rate using both Bazctt’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 conesponding time point in the vehicle-treated control group for two consecutive observation times indicates significant QT Interval prolongation in the individually treated animals. This 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
Fateh clamp assay; 1.-2 mg
QT interval assay; 5 mg/anlmai/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.
Results;
Competition inhibition assay;
MW01-5-188WH, MW01-2-151SRM, and MW01-6-127WH were tested at 10μΜ concentration,
MW01-5-188WH showed 91% inhibition at 10 μΜ. MW01-2-151SRM and MW01-620 127WH were negative, showing only 8% and 19% inhibition, respectively,
Fateh daw Inhibition assay;
MW01-2-151SRM and MW01-6-189WH were tested at three concentrations (0.1,1= 10 μΜ), These compounds showed minimal inhibition, with IC5.3 values of 4.81 pM for MW02-6189WH and 9.21 μΜ for MWG1-2-1S1SRM,
IS Cardiac OT Interval prolongation assay:
MW01-5-188WH and MW01-2-.151SRM were administered PO at 15 mg/kg to 5 guinea pigs (330-3SOg ’weight). QT intervals were obtained at baseline and at. 15 min, 30 min, 45 min, and 60 min after compound athninistration. Neither compound increased cardiac QT interval above the mean + 2SD of corresponding values in the vehicle control group. There were also no significant effects on mean blood pressure or heart rate after compound administration.
„ Pyepppqf Ion. of 3dd-fephepywQtil )glpsiyazlu-i AOtiyytiteglpg
Ο co
2016203312 20 May
Hgure 1 depicts a synthetic scheme for the preparation of 2-(4-(6-phenylpyridarin-3-yi)piperaziu-lyOpyrimidine (MW01-3-183WH). Reagent and condition: (a) 1-BuOH, ΝΗΧΠ, and 2-( piperazin-iyRpydmidine, A typical reaction mixture of comprised about 0,01 moi of 3-ehloro-6phenvlpyridazinc by 2-(piperazan-J- y!)pyrimidine, about 0.05 moi of 2-(pjpera2an-l”yl)pyrimidine 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^SO*. Removal of solvent followed by recrystallization from 95% ethanol yielded light yellow crystals, yield 96.4%; HPLC: 97.4% purity; HRMS calculated 3.18,1587, found 318.1579; 1H NMR (CDCI3): S 8.356 (d, J=4.5, 2H), 8.0Il(d, 1=7.5, 11 2H), 7.692. (d, J=9>5, Iff), 7.468 (t, 1=6.0. 2H), 7.417 (d, J=7.5,1H), 7,047 (d, J=9.5, ΪΗ), 6.546 (t, 3=4,5, IH), 4.013 (t, 1=5.0.4H), 3.826 (t, J=5.0,4H).
Example 5, Biolcgkui Activity, Metabolic Stability and Toxiehy of 4»ureWHi-phCTyl-344·· romirmdm»2-vloioeragin-l-yl)ovridazme (W01-2-151SRW
As shown in Figures 6A-D and 7 A-G, the biological activity of MW01-2-151SRM 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. lire compound was a selective suppressor of activated glia responses, especially key proinfiammatory responses that have been linked, to AD pathology. It also showed efficacy in the mouse model of human Αβ-induced neitroinflammation and neuronal injury as shows by die assays of Figure 7 A-G, lire stability of MW01-2-151SRM in nticrosomes, as shows is Figures 47 and 48, was demonstrated for MW01-2-151SSM. The stability of MWO1-5-188WH (1 pM) is a standard 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 eentrifwged at 16 OOOxg for 10 min. 10/d of the supernatant was analyzed by calibrated HPI.C to quantify the percentage of the initial amount of MW1-2-151SKM remaining after the incubation. The HPLC. system (Dicncx Corp., Sunnyvale. CA) includes a Dionex P480 pump, a Phesomenex © Luna 08 colunm (250 x 2,0 mm, 5Dm) with a guard column (Phenojnesex, Torrance, CA) and a Dionex UVD34OU Ultraviolet (UV) detector. The mobile phase consisted of 0.1% formic add as reagent A and 0.08% formic acid'water in 80% acetonitrile as reagent B, at a flow rate of 02. ml per minute. The gradient consisted of the following linear and isocratic gradient dntion 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
2016203312 20 May 2016 quantification was done based on absorption measured at 260 nm relative to a standard curve obtained by using serial dilutions of MW01-2-151SRM.
liver toxicity after chronic in vivo administration of MW01-2451SRM was investigated, see Figure 47 A. Mice were administered by oral gavage either MW01-2-151SRM (2.5 mg/kg/day) or diluent (10% DMSG) in a 0.5% (w/v) carboxymethylcelluiose 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 Dm liver sections were stained with haematoxylin and eosin. Two independent observers blinded to the treatment groups performed microscopic assessment of the tissue for injury,
A synthetic scheme for the prepration of /V-(cyclopropylmetliyI)--6-phenyl-4-(pyri.din-4yl)pyridazin-3-amine (MW01-7-084WH) is depicted in Figure 8. and synthesis was carried out as described herein.
4<hlQro-6-phenyipyridastin-3(2H)<me(h4W01-^-tB3WH)
4-ohJ.oro--6--phenylpw'idazin-3(2H)-one was synthesized according to tire procedure described by Coudert, P. [18],
4--chlori>2-(methoxyiacfeyiLd-phenylpyrifein~3(2H)-Qne(MW01-7-O53WI-D
A uaxture of cWoropyndazinone 1 (25,5 g, 0,12mo.l), 4-N,N-tKmethyiaminopyridine (0,20 g) andiPr2bEt (26.7g, 0,21rool) In anhydrous CHjCl?. (300mL) was stirred at (?C (ice bath) for 30 min. Methoxymethyi chloride (25g, 0.3.bnoi) was added and the mixture was stirred at 0 jaC for Ih and then allowed to warm to room termperature, The reaction was shirred at room temperature till complete. The solvent was then removed in vacuo, tire residue was treated with water, washed with dilute NagCCb solution and extracted with EtOAc. The organic layer was dried over anhydrous Na2SO4, filtered and evaporated. The residue was then purified by recrystaUization from 95% ethanol to give 20.1 light yellow solid. Yield 66.9%.
jS^^yl4~(pyridin-4-yi)pyri<fe^~3(2H)ri3Pe(MW017-O6gWII)
The protected pyridazinone MW0I-7-053WH (I.Oequiv.) was mixed with arylboronic acid (1.37equiv,), PdfFPh3)4 (0.05 equiv.) and. K2CO3 (3.1equiv) and 200 tnL of DMS in a 350ml of pressure vessel, flushed with argon for 3 min, and the mixture was then stirred and refluxed (oil bath. 120C) 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
2016203312 20 May 2016 was dissolved in 200ml of EtOH, 6 N HQ (200 ml.) was added and the reaction mixture was refluxed (oil bath, 120 pIC) for 6 h, then it was allowed to cool to room tennperatare, 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. Xteaystaffization from 90% ethanol provided brown yellow' solid. Yield 80.4%. ESI-MS: m/z 294,3 (M+H+)
3-chloro-6--phwyi-44pyridm-4-yl)p>ii^zme(MW()l-7-076WH)
3-cldoroAl~phenv'l-4--(pyridin-4--yl.)p>'rldazine (MW01-7-O76WH) (66mmol) was suspended in 75ml phosphorus oxychloride and heated with stirring at lOO^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 fiber funnel to yielding a light yellow solid, ESI-MS: ro/z 263,4 (M+1T+).
NVcycjopropyIrnethyft-6-pjienyl-4--(pyridin-4--vl)pvridaz.in-3-armne fMW01-7-O84WIi)
A mixture of N-<cyclopropylmefeyl)-6-pheayl-4-(pyridin-4-yl)pyri<tein~3-atnine (MW01-7084WH) ( O.Smmol), C-Cyclopropyl-mefliylarmne (Z.Ommol) in 3ml. of 1-BuOH was heated with stirring at 1.30°C 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: nvz 330.4 (M+H+).
A mixture of 3-chloro-6phc.nyl-4-(pyridin-4-yl)pyridazine (MWOI-7-076WH) (0.5mmol), 1l-piperazine- (2.0mmol) in 3 ml of 1-BuOH was heated with stirring at 130°C for about 7 The soiveat 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 to yield a brown solid, ESI-MS: ru/z 332.2 (M+H*). A synthetic reaction scheme for the· preparation of 3-(4-methylpiperazin-l-yl)-6(MW01-7-085WH) is depicted in Figure 9.
A mixture of 3-chloro-6-phenyl-4-(pyridin-4-yi)pyridazine (MW0I-7-076WH) (0,5mmol), 2~4~yl-ethyl:nnine (2.0 mmol) in 3ml of 1-BuOH was heated with stirring nt 130°C for days. The solvent was removed by evaporation in vacuo, the residue- was treated with
Example 8. Fmraratioa of A’-i2~.moredrolmoethvI)dhphnm4.-4-ipy?id.m--4-yl)pyridn:ain”3” amfate (Wm-7«OTB
2016203312 20 May 2016 water to give a suspension. The solid was then filtered off, washed with water, them 1:3., Ethyl Acetate: Petroleum ether, dried over filter funnel in vacuo to yield a gray solid. ESI-MS: mZz 362 2 (M+lF). A synthetic reaction scheme for the preparation of 3-(4-.mcthy1piperazin-l yl)Ar-(2-morphoiinoethyl)-6-phenyl-4-(pyridin-4-yl)pyridazin-3-atmne (MWO1-7-091WH) is depicted in Figure 10.
