Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
SELECTIVE D3 DOPAMINE RECEPTOR AGONISTS AND METHODS OF THEIR USE
Document Type and Number:
WIPO Patent Application WO/2017/181004
Kind Code:
A1
Abstract:
The disclosure of a compound of Formula (I) or a pharmaceutically acceptable salt thereof (I) The variables W, R1, R2, R3, and R4 are defined in the disclosure. The disclosure provides a compound or salt of Formula (I) together with a pharmaceutically acceptable carrier. The disclosure also provides methods of treating a patient for Parkinson's disease and related syndromes, dyskinesia, especially dyskinesias secondary to treating Parkinson's disease with L-DOPA, neurodegenerative disorders such as Alzheimer's disease and dementia, Huntington's disease, restless legs syndrome, bipolar disorder and depression, schizophrenia, cognitive dysfunction, or substance use disorders, the methods comprising administering a compound of Formula I or salt thereof to the patient. The disclosure provides combination methods of treatment in which the compound of Formula (I) is administered to the patient together with one or more additional active agents.

Inventors:
SIBLEY DAVID R (US)
MORITZ AMY ELIZABETH (US)
FREE R BENJAMIN (US)
STEINER JOSEPH P (US)
SOUTHALL NOEL TERRENCE (US)
FERRER MARC (US)
HU XIN (US)
WEINER WARREN S (US)
AUBÉ JEFFREY (US)
FRANKOWSKI KEVIN (US)
Application Number:
PCT/US2017/027618
Publication Date:
October 19, 2017
Filing Date:
April 14, 2017
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
US HEALTH (US)
UNIV KANSAS (US)
International Classes:
C07D333/70; A61P25/14; A61P25/16; A61P25/28; C07D209/42; C07D235/24; C07D295/192; C07D307/85; C07D339/06; C07D401/12; C07D405/12; C07D409/12; C07D471/04
Domestic Patent References:
WO2008016973A12008-02-07
WO2003002543A12003-01-09
WO2010123995A12010-10-28
Foreign References:
CN102285962A2011-12-21
EP1484330A12004-12-08
US6444674B12002-09-03
US5932734A1999-08-03
US20090054449A12009-02-26
Other References:
RAMESH M. KANOJIA ET AL: "Synthesis and class III type antiarrhythmic activity of 4-aroyl (and aryl)-1-aralkylpiperazines", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, vol. 10, no. 24, 18 December 2000 (2000-12-18), AMSTERDAM, NL, pages 2819 - 2823, XP055395652, ISSN: 0960-894X, DOI: 10.1016/S0960-894X(00)00581-3
GUO XIAOKE ET AL: "Discovery of 1-aryloxyethyl piperazine derivatives as Kv1.5 potassium channel inhibitors (part I)", EUROPEAN JOURNAL OF MEDICINAL CHEMISTRY, vol. 81, 28 March 2014 (2014-03-28), pages 89 - 94, XP028851807, ISSN: 0223-5234, DOI: 10.1016/J.EJMECH.2014.03.075
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 20 May 2009 (2009-05-20), XP002773047, retrieved from STN Database accession no. 1147661-57-1
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 3 August 2012 (2012-08-03), XP002773048, retrieved from STN Database accession no. 1386162-69-1
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 16 August 2011 (2011-08-16), XP002773049, retrieved from STN Database accession no. 1318530-74-3
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 15 August 2011 (2011-08-15), XP002773050, retrieved from STN Database accession no. 1318173-92-0
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 29 January 2009 (2009-01-29), XP002773051, retrieved from STN Database accession no. 1097612-59-3
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 29 January 2009 (2009-01-29), XP002773052, retrieved from STN Database accession no. 1097492-13-1
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 24 May 2011 (2011-05-24), XP002773053, retrieved from STN Database accession no. 1299403-53-4
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 19 April 2007 (2007-04-19), XP002773054, retrieved from STN Database accession no. 930893-33-7
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 17 April 2007 (2007-04-17), XP002773055, retrieved from STN Database accession no. 930440-50-9
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 25 April 2008 (2008-04-25), XP002773056, retrieved from STN Database accession no. 1017214-96-8
Attorney, Agent or Firm:
MAXWELL, Leslie-Anne (US)
Download PDF:
Claims:
CLAIMS

What is claimed is:

1. A compound of Formula I

or a pharmaceutically acceptable salt thereof, wherein:

W is O or S;

R1 is aryl or heteroaryl, and

R2 is a phenyl, a 5 or 6-membered heteroaryl, having 1, 2, 3, or 4 heteroatoms independently chosen from N, O, and S or a 6,5-bicyclic heteroaryl group, having 1, 2, 3, 4, 5, or 6 heteroatoms independently chosen from N, O, and S, wherein the point of attachment in Formula I is in the 5-membered ring, and the 5-membered ring contains at least one heteroatom; where

R1 and R2 are each optionally substituted with 1 or more substituents independently chosen from halogen, hydroxyl, cyano, nitro, oxo, -CONH2, amino, mono- and di-Ci- C4alkylcarboxamide, (C3-C6cycloalkyl)Co-C2alkyl, and Ci-C6hydrocarbyl, which Ci- C6hydrocarbyl group is a hydrocarbon chain in which the carbon atoms are joined by single, double or triple bonds, and any one carbon atom can be replaced by O, NH, or N(Ci-C4alkyl) and which hydrocarbyl group is optionally substituted with one or more substituents independently chosen from hydroxyl, oxo, halogen, and amino; and

R3 and R4 are independently hydrogen or methyl.

2. The compound or salt of Claim 1, wherein R3 and R4 are both hydrogen.

3. The compound of Claim 1, of the Formula I- A

or a pharmaceutically acceptable salt thereof, wherein:

R2 is a 6,5-bicyclic heteroaryl group, having 1, 2, 3, 4, 5, or 6 heteroatoms independently chosen from N, O, and S, wherein the point of attachment in Formula I is in the 5-membered ring, and the 5-membered ring contains at least one heteroatom; where substituted with 0 or 1 or more substituents independently chosen from halogen, hydroxyl, cyano, nitro, oxo, -CONH2, amino, mono- and di-Ci-C4alkylcarboxamide, (C3- C6cycloalkyl)Co-C2alkyl, and Ci-C6hydrocarbyl, which Ci-C6hydrocarbyl group is a hydrocarbon chain in which the carbon atoms are joined by single, double or triple bonds, and any one carbon atom can be replaced by O, NH, or N(Ci-C4alkyl) and which hydrocarbyl group is optionally substituted with one or more substituents independently chosen from hydroxyl, oxo, halogen, and amino.

4. The compound or salt of Claim 1, wherein

R2 is a 6,5-bicyclic heteroaryl group selected from the following optionally

5. The compound or salt of Claim 1, wherein

R2 is a phenyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, or pyrolyl group, each of which is optionally substituted.

6. The compound or salt of any of Claims 1 to 4, where R2 is a 6,5-bicyclic heteroaryl group substituted with

0 or 1 or more substituents independently chosen from halogen, hydroxyl, cyano, nitro, - CONH2, amino, Ci-C6alkyl, Ci-C6alkoxy, C2-C4alkanoyl, Ci-C4alkylester mono- and di-Ci- C4alkylamino, mono- and di-Ci-C4alkylcarboxamide, (C3-C6cycloalkyl)Co-C2alkyl, and any one carbon atom in a Ci-C6alkyl or Ci-C6alkoxy can be replaced by O, NH, or N(Ci-C4alkyl) and which Ci-C6alkyl or Ci-C6alkoxy is optionally substituted with one or more substituents independently chosen from hydroxyl, halogen, and amino.

7. The compound or salt of any of Claims 1 to 4, wherein

R2 i is substituted with 0 or 1 or more substituents

independently chosen from halogen, hydroxyl, cyano, nitro, -CONH2, amino, Ci-C6alkyl, Ci- C6alkoxy, C2-C4alkanoyl, Ci-C4alkylester mono- and di-Ci-C4alkylamino, mono- and di-Ci- C4alkylcarboxamide, (C3-C6cycloalkyl)Co-C2alkyl, and any one carbon atom in a Ci-C6alkyl or Ci-C6alkoxy can be replaced by O, NH, or N(Ci-C4alkyl) and which Ci-C6alkyl or Ci- C6alkoxy is optionally substituted with one or more substituents independently chosen from hydroxyl, halogen, and amino.

8. The compound or salt of any one of Claims 1 to 4, wherein R2 is a 6,5-bicyclic heteroaryl group substituted with 0 or 1 to 3 substituents independently selected from halogen, Ci-C2alkyl, and Ci-C2alkoxy.

9. The compound or salt of any one of Claims 1 to 8, wherein W is S.

10. The compound or salt of any one of Claims 1 to 8, wherein W is O.

11. The compound or salt of any one of Claims 1 to 10, where

R1 is phenyl or a monocyclic heteroaryl group having 1, 2, 3 or 4 heteroatoms independently chosen from N, O, and S, each of which R1 is optionally substituted.

12. The compound or salt of any one of Claims 1 to 10, wherein

R1 is phenyl or pyridyl, each of which is optionally substituted.

13. The compound or salt of any one of Claims 1 to 10, wherein R1 is a phenyl or pyridyl, each of which is fused to a 5-membered heterocyclic ring, each of which R1 is optionally substituted.

14. The compound or salt of Claim 13, wherein R1 is optionally substituted indole.

15. The compound or salt of any one of Claims 1 to 14, wherein R is substituted with 0 or 1 or more substituents independently chosen from halogen, hydroxyl, cyano, nitro, -CONH2, amino, Ci-C6alkyl, Ci-C6alkoxy, C2-C4alkanoyl, Ci- C4alkylester mono- and di-Ci-C4alkylamino, mono- and di-Ci-C4alkylcarboxamide, (C3- C6cycloalkyl)Co-C2alkyl, and any one carbon atom in a Ci-C6alkyl or Ci-C6alkoxy can be replaced by O, NH, or N(Ci-C4alkyl) and which Ci-C6alkyl or Ci-C6alkoxy is optionally substituted with one or more substituents independently chosen from hydroxyl, oxo, halogen, and amino.

