AU2008200380A1 - Use of GAL3 receptor antagonists for the treatment of depression and/or anxiety and compounds useful in such methods - Google Patents

Use of GAL3 receptor antagonists for the treatment of depression and/or anxiety and compounds useful in such methods Download PDF

Info

Publication number
AU2008200380A1
AU2008200380A1 AU2008200380A AU2008200380A AU2008200380A1 AU 2008200380 A1 AU2008200380 A1 AU 2008200380A1 AU 2008200380 A AU2008200380 A AU 2008200380A AU 2008200380 A AU2008200380 A AU 2008200380A AU 2008200380 A1 AU2008200380 A1 AU 2008200380A1
Authority
AU
Australia
Prior art keywords
branched
straight chained
alkyl
aryl
heteroaryl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
AU2008200380A
Inventor
Thomas P Blackburn
Lakmal W Boteju
Heidi Chen
Hermo Jimenez
Michael Konkel
Bharat Lagu
Kitae Lim
Stewart Noble
Mathivanan Packiarajan
Ian Jamie Talisman
John M Wetzel
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.)
H Lundbeck AS
Original Assignee
H Lundbeck AS
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 AU2002247149A external-priority patent/AU2002247149B2/en
Application filed by H Lundbeck AS filed Critical H Lundbeck AS
Priority to AU2008200380A priority Critical patent/AU2008200380A1/en
Publication of AU2008200380A1 publication Critical patent/AU2008200380A1/en
Abandoned legal-status Critical Current