Figure AU2016203312B2_D0008
A synthetic reaction scheme for tits preparation of 5’(4-Florophenyl)~3-phenyl“6-(4-pyrimidin2-ylpiperazin-l-yi)pyridaziae (MW01~2-G65X,KM) is depicted in Figure 11, and synthesis was carried out as described herein.
3-phenyl-6-(4-pyti.midi n - 2-ylpiperazh·-1 -vDpvri dari.u-5-oi (MWO 1-6-006WH)
This compound was prepared from 3-chloro-5~hydroxy-6-phsnylpyridasine (1.4g, 6.8mmol) In the same manner as described for MW0I-6-121WH, yielding white solid (2.12g, 6.15mmol, 90.4%). MALDI-TQF: m/z 335,7 (M+H+), 1HNMR (DMSO): d 8.433 (t, 1=2,0,1=2.4, 2B), 7.773 (d, 1=3,2, 2H), 7.497 (t, 1=2.0,1=3.6,3H), 7.182 (s, IB), 6.727 (s, IH), 3.949 (s, 4H).
5ChlofQ~3~ t(MW01-6-015WH)
3“CWoro-5-hydroxy-6-phenyipyridazme (66mmol) was suspended in 75ml phosphorus oxychloride and heated with stirring at KKTC 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 yield white solid (98.8%). ESI-MS: ra/z 353.3 (M+H+). IH NMR (CDC13): d 8.375 (d, 1=5.0, 2H), 7.776 (d, 1=7.0, 2H), 7,487 (m, 3H), 7.073 (s, IH), 6,588 (t, 1=4.5, IB), 4,046 (t, 1=4.5,
1=5.5,4H), 3.849 (t, 1=5,5,1=5.0,4H),
5^4rR(^g^^I>35>h^yl-^4rg^jmidin-2~yifflperazin-byi)pyridaziaeiM.W01-24)651.KM)·
This compound was prepared in the same manner as described for MW01-7-069WHWH, yielding a white solid (60.4%). MAWI-TOF: m/z 413.4 (M+H+).
Figure AU2016203312B2_D0009
A synthetic reaction scheme for the preparation of 5-(4-pyridyl)~3-phenyl-6-(4“pyrimidin-2ylpiperazm-l-yl)pyridazlne (MW01-2-069A-SKM) Is depicted in Figure 12, and synthesis was carried out as described herein. This compound was prepared in the same manner as described for MW01-2-065LKM, yielding white solid (65,4%), MALDI-TOF: m/z 396.2 (&M+).
4-ntothyl-6-phenyl-3-(4-pyrimidin-2-ylpiperazin-l -yl)pyridasane (MW01-2-151SRM) prepared by several synthetic schemes as depicted in Figure 13 (Scheme Ik 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.
2016203312 20 May 2016 was
3- cbloro-6-phenylpyridazim4mi was synthesized according to the procedure described by Coudert, P., et al. [18],
6-phen vl-3 -(4-(pyrimidin-2-yDpiperazin-1 - yl)pyrida?ln-4-oi (MW01-7-121WH)
This compound was prepared from 3-chloro»4~hydroxy-6-phenyIpyridazme (14g, 68mmoi) in the same manner as described below, yielding white solid (22,lg, 66mmol, 97,3%), ESI-MS:
m/z 335,2 (M+H+). 1H NMR (DMSO): 1H NMR (DMSG): d 8.406 (d, 1=6.5, 2H), 7.740 (d, 1=4.0, 2H), 7.558 (s, 3H), 6.686 (t, J=4.8,1=4.4,1H), 6.841 (s, 1H), 3.881 (s, 4H), 3.620 (s, 4H), 3.776 (s, 4H),
4- chloro-6-phenyI-3-(4-pyrimidin-2-ylplperazin-l-yr;pyridaziue (MWQ1-6-127WII) 6-phenyI~3<4-pyrinridin-2-ylpiperaziii-l-yi)pyridazin-4-ol (22.0g, 66mmoi) 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 (M+H+). 1HNMR (CDC13): ¢1 8.377 (d, 1=4.5, 2H), 8.036 (d, 3=7,5, 2H), 7.833 (s, IB), 7.508 (m, 3H), 6.564 (t, 3=4.5,1H), 4.073 (t, 1=4.0,1=4.5, 4H), 3.672 (t, 1=4,0,1=4.5,
Into a reaction tube were added MW01-6-127WH (l,4g, 4.0mmoi), K2CO3 powder (1.7g, 12.4mmoi), Pd(dppf)Ci2 (326mg, O.-fmmoI), silver oxide (2.3g, Wmraol), metltylboronic acid (324mg, 5.4mmol) and 20ml of THF. Argon was then flushed through the tube for 3min. The 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, l.Smmol, yield 45.2%). ESI-MS: m/z 333.4 (M+H+). 1H AMR (CDCI3): d
2016203312 20 May 2016
8.380 (d, 1=5.0, 2E1), 7.065 (d, J=7.0, 2H), 7.626 (s, IB), 7.473 (m, 3H), 6.367 (t, J=4.5,5=5,0, IB), 4.056 (t, 5=5.0,4H), 3.475 (t, 1=5.0,4ΪΪ), 2.456 (s, 3H).
Scheme 2 into a reaction tube were added MW01-6-127WH (1.4g, 4.0mmol), K2CO3 powder (1.7g, 5 12.4m.moi), Pd(FFh3)4 (240mg, 0.2nunol), silver oxide (2..3g, lOmmol), juethylboronic acid (324mg, 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 120uC 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 powder solid was obtained (0.64g, 1.93mmol, yield 48.1%). ESI-MS: m/r, 333.4 (Μ+Ή+)..1Η NMR (CDC13): d 8.380 (d, J«5.0,2H), 7.065 (d, 1=7.0, 2H), 7,626 (s, 1H), 7.473 (m, 311), 6.567 <t, 1=4.5,1=5.0, IB), 4.056 (f, 1=5.0,4H), 3.475 (t, 1=5.0, 4H), 2.456 (s, 3H).
Schemes
4»S'-^hydro-4-meth.yl~6-:'pheoylpyridazia-3f2H)-oae (MW01-8-0Q4WH)
7.7 g (40 mmole) of 2-methyl-4-oxo~4-phenylbutanoic acid was added to a 100 ml singlenecked round bottom flask followed by 3,0 ml (60 mmole) of hydrazine roonohydrate 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 fee 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 NaHCOs, 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 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(2H)rQne (MWO1-8-008WEI)
7.0 g (35 mmole) of MW01-8-004WH was placed in a 100 ml single-necked round bottom flask followed by 9.4 g (70 mmole) of anhydrous copper (Π) chloride and then 30 ml of acetonitrile- to give a brown yellow suspension. A reflux condenser was connected to fee flask and a dry tube filled with CaC!2 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 temperature. The mixture was poured on to 300g of crushed ice and stirred vigorously for 10 minutes to give a gray precipitate
2016203312 20 May 2016 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 HCI solution to rid tee 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 6.3 g of a blue gray solid. Yield was 96.7% and confirmed by ESI-MS. ESI-MS: rmz 187.3 (M+H+).
6.0 g (32 mmole) of MW01-S-008WH and 30 ml (320 mmole) of phosphorus oxychloride were placed in a 100 ml single-necked round bottom flask. The flask was connected with a reflux condenser and a dry tubs filled with anhydrous CaCia was fitted to the top of tee condenser. (HCI gas is formed in tee 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. (phosphorus oxychloride can be decomposed by water to give HCI and H3PCfi). The mixture was teen stirred vigorously for 10 minutes to give a white suspension. The suspension was neutralized with a 2N NaOH solution until tee pH of tee 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 by ESI-MS. ESI-MS; m/z 205.4 (M+H+).
2ri4ri4-mgtevl^phgnylpyria^m-3-yDgijto^
0,82 g (4.0mmole) of WH-8-012 was placed in a 30 ml pressure vessel followed by addition of 2.6 g (lg.Ommole) of l-(2-pyrimidyI) piperazine and then 15 ml of 1-BuOH. The vessel was sealed tightly and placed into an oil bate and stirred at 130 C (temperature of oil bath) for 2.5 days. The reaction mixture was then cooled to ambient temperature and transferred to a singlenecked flask for evaporation under reduced pressure. Removal of solvent gave rise to a brownred residue that was treated with 30 mi of water to give a brown sticky oil. The mixture was kept at ambient temperature overnight while tee oil sutidified gradually. The formed solid was then broken into small pieces with a steel spatula. The solid was collected by filtering and 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, WH-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 funner one. So it Is up to die process chemist to decide which procedure to pick, for the
2016203312 20 May 2016 manufactore. The precipilatkin 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 filter funnel in vacuo to give the desired product.) The final compound was confirmed by LSIMS and NMR. ESI-MS: ru/z 333.8 (M+H+). IB NMR (CDC13): d 8.380 (d, 1=5,0, 2H), 7.065 (d, 1=7,0, 2H), 7.626 (s, IK), 7.473 (m, 3B)« 6.567 (t, J=4.5,1=5.0, .IB), 4.056 (t, 1=5.0, 4H), 3.475 (t, J=5,0, 4B), 2.456 (s, 3H).
4,6-diphenyl-3-(4-pyrimidin-2-yipiperazin~l-yl)pyridaz.me (MW01-5-188WH).was prepared by several synthetic schemes as depicted in Figure 16 (Scheme I), Figure 17 (Scheme 2), and Figure 18 (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 MW01-2-188WH.
3- chlpro-6-phenylpyridazin-4--oI was synthesized according to the procedure described by Coudert, P., et al. [18], fophenyi-3.ri4Tpwtrddim2-yi)piperazin-1 -yDpyridazinM-ol (MW01-7--121 WH).