16. The compound or salt of Claim 15, wherein R1 is substituted with 0 or 1 to 3 substituents independently chosen from halogen, Ci-C2alkyl, and Ci-C2alkoxy.

17. The compound or salt of any one of Claims 1 to 16 wherein R1 is 3-pyridyl.

18. The compound or salt of any one of Claims 1 to 16, wherein R1 is 4-methoxy- phenyl.

19. The compound or salt of Claim 3, wherein the compound of Formula (I) is a compound in which -W-R1 and R2 have the following values:

51

52

54

20. A pharmaceutical comprising a compound or salt of any one of Claims 1 to 19, together with a pharmaceutically acceptable carrier.

21. The pharmaceutical composition of Claim 20. wherein the composition is in the form of an oral dosage form.

22. A method of treating a patient suffering from Parkinson's disease and related syndromes, dyskinesia, especially dyskinesias secondary to treating Parkinson's disease with L-DOPA, neurodegenerative disorders such as Alzheimer's disease and dementia,

Huntington's disease, restless legs syndrome, bipolar disorder and depression, schizophrenia, cognitive dysfunction, or substance use disorders, comprising administering a therapeutically effective amount of a compound of any one of Claims 1 to 19 to the patient.

23. The method of Claim 22, wherein the compound or salt of Formula I is a first active agent and is administered together with one or more additional active agents.

24. The method of Claim 23, wherein one of the one or more additional active agents is L-DOPA.

Description:
SELECTIVE D 3 DOPAMINE RECEPTOR AGONISTS AND METHODS OF THEIR USE

STATEMENT OF GOVERNMENT SUPPORT

[0001] This invention was made in part with government support from the US Department of Health and Human Services, National Institutes of Health. The government has certain rights in this invention.

BACKGROUND

[0002] G-protein coupled receptors (GPCRs) are among the most intensely investigated drug targets in the pharmaceutical industry. Over 40% of all FDA approved drugs target these important receptor proteins. Unfortunately, many of the ligands that are used as drugs or pharmacological tools are not selective and exhibit some unintended activity on non-target GPCRs or other proteins. This is because the orthosteric binding site is highly conserved among closely related types of GPCRs.

[0003] Dopamine receptors (DARs) are G-protein coupled receptors (GPCRs) that play a critical role in cell signaling processes and modulation of information transfer within the nervous system. Five functionally active DARs have been identified in the mammalian genome. The DAR subtypes have profoundly diverse physiological effects. Studies investigating compounds that semi-selectively activate the D 3 DAR suggest that D 3 activation may elicit important neuroprotection actions. Despite numerous attempts to produce D 3 selective modulators, current FDA-approved drugs display relatively poor selectivity between the closely related D 2 and D 3 DARs. D 2 and D 3 receptors share 74% homology in their transmembrane spanning domains, and the putative orthosteric binding sites, where the endogenous agonist dopamine (DA) binds, are 94% identical. Therefore, most currently available dopaminergic drugs, including antipsychotics and anti-Parkinson's disease medications, and research tool compounds that modulate either the D 2 or D 3 DARs modulate the other subtype to varying degrees.

[0004] Studies using semi-selective (i.e., ~ 10-fold) D 3 /D 2 DAR orthosteric agonists suggest that D 3 DAR activation may have important therapeutic potential. Compounds that stimulate both the D 2 DAR and D 3 DAR, but have somewhat higher affinity for the D 3 DAR, such as the FDA-approved drugs pramipexole and ropinirole, are effective in the treatment of Parkinson's disease and restless legs syndrome (RLS). These compounds are not only clinically active in relieving motor deficits, but also slow the loss of dopaminergic terminals upon long-term administration to patients with Parkinson's disease. Further, in preclinical animal models, D 3 DAR-pref erring agonists are potent neuroprotective agents. Treatment with D 3 DAR-preferring agonists has been shown to decrease the loss of dopaminergic neurons in animals treated with selective toxins that target these cells, and studies with D 3 DAR knockout mice support that the neuroprotective effects are mediated directly by the D 3 DAR. Pramipexole has also been shown to prevent neurotoxicity induced by oligomers of beta-amyloid and to restore nigrostriatal dopamine in lesioned mice. Although it is difficult to assess neuroprotection in living human subjects, clinical trials assessing the neuroprotective effects of pramipexole in Parkinson's disease have suggested positive effects. D 3 -preferring ligands have clinical potential in treating Parkinson's disease and related syndromes, dyskinesia, especially dyskinesias secondary to treating Parkinson's disease with L-DOPA, neurodegenerative disorders such as Alzheimer's disease and dementia, restless legs syndrome, depression, schizophrenia, cognitive dysfunction, or substance use disorders, including addiction to alcohol, nicotine, cocaine, methamphetamine and opioids. However, conventional D 3 -preferring D 2 /D 3 DAR agonists can induce impulse control disorders, suggesting that D 2 DAR stimulation may be involved in this significant side effect. Thus, there is a need for highly selective D 3 DAR agonists. This disclosure fulfills this need and provides additional advantages.

SUMMARY

[0005] The present disclosure provides a compound of Formula I or a pharmaceutically acceptable salt thereof. In Formula I the variables W, R 1 , and R 2 carry the following definitions.

[0006] W is O or S.

[0007] R 1 is aryl or heteroaryl.

[0008] R 2 is a a phenyl, a 5 or 6-membered heteroaryl, having 1, 2, 3, or 4

heteroatoms independently chosen from N, O, and S or a 6,5-bicyclic heteroaryl group, having 1, 2, 3, 4, 5, or 6 heteroatoms independently chosen from N, O, and S, wherein the point of attachment in Formula I is in the 5-membered ring, and the 5-membered ring contains at least one heteroatom. [0009] R 1 and R 2 are each substituted with 0 or 1 or more substituents independently chosen from halogen, hydroxyl, cyano, nitro, oxo, -CONH2, amino, mono- and di-Ci- C 4 alkylcarboxamide, (C3-C6cycloalkyl)Co-C2alkyl, and Ci-C6hydrocarbyl, which Ci- C6hydrocarbyl group is a hydrocarbon chain in which the carbon atoms are joined by single, double or triple bonds, and any one carbon atom can be replaced by O, NH, or N(Ci-C 4 alkyl) and which hydrocarbyl group is optionally substituted with one or more substituents independently chosen from hydroxyl, oxo, halogen, and amino.

[0010] R 3 and R 4 are independently hydrogen or methyl.

[0011] The disclosure also provides pharmaceutical compositions comprising a compound of Formula I or salt thereof together with a pharmaceutically acceptable carrier.

[0012] The disclosure also includes methods of treating Parkinson's disease and related syndromes, dyskinesia, especially dyskinesias secondary to treating Parkinson's disease with L-DOPA, neurodegenerative disorders such as Alzheimer's disease and dementia, Huntington' s disease, restless legs syndrome, bipolar disorder and depression, schizophrenia, cognitive dysfunction, or substance use disorders, comprising administering a therapeutically effective amount of a compound or salt of Formula I and at least one pharmaceutically acceptable carrier to a patient in need of such treatment. The compound of Formula I may be the only active compound administered to the patient or may be administered together with one or more additional active agents

BRIEF DESCRIPTION OF THE DRAWINGS

[0013] FIGURE 1. A graph of β-arrestin recruitment (% dopamine maximum) versus concentration of Compound 1 (molar, M) illustrating concentration-response curves for Compound 1 in an arrestin recruitment agonist assay for the Di, D 2 , D3, D 4 , and D5 receptors.

[0014] FIGURE 2. Graphs of BRET Ratio (% dopamine response, % DA response) versus concentration (molar, M) of test compound or control. (Fig. 2A) BRET arrestin recruitment assay, (Fig. 2B) G 0 BRET activation assay, and (Fig. 2C) ERK1/2

Phosphorylation assay.

[0015] FIGURE 3. A 3-dimensional plot of β-arrestin recruitment (% activity) versus receptor type (designated by letter and number), illustrating results of Compound 1 in DiscoverX gpcrMAX SM GPCR assay panel as an agonist.

[0016] FIGURE 4. Inhibition binding profiles of Compound 1. (Fig. 4A) The legend of GPCRs tested in the panel. (Fig. 4B) Full numerical results for Compound 1 in the panel. (Fig. 4C) Histogram of the screen shows that Compound 1 displays affinity only for the D3R, 5-HT1A, 5-HT2B, and Sigmal receptor.

[0017] FIGURE 5. Arrestin BRET and Go BRET assays showing the effect of the Y198A and Y365A mutations on Compound 1 and Dopamine potency.

[0018] FIGURE 6. (Fig. 6A). A graph of neuroprotection (% untreated control) versus compound and dose, illustrating a cell-based neuroprotection assay for Compound 1 and pramipexole as a comparator. (Fig. 6B) A graph of neuroprotection (% untreated control) versus concentration of Compound 1 (molar, M), illustrating a cell-based neuroprotection assay for Compound 1.

[0019] FIGURE 7. Mouse Plasma and Brain levels of Compound 1 as a function of time following administration of a single IP dose (20 mg/kg).

DETAILED DESCRIPTION

TERMINOLOGY

[0020] Compounds of the present disclosure are generally described using standard nomenclature.