Links

Description

Australian Patents Act 1990 Regulation 3.2 00 ORIGINAL COMPLETE SPECIFICATION STANDARD PATENT 00 00 N Invention Title "Use of GAL3 receptor antagonists for the treatment of depression and/or anxiety and compounds useful in such methods" The following statement is a full description of this invention, including the best method of performing it known to us:- QA0perKBMUOO8\Jn\4l0127432 Div Filingdoc 00 C Use of GAL3 Receptor Antagonists For The Treatment Of ¢c Depression And/Or Anxiety And Compounds Useful in Such Methods This application is a divisional of Australian Patent 00 Application No. 2002247149, the entire contents of which are incorporated herein by reference.
c 5 Background of the Invention 0 Throughout this application, various publications are referenced in parentheses by author and year. Full citations for these references may be found at the end of the specification immediately preceding the claims. The disclosures of these publications in their entireties are hereby incorporated by reference into this application to describe more fully the art to which this invention.
pertains.
Depression is the most common of mental disorders and yet is often underdiagnosed and undertreated, inflicting substantial morbidity and psychosocial impairment on its sufferers. Depression is mainly characterized by sadness, flatness, loss of feeling, anhedonia (lack of pleasure), tearfulness, agitation or retardation, thoughts of guilt, and worthlessness; in severe cases, suicide, hallucinations and delusions.
Depression can be mainly categorized into bipolar disorders, identifying wide swings of mood; major depressive illness, marked by severe depressive symptoms but without manic swings; and less defined milder forms of bipolar and major depression that fall short of the specific diagnostic criteria e.g. dysthymic disorder (formerly called depressive neurosis). The symptomatology and diagnostic criteria for depression are 00 2 set out in the DSMIV guidelines (American Psychiatric Association (1994) Diagnostic and Statistical Manual of Mental Disorders). Although many patients have single episodes of major depressive illness, the condition also can be repetitive, and this recurrent condition is 00 rnfrequently called unipolar depressive illness.
00 The key features of depressive illness are a markedly gloomy mood in which there is a loss of interest in life, and general feelings of hopelessness and worthlessness.
Depressive symptoms range in severity from mild mood swings to severe delusions about self-worth, accomplishments, and the future.
The "blackness" of the presentation in the depressed patient is most often accompanied by severe motor retardation with profound sleep and appetite disturbance and suicidal ideation. Furthermore, depressive illness can also present in a highly anxious or agitated state.
The degree to which the underlying brain mechanisms in anxiety and depression differ or overlap remains unknown.
The fact, however, that to some extent the same neurotransmitter systems are involved in depression and anxiety does not mean that the mechanisms are identical.
However, the majority of people in an episode of either depression or anxiety also meet criteria for at least one other psychiatric disorder. But by far the strongest comorbidities in both cases are between depression and anxiety disorders. Therefore, it is now becoming common clinical practice to treat both indications with antidepressants such as SSRIs.
00 3
OC
The key clinical features of anxiety disorders relate to CI various combinations of psychological and physical manifestations of anxiety, not attributable to real O 5 danger and occurring either in attacks (panic disorder 00 SPD) or as a persisting state (generalized anxiety disorder -GAD) Other neurotic features may be present 00 (obsessional or hysterical symptoms) but do not dominate Sthe clinical picture.
C- The Pathophysiology of Depression Theories underlying the pathophysiology of depression have developed from several lines of evidence including: 1) changes in neurotransmitter monoamine levels; 2)endocrine imbalance; and 3) electrophysiological studies on sleep functions.
Evidence implicating the role of neurotransmitters in depression, in particular the monoamines serotonin, noradrenaline and dopamine, include the success of pharmacological agents in treating depressive disorders.
Many of the tricylic antidepressants (TCAs), selective serotonin re-uptake inhibitors (SSRIs) and monoamine oxidase inhibitors (MAOIs) effective in the treatment of depression increase the availability of the catecholamines (noradrenaline and dopamine) and indolamines (serotonin) in the central nervous system (CNS). The clinical efficacy of these agents has given rise to the catecholamine-indolamine hypothesis of depression. This theory postulates that a certain level of amines and/or receptor sensitivity to catecholamines functions to generate a normal mood. A receptor 4 0C Sinsensitivity, a depletion of monoamines, or a decrease in their release, synthesis or storage have been CI postulated to lead to depression.
0 5 Current Treatments for Depression C A variety of pharmacological agents have been employed to Streat depression based on the catecholamine-indolamine 00 hypothesis of depression. Drugs used to treat depression include MAOIs, atypical antipsychotics, lithium, TCAs, and SSRIs. In addition, a number of off-label agents such as antiepileptics are used to treat depression in treatment-resistant patients.
Tricyclic antidepressants are about equal to SSRIs in effectiveness against depression thus providing supporting evidence for the catecholamine-indolamine hypothesis of depression. However, SSRIs have largely displaced TCAs because of side effects associated with TCAs and the need to monitor EKG and plasma drug concentration. Although the SSRIs are viewed as an improvement over other antidepressants, they are not without their clinical problems. Adverse effects on sexual function, primarily anorgasmia and delayed ejaculation, have been consistently reported. Other, common side-effects include sleep disorders, yawning, weight changes, suicidal ideation and extrapyramidal-like side-effects such as dystonic reactions. Thus, there clearly remains a medical need for new treatments of depression, without the adverse side-effect profile of existing agents and with improved efficacy.
00 SCurrent treatments for anxiety CM There is now considerable direct evidence for the efficacy of the SSRIs both in depression and in anxiety O 5 disorders.
oC SOf the current SSRIs approved for marketing in the USA 00 all have shown sufficient efficacy to be further approved Sfor the treatment of at least one anxiety disorder, for example; obsessive compulsive disorder (OCD) and generalized anxiety disorder (GAD). Compounds such as paroxetine and sertraline are also indicated for the treatment of panic disorder (PD).
However, it is clear from the issues raised earlier relating to the efficacy and side- effect profile of' SSRIs and for that matter the more widely prescribed benzodiazapines, there still exists a real medical need for novel approaches for the treatment of anxiety and depression.
Discovery Of GAL3 Receptor Subtype And Its Role In Depression and Anxiety The investigations leading to the present invention arose from the discovery that mRNA for the GAL3 receptor is localized to areas of the rat brain associated with mood and emotion (see PCT International Publication No. WO 98/15570, published April 16, 1998), thus supporting the expression of GAL3 in those regions. Protein for the GAL3 receptor is also shown to localize to areas of the rat brain associated with mood and emotion (see Table 11 and discussion herein).
00 6 This discovery led to the hypothesis that the GAL3 receptor may play a role in controlling the activity of catecholamine and indolamine neurons in the CNS. Galanin 00 5 is known to hyperpolarize neurons, including Smonoaminergic neurons (Seutin, et al., 1989) and to have inhibitory effects on 5-HT neurons (Xu, et al., 1998), 00 and dopamine neurons (Gopalan, et al., 1993; De Weille, et al., 1989; Jansson, et al., 1989; Nordstrom, et al., 1987; Weiss, et al., 1998). In light of these reports, a series of in vivo behavioral experiments were carried out to evaluate the antidepressant properties of a selective GAL3 receptor antagonist. The rat Forced Swim Test and the rat Social Interaction Test were employed to evaluate the use of selective GAL3 receptor antagonists to treat depression and anxiety. These models are considered by experts in the field to reflect the potential of agents to treat depression and anxiety.
Rat Forced Swim Test (FST) The rat Forced Swim Test (FST) is a behavioral test that is used to screen compounds for antidepressant efficacy (Porsolt et al., 1977, 1978; Porsolt, 1981). This test is widely used as it is reliable across laboratories, relatively easy to perform and is sensitive to the effects of some of the major classes of antidepressant drugs, including TCAs and MAOIs, and various atypical antidepressants. Furthermore, this test is relatively selective for antidepressant drugs, as few psychoactive drugs produce similar behavioral actions in the FST.
In the rat FST, animals are placed in a cylinder of 00 7 water, from which there is no escape, for an extended Speriod of time. Typically, animals will display a range CA of behaviors such as immobility, climbing, swimming, and diving, with immobility being predominant after several 0 5 minutes of immersion in the water. Consequently, many 00 CMc past studies have only measured or scored immobility Safter the administration of the test agent.
00 Unfortunately, this method does not score any other Sactive behaviors that may be produced by potential antidepressants. Thus, if a particular class of antidepressant were to have very little effect on immobility, yet produce characteristic behaviors during the FST, these behaviors would not be scored and the conclusion would be that the compound in question does not possess antidepressant action.
Recently, however, a sampling technique was developed to score active behaviors in the FST, such as swimming, climbing and diving, in addition to immobility (Detke, et al., 1995; Lucki, 1997; Page, et al., 1999; Reneric and Lucki, 1998). This modified sampling technique has indicated that SSRIs, such as fluoxetine, paroxetine and sertraline, significantly decrease immobility and increase swimming time (Detke, et al., 1995; Page, et al., 1999). In contrast, selective reuptake inhibitors of norepinephrine (NE) increase climbing behavior but do not alter swimming time (Detke, et al., 1995; Page, et al., 1999).
Rat Social Interaction Test (SIT) There are a number of paradigms that have been used to determine whether a compound possesses anxiolytic action.
8
OC
SA number of these tests involve food or water Sdeprivation, punishment or measurement of consummatory C( behavior (see File, et al., 1980; File, 1985; Rodgers, et al., 1997; and Treit, 1985, for review). In addition, in 00 5 these models, prior conditioning reduces the uncertainty M or anxiety. In general, these tests lack ethological 0 validity.
O<
00 SOne model that is based upon an unconditioned response that does not involve punishment or deprivation is the Social Interaction Test (SIT) (File and Hyde, 1978, 1979). In this model, rats previously housed singly are placed in a familiar, dimly lit, test arena with weightmatched, novel partners. The principal anxiogenic stimulus under these conditions is the partner novelty, which involves an unconditioned response to a potential threat. After pharmacological treatments, the following behaviors are scored as active social interaction: grooming, sniffing, biting, boxing, wrestling, following, crawling over and crawling under. A wide range of psychoactive drugs have been examined in this paradigm and it has been shown that the social interaction test can distinguish anxiolytics from antidepressants, antipsychotics, analeptics and sedative agents (File, 1985; Guy and Gardner, 1985). This test can detect anxiolytic agents such as the benzodiazepines (File and Hyde, 1978; File and Hyde, 1979; File, 1980), in addition to non-benzodiazepines, including paroxetine and other SSRIs (Lightowler, et al., 1994). Finally, the social interaction test can detect anxiogenic agents, including the inverse benzodiazepine receptor agonists (File, et al., 1982; File and Pellow, 1983; File and Pellow, 1984; 00 9 File, 1985) C< In an embodiment of the present invention the synthesis of novel pyrimidines which bind selectively to the cloned 0 5 human GAL3 receptor, compared to other cloned human Goo M protein coupled receptors, as measured in in vitro Sassays, is disclosed. In a further embodiment of the 00 present invention the synthesis of indolones which bind 0 selectively to the cloned human GAL3 receptor, compared to other cloned human G-protein coupled receptors, as measured in in vitro assays, is disclosed. The in vitro receptor assays described hereinafter were performed using various cultured cell lines, each transfected with and expressing only a single galanin-type receptor.
From the binding information described hereinafter, it has unexpectedly been discovered that compounds which are specific for the human GAL3 receptor with a binding affinity greater than ten-fold higher than the binding affinity with which the compounds bind to a human GAL1 receptor are effective in animal models of depression and anxiety which are predictive of efficacy in humans.
Thus, we demonstrate that the GAL3 receptor antagonists, which may be classified as neutral antagonists, inverse agonists or allosteric modulators, provide a novel method to treat depressive disorders and/or anxiety.
Summary of the Invention The present invention provides a method of treating a subject suffering from depression which comprises administering to the subject an amount of compound effective to treat the subject's depression wherein the compound has the structure: wherein W is H, -Cl, -Br, CN, methyl, ethyl, propyl, methoxy or ethoxy; wherein X is; NR 1
R
12
R
1 R 1 7
N
or wherein R11 is H, straight chained or branched C1-C, alkyl,
(CH
2 )q-O-(CH2)-CH3, aryl, or aryl (C 1
-C
6 )alkyl; 00 1 wherein R 12 is straight chained or branched Cl-C7 alkyl, (CH2)q-O(CH 2 )m-CH 3 or -(CH 2 )m-Z; wherein R 13 is a bicyclic alkyl ring system, adamantyl, 00 noradamantyl, C 3
-C
10 cyclcalkyl, heteroaryl, aryl, aryl(Cl- CO)alkyl, Q, or Q2; 00 wherein aryl may be substituted with one or more C-~ straight chained or branched alkyl, aryl, heteroaryl, or wherein Q, is
R
22 R2 R 2 2 R 2
R
0
R
2 2 202 wherein each J is independently 0, S, C(R 2 2 2 or NRA; 12 wherein R 4 is H; straight chained or branched Cl-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3
-C
7 cycloalkyl, C 5
-C
7 cycloalkenyl or aryl; wherein Y is NR 14
R
15 ;or u wherein R 14 is H, straight chained or branched Cl-C 6 alkyl,
(CH
2 )qOiCH 2 )m-CH3i C 3 -CG cycloalkyl, or (C(R 1 9 2 )m-Z; wherein R 15 is straight chained or branched C 3
-C
6 alkyl, (CO)q-0 -(CHO) CH 3
C
3
-C
6 cycloalkyl, (C (Rig) 2 (RIO) 2 or (C (Rig) 2) -Z; wherein R 16 is straight chained or branched Cl-C 7 alkyl, straight chained or branched Cl-C 7 monof luoroalkyl, 00 13 stagt cand or bace IC plfurakl straight chained or branched
C-C
7 olyfluosrakyht chained or branched
C
2
-C
7 alkynyl,
CS-C
7 cycloalkenyl,
(CH
2 or (CH 2 )q-01(CH2)m-CH3; 00 M wherein each R 1 7 is independently H; -OR 2 1 -0C0R 21 -C0R 2 1
-NCOR
21
-N(R
2 1 )2 -CON(R 21 2 -C00R 21 straight chained or 00 branched Cl-C 7 alkyl, straight chained or branched Cl-C 7 monofluoroalkyl, straight chained or branched
C-
1
-C
7 (110 polyfluoroalkyl, straight chained or branched
C
2 -C7 alkenyl, straight chained or branched
C
2
-C
7 alkynyl, c 5
-C
7 cycloalkenyl,
-(CH
2 or (CH 2
(CH
2 )m-CH 3 wherein
R
18 is straight chained or branched
C
1
-C
6 alkyl,
(CH
2 or (CH2)q-O
(CH
2 )m-CH 3 wherein each Rig is independently H, or straight chained or branched Cl-C 6 alkyl; wherein each R 20 is independently straight chained or branched Cl-C 7 alkyl, monofluoroalkYl or polyfluoroalkYl; straight chained or branched
C
2
-C
7 alkenyl or alkynyl;
C
3
C
7 cycloalkyl or C5-C 7 cycloalkenyl; -C1, -Br, or -I;
-NO
2
-N
3 -CM; OR 21 -0C0R 21 -C0R 21
-NCOR
21
-N(R
21 2
CON(R
21 2 or -COOR 2 i1; aryl or heteroaryl; or two R 2 0 groups present on adjacent carbon atoms can join together to form a methylenedioxy group; wherein each R21 is independently straight chained or branched Cl-C 7 alkyl, monofluoroalkYl. or polyfluoroalkyl; straight chained or branched
C
2
-C
7 alkenyl or alkynyl;
C.
3
C
7 cycloalkyl,
CS-C
7 cycloalkenyl, aryl, or aryl(Cl- 00 C14
C
6 alkyl; n wherein each R22 is independently H, F, C1 or Ci-C 4 straight chained or branched alkyl; OO wherein each m is an integer from 0 to 4 inclusive; C( wherein each n is an integer from 1 to 4 inclusive; 00 CI 10 wherein p is an integer from 0 to 2 inclusive; wherein q is an integer from 2 to 4 inclusive; wherein t is 1 or 2; wherein U is 0, -NR 16 S, C(R 17 2 or -NSO 2
R
16 wherein Z is C 3 -Cio cycloalkyl,
C
4
-C
7 cyclic ether, C 4 -C7 cyclic thioether, aryl, or heteroaryl; or a pharmaceutically acceptable salt thereof.
The present invention provides a method of treating a subject suffering from depression which comprises administering to the subject an amount of compound effective to treat the subject's depression wherein the compound has the structure: wherein W is H, -Cl, -Br, CN, methyl, ethyl, propyl, methoxy or ethoxy; S wherein X is NR 11
R
12
R
17
R
17
N
R
1 17
R
17 or wherein R 11 is H, straight chained or branched Cl-C 7 alkyl,
(CH
2 )qO-(CH 2 )m-CH3, aryl or aryl(CI-CG)alkyl; wherein R 12 is straight chained or branched
C
1
-C
7 alkyl,
(CH
2
(CH
2 )m-CH3, or (CH 2
)M-Z;
wherein R 13 is a bicyclic alkyl ring system, aryl "or aryl (Cl-C 6 alkyl; wherein Y is NR 14
R
1 5 00 16 R2~ -N Ip ;or 00R2
-NZ-
wherein R 14 is H, straight chained or branched Cl-C 6 alkyl,
(CH
2 )9-O-(CH 2 )M-CH3, C 3 -C6 cycloalkyl, or (C(R 1 9 2 wherein Rls is straight chained or branched C 3
-C
6 alkyl,
(CH
2 )qOi(C 2
).-CH
3 i C 3
-C
6 cycloalkyl, or (C(R 19 2 wherein U is 0, -NR16, S, C(R 1 7 2 or -NS0 2
R
1 6 wherein Z is C 3 -Cl 0 cycloalkyl, aryl, or heteroaryl; wherein R 16 is straight chained or branched Cl-C 7 alkyl, straight chained or branched Cl-C 7 monofluoroalkyl, straight chained or branched Cl-C 7 polyfluoroalkyl, straight chained or branched
C
2
-C
7 alkenyl, straight chained or branched C 2
-C
7 alkynyl, C 5
-C
7 cycloalkenyl,
(CH
2 or
(CH
2 )m-CH 3 00 17 wherein each RI 7 is independently H; -OR 21
-OCOR
21
-COR
21
-NCOR
21
-N(R
21 2
-CON(R
21 2
-COOR
21 straight chained or branched Cl-C 7 alkyl, straight chained or branched Cl-C 7 monofluoroalkYl, straight chained or branched Cl-C 7 polyfluoroalkyl, straight chained or branched
C
2
-C
7 00 alkenyl, straight chained or branched C 2
-C
7 alkynyl, C 5
C.?
cycloalkenyl,
-(CH
2 or (CH 2 )n-OiCH 2 )m-CH 3 00 wherein R18 is straight chained or branched
CI.-C
6 alkyl, CK110
(CH
2 or (CH 2 )cj-O-(CH 2 )rn-CHa; wherein each R 19 is independently H, or straight chained or branched
C
1
-C
6 alkyl; wherein each R 2 0 is independently straight chained or branched Cl-C 7 alkyl, monofluoroalkYl or polyfluoroalkyl; straight chained or branched
C
2 -C7 alkenyl or alkynyl;
C
3 C7 cycloalkyl or Cs5-C 7 cycloalkenyl; -Cl, -Br, or -I;
-NO
2
-N
3 -CN; -0R 2 1, -OCOR 2 1, -C0R 2 1
-NCOR
2 1
-N(R
21 2
CON(R
2 1)2, or -COOR 2 1; aryl or heteroaryl; or two R 2 0 groups present on adjacent carbon atoms can join together to form a methylenedioxY group; wherein each R 21 is independently straight chained or branched Cl-C7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched
C
2 -C7 alkenyl or alkynyl;
C
3 C, cycloalkyl,
CS-C
7 cycloalkenyl, aryl or aryl(Cl- Cd)alkyl; wherein each m is an integer from 0 to 4 inclusive; wherein each n is an integer from 1 to 4 inclusive; 18 wherein p is an integer from 0 to 2 inclusive; wherein q is an integer from 2 to 4 inclusive; wherein t is 1 or 2; or a pharmaceutically acceptable salt thereof.
The present invention provides a method of treating a subject suffering from depression which comprises administering to the subject an amount of compound effective to treat the subject's depression wherein the compound has the structure:
X
W
H
wherein W is H, -Cl, -Br, CN, methyl, ethyl, propyl, methoxy or ethoxy; wherein X is N(CH 3 2 or 00 19
R
17 CI-N 0 00 M wherein R 13 is an aryl, adamantyl, noradamantyl, C 3
-C
10 0 cycloalkyl, heteroaryl, Q, or Q2; 005 wherein aryl. may be substituted with one or more Cl-CI 0 straight. chained or branched alkyl, aryl, heteroaryl, or N (R 19
Z;
wherein Q, is wherein 02 is
R
22 R 22
R
22 t
R
22
PR
20 wherein each JT is independently 0, S, C(R 2 2 2 or NR 4 00 wherein R 4 is straight chained or branched Cl-C7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight V) chained or branched C 2 -C7 alkenyl or alkynyl; C 3 -C7 cycloalkyl, CS-C, cycloalkeny). or aryl; 00 wherein Y is NR 14
R
15 (Ni
R
20 1N 17 /R 19 p o rt- R 1 7
-N
R
20 wherein
R
14 is H, straight chained or branched
C
1
-C
6 alkyl,
(CH
2 )q0I-CH2)m-CH3I
C
3 -C6 cycloalkyl, or (C(R 1 9 2 )m-Z; wherein
R:
15 is straight chained or branched
C
3 -Cs alkyl,
(CH
2 )q-O1CH2)m-CH3,
C
3
-C
6 cycloalkyl, or (C(R 1 9 2 )m-Z; wherein U is 0, -NR16, S, C(Rl 7 2 or -NS0 2
R
1 6 wherein Z is C 3
-C
10 cycloalkyl, aryl, or heteroaryl; 00 21 wherein R 16 is straight chained or branched CI-C 7 alkyl, straight chained or branched
C
1
-C
7 monofluoroalkyl, straight chained or branched Cl-C 7 polyfluoroalkyl, straight chained or branched
C
2
-C
7 alkenyl, straight 00 M chained or branched
C
2
-C
7 alkynyl,
CS-C
7 cycloalkenyl,
(CH
2 or (CH 2 )q0O(CH 2 ).CH3? 00 wherein each R 1 7 is independently H; -OR 21
-OCOR
21
-COR
2 3 1
-NCOR
21
-N(R
2 2 I -CON(R 21 2
-COOR
21 straight chained or branched Cl-C 7 alkyl, straight chained or branched CI-C 7 monofluoroalkyl, straight chained or branched C 1
I-C
7 polyfluoroalkyl, straight chained or branched C 2 -C7 alkenyl, straight chained or branched C 2
-C
7 alkynyl, C.
5
-C
7 1s cycloalkenyl,
(CH
2 or (CH 2 )n0 -(CH 2 )rn-CH 3 wherein R 18 is straight chained or branched
C
1
-C
6 alkyl, (CH2)MnZ, or (CH 2 )q-O1CH 2 )M-CH3; wherein each R 19 is independently H, or straight chained or branched C 1
-C
6 alkyl; wherein each R 20 is independently straight chained or branched Cl-C 1 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3
C
7 cycloalkyl or C 5
-C
7 cycloalkenyl; -C1, -Br, or -I;
-NO
2
-N
3 -CN; -OR 21 -0C0R 21 -C0R 21
-NCOR
21
-N(R
2 2 CON (R 21 2 or -C00R 21 aryl or heteroaryl; or two R 20 groups present on adjacent carbon atoms can join together to form a methylenedioxy group; wherein each R 2 1 is independently straight chained or 00 o 22 Sbranched Ci-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3 SC 7 cycloalkyl, C 5
-C
7 cycloalkenyl, aryl or aryl(C 1 C) alkyl;
S
00 wherein each R 22 is independently H, F, Cl or CI-C 4 0 straight chained or branched alkyl; 00 Swherein each m is an integer from 0 to 4 inclusive; wherein'each n is an integer from 1 to 4 inclusive; wherein p is an integer from 0 to 2 inclusive; wherein q is an integer from 2 to 4 inclusive; wherein t is 1 or 2; or a pharmaceutically acceptable salt thereof.
The present invention provides a method of treating a subject suffering from depression which comprises administering to the subject an amount of compound effective to treat the subject's depression wherein the compound has the structure: 00 23 wherein W is H, -Cl, -Br, CN, methyl, ethyl, propyl, methoxy or ethoxy; 00 M ~wherein X is N(CH 3 2 or 00
R.
-N 0 wherein
R
13 is a bicyclic alkyl ring system, aryl or aryl (C C 6 alkyl; wherein Y is NR 14
R
15 wherein
R
14 is H, straight chained or branched
C
1
-C
6 alkyl,
(CH
2 )qOiCH2).-CH3I
C
3
-C
6 cycloalkyl, or (C(Rlis) 2
)M-Z;
wherein R 15 is (C(Ri 9 2 wherein Z is C 3
-C
10 cycloalkyl, aryl, or heteroaryl; wherein
R
16 is straight chained or branched Cl-C 7 alkyl, straight chained or branched Cl-C7 monofluoroalkyl, straight chained or branched Cl-C7 polyfluoroalkyl, straight chained or branched
C
2 -C7 alkenyl, straight chained or branched
C
2
-C
7 alkynyl,
C
5
-C
7 cycloalkenyl,
(CH
2 or (CH 2 )q01-CH2),mCH3; wherein each R 17 is independently H; -OR 2 1
-OCOR
2 1, -COR21, 00 CD 24
CI
S-NCOR 21
-N(R
21 2 -CON(R21)2, -COOR 21 straight chained or branched C 1
-C
7 alkyl, straight chained or branched Ci-C 7 l monofluoroalkyl, straight chained or branched Ci-C7 polyfluoroalkyl, straight chained or branched C 2 -C7 alkenyl, straight chained or branched C 2
-C
7 alkynyl, C 5
-C
7 0 0 cycloalkenyl, -(CH 2 or (CH 2 )n-O-(CH 2 )m-CH 3 C< wherein each R 19 is independently H, or straight chained 00 Sor branched C 1
-C
6 alkyl; iC wherein'each
R
21 is independently straight chained or branched C 1
-C
7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3
C
7 cycloalkyl,
C
5 -C7 cycloalkenyl, aryl or aryl(C 1
C
6 )alkyl; wherein each m is an integer from 0 to 4 inclusive; wherein each n is an integer from 1 to 4 inclusive; wherein q is an integer from 2 to 4 inclusive; or a pharmaceutically acceptable salt thereof.
The present invention also provides a method of treating a subject suffering from anxiety which comprises administering to the subject an amount of compound effective to treat the subject's anxiety wherein the compound has the structure: 00 x N 7
R
13 00
YNN
00 wherein W is H, -Cl, -Br, CN, methyl, ethyl, CI propyl, methoxy or ethoxy; wherein X is; NR 11
R
12
R
17
R
17
R
17 -N N =0 or
R
1 7,
R
17 -N N-R 1 8 wherein
R
11 is H, straight chained or branched
C
1
-C
7 alkyl,
(CH
2 )q0- CH2)m-CH3, aryl, or aryl (Cl-C 6 alkyl; wherein R12 is straight chained or branched
C
1 -C7 alkyl,
(CH
2 q01 CH2)m-CH3, or wherein
R
13 is a bicyclic alkyl ring system, adamantyl, noradamantyl,
C
3
-C
1 O cycloalkyl, heteroaryl, aryl, aryl(Ci-
C
6 alkyl, Q, or Q2; wherein aryl may be substituted with one or more Cl-Cio straight chained or branched alkyl, aryl, heteroaryl, or N(Rg-Z wherein Q, is Nzz R22 I I wherein Q2 is wherein each J is independently 0, S, C(R 22 )2 or NR 4 wherein R4 is H; straight chained or branched Ci-C-1 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched
C
2
-C
7 alkenyl or alkynyl;
C
3
-C
7 cycloalkyl,
CS-C
7 cycloalkenyl or aryl; wherein Y is NR 14
R
1 5 00 27
R
20
R
17 I or -N u 00 R_1 00
-N
wherein R 14 is H, straight chained or branched Cj-C 6 alkyl,
(CH
2
(CH
2 )m-CH 3
C
3
-C
6 cycloalkyl, or (C (R 19 2 )mZ; wherein R 15 is straight chained or branched
C
3
-C
6 alkyl,
(CH
2 )q0O(CH2)m-CH3I
C
3
-C
6 cycloalkyl, (C (R19) 2
(R.
6 2 or (C (R 19 2) -mZ; wherein R 16 is straight chained or branched Cl-C 7 alkyl, straight chained or branched Cl-C-1 monofluoroalkyl, straight chained or branched Cl-C,7 polyfluoroalkyl, straight chained or branched
C
2 -C7 alkenyl, straight chained or branched
C
2 -C7 alkynyl,
CS-C
7 cycloalkenyl,
(CH
2 or (CH2)q 0
-(CH
2 )m-CH 3 wherein each R 1 7 is independently H; -OR 2 1 -0C0R 21 -COR21, 00 0 28 -NCOR21, -N(R 2 2
,-CON(R
21 2 -C00R 21 straight chained or branched Cl-C 7 alkyl, straight chained or branched Cl-C7 monofluoroalkyl, straight chained or branched C3.- C7 polyfluoroalkyl, straight chained or branched C 2 -C7 aJlkenyl, straight chained or branched C 2 -C7 alkynyl, C 5 C7 00
(CH
2 or (CH 2
CCH
2 )m-CH 3 C-I wherein each R 19 is independently H, or straight chained or branched C 1
-C
6 alkyl; wherein each R 20 is independently straight chained or branched Cl-C7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or alkynyl; C 3
C
7 cycloalkyl or Cs-C,? cycloalkenyl; -C1, -Br, or -I;
-NO
2
-N
3 -CN; -OR 21 -0C0R 2 i, -C0R 21
-NCOR
21
-N(R
21 2
CON(R
21 or -C00R 21 aryl or heteroaryl; or two R 20 groups present on adjacent carbon atoms can join together to form a methylenedioxy group; wherein each R 21 is independently straight chained or branched CI-C7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or alkynyl; C 3 C7 cycloalkyl, CS-C7 cycloalkenyl, aryl, or aryl (C 1
C
6 alkyl; wherein each R 22 is independently H, F, Cl or Cl-C 4 straight chained or branched alkyl; wherein each m is an integer from 0 to 4 inclusive; 29 wherein each n is an integer from 1 to 4 inclusive; wherein p is an integer from 0 to 2 inclusive; wherein q is an integer from 2 to 4 inclusive; wherein t is 1 or 2; wherein U is O, -NR 16 S, C(R 17 2 or -NSO 2
R
16 wherein Z is C 3
-C
1 o cycloalkyl, C 4 -C7 cyclic ether, C 4
-C
7 cyclic thioether, aryl, or heteroaryl; or a pharmaceutically acceptable salt thereof.
The invention provides a method of treating a subject suffering from anxiety which comprises administering to the subject an amount of compound effective to treat the subject's anxiety wherein the compound has the structure:
X
W
N
H
wherein W is H, -Cl, -Br, CN, methyl, ethyl, propyl, methoxy or ethoxy; 00 wherein X is NRllRl 2
R
17 R17R 1 N or -N N-Re 00R1 00 wherein R 11 is H, straight chained or branched Cl-C-7 alkyl,
(CH
2 )q-O1CH2)m-CH3, aryl or aryl(C 1
-C
6 )alkyl; wherein R 12 is straight chained or branched Cl-C7 alkyl,
(CH
2 )q-OiCH2)rnCH3, or -(CH 2 )mZ; wherein R 13 is a bicyclic alkyl ring system, aryl or aryl (C 1
-C
6 alkyl; wherein Y is NR 14
R
1 5 00 31 c-I
R
2
R
17
R
2 0 j
-NUR
1 9 -p ;or 00N M 17>IT-< 00 R2
-N
wherein R 1 4 is H, straight chained or branched Cl-C 6 alkyl,
(CH
2 q0-CH2)mICH3,
C
3
-C
6 cycloalkyl, or (C (R 19 2 )mZ; wherein Ris is straight chained or branched C 3
-C
6 alkyl,
(CH
2 )q-0-(CH2)m.-C3,
C
3
-C
6 cycloalkyl, or (C (R 19 2 wherein U is 0, -NR, 6 S, C(Rl 7 2 or -NS0 2
R
16 s; wherein Z is C 3 -Cl 0 cycloalkyl, aryl, or heteroaryl; wherein Rls is straight chained or branched Cl-C7 alkyl, straight chained or branched Cl-C 7 monofluoroalkyl, straight chained or branched Cl-C 7 polyfluoroalkyl, straight chained or branched C 2
-C
7 alkenyl, straight chained or branched C 2
-C
7 alkynyl, C 5
-C
7 cycloalkenyl,
(CH
2 or (CH 2 )q-O-(CH2),-C 1 3; 00 32 wherein each R17 is independently H; -OR 21 -0COR 21 -COR21,
-NCOR
21
-N(R
21 2
-CON(R
21 2
-COOR
2 1 straight chained or branched C 1
-C
7 alkyl, straight chained or branched C 1
-C
7 monofluoroalkyl, straight chained or branched C 1
-C,
polyfluoroalkyl, straight chained or branched
C
2
-C
7 00 M alkenyl, straight chained or branched
C
2
-C
7 alkynyl, C 5
-C
7 cycloalkenyl,
-(CH
2 or (CH 2 )n-O-CH 2 )m-CH 3 00 wherein R 18 is straight chained or branched C 1
-C
6 alkyl, N 10 (CH 2 or (CH 2
(CH
2 )m-CH3; wherein each R 19 is independently H, or straight chained or branched C 1 alkyl; wherein each R 2 0 is independently straight chained or branched Cl-C, alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched
C
2
-C
7 alkenyl or alkynyl; C 3
C
7 cycloalkyl or Cs-C 7 cycloalkenyl; -C1, -Br, or -I;
-NO
2
-N
3 -CN; -OR 2 1
-OCOR
21 -COR21, -NCOR 2 1
-N(R
21 2
CON(R
2 1)2, or -COOR21; aryl or heteroaryl; or two R 2 0 groups present on adjacent carbon atoms can join together to form a methylenedioxy group; wherein each R 21 is independently straight chained or branched Cl-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched
C
2
-C
7 alkenyl or alkynyl; C 3
C
7 cycloalkyl,
C
5
-C
7 cycloalkenyl, aryl or aryl (C1- Cs) alkyl; wherein each m is an integer from 0 to 4 inclusive; wherein each n is an integer from 1 to 4 inclusive; 33 wherein p is an integer from 0 to 2 inclusive; wherein q is an integer from 2 to 4 inclusive; wherein t is 1 or 2; or a pharmaceutically acceptable salt thereof.
The invention provides a method of treating a subject suffering from anxiety which comprises administering to the subject an amount of compound effective to treat the subject's anxiety wherein the compound has the structure:
X
W
Y N N
H
wherein W is H, -C1, -Br, CN, methyl, ethyl, propyl, methoxy or ethoxy; wherein X is N(CH 3 2 or 34 wherein R 13 is an aryl, adamantyl, noradamantyl, C 3 -C1 0 cycloalkyl, heteroaryl, Q, or 0.2; wherein aryl may be substituted with one or more Cl-Cio straight chained or branched alkyl, aryl, heteroaryl, or N (Rig) Z; wherein Q, is wherein Q2 is wherein each J is independently 0, S, C(R 22 2 or NR 4 wherein R 4 is straight chained or branched C 1
-C
7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
1 alkenyl or alkynyl; C 3
-C
7 cycloalkyl,
CS-C
7 cycloalkenyl or aryl; wherein Y is NR 1 4
R
15
R
17 -N U p- 1 ;or wherein R 1 4 is H, straight chained or branched CI-CG alkyl, (C2)- 0
(CH
2 mCH 3
C
3
-C
6 cycloalkyl, or (C (Rig) 2
M-Z;
wherein R 15 is straight chained or branched C 3
-C
6 alkyl,
(CH
2 )qO(CH2)m-CH3, C 3
-C
6 i cycloalkyl, or (C(Ris) 2 wherein U is 0, -NR~s, S, C(R.
7 2 or -NS0 2 R2 1 6 wherein Z is C 3
-CI
0 cycloalkyl, aryl, or heteroaryl; wherein R 16 is straight chained or branched C 1
C
7 alkyl, straight chained or branched Cl-C 7 monofluoroalkyl, straight chained or branched Cl-C 7 polyfluoroalkyl, 00 36 straight chained or branched C 2 alkenyl, straight chained or branched C 2
-C
7 alkynyl, CS-C 7 cycloalkenyl, tn (CH 2 or (CH 2
(CH
2 )m,-CH 3 wherein each R 17 is independently H; -OR 21
-OCOR
21
-COR
21 00 -NCOR 21
-N(R
21 2
-CON(R
21 2
-COOR
21 straight chained or branched Cl-C 7 alkyl, straight chained or branched Cl-C 7 monofluoroalkyl, straight chained or branched CI.-C 7 00polyfluoroalkyl, straight chained or branched C 2
-C,
alkenyl, straight chained or branched C 2
-C
7 alkynyl, CS-C 7 cycloalkenyl, (CH 2 m-Z, or (CH 2 n-0- (CH 2 m-CH 3 wherein R1B is straight chained or branched C 1
-C
6 alkyl,
(CH
2 or (CH 2 )q-0I(CH 2 )m-CH3; wherein each R 19 is independently H, or straight chained or branched C 1
-C
6 alkyl; wherein each R 20 is independently straight chained or branched CI-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3
C
7 cycloalkyl or C 5
-C
7 cycloalkenyl; -C1, -Br, or -I;
-NO
2
-N
3 -CN; -OR 21
-OCOR
21 -COR2n, -NC0R 21
-N(R
2 2 1 2
CON(R
21 2 or -C00R 21 aryl or heteroaryl; or two R 20 groups present on adjacent carbon atoms can join together to form a methylenedioxy group; wherein each R 21 is independently straight chained or branched C 1 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or alkynyl; C 3
C
7 cycloalkyl, C 5
-C
7 cycloalkenyl, aryl or aryl (CI-
C
6 alkyl; 00 00 00
§D
oos 0q mD 0D 0q 37 wherein each R 22 is independently H, F, C1 or CI-C 4 straight chained or branched alkyl; wherein each m is an integer from 0 to 4 inclusive; wherein each n is an integer from 1 to 4 inclusive; wherein p is an integer from 0 to 2 inclusive; wherein q is an integer from 2 to 4 inclusive; wherein t is 1 or 2; or a pharmaceutically acceptable salt thereof.
The invention provides a method of treating a subject suffering from anxiety which comprises administering to the subject an amount of compound effective to treat the subject's anxiety wherein the compound has the structure: wherein W is H, -Cl, -Br, CN, methyl, ethyl, propyl, methoxy or ethoxy; wherein X is N(CH 3 )2 or 00 38
R
1 7 -N 0 00
R
17 0 wherein R 1 3 is a bicyclic alkyl ring system, aryl or 00 5 aryl (C C 6 alkyl, wherein Y is NR 14 Rls; wherein R 1 4 is H, straight chained or branched CI-Cs alkyl,
(CH
2 )q-OiCH 2
C
3 -C6 cycloalkyl, or (C(Rig) 2 )m-Z; wherein R 15 is (C (R 1 9
(R,
6 2; wherein Z is C 3
-C
10 cycloalkyl, aryl, or heteroaryl; wherein R 16 is straight chained or branched Cl-C 7 alkyl, straight chained or branched Cj 1 -C7 monofluoroalkyl, straight chained or branched
C
1
-C
7 polyfluoroalkyl, straight chained or branched
C
2
-C
7 alkenyl, straight chained or branched
C
2
-C
7 alkynyl,
C
5
-C
7 cycloalkenyl,-
(CH
2 )mZ, or (CH 2 )q,-O-(CH2)m-CH3; wherein each R 17 is independently H; -OR 2 1 -0C0R 21 -C0R 21
-NCOR
2 1
-N(R
2 1 2
-CON(R
2 1 2 -C00R 21 straight chained or branched Cl-C 7 alkyl, straight chained or branched
C
1 -C7 monofluoroalkyl, straight chained or branched
CI-C
7 polyfluoroalkyl, straight chained or branched
C
2
-C
7 alkenyl, straight chained or branched C 2
-C
7 alkynyl,
CS-C
7 cycloalkenyl,
-(CH
2 or (CH 2 )n-O (C 2 )m-C4 3 39 wherein each R 19 is independently H, or straight chained or branched C 1
-C
6 alkyl; wherein each R 21 is independently straight chained or branched Ci-C7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or alkynyl; C 3
C
7 cycloalkyl, Cs-C 7 cycloalkenyl, aryl or aryl(Ci-
C
s )alkyl; wherein each m is an integer from 0 to 4 inclusive; wherein each n is an integer from 1 to 4 inclusive; wherein q is an integer from 2 to 4 inclusive; or a pharmaceutically acceptable salt thereof.
The invention also provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound having the structure: wherein W is H, -Cl, propyl, methoxy or ethoxy; -Br, CN, methyl, ethyl, 00 whereini X is; NR 11
R
12 R R 1 7 -N -N =0 or
R
1 1 7 00 177 00 -N
N-R
1 8 17 wherein Ril is H, straight chained or branched Cl-C 7 alkyl,
(CH
2 )q0O(CH2)m-CH3, aryl, or aryl (C 1
-C
6 )alkyl; wherein R 12 is straight chained or branched Cl-C 7 alkyl,
(CH
2 )q0O(CH 2 )m-CH3i or -C2.Z wherein R.
1 3 is a bicyclic alkyl ring system, adamantyl, noradamantyl,
C
3
-CI
0 cycloalkyl, heteroaryl, aryl, aryl (Cl-
C
6 alkyl, Qi Or Q2 i wherein aryl may be substituted with one or more Cl-Cio0 straight chained or branched alkyl, aryl, heteroarylvor N (R 19
-Z;
wherein Q, is 00 41
J\/R
22 00 wherein Q2 is
R
22 22 00
R
22
R
22
R
22 20
R
20 wherein each J is independently 0, S, C(R 22 2 or NR 4 wherein R 4 is H; straight chained or branched Cl-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl *or alkynyl; C 3 cycloalkyl, Cs-C 7 cycloalkenyl or aryl; wherein Y is NR 14
R
1 5 00 42 002 0 -No- (C R17 9 N)~Z wherein R 1 4 is Hstraight chained or branched Cl-C6 alkyl, straight CH2 -chne or-C brlanched 1 C monofuoroa.kZ; 5i straight chained or branched C 3-C 1 oyforalkyl, whri 1 sstraight chained or branched
CI-C
7 alkyl, rih chained or branched
C
2
-C
7 alkynyl,
C
5
-C
7 cycloalkenyl,
(CH
2 or (CH 2 )q-O-(CH2),-CH3; wherein each R 17 is independently H; -OR 21 -0C0R 2 1
-COR
2 1, 00 43
-NCOR
2 1
N(R
2 1 2 1 -CON(R 21 2 -C00R 21 straight chained or Ct branched Cl-C7 alkyl, straight chained or branched Cl-C 7 monofluoroalkyl, straight chained or branched Cl-C 7 polyfluoroalkyl, straight chained or branched C 2
-C,
alkenyl, straight chained or branched
C
2 -C-1 alkynyl,
C
5
-C
7 00 cycloalkenyl, -(CH 2 or (CH 2 )n0O-(CH2)m-CH3; wherein R 18 is straight chained or branched C 1
-C
6 alkyl, 00 (CH4 2 Or (CH2)q-O (CH 2 )m-CH 3 wherein each R 19 is independently H, or straight chained or branched CI-C 6 alkyl; wherein each R 20 is independently straight chained or branched CI-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3
C
7 cycloalkyl or C 5
-C
7 cycloalkenyl; -C1, -Br, or -I;
-NO
2
-N
3 -CN; -OR 2 1 -0C0R 21 -C0R 21
-NCOR
21
-N(R
21 2
CON(R
2 1 or -C00R 21 aryl or heteroaryl; or two R 2 0 groups present on adjacent carbon atoms can join together to form a methylenedioxy group; wherein each R 2 1 is independently straight chained or branched CI-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3 C, cycloalkyl, C5-C 7 cycloalkenyl, aryl, or aryl(C 1 CE) alkyl; wherein each R 2 2 is independently H, F, C1 or C 1
-C
4 straight chained or branched alkyl; wherein each m is an integer from 0 to 4 inclusive; 00 CD 44 cwherein each n is an integer from 1 to 4 inclusive; wherein p is an integer from 0 to 2 inclusive; 00 wherein q is an integer from 2 to 4 inclusive; Cy wherein t is 1 or 2; 00 C 10 wherein U is O, -NR 16 S, C(R 17 2 or -NSO 2
R
16 wherein Z is C 3 -Cio cycloalkyl, C 4 cyclic ether, C 4
-C
7 cyclic thioether, aryl, or heteroaryl; or a pharmaceutically acceptable salt thereof.
The invention provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound having the structure:
X
W
NN
yR 1 3
H
wherein W is H, -Cl, -Br, CN, methyl, ethyl, propyl, methoxy or ethoxy; 00 wherein X is NRIIR 12
R
17 -N or -NN-* 00 1 00 wherein R 11 is H, straight chained or branched Cl-C 7 alkyl, (CH) q0-(CH 2 )m-CH 3 aryl or aryl (Cl-C 6 alkyl; c-I wherein R 1 2 is straight chained or branche d Cl-C 7 alkyl,
(CH
2 q0-CH 2 mCH3, or CH 2 )mIZ; wherein R 13 is a bicyclic alkyl ring system, aryl or aryl (Cl-C 6 alkyl; wherein Y is NR14Rl5; 00 46
R
2 0 I p or 00 R 19 00
N
-N-
wherein R 14 is H, straight chained or branched C 1
-C
6 alkyl,
(CH
2 )q0O(CH 2 )M-CH3I C 3
-C
6 cycloalkyl, or (C(R 1 9 2 wherein R3.
5 is straight chained or branched C 3
-C
6 alkyl,
(CH
2 )qDO(CH 2 )m-H3, C 3
-C
6 cycloalkyl, or (C(R 1 9 2 wherein U is 0, -NR 16 S, C(R 1 7 2 or -NSO 2
R,
6 wherein Z is C 3 -Cl 0 cycloalkyl, aryl, or heteroaryl; wherein R 16 is straight chained or branched C 1 C7 alkyl, straight chained or branched Cl-C 7 monof luoroalkyl, straight chained or branched Cl-C 7 polyfluoroalkyl, straight chained or branched C 2 -C7 aikenyl, straight chained or branched C 2
-C
7 aikynyl, CS-C 7 cycloalkenyl,
(CH
2 or (CH2)q0 -(CH 2 )m-CH 3 00 47 wherein each R 1 7 is independently H; -OR 2 1
-OCOR
21 -C0R 21
-NCOR
21
-N(R
21 2 I -CON(R 21 2 -C00R 21 straight chained or branched C 1
-C
7 alkyl, straight chained or branched CI-C 7 monofluoroalkyl, straight chained or branched C 1 -C-y polyfluoroalkyl, straight chained or branched C 2
-C
7 00 alkenyl, straight chained or branched C 2
-C
7 alkynyl, C 5
-C
7 cycloalkenyl, (CH 2 1-Z, or (CH 2
(CH
2 )m.-CH 3 00 wherein RIO is straight chained or branched CI-C 6 alkyl,
(CH
2 or (CH1 2 2 )m-CH3; wherein each R 19 is independently H, or straight chained or branched Cl-C 6 alkyl; wherein each R 2 0 is independently straight chained or branched C 1
-C
7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 alkenyl or alkynyl; C 3
C
7 cycloalkyl or C 5
-C
7 cycloalkenyl; -Cl, -Br, or -I;
-NO
2
-N
3 -CN; -OR 21 -0C0R 21 -C0R 21
-NCOR
21
-N(R
2 3.) 2
CON(R
2 1 2 or -COOR 2 aryl or heteroaryl; or two R 20 groups present on adjacent carbon atoms can join together to form a methylenedioxy group; wherein each R 2 1 is independently straight chained or branched Cl-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl, or alkynyl; C 3
C
7 cycloalkyl, CS-C7 cycloalkenyl, aryl or aryl(Cl-
C
6 alkyl; wherein each m is an integer from 0 to 4 inclusive; 48 wherein each n is an integer from 1 to 4 inclusive; wherein p is an integer from 0 to 2 inclusive; wherein q is an integer from 2 to 4 inclusive; wherein t is 1 or 2; or a pharmaceutically acceptable salt thereof.
The invention provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound having the structure: wherein W is H, -Cl, propyl, methoxy or ethoxy; wherein X is N(CH 3 2 or -Br, CN, methyl, ethyl, 00 4 cR17 -N =0 00 wherein R 13 is an aryl, adamantyl, noradamantyl,
C
3
-C
1 o 0 cycloalkyl, heteroaryl, Q, or Q2; 00 wherein aryl may be substituted with one or more Cj-CjO straight chained or branched alkyl, aryl, heteroaryl, or N (R 19
Z;
wherein is J>(R22 wherein Q2 is
R
22 R 22 2 2
R
2 2 t
R
2 2
R
2 2
PR
20 wherein each J is independently 0, S, C (R22) 2 or NR 4 wherein R 4 is straight chained or branched Cl-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3
-C
7 cycloalkyl, C5-C 7 cycloalkenyl or aryl; wherein Y is NR 1 4
R
1 5
R
17 -N U \Et/ 1 ;or wherein R 14 is H, straight chained or branched C].-C 6 alkyl, (CH2)q-O (CH 2 )m-CH 3
C
3
-C
6 cycloalkyl, or (C(R 19 2 )m-Z; wherein R 15 is straight chained or branched C 3
-C
6 alkyl,
(CH
2 )q0I-CH2)m-CH3,
C
3
-C
6 cycloalkyl, or (C(R 1 9 2 wherein U is 0, -NR, 6 S, C(R, 1 7 2 or -NS0 2
R,
6 wherein Z is C 3 -CIO cycloalkyl, aryl, or heteroaryl; 00 51 wherein RI 6 is straight chained or branched Cl-C-7 alkyl, V')straight chained or branched C.I-C 7 monofluoroalkyl, Ntagt cand o rnhd plfurakl straight chained or branched C-C 7 alyfl sraight 00 chained or branched C 2
-C
7 alkynyl, C 5
-C
7 cycloalkenyl,
(CH
2 )ngZ, or (C 2 )q,-O-(CH2)m-CH 3 00 wherein each R 17 is independently H; -OR 2 1 -0C0R 21 -C0R 21
-NCOR
21
-N(R
2 2 -CONq(R 21 2 -C00R 21 straight chained or btanched C 1 -C7 alkyl, straight chained or- branched CI-C 7 monofluoroalkyl, straight chained or branched Cl-C 7 polyfluoroalkyl, straight chained or branched C 2 -c 7 alkenyl, straight chained or branched C 2
-C
7 alkynyl, C.-C 7 cycloalkenyl,
-(CH
2 or (CH 2 )n-O-(CH 2 )m-CH 3 wherein R3.
8 is straight chained or branched C 1
-C
6 5 alkyl,
(CH
2 or (C!I 2 )q-OiCH 2 )m-CH 3 wherein each R 19 is independently H, or straight chained or branched C 1
-C
6 alkyl; wherein each R 20 is independently straight chained or branched Cl-C 7 alkyl, monofluoroalkyl or polytluoroalkyl; straight chained or branched C 2 -C,7 alkenyl or alkynyl; C 3 C,7 cycloalkyl or Cs-C, cycloalkenyl; -Cl, -Br, or -1;
-NO
2
-N
3 -CN; -OR 21 l, -0C0R 21
-COR
21
-NCOR
21
-NCR
21 2
CON(R
2 1 2 or -COOR 21 aryl or heteroaryl; or two R 2 0 groups present on adjacent carbon atoms can join together to form a methylenedioxy group; wherein each R 2 3. is independently straight chained or 00 52 OO 52 branched C 1
-C
7 alkyl, monofluoroalkyl or polyfluoroalkyl; c straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3 tt C 7 cycloalkyl, Cs-C 7 cycloalkenyl, aryl or aryl(C 1 C) alkyl; 00 wherein each R 22 is independently H, F, Cl or C 1
-C
4 straight chained or branched alkyl; 00 wherein each m is an integer from 0 to 4 inclusive; wherein each n is an integer from 1 to 4 inclusive; wherein p is an integer from 0 to 2 inclusive; wherein q is an integer from 2 to 4 inclusive; wherein t is 1 or 2; or a pharmaceutically acceptable salt thereof.
The invention provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound having the structure: 53 wherein W is H, -Cl, -Br, CN, methyl, ethyl, propyl, methoxy or ethoxy; wherein X is N (CH 3 2 or wherein R 13 is a bicyclic alkyl ring system, aryl or aryl (C CO) alkyl; wherein Y is NR 14
R
15 wherein R3.
4 is H, straight chained or branched Cj-C 6 alkyl,
(CH
2 )q0-(C 2 )m-CH3, C 3
-C
6 cycloalkyl, or (C(R 1 9 2 )m-Z; wherein R 15 is (C(Rl 9 2 )m-N(Rl6)2; wherein Z is C 3 -CIO cycloalkyl, aryl, or heteroaryl; wherein R 16 is straight chained or branched
CI-C
7 alkyl, straight chained or branched C3 1
-C
7 monofluoroalkyl, straight chained or branched CI-C7 polyfluoroalkyl, straight chained or branched C 2 -C7 alkenyl, straight chained or branched
C
2
-C-
7 alkynyl, C 5 -C7 cycloalkenyl, 00 54
(CH
2 or (CH2)q-O-(CH 2 )m-CH 3 Ct l wherein each R 17 is independently H; -OR 21
-OCOR
21
-COR
21
C
N
-NCOR
21
-N(R
21 2 -CON(R21) 2
-COOR
21 straight chained or branched C 1
-C
7 alkyl, straight chained or branched Ci-C, 00 monofluoroalkyl, straight chained or branched Ci-C 7 polyfluoroalkyl, straight chained or branched C 2
-C
7 Salkenyl, straight chained or branched C 2 -C7 alkynyl, Cs-C 7 OO cycloalkenyl, -(CH 2 or (CH 2 )n-O-(CH 2 )m-CH 3 wherein each R 19 is independently H, or straight chained or branched CI-C 6 alkyl; wherein each R 21 is independently straight chained or branched CI-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or alkynyl; C 3
C
7 cycloalkyl, Cs-C 7 cycloalkenyl, aryl or aryl(C 1
C
6 alkyl; wherein each m is an integer from 0 to 4 inclusive; wherein each n is an integer from 1 to 4 inclusive; wherein q is an integer from 2 to 4 inclusive; or a pharmaceutically acceptable salt thereof.
The invention provides a compound having the structure: wherein W is H, -Cl, -Br, CN, methyl, ethyl, propyl, methoxy or ethoxy; wherein X is; NRnlR12;
R
17
R
17 -N1 or wherein RI.I is H, straight chained or branched Cl-C 7
(CH
2 )q0I-CH 2 )m-CH3, aryl, or aryl (Cl-C 6 )alkyl; wherein
R
12 is straight chained or branched
C-
1
-C
7
(CH
2 )q-O1(CH2),-CH3I or -(CH 2 )mZ; alkyl, alkyl, wherein R 13 is a bicyclic alkyl ring system, adamantyl, noradamantyl,
C
3
-C
1 o cycloalkyl, heteroaryl, aryl, aryl(Cl-
C
6 alkyl, Q, or Q2; wherein aryl may be substituted with one or more Cl-C,(o straight chained or branched alkyl, aryl, heteroaryl, or N(Rg-Z wherein Q, is wherein Q2 is is wherein each J is independently 0, S, C(R 2 2 2 or NR 4 wherein R 4 is H; straight chained or branched C 1
-C
7 alkyl, monofluoroalkyl, or polyfluoroalkyl; straight chained or branched C 2 -C-y alkenyl or aikynyl; C 3 -C7 cycloalkyl, CS-C7 cycloalkenyl or aryl; wherein Y is NR 14
R
15 00 57 R2 or N U 1 00 [i'RR 19 00
N-
wherein R 1 4 is H, straight chained or branched C 1
-C
6 alkyl, CH)O- -(CH 2 C13 C 3
-C
6 cycl1oalkyl or (C (R- 19 2 Z wherein Rig is straight chained or branched C 3 -C6 alkyl,
(CH
2 )q-O-(CH 2 )m-CH 3
C
3
-C
6 cycloalkyl, (C (Rig) 2
),N(RIO)
2 or wherein R 16 is straight chained or branched Cl-C 7 alkyl, straight chained or branched Cl-C 7 monofluoroalkyl, straight chained or branched Cl-C 7 polyfluoroalkyl, straight chained or branched C 2
-C
7 alkenyl, straight chained or branched C 2
-C
7 alkynyl, C.
5
-C
7 cycloalkenyl,
(CH
2 or (CH 2 )q-O-(CH 2 )m-CH 3 wherein each R 17 is independently H; -0R 2 1, -0C0R 21 -C0R 21 00 58
-NCOR
21
-N(R
21 2
-CON(R
21 2
-COOR
21 straight chained or c branched Ci-C 7 alkyl, straight chained or branched C 1 -C7 V) monofluoroalkyl, straight chained or branched CI-C 7 polyfluoroalkyl, straight chained or branched C 2 -C7 alkenyl, straight chained or branched C 2
-C
7 alkynyl, Cs-C7 00 cycloalkenyl, (CH 2 or (CH 2
(CH
2 )m-CH 3 wherein R 18 is straight chained or branched CI-C 6 alkyl, 00 (CH 2 or (CH2)q-O-(CH 2 )m-CH 3 wherein each R 19 is independently H, or straight chained or branched C 1
-C
6 alkyl; wherein each R 20 is independently straight chained or branched C 1 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or alkynyl; C 3
C
7 cycloalkyl or C 5
-C
7 cycloalkenyl; -C1, -Br, or -I;
-NO
2
-N
3 -CN; -OR 21
-OCOR
21
-COR
21
-NCOR
21
-N(R
21 2
CON(R
21 2 or -COOR21; aryl or heteroaryl; or two R 20 groups present on adjacent carbon atoms can join together to form a methylenedioxy group; wherein each R 21 is independently straight chained or branched C 1 -C7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3 C7 cycloalkyl, Cs-C, cycloalkenyl, aryl, or aryl(C 1 Cs) alkyl; wherein each R 22 is independently H, F, Cl or C 1
-C
4 straight chained or branched alkyl; wherein each m is an integer from 0 to 4 inclusive; 00 59 c wherein each n is an integer from 1 to 4 inclusive; wherein p is an integer from 0 to 2 inclusive; 00 wherein q is an integer from 2 to 4 inclusive; p- wherein t is 1 or 2; 00 wherein U is O, -NR 16 S, C(R17) 2 or -NSO 2
R
1 s; wherein Z is C 3
-C
10 cycloalkyl, C 4
-C
7 cyclic ether, C 4
-C
7 cyclic thioether, aryl, or heteroaryl; or a pharmaceutically acceptable salt thereof.
The invention provides a compound having the structure:
X
NW
NN
H
wherein W is H, -Cl, -Br, CN, methyl, ethyl, propyl, methoxy or ethoxy; wherein X is NR 11
R
1 2
R
1 7 RI
/R
17 -N or -N N-Ri R, 1 wherein Ril is H, straight chained or branched Cl-C7 alkyl, (CH2)q-O (CH 2 )m-CH 3 aryl or aryl (Cl-CG)alkyl; wherein R 1 2 is straight chained or branched Cl-C7 alkyl, (CHO) q- 0
(CH
2 mCH 3 or (CH 2 mZ; wherein R 1 3 is a bicyclic alkyl ring system, aryl or aryl Cc,) alkyl; wherein Y is NR 14
R
15 lp ;or wherein R 1 4 is H, straight chained or branched Cl-.C 6 alkyl,
(CH
2 )q0O(CH 2 )m-CH3, C 3
-C
6 cycloalkyl, or (C(Ri9) 2 wherein R 15 is straight chained or branched C 3
-C
6 alkyl, 00 61
(CH
2 )qO1CH 2 )m-CH3, C 3
-C
6 cycloalkyl, or (C(R 19 2 )m-Z; wherein U is 0, -NR, 6 S, C(R 1 7 2 or -NS0 2
R,
6 wherein Z is C 3 -CIO cycloalkyl, aryl, or heteroaryl; 00 wherein R3.
6 is straight chained or branched CI-C 7 alkyl, straight chained or branched Cl-C 7 monofluoroalkyl, 00 straight chained or branched CI-C 7 polyfluoroalkyl, straight chained or branched C 2
-C
7 alkenyl, straight -chained or branched C 2
-C
7 alkynyl, Cs-C-r cycloalkenyl,
(CH
2 or (CH 2 q 0
-(CH
2 m-CH3; wherein each R 17 is independently H; -OR 2 1 -0C0R 21 -C0R 2 1
-NCOR
21
-N(R
21 2
-CON(R
21 2 -C00R 21 straight chained or branched CI-C 7 alkyl, straight chained or branched CI-C7 monofluoroalkyl, straight chained or branched CI-C-7 polyfluoroalkyl, straight chained or branched C 2 C7 alkenyl, straight chained or branched C 2 -C7 alkynyl, C 5
-C
7 cycloalkenyl, -(CH 2 or (CH 2 )n-0-(CH 2
).-CH
3 wherein R 18 is straight chained or branched CI-C 6 alkyl,-
(CH
2 or (CH 2 )q0O(CH 2 )m-CH 3 wherein each R 19 is independently H, or straight chained or branched Cj-C 6 alkyl; wherein each R 2 0 is independently straight chained or branched C 1 -07 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3
C
7 cycloalkyl or C 5
-C
7 cycloalkenyl; -C1, -Br, or -I;
-NO
2
-N
3 -EN; -OR 2 1 -0C0R 21
-COR
2 1
-NCOR
2 1
-N(R
2 1 2 00 62
CON(R
21 2 or -COOR 21 aryl or heteroaryl; or two R 20 groups c present on adjacent carbon atoms can join together to V) form a methylenedioxy group;
(N
wherein each R 21 is independently straight chained or 00 branched Ci-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; O straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3 C C 7 cycloalkyl, Cs-C 7 cycloalkenyl, aryl or aryl (C- 0 0 Cs) alkyl; *wherein each m is an integer from 0 to 4-inclusive; wherein each n is an integer from 1 to 4 inclusive; wherein p is an integer from 0 to 2 inclusive; wherein q is an integer from 2 to 4 inclusive; wherein t is 1 or 2; or a pharmaceutically acceptable salt thereof.
The invention provides a compound having the structure: wherein W is H, -Cl, -Br, CN, methyl, ethyl, propyl, methoxy or ethoxy; wherein X is N (CH 3 2 or wherein R 1 3 is an aryl, adamantyl, noradamantyl, C 3
-C
10 cycloalkyl, heteroaryl, Q, or 02; wherein aryl may be substituted with one or more Cl-Clo straight chained or branched alkyl, aryl, heteroaryl, or N (R 19
Z;
wherein Q3. is wherein Q2 is wherein each J is independently 0, S, C(R 2 2 2 or NRZ 4 wherein R 4 is straight chained or branched Cl-C, alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3 -C,7 cycloalkyl, C 5
-C
7 cycloalkenyl or aryl; wherein Y is NR 14
R
1 5
R/
-N u p or
R
20
R
20 00 wherein R 14 is H, straight chained or branched Cl-C 6 alkyl,
(CH
2 )q-O(H 2 )m--H3I C 3
-C
6 cycloalkyl, or (C(R 1 9 2 00 wherein Rls is straight chained or branched C 3
-C
6 alkyl,
(CH
2 )q-OiCH2).-CH3,
C
3
-C
6 cycloalkyl, or 00 wherein U is 0, -NR16, S, C(Rl 7 2 or -NS0 2
R,
6 c-i wherein Z is C 3
-C
10 cycloalkyl, aryl, or'heteroaryl; wherein R 1 6 is straight chained or branched Cl-C,? alkyl, straight chained or branched C2 1
-C
7 monofluoroalkyl, straight chained or branched CI-C 7 polyfluoroalkyl, straight chained or branched C 2
-C
7 alkenyl, straight chained or branched C 2 -C7 alkynyl, CS-C 7 cycloalkenyl,
(CH
2 )mZ, or (CH2)q-O (CH 2 )m-CH 3 wherein each R 1 7 is independently H; -OR 2 1 -0C0R 2 1 -C0R 21
-NCOR
21
-N(R
21 2
-CON(R
21 2 -C00R 21 straight chained or branched Cl-C 7 alkyl, straight chained or branched Cl-C 7 monofluoroalkyl, *straight chained or branched Cl-C 7 polyfluoroalkyl, straight chained or branched C 2
-C
7 alkenyl, straight chained or branched C 2
-C
7 alkynyl, CS-C 7 cycloalkenyl, -(CH 2 or (C2)nO-(CH 2
)-CH
3 wherein R 18 is straight chained or branched Cj-C 6 alkyl,
(CH
2 or (CH 2 )q-O1CH 2 )m-CH3; wherein each R].
9 is independently H, or straight chained or branched CI-C 6 alkyl; 00 66 cwherein each R2o is independently straight chained or Sbranched C 1
-C
7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3 C, cycloalkyl or Cs-C, cycloalkenyl; -Cl, -Br, or -I; OO -NO 2
-N
3 -CN; -OR 21
-OCOR
21
-COR
21
-NCOR
21
-N(R
21 2
CON(R
21 2 or -COOR 21 aryl or heteroaryl; or two R 2 0 groups Spresent on adjacent carbon atoms can join together to 0 0 form a methylenedioxy group; -wherein each R 21 is independently st-raight chained or branched C 1 -C7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or alkynyl; C 3 C, cycloalkyl, C 5 -C7 cycloalkenyl, aryl or aryl(C 1
C
6 alkyl; wherein each R 22 is independently H, F, C1 or C 1
-C
4 straight chained or branched alkyl; wherein each m is an integer from 0 to 4 inclusive; wherein each n is an integer from 1 to 4 inclusive; wherein p is an integer from 0 to 2 inclusive; wherein q is an integer from 2 to 4 inclusive; wherein t is 1 or 2; or a pharmaceutically acceptable salt thereof.
The invention provides a compound having the structure: 00 67 x w CI N N
R
1 3 00
YNN
00 wherein W is H, -Cl, -Br, CN, methyl, ethyl, propyl, methoxy or ethoxy; wherein X is N (CH 3 2 or
-N
17 wherein R 13 is a bicyclic alkyl ring system, aryl or aryl (C 1 -CG) alkyl; wherein Y is NR 14
R
1 5 wherein R 14 is H, straight chained or branched Cj-C 6 alkyl,
(CH
2 )q0O(CH2)m-CH3, C 3
-C
6 cycloalkyl, or (C(R 1 9 2 wherein R 15 is (C(R 19 2 )m-N(R 16 2 wherein Z is C 3
-C
10 cycloalkyl, aryl, or heteroaryl; 00 68 wherein R 16 is straight chained or branched Ci-C 7 alkyl, Cstraight chained or branched C-C, monofluoroalkyl, V straight chained or branched Ci-C, polyfluoroalkyl, straight chained or branched C 2
-C
7 alkenyl, straight chained or branched C 2 -C7 alkynyl, Cs-C 7 cycloalkenyl, 00 (CH 2 Or (CH 2 (CH2)m-CH3; 00 C- wherein each Rn7 is independently H; -OR2i, -OCOR21, -COR21, 0 -NCOR 21
-N(R
21 2
-CON(R
21 2
-COOR
21 straight chained or 10 branched Ci-C 7 alkyl, straight chained or branched Ci-C 7 monofluoroalkyl, straight chained or branched C 1
-C
7 polyfluoroalkyl, straight chained or branched C 2
-C
7 alkenyl, straight chained or branched C 2 -C7 alkynyl, C5-C 7 cycloalkenyl, -(CH 2 or (CH 2
(CH
2 )m-CH 3 wherein each R 19 is independently H, or straight chained or branched CI-C 6 alkyl; wherein each R 21 is independently straight chained or branched C 1
-C
7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or alkynyl; C 3 C7 cycloalkyl,
C
5
-C
7 cycloalkenyl, aryl or aryl(C1- Cg) alkyl; wherein each m is an integer from 0 to 4 inclusive; wherein each n is an integer from 1 to 4 inclusive; wherein q is an integer from 2 to 4 inclusive; or a pharmaceutically acceptable salt thereof.
00 69 c The invention also provides a method of treating a Iy subject suffering from depression which comprises C administering to the subject an amount of compound effective to treat the subject's depression wherein the 00 compound has the structure: 00 C(
B
Y2 Y3N
A
Y4 wherein each of Y 1
Y
2
Y
3 and Y 4 is independently H; straight chained or branched C 1 -C7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3
-C
7 cycloalkyl, or C 5
-C
7 cycloalkenyl; -Cl, -Br, or I; -NO 2
-N
3 -CN; -OR 4
-SR
4
-OCOR
4 -COR4, -NCOR 4
N(R
4 2
-CON(R
4 2 or -COOR4; aryl or heteroaryl; or any two of YI, Y 2
Y
3 and Y 4 present on adjacent carbon atoms can constitute a methylenedioxy group; wherein each R 4 is independently straight chained or branched Ci-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3
-C
7 cycloalkyl, C 5 -C7 cycloalkenyl, aryl or aryl alkyl; wherein A is A' Q3, Q4, Q5, straight chained or branched Cl-C 7 alkyl, aryl, heteroaryl, aryl (Cl-
C
6 alkyl, heteroaryl (C 1
-C
6 alkyl, aryl. substituted with an aryl or heteroaryl, heteroaryl substituted with an aryl or heteroaryl; or (CHR 17 -(CHR1 7 n-Z; wherein A' is 0
R,
n CR 2
R
3 or (CH 2
R
wherein Q3 is R17
R
17
R
17
R
17 n wherein Q 4 is wherein Qs is wherein RI and R 2 are each independently H, straight chained or branched C 1
-C
7 alkyl, -Cl, -Br,
NO
2 or -CN; wherein R 3 is H, straight chained or branched C 1
-C
7 alkyl, -Cl, -Br, -NOz, -CN, -OR 6 aryl or heteroaryl; wherein Rs is straight chained or branched Ci-C 7 alkyl, -N(R 4 2
-OR
6 or aryl; 00 72 00 c wherein Re is straight chained or branched Ci-C7 VS alkyl or aryl; wherein each Ri7 is independently H; straight chained b C 1
-C
7 monofluoroalkyl, straight chained or branched 00 CI-C 7 polyfluoroalkyl, straight chained or branched
SC
2
-C
7 alkenyl, straight chained or branched C 2 -C7 alkynyl, Cs-C 7 cycloalkenyl, -(CH 2 or (CH 2 )n-O-
(CH
2 m-CH 3 wherein each R 20 is independently straight chained or branched Ci-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3
-C
7 cycloalkyl or C 5
-C
7 cycloalkenyl; -Cl, -Br, or -NO 2
-N
3 -CN;
OR
21
-OCOR
21
-COR
21
-NCOR
21
-N(R
21 2
-CON(R
21 2 or
-COOR
21 aryl or heteroaryl; or two R2o groups present on adjacent carbon atoms can join together to form a methylenedioxy group; wherein each R 21 is independently straight chained or branched CI-C7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or alkynyl; C 3 -C7 cycloalkyl, Cs-C, cycloalkenyl, aryl or aryl(C 1
-C
6 )alkyl; 00 73 wherein each m is an integer from 0 to 4 inclusive; 1- wherein each n is an integer from 1 to 4 inclusive; 00 5 wherein each p is an integer from 0 to 2 inclusive; 00 wherein U is O, -NR 16 S, C(R 7 2 or -NSO 2 Ri 6 wherein Z is C 3
-C
1 o cycloalkyl, C4-67 cyclic ether,
C
4 cyclic thioether, aryl, or heteroaryl; wherein R 16 is straight chained or branched C 1 -C7 alkyl, straight chained or branched Ci-C7 monofluoroalkyl, straight chained or branched Ci-C7 polyfluoroalkyl, straight chained or branched C 2 -C7 alkenyl, straight chained or branched C 2 -C7 alkynyl, Cs-C7 cycloalkenyl, -(CH 2 or (CH 2 )q-O-(CH 2 )m-CH 3 wherein q is an integer from 2 to 4 inclusive; wherein B is aryl, heteroaryl, aryl substituted with an aryl or heteroaryl, heteroaryl substituted with an aryl or heteroaryl, tricyclic heteroaryl or Qs; provided however, if B is aryl or heteroaryl the carbon atom or carbon atoms ortho to the nitrogen atom of the imine bond may only be substituted with one or more of the following -Cl, -Br, -CN, methyl, ethyl or methoxy; 74 wherein a tricyclic heteroaryl is a fused three member aromatic system in which one or more of the rings is heteroaryl; carbazole; or acridine; wherein Qs is wherein each R 22 is independently H, F, Cl, or straight chained or branched Ci-C 4 alkyl; or a pharmaceutically acceptable salt thereof.
The invention provides a method of treating a subject suffering from depression which comprises administering to the subject an amount of compound effective to treat the subject's depression wherein the compound has the structure: 00 7 wherein each of Y 1
Y
2
Y
3 and Y 4 is independently H; straight chained or branched C 1
-C
7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3
-C
7 00 cycloalkyl, or C 5 -C7 cycloalkenyl; -C1, -Br, or 1; -NO 2
-N
3 -CM; -OR 4
-SR
4
-OCOR
4
-COR
4
-NCOR
4 CI
N(R
4 2
-CON(R
4 2 or -COOR 4 aryl or heteroaryl; or 00 any two of Y 1
Y
2
Y
3 and Y 4 present on adjacent carbon atoms can constitute a methylenedioxy group; wherein each R 4 is independently straight chained or branched C 1 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3
-C
7 cycloalkyl,
C
5
-C
7 cycloalkenyl, aryl or aryl(Cl-C 6 )alkyl; wherein A is straight chained or branched CI-C,7 alkyl, aryl, heteroaryl, aryl (C 1
-C
6 alkyl or heteroaryl (C 1
-C
6 alkyl; 0 0 wherein A' is
R
1 or (H2.R I' CR 2
R
3 00 76 wherein R 1 and R 2 are each independently H, straight 00 chained or branched C 1
-C
7 alkyl, -Cl, -Br,
NO
2 or -CN; 0 00 wherein R 3 is H, straight chained or branched CI-C, alkyl, -Cl, -Br, -NO 2 -CN, -OR aryl or heteroaryl; wherein R 5 is straight chained or branched C 1
-C,
alkyl, -N(R4) 2 -OR6 or aryl; wherein R 6 is straight chained or branched C 1
-C
7 alkyl or aryl; wherein B is aryl, or heteroaryl; provided however, if B is aryl or heteroaryl the carbon atom or carbon atoms ortho to the nitrogen atom of the imine bond may only be substituted with one or more of the following -Cl, -Br, -CN, methyl, ethyl or methoxy; wherein n is an integer from 1 to 4 inclusive; or a pharmaceutically acceptable salt thereof.
The invention provides a method of treating a subject suffering from depression which comprises administering to the subject an amount of compound effective to treat 00 77 the subject's depression wherein the compound has the structure: 00 000 wherein each of YI, Y 2 1 Y 3 and Y 4 is independently H; straight chained or branched Cl-C7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3
-C
7 cycloalkyl, or C 5
-C
7 cycloalkenyl; -Cl, -Br, or- 1; -NO 2
-N
3 -CN; -OR 4
-SR
4
-OCOR
4
-COR
4
-NCOR
4 N(R42 ,-CON(R4) 2 or -COOR 4 aryl or heteroaryl; or any two of Y 1
Y
2
Y
3 and Y 4 present on adjacent carbon atoms can constitute a methylenedioxy group; wherein each R 4 is independently straight chained or branched Cl-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched Ci-C 7 alkenyl or alkynyl; C 3
-C
7 cycloalkyl, C 5
-C
7 cycloalkenyl, aryl or aryl(Cl-C 6 )alkyl; wherein A is straight chained or branched Cl-C7 alkyl, aryl, heteroaryl, aryl -C 6 alkyl or heteroaryl (Cl-C 6 alkyl; wherein A' is 0 C
RI
Sor -J(CH2) or R4 wherein B is aryl substituted with an aryl or heteroaryl, heteroaryl substituted with an aryl or heteroaryl, tricyclic heteroaryl or Q6; wherein a tricyclic heteroaryl is a fused three ring aromatic system in which one or more of the rings is heteroaryl; carbazole; or acridine; wherein Q6 is wherein n is an integer from 1 to 4 inclusive; wherein each R 22 is independently H, Cl, or straight chained or branched C 1
-C
4 alkyl; or a pharmaceutically acceptable salt thereof.
The invention provides a method of treating a subject suffering from depression which comprises administering to the subject an amount of compound effective to treat the subject's depression wherein the compound has the structure: wherein each of Y 1
Y
2
Y
3 and Y 4 is independently H; straight chained or branched C 1
-C
7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3
-C
7 cycloalkyl, or C 5 -C7 cycloalkenyl; -Cl, -Br, or I; -NO 2
-N
3 -CN; -OR 4
-SR
4 -OCOR4, -COR4, -NCOR4,
N(R
4 2
-CON(R
4 2 or -COOR4; aryl or heteroaryl; or any two of YI, Y 2
Y
3 and Y 4 present on adjacent carbon atoms can constitute a methylenedioxy group; wherein each R 4 is independently straight chained or branched C 1
-C
7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or alkynyl; C 3 -C7 cycloalkyl, Cs-C7 cycloalkenyl, aryl or aryl (C 1
-C
6 alkyl; wherein A is Q3, Q4, Q5, aryl substituted with an aryl or heteroaryl, heteroaryl substituted with an aryl or heteroaryl, or (CHR 1 7
)-(CHR
1 7 )n-Z; wherein Q3 is 17 wherein Q4 is wherein Qs is 00 81
R
1 7 c-I
U
00 00 wherein each R 17 is independently H; straight chained or branched CI-C 7 alkyl, straight chained or branched Cl-C 7 monofluoroalkyl, straight chained or branched
C
1
-C
7 polyfluoroalkyl, straight chained or branched
C
2
-C
7 alkenyl, straight chained or branched C 2
-C
7 alkynyl, C 5
-C
7 cycloalkenyl,
-(CH
2 or (CH 2 )n-O-
(CH
2 m-CH 3 wherein each R 20 is independently straight chained or branched CI.-C7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or alkynyl; C 3
-C
7 cycloalkyl or C5-C 7 cycloalkenyl; -C1, -Br, or -NO 2
-N
3 -CN;
OR
21 -OCOR2 1
-COR
2 1, -NCOR 2 1, -N(R 21 2
-CON(R
21 or -C00R 21 aryl or heteroaryl; or two R 20 groups present on adjacent carbon atoms can join together to form a methylenedioxy group; wherein each R 21 is independently straight chained or branched Cl-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C? 2
-C
7 alkenyl or alkynyl; C 3 -C7 cycloalkyl,
C
5
-C
7 cycloalkenyl or aryl; 00 0 82 Swherein each R 22 is independently H, F, Cl, or straight chained or branched Ci-C4 alkyl; wherein each m is an integer from 0 to 4 inclusive; D M wherein each n is an integer from 1 to 4 inclusive; C wherein each p is an integer from 0 to 2 inclusive; 00 (1 10 wherein U is O, -NR 16 S, C(R 1 7) 2 or -NSO 2
R
16 wherein Z is C 3 -Cio cycloalkyl,
C
4
-C
7 cyclic ether,
C
4
-C
7 cyclic thioether, aryl, or heteroaryl; wherein R 1 6 is straight chained or branched C 1
-C
7 alkyl, straight chained or branched Ci-C 7 monofluoroalkyl, straight chained or branched CI-C 7 polyfluoroalkyl, straight chained or branched
C
2
-C
7 alkenyl, straight chained or branched C 2
-C
7 alkynyl, Cs-C 7 cycloalkenyl,
-(CH
2 or (CH 2 )q-O-(CH2)m-CH3; wherein q is an integer from 2 to 4 inclusive; wherein B is aryl, or heteroaryl; provided however, if B is aryl or heteroaryl the carbon atom or carbon atoms ortho to the nitrogen atom of the imine bond may only be substituted with one or more of the following -Cl, -Br, -CN, methyl, ethyl or methoxy; or a pharmaceutically acceptable salt thereof.
00 83 The invention provides a method of treating a subject c suffering from anxiety which comprises administering to the subject an amount of compound effective to treat the subject's anxiety wherein the compound has the structure: 00 Y1 N Y3 N 0y
A
Y4 wherein each of Yi, Y 2
Y
3 and Y 4 is independently H; straight chained or branched Ci-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3
-C
7 cycloalkyl, or Cs-C 7 cycloalkenyl; -Cl, -Br, or I; -NO 2
-N
3 -CN; -OR 4
-SR
4 -OCOR4, -COR 4
-NCOR
4
N(R
4 2
-CON(R
4 2 or -COOR 4 aryl or heteroaryl; or any two of YI, Y 2
Y
3 and Y 4 present on adjacent carbon atoms can constitute a methylenedioxy group; wherein each R 4 is independently straight chained or branched Ci-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or alkynyl; C 3 -C7 cycloalkyl, Cs-C 7 cycloalkenyl, aryl or aryl(CI-C 6 alkyl; wherein A is Q 3 Q4, Qs, straight chained or 84 branched Cl-C 7 alkyl, aryl, heteroaryl, aryl (Cl-
C
6 alkyl, heteroaryl (Cl-C 6 alkyl, aryl substituted with an aryl or heteroaryl, heteroaryl substituted with an aryl or heteroaryl; or (CHR 17
-(CHR
17 n-Z; wherein A' is 0 n CR 2
R
3 or (CI2)r -nR wherein Q3 is R, e7 wherein Q4 is wherein Q5 is wherein chained
NO
2 or RI and R 2 are each independently H, straight or branched CI-C 7 alkyl, -Cl, -Br,
-CN;
wherein R 3 is H, straight chained or branched CI-C 7 alkyl, -Cl, -Br, -NOz, -CN, -ORe, aryl or heteroaryl; wherein Rs is straight chained or branched CI-C7 alkyl, -N(R 4 2 -ORe or aryl; 00 86 F wherein Rs is straight chained or branched C 1
-C
7 c alkyl or aryl; wherein each R 17 is independently H; straight chained 005 or branched
C
1
-C
7 alkyl, straight chained or branched
C
1
-C
7 monofluoroalkyl, straight chained or branched Cl-C 7 polyfluoroalkyl, straight chained or branched 00
C
2
-C
7 alkenyl, straight chained or branched
C
2 -C7 alkynyl,
C
5
-C
7 cycloalkenyl,
-(CH
2 or (CH 2 )n-O-
(CH
2 ),-CH3; wherein each R 2 0 is independently straight chained or branched Cl-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched
C
2
-C
7 alkenyl or alkynyl;
C
3
-C
7 cycloalkyl or C 5
-C
7 cycloalkenyl; -C1, -Br, or -NO 2
-N
3 -CN;
OR
2 1
-OCOR
21 -COR21, -NCOR 2 1
-N(R
2 1 2
-CON(R
2 1 or
-COOR
2 1; .aryl or heteroaryl; or two R 20 groups present on adjacent carbon atoms can join together to form a methylenedioxy group; wherein each R 21 is independently straight chained or branched Cl-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched
C
2
-C
alkenyl or alkynyl;
C
3
-C
7 cycloalkyl, Cs-C, cycloalkenyl, aryl or aryl(Cl-Cs)alkyl; wherein each m is an integer from 0 to 4 inclusive; 00 0 87 Swherein each n is an integer from 1 to 4 inclusive; C wherein each p is an integer from 0 to 2 inclusive; 00 M 5 wherein U is O, -NR 16 S, C(R 17 or -NSO 2 Ri 6 00 o wherein Z is C 3 -Clo cycloalkyl, C 4
-C
7 cyclic ether, S
C
4
-C
7 cyclic thioether, aryl, or heteroaryl; wherein
R
16 is straight chained or branched
C
1
-C
7 alkyl, straight chained or branched
C
1
-C
7 monofluoroalkyl, straight chained or branched CI-C 7 polyfluoroalkyl, straight chained or branched
C
2
-C
7 alkenyl, straight chained or branched
C
2
-C
7 alkynyl, Cs-C 7 cycloalkenyl,
-(CH
2 or (CH 2 )q-0-(CH2)m-CH3; wherein q is an integer from 2 to 4 inclusive; wherein B is aryl, heteroaryl, aryl substituted with an aryl or heteroaryl, heteroaryl substituted with an aryl or heteroaryl, tricyclic heteroaryl or Q6; provided however, if B is aryl or heteroaryl the carbon atom or carbon atoms ortho to the nitrogen atom of the imine bond may only be substituted with one or more of the following -Cl, -Br,
-CN,
methyl, ethyl or methoxy; 88 wherein a tricyclic heteroaryl is a fused three member aromatic system in which one or more of the rings is heteroaryl; carbazole; or acridine; wherein Qs is wherein each R 22 is independently H, F, Cl, or straight chained or branched Ci-C 4 alkyl; or a pharmaceutically acceptable salt thereof.
The invention provides a method of treating a subject suffering from anxiety which comprises administering to the subject an amount of compound effective to treat the subject's anxiety wherein the compound has the structure: 00 89 wherein each of Y 1
Y
2
Y
3 and Y 4 is independently H; straight chained or branched CL-C7 al~kyl, (Ni monofluoroalkYl or polyfluoroalkyl; straight chained or branched
C
2
-C
7 alkenyl or alkynyl;
C
3
-C
7 cycloalkyl, or C 5
-C
7 cycloalkenyl; -C1, -Br, or 00 M 1I; -NO 2
-N
3 -CN; -OR 4
-SR
4 -OCOR4, -COR4, -NCOR 4
N(R
4 2
-CON(R
4 or -COOR 4 aryl or heteroaryl; or 00 any two of Y 1
Y
2
Y
3 and Y 4 present on adjacent carbon atoms can constitute a methylenedioxy group; (Ni wherein each R 4 id independently straight chained or branched Cl-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched
C
2 -C7 alkenyl or alkynyl;
C
3
-C
7 cycloalkyl,
C
5
-C
7 cycloalkenyl, aryl or aryl(Cl-C6) alkyl; wherein A is straight chained or branched Cl-C 7 alkyl, aryl, heteroaryl, aryl (C3.-C 6 alkyl or heteroaryl
(C
1
-C
6 alkyl; wherein A, is 0 0 Xtin 00 CD
R
1 CRR or (CH 2 n R4 ,n CR 2
R
3 00 wherein R 1 and R 2 are each independently H, straight 0 chained or branched CI-C 7 alkyl, -Cl, -Br, Cy 5 NO 2 or -CN; 00 q wherein R 3 is H, straight chained or branched Ci-C 7 alkyl, -Cl, -Br, -NO 2 -CN, -OR 6 aryl or heteroaryl; wherein R 5 is straight chained or branched Ci-C 7 alkyl, -N(R 4 2
-OR
6 or aryl; wherein Rs is straight chained or branched Ci-C 7 alkyl or aryl; wherein B is aryl, or heteroaryl; provided however, if B is aryl or heteroaryl the carbon atom or carbon atoms ortho to the nitrogen atom of the imine bond may only be substituted with one or more of the following -Cl, -Br, -CN, methyl, ethyl or methoxy; wherein n is an integer from 1 to 4 inclusive; or a pharmaceutically acceptable salt thereof.
The invention provides a method of treating a subject suffering from anxiety which comprises administering to 00 91 the subject an amount of compound effective to treat the subject's anxiety wherein the compound has the structure: 00 000 wherein each of Y 1
Y
2
Y
3 and Y 4 is independently- H; straight chained or branched Cl-C 1 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched
C
2
-C
7 alkenyl or alkynyl;
C
3
-C
7 cycloalkyl, or C 5
-C
7 cycloalkenyl; -C1, -Br, or 1; -NO 2
-N
3 -CN; -OR 4
-SR
4
-OCOR
4 -COR14, -NCOR 4
N(R
4 2 -CON(R4)2, or -COOR4; aryl or heteroaryl; or any two of Y 1
Y
2
Y
3 and Y 4 present on adjacent carbon atoms can constitute a methylenedioxy group; wherein each R 4 is independently straight chained or branched
C
1
-C
7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched
C
2
-C
7 alkenyl or alkynyl;
C
3
-C
7 cycloalkyl,
C
5
-C
7 cycloalkenyl, aryl or aryl (C 1
-C
6 alkyl; wherein A is straight chained or branched Cl-C 7 alkyl, aryl, heteroaryl, aryl -C 6 alkyl or heteroaryl (Cl-C 6 alkyl; wherein A' is 0 R
R
n CR2R, or (CH2) R4 wherein B is aryl substituted with an aryl or heteroaryl, heteroaryl substituted with an aryl or heteroaryl, tricyclic heteroaryl or Qs; wherein a tricyclic heteroaryl is a fused three ring aromatic system in which one or more of the rings is heteroaryl; carbazole; or acridine; wherein Q 6 is wherein n is an integer from 1 to 4 inclusive; wherein each R 22 is independently H, F, 00 93 Cl, or straight chained or branched C 1
-C
4 alkyl; VS or a pharmaceutically acceptable salt thereof.
00 The invention provides a method of treating a subject 0 suffering from anxiety which comprises administering to S the subject an amount of compound effective to treat the 00 subject's anxiety wherein the compound has the structure:
B
Y 1 N Y2 Y4 wherein each of Y 1
Y
2
Y
3 and Y 4 is independently H; straight chained or branched Ci-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3 -C7 cycloalkyl, or Cs-C 7 cycloalkenyl; -C1, -Br, or I; -NO 2
-N
3 -CN; -OR 4
-SR
4 -OCOR4, -COR 4 -NCORi., N(R4)2 -CON(R 4 2 or -COOR 4 aryl or heteroaryl; or any two of YI, Y 2
Y
3 and Y 4 present on adjacent carbon atoms can constitute a methylenedioxy group; wherein each R 4 is independently straight chained or branched C 1
-C
7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 00 94 alkenyl or alkynyl;
C
3
-C
7 cycloalkyl,
C
5
-C
7 cycloalkelyl 1 aryl or aryl (Cl-Cs) alkyl; wherein A is Q3, Q41 Q 5 aryl substituted with an aryl or heteroaryl, heteroaryl substituted with an 00 M aryl or heteroaryl, or (CHR 1 7
-(CHR
1 wherein Q3 is 00 R17
RV
N
R
17 17
R
17 wherein Q4 is R17
R
17 N n n ~R 2 0 m wheei Os7 is 17 151 wherein each R 17 is independently H; straight chained 00 D Sor branched C 1
-C
7 alkyl, straight chained or branched
CI-C
7 monofluoroalkyl, straight chained or branched SCi-C 7 polyfluoroalkyl, straight chained or branched
C
2
-C
7 alkenyl, straight chained or branched C 2
-C
7 O 5 alkynyl, Cs-C 7 cycloalkenyl,
-(CH
2 or (CH 2 )n-O- 00 CM
(CH
2 )m-CH 3 00 wherein each R 20 is independently straight Schained or branched Ci-C 7 alkyl, monofluoroalkyl or CA 10 polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3
-C
7 cycloalkyl or Cs-C 7 cycloalkenyl; -Cl, -Br, or -NO 2
-N
3 -CN;
OR
21
-R
2 1 -21, -COR 21
-NCOR
21
-N(R
21 2
-CON(R
21 2 or
-COOR
21 aryl or heteroaryl; or two R 20 groups present on adjacent carbon atoms can join together to form a methylenedioxy group; wherein each R 21 is independently straight chained or branched Ci-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3
-C
7 cycloalkyl,
C
5 -C7 cycloalkenyl or aryl; wherein each R 22 is independently H, F, Cl, or straight chained or branched Ci-C 4 alkyl; wherein q is an integer from 2 to 4 inclusive; wherein each m is an integer from 0 to 4 inclusive; wherein each n is an integer from 1 to 4 inclusive; 00 96 wherein each p is an integer from 0 to 2 inclusive; I/ wherein U is O, -NR16, S, C(R 17 2 or -NSO 2 R16; wherein Z is C 3
-C
0 cycloalkyl, C 4 -C7 cyclic ether, 00 C 4
-C
7 cyclic thioether, aryl, or heteroaryl; wherein R 16 is straight chained or branched Ci-C 7 0 alkyl, straight chained or branched C 1 -C7 monofluoroalkyl, straight chained or branched Ci-C 7 polyfluoroalkyl, .straight chained or branched C 2 -C7 alkenyl, straight chained or branched C 2
-C
7 alkynyl, Cs-C 7 cycloalkenyl, -(CH 2 or (CH 2 (CH2)m-CH3; wherein B is aryl, or heteroaryl; provided however, if B is aryl or heteroaryl the carbon atom or carbon atoms ortho to the nitrogen atom of the imine bond may only be substituted with one or more of the following -Cl, -Br, -CN, methyl, ethyl or methoxy; or a pharmaceutically acceptable salt thereof.
The invention provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound having the structure: 00 9 000
(NN
wherein each of Y 1
Y
2
Y
3 and Y 4 is independentlystraight chained or branched C 1 -C7 alkyl, monofluoroalky. or polyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or alkynyl; C 3
-C
7 cycloalkyl, or CS-C 7 cycloalkenyl; -C1, -Br, or 1; -NO 2
-N
3 -CN; -OR 4
-SR
4
-OCOR
4
-COR
4
-NCOR
4
N(R
4 2
-CON(R
4 2 or -COOR4* aryl or heteroaryl; or any two of Y 1
Y
2
Y
3 and Y 4 present on adjacent carbon atoms can constitute a methylenedioxy group; wherein each R 4 is independently straight chained or branched Cl-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3
-C
7 cycloalkyl, C 5 -C7 cycloalkenyl, aryl or aryl (Cl-C 6 alkyl; wherein A is A' Q3, Q4, Q-9, straight chained or branched C 1 -C7 alkyl, aryl, heteroaryl, aryl (Cl- CO) alkyl, heteroaryl (C 1
-C
6 alkyl, aryl substituted with an aryl or heteroaryl, heteroaryl substituted with an aryl or heteroaryl; or (CHR 17
-(CHR
17
Z;
wherein A' is 0 n CR 2
R
3 or -(CH 2 n C wherein Q3 is
R
17 R1 N 1 R17 )R 17
R
17 wherein Q4 is wherein Qs is wherein Ri and R 2 are each independently H, straight chained or branched C 1 -C7 alkyl, -Cl, -Br,
NO
2 or -CN; wherein R 3 is H, straight chained or branched C 1
-C
7 alkyl, -Cl, -Br, -NOz, -CN, -OR 6 aryl or heteroaryl; wherein Rs is straight chained or branched Ci-C 7 alkyl, -N(R 4 2
-OR
6 or aryl; 00 100 -n wherein R 6 is straight chained or branched Cl-C.? alkyl or aryl; 00 5 wherein each R 1 7 is independently H; straight chained or branched C 1
-C
7 alkyl, straight chained or branched Cl-C 7 monofluoroalkyl, straight chained or branched 00 IC oyloolysrihchieorbace C2-C 7 poyuralkyl, straight chained or branched alkynyl, C 5
-C
7 cyclpalkenyl, -(CH 2 1 or (CH 2
(CH
2 mCH 3 wherein each R 2 0 is independently straight chained or branched Cl-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3
-C
7 cycloalkyl or C 5
-C
7 cycloalkenyl; -Cl, -Br, or -NO 2
-N
3 -CN;
OR
2 1 -0C0R 21 -C0R 2 1
-NCOR
21
-N(R
21 2 I -CON(R 21 2 or -C00R 2 1 aryl or heteroaryl; or two R 20 groups present on adjacent carbon atoms can join together to form a met hylenedioxy group; wherein each R 2 1 is independently straight chained or branched Cl-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3
-C
1 cycloalkyl, C 5
-C
7 cycloalkenyl, aryl or aryl(Cl-C 6 )alkyl;
I
00 101 wherein each m is an integer from 0 to 4 inclusive; CI wherein each n is an integer from 1 to 4 inclusive; 0 00 5 wherein each p is an integer from 0 to 2 inclusive; Cq 00 wherein U is O, -NR 16 S, C(R 17 2 or -NSO 2
R
1 6 wherein Z is C 3 -CIo cycloalkyl, C 4 -C7 cyclic ether,
C
4
-C
7 cyclic thioether, aryl, or heteroaryl; wherein R 16 is straight chained or branched Ci-C 7 alkyl, straight chained or branched C 1
-C
7 monofluoroalkyl, straight chained or branched CI-C 7 polyfluoroalkyl, straight chained or branched C 2 -C7 alkenyl, straight chained or branched C 2 -C7 alkynyl, Cs-C 7 cycloalkenyl,
-(CH
2 or (CH2)q-O-(CH2 )m-CH 3 wherein q is an integer from 2 to 4 inclusive; wherein B is aryl, heteroaryl, aryl substituted with an aryl or heteroaryl, heteroaryl substituted with an aryl or heteroaryl, tricyclic heteroaryl or Q6; provided however, if B is aryl or heteroaryl the carbon atom or carbon atoms ortho to the nitrogen atom of the imine bond may only be substituted with one or more of the following -Cl, -Br, -CN, methyl, ethyl or methoxy; 102 wherein a tricyclic heteroaryl is a fused three member aromatic system in which one or more of the rings is heteroaryl; carbazole; or acridine; wherein Q6 is wherein each R 22 is independently H, F, Cl, or straight chained or branched C 1
-C
4 alkyl; or a pharmaceutically acceptable salt thereof.
The invention provides a pharmaceutical composition comprising a pharmaceutically acceptable garrier and a compound having the structure: 1 N Y2
A
Y4 wherein each of Yi, Y 2
Y
3 and Y 4 is independently 00 103 H; straight chained or branched Cl-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained CIor branched C 2
-C
7 alkenyl or alkynyl; C 3
-C
7 cycloalkyl, or C 5
-C'
7 cycloalkenyl; -C1, -Br, or 1; -NO 2
-N
3 -CN; -OR 4
-SR
4
-OCOR
4
-CQR
4
-NCOR
4 00 M
N(R
4 2
-CON(R
4 2 or -COOR 4 aryl or heteroaryl; or any two of YI, Y 2
Y
3 and Y 4 present on adjacent 00 carbon atoms can constitute a methylenedioxy group; wherein each R 4 is independently straight chained or branched Cl-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3
-C
7 cycloalkyl, C 5
-C
7 cycloalkenyl, aryl or aryl(Cx-C 6 )alkyl; wherein A is straight chained or branched Cl-C 7 alkyl, aryl, heteroaryl, aryl1(C CO)a 1kyl or heteroaryl (C-CE) alkyl; wherein A' is 0 n nn RUn 'CRR3or -(CH 2
R
00 0 104 wherein Ri and R 2 are each independently H, straight chained or branched
CI-C
7 alkyl, -C1, -Br, 00 M NO2, or -CN; ~0 wherein R 3 is H, straight chained or branched
CI-C
7 alkyl, -Cl, -Br,
-NO
2 -CN, -OR 6 aryl or heteroaryl; wherein Rs is straight chained or branched
CI-C
7 alkyl,
-N(R
4
-OR
6 or aryl; wherein
R
6 is straight chained or branched Ci-C7 alkyl or aryl; wherein B is aryl, or heteroaryl; provided however, if B is aryl or heteroaryl the carbon atom or carbon atoms ortho to the nitrogen atom of the imine bond may only be substituted with one or more of the following -Cl, -Br, -CN, methyl, ethyl or methoxy; wherein n is an integer from 1 to 4 inclusive; or a pharmaceutically acceptable salt thereof.
The invention provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound having the structure: 00 105 000 00 wherein each of Y1, Y 2 1 Y 3 and Y 4 is independently- H; straight chained or branched C, -C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3 -C7 cycloalkyl, or CS-C 7 cycloalkenyl; -C1, -Br, or I; -NO 2
-N
3 -CN; -OR4, -SR 4
-OCOR
4
-COR
4 -NCOR,, N(R42 ,-CON(R 4 2 or -COOR 4 aryl or heteroaryl; or any two of Y 1
Y
2
Y
3 and Y 4 present on adjacent carbon atoms can constitute a methylenedioxy group; wherein each R 4 is independently straight chained or branched Cl-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3 -C7 cycloalkyl, C C-7 cycloalkenyl, aryl or aryl (Cl-C 6 )alkyl; wherein A is straight chained or branched CI-C 7 alkyl, aryl, heteroaryl, aryl (Cl-C 6 alkyl or heteroaryl (C 1
-C
6 alkyl; wherein A' is 0 IM. R 5
I
n CR 2
R
3 or (CH 2 n R 4 wherein B is aryl substituted with an aryl or heteroaryl, heteroaryl substituted with an aryl or heteroaryl, tricyclic heteroaryl or Qs; wherein a tricyclic heteroaryl is a fused three ring aromatic system in which one or more of the rings is heteroaryl; carbazole; or acridine; wherein Q6 is wherein n is an integer from 1 to 4 inclusive; wherein each R 22 is independently H, F, 0 0107 SCl, or straight chained or branched CI-C 4 alkyl; V or a pharmaceutically acceptable salt thereof.
0 The invention provides a pharmaceutical composition 00 M comprising a pharmaceutically acceptable carrier and a compound having the structure: 00 0B ro Y1 N
Y
wherein each of YI, Y 2
Y
3 and Y 4 is independently H; straight chained or branched CI-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched Cz-C 7 alkenyl or alkynyl; C 3
-C
7 cycloalkyl, or C 5 cycloalkenyl; -Cl, -Br, or I; -NO 2
-N
3 -CN; -OR 4 -SR4, -OCOR4, -COR4, -NCOR 4
N(R
4 2
-CON(R
4 2 or -COOR 4 aryl or heteroaryl; or any two of Y 1
Y
2
Y
3 and Y 4 present on adjacent carbon atoms can constitute a methylenedioxy group; wherein each R 4 is independently straight chained or branched C 1
-C
7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3
-C
7 cycloalkyl, Cs-C7 00 108 cycloalkeflyl, aryl or aryl (C 1
-C
6 alkyl; wherein A is Q 3 Q41 Q5, aryl substituted with an aryl or heteroaryl, heteroaryl substituted with an aryl or heteroaryl, or (CHR 17
-(CHR
17 )n-Z; 00 0 wherein Q3 is
NN
00R 1 7 R17 wherein Q4 is RV R 1 7 N n nn wherein Qs is 00 109 00
R
17 00 1 00 wherein each R 17 is independently straight chained or branched C 1
I-C
7 alkyl, straight chained or branched Cl-C 7 monofluorpalkyl, straight chained or branched Cl-C 7 polyfluoroalkyl, straight chained or branched
C
2
-C
7 alkenyl, straight chained or branched C 2
-C
7 alkynyl, C 5
-C
7 cycloalkenyl,
-(CH
2 Or (CH 2
(CH
2
CH
3 wherein each R 20 is independently straight chained or branched
C
1
-C
7 alkyl, monofluoroalkyl or polyfluoroalkyl; Btraight chained or branched C 2
-C
7 alkenyl or alkynyl i C 3
-C
7 cycloalkyl or CS-C 7 cycloalkenyl; -CI, -Br, or -NO 2
-N
3 -CM;
OR
21 -0C0R 21 -C0R 21
-NCOR
2 1 -NCR2l)2 -CON(R 21 2 or -C00R 21 aryl or heteroaryl; or two R 2 0 groups present on adjacent carbon atoms can join together to form a methylenedioxy group; wherein each R 2 1 is independently straight chained or branched Cl-C 7 alkyl, monofluoroalkyl or polyf luoroalkyl; straight -chained or branched C 2
-C
7 alkenyl or alkynyl;
C
3 -C7 cycloalkyl,
C
5
-C
7 cycloalkenyl or aryl;
CN
00) 110 wherein each R 22 is independently H, F, 00 wherein each m is an integer from 0 to 4 inclusive; C0 wherein each n is an integer from 1 to 4 inclusive; 00 o0 wherein each p is an integer from 0 to 2 inclusive; 0D wherein U is O, -NR1s, S, C(Ri 7 2 or -NSO 2
R
16 wherein Z is C 3 -Clo cycloalkyl, C 4
-C
7 cyclic ether,
C
4
-C
7 cyclic thioether, aryl, or heteroaryl; wherein R 16 is straight chained or branched C 1
-C
7 alkyl, straight chained or branched CI-C 7 monofluoroalkyl, straight chained or branched Ci-C 7 polyfluoroalkyl, straight chained or branched C 2 -C7 alkenyl, straight chained or branched C 2
-C
7 alkynyl, Cs-C7 cycloalkenyl,
-(CH
2 or (CH 2 )q-O-(CH 2 )m-CH3; wherein B is aryl, or heteroaryl; provided however, if B is aryl or heteroaryl the carbon atom or carbon atoms ortho to the nitrogen atom of the imine bond may only be substituted with one or more of the following -Cl, -Br, -CN, methyl, ethyl or methoxy; or a pharmaceutically acceptable salt thereof.
00 111 00 MThe invention provides a compound having the structure: 00
B
YY
A N
-O
N"
Y4 wherein each of YI, Y 2
Y
3 and Y4 is independently H; straight chained or branched
C
1
-C
7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched
C
2 -C7 alkenyl or alkynyl;
C
3 -C7 cycloalkyl, or Cs-C 7 cycloalkenyl; -CI, -Br, or I; -NO 2
-N
3 -CN; -OR 4
-SR
4
-OCOR
4
-COR
4 -NCOR4, N(R4)2 -CON(R 4 2 or -COOR 4 aryl or heteroaryl; or any two of Y 1
Y
2
Y
3 and Y 4 present on adjacent carbon atoms can constitute a methylenedioxy group; wherein each R4 is independently straight chained or branched C 1
-C
7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched
C
2
-C
7 alkenyl or alkynyl;
C
3
-C
7 cycloalkyl,
C
5
-C
7 I00 112 cycloalkenyl, aryl or aryl (Cl-C 6 alkyl; wherein A is 03, 04, Q5, straight chained or branched Cl-C 7 alkyl, aryl, heteroaryl, aryl(Cl-
C
6 alkyl, heteroaryl (Cl-C 6 alkyl, aryl substituted with an aryl or heteroaryl, heteroaryl substituted with an aryl or heteroaryl; Or (CHR 17
-(CHR
17
Z;
wherein A' is 0 in n CR 2
R
3 ;or (H )r C wherein 03 is
R
17
R
17 N
_R
17
KR
17 n 113 wherein Q4 is 0 00 wherein Q5 is
R
17
U
17 wherein R, and R 2 are each independently H, straight chained or branched Cl-Cy alkyl, -Cl, -Br,
NO
2 or -CN; wherein R 3 is H, straight chained or branched CI-C 7 alkyl, -Cl, -Br, -NO 2 -CN, -OR 6 aryl or heteroaryl; 00 114 wherein R 5 is straight chained or branched Cl-C 7 alkyl, -N (R 4 2 1 -OR 6 or aryl; 00 5 wherein R 6 is straight chained or branched C 1
-C
7 alkyl or aryl; 00 wherein each R 17 is independently H; straight chained or branched CI-C7 alkyl, straight chained or branched Cl-C 7 mono fluoroalkyl, straight chained or branched Cl-C7 polyfluoroalkyl, straight chained or branched
C
2
-C
7 alkenyl, straight chained or branched
C
2
-C
7 alkynyl, Cs-C.,cycloalkenyl,
-(CH
2 )m1Z, or (CH 2 )1-O-
(CH
2 mCH 3 wherein each R 20 is independently straight chained or branched Cl-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched
C
2
-CI
alkenyl or alkynyl;
C
3
-C-
7 cycloalkyl or CS-C 7 cycloalkenyl; -C1, -Br, or -NO 2
-N
3 -CN;
OR
2 1
-OCOR
21 -C0R 21
-NCOR
21
-N(R
2 3 1 2
-CON(R
2 1 or -C00R 2 aryl or heteroaryl; or two R 20 groups present on adjacent carbon atoms can join together to form a methylenedioXY group; wherein each R 21 is independently straight chained or branched CI-C7 alkyl, monofluoroalkYl or polyfluoroalkyl; straight chained or branched
C
2
-C
7 alkenyl or alkynyl; C 3
-C
7 cycloalkyl,
C
5
-C
7 cycloalkenyl, aryl or aryl(Cl-C6 )alkyl; 00 115 wherein each m is an integer from 0 to 4 inclusive; wherein each n is an integer from 1 to 4 inclusive; 00 wherein each p is an integer from 0 to 2 inclusive; wherein U is O, -NR 16 S, C (R)o 2 or -NS0 2
R
1 us wherein Z is C 3
-C
1 o cycloalkyl, C 4 cyclic ether,
C
4 cyclic thioether, aryl, or heteroaryl; wherein Rse is straight chained or branched Clu-C alkyl, straight chained or branched Cj-C, monofluoroalkyl, straight chained or branched C 1
-C,
polyfluoroalkyl, straight chained or branched
C
2
-C,
alkenyl, straight chained or branched C 2 alkynyl,
C
5 cycloalkenyl,
-(CH
2 )mZ, Or (CH 2 (CH2),mCH3; wherein q is an integer from 2 to 4 inclusive; wherein B is aryl, heteroaryl, aryl substituted with an aryl or heteroaryl, heteroaryl substituted with an aryl or heteroaryl, tricyclic heteroaryl or Qs; provided however, if B is aryl or heteroaryl the carbon atom or carbon atoms ortho to the nitrogen atom of the imine bond may only be substituted with 00 00
C
00
§D
oos 0q mD 0D 0q 116 one or more of the following -Cl, -Br,
-CN,
methyl, ethyl or methoxy; wherein a tricyclic heteroaryl is a fused three member aromatic system in which one or more of the rings is heteroaryl; carbazole; or acridine; wherein Q6 is wherein each R 22 is independently H, F, Cl, or straight chained or branched
CI-C
4 alkyl; or a pharmaceutically acceptable salt thereof.
The invention provides a compound having the structure:
B
wherein each of Yi, Y 2
Y
3 and Y 4 is independently H; straight chained or branched
C
1 -C7 alkyl, 00 117 monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3
-C
7 cycloalkyl, or C5-C 7 cycloalkenyl; -C1, -Br, or 1; -NO 2
-N
3 -CN; -OR 4
-SR
4
-OCOR
4
-COR
4
-NCOR
4
N(R
4 2
-CON(R
4 2 or -COOR 4 aryl or heteroaryl; or 00 M~f any two of Y 1
Y
2
Y
3 and Y 4 present on adjacent carbon atoms can constitute a methylenedioxy group; 00 wherein each R 4 is independently straight chained or branched CI-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3 -C-7 cycloalkyl,
C
5
-C
7 cycloalkenyl, aryl or aryl(Cl-C 6 )alkyl; wherein A is straight chained or branched CI-C 7 alkyl, aryl, heteroaryl, aryl (Cl-C 6 alkyl or heteroaryl (C 1 -Cs) alkyl; wherein A' is 0 0 IIn
R
1 'C23or -(CH 2
R
4 118 wherein Ri and R 2 are each independently H, straight chained or branched Ci-C 7 alkyl, -Cl, -Br, -I,
NO
2 or -CN; 00 M wherein R 3 is H, straight chained or branched C 1
-C
7 alkyl, -Cl, -Br,
-NO
2 -CN, -OR 6 aryl or 00 heteroaryl; C< 10 wherein Rs is straight chained or branched Ci-C 7 alkyl, 2 -ORe or aryl; wherein Rs is straight chained or branched CI-C 7 alkyl or aryl; wherein B is aryl, or heteroaryl; provided however, if B is aryl or heteroaryl the carbon atom or carbon atoms ortho to the nitrogen atom of the imine bond may only be substituted with one or more of the following -Cl, -Br, -CN, methyl, ethyl or methoxy; wherein n is an integer from 1 to 4 inclusive; or a pharmaceutically acceptable salt thereof.
The invention provides a compound having the structure: 00 119 000 00 wherein each of Y 1
Y
2
Y
3 and Y 4 is independently- H; straight chained or branched Cl-C 7 alkyl, monofluoroalky. or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkeny. or alkynyl; C 3
-C
7 cycloalkyl, or CS-C 7 cycloalkenyl; -C1, -Br, or 1; -NO 2
-N
3 -C2N; -OR 4 -SR4, -OCOR4, -COR4, -NCOR 4
N(R
4 2 I -CON(R4) 2 or -COOR 4 aryl or heteroaryl; or any two of Y1, Y 2
Y
3 and Y 4 present on adjacent carbon atoms can constitute a methylenedioxy group; wherein each R 4 is independently straight chained or branched C 1 -C7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C,
alkenyl or alkynyl; C 3
-C
7 cycloalkyl, C 5
-C
7 cycloalkenyl, aryl or aryl (Cl-C 6 alkyl; wherein A is straight chained or branched CI-C7 alkyl, aryl, heteroaryl, aryl (Cl-C 6 alkyl or heteroaryl (Cl-C 6 alkyl; wherein A' is 0 n R R RI or (CH2)n R4, wherein B is aryl substituted with an aryl or heteroaryl, heteroaryl substituted with an aryl or heteroaryl, tricyclic heteroaryl or Qs; wherein a tricyclic heteroaryl is a fused three ring aromatic system in which one or more of the rings is heteroaryl; carbazole; or acridine; wherein Q6 is wherein n is an integer from 1 to 4 inclusive; 121 wherein each R 22 is independently H, Cl, or straight chained or branched CI-C 4 alkyl; or a pharmaceutically acceptable salt thereof.
The invention provides a compound having the structure: wherein each of Yi, Y 2
Y
3 and Y4 is independently H; straight chained or branched Ci-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3 -C7 cycloalkyl, or Cs-C 7 cycloalkenyl; -C1, -Br, or I; -NO 2
-N
3 -CN; -OR 4
-SR
4
-OCOR
4
-COR
4 -NCOR,
N(R
4 2
-CON(R
4 2 or -COOR 4 aryl or heteroaryl; or any two of Yi, Y 2
Y
3 and Y 4 present on adjacent carbon atoms can constitute a methylenedioxy group; wherein each R 4 is independently straight chained or branched Ci-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 122 alkeny. or alkynyl; C 3
-C
7 cycloalkyl, cycloalkenyl, aryl or aryl (Cl-C 6 alkyl; C C 7 wherein A is Q 3 Q4, Q5, aryl. substituted with an aryl. or heteroaryl, heteroaryl substituted with an aryl or heteroaryl, or (CHR, 7
-(CHR,
7 wherein Q3 is wherein Q4 is wherein Qs is 123
R
1 7 c-I
U
00 17 00 wherein each R 17 is independently H; straight chained or branched C3 1 alkyl, straight chained or branched Cl-C 7 mono fluoroal kyl, straight chained or branched Cl-C 7 polyfluoroalkYl, straight chained or branched
C
2
-C
7 alkenyl, straight chained or branched
C
2
-C-I
alkynyl,
C
5
-C
7 cycloalkenyl,
-(CH
2 or (CH 2 )n-O-
(CH
2 mCH 3 wherein each R 2 0 is independently straight chained or branched C3 1
-C
7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched
C
2
-C
7 alkenyl or alkynyl;
C
3
-C
7 cycloalkyl or Cs- C 7 cycloalkelyl; -C1, -Br, or
-NO
2
-N
3 -CN;
OR
2 1 -0C0R 2 1 -C0R 21
-NCOR
21
-N(R
21
-CON(R
21 or
-COOR
2 1; aryl or heteroaryl; or two R 2 0 groups present on adjacent carbon atoms can join together to form a methylenedioxCy group; wherein each R 21 is independently straight chained or branched Cl-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched
C
2 -C7 alkenyl or alkynyl;
C
3
-C
7 cycloalkyl,
C
5
-C
7 cycloalkenyl or aryl; 00 S124 Swherein each R 22 is independently H, F, Cl, or straight chained or branched Ci-C 4 alkyl; wherein q is an integer from 2 to 4 inclusive;
S
M wherein each m is an integer from 0 to 4 inclusive; 00 wherein each n is an integer from 1 to 4 inclusive; o 10 wherein each p is an integer from 0 to 2 inclusive; wherein U is O, -NR16, S, C(R 17 2 or -NSO 2
R
1 6 wherein Z is C 3 -Cio cycloalkyl,
C
4 cyclic ether,
C
4 -C7 cyclic thioether, aryl, or heteroaryl; wherein R 16 is straight chained or branched Cl-C7 alkyl, straight chained or branched
C
1
-C,
monofluoroalkyl, straight chained or branched C 1
-C,
polyfluoroalkyl, straight chained or branched C 2 -C7 alkenyl, straight chained or branched C 2
-C
7 alkynyl,
C
5
-C
7 cycloalkenyl,
-(CH
2 or (CH 2 (CH2)m-CH3; wherein B is aryl, or heteroaryl; provided however, if B is aryl or heteroaryl the carbon atom or carbon atoms ortho to the nitrogen atom of the imine bond may only be substituted with one or more of the following -Cl, -Br, -CN, methyl, ethyl or methoxy; or a pharmaceutically acceptable salt thereof.
00 125
CI
SThe invention provides a method of treating depression in a subject which comprises administering to the subject a V composition comprising a pharmaceutically acceptable carrier and a therapeutically effective amount of a GAL3 receptor antagonist, wherein: 00 the GAL3 receptor antagonist binds to the human SGAL3 receptor with a binding affinity at least C ten-fold higher than the binding affinity with 00 Swhich it binds to the human GAL1 receptor; the GAL3 receptor antagonist does not inhibit the activity of central monoamine oxidase A greater than 50 percent, at a concentration of 10M; and the GAL3 receptor antagonist does not inhibit the activity of central monoamine oxidase B greater than 50 percent, at a concentration of and the GAL3 receptor antagonist binds to the human GAL3 receptor with a binding affinity at least tenfold higher than the binding affinity with which it binds to each of the following transporters: serotonin transporter, norepinephrine transporter, and dopamine transporter.
The invention provides a method of treating anxiety in a subject which comprises administering to the subject a composition comprising a pharmaceutically acceptable carrier and a therapeutically effective amount of a GAL3 receptor antagonist, wherein: the GAL3 receptor antagonist binds to the human GAL3 0 0
O
0 126 Sreceptor with a binding affinity at least ten-fold higher than the binding affinity with which it binds V' to the human GAL1 receptor; and
(N
the GAL3 receptor antagonist binds to the human GAL3 receptor with a binding affinity at least ten-fold 00 higher than the binding affinity with which it binds Sto each of the following transporters: serotonin C1 transporter, norepinephrine transporter, and 00 Sdopamine transporter.
0D
(N
00 127 SBrief Description of the Figures C< Figure 1: Rat Forced Swim Test Results (Immobility: Normal Rats) 00 5 Vehicle and test compounds (F10 fluoxetine at C mg/kg ip; Cl, C3, C10 or C30 Example 92 at 1, 3, 10 or 0 30 mg/kg ip) were injected into normal rats by OO intraperitonal administration (n 5 for each treatment Scondition). One hour later, rats were examined in a minute forced swim test. For each treatment condition, the number of 5-sec intervals culminating with a display of immobility was derived and plotted as the average S.E.M. A significant decrease in immobility was observed for rats injected with fluoxetine at 10 mg/kg, or with Example 92 at 3 and 10 mg/kg, relative to vehicle injected controls (p 0.01, ANOVA and Student-Nerman- Keuls).
Figure 2: Rat Forced Swim Test Results (Climbing: Normal Rats) Vehicle and test compounds (F10 fluoxetine at mg/kg ip; C1, C3, C10 or C30 Example 92 at 1, 3, 10 or mg/kg ip) were injected into normal rats by intraperitonal administration (n 5 for each treatment condition). One hour later, rats were examined in a minute forced swim test. For each treatment condition, the number of 5-sec intervals culminating with a display of climbing was derived and plotted as the average S.E.M. A significant increase in climbing was observed for rats injected with Example 92 at 10 mg/kg, relative to vehicle injected controls (p 0.01, ANOVA and 00 128 Student-Nerman-Keuls), but not in rats dosed with Example S92 at 30 mg/kg ip.
Figure 3: Rat Forced Swim Test Results (Swimming: Normal 00 5 Rats) 3 Vehicle and test compounds (F10 fluoxetine at mg/kg ip; C1, C3, C10 or C30 Example 92 at 1, 3, 10 or 00 30 mg/kg ip) were injected into normal rats by Sintraperitonal administration (n 5 for each treatment condition). One hour later, rats were examined in a minute forced swim test. For each treatment condition, the number of 5-sec intervals culminating with a display of swimming was derived and plotted as the average S.E.M. A significant increase in swimming was observed for rats injected with fluoxetine at 10 mg/kg ip or with Example 92 at 30 mg/kg, relative to vehicle injected controls (p 0.01, ANOVA and Student-Nerman-Keuls).
Figure 4: Social Interaction Test Results (Social Interaction: Unfamiliar Rats) Vehicle and test compounds (CLD 5 chlordiazepoxide at 5 mg/kg ip; C10, C30 or C100 Example 92 at 10, 30 or 100 mg/kg ip) were injected into normal rats by intraperitonal administration (n 5 for each treatment condition). One hour later, unfamiliar rats were examined in a 15 minute social interaction test. For each treatment condition, the amount of time spent in social interaction was derived and plotted as the average S.E.M. A significant increase in social interaction was observed for rats injected with chlordiazepoxide at mg/kg i.p. or with Example 92 at 10 mg/kg ip (p 0.05) as well as 30 mg/kg (p 0.01). When the dose of Example 00 129 92 was increased to 100 mg/kg, the amount of social interaction time was significantly less than measured
C
N after chlordiazepoxide at 5 mg/kg ip or Example 92 at mg/kg ip (p 0.01). Significance in all cases was 00 5 determined by ANOVA and Student-Nerman-Keuls.
0 Figure 5: Western Blot Results 00 In order to establish the specificity of the anti-GAL3 Santiserum, membranes prepared from COS-7 cells transiently transfected with the rat recombinant GAL3 (Borowsky et al., 1999) .(Lane 2) or mock-transfected (vector only) (Lane 3) were applied to an SDS-PAGE gel and blotted using the GAL3 receptor polyclonal antibody.
Lane 1 corresponds to molecular weight marker. The anti- GAL3 antiserum labeled proteins in membranes only from rat GAL3-transfected cells (Lane a predominant band was evident with an apparent molecular weight of approximately 56 kDa, (somewhat higher than the amino acid-derived value of, 40.4 kDa). The apparently high molecular weight observed for rat GAL3 very likely reflects post-translational processing such as glycosylation; note that rat GAL3 contains multiple
N-
terminal glycosylation sites (Smith et al., 1998).
Relative to the predominant band, additional species of higher molecular weight as well as lower molecular weight were labeled by the GAL3 antiserum. These are interpreted as protein aggregates of C-terminal fragments, as they are absent in mock-transfected cells.
130 Detailed Description of the Invention The present invention provides a method of treating a subject suffering from depression which comprises administering to the subject an amount of compound effective to treat the subject's depression wherein the compound has the structure:
X
Y N N
H
wherein W is H, -Cl, -Br, CN, methyl, ethyl, propyl, methoxy or ethoxy; wherein X is; NR 11
R
12 R17
R
1 7
N
or wherein
R
1 3 is H, straight chained or branched
C
1
-C
7 alkyl, 131
(CH
2 )q0-(CH 2 mCH3, aryl, or aryl. (Cl-C 6 alkyl; wherein R 1 2 is straight chained or branched C 1
-C
7 alkyl,
(CH
2 )q-O4CH2)m-CH3, or -(CH 2 )M-Zj wherein R- 1 3 is a bicyclic alkyl ring system, adamantyl, noradamantyl,
C
3
-C
10 cycloalkyl, heteroaryl, aryl, aryl(Cl-