The compound was prepared from 3-chloro-4-hydroxy-6-phenylpyridazine (14g, 68mmol), A mixture of 3-cMoro-4,6-diphenylpyridazine (267mg, l.Ommol), l-(2-pyrimidyl)piperazme (656mg, 4.0mmol) in 3ml of 1-BuOH was heated with stirring at !30*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 ¢22. lg,. 66mmol, 97.3%). ESI-MS: m/z 335.2 (MTH+). 1H NMR (DMSO): 1H NMR (DMSO): d 8.406 (d, 1=6.5, 2B), 7.740 (d, 1=4.0, 2H), 7,558 (s, 3H), 6,686 (t, 1=4,8,1=4.4, IB), 6.841 (s, IB), 3.881 (s, 4H), 3.620 (s, 4H), 3.776 (s, 4B).
4- chlQrp-6-phenyl-3-(4“pyrimidin-2-ylpiperarin-lyl)pvridazine (MW01-6rl27WH).
6-ph£nyl~3’(4-pyrimidin”2-ylpipemzIn-l-yi)pytidazin-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 crashed 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
2016203312 20 May 2016 (M+H*). 1HNMR (CDCI3): d 8.377 (d, 1=4,5, 2H), 8.036 (d, J=7.5S 2H), 7.833 (s, IH), 7.508 (m, 3H), 6.564 (t, 1=4.5,1H), 4.073 (t, 3=4,0, 3=4.5,4H), 3.672 (t, £4.0,7=4,5, 4H).
4,6--diphenyl-3dl4-p5Tinndin-2--ylpiperaan-i-yl)pyridagine (MWQ.fej-lSS.WK)
A mixture of 3-chloK>4,6diphenylpyridazine (267mg, l.Ommot), l-(2-pyrimidyi)piperazine (656mg, 4.0mmol) in 3ml of 1-BuOH was heated with stirring at 130J>C for 3days. The solvent was removed hv 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: na/z 395.5 (M+H+). HRMS calcd 395.1979, found 395.1973; 1H NMR (CDCI3): d 8.329 (d, 1=5.0, 2H), 8.101 (d, 1=7.5, 2ΙΓ),
7.734 (d, 1=7.5, 2H), 7.655 (s, IH), 7,509 (m, 6H), 6.530 (t, 1=4.5, IH), 3.836 (t, 3=4.5, 3=5.0, ; 3.394 ft, 3=5.0,1=4.5,
Figure AU2016203312B2_D0010
Lagin-3(2H)-one
135 ml ¢135 mmole) of a solution of phenylmagnesium bromide (1M) in THF was added to a 15 hot suspension of 6-phenylpyridazinone compound 7.8g (45 mmole) 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 frit sintered glass funnel in vacuo to give 5.6 g of white crystals. Yield was 50%, confirmed by E5Is. ESI-MS: m/z 250.1 (M+H+).
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 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 (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 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.
2016203312 20 May 2016
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-h).
Stehlpro-b^dicnyi-d-aheaylpyridazme
2.0 g (8 mmole) of 6-phenylpyridazmone obtained above aad 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 tribe filled with CaC12 was fitted to the top of the condenser. (HQ gas is formed in the reaction so a basic solution such as NaOH may be needed to absorb HQ 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 HC1 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 1.8 g of a light pink powder. Yield was 85%, confirmed by ESI-MS. ESI-MS: m/z 266.4 (M+H+).
1.1 g (4.0mmole) of 3-chIoropyridazine obtained above was placed in a 30 ml pressure vessel followed by addition of 2,6 g (IhOnunole) of l-(2-pyrimidyl) piperazine and then 15 ml of 120 BuOH (reagent grade). The vessel was sealed rightly and placed into an oil bath and stirred at 130®C (temperature of oil bath) for 3 days. The reaction mixture was then cooled Ιο 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 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+). HUMS calcd 395.1979, found 395,1973; 1HNMR (CDC13); d 8.329 (d, 1=5,0, 2H), 8,101 (d, 1=7.5, 2H), 7.734 (d, 1=7.5, 2H), 7.655 (s, IH), 7.509 (m, 6H), 6,530 (t, 1=4.5, IH), 3.836 (t 1=4.5,1=5.0,4H), 3.394 (t, 1=5,0,1=4.5,4H).
Scheme 3
Ti 3-cldoro-ri-phenylpyridazm4-ol was synthesized according to the procedure described by Condert, P., et al. [18].
4,6-diphenyl-3-i4-pyrimidin-2-y]piperazin-l-vilpyridazine (MW01--5-188WH)
A mixture of 3-chloro-4,6-diphenylpyridazine (267mg, l.Ommol), l~(2~pyrimldyl)piperazine (656mg. 4.0mmol) in 3ml of 1-BuOH was heated with stirring at 130 C for 3days. The solvent
2016203312 20 May 2016 was removed by evaporation in vacuo, the residue was treated wife 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, O.SlmmoI, yield 81,1%). ESI-MS: xn/z 395.5 (M+H+). FIRMS ealed 395.1979, found 395.1973; IH NMR (CDC13): d 8.329 (d, 1=5,0, 2H), 8.101 (d, 1=7,5, 2H),
7.734 (d, 1=7,5, 2H), 7,655 (s, IH), 7.509 (m, 6H), 6.530 (t, 1=4,5, IH), 3.836 (t, 1=4,5,1=5,0,
4H), 3.394 (t, 1=5.0,3=4.5,4H).
Figure AU2016203312B2_D0011
4-pyridyl '6-phenyl-3“(4~pyrimifen~2*ylpipeinzin-1 ~yl)pyridaxine (MW01-6-189WH) 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 fee preparation of MWG1 -2-189WH.
3<ddoro-6-phenytoytidnzi'n-4rol was synthesized according to the procedure described by Coudert, P., et al, [18],
Tins compound was prepared from 3-chlaK>-4-hydcoxy-6-phenyipyridaane (14g, 68mmoi) A mixture of 3-chloro-4,6-diphenylpyridazine (267mg, l.Qmmoi), l~(2-pyrimidyl)piperazine (656mg, 4,0xnmol) in 3ml of 1-BuOH was heated with stirring at 130C &r Sdays. The solvent was removed by evaporation in vacuo, fee residue was treated with water to give- a suspension. The solid was feen filtered off, washed with water, dried over filter funnel In vacuo to give light pink solid, yielding white solid (22. !g, 66mmoL 97.3%), ES1-MS; m/z 335.2 (M4H+). IH
HMR (DMSO): IH NMR (DMSO): d 8,406 (d, 1=6.5, 2H), 7,740 (d, 1=4.0, 2H), 7.558 (s, 3H), 6.686 (t, 1=4.8,1=4.4, IH), 6,841 (s, IH), 3,881 (s, 4H), 3.620 (s, 4H), 3,776 (s, 4H). 4<hloro-6^hgnyl-3-(4-pyri3mdia-2-yipiperazin-l-“yl)pyridazine (MW01-6-i27WTj 6-phenyl-3-(4-pyrmtidin-2-ylpiperazin--l-yl)pyridazin-4~oI lb. (22.0g, 66mmol) was suspended in 75ml phosphorus oxychloride and heated with stirring at 100 for 3h. After cooling to room temperature the mixture was poured onto crashed ice. The mixture was then neutralized wife NaOH solution to give white suspension. The precipitation was filtered off, washed with water, dried over filter funnel to provide white solid (21,3g, 60.3rnm.ol, 91.4%). ESTMS: m/z 353.4 (M+H+). IH NMR (CDC13): d 8,377 (d, J=4.5, 2H), 8.036 (d, 1=7,5, 2H). 7-833 Ob IH), 7.508 (m, 3H), 6.564 (t, 1=4.5, IH), 4.073 (t, 1=4.0,1=4.5, 4H), 3.672 (t, 1=4.0,1=4.5,4H),
2016203312 20 May 2016 vi-3r(4-pyrirmdrn-2-ylpip^azin-l-yl)pyridazrne (MW01-6-189WH)
Into a reaction tube were added WH-6-127 (1.4g, 4,0mmol), K2CO3 powder (1.7g, 12.4mmol), Pd(PPh3)4 (24Grmg, 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 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 (Q.65g, 1.65mmol, yield 41.2%). Confirmed by ESJ-MS and NMR. ESJ-MS: m/z 396.2 (M+H+). 1HNMR (CDC13): d 8.809 (d, >6.0, 2H), 8.335 (d, >5.0, 2H), 8.090 (d, >7.5, 2H), 7.750 (m, 6H), 6,543 (t, >4.5, 1H), 3.868 (t, >5.0,4H), 3.404 (t, >5.0,4H),
4.5-dihydro-6-phenyl4-fpyridin4-yi)pyridazitt-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 mmole) of 4-bronxopyridine and 70 of anhydrous THF. The system was oven-dried and flushed with argon before use. 135 ml (135 mmole) of THF solution of phenyhnagnesium 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, die reaction was stirred for 15 minutes for completion. The solution of Grignard reagent, was then obtained. A solution of 410 pyridyl magnesium bromide obtained above was added to a hot suspension of 6phenylpyridazinone compound 7.8g (45 mmole) in dry toluene (50 mi). 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, aud the aqueous layer was extracted with 100ml of ethyl acetate. The solvent was removed and the residue was crystallized from ethanol. Tbs crystals were collected by filtering and dried over a medium frit sintered glass funnel in vacuo to give 5.6 g of white crystals. Yield was 50%, confirmed by ESI-MS. ESJ-MS: m/z 252.1 (M+H+).
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 CaC12 was fitted to the top of the condenser. The reaction mixture was heated to reflux in an oil bath (rifi C) for 3 h. The color of the reaction suspension changed to dark yellow one© the reflux started. After the completion of the reaction
2016203312 20 May 2016 (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 crashed 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 I,5-2,0), and washed with 50 ml of a IN HC1 solution to rid the solid of any remaining copper byproducts. This is followed by washing with 100 ml of Nfilli-Q water to get rid of the acid in tire solid, and is monitored by checking tire 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-HH-s.