[0021] The terms "a" and "an" do not denote a limitation of quantity, but rather denote the presence of at least one of the referenced item. The term "or" means "and/or." The open-ended transitional phrase "comprising" encompasses the intermediate transitional phrase "consisting essentially of and the close-ended phrase "consisting of." Claims reciting one of these three transitional phrases, or with an alternate transitional phrase such as "containing" or "including" can be written with any other transitional phrase unless clearly precluded by the context or art. Recitation of ranges of values are merely intended to serve as a shorthand method of referring individually to each separate value falling within the range, unless otherwise indicated herein, and each separate value is incorporated into the

specification as if it were individually recited herein. The endpoints of all ranges are included within the range and independently combinable. All methods described herein can be performed in a suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language (e.g., "such as"), is intended merely to better illustrate the disclosure and does not pose a limitation on its scope unless otherwise claimed. No language in the specification should be construed as indicating any non-claimed element as essential to the practice of the invention as used herein. Unless defined otherwise, technical and scientific terms used herein have the same meaning as is commonly understood by one of skill in the art to which this disclosure belongs.

[0022] "Formula I" includes compounds and salts of certain subformulae.

[0023] In certain situations, the compounds of Formula I may contain one or more asymmetric elements such as stereogenic centers, stereogenic axes and the like, e.g.

asymmetric carbon atoms, so that the compounds can exist in different stereoisomeric forms. Formula I includes all stereoisomeric forms, including racemates, optically enriched, and optically pure forms. In addition, compounds with carbon-carbon double bonds may occur in Z- and E-forms, with all isomeric forms of the compounds being included in the present disclosure. In these situations, the single enantiomers, i.e., optically active forms can be obtained by asymmetric synthesis, synthesis from optically pure precursors, or by resolution of the racemates. Resolution of the racemates can also be accomplished, for example, by conventional methods such as crystallization in the presence of a resolving agent, or chromatography, using, for example a chiral HPLC column.

[0024] The disclosure of Formula I include all isotopes of atoms occurring in the present compounds. Isotopes include those atoms having the same atomic number but different mass numbers. By way of general example, and without limitation, isotopes of hydrogen include tritium and deuterium and isotopes of carbon include n C, 13 C, and 14 C and isotopes of fluorine including 19 F.

[0025] Certain compounds are described herein using a general formula that includes variables, e.g. W, R 1 , and R 2 . Unless otherwise specified, each variable within such a formula is defined independently of other variables. Thus, if a group is said to be substituted, e.g. with 0-2 R*, then said group may be substituted with up to two R* groups and R* at each occurrence is selected independently from the definition of R*. When a group is substituted by an "oxo" substituent a carbonyl bond replaces two hydrogen atoms on a carbon. An "oxo" substituent on an aromatic group or heteroaromatic group destroys the aromatic character of that group, e.g. a pyridyl substituted with oxo is a pyridone.

[0026] Combinations of substituents and/or variables are permissible only if such combinations result in stable compounds. A stable compound or stable structure is meant to imply a compound that is sufficiently robust to survive isolation from a reaction mixture, and subsequent formulation into an effective therapeutic agent.

[0027] The term "substituted" means that any one or more hydrogen atoms bound to the designated atom or group is replaced with a selection from the indicated group, provided that the designated atom's normal valence is not exceeded. Unless otherwise specified substituents are named into the core structure. For example, it is to be understood that when (cycloalkyl)alkyl is listed as a possible substituent the point of attachment of this substituent to the core structure is in the alkyl portion.

[0028] Substituents are named into the ring unless otherwise indicated. A dash ("-") or a double bond ("=") that is not between two letters or symbols indicates the point of attachment for a substituent. For example, -CONH2 is attached through the carbon atom.

[0029] An "active agent" means a compound (including a compound disclosed herein), element, or mixture that when administered to a patient, alone or in combination with another compound, element, or mixture, confers, directly or indirectly, a physiological effect on the subject. The indirect physiological effect may occur via a metabolite or other indirect mechanism. The "active agent" may also potentiate, or make more active another active agent. For example the compounds of Formula I potentiate the activity of other active agents when given in combination with another active agent, for example by lowering the effective dose of the other active agent.

[0030] An "aliphatic group" is a non-aromatic hydrocarbon group having the indicated number of carbon atoms. Aliphatic groups may be saturated, unsaturated, or cyclic.

[0031] "Alkyl" includes both branched and straight-chain saturated aliphatic hydrocarbon groups, having the specified number of carbon atoms. Thus, the term Ci- C6alkyl includes alkyl groups having from 1 to about 6 carbon atoms. When Co-C n alkyl is used herein in conjunction with another group, for example, (cycloalkyl)Co-C2 alkyl, the indicated group, in this case cycloalkyl, is either directly bound by a single covalent bond (Co), or attached by an alkyl chain having the specified number of carbon atoms, in this case from 1 to about 2 carbon atoms. Examples of alkyl include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, n-pentyl, and sec-pentyl. Ci-C6alkyl includes alkyl groups having 1, 2, 3, 4, 5, or 6 carbon atoms.

[0032] "Alkoxy" is an alkyl group as defined above with the indicated number of carbon atoms attached through an oxygen bridge. Examples of alkoxy include, but are not limited to, methoxy, ethoxy, n-propoxy, i-propoxy, n-butoxy, 2-butoxy, t-butoxy, n-pentoxy, 2-pentoxy, 3-pentoxy, isopentoxy, neopentoxy, n-hexoxy, 2-hexoxy, 3-hexoxy, and 3- methylpentoxy.

[0033] "Alkanoyl" is an alkyl group as defined above with the indicated number of carbon atoms covalently bound to the group it substitutes through a carbonyl (C=0) bridge. The carbonyl carbon is included in the number of carbons, that is Oalkanoyl is a CH 3 (C=0)- group. [0034] "Alkylester" is an alkyl group as defined herein covalently bound to the group it substitutes by an ester linkage. The ester linkage may be in either orientation, e.g., a group of the formula -0(C=0)alkyl or a group of the formula -(C=0)Oalkyl.

[0035] "Aryl" indicates aromatic groups containing only carbon in the aromatic ring or rings. Typical aryl groups contain 1 to 3 separate, fused, or pendant rings and from 6 to about 18 ring atoms, without heteroatoms as ring members. When indicated, such aryl groups may be further substituted with carbon or non-carbon atoms or groups. Aryl groups include, for example, phenyl, naphthyl, including 1- naphthyl, 2-naphthyl, and bi-phenyl.

[0036] "Mono- and/ or di-alkylamino" are secondary or tertiary alkyl amino groups, wherein the alkyl groups are as defined above and have the indicated number of carbon atoms. The point of attachment of the alkylamino group is on the nitrogen. Examples of mono- and di-alkylamino groups include ethylamino, dimethylamino, and methyl-propyl- amino. A "(mono- and/or di-alkylamino)Co-C2alkyl group is a mono and/or dialkylamino group as defined that is directly bound to the group it substitutes (Coalkyl) or attached to the group it substitutes via a 1 to 2 carbon alkyl group linker.

[0037] "Cycloalkyl" is a saturated hydrocarbon ring groups, having the specified number of carbon atoms, usually from 3 to 7 carbon atoms. Examples of cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl as well as bridged or caged saturated ring groups such as norborane or adamantane. In the term "(cycloalkyl)alkyl," cycloalkyl and alkyl are as defined above, and the point of attachment in on the alkyl group.

[0038] "Halo" or "halogen" as used herein refers to fluoro, chloro, bromo, or iodo.

[0039] "Heteroaryl" is a stable monocyclic aromatic ring having the indicated number of ring atoms which contains from 1 to 3, or in some embodiments from 1 to 2, heteroatoms chosen from N, O, and S, with remaining ring atoms being carbon, or a stable bicyclic or tricyclic system containing at least one 5- to 7-membered aromatic ring which contains from 1 to 3, or in some embodiments from 1 to 2, heteroatoms chosen from N, O, and S, with remaining ring atoms being carbon. Monocyclic heteroaryl groups typically have from 5 to 7 ring atoms. In some embodiments bicyclic heteroaryl groups are 9- to 10-membered heteroaryl groups, that is, groups containing 9 or 10 ring atoms in which one 5- to 7-member aromatic ring is fused to a second aromatic or non-aromatic ring. When the total number of S and O atoms in the heteroaryl group exceeds 1, these heteroatoms are not adjacent to one another. It is preferred that the total number of S and O atoms in the heteroaryl group is not more than 2. It is particularly preferred that the total number of S and O atoms in the aromatic heterocycle is not more than 1. Examples of heteroaryl groups include, but are not limited to, oxazolyl, pyranyl, pyrazinyl, pyrazolopyrimidinyl, pyrazolyl, pyridizinyl, pyridyl, pyrimidinyl, pyrrolyl, quinolinyl, tetrazolyl, thiazolyl, thienylpyrazolyl, thiophenyl, triazolyl, benzo[<JJ oxazolyl, benzofuranyl, benzothiazolyl, benzothiophenyl, benzoxadiazolyl, dihydrobenzodioxynyl, furanyl, imidazolyl, indolyl, and isoxazolyl.

[0040] A "hydrocarbyl" group is a hydrocarbon chain having the specified number of carbon atoms in which carbon atoms are joined by single, double or triple bonds, and any one carbon atom can be replaced by O, NH, or N(Ci-C 4 alkyl).

[0041] "Pharmaceutical compositions" are compositions comprising at least one active agent, such as a compound or salt of Formula I and at least one other excipient.

"Carriers" are any inactive materials, including excipients and diluents, which may be added to the pharmaceutical compositions including carriers and diluents. Pharmaceutical compositions meet the U.S. FDA's GMP (good manufacturing practice) standards for human or non-human drugs.

[0042] "Pharmaceutically acceptable salts" includes derivatives of the disclosed compounds wherein the parent compound is modified by making non-toxic acid or base salts thereof, and further refers to pharmaceutically acceptable hydrates or solvates of such compounds and such salts. Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like. The pharmaceutically acceptable salts include the conventional non-toxic salts and the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. For example, conventional non-toxic acid salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like; and the salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxylmaleic, phenylacetic, glutamic, benzoic, salicylic, mesylic, esylic, besylic, sulfanilic, 2-acetoxybenzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, HOOC-(CH2) n -COOH where n is 0-4, and the like.