C
6 alkyl, Q, or Q2; wherein aryl may be substituted with one or more Cj-C.O straight chained or branched alkyl, aryl, heteroaryl, of N (R 19
Z;
wherein Q, is wherein Q2 is
R
22
R.
R
22 wherein each J is independently 0, S, C(R 2 2 2 or NR 4 132 wherein R 4 is H; straight chained or branched Cl-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 alkenyl or alkynyl; C 3
-C
7 cycloalkyl, C.5-C 7 cycloalkenyl or aryl; wherein Y is NR 1 4
R
1 5 ;or wherein R 14 is H, straight chained or branched Cl-C 6 alkyl,
(CH
2 )q0- CH2) m-CH3I C 3
-C
6 cycloalkyl, or (C (Rig) 2
-Z;
wherein R 15 is straight chained or branched C 3
-C
6 alkyl,
(CH
2 )q0O(CH 2 )m-CH3i C 3 -CIS cycloalkyl, (C (Rig) 2 )MN (R, 6 2 or (C (Rig) 2 -Z wherein R 16 is straight chained or branched Cl-C7 alkyl, 00 133 straight chained or branched C 1 monofluoroalkyl, straight chained or branched C 1 polyfluoroalkyl, straight chained or branched C 2 alkenyl, straight chained or branched C 2 alkynyl, C 5 cycloalkenyl,
(CH
2 or (CH 2 ),q0ICH 2 )m-CH3; 00 wherein each R 1 7, is independently H; -OR 21 -0C0R 21 -CaR 21 00 -NCOR2 1
-N(R
2 1 2
-CON(R
21 b2, -C00R 21 straight chained or branched Cl-C., alkyl, straight chained or branched Cl-C, monofluoroalkyl, straight chained or branched CI-C., polyfluoroalkyl, straight chained or branched C 2 -Ci alkenyl, straight chained or branched C 2 alkynyl, Cs-C, cycloalkenyl,
-(CH
2 or (CH 2 )1n-0-(CH 2 )m,-CH 3 wherein R 18 is straight chained or branched
CI-C
6 alkyl,
(CH
2 or (CH 2 )q-O-(CH 2 ),-CH3; wherein each Rig is independently H, or straight chained or branched C 1
-C
6 alkyl; wherein each R 2 0 is independently straight chained or branched Cl-C, alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 alkenyl or alkynyl; C 3 cycloalkyl or C 5 cycloalkenyl; -C1, -Br, or -I;
-NO
2 -N3; -CN; -OR 2 1 -OCOR2., -COR 21
-NCOR
21
-N(R
21 2 CON (R 2 1 2 or -C00R 21 aryl or heteroaryl; or two R 20 groups present on adjacent carbon atoms can join together to form a methylenedioxY group; wherein each R 21 is independently straight chained or branched CI-C, alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 alkenyl or alkynyl; C 3 00 134 SC 7 cycloalkyl, Cs-C 7 cycloalkenyl, aryl, or aryl (C 1
C
6 )alkyl; wherein each R 22 is independently H, F, Cl or Ci-C 4 0 5 straight chained or branched alkyl; 00 Swherein each m is an integer from 0 to 4 inclusive; 00 Swherein each n is an integer from 1 to 4 inclusive; I wherein p is an integer from 0 to 2 inclusive; wherein q is an integer from 2 to 4 inclusive; wherein t is 1 or 2; wherein U is O, -NR 16 S, C(Rr7) 2 or -NSO 2
R
16 wherein Z is C 3 -Cio cycloalkyl,
C
4
-C
7 cyclic ether, C 4
-C
7 cyclic thioether, aryl, or heteroaryl; or a pharmaceutically acceptable salt thereof.
The invention provides a method of treating a subject suffering from depression which comprises administering to the subject an amount of compound effective to treat the subject's depression wherein the compound has the structure: 00 00 00 135 wherein W is H, -Cl, -Br, CN, methyl, ethyl, propyl, methoxy or ethoxy; wherein X is NR 11
R
12
R
1 R17 -N ;or -N N-R
R
17 wherein R 1 3. is H, straight chained or branched Cl-C 7 alkyl, (CH2)q-O
(CH
2 )m-CH 3 aryl or aryl (Cl-Cs) alkyl; wherein R 12 is straight chained or branched Cl-C7 alkyl,
(CH
2 )q0I-CH2)m-CH3, or -(CH 2
)M-Z;
wherein
R
13 is a bicyclic alkyl ring system, aryl or aryl (Cl-C 6 alkyl; wherein Y is NR 14
R
15 00 136
R,
0
R
2 0 0- N U R p o 00
R
2 0
-N-
wherein R 1 4 is H, straight chained or branched Cl-C 6 alkyl,
(CH
2 )q0O(CH2),-CH3I
C
3
-C
6 cycloalkyl, or (C(Rig) 2 wherein R 15 is straight chained or branched
C
3
-C
6 alkyl,
(CH
2 )q01-CH 2 )CCH3: C 3
-C
6 cycloalky., or (C(R 19 2 wherein U is 0, -NRh.6, S, C(R 1 7 2 or -NS0 2 R6; wherein Z is C 3 -CIO cycloalkyl, aryl, or heteroaryl; wherein R 16 is straight chained or branched
C-
1
-C
7 alkyl, straight chained or branched
CI-C
7 monofluoroalkyl, straight chained or branched Cl-C 7 polyfluoroalkyl, straight chained or branched
C
2
-C
7 alkenyl, straight chained or branched
C
2
-C
7 alkynyl, C5-C 7 cycloalkenyl,
(CH
2 or (CH 2 )q0(C2)m-CH3; 00 137 wherein each R 1 is independently H; -OR 21 -OCOR21, -COR 2 1
-NCOR
21
-N(R
21 2
-CON(R
21 2
-COOR
21 straight chained or CI branched C 1 alkyl, straight chained or branched C 1
-C,
monofluoroalkyl, straight chained or branched C 1
-C
7 00 5 polyfluoroalkyl, straight chained or branched C 2
-C,
0C M alkenyl, straight chained or branched C 2 -C7 alkynyl, C 5
-C
7 cycloalkenyl,
-(CH
2 or (CH 2 )n-O-(CH 2 )m-CH 3 00 O wherein R 1 8 is straight chained or branched C 1
-C
6 alkyl,
(CH
2 or (CH 2 (CH2)m-CH3; wherein each R 19 is independently H, or straight chained or branched C 1 -Cs alkyl; wherein each R 20 is independently straight chained or branched C 1 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or alkynyl; C 3 C, cycloalkyl or C 5 cycloalkenyl; -C1, -Br, or -I;
-NO
2
-N
3 -CN; -OR 21 -OCOR21, -COR 21
-NCOR
2 1
-N(R
21 2
CON(R
2 1 or -COOR21; aryl or heteroaryl; or two R 2 0 groups present on adjacent carbon atoms can join together to form a methylenedioxy group; wherein each R 2 1 is independently straight chained or branched C 1 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 alkenyl or alkynyl; C3- C, cycloalkyl,
C
5 cycloalkenyl, aryl or aryl(C 1
C
6 alkyl; wherein each m is an integer from 0 to 4 inclusive; wherein each n is an integer from 1 to 4 inclusive; 138 wherein p is an integer from 0 to 2 inclusive; wherein q is an integer from 2 to 4 inclusive; wherein t is 1 or 2; or a pharmaceutically acceptable salt thereof.
The invention provides a method of treating a subject suffering from depression which comprises administering to the subject an amount of compound effective to treat the subject's depression wherein the compound has the structure: wherein W is H, -Cl, -Br, CN, methyl, ethyl, propyl, methoxy or ethoxy; wherein X is N(CH 3 2 or 139 wherein R- 1 3 is an aryl, adamantyl, noradamantyl,
C
3
-C
10 cycloalkyl, heteroaryl, Q, or Q2; wherein aryl, may be substituted with one or more C 1 Ci 0 straight chained or branched alkyl, aryl, heteroaryl, *or N (Rig) Z, wherein Q, is wherein Q2 is
*R
20 wherein each J is independently 0, S, C(R 2 2 2 or NR 4 140 wherein R 4 is straight chained or branched Ci-C-7 alkyl, monof2.uoroalkyl or polyfluoroalkyl; straight chained or branched
C
2 -C7 alkenyl or alkynyl;
C
3
-C
7 cycloalkyl,
C
5
-C
7 cycloalkenyl or aryl; wherein Y is NR 14
R
15
R
17 -N U /p 1 or wherein
R
14 is H, straight chained or branched Cl-C 6 alkyl,
(CH
2 )q0I-CH2)m-CH3,
C
3
-C
6 cycloalkyl, or (C(R 1 9 2
)M-Z;
wherein Ris is straight chained or branched
C
3
-C
6 alkyl,
(CH
2 )q0'-(CH 2 )m.CH3I C 3
-C
6 cycloaJlkyl, or (C(Rlg)2)m,-Z; wherein U is 0, -NR16, S, C(Rl 7 or -NS0 2
R,
6 wherein Z is C 3 -Cl 0 cycloalkyl, aryl, or heteroaryl; 00 141 wherein R 16 is straight chained or branched C 1 alkyl, Cl straight chained or branched C 1 monofluoroalkyl, straight chained or branched C 1 polyfluoroalkyl, 00 5 straight chained or branched C 2 -C7 alkenyl, straight 0C Mr chained or branched C 2 -C7 alkynyl, C 5
-C
7 cycloalkenyl,
(CH
2 or (CH 2
(CH
2 ),-CH3; 00 wherein each R17 is independently H; -OR 21
-OCOR
2 1
-COR
21 -NCOR21, -N(R 21 2
-CON(R
2 1 2
-COOR
2 1 straight chained or branched C 1 alkyl, straight chained or branched C 1
-C,
monofluoroalkyl, straight chained or branched Cl-C, polyfluoroalkyl, straight chained or branched
C
2 -C7 alkenyl, straight chained or branched C 2 -C7 alkynyl, C 5
-C,
cycloalkenyl,
-(CH
2 or (CH 2 )n-O-(CH 2 )m-CH 3 wherein R 18 is straight chained or branched C 1
-C
6 alkyl,
(CH
2 Or (CH 2 )q-O-(CH2)mCH3; wherein each R 19 is independently H, or straight chained or branched Cl-Cs alkyl; wherein each R 20 is independently straight chained or branched C 1 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 alkenyl or alkynyl; C 3 C, cycloalkyl or C 5 cycloalkenyl; -C1, -Br, or -I; -NOz 2
-N
3 -CN; -OR 21 -0COR 2 1
-COR
21
-NCOR
21
-N(R
21 2
CON(R
2 1 or -COOR 21 aryl or heteroaryl; or two R 20 groups present on adjacent carbon atoms can join together to form a methylenedioxy group; wherein each R 21 is independently straight chained or 00 142 Sbranched
C
1 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3 Cl C, cycloalkyl, Cs-C, cycloalkenyl, aryl or aryl(C 1
C
6 alkyl; 0 0 wherein each R 22 is independently H, F, Cl or C1-C4 O straight chained or branched alkyl; 0 0 wherein each m is an integer from 0 to 4 inclusive; wherein each n is an integer from 1 to 4 inclusive; wherein p is an integer from 0 to 2 inclusive; wherein q is an integer from 2 to 4 inclusive; wherein t is 1 or 2; or a pharmaceutically acceptable salt thereof.
The invention provides a method of treating a subject suffering from depression which comprises administering to the subject an amount of compound effective to treat the subject's depression wherein the compound has the structure: wherein W is H, -Cl, -Br, CN, methyl, ethyl, 00 143 propyl, methoxy or ethoxy; CI wherein X is N,(CH 3 2 or
R
17 00
R
17 wherein R13 is a bicyclic alkyl ring System, aryl *or aryl (C 1
-C
6 alkyl; wherein Y is NR 14
R
1 5 wherein R 14 is H, straight chained or branched
CI-C
6 alkyl,
(CH
2 )q-O(CH 2 )m-CH3, C 3
-C
6 cycloalkyl, or (C(R 1 9 2 )m-Z; wherein R 15 s is (C(R 1 9 2 ).-N(Rl6)2; wherein Z is C 3
-C
1 o cycloalkyl, aryl, or heteroaryl; wherein
R
16 is straight chained or branched Cl-C 7 alkyl, straight chained or branched CI-C7 monofluoroalkyl, straight chained or branched Cl-C, polyfluoroalkyl, straight chained or branched
C
2 -C,7 alkenyl, straight chained or branched
C
2
-C
7 alkynyl, Cs-C7 cycloalkenyl,
(CH
2 or (CH2)q0 O(CH 2 )m-CH 3 wherein each R 17 is independently H; -OR 21 -0C0R 21 -C0R 21
-NCOR
21
-N(R
21 2
-CON(R
2 1D2, -C00R 21 straight chained or branched
CI-C
7 alkyl, straight chained or branched Cl-C 7 monofluoroalkyl, straight chained or branched Cl-C 7 00 144 Spolyfluoroalkyl, straight chained or branched C 2 -C7 alkenyl, straight chained or branched C 2 -C7 alkynyl, Cs-C, CN cycloalkenyl, -(CH 2 or (CH 2
(CH
2 )m-CH 3 O 5 wherein each R 19 is independently H, or straight chained 00 M or branched Ci-C 6 alkyl; OO wherein each R 21 is independently straight chained or branched Ci-C7 alkyl, monofluoroalkyl or polyfluoroalkyl; C 10 straight chained or branched C 2 alkenyl or alkynyl; C3- C, cycloalkyl, Cs-C7 cycloalkenyl, aryl or aryl(C 1 Cs)alkyl; wherein each m is an integer from 0 to 4 inclusive; wherein each n is an integer from 1 to 4 inclusive; wherein q is an integer from 2 to 4 inclusive; or a pharmaceutically acceptable salt thereof.
As used in the present invention, the term "bicyclic alkyl ring systems" includes, but is not limited to, bicyclo[2.2.1]heptane, bicyclo[3.1.1]heptane and bicyclo[2.2.2]octane. In addition, the bicyclic alkyl ring systems may be substituted with one or more of the following:
-NO
2 -CN, straight chained or branched Ci-C, alkyl, straight chained or branched Ci-C7 monofluoroalkyl, straight chained or branched C 1 -C7 polyfluoroalkyl, straight chained or branched C 2 -C7 alkenyl, straight chained or branched C 2
-C
7 alkynyl, C 3 -C7 cycloalkyl, C 5 cycloalkenyl,
-N(R
21 2
-OR
21
-COR
21 00 145 C0 2
R
2 1
-CON(R
21 2 or (CH 2 n-O- (CH 2 )m CH 3 CI As used in the present invention, the term "cycloalkyl" includes, C 3
-C
7 cycloalkyl moieties which may be 0 5 substituted with one or more of the following: -NO 2 00 Mr) -CN, straight chained or branched C 1
-C
7 alkyl, straight 0chained or branched C 1
-C
7 monofluoroalkyl, straight 00 chained or branched C 1 -C7 polyfluoroalkyl, straight 00taih chained or bace oyloolysrih chained or branched C 2
-C
7 alkenyl, straight chained or branched C 2 C7 alkynyl, C 3
-C
7 cycloalkyl, C 3
-C,
monofluorocycloalkyl,
C
3 polyfluorocycloalkyl,
C
5
-C
7 cycloalkenyl,
-N(R
4 2
-OR
4 -COR4, -NCOR 4 -CO2R 4
CON(R
4 2 or (CH 2
(CH
2 )m-CH 3 As used in the present invention, the term "cyclohexyl" includes, cyclohexyl groups which may be substituted with one or more of the following: -NO 2 -CN, straight chained or branched C 1
-C
7 alkyl, straight chained or branched C 1
-C
7 monofluoroalkyl, straight chained or branched C 1 polyfluoroalkyl, straight chained or branched C 2 alkenyl, straight chained or branched C 2 -C7 alkynyl, C 3
-C
7 cycloalkyl, C 3 -C7 monofluorocycloalkyl,
C
3
-C,
polyfluorocycloalkyl, Cs-C, cycloalkenyl,
-N(R
4 2 -OR4,
COR
4 -NCOR4, -CO 2 R4, -CON(R4) 2 or (CH 2
(CH
2 )m-CH 3 As used in the present invention, the term "cycloalkenyl" includes, Cs-C 7 cycloalkenyl moieties which may be substituted with one or more of the following: -Cl, -Br, -NO 2 -CN, straight chained or branched C 1
-C
7 alkyl, straight chained or branched C 1
-C
7 monofluoroalkyl, straight chained or branched C 1
-C
7 polyfluoroalkyl, straight chained or branched C 2
-C
7 alkenyl, straight 00 146 chained or branched C 2
-C
7 alkynyl, C 3
-C
7 cycloalkyl,
C
3
-C
7 monofluorocycloalkyl,
C
3
-C
7 polyfluorocycloalkyl,
CS
5
C
7 CIcycloalkenyl,
N(R
4 2
OR
4 COR4, -NCOR 4 -C0 2 R4,-
CON(R
4 2 or (CH 2 )n-O-(CH 2 )m-CH 3 00 M In the present invention, the term "heteroaryl" is used to include five and six membered Unsaturated rings that 00 may contain one or more oxygen, sulfur, or nitrogen atoms. Examples of heteroaryl groups include, but are not limited to, furanyl, thienyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazoiyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, and triazinyl.
In addition the term "heteroaryl" is used to include fused bicyclic ring systems that may contain one or more heteroatoms such as oxygen, sulfur and nitrogen. Examples of such heteroaryl groups include, but are not limited to, indolizinyl, indolyl, isoindolyl, benzolfuranyl, benzo Eb] thiophenyl, indazolyl, benzimidazolyl, purinyl, benzoxazolyl, benzisoxazolyl, benzo Eb]thiazolyl, imidazo i-b] thiazolyl, cinnolinyl, quinazolinyl, quinoxalinyl, 1, 8-naphthyridinyl, pteridinyl, quinolinyl, isoquinolinyl, phthalimidyl and 2,l,3-benzothiazolyl.
The term "heteroaryl" also includes those chemical moieties recited above which may be substituted with one or more of the following: -Cl, -Br,
-NO
2
-CM,
straight chained or branched Cl-C 7 alkyl, straight chained or branched
C
1
-C
7 mono fluoroa lkyl, straight chained or branched Cl-C 7 polyfluoroalkyl, straight chained or 147 branched C 2 alkenyl, straight chained or branched C 2
-C
7 alkynyl, C 3
-C
7 cycloalkyl, C 3
-C
7 monofluorocycloalkyl,
C
3
-C,
CN polyfluorocycloalkyl, Cs-C7 cycloalkenyl, -N(R 4 2
-OR
4
COR
4
-NCOR
4 -C0 2
R
4
-CON(R
4 2 or (CH 2
(CH
2 )m-CH 3 00 SThe term "heteroaryl" further includes the N-oxides of 0 those chemical moieties recited above which include at 00 least one nitrogen atom.
In the present invention the term "aryl" is phenyl or naphthyl. The term "aryl" also includes phenyl and naphthyl which may be substituted with one or more of the following: -Cl, -Br, -N02, -CN, straight chained or branched CI-C 7 alkyl, straight chained or branched CI-C7 monofluoroalkyl, straight chained or branched C 1 -C7 polyfluoroalkyl, straight chained or branched
C
2
-C
7 alkenyl, straight chained or branched C 2 alkynyl, C 3 -C7 cycloalkyl,
C
3
-C
7 monofluorocycloalkyl,
C
3
-C
7 polyfluorocycloalkyl, Cs-C 7 cycloalkenyl,
-N(R
4 2
-OR
4 SR4, -OCOR4, -COR4, -NCOR 4
-CO
2
R
4
-CON(R
4 2 or (CH 2 )n-O-
(CH
2 )m-CH 3 In one embodiment of any of the methods described herein, the compound is enantiomerically and diasteriomerically pure. In one embodiment, the compound is enantiomerically or diasteriomerically pure.
In one embodiment of any of the methods described herein, the compound can be administered orally.
In one embodiment, X is: 148 R17 R 17
-N
R17 -N N-RIB 17 or In one embodimenlt, X is NRllRl2 and Ril is H or straight chained or branched Cl-C-? alkyl.
In one embodiment, the compound has the structure: In one embodiment,
R
13 is a bicyclic cyclohexyl or aryl.
In one embodiment,
R
1 4 is H, straight Cl-C 6 alkyl or (CH 2 )q0O(CH2)m-CH3- In one embodiment,
R
14 is H, straight Cl-C 6 alkyl or (CH 2 )qO(C2)MCH3alkyl ring system, chained or branched chained or branched 00 149 In one embodiment, the compound is selected from the group consisting of: 00Q 00ON AN$ 0
Q
25 Jan 2008 2008200380
Z
zZ
*O
2008200380 25 Jan 2008 U' H 0 2008200380 25 Jan 2008
F
zO ,-z z~ 00 153
R
17 00 1 00 In one embodiment, U is NR 1 s.
In one emnbodiment,
R
16 is (CH 2
MZ.
In one embodiment, Z is aryl or heteroaryl.
In one embodiment, the compound is selected from the lo group consisting of: Qo 2008200380 25 Jan 2008
U,
Z.>
2008200380 25 Jan 2008 z K\
Z
131~!
C
156 In one embodiment, the compound is selected from the group consisting of:
N
Cl 'k
N
N N NZ.- 2008200380 25 Jan 2008 00 In one embodiment, Y is 00 N U 00 In one embodiment, U is NR 1 6 In one embodiment, the compound is 11. ;i ,or 159 In one embodiment, the compound is In one embodiment, the compound is selected from the group consisting of: 160
N
N
N C1 N N C 1
JN
NN
1" CI N' KNNQ' 1N"- N CI N N ;and In one embodiment, the compound is selected from the group consisting of: 00 16.
N~ N rNN N rN'IIN'" C,
''N
00
NN
C-I V0,F 3 c-IN 00
NNN
NN
(N4" QOO Q-cA-NQON eyN,
NO
In one embodiment, X is N(CH 3 2 In one emibodiment, Y is 00 00 j In one embodiment, R 13 is an aryl substituted with a C 1 -Cio straight chained alkyl.
In one embodiment, the compound is selected from a group consisting of:
NN
NN
r N" NN ;and
\N
InoeebdmnRi sa rlsbtiue ihaC-i 163 The invention provides a method of treating a subject suffering from anxiety which comprises administering to the subject an amount of compound effective to treat the subject's anxiety wherein the compound has the structure: wherein W is H, -Cl, -Br, CN, methyl, ethyl, propyl, methoxy or ethoxy; wherein X is; NR11Ria;
R
17 R 17
N
R
17 or N-Rial wherein R 11 is H, straight chained or branched Ci-C 7 alkyl,
(CH
2 )q-O-(CH2 )m-CH3, aryl, or aryl (C 1 -Cs)alkyl; 164 wherein R 12 is straight chained or branched Cl-C-7 alkyl, (CH2) q-0 (CH 2
-CH
3 or (CH 2 MnZ; wherein R 1 3 is a bicyclic alkyl ring system, adamantyl, noradamantyl,
C
3 -Cl 0 cycloalkyl, heteroaryl, aryl, aryl (Cl-
C
6 alkyl, Q, or Q2; wherein aryl may be substituted with one or more Cl-Clo straight chained or branched alkyl, aryl, heteroaryl, or
'N(R
1 9 wherein Q, is IIR22 wherein Q2 is wherein each J is independently 0, S, C (R 2 2 2 Or NR 4 wherein R 4 is H; straight chained or branched Cl-C 7 alkyl, 00 165 monof luoroalkyl or polyf luoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3
-C
7 cycloalkyl,
CS-C
7 cycloalkenyl or aryl; 00 wherein Y is NR 1 4
R
1 5 00 1 or uI 17
R
19
N
t
R
2 0
-N
\z
R~R
20 wherein R3.
4 is H, straight chained or branched Cl-C 6 alkyl,
(CH
2 )q-01(CH2)m-CH3,
C
3
C
6 cycloalkyl, or (C(Ri,) 2 wherein
R,
5 is straight chained or branched
C
3
-C
6 alkyl, 1s (CH 2 )q0O(CH2)m-CH3I
C
3
-C
6 cycloalkyl, (C (R 19 2 m(RIO)2 or (C (R 19 2
Z;
wherein
R
16 is straight chained or branched
C
1
-C
7 alkyl, straight chained or branched Cl-C 7 monofluoroalkyl, straight chained or branched Cl-C7 polyfluoroalky)-, 00 ^C 166 straight chained or branched C 2
-C
7 alkenyl, straight n chained or branched C 2
-C
7 alkynyl, Cs-C 7 cycloalkenyl,
(CH
2 or (CH 2 )q-0-(CH2)m-CH3; 00 wherein each R 17 is independently H; -OR 2 1
-OCOR
21
-COR
21 O -NCOR 21
-N(R
21 2
-CON(R
21 2
-COOR
21 straight chained or C branched Ci-C 7 alkyl, straight chained or branched Ci-C 7 00 Smonofluoroalkyl, straight chained or branched Ci-C 7 CA polyfluoroalkyl, straight chained or branched C2-C7 "alkenyl, straight chained or branched C 2
-C
7 alkynyl, C 5
-C
7 cycloalkenyl,
-(CH
2 or (CH 2 (CH2)m-CH3; wherein R 18 is straight chained or branched CI-C 6 alkyl,
(CH
2 Or (CH2)q-O-(CH 2 )m-CH3; wherein each R 19 is independently H, or straight chained or branched CI-C 6 alkyl; wherein each R 20 is independently straight chained or branched Ci-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3
C
7 cycloalkyl or Cs-C 7 cycloalkenyl; -C1, -Br, or -I;
-NO
2
-N
3 -CN; -OR 2 1
-OCOR
21
-COR
21
-NCOR
21
-N(R
21 2
CON(R
21 2 or -COOR 21 aryl or heteroaryl; or two R 2 0 groups present on adjacent carbon atoms can join together to form a methylenedioxy group; wherein each R 21 is independently straight chained or branched CI-C7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3
C
7 cycloalkyl, Cs-C 7 cycloalkenyl, aryl, or aryl (C- Cs) alkyl; 00 (C 167 Swherein each R 22 is independently H, F, Cl or C 1
-C
4 straight chained or branched alkyl; 00 wherein each m is an integer from 0 to 4 inclusive; C wherein each n is an integer from 1 to 4 inclusive; 00 Cl wherein p is an integer from 0 to 2 inclusive; wherein q is an integer from 2 to 4 inclusive; wherein t is 1 or 2; wherein U is O, -NR 16 S, C(R 17 2 or -NSO 2
R
6 wherein Z is C 3 -Co 0 cycloalkyl, C 4 cyclic ether, C 4
-C
7 cyclic thioether, aryl, or heteroaryl; or a pharmaceutically acceptable salt thereof.
The invention provides a method of treating a subject suffering from anxiety which comprises administering to the subject an amount of compound effective to treat the subject's anxiety wherein the compound has the structure: 168 wherein W is H, -Cl, -Br, CN, methyl, ethyl, propyl, methoxy or ethoxy; wherein X is NRjjR 12
R
1 7
R
1 -N17 -N
/N-R
1 8
R
17 or wherein R 11 is H, straight chained or branched C 1
-C
7 alkyl, (CH2)q-O (CH 2 )m-CH 3 aryl or aryl(Cl-C 6 )alkyl; wherein R3.
2 is straight chained or branched Cl-C 7 alkyl, (CH2)q-O (CH 2 )m-CH 3 or -(CH 2 )m-Z; wherein R 13 is a bicyclic alkyl ring system, aryl or aryl (C 1 CO)alkyl; wherein Y is NR 14
R
15 00 169 R 0 c-I R 20 00-NI M NuR 19 p ;or
N
00 vr >t1 wherein R 14 is H, straight chained or branched Cl-C 6 alkyl,
(CH
2 )qOiCH 2 )m-CH 3
C
3
-C
6 cycloalkyl, or (C(Rxg) 2 )m-Z; wherein Ris is straight chained or branched C 3
-C
6 alkyl,
(CH
2 )qOicH 2 )M&H3, C 3 -CG cycloalkyl, or CR92,Z wherein U is 0, -NR16, S, C(Rl 7 2 or -NS0 2
R,
6 wherein Z is C 3
-CI
0 cycloalkyl, aryl, or heteroaryl; wherein R 16 is straight chained or branched CI-C 7 alkyl, straight chained or branched Cl-C, monofluoroalkyl, straight chained or branched C- 1 -C7 polyfluoroalkyl, straight chained or branched C 2
-C
7 alkenyl, straight chained or branched C 2
-C
7 alkynyl, CS-C 7 cycloalkenyl,
(CH
2 or (CH 2 )q01-CH 2 )m-CH 3 00 170 wherein each R 1 7 is independently H; -OR 2 1 -0C0R 21 -C0R 21
-NCOR
21
-N(R
21 2
-CON(R
2 1) 2 -C00R 21 straight chained or branched C 1 -C-y alkyl, straight chained or branched C 1
-C
7 monofluoroalkyl, straight chained or branched C 1
I-C
7 00 M~f 5 polyfluoroalkyl, straight chained or branched
C
2
-C
7 alkenyl, straight chained or branched C 2
-C
7 alkynyl, CS-C 7 00 cycloalkenyl,
-(CH
2 or (CH 2 )n-O-ICH 2 ),-CH3; wherein R 1 8 is straight chained or branched C 1
-C
6 alkyl,
(CH
2 )mZ I or (CH2) qO (CH 2 M- CH 3 wherein each R 19 is independently H, or straight chained or branched C 1
-C
6 alkyl; wherein each R 2 0 is independently straight chained or branched C3 1 -C,7 alkyl, monofluoroalkYl. or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl. or alkynyl; C 3 C7 cycloalkyl or C 5
-C
7 cycloalkenyl; -CI, -Br, or -I;
-NO
2
-N
3 -0R 21 -0C0R 21 -C0R 21
-NCOR
21
-N(R
2 2
CON(R
2 1 2 or -COOR 2 1; aryl or heterojryl; or two R 2 0 groups present on adjacent carbon atoms can join together to form a methylenedioxy group; wherein each R 2 is independently straight chained or branched Cl-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3
C
7 cycloalkyl, C5-C 7 cycloalkenyl, aryl or aryl (Cl-
C
6 alkyl; wherein each m is an integer from 0 to 4 inclusive; wherein each n is an integer from 1. to 4 inclusive; 171 wherein p is an integer from 0 to 2 inclusive; wherein q is an integer from 2 to 4 inclusive; wherein t is 1 or 2; or a pharmaceutically acceptable salt thereof.
The invention provides a method of treating a subject suffering from anxiety which comprises administering to the subject an amount of compound effective to treat the subject's anxiety wherein the compound has the structure: wherein W is H, -Cl, propyl, methoxy or ethoxy; wherein X is N(CH 3 2 or -Br, CN, methyl, ethyl, 172 wherein
R
13 is an aryl, adamantyl, noradamantyl,
C
3
-C
10 cycloalkyl, heteroaryl, Q, or Q2; wherein aryl may be substituted with one or more CI-Clo straight chained or branched alkyl, aryl, heteroaryl, or N (Rig) Z; wherein Q, is wherein Q2 is wherein each J is independently 0, S, C (R 2 2 2 or NR 4 wherein R 4 is straight chained or branched Cl-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched
C
2
-C
7 alkenyl or alkynyl;
C
3
-C
7 cycloalkyl,
C
5 cycloalkenyl or aryl; 00 C-I 173 wherein Y is NR 14
R
15 00 M R 17 8R2 00I\ -N o C-I p R 17
R
20
-N
wherein R 14 is H, straight chained or branched C 1
-C
6 alkyl,
(CH
2 )q-O-ICH2)m-CH3,
C
3
-C
6 cycloalkyl, or (C(Rig) 2 )m-Z; wherein R 15 is straight chained or branched C 3
-C
6 alkyl, (CH2)q-O-(CH 2 ),-CH3s C 3
-C
6 cycloalkyl, or (C(R 1 9 2 )m-Z; wherein U is 0, -NR 1 6 S, C(R17)2, or -NSO 2 Rx 6 wherein Z is C 3 -Cl 0 cycloalkyl, aryl, or heteroaryl; wherein R 16 is straight chained or branched C 1
-C
7 alkyl, straight chained or branched
C
1
-C
7 monofluoroalkyl, straight chained or branched
C
1
-C
7 polyfluoroalkyl, 00 174 straight chained or branched C 2 -C7 alkenyl, straight chained or branched C 2
-C
7 alkynyl, C 5
-C
7 cycloalkenyl,
(CH
2 or (CH 2 )q-O-(CH 2 )u.-CH3; 00 5 wherein each R 17 is independently H; -OR 2 2, -0C0R 21 -C0R 21
-NCOR
21
-N(R
2 1) 2
-CON(R
21 2 -C00R 21 straight chained or CI branched CI-C 1 alkyl, straight chained or branched Cl-C 7 00 monofluoroalkyl, straight chained or branched CI-C 7 polyfluoroalkyl, straight chained or branched C 2
-C
7 alkenyl, straight chained or branched 9 2
-C
7 alkynYl, CS-C7 cycloalkenyl,
(CH
2 or (CH 2
(CH
2 )ii-CH 3 wherein RIG is straight chained or branched CI-C 6 alkyl,
(CH
2 Or (CH 2 )q-O-(CH 2 )m-CH3; wherein each R- 19 is independently H, or straight chained or branched C 1
-C
6 alkyl; wherein each R 20 is independently straight chained or branched CI-C7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3 C7 cycloalkyl or C 5
-C
7 cycloalkenyl; -C1, -Br, or -I;
-NO
2
-N
3 -CN; -OR 2 1 -0C0R 21 -CaR 21
-NCOR
21
-N(R
21 2 CON (R 21 2, or -COOR 21 aryl or heteroaryl; or two R 20 groups present on adjacent carbon atoms can join together to form a methylenedioxy group; wherein each R 2 1 is independently straight chained or branched Cl-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3
C
7 cycloalkyl,
CS-C
7 cycloalkenyl, aryl or aryl(Ci-
C
6 alkyl; 175 wherein each R 22 is independently H, F, Cl or C 1
-C
4 straight chained or branched alkyl; wherein each m is an integer from 0 to 4 inclusive; wherein each n is an integer from 1 to 4 inclusive; wherein p is an integer from 0 to 2 inclusive; wherein q is an integer from 2 to 4 inclusive; wherein t is 1 or 2; or a pharmaceutically acceptable salt thereof.
The invention provides a method of treating a subject suffering from anxiety which comprises administering to the subject an amount of compound effective to treat the subject's anxiety wherein the compound has the structure: wherein W is H, -C1, -Br, CN, methyl, ethyl, propyl, methoxy or ethoxy; wherein X is N(CH 3 2 or 00 176
R
17 -N 0
R
17 00 wherein R 1 3 is a bicyclic alkyl ring system, aryl or 00 aryl (C- 1
-C
6 alkyl; wherein Y is NR1 4 R:m; wherein R 14 is H, straight chained or branched
C
1
-C
6 alkyl,
(CH
2 )q0O(CH 2 )m-CH3, C 3
-C
6 cycloalkyl, or (C(R 1 9 2 wherein R3.
5 is (C(Rig9) 2 )m-N(RlE6)2; wherein Z is C 3 -CIO cycloalkyl, aryl, or heteroaryl; wherein R 16 is straight chained or branched
CI-C
7 alkyl, straight chained or branched
CI-C
7 mono fluoroa lkyl, straight chained or branched
C
1
I-C
7 polyfluoroalkyl, straight chained or branched
C
2 -07, alkenyl, straight chained or branched
C
2
-C
7 alkynyl, Cs-C, cycloalkenyl,
(CH
2 or (CH2)q-O- (CH 2 )m-CH 3 wherein each R 1 7 is independently H; -OR 21 -0C0R 21 -C0R 21
-NCOR
21
-N(R
21 )2 -CON(R 21 2 -C00R 21 straight chained or branched CI-C7 alkyl, straight chained or branched Cl-C7 monofluoroalkyl, straight chained or branched Cl-C7 polyfluoroalkyl, straight chained or branched
C
2 -C7 alkenyl, straight chained or branched
C
2 -C7 alkynyl, C 5
-C
7 cycloalkenyl,
-(CH
2 Or (CH 2 )n-O (CH 2 )mCH 3 00 C 177 wherein each R 19 is independently H, or straight chained n or branched Ci-C 6 alkyl; O wherein each R 21 is independently straight chained or 0 0 5 branched C 1
-C
7 alkyl, monofluoroalkyl or polyfluoroalkyl; 0 straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3 C
C
7 cycloalkyl, Cs-C 7 cycloalkenyl, aryl or aryl(C 1 0 Cs 6 )alkyl; wherein each m is an integer from 0 to.4 inclusive; wherein each n is an integer from 1 to 4 inclusive; wherein q is an integer from 2 to 4 inclusive; or a pharmaceutically acceptable salt thereof.
As used in the present invention, the term "bicyclic alkyl ring systems" includes, but is not limited to, bicyclo[2.2.1 heptane, bicyclo[3.1.1]heptane and bicyclo[2.2.2]octane. In addition, the bicyclic alkyl ring systems may be substituted with one or more of the following:
-NO
2 -CN, straight chained or branched
C
1
-C
7 alkyl, straight chained or branched CI-C 7 monofluoroalkyl, straight chained or branched CI-C 7 polyfluoroalkyl, straight chained or branched C 2
-C
7 alkenyl, straight chained or branched C 2 -C7 alkynyl, C 3
-C
7 cycloalkyl, C 5
-C
7 cycloalkenyl,
-N(R
21 2
-OR
21
-COR
21 CO0R 21
-CON(R
21 )2 or (CH 2 )n-O-(CH 2 )m-CH3.
As used in the present invention, the term "cycloalkyl" includes, C 3 -C7 cycloalkyl moieties which may be 00 178 substituted with one or more of the following:
-NO
2 -CM, straight chained or branched Cl-C 7 alkyl, Btraight chained or branched
CI-C
7 monofluoroalkyl, straight chained or branched
CI-C
7 polyfluoroalkyl, straight 00 M 5 chained or branched
C
2
-C
7 alkenyl, straight chained or branched
C
2 -C7 alkynyl,
C
3
-C
7 cycloalkyl,
C
3 -C7 00 monofluorocycloalkyl,
C
3
-C
7 polyfluoroCccoalkyl,
C
5 -C-7 cycloalkenyl,
-N(R
4 2
-OR
4 -COR4, -NCOR4 -C02R4,
CON(R
4 2 or (CH 2 )n-O-(CH 2 )m-CH3.
jo As used in the present invention, the term "cyclohexyl", includes, cyclohexyl groups which may be substituted with one or more of the following:
-NO
2 -CN, straight chained or branched Cl-C 7 alkyl, straight chained or branched
C
1
-C
7 monofluoroalkyl, straight chained or branched Cl-C 7 polyfluoroalkyl, straight chained or branched
C
2 -C7 alkenyl, straight chained or branched
C
2
-C
7 alkyIny-,
C
3
-C
7 cycloalkyl,
C
3
-C
7 monofluorocycloalkyl, c 3 -c 7 polyfluocYcloalkyl,
C
5 -C7 cycloalkenyl,
-M(R
4 2 -OR4,- COR4, -NCOR 4 -C0 2
R
4 1 -CON(R4) 2 or (CH 2 )n-O-(CH2 )M-CH3.
As used in the present invention, the term "cycloalkenyl", includes,
C
5
-C
7 cycloalkenYl moieties which may be substituted with one or more of the following: -Cl, -Br, -I1, -NO 2 -CM, straight chained or branched Cl-C 7 alkyl, straight chained or branched
C
1
-C
7 monofluoroalkYl, straight chained or branched Cl-C, polyfluoroalkYl, straight chained or branched
C
2
-C
7 alkenyl, straight chained or branched
C
2
-C
7 alkynyl,
C
3 -C7 cycloalkyl,
C
3
-C
7 monofluorocycloalkyl,
C
3
-C
7 polyfluorocycloalkyl,
C
5
-C
7 cycloalkenyl,
-N(R
4 2
-OR
4
-COR
4
-NCOR
4 -C02R4,-
CON(R
4 )2 or (CH 2
)O-(C
2 )m-CH3- 00 C\ 179 CQ In the present invention, the term "heteroaryl" is used to include five and six membered unsaturated rings that O may contain one or more oxygen, sulfur, or nitrogen 00 atoms. Examples of heteroaryl groups include, but are not limited to, furanyl, thienyl, pyrrolyl, oxazolyl, 00 thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl, C( pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, and i0 triazinyl.
In addition the term "heteroaryl" is used to include fused bicyclic ring systems that may contain one or more heteroatoms such as oxygen, sulfur and nitrogen. Examples of such heteroaryl groups include, but are not limited to, indolizinyl, indolyl, isoindolyl, benzo[b]furanyl, benzo[b]thiophenyl, indazolyl, benzimidazolyl, purinyl, benzoxazolyl, benzisoxazolyl, benzo[b]thiazolyl, imidazo[2,1-b]thiazolyl, cinnolinyl, quinazolinyl, quinoxalinyl, 1,8-naphthyridinyl, pteridinyl, quinolinyl, isoquinolinyl, phthalimidyl and 2,1,3-benzothiazolyl.
The term "heteroaryl" also includes those chemical moieties recited above which may be substituted with one or more of the following: -Cl, -Br, -NO 2
-CN,
straight chained or branched
CI-C
7 alkyl, straight chained or branched
CI-C
7 monofluoroalkyl, straight chained or branched
C
1
-C
7 polyfluoroalkyl, straight chained or branched C 2
-C
7 alkenyl, straight chained or branched C 2
-C
7 alkynyl, C 3 cycloalkyl,
C
3
-C
7 monofluorocycloalkyl,
C
3
-C
7 polyfluorocycloalkyl, Cs-C 7 cycloalkenyl,
-N(R
4
-OR
4
COR
4 -NCOR4, -CO 2
R
4 -CON(R4) 2 or (CH 2 )n-O-(CH 2 )m-CH 3 00 C 180 CQ The term "heteroaryl" further includes the N-oxides of those chemical moieties recited above which include at least one nitrogen atom.
00 C In the present invention the term "aryl" is phenyl or 00 naphthyl. The term "aryl" also includes phenyl and Snaphthyl which may be substituted with one or more of the (C following: -Cl, -Br, -NO 2 -CN, straight chained or branched Ci-C 7 alkyl, straight chained or branched Ci-C 7 monofluoroalkyl, straight chained or branched
C
1
-C,
polyfluoroalkyl, straight chained or branched
C
2
-C
7 alkenyl, straight chained or branched C 2
-C
7 alkynyl, C 3
-C
7 cycloalkyl,
C
3
-C
7 monofluorocycloalkyl, C3-C7 polyfluorocycloalkyl, Cs-C 7 cycloalkenyl,
-N(R
4 2
-OR
4
SR
4
-OCOR
4
-COR
4
-NCOR
4 -C0 2
R
4
-CON(R
4 2 or (CH 2 )n-O-
(CH
2 )m-CH 3 In one embodiment of any of the methods described herein, the compound is enantiomerically and diasteriomerically pure. In one embodiment, the compound is enantiomerically or diasteriomerically pure.
In one embodiment, the compound can be administered orally.
In one embodiment, X is: 181
R
1 17 -N 17 or In one embodiment, X is NR 11 Rl 2 and R 11 is H or straight chained or branched Cl-C 7 alkyl.
In one embodiment, the compound has the structure: w
N
In one embodiment, R 13 is a bicyclic alkyl. ring system, cyclohexyl or aryl.
In one embodiment, R 1 4 is H, straight chained or branched
C
1
-C
6 alkyl or (CH 2 )q0O-(CH 2 )m-CH3.
1 00 C. 182 In one embodiment, the compound is selected from the Sgroup consisting of: 00
O
N N N 00 &N N 0 Q 2008200380 25 Jan 2008
Z
z
F
ZOCC
2008200380 25 Jan 2008 z
N-
-0 2008200380 25 Jan 2008 I-o (Ln 0z ,C0 00 186 -N U 00 R 1 00
C(
0 In 'one embodiment, U is NR16.
0 5 In one embodiment, R 16 is (CH 2 )m-Z.
In one embodiment, Z is aryl or heteroaryl.
In one embodiment, the compound is selected from the group consisting of: I 0 2008200380 25 Jan 2008
-C,
200820038025Jn08 25 Jan 2008
'X
0~
K
189 In one embodiment, the compound is selected from the group consisting of: ClC N N N N Cl I
N
N N N11 2008200380 25 Jan 2008
I-.
'0 0 00 c-I 191
-N
00 In one embodiment, U is NR6.
In one embodiment, the compound is N)I) rY, or 192 In one embodiment, the compound is In one embodiment, the compound is selected from the group consisting of: 193 0
NN
NN)
N CI
NN
~NN
N N6
C
N N
\N
N NaC N C1 0NN N l
'N
NN
;and In one embodiment, the compound is selected from the group consisting of: 194
'N
N SCF3 A'l
J
r'N 'N Ni 0
N
N CK~~<F 3
NN
NIN oe F3 c: and N N N
N
In one embodiment, X is N(CH 3 2 In one embodiment, Y is 00 1.95 R 7 -N U 00 1 00 In one embodiment,
R
13 is an aryl substituted with a Cj-Cj 0 straight chained alkcyl.
In one embodiment, the compound is selected from a group consisting of:
N
NN
1 A N' N' 00 196 The invention provides a pharmaceutical Composition comprising a pharmaceutically acceptable carrier and a compound having the structure: 00 x 00N c~KIY N R 13 wherein W is H, -Cl, -Br, CN, methyl, ethyl, propyl, methoxy or ethoxy; wherein X is; NR 11
R
12
R
17 R17R 1 R17
C
RR17 -N
N-R
18
R
17 wherein R 11 is H4, straight chained or branched CI-C 7 alkyl,
(CH
2 2 aryl, or aryl (Cl-C 6 )alkyl; wherein R 12 is straight chained or branched Cl-C 7 alkyl, 197
(CH
2 ),q0(CH 2 )m-CH3I or -(CH 2 )m-Z; wherein
R
13 is a bicyclic alkyl ring system, adamantyl, noradamantyl,
C
3
-C
1 0 cycloalkyl, heteroaryl, aryl, aryl (Cl- CO)alkyl, Q, or Q2; wherein aryl may be substituted with one or more CI-Cl 0 straight chained or branched alkyl, aryl, heteroaryl, or N (R 19
-Z;
wherein Q, is wherein Q2 is wherein each J is independently 0, S, C(R 2 2 2 or NR 4 wherein
R
4 is H; straight chained or branched
C
1
-C
7 alkyl, monofluoroalkYl or polyfluoroalkyl; straight chained or 198 branched C 2
-C
7 alkenyl or alkynyl; C 3
-C
7 cycloalkyl,
C
5
-C
7 cycloalkenyl or aryl; wherein Y is NR 14 R2 5 ;or wherein R 14 is H, straight chained or branched
CI-C
6 alkyl,
(CH
2 )q-OiCH 2 )m-CH3, C 3
-C
6 cycloalkyl, or (C(Rlg) 2 wherein R 15 is straight chained or branched C 3
-C
6 alkyl,
(CH
2 q -0 CH 2 m CH 3
C
3
-C
6 S cycloalkyl, (C (R 1 9 2 N (R 16 2 or (C (R19) 2 )m -Z; wherein R 16 is straight chained or branched Cl-C 7 alkyl, straight chained or branched
CI-C
7 monofluoroalkyl, straight chained or branched Cl-C 7 polyf luoroalkyl, straight chained or branched
C
2
-C
7 alkenyl, straight 00 199 chained or branched C 2 -C7 alkynyl, Cs-C, cycloalkenyl,
(CH
2 )mZ, Or (CH 2 )q-O-(CH 2 )m-CH3; wherein each R 1 is independently H; -ORz 1 -0COR 21
-COR
21 00
-NCOR
21
-N(R
21 2
-CON(R
21 2
-COOR
21 straight chained or branched C1-C, alkyl, straight chained or branched C 1
-C,
0 monofluoroalkyl, straight chained or branched C 1
-C,
0 polyfluoroalkyl, straight chained or branched C 2 -C7 C alkenyl, straight chained or branched C 2 -C7 alkynyl, Cs 5
-C,
cycloalkenyl,
-(CH
2 or (CH 2
(CH
2
)-CH
3 wherein R 18 is straight chained or branched C 1
-C
6 alkyl,
(CH
2 or (CH2)q-O- (CH 2 )m-CH3; wherein each R] 9 is independently H, or straight chained or branched C 1
-C
6 alkyl; wherein each R 20 ao is independently straight chained or branched C 1 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 alkenyl or alkynyl; C 3 C, cycloalkyl or Cs-C, cycloalkenyl; -Cl, -Br, or -I;
-NO
2
-N
3 -CN; -OR 2 1
-OCOR
2 1 -COR21, -NCOR 21
-N(R
21 2 CON(R2 1 or -COOR21; aryl or heteroaryl; or two R 20 groups present on adjacent carbon atoms can join together to form a methylenedioxy group; wherein each R 21 is independently straight chained or branched C 1 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 alkenyl or alkynyl; C 3 C, cycloalkyl, Cs-C, cycloalkenyl, aryl, or aryl(C 1
C
6 alkyl; 00 200 c wherein each R 22 is independently H, F, Cl or C 1
-C
4 I straight chained or branched alkyl; wherein each m is an integer from 0 to 4 inclusive; 00 0 wherein each n is an integer from 1 to 4 inclusive; 00 Swherein p is an integer from 0 to 2 inclusive; wherein q is an integer from 2 to 4 inclusive; wherein t is 1 or 2; wherein U is O, -NR 16 S, C(RI 7 2 or -NSOa 2 Rs; wherein Z is C 3 -Co cycloalkyl, C 4 -C7 cyclic ether, C 4
-C
7 cyclic thioether, aryl, or heteroaryl; or a pharmaceutically acceptable salt thereof.
The invention provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound having the structure: 00 201 wherein W is H, -Cl, -Br, CN, methyl, ethyl, propyl, methoxy or ethoxy; wherein X is NR,,R 12 00
R
17
R
17 00
N
)or -NN-R 8
R
17 wherein R3.
1 is H, straight chained or branched Cl-C 7 alkyl,
(CH
2 )q-O (CH 2
)M-CH
3 aryl or aryl (Cl-C 6 alkyl; wherein
R
12 is straight chained or branched Cl-C 7 alkyl,
(CH
2
(CH
2 ),vCH3, or -(CH 2 wherein
R
13 is a bicyclic alkyl ring system, aryl or aryl (Cl-C6) alkl; is wherein Y is NR 14
R
15 00 202 00 -N u or 00 1
R
20
-N-
wherein R 14 is H, straight chained or branched Cl-C 6 alkyl, (CH2)q 0
-(CH
2
)M-CH
3
C
3
-C
6 cycloalkyl, or (C(Ri 9 2 )m-Z; wherein R 15 is straight chained or branched C 3
-C
6 alkyl,
(CH
2 )q0.i(CH 2 )m-CH 3
C
3
-C
6 cycloalkyl, or (C(Rig) 2 )m-Z; wherein U is 0, -NR16, S, C(Rl 7 or -NS0 2
R
16 s; wherein Z is C 3 -Cl 0 cycloalkyl, aryl, or heteroaryl; wherein R 16 is straight chained or branched Cl-C 7 alkyl, straight chained or branched CI-C 7 monofluoroalkyl, straight chained or branched Cl-C 7 polyfluoroalkyl, straight chained or branched C 2 -C7 alkenyl, straight chained or branched
C
2
-C
7 alkynyl, CS-C 7 cycloalkenyl, (C4 2 or (CH 2 ),q0(CH 2 )m,,CH3; 00 0203 Swherein each Ri, is independently H; -OR 21
-OCOR
2 1
-COR
21 in -NCOR 21
-N(R
21 2
-CON(R
2 1 2
-COOR
21 straight chained or branched C 1
-C
7 alkyl, straight chained or branched C 1
-C
7 monofluoroalkyl, straight chained or branched C
I
-C
7 00 5 polyfluoroalkyl, straight chained or branched C 2 -C7 0 alkenyl, straight chained or branched C 2
-C
7 alkynyl, Cs-C 7 CI cycloalkenyl,
-(CH
2 or (CH 2 )n-O-(CH 2
).-CH
3 00 CS wherein R 18 is straight chained or branched CI-C 6 alkyl,
(CH
2 or (CH 2 )q-O-(CH2)m-CH3; wherein each R 19 is independently H, or straight chained or branched C 1
-C
6 alkyl; wherein each R 20 is independently straight chained or branched C 1
-C
7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3 C7 cycloalkyl or Cs-C7 cycloalkenyl; -C1, -Br, or -I;
-NO
2
-N
3 -CN; -OR 21
-OCOR
21
-COR
21
-NCOR
21
-N(R
21 2 CON(R21)2, or -COOR 21 aryl or heteroaryl; or two R 20 groups present on adjacent carbon atoms can join together to form a methylenedioxy group; wherein each R 21 is independently straight chained or branched C 1
-C
7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or alkynyl; C 3 C7 cycloalkyl, Cs-C7 cycloalkenyl, aryl or aryl(Ci- Cs) alkyl; wherein each m is an integer from 0 to 4 inclusive; 00 204 n wherein each n is an integer from 1 to 4 inclusive; o wherein p is an integer from 0 to 2 inclusive; 00 Swherein q is an integer from 2 to 4 inclusive; 00 Swherein t is 1 or 2; or a pharmaceutically acceptable salt thereof.
The invention provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound having the structure:
X
NI
W
N
H
wherein W is H, -Cl, -Br, CN, methyl, ethyl, propyl, methoxy or ethoxy; wherein X is N(CH 3 2 or 00 205 -N 0 00 whMi 1 sa rl dmnynrdmny, c- 1 0 00 cycloalkyl, heteroaryl, Q, or Q2; wherein aryl may be substituted with one or more Cl-CI 0 straight chained or branched alkyl, aryl, heteroaryl, or NC(Rig) Z; wherein Q, is 22 wherein Q2 is R22 t R22 P
R
I R 20 wherein each J is independently 0, S, C(R 2 2 2 or NR 4 00 206 wherein R 4 is straight chained or branched Cl-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3
-C
7 cycloalkyl, CS-C 7 cycloalkenyl or aryl; 00 wherein Y is NR 1 4
R
1 5 00R2
CIR
17 R2 -N U 1 p ;or R17 R 2 0
N-
R
20 1o wherein R 14 is H, straight chained or branched Cl-CE alkyl,
(CH
2 q-0- (CI4 2
CH
3
C
3
-C
6 cycloalkyl, or (C(Rig) 2
)M-Z;
wherein R 15 is straight chained or branched C 3
-C
6 alkyl,
(CH
2 9O- CH 2 MCIH3, C 3 -C6 cycloalkyl, or (C(Rig) 2 )mZ; wherein U is 0, -NR 16 S, C(Rl 7 2 or -NS0 2
R,
6 wherein Z is C 3
-C
10 cycloalkyl, aryl, or heteroaryl; 00 207 wherein R 16 is straight chained or branched Cl-C 7 alkyl, straight chained or branched Cl-C 7 monofluoroalkyl, straight chained or branched Cl-C 7 polyfluoroalkyl, 00 5 straight chained or branched C 2
-C
7 alkenyl, straight chained or branched C 2 -C7 alkynyl, C 5 -C7 cycloalkenyl,
(CH
2 or (CH 2 )q-O-(CH 2 )m-CH 3 i 00 wherein each R 1 7 is independently H; -OR 2 1 -0C0R 21 -C0R 2 1
-NCOR
21
-N(R
21 2
-CON(R
21 2 -jC00R 21 straight chained or branched C1-C-7 alkyl, straight chained or branched C 1
I-C
7 monofluoroalkyl, straight chained or branched CI-C 7 polyfluoroalkYl, straight chained or branched
C
2
-C
7 alkenyl, straight chained or branched C 2
-C
7 alkynyl, CS-C 7 cycloalkenyl, 4(CH 2 or (CH 2 )n-O-(CH 2 )m-CH 3 wherein Ris is straight chained or branched
C
1
-C
6 alkyl,
(CH
2 or (CH 2 (CH2)M-CH 3 wherein each Rig is independently H, or straight chained or branched C 1
-C
6 alkyl; wherein each R 20 is independently straight chained or branched
CI-C
7 alkyl., monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3
C
7 cycloalkyl or C 5
-C
7 cycloalkenYl; -CI, -Br, or -1;
-NO
2
-N
3 -CN; -OR 2 1 -0C0R 2 1 -C0R 2 1
-NCOR
21
-NCR
21 2
CON(R
2 1 2 or -C00R 21 aryl or heteroaryl; or two R 20 groups present on adjacent carbon atoms can join together to form a methylenedioXY group; wherein each R 2 1 is independently straight chained or 00 208 branched C 1
-C
7 alkyl, monofluoroalkyl or polyfluoroalkyl; I straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3 6 C 7 cycloalkyl,
C
5
-C
7 cycloalkenyl, aryl or aryl(C 1
C
6 alkyl; 00
M
n wherein each R22 is independently H, F, Cl or Ci-C 4 O straight chained or branched alkyl; O< 00 Swherein each m is an integer from 0 to 4 inclusive; wherein each n is an integer from 1 to 4 inclusive; wherein p is an integer from 0 to 2 inclusive; wherein q is an integer from 2 to 4 inclusive; wherein t is 1 or 2; or a pharmaceutically acceptable salt thereof.
The invention provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound having the structure:
X
209 wherein W is H, -Cl, -Br, CN, methyl, ethyl, propyl, methoxy or ethoxy; 0 00 wherein X is N (CH 3 2 or wherein R 13 is a bicyclic alkyl ring system, aryl or aryl (Cl-C 6 alkyl; wherein Y is N~R 14
R
15 wherein R 14 is H, straight chained or branched Cl-C 6 alkyl,
(CH
2
)QOICH
2
)M-CH
3
C
3
-C
6 S cycloalkyl, or (C(Rig) 2 wherein R2 15 is (C (R39) 2 m-N(R 1 6 2 wherein Z is C 3 -Clo cycloalkyl, aryl, or heteroaryl; wherein R 16 is straight chained or branched Cl-C 7 alkyl, straight chained or branched Cl-C 7 monofluoroalkyl, straight chained or branched Cl-C 7 polyf luoroalkyl, straight chained or branched C 2
-C
7 alkenyl, straight chained or branched C 2
-C
7 alkynyl, CS-C 7 cycloalkenyl,
(CH
2 or (CH 2 )q-O-(CH 2 )m-CH 3 wherein each R 1 7 is independently H; -OR 2 1 -0C0R 21 -C0R 21 00 210
-NCOR
21
-N(R
21 2
-CON(R
21 2
-COOR
21 straight chained or branched C 1
-C
7 alkyl, straight chained or branched Ci-C 7 monofluoroalkyl, straight chained or branched C 1
-C
7 polyfluoroalkyl, straight chained or branched C 2
-C,
00 5 alkenyl, straight chained or branched C 2
-C
7 alkynyl, Cs-C7 O cycloalkenyl, -(CH 2 or (CH 2 )n-O-(CH 2 )m-CH 3 0 0 0 wherein each R 19 is independently H, or straight chained or branched Ci-C 6 alkyl; wherein each R21 is independently straight chained or branched C 1
-C
7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3
C
7 cycloalkyl, C 5
-C
7 cycloalkenyl, aryl or aryl(C 1
C
6 )alkyl; wherein each m is an integer from 0 to 4 inclusive; wherein each n is an integer from 1 to 4 inclusive; wherein q is an integer from 2 to 4 inclusive; or a pharmaceutically acceptable salt thereof.
As used in the present invention, the term "bicyclic alkyl ring systems" includes, but is not limited to, bicyclo[2.2.1]heptane, bicyclo[3.1.1]heptane and bicyclo[2.2.2]octane. In addition, the bicyclic alkyl ring systems may be substituted with one or more of the following:
-NO
2 -CN, straight chained or branched Ci-C 7 alkyl, straight chained or branched C 1
-C
7 monofluoroalkyl, straight chained or branched CI-C7 00 211 polyfluoroalkyl, straight chained or branched C 2
-C
7 alkenyl, straight chained or branched C 2
-C
7 alkynyl, C 3
-C
7 cycloalkyl, C 5
-C
7 cycloalkenyl, -N(R 21 2
-OR
21
-COR
2 1 C0 2
R
21
-CON(R
2 1 2 or (CH 2 )n-O-(CH 2 )m-CH 3 00 As used in the present invention, the term "cycloalkyl" CI includes, C 3
-C
7 cycloalkyl moieties which may be 00 substituted with one or more of the following:
-NO
2 -CN, straight chained or branched C 1 -C alkyl, straight chained or branched C 1
-C
7 .monofluoroalkyl, straight chained or branched CI-C7 polyfluoroalkyl, straight chained or branched C 2 -C7 alkenyl, straight chained or branched C 2
-C
7 alkynyl, C 3
-C
7 cycloalkyl, C 3
-C
7 monofluorocycloalkyl, C 3
-C
7 polyfluorocycloalkyl, Cs-C? cycloalkenyl, -N(R 4 2
-OR
4
-COR
4
-NCOR
4
-CO
2
R
4
CON(R
4 2 or (CH 2 )n-O-(CH 2 )m-CH 3 As used in the present invention, the term "cyclohexyl" includes, cyclohexyl groups which may be substituted with one or more of the following: -N0 2 -CN, straight chained or branched C 1
-C
7 alkyl, straight chained or branched CI-C 7 monofluoroalkyl, straight chained or branched C 1
-C
7 polyfluoroalkyl, straight chained or branched C 2
-C
7 alkenyl, straight chained or branched C 2
-C
7 alkynyl, C 3
-C
7 cycloalkyl, C 3
-C
7 monofluorocycloalkyl, C 3
-C
7 polyfluorocycloalkyl, Cs-C 7 cycloalkenyl, -N(R 4 2
-OR
4
COR
4 -NCOR4, -C0 2 R4, -CON(R4)2 or (CH 2 )n-O-(CH 2 )m-CH 3 As used in the present invention, the term "cycloalkenyl" includes, C 5
-C
7 cycloalkenyl moieties which may be substituted with one or more of the following: -Cl, -Br, -NO 2 -CN, straight chained or branched C 1
-C
7 00 212 alkyl, straight chained or branched CI-C 7 monofluoroalkyl, straight chained or branched CI-C 7 polyfluoroalkyl, straight chained or branched C 2
-C
7 alkenyl, straight chained or branched C 2
-C
7 alkynyl, C 3
-C
7 cycloalkyl, C 3
-C
7 00 5 monofluorocycloalkyl, C 3
-C
7 polyfluorocycloalkyl, Cs-C 7 cycloalkenyl, -N(R 4 2
-OR
4
-COR
4
-NCOR
4 -C0 2
R
4 C CON(R 4 )2 or (CH 2 )n-O-(CH 2 )m-CH 3 00 In the present invention, the term "heteroaryl" is used to include five and six membered unsaturated rings that may contain one or more oxygen, sulfur, or nitrogen atoms. Examples of heteroaryl groups include, but are not limited to, furanyl, thienyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, and triazinyl.
In addition the term "heteroaryl" is used to include fused bicyclic ring systems that may contain one or more heteroatoms such as oxygen, sulfur and nitrogen. Examples of such heteroaryl groups include, but are not limited to, indolizinyl, indolyl, isoindolyl, benzo[blfuranyl, benzo[b]thiophenyl, indazolyl, benzimidazolyl, purinyl, benzoxazolyl, benzisoxazolyl, benzo[b] thiazolyl, imidazo[2,1-b]thiazolyl, cinnolinyl, quinazolinyl, quinoxalinyl, 1,8-naphthyridinyl, pteridinyl, quinolinyl, isoquinolinyl, phthalimidyl and 2,1,3-benzothiazolyl.
The term "heteroaryl" also includes those chemical moieties recited above which may be substituted with one or more of the following: -Cl, -Br, -NO 2
-CN,
00 213 c straight chained or branched CI-C 7 alkyl, straight chained Sor branched Cl-C 7 monofluoroalkyl, straight chained or branched C-C 7 polyfluoroalkyl, straight chained or branched C 2
-C
7 alkenyl, straight chained or branched C 2 -C7 00 5 alkynyl, C 3
-C
7 cycloalkyl, C 3
-C
7 monofluorocycloalkyl, C 3
-C
7 D polyfluorocycloalkyl, Cs-C 7 cycloalkenyl, -N(R 4 2
-OR
4 C COR 4
-NCOR
4 -C0 2
R
4
-CON(R
4 2 or (CH 2 )n-O-(CH 2 )m-CH 3 00 OThe term "heteroaryl" further includes the N-oxides of those chemical moieties recited above which include at least one nitrogen atom.
In the present invention the term "aryl" is phenyl or naphthyl. The term "aryl" also includes phenyl and naphthyl which may be substituted with one or more of the following: -Cl, -Br, -NO 2 -CN, straight chained or branched Ci-C 7 alkyl, straight chained or branched C 1
-C
7 monofluoroalkyl, straight chained or branched Ci-C7 polyfluoroalkyl, straight chained or branched C 2 -07 alkenyl, straight chained or branched C 2
-C
7 alkynyl, C 3
-C
7 cycloalkyl, C 3
-C
7 monofluorocycloalkyl, C 3 -C7 polyfluorocycloalkyl, Cs-C 7 cycloalkenyl, -N(R 4 2
-OR
4
SR
4
-OCOR
4
-COR
4
-NCOR
4 -C0 2
R
4
-CON(R
4 2 or (CH 2 )n-O-
(CH
2 )m-CH 3 In one embodiment of any of the pharmaceutical compositions described herein, the compound is enantiomerically and diasteriomerically pure. In one embodiment the compound is enantiomerically or diasteriomerically pure.
214 In one embodiment of any of the pharmaceutical compositions described herein, the compound can be administered orally.
In one embodiment, X is: R17 R,
N
R17
R
1 7, -N N-R1 8 17 or In one embodiment, X is NRI 1
R
12 and R 11 is H or straight chained or branched Ci-C 7 alkyl.
In one embodiment, the compound has the structure: 00 215 In one embodiment, R 13 is a bicyclic alkyl ring system, cyclohexyl or aryl.
In one embodiment, R 1 4 is H, straight chained or branched 00 5 cl-c 6 alkyl or (CH 2 )q-O-(CH 2 )m-CH 3 In one embodiment, Y is 00 -N In one embodiment, U is NR 16 In one embodiment, R 16 is (CH2)m,,Z.
In one embodiment, Z is aryl or heteroaryl.
is In one embodiment, Y is
R
17 -N In one embodiment, U is NR 16 216 In one embodiment, the compound is selected from the group consisting of: C1
NC
N N N& C1
N
N N CN N N CI and N~ CI
N
rNN Nb
N,_
N NI yN 217 In one embodiment, compound is selected from the group consisting of: (NN rnO
K.NX
a"N" N N Nl
NN
0-1ryw N,) (CK> CF 3
N
NNai N NQ 00 218 In one embodiment, X is N (CH 3 2 In one embodiment, Y is 00 1
-N
E\-tpR 17 in one embodiment, R 1 3 is an aryl substituted with a C 1 -Cl 0 straight chained alkyl.
00 219 In one embodiment, the compound is selected from a group consisting of: 00N 00 r N c-I
N
~N~N
N'N N and
N
00 0 220 The invention provides a compound having the struacture: c-I x 00
N
Y N N R 1 3 00 wherein W is H, -Cl, -Br, CN, methyl, ethyl, propyl, methoxy or ethoxy; wherein X is; NR11R 12 R1 -N N =0or
R
17 1~ 7 -N
N-R
1 8
R
17 wherein R 11 is H, straight chained or branched C 1 -07 alkyl,
(CH
2 )q0O(CH 2 )m-CH3, aryl, or aryl (Cl-C 6 )alkyl; wherein R 12 is straight chained or branched Cl-C 7 alkyl,
(C
2 )q-O(CH 2 )m-CH 3 or (CH 2 m-Z; wherein' R 13 is a bicyclic alkyl ring system, adamantyl, 00 c-I 221 noradamantyl,
C
3
-C
10 cycloalkyl, heteroaryl, aryl, aryl(Clci
C
6 alkyl, Q, or Q2; 00 wherein aryl may be substituted with one or more Cl-Ci 0 M 5 straight chained or branched alkyl, aryl, heteroaryl, or N (Rig) -Z; 00 wherein Q, is wherein Q2 is to wherein each J is independently 0, S, C(R22)2 or IN.R 4 wherein
R
4 is H; straight chained or branched Cl-C7 alkyl, monofluoroalkyl or polyfluoroalkYl; straight chained or 2o branched
C
2 -C7 alkenyl or alkynyl; C3-C7 cycloalkyl,
CS-C
7 cycloalkeflYl or aryl; 222 wherein Y is NR 14 Rls; ;or wherein R 1 4 is H, straight chained or branched Cl-C 6 alkyl,
(CH
2 )qO1CH 2 )m-CH3, C 3
-C
6 cycloalkyl, or (C(Rig) 2 wherein R 1 5 is straight chained or branched C 3
-C
6 alkyl,
(CH
2 q 0
(CH
2 rnCH 3
C
3
-C
6 cycloalkyl, (C (Rig) 2 1 mN (R 1 6) 2or (C (Rig) 2 )m-Z; wherein R 16 is straight chained or branched Cl-C 7 alkyl, straight chained or branched Cl-C 7 monofluoroalkyl, straight chained or branched
C
1 -C7 polyfluoroalkyl, straight chained or branched
C
2
-C
7 alkenyl, straight chained or branched C 2 -C7 alkynyl, C 5
-C
7 cycloalkenyl,
(CH
2 MZ, or (CH 2 q 0
(CH
2 r-CM 3 00 C- 223 wherein each R 1 is independently H; -OR 2 1 -OCOR21, -COR 21 C1
-NCOR
2 i, -N(R 2 1 2
-CON(R
21 2
-COOR
21 straight chained or branched C 1
-C
7 alkyl, straight chained or branched Ci-C 7 0 monofluoroalkyl, straight chained or branched C 1
-C
7
O
c 5 polyfluoroalkyl, straight chained or branched C 2
-C
7 Salkenyl, straight chained or branched C 2
-C
7 alkynyl, C 5
-C
7 00 cycloalkenyl,
-(CH
2 or (CH 2 )n-O-(CH 2 )m-CH 3 wherein R 1 e is straight chained or branched Ci-C 6 alkyl,
(CH
2 Or (CH 2 )q-O-(CH2)m-CH3; wherein each R 19 is independently H, or straight chained or branched C 1
-C
6 alkyl; wherein each Rao is independently straight chained or branched Ci-C7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3
C
7 cycloalkyl or Cs-C 7 cycloalkenyl; -C1, -Br, or -I; -N02; -N3; -CN; -OR 21
-OCOR
2 1, -COR 2 1, -NCOR21, -N(R 21 2 I CON(R21)2, or -COOR21; aryl or heteroaryl; or two R 20 groups present on adjacent carbon atoms can join together to form a methylenedioxy group; wherein each R21 is independently straight chained or branched CI-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3
C
7 cycloalkyl,
C
5
-C
7 cycloalkenyl, aryl, or aryl(Ci- C) alkyl; wherein each R 22 is independently H, F, C1 or Ci-C 4 straight chained or branched alkyl; 00 00 M 5 wherein p is an integer from 0 to 2 inclusive; 00 wherein q is an integer from 2 to 4 inclusive; wherein t is .1 or 2; wherein U is O, -NR 16 S, C(R 17 2 or -NSO 2
R
16 wherein Z is C 3 -Ci0 cycloalkyl,
C
4
-C
1 cyclic ether, C 4
-C
7 cyclic thioether, aryl, or heteroaryl; or a pharmaceutically acceptable salt thereof.
The invention provides a compound having the structure:
X
NI
R13 Y N N wherein W is H, -Cl, -Br, CN, methyl, ethyl, propyl, methoxy or ethoxy; 225 wherein X i s NR 11
R
12 R17
R
17
-N
R
17
I-
R
17 or wherein R3 11 is H, straight chained or branched
C
1
-C
7 alkyl, (CH2)q-O
(CH
2 ),-fCH 3 aryl or aryl (Cl-C 6 alkyl; wherein
R
1 2 is straight chained or branched C3 1
-C
7 alkyl., (CH2)q-O
(CH
2 )m-CH 3 or -(CH 2 )m-Z; wherein
R
13 is a bicyclic alkyl ring system, aryl or aryl(Cl-C6 )alkyl; wherein Y is NR 14
R
15 00 226
R
20 00 m-N U _p ;or
R
19 c-I P R 1 7 00
R
20
-N-
wherein
R
14 is H, straight chained or branched
CI-C
6 alkyl, (CH2)q-O (CH 2 )m-CH3, C3-C 6 cycloalkyl, or CR92.Z wherein
R
15 is straight chained or branched
C
3
-C
6 alkyl,
(CH
2
(CH
2 )m1,CH 3
C
3
-C
6 cycloalkyl, or (C(Rig) 2 wherein U is 0, -NR 16 S, C(Rl 7 2 or -NS0 2
R,
6 wherein Z is C 3 -Cl 0 cycloalkyl, aryl, or heteroaryl; wherein R 16 is straight chained or branched
C
1
I-C
7 alkyl, straight chained or branched Cl-C 7 monofluoroalkyl, straight chained or branched Cl-C 7 polyfluoroalkyl, straight chained or branched
C
2
-C
7 alkenyl, straight chained or branched
C
2
-C
7 alkynyl,
C
5
-C
7 cycloalkenyl,
(CH
2 or (CH 2 )q0I-CH 2 )m-CH3; 00 C-I 227 wherein each R 1 is independently H; -OR 2 1
-OCOR
2 1
-COR
21 c
-NCOR
2
-N(R
2 1 2
-CON(R
2 1 2 -COOR21, straight chained or branched
C
1
-C
7 alkyl, straight chained or branched
C
1
-C
7 00 monofluoroalkyl, straight chained or branched
C
1
-C,
0C r 5 polyfluoroalkyl, straight chained or branched
C
2
-C
7 alkenyl, straight chained or branched
C
2
-C
7 alkynyl,
C
5
-C
7 00 cycloalkenyl,
-(CM
2 or (CH 2
(CH
2 )m-CH 3 wherein Rie is straight chained or branched
C
1
-C
6 alkyl,
(CH
2 or (CH2)q-O-(CH2 )mCH 3 wherein each R 19 is independently H, or straight chained or branched
C
1
-C
6 alkyl; wherein each R 20 is independently straight chained or branched Cl-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched
C
2 -C7 alkenyl or alkynyl;
C
3
C
7 cycloalkyl or C 5
-C
7 cycloalkenyl; -C1, -Br, or -I;
-NO
2 -N3; -CN; -OR 2 1
-OCOR
2 1, -COR 21 -NCOR2l,
-N(R
21 )2 CON(R21)2, or -COOR 2 1 aryl or heteroaryl; or two R 2 0 groups present on adjacent carbon atoms can join together to form a methylenedioxy group; wherein each R 21 is independently straight chained or branched
C
1
-C
7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched
C
2
-C
7 alkenyl or alkynyl;
C
3 cycloalkyl,
C
5 cycloalkenyl, aryl or aryl(C,-
C
s )alkyl; wherein each m is an integer from 0 to 4 inclusive; 00 00 M 5 r wherein q is an integer from 2 to 4 inclusive; 0 wherein p is an integer from 0 to 2 inclusive; 00 0 O wherein t is 1 or 2; or a pharmaceutically acceptable salt thereof.
The invention provides a compound having the structure:
W
Y N N
H
wherein W is H, -Cl, -Br, CN, methyl, ethyl, propyl, methoxy or ethoxy; wherein X is N(CH 3 2 or 229 wherein R 13 is an aryl, adamantyl, noradamantyl, C 3 -Cj 0 cyc2.oalkyl, heteroaryl, Q, or Q21 wherein aryl. may be substituted with one or more Cl-Cl 0 straight chained or branched alkyl, aryl., heteroaryl, or
N(R
19
-Z;
wherein Q, is wherein Q2 is wherein each J is independently 0, S, C(R 2 2 2 or NR 4 wherein R 4 is straight chained or branched Cj-C 7 alkyl, monofluoroalkyl or polyfluoroalky-; straight chained or branched C 2 alkeny. or alkynyl; C 3
-C
7 cycloalkyl, C 5
-C
7 cycloalkeny. or aryl; 230.
wherein Y is NR 14
R
15 -N \\u R1 ;or wherein R 14 is H, straight chained or branched Cj-C 6 al.kyl,
(CH
2 )q0O(CH2)m-CH3,
C
3 -Cs cycloalkyl, or (C(R 1 s) 2 wherein R15 is straight chained or branched
C
3
-C
6 alkyl,
(CH
2 )qO1CH2)fmCH3I C3-C 6 cycloalkyl, or (C(R 1 9 2
)M-Z;
wherein U is 0, -NR 16 S, C(R 1 7 2 or -NS0 2
R,
6 wherein Z is C 3 -CIO cycloalkyl, aryl, or heteroaryl; is wherein R 16 is straight chained or branched Cl.-C 7 alkyl, straight chained or branched Cl-C 7 monofluoroalky-, straight chained or branched Cl-C 7 polyfluoroalkyl, straight chained or branched
C
2
-C
7 alkenyl, straight 00 231 chained or branched
C
2
-C
7 alkynyl,
C
5
-C
7 cycloalkelyl,
(CH
2 or (CH 2 )q0O-(CH2)m-CH3; wherein each R 17 is independently H; -OR 2 1 -0C0R 21 -C0R 21 00 M 5 -NCOR 21
-N(R
21 2 I -CON(R 21
-COOR
21 straight chained or branched
C
1
-C
7 alkyl, straight chained or branched Cl-C 1 00monofluoroalkYl, straight chained or branched
CI-C
7 polyfluoroalkyl, straight chained or branched
C
2
-C
7 CIalkenyl, straight chained or branched
C
2
-C
7 alkynyl,
C
5
-C
7 cycloalkenYl,
-(CH
2 or (C1 2 2
),-CH
3 wherein R18 is straight chained or branched
CI-C
6 alkyl,
(CH
2 Or (CH2)q-O(C2)m-H3; wherein each Rig is independently H, or straight chained or branched
CI-C
6 alkyl; wherein each R 2 0 is independently straight chained or branched Cl-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched
C
2
-C
7 alkenyl or alkynyl;
C
3 C7 cycloalkYl or C 5
-C
7 cycloalkenyl; -C1, -Br, or -1; -N0 2
-N
3
-OR
2 1 -0C0R 2 1
-COR
2 1, -NCOR 21
-N(R
2 1 2 CON(R21)2, or -C00R 21 aryl or heteroaryl; or two R 2 0 groups present on adjacent carbon atoms can join together to form a methylenedio2xy group; wherein each R 2 1 is independently straight chained or branched Cl-C7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched
C
2 -C7 alkenyl or alkynyl;
C
3
C
7 cycloalkyl,
C
5
-C
7 cycloalkenyl, aryl or aryl (Ci- Cc,)alkyl; 00 0 232 Swherein each R 22 is independently H, F, Cl or C 1
-C
4 straight chained or branched alkyl;
(N
wherein each m is an integer from 0 to 4 inclusive;
S
00 wherein each n is an integer from 1 to 4 inclusive; 0 wherein p is an integer from 0 to 2 inclusive; 00 wherein q is an integer from 2 to 4 inclusive;
(N
wherein t is 1 or 2; or a pharmaceutically acceptable salt thereof.
The invention provides a compound having the structure:
X
W
NR
H
wherein W is H, -Cl, -Br, CN, methyl, ethyl, propyl, methoxy or ethoxy; wherein X is N(CH 3 2 or 00 233 (Ni wherein R 13 is a bicyclic alkyl ring system, aryl or aryl (Cl-C 6 alkyl; 00 5 wherein Y is NR 1 4
R
1 5 00 wherein R 14 is H, straight chained or branched Cj-C 6 alkyl,
(CH
2 )qj0(CH2)m-CH3,
C
3 -Cr, cycloalkyl, or (C(R, 9 2 )m-Z; wherein R 15 is (C (R 19 2 m-N(R 1 6 2 wherein Z is C 3 -CILO cycloalkyl, aryl, or heteroaryl; wherein R 16 is straight chained or branched Cl-C 7 alkyl, straight chained or branched Cl-C 7 monofluoroalkyl, straight chained or branched Cl-C 7 polyfluoroalkyl, straight chained or branched
C
2
-C
7 alkenyl, straight chained or branched
C
2 -C7 alkynyl,
C
5 -C7 cycloalkenyl,
(CH
2 or (CH 2 )qO-(CH 2 )m-CH3; wherein each R 1 ,7 is independently H; -OR 2 1
-OCOR
2 1, -C0R 21 -iqCOR 21
-N(R
21 2
-CON(R
21 2 -C00R 21 straight chained or branched
CI-C
7 alkyl, straight chained or branched CI-C 7 monofluoroalkYl, straight chained or branched Cj-C7 polyfluoroalkYl, straight chained or branched
C
2
-C
7 alkenyl, straight chained or branched
C
2
-C
7 alkynyl, C 5
-C
7 cycloalkenyl.
-(CH
2 or (CH 2 )n0-O(C 2 )m-CH 3 wherein each R 19 is independently H, or straight chained or branched Cj-C 6 alkyl; wherein each R 21 is independently straight chained or r 00 234 branched CI-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; Sstraight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3 C7 cycloalkyl, Cs-C 7 cycloalkenyl, aryl or aryl(C 1 Cs)alkyl; 00 C 0 wherein each m is an integer from 0 to 4 inclusive; Swherein each n is an integer from 1 to 4 inclusive; wherein q is an integer from 2 to 4 inclusive; or a pharmaceutically acceptable salt thereof.
As used in the present invention, the term "bicyclic alkyl ring systems" includes, but is not limited to, bicyclo[2.2.1]heptane, bicyclo[3.1.1]heptane and bicyclo[2.2.2]octane. In addition, the bicyclic alkyl ring systems may be substituted with one or more of the following:
-NO
2 -CN, straight chained or branched
C
1
-C
7 alkyl, straight chained or branched Ci-C 7 monofluoroalkyl, straight chained or branched C 1
-C
7 polyfluoroalkyl, straight chained or branched
C
2
-C
7 alkenyl, straight chained or branched C 2
-C
7 alkynyl, C 3
-C
7 cycloalkyl, Cs-C7 cycloalkenyl,
-N(R
21 2
-OR
21
-COR
21 C0 2
R
21
-CON(R
2 1 )2 or (CH 2 )n-O-(CH 2 )m-CH3.
As used in the present invention, the term "cycloalkyl" includes,
C
3 -C7 cycloalkyl moieties which may be substituted with one or more of the following:
-NO
2 -CN, straight chained or branched Ci-C 7 alkyl, straight chained or branched Ci-C 7 monofluoroalkyl, straight chained or branched CI-C7 polyfluoroalkyl, straight 00 235 chained or branched C 2
-C
7 alkenyl, straight chained or branched C 2
-C
7 alkynyl, C 3
-C
7 cycloalkyl, 3C monof luorocycloalkyl,
C
3 polyf luorocycloalkyl,
C
5
-C,
cycloalkenyl, -N (R 4 2
-OR
4 -COR4, -NCOR4, -C0 2 R4, 00 5 qON(R 4 2 or (CH 2 )n-O(CH 2
)MCH
3 CIAs used in the present invention, the term "cyclohexyl", 00 includes, cyclohexyl groups which may be substituted with c-ione or more of the following:
-NO
2 -CN, straight chained or branched CI-C7 alkyl, straight chained or branched Cl-C7 monofluoroalkyl, straight chained or branched -C 1
-C
7 polyfluoroalkyl, straight chained or branched C 2 alkenyl, straight chained or branched C 2 -C7 alkynyl, C 3
-C
7 cycloalkyl, C 3 monof luorocycloalkyl, C 3 polyfluorocycloalkyl, C 5 -C7 cycloalkenyl, -N(R4)2, -OR 4
COR
4 -NCOR4, -C0 2 R4, -CON(R.,) 2 or (CH 2 )n-01(CH 2 )M-CH3.
As Used in the present invention, the term "cycloalkenyl" includes, Cs-C7 cycloalkenyl moieties which may 'be substituted with one or more of the following: -Cl, -Br, -NO 2 -EN, straight chained or branched C 1
-C,
alkyl, straight chained or branched Cl-C7 mono fluoroalkyl, straight chained- or branched C2-C., polyfluoroalkyl, straight chained or branched C 2 alkenyl, straight chained or branched C 2 alkynyl, C 3 cycloalkyl, C 3
-C
7 monofluorocycloalkyl, C 3 polyfluorocycloalkyl, C 5 cycloalkenyl, N(RO)2, -OR4, -COR4, -NCOR4, -C0 2
R
4
CON(R
4 2 or (CH 2 )n-O-WCH 2
)M-CH
3 In the present invention, the term "heteroaryl" is used to include five and six memnbered unsaturated rings that may contain one or more oxygen, sulfur, or nitrogen 00 236 atoms. Examples of heteroaryl groups include, but are not limited to, furanyl, thienyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl, 00 M pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, and triazinyl.
00 in addition the term "heteroaryll is used to include fused bicyclic ring systems that may contain one or more heteroatoms such as oxygen, sulfur and nitrogen. Examples of such heteroaryl groups include, but are not limited to, indolizinyl, indolyl, isoindolyl, benzo~blfuranyl, benzo~b) thiophenyl, indazolyl, benzimidazolyl, purinyl, benzoxazolYl, benzisoxazolyl, benzo thiazolyl, imidazo thiazolyl, cinnolinyl, quinazolinyl, quinoxalinyl, 1, 8-naphthyridilyl, pteridinyl, quinolinyl, isoquinolinyl, phthalimidyl and 2,1, 3-benzothiazolyl.
The term "heteroaryl" also includes those chemical moieties recited above which may be substituted with one or more of the following: -Cl, -Br, -N0O2, -CN, straight chained or branched Cl-C7 alkyl, straight chained or branched Cl-C7 monofluoroalkyl, straight chained or branched Cl-C7 polyfluoroalkyl, straight chained or branched
C
2 -C7 alkenyl, straight chained or branched
C
2 -C7 alkynyl,
C
3
-C
7 cycloalkyl,
C
3 monofluorocycloalkyl,
C
3 -C7 polyfluorocycloalkyl,
C
5 cycloalkenyl, -N(R4)2,
-OR
4
COR
4
-NCOR
4 -C0 2
R
4
-CON(R
4 2 or (CH 2 )n,-O-(CH 2
),-CH
3 The term "heteroaryl" further includes the N-oxides of those chemical moieties recited above which include at least one nitrogen atom.
00 237 CI In the present invention the term "aryl" is phenyl or naphthyl. The term "aryl" also includes phenyl and naphthyl which may be substituted with one or more of the 00 M 5 following: -Cl, -Br,
-NO
2 -CN, straight chained or branched
C
1
-C
7 alkyl, straight chained or branched Cl-C 7 00 monofluoroalkyl, straight chained or branched C3.- C 7 polyfluoroalkyl, straight chained or branched
C
2
-C
7 alkenyl, straight chained or branched
C
2
-C
7 alkynyl,
C
3
-C
7 cycloalkyl,
C
3
-C
7 'monofluorocycloalkyl, 3C polyfluorocycloalkyl,
C
5
-C
7 cycloalkenyl,
-N(R
4 2 -0R4, SR,, -OCQR 4
-COR
4
-NCOR
4 -C0 2
R
4
-CON(R
4 2 or (CH 2
(CH
2 m-CH 3 In one embodiment of any of the compounds described herein, the compound is enantiomerically or diasteriomerically pure. In one embodiment of any of the compounds described herein, the compound is enantiolnericallY and diasteriomerically pure.
In one embodiment, X is:
R
17 -N or
NN-R
1 8 238 In one embodiment, X is NRRn 1 and R 11 is H or straight chained or branched C 1
-C
7 alkyl.
In one embodiment, the compound has the structure: In one embodiment,
R
13 is a bicyclic alkyl ring system, cyclohexyl or aryl.
In one embodiment,
R
1 4 is H, straight chained or branched
C
1
-C
6 alkyl or (CH 2 q-O-(CH2)m-CH3 In one embodiment, Y is -N
U
R
17 In one embodiment, U is NR16.
00 239 In one embodiment, R 16 is (CH 2 In one embodiment Z is aryl or heteroaryl.
00 5 In one embodiment, Y is 00
R
17 _N U In one embodiment, U is Nfl 16 In one embodiment, the compound is selected from the group consisting of: N CI N/ CI
N
N CI ONPNN N'o
N
NN aCI NNN N C CI A. ftC ;and In one embodiment, the compound is selected from the group consisting of: 200820038025Jn08 25 Jan 2008
Q
zz z z -nI
CL
z z z 0 01 n 0 >/z 00 242 In one embodiment, Y is c-I
R
17 00 M-N
U
17 00 In one embodiment, R 13 is an aryl substituted with a Cl-Cl 0 straight chained alkyl.
00 0243 SIn one embodiment, the compound is selected from a group n consisting of: 00 M
N
00 NN'N SN N
\N
s dd h n a'N and acceptable carrier.
N
The invention provides a pharmaceutical composition comprising a therapeutically effective amount of any of the compounds described herein and a pharmaceutically acceptable carrier.
The invention provides a pharmaceutical composition made by combining a therapeutically effective amount of any of the compounds described herein and a pharmaceutically acceptable carrier.
00 C 244 Cl The invention provides a process for making a pharmaceutical composition comprising combining a therapeutically effective amount of any of the compounds 00 described herein and a pharmaceutically acceptable carrier.
00 SThe invention provides a method of treating a subject suffering from depression which comprises administering to the subject an amount of any of the compounds described herein effective to treat the subject's depression.
The invention provides a method of treating a subject suffering from anxiety which comprises administering to the subject an amount of any of the compounds described herein effective to treat the subject's anxiety.
The invention provides a method of treating a subject suffering from depression and anxiety which comprises administering to the subject an amount of any of the compounds described herein effective to treat the subject's depression and anxiety.
00 245 c' The invention provides a method of treating a subject C-i suffering from depression which comprises administering to the subject an amount of compound effective to treat the subject's depression wherein the compound has the 00 Cc structure: 00 B
CY
Y3
N
A
Y4 wherein each of Y 1
Y
2
Y
3 and Y 4 is independently H; straight chained or branched C 1
-C
7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3
-C
7 cycloalkyl, or Cs-C 7 cycloalkenyl; -Cl, -Br, or I; -NO 2
-N
3 -CN; -OR4, -SR 4 -OCOR4, -COR 4
-NCOR
4 N(R4)2 -CON(R 4 2 or -COOR 4 aryl or heteroaryl; or any two of Yi, Y 2
Y
3 and Y 4 present on adjacent carbon atoms can constitute a methylenedioxy group; wherein each R 4 is independently straight chained or branched Ci-C7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3
-C
7 cycloalkyl, Cs-C 7 cycloalkenyl, aryl or aryl(Ci-C 6 )alkyl; 246 wherein A is A' Q3, Q4, Q5, straight chained or branched CI-C 7 alkyl, aryl, heteroaryl, aryl(Cl- CO) alkyl, heteroaryl (Cl-C 6 alkyl, aryl substituted with an aryl or heteroaryl, heteroary. substituted with an aryl or heteroaryl; or (CHR 17
)-(CHR
17 wherein A' is 0
R
1 n CR 2
R
3 ;or (CH 2 n 1 wherein Q 3 is r 247 wherein Q4 is wherein Qs is wherein Ri and R 2 are each independently H, straight chained or branched Ci-C7 alkyl, -Cl, -Br,
NO
2 or -CN; wherein R 3 is H, straight chained or branched C 1
-C
7 alkyl, -C1, -Br, -NO 2 -CN, -OR 6 aryl or heteroaryl; wherein Rs is straight chained or branched C 1 -C7 alkyl, -N(R 4 2
-OR
6 or aryl; wherein R 6 is straight chained or branched Ci-C 7 alkyl or aryl; 00 248 wherein each R 17 is independently H; straight chained eC or branched C 1
-C
7 alkyl, straight chained or branched
C
1
-C
7 monofluoroalkyl, straight chained or branched Ci-C 7 polyfluoroalkyl, straight chained or branched 00 CrM C 2
-C
7 alkenyl, straight chained or branched C 2
-C
7 alkynyl, Cs-C 7 cycloalkenyl, -(CH 2 or (CH 2 )n-0- 00
(CH
2
).-CH
3 C 10 wherein each R 20 is independently straight chained or branched Ci-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3 -C7 cycloalkyl or C 5
-C
7 cycloalkenyl; -Cl, -Br, or -NO 2
-N
3 -CN;
OR
21
-OCOR
21
-COR
2 1
-NCOR
21
-N(R
21 2
-CON(R
21 2 or
-COOR
21 aryl or heteroaryl; or two R 20 groups present on adjacent carbon atoms can join together to form a methylenedioxy group; wherein each R 21 is independently straight chained or branched CI-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3 -C7 cycloalkyl, C 5
-C
7 cycloalkenyl, aryl or aryl(C 1
-C
6 )alkyl; wherein each m is an integer from 0 to 4 inclusive; wherein each n is an integer from 1 to 4 inclusive; wherein each p is an integer from 0 to 2 inclusive; wherein U is O, -NR16, S, C(R 7 or -NSO 2 R16; 00 S249 wherein Z is C 3
-C
1 0 cycloalkyl, C 4 -C7 cyclic ether, Ci
C
4
-C
7 cyclic thioether, aryl, or heteroaryl; wherein R 16 is straight chained or branched Ci-C 7 00 M alkyl, straight chained or branched Ci-C 7 monofluoroalkyl, straight chained or branched Ci-C 7 C polyfluoroalkyl, straight chained or branched C 2 -C7 00 Salkenyl, straight chained or branched C 2
-C
7 alkynyl, C< 10 C 5 -C7 cycloalkenyl, -(CH 2 or (CH 2
)-O-(CH
2 )m-CH 3 wherein q is an integer from 2 to 4 inclusive; wherein B is aryl, heteroaryl, aryl substituted with an aryl or heteroaryl, heteroaryl substituted with an aryl or heteroaryl, tricyclic heteroaryl or Q6; provided however, if B is aryl or heteroaryl the carbon atom or carbon atoms ortho to the nitrogen atom of the imine bond may only be substituted with one or more of the following -Cl, -Br, -CN, methyl, ethyl or methoxy; wherein a tricyclic heteroaryl is a fused three member aromatic system in which one or more of the rings is heteroaryl; carbazole; or acridine; wherein Q6 is 00 250 wherein each R 22 is independently H, F, CI Cl, or straight chained or branched C 1
-C
4 alkyl; or a pharmaceutically acceptable salt thereof.
00 The invention provides a method of treating a subject
C
suffering from depression which comprises administering Sto the subject an amount of compound effective to treat C 10 the subject's depression wherein the compound has the structure:
B
Y/
N
Y2
A
Y4 wherein each of Y 1
Y
2
Y
3 and Y 4 is independently H; straight chained or branched C 1
-C
7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C3-C7 cycloalkyl, or Cs-C 7 cycloalkenyl; -Cl, -Br, or I; -NO 2
-N
3 -CN; -OR 4 -SR4, -OCOR 4 -COR4, -NCOR4,
N(R
4 2
-CON(R
4 2 or -COOR 4 aryl or heteroaryl; or any two of Y 1
Y
2
Y
3 and Y 4 present on adjacent carbon atoms can constitute a methylenedioxy group; wherein each R 4 is independently straight chained or branched CI-C7 alkyl, monofluoroalkyl or 251 polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3 C- cycloalkyl, C 5
-C
7 cycloalkenyl, aryl or aryl(Cl-C 6 )alkyl; wherein A is straight chained or branched CI-C 7 alkyl, aryl, heteroaryl, aryl (CI -CO)alkyl or heteroaryl (Cl-C 6 alkyl; nI wherein A' is
R
1 n
CR
2
R
3 or -(CH 2 n R wherein R, and R 2 are each independently H, straight chained or branched CI 1
-C
7 alkyl, -Cl, -Br,
NO
2 or -CN; wherein R 3 is H, straight chained or branched Cl-C 7 alkyl, -Cl, -Br, -NO 2 -CN, -OR 6 aryl or heteroaryl; wherein R 5 is straight chained or branched Cj-C 7 252 alkyl, -N(R 4 2
-OR
6 or aryl; wherein Rs is straight chained or branched Ci-C7 alkyl or aryl; wherein B is aryl, or heteroaryl; provided however, if B is aryl or heteroaryl the carbon atom or carbon atoms ortho to the nitrogen atom of the imine bond may only be substituted with one or more of the following -C1, -Br, -CN, methyl, ethyl or methoxy; wherein n is an integer from 1 to 4 inclusive; or a pharmaceutically acceptable salt thereof.
The invention provides a method of treating a subject suffering from depression which comprises administering to the subject an amount of compound effective to treat the subject's depression wherein the compound has the structure: wherein each of Y 1
Y
2
Y
3 and Y 4 is independently H; straight chained or branched Ci-C 7 alkyl, 00 253 monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or alkynyl; C 3
-C
7 cycloalkyl, Or C5-C 7 cycloalkenyl; -C1, -Br, or 1; -NO 2
-N
3 -CN; -OR 4 -SR4, -OCOR4, -COR 4
-NCOR
4
N(R
4 2 I -CON(R 4 2 or -COOR 4 aryl or heteroaryl; or 00 any two of Y 1
Y
2
Y
3 and Y 4 present on adjacent carbon atoms can constitute a methylenedioxy group; 00 wherein each R 4 is independently -H1; straight chained or branched Cl-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; "straight chained or branched C 2 -C7 alkenyl or alkynyl; C 3
-C
7 cycloalkyl, C 5
-C
7 cycloalkenyl, aryl or aryl(Cl-Cf 6 )alkyl; wherein A is straight chained or branched Cl-C 7 alkyl, aryl, heteroaryl, aryl (C C 6 alkyl or heteroaryl (Cl-C 6 alkyl; wherein A' is 0 0 nn' n CR2R or -(CH 2 nR 00 S254 l r wherein B is aryl substituted with an aryl or heteroaryl, heteroaryl substituted with an aryl or heteroaryl, tricyclic heteroaryl or Qs; 00 Swherein a tricyclic heteroaryl is a fused three ring 0 aromatic system in which one or more of the rings is 00 Sheteroaryl; carbazole; or acridine; wherein Qs is 0 R2 0 R22 wherein n is an integer from 1 to 4 inclusive; wherein each R 22 is independently H, F, Cl, or straight chained or branched Ci-C 4 alkyl; or a pharmaceutically acceptable salt thereof.
The invention provides a method of treating a subject suffering from depression which comprises administering to the subject an amount of compound effective to treat the subject's depression wherein the compound has the structure: 00 255 000 wherein each of Y 1
Y
2
Y
3 and Y 4 is independently H; straight chained or branched C 1
-C
7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3
-C
7 cycloalkyl, or CS-C 7 cycloalkenyl; -C1, -Br, or 1; -NO 2
-N
3 -CN; -OR 4 -SR4, -OCOR4, -COR4, -NCOR 4 N(R42 ,-CON(R 4 2 or -COOR4; aryl or heteroaryl; or any two of Y 1
Y
2
Y
3 and Y 4 present on adjacent carbon atoms can constitute a methylenedioxy group; wherein each R 4 is independently straight chained or branched CI-C7 alkyl, ronofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3
-C
7 cycloalkyl, C 5
-C
7 cycloalkenyl, aryl or aryl (Cl-Cs) alkyl; wherein A is Q3, Q4~, Q5, aryl substituted* with an aryl or heteroaryl, heteroaryl substituted with an.
aryl or heteroaryl, or (CHR 17
-(CHR
1 7 wherein 03 is 256 wherein Q4 iB wherein Q5 is wherein each R 17 is independently H; straight chained or branched Cl-C 7 alkyl, straight chained or branchied Cl-C 7 mono fluoroal kyl, straight chained or branched cl-C 7 polyfluoroalkyl, straight chained or branched
C
2 -C-7 alkenyl, straight chained or branched C 2
-C?
alkynyl, C 5 -C7 cycloalkenyl, -(C1 2 )mZ, or (CH 2
(CH
2
)~.CH
3 00 S257 Swherein each R 20 is independently -HI straight chained or branched Ci-C7 alkyl, monofluoroalkyl or V) polyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or alkynyl; C 3 -C7 cycloalkyl or Cs-C7 cycloalkenyl; -C1, -Br, or -NO 2 -N3; -CN; 00
OR
2 1 -0COR 21
-COR
21
-NCOR
21
-N(R
21 2
-CON(R
21 2 or
S-COOR
21 aryl or heteroaryl; or two R 20 groups present C( on adjacent carbon atoms can join together to form a 00 Smethylenedioxy group; 010 wherein each R 21 is independently straight chained or branched C 1 -C7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or alkynyl; C 3
-C
7 cycloalkyl, Cs-C7 cycloalkenyl or aryl; wherein each R 22 is independently H, F, Cl, or straight chained or branched C 1
-C
4 alkyl; wherein q is an integer from 2 to 4 inclusive; wherein each m is an integer from 0 to 4 inclusive; wherein each n is an integer from 1 to 4 inclusive; wherein each p is an integer from 0 to 2 inclusive; wherein U is O, -NR16, S, C(R 17 2 or -NS0 2
R
16 wherein Z is C 3 -Co 0 cycloalkyl, C 4 -C7 cyclic ether,
C
4 -C7 cyclic thioether, aryl, or heteroaryl; 00 0 258
O
(N
Swherein R 16 is straight chained or branched Ci-C 7 calkyl, straight chained or branched C 1
-C,
Smonofluoroalkyl, straight chained or branched C 1 -07 polyfluoroalkyl, straight chained or branched C 2
-C
7 alkenyl, straight chained or branched C 2 -C7 alkynyl, 00 Cs 5
-C
7 cycloalkenyl, -(CH 2 or (CH 2 )q-O-(CH 2 )m-CH 3 C wherein B is aryl, or heteroaryl; provided however, 00 Sif B is aryl or heteroaryl the carbon atom or carbon N 10 atoms ortho to the nitrogen atom of the imine bond may only be substituted with one or more of the following -Cl, -Br, -CN, methyl, ethyl or methoxy; or a pharmaceutically acceptable salt thereof.
As used in the present invention, the term "cycloalkyl" includes C 3
-C
7 cycloalkyl moieties which may be substituted with one or more of the following:
-NO
2 -CN, straight chained or branched Ci-C 7 alkyl, straight chained or branched Ci-C 7 monofluoroalkyl, straight chained or branched
CI-C
7 polyfluoroalkyl, straight chained or branched
C
2 -C7 alkenyl, straight chained or branched C 2
-C
7 alkynyl, C 3 -C7 cycloalkyl, C 3
-C
7 monofluorocycloalkyl,
C
3
-C
7 polyfluorocycloalkyl, Cs-C 7 cycloalkenyl, N(R4)2, -OR4, -COR 4
-NCOR
4
-CO
2
R
4
-CON(R
4 2 or
(CH
2
(CH
2
CH
3 As used in the present invention, the term "cycloalkenyl" includes Cs-C, cycloalkenyl moieties which may be substituted with one or more of the 00 259 f ol lowing: -Cl, -Br, -NO 2 -CN, straight chained or branched Cl-C7 alkyl, straight chained or branched C 1 -C7 mono fluoroal kyl, straight chained or branched Cl-C7 polyfluoroalkyl, straight chained or branched C 2 -C7 alkenyl, straight chained or branched 00 M 2- 7 alkynyl, C 3 -C7 cycloalkyl, 3C monofluorocycloalky., C 3 -C7 polyfluorocycloalkyl, C 5 00 C7 cycloalkenyl, -N(R 4 2
-OR
4
-COR
4
-NCOR
4 C0 2 R4, -CON(R 4 2 or (CH 2 )n0O(CH 2 )m-CH 3 In the present -invention, the term "heteroaryl" is used to include five and six membered unsaturated rings that may contain one or more oxygen, sulfur, or nitrogen atoms. Examples of heteroaryl groups include, but are not limited to, furanyl, thienyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, and triazinyl.
In addition the term "heteroaryl" is used to include fused bicyclic ring systems that may contain one or more heteroatoms such as oxygen, sulfur and nitrogen. Examples of such heteroaryl groups include, but are not limited to, indolizinyl, indolyl, isoindolyl, benzo [bi furanyl, benzo (bithiophenyl, indazolyl, benzimidazolyl, purinyl, benzoxazolyl, benzisoxazolyl, benzo thiazolyl, imidazo thiazolyl, cininoliiyl, quinazolinyl, quinoxalinyl, 1,8naphthyridinyl, pteridinyl, quinolinyl, isoquinolinyl, phthalimidyl and 2,1,3- 00 260 benzothiazolyl.
The term "heteroaryl" also includes those chemical moieties recited above which may be substituted with one or more of the following: -Cl, -Br, 00 M
NO
2 -CN, straight chained or branched C 1 -C7 alkyl, straight chained or branched Cl-C7 mono fluoroalkyl, 00 straight chained or branched Cl-C7 polyfluoroalkyl, straight chained or branched C 2 -C7 alkenyl, straight chained or branched C 2 alkynyl, C3-C? cycloalkyl, C3-C7 monofluorocycloalkyl, 3C polyfluorocycloalkyl,
C
5 -C,7 cycloalkenyl,
-N(R
4 2
OR
4 -COR4, -NCOR4, -C0 2 R4, -CO1N(R 4 2 Or (CH 2 )n-O-
(CH
2 )m-CH 3 The term "heteroaryl" further includes the N-oxides of those chemical moieties recited above which include at least one nitrogen atom.
In the present invention the term "aryl" is phenyl or naphthyl. The term "'aryl"' also includes phenyl and naphthyl which may be substituted with one or more of the following: -Cl, -Br, -NO 2
-CN,
straight chained or branched C 1 -C7 alkyl, straight chained or branched CI-C7 monofluoroalkyl, straight chained or branched C 1 I-C7 polyfluoroalkyl, straight chained or branched C 2 -C7 alkenyl, straight chained or branched C 2 -C7 alkynyl, C 3 -C7 cycloalkyl, C 3 -C7 monofluorocycloalkYl,
C
3 -C7 polyfluorocycloalkyl, C,7 cycloalkenyl,
-N(R
4 2 -OP4, -SR4, -OCOR 4
COR
4 NCOR4, -C0 2 R4, -CON(R4)2 or (CH 2 )n0I-CH 2 )m-CH 3 261 The present invention also provides a method of treating a subject suffering from depression which compromises administering to the subject an amount of compound effective to treat the subject's .depression, wherein the compound has the structure: 00 00 0D wherein each R 24 is independently one or more of the following: H, F, Cl, Br, I, CF 3 OCH3 or NO 2 wherein R 25 is methyl, ethyl, allyl, phenyl and the phenyl is optionally substituted with a F, Cl, Br,
CF
3
NO
2 In one embodiment of any one of the methods described herein, the compound is enantiomerically or diastereomerically pure. In one embodiment of any of the methods described herein, the compound is enantiomerically and diastereomerically pure.
In one embodiment, the compound is a pure Z imine isomer 262 or a pure Z alkene isomer. In one embodiment, the compound is a pure E imine isomer or a pure E alkene isomer.
In one embodiment, the compound is administered orally.
In one embodiment, the compound has the structure:
B
Y
p N N Y2 1
A
Y,4 wherein each of Y 1
Y
2
Y
3 and Y 4 is independently H; straight chained or branched CI-C 7 alkyl, -CF 3 F, -Cl, -Br, -OR 4 -N(R4) 2 or -CON(R 4 2 wherein each R 4 is independently straight chained or branched C 1
-C
7 alkyl, -CF 3 or phenyl; wherein A is straight chained or branched C-C 7 alkyl, aryl, heteroaryl, aryl (Ci-C) alkyl or heteroaryl(Ci-C 6 )alkyl; and wherein A' is 263 CR 2 n CR2R3 In one embodiment, B is heteroaryl. In one embodiment, B is aryl.
In one embodiment, B 'is phenyl and the phenyl is optionally substituted with one or more of the following: -Cl, -Br, -CF3, straight chained or branched CI-C 7 alkyl, -N(R 4 2
-OR
4 -COR, -NCOR 4 -C0 2
R
4 or -CON(R 4 2 In one embodiment, heteroaryl.
In one embodiment, group consisting of:
F
A is aryl. In one embodiment, A is the compound is selected from the
CI
N Cl 1, and
CN
h I
S
264 In one embodiment, the compound is selected from the group consisting of:
F
F
F
N
ii
N
\\y
F
F
N
0 2008200380 25 Jan 2008 Ul 0- 266 00
NC
N 0 Cl 00 M 6 00 In one embodiment, A is A' and A, is n CR 2
R
In one embodiment, the compound is: Cl
N
or Cl Cl
N
00 267 In one embodiment, B is Q6.
In one embodiment, A is aryl.
00 MIn one embodiment, the compound has the structure: 00 oot In one embodiment, the compound is:
OF
In one embodiment, B is aryl.
00 268 c In one embodiment, A is (CHR 17
(CHR
17 )n-Z.
l t In one embodiment, the compound is: 00 CcC Clc 00 d
CC
-N
The invention provides a method of treating a subject suffering from anxiety which comprises administering to the subject an amount of compound effective to treat the subject's anxiety wherein the compound has the structure:
B
Y1 N Y4
A
Y4 wherein each of Y 1
Y
2
Y
3 and Y 4 is independently 00 269 H; straight chained or branched
CI-C
7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched
C
2
-C
7 alkenyl or alkynyl;
C
3
-C
7 cycloalkyl, or Cs-C7 cycloalkelyl; -C1, -Br, or 1; -NO 2
-N
3 -CN; -OR 4
-SR
4 -OCOR4, -COR4, -NCOR 4 00 M ~N(R 4 2 -CON(R4 2 or -COOR4; aryl or heteroaryl; or any two of Y 1
Y
2
Y
3 and Y 4 present on adjacent 00 carbon atoms can constitute a methylenedioxy group; wherein each R 4 is independently straight chained 'or branched
C
1
-C
7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl;
C
3
-C
7 cycloalkyl,
C
5
-C
7 cycloalkenyl, aryl or aryl(Cl-C6alkyl; wherein A is Q3, Q4, Q5, straight chained or branched
C
1
-C
7 alkyl, aryl, heteroaryl, aryl (Cl-
C
6 )alkyl, heteroaryl alkyl, aryl substituted with an aryl or heteroaryl, heteroaryl substituted with an aryl or heteroaryl; or (CHR 1
(CHR
17
Z;
wherein A' is 0 0 n I
R
1 n
CR
2
R
3 or(H)'4 270 wherein Q3 is wherein Q4 is wherein Qs is 00 D 271 c' wherein Ri and R 2 are each independently H, straight n chained or branched CI-C 7 alkyl, -Cl, -Br,
NO
2 or -CN; 00 0 wherein R 3 is H, straight chained or branched C 1
-C
7 Oalkyl, -Cl, -Br, -NO 2 -CN, -OR 6 aryl or C heteroaryl; 00 CI 10 wherein Rs is straight chained or branched C 1
-C
7 .alkyl, -N(R 4 2 -ORe or aryl; wherein Rs is straight chained or branched Ci-C 7 alkyl or aryl; wherein each R 17 is independently H; straight chained or branched C 1
-C
7 alkyl, straight chained or branched Ci-C 7 monofluoroalkyl, straight chained or branched Ci-C 7 polyfluoroalkyl, straight chained or branched
C
2 -C7 alkenyl, straight chained or branched C 2
-C
7 alkynyl, Cs-C 7 cycloalkenyl,
-(CH
2 or (CH 2 )n-O-
(CH
2 )m-CH 3 wherein each R 20 is independently straight chained or branched C 1
-C
7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched
C
2
-C
7 alkenyl or alkynyl; C 3
-C
7 cycloalkyl or Cs-C 7 cycloalkenyl; -C1, -Br, or -NO 2
-N
3 -CN;
OR
21
-OCOR
21
-COR
2 1
-NCOR
21
-N(R
2 1)2 -CON(R21)2, or
-COOR
21 aryl or heteroaryl; or two R 2 0 groups present on adjacent carbon atoms can join together to form a methylenedioxy group; 00 0 272 c- wherein each R 21 is independently straight chained or branched Ci-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3 -C7 cycloalkyl, C 5
-C
7 00 00 cycloalkenyl, aryl or aryl(C 1
-C
6 )alkyl; Cl wherein each m is an integer from 0 to 4 inclusive; 00 Ci 10 wherein each n is an integer from 1 to 4 inclusive; wherein each p is an integer from 0 to 2 inclusive; wherein U is O, -NR 16 S, C(Ri 7 2 or -NSO 2
R
16 wherein Z is C 3 -Cio cycloalkyl, C 4 -C7 cyclic ether,
C
4 -C7 cyclic thioether, aryl, or heteroaryl; wherein Rs 1 is straight chained or branched Ci-C 7 alkyl, straight chained or branched C 1
-C
7 monofluoroalkyl, straight chained or branched Ci-C 7 polyfluoroalkyl, straight chained or branched C 2 -C7 alkenyl, straight chained or branched C 2
-C
7 alkynyl, Cs-C 7 cycloalkenyl, -(CH 2 or (CH 2 )q-0-(CH2)m-CH3; wherein q is an integer from 2 to 4 inclusive; wherein B is aryl, heteroaryl, aryl substituted with an aryl or heteroaryl, heteroaryl substituted with an aryl or heteroaryl, tricyclic heteroaryl or Q6; provided however, if B is aryl or heteroaryl the carbon atom or carbon atoms ortho to the nitrogen 00 0 273 Satom of the imine bond may only be substituted with Sone or more of the following -Cl, -Br, -CN, n methyl, ethyl or methoxy; wherein a tricyclic heteroaryl is a fused three 0 0 member aromatic system in which one or more of the Srings is heteroaryl; carbazole; or acridine; 00 Swherein Q6 is to O Rz o0 R22 wherein each R 22 is independently H, F, Cl, or straight chained or branched C 1
-C
4 alkyl; or a pharmaceutically acceptable salt thereof.
The invention provides a method of treating a subject suffering from anxiety which comprises administering to the subject an amount of compound effective to treat the subject's anxiety wherein the compound has the structure:
B
I N Y2f 0 00 274 wherein each of YI, Y 2
Y
3 and Y 4 is independently H; straight chained or branched -Cl-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3
-C
7 00 cycloalkyl, or C.S-C 7 cycloalkenyl; -C1, -Br, or 1; -NO 2
-N
3 -CN; -OR 4 -SR4, -OCOR 4 -COR4, -NCOR,, N(R42 ,-CON(R 4 2 or -COOR 4 aryl or heteroaryl; or 00 any two of Y 1
Y
2
Y
3 and Y 4 present on adjacent carbon atoms can constitute a methylenedioxy group; wherein each R 4 is independently straight chained or branched C 1
-C
7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C,
alkenyl or alkynyl; C 3
-C
7 cycloalkyl, C 5 -C7 cycloalkenyl, aryl or aryl(Cl-C 6 )alkyl; wherein A is straight chained or branched Cl-C 7 alkyl, aryl, heteroaryl, aryl (Cl-C 6 alkyl or heteroaryl (Cl-C 6 alkyl; wherein A' is 0 0 n 5n I in 00 275
RI
CR R or (CH2)n 4 n CR 2
R
3 0 0 0 wherein RI and R 2 are each independently H, straight Schained or branched C 1
-C
7 alkyl, -Cl, -Br, S5 NO 2 or -CN; 00 CI wherein R 3 is H, straight chained or branched C 1
-C
7 alkyl, -Cl, -Br, -NO 2 -CN, -OR aryl or heteroaryl; wherein Rs is straight chained or branched Ci-C 7 alkyl, -N(R 4 2
-OR
6 or aryl; wherein R 6 is straight chained or branched C 1
-C
7 alkyl or aryl; wherein B is aryl, or heteroaryl; provided however, if B is aryl or heteroaryl the carbon atom or carbon atoms ortho to the nitrogen atom of the imine bond may only be substituted with one or more of the following -Cl, -Br, -CN, methyl, ethyl or methoxy; wherein n is an integer from 1 to 4 inclusive; or a pharmaceutically acceptable salt thereof.
The invention provides a method of treating a subject suffering from anxiety which comprises administering to 00 276 the subject an amount of compound effective to treat the subject's anxiety wherein the compound has the structure: 00 000 wherein each of Y 1
Y
2
Y
3 and Y 4 is independently- H; straight chained or branched
C
1 -C,7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched
C
2 alkenyl or alkynyl;
C
3
-C
7 cycloa.kyl, or CB-C 7 cycloalkenyl; -C1, -Br, or 1; -NO 2 -N3; -CN; -OR 4
-SR
4
-OCOR
4 -COR4, -NCOR 4 N(R4) 2 -CON(Rd) 2 or -COOR4; aryl or heteroaryl; or any two Of YI, Y 2
Y
3 and Y 4 present on adjacent carbon atoms can constitute a methylenedioxy group; wherein each R 4 is independently straight chained or branched
C
1
-C
1 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched
C
2
-C
7 aJlkenyl or alkynyl;
C
3
-C
7 cycloalkyl,
C
5
C
7 cycloalkenyl, aryl or aryl(Cl-C6 )alkyl; wherein A is straight chained or branched Cl-C 7 alkyl, aryl, heteroaryl, aryl (C 1
-C
6 alkyl. or heteroaryl (Cl-C 6 alkyl; wherein A' is C00 cv 0q 277 0 R3.
n CR 2
R
3 Sor (CH 2 )I
R
4 wherein B is aryl substituted with an aryl or heteroaryl, heteroaryl substituted with an aryl or heteroaryl, tricyclic heteroaryl or Q6; wherein a tricyclic heteroaryl is a fused three ring aromatic system in which one or more of the rings is heteroaryl; carbazole; or acridine; wherein Qs is wherein n is an integer from 1 to 4 inclusive; wherein each R 22 is independently H, Cl, or straight chained or branched C 1
-C
4 alkyl; 00 S278 or a pharmaceutically acceptable salt thereof.
The invention provides a method of treating a subject 0 0 suffering from anxiety which comprises administering to 0 the subject an amount of compound effective to treat the C subject's anxiety wherein the compound has the structure: 00 CD B 1 N Y2
N
3
A
Y4 wherein each of Y 1
Y
2
Y
3 and Y 4 is independently H; straight chained or branched Ci-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3
-C
7 cycloalkyl, or Cs-C 7 cycloalkenyl; -C1, -Br, or I; -NO 2
-N
3 -CN; -OR 4 -SR4, -OCOR 4
-COR
4
-NCOR
4
N(R
4 2 -CON(R4) 2 or -COOR4; aryl or heteroaryl; or any two of Yi, Y 2
Y
3 and Y 4 present on adjacent carbon atoms can constitute a methylenedioxy group; wherein each R 4 is independently straight chained or branched CI-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -07 alkenyl or alkynyl; C 3
-C
7 cycloalkyl, C 5 -C7 cycloalkenyl, aryl or aryl (CI-C 6 alkyl; 279 wherein A is Q3, Q4, Q5, aryl. substituted with an aryl or heteroaryl, heteroaryl substituted with an aryl or heteroaryl, or (CHR 17
)-(CHR
1 7 )n-Z; wherein Q3 N R R1 17
R
17 wherein 04 is wherein Q5 is wherein each R 1 7 is independently H; straight chained or branched Cj-C 7 alkyl, straight chained or branched Cl-C 7 mono fluoroal kyl, straight chained or branched
C
1 I-c 7 polyfluoroalkyl, straight chained or branched
I
0280
SC
2
-C
7 alkenyl, straight chained or branched C 2 -C7 Salkynyl, Cs-C 7 cycloalkenyl, -(CH 2 or (CH 2 mf (CH 2 m-CH 3 00 wherein each R 20 is independently straight 0 chained or branched Ci-C 7 alkyl, monofluoroalkyl or (C polyfluoroalkyl; straight chained or branched C 2 -C7 00 alkenyl or alkynyl; C 3 -C7 cycloalkyl or C 5
-C
7 cycloalkenyl; -C1, -Br, or -NOz; -N 3 -CN;
.OR
21
-OCOR
21
-COR
21 -NCORa 2
-N(R
21 2
-CON(R
21 2 or
-COOR
21 aryl or heteroaryl; or two R 20 groups present on adjacent carbon atoms can join together to form a methylenedioxy group; wherein each R 21 is independently straight chained or branched C 1
-C
7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3
-C
7 cycloalkyl, Cs-C 7 cycloalkenyl or aryl; wherein each R 22 is independently H, F, Cl, or straight chained or branched Ci-C 4 alkyl; wherein q is an integer from 2 to 4 inclusive; wherein each m is an integer from 0 to 4 inclusive; wherein each n is an integer from 1 to 4 inclusive; wherein each p is an integer from 0 to 2 inclusive; 00 281 F wherein U is O, -NR 16 S, C(R 17 2 or -NS0 2
R
16 tn wherein Z is C 3 -Co 0 cycloalkyl, C 4
-C
7 cyclic ether,
C
4
-C
7 cyclic thioether, aryl, or heteroaryl; D 0 0 wherein R 16 is straight chained or branched Ci-C7 0 alkyl, straight chained or branched Ci-C7 C( monofluoroalkyl, straight chained or branched C 1
-C
7 00 Spolyfluoroalkyl, straight chained or branched C 2
-C
7 C 10 alkenyl, straight chained or branched C 2
-C
7 alkynyl, Cs-C 7 cycloalkenyl,
-(CH
2 or (CH2)q-O-(CH 2 )m-CH 3 wherein B is aryl, or heteroaryl; provided however, if B is aryl or heteroaryl the carbon atom or carbon atoms ortho to the nitrogen atom of the imine bond may only be substituted with one or more of the following -Cl, -Br, -CN, methyl, ethyl or methoxy; or a pharmaceutically acceptable salt thereof.
As used in the present invention, the term "cycloalkyl" includes
C
3
-C
7 cycloalkyl moieties which may be substituted with one or more of the following:
-NO
2 -CN, straight chained or branched Ci-C 7 alkyl, straight chained or branched
CI-C
7 monofluoroalkyl, straight chained or branched
C
1
-C
7 polyfluoroalkyl, straight chained or branched
C
2 -C7 alkenyl, straight chained or branched
C
2
-C
7 alkynyl,
C
3
-C
7 cycloalkyl,
C
3
-C
7 monofluorocycloalkyl,
C
3 -C7 polyfluorocycloalkyl, Cs-C 7 cycloalkenyl,
N(R
4 2, -OR4, -COR 4
-NCOR
4
CO
2
R
4
-CON(R
4 2 or 00 S282 (CH2)n-O- (CH 2 )m-CH 3 SAs used in the present invention, the term "cycloalkenyl" includes
C
5 cycloalkenyl moieties which may be substituted with one or more of the 00 straight 00 following: -Cl, -Br, -NO 2 -CN, straight Schained or branched C 1 -C7 alkyl, straight chained or C< branched C 1 -C7 monofluoroalkyl, straight chained or branched C 1 -C7 polyfluoroalkyl, straight chained or branched
C
2 -C7 alkenyl, straight chained or branched C2-C7 alkynyl,
C
3 cycloalkyl, C3-C, monofluorocycloalkyl,
C
3 polyfluorocycloalkyl, Cs- C7 cycloalkenyl,
-N(R
4 2
-OR
4
-COR
4
-NCOR
4 C0 2
R
4
-CON(R
4 2 or (CH 2 )n-0-(CH 2 ),-CH3.
In the present invention, the term "heteroaryl" is used to include five and six membered unsaturated rings that may contain one or more oxygen, sulfur, or nitrogen atoms. Examples of heteroaryl groups include, but are not limited to, furanyl, thienyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, and triazinyl.
In addition the term "heteroaryl" is used to include fused bicyclic ring systems that may contain one or more heteroatoms such as oxygen, sulfur and nitrogen. Examples of such heteroaryl groups include, but are not limited to, indolizinyl, indolyl, isoindolyl, benzo[b]furanyl, benzo[b]thiophenyl, indazolyl, benzimidazolyl, 00 283 purinyl, benzoxazolyl, benzisoxazolyl, benzo b]thiazolyl, imidazo[2,1-b]thiazolyl, cinnolinyl, quinazolinyl, quinoxalinyl, 1,8naphthyridinyl, pteridinyl, quinolinyl, isoquinolinyl, phthalimidyl and 2,1,3- 00 M benzothiazolyl.
C The term "heteroaryl" also includes those chemical 00 moieties recited above which may be substituted with CI 10 one or more of the following: -Cl, -Br,
NO
2 -CN, straight chained or branched C 1
-C
7 alkyl, straight chained or branched C 1 monofluoroalkyl, straight chained or branched C 1 polyfluoroalkyl, straight chained or branched C 2
-C
7 alkenyl, straight chained or branched C 2
-C
7 alkynyl, C 3
-C
7 cycloalkyl,
C
3
C
7 monofluorocycloalkyl,
C
3
-C,
polyfluorocycloalkyl,
C
5 cycloalkenyl,
-N(R
4 2
OR
4
-COR
4 -NCOR4, -C0 2
R
4
-CON(R
4 2 Or (CH 2 )n-O-
(CH
2 )m-CH 3 The term "heteroaryl" further includes the N-oxides of those chemical moieties recited above which include at least one nitrogen atom.
In the present invention the term "aryl" is phenyl or naphthyl. The term "aryl" also includes phenyl and naphthyl which may be substituted with one or more of the following: -Cl, -Br, -NO 2
-CN,
straight chained or branched C 1 alkyl, straight chained or branched C 1
-C
7 monofluoroalkyl, straight chained or branched C 1
-C
7 polyfluoroalkyl, straight chained or branched C 2 alkenyl, straight chained 00 S284 Sor branched C 2
-C
7 alkynyl, C 3 -C7 cycloalkyl, C 3
-C
7 c- monofluorocycloalkyl, C 3
-C
7 polyfluorocycloalkyl, C s C 7 cycloalkenyl, -N(R 4 2
-OR
4 -SR4, -OCOR 4
-COR
4 NCOR4, -C0 2
R
4 -CON(R4) 2 or (CH 2
(CH
2 )m-CH 3 00 The present invention also provides a method of treating a subject suffering from anxiety which compromises administering to the subject an amount 0 0 of compound effective to treat the subject's anxiety where in the compound has the structure:
R
24 N R24 0
R
2 wherein each R 24 is independently one or more of the following: H, F, Cl, Br, I, CF 3
OCH
3 or NO 2 wherein R2s is methyl, ethyl, allyl, phenyl and the phenyl is optionally substituted with a F, Cl, Br,
CF
3
NO
2 In one embodiment of any of the methods described herein, the compound is enantiomerically and diastereomerically pure. In one embodiment of any of the methods described herein, the compound is enantiomerically or 285 diastereomerically pure.
In one embodiment of any of the methods described herein, the compound is a pure Z imine isomer or a pure Z alkene isomer. In one embodiment, the compound is a pure E imine isomer or a pure E alkene isomer.
In one embodiment, the compound has the structure: wherein each of Y 1
Y
2
Y
3 and Y 4 is independently H; straight chained or branched C 1 alkyl, -CF 3 F, -C1, -Br, -OR 4
-N(R
4 2 or -CON(R 4 2 wherein each R 4 is independently straight chained or branched Ci-C 7 alkyl, -CF 3 or phenyl; wherein A is straight chained or branched CI-C 7 alkyl, aryl, heteroaryl, aryl(C 1
-C
6 )alkyl or heteroaryl (Ci-Cs) alkyl; and wherein A' is 00 286
R
1 n- CR 2
R
3 00 M In one embodiment, B is heteroaryl. In one embodiment, B is aryl.
00 In one embodiment, B is phenyl and the phenyl is optionally substituted with one or more of the following: -Cl, -Br, -CF 3 straight chained or branched Cl-C, alkyl, -N(R 4 2
-OR
4 -CaR 4 -NCOR,, -C0 2 or -CON(R 4 2 in one embodiment, A is aryl. In one embodiment, A is heteroaryl.
In one embodiment, the compound is selected from the group consisting of: F C1 -C1 0 F 0~I9= C N Nand Qr/o 287 In one embodiment, the compound is selected from the group consisting of: 0 Cl
F
N
0 b-ss\ v^
C^^
2008200380 25 Jan 2008 m 289 In one embodiment, A is A' and A' is fl CR 2
R
3 In one embodiment, the compound is: ;or NM Cl Cl 00 S290 SIn one embodiment, B is Q6.
In one embodiment, A is aryl.
00 SIn one embodiment, the compound has the structure: 00 R22 ,oJR 22 In one embodiment, the compound is:
F
/F
In one embodiment, B is aryl.
291 In one embodiment, A is (CHR 7
(CHR
1 7 )n-Z.
In one embodiment, the compound is: C1 -C1 Nr
N
The invention provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound having the structure: wherein each of Y 1
Y
2
Y
3 and Y 4 is independently H; straight chained or branched
C
1
-C
7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained 00 292 or' branched C 2 -C-7 alkenyl or alkynyl; C 3
-C
7 cycloalkyl, or C 5
-C
7 cycloalkenyl; -C1, -Br, or 1; -NO 2
-N
3 -CN; -OR 4
-SR
4
-OCOR
4 -COR4, -NCOR 4 N(R4)2 -CON(R), or -COOR 4 aryl or heteroaryl, or 0 5 any two of Y 1
Y
2
Y
3 and Y 4 present on adjacent 00 M carbon atoms can constitute a methylenedioxy group; 00 wherein each R 4 is independently straight chained or branched
C
1 -C7 alkyl, monofluoroalkyl or CIto polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl;
C
3
-C
7 cycloalkyl,
C
5
-C
7 cycloalkenyl, aryl or aryl (C 1
C
6 alkyl; wherein A is A' Q3, Q 4 Q5, straight chained or branched Cl-C7 alkyl, aryl, heteroaryl, aryl(Cl-
C
6 )alkyl, heteroaryl, (Cl-Cc 6 alkyl, aryl substituted with an aryl or heteroaryl, heteroaryl substituted with an aryl or heteroaryl; or (CHRr,) (CHR 17 n-Z7; wherein A' is 0 CR 2R3or -(CH 2 4 00 293 wherein Q3 is R1 R17 00 _1 (R17 00 wherein Q 4 is
R
17 R1 N n
R
17 m wherein Q5 is 00 D 294 c' wherein Ri and R 2 are each independently H, straight Schained or branched Ci-C7 alkyl, -Cl, -Br,
NO
2 or -CN; 00 00 wherein R 3 is H, straight chained or branched C 1 -C7 o alkyl, -Cl, -Br, -NO 2 -CN, -OR 6 aryl or C heteroaryl; 0 10 wherein R 5 is straight chained or branched Ci-C7 alkyl, -N(R4)2, -OR 6 or aryl; wherein R 6 is straight chained or branched C 1 -C7 alkyl or aryl; wherein each R17 is independently H; straight chained or branched C 1 -C7 alkyl, straight chained or branched Ci-C, monofluoroalkyl, straight chained or branched CI-C7 polyfluoroalkyl, straight chained or branched