2.0 g (8 mmole) of 6-pheuylpyridazinone 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 CaC12 was fitted to the top of the condenser. (HCI 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 crashed ice. (phosphorus oxychloride can be decomposed by water to give HCI and H3PO4). The mixture Wits 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 1.8 g of a light pink powder. Yield was 85%. confirmed by ESI-MS. BSI-MS: m/z 268.4 (M+HT).
4-pyridyl-6-phenyl-3A4-nvrimidin-2-vlpiperazin-l--vl)pyridazine (MW01-6-189WH)
1.1 g (4.0mmole) of 3-chloropyridazine obtained above was placed in a 30 ml pressure vessel followed by addition of 2,6 g (16.0mmole) of l-(2-pyrirnidy3) piperazine and then 1.5 ml of 1BuOH (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 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 (CDC13): d 8.809 (d, 1=:6,0,2H), 8.335 (d, 1=5.0, 2H), 8.090 (d, 1=7.5, 2H), 7,750 (ra, 6H), 6,543 (t, 1=4.5,
3.868 (t, 1=5.0,4H), 3.404 (t, 1=5,0,4H),
2016203312 20 May 2016
Figure AU2016203312B2_D0012
A synthetic reaction scheme for the preparation of 4,6-diphenyl-3-(4-phenylpiperazin-l5 yl)pyridazine (MW01-7-029WH) is depicted in Figure 20, and synthesis was carried out as described herein. The compound was prepared from 3-chloro-4,6-diphenylpyridaz:ine (lOOmg,
O.37mmol) in the same manner as described for MW01-7-057WH, yielding white solid (123mg, 0.31ramol, 83.1%). ESI-MS: m/z 393,2 (M+H+). NMR (CDC13): d 8,107 (d, 3=8,0, 2H), 7-736 id, 1=7.5, 2H), 7,65.1 (s, 1H), 7.500 (no, 511), 7,290 (t, J=8.5, 7=6.5, 3H), 6.958 (d, 3=7,5, 2H),
1.0 6.899 (t, 1=7.0,113), 3.487 (s, 4H), 3.214 (s, 4H).
Figure AU2016203312B2_D0013
A synthetic reaction scheme for the preparation of 4,6-diphenyl-3-(4-methyIpiperazin-l15 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-chioro-4,6-diphcnylpyridazine (lOOmg,
0.37mmol) in the same manner as described for MW01-7-GS7WH, yielding white solid (119mg, 0,35mntol, 94.5%), ESI-MS: m/z. 331.1 (M+H+). NMR (CDC13): d 8.089 (d, 3=7,5, 2H), 7,643 (d, 3=7.5,2H), 7.611 (s, IB), 7.510 (m, 6H), 3,365 (s, 3H), 2,472 (s, 4H), 2.337 (s, 4H).
(MWei-3-06SSKM)
A synthetic reaction scheme for the preparation of 4,6“diphenyl-3-(4~cyclohexylpiperazin-l·yl)pyridaane (MW01-3-065SRM) is depicted in Figure 22, and synthesis was carried out as described herein. The compound was prepared from 3-chloro~4,6-diphenyipyridazine (30Qmg,l.lrnmol) in the same manner as described for MW01-7-057WH, yielding white solid (350mg, 0.87mmol, 87%), ESI-MS: m/z 399.2 (M+H+). IH NMR (CDC13): d 8,09 (d, 1=7.5, 2H), 7.68 (d, 3=7.5, 2H), 7.59 (s, IB), 7.56-7.42 (m, 6H), 3.39 (s, 4H), 2.62 (s, 4H), 2.273(s,
IB), 2.01-1.78 (m,4H), 1.63 <d„ 3=12.5, 111), 1.33-1.08 (m, 5H).
17, Reparation o£4.6-diphmYl4K4tisopropvMwra5rinX»y!}pyrldazlm (MW013J66SKMX
A synthetic reaction scheme for the preparation of 4,6~diphenyl-3-(4-isopropylpiperasin-lyl)pyridazine (MW0I-3-066SRM) is depicted in Figure 23, and synthesis was carried out as
2016203312 20 May 2016 described herein. The compound was prepared from 3-chloro-4,6-diphenylpyridazine (300mg, L I mmol) in the same maimer as described for M WO!.-7-057WH, yielding white solid (290mg, O.Slrnmoi, 72%), m/z 359.2 (M+H+). 1HNMR (CDCI3): d 8.09 (d, J= 7.5, 2K), 7.69 (d, 1=7.5, 2H), 7.61 (s, 1H), 7.54-7.46 (m, 6H)> 3.40 (s, 4H), 2.72(m, 1H), 2.59 (s, 4H), 1.10 (d, 1=6, 6H).
A syaiheiic reaction scheme for the preparation of 4,0-diphenyi-3~piperarinyipyridazine (MW01-7-133WH) is depicted in Figure 24, and synthesis was carried out as described herein. The compound was prepared from 3-chioro-4,6-diphenylpyridazine (5'33.m.g, 2Qmmole) in the same manner as described for MW01-7-057WH, yielding light yellow solid (550mg, 17.4mmole, yield 86.9%). ESI-MS: m/z 317.3 (M+H+). 1H NMR (CDC13): d 8.086 (d, 1=7,5, 2H), 7,705 (d, 1=7.5, 2H), 7.619 (s, 1H), 7.498 (m, 6H), 3.318 (d, 1=4.0, 411), 2,932 (d, 1=4.0, 41-Ϊ) 1.896 (s, HI).
Figure AU2016203312B2_D0014
A synthetic reaction scheme for the preparation of 2-(4-(6-phenyl-4-(piperidm~l-yI)pyridazin-3“ yl)piperazin-l-yl)pyriroidine (MWG1-7-107WH) is depicted in Figure 25, and synthesis was carried out as described herein. The compound was prepared from MW01-6-127WH (2O0rag, 0.57rnmole) in the same manner as described for MW01-7-057WH, yielding light yellow solid (220mg, O.55romole, yield 96.3%). ESI-MS: m/z 402.5 (M+H+),
Exsrereto.......20,
Preparation of . .frmethy
A synthetic reaction scheme for tire preparation o.f6-raethyi-4-pheny.l-3-(4~p\rinudi.n-2ylpipetazin-l-yl)pyridazinc (MW01-7-057) is depicted in Figure 26, and synthesis was carried out as described herein, A mixture of 3-chIoro-6-methyI-4-pheny1pyridarine (lOOrng, Orimmol), l-(2~pyri.midyl.)p:iperazine (40()mg, 2.0rmnoi) in 3rnl of 1-BuOH was heated with stirring at 130 C for 7days. The- solvent was removed by evaporation in vacuo, tire 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 to give light, yellow solid (68nag, 0.20mmol, yield 41,7%), Purity >95%; ESI-MS: m/z 333.1 (M+H+). 1H NMR (CDC13); d 8.310 (d, 1=5,0, 2H), 7.678 (d, 1=7.5, 2H), 7,476 (m, 3H)S 7.119 (s, H), 6.509 (t, 1=4.5,1H), 3.785 (t, 1=4.5,1=5.0,4H), 3.277 (t, 3=4.5,1=5.0,4PI), 2.669 (s, 3H).
2016203312 20 May 2016
Figure AU2016203312B2_D0015
Figure AU2016203312B2_D0016
A synthetic reaction scheme for the preparation of 2-(4~(5-phenyl-6-(pyridin-4-yl)pyridnzin-35 yi)p.iperazin-l-yl)pyrimidine (MW01-2-163MAS) is depicted in Figure 27, and synthesis was carried out. as described herein.
4.0 g (23 mmole) of 3-phenyIfumn-2.S-di.one was added to a 100 ml single-necked round bottom flask followed by 2.9g (2.7.6 mmole) of hydrazine monohydrate and then 20 ml of 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 1.1.0 degree (temperature of oil bath) and stirred for
h. The flask was then removed from the oil bath and tire 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 minutes to give a suspension. The precipitate was collected by filtering, washed with 100 mi 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*), .6-dirfoloro-4-pbenvlpyridazine (MW01-2-082A-MAS)
1,5 g (8 mmole) of 6-phenyipyridazinone 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 CaC12 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.
Ϊ5 (phosphorus oxychloride can be decomposed by water to give HC1 and ΙΪ3ΡΟ4). 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 foe 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
ESI-MS. ESI-MS; m/z 226,1 (ΜΗΓ).
Figure AU2016203312B2_D0017
A mixture of 3,6-dichIoro4-phenylpyridazine (1.35g, ommol), I-(2-pyrimidyl)piperazine (E2g, S.Ommol) in 10ml of I-BuOH was heated with stirring at 80°C for 12h. The solvent was removed by evaporation in vacuo, the residue was treated with water to give a suspension, The
2016203312 20 May 2016 solid was then filtered off, washed with water, dried over filter funnel in vacuo to givewhite solid (l,8g, 5.2mmol, yield 86,0%). ESi-MS: m/z 353,9,
2^.(4<5-pl^yI^ipyridin-4-yl)pyridaein--3-yl)pipermin-i-yl>pyrin4d3ne(MW01-2-l!63MAS) into a reaction tube were added. MW01-2-114B-MAS (1 „4g} 4.0imnol)} K2CO3 powder (l,7g,
12.4mmol), Pd(PPh3)4 (240mg, 0,2mmol), 4-pyridineboronic acid (664mg, 5.4mmol) and 2O.mi of DME. Argon was then flashed 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 eelite 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 (0.69g, l„74namol, yield 43.5%), Confirmed by ESI-MS and NMR. ESI-MS: m/z 396.2 (M+H4).
amine (MWOl-7-WTOh
A synthetic reaction scheme for the preparation of /v-(cyclopropyb.uethyi)-6-phenyi-4-(pyridin4-yl)pyridarin-3-amine (MW01-7-084WH) is depicted in Figure 28, and synthesis was carried out as described herein.
•3(2g)-one (MW01~6~093WD was synthesized according to the procedure described by Coudert, P., et al. [18].
4-cidoro-2-(methox.ygiethyl>6-phenyipyridazm-3i'2g)~one 6MW01-7-053WH)