[0043] The term "carrier" applied to pharmaceutical compositions/ combinations of the invention refers to a diluent, excipient, or vehicle with which an active compound is provided.

[0044] A "patient" is a human or non-human animal in need of medical treatment. In some embodiments the patient is a human patient.

[0045] The term "therapeutically effective amount" of a compound of Formula I, or a related formula, means an amount effective, when administered to a patient, to provide a therapeutic benefit such as an amelioration of symptoms, e.g., an amount effective to decrease the symptoms of a central nervous system disorder, and including an amount sufficient to reduce the symptoms of Parkinson's disease, restless legs syndrome, bipolar disorder, hyperprolactinemia, depression, Huntington's chorea, Alzheimer's disease, or the cravings associated with substance abuse. Thus a therapeutically effective amount of a compound is also an amount sufficient to significantly reduce the indicia of the disease or condition being treated. A significant reduction is any detectable negative change that is statistically significant in a standard parametric test of statistical significance, such as Student's t-test, in which p < 0.05.

CHEMICAL DESCRIPTION

[0046] This disclosure provides compounds of Formula I, certain of which exhibit high selectivity for functionally activating the D 3 DAR. In one embodiment, certain compounds of Formula I promote β-arrestin translocation to the D 3 DAR with an EC50 of less than 200 nM and in some embodiments less than 50 nM and can have an efficacy equal to that of dopamine. Certain compounds of Formula I that promote β-arrestin translocation to the D 3 DAR with an EC 50 of less than 200 nM also exhibit minimal effects on D 2 DAR- mediated β-arrestin translocation. Certain compounds of Formula I also lack agonist activity at the D 2 DAR as assessed using a [ 35 S]GTPyS binding assay. Certain compounds of Formula I also exhibit potent agonist activity in D 3 DAR G protein-mediated signaling responses as demonstrated using a Go-BRET assay and ERK1/2 phosphorylation assay. Certain compounds of Formula I have no functional activity at other DAR subtypes except for minimal D 2 DAR inhibition at concentrations over 10 μΜ. Using the β-arrestin translocation assay as an exemplary functional output to screen 168 different G protein- coupled receptors (GPCRs), certain compounds of Formula I exhibit extreme selectivity for activating the D 3 DAR with minimal to no activation of other receptors. Certain compounds of Formula I also exhibit extreme selectivity for the D 3 DAR as assessed in a panel of radioligand binding assays. Certain compounds of Formula I do not compete for orthosteric radioligand binding to the D 3 DAR at concentrations that produce maximal functional stimulation. Without wishing to be bound to any particular theory, these radioligand binding assays suggest that compounds of Formula I exhibit high selectivity and efficacy due to their interaction with the D 3 DAR in a highly unique fashion. Molecular modeling studies and site-directed mutagenesis suggest certain compounds of Formula I bind to the D 3 DAR in a manner distict from the D 2 DAR. Using a neuronal cell culture assay to assess neuroprotective properties against a toxin of monoaminergic neurons, compounds of Formula I exhibit neuroprotection with an efficacy greater than that of pramipexole, a much less selective agonist of the D 3 DAR.

[0047] In addition to compounds and salts of Formula I disclosed in the SUMMARY section, ompounds and salts of Formula I (I).

in which the variables may carry any of the values set forth below. The disclosure includes any combination of the variable definitions so long as a stable compound that is within the scope of Formula (I) results.

[0048] (1) R 3 and R 4 are both hydrogen.

[0049] (2) One of R 3 and R 4 is methyl and the other is hydrogen.

[0050] (3) Formula I- A (I-A)

[0051] R 2 is a 6,5-bicyclic heteroaryl group, having 1, 2, 3, 4, 5, or 6 heteroatoms independently chosen from N, O, and S, wherein the point of attachment in Formula I is in the 5-membered ring, and the 5-membered ring contains at least one heteroatom; where substituted with 0 or 1 or more substituents independently chosen from halogen, hydroxyl, cyano, nitro, oxo, -CONH 2 , amino, mono- and di-Ci-C 4 alkylcarboxamide, (C 3 - C6cycloalkyl)Co-C 2 alkyl, and Ci-C6hydrocarbyl, which Ci-C6hydrocarbyl group is a hydrocarbon chain in which the carbon atoms are joined by single, double or triple bonds, and any one carbon atom can be replaced by O, NH, or N(Ci-C 4 alkyl) and which hydrocarbyl group is optionally substituted with one or more substituents independently chosen from hydroxyl, oxo, halogen, and amino.

[0052] (4) R 2 is a 6,5-bicyclic heteroaryl group selected from the following optionally substituted 6,5-bicyclic heteroaryl groups:

[0053] (5) R 2 is a phenyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, or pyrolyl group, each of which is optionally substituted.

[0054] (6) R 2 is a 6,5-bicyclic heteroaryl group substituted with

0 or 1 or more substituents independently chosen from halogen, hydroxyl, cyano, nitro, -CONH2, amino, Ci-C6alkyl, Ci-C6alkoxy, C2-C 4 alkanoyl, Ci-C 4 alkylester mono- and di-Ci- C 4 alkylamino, mono- and di-Ci-C 4 alkylcarboxamide, (C3-C6cycloalkyl)Co-C2alkyl, and any one carbon atom in a Ci-C6alkyl or Ci-C6alkoxy can be replaced by O, NH, or N(Ci-C 4 alkyl) and which Ci-C6alkyl or Ci-C6alkoxy is optionally substituted with one or more substituents independently chosen from hydroxyl, halogen, and amino.

[0055] (7) R 2 i is which is substituted with 0 or 1 or more substituents independently chosen from halogen, hydroxyl, cyano, nitro, -CONH2, amino, Ci-C6alkyl, Ci- C6alkoxy, C2-C 4 alkanoyl, Ci-C 4 alkylester mono- and di-Ci-C 4 alkylamino, mono- and di-Ci- C 4 alkylcarboxamide, (C3-C6cycloalkyl)Co-C2alkyl, and any one carbon atom in a Ci-C6alkyl or Ci-C6alkoxy can be replaced by O, NH, or N(Ci-C 4 alkyl) and which Ci-C6alkyl or Ci- C6alkoxy is optionally substituted with one or more substituents independently chosen from hydroxyl, oxo, halogen, and amino.

[0056] (8) R 2 is a 6,5-bicyclic heteroaryl group substituted with 0 or 1 to 3 substituents independently selected from halogen, Ci-C2alkyl, and Ci-C2alkoxy.

[0057] (9) W is S.

[0058] (10) W is O.

[0059] (11) R 1 is phenyl or a monocyclic heteroaryl group having 1, 2, 3 or 4 heteroatoms independently chosen from N, O, and S, each of which R 1 is optionally substituted. [0060] (12) R 1 is phenyl or pyridyl, each of which is optionally substituted.

[0061] (13) R 1 is a phenyl or pyridyl, each of which is fused to a 5-membered heterocyclic ring, each of which R 1 is optionally substituted.

[0062] (14) R 1 is optionally substituted indole.

[0063] (15) The composition of any one of embodiments 1 to 11, wherein

[0064] R 1 is substituted with 0 or 1 or more substituents independently chosen from halogen, hydroxyl, cyano, nitro, -CONH2, amino, Ci-C6alkyl, Ci-C6alkoxy, C 2 -C 4 alkanoyl, Ci-C 4 alkylester mono- and di-Ci-C 4 alkylamino, mono- and di-Ci-C 4 alkylcarboxamide, (C3- C6cycloalkyl)Co-C 2 alkyl, and any one carbon atom in a Ci-C6alkyl or Ci-C6alkoxy can be replaced by O, NH, or N(Ci-C 4 alkyl) and which Ci-C6alkyl or Ci-C6alkoxy is optionally substituted with one or more substituents independently chosen from hydroxyl, halogen, and amino.

[0065] (16) R 1 is substituted with 0 or 1 to 3 substituents independently chosen from halogen, Ci-C 2 alkyl, and Ci-C 2 alkoxy.

[0066] (17) R 1 is 3-pyridyl.

[0067] (18) R 1 is 4-methoxy-phenyl.

PHARMACEUTICAL PREPARATIONS

[0068] Compounds disclosed herein can be administered as the neat chemical, but are preferably administered as a pharmaceutical composition. Accordingly, the disclosure provides pharmaceutical compositions comprising a compound or pharmaceutically acceptable salt of Formula I, together with at least one pharmaceutically acceptable carrier. The pharmaceutical composition/ combination may contain a compound or salt of Formula I as the only active agent or may be combined with one or more additional active agents. In certain embodiments the pharmaceutical composition is in a dosage form that contains from about 0.1 mg to about 2000 mg, from about 10 mg to about 1000 mg, from about 100 mg to about 800 mg, or from about 200 mg to about 600 mg of a compound of Formula I.

[0069] Compounds disclosed herein may be administered orally, topically, parenterally, by inhalation or spray, sublingually, transdermally, via buccal administration, or by other means routine in the art for administering pharmaceutical compositions. The pharmaceutical composition may be formulated as any pharmaceutically useful form, e.g., as an aerosol, a cream, a gel, a pill, a capsule, a tablet, a syrup, a transdermal patch, or an ophthalmic solution. Some dosage forms, such as tablets and capsules, are subdivided into suitably sized unit doses containing appropriate quantities of the active components, e.g., an effective amount to achieve the desired purpose.

[0070] Carriers include excipients and diluents and must be of sufficiently high purity and sufficiently low toxicity to render them suitable for administration to the patient being treated. The carrier can be inert or it can possess pharmaceutical benefits of its own. The amount of carrier employed in conjunction with the compound is sufficient to provide a practical quantity of material for administration per unit dose of the compound.