C
2 -C7 alkenyl, straight chained or branched C 2 -C7 alkynyl, C 5 -C7 cycloalkenyl, -(CH 2 or (CH 2 )n-O-
(CH
2 -CH3; wherein each R 2 0 is independently straight chained or branched Ci-C7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or alkynyl; C 3 cycloalkyl or C 5
-C,
cycloalkenyl; -C1, -Br, or -NO 2
-N
3 -CN;
OR
21 -0COR 21
COR
21
-NCOR
21
-N(R
21 2
-CON(R
21 2 or
-COOR
21 aryl or heteroaryl; or two R 20 groups present on adjacent carbon atoms can join together to form a methylenedioxy group; 00 0 295 -s wherein each R 21 is independently straight n chained or branched C 1
-C
7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or alkynyl; C 3 -C7 cycloalkyl, Cs-C, 0- cycloalkenyl, aryl or aryl(C 1 -Cs)alkyl; C-I wherein each m is an integer from 0 to 4 inclusive; 00 cq 10 wherein each n is an integer from 1 to 4 inclusive; wherein each p is an integer from 0 to 2 inclusive; wherein U is 0, -NR 16 S, C(R 17 2 or -NS0 2
R
16 wherein Z is C 3 -Clo cycloalkyl, C 4 -C7 cyclic ether,
C
4 -C7 cyclic thioether, aryl, or heteroaryl; wherein R 16 is straight chained or branched C 1 -C7 alkyl, straight chained or branched C 1 -C7 monofluoroalkyl, straight chained or branched C 1 -C7 polyfluoroalkyl, straight chained or branched C 2 -C7 alkenyl, straight chained or branched C 2 -C7 alkynyl, Cs-C7 cycloalkenyl, -(CH 2 or (CH 2
(CH
2 )m-CH 3 wherein q is an integer from 2 to 4 inclusive; wherein B is aryl, heteroaryl, aryl substituted with an aryl or heteroaryl, heteroaryl substituted with an aryl or heteroaryl, tricyclic heteroaryl or Qs; provided however, if B is aryl or heteroaryl the carbon atom or carbon atoms ortho to the nitrogen 00 296 atom of the imine bond may only be substituted with one or more of the following -Cl, -Br, -CN, methyl, ethyl or methoxy; wherein a tricyclic heteroaryl is a fused three 00 member aromatic system in which one or more of the rings is heteroaryl; carbazole; or acridine; 00 wherein Q6 is C0 R22 xn* 0 R22 wherein each R 22 is independently H, F, Cl, or straight chained or branched C 1
-C
4 alkyl; or a pharmaceutically acceptable salt thereof.
The invention provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound having the structure:
Y
N
Y2 Y3 00 297 wherein each of Y 1
Y
2
Y
3 and Y 4 is independently H; straight chained or branched C C, alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3
-C
7 cycloalkyl, or C 5
-C
7 cycloalkenyl; -C1, -Br, or 00 1; -NO 2
-N
3 -CN; -OR 4 -SR4, -OCOR4, -COR4, -NCOR4, N(R42 ,-CON(R 4 2 or -COOR 4 aryl or heteroaryl; or 00 any two of Y 1
Y
2
Y
3 and Y 4 present on adjacent carbon atoms can constitute a methylenedioxy group; wherein each R 4 is independently -H4; straight chained or branched Cl-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C-7 alkenyl or alkynyl;
C
3 -C7 cycloalkyl,
C
5
-C
7 cycloalkelyl, aryl or aryl(Cl-C 6 )alkyl; wherein A is straight chained or branched Cl-C 7 alkyl, aryl, heteroaryl, aryl (Cl-C 6 alkyl or heteroaryl (Cl-C 6 alkyl; wherein A' is 0 0 n I
R
5 II n R2 or -(CH 2 n
R
4 00 S298 wherein R 1 and R 2 are each independently H, straight chained or branched C 1
-C
7 alkyl, -Cl, -Br, 00 0) NO 2 or -CN; wherein R 3 is H, straight chained or branched C 1
-C
7 00 alkyl, -Cl, -Br, -NO 2 -CN, -OR 6 aryl or CI 10 heteroaryl; wherein R 5 is straight chained or branched C 1
-C
7 alkyl, -N(R 4 2 -OR6 or aryl; wherein R 6 is straight chained or branched C 1
-C
7 alkyl or aryl; wherein B is aryl, or heteroaryl; provided however, if B is aryl or heteroaryl the carbon atom or carbon atoms ortho to the nitrogen atom of the imine bond may only be substituted with one or more of the following -Cl, -Br, -CN, methyl, ethyl or methoxy; wherein n is an integer from 1 to 4 inclusive; or a pharmaceutically acceptable salt thereof.
The invention provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound having the structure: 00 299 200 00 wherein each of Y 1
Y
2
Y
3 1' and Y 4 is independently- H; straight chained or branched Cl-C 7 alkyl, monofluoroalkyl or polyfluoroalky.; straight chained or branched C 2 -C-7 alkenyl or alkynyl; C 3
-C
7 1 cycloalkyl, or CS-C7 cycloalkenyl; -C1, -Br, or 1; -N02; -N 3 -CN; -OR.4, -SR 4
-OCOR
4
-COR
4
-NCOR
4
N(R
4 2
-CON(R
4 2 or -COOR 4 aryl or heteroaryl; or any two of Y 1
Y
2
Y
3 and Y 4 present on adjacent carbon atoms can constitute a methylenedioxy group; wherein each R 4 is independently straight chained or branched Cl-C7 alkyl, monofluoroalkyl or polyf luoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3
-C
7 cycloalkyl, C 5
-C
7 cycloalkenyl, aryl or aryl, (Cl-C 6 alkyl; wherein A is A' straight chained or branched Cl-C 7 alkyl, aryl, heteroaryl, aryl 6 alkyl or heteroaryl (C-COalkyl; wherein A' is 300 0 R RI or
(CH
2 n
R
4 wherein B is aryl substituted with an aryl or heteroaryl, heteroaryl substituted with an aryl or heteroaryl, tricyclic heteroaryl or Q6; wherein a tricyclic heteroaryl is a fused three ring aromatic system in which one or more of the rings is heteroaryl; carbazole; or acridine; wherein Q0 is wherein n is an integer from 1 to 4 inclusive; wherein each R 22 is independently H, Cl, or straight chained or branched Ci-C 4 alkyl; 00 S301 00 -i or a pharmaceutically acceptable salt thereof.
The invention provides a pharmaceutical composition 0 comprising a pharmaceutically acceptable carrier and a M compound having the structure: 3 Y2
A
Y4 wherein each of Y 1
Y
2
Y
3 and Y 4 is independently H; straight chained or branched Ci-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or alkynyl; C 3
-C
7 cycloalkyl, or Cs-C 7 cycloalkenyl; -C1, -Br, or I; -NO 2
-N
3 -CN; -OR 4 -SR4, -OCOR 4 -COR4, -NCOR 4
N(R
4
-CON(R
4 2 or -COOR4; aryl or heteroaryl; or any two of Y 1
Y
2
Y
3 and Y 4 present on adjacent carbon atoms can constitute a methylenedioxy group wherein each R 4 is independently straight chained or branched C 1
-C
7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3
-C
7 cycloalkyl, Cs-C 7 cycloalkenyl, aryl or aryl(Ci-C 6 )alkyl; 302 wherein A is Q3, Q 4 Q5, aryl. substituted with an aryl or heteroaryl, heteroaryl substituted with an aryl or heteroaryl, or (CHR 17
)-(CHR
17 )n-Z; wherein Q3 is wherein Q4 is wherein Q5 is 00 S303 wherein each Rn 1 is independently H; straight chained ^c or branched C 1 alkyl, straight chained or branched
C
1
-C
7 monofluoroalkyl, straight chained or branched
C
1 -C polyfluoroalkyl, straight chained or branched 00 M
C
2
-C
7 alkenyl, straight chained or branched C 2
-C
7 alkynyl, C 5
-C
7 cycloalkenyl,
-(CH
2 or (CH 2 )n-O- C
(CH
2
),-CH
3 00 C- 10 wherein each R 20 is independently straight chained or branched CI-C 7 alkyl, .monofluoroalkyl or polyfluoroalkyl; straight chained or branched
C
2
-C
7 alkenyl or alkynyl;
C
3
-C
7 cycloalkyl or Cs-C 7 cycloalkenyl; -Cl, -Br, or -NO 2
-N
3 -CN;
OR
21
-OCOR
2 1, -COR 21
-NCOR
21
-N(R
21 2
-CON(R
21 2 or
-COOR
21 aryl or heteroaryl; or two R 20 groups present on adjacent carbon atoms can join together to form a methylenedioxy group; wherein each R 21 is independently straight chained or branched Ci-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched
C
2 -C7 alkenyl or alkynyl;
C
3 cycloalkyl, Cs-C 7 cycloalkenyl or aryl; wherein each R 22 is independently H, F, Cl, or straight chained or branched Ci-C 4 alkyl; wherein q is an integer from 2 to 4 inclusive; wherein each m is an integer from 0 to 4 inclusive; 304 Swherein each n is an integer from 1 to 4 inclusive;
V)
n wherein each p is an integer from 0 to 2 inclusive; wherein U is O, -NR 16 S, C(R 17 2 or -NSO 2
R
16 00 0 wherein Z is C 3 -Cio cycloalkyl, C 4 -C7 cyclic ether,
C
0 C4-C 7 cyclic thioether, aryl, or heteroaryl; 00 CI 10 wherein R 16 is straight chained or branched Ci-C 7 alkyl, straight chained or- branched CI-C 7 monofluoroalkyl, straight chained or branched C 1 -C7 polyfluoroalkyl, straight chained or branched C 2
-C
7 alkenyl, straight chained or branched C 2
-C
7 alkynyl, Cs-C 7 cycloalkenyl, -(CH 2 or (CH 2 )q-0-(CH 2 )m-CH 3 wherein B is aryl, or heteroaryl; provided however, if B is aryl or heteroaryl the carbon atom or carbon atoms ortho to the nitrogen atom of the imine bond may only be substituted with one or more of the following -Cl, -Br, -CN, methyl, ethyl or methoxy; or a pharmaceutically acceptable salt thereof.
As used in the present invention, the term "cycloalkyl" includes C 3 -C7 cycloalkyl moieties which may be substituted with one or more of the following:
-NO
2 -CN, straight chained or branched C 1
-C
7 alkyl, straight chained or branched Ci-C7 monofluoroalkyl, straight chained or branched Ci-C7 polyfluoroalkyl, straight chained or branched 00 305
C
2
-C
7 alkenyl, straight chained or branched C 2 -C7 alkynyl, C 3
-C
7 cycloalkyl, C 3 monofluorocycloalkyl,
C
3
-C
7 polyfluorocycloalkyl,
C
5
-C
7 cycloalkenyl, N (R 4 2, -OR 4
-COR
4
-NCOR
4
-CO
2
R
4
-CON(R
4 2 or
(CH
2
(CH
2 m-CH 3 00
C
As used in the present invention, the term C "cycloalkenyl" includes C 5
-C
7 cycloalkenyl moieties 00 which may be substituted with one or more of the CI 10 following: -Cl, -Br, -NOz 2 -CN, straight chained or branched C 1 alkyl, straight chained or branched C 1
-C
7 monofluoroalkyl, straight chained or branched Cl-C 7 polyfluoroalkyl, straight chained or branched C 2 alkenyl, straight chained or branched
C
2
-C
7 alkynyl, C 3
-C
7 cycloalkyl, C3-C7 monofluorocycloalkyl,
C
3
-C
7 polyfluorocycloalkyl,
C
5
C
7 cycloalkenyl,
-N(R
4 2 -OR4, -COR4, -NCOR 4 C0 2
R
4
-CON(R
4 2 or (CH 2 )n-O-(CH 2 )mCH 3 In the present invention, the term "heteroaryl" is used to include five and six membered unsaturated rings that may contain one or more oxygen, sulfur, or nitrogen atoms. Examples of heteroaryl groups include, but are not limited to, furanyl, thienyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, and triazinyl.
In addition the term "heteroaryl" is used to include fused bicyclic ring systems that may contain one or more heteroatoms such as oxygen, sulfur and 0 306 nitrogen. Examples of such heteroaryl groups c- include, but are not limited to, indolizinyl, indolyl, isoindolyl, benzo[b]furanyl, benzo[b]thiophenyl, indazolyl, benzimidazolyl, purinyl, benzoxazolyl, benzisoxazolyl, 0 benzo[b]thiazolyl, imidazo[2,1-b]thiazolyl, cinnolinyl, quinazolinyl, quinoxalinyl, 1,8- C naphthyridinyl, pteridinyl, quinolinyl, 00 isoquinolinyl, phthalimidyl and 2,1,3- CA 10 benzothiazolyl.
The term "heteroaryl" also includes those chemical moieties recited above which may be substituted with one or more of the following: -Cl, -Br,
NO
2 -CN, straight chained or branched Ci-C 7 alkyl, straight chained or branched CI-C7 monofluoroalkyl, straight chained or branched C 1 polyfluoroalkyl, straight chained or branched C 2
-C
7 alkenyl, straight chained or branched C 2
-C
7 alkynyl, C 3
-C
7 cycloalkyl,
C
3
-C
7 monofluorocycloalkyl, C3-C7 polyfluorocycloalkyl, Cs-C7 cycloalkenyl, -N(R 4 2
OR
4
-COR
4
-NCOR
4
-CO
2 R4, -CON(R4) 2 or (CH 2 )n-O-
(CH
2
-CH
3 The term "heteroaryl" further includes the N-oxides of those chemical moieties recited above which include at least one nitrogen atom.
In the present invention the term "aryl" is phenyl or naphthyl. The term "aryl" also includes phenyl and naphthyl which may be substituted with one or more of the following: -Cl, -Br, -NO 2
-CN,
00 307
(N
Sstraight chained or branched Ci-C 7 alkyl, straight chained or branched Ci-C 7 monofluoroalkyl, straight n chained or branched C 1
-C
7 polyfluoroalkyl, straight chained or branched C 2
-C
7 alkenyl, straight chained or branched C 2 -C7 alkynyl, C 3 -C7 cycloalkyl, C 3 -C7 0 0 monofluorocycloalkyl, C 3
-C
7 polyfluorocycloalkyl, C s 0
C
7 cycloalkenyl, -N(R 4 2
-OR
4
-SR
4
-OCOR
4
-COR
4 C0 NCOR 4 -C0 2
R
4
-CON(R
4 2 or (CH 2 )n-O-(CH 2 )m-CH 3 00 In one embodiment of any of the pharmaceutical compositions described herein, the compound is enantiomerically and diastereomerically pure. In one embodiment, the compound is enantiomerically or diastereomerically pure.
In one embodiment, the compound is a pure Z imine isomer or a pure Z alkene isomer.
In one embodiment, the compound is a pure E imine isomer or a pure E alkene isomer.
In one embodiment, the composition can be administered orally.
In one embodiment of the pharmaceutical composition, the compound has the structure: 00 0 308 0 00 wherein each of Y 1
Y
2 1 Y 3 and Y 4 is independently- H; straight chained or branched Cl-C 7 alkyl, -CF 3 F, -Cl, -Br, -OR4, -N(R 4 2 or -CON(R 4 2 wherein each R 4 is independently straight chained or branched Cl-C-7 alkyl, -CF 3 or phenyl; wherein A is straight chained or branched Cl-C 7 alkyl, aryl, heteroaryl, aryl (Cl-C 6 alkyl or heteroaryl (Cl.-C 6 alkyl; and wherein A' is nl CR 2
R
3 In one embodiment, B is heteroaryl.
In one embodiment, B'is aryl.
In one embodiment, B is phenyl and the phenyl is 00 S309 Soptionally substituted with one or more of the following: c- -Cl, -Br, -CF 3 straight chained or branched C 1 -C7 Salkyl, -N(R 4 2
-OR
4
-COR
4 -NCOR4, -C0 2 R4, or -CON(R 4 2 In one embodiment, A is aryl. In one embodiment, A is 00 0 heteroaryl.
00 (N In one embodiment, the compound is selected from the group consisting of: F C3
CI
and rv 0 310 In one embodiment, B is Q6.
In one embodiment, A is aryl.
In one embodiment, the compound has the structure: In one embodiment, the compound is: 00 311.
N /0\ 00 00 In one embodiment, B is aryl.
In one embodiment, A is (CHR 11 7) (C1IR 17 2- Z In one embodiment, the compound is:
CI
CI
0C'So0
CN/
The invention provides a compound having the structure: 00 D 312 n wherein each of Y 1
Y
2
Y
3 and Y 4 is independently H; straight chained or branched Ci-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained Sor branched C 2
-C
7 alkenyl or alkynyl; C 3 -C7 Scycloalkyl, or Cs-C, cycloalkenyl; -Cl, -Br, or C I; -NO 2
-N
3 -CN; -OR 4
-SR
4
-OCOR
4 -COR4, -NCOR 4 00 SN(R 4 2
-CON(R
4 2 or -COOR 4 aryl or heteroaryl; or C 10 any two of Y 1
Y
2
Y
3 and Y 4 present on adjacent carbon atoms can constitute a methylenedioxy group; wherein each R 4 is independently straight chained or branched
C
1
-C
7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched
C
2 -C7 alkenyl or alkynyl; C 3
-C
7 cycloalkyl,
C
5
-C
7 cycloalkenyl, aryl or aryl(Ci-Cs)alkyl; wherein A is Q3, Q4, Qs, straight chained or branched
C
1
-C
7 alkyl, aryl, heteroaryl, aryl (C 1 Cs) alkyl, heteroaryl
(C
1
-C
6 alkyl, aryl substituted with an aryl or heteroaryl, heteroaryl substituted with an aryl or heteroaryl; or (CHR 1 (CHRi 7 )n-Z; wherein A' is 0 Rn n CR 2
R
3 or or (CH 2 n
R
wherein Q.3 is RV R1 N 1
R
17 wherein Q 4 is 00 S314 00 wherein Qs is 710 h O0 0 wherein R, and R2 are each independently H, straight chained or branched Cl-C7 alkyl, -Cl, -Br, NO2, or -CN; wherein R3 is H, straight chained or branched CI-C 7 alkyl, -Cl, -Br, -NO2, -CN, -ORe, aryl or heteroaryl; wherein R 1 is straight chained or branched Cl-C7 alkyl, -OR6 or aryl; wherein R6 is straight chained or branched Cz-C7 alkyl or aryl; wherein each R17 is independently H; straight chained or branched Ci-C7 alkyl, straight chained or branched Cl-C 7 monofluoroalkyl, straight chained or branched
C
1 -C7 polyfluoroalkyl, straight chained or branched
C
2
-C
7 alkenyl, straight chained or branched C 2
-C
7 alkynyl, C 5
-C
7 cycloalkenyl, -(CH 2 or (CH 2 )n-0-
(CH
2 m-CH 3 wherein each R 20 is independently straight chained or branched C 1
-C
7 alkyl, monofluoroalkyl or.
00 315 Spolyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or alkynyl; C 3 -C7 cycloalkyl or Cs-C7 l n cycloalkenyl; -C1, -Br, or -NO 2
-N
3 -CN;
OR
2 1
-OCOR
21
-COR
2 1
-NCOR
2 1
-N(R
2 1 2
-CON(R
2 1 2 or
-COOR
2 1; aryl or heteroaryl; or two R 20 groups present 0 on adjacent carbon atoms can join together to form a Smethylenedioxy group; 00 Swherein each R 21 is independently straight Ci 10 chained or branched C 1 -C7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or alkynyl; C 3 cycloalkyl, Cs-C7 cycloalkenyl, aryl or aryl(Ci-Cs)alkyl; wherein each m is an integer from 0 to 4 inclusive; wherein each n is an integer from 1 to 4 inclusive; wherein each p is an integer from 0 to 2 inclusive; wherein U is O, -NR 16 S, C(R 17 2 or -NS0 2
R
16 wherein Z is C 3 -Cio cycloalkyl, C 4 -C7 cyclic ether,
C
4 -C7 cyclic thioether, aryl, or heteroaryl; wherein R 16 is straight chained or- branched Ci-C7 alkyl, straight chained or branched Ci-C7 monofluoroalkyl, straight chained or branched Ci-C7 polyfluoroalkyl, straight chained or branched C 2 -C7 alkenyl, straight chained or branched C 2 -C7 alkynyl, Cs-C7 cycloalkenyl, -(CH 2 or (CH 2 (CH2)m-CH3; 00 0 316
(N
Swherein q is an integer from 2 to 4 inclusive; r wherein B is aryl, heteroaryl, aryl substituted with an aryl or heteroaryl, heteroaryl substituted with an aryl or heteroaryl, tricyclic heteroaryl or Q6; 00 provided however, if B is aryl or heteroaryl the carbon atom or carbon atoms ortho to the nitrogen C( atom of the imine bond may only be substituted with 00 Sone or more of the following -Cl, -Br, -CN, C1 10 methyl, ethyl or methoxy; wherein a tricyclic heteroaryl is a fused three member aromatic system in which one or more of the rings is heteroaryl; carbazole; or acridine; wherein Q6 is 0 R22 0 R22 wherein each R 22 is independently H, F, Cl, or straight chained or branched CI-C 4 alkyl; or a pharmaceutically acceptable salt thereof.
The invention provides a compound having the structure: 00 317 wherein each of YI, Y 2
Y
3 and Y 4 is independentl.y H; straight chained or branched C 1
-C
7 alkyl, monofluoroalkyl. or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl or alkynyl; C 3
-C
7 00 M cycloalkyl, Or C 5
-C
7 cycloalkenyl; -C1, -Br, or 1; -NO 2
-N
3 -CN; -OR 4
-SR
4 -OCOR4, -COR 4 -NCOR4, 00 NCR 4 2
-CON(R
4 2 or -COOR 4 aryl or heteroaryl; or any two of Y 1
Y
2
Y
3 and Y 4 present on adjacent carbon atoms can constitute a methylenedioxy group; wherein each R 4 is independently straight chained or branched Cl-C7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2
-C
7 alkenyl. or alkynyl; C 3
-C
7 cycloalkyl,
C
5
-C
7 cycloalkenyl, aryl or aryl(Cl-C 6 )alkyl; wherein A is straight chained or branched Cl-C 7 alkyl, aryl, heteroaryl, aryl (C C 6 alkyl or heteroaryl (Cl-C 6 alkyl; wherein A' is 0 0n Un n C23or -(CH 2 R4 00 318 00 M ,NO 2 or -CN; 00 wherein R 3 is H, straight chained or branched CI-C 7 00 10 heteroaryl; wherein R and R2 are eachindependently H, straight 0 5 chained or branched Ci-C7 alkyl, -Cl, -Br, alkyl, or -OR or aryl;-CN; wherein R 6 is straight chained or branched C 1
-C
7 0alkyl, -Cl, -Br, -NO2, -CN, -ORe aryl or CN 10 heteroaryl; wherein Rs is straight chained or branched C1-07 alkyl, -N(R4)2, -ORe or aryl; wherein Re is straight chained or branched Ci-C7 alkyl or aryl; wherein B is aryl, or heteroaryl; provided however, if B is aryl or heteroaryl the carbon atom or carbon atoms ortho to the nitrogen atom of the imine bond may only be substituted with one or more of the following -Cl, -Br, -CN, methyl, ethyl or methoxy; wherein n is an integer from 1 to 4 inclusive; or a pharmaceutically acceptable salt thereof.
00 319 The invention provides a compound having the structure: 000 00
N
wherein each of Y 1
Y
2
Y
3 and Y 4 is independently- H; straight chained or branched CI-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or alkynyl; C 3
-C
7 cycloalkyl, or CS-C 7 cycloalkenyl; -C1, -Br, or 1; -NO 2
-N
3 -CN; -OR 4
-SR
4
-OCOR
4
-COR
4
-NCOR
4
N(R
4 2
-CON(R
4 2 or -COOR 4 aryl or heteroaryl; or any two of Y 1
Y
2
Y
3 and Y 4 present on adjacent carbon atoms can constitute a methylenedioxy group; wherein each R 4 is independently -Hi straight chained or branched Cl-C, alkyl, ronofluoroalkyl or polyf luoroalkyl; straight chained or branched C 2
-C,
alkenyl or alkynyl; C 3 -C7 cycloalkyl, CS--C, cycloalkenyl, aryl or aryl (CI-C 6 alkyl; wherein A is A' straight *chained or branched Cl-C, alkyl, aryl, heteroaryl, aryl (C CO)al kyl or heteroaryl (Cl-C 6 alkyl; wherein A, is 320 0
CRR
3 CRzR3 n or (CH2)n R4 wherein B is aryl substituted with an aryl or heteroaryl, heteroaryl substituted with an aryl or heteroaryl, tricyclic heteroaryl or Q6; wherein a tricyclic heteroaryl is a fused three ring aromatic system in which one or more of the rings is heteroaryl; carbazole; or acridine; wherein Q6 is 0 R22
I/
wherein n is an integer from 1 to 4 inclusive; wherein each R 22 is independently H, F, 00 D 321 SCl, or straight chained or branched Ci-C 4 alkyl; c, n or a pharmaceutically acceptable salt thereof.
O The invention provides a compound having the structure: 00 00 B y3 I
N
Y3N
A
Y4 wherein each of Y 1
Y
2
Y
3 and Y 4 is independently H; straight chained or branched CI-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 C7 alkenyl or alkynyl; C 3 -C7 cycloalkyl, or Cs-C7 cycloalkenyl; -Cl, -Br, or I; -NO 2
-N
3 -CN; -OR 4
-SR
4 -OCOR4, -COR4, -NCOR 4
N(R
4 2
-CON(R
4 2 or -COOR4; aryl or heteroaryl; or any two of Y 1
Y
2
Y
3 and Y 4 present on adjacent carbon atoms can constitute a methylenedioxy group; wherein each R 4 is independently straight chained or branched C 1 -C7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or alkynyl; C 3 -C7 cycloalkyl, Cs-C7 cycloalkenyl, aryl or aryl(Ci-C 6 )alkyl; wherein A is Q3, Q4, Qs, aryl substituted with an 322 aryl or heteroaryl, heteroaryl substituted with an aryl or heteroaryl, or (CHR 17
)-(CHR
17 )n-Z; wherein Q3 is wherein 04 is wherein Q5 is 0 323 wherein each R17 is independently H; straight chained c' or branched C 1 alkyl, straight chained or branched SCi-C7 monofluoroalkyl, straight chained or branched Cl-C7 polyfluoroalkyl, straight chained or branched
C
2 -C7 alkenyl, straight chained or branched Cz-C7 00 M alkynyl, Cs-C 7 cycloalkenyl,
-(CH
2 or (CH 2 )n-O-
(CH
2 )m-CH3; 00 0 wherein each R 20 is independently straight N 10 chained or branched Ci-C7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched
C
2 -C7 alkenyl or alkynyl; C 3 -C7 cycloalkyl or C 5 -C7 cycloalkenyl; -C1, -Br, or -NO 2
-N
3 -CN;
OR
21
-OCOR
21
-COR
21
-NCOR
21
-N(R
2 1 2
-CON(R
21 2 or
-COOR
21 aryl or heteroaryl; or two R 20 groups present on adjacent carbon atoms can join together to form a methylenedioxy group; wherein each R 21 is independently straight chained or branched CI-C7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched
C
2 -C7 alkenyl or alkynyl;
C
3
-C
7 cycloalkyl,
C
5
-C
7 cycloalkenyl or aryl; wherein each R 22 is independently H, F, Cl, or straight chained or branched Ci-C 4 alkyl; wherein q is an integer from 2 to 4 inclusive; wherein each m is an integer from 0 to 4 inclusive; wherein each n is an integer from 1 to 4 inclusive; S324 c- wherein each p is an integer from 0 to 2 inclusive; ci wherein U is O, -NR 16 S, C(R 17 2 or -NS0 2
R
1 6 00 M wherein Z is C 3 -Cio cycloalkyl, C 4
-C
7 cyclic ether,
C
4
-C
7 cyclic thioether, aryl, or heteroaryl; 00 Swherein R 16 is straight chained or branched C 1
-C
7 C1 10 alkyl, straight chained or branched Ci-C 7 monofluoroalkyl, straight chained or branched Ci-C7 polyfluoroalkyl, straight chained or branched C 2 -C7 alkenyl, straight chained or branched C 2 -C7 alkynyl,
C
5
-C
7 cycloalkenyl, -(CH 2 or (CH2)q-- (CH 2 )m-CH 3 wherein B is aryl, or heteroaryl; provided however, if B is aryl or heteroaryl the carbon atom or carbon atoms ortho to the nitrogen atom of the imine bond may only be substituted with one or more of the following -Cl, -Br, -CN, methyl, ethyl or methoxy; or a pharmaceutically acceptable salt thereof.
As used in the present invention, the term "cycloalkyl" includes C 3
-C
7 cycloalkyl moieties which may be substituted with one or more of the following:
-NO
2 -CN, straight chained or branched C 1
-C
7 alkyl, straight chained or branched Ci-C 7 monofluoroalkyl, straight chained or branched
C
1
-C
7 polyfluoroalkyl, straight chained or branched
C
2
-C
7 alkenyl, straight chained or branched C 2 -C7 00 325 alkynyl, C 3
-C
7 cycloalkyl, C 3
-C
7 monofluorocycloalkyl,
C
3
-C
7 polyfluorocycloalkyl, Cs-C7 cycloalkenyl, N (R 4 2, -OR 4 -COR4, -NCOR4, -COaR 4
-CON(R
4 2 or
(CH
2
(CH
2 )m-CH 3 5 00 neto t tr 00As used in the present invention, the term "cycloalkenyl" includes Cs-C 7 cycloalkenyl moieties C- which may be substituted with one or more of the 00 following: -Cl, -Br, -NO2, -CN, straight CI 10 chained or branched C 1
-C
7 alkyl, straight chained or branched C 1
-C
7 monofluoroalkyl, straight chained or branched C 1
-C
7 polyfluoroalkyl, straight chained or branched C 2 alkenyl, straight chained or branched C2-C C alkynyl, C 3
-C
7 cycloalkyl, C3-C7 monofluorocycloalkyl, C 3
-C
7 polyfluorocycloalkyl, Cs 5
C
7 cycloalkenyl, -N(R4) 2
-OR
4
-COR
4 -NCOR4, C0 2 R4, -CON(R4) 2 or (CH 2
(CH
2 )m-CH 3 In the present invention, the term "heteroaryl" is used to include five and six membered unsaturated rings that may contain one or more oxygen, sulfur, or nitrogen atoms. Examples of heteroaryl groups include, but are not limited to, furanyl, thienyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, and triazinyl.
In addition the term "heteroaryl" is used to include fused bicyclic ring systems that may contain one or more heteroatoms such as oxygen, sulfur and nitrogen. Examples of such heteroaryl groups 00 326 include, but are not limited to, indolizinyl, indolyl, isoindolyl, benz-otblfuranyl, V)benzolthiophenyl, indazolyl, benzimidazolyl, purinyl, benzoxazolyl, benzisoxazolyl, benzo~blthiazolyl, imidazo [2,1-b]thiazolyl, 00 cinnolinyl, quinazolinyl, quinoxalinyl, 1,8naphthyridinyl, pteridinyl, quinolinyl, 00isoquinolinyl, phthalimidyl and 2,1,3benzothiazolyl.
(Ni The term "heteroaryl" also includes those chemical moieties recited above which may be substituted with one or more of the following: -Cl, -Br, N0 2 -CN, straight chained or branched C 1
-C
7 alkyl, straight chained or branched Cl-C 7 monofluoroalkyl, straight chained or branched Cj-C7 polyfluoroalkyl, straight chained or branched C 2
-C
7 alkenyl, straight chained or branched C 2
-C
7 alkynyl, C 3
-C
7 cycloalkyl, C-7monofluorocycloalkyl,
C
3
-C
7 polyfluorocycloalkyl, CS-C 7 cycloalkenyl, -N(R 4 2
OR
4
-COR
4
NCOR
4 -C0 2
R
4
-CON(R
4 2 or (CH 2
(CH
2
-CH
3 The term "heteroaryl" further includes the N-oxides of those chemical moieties recited above which include at least one nitrogen atom.
In the present invention the term "aryl"' is phenyl or naphthyl. The term "aryl", also includes phenyl and naphthyl which may be substituted with one or more of the following: -Cl, -Br, -NO 2
-CN,
straight chained or branched Cl-C 7 alkyl, straight 00 S327 Schained or branched CI-C 7 monofluoroalkyl, straight c- chained or branched Ci-C7 polyfluoroalkyl, straight n chained or branched C 2
-C
7 alkenyl, straight chained or branched C 2 -C7 alkynyl, C 3 -C7 cycloalkyl, C 3 -C7 monofluorocycloalkyl, C 3
-C
7 polyfluorocycloalkyl, C s 00 C7 cycloalkenyl, -N(R 4 2
-OR
4
-SR
4
-OCOR
4
-COR
4
SNCOR
4 -CO2R 4
-CON(R
4 2 or (CH 2 2 )m-CH 3 00 o CI In one embodiment of any of the compounds described herein, the compound is -enantiomerically and diastereomerically pure. In one embodiment, the compound is enantiomerically or diastereomerically pure.
In one embodiment, the compound is a pure Z imine isomer or a pure Z alkene isomer.
In one embodiment, the compound is a pure E imine isomer or a pure E alkene isomer.
In one embodiment, the compound can be administered orally.
In one embodiment, the compound has the structure: 00 328 000 Y
A
00 wherein each of Y 1
Y
2
Y
3 and Y 4 is independently- H; straight chained or branched C 1
-C
7 alkyl, -CF 3 F, -C1, -Br, -OR 4
-N~(R
4 2 or -O(42 wherein each R 4 is independently straight chained or branched CI-C,7 alkyl, -CF 3 or phenyl; wherein A is A' straight chained or branched Cl-C 7 alkyl, aryl, heteroaryl, aryl (Cl-C 6 alkyl or heteroaryl (Cl-C 6 alkyl; and wherein A' is
R,
n1 CR 2
R
3 In one embodiment, B is heteroaryl.
In one embodiment, B is aryl.
In one embodiment, In oe emodimnt, B is phenyl. and the phenyl is 329 3 optionally substituted with one or more of the following: -Cl, -Br, -CF 3 straight chained or branched Cz-C, c alkyl, -N(R 4 2
-OR
4 -COR4, -NCOR4, -CO 2
R
4 or -CON(R 4 2 O 5 In one embodiment, A is aryl.
O In one embodiment, A is heteroaryl.
00 0In one embodiment, the compound is selected from the group consisting of: F
CI
SN and 0 1 N C In one embodiment, B is Q6.
In one embodiment, A is aryl.
330 In one embodiment, the compound has the structure: In one embodiment, the compound is: In one embodiment, B is aryl.
In one embodiment, A is (CHR 7
(CHR
7 )n-Z.
In one embodiment, the compound is: 00 331 C1
(N
00 8I
N
00 In one embodiment, the compound is a pure Z imine isomer.
In one embodiment, the compound is a pure E imine isomer.
The invention provides a pharmaceutical composition comprising a therapeutically effective amount of any of the compounds described herein and a pharmaceutically acceptable carrier.
The invention provides a pharmaceutical composition made by combining a therapeutically effective amount of any of the compounds described herein and a pharmaceutically acceptable carrier.
The invention provides a process for making a pharmaceutical composition comprising combining a therapeutically effective amount of any of the compounds described herein and a pharmaceutically acceptable carrier.
The invention provides a method of treating a subject suffering from depression which comprises administering to the subject an amount of any of the compounds described herein effective to treat the subject's 00 332 c depression.
The invention provides a method of treating a subject suffering from anxiety which comprises administering to 0 5 the subject an amount of any of the compounds described 00 M herein effective to treat the subject's anxiety.
00 The invention provides a method of treating a subject suffering from depression and anxiety which comprises administering to the subject an amount of any of the compounds described herein effective to treat the subject's depression and anxiety.
00 333 C The invention provides for each pure stereoisomer of any of the compounds described herein. Such stereoisomers may CI include enantiomers, diastereomers, or E or Z alkene or imine isomers. The invention also provides for O 5 stereoisomeric mixtures, including racemic mixtures, 00 M diastereomeric mixtures, or E/Z isomeric mixtures.
SStereoisomers can be synthesized in pure form (N6grAdi, 00 Stereoselective Synthesis, (1987) VCH Editor Ebel, H.
Sand Asymmetric Synthesis, Volumes 3 5, (1983) Academic Press, Editor Morrison, or they can be resolved by a variety of methods such as crystallization and chromatographic techniques (Jaques, Collet, A.; Wilen, Enantiomer, Racemates, and Resolutions, 1981, John Wiley and Sons and Asymmetric Synthesis, Vol. 2, 1983, Academic Press, Editor Morrison, J).
In addition the compounds of the present invention may be present as enantiomers, diasteriomers, isomers or two or more of the compounds may be present to form a racemic or diastereomeric mixture.
The compounds of the present invention are preferably pure, more preferably 90% pure, and most preferably pure.
Included in this invention are pharmaceutically acceptable salts and complexes of all of the compounds described herein. The acids and bases from which these salts are prepared include but are not limited to the acids and bases listed herein. The acids include, but are not limited to, the following inorganic acids: hydrochloric acid, hydrobromic acid, hydroiodic acid, 334 Ssulfuric acid and boric acid. The acids include, but are not limited to, the following organic acids: acetic acid, C malonic acid, succinic acid, fumaric acid, tartaric acid, maleic acid, citric acid, methanesulfonic acid, benzoic acid, glycolic acid, lactic acid and mandelic acid. The M bases include, but are not limited to ammonia, methylamine, ethylamine, propylamine, dimethylamine, 00 diethylamine, trimethylamine, triethylamine, Sethylenediamine, hydroxyethylamine, morpholine, C 10 piperazine and guanidine. This invention further provides for the hydrates and polymorphs of all of the compounds described herein.
The present invention includes within its scope prodrugs of the compounds of the invention. In general, such prodrugs will be functional derivatives of the compounds of the invention which are readily convertible in vivo into the required compound. Thus, in the present invention, the term "administering" shall encompass the treatment of the various conditions described with the compound specifically disclosed or with a compound which may not be specifically disclosed, but which converts to the specified compound in vivo after administration to the patient. Conventional procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in Design of Prodrugs, ed. H.
Bundgaard, Elsevier, 1985.
The present invention further includes metabolites of the compounds of the present invention. Metabolites include active species produced upon introduction of compounds of this invention into the biological milieu.
00 335 Throughout the invention, the term "binding affinity" C' describes the concentration of a compound required to occupy one-half of the binding sites in a receptor 00 5 population, as detectable by radioligand binding. Binding M affinity concentration can be represented as Kj, inhibition constant, or KD, dissociation constant.
00 The term "selectivity of binding affinity" refers to the ability of a chemical compound to discriminate one receptor from another. For example, a compound showing selectivity for receptor A versus receptor B will bind receptor A at lower concentrations than those required to bind receptor B.
Therefore, the statements of the form "binds to the GAL3 receptor with a binding affinity at least ten-fold higher than" a named receptor, indicates that the binding affinity at the GAL3 receptor is at least ten-fold greater than that for a named receptor, and binding affinity measurements Ki or KD) for the compound are at least ten-fold lower in numerical value.
The present invention provides a method of treating depression in a subject which comprises administering to the subject a composition comprising a pharmaceutically acceptable carrier and a therapeutically effective amount of a GAL3 receptor antagonist, wherein: the GAL3 receptor antagonist binds to the human GAL3 receptor with a binding affinity at least ten-fold higher than the binding affinity with which it binds to the human GALl receptor; 00 336 the GAL3 receptor antagonist does not inhibit the activity of central monoamine oxidase A C greater than 50 percent, at a concentration of and 00 5 the GAL3 receptor antagonist does not inhibit r the activity of central monoamine oxidase B Sgreater than 50 percent, at a concentration of 00 10pM; and the GAL3 receptor antagonist binds to the human GAL3 receptor with a binding affinity at least ten-fold higher than the binding affinity with which it binds to each of the following transporters: serotonin transporter, norepinephrine transporter, and dopamine transporter.
The present invention provides a method of treating anxiety in a subject which comprises administering to the subject a composition comprising a pharmaceutically acceptable carrier and a therapeutically effective amount of a GAL3 receptor antagonist, wherein: the GAL3 receptor antagonist binds to the human GAL3 receptor with a binding affinity at least ten-fold higher than the binding affinity with which it binds to the human GAL1 receptor; and the GAL3 receptor antagonist binds to the human GAL3 receptor with a binding affinity at least ten-fold higher than the binding affinity with which it binds to each of the following transporters: serotonin transporter, norepinephrine transporter, and dopamine transporter.
00 337 c In some embodiments of this invention, the GAL3 receptor antagonist binds to the human GAL3 receptor with a binding affinity at least 30-fold higher than the binding affinity with which it binds to the human GAL1 receptor.
00 c In further embodiments of the invention, the GAL3 receptor antagonist binds to the human GAL3 receptor with 00 a binding affinity at least 50-fold higher than the binding affinity with which it binds to the human GAL1 receptor.
In other embodiments of the invention, the GAL3 receptor antagonist binds to the human GAL3 receptor with a binding affinity at least 100-fold higher than the binding affinity with which it binds to the human GAL1 receptor.
In still other embodiments of the invention, the GAL3 receptor antagonist binds to the human GAL3 receptor with a binding affinity at least 200-fold higher than the binding affinity with which it binds to the human GAL1 receptor.
For the purposes of this invention the term "pharmaceutically acceptable carrier" has been defined herein.
The term "antagonist" refers to a compound which binds to, and decreases the activity of, a receptor in the presence of an agonist. In the case of a G-protein coupled receptor, activation may be measured using an appropriate second messenger system which is coupled to 00 338 Sthe receptor in a cell or tissue in which the receptor is expressed. Some specific but by no means limiting CI examples of well-known second messenger systems are adenylate cyclase, intracellular calcium mobilization, O 5 ion channel activation, guanylate cyclase, inositol 00 M phospholipid hydrolysis, and MAP kinase activation.
SConversely, the term "agonist" refers to a compound which 00 binds to, and increases the activity of, a receptor as Scompared with the activity of the receptor in the absence of any agonist. Methods to perform second messenger assays are described in PCT International Publication No.
97/46250 and in PCT International Publication No.
98/15570, the contents of which are hereby incorporated by reference.
In the case that a receptor has activity in the absence of an agonist (constitutive receptor activity) the antagonist may act as an inverse agonist or an allosteric modulator, as opposed to a neutral antagonist, and suppress receptor signaling independent of the agonist (Lutz and Kenakin, 1999). The categories of "antagonist compounds" are therefore seen to include 1) neutral antagonists (which block agonist actions but do not affect constitutive activity); 2) inverse agonists (which block agonist actions as well as constitutive activity by stabilizing an inactive receptor conformation); 3) and allosteric modulators (which block agonist actions to a limited extent and which may also block constitutive activity through allosteric regulation). The probability that an antagonist is neutral and therefore of "zero efficacy" is relatively low, given that this would require identical affinities for different tertiary 00 0339 cM conformations of the receptor. Thus, Kenakin proposed in S1996 that, "with the development of sensitive test 0C systems for the detection of inverse agonism will come a reclassification of many drugs. It might be observed that 00 5 numerous previously classified neutral antagonists may be C) inverse agonists" (Kenakin, 1996). Indeed, there is now C evidence from studies with known pharmacological agents 00 to support the existence of inverse agonists for numerous Sreceptors, including histamine, 5HTA, 5HT 2 c, cannabinoid, dopamine, calcitonin and human formyl peptide receptors, among others (de Ligt, et al, 2000; Herrick-Davis, et al, 2000; Bakker, et al, 2000). In the case of the 5HT 2 c receptor, clinically effective atypical antipsychotics drugs such as sertindole, clozapine, olanzapine, ziprasidone, risperidone, zotepine, tiospirone, fluperlapine and tenilapine displayed potent inverse activity whereas typical antipsychotic drugs such as chlorpromazine, thioridazine, spiperone and thiothixene were classified as neutral antagonists (Herrick-Davis et al, 2000). In the case of the histamine Hi receptor, the therapeutically used anti-allergics cetirizine, loratadine and epinastine were found to be inverse agonists. These findings further extend the idea that many compounds previously thought of as neutral antagonists will be reclassified as inverse agonists when tested in a constitutively active receptor system (de Ligt et al, 2000).
For the purpose of the claimed invention, a GAL3 antagonist useful in the treatment of depression is one which a) selectively binds to the GAL3 receptor, and b) displays antidepressant activity in the rat Forced Swim 00 S340 C3 Test. Furthermore, a GAL3 antagonist useful in the I treatment of anxiety is one which a) selectively binds to the GAL3 receptor, and b) displays anxiolytic activity in the rat Social Interaction. Also for the purpose in the 00 5 present invention, a GAL3 antagonist useful in the Streatment of depression and anxiety, is one which a) O selectively binds to the GAL3 receptor, b) displays 00 antidepressant activity in the rat Forced Swim Test, and c) displays anxiolytic activity in the rat Social Interaction Test.
In order to test compounds for selective binding to the human GAL3 receptor the cloned cDNAs encoding both the human and rat GAL1 and GAL2 receptors have been used. The cloning and assay methods for the human and rat GAL1 receptors may be found in PCT International Publication No. WO 95/22608, the contents of which are hereby incorporated by reference. The cloning and assay methods for the human and rat GAL2 receptors may be found in PCT International Publication No. WO 97/26853, the contents of which are hereby incorporated by reference.
The present invention provides for a method of determining the binding affinity of a GAL3 antagonist, wherein the GAL3 antagonist is dissolved in a "suitable solvent". A "suitable solvent" means one which permits the measurement of binding affinity of the GAL3 antagonist to the human GAL3 receptor at concentrations less than 1 uM, preferably less than 100 nM. Examples of solvents include, but are not limited to, DMSO, ethanol, N,N-dimethylacetamide, or water. For indolones, the 00 S341 C, preferred solvent is 3% DMSO (final concentration in the VI assay). For pyrimidines, the preferred solvent is 1% C ethanol/0.09% polypuronic acid F-127 (final concentration in the assay). For any other type of compounds, the 00 5 preferred solvent is the solvent which permits the O measurement of binding affinity of a GAL3 antagonist at Sthe lowest concentration. Once a suitable solvent is 00 ascertained for the binding assay of the human GAL3 Sreceptor, the same solvent is used in assays to determine the binding affinity at the GAL1 receptor, the serotonin transporter, the norepinephrine transporter, and the dopamine transporter. A solvent of 0.4% DMSO is used in the central monoamine oxidase enzyme assay.
In certain embodiments, the aforementioned GAL3 receptor antagonist additionally binds to the human GAL3 receptor with a binding affinity at least ten-fold higher than the binding affinity with which it binds to the human GAL2 receptor.
In other embodiments, the GAL3 receptor antagonist additionally binds to the human GAL3 receptor with a binding affinity at least 30-fold higher than the binding affinity with which it binds to the human GAL2 receptor.
In still other embodiments, the GAL3 receptor antagonist additionally binds to the human GAL3 receptor with a binding affinity at least 50-fold higher than the binding affinity with which it binds to the human GAL2 receptor.
In some embodiments, the GAL3 receptor antagonist additionally binds to the human GAL3 receptor with a binding affinity at least 100-fold higher than the 00 S342 C binding affinity with which it binds to the human GAL2 receptor.
(N
In further embodiments, the GAL3 receptor antagonist 0 5 additionally binds to the human GAL3 receptor with a c binding affinity at least 200-fold higher than the Sbinding affinity with which it binds to the human GAL2 00 receptor.
In other embodiments, the receptor antagonist also binds to the human GAL3 receptor with a binding affinity at least ten-fold higher than the binding affinity with which it binds to each of the human 5HTiB, human human 5HT1E, human 5HT 1 p, human 5HT2A, rat 5HT 2 c, human 5HT 6 and human 5HT, receptors.
In still another embodiment, the receptor antagonist also binds to the human GAL3 receptor with a binding affinity at least ten-fold higher than the binding affinity with which it binds to the human histamine Hi receptor.
In still another embodiment, the receptor antagonist also binds to the human GAL3 receptor with a binding affinity at least ten-fold higher than the binding affinity with which it binds to the human dopamine D 1
D
2
D
3
D
4 and Ds receptors.
In a further embodiment, the receptor antagonist also binds to the human GAL3 receptor with a binding affinity at least ten-fold higher than the binding affinity with which it binds to the human alA adrenoceptor, the human auB adrenoceptor and the human aD adrenoceptor.
00 343 In another embodiment, the receptor antagonist also binds CI to the human GAL3 receptor with a binding affinity at least ten-fold higher than the binding affinity with 005 which it binds to the human a2A adrenoceptor, the human ca2B adrenoceptor and the human 2c adrenoceptor.
0 00 In certain embodiments, the GAL3 receptor antagonist also Sbinds to the human GAL3 receptor with a binding affinity less than ten-fold higher than the binding affinity with which it binds to the human 5HT 4 receptor.
In further embodiments, the GAL3 receptor antagonist also binds to the human GAL3 receptor with a binding affinity less than ten-fold higher than the binding affinity with which it binds to the human 5HTj receptor.
In some embodiments the receptor antagonist does not inhibit the activity of central monoamine oxidase A greater than 30 percent. In further embodiments the receptor antagonist does not inhibit the activity of central monoamine oxidase B greater than 30 percent. In other embodiments the receptor antagonist does not inhibit the activity of central monoamine oxidase A greater than 15 percent. In still other embodiments the receptor antagonist does not inhibit the activity of central monoamine oxidase B greater than 15 percent. In still other embodiments the receptor antagonist does not inhibit the activity of central monoamine oxidase A and/or central monoamine oxidase B greater than percent.
00 344 c The binding properties of compounds at different receptors were determined using cultured cell lines that C selectively express the receptor of interest. Cell lines were prepared by transfecting the cloned cDNA or cloned O 5 genomic DNA or constructs containing both genomic DNA and 00 M cDNA encoding the receptors as further described in the Experimental Details herein below. Furthermore, the 00 binding interactions of compounds at different transporters and enzymes were determined using tissue preparations and specific assays as further described in the Experimental Details herein below.
In connection with this invention, a number of cloned receptors discussed herein, as stably transfected cell lines, have been made pursuant to, and in satisfaction of, the Budapest Treaty on the International Recognition of the Deposit of Microorganisms for the Purpose of Patent Procedure, and are made with the American Type Culture Collection, 10801 University Blvd., Manassas, Virginia 20110-2209. Specifically, these deposits have been accorded ATCC Accession Numbers as follows: 345 ATCC Deposits: Designation Receptor ATCC Date of Accession Deposit No.
human GAL1 CRL-1650 (CHO)hGalR2- human GAL2 CRL 12379 07/22/1997 264 L-hGalR3-228 human GAL3 CRL-12373 07/01/1997 =HT1A-3 human 5-HTIA CRL 11889 05/11/1995 Ltk-11 human S-HTIB CRL 10422 04/17/1990 (formerly human 5-HTlD2) Ltk-8-30-84 human 5-HTID CRL 10421 04/17/1990 (formerly human5-HTD1) 5HTLE-7 human 5-HTlE CRL 10913 11/06/1991 human 5-HTF CRL 10957 12/27/1991 2 human 5-HT 2 A CRL 10287 10/31/1989 (formerly humanS-HT2) pSr-1c rat 5-HT 2 C 67636 (formerly pBluescript- human 5-HT 4 75392 12/22/1992 L-5HT-4B human 5-HT 7 CRL 11166 10/20/1992 (formerly human_5-HT4B) L-cclc human a1A CRL11140 09/25/1992 (formerly human alC)
L-CL
1 j human cal CRL11139 09/25/1992 L-clA human alD CRL11138 09/25/1992 (formerly hum alA) L xA human a2A CRL11180 11/06/1992 L-NGC-cL2B human a2B CRL10275 10/25/1989 L-(C2C human cCa CRL11181 11/06/1992 pDopD 1 -GL-30 human D 5 40839 07/10/1990 (formerly hum DIP) pCEXV-H 1 human H 1 75346 11/06/1992 0 The "5-HTic", "5-HTjm 1 fl 115 IMDl2 1 1 "5-HT 4 3 11 and "5-HT 2 1 receptors were renamed the "5-HT 2 "5-HT1D", '"5-HT10, 7 11 and "5-HT 2 A" receptors, respectively, by the Serotonin Receptor Nomenclature Committee of the
IUPHAR.
a The "human aic", "human amA", and "human Dip" were renamed the "human amA", "human czD", and "human D 5 1 respectively.
346 The following receptor sequences have been deposited with the GenBank DNA database, which is managed by the National Center for Biotechnology (Bethesda, MD).
GENBANK DEPOSITS DESIGNATION RECEPTOR GENBANK No.
human mRNA for human D, D-1 receptor (formerly human X58987 1 human dopamine D2 receptor human D 2 M29066 (DRD2) mRNA complete cds Rat mRNA for dopamine D3 rat D 3 X53 944 receptor Homo sapiens dopamine D4 human D 4 L12397 receptor (DRD4) gene (D4.4) sequence The "human D 1 receptor was receptor.
renamed the "human D 1 1 1 00 347 F1 C This invention further provides a pharmaceutical composition comprising a therapeutically effective amount of the compound of the invention and a pharmaceutically acceptable carrier. In one embodiment, the amount of the 00 5 compound is an amount from about 0.01 mg to about 800 mg.
In another embodiment, the amount of the compound is an amount from about 0.01 mg to about 500 mg. In another 00 embodiment, the amount of the compound is an amount from about 0.01 mg to about 250 mg. In another embodiment, the amount of the compound is an amount from about 0.1 mg to about 60 mg. In another embodiment, the amount of the compound is an amount from about 1 mg to about 20 mg. In a further embodiment, the carrier is a liquid and the composition is a solution. In another embodiment, the carrier is a solid and the composition is a powder or tablet. In a further embodiment, the carrier is a gel and the composition is a capsule or suppository.
This invention provides a pharmaceutical composition made by combining a therapeutically effective amount of the compound of this invention and a pharmaceutically acceptable carrier.
This invention provides a process for making a pharmaceutical composition comprising combining a therapeutically effective amount of the compound of this invention and a pharmaceutically acceptable carrier.
In the subject invention a "therapeutically effective amount" is any amount of a compound which, when administered to a subject suffering from a disease against which the compounds are effective, causes 00 348 reduction, remission, or regression of the disease. In the subject application, a "subject" is a vertebrate, a Cl mammal, or a human.
00 5 The present invention provides for a method of treating a 0o M subject suffering from depression which comprises administering to the subject an amount of a compound 00 provided in the present invention effective to treat the subject's depression. The present invention also provides C 10 for a method of treating a subject suffering from anxiety which comprises administering to the subject an amount of a compound provided in the present invention effective to treat the subject's anxiety. The present invention further provides for a method of treating a subject suffering from depression and anxiety which comprises administering to the subject an amount of a compound described in the present invention effective to treat the subject's depression and anxiety.
The present invention provides for the use of any of the chemical compounds disclosed herein for the preparation of a pharmaceutical composition for treating an abnormality. The invention also provides for the use of a chemical compound for the preparation of a pharmaceutical composition for treating an abnormality, wherein the abnormality is alleviated by decreasing the activity of a human GAL3 receptor. In one embodiment, the abnormality is depression. In one embodiment, the abnormality is anxiety. In one embodiment, the abnormality is depression and anxiety.
In one embodiment, the chemical compound is a GAL3 349
OC
c receptor antagonist, wherein: the GAL3 receptor antagonist binds to the human CI GAL3 receptor with a binding affinity at least ten-fold higher than the binding affinity with O 5 which it binds to the human GAL1 receptor; 00 the GAL3 receptor antagonist does not Sinhibit the activity of central monoamine 00 oxidase A greater than 50 percent, at a Sconcentration of 10pM; and the GAL3 receptor antagonist does not inhibit the activity of central monoamine oxidase B greater than 50 percent, at a concentration of the GAL3 receptor antagonist binds to the human GAL3 receptor with a binding affinity at least ten-fold higher than the binding affinity with which it binds to each of the following transporters: serotonin transporter, norepinephrine transporter, and dopamine transporter.
In one embodiment, the chemical compound is a GAL3 receptor antagonist, wherein: the GAL3 receptor antagonist binds to the human GAL3 receptor with a binding affinity at least ten-fold higher than the binding affinity with which it binds to the human GAL1 receptor; and the GAL3 receptor antagonist binds to the human GAL3 receptor with a binding affinity at least ten-fold higher than the binding affinity with which it binds to each of the following transporters: serotonin transporter, norepinephrine transporter, and dopamine 00 S350 c transporter.
CI In the present invention the term "pharmaceutically acceptable carrier" is any pharmaceutical carrier known 0 5 to those of ordinary skill in the art as useful in 00 Cc formulating pharmaceutical compositions. On December 24, S1997 the Food and Drug Administration of the United 00 States Department of.Health and Human Services published a guidance entitled "Q3C Impurities: Residual Solvent".
The guidance recommends acceptable amounts of residual solvents in pharmaceuticals for the safety of the patient, and recommends the use of less toxic solvents in the manufacture of drug substances and dosage forms.
Table 1 of the guidance lists "Class 1 Solvents". The guidance then states that the use of Class 1 Solvents should be avoided in the production of drug substances, excipients, or drug products unless their use can be strongly justified in a risk-benefit assessment. The guidance further states that Class 2 Solvents should be limited in order to protect patients from potentially adverse effects. The guidance characterized the following solvents as Class 1 Solvents: benzene, carbon tetrachloride, 1,2-dichloroethane, 1,1-dichloroethene, and l,1,l-trichloroethane. The guidance characterized the following solvents as Class 2 Solvents: acetonitrile, chlorobenzene, chloroform, cyclohexane, 1,2dichloroethene, dichloromethane, 1,2-dimethoxyethane, N,N-dimethylacetamide, N,N-dimethylformamide, 1,4dioxane, 2-ethoxyethanol, ethyleneglycol, formamide, hexane, methanol, 2-methoxyethanol, methylbutyl ketone, methylcyclohexane, N-methylpyrrolidone, nitromethane, pyridine, sulfolane, tetralin, toluene, 1,1,2- 00 351 c trichloroethene and xylene. As used in this invention the term "pharmaceutically acceptable carrier" shall not CI include Class 1 or Class 2 Solvents.
O 5 In an embodiment of the present invention, the 00 Mpharmaceutical carrier may be a liquid and the pharmaceutical composition would be in the form of a 00 solution. In another embodiment, the pharmaceutically acceptable carrier is a solid and the composition is in the form of a powder or tablet. In a further embodiment, the pharmaceutical carrier is a gel and the composition is in the form of a suppository or cream. In a further embodiment the compound may be formulated as a part of a pharmaceutically acceptable transdermal patch. In yet a further embodiment, the compound may be delivered to the subject by means of a spray or inhalant.
A solid carrier can include one or more substances which may also act as endogenous carriers nutrient or micronutrient carriers), flavoring agents, lubricants, solubilizers, suspending agents, fillers, glidants, compression aids, binders or tablet-disintegrating agents; it can also be an encapsulating material. In powders, the carrier is a finely divided solid which is in admixture with the finely divided active ingredient.
In tablets, the active ingredient is mixed with a carrier having the necessary compression properties in suitable proportions and compacted in the shape and size desired.
The powders and tablets preferably contain up to 99% of the active ingredient. Suitable solid carriers include, for example, calcium phosphate, magnesium stearate, talc, sugars, lactose, dextrin, starch, gelatin, cellulose, 0 352 polyvinylpyrrolidine, low melting waxes and ion exchange -s resins.
Liquid carriers are used in preparing solutions, O 5 suspensions, emulsions, syrups, elixirs and pressurized 00 compositions. The active ingredient can be dissolved or Ssuspended in a pharmaceutically acceptable liquid carrier C such as water, an organic solvent, a mixture of both or 00 Spharmaceutically acceptable oils or fats. The liquid C1 10 carrier can contain other suitable pharmaceutical additives such as solubilizers, emulsifiers, buffers, preservatives, sweeteners, flavoring agents, suspending agents, thickening agents, colors, viscosity regulators, stabilizers or osmoregulators. Suitable examples of liquid carriers for oral and parenteral administration include water (partially containing additives as above, e.g. cellulose derivatives, preferably sodium carboxymethyl cellulose solution), alcohols (including monohydric alcohols and polyhydric alcohols, e.g.
glycols) and their derivatives, and oils (e.g.
fractionated coconut oil and arachis oil). For parenteral administration, the carrier can also be an oily ester such as ethyl oleate or isopropyl myristate.
Sterile liquid carriers are useful in sterile liquid form compositions for parenteral administration. The liquid carrier for pressurized compositions can be halogenated hydrocarbon or other pharmaceutically acceptable propellent.
Liquid pharmaceutical compositions which are sterile solutions or suspensions can be utilized by for example, intramuscular, intrathecal, epidural, intraperitoneal or 00 353 subcutaneous injection. Sterile solutions can also be administered intravenously. The compounds may be V)prepared as a sterile solid composition which may be dissolved or suspended at the time of administration O 5 using sterile water, saline, or other appropriate sterile 00 injectable medium Carriers are intended to include necessary and inert binders, suspending agents, C lubricants, flavorants, sweeteners, preservatives, dyes, 00 and coatings.
C- The compound can be administered orally in the form of a sterile solution or suspension containing other solutes or suspending agents (for example, enough saline or glucose to make the solution isotonic), bile salts, acacia, gelatin, sorbitan monoleate, polysorbate (oleate esters of sorbitol and its anhydrides copolymerized with ethylene oxide) and the like.
The compound can also be administered orally either in liquid or solid composition form. Compositions suitable for oral administration include solid forms, such as pills, capsules, granules, tablets, and powders, and liquid forms, such as solutions, syrups, elixirs, and suspensions. Forms useful for parenteral administration include sterile solutions, emulsions, and suspensions.
Optimal dosages to be administered may be determined by those skilled in the art, and will vary with the particular compound in use, the strength of the preparation, the mode of administration, and the advancement of the disease condition. Additional factors depending on the particular subject being treated will 00 0 354 Sresult in a need to adjust dosages, including subject age, weight, gender, diet, and time of administration.
In This invention will be better understood from the o 5 Experimental Details which follow. However, one skilled 00 00in the art will readily appreciate that the specific Smethods and results discussed are merely illustrative of C the invention as described more fully in the claims which 00 follow thereafter.
I
00 355 Experimental Details I. Synthesis of Chemical Compounds The following examples are for the purpose of 0 illustrating methods useful for making compounds of this 00 C invention.
00 10 General Methods: All reactions were performed under an SArgon atmosphere and the reagents, neat or in appropriate C solvents, were transferred to the reaction vessel via syringe and cannula techniques. Anhydrous solvents were purchased from the Aldrich Chemical Company and used as received. The examples described in the patent were named using the ACD/Name Program (version 4.01, Advanced Chemistry Development Inc., Toronto, Ontario, M5H2L3, Canada). The 1H NMR and 13C NMR spectra were recorded at either 300 MHz (GEQE Plus) or 400 MHz (Bruker Avance) in CDC13 as solvent and tetramethylsilane as the internal standard unless otherwise noted. Chemical shifts are expressed in ppm, coupling constants are expressed in Hz, and splitting patterns are described as follows: s singlet; d doublet; t triplet; q quartet; quintet; sextet; septet; br broad; m mutiplet; dd doublet of doublets; dt doublet of triplets.. Elemental analyses were performed by Robertson Microlit Laboratories, Inc.
Unless otherwise, mass spectra were obtained using electrospray ionization (ESI, Micromass Platform II) and MH* is reported. Thin-layer Chromatography (TLC) was carried out on glass plates pre-coated with silica gel
F
254 (0.25 mm, EM Separations Tech.). Preparative TLC was carried out on glass sheets pre-coated with silica gel GF (2 mm, Analtech). Flash column chromatography was 00 356 0 Sperformed on Merck silica gel 60 (230 -400 mesh).
c3 Melting points (mp) were determined in open capillary tubes on a Mel-Temp apparatus and are uncorrected.
(N
The following additional abbreviations are used: HOAc, 0 00 acetic acid; DIPEA, diisopropylethylamine; DMF, N,N- Cc Sdimethylformamide; EtOAc, ethyl acetate; MeOH, methanol; OO TEA, triethylamine; THF, tetrahydrofuran; All solvent OC ratios are volume/volume unless stated otherwise.
A. General Procedures for Preparing Pyrimidines The compounds of this invention were prepared by sucessively displacing the three chlorine atoms of a 2,4,6-trichloropyrimidine with amines. It was found that some amines anilines) selectively displace the 2position chlorine of 2,4,6-trichloropyrimidine, whereas other amines piperidine) selectively displace the 4- or 6-position chlorine first (note that the 4- and 6positions are chemically equivalent). Some amines react non-selectively at both the 2- and 4- positions of 2,4,6trichloropyrimidine. It was also found that if the pyrimidine is substituted at the 4- or 6-position with an amine (mono- or di-substituted, or unsubstituted), then the next amine (mono- or di-substituted) undergoes substitution at the 2-position of the pyrimidine. Thus, several different Procedures were used to obtain the compounds described by this invention. The following Procedures are representative of the methods that are useful for making compounds of this invention.
00 357 Procedure A: c3 4,6-DICHLORO-N-PHENYL-2-PYRIMIDINAMINE: A solution of I/ 2,4,6-trichloropyrimidine (5.5 g, 30 mmol) in tetrahydrofuran (15 mL) was added dropwise to a solution of aniline (2.8 mL, 1 equivalent) in tetrahydrofuran 00 mL). N,N-diisopropylethylamine (5.2 mL) was added and the solution was stirred at room temperature overnight.
SThe solvent was removed and the crude material was 0 0 purified by flash chromatography on silica gel. The column was eluted with 3% ethyl acetate in hexane, followed by 15% ethyl acetate in hexane. The eluent was removed, giving 4,6-dichloro-N-phenyl-2-pyrimidinamine (1.11 g, 4.6 mmol, 15%, Rf 0.4 in 3% ethyl acetate in hexane).
Procedure B: 4,6-DICHLORO-N-(3,4-DICHLOROPHENYL)-2-PYRIMIDINAMINE:
A
solution of 2,4,6-trichloropyrimidine (5.00 3,4dichloroaniline (4.45 g, 1 equivalent) in 1,4-dioxane mL) and N,N-diisopropylethylamine (10 mL) was heated at reflux with stirring for 3 hours. The solvent was removed and the crude material was purified by flash chromatography on silica gel. The column was eluted with a gradient of cyclohexane to ethyl acetate/cyclohexane The eluent was removed, giving 4,6-dichloro-N- (3,4-dichlorophenyl)-2-pyrimidinamine (1.83 g, 58%, Rf 0.39 in ethyl acetate/cyclohexane, 2:3).
Procedure C: 6-CHLORO-!,-N4-DIMETHYL-N2-PHENYL-2,4-PYRIMIDINEDIAMINE:
I
00 358 SDimethylamine in tetrahydrofuran (2M, 15 mL) was added to a solution of 4,6-dichloro-N-phenyl-2-pyrimidinamine V (0.715 g, 2.97 mmol) in tetrahydrofuran (30 mL) and N,Ndiisopropylethylamine (0.52 mL). The resulting mixture was stirred at room temperature overnight. The solvent 00 was removed and the crude material was purified by flash Cc chromatography on silica gel, eluting with ethyl Sacetate/hexane The eluent was removed, giving 6- 0 0 chloro-N 4
,N
4 -dimethyl-N -phenyl-2,4-pyrimidinediamine (0.592 g, 2.39 mmol, 80%, Rf 0.3).
Procedure D: 2,4-DICHLORO-6-(1-PIPERIDINYL)PYRIMIDINE: A mixture of 2,4,6-trichloropyrimidine (5.0 g, 27 mmol) and piperidine (2.3 g, 27 mmol) in tetrahydrofuran (50 mL) and N,Ndiisopropylethylamine (3.5 g, 27 mmol) was stirred at room temperature for 24 hours. The solvent was removed and the crude material was purified by flash chromatography on silica gel. The column was eluted with a gradient of hexane to yield ethyl acetate/hexane The eluent was removed, giving 2,4-dichloro-6-(1piperidinyl)pyrimidine (3.67 g, 15.8 mmol, 59%, Rf 0.58 in ethyl acetate/hexane, 1:4).
Procedure E: 4-CHLORO-6-(1-PIPERIDINYL)-2-{4-[3-(TRIFLUOROMETHYL)-2- PYRIDINYL]-1-PIPERAZINYL)PYRIMIDINE: A mixture of 2,4dichloro-6-(l-piperidinyl)pyrimidine (100 mg, 0.43 mmol) and 1-[3-(trifluoromethyl)pyrid-2-yl]piperazine (119 mg, 0.52 mmol) in chlorobenzene(1 mL) was heated at 140 0 C in a sealed tube for 24 hours. The solvent was removed and 00 359 the crude material was purified by preparative TIC, eluting with hexane/ethyl acetate 4-chloro-6- tfl piperidinyl) (trifluoromethyl) -2-pyridinyl] -1pipera zinyl) pyrimi dine was obtained as a solid (79 mg, 0.19 mmol, 44%).
00 Procedure F: N-(4-METHYLPHENYL)-6-C1-PIPERIDINYL)-2-(4-[3- 00(TRIFLUOROMETHYL) -2-PYRIDINYL) -1-PIPERAZINYL} -4- PYRIMIDINAMINE: A mixture of 4-chloro-6- (1-piperidinyl) (trifluoromethyl) -2-pyridinyl] -1piperazinyllpyrimidine (75.0 mg, 0.176 mmol), p-toluidine (23.1 mg, 0.216 mmol), l,l'-(bisdiphenylphosphino)-1,1'binaphthol (8.4 mg), tris(dibenzylidene acetone) dipalladium(0) (8.2 mg), and sodium tert-butoxide (86.4 mg) in dry toluene (1 mL) was heated at 90 0 C in a sealed tube for 90 minutes. The solvent was removed and the crude material was purified by preparative TLC, eluting with hexane/ethyl acetate N-(4- Methylphenyl)-6-(l-piperidinyl)-2-{4-[3- (trifluoromethy.) -2-pyridinyl] -1-piperazinyl} -4pyrimidinamine was obtained, from the band at Rf 0.4, as a solid (59.5 mg, 0.119 mmol, 68%).
Procedure G: R'-ETHYL-le- (1H-3-INDOLYL) ETHYL) (4-METHYLPHENYL)-- PIPERIDINO-2,4-PYRIMIDINEDIAMINE: A mixture of N- [4chloro-6- (1-piperidinyl) -2-pyrimidinyl] -N-ethyl-N- (lHindol-3-yl)ethyllamine (33.4 mg, 0.087 mmol) and ptoluidine (47 mg, 0.43 rruol) was heated neat under argon at 1600C in a sealed tube for 12 hours.. The crude material was purified by preparative TLC, eluting with 00 360 hexane/ethyl acetate
N
2 -Ethyl-N2-[2-(1H-3indolyl) ethyl] -N 4 (4-methylphenyl) -6-piperidino-2,4n pyrimidinediamine was obtained, from a band at Rf 0.37, as a solid (15 mg, 0.033 mmol, 38%).
00 Procedure H: 2, Sodium 2,6-DICHLORO-N,N-DIMETHYL-4-PYRIMIDINAMINE: Sodium Shydride (0.13 g, 0.79 mmol) was added to a solution of 0 0 2,6-dichloro-4-pyrimidinamine (0.40 g, 0.95 mmol) in dry tetrahydrofuran (5 mL) and stirred for 10 minutes, at which point gas evolution had ceased. Methyl iodide (0.06 mL, 0.95 mmol) was added and the resulting solution was stirred for 3 hours at room temperature. The solution was quenched with aqueous ammonium chloride/ammonium carbonate. The solution was extracted with ethyl acetate and the extracts were dried over sodium sulfate. The solvent was removed and the resulting crude product was purified by flash chromatography over silica gel, eluting with hexane/ethyl acetate The desired product (Rf 0.55) was obtained as a white powder (70 mg, 0.36 mmol, 46%).
Procedure I: N-ETHYL-2-(1H-INDOL-3-YL)ETHANAMINE: Step 1. Acetic anhydride (1.02 g) was added dropwise to a stirring solution of tryptamine (1.60 g) in tetrahydrofuran (5 mL) at 0OC and then brought to room temperature. After 2 hours, the solvent was removed and the residue was taken up into ethyl acetate. The solution was filtered through a plug of silica gel and the solvent removed, giving N- [2-(iH-indol-3-yl)ethylacetyltryptamineacetamide (1.65 g, 100%).
00 C Step 2. Lithium aluminum hydride in tetrahydrofuran (1M, mL) was added dropwise to a stirring solution of N-[2- (1H-indol-3-yl)ethylacetyltryptamineacetamide (2.02 g) in tetrahydrofuran (10 mL) at 0 C. The solution was then 00 heated at reflux overnight. The solution was cooled to S0C and water was very carefully added dropwise. The Swhite solid was filtered and rinsed with ether/methanol O 2 X 25 mL). The solvent was removed from the filtrate, giving N-ethyl-2-(1H-indol-3-yl)ethanamine as a viscous pale yellow oil (1.75 g, 93%).
Procedure J: 4-CHLORO-N-[2-(1H-INDOL-3-YL)-1-METHYLETHYL]-6-(1- PIPERIDINYL)-2-PYRIMIDINAMINE: A mixture of 2,4dichloro-6-(1-piperidinyl)pyrimidine (80 mg, 0.34 mmol), a-methyltryptamine (59 mg, 0.34 mmol), and potassium carbonate (47 mg, 0.34 mmol) in chlorobenzene(1 mL) was heated at 150°C in a sealed tube for 16 hours. The solvent was removed and the crude material was purified by preparative TLC, eluting with cyclohexane/ethyl acetate 4-Chloro-N-[2-(1H-indol-3-yl)-1methylethyl]-6-(1-piperidinyl)-2-pyrimidinamine (R 0.19) was obtained as a solid (64.5 mg, H NMR (300 MHz, CDC13) 6 8.29 (br s, 1H), 7.68 (br d, 1H, J 7.32 1H, J 7.16 1H, J 7.12 (t, 1H, J 6.95 1H, J 5.87 1H), 4.89 (br d, 1H, J 4.36 (sextet, 1H, J 3.58 3.50 4H), 3.07 (dd, 1H, J 14.4, 2.83 (dd, 1H, J 14.1, 1.70 1.55 6H), 1.16 3H, J 6.6).
00 Procedure K: N-(4-METHYLPHENYL)-2-(1-PIPERAZINYL)-6-(1-PIPERIDINYL)-4- PYRIMIDINAMINE: A solution of 2-(4-benzyl-1piperazinyl) -N-(4-methylphenyl)-6-(1-piperidinyl)-4pyrimidinamine (0.40 g, 0.90 mmol) and ammonium formate 0 (0.28 g, 4.5 mmol) in methanol over Spalladium/charcoal was stirred at 70 0 C for 3 hours. The 0 solution was cooled and passed through celite. The 00 solvent was removed, giving the desired product as a solid (0.21 g, 0.60 mmol, 66%).
Procedure L: N-(4-METHYLPHENYL)-2-[4-(3-METHYL-2-PYRIDINYL)-1- PIPERAZINYL]-6-(1-PIPERIDINYL)-4-PYRIMIDINAMINE:
A
mixture of N-(4-methylphenyl)-2-(1-piperazinyl)-6-(1piperidinyl)-4-pyrimidinamine (100 mg, 0.284 mmol), 2bromo-3-methylpyridine (54 mg, 0.312 mmol), 1,1'- (bisdiphenylphosphino)-1,1'-binaphthol (13 mg), tris(dibenzylidene acetone)dipalladium(0) (13 mg), and sodium tert-butoxide (136 mg) in dry toluene (4 mL) was heated at 90 0 C in a sealed tube for 2 hours. The reaction was quenched with water and the solution was extracted three times with ethyl acetate. The solvent was dried and removed. The crude material was purified by preparative TLC, eluting with hexane/ethyl acetate N-(4-methylphenyl)-2-[4-(3-methyl-2-pyridinyl)-1piperazinyl]-6-(1-piperidinyl)-4-pyrimidinamine was obtained, from the band at Rf 0.46, as a solid (17.1 mg, 0.0385 mmol, 14%).
Procedure M: 4,6-DICHLORO-2-{4-[3-(TRIFLUOROMETHYL)-2-PYRIDINYL]-1- PIPERAZINYLIPYRIMIDINS and 2,4-DICHLORO-6-{4- (3- (TRIFLUOROMETHYL) -2-PYRIDINYL] -1-PIPERAZINYLIPYRIMIDINE: A solution of 4- (trifluoromethyl) -2-pyridinyl] -1piperazine (127 mg, 0.66 mmol), 2,4,6-trichloropyrimidile (100 mg, 0.55 mmol) and N,N-diisopropylethylamile (95 g.L) 00 in tetrahydrofuran (1 mL) was Btirred at 0 0 C for minutes. At this time, the starting material could no longer be detected by TLC. The solvent was removed and 00 the crude material was purified by preparative TLC, eluting with ethyl acetate/hexane Two bands were removed giving 4,6-dichloro-2-{4-[3- (trifluoromethyl) -2pyridinyl] -i-piperazinyl)pyrimidine (41.7 mg, 0.110 mmol, 17%, Rf 0.41), and 2,4-dichloro-6-{4-[3- (trifluoromethyl) -2-pyridinyl] -i-piperazinyl~pyrimidine (162 mg, 0. 429 mmol, 65%, Rf 0. 10) Procedure N: 4- 4- 4-CHLORO-6- (DIMETHYLAMINO) -2-PYRIMIDINYL] -1- PIPERAZINYLIPHENOL: DIPEA (4.535 g, 0.0260 mol) was added to a stirred solution of 4-N,N-dimethylamino-2,6dichloropyrimidine (2.00 g, 0.0104 mol) and 4-(1piperazinyl)phenol (2.23 g, 0.0125 mol) in TMF (50 mL) at room temperature under argon. The resulting mixture was ref luxed for 48 h, cooled to room temperature, quenchied with water (100 mL), concentrated under reduced pressure and the crude product was redissolved in EtOAc. The organic layer was separated and washed with water (2 X 100 mL) brine (2 X 100 mL) and purified by column 00 364 chromatography on silica using EtOAc/Hexane giving the desired product (2.77 g, Procedure 0: 00 5 A solution of p-toludine (0.2 g, 1.87 mmol) in THF (2 mL) O was added to a stirred suspension of NaH (0.11 g, 2.79 00 mmol) in anhydrous THF (2 mL) at room temperature. The O resulting mixture was heated at 40 OC for 15 minutes under argon and cooled to room temperature. 6-Chloropyrimidine (0.34 g, 1.03 mmol) in THF (25 mL) was added to the above mixture and the resulting mixture was heated at refluxed for 15 h. The reaction mixture was then cooled to room temperature and quenched with saturated. NH 4 Cl(2 drops).
The crude product was concentrated under reduced pressure and redissolved in EtOAc. The organic layer was separated and washed with aqueous citric acid (2 X 100 mL), water (2 X 100 mL) and brine (2 X 100 mL). The crude product was purified by column chromatography on silica using EtOAc/hexanes giving the desired product (0.23 g, Procedure P: 2- (4-BENZYL-1-PIPERAZINYL)
-N
4 -(3,4-DICHLOROPHENYL) -N,N 6 DIMETHYL-4,6-PYRIMIDINEDIAMINE: Potassium tert-butoxide (1.6 mmol, 1 M in 2-methyl 2-propanol) was added to a 00 365 solution of N-[2-(4-benzyl-1-piperazinyl)-6-chloro-4pyrimidinyl -N,N-dimethylamine (0.331 g, 0.997 mmol) and C1 3,4 dichloroaniline (0.178 g, 1.10 mmol) in dioxane (2 mL). Subsequently, tris(dibenzylidineacetone)dipalladium 00 5 (40 mg, 0.04 -mmol) and 2,2'-Bis(diphenylphosphino)- 1,1'binapthyl (44 mg, 0.070 mmol) were added and the 00 mixture was stirred for 7 h at 110 The resulting Smixture was cooled to room temperature and concentrated under reduced pressure. The residue was treated with saturated NaHCO 3 (50 mL) and extracted with CH 2 C1 2 (3 X mL). The organic layer was washed with brine (2 X 100 mL), dried over Na 2
SO
4 concentrated in vacuo, and purified by preparative TLC using hexane/EtOAc to give the desired product (300 mg, 65 Procedure Q: N-[2-(4-BENZYL-1-PIPERAZINYL)-6-CHLORO-4-PYRIMIDINYL DIPEA (5.00 g, 40.0 mmol) was added dropwise to a solution of the N-(2,6-dichloro-4-pyrimidinyl)-N,Ndimethylamine (5.70 g, 29.6 mmol) and benzyl piperazine (6.00 g, 34.0 mmol) in m-xylene (15 mL). The mixture was stirred overnight at 130 oC, cooled to room temperature, treated with saturated NaHCO 3 (50 mL) and then extracted with CH 2 C1 2 (3 X 50 mL). The organic layer 00 0 was washed with brine (2 X 100 mL) dried over Na 2
SO
4 and concentrated in vacuc. The crude product was purified by CI chromatography on silica using EtOAc/hexane giving the desired product (6.8 g, 20 mmol, 67%).
0 Procedure R: 00 1,IJ-DIMETHYL-N4- (4-METHYLPHENYL) (2-PHENYLETHYL) 2,4,6-PYRIMIDINETRIAMINE: A mixture of N- IA- (dimethylamino) (4-toluidino) -2-pyrirnidinyl] -2phenylacetamide (60 mg, 0. 166 mmol) and LAH (lmL, 1M in THF) in THF (10 mL) was refluxed for 3h.
The crude product was concentrated in vacuo and treated with saturated NaHCO 3 (50 mL) and extracted with CH 2 Cl 2 (3 X 50 mL). The organic layer was washed with brine (2 X 100 mL) dried over Na 2
SO
4 filtered, and concentrated in vacuo. The crude product was purified by preparative TLC using hexane/EtOAc giving the desired product mg, 52 Procedure S: N- 4- (DIMETHYLAAMINO) (4-TOLUIDINO) -2-PYRIMIDINYL] -2- PHENYLACETAMIDE: A mixture of 10, 10- dimethyl -le- (4 methyiphenyl) 6-pyrimidinetriamine (122 mg, 0.50 mmol), phenylacetyl chloride (84 mg, 0.55 mmol), and 00 367 0 C triethylamine (100 mg, 1.00 mmol) in CH 2 C1 2 was stirred at Sroom temperature for 16h. The crude product was Cy concentrated in vacuo and treated with saturated NaHCO 3 mL) and extracted with CH 2 C1 2 (3 X 50 mL). The O 5 organic layer was washed with brine (2 X 100 mL), dried over Na 2 S0 4 filtered, and concentrated in vacuo. The
(N
00 crude product was purified by preparative TLC using Shexane/EtOAc giving the desired product (60 mg, 33 Procedure T: A mixture of N 4 -(3-methoxyphenyl)-6 ,l-dimethyl-2-[4-(2thienylcarbonyl)-1-piperazinyl]-4,6-pyrimidinediamine (28 mg, 0.06 mmol) and LAH (300 uL 1M, 0.3 mmol) in THF mL) was refluxed for 16 h. The crude product was concentrated in vacuo and treated with saturated NaHC03 mL) and extracted with EtOAc (3 X 50 mL). The organic layer was washed with brine (2 X 100 mL), dried over Na 2 S0 4 filtered, and concentrated in vacuo. The crude product was purified by preparative TLC using hexane/EtOAc giving the desired product (20 mg, 39 Procedure U: 00 368 2- (3-METHOXYBENZYL) -1-PIPERAZINYL) (3- METHOXYPHENYL) If4,N-DIMETHYL-4,6-PYRIMIDINEDIAMINE:
A
CIsolution of N' 1 (3-methoxyphenyl) -A'f,AO-dimethyl-2- (1piperazinyl) 6-pyrimidinediamile (36 mg, 0.1 mmol), 005 DIPEA (52 mg, 0.4 mmol), and 1-(chloromethyl)-3methoxybenzene (20 mg, 0.13 mmol) in 5 mL of dioxane was 00 stirred at 100 0 C for 16 h. The crude product was CI concentrated in vacuo and treated with saturated NaHCO 3 mL) and extracted with CH 2 Cl 2 (3 X 50 mL) The organic layer was washed with brine (2 X 100 mL), dried over Na 2
SO
4 and concentrated in vacuo. The crude product was purified by chromatography on silica using hexane/EtOAc giving the desired product (32 mg, Procedure V: 6-CHLORO-N1- (4-METHYLPHENYL) -2,4-PYRIMIDINEDIAMINE:
A
mixture of 4,6-dichloro-2-pyrimidinamine (1.64 g, 0.01 mol) p-toluidine 07 g, 0. 01 mol) in dioxane (2 mL) was heated in a sealed tube for 30 minutes at 140 0 C. The crude product was treated with NaCH (50 ml, 2M) and extracted with CH 2 Cl 2 (3 X 50 mL) The organic layer was washed with brine (2 X 100 mL) dried over Na 2
SO
4 filtered, and concentrated in vacuo. The crude product 00 369 was purified by chromatography on silica using hexane/EtOAc giving the desired product (2 g, 78 00 00 5 Procedure W:
SN
4 (3-METHOXYPHENYL) -N 6 -DIMETHYL-2- 0 0 THIENYLCARBONYL)-1-PIPERAZINYL]-4,6-PYRIMIDINEDIAMINE: A CI mixture of 2-thiophenecarboxylic acid (15 mg, 0.12 mmol), DIPEA (129 mg, 1.00 mmol) and 0-(7azabenzotriazol-l-yl)N,N,N',N'-tetramethyluronium hexafluorophosphate (44 mg, 0.12 mmol) in DMF (5 mL) was stirred at room temperature for 30 minutes. 2N-(3methoxyphenyl) -N4,1N-dimethyl-2- (1-piperazinyl) -4,6pyrimidinediamine (36 mg, 0.10 mmol) was added to the above mixture and stirred at room temperature for 16 h.
The crude product was treated with saturated NaHCO3 mL) and extracted with EtOAC (3 X 50 mL). The organic layer was washed with brine (2 X 100 mL), dried over Na 2
SO
4 filtered, and concentrated in vacuo. The crude product was purified by chromatography on silica using hexane/EtOAc giving the desired product (25 mg, 57 Procedure X: 00 370 2 (4 -BENZYL- 1 -PI PERAZINYL) -N4 (3 -METHOXYPHENYL) -Al,N 6 f DIMETHYL-4,6-PYRIMIDINEDIAMINE: A mixture of N 4 (3- CI methoxyphelyl) -N6,14-dimethyl-2- (1-piperazinyl) -4,6pyrimidinediamile (36 mg, 0.10 mmol) and benzaldehyde (11 00 5 mg, 0.1 mmcl) in a solution of methanol (5 mL) and acetic acid (0.5 rnL) was stirred at room temperature for 1 h.
00 Sodium cyanoborohydride (7 mg, 0.1 mmol) was added to the CI above solution and stirred at room temperature for 16 h.
The crude product was treated with saturated NaHCO 3 mL) and extracted with EtOAC (3 X 50 mL) The organic layer was washed with brine (2 X 50 mL), dried over Na 2
SO
4 filtered, and concentrated in vacuo. The crude product was purified by chromatography on silica using hexane/EtOAc giving the desired product (8 mg, Procedure Y: 2- (4-BROMOPHENYL) -1-PIPERAZINYL] (3-METHOXYPHENYL) N6,N 6 -DIMETHYL-4, 6-PYRIMIDIWEDIAMINE: A mixture of N4- (3methoxyphenyl) -N6,N6-dimethyl-2- (1-piperazinyl) -4,6pyrimidinediamine (36 Mgt 0.1 mmol), l-bromo-4fluorobenzene (20 mg, 0.13 mmcl) was-heated at 100 0 C for 1 h. The crude product was dissolved in CH 2 Cl 2 (0.5 mL) 00 and purified by preparative TLC using 5 methanol in EtOAc, giving the desired product (20 mg, 40 Procedure Z: 00 OO 5 2- (2-METHOXYBENZYL) -1-PIPERAZINYL] -N,N -DIMETHYL-N 6 (4-METHYLPHENYL)-4,6-PYRIMIDINEDIAMINE: A mixture of
(N
00 N 4 ,N -dimethyl-N 6 (4-methylphenyl)-2-(1-piperazinyl)-4,6- C) pyrimidinediamine (30 mg, 0.086 mmol), 1-(chloromethyl)- 2-methoxybenzene (17 mg, 0.1 mmol) and triethylaminie (200 mg, 2 mmol) in 1 DMF (1 mL) heated by microwave at 200 oC for 12 minutes. The crude product was treated with saturated NaHCO 3 (50 mL) and extracted with EtOAC (3 X mL). The organic layer was washed with brine (2 X 100 mL), dried over Na 2
SO
4 filtered, and concentrated in vacuo. The crude product was purified by chromatography on silica using hexane/EtOAc giving the desired product (10 mg, 27 Procedure AA: N- (3-METHOXYPHENYL) I-DIMETHYL-2- (2- THIENYLCARBONYL)-1-PIPERAZINYL]-4,6-PYRIMIDINEDIAMINE: A solution of J- (3-methoxyphenyl) -N6,1,-dimethyl-2- (1piperazinyl)-4,6-pyrimidinediamine (33 mg, 0.1 mmol), 2thiophenecarbonyl chloride (20 mg, 0.14 mmol), and 00 372 triethylamine (40 mg, 0.4 mmol) in CH 2 C12 (5 mL) was stirred at room temperature for 16 h. The crude product C- was concentrated in vacuo and treated with saturated NaHC03 (50 mL) and extracted with CH 2 C1 2 (3 X 50 mL). The 00 5 organic layer was washed with brine (2 X 100 mL), dried Cc O over Na 2 S04, filtered, and concentrated in vacuo. The c- 00 crude product was purified by chromatography on silica Cq using hexane/EtOAc giving the desired product as a pale red oil (35 mg, 80 Procedure BB:
N
4 I4-DIMETHYL-N- (4-METHYLPHENYL) -2,4,6- PYRIMIDINETRIAMINE: A mixture of 6-chloro-N4- (4methylphenyl)-2,4-pyrimidinediamine (1.5 g, 6.4 mmol), and N,N-dimethylamine hydrochloride (0.56 g, 7 mmol) and triethylamine (1.4 g, 14 mmol) in DMF (2 mL), was heated at 170 oC for 16 h. The product was filtered out and the organic layer was treated with saturated NaHC03 (50 mL) and extracted with EtOAC (3 X 50 mL). The organic layer was washed with brine (2 X 100 mL), dried over Na 2
SO
4 filtered, and concentrated in vacuo. The crude product was purified by chromatography on silica using hexane/EtOAc giving the desired product (0.6 g, 00 373 -s Procedure CC: C( N-(4-METHYLPHENYL)-2-[4-(1-OXIDO-2-PYRIDINYL)-1- PIPERAZINYL]-6-(1-PIPERRDINYL)-4-PYRIMIDINAMINE:
A
00 0 5 solution of 3- cholorperbenzoic acid (450 mg, 2.6 mmol), 0 and 30 H202 (0.1 mL) in CH 2 C12 (2 mL) was added to a 0 0 solution of N-(4-methylphenyl)-6-(l-piperidinyl)-2-[4-(2eC pyridinyl)-1-piperazinyl]-4-pyrimidinamine (150 mg, 0.300 mmol) in CH 2 C12 at 0 OC. The resulting mixture was gradually warmed to room temperature and stirred for 24 h, crude product was treated with saturated NaHCO 3 (50 mL) and extracted with EtOAC (3 X 50 mL). Combined organic layers were washed with brine (2 X 50 mL), dried over Na 2
SO
4 filtered, concentrated in vacuo, and purified by chromatography on silica using hexane/EtOAc to give the desired product.
Piperazines that were not commercially available were synthesized according to the method previously described (Ennis and Ghazal, 1992).
The following are examples to illustrate the compounds of this invention. Procedures A BB as described above, were used and any modifications are noted in parentheses.
Example 1: JN-CYCLOHEXYL-1f-METHYL-N4-(4-METHYLPHENYL)-6- 00 (1-PIPERIDINYL)-2,4-PYRIMIDINEDIAMINE: Prepared by c Procedures D, G (for substitution with cyclohexylamine), In and G. 'H NMR (300 MHz, CDC1 3 6 7.22 2H, J 7.8), 7.12 2H, J 5.29 1H), 4.43 (br s, 1H), 3.55 3.44 5H), 3.01 3H), 2.33 3H), 2.00 00 1.05 16H).
o Example 2: N 2 -CYCLOHEXYL-.N- (2-METHOXYETHYL) (4oO METHYLPHENYL)-6-(1-PIPERIDINYL)-2,4-PYRIMIDINEDIAMINE: Prepared by Procedures D, J (1300C), and F (2 hours). 'H NMR (300 MHz, CDC13) 6 7.25 2H, J 7.10 2H, J 6.17 (br s, 1H), 5.31 1H), 4.58 4.43 (m, 1H), 3.61 3.57 4H), 3.52 3.48 4H), 3.39 (s, 3H), 2.31 3H), 1.83 1.75 4H), 1.70 1.50 (m, 7H), 1.43 1.37 4H), 1.19 1.05 1H); ESI-MS m/z 424 Example 3: N 4 -(4-METHYLPHENYL) -D-PHENYL-6- (1- PIPERIDINYL)-2,4-PYRIMIDINEDIAMINE: Prepared by Procedures A, B (for substitution with aniline), and E (100°C, for substitution with piperidine). 1H NMR (300 MHz, CDC1 3 6 7.58 2H, J 7.26 2H, J 7.19 2H, J 7.15 2H, J 6.95 1H, J 6.82 (br s, 1H), 6.48 (br s, 1H), 5.49 1H), 3.56 3.46 4H), 2.34 3H), 1.67 1.52 6H); ESI-MS m/z 360 (MH).
Example 4: J ,N-DI(4-METHYLPHENYL)-6-PIPERIDINO-2,4- PYRIMIDINEDIAMINE: Prepared by Procedures D and G (100 0
C,
12 hours, for substitution of p-toludine at C2 and C4 of the pyrimidine) H NMR (300 MHz, CDC1 3 6 7.47 2H, J 7.20 2H, J 7.15 2H, J 8.3), 00 375 7.10 2H, J 6.79 (br s, 1H), 6.46 (br s, 1H), F 5.52 1H), 3.51 4H, J 2.36 3H), 2.31 ID 3H), 1.69 1.53 6H); ESI-MS m/z 374 (MH).
Example 5: (4-CHLOROPHENYL) (4-METHYLPHENYL) 00 PIPERIDINYL)-2,4-PYRIMIDINEDIAMINE: Prepared by Procedures D, G (for substitution with 4-chloroaniline), O and G (3.5 hours). H NMR (300 MHz, CDC1 3 6 8.79 (br s, 0 0 1H), 7.72 (br s, 1H), 7.54 2H, J 7.28 7.17 6H), 5.36 1H), 3.61 3.46 4H), 2.36 3H), 1.76 1.53 6H); ESI-MS m/z 393 (MH+ with 3 C1), 395 (MH with 7"Cl).
Example 6: Nf-METHYL-N'- (4-METHYLPHENYL) -AP-PHENYL-6- (1- PIPERIDINyl)-2,4-PYRIMIDINEDIAMINE: Prepared by Procedures D, G (140 0 C, 90 minutes, for substitution with aniline), and G (3.5 hours). 1 H NMR (300 MHz, CDC13) 6 7.42 7.33 4H), 7.18 7.14 (overlapping t at 7.16 d at 7.15, 3H), 7.07 2H, J 6.25 (br s, 1H), 5.41 1H), 3.54 3H), 3.50 3.42 4H), 2.33 (s, 3H), 1.68 1.50 6H); ESI-MS m/z 374 (MH).
Example 7: R2-METHYL-N, N-DI (4-METHYLPHENYL) PIPERIDINYL)-2,4-PYRIMIDINEDIAMINE: Prepared by Procedures D, G (180°C, 10 hours, for substitution with N-methyl-p-toluidine), and G (140 0 1H NMR (300 MHz, CDC13) 5 7.27 7.04 8H), 6.19 (br s, 1H), 5.38 (s, 1H), 3.52 3H), 3.48 3.41 4H), 2.38 3H), 2.31 3H), 1.67 1.49 6H); ESI-MS m/z 388 Example 8: N 2 [2-(5-METHYL-lH-3-INDOLYL) ETHYL] N-(4- 00 376 METHYLPHENYL)-6-(1-PIPERIDINYL)-2,4-PYRIMIDINEDIAMINE: Prepared by Procedures D, J, and G (160 0 C, 12 hours). lH kn NMR (300 MHz, CDC1 3 6 8.05 (br s, 1H), 7.43 1H), 7.23 1H, J 7.15 2H, J 7.10 2H, J 7.00 1H, J 6.98 1H), 6.43 (br s, 00 1H), 5.37 1H), 4.86 (br t, 1H, J 3.70 2H, J 3.52 3.43 4H), 3.02 2H, J 7.1), 2.46 3H), 2.32 3H), 1.67 1.49 6H) ESI-MS 00 m/z 441 (MW).
Example 9: N- (5-METHOXY-1H-3-INDOLYL) ETHYL] N- (4- METHYLPHENYL)-6-(1-PIPERIDINYL)-2,4-PYRIMIDINEDIAMINE: Prepared by Procedures D, E (1600C, 36 hours), and G. H NMR (300 MHz, CDC1 3 6 8.00 (br s, 1H), 7.15 2H, J 7.12 2H, J 7.08 7.04 3H), 6.85 (dd, 1H, J 8.8, 6.48 (br s, 1H), 5.36 1H), 4.96 (br s, 1H), 3.85 3.72 3.67 2H), 3.55 3.45 4H), 3.02 2H, J 2.32 3H), 1.68 1.49 6H); ESI-MS m/z 457 Example 10: N2- (1H-3-INDOLYL) ETHYL] -N4- (4- METHYLPHENYL)-6-(1-PIPERIDINYL)-2,4-PYRIMIDINEDIAMINE: Prepared by Procedures D, E (100 0 and G (1500C). 'H NMR .(300 MHz, CDC13) 6 8.34 (br s, 1H), 7.63 1H, J 7.31 1H, J 7.23 7.09 6H), 6.94 1H), 6.60 (br a, 1H), 5.36 1H), 4.95 1H, L 3.68 (dt, 2H, J 6.3, 3.48 3.44 4H), 3.01 2H, J 2.31 3H), 1.65 1.48 6H); ESI-MS m/z 427 (MH+) Example 11: N 2 (2-(1H-3-INDOLYL)ETHYL]-N-METHYL-N'-(4- METHYLPHENYL)-6-(1-PIPERIDINYL)-2,4-PYRIMIDINEDIAMINE: 00 377 Prepared by Procedures D, E (1600C, 4 hours) and F (12 hours). 1 H NMR (300 MHz, CDC1 3 6 8. 02 (br s, 1H) 7. 71 1H, JT 7.36 1H, LT 7.22 2H, J 7.20 1H, J 7.17 .7.09 3H) 7.03 1H) 6.40 (br s, 1H) 5.35 1H) 3.91 2H, J 00 7.8) 3.56 3.46 (in, 4H-) 3.16 3H) 3.09 2H, J 2.33 3H), 1.70 1.52 6H); ESI-MS m/z 441 (MW) 00 Example 12: (lH-INDOL-3-YL) ETHYL JIeMETHYL-N4- PHENETHYL-S- (1-PIPERIDINYL) 4-PYRIMIDINEDIAMINE: Prepared by Procedures D, E (1600C, 12 hours) and G. lH NMR (300 MHz, CDC1 3 6 8.-00 (br s, 1H) 7.71 1H, LJ 7.34 2H, J 7.24 7.15 Cm, 5H), 7.08 1H, J 6.98 1H), 4.95 Cs, 1H), 4.39 (br s, 1H), 3.88 t, 2H, J 3.57 3.48 (in, 6H), 3.12 (s, 3H) 3.05 2H, J 2.89 t, 2H, J 1.68 1.53 (mn, 6H); ESI-MS m/z 455 (MHW).
Example 13: (H-INDOL-3-YL) ETYL] -9-METHYL-Nbi2- NAPETHYL) (1-PIPERIDINYL) 4-PYRIMIDINEDIAMINE: Prepared by Procedures D, E (1600C, 12 hours, for substitution with N-methyltryptamine) and E (160 0 C, 12 hours). 'H NMR (300 MHz, CDC1 3 6 7.95 (br s, 1H), 7.92 1H), 7.78 7.75 (in, 3H), 7.72 1H, J 7.46 7.41 (mn, 2H) 7.37 2H, J 7.20 iN, j 7.11 1H, J 7.01 1H), 6.42 (br s, 1H), 5.45 1H), 3.95 2H, J 3.56 3.49 (in, 4H) 3.19 Cs, 3H) 3. 11 2H, J 7.8) 1.62 1.59 (in, 6H); ESI-MS m/z 477 (MHW) Example 14: (3-FLUOROPHENYL) -AR- (lH-INDOL-3- Example 14: (3-FLUOROPHENYL) -Ii- (1H-INDOL-3- 00 378 cIYL) ETHYL] -R2 -METH- 6 PI PERIDINYL) 2,4-7 PYRIMIDINEDIAMINE: Prepared by Procedures D, E (1600C, 12 hours, for substitution with 'N-methyltryptamine) and G.
1 H NNR (300 MHz, CDC1 3 5 7.97 (br s, 1H) 7.71 1H, J 7.41 Cdt, 1H, J 9.5, 7.34 1H, J= 00 7.22 7.06 Cm, 4H), 7.02 7.00 Cs at 7.02 dat 7.01 overlapping, 2H), 7.01 1H), 6.33 (br s, 1H), 34 1H) 3. 90 Ct, 2H, LT 7. 8) 3. 58 3. 50 Cm, 4H) 00 3.16 3H) 3.08 Ct, 2H, LT 1.70 1.54 (mn, 6H); ESI-MS m/z 445 (MH 4 Example 15: ZO- (3,4-DIFLUOROPHENYL) (1.H-INDOL-3- YL) ETHYL] -PP-METHYL-6- CI-PIPERIDINYL) -2,4- PYRIMIDINEDIAMINE: Prepared by Procedures D, E (1600C, 12 hours, for substitution with N-methyltryptamine), and G.
1H &MR (300 MHz, CDC1 3 7.99 (br s, 1H), 7.68 1H, J 7.51 (add, 1H, JL= 9.5, 7.8, 7.35 Cd, 1H, LT 7.19 1K, JLT 7.11 Ct, 1H, LJ 7. 8) 7.07 6.90 Cm, 3H), 7.01 Cs, 1H), 6.22 Cbr s, 1H), 5.23 Cs, 1H) 3.89 Ct, 2H, LT 3.57 3.49 Cm, 4H), 3.15 3H), 3.07 Ct, 2H, J 1.68 1.53 Cm, 6H); ESI-MS m/z 463 Example 16: N 4 -(3-CKLORO--4-METHYLPHENYL)-l- [2-(C1I-INDOL- 3 -YL) ETHYL] -METHYL- 6 -PI PERIDINYL) -2,4- PYRIMIDINEDIANINE: Prepared by Procedures D, E (1600C, 12 hours, for substitution with N-methyltryptamine) and G.
1H NMR (300 MHz, CDC1 3 6 7.96 (br s, 1H), 7.69 1H, J 7.51 Cs, 1H), 7.36 Cd, 1H, J 7.19 1H, J 7.14 7.06 (in, 3H), 7.01 1H), 6.18 (br s, 1K), 5.29 1K), 3.89 Ct, 2H, J 3.53 3.48 (in, 4H) 3.13 3H) 3.07 2H, LT 2.31 Cs, 3H) 00 379 1.70 1.55 6H); ESI-MS m/z 475 SExample 17: le-[2-(1H-INDOL-3-YL)ETHYL]-N METHOXYPHENYL) -lI-METHYL-6- (1-PIPERIDINYL) -2,4- PYRIMIDINEDIAMINE: Prepared by Procedures D, E (1600C, 12 00 hours, for substitution with N-methyltryptamine), and G.
1 H NMR (300 MHz, CDC13) 6 8.02 (br s, 1H), 7.71 1H, J 7.34 1H, J 7.25 7.04 4H), 7.01 00 1H), 6.89 1H, J 6.57 (dd, 1H, J 8.3, 6.30 (br s, 1H), 5.42 1H), 3.91 2H, J 3.76 3H), 3.57 3.49 4H), 3.16 3H), 3.08 2H, J 1.70 1.53 6H); ESI-MS m/z 457 Example 18: g-ETHYL-e- [2-(1H-INDOL-3-YL)ETHYL] (4- METHYLPHENYL)-6-(1-PIPERIDINYL)-2,4-PYRIMIDINEDIAMINE: Prepared by Procedures D, E (160 0 C, 12 hours, for substitution with N-ethyltryptamine), and G. 1 H NMR (300 MHz, CDC13) 6 7.97 (br s, 1H), 7.71 1H, J 7.35 1H, J 7.25 7.16 (overlapping d at 7.23 t at 7.22, 3H), 7.14 1I, J 7.08 2H, J 7.02 1H), 6.19 (br s, 1H), 5.34 1H), 3.82 2H, J 3.61 2H, J= 3.55 3.45 (m, 4H), 3.08 2H, J 2.30 6H), 1.68 1.50 (m, 6H), 1.18 3H, J ESI-MS m/z 455 Example 19: [f-[2-(1H-INDOL-3-YL)ETHYL] METHOXYETHYL) (4-METHYLPHENYL) (1-PIPERIDINYL) -2,4- PYRIMIDINEDIAMINE: Prepared by Procedures D, E (1600C, 12 hours, for substitution with N-methoxyethyltryptamine), and G. 1 H NMR (300 MHz, CDC1 3 5 7.96 (br s, 1H), 7.72 (d, 1H, J 7.35 1H, J 7.27 7.07 6H), 00 380 7.02 1H), 6.19 (br s, 1H), 5.35 1H), 3.88 (dd, c 2H, J 9.9, 3.74 2H, J 3.60 (dd, 2H, J tn 10.5, 3.57 3.46 4H), 3.34 3H), 3.12 3.07 2H), 2.32 6H), 1.70 1.58 6H); ESI-MS m/z 485 (MH).
00 O Example 20: 2- [2-(1H-3-INDOLYL)-1-METHYLETHYL]-N- (4- METHYLPHENYL)-6-(1-PIPERIDINYL)-2,4-PYRIMIDINEDIAMINE: 0 0 Prepared by Procedures D, J, and G. lH NMR (300 MHz, CDC1 3 6 8.10 (br s, 1H) 7.70 1H, J 7.36 (d, 1H, J 7.19 6.98 7H), 6.60 (br s, 1H), 5.35 1H), 4.89 (br s, 1H), 4.44 4.36 1H), 3.55 3.45 4H), 3.14 (dd 1H, J 14.1, 2.84 (dd, 1H, J 14.1, 2.33 3H), 1.62 1.50 6H), 1.18 3H, J ESI-MS m/z 441 (MH).
Example 21: N- [2-(1H-INDOL-3-YL)-1-METHYLETHYL] METHYL-.N- (4-METHYLPHENYL) (1-PIPERIDINYL) -2,4- PYRIMIDINEDIAMINE: Prepared by Procedures D, E (160°C, 12 hours, for substitution with N,a-dimethyltryptamine), and G. 1 H NMR (300 MHz, CDC1 3 6 7.92 (br s, 1H) 7.73 1H, J 7.34 1H, J 7.19 7.09 6H), 7.03 1H), 6.17 (br s, 1H), 5.34 1H), 3.51 3.44 5H), 3.11 3.05 1H), 3.02 2H), 2.90 (dd, 1H, J 14.7, 2.32 3H), 1.65 1.49 6H), 1.18 3H, J ESI-MS m/z 455 (MH).
Example 22: J -METHYL-N- (4-METHYLPHENYL) -'2-PHENETHYL-6- (1-PIPERIDINYL)-2,4-PYRIMIDINEDIAMINE: Prepared by Procedures D, E (160°C, 12 hours, for substitution at C2 of the pyrimidine), and G. ESI-MS m/z 402 00 Example 23: 2- (4-BENZYL-1-PIPERAZINYL) METHYLPHENYL)-6-(1-PIPERIDINYL)-4-PYRIMIDINAMINE: Prepared by Procedures D, I (1400C, overnight, for substitution with N-benzylpiperazine) and F (2 hours).
'H NMR (300 MHz, CDC13) 6 7.38 7.26 5H) 7.18 (d, 00 1H, J 7.12 1H, J 6.18 (br s, 1H), 5.34 1H), 3.93 3.87 4H), 3.77 4H, J 3.55 2H), 3.48 3.42 4H), 2.49 4H, J 00 2.31 3H), 1.66 1.49 6H); ESI-MS m/z 443 Example 24: N-(4-METHYLPHENYL)-2-(4-PHENYL-1- PIPERIDINYL)-6-(1-PIPERIDINYL)-4-PYRIMIDINAMINE: Prepared by Procedures D, E (16 hours, for substitution with 4phenylpiperidine), and F (1 hour). 'H NMR (300 MHz, CDC13) 6 7.34 7.24 5H), 7.19 2H, J 7.12 2H, J 6.22 (br s, 1H), 5.36 1H), 4.89 (d with fine splitting, 2H, J 13.0), 3.52 3.42 4H), 2.86 (dt, 2H, J 1.0, 13.0), 2.73 (tt, 1H, J 11.6, 2.32 3H), 1.89 (d with fine splitting, 2H, J 12.0), 1.74 (ddd, 2H, J 13.0, 12.0, 1.67 1.52 6H); ESI-MS m/z 428 Example 25: N-(4-METHYLPHENYL)-2-(4-PHENYLPIPERAZINYL)-6- (1-PIPERIDINYL)-4-PYRIMIDINAMINE: Prepared by Procedures D, G (180 0 C, 2.5 hours, for substitution with Nphenylpiperazine), and G (140 0 C, overnight). 1H NMR (100 MHz, CDC13) 6 7.28 2H, J 7.19 2H, J 7.13 2H, J 6.99 2H, J 6.89 1H, J 6.23 (br s, 1H), 5.38 1H), 3.91 (t, 2H, J 3.54 3.44 4H), 3.23 2H, J 4.6), 2.34 3H), 1.71 1.51 6H); ESI-MS m/z 429 00 382 Example 26: 2-14-(2-ETHYLPHENYL)-1-PIPERAZINYL-N-(4- METHYLPHENYL)-6-(1-PIPERIDINYL)-4-PYRIMIDINAMINE: Prepared by Procedures D, E (1200C), and F. 1H NMR (300 MHz, CDC1 3 5 7.28 1H, J 7.24 7.08 7H), 00 6.37 (br s, 1H), 5.41 1H), 3.98 3.90 4H), 3.53 3.47 4H), 2.99 2.92 4H), 2.80 2H, J 2.35 3H), 1.69 1.54 6H), 1.31 3H, J 00 ESI-MS m/z 457 (ME).
Example 27: 2-[4-(2,6-DIMETHYLPHENYL)-1-PIPERAZINYL]-N- (4-METHYLPHENYL)-6-(1-PIPERIDINYL)-4-PYRIMIDINAMINE: Prepared by Procedures D, E (120 0 and F. 'H NMR (300 MHz, CDC1 3 6 7.22 2H, J 7.15 2H, J 7.05 7.95 3H), 6.30 (br s, 1H), 5.39 1H), 3.88 4H, J 3.53 3.47 4H), 3.15 4H, J 2.37 6H), 2.34 3H), 1.68 1.53 (m, 6H); ESI-MS m/z 457 (MH 4 Example 28: N-{2-[4-(2,4-DIMETHOXYPHENYL)PIPERAZINYL]-6- (1-PIPERIDINYL)-4-PYRIMIDINYL}-N-(4-METHYLPHENYL)AMINE: Prepared by Procedures D, E (150OC, 16 hours), and F hours). 'H NMR (300 MHz, CDC1 3 5 7.18 2H, J 8.1), 7.12 2H, J 6.88 1H, J 6.50 (d, 1H, J 6.43 (dd, 1H, J 8.7, 6.23 (br s, 1H), 5.36 1H), 3.94 4H, J 3.87 314), 3.79 3H), 3.52 3.44 4H), 3.03 4H, J 2.33 3H), 1.65 1.52 6H); ESI-MS m/z 488 Example 29: N-(4-METHYLPHENYL) (1-PIPERIDINYL)-2-(4-[3- (TRIFLUOROMETHYL)PHENYL]-1-PIPERAZINYLI-4-PYRIMIDINAMINE: Prepared by Procedures D, E (1200C, 16 hours), and F. 1H 00 383 00 NMR (300 MHz, CDC13) 5 7.36 1H, J 7.20 7.09 7H), 6.25 (br s, 1H), 5.37 1H), 4.93 4H, J lf 3.52 3.45 4H), 3.26 4H, J 2.34 3H), 1.66 1.52 6H); ESI-MS m/z 497 (MH).
00 Example 30: N-(4-METHYLPHENYL) (1-PIPERIDINYL)-2- (2- PYRIDYL) -1-PIPERAZINYL]-4-PYRIMIDINAMINE: Prepared by SProcedures D, G (1200C, 12 hours, for substitution with 0 0 N-pyrid-2-ylpiperazine), and G (1400C). lH NMR (300 MHz, CDC1 3 5 8.22 (dd, 1H, J 4.4, 7.50 (dd, 1H, J 7.8, 7.20 2H, J 7.13 2H, 8.1), 6.69 1H, J 6.63 1H, J 6.26 (br s, 1H), 5.38 1H), 3.89 4H, J 3.62 4H, J 3.55 3.45 4H), 2.33 3H), 1.70 1.52 6H); ESI-MS m/z 430 (MH).
Example 31: N- (4-METHYLPHENYL) (3-METHYL-2- PYRIDINYL)-1-PIPERAZINYL -6-(1-PIPERIDINYL)-4- PYRIMIDINAMINE: Prepared from 2-(4-benzyl-1-piperazinyl)- N-(4-methylphenyl)-6-(1-piperidinyl)-4-pyrimidinamine by Procedures K and L. 1H NMR (300 MHz, CDC1 3 6 8.19 (dd, 1H, J 4.4, 7.42 (dd, 1H, J 7.8, 7.19 (d, 2H, J 7.12 2H, J 6.85 (dd, 1H, J 7.8, 6.20 (br s, 1H), 5.38 1H), 3.93 3.87 (m, 4H), 3.53 3.48 4H), 3.24 3.18 4H), 2.33 (s, 3H), 1.67 1.53 6H); ESI-MS m/z 444 (MH).
Example 32: N-(4-METHYLPHENYL)-6-(1-PIPERIDINYL)-2-{4- [4- (TRIFLUOROMETHYL)-2-PYRIDINYL]-1-PIPERAZINYL)-4- PYRIMIDINAMINE: Prepared by Procedures D, E (16 hours), and F. ESI-MS m/z 498 (MH
I
00 384 Example 33: N-(4-METHYLPHENYL)-6-(1-PIPERIDINYL)-2-(4-[6- (TRIFLUOROMETHYL)-2-PYRIDINYL]-1-PIPERAZINYLI-4- PYRIMIDINAMINE: Prepared by Procedures D, E (16 hours), and F. 'H NMR (300 MHz, CDC1 3 6 7.56 1H, J 8.1), 7.19 2H, J 7.14 2H, J 6.94 (d, 00 1H, J 6.80 1H, J 6.23 (br s, 1H), 5.37 1H), 3.90 3.87 4H), 3.69 3.66 4H), 3.50 4.46 4H), 2.34 3H), 1.67 1.53 6H); 00 ESI-MS m/z 498 (MH).
Example 34: N-(4-METHYLPHENYL)-6-(1-PIPERIDINYL)-2-(4-[3- (TRIFLUOROMETHYL)-2-PYRIDINYL]-1-PIPERAZINYL}-4- PYRIMIDINAMINE: Prepared by Procedures D, E (16 hours), and F. 'H NMR (300 MHz, CDC1 3 6 8.43 (dd, 1H, J 4.4, 7.87 (dd, 1H, J 7.8, 7.19 2H, J 8.1), 7.13 2H, J 6.99 (dd, 1H, J 7.8 6.23 (br s, 1H), 5.37 1H), 3.89 4H, J 3.53 3.48 4H), 3.36 4H, J 2.33 3H), 1.67 1.53 6H); ESI-MS m/z 498 (MH).
Example 35: N-CYCLOHEXYL-6-(1-PIPERIDINYL)-2-(4-[3- (TRIFLUOROMETHYL)-2-PYRIDINYL]-1-PIPERAZINYL}-4- PYRIMIDINAMINE: Prepared by Procedures M, E (1200C, for addition of piperidine), and F (3 hours). 1 H NMR (300 MHz, CDC13) 6 8.43 1H, J 7.84 1H, J 6.95 (dd, 1H, J 7.4, 4.95 1H), 4.34 (br a, 1H), 3.84 4H, J 3.55 3.38 5H), 3.34 4H, J 2.02 (dd, 2H, J 12.0, 1.79 1.71 2H), 1.69 1.52 6H), 1.44 1.10 6H); ESI-MS m/z 490 (MH).
Example 36: N-BICYCLO[2.2.1]HEPT-2-YL-6- (1-PIPERIDINYL)- 00 385 2-f4- [3-(TRIFLUOROMETHYL)-2-PYRIDINYL]-1-PIPERAZINYL)-4- PYRIMIDINAMINE: Prepared by Procedures M, E (120 0 C, for addition of piperidine), and F (3 hours). 1 H NMR (300 MHz, CDC1 3 6 8.42 1H, J 7.86 1H, J 6.95 (dd, 1H,.J 7.4, 4.95 1H), 4.37 (br 00 s, 1H), 3.84 4H, J 3.57 3.47 4H), 3.40 3.31 5H), 2.25 (br s, 2H), 1.78 (ddd, 2H, J 13.0, 4.6, 1.67 1.42 9H), 1.25 1.12 4H); 00 ESI-MS m/z 502 (MH).
Example 37: N-(4-METHYLPHENYL)-6-(1-PIPERIDINYL)-2-[4-(2- PYRIMIDINYL)-1-PIPERAZINYL]-4-PYRIMIDINAMINE: Prepared by Procedures D, G (1200C, 12 hours, for substitution with N-pyrimid-2-ylpiperazine), and G (1500C, 24 hours). IH NMR (300 MHz, CDC1 3 6 8.33 2H, J 7.19 2H, J 7.13 2H, J 6.50 1H, J 7.8), 6.23 (br s, 1H), 5.37 1H), 3.97 3.82 8H), 3.56 3.44 4H), 2.34 3H), 1.70 1.53 6H); ESI- MS m/z 431 Example 38: N-(4-METHYLPHENYL)-6-(1-PIPERIDINYL)-2-(1- PYRROLIDINYL)-4-PYRIMIDINAMINE: Prepared by Procedures D, G (120 0 C, 3 hours, for substitution with pyrrolidine), and G (140 0 C, 12 hours). 1 H NMR (300 MHz, CDC1 3 6 7.20 2H, J 7.11 2H, J 6.39 (br s, 1H), 5.34 1H), 3.56 4H, J 3.53 3.44 48), 2.33 3H), 1.91 (quintet, 4H, J 1.67 1.50 6H); ESI-MS m/z 338 (MH).
Example 39: N-[2-(2,3-DIHYDRO-1H-INDOL-1-YL)-6-(1- PIPERIDINYL)-4-PYRIMIDINYL]-N-(4-METHYLPHENYL)AMINE: Prepared by Procedures D, E (16 hours), and F. 1 H NMR 00 386 (300 MHz, CDC13) 6 8.31 1H, J 7.28 7.15 (m, M 6H), 6.86 1H, J 6.31 (br s, 1H), 5.49 (s, 1H), 4.22 4H, J 3.59 3.53 4H), 3.13 (t, c 4H, J 2.35 3H), 1.70 1.55 6H); ESI-MS m/z 386 (MH).
00 Example 40: N-(4-METHYLPHENYL) (6-(1-PIPERIDINYL) -2- (1,2,3,4-TETRAHYDRO-1-QUINOLINYL)-4-PYRIMIDINYL
AMINE:
00 Prepared by Procedures D, G (180 0 C, 3 hours, for substitution with 1,2,3,4-tetrahydroquinoline), and G (1400C, 12 hours). 'H NMR (300 MHz, CDC1 3 6 7.87. 1H, J 7.19 2H, J 7.15 7.07 4H), 6.93 1H, J 6.33 (br s, 1H), 5.49 1H), 4.04 2H, J= 3.54 3.44 4H), 2.79 2H, J 2.34 3H), 1.98 (pentet, 2H, J 1.69 1.52 6H); ESI-MS m/z 400 (MH).
Example 41: N-(4-METHYLPHENYL)-N- (1-PIPERIDINYL) -2- (1,2,3,4-TETRAHYDRO-2-ISOQUINOLINYL)-4-PYRIMIDINYL]AMINE: Prepared by .Procedures D, G (180 0 C, 3 hours, for substitution with 1,2,3,4-tetrahydroisoquinoline), and G (140 0 C, 12 hours). 'H NMR (300 MHz, CDC1 3 6 7.56 1H, J 7.26 7.06 7H), 6.37 (br s, 1H), 5.35 (s, 1H), 4.89 2H), 4.00 2H, J 3.58 3.44 (m, 4H), 2.91 2H, J= 2.32 3H), 1.68 1.47 (m, 6H); ESI-MS m/z 400 (MW).
Example 42: N-[2-(6,7-DIMETHOXY-3,4-DIHYDRO-2(1H)- ISOQUINOLINYL)-6-(1-PIPERIDINYL)-4-PYRIMIDINYL]-N-(4- METHYLPHENYL)AMINE: Prepared by Procedures D, E (1600C, 12 hours), and F (5 hours). 1H NMR (300 MHz, CDC13) 6 7.19 2H, J 7.13 2H, J 6.70 (s, 00 387 00 1H), 6.64 1H), 6.25 (br s, 1H), 5.36 1H), 4.82 c¢ 2H), 4.01 2H, J 3.89 3H), 3.87 (s, 3H) 3.58 3.44 4H), 2.84 2H, J 2.33 (s, C3H), 1.68 1.52 6H); ESI-MS m/z 460 (MH).
00 Example 43: N-[2-(2,3-DIHYDRO-1H-BENZO[DE]ISOQUINOLIN-2- YL) (1-PIPERIDINYL) -4-PYRIMIDINYL] (4- O METHYLPHENYL)AMINE: Prepared by Procedures D, E (1600C, 00 12 hours), and G. ESI-MS m/z 436 Example 44: 4-PHENYL-1-[4-(1-PIPERIDINYL)-6-(4- TOLUIDINO)-2-PYRIMIDINYL]-4-PIPERIDINOL: Prepared by Procedures D, E (23 hours), and F. H NMR (300 MHz, CDC1 3 6 7.51 2H, J 7.36 2H, J 7.8), 7.26 1H CHC1 3 J 7.19 2H, J 7.12 2H, J 6.20 (br s, 1H), 5.36 1H), 4.67 (br d, 2H, J 13.5), 3.50 3.45 4H), 4.67 (br t, 2H, J 13.1), 2.33 3H), 2.10 (dt, 2H, J 4.2, 12.6), 1.78 (br d, 2H, J 13.5), 1.65 1.53 6H); ESI-MS m/z 444 Example 45: N 2 f-BIS (2-METHOXYETHYL) -N 4 (4-METHYLPHENYL) 6-(1-PIPERIDINYL)-2,4-PYRIMIDINEDIAMINE: Prepared by Procedures D, G [1400C, 2 hours, for substitution with bis(methoxyethyl)amine], and G (140 OC, 1.5 hours). 1H NMR (300 MHz, CDC13) 6 7.20 2H, J 7.10 2H, J 6.20 (br s, 1H), 5.33 1H), 3.77 4H, J 3.59 4H, J 3.47 3.40 4H) 3.36 6H), 1.64 1.49 6H); ESI-MS m/z 400 Example 46: N-(4-METHYLPHENYL)-2-(3-PHENYL-4- MORPHOLINYL)-6-(1-PIPERIDINYL)-4-PYRIMIDINAMINE: Prepared 00 388 by Procedures D, E (16 hours), and F (1 hour). 'H NMR (300 MHz, CDC1 3 7.51 2H, J 7.31 2H, J 7.23 (t, 1H, J 7.15 2H, J 7.10 2H, J 6.22 (br s, 1H), 5.84 1H, J 5.36 (s, 0 1H), 4.51 4.42 2H), 3.94 2H), 3.66 (dt, 1H, J 11.5), 3.49 3.43 4H), 3.24 (dt, 1H, J 11.5), 2.32 3H), 1.64 1.47 6H); ESI-MS m/z 430 00 (MH C Example 47: N-(4-METHYLPHENYL)-2-(2-PHENYL-4- MORPHOLINYL) (1-PIPERIDINYL) -4-PYRIMIDINAMINE: Prepared by Procedures D, E (14 hours), and F (100 0 C, 2 hours). lH NMR (300 MHz, CDC1 3 1H NMR (300 MHz, CDC1 3 6 7.46 (d, 2H, J 7.38 2H, J 7.34 1H, J 7.18 2H, J 7.13 2H, J 6.19 (br s, 1H), 5.38 1H), 4.70 (br d, 1H, J 12.6), 4.58 4.51 1H), 4.11 (dd, 1H, J 10.2, 3.80 (dt, 1H, J 2.7, 11.7), 3.50 3.43 4H), 3.10 (dt, 1H, J 2.1, 12.8), 2.89 (dd, 1H, J 13.2, 10.2), 2.33 (s, 3H), 1.66 1.50 6H); ESI-MS m/z 430 Example 48: N- (4-METHYLPHENYL) [(2S,3R) -3-METHYL-2- PHENYLMORPHOLINYL]-6-(1-PIPERIDINYL)-4-PYRIMIDINAMINE: Prepared by Procedures D, E (1200C), and F (1 hour). 1H NMR (300 MHz, CDC1 3 6 7.42 2H, J 7.39 2H, J 7.27 1H, J 7.20 2H, J 7.8), 7.14 2H, J 6.25 *(br s, 1H), 5.39 1H), 4.99 4.90 1H), 4.77 1H, J 4.39 (dd, 1H, J 13.0, 4.15 (dd, 1H, J 8.3, 3.80 (dt, 1H, J 3.7, 11.6), 3.53 3.45 4H), 3.26 (dt, 1H, J 3.7, 13.0), 2.33 3H), 1.68 1.52 6H), 0.90 (d, 00 3H, J ESI-MS m/z 444 (MH).
Example 49: 2-[(2R,3R)-3-(METHOXYMETHYL)-2- PHENYLMORPHOLINYL)-N-(4-METHYLPHENYL)-6-(1-PIPERIDINYL)- 4-PYRIMIDINAMINE: Prepared by Procedures D, E, and F (3 00 hours). 1 H NMR (300 MHz, CDC13) 5 7.56 2H, J 7.8), 7.31 2H, J 7.27 7.20 3H), 7.13 2H, J 6.31 (br a, 1H), 5.84 1H, J 5.35 00 (dd, 1H, J 9.3, 5.11 1H), 4.28 (d with splitting, 1H, J 13.0), 4.01 1H, J 3.58 3.46 6H), 3.40 3H), 3.27 3.15 1H),.2.31 (s, 3H), 1.69 1.50 6H); ESI-MS m/z 473 Example 50: N, 4 -DIMETHYL- i,N6-DIPHENYL-2,4,6- PYRIMIDINETRIAMINE: Prepared by Procedures A, C, and G (1400C, overnight) 1H NMR (300 MHz, CDC1 3 6 7.68 (d, 2H, J 7.38 7.27 6H), 7.11 7.04 1H), 6.95 1H, J 6.75 (br s, 1H), 6.38 (br s, 1H), 5.45 1H), 3.06 6H); ESI-MS m/z 306 (MW).
Example 51: N',n 4 -DIMETHYL-N- (2-METHYLPHENYL) -1?-PHENYL- 2,4,6-PYRIMIDINETRIAMINE: Prepared by Procedures A, C, and G (1400C, overnight). 1 H NMR (300 MHz, CDC1 3 6 7.63 2H, J 7.43 1H, J 7.31 7.24 (m, 3H), 7.21 1H, J 7.11 1H, J 6.96 1H, J 6.73 (br s, 1H), 6.12 (br a, 1H), 5.16 1H), 3.01 6H), 2.29 3H); ESI-MS m/z 320 (MH Example 52: 1e ,N 4 -DIMETHYL-1 (3-METHYLPHENYL) -1-PHENYL- 2,4,6-PYRIMIDINETRIAMINE: Prepared by Procedures A, C, and G (140 0 C, overnight). 1 H NMR (300 MHz, CDC1 3 7.63