A mixture of cMoropyridazinone 1 (25,5 g, 0.12mol), 4-N,N-dimethylamin0pyridine (0.20 g) and i~Pr2NEt (26.7g, 0.21mol) in anhydrous CH2C12 (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 Ih and then allowed to warm to r, t. The reaction was starred at r.t, till it complete. The solvent was then removed in vacuo, die residue was treated with water, washed with dilute Nt^CO* solution and extracted with EtOAc. The organic layer was dried over anhydrous NajSOi, filtered and evaporated. The residue was then purified by reorystallization from 95% ethanol to give 20.1 light yellow solid. Yield 66.9%.
The protected pyridazinone MW01-7-053WH (LOequiv.) was mixed with arylboronie cid (1.37equiv.), Pd(PPh3)4 (0.05 equiv.) and K2CO3 (3.1eqniv) 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
2016203312 20 May 2016 off. The filler cake was washed with water over filter Tunnel and teen used for next step directly. The residue obtained above was dissolved in 200ml of EtOH. 6 N HCI (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 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 (MW)
3-chlpro-6-phenyl-4-(pyridin-4-yl)pyridazme (MWO1-7-076WH)
This compound was prepared from MW01-7-O69WH in the same manner as described for MW01-6-127WH, yielding light yellow solid. ESI-MS: m/z 268,4 (MMT). ?/-(byclopropylmethyl)-6-phcnvl“4(pyridln-4-y1)pyridazin-3-amine fMW01-7-O84WH)
This compound was prepared from MW01-7-076WH in the same manner as described for MWOl-7-057WH, yielding gray solid. ESI-MS; m/z 330.4 (M-rH4).
?0
The present invention is not to be limited fn. scope by the specific embodiments described herein, since such embodiments are intended as but single illustrations of one aspect of the invention anti any functionally equivalent embodiments are within the scope of this invention. Indeed, various modifications of the invention in addition to those shown and described herein will become apparent to those skilled in the art from the foregoing description and accompanying drawings. Such modifications arc 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 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
K> of prior invention.
2016203312 20 May 2016
Mel
Figure AU2016203312B2_D0018
2016203312 20 May 201
14 0 Η Λ. 0^0 Ν 5 X J·', 0 υ MW01-ES61
16 0 ΤΑ , A Vv° Ο X V 0 MW01-ES75
17 0 ϊ MW01-ES81
“'M'S ff.
” IS ό
ο κ„ X ν ΐί α νΧΑΑοη MW01-ES91
χ 0
20 MW0.M-04-L-D04
*Μγύ *'Άα
23 χ~χ MW01~l~15-L-H07
/ Ά ) ' /^\ /~~~ζ Η·™7 4 #—(\ //™NH ^-7 ---7 Ν_Ν·
24 MW01-1-16-L-F0S
ΑίΧ WH*
MW01-1-18-L-B09
Figure AU2016203312B2_D0019
Ν'
2016203312 20 May 2016
Figure AU2016203312B2_D0020
2016203312 20 May 201 \
/=f
V/ \
ΛΕ— λί \=Ζ
A
V?
\ \
/”
Figure AU2016203312B2_D0021
/ \\
Figure AU2016203312B2_D0022
Ν—Ν ζ \ // % Λ Α~~\ ν—Ν ν
MW01-5-I.44C.~Z
MWQI-5-144D-Z .MW01-5-145A~Z
MWG1-5-IS9Z
Ηε,Ν”™^
Ν—Ν
MW01-5-202R-Z
Figure AU2016203312B2_D0023
Ό
Ο
CO ο
co
Figure AU2016203312B2_D0024
M4V01-1-02-L-E08
MW01-1-03-L-D03
MW01-1-03-L-F03
MW01-1-03-L-GW
Β
Figure AU2016203312B2_D0025
2016203312 20 May 2016
Figure AU2016203312B2_D0026
2016203312 20 May 2016
Figure AU2016203312B2_D0027
ο co ο
CM
“]”MW0I-9-041Z
Figure AU2016203312B2_D0028
Ο
ΓΗ £
ο
ΓΗ
ΓΗ
ΓΊ
ΓΊ
Ο
ΓΗ
Ο
ΓΗ
Figure AU2016203312B2_D0029
2016203312 20 May 20
Figure AU2016203312B2_D0030
2016203312 08 Aug 2016
KS.
\
Figure AU2016203312B2_D0031
Figure AU2016203312B2_D0032
Figure AU2016203312B2_D0033
Figure AU2016203312B2_D0034
Figure AU2016203312B2_D0035
2016203312 20 May 2
101
Figure AU2016203312B2_D0036
2016203312 20 May 20
102
Figure AU2016203312B2_D0037
2016203312 20 May 20
103
205 HsC^-CHs kJ T H T r Y θΛ,-Ν MW01-1-18-L-B07
208 Z~~NZ αγ% L? MW01-L03-L-G03
210 0 N YYQ'J σνΝ MXV014-04~I.-C03
217 CHg Ov ) HN MW01-1-02-L-E03
218 CHg / s Hs%An XY-N MW014-02-L-E06
2016203312 20 May 20
104
Figure AU2016203312B2_D0038
Ό
Ο <Ν ο
105
229 α s ό MWO1-1-03-1/-1309
230 ,-ΟΗ ι Ίμη Η3<1νΛΝ Μ 0 MW01-1-03-L-B10 ..... ί
231 Br h3c^A Μ ό MW01-1-03-L-C03
233 A Q ΑΑ Κα Λ MW01-1-03-QC08
235 Η*Ν'ΝΗ Η3 MW01-1-03-L-E08
236 νη2 α A 1 0 J N j; A MWO1-1-03-L-EO9 ..............................................I
2016203312 20 May 20
106
240 ho^,ch3 I m MW0M-04-L-A06
242 XJ ό MW01-1-04-L-.D10
250 rO H3c Λν c5 MW01-1-05-L-B11
251 Η3θ^4Ν (Γί MWOM-OS-L-COZ
254 <j>-fo Ύ Ί v HgC. ή Xrx ; MW01-A05-L-G11
Ο
CO ο
co
107
255 g.CHg XHg ό MW01-1-05-L-H05
266 r0 j HN Η3%Αν ifi MW01-1-08-L-D09
268 X) CHgHNT SiS MW01-1-09-L-C06
270 ΐ FFl nh2 0 ό MWOl-l-OP-L-GOS
i 271 : ?HS θ'Υ^ hh2 Hi H 0 ά MWG1-1-09--1.-607
2016203312 20 May 2016
Figure AU2016203312B2_D0039
W'Oi-w-iXjO?
MWO1-1-09-L--H07 ilW01-i-i5-L-A04 j
i
MW01-1-15-L-BW
MW01-M5-I.Z-C04 ~1 MW0l-i-l5-L-D03 .109
2016203312 20 May 20
Figure AU2016203312B2_D0040
MWOl-MS-L-GlO
''xs MW01-1-17-L-A09
AUY014-17-L-A11
MWQ14-17-L-B02
Figure AU2016203312B2_D0041
2016203312 20 May 20
110
299 ύ 9Hs η V w MW01-1-18-VA09
308 /θ\ F i ’ T F H JyX θΧΝ MW01-2-O3-L-BO8
310 % 0 Ί h y jPVT<N MW01-2-03-L-C05
313 ^OH C } XX fj'X'N-* MW0I-2-03~L-G07
318 ,o, H -0 Q AyX Xn-n MWO1-2-10.1-L-H08
3.19 X, ϋ cooy. X JVX ipXA XX_ MW0.1-2~10-L-E05
so
2016203312 20 May 201 in_ __
| 320 0 Ci u ΊΨ i Η i p/V MW0i-2»10~L-E00
321 MWO1-2-20-L-BO2
323 NOg H A „N k ,0 u MW01-2-20-L-D05
324 i ; H ργΑΝ.Ν <,0 MW0i-2--20-L*E09 •
326 c? i 0 -A T H i Α'γ* S/A/ (jA MW01-2-25-L-H06
328 CH jA ΓηΑΑ θχχ MW01-3-01-L-G03
329 ?Hs rr m qV MW01-3-01-L-GG4
2016203312 20 May 201
112
331 ch3 Ό MWO1-3-O1-L-GO8
ΓΥ CHS 'x i 0 ch3 fT Sr*
332 HgGs^ 9¾ „/sOch3 A. kA* 1 pif ch« MW01-3-01-L-G09
a A'n
335 ΓΥ 'tTTTCZY Ό gh3 AsAcHs 0 VA3 b K4W01-3-06-L-E09
337 α Cr KrN N MW01-1-07-L-G07
339 ch3 MWOkl-lS-L-Cll
k rbA
h s k-N
W
340 CX H MWOI-i-lS-L-EOS
O' N
341 H H»CYiYN'NHi MW0M46-L-B11
2016203312 20 May 201
113
Figure AU2016203312B2_D0042
2016203312 20 May 20
Π4
361 HSC, O 0 Υχθ MW01-1-03-L-H08
w
s 362 CoS ό MWG1-1-01-L-HD4
363 0 γ 0 ΐί^ΝΗ X MWOl-l-Ol-L-HOe
366 H’%AH Μ X Γ ® V Ci MW9I4-03-L-EQ7
367 νο2 / ? ΗΐΚ, Μ1 MW01-1-05-L-E05
368 Ο Η^γ\Η MW01-1-03-L-B03
I zvS,
Ό
Ο
CM s
ο
Γ4 ns
Figure AU2016203312B2_D0043
gh3 X» pN' MWQ1~I~O1~L~G10
]WG1~14B-L-G1.X
2016203312 20 May 2016
116
Figure AU2016203312B2_D0044
2016203312 20 May 2016
117
Compound Number Compound Stracture Synthetic Cede
22 pHs A 4-ζ CHa N-N MW01A15AE03
26 A'\ A A /a %_2z jh~4 /} OH MW01-2-O2-B-K09
29 A Νζγί,Ν ,6L o hi A pi-p A A MW01-1-Q30A-LKM
30 Br r,v MW01-1-030B-LKM
32 Br ,, ί H h ί N N • o jL fj MW01-1-048ABLKM
2016203312 20 May 20
Figure AU2016203312B2_D0045
Figure AU2016203312B2_D0046
Figure AU2016203312B2_D0047
MW01.-2-O65LKM
MW01-2-127LKM
MW01-2-134LKM
MW01-2-I46LKM
2016203312 20 May 2016
119
! 37 0 1 ΝγΝ N Μ h2n Η 1 ki MW01-2-147TJSM 1
38 ch3 4 )—4 S~N ΜΗ w -ij„4 \..../ MWG1-1'-O2-L“B11
39 H3C-O pHg )-0 C jH /T~Gi v--z f-l-N MWOl-l-OAL-FW 1
4-2 H3C pHg ,O X-o-A HSC N“-\ ί ^1,,,··.^ λλλ**-. 'A I—z \ N~N MW01-2-33-L-A1X
45 W z/ y / y~NH V=/ N~N \ o Xwwwww M yr~-.--v MW01-1-17-L-E06
2016203312 20 May 20
Figure AU2016203312B2_D0048
2016203312 20 May 201
Figure AU2016203312B2_D0049
2016203312 20 May 20
122
69 OH c NH 1 ffri MW01-1-03-L-B08
72 urn „.Sr Λ i i 0 MW01-1-03-L-G09
87 Br ijX s^o γΑν ί'Ί MW01-1-08-L-E11
93 Ηϊ> HO \ Ν'- ,,/ Λ 0 HH MW01-M3-L-G06
2016203312 20 May 20
123
Figure AU2016203312B2_D0050
2016203312 20 May 201
124
124 MW01-1-17-L-W2
ΤΊ ΗΝ ί] V
126 HgC^N^ J ΗΝ V^N 0 MW014-17-VH07
128 α, r% V Λν <s w·· MWO1-1-18-L-A02
2016203312 20 May 2016
125
Figure AU2016203312B2_D0051
2016203312 20 May 20
126
148 kr- Jk. N' n η Cl U MWQ1--2477B~WH
153 0 ., Cl Η H i „ X. ι'Χχ N -v raX. 0 N fj N XH M ό MW01-2484WH
1SS ΝγΜ X o A 8 I .X H M O -¾^ MW0I-249IA-WH
156 S' θγΟ KNX \ NH Λ H if \1χθγ. N N Αγ; N o MWQ1-24.93B-WH 1
157 HO /7~4 X-, /~~\ (/ >~N N_^ j, MW01-3-003WH
160 / -N /=\ y--ξ N= (χ /7-C ί'Τ’Κ ,ΝΗχ $ MW01-3-019A-WH
Ό
Ο (Μ £
ο (Μ
1.27
Figure AU2016203312B2_D0052
MW0P3417WH
MWQ1-3-11SWH
Ν 'i—' MW0.1~3--183WH
MW01-2-03-L-G03
MWO1~2~03~L~CO4
Λ ΙΜ~~\ ^~ο?
MWO1-2-O34.X303
ρ·°
/--
Ο
CO
128
177 /01-2-21-L-F04 ο
co
178
MW0I-2-24-L-GQ9
ΪΪΪΑ
Figure AU2016203312B2_D0053
II
W”
Figure AU2016203312B2_D0054
X /Γ\ /^\ /™λ \ /—4· /“Κ ^”4 # \Z22222f Ε<ΐ™>ϊΐ5
Figure AU2016203312B2_D0055
ι
MW01-5-188WH
MW01-6-OO3WH
2016203312 20 May 20
129
191 AX AH Ubx MW01-6-046WH
γΊ
200 HO. XK3 MW01-1-01-L-C06