[0071] Classes of carriers include, but are not limited to binders, buffering agents, coloring agents, diluents, disintegrants, emulsifiers, flavorants, glidants, lubricants, preservatives, stabilizers, surfactants, tableting agents, and wetting agents. Some carriers may be listed in more than one class, for example vegetable oil may be used as a lubricant in some formulations and a diluent in others. Exemplary pharmaceutically acceptable carriers include sugars, starches, celluloses, powdered tragacanth, malt, gelatin; talc, and vegetable oils. Optional active agents may be included in a pharmaceutical composition, which do not substantially interfere with the activity of the compound of the present invention.

[0072] The pharmaceutical compositions/combinations can be formulated for oral administration. These compositions contain between 0.1 and 99 weight % (wt.%) of a compound of Formula I and usually at least about 5 wt.% of a compound of Formula I. Some embodiments contain from about 25 wt.% to about 50 wt. % or from about 5 wt.% to about 75 wt.% of the compound of Formula I.

METHODS OF TREATMENT

[0073] The disclosure provides methods of treating central nervous system disorders, including Parkinson's disease and related syndromes, dyskinesia, especially dyskinesias secondary to treating Parkinson's disease with L-DOPA, neurodegenerative disorders such as Alzheimer's disease and dementia, Huntington's disease, restless legs syndrome, bipolar disorder and depression, schizophrenia, cognitive dysfunction, or substance use disorders comprising administering an effective amount of a compound of Formula I to a patient having one of these disorders.

[0074] A compound of Formula I may be the only active agent administered

(monotherapy) or may be combined with one or more other active agents (combination, adjunct, or augmentation therapy).

[0075] In another embodiment the invention provides a method of treating depression comprising (i) diagnosing a patient as having depression and (ii) providing an effective amount of compound of Formula I to the patient, wherein the compound of Formula I is provided as the only active agent or is provided together with one or more additional active agents.

[0076] Psychosocial intervention may play an important role in treatment of any central nervous system disorder. Psychosocial intervention includes cognitive-behavior therapy, dialectical-behavior therapy, interpersonal therapy, psychodynamic therapy, and group therapy. In another embodiment, the disclosure provides a method of treating a central nervous system disorder in a patient including administration of an effective amount of a compound of Formula I to the patient, the method further including providing psychosocial intervention to the patient.

[0077] In another embodiment the disclosure provides a method to slow or reverse the progressive loss of neurons seen in Parkinson's disease, Alzheimer's disease, or other disorders involving neurodegeneration, by providing an effective amount of compound of Formula I to the patient, wherein the compound of Formula I is provided as the only active agent or is provided together with one or more additional active agents. For example an effective amount of a compound of Formula I is an amount sufficient to decrease depression symptoms or Parkinson's disease symptoms. Preferably the decrease in depression symptoms or Parkinson's disease symptoms is a 50% or greater reduction of symptoms identified on symptom rating scale for these disorders. For example an effective amount may be an amount sufficient to decrease the patient's score on a psychiatric symptoms rating scale such as the Brief Psychiatric Rating Scale, the Clinical Global Impression, or the Positive and Negative Syndrome Scale or the Movement Disorder Society Unified Parkinson's Disease Rating Scale (MDS-UPDRS).

EXAMPLES

GENERAL METHODS FOR CHEMISTRY

[0078] All reagents were used as received from the following suppliers: Alfa Aesar, Ark Pharm, Aldrich, and Fisher Scientific. Acetonitrile and THF were purified using the Innovative Technology PureSolv solvent purification system. The l H and 13 C NMR spectra were recorded on either a Bruker Avance 400 MHz or 500 MHz spectrometer. Chemical shifts are reported in parts per million and were referenced to residual proton solvent signals. When indicated, 13 C multiplicities were determined with the aid of an APT pulse sequence, differentiating the signals for methyl and methane carbons as "d" from methylene and quarternary carbons as "u". [0079] The infrared (IR) spectra were acquired as thin on a PerkinElmer Spectrum One FT-IR spectrometer equipped with a universal ATR sampling accessory and the absorption frequencies are reported in cm "1 .

[0080] Flash column chromatography separations were performed using the Teledyne Isco CombiFlash R F using RediSep R F silica gel columns. TLC was performed on Analtech UNIPLATE silica gel GHLF plates (gypsum inorganic hard layer with fluorescence). TLC plates were visualized under a long wave/short wave UV lamp.

[0081] Automated preparative RP HPLC purification was performed using an Agilent 1200 Mass-Directed Fractionation system (Prep Pump G1361 with gradient extension, makeup pump G1311A, pH modification pump G1311A, HTS PAL autosampler, UV-DAD detection G1315D, fraction collector G1364B, and Agilent 6120 quadrapole spectrometer G6120A).

[0082] The preparative chromatography conditions included a Waters X-Bridge C 18 column (19 x 150 mm, 5 μιη, with 19 x 10-mm guard column), elution with a water and acetonitrile gradient, which increases 20% in acetonitrile content over 4 min at a flow rate of 20 mL/min (modified to pH 9.8 through addition of NH 4 OH by auxiliary pump), and sample dilution in DMSO. The preparative gradient, triggering thresholds, and UV wavelength were selected according to the analytical RP HPLC analysis of each crude sample. The analytical method used an Agilent 1200 RRLC system with UV detection (Agilent 1200 DAD SL) and mass detection (Agilent 6224 TOF). The analytical method conditions included a Waters Aquity BEH Cis column (2.1 x 50 mm, 1.7 μιη) and elution with a linear gradient of 5% acetonitrile in pH 9.8 buffered aqueous ammonium formate to 100% acetonitrile at 0.4 mL/min flow rate.

[0083] Compound purity was measured on the basis of peak integration (area under the curve) from UV/Vis absorbance (at 214 nm), and compound identity was determined on the basis of mass analysis. All compounds used for biological studies have purity >90%.

EXAMPLE 1. SYNTHESIS OF (1H-INDOL-2-YL)(4-(2-(4-METHOXYPHENOXY)ETHYL)PIPERAZIN- 1-YL)METHANONE (Compound 1)

[0085] Compound 1 was prepared according to the method shown in Scheme I. Reagents and conditions: a) K2CO3, potassium iodide, MeCN, 70 °C; b) CF3CO2H, Et 3 SiH, CH2CI2, rt; c) PyBop, i-Pr 2 EtN, indole-2-carboxylic acid, DMF, rt.

[0086] Step 1. Synthesis of ie/t-Butyl 4-(2-(4-methoxyphenoxy)ethyl)piperazine-l- carboxylate (Reaction a). A mixture of ieri-butyl piperazine-l-carboxylate (490 mg, 2.63 mmol), l-(2-bromoethoxy)-4-methoxybenzene (608 mg, 2.63 mmol), potassium carbonate (727 mg, 5.26 mmol) and potassium iodide (44 mg, 0.263 mmol) in acetonitrile (35 mL) was heated at 70 °C for 16 h. The reaction was cooled to rt, filtered and the solids washed with acetonitrile (2 x 15 mL). The combined organics were adsorbed onto celite and purified by silica chromatography to afford the ether product as a colorless oil (622 mg, 1.85 mmol, 70% yield). R = 0.57 (EtOAc); l H NMR (400 MHz, CDC1 3 ) δ 1.46 (s, 9 H), 2.51 (t, = 4.8 Hz, 4 H), 2.79 (t, J = 6.0 Hz, 2 H), 3.45 (t, J = 4.8 Hz, 4 H), 3.76 (s, 3 H), 4.05 (t, J = 6.0 Hz, 2 H), 6.83 (d, J = 2.4 Hz, 4 H); 13 C NMR (101 MHz, APT pulse sequence, CDC1 ) δ d 28.5, 55.8, 114.7, 115.7; u 53.5, 57.5, 66.7, 79.6, 153.0, 154.1, 154.8; HRMS (m z): calcd. for

C18H29N2C [M + H] + 337.2122; found 337.2122; HPLC purity: 99.0%.

[0087] Step 2. l-(2-(4-Methoxyphenoxy)ethyl)piperazine. To a solution of tert-butyl 4-(2-(4-methoxyphenoxy)ethyl)piperazine- l-carboxylate (185 mg, 0.55 mmol) and triethylsilane (96 mg, 0.825 mmol) in CH2CI2 (5 mL) was added trifluoroacetic acid (0.85 mL, 1,254 mg, 11.00 mmol). The reaction was stirred at rt for 4 h and concentrated under vacuum. The residue was partitioned between aqueous sodium bicarbonate (10 mL) and CH2CI2 (3 x 10 mL). The combined organic layers were concentrated under vacuum to afford the piperazine product as a colorless oil (118 mg, 0.499 mmol, 91% yield). X H NMR (400 MHz, CDC1 ) δ 2.54-2.60 (m, 4 H), 2.78 (t, J = 6.0 Hz, 2 H), 2.94 (t, J = 4.8 Hz, 4 H), 3.76 (s, 3 H), 4.05 (t, J = 6.0 Hz, 2 H), 6.83 (d, J = 2.8 Hz, 4 H); 13 C NMR (101 MHz, APT pulse sequence, CDCb) δ d 55.8, 114.8, 115.7; u 45.9, 54.6, 58.0, 66.6, 153.0, 154.0; HRMS (m/z): calcd. for C13H21N2O2 [M + H] + 237.1598; found 237.1573; HPLC purity: 94.1%.