I
00 390 2H, J 7.29 2H, J 7.21 1H, J 7.16 7.11 2H), 6.97 1H, J 6.91 1H, J 6.78 (br s, 1H), 6.38 (br s, 1H), 5.44 1H), 3.05 6H), 2.35 3H); ESIvMS m/z 320
(MH)
00 Example 53: N, -DIMETHYL-N- (3-METHYLPHENYL) (4- METHYLPHENYL)-2,4,6-PYRIMIDINETRIAMINE: Prepared by 00 Procedures A, C, and G (overnight) 'H NMR (300 MHz, CDC13) 6 7.50 2H, J 7.25 7.08 5H), 6.90 1H, -J 6.86 (br s, 1H), 6.54 (br s, 1H), 5.44 1H), 3.05 6H), 2.34 3H), 2.31 3H); ESI- MS m/z 334 Example 54: N, -DIMETHYL-N6-(4-METHYLPHENYL)
-I-PHENYL-
2,4,6-PYRIMIDINETRIAMINE: Prepared by Procedures A, C, and G (140 0 C, overnight). 1H NMR (300 MHz, CDC1 3 6 7.63 2H, J= 7.28 2H, J 7.21 2H, J 7.15 2H, J 6.96 1H, J 6.71 (br s, 1H), 6.29 (br s, 1H), 5.39 1H), 3.04 6H), 2.34 3H); ESI-MS m/z 320 Example 55: N2- (3,4-DICHLOROPHENYL) -N 4 n-DIMETHYL-10N- (4- METHYLPHENYL)-2,4,6-PYRIMIDINETRIAMINE: Prepared by Procedures B, C, and G (1800C, 3 hours). 'H NMR (300 MHz, CDC1 3 8 8.04 1H, J 7.27 1H, J 7. 7.24 (dd, 1H, J 7.8, 7.19 2H, J 7.15 2H, J 7.01 (br s, 1H), 6.59 (br s, 1H), 5.39 1H), 3.04 6H), 2.35 3H); ESI-MS m/z 388 (MH with 35 C1, s 35 C1) 390 (MH+ with 35 C1, 3"Ci),392 (MH+ with "cl, "C1).
00 391 Example 56: N 4
,N
4 -DIMETHYL- N2,N6-BIS(4-METHYLPHENYL) -2,4,6- PYRIMIDINETRIAMINE: Prepared by Procedures B, C, and G In (1800C, 3 hours). H NMR (300 MHz, CDC13) 6 7.49 2H, J 7.19 2H, J 7.14 2H, J 8.4), 7.08 2H, J 6.73 (br s, 1H), 6.39 (br s, 1H), 00 5.37 1H), 3.02 6H); ESI-MS m/z 334 (MH 4 Example 57: N 4 (3-FLUOROPHENYL) -N,N-DIMETHYL-N2-PHENYLoo00 2,4,6-PYRIMIDINETRIAMINE: Prepared by Procedures A, C, and G (1400C, overnight).. 1H NMR (300 MHz, CDC1 3 6 7.62 2H, -J 7.34 7.23 5H), 7.01 1H, J 6.77 (br s, 1H), 6.38 (br a, 1H), 5.43 1H), 3.07 6H); ESI-MS m/z 324 Example 58: N 2 -(4-CHLOROPHENYL)-N 6 ,N6-DIMETHYL-N2-PHENYL- 2,4,6-PYRIMIDINETRIAMINE: Prepared by Procedures A, C, and G (150OC, overnight). 'H NMR (300 MHz, CDC1 3 6 7.60 2H, J 7.32 7.26 6H), 6.96 1H, J 6.77 (br s, 1H), 6.34 (br a, 1H), 5.34 1H), 3.04 6H); ESI-MS m/z 340 (MH' with 35 C1), 342 (MN 4 with "C1).
Example 59: N4- (4-BROMOPHENYL) -N6, N6-DIMETHYL-N2-PHENYL- 2,4,6-PYRIMIDINETRIAMINE: Prepared by Procedures A, C, and G (150 0 C, overnight). 1H NMR (300 MHz, CDC1 3 6 7.59 2H, J 7.42 2H, J 7.31 7.22 (m, 4H), 6.98 1H, J 6.92 Cbr s, 1H), 6.48 (br a, 1H), 5.35 1H), 3.05 6H), ESI-MS m/z 384 (MH+ with "79Br), 386 (MH+ with 'Br) Example 60: 1N'- (3,4-DICHLOROPHENYL) N,1 -DIMETHYL-N2- PHENYL-2,4,6-PYRIMIDINETRIAMINE:- Prepared by Procedures 00 392 A, C, and G (0.5mL diisopropylethylamine added, 1500C, overnight). 'H NMR (300 MHz, CDC1 3 6 7.61 (d with s at the center, 3H, J 7.34 2H, J 7.29 (d, 1H, J 7.17 (dd, 1H, J 8.7, 6.98 1H, J 6.80 (br s, 1H), 6.33 (br s, 1H), 5.33 1H), 00 3.07 6H); ESI-MS m/z 373 (MH).
Example 61: 4- (4-CHLORO-3-METHYLPHENYL)-N6,N4-DIMETHYL-N 2 00 PHENYL-2,4,6-PYRIMIDINETRIAMINE: Prepared by Procedures A, C, and G (1500C, 1 hour). 'H NMR (300 MHz, CDC1 3 6 7.61 (dd, 2H, J 7.4, 7.30 7.25 2H), 7.19 1H, J 7.12 (dd, 1H, J 8.5, 6.97 (t, 1H, J 6.88 (br a, 1H), 6.44 (br B, 1H), 5.35 (s, 1H), 3.05 6H), 2.35 3H); ESI-MS m/z 454(MH* with s 35 C1) 456 (MH+ with "C1).
Example 62: N 4 (3-CHLORO-4-METHYLPHENYL)-N,N-DIMETHYL-N PHENYL-2,4,6-PYRIMIDINETRIAMINE: Prepared by Procedures A, C, and F (1000C, 3 hours). 'H NMR (300 MHz, CDC1 3 6 7.63 2H, J 7.41 1H, J 7.30 (t, 2H, J 7.18 1H, J 7.09 (dd, 1H, J 7.8, 6.98 1H, J 6.67 (br a, 2H), 5.35 1H), 3.07 6H), 2.37 3H); ESI-MS m/z 454(MH' with 35 C1) 456 (MH+ with "C1).
Example 63: N4- (4-tert-BUTYLPHENYL) -N,N6-DIMETHYL- PHENYL-2,4,6-PYRIMIDINETRIAMINE: Prepared by Procedures A, C, and G (150 0 C, 5 hours). 1H NMR (300 MHz, CDC1 3 6 7.62 2H, J 7.36 2H, J 7.29 (d, 2H, J 7.25 2H, J 6.95 1H, J 6.69 (br a, 1H), 6.30 (br a, 1H), 5.44 1H), 3.05 6H), 1.33 9H); ESI-MS m/z 362 00 393 0D rsl Example 64: ZN 4
-DIMETHYL-N
l (4-PHENOXYPHENYL) -1 -PHENYLn 2,4,6-PYRIMIDINETRIAMINE: Prepared by Procedures A, C, Sand G (150°C, 2 hours). 1 H NMR (300 MHz, CDC13) 6 7.61 2H, J 7.35 2H, J 7.31 7.24 (m, 00 3H), 7.12 2H, J 7.08 7.04 3H), 6.98 (t, S1H, J 6.74 (br s, 1H), 6.71 (dd, 1H, J 7.8, 6.43 (br s, 1H), 5.41 1H), 3.03 6H); ESI- 00 MS m/z 398 (MHW).
CD Example. 65: N 4 4-DIMETHYL-N 6 -(2-NAPHTHYL) PHENYL-2,4,6- PYRIMIDINETRIAMINE: Prepared by Procedures A, C, and G (150 0 C, 2 hours). 'H NMR (300 MHz, CDC13) 6 7.81 1H), 7.80 1H, J 7.75 2H, J 7.65 (d, 2H, J 7.49 7.37 3H), 7.29 2H, J 6.98 1H, J 6.85 (br s, 1H), 6.59 (br s, 1H), 5.51 1H), 3.06 6H); ESI-MS m/z 356 (MH).
Example 66: N4-CYCLOHEXYL-N 6 N-DIMETHYL- a-PHENYL-2,4,6- PYRIMIDINETRIAMINE: Prepared by Procedures A, C, and G (1400C, 2 days). lH NMR (300 MHz, CDC13) 6 7.62 2H, J 7.26 2H, J 6.92 1H, J 8.1), 6.64 (br s, 1H), 4.96 1H), 4.39 (br d, 1H, J 8.1), 3.53 3.44 1H), 3.05 6H), 2.09 1.99 2H), 1.80 1.55 4H), 1.44 1.11 4H); ESI-MS m/z 312 Example 67: N 4 N-DIMETHYL-4- (4-METHYLCYCLOHEXYL)- -N- PHENYL-2,4,6-PYRIMIDINETRIAMINE: Prepared by Procedures A, C, and G (1500C, overnight). ESI-MS m/z 326 Example 68: N 4 tert-BUTYLCYCLOHEXYL) -i f, -DIMETHYL-N2- 00 394 0 PHENYL-2,4,6-PYRIMIDINETRIAMINE: Prepared by Procedures o A, C, and G (1500C, overnight). 1H NMR (300 MHz, CDC1 3 6 l 7.62 2H, J 7.26 2H, J 6.92 (t, 1H, J 6.61 (br s, 1H), 4.96 1H), 4.32 (br d, 1H J 3.46 3.37 1H), 3.06 6H), 1.88 00 1.80 2H), 1.29 1.20 1H), 1.19 0,97 4H), O 0.87 9H); ESI-MS m/z 368 (MH).
0 00 Example 69: I*-BICYCLO [2.2.1]HEPT-2-YL-N 6
N-DIMETHYL-N-
PHENYL-2,4,6-PYRIMIDINETRIAMINE: Prepared by Procedures A, C, and G (140°C). 'H NMR (300 MHz, CDC1 3 5 7.62 (d, 2H, J 7.26 2H, J 6.92 1H, J 6.62 (br s, 1H), 4.94 1H), 4.42 (br d, 1H, J 3.45 3.37 1H), 3.06 6H), 2.33 2.27 (m, 1H), 1.82 (dd, 1H, J 12.3, 1.56 1.42 2H) 1.30 1.14 5H), 0.91 0.85 1H); ESI-MS m/z 324
(MH
4 Example 70: N1, N 4 -DIMETHYL- 2 -PHENYL-N 7,7- TRIMETHYLBICYCLO[2.2.1]HEPT-2-YL)-2,4,6- PYRIMIDINETRIAMINE: Prepared by Procedures A, C, and G (overnight). 'H NMR (300 MHz, CDC1 3 6 7.62 2H, J 7.26 2H, J 6.93 1H, J 6.87 (br s, 1H), 4.95 1H), 4.80 (br d, 1H, J 3.94 3.84 1H), 3.06 6H), 2.45 2.34 1H), 1.82 1.62 3H), 1.46 1.32 1H), 1.29 1.16 2
H),
0.99 3H), 0.90 3H), 0.89 3H) ESI-MS m/z 366 (MH) Example 71: 1 ,l-DIMETHYL-1e-PHENYL-N6-[(2R,3S)-3,6,6- TRIMETHYLBICYCLO[3.1.1]HEPT-2-YL]-2,4,6- PYRIMIDINETRIAMINE: Prepared by Procedures A, C, and G 00 395 hours). 1H NMR (300 MHz, CDC1 3 6 7.64 2H, J 8.1), 7.26 2H, J 6.92 1H, J 6.72 (br s, n 1H), 4.99 1H), 4.47 (br d, 1H, J 4.05 3.91 1H), 3.06 6H), 2.72 2.62 1H), 2.46 2.36 1H), 2.00 1.45 5H), 1.25 3H), 1.16 3H, 00 J 1.10 3H); ESI-MS m/z 366 (MH).
Example 72: I, N 4 -TRIMETHYL-I ,1 -BIS (4-METHYLPHENYL)- 00 2,4,6-PYRIMIDINETRIAMINE: Prepared by Procedures D, E (150OC, 16 hours), and F (5 hours). 1 H NMR (300 MHz, CDC1 3 -6 7.26 2H, J 7.15 (br d, 4H, J 8), 7.04 2H, J 6.19 (br s, 1H), 5.29 1H), 3.50 3H), 2.94 6H), 2.36 3H), 2.29 3H); ESI-MS m/z 348 (MH).
Example 73: N2-CYCLOHEXYL-N, N, N4-TRIMETHYL- N- (4- METHYLPHENYL)-2,4,6-PYRIMIDINETRIAMINE: Prepared by Procedures D, E (150 0 C, 12 hours), and F (5 hours). 1H NMR (300 MHz, CDC1 3 6 7.25 2H, J 7.10 2H, J 6.26 (br a, 1H), 5.22 1H), 4.66 4.52 (m, 1H), 3.01 3H), 2.99 6H), 2.32 3H), 1.87 1.64 1.52 1.35 4H), 1.22 1.06 1H); ESI-MS m/z 340 Example 74: N2-CYCLOHEXYL-- (2-METHOXYETHYL)-N, N 4 DIMETHYL-N4-(4-METHYLPHENYL)-2,4,6-PYRIMIDINETRIAMINE: Prepared by Procedures H, J (overnight), and F (2 hours).
'H NMR (300 MHz, CDC1 3 6 7.28 2H, J 7.11 (d, 2H, J 6.19 (br s, 1H), 5.22 1H), 4.60 4.50 1H), 3.64 3.55 4H), 3.39 3H), 2.99 6H), 2.31 3H), 1.83 1.75 4H), 1.73 1.63 1H), 1.52 1.38 4H), 1.19 1.05 1H); ESI-MS m/z 384 00 396 In Example 75: 2- 3-DIHYDRO-lH-INDOL-1-YL) -N%,N 4
-DIMETHYL-
N
6 (4-METHYLPHENYL) 6-PYRIMIDIN'EDIAMINE: Prepared by Procedures H, E (1500C, 16 hours), and F (2 hours) IH 00 N'MR (300 MHz, CDCl 3 6 8.37 1H, J 7.8) 7.26 2H, J 7.20 -7.11 (in, 4H), 6.86 1H, J 7.8), 6.31 (br s, 1H), 5.39 1H) 4.24 4H, J 8.3), 00 3.13 4H, LT S. 83) 3.07 6H) 2.35 3H); ESI- MS n,/z 346 Example 76: fl- 2- (1.H-3-INDOLYL) ETHYL] -10, N-DIMETHYL-N6- (4-METHYLPHENYL) 6-PYRIMIDINETRIAMINE: Prepared by Procedures H, J, and G. 1H NIVR (300 MHz, CDCl 3 6 8.19 (br B, 1H) 7. 65 (d 1H, J 7. 8) 7. 36 1H, J 7. 8) 7.21 7.09 (in, 6H) 7.04 1H) 6.52 (br s, 1H) 5.28 1H) 4.95 (br d, 1H, J 3.72 2H, J 7.2), 3 .06 2H, LT 7. 8) 2. 99 6H) 2 .32 3H); ESI-MS m/z 3 87 Example 77: 9'J- (1H-INDOL-3-YL) ETHYL] -V2J,N1,N6-TRIMETHYL- N6- (4-METHYLPHENYL) 6-PYRIMIDINETRIAMINE: Prepared by Procedures H, J, and G or F. 'H NMR (300 MHz, CDC1 3 6 8.14 (br B, 1H) 7.70 (d 1H, LT 7.32 1H, J 7.22 2H, J 7.17 1H, J 7.12 1H, J 7.08 2H, J 6.98 114), 6.36 (br a, lH), 5.25 1H), 3.90 2H, LTJ 7.8), 3 .14 3H) 3. 07 2H, LJ 7. 8) 2.99 6H) 2. 3H) ;ESI-MS m/z 401 (MH 4 Example 78: 10- (3,4-DICHLOROPHENYL) (lH-3- INDOLYL) ETHYL] bf N6 -TRIMETHYL 4,6 00 397 PYRIMIDINETRIAMINE: Prepared by Procedures H, J, and G.
1H NMR (300 MHz, CDC13) 6 8.00 (br s, 1H), 7.75 1H), n 7.68 (d 1H, J 7.35 1H, J 7.24 7.15 3H), 7.10 1H, J 7.00 1H) 6.23 (br s, 1H), 5.15 1H), 3.90 2H, J 3.14 3H), 00 3.08 2H, J 3.03 6H); ESI-MS m/z 455(MH+ with 35 C1), 457 (MH+ with 37 C1).
00 Example 79: 9d- (1H-INDOL-3-YL) ETHYL] -N2,N, Al-TRIMETHYL- (2-NAPHTHYL)-6-(1-PIPERIDINYL)-2,4,6-PYRIMIDINETRIAMINE: Prepared by Procedures D, E (1600C, 28 hours), and G. NMR (300 MHz, CDC1 3 6 8.18 (br s, 1H), 7.92 1H), 7.90 7.03 10H), 6.95 1H) 6.84 (br s, 1H), 5.34 (s, 1H), 3.90 2H, J 3.17 3H), 3.07 2H, J 2.96 6H); ESI-MS m/z 437 (MH).
Example 80: 1-[4-(DIMETHYLAMINO)-6-(4-TOLUIDINO)-2- PYRIMIDINYL]-4-PHENYL-4-PIPERIDINOL: Prepared by Procedures H, E (150 0 C, 10 hours), and F (3 hours). 1H NMR (300 MHz, CDC13) 5 7.43 2H, J 7.35 2H, J 7.27 7.21 3H), 7.14 2H, J 7.8), 6.24 (br s, 1H), 6.18 (br s, 1H), 5.28 1H), 4.43 4.37 2H), 4.03 2H, J 3.06 2.97 (m with a at 3.03, 8H), 2.66 2.58 2H), 2.34 3H).
Example 81: IV, t-DIMETHYL-PN- (4-METHYLPHENYL) PHENYL-1-PIPERIDINYL)-4,6-PYRIMIDINEDIAMINE: Prepared by Procedures H, E (1500C, 16 hours), and F (4 hours). 'H NMR (300 MHz, CDC1 3 8 7.34 7.18 7H), 7.13 2H, J 6.25 (br s, 1i), 5.28 1H), 4.94 (d with fine splitting, 2H, J 13.0), 3.01 6H), 2.87 (dt, 2H, J 13.0), 2.74 (tt, 1H, J 11.6, 2.32 3H), 00 398 1.90 (d with fine splitting, 2H, LTJ 12.0), 1.72 (ddd, 2H, LT 13.0, 12.0, ESI-MS m/z 388 ClExample 82: N 4 1
N
4 "-DIMETHYL-N$- (4-METHYLPHENYL) (3- PHENYL-4-MORPHOLINYL) 6-PYRIMIDINEDIAMINE: Prepared by 00Procedures H, E (150 0 C, 20 hours), and F (3 hours). 1H NMR (300 MHz, CDCl 3 6 7.51 2H, J 7.32 Ct, 2H, LJ 7.23 1H, LT 7.17 Cd, 2H, LT 7.8), 00 7.09 2H, LTJ 6.25 (br s, 1H) 5.88 1H, J= 1. 0) 5.27 1H) 4.49 Ct, 2H, LT 13.2) 3.94 (in, 2H) 3.66 (dt, IH, J 1.0, 11.5) 3.24 (dt, 1H,. J 11.5), 2.97 6H), 2.32 3H); ESI-MS m/z 390 CMH*).
Example 83: 0,N'-DIMETHYL-'7'- (4-METHYLPHENYL) (2- PHENYL-4-MORPHOLINYL) -A,6-PYRIMIDIEDIAMvINE: Prepared by Procedures H, E (1500C, 20 hours) and F (3 hours) 1H NMR (300 MHz, CDCl 3 6 7.47 Cd, 2H, J 7.38 Ct, 2H, LT 7.33 Ct, 1H, J 7.19 Cd, 2H, J 7.8), 7.11 2H, JLT 6.22 (br s, 1H), 5.29 Cs, 1H), 4.74 (dd, 1H, LT 13.2, 1.0) 4.59 4.51 Cm, 2H) 4.16 4. 08 Cm, 1H) 3. 80 Cdt, 1H, LT 1. 0, 11. 9) 3. 11 (dt, 1H, J 1.5, 12.4), 2.98 6H), 2.90 (dd, 1H1, LTJ 10.6, 11.9) 2.33 3H) ESI-MS znhz 390 Example 84: 10, 10-DIMETHYL-.t- (4 -METHYLPHENYL) -2 j4- (4- METHYLPHENYL) SULFONYL] -l-PIPERAZINYLI 6- PYRIMIDINEDIAMINE: Prepared by Procedures H, E (1500C, overnight), and F (3 hours). 1H NMR (300 MHz, CDCl 3 6 7.65 Cd, 2H, LT 7.31 Cd, 2H, J 7. 15 Cd, 2H, J 7.11 2H, J 6.20 (br s, 1H), 5.22 Cs, 1H), 3.87 4H, LT= 3.02 4H, LT 2.95 6H), 2.43 Cs, 3H), 2.33 Cs, 3H); ESI-MS 00 399 m/z 467 Example 85: N 4 %-DIMETHYL-N6- (4-METHYLPHENYL) (2- METHYLPHENYL)-1-PIPERAZINYL]-4,6-PYRIMIDINEDIAMINE: Prepared by Procedures D, E (1600C, 12 hours), and F (12 00 hours). H NMR (300 MHz, CDC13) 6 7.23 7.10 SH), 7.02 6.96 2H), 6.28 (br s, 1H), 5.28 1H), 3.95 3.86 4H), 2.99 6H), 2.96 2.92 4H), 2.36 00 3H), 2.32 3H); ESI-MS m/z 403 (MH).
Example 86: N4,NG-DIMETHYL-N- (4-METHYLPHENYL)-2- METHYLPHENYL)-1-PIPERAZINYL]-4,6-PYRIMIDINEDIAMINE: Prepared by Procedures D, E (1600C, 12 hours), and F (12 hours). 1 H NMR (300 MHz, CDC13) 5 7.19 2H, J 7.8), 7.17 1H, J 7.11 2H, J 6.91 (s, 1H), 6.89 1H, J 6.69 1H, J 6.33 (br s, 1H), 5.29 1H), 3.93 4H, J 3.22 (t, 4H, J 3.01 6H), 2.33 6H); ESI-MS m/z 403 (MH) Example 87: N,N-DIMETHYL-Ni- (4-METHYLPHENYL)-2-[4-(4- METHYLPHENYL)-1-PIPERAZINYL-4,6-PYRIMIDINEDIAMINE: Prepared by Procedures D, E (1600C, 36 hours), and F (8 hours). 1 H NMR (300 MHz, CDC1 3 6 7.19 2H, J 7.16 2H, J 7.10 2H, J 6.90 (d, 2H, J 6.24 (br s, 1H), 5.27 1H), 3.93 4H, J 3.18 4H, J 3.00 6H), 2.33 (s, 3H), 2.28 3H); ESI-MS m/z 403 Example 88: N 4 -DIMETHYL-N6-(4-METHYLPHENYL)-2-{4-[3- (TRIFLUOROMETHYL)-2-PYRIDINYL]-1-PIPERAZINYL}-4,6- PYRIMIDINEDIAMINE: Prepared by Procedures H, E (16 00 400 hours), and F. 'H NMR (300 MHz, CDC1 3 5 8.57 (dd, 1H, J 4.4, 7.87 (dd, 1H, J 7.8, 7.20 2H, J 7.13 2H, J 6.98 (dd, 1H, J 7.8, 6.24 (br s, 1H), 5.28 1H), 3.90 4H, J 3.36 4H, J 3.00 6H), 2.32 3H); 00 ESI-MS m/z 458 Example 89: N-(4-METHYLPHENYL)-2-(1-PIPERIDINYL)-6-{4-[3- 00 (TRIFLUOROMETHYL)-2-PYRIDINYL]-1-PIPERAZINYL}-4- PYRIMIDINAMINE: Prepared by Procedures M, E (1200C, for addition of piperidine), and F. 'H NMR (300 MHz, CDC1 3 6 8.43 (dd, 1H, J 4.4, 7.87 (dd, 1H, J 7.8, 7.19 2H, J 7.12 2H, J 6.99 (dd, 1H, J 6.28 (br s, 1H), 5.35 1H), 3.77 3.72 4H), 3.62 4H, J 3.33 4H, J 2.33 3H), 1.69 1.52 6H); ESI-MS m/z 498 (MH).
Example 90: 6-[2-(METHOXYMETHYL)-1-PIPERIDINYL]-N-(4- METHYLPHENYL)-2-(4- [3-(TRIFLUOROMETHYL) -2-PYRIDINYL] -1- PIPERAZINYL)-4-PYRIMIDINAMINE: Prepared by Procedures D, J (900C, overnight), and F (2 hours). 1H NMR (300 MHz, CDC1 3 6 8.44 (dd, 1H, J 4.4, 7.88 (dd, 1H, J 7.8, 7.20 2H, J 7.12 2H, J 8.1), 6.99 (dd, 1H, J 7.8, 6.23 (br s, 1H), 5.38 (s, 1H), 4.68 4.54 1H), 4.15 4.03 1H), 3.90 (t, 4H, J 3.57 1H, J 3.44 3.35 3.34 3H), 2.81 1H, J 12.0), 2.33 3H), 1.93 1.86 1H), 1.72 1.41 3H), 1.29 1.25 1H), 0.91 0.86 1H); ESI-MS m/z 542 (MH).
I
00 4ul Example 115: N-4- (BENZYLOXY)PHENYL] -N-6-,N-6-DIMETHYL- 2-(4-(2-PYRIDINYL)-l-PIPERAZINYL]-4,6-PYRIMIDINEDIAMINE: cI Prepared by Procedures A'(CH 2 C1 2 Et 3 N, Me 2 NHHC1, stirred h at -78 0 C, warmed to 0 OC and stirred 3 N, and 00 0. lH NMR (400 MHz, CDC1) 8 8.23 8.19 1H), 7.52 (dt, 1H, J 1.9, 7.43 7.20 Cm, 7H), 6.96 Cs, 00 1H), 6.88 Cd, 1H, J 6.80 Cd, 1H, J 6.69 C~ 6.63 2H), 5.34 1H), 5.03 2H), 4.03 3.97 (m, 4H), 3.66 4H, J 3.02 6H); ESI-MS M/z 482
CMH+).
Example 116: (DIMETHYLAMINO) (4-TOLUIDINO)-2- PYRIMIDINYL -1-PIPERAZINYL) PHENOL: Prepared by Procedures A (CH 2 C1 2 Et 3 N, Me 2 NHHCl, stirred 3.5 h at -78 OC, warmed to 0 OC and stirred 3 N, and 0. lH NMR (400 MHz,
CDC
3 5 10.04 Cs, 1H), 7.19 7.14 4H), 6.85 6.79 4H), 5.31 Cs, 1H), 5.22 Cs, 1H), 3.96 t, 4H, J 3.05 t, 4H, J 3.03 6H), 2.34 3H); FIAMS m/z 405 (MHW).
Example 117: N 4 BENZYLOXY)PHENYLI-N4,I$-DIMETHYL-2-[4- (2-PYRIDINYL)-1-PIPERAZINYL-4,6-PYRIMIDINEDIAMINE: Prepared by Procedures A CCH 2 C1 2 Et 3 N, Me 2 NHHC1, stirred h at -78 0 C, warmed to 0 0 C and stirred 3 N, and 00 402 0. 'H NMR (400 MHz, CDC1 3 6 8.21 (dd, 1H, J 1.9, 5.6), c 7.55 7.27 7H), 7.24 7.16 2H), 7.04 6.91 (m, c 2H), 6.69 6.64 2H), 5.06 2H), 5.05 1H), 4.08 3.97 4H), 3.69 4H, J 3.03 6H); 00 5 ESI-MS m/z 482 (MH 00Example 118: Pf- (1,3-BENZODIOXOL-5-YL) -16,19-DIMETHYL-2- Ci [4-(2-PYRIDINYL)-1-PIPERAZINYL]-4,6-PYRIMIDINEDIAMINE: Prepared by Procedures A (CH 2 C1 2 Et 3 N, Me 2 NHHC1, stirred 3.5 h at -78 0 C, warmed to 0 OC and stirred 3 N, and O. 'H NMR (400 MHz, CDC1 3 5 8.24 8.18 1H), 7.48 (dt, 1H, J 1.9, 6.92 1H, J 6.75 (d, 1H, J 6.74 6.54 3H), 6.41 (br s, 1H), 5.95 2H), 5.16 1H), 3.89 4H, J 3.60 (t, 4H, J 2.99 6H)l ESI-MS m/z 420 (MH).
Example 119: N4'-(2,3-DIHYDRO-1,4-BENZODIOXIN-6-YL)--N#,Nt- DIMETHYL-2-[4-(2-PYRIDINYL)-1-PIPERAZINYL]-4,6- PYRIMIDINEDIAMINE: Prepared by Procedures A (CH 2 C1 2 Et 3
N,
Me 2 NHHC1, stirred 3.5 h at -78 OC, warmed to 0 0 C and stirred 3 N, and 0. 1H NMR (400 MHz, CDC1 3 6 8.24 8.18 1H), 7.49 (dt, 1H, J 2.1, 6.89 1H, J 6.81 1H, J 6.76 1H, J 2.4), 6.68 1H, J 6.62 (dd, 1H, J 4.6, 6.18 403 (br s, 1H), 5.21 1H), 4.33 4.15 4H), 3.89 (t, 4H, J 3.61 4H, J 3.00 6H); ESI-MS m/z 434 (MH).
Example 120: NA4- (4-ISOQUINOLINYL)-R6,Zt-DIMETHYL-2-[4-(2- PYRIDINYL)-1-PIPERAZINYL)-4,6-PYRIMIDINEDIAMINE: Prepared by Procedures A (CH 2 C1 2 Et 3 N, Me 2 NHHC1, stirred 3.5 h at 78 OC, warmed to 0 *C and stirred 3 N, and 0. 1 H NMR (400 MHz, CDC1 3 8 8.93 1H, J 8.31 1H, J 8.27 8.19 1H), 8.01 1H, J 7.70 1H, J 7.59 7.52 1H), 7.51 7.45 (m, 2H), 6.78 (br s, 1H), 6.68 1H, J 6.63 (dd, 1H, J 5.0, 5.29 1H), 3.94 4H, J 3.63 4H, J 3.01 6H); ESI-MS m/z 427 (MH).
Example 121: N 4 (4-CYCLOHEXYLPHENYL) -'Af ,N-DIMETHYL-2- [4- (2-PYRIDINYL)-1-PIPERAZINYL]-4,6-PYRIMIDINEDIAMINE: Prepared by Procedures A (CH 2 C1 2 Et 3 N, Me 2 NHHC1, stirred h at -78 OC, warmed to 0 OC and stirred 3 N, and 0. 'H NMR (400 MHz, CDC1 3 8 8.25 8.19 1H), 7.49 (dt, 1H, J 2.0, 7.22 2H, J 7.16 (d, 2H, J 6.68 1I, J 6.66 6.60 (mn, 1H), 6.21 (br s, 1H), 5.30 1H), 3.99 3.91 4H), 3.63 4H, J 3.02 6H), 2.53 2.42 1H), 1.92 00 404 -1.79 Cm, 4H) 1.48 -1.32 Cm, 4H) 1.31 1.19 (in, 2H); n ES I -MS m/z 4 58 00 Example 122: N 4
,N
4 -DIMETHYL-2- (2-PYRIDINYL) -1- PIPERAZINYL] (5,6,7,8B-TETRAHYDRO-1-NAPHTHALENYL) -4,6- 00 PYRIMIDINEDIAMINE: Prepared by Procedures A (CH 2 C1 2 Et 3
N,
Me 2 NHHC1, stirred 3.5 h at -78 0 C, warmed to 0 OC and stirred 3 h) N, and 0. lH NMR (400 MHz, CDCl 3 8 8.20 (dd, 1H, J 1.3, 7.50 (dt, 1H, L 2.2, 7.17 1KH, J 5) 7.09 Ct, 1H, J 7. 6) 6. 94 1KH, LT 7.7) 6.73 -6.62 2H) 5.06 1H), 4.08 3.93 (m, 4H), 3.66 t, 4H, J 5. 3) 3. 00 Cs, 6H) 2. 79 (t 2H, J1 2.72 t, 2H, LT 5. 9) 1.88 1.67 (mn, 4H), NH (1H, unobserved); ESI-MS m/z 430 Example 123: 3-DIHYDRO-1H- INDEN-5-YL) -N6, bf- DIMETHYL-2- (2-PYRIDINYL) -1-PIPERAZINYL] -4,6- PYRIMIDINEDIAMINE: Prepared by Procedures A (CH 2 C1 2 Et 3
N,
Me 2 NHHC1, stirred 3.5 h at -78 OC, warmed to 0 OC and stirred 3 h) N, and 0. 'H NMR (400 MHz, CDC1 3 8 8.20 (d, 1K, LTJ 7.51 Cdt, 1K, J 1.8, 7.19 Cd, 1H, J 7.14 1H), 7.04 Cdd, 1H, J 1.7, 6.73 00 405 6.61 2H), 5.23 1H), 4.09 3.94 4H), 3.68 (t, .4H, J= 3.04 6H), 2.89 4H,.J 2.16 c- 2.01 2H), NH (1H, unobserved); ESI-MS m/z 416 005 Example 124: N-(3,4-DICHLOROPHENYL) -DI,#-DIMETHYL-2-[4- (2-PYRIDINYL)-1-PIPERAZINYL]-4,6-PYRIMIDINEDIAMINE: 00 Prepared by Procedures A (CH 2 C1 2 Et 3 N, Me 2 NHHC1, stirred Ci 3.5 h at -78 OC, warmed to 0 "C and stirred 3 N, and O. 'H NMR (400 MHz, CDC13) 8 8.31 8.20 1H), 7.79 7.69 1H), 7.61 7.44 1H), 7.42 7.28 1H), 7.25 7.11 1H), 6.79 6.61 2H), 6.42 (br s, 1H), 5.22 1H), 3.98 3.82 4H), 3.65 3.56 (m, 4H), 3.02 6H); ESI-MS m/z 444 (MH+ with 35 C1, 35 C1), 446 (MH* with 3 sC1, "C1) 448 (MH+ with "C1, "C1) Example 125: N 4
N
4 -DIMETHYL-2- (2-PYRIDINYL) -1- PIPERAZINYL] (TRIFLUOROMETHYL) PHENYL] -4,6- PYRIMIDINEDIAMINE: Prepared by Procedures A (CH 2 C1 2 Et 3
N,
Me 2 NHHC1, stirred 3.5 h at -78 warmed to 0 OC and stirred 3 N, and 0. 'H NMR (400 MHz, CDC1 3 8 8.59 (br a, 1H), 8.24 8.18 1H), 7.86 1H), 7.78 7.22 4H), 6.65 2H, J 5.29 1H), 3.96 (t, 4H, J 3.64 4H, JLT 3.03 6H); ESI-MS m/z 444 00 406 -s Example 126: 2-(4-BENZYL-1-PIPERAZINYL) -N 4 [3- CI (DIMETHYLAMINO) PHENYL] -N6,f -DIMETHYL-4,6- PYRIMIDINEDIAMINE: Prepared by Procedures P (toluene, 00 0 0 5 16 Q (dioxane, 120 and A. H NMR (400 MHz, 0 CDC1 3 8 7.52 7.37 7H), 7.25 1H, J 7.14
(C
0 (dd, 1H, J 1.5, 7.05 (dd, 1H, J 2.5, 4.36 C0N 2H), 3.98 (br s, 4H), 3.36 4H), 3.11 6H), 3.05 6H), 2.60 1H); ESI-MS m/z 432 (MH).
Example 127: 2-(4-BENZYL-1-PIPERAZINYL) -DIMETHYL-iN- (2-METHYL-1,3-BENZOTHIAZOL-5-YL)-4,6-PYRIMIDINEDIAMINE: Prepared by Procedures P (130 13 Q, and A. 'H NMR (400 MHz, CDC1 3 6 8.12 1H), 7.87 1H, J 8.8), 7.52 7.38 6H), 5.58 1H), 4.58 1H), 4.30 (s, 2H), 3.79 3.42 4H), 3.22 2.91 4H), 3.09 (s, 6H), 2.98 3H); ESI-MS m/z 460 (MH), Example 128: 2- (4-BENZYL-1-PIPERAZINYL) -bN-CYCLOHEPTYL-
N
6 N6-DIMETHYL-4,6-PYRIMIDINEDIAMINE: Prepared by Procedures P (140 toluene, 6 Q, and A. 'H NMR (400 MHz, CDC1 3 8 7.20 7.09 5H), 4.78 1H), 4.18 (br s, 1H), 3.74 4H, J 3.52 2H), 2.99 (s, 00 407 6H) 2.46 t, 4H, J 5.1) 2.03 -1.92 Cm, 2H) 1. 87 1. 68 1 1H) ES I -MS m/z 4 09 (MH+) Example 129: [2-(4-BENZYL-1-PIPERAZINYL)-6- 00 5 (DIMETHYLANINO) -4-PYRIMIDINYL]AMINO)-2- CHLOROBENZONITRILE: Prepared by Procedures P (toluene, 00 0 C, 16 h) Q (dioxane, 120 OC) and A. 1H NMR (400 MHz, CDCl 3 8 7.88 1H, J 7.48 Cd, 1H, LT 7.42 7.22 (in, 6H) 6.45 1H) 5.20 1W), 3.79 t, 4 H, J 3. 55 Cs, 2 H) 3. 02 6 H) 2. 51 4 H, LJ ESI-MS m/z 448 (MH 4 with 3 1Cl) 450 (MW+ with 7Ci) Example 130: 2-(4-BENZYL-1-PIPERAZINYL)-N 4 1 N"'-DIMETHYL-R6- (1,3,3-TRIMETHYLBICYCLO[2.2.1IHEPT-2-YL)-4,6- PYRIMIDINEDIA4INE: Prepared by procedures P (toluene, OC, 16 h) 0 Cdioxane, 120 0 C) and A. 'H NMR C400 MHz, CDC1 3 87. 38 7.22. C m, 6H) 4. 87 Cs, 1H) 3. 79 3 .6 9 Cm, 4H), 3.53 Cs, 2H) 3.46 Cs, 1H) 2.98 Cs, 6H) 2.46 4H, J 5. 1) 1. 71 Cs, 1W) 1. 69 1. 62 Cm, 2H) 1.4 8 1.35 Cm, 2H), 1.20 Cd, 1H, LTJ 10.2), 1.19 1.02 Cm, 1H), 1.14 Cs, 3H), 1.07 Cs, 3H), 0.79 3H); ESI-MS m/z 449 (MHW).
408 Example 131: 2-(4-[3-(BENZYLOXY)PHENYL]-1-PIPERAZINYL)- N4,N4-DIMETHYL-16- (4-METHYLPHENYL) -4,6-PYRIMIDINEDIAMINE: Prepared by Procedures A (CH 2 C12, TEA, 3 4 h at -78 OC, then 3 4 h at 0 OC), N, and 0. lH NMR (400 MHz, CDC1 3 8 7.44 2H, J 7.36 2H, J 7.29 (d, 1H, J 7.22 7.04 5H), 6.58 6.52 2H), 6.48 1H, J 5.29 1H), 5.21 1H), 5.03 2H), 3.89 3.80 4H), 3.28 3.15 4H), 3.00 6H), 2.30 3H); ESI-MS m/z 495 Example 132: N 4 1 N-DIMETHYL-2-[4-(2-PYRIDINYL)-1- PIPERAZINYL] (3-QUINOLINYL) 6-PYRIMIDINEDIAMINE: Prepared by Procedures A (CH 2 C1 2 TEA, 3 4 h at -78 OC, then 3 4 h at 0 OC), N, and 0. 'H NMR (400 MHz, CDC1 3 8 8.93 1H, J 8.31 1H, J 8.26 8.18 1H), 8.02 1H, J 7.71 1H, J 7.7), 7.57 (dt, 1H, J 1.5, 7.53 7.46 2H), 6.68 1H, J 6.64 (dd, 1H, J 4.9, 5.30 (d, 2H, J 3.94 4H, J 3.64 4H, J 3.03 6H); ESI-MS m/z 427 (Mli).
Example 133: Zif-[4-BROMO-3-(TRIFLUOROMETHYL)PHENYL-IG,N 6 DIMETHYL-2-[4-(2-PYRIDINYL)-1-PIPERAZINYL]-4,6- 00 409 PYRIMIDINEDIAMINE: Prepared by Procedures A (CH 2 C1 2
TEA,
3 4 h at -78 OC, then 3 4 h at 0 N, and 0. 1H NMR Cr (400 MHz, CDC1 3 8 8.23 8.19 1H), 8.17 1H, J 7.57 1H, J 7.53 7.47 1H), 7.39 00 00 5 1H, J 6.69 1H, J 6.64 1H, J 6.27 1H), 5.19 1H), 3.94 3.87 4H), 00 3.65 3.59 4H), 3.04 6H); ESI-MS m/z 522 (MH' A with "Br) 524 (MH 4 with 1 Br) Example 134: 3-CHLORO-4- [(TRIFLUOROMETHYL) SULFANYL PHENYL)-eN,bN-DIMETHYL-2- (2- PYRIDINYL)-1-PIPERAZINYL] -4,6-PYRIMIDINEDIAMINE: Prepared by Procedures A (CH 2 C1 2 TEA, 3 4 h at -78 OC, then 3 4 h at 0 OC), N, and 0. 1H NMR (400 MHz, CDC1 3 8 8.23 8.19 1H), 7.91 1H, J 7.61 1H, J 7.50 (dt, 1H, J 2.1, 7.30 7.20 1H), 6.70 1H, J= 6.64 (dd, 1H, J 4.7, 6.35 (br s, 1H), 5.26 1H), 3.92 4H, J 3.64 (t, 4H, J 3.06 6H); ESI-MS m/z 510 (MH+ with 3 sC1) 512 (MH+ with 37 C1) Example 135: N'-(3-ETHOXYPHENYL)-Ns 6 ,N-DIMETHYL-2- (2- PYRIDINYL)-1-PIPERAZINYL]-4,6-PYRIMIDINEDIAMINE: Prepared by Procedures A (CH 2 C1 2 TEA, 3 4 h at -78 oC, then 3 00 410 4 h at 0 oC), N, and 0. -1H NMR (400 MHz, CDC1 3 5 8.28 8.19 1H), 7.50 (dt, 1H, J 2.1, 7.19 1H, J 6.96 1H, J 6.85 1H, J 8.2), 6.68 1H, J 6.63 6.56 1H), 6.35 (br s, 00 m 5 1H), 5.36 1H), 4.09 3.98 2H), 3.91 4H, J 3.61 4H, J 3.02 6H), 1.39 3H, J 00 ESI-MS m/z 420 (MH).
Example 136: N4-[2-CHLORO-4-(TRIFLUOROMETHYL)PHENYL]- 16,IA-DIMETHYL-2-[4-(2-PYRIDINYL)-1-PIPERAZINYL]-4,6- PYRIMIDINEDIAMINE: Prepared by Procedures A (C 2 C12, TEA, 3 4 h at -78 OC, then 3 4 h at 0 OC), N, and 0.
1 H NMR (400 MHz, CDC1 3 8 8.23 8.15 1H), 8.15 (d, 1H, J 7.50 (dt, 1H, J 2.0, 7.42 7.33 2H), 6.69 1H, J 6.64 (dd, 1H, J 4.8, 6.28 1H), 5.18 1H), 3.91 4H, J 3.62 4H, J 3.04 6H); ESI-MS m/z 478 (MH+ with 3 sC1) 480 (MH+ with 37 Cl) Example 137: N-4-(2-ADAMANTYL)-2-(4-BENZYL-1- PIPERAZINYL)-N-6-N-6-DIMETHYL-4,6-PYRIMIDINEDIAMINE: Prepared by Procedures P (toluene, 90 Q, and A. 'H NMR (400 MHz, CDC1 3 8 7.39 7.21 5H), 4.83 1H), 4.72 (br s, 1H), 3.74 3H), 3.52 2H), 2.98 (s, 00 411 6H), 2.46 4H, J 2.05 1.53 Cm, 13H); ESI-MS m/z: 433 Example 138: N-4-(1-NORADAMANTYL)-2- (4-BENZYL-1- 00 5 PIPERAZINYL) -N-6-N-6-DIMETHYL-4, 6-PYRIMIDINEDIAMINE: Prepared by Procedures P (toluene, 90 OC), Q, and A. 1H 00 NMR (400 MHz, CDC1 3 8 7.38 7.20 Cm, 5H), 4.97 1H), 4.67 (br s, 1H), 3.74 4H), 3.52 2H), 2.99 (s, 6H), 2.46 4H, J 2.32 1.51 Cm, 15H); ESI-MS m/z: 447 CMH+).
Example 139: 2-(4-BENZYL-1-PIPERAZINYL)- N' N-DIMETHYL-ZV6- [(1S,2R,3R,5S)-2,6,6-TRIMETHYLBICYCLO[3.1.1)HEPT-3-YL)- 4,6-PYRIMIDINEDIAMINE: Prepared by Procedures P (toluene, 150 OC, 4 Q (neat, 130 OC), and A. 'H NMR (400 MHz, CDC1 3 8 7.38 7.21 5H), 4.86 1H), 4.35 (br a, 1H), 3.75 4H, J 3.53 2H), 2.99 6H), 2.66 2.56 Cm, 1H), 2.47 4H, J 2.41 2.33 1H), 1.98 1.92 1H), 1.83 1H, J 1.68 1.60 Cm, 2H), 1.23 3H), 1.14 3H, J 1.05 3H), 0.92 2H); _ESI-MS m/z: 449 CMH+).
00 412 Example 140: 2- [4-(5-BROMO-2-PYRIDINYL)--PIPERAZINYL]- AZ, 4 -DIMETHYL-If (4-METHYLPHENYL) 6-PYRIMIDINEDIANINE: c- Prepared using Procedure Y (DMF). 'H NNR (400 MHz, CDCl 3 8 8.21 1H, J 7.53 (dd, 1H, J 2.6, 8.8), 00 00 5 7.19 2H, J 7.12 d, 2H, JL 6.21 s, 1H), 5.28 1H), 3.88 Ct, 4H, J 3.58 4H, J 00 0 3.00 Cs, 6H), 2.33 3H); ESI-MS mhz: 468 MH* with 470 (MH+ with 81 Br) Example 141: 6-{4-[4-(DIMETHYLAMINO)-6-(4-TOLUDINO)-2- PYRIMIDINYL-1-PIPERAZINYL)NICOTINAIDE: Prepared by Procedure Y CDMF). 'H NMR (400 MHz, CDC1 3 8 8.13 1H), 7.30 7.25 4H), 7.17 2H, J 7.13 Cd, 2H, J 6.18 (br a, 1H), 5.28 1H), 3.82 Ct, 2H, J 3.79 2H, J 3.60 Ct, 2H, J 3.41 2H, J 2.99 6H) 2.33 Cs, 3H); ESI-MS r/z: 433 (MH+) Example 142: 2-[4-(3-METHOXYBENZYL) -1-PIPERAZINYL) -N 4
,NI
4
DIMETHYLN
6 C4-METhYLPHENYL) -4,6-PYRIMIDINEDIAMINE: Prepared by Procedure Z (DIEA) 'H NMR (400 MHz, CDC1 3
B
7.22 Cd, 1H, J 7.17 Cd, 2H, J 7.10 (d, 00 413 2H, J 6.93 1H, LT 6.92 1H, L 2. 6. 80 (dd, 1H, LT 2. 0, 7. 6) 6. 18 (br s, 1H) 5.2 (K s, 1H) 3 .82 3H) 3. 78 4H, J. 5. 1) 3.52 (s, 2H) 2.97 6H) 2.49 4H, LT 5. 2.31. 3H); 005 ESI-MS m/z: 433 (MHi+) 00 Example 143: 2- [4-(5-BROMO-2-PYRIDINYL)-1-PIPERAZINYL]
-N
4 (3-METHOXYPHENYL) -N6,N4-DIMETHYL-4, 6-PYRIMIDINqEDIANINE: Prepared by Procedure Y. 1H NMR (400. MHz, CDC1 3 8 8.21 1H, J 7.53 (dd, 1H, LJ 2.5, 7.20 Ct, 1H, LT 8. 1) 7. 00 1H, LT 6.85 (dd, 1H, JL= 6.62 6.54 (mn, 2H) 6.29 1H) .5.36 (s, 1H) 3. 89 4H, LJ 5. 1) 3.80 Cs, 3H) 3.58 4H, LT 3.02 Cs, 6H); ESI-MS m/z: 484 (MH+ with 79Br) 486 (MH+ with 8 1 Br) Example 144: tl4-(3-METHOXYPHENYL)-6N 6 -NDIMFTHYL-2-[4-(2- PYRIDINYLMETHYL) -1-PIPERAZINYL) 6-PYRIMIDINEDIAMINE: Prepared by Procedure X. 1 H NMR (400 M4Hz, CDC+1 3 8 8.61 8. 54 Cm, 1H) 7. 66 (dt, 1H, LJ 1. 8, 7. 8) 7. 45 1H, LT 7.23 7.14 (in, 2H) 7.00 1H, LT 6.87 -6.78 Cm, 1H) 6.61 6.54 1H) 6.26 Cbr s, 1H) 00 414
C<-
5.33 1H), 3.82 4H, J 3.78 3H), 3.70 2H), 2.99 6H), 2.56 4H, J ESI-MS m/z: C1 420 00 Example 145: 2-[4-(CYCLOHEXYLMETHYL)-1-PIPERAZINYL] -N 4 00 METHOXYPHENYL) N 6 f -DIMETHYL-4, 6-PYRIMIDINEDIAMINE: Prepared by Procedure T. 'H NMR (400 MHz, .CDC1 3 8 7.21 1H, J 7.00 6.95 1H), 6.85 1H, J 6.59 1H, J 6.32 1H), 5.36 1H), 3.82 3.71 4H), 3.79 3H), 3.69 3.62 2H), 3.58 3.50 2H), 3.01 6H), 2.54 2.45 1H), 1.87 1.48 8H), 1.45 1.29 4H); ESI-MS m/z: 425 Example 146: 0- (3-METHOXYPHENYL) -N6,l-DIMETHYL-2-[4-(3- THIENYLMETHYL)-1-PIPERAZINYL] -4,6-PYRIMIDINEDIAMINE: Prepared by Procedures T (reduction 4 h) and W. 'H NMR (400 MHz, CDC1 3 8 7.27 (dd, 1H, J 3.2, 7.19 (t, 1H, J 7.16 7.11 1H), 7.08 (dd, 1H, J 1.3, 7.00 1H, J 6.82 (dd, 1H, J 2.0, 8.3), 6.57 (dd, 1H, J 2.5, 6.25 1H), 5.33 1H), 00 415 3.79 4H, LJ 3 .78 3H), 3.57 2H), 2.99 6H) 2.48 4H, J 5.2) ESI-MS mhz: 425 00 Example 147: N 4 (3-METHOXYPHENYL) -N,N6-DIMETHYL-2- C4- (4- 00 PYRIDINYLMETHYL) -1-PIPERAZINYLI -4,6-PYRIMIDINEDIAMINE: prared by Procedure T (acylation with DIE) 1
M
(400 MHz, CDCl 3 8 8. 55 (dd, 2H, LT 1. 5, 7.31 (d, 2H, LT 7.19 1H, LT 6.99 Ct, 1 H, J 6.83 (dd, 1H, LTJ 1.5, 6.58 (dd, 1H, J 2. 0, 6.28 (br s, 1H) 5.34 1H) 3. 80 4H, LTJ 3.78 3.54 2H), 3.00 6H), 2.49 Ct, 4H, J ESI-MS m/z: 420 Example 148: 2-[4-(3-METHOXYBEN'ZYL)-1-PIPERAZINYL]-N4-C3- METHOXYPHENYL) -1N4, N -DIMETHYL- 4, 6- PYRIMIDINEDIAMINE: Prepared by Procedure S. lH NMR (400 MHz, CDC1 3 8 7,22 1H, LT 7.17 Ct, 1H, JL= 6.99 Ct, 1H, LJ 2. 6. 95 6. 84 Cm, 2H) 6. 86 78 Cm, 2H) 6. 59 6.55 Cm, 1H), 6.29 Cbr s, 1H) 5.32 Cs, 1H) 3.82 (s, 3H) 3.79 4H, JT 3.77 3H) 3.52 Cs, 2H) 00 416 2. 99 6H), 2.49 4H, J ESI-MS MhZ: 449 00 Example 149: N2- (3-METHOXYPHENYL) ETHYL) -N,N-DIMETHYLiVt- (4-METHYLPHENYL) -2,4,6-PYRIMIDINETRIAMINE: 00 Prepared by Procedure F (dioxane, potassium tertbutoxide, 120 0 C, 16 h) Q (toluene, TEA, 120 OC) A
(CH
2 C1 2 A, TEA) 1H NNR (400 MHz, CDCl 3 8 7.22 1H, J 7.18 2H, J 7.12 2H, J 8.3), 6. 84 1N, J 7.6) 6.82 6.74 (in, 2H) 6.28 (br s, 1N) 5.28 1H) 4.77 1H) 3.80 3H) 3.63 (q, 2H, J 7) 2. 99 6H) 2. 89 2H, J 7. 4) 2.3 2 3H) ;ESI-MS m/z: 378 Example 150: bP- (2-METHOXYPHENYL) ETHYL) -N 4 Nf- (4 -METHYLPHENYL) 6-PYRIMIDINETRIAMINE: Prepared by Procedures F (dioxane, potassium ter'tbutoxide, 140 OC, 16 h) Q (toluene), and A (CH 2 C1 2
A,
TEA) 1H NMR (400 MHz, CDC1 3 8 7.23 7.12 4H) 7.12 2H, J 8.1) 6.89 1N, J 6.86 1H, J 7. 6. 61 1N, LT 8 6. 50 (br s, 1N) 5. 25 (s, 00 417 1H), 3.84 3H), 3.60 2H, LT 3.00 6H), 2.93 Ct, 2H, J 2.33 3H) ESI-MS m/z: 378 00 Example 151: 2- (4-BENZYL-1-PIPERAZINYL) 4- 00 DICHLOROPHENYL) -N6,1 -DIMETHYL- 4, 6 PYRIMIDINEDIANINE: Prepared by Procedures P (toluene, 140 OC, 6 hQ (dioxane, 120 OC) and A. NMR (400 MHz, CDCl 3 87.65 1H, J 7.35 7.30 Cm, 4H) 7.29 7.22 Cm, 2H) 7.13 Cdd, 1H, JT 1. 5, 8.5) 6.19 (br s, 1H) 5. 21 1H) 3.78 Ct, 4H, J 3.55 Cs, 2H) 3.00 Cs, 6H) 2.49 t, 4H, J 5.0) ESI-MS m/z: 457 (MH* with 35 C1, 35 C1) 459 (MH+ with 3 5 C1 37 C1) 461 (MH+ with 37Cl, 37cl) Example 152: PJ (BENZYLOXY)CYCLOHEXYLJ (4-BENZYL-1- PIPERAZINYL) -IV, -DIMETHYL- 4, 6 -PYRIMIDINEDIAMINE: Prepared by Procedures P (16 Q, and A. 1H NMR (400 MHz, CDC1 3 8 7.42 7.18 Cm, 10H) 4.94 1H), 4.61 (d, 1H, LT 11. 8) 4.51 Cd, 1H, J. 11. 8) 4.3 9 Cbr s, 1H) 3.75 4H, LT 3.53 2H) 3.31 Cdt, 1H, J= 5.3, 2.95 Cs, 6H), 2.46 t, 4H, J 2.19- 00 418 2. 11 1H), 2. 07 98 (in, 1H1), 1. 79 1.56 (mn, 3H), 1. 53 -1.41 (in, 1H), 1. 40 -1.21 3H) ESI-MS m/z: 501 CI (MH4).
00 Excample 153: 2- (4-BENZYL-1-PIPERAZI1NYL)-N 4 1
N
4 DIMETHYL-bf- 00 [(1R,2R,4R)-1,7,7-TRIMETHYLBICYCLO[2.2.1]HEPT-2-YL]-4,6- PYRIMIDINEDIAMINE: Prepared by Procedures P (90 OC, 16 h) Q, and A. 1 H NMR (400 MHz, CDC1 3 8 7.4 4 7.22 (m, 6H) 4.81 Cs, 1H) 4.36 Cd, 1H, J 3.74 4H) 3.53 .2H) 2.98 6H) 2.46 Ct, 4H, J 1.84 (dd, 1H, LT 8. 9, 12.9), 1.78 1.52 Cm, 4H) 1. 29 1. 11 Cm, 2H), 0.97 3H), 0.89 3H), 0.83 CB, 3H); ESI-MS m/z: 449 CMH*) Example 154: N 4 ,.NO-DIMETHYL-bfl-(4-METHYLPHENYL) [4- (TETRAHYDRO-2 -FURANYLMETHYL) -1-PIPERAZINYL] -4,6- PYRIMIDINEDIAMINE: Prepared by Procedures A, P (16 h) and Q (dioxane, 120 OC). 'H NMR (400 MHz, CDC1 3 8 7.17 Cd, 2H, LT 8. 4) 7. 11 2 H, LT 8 6. 22 Cbr s, 1H) 5.29 Cs, 1H), 4.12 4.03 (in, 1H), 3.91 1H, J 6.7), 3.80 4H, LT 3.76 1H, LT= 2.98 Cs, 6H) 2.57 Ct, 4H, J 2.56 2.40 C,2H) 2.32 (s, 00 419 3H), 2.05 1.96 1H), 1.94 1.80 2H), 1.57 1.45 1H); ESI-MS m/z: 397 Example 155: 3-([2-(4-BENZYL-1-PIPERAZINYL)-6- 00 (DIMETHYLAMINO)-4-PYRIMIDINYL)AMINOIPHENOL: Prepared By Procedures P (Toluene, 120 OC, 40 Q (dioxane, 120 00 OC), AND A. -H NMR (400 MHz, CDC1 3 6 7.38 7.29 4H), 7.28 7.26 1H), 7.13 1H, J 6.84 1H, J 6.80 (ddd, 1H, J 0.7, 2.0, 6.48 (ddd, 1H, J 0.7, 2.1, 6.32 (br a, 1H), 5.32 1H), 3.79 4H, J 3.55 2H), 3.49 1H), 2.99 6H) 2.50 4H, J 5.0) ESI-MS m/z: 405 Example 156: 2- (4-BENZYL-1-PIPERAZINYL) -n0- (4- FLUOROPHENYL) -N6,bN-DIMETHYL-4, 6-PYRIMIDINEDIAMINE: Prepared by Procedures P (toluene, sodium tert-butoxide, 120 OC, 16 Q (dioxane, 120 OC) and A. 'H NNR (400 MHz, CDC1 3 6 7.37 7.30 Cm, 4H), 7.29 7.21 3H), 6.99 2H, -J 6.14 (br a, 1H), 5.13 11), 3.77 4H, J 3.54 2H), 2.97 6H), 2.48 4H, J ESI-MS m/z: 407 Example 157: 2- (4-BENZYL-1-PIPERAZINYL) -N4,N-DIMETHYL-N6- (4-METHYLCYCLOHEXYL) -4,6-PYRIMIDINEDIAMINE: Prepared by 00 420 Procedures P (sodium tert-butoxide, toluene, 120 16 Q (dioxane, 120 oC), and A. 'H NMR (400 MHz, CDC1 3 6 C' 7.35 7.10 6H), 4.82 1H, J 3.81 3.61 5H) 3.53 2H), 2.99 6H), 2.46 4H, J 00 5 1.79 1.46 7H), 1.29 0.98 2H), 0.90 (d, 0 3H, J ESI-MS m/z: 409 00 Example 158: 2- (4-BENZYL-1-PIPERAZINYL) -N 4 [4- (DIMETHYLAMINO) PHENYL] -I i -DIMETHYL-4,6- PYRIMIDINEDIAMINE: Prepared by Procedures P (sodium tertbutoxide, toluene, 120 OC, 16 Q (neat, 130 oC), and A. 1H NMR (400 MHz, CDC1 3 8 7.39 7.22 5H), 7.14 (d, 2H, J 6.71 2H, J 6.04 (br s, 1H), 5.08 1H), 3.85 3.74 4H), 3.54 2H), 2.94 (s, 6H), 2.93 6H), 2.48 4H, J ESI-MS m/z: 432 Example 159: N 4
N-DIMETHYL-A
6 (4-METHYLPHENYL) (2- PHENYLETHYL)-1-PIPERAZINYL]-4,6-PYRIMIDINEDIAMINE: Prepared by Procedure S (toluene, 120 OC). 'H NMR (400 MHz, CDC1 3 8 7.34 7.20 5H), 7.18 2H, J 7.12 2H, J 6.21 (br s, 1H), 5.26 1H), 00 421 00 3.88 3.79 4H), 2.99 6H), 2.90 2.83 2H), 2.68 2.63 2H), 2.60 4H, J 2.32 3H); CI ESI-MS m/z: 417 (MH 00 Example 160: 2- (4-BENZYL-1-PIPERAZINYL) (3- 00 CHLOROPHENYL) -Zf,N -DIMETHYL-4,6-PYRIMIDINEDIAMINE: SPrepared by Procedures P (toluene, sodium tert-butoxide, 120 OC, 40 Q (dioxane, 120 oC), and A. 'H NMR (400 MHz, CDC1 3 6 7.48 1H, J 7.38 7.23 7.20 7.11 2H), 6.95 (ddd, 1H, J 1.2, 1.9, 7.6), 6.28 (br s, 1H), 5.24 1H), 3.79 4H, J 3.54 2H), 3.00 6H), 2.49 4H, J ESI-MS m/z: 423 (MH* with 3 C1) 425 (MH* with "Cl).
Example 161: N N 4
-TRIMETHYL-N
6 (4-METHYLPHENYL) -N 1 [2- (2-PYRIDINYL) ETHYL] -2,4,6-PYRIMIDINETRIAMINE: Prepared by Procedures F (dioxane, potassium tert-butoxide, 140 OC, 16 Q, and A (CH 2 C1 2 A, TEA). 1H NMR (400 MHz, CDC1 3 8 8.54 (ddd, 1H, J 1.2, 2.1, 7.57 (dt, 1H, J 1.7, 7.23 2H, J 7.18 1H, J 7.14 7.09 1H), 7.10 2H, J 6.29 (br s, 1H), 5.24 1H), 3.93 (dd, 2H, J 5.9, 3.11 (dd, 2H, 00 422 J 6.0, 3.08 3H), 3.00 6H), 2.32 3H); ESI-MS m/z: 363 (MH).
00 Example 162: A, -DIMETHYL- (4-METHYLPHENYL) -N2- (3- Cr PHENYLPROPYL)-2,4,6-PYRIMIDINETRIAMINE: Prepared using 00 Procedures R, S, and V. IH NMR (400 MHz, CDC1 3 6 7.25 2H, J 7.22 7.14 5H), 7.11 2H, J 6.41 (br s, 1H), 5.27 1H), 4.76 1H, J 3.41 (dd, 2H, J 7.0, 12.9), 2.96 6H), 2.70 2H, J 2.31 3H), 1.91 2H, J ESI-MS m/z: 362 Example 163: 2-(4-CYCLOHEXYL- -PIPERAZINYL) (3- METHOXYPHENYL) JN-DIMETHYL-4, 6-PYRIMIDINEDIAMINE: Prepared using Procedures P (16 Q (dioxane, 120 OC), and A. 1 H NMR (400 MHz, CDC13) 6 7.11 1H, J 8.3), 6.92 1H, J 6.78 6.73 1H), 6.53 6.48 1H), 6.39 (br s, 1H), 5.27 1H), 3.72 4H, J 3.71 3H), 2.92 6H), 2.55 4H, J 5.1), 2.28 2.18 1H), 1.87 1.79 2H), 1.77 1.68 (m, 2H), 1.56 1H, J 12.4), 1.24 1.08 4H), 1.08 0.97 1H); ESI-MS m/z: 411 (MH).
00 423 Example 164: 2- (4-BENZYL-1-PIPERAZINYL) FLUOROPHENYL) -1t, 1V-DIMETHYL-4,6-PYRIMIDINEDIAMINE: 00 Prepared by Procedures P (140 OC, 4 Q (neat, 130 OC), 00 and A. 'H NMR (400 MHz, CDC1 3 8 7.37 7.31 5H) 7.28 7.17 Cm, 2H), 6.98 (ddd, 1H, J 0.7, 2.0, 6.67 00 (ddt, 1H, J 0.9, 2.0, 6.30 (br a, 1H), 5.27 (s, 1H), 3.79 4H, J= 3.55 2H) 3.00 6H), 2.50 4H, J ESI-MS m/z: 407 (MH+) Example 165: A -(3-METHOXYPHENYL)-A, N6-DIMETHYL-2-4-(2- THIENYLMETHYL)-1-PIPERAZINYL-4,6-PYRIMIDINEDIAMINE: Prepared by Procedure T. 'H NMR (400 MHz, CDC1 3 8 7.24 Cdd, 1H, J 1.2, 7.19 Ct, 1H, J 6.99 (t, 1H, J 6.96 6.91 2H), 6.83 Cddd, 1H, J 0.8, 1.7, 6.57 (dd, 1H, J 2.0, 6.25 (br s, 1H), 5.33 1H), 3.81 Ct, 4H, J= 3.78 3H), 3.76 Cs, 2H), 2.99 6H), 2.53 4H, J ESI-MS m/z: 425 (MH)) Example 166: 2- (2-METHOXYBENZYL) -1-PIPERAZINYL]-N 4 (3- METHOXYPHENYL) -N6,N 6 -DIMETHYL-4,6-PYRIMIDINEDIAMINE: 00 424 SPrepared by Procedure T (reduction 3 H NMR (400 MHz, CDC1 3 8 7.40 (dd, 1H, J 1.6, 7.23 (dd, 1H, J 1.2, 7.19 1H, J 7.01 1H, J 1.9), O 6.95 (dt, 1H, J 1.0, 6.87 (dd, 1H, J 1.1, 8.3), 00 O 5 6.82 (ddd, 1H, J 1.0, 2.0, 6.57 (ddd, 1H, J 00 C, 0.7, 2.5, 6.26 (br s, 1H), 5.32 1H) 3.82 (s, O 3H), 3.81 4H, J 3.78 3H), 3.62 2H), 2.99 6H), 2.55 4H, J ESI-MS m/z: 449 (MH) Example 167: 2-(4-BENZYL-1-PIPERAZINYL) ~-DIMETHYL-Nf- [(1R, 2 S)-1,7,7-TRIMETHYLBICYCLO[2.2.1]HEPT-2-YL]-4,6- PYRIMIDINEDIAMINE: Prepared by Procedures P (toluene, 120 oC, 16 Q (neat, 130 oC), and A. 'H NMR (400 MHz, CDC1 3 8 7.37 7.22 5H), 4.82 1H), 4.51 (br s, 1H), 3.74 4H), 3.53 2H), 2.97 6H), 2.47 (t, 4H, J 2.39 2.30 1H), 1.76 1.68 4H), 1.66 1H, J 1.41 1.31 2H), 0.96 3H), 0.88 3H), 0.86 3H); ESI-MS m/z: 449 Example 168: N4-(2-ADAMANTYL)-2-(4-BENZYL-1-PIPERAZINYL) N6,N6-DIMETHYL-4,6-PYRIMIDINEDIAMINE: Prepared by 00 425 Procedures P (90 OC, toluene), Q, and A. H NMR (400 MHz, CDC1 3 6 7.39 7.21 5H), 4.83 1H), 4.72 (br s, N 1H), 3.74 5H), 3.52 2H), 2.98 6H), 2.46 (t, o 4H, J 2.05 1.53 14H); ESI-MS m/z: 447 (MH) 00 rc 0 0 Example 169: 2- (4-BENZYL-1-PIPERAZINYL) (4-TERT- C( BUTYLCYCLOHEXYL) -N 6 6 -DIMETHYL-4,6-PYRIMIDINEDIAMINE: Prepared by Procedures P (toluene, 16 Q (neat, 130 oC), and A. 'H NMR (400 MHz, CDC1 3 8 7.36 7.22 4.82 1H), 3.74 4H, J 3.53 2H), 3.33 1H), 2.98 6H), 2.46 4H, J 1.15 0.91 9H), 0.86 9H); ESI-MS m/z: 451 (MH Example 170: 2- (4-BENZYL-1-PIPERAZINYL)
-IN-CYCLOOCTYL-
N6 -DIMETHYL-4,6-PYRIMIDINEDIAMINE: Prepared by Procedures P (16 Q, and A. 'H NMR (400 MHz, CDC1 3 6 7.39 7.21 5H), 4.79 1H), 4.34 1H), 3.74 (t, 4H, J 3.53 2H), 2.99 6H), 2.40 4H, J 1.93 1.49 15H); ESI-MS m/z: 423 00 426 Example 171: 2- (4-BENZYL-1-PIPERAZINYL)
-N
4 (4cHLOROPHENYL)
N
6 N-DIMETHYL-4, 6-PYRIMIDINEDIAMINE: Prepared by Procedures P (140 OC, Q (neat, 130 DC), and A. 1 H NMR (400 MHz, CDC1 3 8 7.38 7.22 9H), 6.31 (br 00 s, 1H), 5.21 1H), 3.78 4H, J 5.1 Hz), 3.55 (s, 2H), 2.99 6H), 2.49 4H, J ESI-MS m/z: 423 00 (MH+ with 3 5 C1) 425 (MHW with 3C) Example 172: 2-(4-BENZYL1PIPERAZINYL)
-N
4 (3-CHLORO-4- METHYLPHENYL) -N6, N6-DIMETHYL-4, 6-PYRIMIDINEDIAMINE: Prepared by Procedures P (toluene, 120 OC, 16 Q (neat, 130 OC), and A. 1H NNR (400 MHz, CDC1 3 8 7.43 1H, J 7.38 7.09 5H), 7.07 1H, J 7.05 1H, J 6.02 1H), 5.21 1H), 3.78 4H, J 3.54 2H), 2.99 6H), 2.49 4H, J 2.31 3H); ESI-MS m/z: 437 (MH+ with 35 C1) 439 (MH* with 37Cl) Example 173: 2-(4-BENZYL-1-PIPERAZINYL)N4,N-DIMETHYL- (l, 2 ,3,4-TETRAHYDRO-2-NAPHTHALENYL)-4,6- PYRIMIDINEDIAMINE: Prepared by Procedures p (16 0, and A. 'H NMR (400 MHz, CDC1 3 6 7.41 7.04 9H), 4.99 00 427 1H), 4.91 1H), 3.74 4H), 3.53 2H), 3.47 1H), 2. 99 sH), 2.90 2.69 2H), 2.49 (m, 4H), 2.09 1.71 4H); ESI-MS m/z: 443 00 Example 174: AN-DIMETHYL- (4-METHYLPHENYL) (2- 00
RMDNDAIE
O THIENYLMETHYL)-1-PIPERAZINYL)-4,6-PYRIMIDINEDIAMINE: C Prepared by Procedure X (NaBH(OAc)3,
CH
2 C1 2 molecular sieves). 'H NMR (400 MHz, CDC1 3 8 7.17 2H, J 8.3), 7.15 7.09 2H), 7.03 6.94 3H), 5.22 (br a, 1H), 4.85 1H), 3.86 3.79 4H), 3.77 2H), 2.98 6H), 2.62 2.53 4H), 2.32 3H); ESI-MS m/z: 409 (MH).
Example 175: 2-[4-(2-METHOXYBENZYL)-1PIPERAZINYLl DIMETHYL-N6- (4-METHYLPHENYL)-4 6PYRIMIDINEDIAINE: Prepared by Procedure Z. 'H NMR (400 MHz, CDC1 3 6 7.40 (dd, 1H, J 1.6, 7.23 (dt, 1H, J 1.4, 7.17 2H, J 7.10 2H, J 6.94 1H, J 6.87 1H, J 6.17 (br a, 1H), 5.24 (s, 1H), 3.82 3H), 3.79 4H, J 3.62 2H), 2.97 6H), 2.55 4H, J 2.31 3H); ESI-MS m/z: 433 (MH).
00 428 Example 176: t32- (2-ANILINOETHYL) N1 4 ,N-DIMETHYL-Nf-(4- METHYLPHENYL)-2,4, 6-PYRIMIDINETRIAMINE: Prepared by 00 Procedures A, Q (toluene, 100 aC), and F (potassium tert- 00 butoxide, 110 OC, 16 h) 'H NMR (400 MHz, CDC 3 8 7.19 00 7.10 (in, 6H), 6.67 (dt, 1H, J 0.8, 6.59 (dd, 2H, J 0.8, 6.31 (br s, 1H), 5.28 1H), 4.99 (s, 3.66 2H, J 3.49 3.37 2H, J 3.00 6H), 2.33 3H); ESI-MS m/z: 363 Example 177: N4 (3-METHOXYPHENYL) ,1fN6,N-TRIMETHYLN 2 [2- (2-PYRIDINYL) ETHYL] -2,4,6-PYRIMIDINETRIAMINE: Prepared by Procedures F (dioxane, 140 OC, 15 A (CH 2 C1 2 A, TEA), and Q (toluene, TEA, A, 40 'H NMR (400 MHz, CDC1 3
S
8.55 1H, J 7.58 1H, J 7.25 7.16 2H), 7.15 7.06 2H), 6.89 1H, J 6.57 Cd, 1H, J 6.30 (br a, 1H), 5.31 1H), 3.95 (t, 2H, J 3.78 3H), 3.18 3.06 Cm, 5H), 3.02 6H); ESI-MS m/z: 379 Example 178: 1 (4-CLOHEXYLPHENYL)-NfD6-DIMETHYL-2- 4- (2-PYRAZINYL)-1-PIPERAZINYLI-4,6-PYRIMIDINEDIAMINE: 00 429 SPrepared by Procedures A (CH 2 C12, Et 3 N, Me 2 NHHC1, -78 °C for 3.5 h, warmed from -78 OC to 0 OC and stirred for 3 C N, and 0. H NMR (400 MHz, CDC1 3 8 9.90 (br s, 1H), O 8.19-8.16 1H), 8.09-8.06 1H), 7.89-7.85 1H), 00 mC 5 7.20-7.18 4H), 5.28 1H), 3.99 4H, J 5.3), ^C 3.73 4H, J 3.04 6H), 2.53-2.44 1H), 00 0 1.91- 1.71 4H), 1.46-1.71 6H); ESI-MS m/z: 459 CNI Example 179: N-[3-(BENZYLOXY)PHENYL]-
-N
6 ,-DIMETHYL-2-[4- (2-PYRAZINYL)-1-PIPERAZINYL)-4,6-PYRIMIDINEDIAMINE: Prepared by Procedures A (CH 2 C1 2 Et 3 N, Me 2 NHHCi, -78 OC for 3.5 h, warmed from -78 oC to 0 OC and stirred for 3 N, and 0. 1 H NMR (400 MHz, CDC1 3 8 9.82 (br s, 1H), 8.17-8.15 1H), 8.09-8.06 1H), 7.89 1H, J 7.45-7.29 9H), 5.32 1H), 5.05 2H), 4.03 4H, J 3.74 4H, J 3.05 6H); ESI-MS m/z: 483 Example 180: N 4 (2,3-DIHYDRO-1H-INDEN-5-YL)
-N
6 6 DIMETHYL-2-[4-(2-PYRAZINYL)-1-PIPERAZINYL]-4,6- PYRIMIDINEDIAMINE: Prepared by Procedures A (CH 2 C1 2 Et 3
N,
00 430 Me 2 NHHCr1, -78 oC for 3.5 h, warmed from -78 OC to 0 OC and stirred for 3 N, and 0. 'H NMR (400 MHz, CDC1 3 8 10.01 (br s, 1H), 8.16 1H), 8.10-8.97 1H), 7.91- 7.87 1H), 7.19 1H, J 7.13 1H), 7.04 00 1H, J 5.23 1H), 4.03 4H, J 5.2), 3.74 4H, J 3.05 6H), 2.89 2H, J 00 2.14-2.04 4H); ESI-MS m/z: 417 Example 181: N, N-DIMETHYL-- (4-METHYLPHENYL) 2 PYRAZINYL) -1-PIPERAZINYL] -4,6-PYRIMIDINEDIAMINE: Prepared by Procedures A (CH 2 C1 2 Et 3 N, Me 2 NHHC1, -78 oC for 3.5 h, warmed from -78 OC to 0 OC and stirred for 3 N, and O. 1H NMR (400 MHz, CDC1 3 8 10.01 1H), 8.17 1H), 8.12 8.09 1H), 7.90 1H, J 7.18 2H, J 7.16 2H, J 5.19 1H), 4.18 4.02 4H), 3.77 4H, J 3.20 (br s, 3H), 2.99 (br a, 3H), 2.35 3H); ESI-MS m/z: 391 Example 183: N- 4-DIMETHYLPHENYL) -DIMETHYL-2-[4- (2-PYRAZINYL)-1-PIPERAZINYL] -4,6-PYRIMIDINEDIAMINE: Prepared by Procedures A (CH 2 C1 2 Et 3 N, Me 2 NHHC1, -78 OC for 3.5 h, warmed from -78 OC to 0 OC and stirred for 3 00 431 N, and 0. H NMR (400 MHz, CDC13) 8 8.75 (br s, 1H), 8.16 1H, J 8.08 (dd, 1H, J 1.5, 7.88 1H, J 7.10 1H, J= 7.08 7.00 (m, 2H), 5.26 1H), 4.00 4H, J 3.72 4H, J 00 C 5 3.03 6H), 2.24 6H); ESI-MS m/z: 405 00 SExample 184: 1- (4-BENZYL-1-PIPERAZINnL) 4 TOLUIDINO)-4-PYRIMIDINYL]-4-PIPERIDINONE: Prepared by Procedures a (Ch 2 Cl12, -78 OC, 4 N (24 and 0. 'H NMR (400 MHz, CDC13) 8 7. 38- 7.30 5H), 7,19-7,10 (m, 4H), 6.24 1H), 5.40 1H), 3.84-3.75 8H), 3.56 2H), 2.54-2.43 8H), 2.32 3H); ESI-MS m/z: 457 Example 185: ,N4-dimethyl-V- (2-propylphenyl)-2- [4- (2-pyridiyl)-i-piperazinyl-4,6-pyrimidinediaine: prepared by Procedures A (Ch 2 cl12, Tea, 3 4 H at -78 OC, then 3 4 H at 0 OC), N, and 0. 'H NMR (400 MHz, CDC1 3 8 8.22 8.18 1H), 7.56 7.40 2H), 7.25 7.07 (m, 3H), 6.75 6.60 2H), 6.04 1H), 5.04 1H), 3.91 4H), 3.62 4H), 2.96 6H), 2.60 2H, J 1.62 2H), 0.96 3H, J ESI-MS M/Z: 418 Example 186: 0- (2-BENZYLPHENYL) V-DIMETHYL-2- (2- PYRIDINYL)-1-PIPERAZINYL]-4,6-PYRIMIDINEDIAMINE: Prepared 00 432 3 by Procedures A (CH 2
CL
2 TEA, 3 4 H at -78 OC, then 3 4 H at 0 OC), N, AND 0. H NMR (400 MHZ, CDCL) 6 8.20 n 8.18 1H), 7.54 7.45 1H), 7.34 7.04 9H), 6.73 6.59 2H), 5.99 (BR S, 1H), 5.01 1H), 3.99 0 5 2H), 3.93 3.83 4H), 3.66 3.57 4H), 2.96 00 6H); ESI-MS M/Z: 466 (MH).
0 OO Example 187: N 4 (4-HEXYLPHENYL) -DIMETHYL-2- (2- <D4 PYRIDINYL)-1-PIPERAZINYL]-4,6-PYRIMIDINEDIAMINE: Prepared by Procedures A (CH 2 C1 2 TEA, 3 4 h at -78 then 3 4 h at 0 OC), N, and 0. ESI-MS m/z: 460 (MH 4 Example 188: N 4 (4-BENZYLPHENYL)
-N
6
,N
6 -DIMETHYL-2- (2- PYRIDINYL)-1-PIPERAZINYL]-4,6-PYRIMIDINEDIAMINE: Prepared by Procedures A (CH 2 Cl 2 TEA, 3 4 h at -78 OC, then 3 4 h at 0 OC), N, and 0. 1H NMR (400 MHz, CDC13) 6 8.22 8.18 1H), 7.52 7.45 1H), 7.32 7.09 9H), 6.78 1H, J 6.65 6.59 1H), 6.24 (br s, 1H), 5.29 1H), 3.96 2H), 3.91 3.83 4H), 3.63 3.55 4H), 3.00 6H); ESI-MS m/z: 466 (MH Example 189: N- (4-HEPTYLPHENYL)
-N
6 ,I-DIMETHYL-2- (2- PYRIDINYL)-1-PIPERAZINYL]-4,6-PYRIMIDINEDIAMINE: Prepared by Procedures A (CH 2 C1 2 TEA, 3 4 h at -78 OC, then 3 00 433 4 h at 0 OC), N, and 0. 1 H NMR (400 MHz, CDC13) 8 8.25 8.18 1H), 7.57 7.44 1H), 7.38 7.08 4H), 6.75 6.57 2H), 6.26 (br a, 1H), 5.29 1H), 3.95 3.85 4H), 3.71 3.56 4H), 3.00 6H), 2.57 00 S 5 2H, J 1.84 1.51 4H), 1.40 1.16 (m, 6H) 0.93 0.82 3H); ESI-MS m/z: 474 00 Example 190: N- (3,4-DIMETHYLPHENYL) -N6, N6-DIMETHYL-2- [4- (2-PYRIDINYL)-1-PIPERAZINYL]-4,6-PYRIMIDINEDIAMINE: Prepared by Procedures A (CH 2 C1 2 TEA, 3 4 h at -78 OC, then 3 4 h at 0 OC), N, and 0. 1H NMR (400 MHz, CDC1 3 8 8.25 8.19 1H), 7.55 7.44 1H), 7.31 7.23 (m, 1H), 7.14 7.02 2H), 6.73 6.59 2H), 6.18 (br a, 1H), 5.29 1H), 3.95 3.85 4H), 3.67 3.55 4H), 3.00 6H), 2.24 3H), 2.23 3H), ESI-MS m/z: 404 (MH).
Example 191: N- (3-ISOPROPYLPHENYL) -bN,N-DIMETHYL-2- [4- (2-PYRIDINYL)-1-PIPERAZINYL)-4,6-PYRIMIDINEDIAMINE: Prepared by Procedures A (CH 2 C1 2 TEA, 3 4 h at -78 OC, then 3 4 h at 0 OC), N, and 0. 1H NMR (400 MHz, CDC1 3
S
8.25 8.19 1H), 7.54 7.45 1H), 7.31 7.21 (m, 00 434 2H), 7.13 7.08 1H), 6.95 6.88 1H), 6.74 6.60 2H), 6.29 (br s, 1H), 5.37 5.34 1H), 3.96 3.87 4H), 3.68 3.57 4H), 3.00 6H), 2.95 2.85 1H), 1.36 1.19 6H); ESI-MS m/z: 418 (MH).
00
C
00Example 192: N,N-DIMETHYL-b (4-OCTYLPHENYL)-2- (2- CI PYRIDINYL)-1-PIPERAZINYL)-4,6-PYRIMIDINEDIAMINE: Prepared by Procedures A (CH 2 C1 2 TEA, 3 4 h at -78 oC, then 3 4 h at 0 oC), N, and 0. 'H NMR (400 MHz, CDC1 3 8 8.22 (s, 1H), 7.55 7.44 1H), 7.37 7.07 4H), 6.76 6.59 2H), 6.28 (br s, 1H), 5.29 1H), 3.96 3.86 4H), 3.69 3.56 4H), 3.00 6H), 2.57 2H, J 1.74 1.51 4H), 1.41 1.08 8H), 0.93 0.82 3H); ESI-MS m/z: 488 Example 193: N- (3-IODOPHENYL) N-DIMETHYL-2- (2- PYRIDINYL)-1-PIPERAZINYL] -4,6-PYRIMIDINEDIAMINE: Prepared by Procedures A (CH 2 C1 2 TEA, 3 4 h at -78 OC, then 3 4 h at 0 OC), N, and 0. 1H NMR (400 MHz, CDC1 3 8 8.29 8.18 1H), 8.01 7.93 1H), 7.56 7.45 1H), 7.39 7.29 1H), 7.11 6.95 2H), 6.78 6.56) 2H), 6.42 6.25 1H), 5.34 1H), 3.95 3.85 00 435 S(m, 4H), 3.65 3.56 4H), 3.00 6H); ESI-MS m/z: C 502 (MH 00 SExample 194: N 4 (4-CHLOROPHENYL) -N,N6-DIMETHYL-2- (2- PYRIDINYL)-1-PIPERAZINYL]-4,6-PYRIMIDINEDIAMINE: Prepared 0 0 by Procedures A (CH 2 C1 2 TEA, 3 4 h at -78 OC, then 3 OC 4 h at 0 oC), N, and 0. 1 H NMR (400 MHz, CDC13) 8 8.28 (s, 1H), 7.53 7.42 1H), 7.35 7.24 2H), 7.11 6.95 2H), 6.76 6.57 2H), 6.21 1H), 5.29 (s, 1H), 3.97 3.86 4H), 3.67 3.57 4H), 3.00 (s, 6H); ESI-MS m/z: 410. (MH).
Example 195: N6- (2-CHLOROPHENYL)
-N
4 N4-DIMETHYL-2- (2- PYRIDINYL)-1-PIPERAZINYL]-4,5-PYRIMIDINEDIAMINE: Prepared by Procedures A (CH 2 C1 2 TEA, 3 4 h at -78 OC, then 3 4 h at 0 OC), N, and 0. 1 H NMR (400 MHz, CDC13) 8 8.50 8.10 2H), 7.55 7.12 4H), 7.05 6.90 2H), 6.61 1H), 5.31 1H), 3.95-3.85 4H), 3.65 3.54 4H), 3.00 6H); ESI-MS m/z: 410 436 Example 196: N4-(3,4-DIFLUOROPHENYL)-, N6-DIMETHYL-2- (4- (2-PYRIDINYL)-1-PIPERAZINYL] -4,6-PYRIMIDINEDIANINE: Prepared by Procedures A (CH 2 C12, TEA, 3 4 h at -78 OC, 00ethen 3 4 h at 0 OC), N, and 0. 'H NMR (400 MHz, CDC1 3 8 8.31 1H), 7.59 6.95 4H), 6.68 6.54 2H), 00 6.29 1H), 5.27 1H), 3.94 3.82 4H), 3.63 3.51 4H), 3.01 6H); ESI-MS mhz: 412 Example 197: N 4 [3-METH0XY-5- (TRIFLUOROMETHYL)
PHENYL]-
D6,N6-DIMETHYL-2- (2-PYRIDINYL) -1-PIPERAZINYLI -4,6- PYRIMIDINEDIAMINE: Prepared by Procedures A (CH 2 C1 2
TEA,
3 4 h at -78 OC, then 3 4 h at 0 OC) N, and 0. 'H NMR (400 MHz, CDCl 3 8 8.26 8.18 1H), 7.58 7.11 (m, 3H) 6.77 6.38 3H), 6.34 1H), 5.25 1H), 3.96 3.88 4H), 3.85 3H), 3.69 3.55 4H), 3.00 6H); ESI-MS m/z: 474 Example 198: N 4 ,N-DIMETHYL-2- 4-(2-PYRIDINYL) -1- PIPERAZINYL -N4- (2,3,4-TRIFLJOROPHENYL) -4,6- PYRIMIDINEDIAMINE: Prepared by Procedures A (CH 2 Cl 2
TEA,
3 4 h at -78 OC, then 3 4 h at 0 OC), N, and 0. 'H NMR (400 MHz, CDC1 3 8 8.26 8.18 1H), 7.58 7.11 (m, 00 437 3H), 6.77 6.38 2H), 6.34 1H), 5.25 1H), 3.96 3.88 4H), 3.85 3H), 3.69 3.55 4H),
C
N 3.00 6H); ESI-MS m/z: 430 00 C Example 199: NA- (4-BROMO-2-FLUOROPHENYL) -N4,N-DIMETHYL- 0 0 2-[4-(2-PYRIDINYL)-1-PIPERAZINYL] -4,6-PYRIMIDINEDIAMINE: Cq Prepared by Procedures A (CH 2 C1 2 TEA, 3 4 h at -78 OC, then 3 4 h at 0 OC), N, and 0. 'H NMR (400 MHz, CDC13) 8 8.27 8.17 1H), 7.61 7.01 4H), 6.75 6.57 (m, 2H), 6.34 (br s, 1H), 5.23 1H), 3.95 3.85 4H), 3.68 3.59 4H), 3.00 6H); ESI-MS m/z: 472 (MH).
Example 200: N 4 (4-FLUORO-3-METHYLPHENYL) -N,N6-DIMETHYL- 2-[4-(2-PYRIDINYL)-1-PIPERAZINYL -4,6-PYRIMIDINEDIAMINE: Prepared by Procedures A (CH 2 C1 2 TEA, 3 4 h at -78 OC, then 3 4 h at 0 OC), N, and 0. 'H NMR (400 MHz, CDC1) 8 8.27 8.17 1H), 7.56 7.47 1H), 7.21 6.89 (m, 3H), 6.75 6.58 2H), 6.24 (br a, 1H), 5.18 1H), 3.95 3.84 4H), 3.69 3.55 4H), 3.00 6H), 2.25 3H); ESI-MS m/z: 408 (MH).
00 438 Example 201: 5 -DIMETHOXYPHENYL) -N6, N-DIMETHYL-2 (4-(2-PYRIDINYL)-1-PIPERAZINYL-4,6-PYRIMIDINEDIAMINE: Prepared by Procedures A (CH 2 C1 2 TEA, 3 4 h at -78 OC, then 3 4 h at 0 OC), N, and 0. 'H NIR (400 MHz, CDC13) 8 00 8.27 8.16 1H), 7.96 7.86 1H), 7.56 -7.43 (m, 1H), 6.93 6.42 5H), 5.31 Cs, 1H), 4.01 3.90 (m, 00 4H) 3.84 3H) 3.79 3H), 3.70 3.54 4H) 3.04(s, 6H); ESI-MS m/z: 436 (MH+) Example 202: N4-(3,5-DIMETHOXYPHENYL)-N6,N-DIMETHYL-2- 4- (2-PYRIDINYL)-1-PIPERAZINYL -4,6-PYRIMIDINEDIAMINE: Prepared by Procedures A (CH 2 C1 2 TEA, 3 4 h at -78 OC, then 3 4 h at 0 0 N, and 0. 'H NMR (400 MHz, CDC1 3
S
8.26 8.17 1H), 7.55 7.44 1H), 6.73 6.58 (m, is 2H), 6.59 6.53(m, 2H), 6.23 (br s, 1H) 5.37 1H), 3.98 3.88 Cm, 4H), 3.77 6H), 3.62 3.58 4H), 3.01 6H); ESI-MS m/z: 436 Example 203: N4-[3-BENZYLOXY)PHENYL-2-[4-(3- BROMOPHENYL) -1-PIPERAZINYL] -N,N-DIMETHYL-4, 6- PYRIMIDINEDIAMINE: Prepared by Procedures A (CH 2 C1 2
TEA,
3 4 h at -78 OC, then 3 4 h at 0 OC), N (TEA), and 0.
00 439 S'H NMR (400 MHz, CDC1 3 8 7.55 6.26 14H), 5.29 (s, 1H), 5.06 2H), 3.97 3.82 4H), 3.21 3.14 (m, 4H), 3.01 6H); ESI-MS m/z: 560 00 C Example 204: N 4 (2-BROMO-4-METHYLPHENYL) N-DIMETHYL-2- 00 [4-(2-PYRIDINYL)-1-PIPERAZINYL]-4,6-PYRIMIDINEDIAMINE: Prepared by Procedures A (CH 2 C1 2 TEA, 3 4 h at -78 oC, then 3 4 h at 0 OC), N, and 0. 'H NMR (400 MHz, CDC1 3 6 8.26 8.16 1H), 7.81 1H, J 7.52 7.44 1H), 7.38 1H, J 7.08 1H, J 6.72 2H), 6.47 (br s, 1H), 5.24 1H), 3.90 4H, J 3.61 4H, J 3.01 6H), 2.28 (s, 3H); ESI-MS m/z: 468 Example 205: AN- (2,4-DICHLOROPHENYL) -N',-DIMETHYL-2- [4- (2-PYRIDINYL)-1-PIPERAZINYL]-4,6-PYRIMIDINEDIAMINE: Prepared by Procedures A (CH 2 C1 2 TEA, 3 4 h at -78 OC, then 3 4 h at 0 OC), N, and 0. 'H NMR (400 MHz, CDC1 3 8 8.25 8.17 1H), 8.21 1H, J 7.49 1H, J 7.38 7.16 2H), 6.71 6.59 2H), 6.57 (br s, 1H), 5.25 1H), 3.93 3.85 4H), 3.65 3.55 4H), 3.03 6H); ESI-MS m/z: 444 440 Example 206: N 4 (3-FLUOROPHENYL) -I6,fl-DIMETMY-2- (2pyRIDINYL) -1-PIPERAZINYL] -4,6-PYRIMIDINEDIAMINE: Prepared by Procedures A (CH 2 Cl 2 TEA, 3 4 h at -78 OC, then 3- 4 h at 0 OC) N, and 0. 'H NMR (400 MHz, CDCl 3 88.25- 00 6.39 (in, 9H) 5.30 lH) 3.97 3.85 (mn, 4H), 3.74- 3.58 (mn, 4H), 3.01 6H) ESI-MS rn/z: 394 Example 207: B1 4 N'-DI METHYL 2- (2 -PYRI DINYL) -1I- PI PERAZ INYL] -AAt- (TRIFLUOROMETHOXY) PHENYL] 4,6 PYRIMIDINEDIAMINE: Prepared by Procedures A (CH 2 Cl 2
TEA,
3 4 h at -78 OC, then 3 4 h at 0 OC) and 0. ESI- MS m/z: 460 Example 208: it- (2,5-DICHLOROPHENYL)-N,N 6 -DIMETHYL-2- [4- (2 -PYRIDINYL) -1-PIPERAZINYLI 6-PYRIMIDINEDIAMINE Prepared by Procedures A (CH 2 Cl 2 TEA, 3 4 h at -78 OC, then 3 4 hi at 0 CC), N, and 0. ESI-MS m/z: 445 00 441.
Example 209: N',lN 4
-DIMETHYL-N
6 (4-PROPYLPHENYL) (2- PYRIDINYL) -1-PIPERAZINYL] 6-PYRIMIDINEDIAMINE: Prepared by Procedures A (CH 2 C1 2 TEA, 3 4 h at -78 IOC, then 3 4 h at 0 OC), N, and 0. ESI-MS m/z: 418 00 00 Example 210; AN4,N4-DIMETHYL-10- (4-PENTYLPHENYL) (2- PYRIDINYL) -1-PIPERAZINYL] 6-PYRIMIDINEDIAMINE: Prepared by Procedures A (CH 2 C1 2 TEA, 3 4 h at -78 0 C, then 3 4 h at 0 OC), N, and 0. ESI-MS mhz: 446 Example 211: V 4 (4-SEC-13UTYLPHENYL) -N6,N6-DIMETHYL-2- [4- (2-PYRIDINYL) -1-PIPERAZINYL] 6-PYRIMIDINEDIAMINE: Prepared by Procedures A (CH 2 Cl 2 TEA, 3 4 h at -78 OC, then 3 4 h at 0 N, and 0. ESI-MS m/z: 432 (MH4).
Example 212: N4-(2-TBRT-BUTYLPHENYL)- t,I#-DIMETHYL-2- t4- (2-PYRIDINYL) -1-PIPERAZINYL] 6-PYRIMIDINEDIAMINE Prepared by Procedures A (CH 2 C1 2 TEA, 3 4 h at -78 OC, then 3 4 h at 0 CC), N, and 0. ESI-MS m/z: 432 (MW).
00 442 Example 213: N4- (2,5-DIMETHYLPHENYL)-Nt,Ais-DIMETHYL-2- [4- (2-PYRIDINYL) -1-PIPERAZINYL] 6-PYRIMIDINEDIANINE: Prepared by Procedures A (CH 2 Cl 2 TEA, 3 4 h at -78 0
C,
00 then 3 4 h at 0 OC), N, and 0. ESI-MS m/z: 404 (MH+) 00 Example 214: N 4 (3,5-DIMETHYLPHENYL)-4f,N--DIMETHYL-2- [4- (2-PYRIDINYL) -1-PIPERAZINYL) 6-PYRIMIDINEDIAMINE: Prepared by Procedures A (CH 2 Cl 2 TEA, 3 4 h at -78 OC, then 3 4 h at 0 IC), N, and 0. ESI-MS m/z: 404 (MR+) Example 215: N 4 -(2,3-D)IMETI{YLP1HENYL)-N6,N4-DIMETHYL-2- [4- (2-PYRIDINYL) -1-PIPERAZINYL] 6-PYRIMIDINEDIAMINE: Prepared by Procedures A (CH 2 Cl 2 TEA, 3 4 h at -78 OC, then 3 4 h at 0 OC), N, and 0. ESI-MS m/z: 404 Example 216: N4- (3-B3ENZYLPHENYL) -N6,Nt-DIMETHYL-2- (2- PYRIDINYL) -1-PIPERAZINYL] 6-PYRIMIDINEDIAMINE: Prepared by Procedures A (CH 2 C1 2 TEA, 3 4 h at -78 OC, then 3 4 h at 0 OC), N, and 0. ESI-MS m/z: 466 (MW).
00 443 Example 217: N0-(4-ROMO-2-CHLOROPHENYL),6DIMETHYL-2- (2-PYRIDINYL) -1-PIPERAZINYL] 6-PYRIMIDINEDIAMINE: (1 Prepared by Procedures A (CH 2 C1 2 TEA, 3 4 h at -78 OC, then 3 4 h at 0 OC), N, and 0. ESI-MS m/z: 489 (MHW) 00 00Example 218: N4- (2,3-DICHLOROPHENYL) -DN 6 I-DIMETHYL-2- [4- (2-PYRIDINYL) -1-PIPERAZINYL] 6-PYRIMIDINEDIAMINE: Prepared by Procedures A (CH 2 Cl 2 TEA, 3 4 h at -78 OC, then 3 4 h at 0 OC), N, and 0. ESI-MS m/z: 445 (MW).
Example 219: N',N74-DIMETHYL-2- (2-PYRIDINYL) -1- PIPERAZINYL] -Nt- (2,4,5-TRIFLUOROPHENYL) 6- PYRI MIDI NEDIAMINE: Prepared by Procedures A (CH 2 Cl 2
TEA,
3 4 h at -78 OC then 3 4 h at 0 OC), N, and 0. ESI- MS mhZ: 4 30 (MH 4 Example 220: 4V4- (5-CHLORO-2-METH0XYPHENYL) -IQ6,-DIMETHYL- 2- (2-PYRIDINYL) -l-PIPERAZINYL] 6-PYRIMIDINEDIANINE: Prepared by Procedures A (CH 2 Cl 2 TEA, 3 4 h at -78 OC, then 3 4 h at 0 OC), N, and 0. ESI-MS m/z: 440 (MW) 00 444 Example 221: 10N 4 N- DIMETHYL 2- 4- (2 -PYRIDINYL) -1I- PI PERMZINYL] bf- 4, 5 -TRI FLUOROPHENYL) 4, 6 PYRIMIDINEDIAMINE: Prepared by Procedures A (CH 2 Cl 2
TEA,
0 3 4 h at -78 OC, then 3 -4 hi at 0 OC) N, and 0. ESI- 00 5 MS m/z: 4 30 00 Example 222: N 4 (2-CHLORO-5-FLUOROPHENYL)
-N
6
,N-DIMETHYL-
2- (2-PYRIDINYL) -1-PIPERAZINYL) -4,6-PYRIMIDINEDIAMINE: Prepared by Procedures A (CH 2 Cl 2 TEA, 3 4 h at -78 OC, then 3 4 h at 0 OC), N, and 0. ESI-MS 428 Example 223: 10- (2-CHLORO-4-METHYLPHENYL) -ZN,N-DIMETHYL- 2- (2-PYRIDINYL) -1-PIPERAZINYL] 6-PYRIMIDINEDIAMINE: Prepared. by Procedures A (CH 2 Cl 2 TEA, 3 4 h at -78 C, then 3 4 h at 0 OC), N, and 0. ESI-MS m/z: 424 (MH+) Example 224: 10- (3-CHLOROPHENYL) -N6,N4f-DIMETHYL-2- 4-12- PYRIDINYL) -1-PIPERAZINYLI 6-PYRIMIDINEDIAMINE: Prepared by Procedures A (CH 2 Cl 2 TEA, 3 4 h at -78 0 C, then 3 4 h at 0 N, and 0. ESI-MS m/z: 410 00 445 Example 225: 2-(4-BENZYL-1-PIPERAZINYL)-N 4 (TRIFLUOROMETHYL) PHENYL] Nt-DIMETHYL-4, 6- PYRIMIDINEDIAMINE: Prepared by Procedures 0 (toluene, OC) ,Q (toluene, 120 OC) and A. ESI-MS m/z: 487 00 00Example 226: 2- (4-BENZYL-1-PIPERAZINYL) (TRIFLUOROMETHYL) PHENYL) -iN, 16-DIMETHYL-4, 6- PYRIMIDINEDIAMINE: Prepared by Proced ures 0, Q (dioxane, 120 OC), and A. ESI-MS m/z: 487 (MH+) Example 227: 2- (4-BENZYL-1-PIPERAZINYL) -10- DIMETHOXYPHENYL) -N6, IN6- DIMETHYL- 4,6 -PYRIMIDINEDIAMINE: Prepared by Procedures 0, Q (dioxane, 120 OC) and A.
ESI-MS m/z: 449 (MW).
Example 228: N [3-(BENZYL0XY)PHENYL]-2-(4-BENZYL-1- PIPERAZINYL) -I ,N~-DIMETHYL-4, 6-PYRIMIDINEDIAMINE: Prepared by Procedures 0, Q (toluene, 120 OC) and A.
ESI-MS m/z: 495 (MW) 00 446 Example 229: 2-(4-BENZYL-1-PIPERAZINYL) -N 4 1
N
4 -DIMETHYL-Ns- (TRIFLUOROMETHYL) PHENYL) 6-PYRIMIDINEDIAMINE: (I Prepared by Procedures P (toluene, 105 OC), Q (toluene, 120 OC), and A. ESI-MS m/z: 457 (MvH+).
00 Example 230: 2-(4-BENZYL-l-PIPERAZINYL)-,N 4 (1 4-TRICHLOROPHENYL) 6-PYRIMIDINEDIANINE: Prepared 00 by Procedures 0 (60 OC), Q (toluene, 120 OC), and A. ESI- MS m/z: 492 Example 231: (2-FURYLMETHYL) -1-PI-PERAZINYL) -N 4
,N
4 DIMETHYL-If (4-METHYLPHENYL) 6-PYRIMIDINEDIAMINE: Prepared by Procedures R (16 P (sodium tert-butoxide, toluene, 120 OC) N (TEA, toluene ref lux) and A. ESI-MS m/z: 3 93 (MH~) Example 232: A1 2 (4-METI{OXYPHENYL) ETHYL) -AN 4 1
N-DIMETHYL-
it- (4-METHYLPHENYL) 6-PYRIMIDINETRIAMINE: Prepared by Procedures V, R, and S (DIEA, DMAP). ESI-MS m/z: 378 00 447 Example 233: Al-(3 -METHOXPHENYL) -IN, N6- D IMETHYL 2-[4 (TETRAHYDRO- 2- FURANYLMETHYL) P1PERAZINYL] 6- CI PYRIMIDINEDIAMvINE: Prepared by Procedures A, P (16 h), and Q (dioxane, 120 OC). ESI-MS m/z: 413 00 00Example 235: 2- (4-METHOXYBENZYL) -1-PIPERAZINYL] -N',N 4 DIMETHYL-N'- (4-METHYLPHENYL) 6-PYRIMIDINEDIAMINE: Prepared by Procedure Z. ESI-MS m/z: 433 Example 237: AN'A7-DIMETHYL-N'6-(4-METHYLPHENYL)-D'- 2-(2- THIENYL) ETHYL) 6-PYRIMIDINETRIAMINE: Prepared by Procedures R, S, and V. ESI-MS m/z: 354 Example 238: A14, N'-DIMETHYL-N6 (4 -METHYLPHENYL) 2- [4 THIENYLMETHYL) -1-PIPERAZINYL) 6-PYRIMIDINEDIAMINE: Prepared by Procedures AA, T (2 h) and W. ESI-MS mhz: 409 (MH*) Example 239: 2- (4-BENZYL-l-PIPERAZINYL) -N4- [4-CHLORO-2- (TRI FLUOROMETHYL) PHENYL) -N6, N6- DIMETHYL- 4, 6 448 PYRIMIDINEDIAMINE: Prepared by Procedures 0 (100 OC, Q (toluene, 120 OC), and A. ESI-MS m/z: 491 (MH') Example 240: N 4 (3-BROMO-4-METHYLPHENYL) -N6,N6-DIMETHYL-2- (2-PYRIDINYL) -1-PIPERAZINYLI 6-PYRIMIDINEDIANINE: Prepared by Procedures 0 (80 OC) Q (toluene, 120 OC) and A. ESI-MS mhz: 469 Example 241: (DIMETHYLAMINO) (4-TOLUIDINO) -2- PYRIMIDINYL] -1-PIPERAZINYL}NICOTINONITRILE: Prepared by Procedures 0, Q (tyoluene, 120 OC) ,and A. ESI-MS m/z: 415 (MH+) Example 242: N4,N4-DIMETHYL-fl- [4-METHYL-3- (2- PYRIDINYLAMIN0)PHENYL) (2-PYRIDINYL) -1-PIPERAZINYL] 4,6-PYRIMIDINEDIAMINE: Prepared by Procedures
P
(toluene) Q (toluene, 120 OC) ,and A. ESI-MS m/z: *482 Example 243:* R4- (3 -BROMOPHENYL) N, N-DIMETHYL- 2- 4- (2 PYRIDINYL) PIPERAZINYL) 6-PYRIMIDINEDIANINE: Prepared 00 449 by Procedures 0 (85 OC), Q (toluene, 120 OC) and A. ESI- MS m/z: 455 (MW).
00 Example 244: 2- (4-BENZYL-1-PIPERAZINYL)
-N
4 (2-CHLORO-4- (TRIFLUOROMETHYL) PHENYL] -N6,1 6 -DIMETHYL-4, 6- 00 PYRIMIDINEDIAMINE: Prepared by Procedures P (16 h, CItoluene) Q (toluene, 120 OC) ,and A. ESI-MS m/z: 491 Example 245: N4-(3-METHOXYPHENYL)-N4,,f-DIMETHYL-2- 4-(2- PYRIDINYL) -1-PIPERAZINYL) 6-PYRIMIDINEDIANINE: Prepared by Procedures A, N, and P. ESI-MS m/z: 406 (MW).
Example 246: N'4-(3-METHOXYPHENYL)-N4,?t-DIMETHYL-2-14- 2-.
(TRIFLUOROMETHYL) PHENYL] PIPERAZINYLI -4,6- PYRIMIDINEDIAMINE: Prepared by Procedures A, N, and P.
ESI-MS ni/z: 473 Example 247: N4- (3-METHOXYPHENYL) -,f-DIMETHYL-N2- (2- PHENYLETHYL) 6-PYRIMIDINETRIA'INE: Prepared by Procedures A, N, and P. ESI-MS rn/z: 364 450 ExaMple 248: B2N4-TRIMETHYL--(4-METHYLPHENYL)-N 2 PH-ENYLETHYL) 6- PYRIMIDINETRIAMINE: Prepared by Procedures A, N, and P. ESI-MS m/z: 362 (MH 4 Example 249: N-(4-METHYLPHENYL)-2-{4- fl-OXIDO-3- (TRIFLUOROMETHYL) -2-PYRrDINYL] -l-PIPERAZINYL}-6- (1- PIPERIDINYL)-4-PYRIMIDINAMINE: Prepared by Procedure CC.
ESI-MS m/z: 514 (MI-I).
Example 250: le, -DIMETHYL-Nf 4 PHENYLETHYL) 6-PYRIMIDINETRIAMINE: METHYLPHENYL) Ai2- (2 Prepared Procedures R and S. ESI-MS m/z: 348 (MW) Example 251: N 4 (3-METHOXYPHENYL) -is, IN-TRIMETHYL-f2- (2- PHENYLETHYL) 6-PYRIMIDINETRIAMINE: :Prepared by Procedures A, N, and P. ESI-MS m/z: 378 (MH 4 00 451 Example 252: 2-(4-BENZYL-1-PIPERAZINYL) -N 4 (3- METHOXYPHENYL) ,N 6 -DIMETHYL-4,6-PYRIMIDINEDIAMINE: Prepared by Procedures A, N, and P. ESI-MS m/z: 419 (MH).
00 Example 253: 2-(4-BENZYL-1-PIPERAZINYL)-N4,N4-DIMETHYL-N 6 0 (4-METHYLPHENYL)-4,6-PYRIMIDINEDIAMINE: Prepared by C1 Procedures A, N, and P. ESI-MS m/z: 403 (MlH).
Examples 1-90 and 115-253 as described above are merely illustrative of the methods used to synthesize pyrimidine derivatives. Further derivatives may be obtained utilizing methods shown in Schemes 1-5b. The substituents in Schemes 1-5b are described in the Detailed Description.
It may be necessary to incorporate protection and deprotection strategies for substituents such as amino, amido, carboxylic acid, and hydroxyl groups in the synthetic methods described above to form pyrimidine derivatives. Methods for protection and deprotection of such groups are well-known in the art, and may be found, for example in Green, T. W. and Wuts, P.G. M. (19.91) Protection Groups in Organic Synthesis, 2nd Edition John Wiley Sons, New York.
00 452 Scheme 1. Synthesis of Substituted Ct Triaminopyrimidines xR R R
NH->
00
R
RN RNH2 X leaving group such halogen OTf or OTs 453 Scheme 2. Alternate Synthesis of Substituted Triaminopyrimidines 1. NaH 2. R-X
R,R
x N R
RN
R-,N -R
RN,
I.
RNHH
R
X =leaving group such halogen OTf or OTs 454 Schemse 3. Alternate Synthesis of Substituted Trisininopyrimidines x x 1. NaH N 2. R-X R 10 3. NaH Ix 00 HM x 4. R-X R ,.R 00
RH
R___R
R N X -leaving group such halogen OTf or OT9 Alternatively, S N X
,R
R
NNS
455 Scheme 4. Synthesis of Morpholine intermediates
OH
R-NHr 2 C'CH2MC' C1CH 2 COC1NaH
H
0 R LiA1H 4 N
R
H
Scheme 5. Synthesis of N-Alkylamine Intermediates EtOCHO RNor RCOC1 LiAlH 4
RINHCH
2
R
456 Scheme Sa. Synthesis of Triaminopyrimidines from 2 -Amidopyrimidines Cl
N
H
2 N)N"C 1
RNH
2
R
RNH-N
N
RCOCl ~R K LiAlH 4 457 Scheme 5b. Substitution on the Piperazine Moiety of 2- (Piperazixi-1-yl)pyrimidines R R ArB (OH) Pd, (dba f 3 0 00
BINAP
RX
R
N
N N R-'e
H
RCOCl or
RCO
2 H and a peptide coupling reagent such as HATU LiAlH 4 H rH x is a leaving group such as a halogen or tosylate; HATU is 0- (7azabenzenzotriazol- I-yl) N, N N I -tetramethyluronium hexaf luorophosphate; ciba is dibenzylideneace tone; BINAP is 2,2'-bis(diphenylphosphino) -1,3.'-binaphthyl.
00 S458 SRadioligand Binding of Pyrimidines at Cloned Galanin Receptors n The binding properties of the pyrimidines of the present invention were evaluated at the cloned human galanin receptors, GAL1, GAL2, and GAL3, using protocols 00 Sdescribed herein.
C Radioligand Binding Assay Results 00 SThe pyrimidines described in Examples 1-90 and 115-253 y 10 were assayed using cloned human galanin receptors. The compounds were found to be selective for the GAL3 receptor. The binding affinities of the compounds of Examples 1-90 and 115-253 are illustrated in Tables l-3a.
459 TABLE I Wi Ki substitution (nM) Example Rli R2 GalR. Ga1R2 GalR3 1 Cs- 668 188 2 a m2818 562 26 3 H>5000 >5000 163 450 HN' >5000 627 Ia H a >5000 >5000 345 6 HN- >5000 2157 248 7HN)N 1107 775 177 8 HN'I >5000 795 264
HN
460
Q
R1 N R2 Table 1 continued Ki substitution (nM) Example R1 R2 GalR1 GalR2 GalR3 18 i 3589 543 19 H >5000 1771 79 >5000 >5000 164 21 4786 1096 49 22 442 176 28 23 HN.Q( >5000 >5000 24 H'oN >5000 396 1 210 Nc >5000 1497 548 255000 4049 26 f -N >5000 4049
II
462 463
QN
RI N R2 Table 1 continued Ki substitution Example R1 R2 GaiR1 GalR2 GalR3 37 HVJCN >5000 903 343 38 CY HN2901 516 320 39 5000 >5000 128 HNJ: >5000 2623 164 41 JGNHN r 2131 840 151 42 ffci, >5000 1137 275 43 y>5000 >5000 107 44 O_ N>5000 1023 133 NI ",>5000 >5000 505 CI N
N
00 R1 N R2 _______substitution 00 o Example R1 Table 1.
continued 465 TABLE 2
N
Rl N R2 Ki substitution (rM) Example Rl R2 GalRI GalR2 GalR3 OJK >5000 >5000 699 51 >5000 >5000 987 52 OK'H HN' >5000 >5000 570 53 >5000 >5000 980 54HN/ >5000 >5000 132 2 1 >5000 >5000 48 56 HNO >5000 >5000 794 57 0 'H HNla F >5000 >5000 360 58 ONNH >5000 >5000 783 59 IZI1NH >5000 >5000 566 NA >5000 >5000 86 00 466 Table 2 tr~ continued R1 N R2 Ki 00 Bubstitutiofl (rim) Example R1 R2 GaiR1 GalR2 00 61 1N LK >5000 >5000 c-Iryi >5000 >5000 467 RI N R2 Table 2 continued 468 RlI R2 Table 2 continued Ki substitution (nM) Example R1 R2 GalR1 GalR2 GalR3 79 3802 1656 190 PhO >5000 2478 615
HO
81 >5000 4789 160 82 5 >5000 >5000 232 83 HN'O >5000 >5000 160 84 JJ>J >5000 >5000 261 I s H>5000 4228 72 86 b0. >5000 >5000 227 87 >5000 4617 157 88 N 2188 355 39 Key: Ph Phenyl 469 TAB3LE 3 R2 RI AeR 3 Ki _______substitution Example X1 Rl R2 R3 GaiRl GalR2 GalR3 89 H CF N1122 1274 105
CF,
H HIr>5000 2460 105 470 Table 3a.
471 00 00 00 472
ND
473 474 475 00 476 Table 3a.
N
147 N640 00 0- N/ M 148 N N O 276 I Oa 00 0 N 149 N N 138*
N
150 N N 180 NN N"y N CI 151 CI 11 151 r 0 N
N
.N
152 Q 172 0, IN
N
153 1"55
H
The binding assay normally used for the indolone compounds was used to test this compound.
477 Table 3a.
00 478 Table 3a.
NN C1 160 26 QO Nb 00
NN
0 N 114 161 00 NN N 162 N N 42 N O 163 N N 500
NI-
N
16 4 r0jN j j
N
165 N139* The binding assay normally used for the indolone compounds was used to test this compound.
479 Table 3a. 166 f NN' 'N 263 4- 167 168 1"50 169 1N 77
N,,N
171 00 480 Table 3a.
N CI 172 N 00 N 173 117 00 (N
NN
174 N 325*
\N
175 N) -S 56
\N
-0 176 N N 608 N O 177 N N 1 4 2 The binding assay normally used for the indolone compounds was used to test this compound.
481 00 482 Table 3a.
0 184 C 00 NN 00 185 N 'lN 'y 00 483 7 "'N~Table 3a.
189 2 00 00 N153 190
I
NrN
N
N-
191
N,_
192 118 N NC 193 N~N N244 484 Table 485 486 487 Table 3a.
489 Table 3a._ 219 N~'I~F 558 00 220 ('NN 708 N F 221 NF213 N~ N N F 223 IN'I
QNN
490 00 491 Table 3a.
N'
CI
230 N NN N C C 00
N
231 136 00 232 N 155 233 869 Q%%NIN
N
rNN
N
237 AN404
SN'
238 331 S FNIN' N'O The binding assay normally used for the indolone compounds was used to test this compound.
492 493 Table 3a.
494 00 o 495 B. General Procedure for Preparing Indolones General Procedure for Synthesis of Iminoisatins. The appropriately substituted isatin (10 mg 10 g) was Q 5 placed in a flask and the appropriate aniline (1.0 1.1 00 equivalents) was added and the' mixture was stirred to Ohomogeneity. The mixture was then heated to 110 oC for 2- C 7 hours and then cooled. Solids were crystallized from 00 Shot methanol and filtered, giving the desired products C 10 (usually as an inseparable interconverting mixture of E/Z isomers).
Procedure A: 1-(3-THIENYL)-1H-INDOLE-2,3-DIONE: Triethylamine (56.9 mL, 0.408 mol), was added to a mixture of 1H-indole-2,3dione (15.0 g, 0.102 mol), copper (II) acetate (46.0 g, 0.255 mol), and 3-thienylboronic acid (19.6 g, 0.153 mol) in CH 2 C1 2 (500 mL). The reaction mixture was stirred overnight, filtered through Celite, rinsed with EtOAc/hexane 300 mL), and concentrated in vacuo.
The crude product was purified by column chromatography on silica using Hexane/EtOAc giving the desired product (1.1 g, 50 Procedure B: (3E)-3-[(4-METHYLPHENYL)IMINO]-1-(3-THIENYL)-1,3-DIHYDRO- 2H-INDOL-2-ONE: A solution of 1-(3-Thienyl)-lH-indole- 496 2,3-dione (20 mg, 0.087 nunol) in 1% H-OAc/MeOH (8 mL) was added to a solution of p-toluidine (19 mg, 0.18 mmol) in 1% HOAc/MeOH (B mL). The reaction mixture was stirred for 12 h at room temperature, heated at 50 0 C for 1 h, and concentrated in vacuo. The residue was purified by preparative TLC on silica using EtOAc/hexanes 0.1% TEA) giving the desired product (14 mg, Procedure C: (3Z) -1-PHENYL-3-( (3-THIENYL)PHENYL)IMINO}-1,3-DIHYDRO- 2H-INDOL-2-ONE: A mixture of (3Z) bromophenyl) imino) -1-phenyl 3 -dihydro-2H- indol- 2 -one (50.0 mg, 0.133 mmol) thiophene-3-boronic acid (26.0 mg, 0.199 mmol), tetrakis (triphenylphOsphine)palladium(0) (31.0 mg, 0.0268 mmol in THF (5 mL), and aqueous Na 2
CO
3 (2M, 100 4.L) was heated at 67 'C for 24 h. The crude product was concentrated in vacuc and the residue was extracted with CH 2 C1 2 (3 x 1 ml), and concentrated. The crude product was purified by preparative TLC using methanol in CHC1 3 giving the desired product (18 mg, 00 497 In Procedure D: (3Z) -5-B3ROMO-3-{([3- (TRIFLUOROMETHYL)PHENYL) IMINO}-1,3- DIHYDRO-2H-INDOL-2 -ONE: A mixture of 00 2,3-dione (1.0 g, 0.442 Mmol) and 3trifluoromethylanilile (0.993 g, 6.2 mmol)in a solution 00 of 1% acetic acid in methanol was stirred at 50 0 C for 12 h. The crude product was concentrated in vacuo, giving the desired crude product (640 mg, Procedure
E:
(3Z) -5-BROMO-1-PHENYL-3-( 3- (TRIFLUOROMETHYL) PHENYL] IMINsO) 3-DIHYDRO--21-INDOL-2- ONE: A mixture of (3z)-5-bromo-3-{[3- (trifluoromethyl)phenyl] imino)-1, 3-dihydro-2h-iYndol-2-one (100 mg, 0.272 mmol), copper (II) acetate (54 mg, 0.33 mmol) triethylamine (82.8 mg, 0.817 mmol) and benzene boronic acid (40 mg, 0.325 mmol) in 5 nIL of CH 2 Cl 2 was stirred at room temperature for 12 h. The crude mixture was concentrated in vacuo and purified by preparative
TLC
using EtOAc:hexane 1% triethylamine), giving the desired product (22 mng, 00 498 Procedure F: tf~ (3Z) 1,5-DIPHENYL3{ t3 (TRIFLUOROMETHYL)PHEN'YL)IMINO}- 1, 3 DIHYDRO-2H- IND0L-2 -ONE: A mixture of phenyl-3- (trifluoromethYl)phellimino) 3-dihydro- 00 2H-indol-2-one (22 Mg, 0.05 mmol), tetrakis (triphenylphosphifle) palladium (12.0 mg, 0.01 00 mmol), benzene boronic acid (10 mg, 0.08 mmcl) in THF mL) and aqueous Na 2
CO
3 C2M, 100 RL) was heated at 67 0
C
f or 24 h. The crude product was concentrated in vacuo and the residue was extracted with CH 2 C1 2 (3 x 1 ml) concentrated, and purified by preparative TLC using methanol in CHCl 3 giving the desired product (4 mg, 18%).
Procedure G: ETHYL 5-( 2 3 DIOXO2,3DIHDROlHINDOL-1YL)METHYL] -2- FIJROATE: A mixture of ethyl 5-(chloromethy1)-2-furoate (148 mg, 1.01 tmol) in dioxane (15 ml) was added to a mixture of NaH (48 mg, 1.20 mmol in dioxane (10 tuL) under argon at 0 The mixture was stirred f or 1 h at room temperature, ref luxed under argon for 16 h, cooled to room temperature, and then concentrated in vacuo. The residue was purified by preparative TLC using EtOAc/hexane giving the desired product (56 mg, 19 00 0 499 Procedure H: ETHYL 5- [3- O (TRIFLUOROMETHYL)PHENYL]IMINO}-2,3-DIHYDRO-1H-INDOL-1- 00 5 YL)METHYL]-2-FUROATE: A mixture of ethyl 5-[(2,3-dioxo- C) 2,3-dihydro-lH-indol-l-yl)methyl]-2-furoate (60 mg, 0.200 00 Smmol) and 3-trifluromethylaniline (32 mg, 0.200 mmol) was heated at 140 OC for 2 h. The residue was dissolved in CHC1 3 (1 mL) and purified by preparative TLC using EtOAc/hexane giving the desired product (20 mg, 23 Procedure I: 6-METHOXY-1-PHENYL-1H-INDOLE-2,3-DIONE: A solution of N- (3-methoxyphenyl)-N-phenylamine (1.14 g, 5.72 in ether (3 mL) was added to a solution of oxylyl chloride (728 g, 5.75 mmol)and heated at reflux for 1 h. The resulting mixture was cooled to room temperature, concentrated to dryness, and redissolved in nitrobenzene (35 mL). The solution was added to a solution of AlC1 3 in nitrobenzene (0.762 g, 5.72 mmol), and the resulting mixture was heated at 70 OC for 16 h. The crude product was concentrated in vacuo and purified by column 500 chromatography using EtOAc/hexane giving the desired product 60, mg, 50 Procedure J: 00 (3Z)-1-(4-BROMOPHENYL)-31E[3- (TRIFLUOROMETHYL) PHENYL] IMINO) -1 ,3-DIH4YDRO-2H-INDOL-2- 00 ONE: A solution of (trifluoromethyl)phenyl] imino}-1,3-dihydro-2H-indol-2-one (100 mg, 0.344 mmol) copper (II) acetate (93 mg, 0.516 mmol), triethylamine (105 mg, 1.03 mmol) ,and 4bromobenzene boronic acid (104 mg, 0.516 mmol) in 5 mL of
CH
2 C1 2 was stirred at room temperature for 12 h. The crude mixture was concentrated in vacuo and purified by preparative TLC using EtOAc:hexane 1% triethylamine), giving the desired product (65 mg, 42%).
Procedure K: A solution of (3Z) -1-(4-bromOpheny)3-{(3- (trifluoromethyl)phenyl) imino} 3-dihydro-2H-indo1-2-one (30 mg, 0.068), tetrakis (triphenylphosphine) palladium (0) (16.0 mg, 0.014 mmol), benzene boronic acid (13 mg, 0. 101 mmol) in THF (5 mL) and aqueous Na 2
CO
3 (0.45 M, 300 jiL) was heated at 67 0 C for 40 h. The crude product was concentrated in vacuo and the residue was extracted with 00 501
CH
2 Cl 2 (3 x 1 ml), concentrated, and purified by preparative TLJC using 10 methanol in CHC1 3 1 giving the desired product (5 mg, 16%).
005 The compounds of Examples 92 107, inclusive, were purchased from Bionet Research Ltd., 3 Highfield Industrial Estate, Camelford, Cornwall PL32 9QZ, UK.
00 These compounds can also be synthesized using the (1 procedure described above.
Example 91: 3-1 (2-METHOXYPHENYL) IMINO] -l-PHENYL-l, 3- DIHYDRO-2H- INDOL- 2-ONE Example 92: 1-PHENYL-3- (TRIFLUOROMETHYL) PHENYL) IMINO) 3 -DIHYDRO-2H-INDOL-2
-ONE
Example 93: 3- t(3-METHYLPHENYL)IMINO) -1-PHENYL-1,3- DIHYDRO- 2H- INDOL- 2-ONE Example 94: 3- [(3-CHLOROPHENYL)IMINO] -1-PHENYL-l,3- DIHYDRO-2H- INDOL-2 -ONE Example 95: 1-PHENYL-3-1A14- (TRIFLUOROMETHYL) PHENYL) IMINO) -1,3-DIHYDRO-2H-INDOL-2-ONE Example 96: 3- [(4-METHYLPHENYL)IMINO) -1-PHENYL-1,3- DIHYDRO-2H- INDOL-2 -ONE Example 97: 3-1 (4-CHLOROPHENYL) IMINO) -1-PHENYL-1, 3- DIHYDRO-2H-INDOL-2-ONE 00 502 Example 98: 3-[(4-BROMOPHENYL) IMINO) -1-PHENYL-l, 3- DIHYDRO-2H-INDOL-2-ONE Example 99: 3- I(4-FLUOROPHENYL)IMINO) -1-PHENYL-l,3- DIHYDRO-2H-INDOL-2-ONE 00 Example 100: 3-[(4-PHENOXYPHENYL)IMINO) -l-PHENYL-l,3- DIHYDRO-2H-INDOL-2-ONE 00 Example 101: 3-((4-ETHOXYPHENYL)IMINO]-1-PHENYL-l, 3 DIHYDRO-2H- INDOL-2-ONE Example 102: 3- ((4-METHOXYPHENYL) IMINO) -1-PHENYL-l, 3 DIHYDRO-2H-INDOL-2-ONE Example 103: 3-[(3,5-DICHLOROPHENYL)IMINO1 -PHENYL-l,3- DIHYDRO-2H- INDOL-2 -ONE Example 104: 3-[(3,5-DIMETHYLPHENYL)IMINO) -1-PHENYL-1,3- DIHYDRO-2H-INDOL-2-ONE Example 105: l-ALLYL-3-[U3,4-DICHLOROPHENYL)IMINO)-l, 3 DIHYDRO-2H- INDOL-2 -ONE Example 106: l.ALLYL3-[(3,5-DICHLOROPHENYL)IMINOI-1, 3 DIHYDRO- 2H- INDOL- 2-ONE Example 107: 3- [(4-BROMOPHENYL) IMINO) -l-ISOPROPYL-1,3- DIHYDRO-2H-INDOL-2-ONE- The methods that follow demonstrate procedures useful for synthesizing compounds of this invention (illustrated in 00 503 SSchemes 6 and Substituted isatins useful for synthesizing compounds of this invention can n alternatively be obtained using the procedures described in the following references: O 5 Garden, S. Da Silva, L. Pinto, Synthetic O0 Communications, 1998, 28, 1679 1689.
SCoppola, Journal of Heterocyclic Chemistry, 1987, 00 24, 1249.
00 Hess, B.A. Jr; Corbino, Journal of Heterocyclic C 10 Chemistry, 1971, 8, 161.
Bryant, W. M. III;- Huhn, Jensen, Pierce, M.
Stammbach, Synthetic Communications, 1993, 23, 1617 1625.
Example 108: 1-[(5-CHLORO-2-THIENYL)METHYL]-3-( [3- (TRIFLUOROMETHYL) PHENYL] IMINO)-1,3-DIHYDRO-2H-INDOL-2- ONE: A mixture of 1-[(5-chloro-2-thienyl)methyl]-2Hindole-2,3-dione (25 mg, 0.09 mmol) (prepared as described below) and 3-trifluoromethylaniline (11.3 pL, 0.09 mmol) was heated neat at 140 oC for 2 h. The crude material was purified by preparative TLC using a mixture of 3:7 ethyl acetate in hexane as the eluent, giving the desired product (23 mg 0.05 mmol, 61 1 H NMR (400 MHz): 6 (major isomer) 7.57 J 7.7, 1H), 7.53
J
7.8, 1H), 7.33 J 7.8, 1H), 7.28 1H), 7.19 (d, J 7.6, 2H), 6.94 6.72 4H), 6.56 J 7.7, 1H), 5.02 2H); ESI-MS m/z found 421 1-[(5-CHLORO-2-THIENYL)METHYL]-2H-INDOLE-2,3-DIONE:
A
solution of isatin (125 mg, 0.85 mmol) in anhydrous dioxane (10 mL) was added dropwise to a solution of sodium hydride (60% dispersion in mineral oil, 24 mg, 00 S504 S0.62 mmol) in anhydrous dioxane (10 mL) at 0 oC under argon. The mixture was allowed to stir for 5 minutes and l n then 2-chloro-5-(chloromethyl)thiophene (0.12 mL, 1.02 mmol) in dioxane (10 mL) was added dropwise to the resulting mixture. The reaction mixture was heated at 0 reflux under argon for 16 h and concentrated in vacuo.
SThe crude material was purified preparative TLC using C1 1:24 methanol in chloroform as the eluent, giving the 00 Sdesired product as a yellow solid (53 mg, 0.19 mmol, 22 CN 10 'H NMR (400 MHz): 8 7.62 J 7.4, 1H), 7.56 J 7.8, 1H), 7.14 J 7.7, 1H), 6.94 J 8.0, 1H), 6.90 J 3.2, 1H), 6.78 J 3.7, 1H), 4.90 (s, 2H).
Example 109: 1-(3-THIENYL)-3-{[3- (TRIFLUOROMETHYL)PHENYL]IMINO)-1,3-DIHYDRO-2H-INDOL-2- ONE: A mixture of 1-(3-thienyl)-2H-indole-2,3-dione mg, 0.11 mmol) (prepared as described below) and 3trifluoromethylaniline (14 uL, 0.11 mmol) was heated neat at 140 oC for 2 h. The crude material was purified by preparative TLC using a mixture of 3:7 ethyl acetate and hexane as the eluent, giving the desired product as a yellow solid (7.3 mg, 0.02 mmol, 22 1H NMR (400 MHz) 7.62 7.19 9H), 6.94 J 8.0, 1H), 6.76 J 7.6, 1H); ESI-MS m/z found 373 1-(3-THIENYL)-2H-INDOLE-2,3-DIONE: Copper(II) acetate monohydrate (4.25 g, 23.4 mmol) was heated at reflux in acetic anhydride (30 mL) for 2 h. The mixture was filtered and washed with anhydrous ether (500 mL). The solid was dried in vacuo at 55 OC for 16 h.
Dichloromethane (1 mL) was added to a mixture of 00 505 F copper(II) acetate (62 mg, 0.34 mmol), isatin (50 mg, S0.34 mmol), and thiophene-3-boronic acid (87 mg, 0.68 Smmol), followed by triethylamine (0.10 mL, 0.68 mmol) under argon. The resulting solution was stirred for 16 h at room temperature. The reaction mixture was then 00 M recharged with 0.10 mmol copper(II) acetate, 0.10 mmol of S3-thiophene boronic acid, and 1 drop of triethylamine, C and the mixture was heated at 50 oC for 6 h. The crude 00 0 material was purified by preparative TLC using 3:97 Cl 10 methanol in chloroform as the eluent, giving the desired product as a yellow solid (25 mg, 0.11 mmol, 33 1H NMR (400 MHz): 5 7.70 J 7.5, 1H), 7.58 J 7.8, 1H), 7.50 J 5.1, 1H), 7.48 1H), 7.24 J 5.1, 1H), 7.18 J 7.51, 1H), 7.05 J 8.0, 1H).
Example 110: 2-METHYL-5-[(2-OXO-1-PHENYL-1,2-DIHYDRO-3H- INDOL-3-YLIDENE)AMINO]-2H-ISOINDOLE-1,3(2H)-DIONE:
A
mixture of 1-phenylisatin (50 mg, 0.22 mmol) and 4-amino- N-methylpthalimide (40 mg, 0.22 mmol) was heated neat at 215 OC for 2 h. The crude material was purified by preparative TLC using a mixture of 3:7 ethyl acetate and hexane as the eluent, giving the desired product as a yellow solid (8 mg, 0.02 mmol, 10 H NMR (400 MHz) 8 7.88 J 7.8, 1H), 7.83 7.80 1H), 7.51 J 1H), 7.47 7.18 6H), 7.02 J 8.0, 1H), 6.91 6.79 2H), 6.58 J 7.5, 1H), 3.22 3H); ESI-MS m/z found 382 Example 111: 1- [(5-CHLORO-1-BENZOTHIEN-3-YL)METHYL]-3- {[3-(TRIFLUOROMETHYL)PHENYL]IMINO}-1,3-DIHYDRO-2H-INDOL- 00 S506 S2-ONE: A mixture of 1-[(5-chloro-l-benzothien-3c- yl)methyl]-2H-indole-2,3-dione (50 mg, 0.15 mmol) C (prepared as described below) and 3trifluoromethylaniline (0.020 mL, 0.15 mmol) was heated neat at 140 °C for 2 h. The crude material was purified 00 c by preparative TLC using a mixture of 1:3 ethyl acetate Sand hexane as the eluent giving the desired product as a C yellow solid (13 mg, 0.030 mmol, 1H NMR (400 MHz): 0 8 7.98 J 2.0, 1H), 7.80 J 8.6, 1H), 7.58 (t, J 7.7, 1H), 7.52 J 8.1, 1H), 7.43 1H), 7.38 (dd, J 8.6, 7.31 (overlapping singlet and dt, J 1.2, 7.8, 2H), 7.24 J 7.8, 1H), 6.87 J 7.9, 1H), 6.77 J 7.7, 1H), 6.59 J 7.7, 1H), 5.20 2H). ESI-MS m/z found 471 (MH with 5 C1) 473 (MH with 1-[(5-CHLORO-1-BENZOTHIEN-3-YL)METHYL]-2H-INDOLE-2,3dione: A solution of isatin (125mg, 0.85 mmol) in anhydrous dioxane (10 mL) was added dropwise to a solution of sodium hydride (60% dispersion in mineral oil, 25 mg, 0.62 mmol) in anhydrous dioxane (10 mL) at 0 oC under argon. The mixture was allowed to stir for minutes and then a solution of chlorobenzo[b]thiophene (267 mg, 1.02 mmol) in dioxane (10 mL) was added dropwise to the reaction mixture. The reaction mixture was heated at reflux under argon for 16 h and concentrated in vacuo. The crude material was purified by preparative TLC using 1:24 methanol in chloroform as the eluent, giving the desired product as a yellow solid (125 mg, 0.38 mmol, 1 H NMR (400 MHz): 8 7.89 1H), 7.79 J 8.5, 1H), 7.65 J 1H), 7.54 J 8.0, 1H), 7.42 1H), 7.38 J 00 507 8. 5, 114), 7. 14 J 7. 5, 1H) 6. 88 J 7. 8, 114), 5.13 2H).
Example 112: 3-(lH-INDOL5YLIMINO)1l-PHENYL-1,3-DIHYDRO- 2H-INDOL-2-ONE.: 1-pheflylisatin (51.8 mg, 0.23 mmol) and 00 5-aminoindole (31 mg, 0.23 mmol) were mixed and heated at 140 0 C for 2 h. The resulting crude product was purified 00 by preparative TLC using ethyl acetate/hexane as the eluent, giving the desired product as a yellow solid (10. 8 mg, 14%) 1H NMvR (400 MHz): 8.28 1H4), 7.57 (t, J 7.7, 2H4), 7.49 7.40 (in, 6H), 7.29 7.23 (mn, 1H) 7.03 (dd, J 8.5, 1.7, 114), 6.98 J 7.6, 1H), 6.83 J 8.0, 114), 6.74, TJ 7.6, 114), 6.59 114); ESI- MS m/z found 338 Example 113: 3- [(6-CLORO-3-'PYRIDINYL) IMINO) -1-PHENYL- 1,3-DIHYDRO-2.H-INDOL-2-ONE: 1-phenylisatin (23.0 mg, 0.10 mmol) and 5-amino-2-chloropyridile (12.8 mng, 0.10 minol) were mixed and heated at 140 0 C for 7 h. The resulting crude product was purified by preparative
TLC
using hexane/ethyl acetate as the eluent, giving the desired product as a yellow solid (19.7 mg,
'H
NMR (400 MHz) 8 8.15 J 8, 1H), 7.6 7.2 (in, 914), 6.85 6.75 (in, 2H); ESI-MS m/z found 334 (Mlii.
Example 114: 3- [(2METHYL1,3BENZOT1IAZOL5YL) IMINO) l-PHENYL-1,3-DI14YDRO-2H-INDOL-2-ONE: 5-amino-2methylbenzcthiazcle (52.2 mng, 0.31 mmmol) was mixed with 1-phenylisatin (69.7 mng, 0.31 mmcl) and heated at 140 0
C
for 3 h. The resulting crude product was purified by preparative TLC using ethyl acetate/hexane as the 00 508 eluent to give the desired product as a yellow solid (36.9 mg, 32.3 'H NMR Data: 5 7.9-6.7 12H), 2.9 3H). ESI-MS m/z found 370 (MH).
Example 254: (3Z)-3-[(3,4-DICHLOROPHENYL)IMINO]-1-(2- 00 r PYRIDINYLMETHYL)-1, 3-DIHYDRO-2H-INDOL-2-ONE: Prepared by Procedures H and K (for substitution of 2-picolyl 00 chloride). 1H NMR (400 MHz, CDC13) 8 8.51 8.46 1H), 7.87 7.78 1H), 7.64 1H, J 7.53 7.31 5H), 7.28 1H, J= 7.12 1H, J 8.1), 6.58-6.53 1H), 5.51 2H); ESI-MS m/z 381 Example 255: (3Z)-3-[(3,4-DICHLOROPHENYL)IMINO]-1-[(3,5- DIMETHYL-4-ISOXAZOLYL)METHYL]) -1,3-DIHYDRO-2H-INDOL-2-ONE: Prepared by Procedure B (microwave heating). 'H NMR (400 MHz, CDC13) 6 7,63 1H, J 7.46 (dt, 1H, J 8.1, 7.28 1H, J 7.02 1H, J= 6.88 (dt, 1H, J 8.0, 6.74 6.72 1H), 6.72 6.70 1H), 5.53 2H), 2.50 3H), 2.24 3H); ESI-MS m/z 399 Example 256: (3Z)-3-[(3,4-DICHLOROPHENYL)IMINO]-1- [3- (TRIFLUOROMETHYL)PHENYL -1,3-DIHYDRO-2H-INDOL-2-ONE: Prepared by Procedures A and B. 1H NMR (400 MHz, CDC13) 7.90 7.87 1H), 7.83 7.79 1H), 7.67 1H, J 00 509 8) 7.46 -7.40 (in, 1H) 7.33 1H, J 7.08 7.05 (mn, 1H), 6.96 6.80 (in, 5H) ESI-MS m/z 435 Example 257: (3Z)-l-(3,5-DICHLOROPHENYL)-3-[(3,4- 00 5 DICHLOROPHENYL) IMINO) 3-DIHYDRO-2H-INDOL-2-ONE: Prepared by Procedures A and B. 'H NNR (400 MHz, CDC1 3 8 00 7. 93 1H, J B. 81) 7. 79 1H, J 0) 7. 72 7. 68 (mn, 1H) 7.59 -7.45 (mn, 1H) 7.46 1H, J 8.1) 7.32 (dt, 1H, J 8.0, 7.23 1H, JT 6.97 (ad, 1H, J 8.0, 6.92 6.87 (mn, 1H) 6.85 6.81 (in, 1H) ESI-MS m/z 435 (MH 4 Example 258: (3Z) -3-[(3,4-DICHLOROPHENYL)IMINO) -6- METHOXY-1-PHENYL-1, 3-DIHYDRO-2H-INDOL-2 -ONE: Prepared by Procedures K, L, and B. 'H NMR (400 MHz, CDCl 3 8 7.69 7. 54 (mn, 1H) 7. 53 7. 38 (in, 3H) 7. 29 1H, LT 2. 0) 7.17 1H, LT 7.12 1H, J 6.84 (d, 1H, J 2.5) 6.78 1H, J 8) 6.6 (dd, 2H, J3=8.0, 6.55 (rdd, 2H, LT 8.1, ESI-MS m/z (398 MH+) Example 259: (3Z)-3-E(4CHLORO-3METHYLPHENYL)IMINO) -1- (3 -THIENYL) 3-DIHYDRO-2H-INDOL-2-ONE: Prepared by Procedures A and B (80 oC) 'H NMR (400 MHz, CDC1 3 8 7.69 7.62 2H) 7.49 1H) 7.47 1H), 7.41 (at, 1H1, 0O 00 510 J 7.1, 7.3 (dd, 1H, J 5.0, 7.05 6.97 i 1H, 6.93 6.86 1H), 6.77 1H), 6.56 1H), (c 2.53 3H); ESI-MS m/z 353 00 Example 260: (3Z)-3-(2-NAPHTHYLIMINO)-1-(3-THIENYL)-1,3- DIHYDRO-2H-INDOL-2-ONE: Prepared by Procedures A and B 00 1H NMR (400 MHz, CDC1 3 6 8.15 1H, J 9.1), C 8.06 7.99 1H), 7.89 7.80 1H), 7.78 7.71 (m, 1H), 7.71 7.47 4H), 7.41 7.35 1H), 7.33 (d, 1H, J 7.28 1H, J 7.00 1H, J 6.76 1H, J 6.67 1H, J ESI- MS m/z 355 Example 261: [(4-CHLOROPHENYL) IMINO]-1-(3- THIENYL)-1,3-DIHYDRO-2H-INDOL-2-ONE: Prepared by Procedures A and B (80 'H NMR (400 MHz, CDC13) 5 7.69 7.56 2H), 7.54 7.48 1H), 7.41 (dt, 1H, J 8, 7.32 7.28 1H), 7.11 6.99 3H), 6.89 (dt, 1H, J 6.77 6.73 1H), 6.66 6.33 1H); ESI-MS m/z 339 (MH 4 Example 262: (3Z)-3-[(4-IODOPHENYL)IMINO] -1-(3-THIENYL)- 1,3-DIHYDRO-2H-INDOL-2-ONE: Prepared by Procedures A and B HOAc in MeOH). 1 H NMR (400 MHz, CDC13) 8 7.79 7.74 00 511 2H), 7.53 7.48 2H), 7.35 (dt, IH, J 7.29 7.24 1H), 6.98 1H, J 6.89 6.75 4H) ESI-MS m/z 431 (MH+) 00 Example 263: (3Z)-3-[(4-METHYLPHENYL)IMINO]-1-(3- 00 rpr b THIENYL) 3-DIHYDRO-2H-INDOL-2 -ONE: Prepared by Procedures A and B HOAc in MeOH). 1 H NNR (400 MHz, CDC1 3 8 7.52 7.44 2H), 7.35 7.22 4H), 6.99 6.93 3H), 6.87 6.78 2H), 2.42 3H); ESI-MS n/z 319 Example 264: (3Z)-3-[(3,5-DIFLUOROPHENYL)IMINOI 1- (3- THIENYL) -1,3-DIHYDRO-2H-INDOL-2-ONE: Prepared by Procedures A and B HOAc in MeOH). 1 H NNR (400 MHz, CDC1 3 8 7.54 7.16 4H) 6.99 (dt, 1H, J 8.2, 0.8), 6.89 (dt, 1H, J 7.7, 6.76 1H, J 6.71 (tt, 1H, J 9.3, 6.64 6.57 2H); ESI-MS m/z 341 Example 265: BIPHENYL>-4-YLIMINO)-l1(3- THIENYL) -1,3-DIHYDRO-2H-INDOL-2-ONE: Prepared by Procedures A and B HOAc in MeOH). 1 H NMR (400 MHz, 512 CDC1 3 8 7.73 7.12 (in, 1311), 6. 99 1H1, J 8 8.0) 6. 89 1H, JLT 6.82 (dt, 1H, J ESI-MS m/z 381 (MH+) 00 Example 266: ETHYL 3-{[(3Z)-2-OX(>1-(3-THIENYL)-1,2- 00 DIHYDRO-3H-INDOL-3-YLIDENEJ AMNO)BENZOATE: Prepared by Procedures A and B HOAc in MeOH) 111 NMR (400 MHz, CDC1 3 8 7.96 111, J 7.75 7.17 (mn, 6H1), 6.98 1H, J 8 8.0) 6. 87 78 (in, 2H1), 6. 63 1H1, J 7.8) 4.45 -4.32 (in, 2H) 1.43 1.33 Cm, 3H); ESI-MS m/z 377 Example 267: t(6-CHLORO-3-PYRIDINYL)IMINO) THIENYL) -1,3-DIHYDRO-2H-INDOL-2-ONE: Prepared by Procedures A and B (1%6 HOAc in MeOH) 'H NMR (400 MHz, CDC1 3 8 8.21 6. 81 (in, 1011); ESI-MS m/z 340.13 Example 268: 3Z)-3-((4-PHENOXYPHENYL)IMINO)-1(3- THIENYL) 3-DIHYDRO-2H-INDOL-2-ONE: Prepared by Procedures A and B HOAc in MeOH) 'H NNR (400 MHz, CDC1 3 8 7.85 6.70 (in, 1611); ESI-MS m/z 397 00 513 Example 269: [(4-BROMOPHENYL)IMINOJ -1-(3-THIENYL)- 1, 3-DIHYDRO-2H- INDOL-2 -ON~E: Prepared by Procedures A and 00 H. 'H NMR (400 MHz, CDCl 3 8 7.82 6.55 (in, 11H) ESI-MS zn/z 3 83 (MH*) 00 ExaMple 270:- (3Z)-3-(3-CHLOROPHENYL)IMINO]-l- THIENYL) -1,3-DIHYDRO-2H-INDOL-2-ONE: Prepared by Procedures A and H. "H NNR (400 MHz, CDC1 3 8 7.55 6-50 (mn, 11H) ESI-MS m/z 339 (MHW).
Example 271: (3Z) ((3-METHYLPHENYL) IMINO] (3- THIENYL) -1,3-DIHYDRO-2H-INDOL-2-ONE: Prepared by Procedures A and B HOAc in MeOH). lH NNR (400 MHz, CDCl 3 8 7.67 6.78 (in, 11H) 2.39 3H) ESI-MS mZn/ 319 (MH+) Example 272: [(3,4-DICHLOROPHENYL)IMINO]-1-(3- THIENYL) 3-DIHYDRO-2H-INDOL-2-ONE: Prepared by Procedures A and B HOAc in MeOH) NMR (400 MHz, CDC1 3 8 7.82 80 (mn, 10H) ESI-MS m/z 373 (MW).
00 514 t~ Example 273: (3Z)-l-(2-PYRIDINYLMETHYL)-3-{ [3- (TRIFLUOROMETHYL) PHENYL] EMINO}-1,3-DIHYDRO-2H-INDOL-2- 00ONE: Prepared by Procedure B. ESI-MS m/z 382 (MHW).
005 00 Example 274: (3Z)-3-[(3,5-DICHLOROPHENYL)IMINOJ (2- PYR IDINYLMETHYL) -1,3 -DIHYDRO' 2H- INDOL- 2-ONE: Prepared by Procedure B. ESI-MS m/z 382 Example 275: ((3,5-DIMETHYL-4-ISOXAZOLYL)METHYLJ (TRIFLUOROMETHYL)PHENYL) IMINO-1,3-DIHYDRO-2H- INDOL-2-ONE: Prepared by Procedure B. ESI-MS m/z 400 Example 276: (3Z)-3-[(3,4-DIFLUOROPHENYL)IMINO]11-3- PYRIDINYLVETHYL) 3-DIHYDRO-2H-INDOL-2 -ONE Prepared by Procedure B. ESI-MS m/z 350 (MW) 00 515 Example 277: (3Z)-1-(3-PYRIDINLMETHYL)-3i [3- (TRIFLUOROMETHYL) PHENYL] IMINO} 3-DIHYDRO-2H-INDOL-2- ONE: Prepared by Procedure B. ESI-MS m/z 382 ((MHW).
00 Example 278: [(3,4-DIFLUOROPHENYL)IMINO]-1- (2- 00 PYRIDINYLMETHYL) -1 ,3-DIHYDRO-2H-INDOL-2-ONE: Prepared by Procedure B. ESI-MS m/z 350 (MH+) Example 279: (3Z) [(3,5-DICHLOROPHENYL)IMINO]-1- (3- PYRIDINYLMETHYL) 3-DIHYDRO-2H-INDOL-2-ONE: Prepared by Procedure B. ESI-MS m/z 384 Example 280: (3Z) -3-((3,5-DICHLOROPHENYL)IMINO>-1-[(3j5- DIMETHYL-4-ISOXAZOLYL) METHYL] 3-DIHYDRO-2H-INDOL-2-ONE: Prepared by Procedure B. ESI-MS zn/z 402 Example 281: (3Z) (9-ETHYL-9H-CARBAZOL3YL) IMINO] -1- PHENYL-.l,3-DIHYDRO-2H-INDOL-20ONE: :Prepared by' Procedure H. NMR (400 MHz, CDCl 3 8 8.28 6.66 (in, 16H), 4.47 4.35 (in, 2H), 1.55 1.44 (in, 3H); ESI-MS m/z 416 00 516 Example 282: (3Z)-1-PHENYL-3-(5--QUINOLINYLIMINO)-1,3- DIHYDRO-2H-INDOL-2-ONE: Prepared by Procedure H. 1H NMR (400 MHz, CDCl 3 8 9.38 9.32 (in, 1H) 8.55 8.50 (mn, 1H), 8.01 6.62 (in, 12H), 6.43 6.35 (in, 1H); ESI- 00 MS m/z 350 00 Example 283: (3Z)-3-[(4-IODOPI4ENYL)IMINOJ-1-PHENYL-1,3- DIHYDRO-2H-INDOL-2 -ONE: Prepared by Procedure B (0.1% HOAc, 80 OC, 92 h, 4 eq RNH 2 3 A molecular sieves) ESI- MS m/z 425 Example 285: (3Z) -3-[(3,4-DIFLUOROPHENYL) IMINO] -1-PHENYL- 1, 3-DIHYDRO-2H-INDOL-2 -ONE: Prepared by Procedure B (0.1 HOAc, 80 0 C, 92 h, 4 eq RNH 2 3 A molecular sieves).
ESI-MS m/z 335 Example 286: ((2-CHLORO-4-METHYLPHENYL)IMINO] -1- PHENYL- 1, 3-DIHYDRO-2B-INDOL-2 -ONE: Prepared by Procedure B 1 %HOAc, 80 0 C, 92 h, 4 eq RNI 2 3 A molecular sieves) .ESI-MS mhz 347 (MH 4 with 35 C1) 349 (MR* with 3 7 cl) Example 287: (3Z)-3-[(2,4-DIMETHOXYPHENYL)IMINOlI-1 PHENYL- 1, 3 -DIHYDRO-2H- INDOL-2 -ONE: Prepared by Procedure 00 517 B 1 HOAc, 80 OC, 92 h, 4 eq RNH 2 3 A molecular sieves) ESI-MS m/z 359 (MW*) Example 288: U3Z)-2-OXO--1-PHENYL-1,2-DIHYDRO-3H- 00 INDOL- 3-YLIDENEI AMINO IBENZONITRILE:. Prepared by Procedure (Ni B 1 HOAc, 80 92 h, 4 eq RNH 2 3 A molecular 00 sieves) ESI-MS m/z 324 Example 289: (3Z)-3-f[2-METHYL-5- (TRIFLUOROMETHYL) PHENYL] IMINO} -1-PHENYL-1, 3-DIHYDRO-2H- INDOL-2-ONE: Prepared by Procedure B (0.1 HOAc, 80 0
C,
92 h, 4 eq RNH 2 3 A molecular sieves) ESI-MS mhz 381
(MH
4 Example 290: (3Z) f(4-CHLORO-3-METHYLPHENYL)IMINO] -1- (3-THIENYL) -l,3-DIHYDRO-2H-INDOL-2-ONE: Prepared by Procedures A and B (80 ESI-MS m/z 353 (MW) Example 291: -(3Z)-3-(6-QUINOLINYLIMINO)-1-(3-THIENYL)- 1, 3-DIHYDRO-2H- INDOL-2 -ONE: Prepared by Procedures A and B (80 OC) ESI-MS m/z 356 (MHW) Example 292: (3Z) (4-CHL (3Z-3-4-CL( ROPHENYL) IMINO] (3- THIENYL) -1,3-DIHYDRO-2H-INDOL-2-ONE: Prepared Procedures A and B (80 OC). ESI-MS m/z 339 Example __295: (3Z)-3-[(3-ISOPRO PYLPHENYL) IMINO] (3- THIENYL) 3-DIHYDRO-2H-INDOL-2-ONE: Prepared Procedures A and B (80 OC) ESI-MS m/z 347 Example 296: [(4-CYCLOHEXYLPHENYL)IMINOJ-1-(3- THIENYL)-1,3-DIHYDRO-2H-INDOL-2-ONE: Prepared by Procedures A and B (80 Oc). ESI-MS mhz 387 Example 297: -2-OXO-1-PHENYL-l,2-DIHYDRO-3H- INDOL-3 -YLIDENE] AIINO} PHENYL) ACETONITRILE: Prepared by Procedure B 1 HOAc, 80 92 h, 4 eq RNH 2 3 A molecular sieves). ESI-MS m/z 339 Example 298: [(2,2-DIFLUORO-l,3-BENZODIOXOL-5- YL) IMINO] -l-PHENYL-l ,3-DIHYDRO-2H-INDQL-2-ONE: Prepared by Procedure B 1 HOAc, 80 92 h, 4 eq RNH 2 3 A molecular sieves). ESI-MS m/z 379(MH+).
00 519 Example 299: (3Z)-3-(1,3-BENZOTHIAZOL-6-YLIMINO) -1- PHENYL- 1, 3 7DIHYDRO-2H- INDOL-2 -ONE: Prepared by Procedure H. ES I -MS m/z 3 56 00 M Example 300: (3Z)-l-TETRAHYDRO-2H-PYRAN-4-YL-3-{[3-- (TRIFLUOROMETHYL) PHENYL] IMINO}-1, 3-DIHYDRO-2H-INDOL-2- 00 ONE: Prepared by Procedures G and H. ESI-MS m/z 375(MH+).
Example 301: (3Z) -3-(1H-INDAZOL-6-YLIMINO) -l-PHENYL- 1,3-DIHYDRO-2H-INDOL-2-ONE: Prepared by Procedure H.
ESI-MS m/z 339 Example 302- (3Z) -3-[(3-CHLOROPHENYL)IMINO] -6-METHOXY-1- PHENYL-1, 3 -DIHYDRO-2H-INDOL-2 -ONE: Prepared by Procedures I and H. ESI-MS mhz 363 (MH 4 Example 303: (3Z)-6-METHOXY-1-PHENYL-3-{ [3- (TRIFLUOROMETHYL) PHENYLI IMINOI 3-DIHYDRO-2H-INDOL-2- ONE: Prepared by Procedures I and H. ESI-MS m/z 397 Example 304: (3Z) -1-PHENYL-3-{[(4- (3- THIENYL) PHENYL] IMINOPI-, 3-DIHYDRO-2H-INDOL-2-ONE: Prepared by Procedures H and C. ESI-MS m/z 381 00 520 Example 305: (3Z)-1-PHENYL-3-{13- (TRIFLUOROMETHYL) [1,1- BIPHENYL] -4-YL]IMINO)-1,3-DIHYDRO-2H-INDOL-2-ONE: Prepared by Procedures H and C. ESI-MS m/z 443 (MH+) 00 00 Example 306: (3Z)-l-PHENYL-3-{t4-(3- PYRIDINYL)PHENYL] IMINO}-1,3-DIHYDRO-2H-INDOL-2-ONE: Prepared by Procedures H and C. ESI-MS m/z 376 (MW).
Example 307: [(3-BROMOPHENYL)IMINO]-1-PHENYL-l,3- DIHYDRO-2H-INDOL-2 -ONE: Prepared by Procedure B. ESI-MS m/z 3 7 8(MH 4 Example 308: (3Z) -1,5-DIPHENYL-3-{ [3- (TRIFLUOROMETHYL) PHENYL) IMINO) -1,3-DIHYDRO-2H-INDOL-2- ONE: Prepared by Procedures D, E, and F. ESI-MS m/z 443
(MH+)
Example 309: (3Z)-l-[1,1"-BIPHENYLJ-4-YL-3-{ (3- (TRIFLUOROMETHYL) PHENYL] IMINOI 3-DIHYDRO-2H-INDOL-2o 521 ONE: Prepared by Procedures H (6 eq of aniline), J, and K. ESI-MS m/z 443 (MH).
Example 310: (3Z)-1-(4-HYDROXYPHENYL)-3-[3- 00 5 (TRIFLUOROMETHYL)PHENYL]IMINO}-1,3-DIHYDRO-2H-INDOL-2- SONE: Prepared by Procedures H (6 eq of aniline) and E.
00 S ESI-MS m/z 383 (MH).
Example 311: (3Z)-3-[(3,4-DICHLOROPHENYL)IMINO PYRIDINYLMETHYL)1, 3-DIHYDRO-2H-INDOL-2-ONE: Prepared by Procedures H (75 o, 2 K (3-picolyl chloride), and B.
ESI-MS m/z 383 (MH Examples 91-114 and 254-311 as described above are merely illustrative of the methods used to synthesize indolone derivatives. Further derivatives may be obtained utilizing methods shown in Schemes 6a, 7a and 8-10. The substituents in Schemes 6a, 7a and 8-10 are described in the Detailed Description.
It may be necessary to incorporate protection and deprotection strategies for substituents such as amino, amido, carboxylic acid, and hydroxyl groups in the synthetic methods described above to form indolone derivatives. Methods for protection and deprotection of such groups are well-known in the art, and may be found, for example in Green, T. W. and Wuts, P.G. M. (1991) Protection Groups in Organic Synthesis, 2nd Edition John Wiley Sons, New York.
522 Scheme 6a 1) base 2) A-X y
I
N
B-NH
2 Y y 4
V
Scheme 72
A-R
Cu(OAc) 2 y y 3
B-NH
2 ay~ ,1 Y Y 4 A, and B are defined as described in the specification. X is a leaving group such as Cl, Br, I, or OTs. R is boric acid or a dialkcylborate group.
523 Scheme 88. Synthesis of Isatins Y 1 Y 3
A,
1. (CO) 2 C1 2 2. AlC1 3 y 3 Y4
A
"Y1 1
Y
2
,Y
3 tY 4 A, and B are defined as described in the specification.
X is a leaving group such as Cl, Br, 1, or OTs. R is boric acid or a dialkylborate group.
524 ,,hee 9 a. Synthesis of Substituted Iminoindolones
B-NH,
Base (such as NaH or K2C03), R-X Or For A aryl or heteroaryli A-B (OH), Cu (OAc) Et 3
N
Base (such as NaM or K2C02), R-X or For A -aryl or heteroarylt A-B(OH) 2
CU(QAC)
2 Et.
3
N
B
Y
B-NHN
Y4 X is a leaving group such as a halogen or tosylate.
ayll Y 2
,Y
3
,Y
4 A, and B are defined as described in the specification.
X is a leaving group such as Cl, Br, I, or OTs. R is boric acid or a dialkylborate group.
525 Scheme 10a. Synthesis of Aryl or Heteroaryl-Substituted Iminoindolones 1 N y 3 I N 4 A Ar-B(OH) 2 Pd(PPH 3 4 1
PA
Y4
A
Ar-B (OH) 2 1 Pd (PPH 3 4 Ar-B(OH) 2 Pd(PPH 3 4 Ar aryl or heteroaryl ay 11Y 2
,Y
3
,Y
4 A, and B are defined as described specification. X is a leaving group such as Cl, OTs. R is boric acid or a dialkylborate group.
in the Br, I, or 526 Radioligand Binding of Indolones at Cloned Galanin Receptors The binding properties of invention were evaluated receptors, GAL1, GAL2, described herein.
the indolones of the present at the cloned human galanin and GAL3, using protocols Radioligand Binding Assay Results The indolones described in Examples 91-114 and 254-311 were assayed using cloned human galanin receptors. The compounds were found to be selective for the GAL3 receptor. The binding affinities of the compounds of Examples 91-114 and 254-311 are illustrated in Tables 4 and 4a.
2008200380 25 Jan 2008 Table 4. Binding Affinities of Indolones at Galanin Rleceptors Q 4 4 aN 0 -ubsitio Ki (rnM) R1 R2 R3 R4 RS GaiRi GaIR2 Ga1R3 91 Ph Me H H H >10000 >10000 527 F 92 Ph H CF, H H >10000 >10000 38 93 Ph- H Me H H >10000 >10000 171 94 Ph H Cl H H >10000 >10000 49 Ph H H CF, H >100001 >10000 29 96 Ph H H Me H >10000 >10000 i11 97 Ph H H Cl H >10000 >10000 51 98 Ph H H Br H >10000 >10000 38 99 Ph H H IF H >10000 >10000 229 100 Ph H H OPh H >10000 >10000 101 Ph H H OEt H >10000 >10000 305 102 Ph H H OMe H >10000 >10000 429 103 Ph H Cl H Cl >10000 >10000 68 104 Ph H Me H Me >10000 >10000 143 105 ally1 H Cl Cl H >10000 >10000 97 106 allyl H Cl H Cl >10000 >10000i 62 107 isopropyl. H H Br H >10000 I>100001 126 Key: Ph- Phenyl Me- methyl, OMe- Methaxy OPh= Phenoxy OEt= Ethoxy 00 00 00 528 Table 4a.
529 Table 4a.
530 Table 4a.
00 00 00 531 00 00 00 532 00 00 00 533 00 534 (Ni Table 27
CF
3 00
F
00 276 Qj~0 00 00 00 535 536 Table 4a.
537 Table 4a. 291 N\/271 292 3 0 295 242
N-
69 82_ 538 Table 4a.
539 Table 4a.
00 00 SOral Compositions c-s As a specific embodiment of an oral composition of a V' compound of this invention, 100 mg of one of the compounds described herein is formulated with sufficient finely divided lactose to provide a total amount of 580 00 to 590 mg to fill a size 0 hard gel capsule.
C I. In-Vivo Models 00 A. Materials and Methods C i. Forced Swim Test (FST) The procedure used in this study was similar to that previously described (Porsolt, et al., 1978), except the water depth (30 cm in this procedure). The greater depth in this test prevented the rats from supporting themselves by touching the bottom of the cylinder with their feet. Swim sessions were conducted by placing rats in individual plexiglass cylinders (46 cm tall x 20 cm in diameter) containing 23-25°C water 30 cm deep (Porsolt, et al. used a depth of only 15 cm; also, see Detke, et al., 1995). Two swim tests were conducted always between 1200 and 1800 hours: an initial 15-min pretest followed 24 h later by a 5-minute test. Drug treatments were administered 60 minutes before the 5-minute test period.
All other test sessions were conducted between 1300 to 1700 hours. Following all swim sessions, rats were removed from the cylinders, dried with paper towels and placed in a heated cage for 15 minutes and returned to their home cages. All test sessions were videotaped using a Panasonic color video camera and recorder for scoring later.
00 542
C-I
SAnimals Male Sprague-Dawley rats (Taconic Farms, NY) were used in all experiments. Rats were housed in pairs and maintained on a 12:12-h light-dark cycle. Rats were handled for 5 minutes each day for 5 days prior to 00 behavioral testing.
CI Behavioral Scoring 00 The rat's behavior was rated at 5 second intervals during Ci 10 the 5 minute test as one of the following: 1. Immobility- rat remained floating in the water without struggling and was only making those movements necessary to keep its head above water; 2. Climbing rat was making active movements with its forepaws in and out of the water, usually directed against the walls; 3. Swimming rat was making active swimming motions, more than necessary to merely maintain its head above water, e.g. moving around in the cylinder; and 4. Diving entire body of the rat was submerged.
All of the behavior scoring was done by a single rater, who was blind to the treatment condition. The rater was also present in the room throughout the entire test period.
Drug Administration Animals were randomly assigned to receive a single i.p.
00 543 Sadministration of Example 92 3, 10 or 30 mg/kg, h- dissolved in 100% DMSO), fluoxetine (10 mg/kg, dissolved Sin distilled water) or vehicle (equal mixture of DMSO and distilled water) 30 minutes before the start of the minute test period. All injections were given using 1 cc 00 tuberculin syringe with 26 3/8 gauge needles (Becton- C0 SDickinson, VWR Scientific, Bridgeport, NJ). The volume C1 of injection was 1 ml/kg.
00 C1 10 In another set of experiments, animals were randomly assigned to receive a single p.o. administration of one of the following treatments: Example 151 3 or mg/kg), fluoxetine (5 or 10 mg/kg) or vehicle (1 ml/kg of 100% N,N-dimethylacetamide) 60 minutes before the start of the 5 minute test period. The drugs were dissolved in 100% N,N-dimethylacetamide. All administrations were given using 1 cc tuberculin syringes, to which a 3 inch, curved, stainless steel gavage needle was attached. The volume of administration was 1 ml/kg.
In other sets of experiments, animals were randomly assigned to receive a single p.o. administration of one of the following treatments: Example 103 10 and mg/kg), fluoxetine (10 mg/kg) or vehicle (1 ml/kg of 100% N,N-dimethylacetamide) 60 minutes before the start of the minute test period; or Example 272 (3 mg/kg), fluoxetine (10 mg/kg) or vehicle (1 ml/kg of 100% N,Ndimethylacetamide) 24 hours before the start of the minute test period; or Example 98 10 and 30 mg/kg), fluoxetine (10 mg/kg) or vehicle (1 ml/kg of 100% N,Ndimethylacetamide) 60 minutes before the start of the minute test period; or Example 34 1, 3 and 544 Smg/kg), fluoxetine (10 mg/kg) or vehicle (1 ml/kg of a 100% solution of dimethylacetamide) 60 minutes before the V start of the 5 minute test period; or Example 49 and 30 mg/kg), fluoxetine (10 mg/kg) or vehicle (1 ml/kg of 100% N,N-dimethylacetamide) 60 minutes before the 00 start of the 5 minute test period; or Example 22 Sand 30 mg/kg), fluoxetine (10 mg/kg) or vehicle (1 ml/kg CI of 100% N,N-dimethylacetamide) 60 minutes before the 00 Sstart of the 5 minute test period. The compounds were Ci 10 dissolved in 100% N,N-dimethylacetamide. All administrations were given using 1 cc tuberculin syringes, to which a 3 inch, curved, stainless steel gavage needle was attached. The volume of administration was 1 ml/kg.
The effect of 5 or 10 mg/kg of fluoxetine was utilized in the FST as a positive control.
Data Analysis The forced swim test data (immobility, swimming, climbing, diving) were subjected to a randomized, one-way ANOVA and post hoc tests conducted using the Student- Newman-Keuls test. The data were analyzed using the GBSTAT program, version 6.5 (Dynamics Microsystems, Inc., Silver Spring, MD, 1997). All data are presented as means S.E.M.
2. Social Interaction Test (SIT) Rats were allowed to acclimate to the animal care facility for 5 days and were housed singly for 5 days prior to testing. Animals were handled for 5 minutes per day. The design and procedure for the Social Interaction 545 STest was carried out as previously described by Kennett, et al. (1997). On the test day, weight matched pairs of rats unfamiliar to each other, were given c-I identical treatments and returned to their home cages.
O 5 Animals were randomly divided into 5 treatment groups, 00 with 5 pairs per group, and were given one of the following i.p. treatments: Example 92 (10, 30 or 100 CI mg/kg), vehicle (1 ml/kg) or chlordiazepoxide (5 mg/kg).
00 Dosing was 1 hour prior to testing. Rats were C- 10 subsequently placed in a white perspex test box or arena (54 x 37 x 26 cm), whose floor was divided up into 24 equal squares, for 15 minutes. An air conditioner was used to generate background noise and to keep the room at approximately 74 0 F. All sessions were videotaped using a JVC camcorder (model GR-SZ1, Elmwood Park, NJ) with either TDK (HG ultimate brand) or Sony 30 minute videocassettes. All sessions were conducted between 1:00 4:30 P.M. Active social interaction, defined as grooming, sniffing, biting, boxing, wrestling, following and crawling over or under, was scored using a stopwatch (Sportsline model no. 226, 1/100 sec. discriminability).
The number of episodes of rearing (animal completely raises up its body on its hind limbs), grooming (licking, biting, scratching of body), and face washing hands are moved repeatedly over face), and number of squares crossed were scored. Passive social interaction (animals are lying beside or on top of each other) was not scored.
All behaviors were assessed later by an observer who was blind as to the treatment of each pair. At the end of each test, the box was thoroughly wiped with moistened paper towels.
OO .546 Animals Male albino Sprague-Dawley rats (Taconic Farms, NY) were V housed in pairs under a 12 hr light dark cycle (lights on at 0700 hrs.) with free access to food and water.
00 Drug Administration 0 Example 92 was dissolved in 100% DMSO (Sigma Chemical SCo., St. Louis, MO). Chlordiazepoxide (purchased from 0 Sigma Chemical Co., St. Louis, MO) was dissolved in O 10 double distilled water. The vehicle consisted of DMSO All drug solutions were made up 10 minutes prior to injection and the solutions were discarded.
Example 34 was dissolved in 5% lactic acid, v/v. The vehicle consisted of 100% dimethylacetamide (DMA) and this was used to make up all drug solutions. All drug solutions were made up fresh each day and any unused solutions were discarded at the end of the test day. The volume of drug solution administered was 1 ml/kg.
Data Analysis The social interaction data (time interacting, rearing and squares crossed) were subjected to a randomized, oneway ANOVA and post hoc tests conducted using the Student- Newman-Keuls test. The data were subjected to a test of normality (Shapiro-Wilk test). The data were analyzed using the GBSTAT program, version 6.5 (Dynamics Microsystems, Inc., Silver Spring, MD, 1997). All data are presented as means S.E.M.
00 547 B. Results n 1. Forced Swim Test A. The Effect Of Vehicle, Fluoxetine and Example 92 On Immobility, Climbing and Swimming In The Forced Swim Test 00 Immobility Statistical analysis indicated that there was a 00 significant drug effect [F(4,45) 12.1, p 0.0001] on immobility. Subsequent post hoc analysis revealed that a single injection of 10 mg/kg i.p. of fluoxetine significantly decreased immobility to 21.0 0.9 (Student-Newman-Keuls value was 36.5, p 0.01) compared to vehicle-treated controls (Table 5 and Figure In addition, a single injection of either 3 or 10 mg/kg i.p.
of Example 92 significantly decreased immobility (24 1.1 24 0.8 counts at each dose, respectively) compared to vehicle-treated controls 30 1.2 (Student- Newman-Keuls values of 16.8 and 15.7, respectively) (Table 5 and Figure No significant effects on immobility were observed with Example 92 at 30 mg/kg i.p.
(Table 5 and Figure 1).
Climbing The statistical analysis of the climbing counts indicated that there was a significant drug effect [F(4,45) 4.4, p 0.004]. Post hoc analysis indicated that a single injection of 10 mg/kg of fluoxetine did not significantly alter climbing counts compared to vehicle-treated animals (Table 5 and Figure In contrast, a single injection of 10 mg/kg of Example 92 produced a significant increase (16.8 0.6) in climbing counts (Student-Newman-Keuls value 11.6, p 0.01) compared to vehicle-treated 0 548 Sanimals (12 Example 92 dosed at 1, 3 30 mg/kg c- did not significantly alter climbing.
