4-PHENYLPIPERAZINE DERIVATIVES WITH FUNCTIONALIZED LINKERS AS DOPAMINE D3 RECEPTOR SELECTIVE LIGANDS AND METHODS OF USE

Dopamine D3 receptor antagonists and partial agonists are known to modulate the reinforcing and drug-seeking effects induced by cocaine and other abused substances. By introducing functionality into the butylamide linking chain of the 4-phenylpiperazine class of ligands, improved D3 receptor affinity and selectivity, as well as water solubility, is achieved. A series of linking-chain derivatives are disclosed wherein functionality such as OH, OAc, and cis or trans-cyclopropyl groups have been introduced into the linking chain. In general, these modifications are well tolerated at D3 receptors and achieve high selectivity over D2 and D4 receptors.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
CROSS-REFERENCE TO RELATED APPLICATIONS

This application is a divisional of U.S. Nonprovisional patent application Ser. No. 12/664,668, having a 371(c) date of Jun. 25, 2010, which is a 371 national stage application of international application number PCT/US07/071412 filed Jun. 15, 2007, both of which are fully incorporated herein by reference.

BACKGROUND

The dopamine receptor system plays a key role in numerous neuropsychiatric and neurological disorders and investigation into mechanistic underpinnings and neuroadaptations within this family of receptors has been the focus of intensive research over the past decade. The dopamine D3 receptor subtype has been hypothesized to play a fundamental role, for example, in the abuse-related effects of cocaine and other drugs of abuse. Hence, there is a well-recognized need to develop novel, selective and bioavailable dopamine D3 receptor ligands.

Further reasons for pursuing dopamine D3 receptor selective ligands as medications for various conditions comes from the brain localization of D3 receptors, which are primarily expressed in limbic regions of the brain, including the nucleus accumbens. D3 receptor blockade may attenuate drug reward and/or reinforcement while avoiding the risk of extrapyramidal side effects associated with the blockade of the more ubiquitous D2 receptors.

The high degree of amino acid homology within the binding sites of the dopamine D2-like receptors, and especially between the D2 and D3 dopamine receptor subtypes, has provided a formidable challenge in the pursuit to discover dopamine D3-selective compounds. Thus far, high dopamine D2/D3 selectivity has generally been achieved with relatively large molecules, characterized, for example, by a heterocyclic moiety bridged by an unsubstituted 4-carbon chain or carbocycle to an extended or substituted arylamide or a corresponding bioisotere.

In addition to optimizing pharmacological selectivity, it is also important that dopamine D3-selective compounds be able to penetrate the blood brain barrier (BBB) and have appropriate pharmacokinetics to facilitate interpretation of in vivo results. However, generally relatively high doses of D3-selective agents have been required for behavioral activity. It is not known whether these required high doses are due to a low permeability surface area product of these agents for crossing the BBB, high peripheral metabolism, large uptake in some other organ or compartment, or is due to some other reason.

Therefore, there is a well-recognized need in the art for highly D3-selective compounds that are able to penetrate the blood brain barrier, and that show activity at relatively low dosages.

BRIEF DESCRIPTION OF THE DRAWINGS

FIG. 1: Synthesis of the 3-hydroxyl amines 27.

FIG. 2: Synthesis of the 2-hydroxyl amines 30.

FIG. 3: Synthesis of Compounds 2-23.

DESCRIPTION OF THE INVENTION

The present invention relates to chemical compounds of the following chemical formula:

wherein:

A=CHR4 or trans CH═CH;

n=0 or 1;

R1 and R2=independently represent hydrogen, halogen, or alkoxy;

R3 and R4=H, OH, OAc, alkoxy, halogen, amino, nitro, alkyl, acyl or pyridyl;

R5=phenyl, indole, thiophene, benzofuran, fluorenyl, or 2-pyridylphenyl; and R5 is optionally substituted with one or more of hydrogen, halogen, amino, nitro, hydroxyl, alkoxy, alkyl, acyl and pyridyl, substitution may occur at any of the ortho, meta, or para positions;

including all enantiomers and pharmaceutical salts.

In particular, when R3 and R4 are not H, chiral centers are present on the butyl amide linking chain and are included within the scope of the invention.

In one embodiment of the invention A in Formula (I) is C.

In one embodiment, the present invention is directed to compounds of the following chemical formula:

wherein

R1 and R2=2-methoxy or 2,3-dichloro;

R3 and R4=OH, OAc, H; and

R5=indole, thiophene, benzofuran, fluorenyl, or 2-pyridylphenyl, and R5 is substituted with one or more of methoxy, fluoro, or iodo;

including all enantiomers and pharmaceutical salts.

Dopamine D3 receptor antagonists and partial agonists are known to modulate the reinforcing and drug-seeking effects induced by cocaine and other abused substances. The introduction of functionality into the butylamide linking chain of the 4-phenylpiperazine class of ligands, improves D3 receptor affinity and selectivity, as well as water solubility. See J. Med. Chem. 2005, 48, 839-848. Along these lines, a series of linking-chain derivatives wherein functionality such as OH, OAc, etc., are introduced into the linking chain is disclosed.

In general, these modifications are well tolerated at D3 receptors (Ki=<1-5 nM) and several analogues demonstrated >100-fold selectivity over D2 and D4 receptors using competition binding assays in HEK 293 cells transfected with either hD2L, hD3 or hD4 dopamine receptors. Furthermore, addition of these groups affected efficacy of the compounds as measured by quinpirole stimulation of mitogenesis at human dopamine D3 receptors transfected into Chinese hamster ovary (CHO) cells. These compounds also provide additional tools with which to elucidate the role of D3 receptors in drug reinforcement in vivo.

The dopamine D3 receptor subtype is a member of the dopamine D2 family of receptors. These receptors are involved in a number of CNS disorders including but not limited to psychostimulant abuse, psychosis, and Parkinson's disease. (Newman, A. H.; Grundt, P.; Nader, M. A., Dopamine D3 receptor partial agonists and antagonists as potential drug abuse therapeutic agents. J. Med. Chem. 2005, 48, 3663-3679.; Heidbreder, C. A.; Andreoli, M.; Marcon, C; Thanos, P. K.; Ashby, C. R.; Gardner, E. L., Role of dopamine D-3 receptors in the addictive properties of ethanol. Drugs Today 2004, 40, 355-365.; Joyce, J. N.; Millan, M. J., Dopamine D-3 receptor antagonists as therapeutic agents. Drug Discov. Today 2005, 10, 917-925; See also, Le Foil, B.; Goldberg, S. R.; Sokoloff, P., The dopamine D-3 receptor and drug dependence: Effects on reward or beyond? Neuropharmacology 2005, 49, 525-541; Luedtke, R. R.; Mach, R. H., Progress in developing D3 dopamine receptor ligands as potential therapeutic agents for neurological and neuropsychiatric disorders Curr. Pharm. Design 2003, 9, 643-671; Boeckler, F.; Gmeiner, P., The structural evolution of dopamine D-3 receptor ligands: Structure-activity relationships and selected neuropharmacological aspects Pharmacol. Ther. 2006, 112, 281-333.)

Compounds that bind with high affinity and selectivity to D3 receptors not only provide important tools with which to study the structure and function of this receptor subtype, but also have a therapeutic effect in the treatment of numerous psychiatric and neurologic disorders.

It is known in the art that the D2 family of receptors (which includes D3) are increased in cocaine addicts and monkeys trained to self-administer cocaine. See Volkow, N. D.; Wang, G. J.; Fowler, J. S.; Logan, J.; Gatley, S. J.; Wong, C; Hitzemann, R.; Pappas, N. R., Reinforcing effects of psychostimulants in humans are associated with increases in brain dopamine and occupancy of D-2 receptors, J. Pharmacol. Exp. Ther. 1999, 291, 409-415; Volkow, N. D.; Fowler, J. S.; Wang, G. J.; Swanson, J. M., Dopamine in drug abuse and addiction: results from imaging studies and treatment implications, MoI. Psychiatry 2004, 9, 557-569; Nader, M. A.; Morgan, D.; Gage, H. D.; Nader, S. H.; Calhoun, T. L.; Buchheimer, N.; Ehrenkaufer, R.; Mach, R. H., PET imaging of dopamine D2 receptors during chronic cocaine self-administration in monkeys, Nat. Neurosci. 2006, 9, 1050-1056.

The 4-phenylpiperazine derivatives are an important class of dopamine D3 selective ligands. However, due to their highly lipophilic nature, these compounds often suffer from solubility problems in aqueous media and reduced bioavailability. To address these problems, functionality (e.g. OH, OCH3, OAc, etc.,) is introduced into the carbon chain linker of these compounds. Compared to currently available dopamine D3 receptor ligands, the resulting compounds show improved pharmacological properties and D3 selectivities, but due to their more hydrophilic nature these derivatives also have improved water solubility and bioavailability.

The more hydrophilic nature of these derivatives is shown by the cLogP (calculated measure of lipophilicity) and polar surface areas (PSA). See Table 1. As expected, the introduction of a hydroxyl group into the butyl linking chain resulted in lower clogP values, showing decreased lipophilicity, compared to the corresponding parent olefinic or aliphatic compounds. The calculated polar surface area (PSA) was significantly increased (by 20 Å2). Values less than 75 Å2 are considered as being favorable for brain penetration. See Grundt, P.; Prevatt, K. M.; Cao, J.; Taylor, J.; Floresca, C. Z.; Choi, J.-K.; Jenkins, B. G.; Luedtke, R. R.; Newman A. H. Heterocyclic Analogues of N-(4-(4-(2,3-Dichlorophenyl)piperazin-1-yl)-butyl)-aryl-carboxamides with Functionalized Linking Chains as Novel Dopamine D3 Receptor Ligands: Potential Substance Abuse Therapeutic Agents. J. Med. Chem. 2007, in press.