[0088] Step 3. (lH-Indol-2-yl)(4-(2-(4-methoxyphenoxy)ethyl)piperazin-l- yl)methanone (Compound 1). To a mixture of l-(2-(4-methoxyphenoxy)ethyl)piperazine (146 mg, 0.618 mmol), indole-2-carboxylic acid (119 mg, 0.741 mmol) and DMAP (8 mg, 0.062 mmol) in THF (10 mL) was added diisopropylcarbodiimide (0.29 mL, 234 mg, 1.85 mmol). The reaction was stirred at rt for 15 h and the solvents removed under vacuum. The residue was purified via silica gel chromatography with CH 2 Cl2/(MeOH containing 2% NH 4 OH) to afford the title compound as an off-white solid. Mp = 163-165 °C; R = 0.54 (MeOH (10%) and NH 4 OH (2%) in CH2CI2); *H NMR (500 MHz, CDCb) δ 2.68 (t, J = 5.0 Hz, 4 H), 2.85 (t, = 5.5 Hz, 2 H), 3.77 (s, 3 H), 3.91-4.06 (m, 4 H), 4.10 (t, = 5.5 Hz, 2 H), 6.78 (s, 1 H), 6.83-6.87 (m, 3 H), 7.12-7.15 (m, 1 H), 7.26-7.30 (m, 2 H), 7.43 (d, J = 8.5 Hz, 1 H), 7.65 (d, / = 8.0 Hz, 1 H), 9.25 (br s, 1 H); 13 C NMR (125 MHz, CDCb) δ 41.0, 53.6, 55.7, 57.3, 66.5, 105.2, 111.7, 114.6, 115.6, 120.6, 121.8, 124.4, 127.4, 129.2, 135.5, 152.8, 154.0, 162.2; HRMS (m/z): calcd. for C22H26N3O3 [M + H] + 380.1969; found

380.1995; HPLC purity: 95.6%; FTIR (neat): 3258, 1597, 1506, 1437 cm 1 .

EXAMPLE 2. ADDITIONAL DOPAMINE D3 RECEPTOR AGONISTS

[0089] The compounds of Formula I shown in Table 1 were prepared by the methods shown in Example 1 for the preparation of Compound 1.

Table 1. Additional Compounds









[0090] The following compounds are also made by the methods used to prepare Compound 1. Those of skill in the art will recognize the routine changes to the procedure for preparing Compound 1 needed to produce the following compounds. Starting material for certain of the compounds in the "additional compounds" table can also be made by the following general schemes. For example starting material for compound 118 can be made by Scheme A and starting material for compounds 120-121 can be made by Scheme B.

Scheme A

121 0

122 0

123

Η 0

124

H 0

125

Analytical Data for select compounds

(4-(2-(pyridin-3-yloxy)ethyl)piperazin-l-yl)(lH-pyrrolo[2,3- b]pyridin-2-yl)methanone (Compound 4).

[0091] l H NMR (500 MHz, CDCb) δ 2.66 - 2.78 (m, 4 H), 2.90 (t, 7 = 5.5 Hz, 2 H), 3.97 (s, 4 H), 4.19 (t, 7 = 5.5 Hz, 2 H), 6.73 (d, 7 = 1.5 Hz, 1 H), 7.14 (dd, 7 = 7.9, 4.7 Hz, 1 H), 7.19 - 7.26 (m, 2 H), 7.99 (dd, 7 = 7.9, 1.4 Hz, 1 H), 8.24 (dd, J = 4.0, 2.0 Hz, 1 H), 8.34 (dd, 7 = 2.4, 1.0 Hz, 1 H), 8.51 (dd, 7 = 4.7, 1.5 Hz, 1 H), 10.81 (s, 1 H). 13 C NMR (125 MHz, CDCb) δ 41.00, 53.62, 57.02, 66.25, 103.40, 116.87, 119.80, 121.27, 123.87, 129.86, 130.36, 137.99, 142.44, 145.98, 147.71, 154.81, 162.04. HRMS (m/z): calcd. for C19H22N5O2 [M + H] + 352.1773; found 352.1769; HPLC purity: 100%; FTIR (neat) : 3055, 2942, 2812, 1615, 1574, 1428, 1225 cm "1 . 6-methoxy-lH-indol-2-yl)(4-(2-(4-methoxyphenoxy)ethyl)pipera zin-l-yl)methanone:

(Compound 32)

[0092] *H NMR (500 MHz, CDC1 3 ) δ 2.63 - 2.72 (m, 4 H), 2.84 (t, J = 5.6 Hz, 2 H), 3.77 (s, 3 H), 3.86 (s, 3 H), 3.93 (d, J = 20.2 Hz, 4 H), 4.09 (t, J = 5.6 Hz, 2 H), 6.72 (dd, J = 2.1, 0.7 Hz, 1 H), 6.77 - 6.90 (m, 6 H), 7.50 (d, J = 8.8 Hz, 1 H), 9.10 (s, 1 H). 13 C NMR (125 MHz, CDCI3) δ 0.00, 53.60, 55.52, 55.73, 57.28, 66.52, 76.76, 77.02, 77.22, 77.27, 93.69, 105.63, 111.91, 114.65, 115.59, 121.81, 122.66, 128.13, 136.51, 152.77, 153.98, 158.22, 162.18; HRMS (m/z): calcd. for C23H28N3O4 [M + H] + 410.2080; found 410.2074; HPLC purity: 99.5%; FTIR (neat): 3265, 2937, 1596, 1505, 1231 cm 1 .

EXAMPLE 3. D3 DAR BETA-ARRESTIN RECRUITMENT ASSAY

[0093] A D3 DAR β-arrestin cell line from DiscoverX (Fremont, CA) was used as described in the protocol below (Table 2) for the screen. A CHO cell line engineered to overexpress the D3 DAR fused with a small 42-amino acid fragment of β-galactosidase called ProLink™ and a fusion protein consisting of β-arrestin and a larger N-terminal deletion mutant of β-galactosidase was used (DiscoverX catalogue number 93-0579C2). When the D3 DAR is activated by dopamine, it stimulates binding of β-arrestin to ProLink- tagged D3 DARs, and the two complementary parts of β-galactosidase form a functional enzyme. When substrate (PathHunter® Detection reagent, DiscoverX 93-0001) is added, β- galactosidase hydrolyzes it and generates a chemiluminescent signal.

EXAMPLE 4. D 2 DAR BETA-ARRESTIN RECRUITMENT ASSAY

[0094] For a secondary- screen and selectivity assays, DAR PathHunter® β-arrestin GPCR cell lines from DiscoverX (Fremont, CA) were used. In the D 2 Receptor PathHunter® β-arrestin GPCR cell line, the D 2 DAR is overexpressed and fused with a small 42-amino acid fragment of β-galactosidase called ProLink™ on a CHO cellular background expressing a fusion protein of β-arrestin and a larger N-terminal deletion mutant of β -galactosidase ("enzyme acceptor"). When DAR is activated by dopamine, it stimulates binding of β- arrestin to the ProLink-tagged DAR and the two complementary parts of β-galactosidase form a functional enzyme. When substrate (PathHunter™ Detection reagent) is added, β- galactosidase hydrolyzes it and generates a chemiluminescent signal. Table 3 summarizes the protocol for the D 2 DAR β-arrestin recruitment assay.

[0095] Table 4 shows the D 3 agonist, D 2 agonist, and D 2 antagonist activity of compounds 1-32 in the beta-arrestin recruitment assays of Examples 3-4. Table 4. Compound Activity

Cmpd. No. D3 EC50 D 3 Emax D 2 EC50 D 2 Emax D 2 IC50 D 2 Imax

(nM) (nM) (nM)

1 36 106 >10,000 Curve >50000 Curve

Interrupted Interrupted

2 9.2 83 1800 49 535 37

3 11.5 95 1000 75 None None

4 13.5 99 210 99 None None

5 33 112 2400 61 None None

6 13.3 102 617 34 2100 60

7 45 98 4100 71 None None

8 81 117 780 66 None None

9 285 120 660 29 2000 53

10 42 105 1700 57 None None

11 50 129 128 76 None None

12 63 108 1800 61 None None

13 5.0 79 790 56 None None

14 Curve

33.8 105 2300 48 >100000

interrupted

15 Curve

126 90 >50000 9700 72

interrupted

16 60.9 84 None None 440 100

17 3.0 96 183 89 None None

18 4.4 107 217 82 None None

19 98 119 1400 90 None None

20 4.3 106 138 74 None None

21 33 127 617 74 None None

22 9.5 94 4400 91 None None

23 6.4 107 364 81 None None

24 6.9 97 180 70 None None

25 13 113 1300 101 None None

26 25.7 108 786 81 None None

27 37.6 112 6200 53 None None

28 59 97 2600 97 None None

29 284 110 1700 67 None None Table 4. Compound Activity

Cmpd. No. D3 EC50 D 3 Emax D 2 EC50 D 2 Emax D 2 IC50 D 2 Imax

(nM) (nM) (nM)

30 266 106 4300 37 744 30

31 208 112 2500 19 4600 64

32 155 91 5200 62 None None

33 710 104 none none 14,000 100

34 278 36 none none 9,000 99

35 curve

2,600 44 none none > 50,000

interrupted

36 1,000 103 none none > 10,000 101

37 none none none none > 50,000 100

38 curve

3,500 50 none none

> 100,000 interrupted

39 310 78 none none > 10,000 87

40 curve

17 123 2,900 96 > 100,000

interrupted

41 548 70 none none > 50,000 100

42 curve

2,500 121 none none > 100,000

interrupted

43 22,000 50 none none none none

44 curve curve

2,100 97 > 50,000 > 100,000

interrupted interrupted

45 curve

980 115 6,000 56 > 100,000

interrupted

46 curve

520 109 7,800 46 > 100,000

interrupted

47 curve

411 116 5,200 33 > 100,000

interrupted

48 curve

473 103 10,100 49 > 100,000

interrupted

49 curve curve

611 119 > 50,000 > 100,000

interrupted interrupted

50 266 106 4,300 37 744 30

51 curve

563 122 13,000 70 > 100,000

interrupted

52 curve

225 130 3,500 58 > 100,000

interrupted Table 4. Compound Activity

Cmpd. No. D3 EC50 D 3 Emax D 2 EC50 D 2 Emax D 2 IC50 D 2 Imax

(nM) (nM) (nM)