0
203 9H3 h O Z%AN'N '-<Xh MW01-2~Q3~L-D09
204 o AfYY^ ^p/ah '^ok MW01-1-01-L-B02
] rX
kJ 0
206 OH <L r-K.J MW01-2-03-L--D09
kJ J y hn
jS X^N
2016203312 20 May 20
MW01-2-03-L-G04
Figure AU2016203312B2_D0056
2016203312 20 May 2016
131
213 CHg SO K5GZXO'AY^^ MW0I-1-01-L-F02
214 oA o jA> Hs°A MW01-1-O1-L-FO3
215 jO A-^ Ai A^.-rA MW01-1-01-L-G08
216 OH r k ^,ΟΗ J G HN h3ckAn γ MW01-1-02-L-D11
219 γ3 ΝΗ?γ° HacA^J An fS _______ A;A _ MW0l~l~Q2-L--EG4 j
__ 132
2016203312 20 May 201
Figure AU2016203312B2_D0057
Figure AU2016203312B2_D0058
Figure AU2016203312B2_D0059
MW01-1-O2-L-FO9
MW01-I-03-VA04
Figure AU2016203312B2_D0060
Figure AU2016203312B2_D0061
2016203312 20 May 2016
Figure AU2016203312B2_D0062
2016203312 20 May 20
134
Figure AU2016203312B2_D0063
2016203312 20 May 20
135
Figure AU2016203312B2_D0064
Ό
Ο ο
cs [“255
136
ι.
MW01-
Figure AU2016203312B2_D0065
co
CD
CD
Ο <Ν
Ο (Μ
5 ηΧ η<Α Gn η XX
253 COOH J ΗΝ HOOC. Λν Αν ο XX MWOl-l-ib-L-GlO
256 ΟΗ 0=8=0 ΝΗΧ] Αν A S, .-X <> MW01-1-05-L-H07
257 Μ HN'NHa [i Μ χ^Ν ό MWO14-05-L-HO9
258 ΝΗ2 H3C%^AskrCHs Jj f An A> ii . i MW01.--1-OS-L-Hll
2016203312 20 May 20
Figure AU2016203312B2_D0066
2016203312 20 May 2016
138
Figure AU2016203312B2_D0067
2016203312 20 May 20
139
269 ί J - ..... .......... 1 nh2 ΧΆ N •^Χζ'γ-θχζ CH3 Lj 0 0 MW01-1-09-L-G04
273 A» A 1 AA 0 MW01-N09-L-G11
277 cs a CI'tA'N V A u MW0M-I5~L-B07
”279 QHg H?cA'A OH Jn A U MW01-1-15-L-B11
281 a ipYv^N aa AJ LA X 1^! M MW01-1-15-L-D02
282 ch3 ci i 1 κ3σ %A 0Ά MW0M-15-L-D03
2016203312 20 May 2016
140
j'’ 283 J N HSCVUK WO1445~L--E1Q
0
| 285 0 Cl MW01445-L-H09
,-A, H 1
w
286 ! 1 .-•'Ύ. -A O Hx xzA 1 MW014-16-L-E05
A
it A Y>'z
287 V'N Cl XAa %^ci MW0M-01-L-FU
288 Η Γ Π 04 MW01-1-17-L-B05
CAVN
290 9¾ xLyNHj, LA A> u Cl CHa V MW01-l~16-L-E08
2016203312 20 May 2016
141
291 q-V 3 Mi ->h3 MW01-1-16-L-GO7
297 A H MWG1-1-17-L-F03
AA < ηρ N k„.>
kA
300 fri MW01-1-1S-L-BO4
T
Ah Af
j AA'H kA
j 301 CHg X MWOM-18-L-B10
Ί η Π JyNAN'
0^n-n HSC J
302 Π MWOl-l-lS-L-Bll
HgCx^-CHg /x^k Ar-^AA
Ζ%Λκ.'Ν if η N
u
303 C! Γ Aa- MW01-1-18-L-CO5
θΑΐ'Ν HO' A L ί
2016203312 20 May 20
Figure AU2016203312B2_D0068
2016203312 20 May 201
143
Figure AU2016203312B2_D0069
2016203312 20 May 2016
330 144 ch3 i CHS u MW01-3-Q1-L-GQ5
333 ch3 kv* OH MW01-3-06-L-B07
334 CK3 /zk. NH2 XV (V ™ MWQ1-3-O6-L-B08
336 Η3οχν^γΝ·-/ ζκ Λ N A-rVCHa ipV^hV cf 0 MW01-1-07-L-G07
338 N Y^Xx'CHg x^v^kx^ vS u u MWO1-1-O8-L-DO3
342 X u u I H i ΧγΧ ¢) MW01-1-10-L-E09
343 X ’ Y r ϋ v%AxN JM...................................... MWOl-l-n-L-CGS
2016203312 20 May 20
145
Figure AU2016203312B2_D0070
Figure AU2016203312B2_D0071
2016203312 20 May 201
Figure AU2016203312B2_D0072
TMWOV2~24-L-E07
MW01-O1-01-L-B07
MW01-7-O84WH
MW01-7-085WH
2016203312 20 May 20
Figure AU2016203312B2_D0073
Ο
CM
149
2016203312 20 May
Figure AU2016203312B2_D0074
MW01-6127WH
Figure AU2016203312B2_D0075
Figure AU2016203312B2_D0076
MW01-6189WH
Figure AU2016203312B2_D0077
MWG1-7107 WH
2016203312 20 May 201
ISO
Figure AU2016203312B2_D0078
2016203312 20 May 201
Figure AU2016203312B2_D0079
2016203312 20 May 20
152
Figure AU2016203312B2_D0080
2016203312 20 May 2016
153
Figure AU2016203312B2_D0081
2016203312 20 May 201
Figure AU2016203312B2_D0082
2016203312 20 May 2016
155
Figure AU2016203312B2_D0083
2016203312 20 May 2016
156
Figure AU2016203312B2_D0084
2016203312 20 May 2016
157
I s
!
I
Figure AU2016203312B2_D0085
MWOVQ2-034.-GM
Figure AU2016203312B2_D0086
Figure AU2016203312B2_D0087
so o
CT
2016203312 20 May
158
Figure AU2016203312B2_D0088
2016203312 20 May 2016
159
Figure AU2016203312B2_D0089
2016203312 20 May 2016
160 / y—N ?
\«««κΖ V»—^z
Figure AU2016203312B2_D0090
Ό
Ο
CM ο
Γ4
2016203
161
Figure AU2016203312B2_D0091
<0
Ο
CM
2016203312 20 May
162
Figure AU2016203312B2_D0092
2016203312 20 May 2016
103
Figure AU2016203312B2_D0093
Ό
Ο
CM
164
2016203312 20 May
Table 5
Figure AU2016203312B2_D0094
2016203312 20 May 2016
165
Figure AU2016203312B2_D0095
Figure AU2016203312B2_D0096
O
C4
2016203312 20 May
Figure AU2016203312B2_D0097
so ο
CT ο
CT
167
Figure AU2016203312B2_D0098
2016203312 20 May 2016
168
Figure AU2016203312B2_D0099
Figure AU2016203312B2_D0100
Figure AU2016203312B2_D0101
Figure AU2016203312B2_D0102
Ό
Ο
CM ο
Γ4
169
201620331
Figure AU2016203312B2_D0103
2016203312 20 May 2016
1.70
Figure AU2016203312B2_D0104
Figure AU2016203312B2_D0105
Figure AU2016203312B2_D0106
2016203312 20 May 2016
Figure AU2016203312B2_D0107
2016203312 20 May 2016
172
Figure AU2016203312B2_D0108
Figure AU2016203312B2_D0109
Figure AU2016203312B2_D0110
so o
CT
173
2016203312 20 May
Figure AU2016203312B2_D0111
2016203312 20 May 2016
174
Figure AU2016203312B2_D0112
2016203312 20 May 2016
175
Figure AU2016203312B2_D0113
2016203312 20 May 2016
Figure AU2016203312B2_D0114
2016203312 20 May 2016
Figure AU2016203312B2_D0115
2016203312 20 May 2016
178
Figure AU2016203312B2_D0116
2016203312 20 May 2016
179
Figure AU2016203312B2_D0117
Figure AU2016203312B2_D0118
Figure AU2016203312B2_D0119
J-
Figure AU2016203312B2_D0120
2016203312 20 May 2016
180
References [I] Akiyama H, Barger S, Bamatn S, BradtB, Bauer J, Cole GM, et al. Inflammation, and Alzheimer’s disease. Neurobiol Aging 21.: 383-421 (2000).
S [2] Mirzoeva S, Sawkar A, ZasadzJd M, Quo L, Velentza AV, Dunlap V, et al. discovery of a 3-amino-6-phenyl-py.ndazine derivative as a new synthetic anti-neuroinflammatory compound. J MedChem45:563-566(2002).
[3] Watterson DM, Haiech J and Van Eldik I J, Discovery of new chemical classes of synthetic ligands that suppress neuroinflammatory responses. J Mol Neurosci 19: 89-94 (2002), [4] Watterson DM, Velcntza AV, Zasadzfd M, Craft JM, Haiech J and Van Eldik LJ. 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: 4.11424(2003).
[5] Craft. JM, Watterson DM, Frautechy SA and Van Eldik LJ. Aminopyridazines inhibit βamyloid 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 murine model of 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 Alzheimer's 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 thschemical and pharmacological dissection of aminopyridazines. J Heterocyclic Chem 35: 1091-1100 (1998).
[9] Frautschy SA. Yang F, Calderon I. and Cols GM Rodent models of Alzheimer's disease: rat A β infusion approaches to amyloid deposits, Neurobiol Aging 17: 311-21 1996), [10] Verier DF, Johnson SR, Cheng HY, Smith BR, Ward KW and Kopple KD Molecular 30 properties that influence the oral bioavailability of drug candidates. I Med Chem 45: 2615-2623
Ό (2002).
[II] Vieth M, Siegel MG, Higgs RE, Watson ΪΑ, Robertson DH, Savin KA, et al (2004) Characteristic physical properties and structural fragments of marketed oral drugs. J Med Chem 47: 224-232 (2004).
2016203312 20 May 2016 [12] Cignarella G, Badocco D, Pinna G, Longa M, Curzu MM, Tofaneiti O, et ah Synthesis and biological evaluation of substituted benzo[A]cinnolinones and 3H-benzo[0,7]cyeIahepta[l,2c]pyridazinones: higher homologues of die antihypertensive and antithrombotic 5H“indeno[l,2c}pyridazinones. J Med Chem 32: 2277-2282 (1989).
[13} Costantino L, Rastelli G, Yeseoyiui 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: 4396-44-05 (1996).
[14] Sotelo E and Ravina E. Efficient aromatization of 4,5-dihydro-3-(2H)-pyridazinones substituted at 5 position by using anhydrous copper (Π) chloride. Synthetic Communications 30: 1-7 (2( [15} Wemsuth CO, Bourguignon JJ, Schlewer G. Gies JP, Schoenfeider A, Meiikiau A, et al. Synthesis and structure-activity relationships of a series of aminopyridazwe derivatives ot yaminobutyric acid acting as selective GABAj, antagonists. J Med Chem 30: 239-249 (1987).
[16] Wennuth 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 Eidik IJ Screening in a cell-based assay for inhibitors of microglial nitric oxide production reveals calmoduiinregulated protein kinases as potential drug discovery targets. Brain Res 844: 126-134- (1999).
[18] Coudert, P,; Couquelet, J.; Trmche, P. A new synthetic route to 4}6-diaxy1pyridazinone3 and some of their derivatives. Journal of Heterocyclic Chemistry, 1988,25(3), 799-802,
2016203312 03 May 2018