According to Lipinski's “rule of 5” cLogP values in the 2-5 range are expected to have drug like properties. See Lipinski, C. A., Drug-like properties and the causes of poor solubility and poor permeability, J. Pharmacol. Toxicol. Methods 2000, 44, 235-249. Comparisons of the hydroxylated analogues with their saturated butyl or olefinic counterparts demonstrate that both PSA and particularly cLogP values are predictive of improved “drug-like”, soluble, bioavailable and CNS penetrant profiles. The PSA values were calculated according to Ertl, P.; Rohde, B.; Selzer, P., Fast, Calculation of molecular polar surface area as a sum of fragment-based contributions and its application to the prediction of drug transport properties, 2000, 43, 3714-3717. CLogP values were calculated using Cambridgesoft ChemDraw Ultra 9.0, 2004.

Based on their neurochemical and behavioral properties, the dopamine D3 receptor selective ligands of the present invention are useful in methods for the treatment of all addictions, especially including nicotine and alcohol, and psychostimulant abuse, such as of cocaine, amphetamine, and derivatives thereof. The dopamine D3 receptor selective ligands of the present invention are also useful in the treatment of schizophrenia and Parkinson's disease and dyskinesias associated with these disorders and treatment thereof. Generally the methods involve administering a pharmaceutically effective amount of a compound of the present invention to a patient in need thereof.

D3 selectivity is expressed as D2/D3; a ratio that is derived from the Ki value at D2 over the Kj value at D3 receptors. Hence, a compound that exhibits higher affinity at D3 than at D2 receptors, has a D2/D3 ratio>1.” See Newman, A. H.; Grundt, P.; Nader, M. A., Dopamine D3 receptor partial agonists and antagonists as potential drug abuse therapeutic agents. J. Med. Chem. 2005, 48, 3663-3679.

The term “pharmaceutically effective amount” as used herein means an amount of the compound that produces any therapeutic effect or imaging effect in a patient. Therapeutic effect in a patient preferably relates to one of the above-mentioned conditions.

The term “alkyl” is used herein to refer to a branched or unbranched, saturated or unsaturated, monovalent hydrocarbon radical having from 1-8 carbons, including arylalkyls. Suitable alkyl radicals include, for example, methyl, ethyl, n-propyl, i-propyl, 2-propenyl (or allyl), n-butyl, t-butyl, i-butyl (or 2-methylpropyl), i-amyl, n-amyl, hexyl, etc. As used herein, the term alkyl encompasses “substituted alkyls.” The term “substituted alkyl” refers to alkyl as just described including one or more functional groups such as lower alkyl, aryl, aralkyl, acyl, halogen (i.e., alkylhalos, e.g., CF3), hydroxyl, amino, acylamino, acyloxy, alkoxyl, mercapto and the like. These groups may be attached to any carbon atom of the lower alkyl moiety.

The term “alkoxy” is used herein to refer to the —OR group, where R is a lower alkyl, substituted lower alkyl, aryl, substituted aryl, aralkyl or substituted aralkyl. Suitable alkoxy radicals include, for example, methoxy, ethoxy, phenoxy, t-butoxy, etc.

The term “lower alkyl” means C1 to C3. The term “halogen” is used herein to refer to fluorine, bromine, chlorine, and iodine atoms. The term “hydroxyl” is used herein to refer to the group —OH.

As used herein, “psychostimulant abuse” has its conventional meaning, i.e., misuse or addiction of a psychostimulant, such as cocaine, amphetamine, and derivatives thereof. Typically, cocaine is taken by a person due to a craving for cocaine generated by its prior use. Cocaine is abused when it is used for gratification, producing effects not required or recommended for therapy. The resultant high use of cocaine produces many serious and adverse side effects. As such, it is highly desirable to reduce the number and/or intensity of episodes in which a person experiences a craving for the substance or, more preferably, to eliminate the craving episodes entirely. Dopamine D3 antagonists or partial agonists have demonstrated utility in reducing craving in animal models (Villa, M. et al. Nature 400:371-375 (1999), Vorel, S. R. et al. J. Neurosci. 22:9595-9603 (2002), DiCiano, P. et al. Neuropsychopharmacology 28:329-338 (2003)).

“Treatment” or “treating,” as used herein, refers to any administration of a compound of the present invention and includes: (i) inhibiting the symptoms of the disease, e.g., cocaine addiction; and/or (ii) lessening or inhibiting the long term effects of the disease, e.g., cocaine addiction. In therapeutic applications, compositions are administered to a patient already suffering from the disease, e.g., cocaine addiction or Parkinson's disease, in a pharmaceutically effective amount.

In conjunction with the foregoing method, the present invention provides pharmaceutical compositions comprising a compound disclosed herein and a pharmaceutically acceptable diluent, carrier or excipient. While it is possible to administer the active ingredient of this invention alone, it is preferable to present it as part of a pharmaceutical formulation. The formulations of the present invention comprise at least one compound described herein in a therapeutically or pharmaceutically effective dose together with a pharmacologically or therapeutically acceptable carrier. The phrase “pharmaceutically or therapeutically acceptable carrier,” as used herein, refers to a carrier medium which does not interfere with the effectiveness of the biological activity of the active ingredients, especially D3 receptor binding of a compound of the present invention, and which is not toxic to the host or patient.

The pharmaceutical compositions of the present invention can be in a variety of forms. These include, for example, solid, semi-solid, and liquid dosage forms, such as tablets, pills, powders, liquid solutions or suspensions, liposomes, injectable and infusible solutions, inhalable preparations, such as aerosols, are also included. Preferred formulations are those directed to oral, intranasal and parenteral applications, but it will be appreciated that the preferred form will depend on the particular therapeutic application at hand. The methods for the formulation and preparation of therapeutic compositions comprising the compounds of the invention are well known in the art and are described in, for example, REMINGTON'S PHARMACEUTICAL SCIENCES (Mack Publishing Company, Philadelphia, Pa., 17th ed. (1985)), THE MERCK INDEX 11th Ed., (Merck & Co. 1989), and Langer, Science 249: 1527-1533 (1990), the teachings of which are incorporated herein by reference.

For parenteral administration, for example, the pharmaceutical compositions comprise a solution of a compound of the present invention, as described above, dissolved or suspended in an acceptable carrier, preferably an aqueous carrier. A variety of aqueous carriers can be used including, for example, water, buffered water, 0.4% saline, 0.3% glycine, hyaluronic acid and the like. These compositions may be sterilized by conventional, well-known sterilization techniques, or they may be sterile filtered. The resulting aqueous solutions may be packaged for use as is or lyophilized, the lyophilized preparation being combined with a sterile solution prior to administration. The compositions may contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions including pH adjusting and buffering agents, wetting agents and the like, such as, for example, sodium acetate, sodium lactate, sodium chloride, potassium chloride, calcium chloride, sorbitan monolaurate, thethanolamine oleate, etc.

For solid compositions, conventional nontoxic solid carriers may be used which include, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharin, talcum, cellulose, glucose, sucrose, magnesium carbonate, and the like. For oral administration, a pharmaceutically acceptable nontoxic composition is formed by incorporating any of the normally employed excipients, such as those carriers previously listed, and generally about 1% to 95%, preferably 10% to about 95% of the active ingredient and, more preferably, about 25% to about 75% of the active ingredient.

For aerosol administration, the compounds of the present invention are preferably supplied in a finely divided form along with a surfactant and propellant. The surfactant must, of course, be nontoxic, and preferably soluble in the propellant. Representative of such agents are the esters or partial esters of fatty acids containing from 6 to 22 carbon atoms, such as caproic, octanoic, lauric, palmitic, stearic, linoleic, linolenic, olesteric and oleic acids with an aliphatic polyhydric alcohol or its cyclic anhydride. Mixed esters, such as mixed or natural glycerides may be employed. A carrier can also be included as desired, as with, e.g., lecithin, for intranasal delivery.

Once improvement of the patient's conditions has occurred, a maintenance dose is administered if necessary. Subsequently, the dosage or the frequency of administration, or both, can be reduced, as a function of the symptoms, to a level at which the improved condition is retained. When the symptoms have been alleviated to the desired level, treatment can cease. Patients can, however, require intermittent treatment on a long-term basis upon any recurrence of the disease symptoms.

In general, a suitable effective dose of the compounds of the present invention will be in the range of 0.05 to 1000 milligram (mg) per recipient per day, preferably in the range of 0.1 to 100 mg per day. The desired dosage is preferably presented in one, two, three, four or more subdoses administered at appropriate intervals throughout the day. These subdoses can be administered as unit dosage forms, for example, containing 0.01 to 1000 mg, preferably 0.01 to 100 mg of active ingredient per unit dosage form. Again, the desired dosage will depend on, for example, the particular compound employed, the disease to be treated, the manner of administration, the weight and general state of health of the patient, and the judgment of the prescribing physician.

Based on their neurochemical and behavioral properties, the D3 receptor selective ligands of the present invention are also useful as imaging probes. The present compounds may be useful for functional MRI imaging of D3 receptors. In addition, the dopamine D3 receptor selective ligands of the present invention are useful as imaging agents for dopamine D3 receptors and as imaging probes for neurodegenerative disorders (e.g., Parkinson's disease). As such, in another aspect, the present invention provides a method of selectively imaging dopamine binding sites of the central nervous system of a subject, such as the brain of a human patient, the method comprising:

(a) administering to a human in need thereof an inventive compound of the present invention; and

(b) detecting the binding of the compound to the central nervous system tissue, such as the dopamine D3 receptors in the brain.