53 2,900 117 none none none none

54 curve

160 122 7,100 57 > 100,000

interrupted

55 617 96 none none 16,000 99

56 167 122 413 94 none none

57 curve

810 105 7,800 21 > 100,000

interrupted

58 curve

576 107 5,500 28 > 100,000

interrupted

59 curve

2,800 103 > 100,000 none none interrupted

60 curve

192 95 none none > 50,000

interrupted

61 3,300 51 none none 5,100 96

62 430 80 none none 7,700 100

63 curve curve

> 50,000 none none > 50,000

interrupted interrupted

64 35 9,700 none none None none

65 curve none none none none > 50,000

interrupted

66 353 104 7,400 54 None none

67 284 110 1,700 67 None none

68 50.0 129 128 76 None none

70 curve

116 102 4,600 52 > 100,000

interrupted

71 6.4 107 364 81 none none

72 59 97 2,600 97 none none

73 25.7 108 786 81 none none

74 4.3 106 138 74 none none

75 13 113 1,300 101 none none

76 6.9 97 180 70 none none

77 33 127 617 74 none none

78 3.0 96 183 89 none none

79 98 119 1,400 90 none none Table 4. Compound Activity

Cmpd. No. D3 EC50 D 3 Emax D 2 EC50 D 2 Emax D 2 IC50 D 2 Imax

(nM) (nM) (nM)

80 4.4 107 217 82 none none

81 37.6 112 6,200 53 none none

82 9.5 94 4,400 91 none none

83 13.5 99 210 99 none none

85 63 108 1,800 61 none none

88 42 104 1,700 57 none none

90 curve

118 110 6,100 34 > 100,000

interrupted

92 curve none none none none > 100,000

interrupted

93 curve

630 89 none none > 100,000

interrupted

94 curve

33.8 105 + 9.3 2,300 48 > 100,000

interrupted

95 curve curve

> 100,000 none none > 100,000

interrupted interrupted

96 curve curve

3,800 100 > 100,000 > 100,000

interrupted interrupted

97 curve curve

151 91 > 50,000 > 100,000

interrupted interrupted

98 curve

1,600 98 none none > 50,000

interrupted

99 curve none none none none > 100,000

interrupted

100 curve 9,700 +

126 90 > 50,000 72 + 4.6 interrupted 5,800

101 curve

114 118 none none > 50,000

interrupted

102 curve

105 106 none none > 100,000

interrupted

103 1,300 92 none none none none

104 curve

710 111 none none > 50,000

interrupted

105 curve curve

78 123 > 50,000 > 50,000

interrupted interrupted

106 8.1 103 252 65 none none Table 4. Compound Activity

Cmpd. No. D3 EC50 D 3 Emax D 2 EC50 D 2 Emax D 2 IC50 D 2 Imax

(nM) (nM) (nM)

107 curve

472 71 none none > 100,000

interrupted

108 5.0 79 790 56 none none

109 60.9 84 none none 440 100

111 curve

9,300 106 none none > 100,000

interrupted

112 none none none none 6,500 97

113 none none none none 6,700 96

114 curve none none none none > 100,000

interrupted

115 none none none none 9,000 107

116 curve

none none > 100,000 none none

interrupted

117 none none none none none none

118 none none none none 18,000 72

119 curve

1,900 45 none none > 100,000

interrupted

120 82 108 3,300 46 none none

121 32 101 1,900 43 none none

122 none none none none 691 97

123 none none none none 1200 97

124 none none none none none none

125 none none none none none none

EXAMPLE 5. D 3 RADIOLIGAND BINDING ASSAY

[0096] Compounds were tested for their ability to compete with the orthosteric radioligand [ 3 H]methylspiperone for binding to the D 3 DAR using stable HEK cell lines expressing the D 3 DAR (Codex Biosciences, Gaithersburg, MD, Catalogue no. CB-80300- 206) as described in the detailed protocol below presented in Table 5. Cells were cultured in Dulbecco's modified Eagle's Medium (Corning, catalogue no. 10-013) containing 10% FBS, 1,000 units/mL Penicillin, 1,000 mg/mL Streptomycin, 100 mM sodium pyruvate, 1 μg/mL Gentamicin, and 250 mg/mL G418. All cells were maintained at 37 °C in 5% C0 2 and 90% humidity. For radioligand binding assays cells were removed mechanically using calcium- free Earle' s balanced salt solution (EBSS). Intact cells were collected by centrifugation and then lysed with 5 mM Tris-HCl and 5 mM MgCk at pH 7.4. Homogenates were centrifuged at 30,000 x g for 30 minutes. The membranes were re-suspended in EBSS (US Biological, catalogue no. E0249-05) pH 7.4 to a concentration of 16 μg/mL. For competition binding studies, membrane preparations were incubated for 90 minutes at room temperature with various concentrations of compound and a single concentration of [ 3 H] methyl spiperone (Perkin Elmer, NET856) in a reaction volume of 250 μί. Non-specific binding was determined in the presence of 4 μΜ (+)-butaclamol (Sigma-Aldrich, catalogue no. D033). Bound ligand was separated from unbound by filtration through GF/C filters using a

PerkinElmer cell harvester and quantified on a Top-count (PerkinElmer). Ki values were determined using Cheng-Prusoff equation from observed IC50 values and ligand Kd values from separate saturation experiments.

EXAMPLE 6. ORTHOGONAL SCREENING ASSAYS

[0097] Orthogonal testing using an unrelated assay of β-arrestin recruitment, as well as assays of G-protein mediated signaling and assays of ERK phosphorylation also show that compound 1 is a potent and fully efficacious D3R-selective agonist. [0098] BRET arrestin recruitment assay. As an orthogonal test using an unrelated assay of D3R-mediated β-arrestin recruitment, an arrestin BRET assay was conducted.

HEK293T cells transiently expressing D2-Rluc8, Arrestin3-mvenus and GRK2, or D3-Rluc8, Arrestin3-mvenus, and GRK3 were harvested with EBSS, plated in 96-well plates in

Dulbecco's phosphate-buffered saline (DPBS) and incubated at rt for 45 min. Cells were incubated with 5 μΜ coelenterazine H (the substrate of Rluc8, Nanolight Technology, Pinetop AZ) for 5 min, then stimulated with the indicated concentrations of either dopamine or a test compound of Formula I for 5 min. BRET signal was determined by quantifying and calculating the ratio of the light emitted by mVenus (525 nm) over that emitted by RLuc8 (485 nm) using a PHERAstar FSX Microplate Reader (BMG Labtech).

[0099] Go BRET activation assay. To determine if a test compound of Formula I displays functional selectivity (the ability to selectively activate one signaling pathway versus another), a G 0 BRET activation assay was utilized as described in the protocol below (Table 6). Briefly ,HEK293T cells transiently expressing either the D2R or D3R and Ga 0 A-Rluc8, untagged-βι, and mVenus-y2 were harvested with EBSS-, plated in 96-well white plates at 20,000 cells/well in DPBS and incubated at RT for 45 min. Cells were incubated with 5 μΜ coelenterazine h (Nanolight Technology, Pinetop, AZ) for 5 min, then stimulated with the indicated concentrations of either dopamine or a test compound of Formula I for 5 min.

BRET signal was determined by quantifying and calculating the ratio of the light emitted by mVenus (525 nm) over that emitted by RLuc8 (485 nm) using a PHERAstar FSX Microplate Reader (BMG Labtech).

[0100] ERK1/2 Phosphorylation assay. ERK1/2 phosphorylation was measured using the Alphascreen SureFire Ultra ERK kit (PerkinElmer, Waltham, USA). CHO-K1 DiscoverX cells stably expressing either the D2R or D3R were seeded into 384-well small volume white plates at a density of 40,000 cells/well in serum-free Ham's F12 media overnight. Cells were stimulated with the indicated concentration of test compound for 15 min, followed by cell lysis as specified by manufacture's protocol. The plate was shaken for 10 min at RT, followed by the addition of Surefire activation buffer, Surefire reaction buffer, Alphascreen acceptor beads, and Alphascreen donor beads in ratios specified by the manufacturer. The plate was incubated in the dark for 2 h, then read using a PHERAstar FSX Microplate Reader (BMG Labtech).

EXAMPLE 7. [ 35 S]GTPyS binding assay

[0101] A [ 35 S]GTPyS (Perkin Elmer NEG030H) binding assay was utilized as described in the protocol below (Table 7). Briefly, cells stably expressing D 2 DARs (Codex Biosolutions CB-80300-206) were lysed with 1 mM EDTA, 20 mM HEPES, pH 7.4.

Homogenates were centrifuged at 30,000 x g for 30 minutes. The membranes were re- suspended in 20 mM HEPES, 10 mM MgCl 2 , 100 mM NaCl, 3 mM EGTA, 0.2 mM sodium metabisulfite, pH 8.0 containing 50 μΜ GDP and incubated with dopamine or Compound 1 and [ 35 S]GTPyS for 1 hour at 30 °C. Basal binding was determined in the presence of 10 μΜ unlabeled GTPyS and non-specific binding was determined in the presence of 4 μΜ (+)- butaclamol (Sigma-Aldrich D033). Bound ligand was separated from unbound by filtration through GF/C filters using a PerkinElmer cell harvester and quantified on a Top-count (PerkinElmer). Data are expressed as % maximum dopamine response. Compound 1 was found not to activate the D 2 DAR in this assay.