Claims (22)

  1. THE CLAIMS DEFINING THE INVENTION ARE AS FOLLOWS:1. A compound of the Formula II:
    wherein R10 and R11 are independently hydrogen, hydroxyl, alkyl, alkenyl, alkynyl, alkylene, alkenylene, alkoxy, alkenyloxy, cycloalkyl, cycloalkenyi, aryl, aryloxy, arylalkoxy, aroyl, heteroaryl, heterocyclic, acyl, acyloxy, sulfonyl, sulfinyl, sulfenyl, amino, imino, azido, thiol, thioalkyl, thioalkoxy, thioaryi, nitro, ureido, cyano, halo, silyi, silyioxy, silylalkyl, silylthio, =0, =S, carboxyl, carbonyl carbamoyl, or carboxamide; or a pharmaceutically acceptable salt thereof.
  2. 2. A compound of claim 1, wherein R10 is hydrogen and R11 is an unsaturated 5 to 6 membered heteromonocyclic group containing 1 to 4 nitrogen atoms.
  3. 3. A compound according to claim 2, wherein R11 is pyrrolyl; pyrrolinyl, imidazoiyl, pyrazolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, pyridinyi, pyrimidinyl, pyrazinyl, pyridazinyl, triazolyl or tetrazolyl.
  4. 4. A compound of claim 1, wherein R11 is not an unsaturated 5 to 6 membered heteromonocyclic group containing 1 to 4 nitrogen atoms.
  5. 5. An isolated and substantially pure compound as defined in any one of claims 1 to 4.
    01/05/18
    2016203312 03 May 2018
    183
  6. 6. A prodrug comprising a compound as defined in any one of claims 1 to 5 wherein the compound comprises one or more cieavabfe groups that are cleaved after administration to a subject to provide a therapeutically effective amount of the compound,
  7. 7. A pharmaceutical composition comprising a compound as defined in any one of claims 1 to 5 or a prodrug of claim 6, and a pharmaceutically acceptable carrier, excipient, or vehicle.
  8. 8. A method for treating an inflammatory disease or a disease involving or characterized by inflammation in a subject, comprising administering to the subject a therapeutically effective amount of a compound of any one of claims
    1 to 5, a prodrug of claim 6 or a pharmaceutical composition of claim 7,
  9. 9. A method for treating a disease requiring modulation of one or more of inflammation, signalling pathways involved in inflammation, cell signalling 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 therapeutically effective amount of a compound of any one of claims 1 to 5, a prodrug of claim 6 or a pharmaceutical composition of claim 7.
  10. 10. A method for treating in a subject a condition associated with neuroinflammation that can be decreased or inhibited with a compound of the Formula II according to any one of claims 1 to 5, comprising administering to
    01/05/18
    2016203312 03 May 2018
    184 the subject a therapeutically effective amount of a compound of any one of claims 1 to 5, a prodrug of claim 6 or a pharmaceutical composition of claim 7.
  11. 11. The method of claim 10, wherein said compound reduces neuroinflammation associated with any one of traumatic brain injury, intracerebral haemorrhage, or Alzheimer’s disease.
  12. 12. A method for reducing or inhibiting kinase activity, glial activation, neuronal cel! damage, and/or neuronal cell death in a subject, comprising administering to the subject a therapeutically effective amount of a compound according to any one of claims 1 to 5, a prodrug of claim 6 or a pharmaceutical composition of claim 7.
  13. 13. A method of inhibiting cell signalling molecule production, comprising administering compositions comprising one or more compounds according to any one of claims 1 to 5, a prodrug of claim 6 or a pharmaceutical composition of claim 7.
  14. 14. The method of claim 9, wherein the signalling molecule is IL- 1β and/or TNFa.
  15. 15. 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 any one of claims 1 to 5, a prodrug of claim 6 or a pharmaceutical composition of claim 7.
    01/05/18
    2016203312 03 May 2018
    185
  16. 16. A method for ameliorating progression of an inflammatory or a neuroinflammatory disease or obtaining a less severe stage of a disease in a subject suffering from such disease, comprising administering to the subject a therapeutically effective amount of a compound according to any one of claims 1 to 5, a prodrug of claim 6 or a pharmaceutical composition of claim 7,
  17. 17. The method of any one of claims 8 to 16, wherein the disease is a dementing disorder, a neurodegenerative disorder, a CNS demyelinating disorder, an autoimmune disorder, or a peripheral inflammatory disease.
  18. 18. The method according to any one of claims 8 to 17, wherein the disease is Alzheimer's disease.
  19. 19. A method of increasing survival of a subject suffering from Alzheimer's disease, comprising administering to the subject a therapeutically effective amount of a compound of the Formula II according to any one of claims 1 to 5, a prodrug of claim 6 or a pharmaceutical composition of claim 7,
  20. 20. A method for treating mild cognitive impairment (MCI) in a subject, comprising administering to the subject a therapeutically effective amount of a compound of any one of claims 1 to 5, a prodrug of claim 6 or a pharmaceutical composition of claim 7.
  21. 21. Use of a compound according to any one of claims 1 to 5, a prodrug of claim 6 or a pharmaceutical composition of claim 7 for the preparation of a medicament for treating an inflammatory or a neuroinflammatory disease.
    01/05/18
    2016203312 03 May 2018
    186
  22. 22, A kit comprising compound according to any one of claims 1 to 5, a prodrug of claim 6 or a pharmaceutical composition of claim 7 for preventing and/or treating an inflammatory or a neuroinflammatory disease, a container, and instructions for use.
AU2016203312A 2004-11-02 2016-05-20 Pyridazine compounds, compositions and methods Ceased AU2016203312B2 (en)