Moreover, in yet another aspect, the present invention provides a method for detecting or monitoring a disease resulting from abnormal distribution and/or density of dopamine D3 receptor in the central nervous system of a subject, comprising:

(a) administering to the subject a detectably labeled compound of the invention;

(b) detecting the binding of that compound to dopamine D3 receptor in the central nervous system;

(c) determining the distribution and/or density of the dopamine D3 receptor in the central nervous system tissue;

(d) comparing the distribution and/or density obtained in (c) with the distribution and/or density of dopamine D3 receptor in a corresponding normal tissue; and

(e) diagnosing a disease state by a difference in the distribution and/or density between the normal tissue and the subject tissue.

In a presently preferred embodiment, the dopamine selective ligands of the present invention are labeled with a radioactive label using standard labeling techniques known to and used by those of skill in the art. Suitable labels include, but are not limited to: 123I, 11C, 18F, or 99Tc. In addition, binding of the dopamine D3 receptor selective ligands to the brain, such as limbic brain regions, including the Nucleus Accumbens and islands of Calleja, is detected using methods known in the art, such as positron emission tomography (PET), single-photon emission computed tomography (SPECT), or magnetic resonance imaging (MRI). (See, e.g., Yokoi F. et al., Neuropsychopharmacology 27(2):248-59(2002); Pilowsky L. S., Nucl Med Commun 22(7):829-33(2001); Soares J C and Innis R B, Biol Psychiatry 46(5):600-15(1999); and Videbaek C, J Cereb Blood Flow Metab 21(1):92-7(2001), the teachings of which are incorporated herein by reference.

Preferably SPECT imaging employs gamma-emitting derivatives of the ligands described herein (e.g., dopamine D3 receptor selective ligands labeled with 123I or 99Tc). Yokoi et al. (supra) have mapped the normal distribution of dopamine D2 and D3 receptors in humans. Using this method, one can diagnose and/or monitor neurodegenerative disorders, such as Parkinson's disease, characterized by the progressive degeneration of dopamergic nerve terminals.

EXAMPLES

All melting points were determined on a Thomas-Hoover melting point apparatus and are uncorrected. The 1H and 13C NMR spectra were recorded on a Varian Mercury Plus 400 instrument. Proton chemical shifts are reported as parts per million (δ ppm) relative to tetramethylsilane (0.00 ppm) as an internal standard. Coupling constants are measured in hertz (Hz). Chemical shifts for 13C NMR spectra are reported as δ relative to the deuterium signal of the solvent (CDCI3, 77.5 ppm, CD3OD 49.3). Microanalyses were performed by Atlantic Microlab, Inc. (Norcross, Ga.) and agree within 0.4% of calculated values. If not stated otherwise all final compounds were purified by column chromatography (silica gel, Merck, 230-400 mesh, 60 Å) or thin layer chromatography (silica gel, Analtech, 1000 micron) using EtOAc/CHCl3/MeOH 5:5:1, 1% triethylamine or CHCI3/MeOH 10:1, 1% triethylamine as an eluent. Microwave reactions were performed in a CEM Discover Labmate system equipped with a 80 mL pressure vessel. Yields and reaction conditions are not optimized. Generally, yields and spectroscopic data refer to the free base.

Methods for performing in vitro dopamine receptor binding studies are described in Huang et al. J. Med. Chem. 44:1815-1826 (2001) and Luedtke et al. Synapse 38:438-439 (2000), the contents of which are hereby incorporated by reference. These papers describe radioactively labeled dopamine receptor selective ligands binding with picomolar affinity and nonselectivity to D2 and D3 dopamine receptors expressed in Sf9 and HEK 293 cells. 125I-IABN binds with 7- to 10-fold lower affinity to human D4.4 dopamine receptors expressed in HEK 293 cells. Dissociation constants (Kd) calculated from kinetic experiments were found to be in agreement with equilibrium Kd values obtained from saturation binding studies. Saturation plots of the binding of 125I-IABN with rat caudate membrane preparations were monophasic and exhibited low nonspecific binding. The pharmacologic profile of the binding of 125I-IABN to rat caudate was found to be consistent with a D2-like receptor, suggesting that in the caudate the ligand binds primarily to D2 dopamine receptors. IABN was found to bind with low affinity to σ1 and σ2 binding sites, as well as to D1a dopamine receptors. Quantitative autoradiographic studies using rat brain indicated that 125I-IABN selectively labels the striatum and the olfactory tubercle area, which is consistent with the labeling receptors expressed in HEK cells. Therefore, 125I-IABN appears to be a high affinity, selective antagonist at D2-like dopamine receptors.

Human dopamine D2-long (D2) and D3 (D3) receptors were expressed in HEK cells. In brief, stably transfected HEK cells expressing the human D2-long and the D3 dopamine receptor were developed using the plRES bicistronic expression vector (CLONTECH; Palo Alto, Calif.). The level of expression of D2 or D3 receptors was determined to be greater than 2,000 fmoles/mg protein. For comparison, human dopamine D4 (D4) receptors were obtained from HEK 293 cells stably transfected with a PCR product of a human cDNA coding for the D4.4 form of the human D4 dopamine receptor. The density of binding sites is approximately 1000 fmol/mg protein.

To measure D2 and D3 stimulation of mitogenesis (agonist assay) or D2 and D3 inhibition of quinpirole stimulation of mitogenesis (antagonist assay), CHOp-cells (human receptor) were seeded in a 96-well plate at a concentration of 5,000 cells/well. The cells were incubated at 37° C. in α-MEM with 10% FBS, 0.05% penicillin-streptomycin, and 200 μg/mL of G418. After 48 hours, the cells were rinsed twice with serum-free α-MEM and incubated for 24 hours at 37° C. In the functional assay for agonism, the medium was removed and replaced with 90 μl of serum-free α-MEM and 10 μl of test compound in sterile water; in the antagonist assay, the test compound was diluted in sterile water plus 30 nM quinpirole. After another 24-hour incubation at 37° C., 0.25 μCi of [3H]thymidine was added to each well and the plates were further incubated for 2 hours at 37° C. The cells were then trypsinized, and the plates were filtered and counted as usual in the art. Quinpirole was run on every plate as an internal standard.

The procedures to determine the binding affinities at the human dopamine D2-like receptors, the binding affinities at the serotonin 5HT1A, 5HT2A and 5HT2c receptors and functional mitogenesis assay are known in the art. See J. Med. Chem. 2005, 48, 839-848. See also, NIDA Research Monograph #178, Proceedings of the College of Drug Dependence, p. 440-466, 1998. Both of these references are incorporated by reference.

The racemic hydroxybutyl amine intermediates needed to prepare the 3-hydroxy derivatives 16-19 and 2-hydroxy analogues 21-23 were synthesized as depicted in FIGS. 1 and 2. In both cases, the synthetic routes used bifunctional 2-(2-bromoethyl)oxirane as starting material. See Cruickshank, P. A.; Fishman, M., “Studies In Alkylation.2. Reactions Of Epoxyalkyl Bromides”, J. Org. Chem. 1969, 34, 4060-4065, incorporated by reference. All key steps were found to be regioselective and only the products depicted were isolated. The amines (FIG. 1) were synthesized via a modified Gabriel synthesis. No side products were observed in the alkylation reaction to form the phthalimide.

The opening of the oxirane moiety occurred selectively at the least substituted side to yield the 3-hydroxy phthalimides, which were then deprotected with hydrazine to afford the hydroxylamines. In the case of the 2-hydroxy amines (30, FIG. 2) the butylpiperazine bond was formed first, followed by a regioselective opening of the epoxide with sodium azide and a Schlesinger-type reduction. The general reaction sequence used to prepare the dopamine D3 receptor preferring analogues 2-23 incorporating a butyl, butenyl or hydroxybutyl linking chain as depicted in FIG. 3. The required carboxylic acids were prepared according to procedures known in the art. See J. Med. Chem. 2001, 44, 3175-3186. See also, Synlett 2000, 829-831. Both of these references are incorporated by reference.

The general synthesis of the butyl amines and the butenyl amines is also known in the art. See J. Med. Chem. 2005, 48, 839-848; J. Med. Chem. 2001, J. Med. Chem. 2003, 46, 3883-3899; and Bioorg. Med. Chem. Lett. 2003, 13, 2179-2183. All of these references are incorporated by reference.

General procedure for the synthesis of carboxylic acid amides. The 1-imidazole adduct appropriate carboxylic acid was reacted with the suitable secondary amine derivative as known in the art. See J. Med. Chem. 2005, 48, 839-848, incorporated by reference. The crude product was purified by chromatography, structurally characterized, and then converted into its oxalate or hydrochloride for biological evaluation.

EXAMPLES Example 1 N-(4-(4-(2-Methoxyphenyl)piperazin-1-yl)butyl)-9H-fluorene-2-carboxamide (7)

Prepared from 9H-fluorene-2-carboxylic acid and 4-(4-(2-methoxyphenyl)-piperazin-1-yl)-butylamine according to the general procedure. Yield: 74%. Mp. (hydrochloride): 208-210° C. 1H NMR (CDCI3): δ 1.67-1.73 (m, 4H), 2.47 (t, J 6.7, 2H), 2.65 (s, 4H), 3.04 (s, 4H), 3.51 (q, J 6.1, 2H), 3.84 (s, 3H), 3.91 (s, 2H), 6.82-6.88 (m, 4H), 6.98 (dt, J 7.8, 4.6, 1H), 7.35 (td, J 7.4, 1.2, 1H), 7.40 (t, J 7.0, 1H), 7.55 (d, J 7.0, 1H), 7.75-7.77 (m, 2H), 7.81 (d, J 8.0, 1H), 7.96 (s, 1H). 13C NMR (CDCI3): δ 25.0, 27.9, 37.3, 40.5, 50.93, 53.9, 55.8, 58.5, 111.5, 118.6, 120.1, 121.0, 121.4, 123.4, 124.4, 125.6, 126.3, 127.4, 128.0, 133.8, 141.2, 141.6, 143.8, 144.4, 145.1, 152.7, 168.6. Anal. (C29H33N3O2 ̂HCIO—SH2O) C, H, N.