7 Reagent ΙΟ μΙ, [ 35 S]GTP Y S

8 Reagent 60 μΐ. Membrane preparation per assay well

9 Time 1 hour Incubate at 30 °C

10 Filter 4 Filter membranes onto GF/C filter plates

washes

11 Reagent 50 μΐ. Perkin Elmer scintillation cocktail

EXAMPLE 8. DISCOVERX GPCRMAX sm GPCR ASSAY PANEL

[0102] To determine if a test compound of Formula I displays selectivity for the D 3 DAR compared to a large panel of GPCRs, Compound 1 was screened in the DiscoverX gpcrMAX SM GPCR Assay Panel of β-arrestin recruitment as an agonist at a single high dose of 10 μΜ. Methods employed in this study performed at DiscoverX have been adapted from the scientific literature to maximize reliability and reproducibility. Reference standards were run as an integral part of each assay to ensure the validity of the results obtained. Assay results are presented as the mean percent activation of indicated GPCRs (for n = 2 replicates) for Compound 1 tested at a concentration of 10 μΜ. For a full description of the DiscoverX gpcrMAX™GPCR Assay Panel see: http://www.DiscoverX.com. The results are shown numerically in Table 8, and are shown graphically in FIG. 3. In FIG. 3, the grid reference numbers 1-21 and reference letters a-h correspond to the cells of Table 8, with each table cell representing a different GPCR and its recruitment activity in %. Compound 1 shows a high degree of selectivity among the 168 GPCRs tested.

The GPCR is listed in text, the activity in %. Grid reference with number and letter is for reference to FIG. 3.

EXAMPLE 9. INHIBITION BINDING PROFILE

[0103] Compound 1 displays limited liability for off-target effects as shown by its inhibition binding profiles. Compound 1 was evaluated for binding affinity in the

comprehensive panel offered by the Psychoactive Drug Screening Program (PDSP) at the University of North Carolina, Chapel Hill. Experimental conditions will be readily apparent to those of skill in the art. Experimental details, including radioligands used and associated Kd values for each individual target, are listed on the PDSP website http://pdsp.med.unc.edu/. Data represent mean % inhibition (n=4) for each compound binding to various receptor subtypes and ion channels: >50% inhibition is considered significant. The default

concentration for primary binding experiments is 10 μΜ. Ki determinations and full receptor binding profiles were performed for those targets that showed significant inhibition in the primary binding experiments (See Table 9). This screen employs radioligand binding assays for a number of receptors, transporters and some ion channels. FIG. 4A provides the legend of GPCRs tested in this panel. Full numerical results of the panel are shown in FIG. 4 B. The histogram of FIG. 4C of the initial screen results show that Compound 1 displays affinity for only the D3R, 5-HT1A, 5-HT2B, and Sigmal receptor. Since the PDSP binding panel showed affinity of Compound 1 at the potentially clinically problematic 5HT2B receptor, and the DiscoverX functional screen lacked the 5HT2B, we tested Compound 1 as both an agonist and antagonist in an HTRF IP1 accumulation assay using a single high concentration of 10 μΜ using the Eurofins Cerep service. No agonist activity was found, but this dose produced an 80% inhibition of a response elicited by a 30 nM concentration of serotonin (data not shown); indicating that Compound 1 is not an agonist of the 5HT2B, although it may be a low affinity antagonist.

N/A b indicates no significant bindind affinity was found in the primary

N/D c indicates the value was not determined.

[0104] The results of this binding panel show that Compound 1 is a highly selective compound as assessed by the PDSP, cross reacting with relatively few other GPCR orthosteric sites, and even then, only at concentrations higher than needed to fully active the D3R. It is more selective than the clinically used D3R-preferring agonist pramipexole.

EXAMPLE 10. MUTAGENESIS STUDIES

[0105] Molecular modeling studies indicated that compound 1 interacts with the D3R in a unique fashion, and identified 2 residues important for the binding of compound 1 to the D3R. Mutagenesis was conducted on these residues (Y198 and Y365) and arrestin BRET and Go BRET assays were conducted. See FIG. 5. While small effects of dopamine potency were caused by the mutants, large effects on potency and/or efficacy of compound 1 were observed. The Y198A mutation reduced the signaling efficacy of compound 1 to 36% that of dopamine in the arrestin BRET assay, and to 64% that of dopamine in the Go BRET assay, while displaying minimal effects on the potency of compound 1. The Y365A mutation had more dramatic effects. In the arrestin assay, the efficacy of compound 1 was reduced to 21% that of dopamine, and the potency of compound 1 was reduced 440-fold. In the Go BRET assay, the efficacy of compound 1 appeared to be unaffected, but the potency was reduced 25,000-fold. The results indicate compound 1 interacts with the D3R in a unique fashion compared to the endogenous agonist dopamine, which could account for its striking D3R selectivity.

EXAMPLE 11. NEUROPROTECTION ASSAY

[0106] Because prior studies have found that partially selective D 3 DAR preferring agonists, such as pramipexole, elicit neuroprotective effects, we sought to examine the potential of a test compound of Formula I to show similar activity in a model of

neurodegeneration. SHSY5Y cells were terminally differentiated for 1 week using 10 μΜ retinoic acid and 80 nM phorbol 12-myristate 13-acetate, after which they display a neuronal phenotype including the expression of D 3 dopamine receptors. Cells were pretreated with either pramipexole or Compound 1 for 24 h followed by 0 μΜ treatment with the neurotoxin 6-hydroxydopamine to induce neuronal insult. Protection against total neurite length decrease was measured 24 h later. For dose-response assays, terminally differentiated SHSY5Y cells were pretreated with Compound 1 for 24 h followed by 10 μΜ treatment with 6-hydroxydopamine, and protection against total neurite length decrease was measured 24h later. The results are shown in FIG. 6A and FIG. 6B. As shown by FIG. 6 A, Compound 1 demonstrates neuroprotection superior to pramipexole under the assay conditions. As shown by FIG. 6B, the neuroprotection provided by Compound 1 shows clear concentration dependent effect.

EXAMPLE 12. AMES TESTING

[0107] The mutagenicity potential of Compound 1 was tested using the Ames reverse mutation assay. Briefly, 10 million bacteria were exposed in triplicate to Compound 1 for 90 min in medium containing a low concentration of histidine. The cultures were then diluted into an indicator medium lacking histidine, dispensed into a 384-well plate, and incubated for 48 h at 37 °C. Cells that have undergone a reversion will grow, resulting in a color change. The number of wells showing growth were counted and compared to the vehicle control. An increase in the number of colonies of at least 2-fold over baseline and a dose response indicated a positive response. Data were analyzed using an unpaired, one-sided Student's T- test. Where indicated, S9 fraction from the livers of Aroclor 1254-treated rats were included in the incubation at a 4.5% final concentration. An NADPH-regenerating system was also included to ensure a steady supply of reducing equivalents. The test results are shown in Table 10.

EXAMPLE 13. CYTOTOXICITY SCREENING PANEL

[0108] HepG2 cells were plated on 384-well tissue culture treated black walled clear bottomed polystyrene plates. The cells were dosed with test compound at a range of concentrations. At the end of the incubation period, the cells were loaded with the relevant dye/antibody for each cell health marker. The plates were then scanned using an automated fluorescent cellular imager, ArrayScan® (Thermo Scientific Cellomics). Cytotoxicity was assessed using a multi-parametric approach using High Content Screening (HCS). The following parameters were screened. [0109] Nuclear size: An increase in nuclear area can indicate necrosis or G2 cell cycle arrest and a decrease can indicate apoptosis.

[0110] DNA structure: An increase in DNA structure can indicate chromosomal instability and DNA fragmentation.

[0111] Cell membrane permeability: An increase in cell membrane permeability is a general indicator of cell death.

[0112] Mitochondrial mass: A decrease in mitochondrial mass indicates loss of total mitochondria and an increase implies mitochondrial swelling or an adaptive response to cellular energy demands.

[0113] Mitochondrial membrane potential (Δψπι): A decrease indicates a loss of mitochondrial membrane potential and mitochondrial toxicity, as well as a potential role in apoptosis signaling, an increase in mitochondrial membrane potential indicates an adaptive response to cellular energy demands.

[0114] Cytochrome c: An increase in cytochrome c release is one of the hallmarks of the apoptosis cascade.

[0115] The results of the cytotoxicity screen are shown in Table 11. All cell health marker assays showed minimal cytoxicity liability of Compound 1, as the AC 50 was greater than 50 μΜ in each assay.

MEC is the Minimum effective concentration thai significantly crosses vehicle control threshold. ACSO is the ccnceniratiof! at which 50% maximum effect is observed for each health parameter.

EXAMPLE 14. PLASMA AND BRAIN TISSUE SAMPLING [0116] The levels of Compound 1 in mouse plasma and brain tissue samples were assessed as follows.

[0117] A single IP dose (20 mg/kg) of Compound 1 was administered to C57BL/6 mice. The formulation consisted of 10% dimethylacetamide (DMA) and 60% PEG400, balanced with 30% saline. Plasma and brain samples were collected across 8 time points (5, 15, 30, 60, 120, 240, 480, and 1440 minutes). Brain samples were homogenized in two volumes (1:2 w/v dilution) of phosphate buffer solution (PBS). Once homogenized, the samples were crashed with three volumes of acetonitrile containing an analytical internal standard (bucetin). Samples were then centrifuged to remove precipitated protein, and the supernatant was analyzed by LC-MS/MS. All brain samples were compared to a calibration curve prepared in mouse blank brain. Plasma samples (5, 15, 30, 60 and 120 min time points) were diluted ten-fold with blank plasma. No dilutions were made for 240, 480 and 1440 min time points plasma samples. All plasma samples were crashed with three volumes of acetonitrile containing an analytical internal standard (bucetin). Samples were then centrifuged to remove precipitated protein, and the supernatant was analyzed by LC-MS/MS. All plasma samples were compared to a calibration curve prepared in mouse blank plasma. Samples were analyzed by LC-MS/MS using Waters Xevo TQ mass spectrometer coupled with an Acquity UPLC and a CTC PAL chilled autosampler, all controlled by MassLynx software (Waters). After separation on a C18 reverse phase HPLC column (Waters Acquity HSS T3 2.1x50mm 1.8 μΜ) using an acetonitrile-water gradient system, peaks were analyzed by mass spectrometry (MS) using ESI ionization in MRM mode. Compound 1 levels in plasma and brain as a function of time are shown in FIG. 7.