ci Swimming O 5 The statistical analysis of the swimming data indicated 00 0 that there was a significant drug effect [F(4,45) 6.6, op 0.0001] (Table 5 and Figure 3) The post hoc test C showed that a single injection of 10 mg/kg i.p. of 00 0 fluoxetine produced a significant increase (25 1.2) in CI 10 swimming counts over the vehicle treated animals, 18 1 (Student-Newman-Keuls value of 19.9, p 0.01): In contrast, a single injection of 1, 3 or 10 mg/kg i.p. of Example 92 did not significantly alter swimming counts 1.1, 21 18 0.9, respectively (Table 5 and Figure 3) (However, at 30 mg/kg i.p. Example 92 significantly increased swim behavior in the rat, comparable to fluoxetine at 10 mg/kg i.p. (27 2.5 vs.
1.2, Table 5 and Figure 3).
Diving This behavior was rarely observed following a single injection of vehicle (0.1 0.1, one animal dove once), fluoxetine (0.1 0.1, one animal out of 10 dove once), 1 mg/kg of Example 92 (0.6 0.2; 5 animals had counts of 2, 1, 1, 1, and 3 mg/kg of Example 92 (0.6 0.3; 3 animals had counts of 3, 2 and. 1) or 10 mg/kg of Example 92 (0.5 0.5; note: only one animal at this dose showed diving behavior and the score was At 30 mg/kg i.p.
of Example 92 diving behavior was only observed in two animals (mean 0.2 Thus there was no significant drug effect on diving [F(4,45) 0.77, p 0.55].
549 Table 5. The effect of a single injection of vehicle, fluoxetine and Example 92 on immobility, climbing and swimming in the rat Forced Swim Test.
Treatment Dose (mg/kg) Immobility Climbing Swimming Vehicle 30 1.2 12.0 0.8 18 1 Fluoxetine 10 21 0.9a 14.3 0.9 25 1.2 b Example 92 1 28 1.0 11.7 1.1 20 1.1 Example 92 3 24 1.1 a 14.6 1.5 21 0.9 Example 92 10 24 0.8 a 16.8 0.6 c 18 0.9 Example 92 30 25 3.5 8.6 1.7 27 2 Each value represents the mean number of counts per seconds S.E.M in a 5 minute observation period.
a Significantly less than Vehicle on immobility scores, p <0.01, ANOVA and Student-Newman-Keuls test.
b Significantly greater than Vehicle and 1,3 10 of Example 92, on swim scores, p 0.01, ANOVA and Student-Newman-Keuls.
c Significantly greater than vehicle and 1, 3 30 mg/kg dose of Example 92 on climbing scores, p 0.01, ANOVA and Student-Newman-Keuls d Significantly greater than Vehicle, 1, 3 and 10 mg/kg i.p. of Example 92 on swim scores,p 0.01, ANOVA and Student-Newman-Keuls test.
00 550 The results of the Forced Swim Test indicate that using a In modified version of the Lucki forced swim test, a single injection of 10 mg/kg i.p. of fluoxetine produced a significant decrease in immobility and an increase in 00 swimming in male Sprague-Dawley rats. This is consistent Swith findings from previous studies using the Lucki CI version (Detke, et al., 1995; Kirby and Lucki, 1997; 00 SLucki, 1997; Page, et al., 1999; Reneric and Lucki, 1998). In addition, the results obtained using fluoxetine are consistent with those using other SSRIs (Detke, et al., 1995). Thus, a modified version of the Lucki forced swim test can consistently detect the antidepressant action of SSRIs such as fluoxetine.
Interestingly, at doses of 3 and 10 mg/kg Example 92, significantly decreased immobility compared to vehicle-treated animals. The magnitude of the decrease was not significantly different than that of fluoxetine.
Thus, based on past interpretations of the Forced Swim Test, our results suggest that Example 92 has antidepressant-like properties.
A single injection of either 1, 3 or 10 mg/kg i.p. of Example 92 did not significantly alter swimming behavior.
This is in contrast to the results obtained with fluoxetine, which increased swimming at 10 mg/kg i.p.
Previously, it has been reported that compounds which selectively block serotonin uptake significantly increase swimming but not climbing whereas selective NE uptake blockers significantly increase climbing but not swimming behavior (Reneric and Lucki, 1998). Thus, the present 00 0 551 Sfindings suggest that Example 92 exhibits a profile similar to NE and selective serotonin reuptake inhibitors I (SSRIs) depending on the dose tested.
Finally, as previously reported by Lucki, diving behavior 00 was rarely observed in vehicle or fluoxetine-treated Sanimals (1 dive in one rat for each group). Example 92 at CI all doses tested did not produce a significant effect on 00 Sdiving behavior. It is possible that antidepressant eC 10 drugs do not induce diving behavior.
In conclusion, compared to vehicle-treated animals, Example 92, at doses of 3 and 10 mg/kg, produced a significant decrease in immobility and a significant increase in climbing at the 10 mg/kg dose. At i.p. Example 92 produced a significant increase in swimming behavior comparable with that observed with the antidepressant fluoxetine, thus supporting the antidepressant-like profile of Example 92.
B. The effect of Example 151, fluoxetine, and vehicle on swimming, climbing, immobility, and diving in the forced swim test.
Immobility Statistical analysis indicated a significant effect of treatment on immobility (ANOVA, F(5,46) 3.5, p 0.0095). Post hoc analyses revealed that a single p.o.
administration of 10 mg/kg of fluoxetine significantly decreased immobility (Fisher's LSD value of 2.9) compared to vehicle-treated animals (Table 5a). In contrast, a single p.o. administration of 5 mg/kg of fluoxetine did 00 S552 Snot significantly alter immobility compared to vehicle- -s treated animals.
A single p.o. administration of 1 mg/kg of Example 151 O 5 did not significantly alter immobility compared to 0 vehicle-treated animals (Table 5a). In contrast, a Ssingle p.o. administration of either 3 or 10 mg/kg of C Example 151 significantly decreased immobility compared 00 Sto animals treated with vehicle (Fisher's LSD values of C( 10 2.8 and 2.6, respectively) or 5 mg/kg p.o. of fluoxetine (Fisher'g LSD values of 2.6 and 2.4, respectively).
There was no significant difference in the reduction in immobility between 10 mg/kg of fluoxetine and 3 and mg/kg of Example 151.
Swimming Statistical analysis indicated a significant treatment effect on swimming behavior (ANOVA, F(5,46) 5.5, p 0.0005). Post hoc analyses revealed that a single p.o.
administration of 10 mg/kg of fluoxetine produced a significant increase in swimming behavior compared to vehicle-treated animals (Student-Newman-Keuls value of 16.8 (Table 5a). In contrast, a single p.o.
administration of 5 mg/kg of fluoxetine did not significantly alter swimming compared to vehicle-treated animals.
A single p.o. administration of either 1, 3 or 10 mg/kg of Example 151 significantly increased swimming (Student- Newman-Keuls values of 6.9, 14.8 and 13.4, respectively) compared to vehicle-treated animals. There was no significant difference in the magnitude of the increase r I 00 S553 in swimming between the doses of Example 151. The 3 and -s 10 mg/kg doses of Example 151 produced a significantly I greater increase in swimming compared to animals treated with 5 mg/kg p.o. of fluoxetine. There was no significant difference in the increase in swimming 00 between animals treated with 10 mg/kg of fluoxetine and those treated with Example 151.
00 SClimbing behavior r- 10 Statistical analysis revealed that climbing was not significantly altered by a single p.o administration of 1, 3 or 10 mg/kg of Example 151 or 5 or 10 mg/kg of fluoxetine compared to vehicle-treated animals (ANOVA, F(5,46) 0.81, p 0.55)(Table Diving Statistical analysis revealed that diving was not significantly altered by a single p.o. administration of 1, 3 or 10 mg/kg of Example 151 or 5 or 10 mg/kg of fluoxetine compared to vehicle-treated animals (ANOVA, F(5,46) 0.36, p 0.87)(Table 554 TABLE 5a. The effect of a single p.o. administration of vehicle, 1, 3 and 10 mg/kg of Example 151 and 5 and mg/kg of fluoxetine on immobility, climbing, diving and swimming in the forced swim test in rats.
male Sprague-Dawley Treatment Immobility Climbing Swimming Diving Vehicle 46 1.8 2.7 0.7 11.4 0.4 1.2 0.4 1 mg/kg EX151 41 2.0 2.3 0.6 16.8 0.2 1.4 d 0.2 3 mg/kg.EX151 38 2.0 a 2.4 0.5 19.5 .0.3 0.2 mg/kg EX151 39 1.Bb 2.2 0.5 18.9 0.3 e 0.2 mg/kg Fluox 45 1.3c 1.2 0.4 13.9 0.0 0.0 mg/kg Fluox 38 2.3 a 2.0 0.6 19.8 0.6 1.8e 0.6 Each value represents animals were examined Fluoxetine, EX151 the mean for each Example S.E.M. A total of 8-9 treatment group. Fluox 151. Experiments were conducted 1 hr. after the appropriate treatment.
aSignificantly less than Vehicle Fisher's protected t test.
bSignificantly less than Vehicle Fisher's protected t test.
(p 0.01), ANOVA and (p 0.05), ANOVA and Significantly greater than 3 and 10 mg/kg of Example 151 and 10 mg/kg of fluoxetine, ANOVA and ANOVA and Fisher's protected t test.
00 555 1 dSignificantly greater than Vehicle (p 0.05) and 5 mg/kg of fluoxetine(p 0.05), ANOVA and Student-Newman-Keuls Stest.
eSignificantly greater than Vehicle (p 0.01) and 5 mg/kg 00 of fluoxetine(p 0.05), ANOVA and Student-Newman-Keuls Stest.
00 SThe results of this study indicate that a single p.o.
administration of Example 151, at doses of 1,3 and mg/kg, produces a significant increase in swimming behavior. There was no significant difference in the magnitude of the increase in swimming between the doses of Example 151, although the 1 mg/kg dose produced a lower increase. In contrast, only the 3 and 10 mg/kg doses of Example 151 significantly decreased immobility compared to vehicle-treated animals. Thus, it appears that a single p.o. administration of either 3 or mg/kg, compared to 1 mg/kg of Example 151, produce a more robust antidepressant profile in the FST in male Sprague- Dawley rats. Our results also indicate that Example 151 produced changes in swimming and immobility that were not significantly different from that of 10 mg/kg p.o. of fluoxetine. This suggests that Example 151 produces behavioral effects similar to that of 10 mg/kg of fluoxetine in the FST.
A single p.o. administration of 5 mg/kg of fluoxetine did not significantly alter swimming, climbing, diving or immobility compared to vehicle treated animals. This finding, together with the data indicating that 10 mg/kg of fluoxetine produces a significant effect on swimming 556 c-I Sand immobility in the FST, suggest that the threshold dose of fluoxetine is greater than 5, but less than mg/kg. This is consistent with ex vivo data indicating that a p.o. dose of 7 mg/kg of fluoxetine is required to inhibit 5-HT uptake in the CNS by 50% (Leonard, 1996).
00 SIn conclusion, the results of this study indicate that a C- single p.o. administration of Example 151 (particularly 00 the 3 and 10 mg/kg doses) produces behavioral effects in C 10 the FST in rats that resemble those of antidepressants.
C. The Effect of a Single P.O. Administration of Example 103, Fluoxetine and Vehicle on Swimming, Immobility, Climbing and Diving in the Forced Swim Test Immobility Statistical analysis indicated a significant effect of treatment on immobility (ANOVA, F(4,40) 6.3, p 0.0005). Post hoc analyses revealed that a single p.o.
administration of 10 mg/kg of fluoxetine significantly decreased immobility (Student-Newman-Keuls value of 8.3) compared to vehicle-treated animals (Table 5b). The decrease in immobility produced by fluoxetine was significantly greater than that of either 3 or 10 mg/kg p.o. of Example 103 (Student-Newman-Keuls values of 9.1 and 6.1,-respectively).
A single p.o. administration of either 3 or 10 mg/kg of Example 103 did not significantly alter immobility compared to vehicle-treated animals. However, the mg/kg dose of Example 103 produced a significant decrease in immobility (Student-Newman-Keuls values of 13.9) 00 0 557 Scompared to vehicle-treated animals. In addition, the h- decrease in immobility produced by 30 mg/kg of Example ln 103 was significantly greater than that of 3 and 10 mg/kg of Example 103 (Student-Newman-Keuls values of 14.4 and 10.6, respectively). There was no significant difference 00 between fluoxetine and 30 mg/kg of Example 103 in the Sreduction of immobility.
00 SSwimming (C 10 Statistical analysis indicated a significant treatment effect "on swimming behavior (ANOVA, F(4,40) p 0.0001). Post hoc analyses revealed that a single p.o.
administration of 10 mg/kg of fluoxetine produced a significant increase in swimming behavior compared to animals treated with vehicle, 3 or 10 mg/kg p.o. of Example 103 (Student-Newman-Keuls values of 14.9, 15.3 and 11.6, respectively) (Table A single p.o. administration of either 3 or 10 mg/kg of Example 103 did not significantly alter swimming behavior compared to vehicle-treated animals. A single p.o.
administration of 30 mg/kg of Example 103 produced a significantly greater increase -in swimming behavior compared to animals treated with either vehicle, 3 or mg/kg of Example 103 (Student-Newman-Keuls values of 18, 18.6 and 14.5 respectively).
Climbing behavior Statistical analysis revealed that diving was not significantly altered by a single p.o. administration of 3, 10 or 30 mg/kg of Example 103 or 10 mg/kg of 00 S558
(N
Sfluoxetine compared to vehicle-treated animals (ANOVA, F(4,40) 1.2, p 0.31) (Table Diving o 5 Statistical analysis revealed that diving was not 00 significantly altered by a single p.o. administration of 3, 10 or 30 mg/kg of Example 103 or 10 mg/kg of C1 fluoxetine compared to vehicle-treated animals (ANOVA, 00 0F(4,40) 1.1, p 0.36) (Table C00
(N
559 TABLE 5b. The effect of a single p.o. administration of vehicle, 10 mg/kg of fluoxetine and 3, 10 or 30 mg/kg of Example 103 on immobility, climbing, diving and swimming in the forced swim test in male Sprague-Dawley rats.
Treatment Immobility Climbing Swimming Diving Vehicle 44 1.7 2.9 13.1 0.4 0.7 1.2 0.2 3 mg/kg EX103 44 2.7 2.8 13.2 0.5 0.6 1.9 0.4 mg/kg EX103 42 2.2 3.5 14.3 0.4 0.6 1.6 0.2 EX103 32 1.8a 4.8 22.7 1.1 0.7 1.1 C mg/kg Fluox 34 2.3 b 3.8 21.8 0.1 0.8 1.4
C
0.1 Each value represents the animals were examined for mean S.E.M. A total each treatment group.
of 8-10 Fluox Fluoxetine, EX103 Example 103. Experiments were conducted 1 hr. after the appropriate treatment.
aSignificantly less than Vehicle, 3 and 10 mg/kg of Example 103, p 0.01, ANOVA and Student-Newman-Keuls test.
bSignficantly less than Vehicle, 3 and 10 mg/kg of Example 103, p 0.05, ANOVA and Student-Newman-Keuls test.
00 0 560 C CSignficantly greater than Vehicle, 3 and 10 mg/kg of Example 103, P 0.01, ANOVA and Student-Newman-Keuls C< test.
0 5 The results of this study indicated that as previously 00 CMr reported, a single p.o. administration of 10 mg/kg of fluoxetine produced a significant increase in swimming 00 and a significant decrease in immobility in male rats in the FST compared to vehicle-treated animals. The
C
10 magnitude of these changes are similar to those reported of our past studies with 10 mg/kg p.o. of fluoxetine. In contrast, neither climbing nor diving behavior was significantly altered by a single p.o. administration of mg/kg of fluoxetine.
A single p.o. administration of either 3 or 10 mg/kg of Example 103 did not significantly alter swimming, climbing, immobility or diving in male rats in the FST, indicating that at these doses using the p.o. route, Example 103 does not exhibit antidepressant action in the FST. In contrast, a single p.o. administration of mg/kg of Example 103 produced a significant increase in swimming and a significant decrease in immobility compared to animals treated with vehicle or 10 mg/kg of Example 103. However, the 30 mg/kg p.o. dose of Example 103 did not significantly alter diving or climbing counts compared to vehicle-treated animals. The increase in swimming counts produced by 30 mg/kg p.o. of Example 103 was comparable to that for 10 mg/kg of fluoxetine.
In conclusion, a single p.o. administration of 30 mg/kg of Example 103 (one hour before the last swim test) 00 S561 Sincreases swimming and decreases immobility counts in the FST, suggesting that Example 103 has antidepressant n properties.
D. Effect of a single p.o. administration of Example 272, OO fluoxetine and vehicle on swimming, climbing, immobility 0 and diving in the forced swim test 00 SImmobility C( 10 Statistical analysis indicated a significant effect of treatmeht on immobility (ANOVA, F(2,27) 5.2, p 0.0126). Post hoc analyses revealed that a single p.o.
administration of 10 mg/kg of fluoxetine and 3 mg/kg of Example 272 significantly decreased immobility (Student- Newman-Keuls values of 5.4 and 9.8, respectively) compared to vehicle-treated animals (Table 5c). There was no significant difference between fluoxetine and 3 mg/kg of Example 272 in the reduction of immobility (Student-Newman-Keuls value of 0.53).
Swimming Statistical analysis indicated a significant treatment effect on swimming behavior (ANOVA, F(2,27) 9.9, p 0.0007). Post hoc analyses revealed that a single p.o.
administration of 10 mg/kg of fluoxetine and Example 272 produced a significant increase in swimming behavior compared to animals treated with vehicle (Student- Newman-Keuls values of 11.9 and 17.5, respectively) (Table 5c). There was no significant difference in the increase in swimming between 10 mg/kg of fluoxetine and 3 mg/kg of Example 272 (Student-Newman-Keuls value of 0.42).
Climbing behavior ln Statistical analysis revealed that diving was not significantly altered by a single p.o. administration of O 5 either 3 mg/kg of Example 272 or 10 mg/kg of fluoxetine 00 compared to vehicle-treated animals (ANOVA, F(2,27) S1.8, p 0.19)(Table 00 SDiving C1 10 Statistical analysis revealed that diving was not significantly altered by a single p.o. administration of 3 mg/kg of Example 272 or 10 mg/kg of fluoxetine compared to vehicle-treated animals (ANOVA, F(2,27) 0.65, p 0.53) (Table 00 S563 STABLE 5c. The effect of a single p.o. administration of vehicle, fluoxetine and Example 272 on immobility, climbing, diving and swimming in the forced swim test in male Sprague-Dawley rats.
0 0 Treatment Immobility Climbing Swimming Diving O Vehicle 43 3.3 2.4 0.4 13.4 2.2 0.2 (C 0.1 00 S3 mg/kg 33 1.8 a 3.9 0.6 22.9 1.3 0.6 C EX272 0.4 mg/kg 35 1.7 a 3.3 0.6 21.4 1.0° 0.2 FLUOX 0.1 Each value represents the mean S.E.M. A total of 9-10 animals were examined for each treatment group.
Abbreviations: FLUOX Fluoxetine, EX272 Example 272.
Animals received 1 p.o. administration of the appropriate treatment 24 hours before the test day.
aSignificantly less than Vehicle, p 0.05, ANOVA and Student-Newman-Keuls test.
bsignificantly less than Vehicle, p 0.01, ANOVA and Student-Newman-Keuls test.
The finding of this study indicate that a single p.o.
administration of 3 mg/kg of the compound Example 272 produced a significant increase in swimming and a significant decrease in immobility 24 hours after administration compared to vehicle-treated animals.
However, the administration of Example 272 did not significantly alter climbing or diving compared to 00 S564 Svehicle-treated animals. These results are similar to -s those of a single p.o. administration of 10 mg/kg of Sfluoxetine. Our finding suggest that a single p.o.
administration of 3 mg/kg of Example 272 has the profile of an antidepressant in male Sprague-Dawley rats in the 0 Lucki version of the FST.
CI E. Effect of a single p.o. administration of Example 98, 00 Sfluoxetine and vehicle on swimming, climbing, immobility C 10 and diving in the forced swim test.
Immobility Statistical analysis indicated a significant effect of treatment on immobility (ANOVA, F(4,43) 7.5, p 0.0001). Post hoc analyses revealed that a single p.o.
administration of 10 mg/kg of fluoxetine significantly decreased immobility (Student-Newman-Keuls value of 23.8) compared to vehicle-treated animals (Table A single p.o. administration of 3, 10 or 30 mg/kg of Example 98 significantly decreased immobility compared to vehicle-treated animals (Student-Newman-Keuls values of 19.3, 9.7 and 13.7, respectively). There was no significant difference between fluoxetine and 3, 10 or mg/kg of Example 98 in the magnitude of the reduction of immobility. There were no significant differences between the doses of Example 98 regarding the magnitude of the decrease in immobility.
Swimming Statistical analysis indicated a significant treatment effect on swimming behavior (ANOVA, F(4,43) 11, p 00 S565 S0.0001). Post hoc analyses revealed that a single p.o.
c- administration of 10 mg/kg of fluoxetine produced a n significant increase in swimming behavior compared to vehicle-treated animals (Student-Newman-Keuls value of 35.1) (Table 00 SA single p.o. administration of 3, 10 or 30 mg/kg of C Example 98 significantly increased swimming compared to 00 vehicle-treated animals (Student-Newman-Keuls values of C< 10 24.4, 14.7 and 25.1, respectively) (Table 5d). There was no significant difference between fluoxetine and 3, 10 or mg/kg of Example 98 in the magnitude of the increase in swimming. There were no significant differences between the doses of Example 98 regarding the magnitude of the increase in immobility.
Climbing behavior There was a significant treatment effect on climbing behavior (ANOVA, F(4,43) 2.8, p 0.04) (Table Post hoc tests indicated that this was the result of the 3 mg/kg dose of Example 98 producing a significantly greater increase in climbing compared to 30 mg/kg of Example 98 (Table 5d; Student-Newman-Keuls value of 8.6).
There was no significant difference in the number of climbing counts between animals treated with vehicle and Example 98.
Diving Statistical analysis revealed that diving was not significantly altered by a single p.o. administration of 3, 10 or 30 mg/kg of Example 98 or 10 mg/kg of fluoxetine 566 compared to vehicle-treated animals (ANOVA, F(4,43) 1.29, p 0.29) (Table TABLE 5d. The effect of a single p.o. administration of vehicle, 10 mg/kg of fluoxetine and 3, 10 or 30 mg/kg of Example 98 on immobility, climbing, diving and swimming in the forced swim test in male Sprague-Dawley rats.
Treatment Immobility Climbing Swimming Diving Vehicle 48 1.2 2.5 0.5 8.8 0.9 0.4 0.3 3 mg/kg EX98 35 2 .6a 4.3 20.4 0.1 0.9 b 1.9 C 0.1 mg/kg 39 I.
1 a 2.4 0.3 17.6 0.8 EX98 1.0c 0.4 mg/kg 38 2.3 a 2.0 0.3 20.3 0.2 EX98 2. c 0.2 mg/kg 34 3.0 a 3.4 0.8 22.8 0.1 Fluox 2.2c 0.1 Each value represents the mean S.E.M. A total of animals were examined for each treatment group, except for the fluoxetine and 3 mg/kg groups, where a total of 9 animals were examined. Vehicle 100% DMA.
Fluox Fluoxetine, EX98 Example 98. Experiments were conducted 1 hr. after the appropriate treatment.
aSignificantly less than Vehicle, p 0.01, ANOVA and Student-Newman-Keuls test.
00 S567 bSignificantly greater than 30 mg/kg of Example 98, p Cl 0.05, ANOVA and Student-Newman-Keuls test.
CSignificantly greater than Vehicle, p 0.01, ANOVA and Student-Newman-Keuls test.
0 00 The results of this study clearly indicate that in male SSprague-Dawley rats, a single p.o. administration of 3, 10 or. 30 mg/kg of Example 98 produces a significant increase in swimming and a significant decrease in immobility compared to vehicle-treated animals in the FST. In addition, the Example 98 induced alterations were similar in magnitude to that of a single p.o.
administration of 10 mg/kg p.o. of fluoxetine. However, neither fluoxetine nor Example 98 produced a significant alteration in climbing or diving compared to vehicletreated animals.
In conclusion, these results indicate that a single p.o.
administration of Example 98 produces a profile in the modified Lucki version of the FST resembling that of the clinically established antidepressant fluoxetine.
F. Effect of a single p.o. administration of Example 34, fluoxetine and vehicle on swimming, climbing, immobility and diving in the forced swim test.
Immobility Statistical analysis indicated a significant effect of treatment on immobility (ANOVA, F(5,44) 18.1, p 0.0001). Post hoc analyses revealed that a single p.o.
00 S568 Sadministration of 10 mg/kg of fluoxetine significantly h- decreased immobility (Student-Newman-Keuls value of 39.6) n compared to vehicle-treated animals (Table Fluoxetine also produced a significantly greater decrease in immobility compared to the 0.3 and 10 mg/kg doses of 0 0 Example 34 (Student-Newman-Keuls values of 15.3 and 29.8, Srespectively). There was no significant difference in the CI magnitude of the decrease in immobility between 00 Sfluoxetine and the 1 and 3 mg/doses of Example 34.
C- A single p.o. administration of 0.3, 1 and -3 mg/kg of Example 34 significantly decreased immobility compared to vehicle-treated animals (Student-Newman-Keuls values of 7.03, 41.6 and 42.0, respectively)(Table 5e). However, a single p.o. administration of 10 mg/kg of Example 34 did not significantly decrease in immobility compared to vehicle-treated animals. The magnitude of the decrease in immobility produced by 1 and 3 mg/kg doses of Example 34 was significantly greater than that for the 0.3 (Student-Newman-Keuls values of 14.5 and 15.3) and mg/kg (Student-Newman-Keuls of 30.6 and 31.3, respectively) doses of Example 34 (Student-Newman-Keuls of 21.3 and 10.8, respectively).
Swimming Statistical analysis indicated a significant treatment effect on swimming behavior (ANOVA, F(5,44) 33.0, p 0.0001). Post hoc analyses revealed that a single p.o.
administration of 10 mg/kg of fluoxetine produced a significant increase in swimming compared to animals treated with vehicle, 0.3 or 10 mg/kg of Example 34 (Student-Newman-Keuls values of 73.7, 30.0 and 53.9, 00 S569 Srespectively) (Table 5e). There was no significant difference in swimming behavior between fluoxetine and
V)
n the 1 and 3 mg/kg p.o. of Example 34.
O 5 A single p.o. administration of either 0.3, 1 or 3 mg/kg 0 0 of Example 34 produced a significant increase in swimming Obehavior compared to vehicle-treated animals (Student- C<1 Newman-Keuls values of 12.1, 72.1 and 80.3, respectively) 00 S(Table 5e). In addition, the magnitude of the increase C' 10 in swimming was greater for the 1 and 3 mg/kg doses (Student-Newman-Keuls values of 50.4 and 57.9, respectively) compared to 0.3 mg/kg of Example 34.
Climbing behavior Statistical analysis indicated a significant treatment effect on swimming behavior (ANOVA, F(5,44) p 0.014) (Table 5e) Post hoc analyses revealed that a single p.o. administration of 1 mg/kg of Example 34 produced a significant increase in climbing compared to vehicle-treated animals (Student-Newman-Keuls value of 9.2) (Table Diving Statistical analysis revealed that diving was not significantly altered by a single p.o. administration of 0.3, 1, 3 or 10 mg/kg of Example 34 or 10 mg/kg' of fluoxetine compared to vehicle-treated animals (ANOVA, F(5,44) 0.75, p 0.59)(Table
L
570 TABLE 5e. The effect of a single p.o. administration of vehicle, 10 mg/kg of fluoxetine and Example 34 on immobility, climbing, diving and swimming in the forced swim test in male Sprague-Dawley rats.
Treatment Immobility Climbing Swimming Diving Vehicle 52 1.3 2.1 0.6 6.0 0.6 0.8 0.7 0.3 mg/kg 45 1.5 a 3.3 0.7 11.6 0.2 EX34 0.9d 0.1 1 mg/kg 35 1.9 5.0 0.8 C 19.6 0.3 EX34 1.
3 d ,e 0.2 3 mg/kg 35 2 .0 4.3 0.8 20.8 0.3 EX34 1.3'd,e 0.3 mg/kg 49 1.4 2.0 0.4 8.2 i 1.2 0.4 EX34 0.3 mg/kg 34 3.3b 4.5 1.2 21.3 1.0 Fluox 1.
8 d e 0.8 Each value represents the mean S.E.M. A total of 9 animals were examined for each treatment group, except for the 3 mg/kg Example 34 and fluoxetine groups, were a total of 8 and 6 animals were examined, respectively.
Fluox Fluoxetine, EX34 Example 34. Experiments were conducted 1 hr. after the appropriate treatment.
aSignificantly less than Vehicle, p 0.05, ANOVA and Student-Newman-Keuls test.
00 0 571 Sbsignificantly less than Vehicle, 0.3 and 10 mg/kg of Example 34, ANOVA and Student-Newman-Keuls test.
CSignificantly greater than Vehicle, p 0.05, ANOVA and 00 5 Student-Newman-Keuls test.
Cq dignificantly greater than Vehicle (p 0.01) and 00 Smg/kg Example 34 (all p 0.01 except for 0.3 mg/kg of CI Example 34, p 0.05), ANOVA and Student-Newman-Keuls test.
eSignificantly greater 0.3 mg/kg of Example 34, p 0.05, ANOVA and Student-Newman-Keuls test.
The results of this study indicate that a single p.o.
administration (one hour before the final swim test) of either 0.3, 1 .or 3 mg/kg of Example 34 produced a significant increase in swimming and a significant decrease in immobility compared to vehicle-treated animals. However, a single p.o. administration of mg/kg of Example 34 did not significantly alter swimming or climbing compared to vehicle-treated animals.
Currently, the explanation for the lack of effect of mg/kg p.o. of Example 34 is unknown. The 1 mg/kg dose of Example 34 produced a significant increase in climbing compared to vehicle-treated animals. The magnitude of the alterations in swimming and immobility produced by 1 and 3 mg/kg p.o. of Example 34 was significantly greater than that for the 0.3 and 10 mg/kg doses of Example 34.
Finally, none of the doses of Example 34 significantly 00 572 Saltered diving behavior compared to vehicle-treated c controls.
As previously reported, a single p.o. administration of 0 5 10 mg/kg of fluoxetine produced a significant increase in 00 M swimming and a significant decrease in immobility compared to vehicle-treated controls. The effect of 00 fluoxetine on swimming and immobility was similar to that Sfor the 1 and 3 mg/kg doses of Example 34 but was C< 10 significantly greater than that of 0.3 and 10 mg/kg of Example 34. A single p.o. administration of 10 mg/kg of fluoxetine did not significantly alter climbing or diving behavior compared to vehicle-treated controls.
In conclusion, these results indicate that a single p.o.
administration of 0.3, 1 or 3 mg/kg Example 34 produces an effect in the FST that resembles that of antidepressants in male Sprague-Dawley rats.
G. Effect of a single p.o. administration of Example 49, fluoxetine and vehicle on swimming, climbing, immobility and diving in the forced swim test.
Immobility Statistical analysis indicated a significant effect of treatment on immobility (ANOVA, F(4,41) 6.5, p 0.0004). Post hoc analyses revealed that a single p.o.
administration of 10 mg/kg of fluoxetine significantly decreased immobility (Student-Newman-Keuls value of 15.6) compared to vehicle-treated animals (Table 00 S573 SA single p.o. administration of either 3 or 10 mg/kg of Example 49 did not significantly alter immobility compared to vehicle-treated animals. However, the mg/kg dose of Example 49 produced a significant decrease O 5 in immobility (Student-Newman-Keuls values of 00 Scompared to vehicle-treated animals. In addition, the Sdecrease in immobility produced by either fluoxetine or C 30 mg/kg of Example 49 was significantly greater than 0that of the 10 mg/kg dose of Example 49. There was no C 10 significant difference between fluoxetine and 30 mg/kg of Example 49 in the reduction-of immobility.
Swimming Statistical analysis indicated a significant treatment effect on swimming behavior (ANOVA, F(4,41) 16.2, p 0.0001). Post hoc analyses revealed that a single p.o.
administration of 10 mg/kg of fluoxetine produced a significant increase in swimming behavior compared to animals treated with vehicle, 3, 10 or 30 mg/kg p.o. of Example 49 (Student-Newman-Keuls values of 42.7, 20.9, 47.5 and 8.4, respectively) (Table A single p.o. administration of either 3 or 10 mg/kg of Example 49 did not significantly alter swimming behavior compared to vehicle-treated animals. A single p.o.
administration of 30 mg/kg of Example 49 produced a significantly greater increase in swimming behavior compared to animals treated with vehicle, 3 or 10 mg/kg of Example 49 (Student-Newman-Keuls values of 14 and 16.9, respectively).
00 0574
(N
SClimbing behavior h- There was a significant treatment effect on climbing t behavior (ANOVA; F(4,42) 5.9, p 0.007). Post hoc tests indicated that this was the results of the vehicle, 3, 10 and 30 mg/kg doses of Example 49 producing a 00 significantly greater increase in climbing counts Scompared to fluoxetine-treated animals (Table C Student-Newman-Keuls values of 7.9, 18.1, 14.05 and 12.9, 00 Srespectively). There was no significant difference in the number of climbing counts between animals treated with vehicle and Example 49.
Diving Statistical analysis revealed that diving was not significantly altered by a single p.o. administration of 3, 10 or 30 mg/kg of Example 49 or 10 mg/kg of fluoxetine compared to vehicle-treated animals (ANOVA, F(4,41) 1.06, p 0.38)(Table 575 TABLE 5f. The effect of a single p.o. administration of vehicle, 10 mg/kg of fluoxetine and 3, 10 or 30 mg/kg of Example 49 on immobility, climbing, diving and swimming in the forced swim test in male Sprague-Dawley rats.
Treatment Immobility Climbing Swimming Diving Vehicle 47 1.2 1.8 0.3 10.6 1.1 0.2 0.2 3 mg/kg EX 43 1.9 3.0 0.7 13.1 1.4 1.0 49 0.7 mg/kg 48 1.7 2.4 0.7 10.0 1.0 0.0 EX49 0.0 mg/kg 41 2.3 0.4 16.7 0.4 EX49 2.0 a 1.3 d 0.4 mg/kg 38 0.0 21.6 0.8 Fluox 1.3 b 0.0 1.1e Each value represents animals were examined the mean S.E.M. A total of for each treatment group, except for the fluoxetine and 3 mg/kg groups, where a total of 9 and 7 animals were examined, respectively.
Fluox Fluoxetine, EX49 Example 49. Experiments were conducted 1 hr. after the appropriate treatment.
"Significantly less than Vehicle and 10 mg/kg of Example 49, p 0.05, ANOVA and Student-Newman-Keuls test.
bSignficantly less than Vehicle and 10 mg/kg of Example 49, p 0.01, ANOVA and Student-Newman-Keuls test.
S576 SCSignficantly less than all other treatment groups, p c' 0.01, ANOVA and Student-Newman-Keuls test.
ci dSignficantly greater than vehicle and 10 mg/kg of Example 49, p 0.01, ANOVA and Student-Newman-Keuls test.
00 0 eSignficantly greater than all other treatment groups, p N0 0.01, ANOVA and Student-Newman-Keuls test.
00 Cl 10 The results of this study indicated that as previously reported, a single p.o. administration of 10 mg/kg of fluoxetine produced a significant increase in swimming and a significant decrease in immobility in male rats in the FST compared to vehicle-treated animals. The magnitude of these changes are similar to those reported of our past studies with 10 mg/kg p.o. of fluoxetine. In contrast, climbing behavior was significantly decreased by a single p.o. administration of 10 mg/kg of fluoxetine compared to all other treatment groups. However, this could be related to the fact that fluoxetine has a much greater effect on swimming than climbing and it is likely that fluoxetine is not producing climbing as opposed to actually decreasing climbing. Finally, fluoxetine, as previously reported, does not significantly alter diving compared to vehicle-treated behavior.
A single p.o. administration of either 3 or 10 mg/kg of Example 49 did not significantly alter swimming, climbing, immobility or diving in male rats in the FST, indicating that at these doses using the p.o. route, Example 49 does not exhibit antidepressant action in the FST. In contrast, a single p.o. administration of 0 577 Smg/kg of Example 49 produced a significant increase in swimming and a significant decrease in immobility Scompared to animals treated with vehicle, or 3 and mg/kg of Example 49. However, the 30 mg/kg p.o. dose of 0 5 Example 49 did not significantly alter diving or climbing 00 M counts compared to vehicle-treated animals. The increase in swimming counts produced by 30 mg/kg p.o. of Example 00 49 was comparable to that of 10 mg/kg of fluoxetine, Salthough Example 49 was less effective than fluoxetine in CO 10 reducing immobility.
In conclusion, a single p.o. administration of 30 mg/kg of Example 49 (one hour before the last swim test) increases swimming and decreases immobility counts in the FST, suggesting that Example 49 may have antidepressant properties in this model.
H. Effect of a single p.o. administration of Example 22, fluoxetine and vehicle on swimming, climbing, immobility and diving in the forced swim test Immobility Statistical analysis indicated a significant effect of treatment on immobility (ANOVA, F(4,44) 20.2, p 0.0001). Post hoc analyses revealed that a single p.o.
administration of 10 mg/kg of fluoxetine significantly decreased immobility (Student-Newman-Keuls value of 20.1) compared to vehicle-treated animals (Table A single p.o. administration of 10 or 30 mg/kg doses of Example 22 produced a significant decrease in immobility compared to vehicle-treated animals (Student-Newman-Keuls 00 578 values of 12.2 and 55.0, respectively). In addition, the decrease in immobility produced the either fluoxetine C or the 10 and 30 mg/kg doses of Example 22 (Student- Newman-Keuls values of 21.2, 13.0 and 56.8, respectively) 0 5 was significantly greater than that of the 3 mg/kg dose Sof Example 22. The decrease in immobility produced by mg/kg i.p. of Example 22 was significantly greater than 00 that of the 10 mg/kg dose (Student-Newman-Keuls value 16.2). In addition, the magnitude of the decrease in immobility produced by 30 mg/kg of Example 22 was significantly greater than that of fluoxetine (Student- Newman-Keuls value of 9.3).
Swimming Statistical analysis indicated a significant treatment effect on swimming behavior (ANOVA, F(4,44) 35.00, p 0.0001). Post hoc analyses revealed that a single i.p.
administration of 10 mg/kg of fluoxetine produced a significant increase in swimming compared to animals treated with vehicle, 3 or 10 mg/kg of Example 22 (Student-Newman-Keuls values of 49.6, 51.3 and 5.8, respectively) (Table A single p.o. administration of 3 mg/kg did not significant alter swimming behavior compared to vehicletreated animals (Table 5g). However, a single p.o.
administration of 30 mg/kg of Example 22 produced a significantly greater increase in swimming behavior compared to animals treated with vehicle, 3 or 10 mg/kg of Example 22 and fluoxetine (Student-Newman-Keuls values of 85.9, 88.1, 22.7 and 5.84, respectively).
00 579 Climbing behavior c' There was a significant treatment effect on climbing C1 behavior (ANOVA, F(4,44) 4.1, p 0.0066). Post hoc tests indicated that a single p.o. administration of mg/kg dose of Example 22 produced a significant increase 00 c in climbing compared to animals treated with vehicle, 3 Sor 10 mg/kg of Example 22 and fluoxetine (Student-Newman- 00 Keuls values of 10.5, 11.1, 5.8 and 11.8, respectively).
C1 10 Diving Statistical analysis revealed that diving was not significantly altered by a single i.p. administration of 3, 10 or 30 mg/kg of Example 22 or 10 mg/kg of fluoxetine compared to vehicle-treated animals (ANOVA, F(4,44) 0.58, p 0.68)(Table
F--
00 S580 STABLE 5g. The effect of a single p.o. administration of vehicle, 10 mg/kg of fluoxetine and Example 22 on C immobility, climbing, diving and swimming in the forced swim test in male Sprague-Dawley rats.
00 M Treatment Immobility Climbing Swimming Diving Vehicle 50 1.6 2.1 0.7 7.8 0.3 C00 1.0 0.3 S3 mg/kg EX22 50 0.9 2.0 0.6 7.6 0.4 0.4 mg/kg EX22 41 1.3c 2.9 0.5 15.3 0.4 0.8 9 0.3 mg/kg EX22 31 2 .8b 5.2 23.2 0.0 a 2.0' 0.0 mg/kg 39 1.7d 1.9 0.5 19.2 0.0 Fluox 1.2e 0.0 Each value represents the mean S.E.M. A total of animals were examined for each treatment group, except for the 30 mg/kg dose of Example 22, where a total of 9 animals were examined.
Fluox Fluoxetine, EX22 Example 22. Experiments were conducted 1 hr. after the appropriate treatment.
asignificantly greater than the vehicle (p 0.01), 3 mg/kg Example 22 (p 0.01), 10 mg/kg Example 22 (p 0.05) and 10 mg/kg of fluoxetine (p 0.05), ANOVA and Student-Newman-Keuls test.
00 D 581 SbSignificantly less than all other treatment groups, p c- 0.01, ANOVA and Student-Newman-Keuls test.
CSignificantly less than vehicle, 3 and 30 mg/kg of Example 22 (p 0.01) and 10 mg/kg of fluoxetine (p M 0.05), ANOVA and Student-Newman-Keuls.
<C dSignificantly less than vehicle, 3 and 30 mg/kg of 00 SExample 22, p 0.01, ANOVA and Student-Newman-Keul s CA 10 test.
eSignificantly greater than the vehicle, 3 and 10 mg/kg of Example 22, p 0.01, ANOVA and Student-Newman-Keuls test.
'Significantly greater than the vehicle, 3 and 10 mg/kg of Example 22, p 0.01 and fluoxetine, p 0.05, ANOVA and Student-Newman-Keuls test.
9 Significantly greater than the vehicle and 3 mg/kg of Example 22, p 0.05, ANOVA and Student-Newman-Keuls test.
The results of this study indicated that as previously reported, a single p.o. administration of 10 mg/kg of fluoxetine produced a significant increase in swimming and a significant decrease in immobility in male Sprague- Dawley rats in the FST compared to vehicle-treated animals. The magnitude of these changes are similar to those reported of our past studies with 10 mg/kg p.o. of fluoxetine. In contrast, neither climbing nor diving 00 S582 Sbehavior was significantly altered by a single i.p.
c- administration of 10 mg/kg of fluoxetine.
A single p.o. administration of 3 mg/kg of Example 22 did O 5 not significantly alter swimming in male rats in the FST.
00 Mg In contrast, a single p.o. administration of 10 or O mg/kg of Example 22 produced a significant increase in C swimming and a significant decrease in immobility 00 0compared to animals treated with vehicle or 3 mg/kg of Cl 10 Example 22. In addition, the magnitude of the increase in swimming behavior produced by 30 mg/kg p.o. of Example 22 was significantly greater than that of 10 mg/kg of Example 22 and 10 mg/kg of fluoxetine. The rank order of the treatments for increasing swimming is: 30 mg/kg Example 22 fluoxetine 10 mg/kg Example 22 3 mg/kg Example 22 Climbing behavior was significantly greater in animals treated with 30 mg/kg p.o. of Example 22 compared to animals treated p.o. with either vehicle, 3 or 10 mg/kg of Example 22 or 10 mg/kg of fluoxetine. None of the other treatments besides 30 mg/kg of Example 22 significantly altered climbing behavior compared to vehicle-treated animals. The rank order of the treatments for increasing climbing is: 30 mg/kg Example 22 3 Mg/kg Example 22 10 mg/kg Example 22 fluoxetine.
A single p.o. administration of 3 mg/kg of Example 22 did not significantly alter swimming compared to vehicletreated animals. However, the 10 and 30 mg/kg doses produced a significantly greater decrease in immobility 00 583 Scompared to vehicle-treated animals, with the effect at c, 30 mg/kg being greater then that of 10 mg/kg.
C- Furthermore, 30 mg/kg p.o. of Example 22 produced a significantly greater decrease in immobility than 0 5 mg/kg p.o. of fluoxetine. The rank order of the 00 M treatments for decreasing immobility is 30 mg/kg Example S22 10 mg/kg Example 22 fluoxetine 3 mg/kg Example 00 22.
00 0C 10 In conclusion, a single p.o. administration of 10 or mg/kg of Example 22 significantly increases swimming and significantly decreases immobility in vehicle-treated male Sprague-Dawley rats. This suggests that at these doses, Example 22 has antidepressant properties.
I. Effect of a single p.o. administration of Example fluoxetine and vehicle on swimming, climbing, immobility and diving in the forced swim test Statistical analysis indicated that a single p.o.
administration of 10 or 30 mg/kg Example 95 significantly increased rat immobility and significantly decreased swim behavior in the rat forced swim test at both doses (Table p <0.01, ANOVA and Student-Newman-Keuls, respectively).
584 TABLE 5h. The effect of a single p.o. administration of vehicle, 10 mg/kg of fluoxetine and 3, 10 or 30 mg/kg of Example 95 on immobility, climbing, diving and swimming in the forced swim test in male Sprague-Dawley rats.
Treatment Immobility Climbing Swimming Diving Vehicle 42 1.7 2.3 0.5 14.7 0.1 0.1 3 mg/kg 40 3.3 2.6 0.8 17.1 0.0 2.5 0.0 mg/kg 52 1.2a 1.3 0.5 6.9 0 .9b 0.1 0.1 54 0.9a 1.0 0.3 4.8 0 .7b 0.0 0.0 mg/kg 38 2.2 1.9 0.6 20.0 0.1 Fluox 1.5c 0.1 Each value represents the mean S.E.M. A total of 8 animals were examined for each treatment group, except for the vehicle, where a total of 10 animals were examined. Fluox Fluoxetine; EX95 Example Experiments were conducted 1 hr. after the appropriate treatment.
aSignificantly less than Vehicle, 3 mg/kg of Example and 10 mg/kg of fluoxetine, p 0.01, ANOVA and Student- Newman-Keuls test.
bSignficantly less than Vehicle, 3 mg/kg of Example 95 and 10 mg/kg of fluoxetine, p 0.01, ANOVA and Student- Newman-Keuls test.
00 S585 CSignficantly greater than Vehicle (p 0.05), 10 and Smg/kg of Example 95 (p 0.01), ANOVA and Student-Newman- Keuls test.
D 00 SA single p.o. administration of 10 mg/kg of fluoxetine C produced a significant increase in swimming behavior compared to vehicle-treated animals. In addition, CN 10 fluoxetine significantly decreased immobility compared to vehicle-treated animals. A single p.o. administration of 3 mg/kg of Example 95 did not significantly alter swimming, climbing, immobility or diving behavior compared to vehicle-treated animals. In contrast, a single p.o. administration of either 10 or 30 mg/kg of Example 95 produced a significant increase in immobility and a significant decrease in swimming behavior compared to vehicle-treated animals. There was no significant difference in the magnitude of change in swimming and immobility between the 10 and 30 mg/kg doses of Example These data indicate that at a doses of 10 and 30 mg/kg Example 95 produced effects opposite of that seen with antidepressants in the rat forced swim test, suggesting that Example 95 does not produce antidepressant-like actions in the forced swim test in male Sprague-Dawley rats.
S586 2. Social Interaction Test A. The Effect Of Example 92 And Chlordiazepoxide On j Behavior In The Rat Social Interaction Test O 5 A single i.p. administration of either 10 or 30 mg/kg of 00 SExample 92 significantly increased social interaction (Table 6 and Figure as did the benzodiazepine C0 anxiolytic, chlordiazepoxide (Student-Newman-Keuls value Sof 31.3) compared to vehicle-treated animals [ANOVA, CA 10 F(4,45) 10.3, p 0.0001; Student-Newman-Keuls values for the 10 and 30 mg/kg doses were 8.61 and 19.55, respectively]. However, the 100 mg/kg i.p. dose of Example 92 did not significantly alter social interaction time compared to vehicle-treated animals (Table 6 and Figure The degree of social interaction was greater in the chlordiazepoxide-treated animals compared to those that received either the 10 or 30 mg/kg doses of Example 92.
587 Table 6. The Effect Of A Single Injection Of Vehicle, Chlordiazepoxide And Example 92 On The Social Interaction And Rearing Of Unfamiliar Cage Mates In A Familiar Arena Drug Social Treatment Interaction (sec)a Vehicle, 1 ml/kg 96 12 Chlordiazepoxide, 5 mg/kg 188 1 5 b Example 92, 10 mg/kg 144 1 2 b Example 92, 30 mg/kg 169 13 c Example 92, 100 mg/kg 117 6 d a Each value represents the mean seconds of social interaction
S.E.M.
b Significantly greater than Vehicle, p 0.05, ANOVA and Student-Newman-Keuls test.
c Significantly greater than Vehicle, p 0.01, ANOVA and Student-Newman-Keuls test.
d Significantly less than 30 mg/kg dose and chlordiazepoxide, p 0.01, ANOVA and Student-Newman- Keuls.
C 588 SB. The Effect Of Example 92 And Chlordiazepoxide On Rearing Behavior, Locomotor Activity And Grooming In The C Rat Social Interaction Test The administration of 10 and 30 mg/kg, but not 100 mg/kg 00 Sof Example 92, significantly increased rearing behavior §compared to either vehicle or chlordiazepoxide
[ANOVA,
C0 F(4,45) 2.6, p 0.046; See Table 13]. In addition, the number of rearings at the 10 mg/kg dose of Example 92 C< 10 was significantly greater than that produced by chlordiazepoxide (Table 13).
The administration of either Example 92 or chlordiazepoxide did not significantly alter the number of grooming bouts compared to vehicle-treated animals [F(4,45) .67, p 0.621.
A single injection of either 10 or 30 mg/kg i.p. of Example 92 or 5 mg/kg i.p. of chlordiazepoxide did not significantly alter the number of squares crossed (Table 13). However, the number of squares crossed following the 100 mg/kg dose of Example 92 was significantly lower than animals treated with either vehicle, 10 mg/kp i.p.
of Example 92, 30 mg/kg i.p. of Example 92 or 5 mg/kg i.p. of chlordiazepoxide. [ANOVA, F(4,43) 6.94, p 0.0002].
589 Table 13. The Effect of a Single Injection of Vehicle, Cllordiazepoxide and Example 92 on the Number of Rearings, Squares Crossed and Grooming Bouts in the Rat Social Interaction Test.
Drug Treatmeflt(i.p.) Rearings Squares Crossed Grooming Bouts All values represent the mean S.E.M.
a significantly greater than chiordiazepoxide, p< 0.05, ANOVA and Student-Newman-Keuls test b Significantly greater than vehicle and chlordiazepoxide, p 0.05, ANOVA and Student-Newmal Keuls test.
CSignificantly less than 10 30 mg/kg of Example 92 (p 0.01), vehicle (p 0.01) and chlordiazepoxide (p 0.05), ANOVA and Student-Newman-Keuls test.
0590 FAt doses of 10 and 30 mg/kg Example 92 produced a -s significant increase in social interaction time in male n rats compared to vehicle-treated animals. Also, the anxiolytic agent (5 mg/kg i.p. chlordiazepoxide) produced O 5 a significant increase in social interaction time compared 00 00 to vehicle-treated animals. The response"produced by the O 30 mg/kg dose of Example 92 was comparable to that of the C positive control, chlordiazepoxide. The 30 mg/kg dose of SExample 92 produced a significant increase in rearing C( 10 compared to vehicle- and chlordiazepoxide-treated animals.
Previously, it has been shown that in the Social Interaction Test, psychostimulants such as amphetamine and caffeine, increase social interaction and locomotor activity, whereas anxiolytics increase social interaction time. (File, 1985; File and Hyde, 1979; Guy and Gardner, 1985). Consistent with an increase in social interaction, Example 92 increased rearing behavior. However, it did not produce an increase in horizontal locomotor activity or grooming bouts. In addition, Example 92 did not elicit stereotypes or produce aggressive behaviors. In fact, locomotor activity as measured by squares crossed was significantly reduced at the 100 mg/kg i.p. dose of Example 92 compared to vehicle-treated animals. This decrease in locomotor activity did not appear to be accompanied by ataxia or sedation. Thus, it is unlikely that Example 92 is producing a non-specific effect on social interaction through motor stimulation. In order to justify this claim, in another study (not reported), the effect of Example 92 was dosed to familiar cage mates in the social interaction test and no significant increase in interaction in this variation of the Social Interaction Test was observed. In this test, the anxiogenic stimulus 00 591 Sof a novel partner is removed and therefore only locomotor c' activity and normal behavior are observed (Guy and V' Gardner, 1985) In conclusion, the results of this study indicate that Example 92, at doses of 10 and 30 mg/kg significantly increases social interaction time 00 M without producing an increase in horizontal locomotor Sactivity or grooming bouts. Furthermore, the effect
C
produced by the 30 mg/kg of Example 92 was comparable to 0 that observed for 5 mg/kg of chlordiazepoxide, the active C< 10 control. No increase in social interaction was observed at the 100 mg/kg dose of Example 92. However, a decrease in the number of squares crossed was observed. In summary, Example 92 has the profile of an anxiolytic drug in the Social Interaction Test.
C. The effect of a single p.o. administration of Example 34, vehicle and chlordiazepoxide on the duration of social interaction in the rat social interaction test.
There was a significant treatment effect on the duration of social interaction (ANOVA, F(5,40) 11.8, p 0.001).
Subsequent post hoc analyses indicated that a single p.o.
administration of either 0.03, 0.1, 0.3 and 1 mg/kg of Example 34 (Student-Newman-Keuls test values of 10.6, 4.3 and 13.2, respectively) significantly increased social interaction, as did chlordiazepoxide (Student- Newman-Keuls value of 57.1), compare to vehicle-treated animals (Table 6a). The duration of social interaction produced by chlordiazepoxide was significantly greater than that of 0.03, 0.1, 0.3 and 1 mg/kg p.o. of Example 34 (Student-Newman-Keuls values of 19.6, 18.6, 26.2 and 17.6, respectively). There was no significant difference 00 592
(N
Sin the duration of social interaction between the various doses of Example 34 (Table 6a).
ci 0 5 Table 6a. The effect of a single p.o. administration of 00 M vehicle, chlordiazepoxide and Example 34 on social Sinteraction time in unfamiliar cage mates in a familiar C arena 00 CA 10 Drug Treatment Social Interaction (sec) Vehicle, 1 ml/kg 27 1.
4
A
Chlordiazepoxide, 5 mg/kg 122 185 Example 34, 0.03 mg/kg 62 11i Example 34, 0.1 mg/kg 66 7* Example 34, 0.3 mg/kg 53 6" Example 34, 1 mg/kg 69 6* Animals received one p.o administration of the appropriate treatment and all experiments were conducted one hour after the last injection.
AEach value represents the mean seconds of social interaction S.E.M. A total of 6-8 animals were examined for each treatment group.
*Significantly greater than Vehicle, p 0.05, ANOVA and Student-Newman-Keuls test.
*Significantly greater than Vehicle, p 0.01, ANOVA and Student-Newman-Keuls test.
SSignificantly greater than all other treatment groups, p 0.01, ANOVA and Student-Newman-Keuls test.
00 S593 SD. The effect of a single p.o. administration of Example c- 34, vehicle and chlordiazepoxide on rearing behavior, l locomotor activity and grooming in the social interaction test.
00 M Statistical analysis indicated a significant effect of Streatment on rearing behavior (ANOVA, F(5,40) 3.5, p c0 0.01; Table 14). Post hoc analyses revealed that the the O0 0 number of rears following 0.3 mg/kg of Example 34 was Cl 10 significantly lower than that of 0.1 and 1 mg/kg p.o. of Example 34 (Student-Newman-Keuls values of 8.8 amd 9.4, respectively).
Statistical analysis indicated a significant effect of treatment on number of squares crossed (F(5,40) 2.9, p 0.03). Post hoc analyses indicated that a single p.o.
administration of 0.3 mg/kg of Example 34 produced a significantly greater effect on the number of squares crossed compared to vehicle-treated animals (Student- Newman-Keuls values of 10.4).
Statistical analysis indicated a significant effect of treatment on grooming behavior (F(5,40) 4.3, p 0.004). Post hoc analyses indicated that the number of grooming episodes was significantly lower in the 0.03 mg/kg group compared to animals treated with 0.1, 0.3 or 1 mg/kg p.o. of Example 34 (Student-Newman-Keuls values of 11, 8 and 9.7, respectively (Table 14). In additon, the number of grooming episodes was significantly greater in animals treated with 0.1 mg/kg p.o. of Example 34 compared to those treated with vehicle (Table 14).
594 Table 14. The effect of a single p.o. administration of vehicle, chlordiazepoxide and Example 34 on the number of rearings, grooming episodes and squares crossed in the social interaction test in unfamiliar cage mates in a familiar arena Drug Treatment Rearing Squares Crossed Grooming bouts Vehicle, 1 ml/kg 34 3 250 31 4.6 0.7 Chlordiazepoxide, 5 35 2 265 30 5.3 0.7 mg/kg Example 34, 0.03 27 3* 312 23 4.0 0.4& mg/kg Example 34, 0.1 40 5 295 40 7.6 mg/kg Example 34, 0.3 27 2 363 17 7.2 1.1 mg/kg Example 34, 1 mg/kg 40 1 343 15® 7.3 0.8 Animals received one p.o administration of the experiments were conducted appropriate treatment and all one hour after the last injection. All values represent the mean S.E.M. A total of 6-8 animals were examined for each treatment group.
'Significantly less than 0.1 mg/kg of Example 34, p 0.05, ANOVA and Student-Newman-Keuls test.
$Significantly less than 0.1 and Img/kg of Example 34, p 0.05, ANOVA and Student-Newman-Keuls test.
00 S595 1 Significantly greater than Vehicle, p 0.05, ANOVA and Student-Newman-Keuls test.
ci 'Significantly less than 0.1, 0.3 and 1 mg/kg of Example 34, p 0.05, ANOVA and Student-Newman-Keuls test.
00 Significantly greater than Vehicle, p 0.05, ANOVA and C Student-Newman-Keuls test.
00 C< 10 One of the main findings of this study was that in paired, unfamiliar male Sprague-Dawley rats, a single p.o. administration of either 0.03, 0.1, 0.3 or 1 mg/kg p.o. of Example 34 produced a significant increase (2-2.6 fold) in the duration of social interaction compared to animals treated with vehicle. In addition, there was no significant difference in the magnitude of increase in social interaction between the various doses of Example 34, i.e. there was no dose-response relationship. As previously reported, a single p.o. administration of mg/kg of chlordiazepoxide produced a significant increase in the duration of social interaction compared to vehicle-treated animals.
Rearing behavior was not significantly altered by any of the doses of Example 34 or by chlordiazepoxide compared to vehicle-treated animals, although there were differences between the doses of Example 34. The number of squares crossed was significantly greater following a single p.o. administration of 1 mg/kg of Example 34 compared to vehicle-treated animals, whereas there were no significant differences between the other doses of Example 34 and vehicle. Thus, overall, Example 34 does 0 596 Snot significantly alter locomotor activity, suggesting that it does not produce locomotor activation or I stimulation.
Grooming behavior following a single p.o. administration 00 Stended to be greater after 0.1, 0.3 and 1 mg/kg of Example 34 compared to animals that had received vehicle, C0 although this was only statistically significant for the 00 S0.1 mg/kg dose. Furthermore, the number of grooming CI 10 episodes was significantly lower after a single p.o.
administration of 0.03- mg/kg of Example 34 compared to 0.1, 0.3 and 1 mg/kg of Example 34.
In conclusion, the above results suggest that a single p.o. administration of 0.03, 0.1, 0.3 or 1 mg/kg of Example 34 produces an anxioltyic action in the social interaction test in male Sprague-Dawley rats.
I
597 SIII Binding Properties of Compounds at Cloned Receptors A. Materials and Methods Cl The binding properties of the compounds of the present invention were evaluated at one or more cloned receptors or native, tissue-derived transporters, using protocols 00 cdescribed below.
00 Cell Culture SCOS-7 cells were grown on 150 mm plates in D-MEM with (C 10 supplements (Dulbecco's Modified Eagle Medium with bovine calf serum, *4 mM glutamine, 100 units/ml penicillin, 100 pg/ml streptomycin) at 37 0 C with 5% CO 2 Stock plates of COS-7 cells were trypsinized and split 1:6 every 3-4 days. Human embryonic kidney 293 cells were grown on 150 mm plates in D-MEM with supplements (minimal essential medium) with Hanks' salts and supplements (Dulbecco's Modified Eagle Medium with 10% bovine calf serum, 4 mM glutamine, 100 units/ml penicillin, 100 pg /ml streptomycin) at 37 0 C with 5% CO 2 Stock plates of 293 cells were trypsinized and split 1:6 every 3-4 days.
Mouse fibroblast LM(tk-) cells were grown on 150 mm plates in D-MEM with supplements (Dulbecco's Modified Eagle Medium with 10% bovine calf serum, 4 mM glutamine, 100 units/mL penicillin, 100 ug/mL streptomycin) at 37 0
C
with 5% COz. Stock plates of LM(tk-) cells were trypsinized and split 1:10 every 3-4 days. Chinese Hamster Ovary (CHO) cells were grown on 150 mm plates in HAM's F12 medium with (HAM's F-12 with 10% bovine calf serum, 4 mM glutamine, 100 units/mL penicillin, 100 gg/mL streptomycin) at 37 0 C with 5% CO 2 Stock plates of CHO cells were trypsinized and split 1:8 every 3-4 days.
0 598 SLM(tk-) cells were stably transfected with the human GAL1 or GAL3 receptor. CHO cells were stably transfected with the human GAL2 receptor.
Stable Transfection 00 ScDNAs for the human and rat GAL1, and human and rat GAL3 receptors were transfected with a G-418 resistant gene C0 into the mouse fibroblast LM(tk-) cell line by a calcium 00 Sphosphate transfection method (Cullen, 1987). Stably C 10 transfected cells were selected with G-418. Human and rat GAL2 receptors were similarly transfected into CHO cells.
Membrane Harvest Membranes were harvested from stably transfected LM(tk-) cells. Adherent cells were washed twice in ice-cold phosphate buffered saline (138 mM NaC1, 8.1 mM Na2HPO4, mM KC1, 1.2 mM KH 2 P04, 0.9 mM CaC12, 0.5 mM MgCl2, pH 7.4) and lysed by sonication in ice-cold sonication buffer (20 mM Tris-HCl, 5 mM EDTA, pH Large particles and debris were cleared by low speed centrifugation (200 x g, 5 min, 4 0 Membranes were collected from the supernatant fraction by centrifugation (32,000 x g, 18 min, 4 0 washed with ice-cold hypotonic buffer, and collected again by centrifugation (32,000 x g, 18 min, 4 0 The final membrane pellet was resuspended by sonication into a small volume of ice-cold binding buffer ml for every 5 plates: 10 mM NaCl, 20 mM HEPES, 0.22 mM KH 2 PO4, 1.26 mM CaC12, 0.81 mM MgS04, pH Protein concentration was measured by the Bradford method (Bradford, 1976) using Bio-Rad Reagent, with bovine serum albumin as a standard. Membranes were held on ice for up to one hour and used fresh, or flash frozen S599 Sand stored in liquid nitrogen. Membranes were prepared similarly from CHO cells.
As described in the Background of the Invention, 0 5 compounds that block the effects of galanin on the GAL3 00 Sreceptor subtype can potentially be used for the Streatment of depression and anxiety. Biogenic amine
C
N transmitter molecules that mediate neuronal signals are O currently known in the art and include among others C 10 serotonin (5HT), norepinephrine and dopamine (DA).
Recent advances in the molecular studies of the mechanisms for these transmitter molecules, together with the characterization of their pharmacological properties, has enabled the identification of numerous potential targets for therapeutic intervention. Inhibitors of the NE and DA transporter systems, and inhibitors of the enzyme, monoamine oxidase, have been widely studied and are known to enhance the action of biogenic amine neurotransmitters. The resultant clinically effective antidepressant drugs are known today as TCAs, SSRIs and MAOIs. (Tatsumi et al., 1997; Iversen, 2000).
In the case of galanin, the evidence presented in this invention suggests that GPCR-targeted molecules that bind to and antagonize the GAL3 receptor may be used for the treatment of depression and/or anxiety disorders. Another approach could involve the administration of an antagonist of the GAL3 receptor, such as those described herein, which also possesses 5HT 4 receptor antagonist properties (Kennett et al., 1997). A further approach could involve the administration of a GAL3 antagonist, such as those described herein, which also possesses 00 600 Sreceptor binding properties (Razani et al., 1997).
c However, in any case the GAL3 antagonist(s) should be C free of activity at the human GAL1 receptor and the NE and DA transporters. Furthermore, the GAL3 antagonist(s) should not inhibit the enzymatic activity 00 M of monoamine oxidase A (MAOA) or monoamine oxidase B S(MAOB) present in the brain central MAO). The design 0 of such compounds can be optimized by determining their binding affinity at the recombinant GAL3, GAL1, 5HT 4 and CI 10 5HTA receptors; and the native 5HT, NE and DA transporters. The design of such compounds can be further optimized by determining their interaction with central MAOA and central MAOB.
Additionally, the GAL3 antagonist(s) would optimally not bind at the following receptors due to possible side effects: human GAL2; human H 1 histamine; human XIA adrenergic, human alB adrenergic, human amu adrenergic, human Ct2A adrenergic, human o2B adrenergic, and human a2c adrenergic; human dopamine DI, D 2
D
3
D
4 and Ds; and the human 5HTis, human 5HTiD, human 5HT 1 E human 5HTIF, human 5HT2A, rat 5HT 2 c, human SHT 6 and human 5HT 7 receptors.
Radioligand Binding Assays and Enzymatic Assays The methods to obtain the cDNA of the receptors, express said receptors in heterologous systems, and carry out assays to determine binding affinity are described as follows.
Galanin Receptors: Binding assays were performed according to the following published methods: human GAL3 (PCT International Publication No. WO 98/15570), human 00 0 601 SGALl (PCT International Publication No. WO 95/2260), human GAL2 (PCT International Publication No. WO CQ 97/26853).
Human 5HTiB, 5HT1D, 5HTlE, 5HTip, and 5HT 7 Receptors: The 00 Scell lysates of LM(tk-) clonal cell line stably with the genes encoding each of these C receptor-subtypes were prepared as described above. Cell Smembranes were suspended in 50mM Tris-HC1 buffer (pH 7.4 Cl 10 at 37°C) containing 10 mM MgC12, 0.2 mM EDTA, 10 M pargyline, and ascorbate. The affinities of compounds were determined in equilibrium competition binding assays by incubation for 30 minutes at 37 oC in the presence of 5 nM 3 H]-serotonin. Nonspecific binding was determined in the presence of 10 plM serotonin. The bound radioligand was separated by filtration through GF/B filters using a cell harvester.
Human 5HT2A Receptor: The coding sequence of the human 5HT2A receptor was obtained from a human brain cortex cDNA library, and cloned into the cloning site of pCEXV-3 eukaryotic expression vector. This construct was transfected into COS-7 cells by the DEAE-dextran method (Cullen, 1987). Cells were harvested after 72 hours and lysed by sonication in 5 mM Tris-HCl, 5 mM EDTA, pH The cell lysates were subjected to centrifugation at 1000 rpm for 5 minutes at 4°C, and the supernatant was subjected to centrifugation at 30,000 x g for 20 minutes at 4 0 C. The pellet was suspended in 50 mM Tris-HCl buffer (pH 7.7 at room temperature) containing 10 mM MgS04, mM EDTA, and 0.1% ascorbate. The affinity of compounds at 5HT2A receptors were determined in equilibrium competition 00 602 Sbinding assays using 3 [H]ketanserin (1 nM). Nonspecific c, binding was defined by the addition of 10 pM mianserin.
CI The bound radioligand was separated by filtration through GF/B filters using a cell harvester.
D 00 cr 5-HTIA Receptor: The cDNA corresponding to the 5-HT1A 0 receptor open reading frames and variable non-coding 00 and 3'-regions, was cloned into the eukaryotic expression vector pCEXV-3. These constructs were transfected transiently into COS-7 cells by the DEAE-dextran method (Cullen, 1987), and harvested after 72 hours. Radioligand binding assays were performed as described above for the 5-HT2 receptor, except that 3 H-8-OH-DPAT was used as the radioligand and nonspecific binding was determined by the addition of 10 pM mianserin.
00 S603 SOther 5-HT Receptors: Other serotonin receptor binding assays were performed according to published methods: rat 5HT 2 c receptor (Julius et al., 1988); and 5-HT 6 (Monsma, et al., 1993). The binding assays using the 5-HT 4 receptor were performed according to the 00 f procedures described in U.S. Patent No. 5,766,879, the Sdisclosure of which is hereby incorporated by reference 00 in its entirety into this application.
C Other receptors: Cell membranes expressing human dopamine
D
1
D
2
D
4 and rat D 3 receptors were purchased through BioSignal, Inc. (Montreal, Canada). Binding assays using the histamine Hi receptor; dopamine receptors; and lA, aIB, and a 2 adrenergic receptors may be carried out according to the procedures described in U.S. Patent No. 5,780,485, the disclosure of which is hereby incorporated by reference in its entirety into this application. Binding assays using the dopamine Ds receptor may be carried out according to the procedures described in U.S. Patent No. 5,882,855, the disclosure of which is hereby incorporated by reference in its entirety into this application. Binding assays for the human alD adrenergic receptor may be carried out according to the procedures described in U.S. Patent No. 6,156,518, the disclosure of which is hereby incorporated by reference in its entirety into this application.
The methods to determine binding affinity at native transporters are described in the following publications: 5HT transporter and NE transporter (Owens S604 Set al., 1997), and DA transporter (Javitch et al, c, 1984).
The methods to determine activity at monoamine oxidase enzymes (for example, central MAOA and MAOB) are 00 Sdescribed by Otsuka and Kobayashi, 1964, and were performed by NovaScreen (Hanover, MD) with the c0 following modifications.
C i10 Central Monoamine Oxidase A Enzyme Assay: Rat brain was used as the enzyme source. The enzyme source was pre-incubated with reference compound (RO 41-1049), test compound (Example 92), and subtype selective blocker (100nM deprenyl) for 60 minutes at 37 0 C in 50 mM
KPO
4 containing 50 M EDTA and 10 pM dithiothreitol (pH 7.2 at 25 0 Substrate ([14C]Serotonin, 45-60 Ci/mmol) was then added and incubated for 30 minutes. The reaction was stopped by the addition of 0.5 ml of 1-2M citric acid. Radioactive product was extracted into xylene/ethyl acetate fluor and compared to control values by scintillation spectrophotometry in order to ascertain any interactions of test compound with central MAOA.
Central Monoamine Oxidase B Enzyme Assay: Rat brain was used as the enzyme source. The assay was performed as described above for central MAOA, except the reference compound was RO 166491 and the subtype selective blocker was 100 nM clorgyline. Also, the substrate [14C]Phenylethylamine, 0.056 Ci/mmol) was added and incubated for 10 minutes.
00 0605 SMaterials Cell culture media and supplements were from Specialty n Media (Lavallette, NJ). Cell culture plates (150 mm and 96-well microtiter) were from Corning (Corning, NY).
Polypropylene 96-well microtiter plates were from Co- 0 0 star (Cambridge, MA). Bovine serum albumin (ultra-fat free, A-7511) was from Sigma (St. Louis, MO). All CI radioligands were from New England Nuclear (Boston, 00 MA) Commercially available peptides and peptide C 10 analogs were either from Bachem California (Torrance, CA) or Peninsula (Belmont, CA). All other materials were reagent grade.
Data Analysis Binding data were analyzed using nonlinear regression and statistical techniques available in the GraphPAD Prism package (San Diego, CA). Enzymatic assay data were derived from a standard curve of reference compound data.
The selectivity ratios for compounds of the claimed invention were calculated from the binding data presented in Tables 1-4, Table 7 and Table 9 of the subject application. More specifically, these ratios were calculated by dividing the binding affinity (Ki value) of said compound to a particular receptor or transporter by the binding affinity (Ki value) of said compound to the human GAL3 receptor. The data presented in Table 8 and Table 10, hereinafter, were calculated using the above described method.
00 D 606 SFor example, the GAL3/GAL1 selectivity ratio of c- recited in claim 110 of the subject application is Scharacteristic of Example 34. This binding ratio was calculated by dividing the Ki value of 912 for the binding of Example 34 to the GAL1 receptor (see Table 00
M
0 1) by the Ki value of 23 for the binding of Example S34 to the human GAL3 receptor, thus obtaining the C result of 39. Therefore the GAL3/GAL1 binding ratio for 00 SExample 34 was determined to be greater than C B. Results The compounds described in the claimed invention were assayed using a panel of cloned receptors and native transporters. The preferred compounds were found to be selective GAL3 antagonists. The binding affinities and selectivity ratios of several compounds are illustrated in Tables 7-10.
2008200380 25 Jan 2008 Table 7: Antagonist binding affinity (Ki) at the human GAL3 receptor vs.
serotonin receptors and several transporters.
Example hGAL3 h5HT h5HT 1 h5HT h5HT 1 E h5HT h5T2, r5HT 2 c h5HT 4 h5HT 6 h5HT 7 r5HT rNE rDA Uptk Uptk Uptk Ki Ki Ki Ki Ki Ki Ki Ki Ki Ki Ki Ki Ki Ki (nM) (nM) (aM) (iM) (raM) (aM) (nM) (aM) (nM) (raM) (aM) (aM) (nM) 11 91 4682 101 102 9174 1780 6708 802 1308 800 1012 1 95 5430 73 5098 487 1272 11038 4192 11270 572 2301 1457 2527 1737 24500 17 87 3477 407 1032 33523 10271 7157 562 2606 711 1797 719 18325 27200 22 28 9714 1981 1852 13230 5773 20689 1717 2457 -2264 2672 8483 13085 7480 34 23 1059 2976 28282 4803 2076 20762 38921 4439 37462 3900 49 211 29187 8447 16872 23886 8894 6687 13230 13 12268 40666 37585 2010 86 33666 5461 9198 1180 2124 26118 1781 1180 47536 3235 25274 46108 14500 77 79 5472 365 716 5888 3237 2242 456 1324 503 1547 821 28083 2790 92 38 11323 32139 18934 5290 ND 72 ND 45111 33879 17800 94 49 3349 10764 25227 5683 4099 4120 3647 8018 12961 4876 2200 29 28288 5226 16018 27211 4446 3471 3031 21507 11638 6101 12000 97 51 5057 14235 22692 4157 1950 2550 29131 11283 36308 4412 8440 A n IA A464 36329 5496 7430 S98 38 124 5 7 6 2419 9118 16240 3359 20 V J. Jv I L- >50000 ND Not determined 2008200380 25 Jan 2008 Table 8: Antagonist selectivity ratios determined for the human GAL3 receptor vs.
serotonin receptors and several transporters.
Example I hGAL3 IhSHi 11 17 22 34 49 77 92 94 97 98 1 1 1 1 1 1 1 1 1 1 1 1 1 >30 >30 >30 >100 >100 >100 >100 >30 >100 >100 i>1 0 0 h,5HTjn h5HTID~ 7 17 5 12 >30 >30 >30 >100 >30 >30 >30 >100 5 -9 >100 >100 >300 >100 >00 >100 >30 >100 >30 >100 h5HITI >100 >100 >100 >100 >100 >100 14 >30 >100 >100 >100 >100 >100
ND
hSHTj, h5HT 21 r5BT2C h5ET 4 i h5HT 6 h5HT 7 r5HT rKiE rDA Uptk Uptk Uptk 20 >30 9 14 9 11 18 >100 >30 >100 8 >30 20 >30 24 >100 >100 >100 >30 6 30 8 21 8 >100 >100 7>100 >100 >30 >30 >30 >30 >100 >100 >100 >100 >100 >30 >100 >100 >100 >100 >100 >100 >30 >100 >30 >30 0 >30 >100 >100 25 >100 21 14 >100 >30 >100 >100 >100 >30 28 6 17 16 20 10 >100 >100 >100 ND 2 >100 ND >100 >100 >100 >100 >100 >30 >30 >30 >100 >100 >30 >130 >100 >100 >100 >100 >100 >100 >100 >100 >100 >30 >100 >30 >30 >100 >100 >100 >30 >100 >30 1>100 >30 -Not determined >30 1 >100 >100 >100 >100 >100 2008200380 25 Jan 2008 Table 9: Antagonist binding affinity (Ki) at the human GAL3 receptor vs.
alpha-adrenergic, dopamine, and histamine receptors.
Example hGAL3 ha hca hrlz ha2 ha 2 B ha 2 C hD, hD 2 rD 3 hD 4 hD 5 hH
X
Ki Ki Ki Ki Ki Ki Ki Ki Ki Ki Ki Ki Ki (niM) (nM) (nM) (nM) (nM) (nM) (nM) (nM) (nM) (nM) (nM) (nM) (nM) 11 91 926 1436 264 1819 10235 3004 79 782 2139 4828 64 ND 73 3392 853 480 14413 24515 8202 344 2184 8809 13151 78 ND 17 87 996 1167 221 3523 38732 10269 516 1808 2477 22227 89 ND 22 28 1278 1582 368 906 5757 2737 128 .1501 5664 11621 63 ND 34 23 3756 15004 1240 3679 15488 8832 290 2500 9922 18716 111 ND 49 211 6646 18852 678 4731 25374 9244 3781 5940 13964 45824 328 ND 86 13604 40615 4231 10838 7200 600 26815 15295 48756 538 39909 77 79 834 452 217 315 7783 634 60 910 2716 504 122 ND 92 38 ND 17175 21943 41369 48180 41369 29290 39909 94 49 12715 31135 4027 12718 45378 47863 2145 6249 423 727 ND 29 13137 32494 3468 30072 48552 4394 9716 466 2590 ND 97 51 16921,45845 6454 13569 25115 9716 10069 ND 98 38 14500 31693 1891 23236 2524 3788 592 1199 ND >50000 ND Not determined 2008200380 25 Jan 2008 Table 10: Antagonist selectivity ratios determined for the human GAL3 receptor vs.
aipha-adrenergic, dopamine, and histamine receptors.
Example kLGAL3hz c hcxljhcz 2 .B hrt 23 2 ha 2 c~ liD, hD 2 rD 3 liD 4 liD 5 hE 1 11 1 10 16 3 20 >100 >30 0.9 9 24 >30 0.7 ND 1 46 12 7 >100 >100 >100 S 30 >100 >30 1 ND 17 1 11 13 3 >30 >100 >100 6 21 28 >100 1 ND 22 1 >30 >30 13 >30 >100 >100 5 >30 >100 >100 2 ND all 34 1 >100 >100 >30 >100 >100 >100 13 >100 >100 >100 S ND C) 49 1 >30 >30 3 22 >100 >30 18 28 >30 >100 2 ND 1 >100 >100 >30 >100 >100 >30 7 >100 >100 >100 6 >100 77 1 11 6 3 4 >30 8 0.8 11 >30 6 2 ND 92 1 ND >100 >100 >100 >100 >100 >100 >100 >100 >100 >100 >100 94 1 >100 >100 >30 >100 >100 >100 >30 >100 9 >100 15 ND 1 >100 >100 >100 >100 >100 >100 >100 >100 16 >100 >30 ND 97 1 >100 >100 >1001 >100 >100 >100 >100 >100 >100 >100 >100 N 98 1 >100 >100 >30 >100 >100 >100 >3 10 1 10 >0 ND ND =Not determined 611 The activity of Example 92 was determined for central MAOA and central MAOB using the methods described hereinabove.
The results, expressed as percent inhibition, are illustrated in Table 11.
Table 11: Percent inhibition of Example 92 in the central monoamine oxidase enzyme assay TARGET SPECIES INHIBITION Monoamine Oxidase A Rat (central) Monoamine Oxidase B Rat 1 (central) 00 612 SIV. GAL3 Receptor Localization n A. Materials And Methods Preparation of the anti-GAL3 Antiserum 00 BioSource International, Hopkinton, MA performed the C immunization and maintenance of rabbits. Following a C pre-immune bleed, one peptide for each GAL receptor was 00 injected into a pair of New Zealand white rabbits. The peptide sequences was chosen based on sequence specificity and immunogenicity. The rabbit anti-GAL3 antiserum were raised against C-terminal epitopes corresponding to amino acids 357 370 (Genbank accession number AF073798). The peptides were conjugated to the carrier KLH (keyhole limpet hemocyanin) by a cross linker and subcutaneously injected into the rabbits. The generation of the anti-GAL3 antiserum required OVA followed by a third series of injections with the GAL3 peptide conjugated to tetanus toxoid (TTOX). All injections were done using the Freund's Adjuvant System.
Once immunoreactivity was established (see below) the antiserum was affinity purified by passing it over an agarose based column thiol coupled to its antigenic peptide. The column was washed and the antiserum was eluted using a low pH glycine buffer. The purified material was dialyzed, the optical density is taken at 280 X and the purified antiserum was frozen.
Characterization of the anti-GAL3 antiserum Recombinant GAL1, GAL2, and GAL3 receptor transfected cells 00 613 (c- STo determine the ability of the GAL3 antiserum to recognize only the GAL3 receptor protein in vitro, COS-7 cells were grown on poly-L-lysine-coated plastic chamber slides (Nalge Nunc International, Naperville, IL) and 0 5 transfected with recombinant rat GAL receptors (Genbank 00 M^ accession numbers U30290, AF010318, AF073798, respectively) or expression vector only (for mock- C transfected cells) as previously described by Borowsky et Sal. (1999). Receptor expression was confirmed by CI 10 radioligand binding. Briefly, a subset of slides was washed three times in binding buffer (50 mM Tris, pH mM MgC1 2 1 mM EDTA, 0.1% bovine serum albumin, and 0.1% bacitracin) and incubated in 500 pl binding buffer containing porcine 12s 5 -galanin (625,000 dpm) plus or minus 10 pM porcine galanin. After incubation at room temperature for 1 hour, the binding buffer was aspirated and slides were rinsed three times in ice cold 50 mM Tris, pH 7.5. Cells were solubilized in 1 ml of 0.1 N NaOH and 0.05% sodium deoxycholate for 30 minutes then transferred to test tubes for gamma counting of 1251. To evaluate antibody activity another subset of slides were washed with phosphate buffered saline (PBS) (Sigma, St.
Louis, MO) to remove the medium and fixed with 4% paraformaldehyde (PFA) (Sigma, St. Louis, MO) then permeabilized using 0.2% Triton X-100/PBS and incubated in 3% normal goat serum for 30 minutes to minimize nonspecific binding of the primary antibody. Cells were incubated overnight at 4 0 C with the anti-GAL3 antiserum (1:1000 dilution). The cells were rinsed three times with PBS, incubated for 30 minutes at 25 0 C with goat antirabbit IgG (1:200 dilution) (Santa Cruz Biotechnology, Santa Cruz, CA), rinsed and processed using the 00 S614 Speroxidase-antiperoxidase (PAP) reaction of Sternberger c' et al. (1982). Control experiments for antibody C specificity were incubation of the cells in primary antiserum that had been preabsorbed with the respective 0 5 antigenic peptide (20 ug/ml), incubation without the 00 c primary antiserum, or incubation with the primary Santiserum replaced by normal goat serum.
00 O Western Blotting C 10 Membranes were prepared from COS-7 cells transiently transfected with the rat recombinant receptors GAL1, GAL2, and GAL3 as previously described (Borowsky et al., 1999). Transfected cells were lysed by sonication in ice-cold sonication buffer (20 mM Tris-HCl, pH 7.7, 5 mM EDTA). Cell lysates were subjected to centrifugation at 4 0 C for 10 minutes at 200 g. The supernatant was then fractionated by centrifugation at 4 0 C for 18 minutes at 32,000 g. The resulting membrane pellet was suspended into 50 mM Tris, pH 7.5, 5 mM MgC1 2 1 mM EDTA. Protein samples (1-10 pg) were solubilized in 2 X Laemmli buffer (Bio-Rad, Hercules, CA) and fractionated by SDS-PAGE in polyacrylamide gels. Proteins were transferred to polyvinylidine difluoride membranes for immunoblot analysis in ice-cold 25 mM Tris, pH 8, 192 mM glycine, 20% methanol as previously described by Harlow and Lane (1999). Blots were incubated" for 1 hour at 25 0 C in blocking buffer composed of 5% non-fat dried milk in TTBS Tween-20, 500 mM NaC1, 20 mM Tris, pH 7.5) then for 16 hours at 25 0 C with the receptor-specific polyclonal antibody (1:1000 dilution in blocking buffer) (0.25 mg/ml for GAL2 or 1.5 mg/ml for GAL3). Immunoreactive bands were detected with the Phototope-HRP Detection Kit for 00 615 SWestern Blotting (New England BioLab, Beverly, MA) according to the protocol. Briefly, the blots were (C incubated with horseradish peroxidase-conjugated goat anti-rabbit IgG then developed with a mixture of LumiGLO plus hydrogen peroxide and recorded by chemiluminescence 00 r on Kodak Biomax-ML film (Kodak, Rochester, NY).
00 Immunohistochemistry SMale Sprague-Dawley rats, (200-250 g; Charles Rivers, C1 10 Rochester, NY) were anesthetized by intraperitoneal injection of ketamine 20 mg/kg (RBI, Natick, MA) and xylazine 0.2 mg/kg (Bayer, Shawnee Mission, KS) then transcardially perfused with 200 ml PBS, pH 7.4 followed by 200 ml 4% PFA in PBS. The brains and spinal cords were removed, blocked, and postfixed in the same fixative for 4 hours at 4 0 C then cryoprotected in 30% sucrose in PBS at 4 0 C for 48 hours before freezing on dry ice.
Coronal brain sections and transverse spinal cord sections were cut at 30 pm using a freezing microtome.
Tissue sections were immediately immersed in PBS and stored at 4°C until use. Sections were processed freefloating according to the protocol outlined in NEN Life Science Products TSA (Tyramide Signal Amplification) Indirect Kit. Briefly, tissue sections were permeabilized in 0.2% Triton X-100 (Sigma, St. Louis, MO)/PBS, incubated in 1% hydrogen peroxide (Sigma, St.
Louis, MO)/PBS to remove endogenous peroxidase activity then blocked in TNB Buffer (0.1 M Tris-HCl, pH 7.5, 0.15 M NaC1, and 0.5% Blocking Reagent. Sections were incubated for 24 hours at 4°C in either the anti-GAL2 or anti-GAL3 antiserum (1:100). Following incubation with the primary antiserum, the tissue sections were washed in S616 STNT Buffer (0.1 M Tris-HCl, pH 7.4, 0.15 M NaCI, 0.05% Tween 20) followed by incubation at 25 0 C for 30 minutes C with horseradish peroxidase (HRP)-conjugated goat antirabbit immunoglobulin (1:200) (Sternberger Monoclonals Inc., Lutherville, MD). Tissue sections were rinsed in 00 STNT Buffer and incubated in a solution containing Sbiotinylated tyramide to amplify the signal then rinsed 00 in TNT buffer and incubated with HRP-conjugated to Sstreptavidin at 25 0 C for 30 minutes. An immunoperoxidase reaction was done by incubating the section in 3,3'diaminobenzidine (DAB) in 0.1 mM Tris, pH 7.4 and adding hydrogen peroxide to 0.006% immediately before use. The reaction was stopped in water and the sections mounted on microscopic slide with mounting medium ethanol: gelatin) and counterstained with Cresyl violet then coverslipped for light microscopy.
Optimal GAL3 antibody concentrations (1:200) for rat brain sections were determined in preliminary titration experiments. Experimental controls in the tissue sections included incubation in normal rabbit serum or omission of the primary antiserum.
Analysis COS-7 cells and tissue sections were examined using a Zeiss Axioscope. A total of 6 male rats were examined with the anti-GAL3 antiserum. The identification of GAL3-LI in the transfected cells and brain regions was based on the presence of immunoreactivity appearing as a brownish precipitate in individual cells and their projections or in the neuropil of the tissue by light 00 D 617 Smicroscopy. The descriptions of neuroanatomic boundaries are based on the atlas of Paxinos and Watson (1998).
B. Results 00 OO Characterization of the GAL3 antiserum Cl Recombinant GAL1, GAL2, and GAL3 receptor transfected 00 0 cells Cy 10 The ability of the anti-GAL3 antiserum to recognize only the GAL3 receptor-protein in vitro was established by performing immunocytochemistry on COS-7 cells transiently transfected with the recombinant receptor proteins for the rat GAL1, GAL2, and GAL3, or mock-transfected with vector only. Specific porcine 1 2 5 I-galanin binding was detected for all transfectants except mock-transfected cells. An immune response was detected only in the COS-7 cells incubated with the antiserum generated for the particular recombinant receptor. Specifically, no immune reaction was observed with the anti-GAL3 antiserum (1:1000) in GAL1 or GAL2 transfected cells. Furthermore, no visible immune reaction was detected in the mocktransfected cells. Incubation of the cells in primary antiserum that had been preabsorbed with the antigenic peptide (20 pg/ml) or without the primary antiserum or with the primary replaced by normal goat serum did not result in an immune response.
Taken together, these data demonstrate that the anti-GAL3 antiserum recognizes the receptor against which it was generated and does not show cross reactivity with other known.GAL receptors.
00 618 c' Western Blots C. To determine the specificity of the anti-GAL3 antiserum, COS-7 cells were transiently transfected either with 5 recombinant rat GAL2 or GAL3 receptors or with expression 00 00 vector only; membranes were then isolated for evaluation Sby immunoblotting (see Figure The anti-GAL3 antiserum labeled proteins in membranes only from rat GAL3- Stransfected cells; a predominant band was evident with an Cl 10 apparent molecular weight of approximately 56 kDa (Figure somewhat higher than the amino acid-derived value of 40.4 kDa. (For comparison, apparent molecular weights determined by SDS-PAGE are 56 kDa (Servin et al., 1987) or 54 kDa (Chen et al., 1992) for native GAL receptors purified from rat brain and 54 kDa (Amiranoff et al., 1989) for native GAL receptors purified from Rin m cells. These values are all higher than the amino acidderived value any known GAL receptor subtype, including the value of 38.9 kDa for rat GAL1 (Parker et al., 1995).
The apparently high molecular weight observed for rat GAL3 very likely reflects post-translational processing such as glycosylation; note that rat GAL3 contains multiple N-terminal glycosylation sites (Smith et al., 1998). Relative to the predominant band, additional species of higher molecular weight as well as lower molecular weight were labeled by the corresponding antiserum (Figure These are presumably receptorrelated species composed of protein aggregates of Cterminal fragments, as they are absent in mocktransfected cells.
00 S619 SImmunohistochemical distribution of GAL3-LI in the CNS GAL3-like immunoreactivity (GAL3-LI) was observed in many C regions of the brain, specifically, the neocortex, septum, hippocampus, amygdala, and brainstem (see Table 0 5 12). Throughout the brain and spinal cord GAL3-LI was 00 Mr\ found to be associated with neuronal profiles however, there was neuropil staining observed in several brain C regions. Several regions of the CNS almost exclusively 00 Sexpressed GAL3-LI, specifically the accumbens nucleus, Cl 10 dorsal raphe, ventral tegmental area (Table 12). There was no observable staining of the fiber tracts.
The specificity of the anti-GAL3 antiserum was determined in tissue sections by omission of the primary antiserum or incubation with normal rabbit serum. No specific staining was observed in either condition.
Preabsorption of the GAL3 primary antiserum with the antigenic peptide (10 gg/ml) decreased but did not completely block staining in the tissue sections as in the transfected cells. This was most likely related to the different localization approaches. In the transiently transfected COS-7 cells the expression of GAL3 receptor protein was relatively high therefore, indirect immunocytochemistry with no amplification was used. In contrast, GAL3 receptor protein expression is presumed to be relatively lower in the tissue sections and for that reason the TSA (amplification) technique was employed.
It is possible that because of the amplification (1000fold) in the TSA technique even small amounts of unabsorbed antiserum may result in a signal.
00 620 SDistribution of GAL3-LI in the rat CNS Cerebral cortex GAL3-LI was widespread in the cerebral cortex and the distribution pattern extended rostrocaudally. A weak to 00 rr moderate GAL3-LI was seen in numerous cell bodies in the anterior cingulate cortex.
00 SSeptal region C 10 An extensive and densely stained fiber network was seen throughout the entire lateral, intermediate and medial septal nuclei. The dorsal division of the lateral septum contained scarce moderately GAL3-like immunoreactive somata.
Basal ganglia Numerous moderately GAL3-like immunoreactive cell bodies and fibers were present in the shell and core of the accumbens nucleus. The cell bodies of the subthalamic nucleus, a relay nucleus in the basal ganglia, contained weak GAL3-LI.
Amygdala and Extended Amygdala In general, GAL3-LI was weak throughout the amygdala.
Scattered cell bodies and fibers exhibited weak staining in several nuclei. Very fine GAL3-like immunoreactive fibers with scattered moderately labeled cells were detected in the central amygdaloid nucleus.
Midbrain/Mesencephalon Labeled cells were detected within the dorsal raphe and projections from these cells were seen converging toward 00 D 621 Sthe midline of the raphe. Moderately immunoreactive scattered cells were evident in the ventral tegmental IC area.
Brain stem 00 SIntense staining was observed in cell bodies in the locus coeruleus.
00 SThe distribution of rat GAL3 protein in the CNS using C< 10 receptor subtype selective polyclonal antibodies and tyramide signal amplification (TSA) immunocytochemistry.
is illustrated in Table 12. These were qualitative evaluations for the rat GAL3 receptor protein distribution based on the relative intensity of the chromogen (3,3'-diaminobenzidine) observed in individual cells at the microscopic level.
A total of 4 rat brains were analyzed for this study. As shown in Table 12, the strength of the signal obtained in various regions of the rat brain was graded as weak or moderate or intense(+++).
622 Table 12 00 00 C 0 oo 0D REGION cells fiber Potential a Therapeutic Application Telencephalon Frontal cortex Anxiety/Depression Cingulate cortex Anxiety/Depression Basal ganglia Accumbens nucleus Treatment of the positive symptoms of schizophrenia Treatment of drug addiction. This region is particularly sensitive to psychoactive drugs.
Anxiety/depression Septal Region Relief of fear Lateral septal nucleus, dorsal Lateral septal nucleus, ventral Intermediate septal nucleus Medial septal nucleus Amygdala and extended Treatment of anxiety, panic Amygdala attack, and depression.
Treatment of disorders of integrated behaviors such.as defense, ingestion, reproduction, and learning.
Central nucleus Fear and anxiety 623 Mesencephalon Dorsal raphe Depression/Analgesi a Ventral tegmental Depression area Brainstem/Pons/Medulla Locus coeruleus Modulation of noradrenergic transmission.
Treatment of depression The GAL3 antiserum was characterized using recombinant GAL receptors in transiently transfected COS-7 cells and Western blot analysis and the specificity of the GAL3 antiserum to recognize only the cognate receptor in vitro was established. The anatomical distribution of the GAL3 receptor protein in the rat CNS was determined using a modified immunohistochemical technique to enhance sensitivity and delectability via tyramide signal amplification (Toda et al., 1999).
The results indicate that the expression GAL3-LI was primarily found in neuronal profiles with neuropil labeling detectable in several areas. In general, the distribution of GAL3-LI is in good agreement with the reported distribution for galanin-LI, galanin binding sites, and GAL3 mRNA in the rat brain (for recent review, Branchek et al., 2000). Overall, GAL3-LI was extensively distributed throughout the brain. Paralleling the distribution of galanin binding sites GAL3-LI was observed in ventral regions of the brain.
00 624 SThe localization of the GAL3 protein in the dorsal raphe and locus coeruleus suggests a potential therapeutic C application of galanin receptor antagonists in the treatment of depression by attenuating galanin's 5 inhibitory tone on both of these regions.
00 SA decrease in central serotonin (5-HT) neurotransmission 00 has been implicated in depression. GAL3 antagonists could Spossibly act via GAL3 receptors on the cell bodies of Cl 10 dorsal raphe neurons to increase firing rate of raphe neurons thus increasing 5-HT release in the telencephalon and diencephalon. Another possible site of action for a GAL3 antagonist could be on postsynaptic GAL3 receptors in the limbic forebrain to block the putative ability of galanin to negatively regulate 5-HTiA receptor transmission (Misane et al, 1998).
Unlike the dorsal raphe cells, the cells of the locus coeruleus express abundant galanin under normal conditions and it has been proposed that galanin may be released from dendrites and soma of the noradrenergic cell bodies (for review, H6kfelt et al., 1998). The ascending afferent projections of the locus coeruleus are extensive throughout the brain. Changes in the noradrenergic system have been hypothesized to be involved in depression-related behaviors and symptoms (for review, Weiss et al., 1998). The ventral tegmental area (VTA) receives projections from the locus coeruleus that have been reported to co-localize galanin and noradrenaline. It has been proposed that in certain pathological states (ex. stress induced depression) galanin released from noradrenergic terminals in the VTA 00 S625 g inhibits dopaminergic neurons in the region that results in decreased dopamine release in the forebrain regions, CI particularly the accumbens nucleus and prefrontal cortex.
This decrease in dopamine release produces a decreased 0 5 motor activation and anhedonia. GAL3 has been identified 00 Sin all of these regions and thus presents itself as a potential therapeutic target in the treatment of 00 depression. Drugs that would effectively decrease galanin's release in the VTA whether at the level of the C 10 locus coeruleus (somatodendritic GAL3 receptors to decrease the activity of LC cells) or in the VTA itself.
(presynaptically on NE/GAL terminals in the VTA or via GAL3 receptors on VTA-DA neurons to prevent the hyperpolarization VTA-DA cells by released galanin) would produce an antidepressant effect.
00 626 SReferences C American Psychiatric Association (1994) Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition.
American Psychiatric Association, Washington, DC.
00 Amiranoff, et al., (1989) Galanin receptor in the rat C pancreatic beta cell line Rin m 5F. Molecular 00 Scharacterization by chemical cross-linking. J. Biol.
C< 10 Chem., 264(34): 20714-20717.
Asymmetric Synthesis (1983) Vols 2-5, Academic Press, Editor Morrison, J.
Bakker, et al., (2000) Constitutive activity of the histamine H1 receptor reveals inverse agonism of histamine HI receptor antagonists. Eur. J. Pharmacol., 387: R5-R7.
Borowsky, et al., (1999) Cloning and characterization of the human galanin GALR2 receptor. Peptides, 19: 1771- 1781.
Bradford, M.M. (1976) A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of the protein-dye binding. Anal.
Biochem., 72: 248-254.
Branchek, et al., (2000) Galanin receptor subtypes.
Trends in Pharm. Sci., 21: 109-116.
Bryant, W.M.III, et al., (1993) Synthetic Communications, 00 D 627 S231 1617-1625.
SChen, et al., (1992) Solubilization and molecular characterization of active galanin receptors from rat brain. Biochemistry, 31(8): 2415-2422.
00 oC SCoppola, G.M. (1987) Journal of Heterocyclic Chemistry, C 24: 1249.
00 Cl 10 Cullen, B. (1987) Use of eukaryotic expression technology in the functional analysis of cloned genes. Methods Enzymol., 152s 685-704.
deLigt, et al., (2000) Inverse agonism at G protein-coupled receptors: (patho)physiological relevance and implications for drug discovery. Br. J. Pharmacol., 130(1): 1-12.
De Weille, et al., (1989) Galanin inhibits dopamine secretion and activates a potassium channel in pheochromocytoma cells. Brain Res., 485: 199-203.
Detke, et al., (1995) Active behaviors in the rat forced swim test differentially produced by serotonergic and noradrenergic antidepressants. Psychopharmacology, 121: 66-72.
Ennis, M. D. and Ghazal, N. (1992) The synthesis of and (-)-flesinoxan: Application of enzymatic resolution methodology. Tetrahedron Lett., 33: 6287-6290.
00 S628 c-I SFile, S.E. (1985) Animal models for predicting clinical efficacy of anxiolytic drugs: social behaviour.
Neuropsychobiology, 13: 55-62.
00 c SFile, S.E. and Pellow, S. (1984) The anxiogenic action of
C
FG 7142 in the social interaction test is reversed by Schlordiazepoxide and Ro-15-1788 but not by CGS 8216.
C< 10 Archs. Int. Pharmacodyn. Ther., 271: 198-205.
File, S.E. and Pellow, S. (1983) The anxiogenic action of a convulsant benzodiazepine: reversal by chlordiazepoxide. Brain Res., 278: 370-372.
File, et al., (1982) The anxiogenic action of benzodiazepine-like antagonists. Neuropharmacology, 21: 1033-1037.
File, S.E. (1980) The use of social interaction as a method for detecting anxiolytic activity of chlordiazepoxide-like drugs. J. Neurosci. Methods, 21 219-238.
File, S.E. and Hyde, J.R.G. (1979) A test of anxiety that distinguishes between the actions of benzodiazepines and those of other minor tranquilisers and of stimulants.
Pharmacol. Behav. Biochem., 11: 65-69.
File, S.E. and Hyde, J.R.G. (1978) Can social interaction be used to measure anxiety? Br. J. Pharmacol., 62: 19-24.
00 629 SGarden, et al., (1998). Synthetic Communications, c- 28: 1679-1689.
ci Glover, V. (1998) Function of endogenous monoamine oxidase inhibitors (tribulin). J. Neural. Transm. Suppl., 00 00 52: 307-13.
C Gopalan, et al., (1993) Neurochemical evidence that 00 0 the inhibitory effect of galanin on tuberoinfundibular Ci 10 dopamine neurons is activity dependent.
Neuroendocrinology, 58: 287-293.
Green, T.W. and Wuts, P.G.M. (1991) Protection groups in Organic Synthesis, second Edition John Wiley Sons, New York.
Guy, A.P. and Gardner, C.R. (1985) Pharmacological characterisation of a modified social interaction model of anxiety. Neuropsychobiology, 13: 194-200.
Harlow, E. and Lane, D. (1999) Immunoblotting. In: Barker, P. editor. Using Antibodies: A Laboratory Manual.
New York: Cold Spring Harbor Laboratory Press. p 267-309.
Herrick-Davis, et al., (2000) Inverse agonist activity of atypical antipsychotic drugs at human Hydroxytryptamine2C receptors. J. Pharmacol. Exp. Ther., 295(1): 226-32.
Hess, B.A. Jr. and Corbino, S. (1971) Journal of Heterocyclic Chemistry, 8: 161.
00 S630 (c- SHikfelt, et al., (1998) Galanin in Ascending Systems.
Annals of the N.Y. Acad. Sci., Ed. T. H6kfelt, Tamas 1C Bartfai and J. Crawley p. 252-263.
Iversen, L. (2000) Neurotransmetter transporters: 00 00 fruitful targets for CNS drug discovery. Mol. Psychiatry, 357-62.
00 SJansson, et al., (1989) Centrally administered C 10 galanin reduces dopamine utilization in the median eminence and increases dopamine utilization in the medial neostriatum of the male rat. Acta Physiol. Scand., 135: 199-200.
Javitch, et al, (1984) 3 H-Mazindol binding associated with neuronal dopamine and norepinephrine uptake sites. Molecular Pharmacology, 26: 35-44.
Jaques, et al., (1981) Enantiomer, Racemates and Resolutions. John Wiley Sons.
Julius, et al., (1988) Molecular characterization of a functional cDNA encoding the serotonin Ic receptor.
Science, 241: 558-564.
Kenakin, T. (1996) The classification of seven transmembrane receptors in recombinant expression systems. Pharmacol. Rev., 48(3): 413-63.
Kennett, et al., (1997) Anxiolytic-like actions of the selective 5-HT4 receptor antagonist SB-20470-A and SB-20766-A in rats. Neuropharmacology, 36(4-5): 707-712.
00 D 631 Kirby, L.G. and Lucki, I. (1997) Interaction between the n forced swimming test and fluoxetine treatment on extracellular 5-hydroxytryptamine and hydroxyindoleacetic acid in the rat. Stress, 2(4) 251- 00 0 263.
CI Leonard BE. (1996) New approaches to the treatment of 00 Sdepression. J Clin Psychiatry. 57(4): 26-33.
C Lightowler, 'et al., (1994) Anxiolytic-like effect of paroxetine in a rat social interaction test. Pharmacol.
Behav. Biochem., 49t 281-285.
Lucki, I. (1997) The forced swimming test as a model for core and component behavioral effects of antidepressant drugs. Behav. Pharmacol., 8s 523-528.
Lutz, M. and Kenakin, T. (1999) Quantitative Molecular Pharmacology and Informatics in Drug Discovery, John Wiley Sons, LTD, West Sussex, England. p. 153.
Misane, et al., (1998) Modulation of a 5-HT1A receptor-mediated beavioral response by the neuropeptide galanin. Ann. N.Y. Acad. Sci., 863: 442-444.
Monsma, F.J. Jr., et al., (1993) Cloning and expression of a novel serotonin receptor with high affinity for tricyclic psychotropic drugs. Mol. Pharmacol., 43: 320- 327.
00 D 632 SN6gradi, M. (1987) Stereoselective Synthesis, VCH, Editor Ebel, H.
Nordstrom, et al., (1987) Evidence for an inhibitory effect of the peptide galanin on dopamine release from 00 M the rat median eminence. Neurosci. Lett., 73: 21-26.
00 Owens, M.J. (1997) Neurotransmitter receptor and Stransporter binding profile of antidepressants and their C< 10 metabolites. J. Pharm. Exp. Ther., 2831 1305-1322.
Otsuka, S. and Kobayashi, Y. (1964) A radioisotopic assay for monoamine oxidase determinations in human plasma.
Biochem. Pharmacol., 13t 995-1006.
Page, et al., (1999) Serotonergic mediation of the effects of fluoxetine, but not desipramine, in the rat forced swim test. Psychopharmacology, 147: 162-167.
Parker, et al., (1995) Cloning and characterization of the rat GALR1 galanin receptor from Rinl4B insulinoma cells. Mol. Brain Res., 34s 179-189.
Paxinos, G. and Watson, C. (1986) The Rat Brain in Stereotaxic Coordinates. San Diego: Academic Press, Inc.
Porsolt, R.D. (1981) Behavioral despair. In Enna, SJ (ed) Antidepressants: neurochemical, behavioral and clinical perspectives. Raven Press, New York, pp. 121- 139.
00 633 SPorsolt, et al., (1978) Behavioral despair in rats: c' a new model sensitive to antidepressant treatments. Eur.
L J. Pharmacol., 47: 379-391.
O 5 Porsolt, et al., (1977) Depression: a new animal 00 0 model sensitive to antidepressant treatments. Nature, 266: 730-732.
00 0 Razani, et al., (1997) 5-HT1A receptor activation: C( 10 short-term effects on the mRNA expression of the 5-HT1A receptor and galanin in the raphe nuclei. Neuroreport, 8(16): 3565-3570 Reneric, J.P. and Lucki, I. (1998) Antidepressant behavioral effects by dual inhibition of monoamine reuptake in the rat forced swim test.
Psychopharmacology, 136: 190-197.
Rodgers, et al., (1997) Animal models of anxiety: an ethological perspective. Braz. J. Med. Biol. Res., 289-304.
Servin, et al., (1987) Identification and molecular characterization of galanin receptor sites in rat brain.
Biochem. Biophys. Res. Commun., 144(1): 298-306.
Seutin, et al., (1989) Galanin decreases the activity of locus coeruleus neurons in vitro. Euro. J. Pharmacol.
164: 373-376.
Smith, et al., (1998) Cloned human and rat galanin GALR3 receptors Pharmacology and activation of G-protein 00 S634 Sinwardly rectifying K+ channels. J. Biol. Chem., c- 273(36): 23321-223326.
Sternberger, L.A. (1982) Neurotypy: regional O 5 individuality in rat brain detected by immunocytochemistry with monoclonal antibodies. Proc.
q0 Natl. Acad. Sci. USA, 79: 1326-1330.
00 Tatsumi, et al., (1997) Pharmacological profile of antidepressants and related compounds at human monoamine transporters. Eur. J. Pharmacol., 340(2-3): 249-258.
Toda, et al., (1999) Application of tyramide signal amplification system to immunohistochemistry: a potent method to localize antigens that are not detectable by ordinary method. Pathol. Int., 49(5): 479-483.
Treit, D. (1985) Animal models for the study of antianxiety agents: a review. Neurosci. Biobehav. Rev., 9: 203-222.
Weiss, et al., (1998) Annals of the N.Y. Acad.
Sci., (Ed. T. Hbkfelt, Tamas Bartfai and J. Crawley) p.
364-382.
Xu, et al., (1998) interactions: Electrophysiological, immunohistochemical and in situ hybridization studies on rat dorsal raphe neurons with a note on galanin R1 and R2 receptors.
2008200380 25 Jan 2008