Example 2 N-(4-(4-(2-Methoxy-phenyl)-piperazin-1-yl)-butyl)-4-pyridin-2-yl-benzamide (8)

Prepared from 4-pyridin-2-yl-benzoic acid hydrochloride and 4-(4-(2-methoxy-phenyl)-piperazin-1-yl)-butylamine according to the general procedure. Yield: 53%. Mp. (oxalate): foam. 1H NMR (CDCI3): δ 1.67-1.74 (m, 4H), 2.49 (t, J 6.84, 2H), 2.67 (s, 2H), 3.07 (s, 2H), 3.51 (q, J 6.0, 2H), 3.84 (s, 3H), 6.83 (m, 5H), 7.26 (m, 1H), 7.73-7.79 (m, 2H), 7.89 (d, J 8.2, 2H), 8.06 (d, J 8.6, 2H), 8.71 (dt, J 4.7, 1.3, 1H). 13C NMR (CDCl3): δ 24.8, 27.9, 40.4, 50.8, 53.9, 55.8, 58.5, 111.6, 118.7, 121.3, 121.4, 123.1, 123.4, 127.4, 127.9, 135.6, 137.3, 142.5, 150.3, 152.7, 156.7, 167.8. Anal. (C27H32N4O2 (COOH)20.75H2O) C, H, N.

Example 3 N-(4-(4-(2,3-Dichloro-phenyl)-piperazin-1-yl)-trans-but-2-enyl)-4-(6-oxo-1,6-dihydro-pyridin-2-yl)-benzamide (9)

A suspension of 0.12 g (0.53 mmol) 4-(6-oxo-1,6-dihydro-pyridin-2-yl)-benzoic acid, 0.13 g (0.64 mmol) dicyclohexylcarbodiimide, 0.1 g (0.7 mmol) 1-hydroxbenzotriazole hydrate in 15 mL DMF was treated at 0° C. with 0.16 g (0.53 mmol) 4-(4-(2,3-chloro-phenyl)-piperazin-1-yl)-trans-but-2-enyl amine and 0.17 mL (1.2 mmol) triethylamine. The reaction mixture was stirred at room temperature for 3 days and filtered. The solvent was removed in vacuo and the residue was taken up in saturated sodium bicarbonate and CHCI3. The combined organics were dried with sodium sulphate, concentrated and the crude product was purified by thin layer chromatography. Yield: 0.10 g (38%). Mp. (oxalate): 153-154° C. 1H NMR (CDCI3): δ 2.67 (s, 4H), 3.06 (s, 6H), 4.11 (m, 2H), 5.74-5.83 (m, 2H), 6.51-6.54 (m, 2H), 6.70 (t, J 5.3, 1 H), 6.95 (dd, J 6.7, 2.9, 1H), 7.11-7.17 (m, 2H), 7.50 (dd, J 9.1, 7.0, 1H), 7.72 (d, J 8.3, 2 H), 7.85 (d, J 8.4, 2 H). 13C NMR (CDCI3): δ 40.6, 50.2, 52.3, 59.4, 105.9, 17.9, 118.1, 124.1, 126.3, 126.6, 126.7, 127.0, 127.3, 130.2, 133.2, 135.0, 135.7, 141.6, 145.6, 150.4, 164.5, 167.0. Anal. (C26H26CI2N4O2 1.5 (COOH)2) C, H, N.

Example 4 N-(4-(4-(2,3-Dichloro-phenyl)-piperazin-1-yl)-trans-but-2-enyl)-4-(6-methyl-pyridin-2-yl)-benzamide (10)

Prepared from 4-(6-methylpyridin-2-yl)benzoic acid hydrochloride and 4-(4-(2,3-chloro-phenyl)-piperazin-1-yl)-trans-but-2-enyl amine according to the general procedure. Yield: 18%. Mp. (oxalate): 125-126° C. 1H NMR (CDCI3): δ 2.63 (s, 7H), 3.08 (s, 6H), 4.12 (m, 2H), 5.78-5.80 (m, 2H), 6.42 (s, br, 1H), 6.95 (d, J 6.7, 2.7, 1H), 7.11-7.16 (m, 3H), 7.55 (d, J 7.7, 1 H), 7.65 (t, J 7.6, 1H), 7.87 (d, J 8.7, 2H), 8.05 (d, J 8.3, 2H). 13CNMR (CDCI3): δ 25.2, 42.0, 51.7, 53.7, 60.7, 118.3, 119.1, 122.8, 125.0, 127.5, 127.8, 127.9, 129.4, 130.2, 134.4, 134.7, 137.5, 143.1, 151.7, 156.0, 159.0, 167.4. Anal. (C27H28CI2N4O) 2(COOH)2): C1H1N.

Example 5 N-(4-(4-(2,3-Dichlorophenyl)piperazin-1-yl)-trans-but-2-enyl)-4-(3-methyl-pyridin-2-yl)-benzamide (11)

Prepared from 4-(3-methylpyridin-2-yl)benzoic acid hydrochloride and 4-(4-(2,3-chloro-phenyl)-piperazin-1-yl)-trans-but-2-enyl amine according to the general procedure. Yield: 55%. Mp. (oxalate): foam. 1H NMR (CDCI3): δ 2.34 (s, 3H), 2.65 (s, 4H), 3.07-3.10 (m, 6H), 6.12 (m, 2H), 5.78-5.82 (m, 2H), 6.49 (t, J 5.5, 1H), 6.95 (dd, J 6.5, 2.9, 1H), 7.12-7.16 (m, 2H), 7.21 (dd, J 7.6, 4.8, 1H), 7.57-7.62 (m, 3H), 7.86 (dt, J 8.6, 2.0, 2H), 8.53 (dt, J 4.2, 0.8, 1H). 13C NMR (CDCI3): δ 20.4, 42.0, 51.7, 53.7, 60.7, 119.1, 123.0, 125.0, 127.3, 127.9, 129.4, 129.7, 130.3, 131.4, 134.2, 134.4, 139.2, 144.1, 147.5, 151.6, 158.0, 167.5. Anal. (C27H28CI2N4O IS(COOH)2O—SC3H7OHÔH2O) C, H, N.

Example 6 N-(4-(4-(2,3-Dichloro-phenyl)-piperazin-1-yl)-trans-but-2-enyl)-4-(pyridin-N-oxide-2-yl)-benzamide (12)

Prepared from 4-(pyridin-N-oxide-2-yl)-benzoic acid and 4-(4-(2,3-dichloro-phenyl)-piperazin-1-yl)-trans-but-2-enylamine according to the general procedure. Yield: 27%. Mp. (oxalate): 153-154° C. 1HNMR (CDCI3): δ 2.91 (s, br, 4H), 3.06 (m, 6H), 4.07 (m, 2H), 5.71-5.82 (m, 2H), 6.93-6.96 (m, 2H), 7.12-7.15 (m, 2H), 7.27-7.31 (m, 1H), 7.36 (t, J 7.54, 1H), 7.45 (d, J 7.83, 1.74, 1H), 7.86 (m, 4H), 8.33 (d, J 6.65, 0.78, 1H). 13C NMR (CDCI3): δ 41.8, 51.4, 53.4, 60.4, 118.9, 124.8, 125.4, 126.5, 127.3, 127.7, 127.7, 128.9, 129.7, 130.3, 134.2, 135.5, 135.6, 140.7, 148.7, 151.39, 166.9. Anal. (C26H26CI2N4O2 1.5 (COOH)2): C1H1N.

Example 7 4-(2,3-Dichlorophenyl)-1-(4-(4-(pyridin-2-yl)benzamido)-trans-but-2-enyl)-piperazine 1-oxide (13)

A solution of PG01037, see U.S. 2006/0106030 (288 mg, 0.60 mmol) in 10 mL dichloromethane was treated at 0° C. with meta-chloroperbenzoic acid (0.16 g, 77%, 0.72 mmol). After stirring for 16 h at room temperature, the reaction mixture was successively washed with saturated sodium bicarbonate solution, H2O and brine and dried with sodium sulphate. The volatiles were removed in vacuo and the residue was purified by preparative thin layer chromatography. Yield: 93 mg (32%). Mp. (hydrochloride): foam. 1H NMR (CD3OD): δ 3.23-3.29 (m, 4H), 3.50-3.61 (m, 4H), 3.99 (d, J 6.5, 1H), 4.13 (d, J 4.6, 1H), 6.04-6.18 (m, 2H), 7.18 (dd, J 7.8, 3.9, 1H), 7.24-7.28 (m, 2H), 7.40 (m, 1H), 7.91-7.93 (m, 2H), 7.99 (d, J 8.4, 2H), 8.06 (d, J 8.7, 2H), 8.64 (dt, J 4.8, 1.2, 1H). 13C NMR (CDCI3): δ 41.0, 45.5, 63.5, 72.0, 119.3, 120.0, 121.6, 123.1, 125.4, 127.0, 127.3, 127.7, 128.0, 133.8, 134.6, 137.8, 137.9, 142.2, 149.3, 150.1, 156.4, 168.3. Anal. (C26H26CI2N4O2:2HCr0.5H2O) C, H, N.