Priority Applications (1)

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

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US60/624,346 2004-11-02
US60/723,124 2005-10-03
AU2005302225A AU2005302225B2 (en) 2004-11-02 2005-11-02 Pyridazine compounds, compositions and methods
US60/723,090 2006-11-01
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

Related Parent Applications (1)

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

Publications (2)

Publication Number Publication Date
AU2016203312A1 AU2016203312A1 (en) 2016-06-09
AU2016203312B2 true AU2016203312B2 (en) 2018-05-24

Family

ID=56116604

Family Applications (1)

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

Country Status (1)

Country Link
AU (1) AU2016203312B2 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0211457A2 (en) * 1985-07-31 1987-02-25 Janssen Pharmaceutica N.V. Novel (4-substituted-piperazinyl)pyridazines
US5484940A (en) * 1994-11-28 1996-01-16 Grant; Francine S. Substituted 3-indolyl-5-pyrazolone compounds
WO2001042241A1 (en) * 1999-12-13 2001-06-14 Merck & Co., Inc. Substituted pyridazines having cytokine inhibitory activity

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0211457A2 (en) * 1985-07-31 1987-02-25 Janssen Pharmaceutica N.V. Novel (4-substituted-piperazinyl)pyridazines
US5484940A (en) * 1994-11-28 1996-01-16 Grant; Francine S. Substituted 3-indolyl-5-pyrazolone compounds
WO2001042241A1 (en) * 1999-12-13 2001-06-14 Merck & Co., Inc. Substituted pyridazines having cytokine inhibitory activity

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
ALLEN, C.F.H. et al, Journal of the American Chemical Society, 1951, vol. 73, pp. 5850-5856 *
CONSTANTINO, L. et al, Il Farmaco, 2000, vol. 55, pp. 544-552 *
GARCIA MERA, G. et al, Anales de Quimica Serie C – Quimica Organica y Bioquimica, 1985, vol. 81, pp. 280-283 *
HOLAVA, H.M. et al, Journal of Medicinal Chemistry, 1971, vol. 14, pp. 262-264 *
LAMANT, M., Compte Rendu Hebdomadaires des Seances de l’Academie des Science, 1973, vol. 277, pp. 319-322 *
SOTELO, E. et al, Synthetic Communications, 2000, vol. 30, pp. 1-7 *
VELENTZA, A.V. et al, Bioorganic & Medicinal Chemistry, 2003, vol. 13, pp. 3465-3470 *
WATTERSON, D.M. et al, Journal of Molecular Science, 2003, vol. 20, pp. 411-423 *

Also Published As

Publication number Publication date
AU2016203312A1 (en) 2016-06-09

Similar Documents

Publication Publication Date Title
CA2589106C (en) Pyridazine compounds, compositions and methods and their use in treating inflammatory diseases
US9408845B2 (en) Formulations containing pyridazine compounds
CA2650711A1 (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
WO2007127448A2 (en) Salts of pyridazine compounds
AU2016203312B2 (en) Pyridazine compounds, compositions and methods
AU2012216322A1 (en) Pyridazine compounds, compositions and methods
TWI725041B (en) Chk1/2 inhibitors for use in the treatment of neuroblastomas and/or soft tissue sarcomas
WO2024107859A1 (en) Cyclopentene-oxymethylene phosphonamidates and related compounds and their use in treating medical conditions

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)
MK14 Patent ceased section 143(a) (annual fees not paid) or expired