Claims (219)

1. A method of treating a subject suffering from depression which comprises administering to the subject an amount of compound effective to treat the subject's depression wherein the compound has the structure: X wherein W is H, -Cl, -Br, CN, methyl, ethyl, propyl, methoxy or ethoxy; wherein X is; NR 11 R 12 R 1 7 R 17 -N or wherein R 11 is H, straight chained or branched CI-C 7 alkyl, (CH 2 )q-O-(CH 2 )m-CH 3 aryl, or aryl (Ci-C)alkyl; 00 637 wherein R 12 is straight chained or branched CI-C 7 alkyl, (CH 2 )q-Oi(CH2)m-CH3, or -(CH 2 )m-Z; wherein R 13 is a bicyclic alkyl ring system, adamantyl, 00 M noradamantyl, C 3 -C 10 cycloaJlkyl, heteroaryl, aryl, aryl (Cl- C 6 alkyl, Q, or Q2; 00 wherein aryl may be substituted with one or more Cl-Cl 0 straight chained or branched alkyl, aryl, heteroaryl, or N(Rj 9 _Z1 wherein Q3. is i X\/22 JR 2 2 wherein Q2 is R 2 2 R2 R 2 t RR22 R 2 2 PR 2 22 0 R 20 wherein each J is independently 0, S, C(R 2 2 2 or NR 4 638 wherein R 4 is H; straight chained or branched Cl-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C 7 alkenyl or alkynyl; C 3 -C 7 cycloalkyl, C 5 -C 7 cycloalkenyl or aryl; wherein Y is NR 14 R, 5 or wherein R- 1 4 is H, straight chained or branched Cl-C 6 alkyl, (CH 2 )q-CiCH2)m-CH3, C 3 -C 6 cycloalkyl, or (C(Rig) 2 wherein R 15 is straight chained or branched C 3 -C 6 alkyl, (C2)q-O (CH2)m-CH 3 C 3 -C 6 cycloalkyl, (C (Rig) 2 )MN (Rl 6 2 or (C (Rig) 2) Z wherein R 16 is straight chained or branched Cl-C 7 alkyl, straight chained or branched Cl-C 7 monofluoroalkyl, 00 C639 straight chained or branched C 1 -C 7 polyfluoroalkyl, straight chained or branched C 2 -C 7 alkenyl, straight chained or branched C 2 -C 7 alkynyl, C 5 -C 7 cycloalkenyl, (CH 2 I-Z, Or (CH 2 q (CH 2 m- CH 3 00 wherein each R 1 7 is independently H; -OR 2 1 -0C0R 21 -C0R 21 -NCOR 21 -N(R 21 2 -CON(R 21 2 -C00R 21 straight chained or CI branched Cl-C 7 alkyl, straight chained or branched Cl-C 7 00 monofluoroalkyl, straight chained or branched CI-C 7 polyfluoroalkyl, straight chained or branched C 2 -C 7 alkenyl, straight chained or branched C 2 -C 7 alkynyl, CS-C 7 cycloalkenyl, *I(CH 2 or (CH- 2 (CH 2 m- CH 3 wherein Ris is straight chained or branched C 1 -C 6 alkyl, (CJ4 2 or (CH 2 )q-O-(CH 2 )m-CH 3 wherein each R 19 is independently H, or straight chained or branched C 1 -C 6 alkyl; wherein each R 2 0 is independently straight chained or branched C 1 -C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C 7 alkenyl or alkynyl; C 3 C 7 cycloalkyl or CS-C 7 cycloalkenyl; -C1, -Br, or -I; -NO 2 -N 3 -CM; -OR 2 1 -0C0R 21 -C0R 21 -NCOR 21 -N(R 21 2 CON(R 2 1 2 or -C00R 21 aryl or heteroaryl; or two R 2 0 groups present on adjacent carbon atoms can join together to form a methylenedioxy group; wherein each R 2 1 is independently straight chained or branched C 1 -C- 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C 7 alkenyl or alkynyl; C 3 C 7 cycloalkyl, C 5 -C 7 cycloalkenyl, aryl, or aryl(Cl- I 00 640 SC 6 )alkyl; C( wherein each R 2 2 is independently H, F, Cl or Ci-C 4 straight chained or branched alkyl; 00 Mq wherein each m is an integer from 0 to 4 inclusive; C wherein each n is an integer from 1 to 4 inclusive; 00 C( 10 wherein p is an integer from 0 to 2 inclusive; wherein q is an integer from 2 to 4 inclusive; wherein t is 1 or 2; wherein U is O, -NR 1 s, S, C(R 17 2 or -NSO 2 RI 6 wherein Z is C 3 -C 10 cycloalkyl, C 4 -C7 cyclic ether, C 4 -C 7 cyclic thioether, aryl, or heteroaryl; or a pharmaceutically acceptable salt thereof.
2. A method of treating a subject suffering from depression which comprises administering to the subject an amount of compound effective to treat the subject's depression wherein the compound has the structure: 641 wherein W is H, -Cl, -Br, CN, methyl, ethyl, propyl, methoxy or ethoxy; wherein X is NRIIR 12 R 1 7 R 1 or -1 R 1 7, wherein R 11 is H, straight chained or branched Cl-C 7 alkyl, (CH 2 )q0I-CH 2 )m-CH 3 aryl or aryl(C- 1 -C 6 )alkyl; wherein R 12 is straight chained or branched C 1 C 7 alkyl, (CH 2 )q-O(CH 2 )m-CH 3 or -(CH 2 )m-Z; wherein R 13 is a bicyclic alkyl ring system, aryl' or aryl (Cl-C 6 alkyl;, wherein Y is NR 1 4 R 15 00 642 R 20 00R19 \1 tp-- -/R17o 00 -N wherein R 14 is H, straight chained or branched Cl-C 6 alkyl, (CH 2 )q0 (CH 2 C 3 -C 6 cycloalkyl, or (C(Ror)2)-Z; wherein R 15 is straight chained or branched C 3 -C 6 alkyl, (CH 2 )q-O-(CH 2 )CH3, C 3 -C 6 cycloalkyl, or (C(R 19 2 wherein U is 0, -NR 16 S, C(Rl 7 2 or -NS0 2 R 16 wherein Z is C 3 -Cl 0 cycloalkyl, aryl, or heteroaryl; wherein R 16 is straight chained or branched C 1 -C 7 alkyl, straight chained or branched C 1 -C 7 monofluoroalkyl, straight chained or branched C 1 -C 7 polyfluoroalkyl, straight chained or branched C 2 -C 7 alkenyl, straight chained or branched C 2 -C 7 alkynyl, C 5 -C7 cycloalkenyl, (CH 2 or (CH 2 )q01 (CH 2 )MrCH3; 643 wherein each RI-, is independently H; -OR 21 -OCOR 21 -C0R 21 -NCOR 2 1 -N(R 2 1 2 I -CON(R 2 1 2 -COOR 21 straight chained or branched Cl-C7 alkyl, straight chained or branched Cl-C7 monofluoroalkyl, straight chained or branched Cl-C7 polyfluoroalkyl, straight chained or branched C 2 -C7 00 M alkenyl, straight chained or branched C 2 -C7 alkynyl, Cs5-C7 C) ~cycloalkenyl, -(CH 2 )mn-Z, or (CH 2 (CH 2 )m-CH 3 00 C) wherein R 18 is straight chained or branched Cl-C 6 alkyl, (CH 2 )ngZ, or (CH 2 (CH 2 )m-CH 3 wherein each Rig is independently or straight chained or branched C 1 -C 6 alkyl; wherein each R 2 0 is independently straight chained or branched C 1 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or alkynyl; C 3 C,7 cycloalkyl Or C5-C-7 cycloalkenyl; -C1, -Br, or -I; -NO 2 -N 3 -CN; -OR 2 1 -OCOR 21 -C0R 21 NCOR 21 -N(R 21 2 CON(R 2 1) 2 or -C00R 21 aryl or heteroaryl; or two R 2 0 groups present on adjacent carbon atoms can join together to form a methylenedioxy group; wherein each R 2 2. is independently straight chained or branched Cl-C7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or alkynyl; C 3 C, cycloalkyl, C 5 -C7 cycloalkenyl, aryl or aryl(Ci- C 6 alkyl; wherein each m is an integer from 0 to 4 inclusive; wherein each n is an integer from 1 to 4 inclusive; 644 wherein p is an integer from 0 to 2 inclusive; wherein q is an integer from 2 to 4 inclusive; wherein t is 1 or 2; or a pharmaceutically acceptable salt thereof.
3. A method of treating a subject suffering from depression which comprises administering to the subject an amount of compound effective to treat the subject's depression wherein the compound has the structure: wherein W is H, -Cl, -Br, CN, methyl, ethyl, propyl, methoxy or ethoxy; wherein X is N(CH 3 2 or 645 wherein R 13 is an aryl, adamantyl, noradamantyl, C 3 -C 10 cycloalkyl, heteroaryl, Q, or Q2; wherein aryl may be substituted with one or more CI-C 10 straight chained or branched alkyl, aryl, heteroaryl, or N (Rig) -Z; wherein is wherein Q 2 is wherein each J is independently 0, S, C(R 2 2 2 or NR 3 646 wherein R 4 is straight chained or branched cl-c 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C 7 alkenyl or alkynyl; C 3 -C 7 Y cycloalkyl, C 5 -C 7 cycloalkenyl or aryl; 00 wherein Y is NR 14 R 15 00 117R20~A SR 1 Jp ;or R 20 -N R 2 0 wherein R 14 is H, straight chained or branched Cl-Cs alkyl, (CH2) q-0 (CH 2 .CH 3 C 3 -C 6 cycloalkyl, or (C (Rig) 2 m-Z; wherein R 15 is straight chained or branched C 3 -C 6 alkyl, (CH 2 )q-O (CH 2 )m-CH 3 C 3 -C 6 cycloalkyl, or (C(R 1 9 2 wherein U is 0, -NR 16 S, C(Rl 7 2 or -NS0 2 R 16 wherein Z is C 3 -Clo cycloalkyl, aryl, or heteroaryl; 00 D 647 c- wherein R 16 is straight chained or branched Ci-C 7 alkyl, Sstraight chained or branched CI-C, monofluoroalkyl, straight chained or branched CI-C 7 polyfluoroalkyl, straight chained or branched C 2 -C 7 alkenyl, straight 00 00 chained or branched C 2 -C 7 alkynyl, C 5 -C 7 cycloalkenyl, (CH 2 or (CH 2 )q-0-(CH 2 )m-CH 3 00 Swherein each R 17 is independently H; -OR 21 -0COR 21 -COR 2 11 C< 10 -NCOR 2 1, -N(R 2 1 2 -CON(R 21 2 -COOR 21 straight chained or branched CI-C, alkyl, straight chained or branched C 1 -C 7 monofluoroalkyl, straight chained or branched C 1 -C 7 polyfluoroalkyl, straight chained or branched C 2 -C 7 alkenyl, straight chained or branched C 2 -C 7 alkynyl, Cs-C, cycloalkenyl, -(CH 2 or (CH 2 )n-O-(CH 2 )m-CH 3 wherein R 18 is straight chained or branched CI-C 6 alkyl, (CH 2 or (CH 2 )q-O-(CH 2 )m-CH3; wherein each R 19 is independently H, or straight chained or branched C 1 -C 6 alkyl; wherein each R 20 is independently straight chained or branched C 1 -C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or alkynyl; C a C 7 cycloalkyl or C 5 -C 7 cycloalkenyl; -Cl, -Br, or -I; -NO 2 -N 3 -CN; -OR 21 -OCOR 21 -COR 21 -NCOR 21 -N(R 21 2 CON(R 21 2 or -COOR 2 1; aryl or heteroaryl; or two R 20 groups present on adjacent carbon atoms can join together to form a methylenedioxy group; wherein each R 21 is independently straight chained or 00 D 648 Sbranched C 1 -C7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or alkynyl; C 3 C, cycloalkyl, C 5 -C7 cycloalkenyl, aryl or aryl(C 1 C) alkyl; 00 M wherein each R 22 is independently H, F, Cl or Ci-C 4 o straight chained or branched alkyl; 00 Swherein each m is an integer from 0 to 4 inclusive; C wherein each n is an integer from 1 to 4 inclusive; wherein p is an integer from 0 to 2 inclusive; wherein q is an integer from 2 to 4 inclusive; wherein t is 1 or 2; or a pharmaceutically acceptable salt thereof.
4. A method of treating a subject suffering from depression which comprises administering to the subject an amount of compound effective to treat the subject's depression wherein the compound has the structure: 00 00 00 649 wherein W is -Cl, propyl, methoxy or ethoxy; wherein X is N (CH 3 2 or -Br, CN, methyl, ethyl, wherein R 1 3 is a bicyclic alkyl ring system, aryl or aryl (Cl-C 6 alkyl; wherein Y is NR 14 R, 5 wherein R 1 4 is H, straight chained or branched Cj-C 6 alkyl, (CH 2 )q-0-(CH 2 C 3 -C 6 cycloalkyl, or (C(R 1 9 2 wherein R 15 is (C(R 19 2 )m-N(R, 6 2 wherein Z is C 3 -CIO cycloalkyl, aryl, or heteroaryl; wherein R 16 is straight chained or branched Cl-C 7 alkyl, straight chained or branched Cl-C 7 'monofluoroalkyl, straight chained or branched C- 1 -C7 polyfluoroalkyl, straight chained or branched C 2 -C 7 alkenyl, straight chained or branched C 2 -C 7 alkynyl, C 5 -C 7 cycloalkenyl, (CH 2 or (CH)q0O(CH 2 )m,,CH 3 wherein each R 17 is independently H; -OR 2 1 -0C0R 21 -C0R 2 1 00 0 650 -NCOR 21 -N(R 21 2 -CON(R 21 2 -COOR 21 straight chained or branched C 1 -C7 alkyl, straight chained or branched CI-C7 n monofluoroalkyl, straight chained or branched C 1 -C 7 polyfluoroalkyl, straight chained or branched C 2 -07 O 5 alkenyl, straight chained or branched C 2 -C 7 alkynyl, C 5 -C? M 0 cycloalkenyl, -(CH 2 or (CH 2 )n-0-(CH 2 )m-CH 3 (C wherein each R19 is independently H, or straight chained 00 Sor branched Ci-C 6 alkyl; C wherein each R 21 is independently straight chained or branched C 1 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C 7 alkenyl or alkynyl; C 3 C 7 cycloalkyl, Cs-C, cycloalkenyl, aryl or aryl(C 1 C 6 )alkyl; wherein each m is an integer from 0 to 4 inclusive; wherein each n is an integer from 1 to 4 inclusive; wherein q is an integer from 2 to 4 inclusive; or a pharmaceutically acceptable salt thereof.
5. The method of claim 1, 2, 3 or 4, wherein the compound is enantiomerically and diasteriomerically pure.
6. The method of claim 1, 2, 3 or 4, wherein the compound is enantiomerically or diasteriomerically pure.
7. The method of claim 1, 2, 3 or 4, wherein the compound can be administered orally. 00 c, 0D 0q (N §q 651
8. The method of claim 1, wherein X is: R 17 R 17 R17 -N R17 or
9. The method of claim 1, wherein X is NR 11 R 12 and R1 is H or straight chained or branched C 1 -C 7 alkyl. The method of claim 9, wherein the compound has the structure:
11. The method of claim 8, wherein alkyl ring system, cyclohexyl or aryl.
12. The method of claim 10, wherein alkyl ring system, cyclohexyl or aryl. R 13 is a bicyclic R1 3 is a bicyclic
13. The method of claim 11, wherein R 14 is H, straight chained or branched CI-Cs alkyl or (CH 2 )q-0-(CH 2 )m-CH3. 00 D 652 c, 14. The method of claim 12, wherein R 14 is H, straight C r chained or branched CI-C 6 alkyl or (CH2)q-- (CH 2 )m-CH 3
15. The method of claim 13, wherein the compound is 00 fr selected from the group consisting of: 00 O 2008200380 25 Jan 2008 zO /O 1711 ft3 2008200380 25 Jan 2008 -z b U, 2008200380 25 Jan 2008 q% Ln uL 3 -c) 00 656 S16. The method of claim 11, wherein Y is cO -NU 00 00 S17. The method of claim 16, wherein U is NR 16
18. The method of claim 17, wherein R 16 is (CH 2 )m-Z.
19. The method of claim 18, wherein Z is aryl or heteroaryl. The method of claim 19, wherein the compound is selected from the group consisting of: N 2008200380 25 Jan 2008 o 2008200380 25 Jan 2008 IZJ Kill) Kill /KZ) z/il 659
21. The method of claim 12, wherein the compound is selected from the group consisting of: Cl 0 0PN Cl M-" N N N NII 2008200380 25 Jan 2008 /2- Ch 00 0 661 00 O
23. The method of claim 22, wherein U is NR 16
24. The method of claim 23, wherein the compound is N II or 0 662 00 N C S00 oo ^L 663
26. The method of claim 23, wherein the compound is selected from the group consisting of: N 0N N N CtI- C Nti N~ N N~ a N O<SQ N N N N crc N N C 0 rNN NN C Nk C N N N& ;and 664
27. The method of claim 23, wherein the compound is selected from the group consisting of: N N N N N N N 3 1% NNN- N.j N CI, .CF3 Nd O ;and
28. The method of claim 3, wherein X is N(CH 3 2 00 S665 F29. The method of claim 28, wherein Y is R 17 -N U 00 00 The method of claim 29, wherein R 13 is an aryl substituted with a CI-Cio straight chained alkyl. 666
31. The method of claim 30, wherein the compound is selected from a group consisting of: (N N.~ [U ;and N N N 667
32. A method of treating a subject suffering from anxiety which comprises administering to the subject an amount of compound effective to treat the subject's anxiety wherein the compound has the structure: N wherein W is H, -Cl, -Br, CN, methyl, ethyl, propyl, methoxy or ethoxy; wherein X is; NR 11 R 1 a; R17 R 1 7 -N R 17 or wherein R 11 is H, straight chained or branched C 1 alkyl, (CH2)q-O-(CH2 )m-CH 3 aryl, or aryl (CI-C 6 )alkyl; wherein R 1 2 is straight chained or branched Ci-C 7 alkyl, 668 (C12)QO 0(CH 2 3 or (CH 2 wherein R 1 3 is a bicyclic alkyl. ring system, adamantyl, noradamantyl, C 3 -C1O cycloalkyl, heteroaryl, aryl, aryl(Ci- CO)alkyl, Q, or Q2; wherein aryl may be substituted with one or more C 1 L-Ci 0 straight chained or branched alkyl, aryl, heteroaryl, or N (Rig) Z; wherein 0, is wherein Q2 is wherein each J is independently 0, S, C(R 2 2 2 or NR 4 wherein R 4 is H; straight chained or branched Cl-C7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or 669 branched C 2 -C7 alkenyl or alkynyl; C 3 -C7 cycloalkyl, CS-C 7 cycloalkenyl or aryl; wherein Y is NR 14 R 1 5 ;or wherein R- 14 is H, straight chained or branched Cl-C 6 alkyl, (CH 2 )q0-(CH2)nrCH3I C 3 -C 6 cycloalkyl, or (C(R, 9 2 )m-Z; wherein R 1 5 is straight chained or branched C 3 -C 6 alkyl, -(CH 2 )m-CH 3 C 3 -C 6 cycloalkyl, (C(Rl 9 2 )mN(RlS)2 or (C (R 19 2 m-Z; wherein R 1 6 is straight chained or branched Cl-C 7 alkyl, straight chained or branched Cl-C 7 monofluoroalkyl, straight chained or branched Cl-C 7 polyfluoroalkyl, straight chained or branched C 2 -C 7 alkenyl, straight 00 670 chained or branched C 2 -C7 alkynyl, Cs-C, cycloalkenyl, (CH 2 or (CH 2 (CH 2 )m-CH 3 wherein each R 1 7 is independently H; -OR 2 1 -OCOR 2 1 -COR 2 1 -NCOR 2 1 -N(R 2 1 2 -CON(R 21 2 -COOR 21 straight chained or 00 branched C 1 alkyl, straight chained or branched C 1 -C, monofluoroalkyl, straight chained or branched Cl-C, 00 polyfluoroalkyl, straight chained or branched C 2 -C7 alkenyl, straight chained or branched C 2 alkynyl, Cs-C, cycloalkenyl, (CH 2 or (CH 2 (CH 2 )m3CH 3 wherein R 18 is straight chained or branched C 1 -C alkyl, (CH 2 or (CH 2 (CH2).-CH3; wherein each R 19 is independently H, or straight chained or branched C 1 -C 6 alkyl; wherein each R 20 is independently straight chained or branched C 1 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or alkynyl; C 3 C, cycloalkyl or Cs-C, cycloalkenyl; -Cl, -Br, or -I; -NO 2 -N 3 -CN; -OR 2 1 -OCOR 21 -COR 21 -NCOR 2 1 -N(R 21 2 CON(R 2 2 or -COOR 21 aryl or heteroaryl; or two R 20 groups present on adjacent carbon atoms can join together to form a methylenedioxy group; wherein each R 21 is independently straight chained or branched C 1 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 alkenyl or alkynyl; C 3 C, cycloalkyl, Cs 5 cycloalkenyl, aryl, or aryl(Cl- C 6 alkyl; 00 671 Swherein each R 22 is independently H, F, Cl or Ci-C 4 straight chained or branched alkyl; wherein each m is an integer from 0 to 4 inclusive; 00 o wherein each n is an integer from 1 to 4 inclusive; 00 wherein p is an integer from 0 to 2 inclusive; wherein q is an integer from 2 to 4 inclusive; wherein t is 1 or 2; wherein U is O, -NRi 6 S, C(R 17 2 or -NSO 2 R 16 wherein Z is C 3 -Cio cycloalkyl, C 4 -C 7 cyclic ether, C 4 -C7 cyclic thioether, aryl, or heteroaryl; or a pharmaceutically acceptable salt thereof.
33. A method of treating a subject suffering from anxiety which comprises administering to the subject an amount of compound effective to treat the subject's anxiety wherein the compound has the structure: 672 wherein W is H, -Cl, -Br, CN, methyl, ethyl, propyl, methoxy or ethoxy; wherein X is NRIR 12 0 00 -N 11 or wherein R 11 is H, straight chained or branched Cl-C 7 alkyl, (CH2)q-O (CH 2 ).-CH 3 aryl or aryl(Cl-Cs)alkyl; wherein R 1 2 is straight chained or branched CI-C 7 alkyl, (CH2)Q-O (CH 2 )M-CH 3 or -(CI- 2 )M-Z; wherein R 13 is a bicyclic alkyl ring system, aryl or ary 1 (C CO) a lkyl; wherein Y is NR 14 R 15 00 673 00-N Uor 00 ~R 19 00 R2 -N wherein R 14 is H, straight chained or branched Cl-C 6 alkyl, (CH 2 q 0 (CH 2 mCH 3 C 3 -C 6 cycloalkyl, or (C (R 19 2 )m-Z; wherein Ris is straight chained or branched C 3 -C 6 alkyl, (CH 2 )qO1CH 2 )m-CH3, C 3 -C6 cycloalkyl, or (C(R 1 9 2 )m-Z; wherein U is 0, -NR, 6 S, C(Rl 7 2 or -NS0 2 R 16 wherein Z is C 3 -Clo cycloalkyl, aryl, or heteroaryl; wherein R 16 is straight chained or branched CI-C7 alkyl, straight chained or branched Cl-C 7 monofluoroalkyl, straight chained or branched Cl-C 7 polyfluoroalkyl, straight chained or branched C 2 -C7 alkenyl, straight chained or branched C 2 -C7 alkynyl, CS-C 7 cycloalkenyl, (CH 2 or (CH 2 )q0O(CH 2 )m-CH3; 00 674 0 wherein each R17 is independently H; -OR 2 1 -OCOR21, -COR 2 1 -NCOR 2 1 -N(R 2 1 2 -CON(R 2 1 2 -COOR 2 1 straight chained or CI branched C 1 alkyl, straight chained or branched C 1 -C, monofluoroalkyl, straight chained or branched C 1 -C 7 polyfluoroalkyl, straight chained or branched C 2 -C, 00 Mr) alkenyl, straight chained or branched C 2 alkynyl, C 5 -C, cycloalkenyl, -(CH 2 or (CH 2 )n-O-(CH 2 )m-CH 3 00 wherein R 18 is straight chained or branched C 1 -C alkyl, (CH 2 m Z, or (CH 2 q-0- (CH 2 m-CH 3 wherein each R 19 is independently H, or straight chained or branched C 1 -C 6 alkyl; wherein each R 20 is independently straight chained or branched C 1 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or alkynyl; C 3 C, cycloalkyl or C 5 cycloalkenyl; -C1, -Br, or -I; -NO 2 -N 3 -CN; -OR 21 -OCOR 21 -COR 2 1, -NCOR 2 1 -N(R 2 1 2 CON(R 21 2 or -COOR 2 1; aryl or heteroaryl; or two R 20 groups present on adjacent carbon atoms can join together to form a methylenedioxy group; wherein each R 21 is independently straight chained or branched C 1 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 alkenyl or alkynyl; C 3 C, cycloalkyl, Cs-C7. cycloalkenyl, aryl or aryl(C 1 C 6 alkyl; wherein each m is an integer from 0 to 4 inclusive; wherein each n is an integer from 1 to 4 inclusive; 675 wherein p is an integer from 0 to 2 inclusive; wherein q is an integer from 2 to 4 inclusive; wherein t is 1 or 2; or a pharmaceutically acceptable salt thereof.
34. A anxiety amount anxiety method of treating a subject suffering from which comprises administering to the subject an of compound effective to treat the subject's wherein the compound has the structure: wherein W is H, -Cl, -Br, CN, methyl, ethyl, propyl, methoxy or ethoxy; wherein X is N(CH 3 )2 or 676 wherein. R 13 is an aryl, adamantyl, noradamantyl, C 3 -Cj 0 cycloalkyl, heteroaryl, Q, or Q2; wherein aryl may be substituted with one or more C 1 -CIO straight chained or branched alkyl, aryl, heteroaryl, or N (R 19 -Z; wherein Q3. is wherein Q2 is wherein each J is independently 0, S, 0(R 2 2 2 or NR 4 wherein R 4 is straight chained or branched Cl-C, alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C 7 alkenyl or alkynyl; C3-C7 cycloalkyl, C 5 -C 7 cycloalkenyl or aryl; 00 677 wherein Y is NR 14 R 15 17 00R2 -N 19 R 1 7 R 20 -N wherein RIA is H, straight chained or branched C 1 -C 6 alkyl, (CHO)q-0- CHO)rnCH3I C 3 -C 6 cycloalkyl, or (C (Rig) 2 wherein R 15 is straight chained or branched C 3 -CG alkyl, (CH 2 )qO1CH2)m-CH3, C 3 -C 6 cycloalkyl, or (C(R 1 9 2 wherein U is 0, -NR16, S, C(R 1 7 2 or -NS0 2 R1 6 wherein Z is C 3 -Cl 0 cycloalkyl, aryl, or heteroaryl; wherein R 16 is straight chained or branched C 1 -C 7 alkyl, straight chained or branched C 1 -C 7 monofluoroalkyl, straight chained or branched Cl-C 7 polyfluoroalkyl, 00 678 straight chained or branched C 2 -C 7 alkenyl, straight chained or branched C 2 -C 7 alkynyl, C 5 -C 7 cycloalkenyl, (CH 2 Or (CH 2 )9-0-(CH 2 )m-CH 3 each R 17 is independently H; -OR 21 -0C0R 21 -CaR 21 00 M~f -NCOR 21 -N(R 21 2 -CON(R 2 1 -C00R 21 straight chained or branched C 1 -C 7 alkyl, straight chained or branched Cl-C 7 00 monofluoroalkyl, straight chained or branched C 1 -C 7 polyfluoroalkyl, straight chained or branched C 2 -C-1 alkenyl, straight chained or branched C 2 -C 7 alkynyl, C 5 -C-7 cycloalkenyl, -(CH 2 or (CH 2 )n-O-(CH 2 )m-CH 3 wherein R 1 8 is straight chained or branched CI-C 6 alkyl, (CH 2 or (CH2)q-O (CH 2 )m-CH 3 wherein each R3. 9 is independently H, or straight chained or branched C 1 -C 6 alkyl; wherein each R 2 o is independently straight chained or branched Cl-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C- 7 alkenyl or alkynyl; C 3 C 7 cycloalkyl Or C 5 -C7 cycloalkenyl; -C1, -Br, or -I; -NO 2 -N 3 -CM; -OR 21 -0C0R 21 -C0R 21 -NCOR 21 0 -N(R 21 2 CON(R 21 2 or -C00R 2 1 aryl or heteroaryl; or two R 20 groups present on adjacent carbon atoms can join together to form a methylenedioxy group; wherein each R 21 is independently straight chained or branched C 1 -C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C 7 alkenyl or alkynyl; C 3 C 7 cycloalkyl, C 5 -C 7 cycloalkenyl, aryl or aryl (Cl- C 6 alkyl i 00 679 Swherein each R 22 is independently H, F, Cl or C 1 -C 4 straight chained or branched alkyl; wherein each m is an integer from 0 to 4 inclusive; 00 M1 wherein each n is an integer from 1 to 4 inclusive; 00 wherein p is an integer from 0 to 2 inclusive; c 10 wherein q is an integer from 2 to 4 inclusive; wherein t is 1 or 2; or a pharmaceutically acceptable salt thereof. A method of treating a subject suffering from anxiety which comprises administering to the subject an amount of compound effective to treat the subject's anxiety wherein the compound has the structure: X W N yR 1 3 H wherein W is H, -Cl, -Br, CN, methyl, ethyl, propyl, methoxy or ethoxy; wherein X is N(CH 3 2 or 00 680 R 17 c-I-N 0 00 wherein R 13 is a bicyclic alkyl ring system, aryl or c-Iaryl (CI-C 6 alkyl; 00 wherein Y is NR 14 R 15 wherein R 1 4 is H1, straight chained or branched Cj-C 6 alkyl, (CH 2 )q0O-(CH 2 )n-CH3, C 3 -C 6 cycloalkyl, or (C(R 1 9 2 )m-Z; wherein.R 15 is (C(R 1 9 2 )m-N(Rl 6 )2; wherein Z is C 3 -C 10 cycloalkyl, aryl, or heteroaryl; wherein R 1 6 is straight chained or branched Cl-C,7 alkyl, straight chained or branched Cl-C7 monofluoroalkyl, straight chained or branched Cl-C7 polyfluoroalkyl, straight chained or branched C 2 -C7 alkenyl, straight chained or branched C 2 -C7 alkynyl, CS-C7 cycloalkenyl, (CH 2 or (CH 2 )q-0-(CH 2 )m-CH3; wherein each R 1 7 is independently H; -OR 2 1 -0C0R 21 -C0R 21 -NCOR 21 -N(R 21 2 -CON(R 2 1D 2 -C00R 21 straight chained or branched Cl-C7 al~kyl, straight chained or branched Cl-C7 monofluoroalkyl, straight chained or branched Cl-C7 polyfluoroalkyl, straight chained or branched C 2 -C7 alkenyl, straight chained or branched C 2 -C7 alkynyl, Cs-C7 cycloalkenyl, -(CH 2 or (CH 2 )n0O(CH 2 )m-CH 3 S681 wherein each R 19 is independently H, or straight chained Cq or branched Ci-C 6 alkyl; wherein each R 21 is independently straight chained or 00 CO branched Ci-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or alkynyl; C 3 00 C 7 cycloalkyl, Cs-C, cycloalkenyl, aryl or aryl(C 1 C) alkyl; C wherein each m is an integer from 0 to 4 inclusive; wherein each n is an integer from 1 to 4 inclusive; wherein q is an integer from 2 to 4 inclusive; or a pharmaceutically acceptable salt thereof.
36. The method of claim 32, 33, 34 or 35, wherein the compound is enantiomerically and diasteriomerically pure.
37. The method of claim 32, 33, 34 or 35, wherein the compound is enantiomerically or diasteriomerically pure.
38. The method of claim 32, 33, 34 or 35, wherein the compound can be administered orally. 682
39. The method of claim 32, wherein X is: R 17 R, -N R1 R 17 N N--R, R 1 7 or The method of claim 32, wherein X is NR 1 R 12 and R 11 is H or straight chained or branched C 1 -C7 alkyl.
41. The method of claim 40, wherein the compound has the structure:
42. The method of claim 39, wherein alkyl ring system, cyclohexyl or aryl.
43. The method of claim 41, wherein alkyl ring system, cyclohexyl or aryl. RI 3 is a bicyclic R 13 is a bicyclic
44. The method of claim 42, wherein R 14 is H, straight chained or branched CI-Cs alkyl or (CH 2 (CH 2 )m-CH 3 00 683
45. The method of claim 43, wherein R 14 is H, straight Ci chained or branched Ci-C 6 alkyl or (CH 2 )q-O-(CH 2 )m-CH 3 O 5 46. The method of claim 44, wherein the compound is 00 Cc selected from the group consisting of: 00 A o0, 2008200380 25 Jan 2008 .zo 2008200380 25 Jan 2008 aD uL /ID \O /O z2 2008200380 25 Jan 2008 U, z*~0 -ZK~ 00 0 687 (N
47. The method of claim 42, wherein Y is R 17 00 -N U M/
48. The method of claim 47, wherein U is NR 16
49. The method of claim 48, wherein R 16 is (CH 2 )1-Z. The method of claim 49, wherein Z is aryl or heteroaryl.
51. The method of claim 50, wherein the compound is selected from the group consisting of: 2008200380 25 Jan 2008 C) O O 2008200380 25 Jan 2008 z/ KID CONC N- /o z~ D /OD 690
52. The method of claim 43, wherein the compound is selected from the group consisting of: Cl N 1C l0 0 1 HNLN N. \N N 2008200380 25 Jan 2008 00 00 00 O- i 0 mq o0 0D 692
53. The method of claim 43, wherein Y is R 17 -N U *P R17
54. The method of claim 53, wherein U is NR16. The method of claim 54, wherein the compound is or 0 0693
56. The method of claim 50, wherein the compound is N N N 0 NJ 00 o 1 694
57. The method of claim 54, wherein the compound is selected from the group consisting of: N O N NI IN 0 N N I N NN N CI N NIL N N CI N CI UNN 0N Cl f^ ^N0 ^N^ ;and 00 695
58. The method of claim 54, wherein the compound is selected from the group consisting of: Ne, 'IN,"N N 00 00 'ySCIF (NN 3' r- N N,) INN N 11 No; F 3 ;and NN
59. The method of claim 34, wherein X is N(CH 3 2 00 696 The method of claim 59, wherein Y is 17 -N U 00 SR R17
61. The method of claim 60, wherein R 13 is an aryl substituted with a CI-Co 0 straight chained alkyl. 697
62. The method of claim wherein the compound is selected from a group consisting of: 'N Y ;and N KNN C-C, IN 698
63. A pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound having the structure: wherein W is H, -Cl, -Br, CN, methyl, ethyl, propyl, methoxy or ethoxy; wherein X is; NR11R12; R 17 R 17 N or -N N-R R 17 wherein R 11 is H, straight chained or branched C 1 -C7 alkyl, (CH 2 (CH 2 )m-CH 3 aryl, or aryl (C-C 6 )alkyl; wherein R 12 is straight chained or branched Cl-C 7 alkyl, (CH 2 )q0-(CH2) .CH3, or (CH 2 )m-Z; 699 wherein R3. 3 is a bicyclic alkyl ring system, adamantyl, noradamantyl, C 3 -CIO cycloalkyl, heteroaryl, aryl, aryl(Cl- CO)alkyl, Q, or Q2; wherein aryl may be substituted with one or more Cl-Clo straight chained or branched alkyl, aryl, heteroaryl, or N (Rig) -Z; wherein Q, is wherein Q2 is wherein each LT is independently 0, S, C(R 2 2)2 or NR 4 wherein R 4 is H; straight chained or branched Cl-C,? alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or alkynyl; C 3 -C 7 cycloalkyl, C 5 -C7 700 cycloalkenyl or aryl; wherein Y is NR 14 R 1 5 ;or wherein R 14 is H, straight chained or branched C 1 -C 6 alkyl, (CH 2 )q01-CH 2 )m-H3: C 3 -c 6 cycloalkyl, or (C(R 1 9 2 )mZ; wherein R 15 is straight chained or branched C 3 -C 6 alkyl, (CH2)q-O'(CH2 )m-CH3, C 3 -C 6 cycloalkyl, (C(R 1 9 2 )mN(R,6)2' Or (C (R 19 2 Z wherein R 16 is straight chained or branched Cl-C7 alkyl, straight chained or branched Cl-C 7 ronofluoroalkyl, straight chained or branched C 7 polyfluoroalkyl, straight chained or branched C 2 -C 7 alkenyl, straight chained or branched C 2 -C 7 alkynyl, C 5 -C 7 cycloalkenyl, 00 701 (CH 2 or (CH2)q-O-(CH2)m-CH3; tfl wherein each R 1 7 is independently H; -OR 21 -0C0R 21 -C0R 21 -NCOR 21 -N(R 21 2 -CON(R 21 2 -C00R 21 straight chained or branched Cl-C7 alkyl, straight chained or branched Cl-C7 00 monofluoroalkyl, straight chained or branched C 1 -C, M polyfluoroalkyl, straight chained or branched C 2 -C7 0 00 cycloalkenyl, -(CH 2 or (CH 2 )n-(CH 2 )m-CH 3 wherein R Is is straight chained or branched C 1 -C 6 alkyl, (CH 2 or (CH 2 )q-O-(CH2)m-CH3; wherein each R 19 is independently H, or straight chained or branched C 1 -Cr, alkyl; wherein each R 20 is independently straight chained or branched CI-C,7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 aJlkenyl or alkynyl; C 3 C7 cycloalkyl or C5-C, cycloalkenyl; -C1, -Br, or -I; -NO 2 -N 3 -CN; -OR 2 1 -0C0R 21 -C0R 21 -NCOR 21 -N(R 21 2 CON(R 2 1 2 or -C00R 21 aryl or heteroaryl; or two R 20 groups present on adjacent carbon atoms can join together to form a methylenedioxy group; wherein each R 2 1 is independently straight chained or branched Cl-C7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or alkynyl; C 3 C, cycloalkyl, C 5 -C7 cycloalkenyl, aryl, or arylCl- CG) alkyl; wherein each R 2 2 is independently H, F, Cl or CI-C 4 00 702 Sstraight chained or branched alkyl; V) wherein each m is an integer from 0 to 4 inclusive; wherein each n is an integer from 1 to 4 inclusive; 00 O wherein p is an integer from 0 to 2 inclusive; 0 0 0 wherein q is an integer from 2 to 4 inclusive; wherein t is 1 or 2; wherein U is 0, -NR6s, S, C(R 17 2 or -NSO 2 R 1 s; wherein Z is C 3 -Clo cycloalkyl, C 4 -C 7 cyclic ether, C 4 -C 7 cyclic thioether, aryl, or heteroaryl; or a pharmaceutically acceptable salt thereof.
64. A pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound having the structure: X N Y NN H wherein W is H, -Cl, -Br, CN, methyl, ethyl, 00 703 propyl, Tnethoxy or ethoxy; wherein X is NR11R12; R1R 1 .R R 1 7 00 z R 17 00R1 wherein R 1 3. is H, straight chained or branched CI-C 7 alkyl, (CH 2 )q0.-(CH 2 )m-CH3, aryl or aryl(Cl-C6)alkyl; wherein R 12 is straight chained or branched Cl-C 7 alkyl, (CH 2 )q0I-CH 2 )m-CH3i or -C2MZ wherein R 13 is a bicyclic alkyl ring system, aryl or aryl (C CG) alkyl, wherein Y is NR 14 R 1 5 00 704 R 0 -N U R 19 p or 00 00 R2 -N wherein R 14 is H, straight chained or branched C 1 -C 6 alkyl, (CH 2 )q-O-0CH 2 )m-CH3i C 3 -C 6 cycloalkyl, or (C(R 1 9 2 )m-Z; wherein R 15 is straight chained or branched C 3 -C 6 alkyl, (CH 2 )q-O-1CH2)CH3I C 3 -C 6 cycloalkyl, or (C(R 1 9 2 )m-Z; wherein U is 0, -NR 16 S, C(R 17 2 or -NS0 2 R 1 6 wherein Z is C 3 -CIO cycloalkyl, aryl, or heteroaryl; wherein R 16 is straight chained or branched C 1 -C7 alkyl, straight chained or branched Cl-C 7 monofluoroalkyl, straight chained or branched C 1 -C 7 polyfluoroalkyl, straight chained or branched C 2 -C7 alkenyl, straight chained or branched C 2 -C 7 alkynyl, Cs-C7 cycloalkenyl, (CH 2 or (CH 2 (CH 2 )m-CH 3 00 705 wherein each RI7 is independently H; -OR 21 -OCOR 21 -C0R 21 NCOR 21 -N(R 21 2 -CON(R 21 2 -COOR 21 straight chained or V) branched Cl-C,? alkyl, straight chained or branched Cl-C7 monofluoroalkyl, straight chained or branched Cl-C7 polyfluoroalkyl, straight chained or branched C 2 -C7 00 alkenyl, straight chained or branched C 2 -C7 alkynyl, CS-C 7 cycloalkenyl, -(CH 2 or (CH 2 (CH 2 )m-CM 3 00 wherein R3. 8 is straight chained or branched C 1 -C 6 alkyl, (CH 2 )mnZ, or (CH2) q-0 (CH 2 )m.CH 3 wherein each RI 9 is independently H, or straight chained or branched C 1 -C 6 alkyl; wherein each R 2 0 is independently straight chained or branched Cl-C7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or alkynyl; C 3 C7 cycloalkyl or C 5 -C7 cycloalkenyl; -C1, -Br, or -I; -NO 2 -N 3 -CN; -OR 2 1 -OCOR 21 -COR21, -NCOR 21 -N(R 21 2 CON(R 2 1 2 or -COOR 21 aryl or heteroaryl; or two R 20 groups present on adjacent carbon atoms can join together to form a methylenedioxY group; wherein each R 21 is independently straight chained or branched Cl-C7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or alkynyl; C 3 C7 cycloalkyl, C 5 -C7 cycloalkenyl, aryl or aryl(Cl- C 6 alkyl; wherein each m is an integer from 0 to 4 inclusive; 00 706 Lc wherein each n is an integer from 1 to 4 inclusive; wherein p is an integer from 0 to 2 inclusive; 00 wherein q is an integer from 2 to 4 inclusive; eg wherein t is 1 or 2; or 00 C 10 a pharmaceutically acceptable salt thereof. A pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound having the structure: x NW N N/R,,13 Y N N wherein W is H, -C1, -Br, CN, methyl, ethyl, propyl, methoxy or ethoxy; wherein X is N(CH 3 )2 or 707 wherein R 13 is an aryl, adamantyl, noradamantyl, C 3 -C1 0 cycloaJlkyl, heteroaryl, Q, or Q2; wherein aryl may be substituted with one or more Cl-Clo straight chained or branched alkyl, aryl, heteroaryl, or N (R 1 i 9 Z; wherein Q, is wherein 02 is wherein each J is independently 0, S, C(R 2 2 2 or NR 4 00 708 wherein R 4 is Straight chained or branched Cl-C7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight V) chained or branched C 2 -C. 7 alkenyl or alkynyl; C 3 -C 7 cycloalkyl, CS-C 7 cycloalkenyl or aryl; 00 wherein Y is NR 14 R 15 R 20 00 R 17 R 2 0',I -N r R 17 R 1 X R 20 -N R 20 wherein R3. 4 is H, straight chained or branched Cl-C 6 alkyl, (CH 2 )q0I-CH 2 )M-CH3, C 3 -C 6 cycloalkyl, or (C(R 1 9 2 )m1,Z; wherein R- 1 5 is straight chained or branched C 3 -C 6 alkyl, (CH 2 )qO4CH2)m-CH3: C 3 -C 6 cycloalkyl, or (C(R 19 2 )m-Z. wherein U is 0, -NR 16 S, C(R, 7 2 or -NS0 2 R, 6 wherein Z is C 3 -ClO cycloalkyl, aryl, or heteroaryl; 00 709 wherein R 1 6 is straight chained or branched C 1 -C7 alkyl, straight chained or branched Cl-C 7 mnono fluoroa lkyl, straight chained or branched Cl-C 7 polyfluoroalkyl, straight chained or branched C 2 -C 7 alkeny)., straight 00 chained or branched C 2 -C 7 alkynyl, C 5 -C7 cycloalkenyl, (CH 2 )mZ, or (CH2)q-O (CH 2 )m-CH 3 00 wherein each R 17 is independently H; -OR 2 1 -0C0R 2 1 -C0R 21 -NCOR 21 -N(R 21 2 -CONCR 2 2 -C00R 21 straight chained or branched Cl-C7 alkyl, straight chained or branched CI-C 7 monofluoroalkYl, straight chained or branched C 1 -C 7 polyfluoroalkyl, straight chained or branched C 2 -C 7 alkenyl, straight chained or branched C 2 -C 7 alkynyl, C 5 -C 7 cycloalkenyl, -(CH 2 or (CH 2 2 )m-CH 3 wherein R 18 is straight chained or branched C 1 -CG alkyl, (CH 2 or (CH2)q-O (CH 2 )m-CH 3 i wherein each Rig is independently H, or straight chained or branched Cl-C 6 alkyl; wherein each R 20 is independently straight chained or branched Cl-C7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C 7 alkenyl or alkynyl; C 3 C 7 cycloalkyl or C 5 -C 7 cycloalkenyl; -CI, -Br, or -I; -NO 2 -N 3 -CM; -OR 21 -OCOR 2 1, -C0R 21 -NCOR 21 -N(R 21 2 CON(R 2 3.) 2 or -COOR 21 aryl or heteroaryl; or two R 20 groups present on adjacent carbon atoms can join together to form a methylenedioxy group; wherein each R 21 is independently straight chained or 0O 0710 Sbranched CI-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or alkynyl; C 3 C 7 cycloalkyl, C 5 cycloalkenyl, aryl or aryl(C 1 C 6 alkyl; 00 wherein each R 22 is independently H, F, Cl or C 1 -C 4 0 straight chained or branched alkyl; 00 Swherein each m is an integer from 0 to 4 inclusive; 4 1 0 wherein each n is an integer from 1 to 4-inclusive; wherein p is an integer from 0 to 2 inclusive; wherein q is an integer from 2 to 4 inclusive; wherein t is 1 or 2; or a pharmaceutically acceptable salt thereof.
66. A pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound having the structure: 711 wherein W is H, -Cl, -Br, CN, methyl, ethyl, propyl, methoxy or ethoxy; wherein X is N(CH 3 2 or wherein R 13 is a bicyclic alkyl ring system, aryl or aryl (Cl-C 6 alkyl; wherein Y is NR 14 R 15 wherein R 1 4 is H, straight chained or branched Cl-C 6 alkyl, (CH 2 q0-CH2) nCCH3, C 3 -C 6 cycloalkyl, or (C (R1 9 2 mZ; wherein R 1 5 is (C(R 1 9 2 )m-N(R 1 6)2; wherein Z is C 3 -C3.0 cycloalkyl, aryl, or heteroaryl; wherein R 16 is straight chained or branched Cl-C 7 alkyl, straight chained or branched Cl-C 7 monofluoroalkyl, straight chained or branched Cl-C 7 polyfluoroalkyl, straight chained or branched C 2 -C 7 alkenyl, straight chained or branched C 2 -C 7 alkynyl, C 5 -C 7 cycloalkenyl, 00 712 (CH 2 or (CH 2 )q-O-(CH 2 )m-CH 3 Ct- Swherein each R17 is independently H; -OR 21 -OCOR 21 -COR 21 -NCOR 21 -N(R 21 2 -CON(R 21 2 -COOR 2 1 straight chained or branched Ci-C7 alkyl, straight chained or branched Ci-C7 00 monofluoroalkyl, straight chained or branched C 1 -C7 polyfluoroalkyl, straight chained or branched C 2 -C 7 Salkenyl, straight chained or branched C 2 -C7 alkynyl, 0 0 cycloalkenyl, -(CH 2 or (CH 2 )n--CH 2 )m-CH 3 wherein each Rs 1 is independently H, or straight chained or branched Ci-C 6 alkyl; wherein each R 21 is independently straight chained or branched CI-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or alkynyl; C 3 C7 cycloalkyl, C 5 -C 7 cycloalkenyl, aryl or aryl(Ci- C 6 alkyl; wherein each m is an integer from 0 to 4 inclusive; wherein each n is an integer from 1 to 4 inclusive; wherein q is an integer from 2 to 4 inclusive; or a pharmaceutically acceptable salt thereof.
67. The pharmaceutical composition of claim 63, 64, or 66, wherein the compound is enantiomerically and diasteriomerically pure.
68. The pharmaceutical composition of claim 63, 64, 713 or 66, wherein the compound is enantiomerically or diasteriomerically pure.
69. The pharmaceutical composition of claim 63, 64, or 66,, wherein the compound can be administered orally. The pharmaceutical composition of claim 63, wherein X is: R 7 R17 R1, N r 17 -N N--R, R17 or
71. The pharmaceutical composition of claim 63, wherein X is NR 11 R 12 and R1 is H or straight chained, or branched Ci-C7 alkyl. 00 714
72. The pharmaceutical composition of claim 71, wherein c the compound has the structure: )N R12 N 00 N OC I N NR13
73. The pharmaceutical composition of claim 70, wherein R 13 is a bicyclic alkyl ring system, cyclohexyl or aryl.
74. The pharmaceutical composition of claim 72, wherein R 13 is a bicyclic alkyl ring system, cyclohexyl or aryl. The pharmaceutical composition of claim 73, wherein R 14 is H, straight chained or branched C 1 -C 6 alkyl or (CH2) q-0- (CH 2 -CH 3
76.The pharmaceutical composition of claim 74, wherein R 14 is H, straight chained or branched Ci-C 6 alkyl. or (CH 2 (CH 2 )m-CH 3 715
77. The pharmaceutical composition of claim 73, wherein Y is R 1 7 U r- J Rl7
78. The pharmaceutical composition of claim U is NR 16 79 The pharmaceutical composition of claim R 16 is (CH 2 )m-Z. LO The pharmaceutical composition of claim Z is aryl or heteroaryl.
81. The pharmaceutical composition of claim Y is 77, wherein 78, wherein 79, wherein 74, wherein R17
82. The pharmaceutical composition of claim 81, wherein U is NR 16 716
83. The pharmaceutical composition of claim 82, wherein the compound is selected from the group consisting of: 00 00 MO 00 N N N N 0 $NN/ r N N- N rN N N N N C I N N $NN N Cf N: QN N N CI N N C1 rN N N "N N NO11 ;and 717
84. The pharmaceutical composition of claim 82, wherein the compound is selected from the group consisting of: N, N SN' N No" N C 'N N N N N N N, N NN Q-~c- ON'N"NKN'O N N (CI c CF3 NN N N N el and 0 0 718 The pharmaceutical composition of claim 65, wherein SX is N(CH 3 2 c,
86. The pharmaceutical composition of claim 85, wherein Y is 00 M R 17 00 S-N U R 1 7
87.The pharmaceutical composition of claim 86, wherein R 13 is an aryl substituted with a Ci-Cio straight chained alkyl. 00 719
88. The pharmaceutical composition of claim 87, wherein the compound is selected from a group consisting of: 00 NN 00 N I-NI Nand NN NINI 720
89. A compound having the structure: wherein W is H, -Cl, propyl, methoxy or ethoxy; wherein X is; NR 11 R 12 R 17 R1 -N R 17 -Br, CN, methyl, ethyl, or 17. -N N-R 1 8 R1 wherein R 11 is H, straight chained or branched Cj-C 7 alkyl, (CH 2 )q-O (CH 2 )m-CH 3 aryl, or aryl (Cl-C 6 alkyl; wherein R 1 2 is straight chained or branched Cl-C 7 alkyl, (CH 2 CH2) M-CH3, or -(CH 2 m-Z; wherein R1 3 is a bicyclic alkyl ring system, adamantyl, 721 noradamantyl, C 3 -C1o cycloalkyl, heteroaryl, aryl, aryl(Cl- C 6 alkyl, Q, or Q2; wherein aryl may be substituted with one or more CI-Clc straight chained or branched alkyl, aryl, heteroaryl, or N (R 19 -Z; wherein Q, is wherein Q2 is wherein each J is independently 0, S, C(R 2 2 )2 or NR 4 wherein R 4 is H; straight chained or branched Cl-C,7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C 7 alkenyl or alkynyl; C 3 -C 7 cycloalkyl, C 5 -C-I cycloalkenyl or aryl; 722 wherein Y is NR 14 Rls; ;or wherein R 1 4 is H, straight chained or branched C 1 I-C 6 alkyl, (CH 2 )qO(C 2 )m-CH3I C 3 -C 6 cycloalkyl, or (C(Ra,) 2 )M-Z; wherein R 15 is straight chained or branched C 3 -C 6 alkyl, (CH 2 )qO0-(CH 2 )nCH3, C 3 -C 6 cycloalkyl, (C (Rig) 2 )MN (Ri6)2 or (C (Rig) 2 Z; wherein R 16 is straight chained or branched Cl-C 7 alkyl, straight chained or branched Cl-C7 monofluoroalkyl, straight chained or branched Cl-C 7 polyfluoroalkyl, straight chained or branched C 2 -C 7 alkenyl, straight chained or branched C 2 -C 7 alkyny., C 5 9-C7 cycloalkenyl, (CH 2 or (CH2)q-O (CH 2 )m-CH 3 00 723 wherein each R 1 7 is independently H; -OR 2 1 -OCOR 21 -C0R 21 -NCOR 21 -N(R 21 2 -CON(R 21 2 -COOR 21 straight chained or branched C 1 -C,7 alkyl, straight chained or branched CI-C7 monofluoroalkyl, straight chained or branched Cl-C7 polyfluoroalkyl, straight chained or branched C 2 -C7 00 alkenyl, straight chained or branched C 2 -C,7 alkynyl, CS-C7 cycloalkenyl, -(CH 2 or (CH 2 )nO-(CH 2 )m-CH 3 00 wherein R 18 is straight chained or branched C 1 -C 6 alkyl, (CH 2 or (CH 2 )q-O-(CH 2 )m,,CH3; wherein each R 19 is independently H, or straight chained or branched Cl-C 6 alkyl; wherein each R 2 0 is independently straight chained or branched C 1 -C,7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or: alkynyl; C 3 C 7 cycloalkyl or Cs-Cy cycloalkenyl; -C1, -Br, or -I; -NO 2 -N 3 -CN; -OR 21 -0C0R 21 -C0R 21 NCOR 21 -N(R 21 2 CON(R 2 1 2 or -C00R 21 aryl or heteroaryl; or two R 2 0 groups present on adjacent carbon atoms can join together to form a methylenedioxy group; wherein each R 21 is independently straight chained or branched Cl-C7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C,7 alkenyl or alkynyl; C3- C,7 cycloalkyl, C 5 -C7 cycloalkenyl, aryl, or aryl (Cl- CO) alkyl; wherein each R 2 2 is independently H, F, C1 or CJ-C 4 straight chained or branched alkyl; 00 724 wherein each m is an integer from 0 to 4 inclusive; Vn wherein each n is an integer from 1 to 4 inclusive; wherein p is an integer from 0 to 2 inclusive; 00 C^ 0 wherein q is an integer from 2 to 4 inclusive; 00 0wherein t is 1 or 2 wherein U is O, -NR16, C(R 17 or -NSOzR 16 wherein Z is C 3 -C0 cycloalkyl, C 4 cyclic ether, C 4 -C 7 cyclic thioether, aryl, or heteroaryl; or a pharmaceutically acceptable salt thereof. A compound having the structure: X W N R, Y N N H wherein W is H, -Cl, -Br, CN, methyl, ethyl, propyl, methoxy or ethoxy; 00 725 wherein X is NRl 1 Rl 2 InR 1 7 R 17 o r N R s 00 1 00 wherein R11 iB H, straight chained or branched Cl-C, alkyl, (CH 2 q0-(C2)m-CH3, aryl or aryl (Cl-C 6 alkyl; (Ni wherein R 12 is straight chained or branched Cl-C7 alkyl, (C2)q 0 -(CM 2 )m-CH 3 or -(CH 2 )m-Z; wherein R 13 is a bicyclic alkyl ring system, aryl or aryl (Cl-CG) alkyl; wherein Y is NR 14 Ras; 00 726 R 20 R 2 0 R 1 N Up ;or 00 [~R1 1 00 -N wherein R 14 is H, straight chained or branched Cl-C 6 alkyl, (CH 2 )q0O(CH2)M-cH3u C 3 -C6 cycloalkyl,. or (C(R, 9 )m-Z; wherein R 15 is straight chained or branched C 3 -C 6 alkyl, (CH 2 )q0O(ICH 2 ),mCH3, C 3 -C 6 cycloalkyl, or (C(Rig) 2 wherein U is 0, -NR, 6 S, C(Rl 7 2 or -NS0 2 R, 6 wherein Z is C 3 -Cl 0 cycloalkyl, aryl, or heteroaryl; wherein Rls is straight chained or branched Cl-C 7 alkyl, straight chained or branched C 1 I-C 7 monofluoroalkyl, straight chained or branched Cl-C 7 polyfluoroalkyl, straight chained or branched C 2 -C7 alkenyl, straight chained or branched C 2 -C7 alkynyl, C 5 -C 7 cycJloalkenyl, (CH 2 or (CH2)q0 -(CH 2 )m-CH 3 00 727 wherein each R17 is independently H; -OR 21 -OCOR 21 -COR 2 1 -NCOR 21 -N(R 21 2 -CON(R 21 2 -COOR 21 straight chained or In branched C 1 alkyl, straight chained or branched C 1 -C, monofluoroalkyl, straight chained or branched C 1 -C 7 polyfluoroalkyl, straight chained or branched C 2 -C, 00 alkenyl, straight chained or branched C 2 -C7 alkynyl, C 5 -C, 0 cycloalkenyl, -(CH 2 or (CH 2 )n-O-(CH 2 )m-CH3; 00 wherein R 18 is straight chained or branched C 1 -Cs alkyl, (CH 2 or (CH 2 )q-O-(CH2),-mH3; wherein each R 19 is independently H, or straight chained or branched C 1 -C 6 alkyl; wherein each R 20 is independently straight chained or branched C 1 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C 7 alkenyl or alkynyl; C 3 C, cycloalkyl or Cs-C, cycloalkenyl; -Cl, -Br, or -I; -NO 2 -N3; -CN; -OR 2 1 -OCOR 21 -COR 21 -NCOR 2 1, -N(R 2 1) 2 CON(R 2 1 2 or -COOR 21 aryl or heteroaryl; or two R 20 groups present on adjacent carbon atoms can join together to form a methylenedioxy group; wherein each R 21 is independently straight chained or branched C 1 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 alkenyl or alkynyl; C 3 C, cycloalkyl, Cs-C, cycloalkenyl, aryl or aryl(Cl- C 6 )alkyl; wherein each m is an integer from 0 to 4 inclusive; 00 oO 728 C3 wherein each n is an integer from 1 to 4 inclusive; a pharmaceutically acceptable salt thereof.
91. A compound having the structure: N wherein W is H, -C1, -Br, CN, methyl, ethyl, propyl, methoxy or ethoxy; wherein X is N(CH 3 2 or wherein X is N(CH13)2 or 729 wherein R 13 is an aryl, adamantyl, noradamantyl, C 3 -CIO cycloalkyl, heteroaryl, Q3. or Q2; wherein aryl may be substituted with one or more C 1 -Cl 0 straight chained or branched alkyl, aryl, heteroaryl, or N (Rig) Z; wherein Q, is wherein Q2 is !0 wherein each J is independently 0, S, C(R 2 2 2 or NR 4 wherein R 4 is straight chained or branched Cl-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 a lkenyl or alkynyl; C 3 -C 7 cycloalkyl, CS-C 7 cycloalkenyl or aryl; 00 730 wherein Y is NR 14 R, 5 c-I R 20 R 17 00 R 2 0 -N /u 00 R, 9 R 2 0 -N wherein R 1 4 is H, straight chained or branched Cl-C 6 alkyl, (CH 2 )q0O-(CH 2 C 3 -C6 cycloalkyl, or (C(Ri 3 9)m,,Z; wherein R3. 5 is straight chained or branched C 3 -C 6 alkyl, (CH 2 )qO-(H 2 )m-CH 3 C 3 -C 6 cycloalkyl, or (C(R 1 9 2 wherein U is 0, -NR 16 S, C(Rl 7 2 or -NS0 2 R 16 wherein Z is C: 3 -Cl 0 cycloalkyl, aryl, or heteroaryl; wherein R 1 6 is straight chained or branched Cl-C 7 alkyl, straight chained or branched C 1 -C 7 monofluoroalkyl, straight chained or branched CI-C7 polyfluoroalkyl, OC 00 731 straight chained or branched C 2 -C7 alkenyl, straight chained or branched C 2 alkynyl, C 5 -C 7 cycloalkenyl, c- (CH 2 or (CH2)q-0- (CHa)m-CH 3 wherein each R 1 7 is independently H; -OR 2 1 -OCOR 21 -COR 21 00 -NCOR 21 -N(R 2 1 2 -CON(R 21 2 -C00OR 21 straight chained or branched C 1 alkyl, straight chained or branched Ca-C, Smonofluoroalkyl, straight chained or branched C 1 -C, 00 0 polyfluoroalkyl, straight chained or branched C 2 -C7 alkenyl, straight chained or branched C 2 alkynyl, C 5 -C 7 cycloalkenyl, -(CH 2 or (CH 2 (CH 2 )m-CH 3 wherein RiB is straight chained or branched C 1 -C 6 alkyl, (CH 2 or (CH 2 2 )m-CH 3 wherein each Rl 9 is independently H, or straight chained or branched C 1 -Cs alkyl; wherein each R 20 is independently straight chained or branched C 1 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 alkenyl or alkynyl; C 3 C, cycloalkyl or C 5 cycloalkenyl; -C1, -Br, or -I; -NO 2 -N 3 -CN; -OR 2 1 -OCOR 2 1 -COR 21 -NCOR 2 1 -N(R 2 1 2 CON(Ra 21 or -COOR 21 aryl or heteroaryl; or two R 20 groups present on adjacent carbon atoms can join together to form a methylenedioxy group; wherein each R 21 is independently straight chained or branched C 1 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 alkenyl or alkynyl; C 3 C, cycloalkyl, C 5 cycloalkenyl, aryl or aryl(Cl- C 6 alkyl; 732 wherein each R 22 is independently H, F, straight chained or branched alkyl; wherein each m is an integer from 0 to 4 inc wherein each n is an integer from 1 to 4 inc wherein p is an integer from 0 to 2 inclusi\ wherein q is an integer from 2 to 4 inclusi wherein t is 1 or 2; or a pharmaceutically acceptable salt thereof.
92. A compound having the structure: Cl or C 1 -C 4 :lusive; :lusive; re; wherein W is H, -Cl, -Br, CN, methyl, ethyl, propyl, methoxy or ethoxy; wherein X is N(CH 3 2 or 00 733 cR17 -N a R 17 00 M wherein R 13 is a bicyclic alkyl. ring system, aryl or aryl (C 1 -C 6 alkcyl; 00 wherein Y is NR 14 R 15 wherein R 14 is H, straight chained or branched Cj-C 6 alkyl, (CH 2 )q01-CH2)m-CH3D C 3 -C 6 cycloalkyl, or (C(Rig) 2 )m-Z; wherein R15 is (C(Ri 9 2 )m-N(Rir 6 2 wherein Z is C 3 -CI 0 cycloalkyl, aryl, or heteroaryl; wherein R 16 is straight chained or branched Cl-C 7 alkyl, straight chained or branched C 1 -C 7 monofluoroalkyl, straight chained or branched Cl-C 7 polyfluoroalkyl, straight chained or branched C 2 -C 7 alkenyl, straight chained or branched C 2 -C 7 alkynyl, CS-C 7 cycloalkenyl, (CH 2 or (CH2)q-O-(C1 2 )M-CH3; wherein each R 1 7, is independently H; -OR 21 -0C0RT 1 -C0R 21 -NCOR 21 -N(R 21 2 -CON(R 21 2 -C00R 21 straight chained or branched Cl-C 7 alkyl, straight chained or branched Cl-C 7 monofluoroalkyl, straight chained or branched Cl-C 7 polyfluoroalkyl, straight chained or branched C 2 -C 7 alkenyl, straight chained or branched C 2 -C 7 alkynyl, CS-C 7 cycloalkenyl, -(CH 2 )mZ, Or (CH 2 (CH 2 )m1,CH 3 r 734 wherein each R 19 is independently H, or straight chained or branched CI-C 6 alkyl; wherein each R 21 is independently straight chained or branched Ci-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or alkynyl; C 3 C7 cycloalkyl, Cs-C7 cycloalkenyl, aryl or aryl(C 1 Cs) alkyl; wherein each m is an integer from 0 to 4 inclusive; wherein each n is an integer from 1 to 4 inclusive; wherein q is an integer from 2 to 4 inclusive; or a pharmaceutically acceptable salt thereof.
93. An enantiomerically and diasteriomerically pure compound of claim 89, 90, 91, or 92.
94. An enantiomerically or diasteriomerically pure compound of claim 89, 90, 91, or 92. The compound of claim 89, wherein X is: R 17 R ~CR17 17 N R R17 or 735
96. The compound of claim 88, wherein X is NR 1 Ri2 and R 11 VB is H or straight chained or branched Ci-C 7 alkyl.
97. The compound of claim 96, wherein the compound has OO the structure: ,R 1 2 0N/R" y
98. The compound of claim 95, wherein R 13 alkyl ring system, cyclohexyl or aryl.
99. The compound of claim 97, wherein R 13 alkyl ring system, cyclohexyl or aryl. is a bicyclic is a bicyclic
100. The compound of claim 98, wherein R 14 is H, straight chained or branched Ci-C 6 alkyl or (CH 2 )q-O-(CH 2 )m-CH 3
101. The compound of claim 99, wherein R 14 is H, straight chained or branched Ci-C 6 alkyl or (CH 2 )q-0-(CH 2 )m-CH3. 00 00 736
102. The compound of claim 98, wherein Y is -N U R:7R 00 00
103. The compound of claim 102, wherein U is NR 1 6
104. The compound of claim 103, wherein R 16 is (CH2)m,-Z.
105. The compound of claim 104, wherein Z is aryl or heteroaryl. N U
106. The compound of claim 99, wherein Y is gR 17 0r
107. The compound of claim 106, wherein U is NR 16 737
108. The compound of claim 107, wherein the compound is selected from the group consisting of: \0 c I K N~ N ~C1 C1 Q(N)N N N "I N CI N Q11"NfDNN b N/ ~,NNN Q~r~JNN' N CI NN NO NIt N bCI (N N N ;and IN N' N i ONN 738
109. The compound of claim 107, wherein the compound is selected from the group consisting of: N C1 N N SCF N, NQ NN N,, N N(an N, Nan N -F 0739
110. The compound of claim 89, wherein X is N(CH 3 2
111. The compound of claim 110, wherein Y is 17 00 740
112. The compound of claim 111, wherein the compound is V) selected from a group consisting of: N N N N CN
113. A pharmaceutical composition comprising a therapeutically effective amount of the compound of claim 89, 90, 91, or 92 and a pharmaceutically acceptable carrier.
114. A pharmaceutical composition made by combining a therapeutically effective amount of the compound of claim 89, 90, 91, or 92 and a pharmaceutically acceptable carrier. 00 741 F1
115. A process for making a pharmaceutical composition Scomprising combining a therapeutically effective amount of the compound of claim 89, 90, 91, or 92 and a pharmaceutically acceptable carrier. 00
116. A method of treating a subject suffering from C depression which comprises administering to the 00 subject an amount of the compound of claim 89, 91, or 92 effective to treat the subject's depression.
117. A method of treating a subject suffering from anxiety which comprises administering to the subject an amount of the compound of claim 89, 90, 91, or 92 effective to treat the subject's anxiety.
118. A method of treating a subject suffering from depression and anxiety which comprises administering to the subject an amount of the compound of claim 89, 90, 91, or 92 effective to treat the subject's depression and anxiety. 00 742 00 00 Y3 Y4 wh119. A method of treating, Y, Y a subject sufferin g fromdependently H;depression which comprises administering to the subjectalkyl, an amount ofl uoroalkyl or polyfluoroalkyl; straight chainedt's or branched C 2 -C 7 alkenyl or alkynyl; C 3 -Cp cycloalky, or C5-C 7 cycloalkenyl; -C1, -Br, or I; -NO 2 -N 3 -CN; -OR 4 -SR 4 -OCOR 4 -COR 4 NCOR 4 N R4 2, -CON(R 4 2 or -COOR 4 aryl or heteroaryl; or any two of Y 1 Y 2 Y 3 and Y 4 present on adjacent cadepression atomswherein the compound has mthylenedioxy group; wherein each R 4 is independently straight chained 0 H; straight chained or branched C-Cluoroalkyl or monofluoroalky or polyfluoroalky; straight chained or branched or branched alkenyl or akynyl; C-C cycloalkyl, -C 7 cycloalkenyl, aryl or r cycloalkenyl; -Br, or I; -NO2; -N3; -CN; -OR4, -SR4, -OCOR4, -COR4, NCOR4, N(R)2 -CON(R4)2, or -COOR4; aryl or heteroaryl; or any two of Yi, Y2, Y3 and Y4 present on adjacent carbon atoms can constitute a methylenedioxy group; wherein each R4 is independently straight chained or branched Ci-C7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C2-C7 alkenyl or alkynyl; C3-C7 cycloalkyl, Cs-C7 cycloalkenyl, aryl or aryl(Ci-Cs)alkyl; wherein A is Q3, Q4, Qs, straight chained or 743 branched Cl-C 7 alkyl, aryl, heteroaryl, aryl (Cl- CO) alkyl, heteroaryl (Cl-C 6 alkyl, aryl substituted with an aryl or heteroaryl, heteroaryl substituted with an aryl or heteroaryl; or (CHR 1 7 ,)-(CHR 17 wherein A' is 0 In n R R 3 ;or H 2 n 1 wherein Q3 is N 1 17 744 wherein Q4 is wherein Qs is wherein Ri and R 2 are each independently H, straight chained or branched Ci-C 7 alkyl, -Cl, -Br, NO 2 or -CN; wherein R 3 is H, straight chained or branched C 1 -C, alkyl, -C1, -Br, -NO 2 -CN, -OR 6 aryl or heteroaryl; wherein Rs is straight chained or branched C 1 -C 7 alkyl, -N(R 4 2 -OR 6 or aryl; wherein R 6 is straight chained or branched Ci-C 7 alkyl or aryl; wherein each R 17 is independently H; straight chained 00 745 or branched CI-C 7 alkyl, straight chained or branched c Ci-C 7 monofluoroalkyl, straight chained or branched l Ci-C7 polyfluoroalkyl, straight chained or branched C 2 -C 7 alkenyl, straight chained or branched C 2 -C 7 alkynyl, Cs-C 7 cycloalkenyl, -(CH 2 or (CH 2 )n-O- 00 (CH 2 )m-CH 3 O wherein each R 20 is independently straight 0 0 chained or branched C 1 -C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or alkynyl; C 3 -C7 cycloalkyl or C 5 -C 7 cycloalkenyl; -C1, -Br, or -NO 2 -N 3 -CN; OR 21 -OCOR 21 -COR 21 -NCOR 21 -N(R 2 1 2 -CON(R2i)2, or -COOR 21 aryl or heteroaryl; or two R 20 groups present on adjacent carbon atoms can join together to form a methylenedioxy group; wherein each R 21 is independently straight chained or branched Ci-C7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C 7 alkenyl or alkynyl; C 3 -C7 cycloalkyl, Cs-C7 cycloalkenyl, aryl or aryl(C 1 -C 6 )alkyl; wherein each m is an integer from 0 to 4 inclusive; wherein each n is an integer from 1 to 4 inclusive; wherein each p is an integer from 0 to 2 inclusive; wherein U is O, -NR 16 S, C(R 17 2 or -NSO 2 Ri 6 wherein Z is C 3 -CIo cycloalkyl, C 4 -C7 cyclic ether, 00 746 SC 4 -C7 cyclic thioether, aryl, or heteroaryl; Ct kn wherein R 16 is straight chained or branched Ci-C 7 alkyl, straight chained or branched Ci-C 7 monofluoroalkyl, straight chained or branched CI-C 7 00 polyfluoroalkyl, straight chained or branched C 2 -C 7 Salkenyl, straight chained or branched C 2 -C 7 alkynyl, SCs-C cycloalkenyl, -(CH 2 or (CH 2 (CH2) -CH3; 00 wherein q is an integer from 2 to 4 inclusive; wherein B is aryl, heteroaryl, aryl substituted with an aryl or heteroaryl, heteroaryl substituted with an aryl or heteroaryl, tricyclic heteroaryl or Qs; provided however, if B is aryl or heteroaryl the carbon atom or carbon atoms ortho to the nitrogen atom of the imine bond may only be substituted with one or more of the following -Cl, -Br, -CN, methyl, ethyl or methoxy; wherein a tricyclic heteroaryl is a fused three member aromatic system in which one or more of the rings is heteroaryl; carbazole; or acridine; wherein Q6 is 0 R22 O R22 wherein each R 22 is independently H, F, Cl, or straight chained or branched Ci-C 4 alkyl; 00 747 os cor a pharmaceutically acceptable salt thereof.
120. A method of treating a subject suffering from depression which comprises administering to the 00 subject an amount of compound effective to treat the S subject's depression wherein the compound has the Sstructure: 00 SB Yz YY wherein each of Y 1 Y 2 Y 3 and Y 4 is independently H; straight chained or branched C 1 -C7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C 7 alkenyl or alkynyl; C 3 -C 7 cycloalkyl, or C 5 -C7 cycloalkenyl; -C1, -Br, or I; -NO 2 -N 3 -CN; -OR 4 -SR 4 -OCOR 4 -COR 4 -NCOR 4 N(R 4 2 -CON(R 4 2 or -COOR 4 aryl or heteroaryl; or any two of Yi, Y 2 Y 3 and Y 4 present on adjacent carbon atoms can constitute a methylenedioxy group; wherein each R 4 is independently straight chained or branched Ci-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C 7 alkenyl or alkynyl; C 3 -C7 cycloalkyl, Cs-C7 cycloalkenyl, aryl or aryl(Ci-Cs)alkyl; 748 wherein A is straight chained or branched CI-C7 alkyl, aryl, heteroaryl, aryl(C 1 alkyl or heteroaryl (Ci-C 6 alkyl; 0 Jn wherein A' is R 1 CR 2 R 3 or (CH2 n R 4 wherein Ri and R 2 are each independently H, straight chained or branched Ci-C7 alkyl, -Cl, -Br, NO 2 or -CN; wherein R 3 is H, straight chained or branched CI-C7 alkyl, -Cl, -Br, -NO 2 -CN, -OR 6 aryl 'or heteroaryl; wherein Rs is straight chained or branched Ci-C7 alkyl, -N(R 4 2 -OR 6 or aryl; wherein R 6 is straight chained or branched Ci-C7 00 749 alkyl or aryl; wherein B is aryl, or heteroaryl; provided however, if B is aryl or heteroaryl the carbon atom or carbon atoms ortho to the nitrogen atom of the imine bond O may only be substituted with one or more of the 00 M n following -Cl, -Br, -CN, methyl, ethyl or Smethoxy; 00 wherein n is an integer from 1 to 4 inclusive; or a pharmaceutically acceptable salt thereof.
121. A method of treating a subject suffering from depression which comprises administering to the subject an amount of compound effective to treat the subject's depression wherein the compound has the structure: B 1 N Y2 0 Y 31#N Y4 wherein each of Y 1 Y 2 Y 3 and Y 4 is independently H; straight chained or branched CI-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C 7 alkenyl or alkynyl; C 3 -C 7 cycloalkyl, or C 5 -C 7 cycloalkenyl; -C1, -Br, or 00 750 1; -NO 2 -N 3 -CM; -OR 4 -SR4, -OCOR,-O 4 -NO,- N(R42 ,-CON(Ri)2, or -COOR4; aryl or heteroaryl; or any two of Y 1 Y 2 Y 3 and Y 4 present on adjacent carbon atoms can constitute a methylenedioxy group; 00 wherein each R 4 is independently straight chained or branched Cl-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C 7 00alkenyl or alkynyl; C 3 -C 7 cycloalkyl, C 5 -C 7 cycloalkenyl, aryl or aryl(Cl-C 6 )alkyl; wherein A is straight chained or branched Cl-C 7 alkyl, aryl, heteroaryl, aryl (C CO)alkyl or heteroaryl (Cl-C 6 alkyl; wherein A' is 0 0 R 5 n I CR2R3or (CH 2 R wherein B is aryl substituted with an aryl or heteroaryl, heteroaryl substituted with an aryl or 00 751 C heteroaryl, tricyclic heteroaryl or Q6; Swherein a tricyclic heteroaryl is a fused three ring aromatic system in which one or more of the rings is heteroaryl; carbazole; or acridine; 00 Swherein Q6 is 0 R 22 00 O R22 wherein n is an integer from 1 to 4 inclusive; wherein each R 22 is independently H, F, Cl, or straight chained or branched Ci-C 4 alkyl; or a pharmaceutically acceptable salt thereof.
122. A method of treating a subject suffering from depression which comprises administering to the subject an amount of compound effective to treat the subject's depression wherein the compound has the structure: B Y N Y3 2 00 752 wherein each of Y 1 Y 2 Y 3 and Y 4 is independently InH; straight chained or branched Cl-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C 7 alkenyl or alkynyl; C 3 -C 7 00 cycloalkyl, or CS-C 7 cycloalkenyl; -C1, -Br, or 1; -NO 2 -N 3 -CN; -OR 4 -SR 4 -OCOR 4 -COR 4 -NCOR 4 N(R42 ,-CON(R 4 2 or -COOR 4 aryl or heteroaryl; or 00 any two of Y 1 Y 2 Y 3 and Y 4 present on adjacent carbon atoms can constitute a methylenedioxy group; wherein each R 4 is independently straight chained or branched Cl-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C 7 alkenyl or alkynyl; C 3 -C 7 cycloalkyl, C 5 -C 7 cycloalkenyl, aryl or aryl(CI-C 6 )alkyl; wherein A is Q3, Q41 Q5, aryl substituted with an aryl or heteroaryl, heteroaryl substituted with an aryl or heteroaryl, or (CHR 17 -(CHR 1 7 wherein Q3 is R 17 R1 N -1 R17 R17 '-n wherein Q4 is 00 753 CIR 17 R 17 n N n C-I R 20 R 1 7 M 00 C-I wherein 05 is 00R1 IU 51 wherein each R 1 7, is independently H; straight chained or branched Cl-C 7 alkyl, straight chained or branched CI-C. 7 monofluoroalkyl, straight chained or branched Cl-C 7 polyfluoroalkyl, straight chained or branched C 2 -C 7 alkenyl, straight chained or branched C 2 -C 7 alkynyl, C 5 -C 7 cycloalkenyl, -(CH 2 or (CH 2 (CH 2 m -CH 3 wherein each R 20 is independently straight chained or branched Cl-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C 7 alkenyl or alkynyl; .C 3 cycloalkyl or C 5 -C 7 cycloalkenyl; -C1, -Br, or -NO 2 -N3; -CM; OR 21 -0C0R 21 -C0R 21 -NCOR 21 -N(R 21 2 -CON(R 2 2 or -C00R 21 aryl or heteroaryl; or two R 20 groups present on adjacent carbon atoms can join together to form a 00 754 methylenedioxy group; wherein each R 21 is independently straight ^C chained or branched Ci-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C 7 00 alkenyl or alkynyl; C 3 -C 7 cycloalkyl, Cs-C7 Scycloalkenyl or aryl; 00 wherein each R 22 is independently H, F, Cl, or straight chained or branched CI-C 4 alkyl; wherein q is an integer from 2 to 4 inclusive; wherein each m is an integer from 0 to 4 inclusive; wherein each n is an integer from 1 to 4 inclusive; wherein each p is an integer from 0 to 2 inclusive; wherein U is O, -NR 16 S, C(R17) 2 or -NSO 2 Ris; wherein Z is C 3 -Clo cycloalkyl, C 4 -C 7 cyclic ether, C 4 -C7 cyclic thioether, aryl, or heteroaryl; wherein R 16 is straight chained or branched Ci-C7 alkyl, straight chained or branched Ci-C7 monofluoroalkyl, straight chained or branched Ci-C7 polyfluoroalkyl, straight chained or branched C 2 -C7 alkenyl, straight chained or branched C 2 -C7 alkynyl, Cs-C7 cycloalkenyl, -(CH 2 or (CH 2 (CH 2 )m-CH 3 wherein B is aryl, or heteroaryl; provided however, 00 755 C if B is aryl or heteroaryl the carbon atom or carbon c atoms ortho to the nitrogen atom of the imine bond Smay only be substituted with one or more of the C following -Cl, -Br, -CN, methyl, ethyl or methoxy; 00 C or a pharmaceutically acceptable salt thereof. 00 10 123. The method of claim 119, 120, 121, or 122, wherein the compound is enantiomerically and diastereomerically pure.
124. The method of claim 119, 120, 121, or 122, wherein the compound is enantiomerically or diastereomerically pure.
125. The method of claim 119, 120, 121, or 122, wherein the compound is a pure Z imine isomer or a pure Z alkene isomer.
126. The method of claim 119, 120, 121, or 122, wherein the compound is a pure E imine isomer or a pure E alkene isomer.
127. The method of claim 119, 120, 121, or 122, wherein the compound is administered orally. 756
128. The method of claim 119 or 120, wherein the compound has the structure: wherein each of Yi, Y 2 Y 3 and Y 4 is independently H; straight chained or branched C 1 -C 7 alkyl, -CF 3 F, -Cl, -Br, -OR 4 -N(R 4 2 or -CON(R 4 2 wherein each R 4 is independently straight chained or branched C 1 -C 7 alkyl, -CF 3 or phenyl; wherein A is straight chained or branched CI-C7 alkyl, aryl, heteroaryl, aryl (Ci-C) alkyl or heteroaryl (Ci-C 6 alkyl; and wherein A' is CR 2 R 3
129. The method of claim 119, 120 or 122, wherein B is heteroaryl. 757
130. The method of claim 119 or 120, wherein B is aryl.
131. The method of claim 130, wherein B is phenyl and the phenyl is optionally substituted with one or more of the following: -Cl, -Br, -CF 3 straight chained or branched Ci-C 7 alkyl, -N(R 4 2 -OR 4 -COR4, -NCOR4, -C0 2 R 4 or -CON(R4) 2
132. The method of claim 131, wherein A is aryl.
133. The method of claim 131, wherein A is heteroaryl.
134. The method of claim 133, wherein the compound is selected from the group consisting of: F OC~=oF 0 ts~o C1 CI W.\ and Ks-N S,,C 758
135. The method of claim 132, wherein the compound is selected from the group consisting of: N 0-e \N/50 F N 0 2008200380 25 Jan 2008 -3' F- 760
136. The method of claim 130, wherein A is A' and A' is R, RR 3
137. The method of claim 136, wherein the compound is: NCl 0 Cl or 00 761
138. The method of claim 121, wherein B is Q6.
139. The method of claim 138, wherein A is aryl. 00 0 r n
140. The method of claim 139, wherein the compound has the structure: 00 SR22 N R22 O
141. The method of claim 140, wherein the compound is: 0 F
142. The method of claim 122, wherein B is aryl. 762
143. The method of claim 142, wherein A is (CHR 17 (CHR17) n-Z.
144. The method of claim 143, wherein the compound is: 0 00 00 o0 0D Cl NCI O N
145. The method of claim 119, wherein the compound has the structure: wherein each R 24 is independently one or more of the following: H, F, Cl, Br, I, CF 3 OCH 3 or NOz; and S763 wherein R2s is methyl, ethyl, allyl, phenyl and the n phenyl is optionally substituted with a F, Cl, Br, CF 3 NO 2 S 0 0
146. A method of treating a subject suffering from O anxiety which comprises administering to the subject an C< amount of compound effective to treat the subject's 00 Sanxiety wherein the compound has the structure: B YY Y4 wherein each of Y 1 Y 2 Y 3 and Y 4 is independently H; straight chained or branched C 1 -C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C 7 alkenyl or alkynyl; C 3 -C7 a cycloalkyl, or C 5 -C 7 cycloalkenyl; -Cl, -Br, or I; -NO 2 -N 3 -CN; -OR 4 -SR4, -OCOR4, -COR 4 -NCOR4, N(R 4 2 -CON(R 4 2 or -COOR4; aryl or heteroaryl; or any two of YI, Y 2 Y 3 and Y 4 present on adjacent carbon atoms can constitute a methylenedioxy group; wherein each R 4 is independently straight chained or branched Ci-C 7 alkyl, monofluoroalkyl or 0076 polyfluoroalkyl; straight chained or branched C 2 -C 7 a2.kenyl or alkynyl; C 3 -C 7 cycloalkyl, C 5 -C 7 cycloalkenyl, aryl or aryl (Cl-C 6 a2kyl; wherein A is A' Q3, Q4, Q5, straight chained or branched C 1 -C 7 alkyl, aryl, heteroaryl, aryl (Cl- CO) alkyl, heteroaryl (Cl-C 6 alkyl, aryl substituted with an aryl or heteroaryl, heteroaryl substituted with an aryl or heteroaryl; or (CHR 1 7) -(CHR 17 Z; wherein A' is 0 CR 2 R ;or (CH2) It4 wherein Q3 is 765 0 00 00 wherein Q4 is wherein Q5 is wherein Ri and R 2 are each independently H, straight chained or branched C 1 -C 7 alkyl, -Cl, -Br, NO 2 or -CN; wherein R 3 is H, straight chained or branched Ci-C 7 alkyl, -Cl, -Br, -NO 2 -CN, -OR 6 aryl or heteroaryl; wherein R 5 is straight chained or branched Ci-C alkyl, -N(R4)2, -ORs or aryl; 766 h- wherein R 6 is straight chained or branched C 1 -C 7 l alkyl or aryl; wherein each R 17 is independently H; straight chained 00 or branched C 1 -C 7 alkyl, straight chained or branched SCi-C 7 monofluoroalkyl, straight chained or branched 00 CN Ci-C7 polyfluoroalkyl, straight chained or branched SC 2 -C 7 alkenyl, straight chained or branched C 2 -C 7 alkynyl, C 5 -C 7 cycloalkenyl, -(CH 2 or (CH 2 )n-O- (CH 2 -CH 3 wherein each R 20 is independently straight chained or branched CI-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C 7 alkenyl or alkynyl; C 3 -07 cycloalkyl or C 5 -C 7 cycloalkenyl; -C1, -Br, or -NO 2 -N 3 -CN; OR 21 -OCOR 21 -COR 21 -NCOR 21 -N(R 21 2 -CON(R 21 2 or -COOR 21 aryl or heteroaryl; or two R 2 0 groups present on adjacent carbon atoms can join together to form a methylenedioxy group; wherein each R 2 1 is independently straight chained or branched CI-C7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or alkynyl; C 3 -C7 cycloalkyl, C5-C 7 cycloalkenyl, aryl or aryl(Ci-Cs)alkyl; wherein each m is an integer from 0 to 4 inclusive; wherein each n is an integer from 1 to 4 inclusive; 00 767 Swherein each p is an integer from 0 to 2 inclusive; Ct n wherein U is 0, -NR 16 S, C(RI 7 2 or -NSOzR 16 wherein Z is C 3 -C 1 cycloalkyl, C 4 -C 7 cyclic ether, 0 0 C 4 -C 7 cyclic thioether, aryl, or heteroaryl; CI wherein R 16 is straight chained or branched C 1 -C 7 00 alkyl, straight chained or branched C 1 -C 7 CI 10 monofluoroalkyl, straight chained or branched Ci-C7 polyfluoroalkyl, straight chained or branched C 2 -C 7 alkenyl, straight chained or branched C 2 -C 7 alkynyl, Cs-C 7 cycloalkenyl, -(CH 2 m-Z, or (CH 2 q-O- (CH 2 m-CH3; wherein q is an integer from 2 to 4 inclusive; wherein B is aryl, heteroaryl, aryl substituted with an aryl or heteroaryl, heteroaryl substituted with an aryl or heteroaryl, tricyclic heteroaryl or Q6; provided however, if B is aryl or heteroaryl the carbon atom or carbon atoms ortho to the nitrogen atom of the imine bond may only be substituted with one or more of the following -Cl, -Br, -CN, methyl, ethyl or methoxy; wherein a tricyclic heteroaryl is a fused three member aromatic system in which one or more of the rings is heteroaryl; carbazole; or acridine; wherein Q6 is S768 0 R 2 2 n 0 R22 wherein each R 22 is independently H, F, M Cl, or straight chained or branched C 1 -C 4 alkyl; 00 or a pharmaceutically acceptable salt thereof.
147. A method of treating a subject suffering from anxiety which comprises administering to the subject an amount of compound effective to treat the subject's anxiety wherein the compound has the structure: B Y1 N Y wherein each of Y 1 Y 2 Y 3 and Y 4 is independently H; straight chained or branched CI-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C alkenyl or alkynyl; C 3 -C, cycloalkyl, or C 5 -C 7 cycloalkenyl; -C1, -Br, or I; -NO 2 -N 3 -CN; -OR 4 -SR 4 -OCOR4, -COR4, -NCOR4, N(R4)2 -CON(R 4 2 or -COOR4; aryl or heteroaryl; or any two of YI, Y 2 Y 3 and Y 4 present on adjacent 769 carbon atoms can constitute a methylenedioxy group; wherein each R 4 is independently straight chained or branched Cl-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C 7 alkenyl or alkynyl; C 3 -C7 cycloalkyl, CS-C7 cycloalkenyl, aryl or aryl (C 1 -C 6 alky.; wherein A is straight chained or branched alkyl, aryl, heteroaryl, aryl (C C 6 alkyl heteroaryl (Cl-C 6 alkyl; Cl -C 7 or wherein A' is 0 n CR 2 R 3 or -(CH 2 n R wherein R, and R 2 are each independently H, straight chained or branched Cl-C 7 alkyl, -Cl, -Br, NO 2 or -CM; wherein R 3 is H, straight chained or branched Cl-C 7 alkyl, -Cl, -Br, -NO 2 -CN, -OR 6 aryl or 00 770 Sheteroaryl; wherein Rs is straight chained or branched Ci-C 7 alkyl, -N(R 4 2 -OR 6 or aryl; 00 wherein R 6 is straight chained or branched Ci-C7 0 alkyl or aryl; 00 Swherein B is aryl, or heteroaryl; provided however, C- 10 if B is aryl or heteroaryl the carbon atom or carbon atoms ortho to the nitrogen atom of the imine bond may only be substituted with one or more of the following -Cl, -Br, -CN, methyl, ethyl or methoxy; wherein n is an integer from 1 to 4 inclusive; or a pharmaceutically acceptable salt thereof.
148. A method of treating a subject suffering from anxiety which comprises administering to the subject an amount of compound effective to treat the subject's anxiety wherein the compound has the structure: B Y.1 Y2i 00 771 wherein each of Y1, Y 2 Y 3 and Y 4 is independently H; straight chained or branched Cl-C7 alkyl, monofluoroalkyl. or polyfluoroalkyl; straight chained or branched C 2 C alkenyl or alkynyl; C 3 -C 7 00 cycloalkyl, or CS-C 7 cycloalkenyl; -C1, -Br, or 1; -NO 2 -N 3 -CN; -OR 4 -SR 4 -OCOR 4 -COR 4 -NCOR 4 N(R 4 2 -CON(R 4 2 or -COOR 4 aryl or heteroaryl; or 00 any two of Y 1 Y 2 Y 3 and Y 4 present on adjacent carbon atoms can constitute a methylenedioxy group; wherein each R 4 is independently straight chained or branched Cl-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C 7 alkenyl or alkynyl; C 3 -C 7 cycloalkyl, C 5 -C 7 cycloalkenyl, aryl or aryl(Cl-C 6 )alkyl; wherein A is straight chained or branched C 1 -C 7 alkyl, aryl, heteroaryl, aryl C 6 alkyl or heteroaryl (Cl-C 6 alkyl; wherein A' is 0 0 -t R 5 nn 772 or (CH2) n R4 R n CR2R3 wherein B is aryl substituted with an aryl or heteroaryl, heteroaryl substituted with an aryl or heteroaryl, tricyclic heteroaryl or Qs; wherein a tricyclic heteroaryl is a fused three ring aromatic system in which one or more of the rings is heteroaryl; carbazole; or acridine; wherein Qs is wherein n is an integer from 1 to 4 inclusive; wherein each R 22 is independently H, Cl, or straight chained or branched C 1 -C 4 alkyl; or a pharmaceutically acceptable salt thereof.
149. A method of treating a subject suffering from anxiety which comprises administering to the subject an amount of compound effective to treat the 00 773 Ssubject's anxiety wherein the compound has the -s structure: B S N 00 M¢3 Y2 00 Y4 wherein each of Y 1 Y 2 Y 3 and Y 4 is independently H; straight chained or branched C 1 -C7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C 7 alkenyl or alkynyl; C 3 -C 7 cycloalkyl, or Cs-C 7 cycloalkenyl; -C1, -Br, or I; -NO 2 -N 3 -CN; -OR 4 -SR4, -OCOR4, -COR4, -NCOR4, N(R 4 2 -CON(R 4 2 or -COOR4; aryl or heteroaryl; or any two of Y 1 Y 2 Y 3 and Y 4 present on adjacent carbon atoms can constitute a methylenedioxy group; wherein each R 4 is independently straight chained or branched Ci-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or alkynyl; C 3 -C 7 cycloalkyl, Cs-C 7 cycloalkenyl, aryl or aryl(Ci-C6)alkyl; wherein A is Q0, Q4, Qs, aryl substituted with an aryl or heteroaryl, heteroaryl substituted with an aryl or heteroaryl, or (CHR 17 )-(CHR17)n-Z; wherein Q3 is 0 00 o 0 oq wherein Q4 is wherein Qs is wherein each R 1 1 is independently H; straight chained or branched C 1 -C 7 alkyl, straight chained or branched C 1 -C 7 monofluoroalkyl, straight chained or branched Ci-C 7 polyfluoroalkyl, straight chained or branched C 2 -C7 alkenyl, straight chained or branched Ca-C 7 alkynyl, Cs-C 7 cycloalkenyl, -(CH2)m-Z, or (CH 2 )n-0- (CH2),-CH 3 wherein each R 20 is independently straight 00 S775 Schained or branched C 1 -C7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C 7 alkenyl or alkynyl; C 3 -C7 cycloalkyl or C 5 -C 7 cycloalkenyl; -Cl, -Br, or -NO 2 -N 3 -CN; OR 21 -OCOR 21 -COR 21 -NCOR 21 -N(R 21 2 -CON(R 21 2 or 0 0 -COOR 21 aryl or heteroaryl; or two R 20 groups present Son adjacent carbon atoms can join together to form a CI methylenedioxy group; 00 C- 10 wherein each R 2 1 is independently straight chained or branched Ci-C7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or alkynyl; C 3 -C7 cycloalkyl, Cs-C7 cycloalkenyl or aryl; wherein each R 22 is independently H, F, Cl, or straight chained or branched Ci-C 4 alkyl; wherein q is an integer from 2 to 4 inclusive; wherein each m is an integer from 0 to 4 inclusive; wherein each n is an integer from 1 to 4 inclusive; wherein each p is an integer from 0 to 2 inclusive; wherein U is O, -NR16, S, C(R 17 2 or -NSO 2 Ris; wherein Z is C 3 -C 0 o cycloalkyl, C 4 -C7 cyclic ether, C 4 -C7 cyclic thioether, aryl, or heteroaryl; wherein Rig is straight chained or branched CI-C7 00 776 alkyl, straight chained or branched C 1 -C 7 h- monofluoroalkyl, straight chained or branched C 1 -C 7 l polyfluoroalkyl, straight chained or branched C 2 -C7 alkenyl, straight chained or branched C2-C 7 alkynyl, Cs-C7 cycloalkenyl, -(CH 2 or (CH 2 (CH 2 )-CH 3 00 wherein B is aryl, or heteroaryl; provided however, 00 CA if B is aryl or heteroaryl the carbon atom or carbon Satoms ortho to the nitrogen atom of the imine bond may only be substituted with one or more of the following -Cl, -Br, -CN, methyl, ethyl or methoxy; or a pharmaceutically acceptable salt thereof.
150. The method of claim 146, 147, 148, or 149, wherein the compound is enantiomerically and diastereomerically pure.
151. The method of claim 146, 147, 148, or 149, wherein the compound is enantiomerically or diastereomerically pure compound.
152. The method of claim 146, 147, 148, or 149, wherein the compound is a pure Z imine isomer or a pure Z alkene isomer.
153. The method of claim 146, 147, 148, or 149, wherein the compound is a pure E imine isomer or a pure E alkene isomer. 777
154. The method of claim 146 or 147, wherein the compound has the structure: wherein each of Y 1 Y 2 Y 3 and Y 4 is independently H; straight chained or branched Cl-C 7 alkyl, -CF 3 F, -C1, -Br, -OR 4 -N(R4)2, or -CON(R4) 2 wherein each R 4 is independently straight chained or branched Cl-C 7 alkyl, -CF 3 or phenyl; wherein A is A' straight chained or branched C 1 -C 7 alkyl, aryl, heteroaryl, aryl (Cl Cdalkyl or heteroaryl (Cl-C 6 alkyl; and wherein A' is n CR 2 R 3 00 S778
155. The method of claim 146 or 147, wherein B is heteroaryl.
156. The method of claim 146 or 147, wherein B is aryl. 0 00 0
157. The method of claim 156, wherein B is phenyl and the phenyl is optionally substituted with one or more of the following: -Cl, -Br, -CF 3 straight chained or branched CI-C 7 alkyl, -N(R 4 2 -OR 4 -COR 4 -NCOR 4 -C0 2 R 4 or -CON(R 4 2
158. The method of claim 157, wherein A is aryl.
159. The method of claim 157, wherein A is heteroaryl.
160. The method of claim 159, wherein the compound is selected from the group consisting of: F NQ 6 N'CI NCl r/ S and rN hOjI SL 779
161. The method of claim 158, wherein the compound is selected from the group consisting of: F F 2008200380 25 Jan 2008 0 00 781 (N 0 Cl 00 00 O 162. The method of claim 156, wherein A is and A' is n -CR 2 R 3
163. The method of claim 162, wherein the compound is: S; or Cl 782
164. The method of claim 148, wherein B is Q6.
165. The method of claim 164, wherein A is aryl.
166. The method of claim 165, wherein the compound has the structure:
167. The method of claim 166, wherein the compound is:
168. The method of claim 149, wherein B is aryl. 783
169. The method of claim 168, wherein A is (CHR 17 (CHR17)n-Z.
170. The method of claim 169, wherein the compound is: Cl N' l -N
171. The method of claim 146, wherein the compound has the structure: wherein each R24 is independently one or more of the following: H, F, Cl, Br, I, CF 3 OCH 3 or NO 2 and 784 wherein R 25 is methyl, ethyl, allyl, phenyl and the phenyl is optionally substituted with a F, Cl, Br, CF 3 NO 2
172. A pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound having the structure: wherein each of Y 1 Y 2 Y 3 and Y 4 is independently H; straight chained or branched Ci-C7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C 7 alkenyl or alkynyl; C 3 -C 7 cycloalkyl, or Cs-C 7 cycloalkenyl; -C1, -Br, or I; -NOz; -N 3 -CN; -OR 4 -SR4, -OCOR4, -COR4, -NCOR4', N(R 4 2 -CON(R 4 2 or -COOR4; aryl or heteroaryl; or any two of Y 1 Y 2 Y 3 and Y 4 present on adjacent carbon atoms can constitute a methylenedioxy group; wherein each R 4 is independently straight chained or branched Ci-C 7 alkyl, monofluoroalkyl or 00 785 polyf luoroalkyl; straight chained or branched C 2 -C 7 alkenyl or alkynyl; C 3 cycloalkyl, C 5 -C 7 cycloalkenyl, aryl or aryl(Cl-C 6 )alkyl; wherein A is Q3, Q4, Q5, straight chained or 00 branched Cl-C 7 alkyl, aryl, heteroaryl, aryl (Cl- CO) alkyl, heteroary). (Cl-C 6 alkyl, aryl substituted with an aryl or heteroaryl, heteroaryl substituted 00 with an aryl or heteroaryl; or (CHR 17 -(CHR 17 -Z; wherein A' is 0 0 or -(CH 2 n CR 2 R 3 4 wherein Q3 is RW R 17 N R 17 R 17 U R 17 786 wherein Q4 is wherein Qs is wherein RI and R 2 are each independently H, straight chained or branched C 1 -C7 alkyl, -Cl, -Br, NO 2 or -CN; wherein R 3 is H, straight chained or branched Ci-C 7 alkyl, -Cl, -Br, -NO 2 -CN, -OR 6 aryl or heteroaryl; 00 S787 Swherein Rs is straight chained or branched CI-C 7 alkyl, -N(R 4 2 -OR 6 or aryl; wherein R 6 is straight chained or branched C 1 -C 7 alkyl or aryl; 00 Swherein each R 17 is independently H; straight chained CA or branched C 1 -C 7 alkyl, straight chained or branched 00 0 Ci-C 7 monofluoroalkyl, straight chained or branched Cq 10 C 1 -C 7 polyfluoroalkyl, straight chained or branched C 2 -C 7 alkenyl, straight chained or branched C 2 -C 7 alkynyl, C 5 -C7 cycloalkenyl, -(CH 2 or (CH 2 )n-O- (CH 2 m-CH 3 wherein each R 20 is independently straight chained or branched Ci-C7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C 7 alkenyl or alkynyl; C 3 -C 7 cycloalkyl or C s -C 7 cycloalkenyl; -Cl, -Br, or -NO 2 -N 3 -CN; OR 21 -OCOR 21 -COR 21 -NCOR 21 -N(R 21 2 -CON(R 21 2 or -COOR 21 aryl or heteroaryl; or two R 20 groups present on adjacent carbon atoms can join together to form a methylenedioxy group; wherein each R 21 is independently straight chained or branched C 1 -C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C 7 alkenyl or alkynyl; C 3 -C 7 cycloalkyl, Cs-C 7 cycloalkenyl, aryl or aryl(Ci-C 6 )alkyl; wherein each m is an integer from 0 to 4 inclusive; 00 S788 wherein each n is an integer from 1 to 4 inclusive; Ct V) wherein each p is an integer from 0 to 2 inclusive; wherein U is O, -NR 16 S, C(R 17 2 or -NSO 2 R 16 00 0wherein Z is C 3 -C 1 0 cycloalkyl, C 4 -C7 cyclic ether, Cq C 4 -C 7 cyclic thioether, aryl, or heteroaryl; 00 wherein R 16 is straight chained or branched C 1 -C 7 alkyl, straight chained or branched Ci-C7 monofluoroalkyl, straight chained or branched C 1 -C 7 polyfluoroalkyl, straight chained or branched C 2 -C7 alkenyl, straight chained or branched C 2 -C 7 alkynyl, Cs-C 7 cycloalkenyl, -(CH 2 or (CH 2 )q-O-(CH 2 )m-CH 3 wherein q is an integer from 2 to 4 inclusive; wherein B is aryl, heteroaryl, aryl substituted with an aryl or heteroaryl, heteroaryl substituted with an aryl or heteroaryl, tricyclic heteroaryl or Q6; provided however, if B is aryl or heteroaryl the carbon atom or carbon atoms ortho to the nitrogen atom of the imine bond may only be substituted with one or more of the following -Cl, -Br, -CN, methyl, ethyl or methoxy; wherein a tricyclic heteroaryl is a fused three member aromatic system in which one or more of the rings is heteroaryl; carbazole; or acridine; wherein Q6 is 00 789 1 O R 22 0 R22 00 wherein each R 22 is independently H, F, Cl, or straight chained or branched C 1 -C 4 alkyl; 00 or a pharmaceutically acceptable salt thereof.
173. A pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound having the structure: B Y/ rN Y4 wherein each of Y 1 Y 2 Y 3 and Y 4 is independently H; straight chained or branched Ci-C7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C 7 alkenyl or alkynyl; C 3 -C 7 cycloalkyl, or Cs-C7 cycloalkenyl; -C1, -Br, or I; -NO 2 -N 3 -CN; -OR 4 -SR4, -OCOR 4 -COR 4 -NCOR 4 N(R 4 2 -CON(R 4 2 or -COOR4; aryl or heteroaryl; or any two of Yi, Y 2 Y 3 and Y 4 present on adjacent carbon atoms can constitute a methylenedioxy group; 790 wherein each R 4 is independently straight chained or branched Cl-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 alkenyl or alkynyl; C 3 -C 7 cycloalkyl, C 5 -c 7 cycloalkenyl, aryl or aryl(Cl-Cs)alkyl; wherein A is straight chained or branched alkyl, aryl, heteroaryl, aryl (C C 6 alkyl heteroaryl (Cl-C 6 )alkyl; cI-C 7 or wherein A' is In CR 2 R or -(CH 2 n R wherein R3. and R 2 are each independently H, straight chained or branched Cl-C 7 alkyl, -Cl, -Br, NO 2 or -CN; wherein R 3 is H, straight chained or branched C 1 -C 7 alkyl, -Cl, -Br, -NO 2 -CN, -OR 6 aryl or 00 791 Sheteroaryl; V wherein Rs is straight chained or branched C 1 -C 7 alkyl, -N(R 4 2 -OR 6 or aryl; 00 wherein R 6 is straight chained or branched C 1 -C 7 0 alkyl or aryl; 00 Swherein B is aryl, or heteroaryl; provided however, C- 10 if B is aryl or heteroaryl the carbon atom or carbon atoms ortho to the nitrogen atom of the imine bond may only be substituted with one or more of the following -Cl, -Br, -CN, methyl, ethyl or methoxy; wherein n is an integer from 1 to 4 inclusive; or a pharmaceutically acceptable salt thereof.
174. A pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound having the structure: 00 792 wherein each of Y 1 Y 2 Y 3 and Yq is independently H; straight chained or branched CI-C7 alkyl, V) monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or alkynyl; C 3 -C 7 cycloalkyl, or C 5 -C 7 cycloalkenyl; -Cl, -Br, or 00 1; -NO 2 -N 3 -CN; -OR 4 -SR4, -OCOR4, -COR4, -NCOR 4 N(R 4 2 -CON(R4) 2 or -COOR4; aryl or heteroaryl; or ci any two of Y 1 Y 2 Y 3 and Y 4 present on adjacent 00 carbon atoms can constitute a methylenedioxy group; wherein each R 4 is independently straight chained or branched C. 1 C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C,7 alkenyl or alkynyl; C 3 -C 7 cycloalkyl, C5-C 7 cycloalkenyl, aryl or aryl(Cl-C 6 )alkyl; wherein A is straight chained or branched CI-C7 alkyl, aryl, heteroaryl, aryl (C1-C 6 alkyl or heteroaryl. (Cl-C 6 alkyl; wherein A' is 0 0 n I. n CR 2 R 3 or -(CH 2 ).R wherein B is aryl substituted with an aryl or heteroaryl, heteroaryl substituted with an aryl or heteroaryl, tricyclic heteroaryl or Qs; wherein a tricyclic heteroaryl is a fused three ring aromatic system in which one or more of the rings is heteroaryl; carbazole; or acridine; wherein Q6 is wherein n is an integer from 1 to 4 inclusive; wherein each R 22 is independently H, Cl, or straight chained or branched C 1 -C 4 alkyl; or a pharmaceutically acceptable salt thereof. 00 o 794
175. A pharmaceutical composition comprising a V' pharmaceutically acceptable carrier and a compound (N having the structure: 0 00 B Y4 00 wherein each of Y 1 Y 2 Y 3 and Y 4 is independently H; straight chained or branched C 1 -C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C 7 alkenyl or alkynyl; C 3 -C 7 cycloalkyl, or Cs-C 7 cycloalkenyl; -C1, -Br, or I; -NO 2 -N 3 -CN; -OR 4 -SR4, -OCOR4, -COR 4 -NCOR4, N(R 4 2 -CON(R 4 2 or -COOR4; aryl or heteroaryl; or any two of Y 1 Y 2 Y 3 and Y 4 present on adjacent carbon atoms can constitute a methylenedioxy group; wherein each R4 is independently straight chained or branched Ci-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C, alkenyl or alkynyl; C 3 -C7 cycloalkyl, Cs-C 7 cycloalkenyl, aryl or aryl(CI-C 6 )alkyl; wherein A is Q3, Q4, Qs, aryl substituted with an aryl or heteroaryl, heteroaryl substituted with an 795 aryl or heteroaryl, or (CHR 17 -(CHR 1 7 wherein Q3 is R17 R 17 N171 17R 17 n wherein Q 4 is wherein Q5 is wherein each R 17 is independently H; straight chained 0 (N 0796 Sor branched CI-C7 alkyl, straight chained or branched SCi-C7 monofluoroalkyl, straight chained or branched SCi-C7 polyfluoroalkyl, straight chained or branched c-I C 2 -C7 alkenyl, straight chained or branched C 2 -C7 alkynyl, Cs-C7 cycloalkenyl, -(CH 2 or (CH 2 )n-O- 00 (CH 2 )m-CH 3 00 CA wherein each R2o is independently straight Schained or branched CI-C7 alkyl, monofluoroalkyl or 10 polyfluoroalkyl; straight chained or branched C 2 -C, alkenyl or alkynyl; C 3 -C 7 cycloalkyl or Cs-C7 cycloalkenyl; -C1, -Br, or -NO 2 -N 3 -CN; OR 21 -OCOR 21 -COR 21 -NCOR 21 -N(R 21 2 -CON(R 21 2 or -COOR 21 aryl or heteroaryl; or two R 20 groups present on adjacent carbon atoms can join together to form a methylenedioxy group; wherein each Rz 2 is independently straight chained or branched C 1 -C7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or alkynyl; C 3 -C7 cycloalkyl, Cs-C7 cycloalkenyl or aryl; wherein each R 22 is independently H, F, Cl, or straight chained or branched C 1 -C 4 alkyl; wherein q is an integer from 2 to 4 inclusive; wherein each m is an integer from 0 to 4 inclusive; wherein each n is an integer from 1 to 4 inclusive; 797 Swherein each p is an integer from 0 to 2 inclusive; wherein U is O, -NR 16 S, C(R 17 2 or -NSOzR 1 6 wherein Z is C 3 -C 1 o cycloalkyl, C 4 -C 7 cyclic ether, 0 0 C 4 -C 7 cyclic thioether, aryl, or heteroaryl; CI wherein R 16 is straight chained or branched Ci-C, 00 alkyl, straight chained or branched C 1 -C 7 C- 10 monofluoroalkyl, straight chained or branched Ci-C 7 polyfluoroalkyl, straight .chained or branched C 2 -C 7 alkenyl, straight chained or branched C 2 -07 alkynyl, Cs-C7 cycloalkenyl, -(CH 2 or (CH 2 )q-O-(CH 2 )m-CH 3 wherein B is aryl, or heteroaryl; provided however, if B is aryl or heteroaryl the carbon atom or carbon atoms ortho to the nitrogen atom of the imine bond may only be substituted with one or more of the following -Cl, -Br, -CN, methyl, ethyl or methoxy; or a pharmaceutically acceptable salt thereof.
176. The pharmaceutical composition of claim 172, 173, 174, or 175, wherein the compound is an enantiomerically and diastereomerically pure compound.
177. The pharmaceutical composition of claim 172, 173, 174, or 175, wherein the compound is an enantiomerically or diastereomerically pure compound. 798
178. The pharmaceutical composition of claim 172, 173, 174, or 175, wherein the compound is a pure Z imine isomer or a pure Z alkene isomer.
179. The pharmaceutical composition of claim 172, 173, 174, or 175, wherein the compound is a pure E imine isomer or a pure E alkene isomer.
180. The pharmaceutical composition of claim 172, 173, 174, or 175, wherein the composition can be administered orally.
181. The pharmaceutical composition of claim 172 or 173, wherein the compound has the structure: wherein each of Yi, Y 2 Y 3 and Y 4 is independently H; straight chained or branched CI-C? alkyl, -CF 3 F, -C1, -Br, -OR 4 -N(R 4 2 or -CON(R4) 2 wherein each R 4 is independently straight chained or branched CI-C 7 alkyl, -CF 3 or phenyl; wherein A is straight chained or branched C 1 -C 7 00 S799 F3 alkyl, aryl, heteroaryl, aryl(Ci-C 6 )alkyl or heteroaryl(Ci-C6) alkyl; and wherein A' is 00 CR 2 R 3 00 C 182. The pharmaceutical composition of claim 172, 173 or 175, wherein B is heteroaryl.
183. The pharmaceutical composition of claim 172 or 173, wherein B is aryl.
184. The pharmaceutical composition of claim 183, wherein B is phenyl and the phenyl is optionally substituted with one or more of the following: -Cl, -Br, CF 3 straight chained or branched Ci-C 7 alkyl, N(R 4 2 -OR 4 -COR 4 -NCOR4, -CO2R 4 or -CON(R) 2
185. The pharmaceutical composition of claim 184, wherein A is aryl.
186. The pharmaceutical composition of claim 184, wherein A is heteroaryl. 800
187. The pharmaceutical composition of claim 186, wherein the compound is selected from the group consisting of: F ff^T CI N& and N'"CI
188. The pharmaceutical composition of claim 174, wherein B is Q 6
189. The pharmaceutical composition of claim 188, wherein A is aryl. 00 S801 0 (N S190. The pharmaceutical composition of claim 189, wherein the compound has the structure: 0 N
191. The pharmaceutical composition of claim 190, wherein the compound is: 00F
192. The pharmaceutical composition of claim 175, wherein B is aryl.
193. The pharmaceutical composition of claim 192, wherein A is (CHR 17 )-(CHR 17 )n-Z. 802
194. The pharmaceutical composition of claim 193, wherein the compound is: C1 C NCI (N"
195. A compound having the structure: wherein each of Y 1 Y 2 Y 3 and Y 4 is independentl y H; straight chained or branched Cl-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 alkenyl or alkynyl; C 3 -C 7 cycloalkyl, or C, 5 -C 7 cycloalkenyl; -C1, -Br, or 1; -NO 2 -N 3 -CN; -OR 4 -SR 4 -OCOR4, -COR4, -NCOR4, N(R 4 2 -CON(R4) 2 or -COOR4; aryl or heteroaryl; or 803 any two of Y 1 Y 2 Y 3 and Y 4 present on adjacent carbon atoms can constitute a methylenedioxy group; wherein each R 4 is independently straight chained or branched Cl-C 7 alkyl, monofluoroalkyl or 00 polyfluoroalkyl; straight chained or branched C 2 -C 7 alkenyl or alkynyl; C 3 -C 7 cycloalkyl, C 5 -C 7 (1 cycloalkenyl, aryl or aryl(Cl-C 6 )alkyl; 00 wherein A is A' Q3, Q4~, Q5, straight chained or branched Cl-C 7 alkyl, aryl, heteroaryl, aryl(Cl- C 6 a lkyl, heteroaryl (Cl-C 6 alkyl, aryl substituted with an aryl or heteroaryl, heteroaryl substituted with an aryl or heteroaryl; or (CHR 17 (CHR 1 7 -Z; wherein A' is 0 0 R 5 n Ii CRR3or -(CH 2 t 4 wherein Q3 is wherein Q 4 is wherein Qs is wherein R 1 and R 2 are each independently H, straight S805 Schained or branched Ci-C7 alkyl, -Cl, -Br, NO 2 or -CN; (N wherein R 3 is H, straight chained or branched C 1 -C 7 alkyl, -Cl, -Br, -NO 2 -CN, -OR 6 aryl or 0 0 heteroaryl; C^ C wherein R 5 is straight chained or branched Ci-C 7 00 Salkyl, -N(R 4 2 -OR 6 or aryl; ^C wherein Rg is straight .chained or branched C 1 -C 7 alkyl or aryl; wherein each R17 is independently H; straight chained or branched Ci-C 7 alkyl, straight chained or branched C 1 -C 7 monofluoroalkyl, straight chained or branched CI-C7 polyfluoroalkyl, straight chained or branched C 2 -C 7 alkenyl, straight chained or branched C 2 -C 7 alkynyl, Cs-C 7 cycloalkenyl, -(CH 2 or (CH 2 )n-O- (CH 2 )m-CH 3 wherein each R 20 is independently straight chained or branched Ci-C7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C 7 alkenyl or alkynyl; C 3 -C7 cycloalkyl or Cs-C 7 cycloalkenyl; -C1, -Br, or -NO 2 -N 3 -CN; OR 21 -OCOR 21 -COR 21 -NCOR 21 -N(R 21 2 -CON(R 21 2 or -COOR 21 aryl or heteroaryl; or two R 20 groups present on adjacent carbon atoms can join together to form a methylenedioxy group; wherein each R 21 is independently straight 00 S806 Schained or branched C 1 -C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C 7 Salkenyl or alkynyl; C 3 -C 7 cycloalkyl, C 5 -C 7 cycloalkenyl, aryl or aryl(C 1 -C 6 )alkyl; 0 OO wherein each m is an integer from 0 to 4 inclusive; Cc C1 wherein each n is an integer from 1 to 4 inclusive; 00 C1 10 wherein each p is an integer from 0 to 2 inclusive; wherein U is 0, -NR 16 S, C(R 17 2 or -NSOzR 16 wherein Z is C 3 -Cio cycloalkyl, C 4 -C cyclic ether, C 4 -C 7 cyclic thioether, aryl, or heteroaryl; wherein R 16 is straight chained or branched Ci-C 7 alkyl, straight chained or branched CI-C 7 monofluoroalkyl, straight chained or branched C 1 -C7 polyfluoroalkyl, straight chained or branched C 2 -C7 alkenyl, straight chained or branched C 2 -C 7 alkynyl, Cs-C 7 cycloalkenyl, -(CH 2 or (CH 2 (CH 2 )-CH 3 wherein q is an integer from 2 to 4 inclusive; wherein B is aryl, heteroaryl, aryl substituted with an aryl or heteroaryl, heteroaryl substituted with an aryl or heteroaryl, tricyclic heteroaryl or Qs; provided however, if B is aryl or heteroaryl the carbon atom or carbon atoms ortho to the nitrogen atom of the imine bond may only be substituted with one or more of the following -Cl, -Br, -CN, 807 methyl, ethyl or methoxy; wherein a tricyclic heteroaryl is a fused three member aromatic system in which one or more of the rings is heteroaryl; carbazole; or acridine; wherein Q6 is wherein each R 22 is independently H, Cl, or straight chained or branched Ci-C 4 alkyl; or a pharmaceutically acceptable salt thereof.
196. A compound having the structure: B Y/ 1 N Y2 -0 Y4 wherein each of Y 1 Y 2 Y 3 and Y 4 is independently H; straight chained or branched Ci-C7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C 7 alkenyl or alkynyl; C 3 -C 7 cycloalkyl, or Cs-C 7 cycloalkenyl; -C1, -Br, or F-- 808 1; -NO 2 -N 3 -CN; -OR 4 -SR4, -OCOR4, -COR4, -NCOR4, N(R42 ,-CON(R 4 2 or -COOR4; aryl or heteroaryl; or any two of Y. 1 Y 2 Y 3 and Y 4 present on adjacent carbon atoms can constitute a methylenedioxy group; wherein each R 4 is independently straight chained or branched C 1 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C, alkenyl or alkynyl; C 3 cycloalkyl, C5s-C-7 cycloalkenyl, aryl or aryl(Cl-C 6 alkyl; wherein A is straight chained or branched alkyl, aryl, heteroaryl, aryl -C 6 alkyl heteroaryl (Cl-C 6 alkyl; C1 C7 or wherein A' is RS n CR 2 R 3 ;or -(CH 2 n R 4 wherein R, and R 2 are each independently H, straight chained or branched Cl-C., alkyl, -Cl, -Br, r S809 SNO 2 or -CN; r wherein R 3 is H, straight chained or branched C 1 -C 7 alkyl, -Cl, -Br, -NO 2 -CN, -OR 6 aryl or heteroaryl; 00 Swherein Rs is straight chained or branched C 1 -C 7 CI alkyl, -N(R 4 2 -ORs or aryl; 00 C- 10 wherein R 6 is straight chained or branched C 1 -C7 alkyl or aryl; wherein B is aryl, or heteroaryl; provided however, if B is aryl or heteroaryl the carbon atom or carbon atoms ortho to the nitrogen atom of the imine bond may only be substituted with one or more of the following -Cl, -Br, -CN, methyl, ethyl or methoxy; wherein n is an integer from 1 to 4 inclusive; or a pharmaceutically acceptable salt thereof. 00 810
197. A compound having the structure: 00 Y 000 wherein each of YI, Y 2 Y 3 and Y 4 is independently- H; straight chained or branched Cl-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or alkynyl; C 3 -C 7 cycloalkyl, or CS-C 7 cycloalkenyl; -C1, -Br, or 1; -NO 2 -N3; -CN; OR 4 -SR 4 0OC0R 4 -COR 4 -NCOR 4 N(R42 ,-CON(R 4 2 or -COOR 4 aryl or heteroaryl; or any two of Y 1 Y 2 Y 3 and Y 4 present on adjacent carbon atoms can constitute a methylenedioxy group; wherein each R 4 is independently straight chained or branched C 1 -C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C 7 alkenyl or alkynyl; C 3 -C 7 cycloalkyl, C 7 cycloalkenyl, aryl or aryl(Cl-C 6 )alkyl; wherein A is straight chained or branched Cl-C 7 alkyl, aryl, heteroaryl, aryl (C 1 -C 6 alkyl or heteroaryl (Cl-C6) alkyl; wherein A' is 00 O 00 O 00 0O (N 0 n R s n CR2R3, or (CH2) n R wherein B heteroaryl, heteroaryl, is aryl substituted with an aryl heteroaryl substituted with an aryl tricyclic heteroaryl or Q6; wherein a tricyclic heteroaryl is a fused three ring aromatic system in which one or more of the rings is heteroaryl; carbazole; or acridine; wherein Q6 is wherein n is an integer from 1 to 4 inclusive; 00 812 C(N wherein each R 22 is independently H, F, Cl, or straight chained or branched Ci-C 4 alkyl; or a pharmaceutically acceptable salt thereof. 00 Mr)
198. A compound having the structure: B 00 Y2 1 N T N Y2Y A Y4 wherein each of Y 1 Y 2 Y 3 and Y 4 is independently H; straight chained or branched C 1 -C7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched Cz-C, alkenyl or alkynyl; C 3 -C 7 cycloalkyl, or C5-C 7 cycloalkenyl; -C1, -Br, or I; -NO 2 -N 3 -CN; -OR 4 -SR4, -OCOR 4 -COR4, -NCOR 4 N(R4)2 -CON(R 4 2 or -COOR4; aryl or heteroaryl; or any two of Y 1 Y 2 Y 3 and Y 4 present on adjacent carbon atoms can constitute a methylenedioxy group'; wherein each R 4 is independently straight chained or branched Ci-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C 7 alkenyl or alkynyl; C 3 -C 7 cycloalkyl, C 5 -C 7 cycloalkenyl, aryl or aryl(Ci-C) alkyl; 7 813 wherein A is Q3, Q4, Q5, aryl substituted with an aryl or heteroaryl, heteroaryl substituted with an aryl or heteroaryl, or (CHR 17 -(CHR,1 7 )n-ZF wherein Q3 is wherein Q4 is wherein Q5 is 0 00 814 h- wherein each R 17 is independently H; straight chained Sor branched CI-C 7 alkyl, straight chained or branched CC-C 7 monofluoroalkyl, straight chained or branched CI-C 7 polyfluoroalkyl, straight chained or branched 00 C 2 -C 7 alkenyl, straight chained or branched C 2 -C7 Salkynyl, Cs-C 7 cycloalkenyl, -(CH 2 or (CH 2 )m-CH 3 00 C- 10 wherein each R 20 is independently straight chained or branched C1-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C7 alkenyl or alkynyl; C 3 -C7 cycloalkyl or C 5 -C 7 cycloalkenyl; -Cl, -Br, or -NO 2 -N 3 -CN; OR 21 -OCOR 21 -COR 21 -NCOR 21 -N(R 21 2 -CON(R 2 1) 2 or -COOR 21 aryl or heteroaryl; or two R 20 groups present on adjacent carbon atoms can join together to form a methylenedioxy group; wherein each R 21 is independently straight chained -or branched Ci-C 7 alkyl, monofluoroalkyl or polyfluoroalkyl; straight chained or branched C 2 -C 7 alkenyl or alkynyl; C 3 -C7 cycloalkyl, C 5 -C7 cycloalkenyl or aryl; wherein each R 22 is independently H, F, C1, or straight chained or branched C 1 -C 4 alkyl; wherein q is an integer from 2 to 4 inclusive; wherein each m is an integer from 0 to 4 inclusive; 00 815 Swherein each n is an integer from 1 to 4 inclusive; Vt wherein each p is an integer from 0 to 2 inclusive; wherein U is O, -NR 16 S, C(R 17 2 or -NSO 2 R 16 00 Swherein Z is C 3 -C 1 0 cycloalkyl, C 4 -C7 cyclic ether, C< C 4 -C 7 cyclic thioether, aryl, or heteroaryl; 00 10 wherein R 16 is straight chained or branched Ci-C 7 alkyl, straight chained or branched C 1 -C 7 monofluoroalkyl, straight chained or branched Ci-C 7 polyfluoroalkyl, straight chained or branched C 2 -C 7 alkenyl, straight chained or branched C 2 -C 7 alkynyl, Cs-C7 cycloalkenyl, -(CH 2 or (CH 2 )q-O-(CH 2 )m-CH3; wherein B is aryl, or heteroaryl; provided however, if B is aryl or heteroaryl the carbon atom or carbon atoms ortho to the nitrogen atom of the imine bond may only be substituted with one or more of the following -Cl, -Br, -CN, methyl, ethyl or methoxy; or a pharmaceutically acceptable salt thereof.
199. An enantiomerically and diastereomerically pure compound of claim 195, 196, 197, or 198.
200. An enantiomerically or diastereomerically pure compound of claim 195, 196, 197, or 198.
201. A pure Z imine isomer or a pure Z alkene isomer of 816 the compound of claim 195, 196, 197, or 198.
202. A pure E imine isomer or a pure E alkene isomer of the compound of claim 195, 196, 197, or 198.
203. The compound of claim 195, 196, 197, or 198, wherein the compound can be administered orally.
204. The compound of claim 195 or 196, wherein the compound has the structure: wherein each of Yi, Y 2 Y 3 and Y 4 is independently H; straight chained or branched Ci-C 7 alkyl, -CF 3 F, -C1, -Br, -OR 4 -N(R 4 2 or -CON(R 4 2 wherein each R 4 is independently straight chained or branched Ci-C 7 alkyl, -CF 3 or phenyl; wherein A is straight chained or branched C 1 -C alkyl, aryl, heteroaryl, aryl(Ci-Cs)alkyl or heteroaryl (C 1 -C 6 alkyl; and wherein A' is 817 n CR 2 R 3 00 Mn 205. The compound of claim 195, 196 or 198, wherein B is Sheteroaryl. 00 O 206. The compound of claim 195 or 196, wherein B is aryl.
207. The compound of claim'206, wherein B is phenyl and the phenyl is optionally substituted with one or more of the following: -Cl, -Br, -CF 3 straight chained or branched Ci-C? alkyl, -N(R 4 2 -OR 4 -COR 4 -NCOR 4 -CO2R4, or -CON(R 4 2
208. The compound of claim 207, wherein A is aryl.
209. The compound of claim 207, wherein A is heteroaryl. 818
210. The compound of claim 209, wherein the compound is selected from the group consisting of: F O F CI N4j 1 and criiJo NC
211. The compound of claim 197, wherein B is Q
0212. The compound of claim 211, wherein A is aryl. 212. The compound of claim 211, wherein A is aryl.
213. The compound of claim 212, wherein the compound has the structure: 6°R 22 R22 N- aO 00 oo
215. The compound of claim 19 I214. The compound of claim 213, wherein the compound is: cC SFC N0 0 215. The compound of claim 198, wherein B is aryl.
216. The compound of claim 215, wherein A is (CHRi7)- (CHRi7) n-Z.
217. The compound of claim 215, wherein the compound is: Cl
218. A pure Z imine isomer of the compound of claim 195, 196, 197 or 198.
219. A pure E imine isomer of the compound of claim 195, 196, 197 or 198. oo 00 820
220. A pharmaceutical composition comprising a therapeutically effective amount of the compound of V)claim 195, 196, 197 or 198, and a pharmaceutically acceptable carrier. 00 221. A pharmaceutical composition made by combining a therapeutically effective amount of the compound of CI claim 195, 196, 197 or 198, and a pharmaceutically 00 acceptable carrier. C
222. A process for making a pharmaceutical composition comprising combining a therapeutically effective amount of the compound of claim 195, 196, 197 or 198, and a pharmaceutically acceptable carrier.
223. A method of treating a subject suffering from depression which comprises administering to the subject an amount of the compound of claim 195, 196, 197 or 198 effective to treat the subject's depression.
224.* A method of treating a subject suffering from anxiety which comprises administering to the subject an amount of the compound of claim 195, 196, 197 or 198 effective to treat the subject's anxiety.
225. A method of treating a subject suffering from depression and anxiety which comprises administering to the subject an amount of the compound of claim 195, 196, 197 or 198 effective to treat the subject's depression and anxiety. 821
226. A method of treating depression in a subject which V comprises administering to the subject a composition comprising a pharmaceutically acceptable carrier and a therapeutically effective amount of a GAL3 receptor 00 antagonist, wherein: the GAL3 receptor antagonist binds to the human GAL3 Sreceptor with a binding affinity at least ten-fold 00 higher than the binding affinity with which it binds to the human GAL1 receptor; the GAL3 receptor antagonist does not inhibit the activity of central monoamine oxidase A greater than percent, at a concentration of 10pM; and the GAL3 receptor antagonist does not inhibit the activity of central monoamine oxidase B greater than percent, at a concentration of 10pM; and the GAL3 receptor antagonist binds to the human GAL3 receptor with a binding affinity at least ten-fold higher than the binding affinity with which it binds to each of the following transporters: serotonin transporter, norepinephrine transporter, and dopamine transporter.
227. The method of claim 226, wherein the receptor antagonist binds to the human GAL3 receptor with a binding affinity at least 30-fold higher than the binding affinity with which it binds to the human GAL1 receptor.
228. The method of claim 227, wherein the receptor antagonist binds to the human GAL3 receptor with a binding affinity at least 50-fold higher than the binding affinity with which it binds to the human GAL1 receptor.
229. The method of claim 228, wherein the receptor S822 Santagonist binds to the human GAL3 receptor with a binding affinity at least 100-fold higher than the binding affinity V) with which it binds to the human GAL1 receptor.
230. The method of claim 229, wherein the receptor 00 antagonist binds to the human GAL3 receptor with a binding O affinity at least 200-fold higher than the binding affinity Swith which it binds to the human GAL1 receptor. 00
231. The method of claim 226, wherein the receptor antagonist additionally binds to the human GAL3 receptor with a binding affinity at least ten-fold higher than the binding affinity with which it binds to the human GAL2 receptor.
232. The method of claim 226, wherein the receptor antagonist also binds to the human GAL3 receptor with a binding affinity at least ten-fold higher than the binding affinity with which it binds to each of the human human 5HT1D, human 5HTIE, human 5HTiF, human 5HT 2 A, rat 5HT 2 c, human 5HTs and human 5HT 7 receptors.
233. The method of claim 226, wherein the receptor antagonist also binds to the human GAL3 receptor with a binding affinity at least ten-fold higher than the binding affinity with which it binds to the human histamine H 1 receptor.
234. The method of claim 226, wherein the receptor antagonist also binds to the human GAL3 receptor with a binding affinity at least ten-fold higher than the binding affinity with which it binds to the human dopamine DI, D 2 D 3 D 4 andDs receptors. 823
235. The method of claim 226, wherein the receptor antagonist also binds to the human GAL3 receptor with a binding affinity at least ten-fold higher than the binding affinity with which it binds to the human aAA adrenoceptor, the human aB adrenoceptor and the human aiD adrenoceptor.
236. The method of claim 226, wherein the receptor antagonist also binds to the human GAL3 receptor with a binding affinity at least ten-fold higher than the binding affinity with which it binds to the human a2A adrenoceptor, the human 2B 3 adrenoceptor and the human a2c adrenoceptor.
237. The method of claim 226, wherein the receptor antagonist also binds to the human GAL3 receptor with a binding affinity less than ten-fold higher than the binding affinity with which it binds to the human 5HT 4 receptor.
238. The method of claim 226, wherein the receptor antagonist also binds to the human GAL3 receptor with a binding affinity less than ten-fold higher than the binding affinity with which it binds to the human 5HTA receptor.
239. The method antagonist does monoamine oxidase of claim 226, wherein the receptor not inhibit the activity of central A greater than 30 percent.
240. The method antagonist does monoamine oxidase
241. The method antagonist does monoamine oxidase of claim 226, not inhibit the B greater than 30 wherein the activity of percent. receptor central of claim 226, wherein the receptor not inhibit the activity of central A greater than 15 percent. 00 824 c-s 242. The method of claim 226, wherein the receptor lr antagonist does not inhibit the activity of central monoamine oxidase B greater than 15 percent. 00 243. A method of treating anxiety in a subject which comprises administering to the subject a composition C1 comprising a pharmaceutically acceptable carrier and a 00 therapeutically effective amount of a GAL3 receptor. antagonist, wherein: the GAL3 receptor antagonist binds to the human GAL3 receptor with a binding affinity at least ten-fold higher than the binding affinity with which it binds to the human GALl receptor; and the GAL3 receptor antagonist binds to the human GAL3 receptor with a binding affinity at least ten-fold higher than the binding affinity with which it binds to each of the following transporters: serotonin transporter, norepinephrine transporter, and dopamine transporter.
244. The method of claim 243, wherein the receptor antagonist binds to the human GAL3 receptor with a binding affinity at least 30-fold higher than the binding affinity with which it binds to the human GAL1 receptor.
245. The method of claim 244, wherein the receptor antagonist binds to the human GAL3 receptor with a binding affinity at least 50-fold higher than the binding affinity with which it binds to the human GAL1 receptor.
246. The method of claim 245, wherein the receptor antagonist binds to the human GAL3 receptor with a binding affinity at least 100-fold higher than the binding affinity 00 0 825 Swith which it binds to the human GAL1 receptor. Vt 247. The method of claim 246, wherein the receptor antagonist binds to the human GAL3 receptor with a binding affinity at least 200-fold higher than the binding affinity OO with which it binds to the human GAL1 receptor. S248. The method of claim 243, wherein the receptor 00 antagonist additionally binds to the human GAL3 receptor with a binding affinity at least ten-fold higher than the C binding affinity with which it binds to the human GAL2 receptor.
249. The method of claim 243, wherein the receptor antagonist also binds to the human GAL3 receptor with a binding affinity at least ten-fold higher than the binding affinity with which it binds to each of the human human 5HT 1 o, human 5HT1E, human 5HTpF, human 5HTA, rat 5HT 2 c, human 5HTs and human 5HT 7 receptors.
250. The method of claim 243, wherein the receptor antagonist also binds to the human GAL3 receptor with a binding affinity at least ten-fold higher than the binding affinity with which it binds to the human histamine H, receptor.
251. The method of claim 243, wherein the receptor antagonist also binds to the human GAL3 receptor with a binding affinity at least ten-fold higher than the binding affinity with which it binds to the human dopamine Di, D 2 D 3 D 4 and Ds receptors.
252. The method of claim 243, wherein the receptor antagonist also binds to the human GAL3 receptor with a 00 S826 Sbinding affinity at least ten-fold higher than the binding c-s affinity with which it binds to the human XA adrenoceptor, V the human aiB adrenoceptor and the human a- adrenoceptor.
253. The method of claim 243, wherein the receptor 00 antagonist also binds to the human GAL3 receptor with a Sbinding affinity at least ten-fold higher than the binding CI affinity with which it binds to the human oZA adrenoceptor, 00 Sthe human o2B adrenoceptor and the human a2c adrenoceptor. C
254. The method of claim 243, wherein the receptor antagonist also binds to the human GAL3 receptor with a binding affinity less than ten-fold higher than the binding affinity with which it binds to the human 5HT 4 receptor.
255. The method of claim 243, wherein the receptor antagonist also binds to the human GAL3 receptor with a binding affinity less than ten-fold higher than the binding affinity with which it binds to the human 5HT1A receptor.
AU2008200380A 2001-01-31 2008-01-25 Use of GAL3 receptor antagonists for the treatment of depression and/or anxiety and compounds useful in such methods Abandoned AU2008200380A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2008200380A AU2008200380A1 (en) 2001-01-31 2008-01-25 Use of GAL3 receptor antagonists for the treatment of depression and/or anxiety and compounds useful in such methods