Example 8 N-(4-(4-(2-Methoxy-phenyl)-piperazin-1-yl)-trans-but-2-enyl)-4-pyridin-2-yl-benzamide (14)

Prepared from 4-pyridin-2-yl-benzoic acid hydrochloride and 4-(4-(2-methoxyphenyl)-piperazin-1-yl)-trans-but-2-enyl amine according to the general procedure. Yield: 65%. Mp. (hydrochloride): 168-170° C. 1H NMR (400 MHz, CDCI3): δ 2.70 (s, 4H), 3.12 (s, 6H), 3.86 (s, 3H), 4.13 (m, 2H), 5.78-5.85 (m, 2H), 6.43 (m, 1H), 6.85-7.02 (m, 4H), 7.27 (s, 2H), 7.77 (s, 2H), 7.90 (d, J 7.5, 2H), 8.06 (d, J 8.1, 2H), 8.72 (d, J 3.1, 1H). 13C NMR (101 MHz, CDCI3): δ 41.0, 50.9, 53.7, 55.8, 60.7, 111.6, 118.7, 121.3, 121.4, 123.2, 123.5, 127.5, 127.9, 129.1, 130.5, 135.0, 137.4, 141.6, 142.7, 150.3, 152.7, 156.7, 167.4. Anal. (C27H3oN4O2-3HC|-4H2O) C, H, N.

Example 9 N-(4-(4-(2-methoxyphenyl)piperazin-1-yl)-trans-but-2-enyl)-9H-fluorene-2-carboxamide (15)

Prepared from 9H-fluorene-2-carboxylic acid and 4-(4-(2-methoxyphenyl)-piperazin-1-yl)-trans-but-2-enyl amine according to the general procedure. Yield: 61%. Mp. (hydrochloride): 224-226° C. 1H NMR (400 MHz, CDCI3): d 2.68 (s, 4H), 3.09-3.10 (m, 6H), 3.86 (s, 3H), 3.94 (s, 2H), 4.13 (m, 2H), 5.81-5.83 (m, 2H), 6.32 (t, J 5.4, 1H), 6.85 (dd, J 8.2, 1.2, 1H), 6.89-6.96 (m, 2H), 7.00 (m, 1H), 7.35 (td, J 7.3, 1.3, 1H), 7.40 (td, J 7.4, 1.6, 1H), 7.57 (d, J 7.4, 1H), 7.78-7.83 (m, 3H), 7.99 (s, 1H). 13C NMR (101 MHz, CDCI3): d 37.4, 42.0, 51.0, 53.8, 55.8, 60.8, 111.6, 118.7, 120.2, 121.0, 121.4, 123.4, 124.3, 125.7, 126.2, 127.5, 128.1, 129.3, 130.4, 133.1, 141.1, 141.7, 143.9, 144.5, 145.4, 152.7, 168.0. Anal. (C29H31N3O2—SHCI) C, H1 N.

Example 10 N-(4-(4-(2,3-Dichlorophenyl)piperazin-1-yl)-3-hydroxybutyl)-4-pyridin-2-yl-benzamide (16)

Prepared from 4-pyridin-2-yl-benzoic acid hydrochloride and 27a according to the general procedure. Yield: 52%. Mp. (hydrochloride): foam. 1H NMR (CDCI3): δ 1.62 (m, 1H), 1.83 (m, 1H), 2.39-2.46 (m, 2H), 2.58 (m, 2H), 2.82 (m, 2H), 3.05 (s, 4H), 3.45 (m, 1H), 3.89 (m, 2H), 4.00 (s, 1H), 6.91 (dd, J 7.1, 2.5, 1H), 7.08-7.15 (m, 2H), 7.23 (ddd, J 5.89, 4.83, 2.58, 1H), 7.50 (dd, J 5.9, 3.7, 1H), 7.70-7.76 (m, 2H), 7.88 (d, J 8.3, 2H), 8.02 (d, J 8.3, 2H), 8.67 (dt, J 4.8, 1.2, 1H). 13C NMR (CDCI3): δ 33.7, 38.8, 51.6, 53.5, 63.9, 66.6, 118.3, 120.5, 122.3, 124.3, 126.6, 127.1, 127.1, 133.6, 134.4, 136.4, 141.5, 149.3, 150.5, 155.7, 166.3. Anal. (C26H28CI2N4O2-2HCI O.52-PrOH I 0.5H2O) C1 H, N.

Example 11 N-(3-(4-(2,3-Dichlorophenyl)-piperazin-1-yl)-3-hydroxybutyl)-9H-fluorene-2-carboxamide (17)

Prepared from 9H-fluorene-2-carboxylic acid and 27a according to the general procedure. Yield: 58%. Mp. (oxalate): 188-190° C. 1H NMR (oxalate, CDCI3, 5% D2O) δ 1.60-1.69 (1H, m), 1.83-1.87 (1H, m), 2.42-2.50 (2H, m), 2.61 (2H, m), 2.86-2.87 (2H, m), 3.07 (4H, s), 3.45-3.52 (1H, m), 3.87-3.94 (4H, m), 6.94-8.00 (10Hi m). 13C NMR (oxalate, CDCI3, 5% D2O) δ 33.6, 37.2, 38.6, 51.6, 53.5, 63.9, 66.7, 118.8, 119.9, 120.8, 124.1, 125.0, 125.4, 126.0, 127.2, 127.7, 127.8, 127.8, 133.1, 134.3, 141.0, 143.6, 144.3, 145.0, 151.3, 167.4, 167.8. Anal. (C28H29CI2N3O2 (COOH)2), C1H, N.

Example 12 N-(3-Hydroxy-4-(4-(2-methoxy-phenyl)-piperazin-1-yl)-butyl)-4-pyridin-2-yl-benzamide (18)

Prepared from 4-pyridin-2-yl-benzoic acid hydrochloride and 27b according to the general procedure. Yield: 47%. Mp. (oxalate): foam. 1H NMR (CDCI3): δ 1.63 (m, 1H), 1.84 (m, 1H), 2.43 (m, 2H), 2.64 (m, 2H), 2.88 (m, 2H), 3.11 (s, 4H), 3.46 (m, 1H), 3.86 (s, 3H), 4.94 (m, 2H), 6.86 (dd, J 7.4, 1.2, 1H), 6.92-6.94 (m, 2H), 7.01 (m, 1H), 7.23 (m), 7.52 (dd, J 5.9, 3.7, 1H), 7.74-7.79 (m, 2H), 7.91 (d, J 8.3, 2H), 8.05 (d, J 8.3, 2H), 8.71 (dt, J 4.8, 1.2, 1H). 13C NMR (CDCI3): δ 33.0, 38.3, 50.5, 53.2, 55, 63.5, 66.3, 110.9, 117.9, 120.6, 120.7, 122.4, 122.8, 126.7, 127.2, 134.6, 136.6, 140.8, 141.7, 149.6, 152.0, 156.1, 166.7. Anal. (C27H32N4O22(COOH)20.5H2O) C, H, N.

Example 13 N-(3-Hydroxy-4-(4-(2-methoxyphenyl)piperazin-1-yl)-butyl)-9H-fluorene-2-carboxamide (19)

Prepared from 9/−/-fluorene-2-carboxylic acid and 27b according to the general procedure. Yield: 45%. Mp. (oxalate): foam. (1H NMR (CDCI3): δ 1.64 (m, 1H), 1.83 (m, 1H), 2.43 (m, 2H), 2.87 (m, 2H), 3.10 (s, 4H), 3.47 (m, 1H), 3.86 (s, 3H), 3-89-3.99 (m, 6H), 6.86 (d, J 7.4, 1H), 6.91-6.94 (m, 2H), 7.01 (m, 1H), 7.32-7.41 (m, 2H), 7.45 (dd, J 5.9, 3.5, 1H), 7.55 (d, J 7.4, 1H), 7.77-7.83 (m, 3H), 8.00 (s, 1H). 13C NMR (CDCI3): δ 33.3, 36.9, 38.5, 50.7, 55.3, 63.8, 66.6, 111.1, 118.1, 119.6, 120.5, 120.9, 123.1, 123.8, 125.2, 125.8, 126.9, 127.5, 133.0, 140.7, 141.0, 143.3, 144.0, 144.6, 152.2, 167.5. Anal. (C29H33N3O3 (COOH)2 H2O) C, H, N.

Example 14 1-(4-(2,3-Dichlorophenyl)piperazin-1-yl)-4-(4-(pyridin-2-yl)-benzamido)-butan-2-yl acetate (20)

A solution of 16 (0.25 g, 0.5 mmol) in 10 ml of CH2CI2 was treated with 70 μl (0.75 mmol) acetic anhydride followed by 140 μL (1.0 mmol) triethylamine. After stirring for 16 h, the mixture was washed with sodium bicarbonate solution, dried with sodium sulphate, and purified by flash chromatography. Yield: 0.22 g (82%). Mp. (oxalate): foam. 1H NMR (CDCI3): δ 1.84 (m, 1H), 2.05 (m, 1H), 2.12 (s, 3H), 2.52 (dd, J 13.2, 5.2, 1H), 2.65 (m, 5H), 2.99 (s, 4H), 3.25 (dq, J 9.7, 5.00, 1H), 3.81 (dt, J 12.3, 5.7, 1H), 5.19 (m, 1H), 6.89 (dd, J 7.7, 1.8, 1 H), 7.06-7.17 (m, 3H), 7.26 (ddd, J 6.1, 4.8, 2.44, 1H), 7.73-7.76 (m, 2H), 7.93 (d, J 8.6, 2H), 8.07 (d, J 8.6, 2H), 8.70 (td, J 4.83, 1.50, 1.50, 1H). 13C NMR (CDCI3): δ 21.0, 32.4, 36.1, 51.3, 53.7, 61.5, 69.6, 118.6, 120.8, 122.7, 124.5, 127.0, 127.4, 127.5, 133.9, 134.7, 136.9, 142.1, 149.8, 151.1, 156.2, 167.1, 171.5, Anal. (C28H3OCI2N4O3 IS(COOH)2—H2O) C, H, N.

Example 15 N-(4-(4-(2,3-Dichlorophenyl)piperazin-1-yl)-2-hydroxybutyl)-4-(pyridin-2-yl)-benzamide (21)

Prepared from 4-pyridin-2-yl-benzoic acid and 30a according to the general procedure. Yield: 52%. Mp. (oxalate): foam. 1H NMR (CDCI3): δ 1.63 (ddd, J 14.6, 6.0, 3.4, 1 H), 1.81 (dtd, J 14.5, 10.9, 10.8, 3.9, 1H), 2.72 (m, 6H), 3.07 (s, 4H), 3.34 (ddd, J 13.4, 7.6, 4.6, 1H), 3.77 (ddd, J 13.4, 6.7, 3.4, 1H), 4.07 (m, 1H), 6.77 (t, J 5.3, 1H), 6.93 (dd, J 7.4, 2.2, 1H), 7.13-7.19 (m, 2H), 7.28 (m, 1H), 7.76-7.81 (m, 2H), 7.91 (d, J 8.6, 2H), 8.07 (d, J 8.6, 2H), 8.72 (td, J 4.8, 1.4, 1.4, 1H). 13C NMR (CDCI3): δ 28.8, 45.8, 51.4, 53.3, 57.5, 72.7, 118.7, 120.9, 122.8, 125.0, 127.1, 127.5, 127.6, 127.7, 134.2, 134.8, 137.0, 142.2, 149.9, 150.9, 156.3, 167.3. Anal. (C26H28CI2N4O2 (COOH)2 H2O), C, H, N.

Example 16 N-(4-(4-(2,3-Dichlorophenyl)piperazin-1-yl)-2-hydroxybutyl)-9H-fluorene-2-carboxamide (22)

Prepared from 9H-fluorene-2-carboxylic acid and 30a according to the general procedure. Yield: 68%. Mp. (oxalate): foam. 1H NMR (CDCI3): δ 1.63 (m, 1H), 1.78 (m, 1H), 2.63 (s, 2H), 2.72-2.84 (m, 2H), 2.87 (s, 2H), 3.07 (s, 4H), 3.34 (m, 1H), 3.77 (ddd, 13.7, 7.0, 3.5, 1H), 3.94 (s, 2H), 4.07 (m, 1H), 6.90 (dd, J 5.4, 2.0, 1H), 7.12-7.18 (m, 2H), 7.36 (t, J 5.3, 1H), 7.37 (t, J 5.4, 1H), 7.41 (d, J 7.0, 1H), 7.81-7.82 (m, 3H), 8.00 (s, 1H). 13C NMR (CDCI3): δ 28.9, 37.0, 45.9, 51.4, 53.4, 57.5, 72.8, 118.7, 119.8, 120.7, 124.0, 125.0, 125.3, 126.0, 127.1, 127.6, 127.7, 127.8, 133.0, 134.2, 140.8, 143.6, 144.2, 145.0, 150.9, 168.0. Anal. (C28H29CI2N3O2-2.5HCIO.5EtOAσ1.75H2O) C, H, N.

Example 17 N-(4-(4-(2-Methoxyphenyl)piperazin-1-yl)-2-hydroxybutyl)-9H-fluorene-2-carboxamide (23)

Prepared from 9H-fluorene-2-carboxylic acid and 30b according to the general procedure. Yield: 42%. Mp. (oxalate): foam. 1H NMR (CDCI3): δ 1.62 (ddd, J 9.9, 6.1, 3.4, 1H), 1.80 (m, 1H), 2.63 (s, 2H), 2.76 (m, 2H), 2.88 (s, 2H), 3.10 (s, 4H), 3.33 (ddd, J 13.4, 7.7, 4.5, 1 H), 3.78 (ddd, J 13.4, 6.8, 3.4, 1H), 3.80 (s, 3H), 3.91 (s, 2H), 4.06 (m, 1H), 6.83-6.87 (m, 2H), 6.89-6.92 (m, 2H), 7.01 (ddd, J 8.0, 5.8, 3.3, 1H), 7.34 (td, J 7.4, 1.3, 1H), 7.39 (td, J 7.5, 1.3, 1 H), 7.55 (d, J 7.5, 1H), 7.79 (dd, J 7.9, 0.5, 1H), 7.82 (dd, J 8.1, 1.5, 1H), 8.00 (d, J 0.7, 1H). 13C NMR (CDCI3): δ 28.8, 36.9, 45.8, 50.6, 53.5, 55.4, 57.4, 72.6, 111.2, 118.2, 119.7, 120.6, 121.0, 123.2, 123.9, 125.2, 125.9, 127.0, 127.6, 132.9, 140.7, 140.8, 143.4, 144.0, 144.8, 152.2, 167.9. Anal. (C29H33N3O3—(COOH)2—ÎH2O) C, H, N.

Example 18 2-(Oxiran-2-yl)-ethyl-isoindoline-1,3-dione (25)

A suspension of 1.84 g (10.0 mmol) phthalimide potassium salt in 20 ml DMF was treated with 2.27 g (15.0 mmol) 24 in the microwave (pressure vessel, Pmax 150 W, cooling, 100° C., 20 min). See J. Org. Chem. 1969, 34, 4060-4065. The cooled reaction mixture was filtered, diluted with EtOAc (20 mL) and was washed with H2O (2×10 mL). The organic phase was dried with sodium sulphate and the volatiles were removed in vacuo to give 25 (1.68 g, 78%) as a foam, which was used without further purification. 1H NMR (CDCI3): δ 1.86 (m, 1H), 2.00 (m, 1H), 2.46 (m, 1H), 2.73 (t, J 3.9, 1H), 3.00 (m, 1H), 3.89 (m, 2H), 7.70-7.74 (m, 2H), 7.83-7.87 (m, 2H). 13C NMR (CDCI3): δ 31.7, 35.2, 46.5, 50.4, 123.4, 132.2, 134.1, 168.4.

Example 19 2-(4-(4-(2,3-Dichlorophenyl)-piperazin-1-yl)-3-hydroxybutyl)-isoindoline-1,3-dione (26a)

A sample of 2.1 g (9.0 mmol) 1-(2,3-dichlorophenyl)-piperazine in 40 mL 2-PrOH was reacted in the microwave (pressure vessel, Pmax 150 W, cooling, 90° C., 20 min) with 2.0 g (9.0 mmol) 25. The solvent was removed in vacuo and the foamy residue was washed with 10 mL 2-PrOH. Yield: 2.96 g (73%). 1H NMR (CDCI3): δ 1.79 (m, 2H), 2.42 (m, 2H), 2.56 (s, 2H), 2.79 (m, 2H), 3.02 (s, 4H), 3.60 (s, 1H), 3.74-3.83 (m, 3H), 6.90 (m, 1H), 7.06-7.11 (m, 2H), 7.67 (m, 2H), 7.81 (m, 2H). 13C NMR (CDCI3): δ 33.9, 35.3, 51.4, 53.4, 63.8, 64.5, 118.2, 122.7, 124.1, 127.0, 131.6, 133.4, 133.4, 150.4, 167.7.

Example 20 2-(4-(4-(2-Methoxyphenyl)piperazin-1-yl)-3-hydroxybutyl)isoindoline-1,3-dione (26b)

Prepared from 1-(2-methoxyphenyl)piperazine and 25 in a similar fashion as described above for 26a. Yield: 28%. Mp.: 192-194° C. 1H NMR (CDCI3): δ 1.79 (q, J 6.8, 2H), 2.41 (m, 2H), 2.61 (s, 2H), 2.85 (s, 2H), 3.07 (s, 4H), 3.77 (m, 1H), 3.85 (s, 3H), 3.91 (m, 2H), 6.86 (d, J 7.0, 1H), 6.91-6.95 (m, 2H), 7.00 (m, 1H), 7.85 (dd, J 5.5, 3.0, 2H), 7.71 (dd, J 5.4, 3.1, 2H). 13C NMR (CDCI3): δ 33.8, 35.4, 50.9, 53.6, 55.6, 64.1, 64.7, 111.3, 118.4, 121.2, 123.2, 123.4, 132.4, 134.1, 141.4, 152.4.

Example 21 4-Amino-1-(4-(2,3-dichlorophenyl)piperazin-1-yl)-butan-2-ol (27a)

A sample of 4.48 g (10.0 mmol) 26a was fully dissolved in 25 ml EtOH and treated with 0.48 g (15.0 mmol) hydrazine in the microwave (pressure vessel, Pmax 150 W, cooling, 90° C., 20 min). The cooled reaction mixture was filtered and the filtrate was evaporated in vacuo. Both the residue from the evaporation and the initial precipitate were partitioned between CHCI3 and 20% potassium carbonate solution. The layers were separated and the aqueous layer was dried with sodium sulphate to give the title compound as an oil, which was used without further purification. Yield: 2.25 g (71%). 1H NMR (CDCI3): δ 1.56 (m, 2H), 2.40 (m, 2H), 2.61 (s, 2H), 2.80 (s, 2H), 2.97-3.05 (m, 9H), 3.89 (m, 1H), 6.92 (dd, J 6.3, 3.1, 1H), 7.09-7.14 (m, 2H). 13C NMR (CDCI3): δ 37.4, 39.7, 51.5, 53.6, 64.5, 66.3, 118.3, 124.2, 127.1, 133.5, 150.6.