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US09/775,341 2001-01-31
AU2002247149A AU2002247149B2 (en) 2001-01-31 2002-01-31 Use of GAL3 receptor antagonists for the treatment of depression and/or anxiety and compounds useful in such methods
AU2008200380A AU2008200380A1 (en) 2001-01-31 2008-01-25 Use of GAL3 receptor antagonists for the treatment of depression and/or anxiety and compounds useful in such methods

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU2002247149A Division AU2002247149B2 (en) 2001-01-31 2002-01-31 Use of GAL3 receptor antagonists for the treatment of depression and/or anxiety and compounds useful in such methods

Publications (1)

Publication Number Publication Date
AU2008200380A1 true AU2008200380A1 (en) 2008-02-21

Family

ID=39233157

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2008200380A Abandoned AU2008200380A1 (en) 2001-01-31 2008-01-25 Use of GAL3 receptor antagonists for the treatment of depression and/or anxiety and compounds useful in such methods

Country Status (1)

Country Link
AU (1) AU2008200380A1 (en)

Similar Documents

Publication Publication Date Title
US7465750B2 (en) Use of GAL3 antagonist for treatment of depression and/or anxiety and compounds useful in such methods
US20030078271A1 (en) Use of GAL3 receptor antagonists for the treatment of depression and/or anxiety and compounds useful in such methods
EP1300398B1 (en) Propane-1,3-dione derivatives
US7868034B2 (en) Use of GALR3 receptor antagonists for the treatment of depression and/or anxiety and compounds useful in such methods
AU2008200385B2 (en) Use of GAL3 receptor antagonists for the treatment of depression and/or anxiety and compounds useful in such methods
US7220775B2 (en) Compound useful for the treatment of neuropathic pain
US7166635B2 (en) 3-imino-2-indolones for the treatment of depression and/or anxiety
AU2002247149A1 (en) Use of GAL3 receptor antagonists for the treatment of depression and/or anxiety and compounds useful in such methods
US20040110821A1 (en) GAL3 receptor antagonists for the treatment of affective disorders
WO2004014307A2 (en) Gal3 antagonists for the treatment of neuropathic pain
WO2004014376A1 (en) Gal3 receptor antagonists for the treatment of affective disorders
AU2008200380A1 (en) Use of GAL3 receptor antagonists for the treatment of depression and/or anxiety and compounds useful in such methods
US6936607B2 (en) 2,4,6-Triaminopyrimidines for the treatment of depression and/or anxiety
WO2004014854A1 (en) 3-imino-2-indolones for the treatment of depression and/or anxiety
WO2004034967A2 (en) 2,4,6-triaminopyrimidines for the treatment of depression and/or anxiety

Legal Events

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