Example 22 4-Amino-1-(4-(2-methoxy-phenyl)-piperazin-1-yl)-butan-2-ol (27b)

Prepared from 26b in a similar fashion as described above for 27a. Yield: 69%. Wax. 1H NMR (CDCI3): δ 1.59 (m, 2H), 2.41 (m, 2H), 2.57 (m, 2H), 2.79-2.81 (m, 9H), 3.86 (s, 3H), 3.91 (m, 1H), 6.86 (d, J 7.5, 1H), 6.91-6.96 (m, 2H), 6.98-7.03 (m, 1H). 13C NMR (CDCI3): δ 37.3, 39.7, 50.9, 53.6, 55.5, 64.5, 66.2, 111.2, 118.3, 121.1, 123.1, 141.3, 152.3.

Example 23 1-(2,3-Dichlorophenyl)-4-(2-(oxiran-2-yl)ethyl)piperazine (28a)

2.31 g (10.0 mmol) 1-(2,3-dichlorophenyl)piperazine was added to a suspension of 2.27 g (15.0 mmol) 24, 4.15 g (30.0 mmol) potassium carbonate in 150 ml acetone and the reaction mixture was refluxed for 24 h. The reaction mixture was filtered and the volatiles were removed in vacuo to give an oil (2.86 g, 95%), which was used without further purification. 1H NMR (CDCI3): δ 1.72 (m, 1H), 1.83 (m, 1H), 2.53 (dd, J 5.0, 2.7, 1H), 2.61 (ddd, J 8.3, 6.5, 3.0, 2H), 2.66 (s, 4H), 2.79 (dd, J 4.9, 4.0, 1H), 3.01 (m, 1H), 3.07 (s, 4H), 6.96 (dd, J 6.5, 3.1, 1H), 7.11-7.19 (m, 2H). 13C NMR (CDCI3): δ 30.2, 47.1, 51.0, 51.4, 53.3, 55.0, 118.6, 124.6, 127.5, 134.0, 151.3.

Example 24 1-(2-Methoxyphenyl)-4-(2-(oxiran-2-yl)ethyl)piperazine (28b)

Prepared from 24 and 1-(2-methoxyphenyl)piperazine in a similar fashion as described above for 28a. Yield: 87%. 1H NMR (CDCI3): δ 2.60 (m, 2H), 2.67 (s, 4H), 2.78 (dd, J 4.9, 4.0), 3.00 (m, 1H), 3.10 (s, 4H), 3.86 (s, 3H), 6.86 (dd, J 7.8, 1.3, 1H), 6.88-7.03 (m, 3H). 13C NMR (CDCI3): δ 30.2, 47.2, 50.7, 51.0, 53.5, 55.2, 55.4, 111.2, 118.3, 121.1, 123.0, 141.4, 152.3.

Example 25 1-Azido-4-(4-(2,3-dichlorophenyl)piperazin-1-yl)butan-2-ol (29a)

A suspension of 0.75 g (2.5 mmol), 0.24 g (3.8 mmol) sodium azide and 0.27 g (5.0 mmol) ammonium chloride in 5 imL DMF was heated at 100° C. for 5 h. The reaction mixture was partitioned between 10 ml CHCI3 and 10 ml H2O. The aqueous organic layer was extracted twice with 10 ml CHCI3, dried over sodium sulphate and the volatiles were removed in vacuo. The residue was purified by flash chromatography to give the 29a as an oil. Yield: 0.44 g (51%). 1H NMR (CDCI3): δ 1.56 (ddd, J 14.7, 6.5, 3.4, 1H), 1.81 (m, 1H), 2.62 (s, 2H), 2.75 (m, 2H), 2.87 (s, 2H), 3.07 (s, 4H), 3.27 (dd, J 5.1, 1.4, 2H), 4.04 (dtd, J 7.9, 5.3, 2.6, 1H), 6.52 (s, 1H), 6.93 (dd, J 7.2, 2.4, 1H), 7.12-7.19 (m, 2H). 13C NMR (CDCI3): δ 28.4, 51.3, 53.3, 56.5, 57.3, 73.0, 118.7, 124.9, 127.6, 134.1, 150.9.

Example 26 1-Azido-4-(4-(2-methoxyphenyl)piperazin-1-yl)butan-2-ol

Prepared from 28b in a similar fashion as described above for 29a. Yield: 15%. 1H NMR (CDCI3): δ 1.55 (ddd, J 14.6, 6.7, 3.6, 1H), 1.80 (m, 1H), 2.62 (s, 2H), 2.74 (m, 2H), 2.89 (s, 2H), 3.09 (s, 4H), 3.24 (dd, J 11.6, 4.1, 1H), 3.29 (dd, J 11.6, 4.9, 1H), 3.86 (s, 3H), 4.03 (dtd, J 9.8, 5.5, 2.5, 1H), 6.86 (d, J 8.0, 1H), 6.89-6.95 (m, 2H), 7.01 (ddd, J 8.0, 5.1, 4.1, 1H). 13C NMR (CDCI3): δ 28.4, 50.7, 53.5, 53.5, 55.4, 56.6, 57.4, 73.0, 111.2, 118.3, 121.1, 123.2, 140.9, 152.3.

Example 27 1-Amino-4-(4-(2,3-dichlorophenyl)piperazin-1-yl)butan-2-ol (30a)

A solution of 29a (0.41 g, 1.2 mmol) and 1.87 g (7.2 mmol) triphenylphosphine in 20 ml of a THF/H2O mixture (10:1 v/v) was stirred at room temperature for 16 h. The volatiles were removed in vacuo and the residue was taken up in 2-PrOH (5 ml) and treated with ethereal hydrochloric acid to give desired amine as a hydrochloride (0.33 g, 77%). 1H NMR (CDCI3): δ 1.53 (ddd, J 14.5, 6.8, 4.0, 1H), 1.71 (m, 1H), 2.61 (s, 2H), 2.65-2.77 (m, 4H), 2.84 (s, 2H), 3.07 (m, 4H), 3.77 (m, 1H), 6.93 (dd, J 7.0, 2.5, 1H), 7.12-7.17 (m, 2H). 13C NMR (CDCI3): δ 29.0, 48.3, 51.3, 53.3, 74.9, 118.6, 124.8, 127.5, 134.0, 150.9.

Example 28 1-Amino-4-(4-(2-methoxyphenyl)piperazin-1-yl)butan-2-oI (30b)

Prepared from 29b in a similar fashion as described above for 30a. Yield: 13%. 1H NMR (CDCI3): δ 1.52 (ddd, J 14.5, 6.5, 3.9, 1H), 1.71 (m, 1H), 2.54-2.77 (m, 4H), 2.86 (s, 2H), 3.09 (S1 4H), 3.77 (m, 1H), 3.87 (s, 3H), 6.86 (d, J 7.9, 1H), 6.90-6.95 (m, 2H), 7.00 (m, 1H). 13C NMR (CDCI3): δ 29.0, 48.4, 50.7, 53.6, 55.4, 57.6, 75.2, 111.2, 118.3, 121.1, 123.2, 141.0, 152.3.

TABLE 1 Human D2-like Family Receptor Subtype Binding Data in HEK Cells D3 Compd. structure D2 SEM Ki [nM] SEM D4 SEM D2/D3 D4/D2  9 23.6 5.7 0.6 0.1 39 10 105 24 1.4 0.3 75 11 92.0 9.4 1.6 0.4 58 12 25.8 3.1 1.1 0.3 23 13 1160 230 49.2 9.3 24 14 69.0 13 2.9 1.1 24 15 48.4 7.3 1.2 0.1 40 16 267 20 3.0 0.2 4620 200 89 1540 17 319 54 1.8 0.0 16400 2600 177 9110 18 284 48 2.8 0.8 1490 150 101 532 19 249 14 1.8 0.3 1230 330 138 683 20 134 28 11.7 1.0 11 21 28.4 6.4 0.5 0.1 57 22 84.0 5.3 2.5 0.3 34 23 68.4 6.4 1.3 0.4 53 34 200 52 0.9 0.3 225 34 Enantiomer A 441 52 1.1 0.3 400 34 Enantiomer B 1083 379 16.5 3.2 65 35 248.6 62.7 1.37 0.2 1919 373 181 1401 36 28.4 6.5 0.26 0.06 109 37 52.5 4.5 0.5 0.03 105 38 47.1 7.4 62.1 3.1 1 39 337 21.7 4.6 0.6 73 42 185 0.98 188 43 233 5.4 43 44 158 2.2 71 45 697 3.5 200 46 245 1.6 149 47 180 0.65 276 48 244 1.3 185 49 174 1.0 172

TABLE 2 In Vitro Functional Data at D2-like Family Receptor for selected ligandsa IC50 (nM) ± S.E.M. Compd. Structure D2 D3  9   41 ± 8  1.0 ± 0.2 10  1300 ± 320  6.7 ± 2.6 11   368 ± 113 25.6 ± 8.5 12  22.9 ± 2.0   231 ± 1 (31)b  1.2 ± 0.0 14   175 ± 17 42.6 ± 7.9 15   179 ± 48 18.9 ± 4.2 16  15.8 ± 2.7 (26)b  1.0 ± 0.1 (48)b 17 ND 42.0 ± 1.3 (26)b 18 ND 12.8 ± 1.3 20 ND  3.9 ± 1.4 (47)b 21  88.3 ± 15.7  5.4 ± 1.8  3.4 ± 0.5 (20)b aData were obtained through the NIDA Addiction Treatment Discovery Program contract with SRI (N01DA-1-8816). bpartial agonist activity: EC50 (% stimulation)

TABLE 3 Binding affinities for a serotonin receptor subtypes for selected ligands.a Ki(nM) ± S.E.M. Compd. Structure 5HT1A 5HT2A 5HT2C D3 5HT1A/D3 5HT2A/D3 5HT2C/D  9  29.7 ± 0.2 15.5 ± 3.8  10.6 ± 0.0  0.6 ± 0.1  50  26  18 10   309 ± 39 92.7 ± 16  96.4 ± 1.1  1.4 ± 0.3  221  66  69 11   92 ± 8.3 46.9 ± 5.4   31 ± 2.3  1.6 ± 0.4  58  29  19 12  60.6 ± 4.3 57.9 ± 0.6  74.3 ± 3  1.1 ± 0.3  55  53  68 14  21.7 ± 3.5 75.3 ± 1.1   256 ± 33  2.9 ± 1.1   7  26  88 15  71.8 ± 2.1 65.7 ± 12   176 ± 14  1.2 ± 0.1  60  55  147 16  34.3 ± 0.3 42.3 ± 9.1   115 ± 20  3.0 ± 0.2  11  14  38 17  1810 ± 350  545 ± 150  2830 ± 360  1.8 ± 0.0 1006 303 1572 18  36.2 ± 7  695 ± 20  3940 ± 130  2.8 ± 0.8  13 248 1407 20   117 ± 12 88.7 ± 7.5  84.3 ± 5.3 11.7 ± 1.0  10  7   7 21  15.1 ± 2.4 15.4 ± 2.8  25.2 ± 3.2  0.5 ± 0.1  30  31  50 aData were obtained through NIDA Addiction Treatment Discovery Program contract with SRI (N01DA-1-8816).

Claims

1. A method of treating an addiction comprising: administering a pharmaceutically effective amount of a compound of the following chemical formula:

wherein: A=CHR4 or trans CH═CH; n=0 or 1; R1 and R2=independently represent hydrogen, halogen, or alkoxy; R3 and R4=H, OH, OAc, alkoxy, halogen, amino, nitro, alkyl having from 2-8 carbons, or pyridyl; R5=phenyl, indole, thiophene, benzofuran, fluorenyl, or 2-pyridylphenyl; and R5 is optionally substituted with one or more of hydrogen, halogen, amino, nitro, hydroxyl, alkoxy, alkyl, and pyridyl, substitution may occur at any of the ortho, meta, or para positions; including all enantiomers and pharmaceutical salts thereof, wherein alkyl is a branched or unbranched, saturated or unsaturated, monovalent hydrocarbon radical having from 1-8 carbons, optionally substituted with one or more C1 to C3 alkyl, halogen, hydroxyl, amino, alkoxyl, or mercapto groups, and wherein alkoxy is —OR group, wherein R is a C1 to C3 alkyl optionally substituted with one or more C1 to C3 alkyl, halogen, hydroxyl, amino, alkoxyl, or mercapto groups; wherein when A=trans CH═CH, then R3=OH, OAc, alkoxy, halogen, amino, nitro, alkyl, or pyridyl; and wherein when A=CHR4, then one of R3 or R4=OH, OAc, alkoxy, halogen, amino, nitro, alkyl, or pyridyl, to a patient in need thereof.

2. The method of claim 1, in which A is CHR4.

3. The method of claim 1, wherein the compound is:

N-(3-hydroxy-4-(4-(2-methoxyphenyl)-piperazin-1-yl)-butyl)-4-pyridin-2-yl-benzamide;
N-(3-hydroxy-4-(4-(2-methoxyphenyl)piperazin-1-yl)-butyl)-9H-fluorene-2-carboxamide;
N-(4-(4-(2,3-dichlorophenyl)piperazin-1-yl)-3-hydroxybutyl)-1H-indole-2-carboxamide;
N-(3-hydroxy-4-(4-(2-methoxyphenyl)piperazin-1-yl)butyl)-1H-indole-2-carboxamide;
N-(4-(4-(2,3-dichlorophenyl)piperazin-1-yl)-3-hydroxybutyl)-5-iodo-benzofuran-2 carboxamide;
N-(4-(4-(2,3-dichlorophenyl)piperazin-1-yl)-3-hydroxybutyl)-5-fluorobenzo furan-2 carboxamide;
5-fluoro-N-(3-hydroxy-4-(4-(2-methoxyphenyl)piperazin-1-yl)butyl)-1H-indole-2-carboxamide;
N-(4-(4-(2,3-dichlorophenyl)piperazin-1-yl)-3-hydroxybutyl)-5-methoxy-1H-indole-2-carboxamide;
(R)—N-(4-(4-(2,3-dichlorophenyl)piperazin-1-yl)-3-hydroxybutyl)-1H-indole-2-carboxamide; or
(S)—N-(4-(4-(2,3-dichlorophenyl)piperazin-1-yl)-3-hydroxybutyl)-1H-indole-2-carboxamide.

4. The method of claim 1, wherein R3=OH, OAc, alkoxy, halogen, amino, nitro, alkyl having from 2-8 carbons, or pyridyl.

5. The method of claim 1, wherein R4=OH, OAc, alkoxy, halogen, amino, nitro, alkyl having from 2-8 carbons, or pyridyl.

6. The method of claim 1, wherein

A=CHR4;
n=1;
R1 and R2=independently represent hydrogen, halogen, or alkoxy;
R3=OH or halogen;
R4=H; and
R5=indole or fluorenyl, and R5 is optionally substituted with alkoxy.

7. The method of claim 1, wherein

A=CHR4;
n=1;
R1 and R2=Cl;
R3=F;
R4=H; and
R5=indole or fluorenyl, and R5 is optionally substituted with methoxy.

8. The method of claim 1, wherein

A=CHR4;
n=1;
R1=H;
R2=methoxy;
R3=OH;
R4=H; and
R5=indole substituted with methoxy.

9. A method of treating schizophrenia or Parkinson's disease or dyskinesias associated with these diseases comprising: administering a pharmaceutically effective amount of a compound of the following chemical formula:

wherein: A=CHR4 or trans CH═CH; n=0 or 1; R1 and R2=independently represent hydrogen, halogen, or alkoxy; R3 and R4=H, OH, OAc, alkoxy, halogen, amino, nitro, alkyl having from 2-8 carbons, or pyridyl; R5=phenyl, indole, thiophene, benzofuran, fluorenyl, or 2-pyridylphenyl; and R5 is optionally substituted with one or more of hydrogen, halogen, amino, nitro, hydroxyl, alkoxy, alkyl, and pyridyl, substitution may occur at any of the ortho, meta, or para positions; including all enantiomers and pharmaceutical salts thereof, wherein alkyl is a branched or unbranched, saturated or unsaturated, monovalent hydrocarbon radical having from 1-8 carbons, optionally substituted with one or more C1 to C3 alkyl, halogen, hydroxyl, amino, alkoxyl, or mercapto groups, and wherein alkoxy is —OR group, wherein R is a C1 to C3 alkyl optionally substituted with one or more C1 to C3 alkyl, halogen, hydroxyl, amino, alkoxyl, or mercapto groups; wherein when A=trans CH═CH, then R3=OH, OAc, alkoxy, halogen, amino, nitro, alkyl, or pyridyl; and
wherein when A=CHR4, then one of R3 or R4=OH, OAc, alkoxy, halogen, amino, nitro, alkyl, or pyridyl,
to a patient in need thereof.

10. The method of claim 9, in which A is CHR4.

11. The method of claim 9, wherein the compound is:

N-(3-hydroxy-4-(4-(2-methoxyphenyl)-piperazin-1-yl)-butyl)-4-pyridin-2-yl-benzamide;
N-(3-hydroxy-4-(4-(2-methoxyphenyl)piperazin-1-yl)-butyl)-9H-fluorene-2-carboxamide;
N-(4-(4-(2,3-dichlorophenyl)piperazin-1-yl)-3-hydroxybutyl)-1H-indole-2-carboxamide;
N-(3-hydroxy-4-(4-(2-methoxyphenyl)piperazin-1-yl)butyl)-1H-indole-2-carboxamide;
N-(4-(4-(2,3-dichlorophenyl)piperazin-1-yl)-3-hydroxybutyl)-5-iodo-benzofuran-2 carboxamide;
N-(4-(4-(2,3-dichlorophenyl)piperazin-1-yl)-3-hydroxybutyl)-5-fluorobenzofuran-2 carboxamide;
5-fluoro-N-(3-hydroxy-4-(4-(2-methoxyphenyl)piperazin-1-yl)butyl)-1H-indole-2-carboxamide;
N-(4-(4-(2,3-dichlorophenyl)piperazin-1-yl)-3-hydroxybutyl)-5-methoxy-1H-indole-2-carboxamide;
(R)—N-(4-(4-(2,3-dichlorophenyl)piperazin-1-yl)-3-hydroxybutyl)-1H-indole-2-carboxamide; or
(S)—N-(4-(4-(2,3-dichlorophenyl)piperazin-1-yl)-3-hydroxybutyl)-1H-indole-2-carboxamide.

12. The method of claim 9, wherein R3=OH, OAc, alkoxy, halogen, amino, nitro, alkyl having from 2-8 carbons, or pyridyl.

13. The method of claim 9, wherein R4=OH, OAc, alkoxy, halogen, amino, nitro, alkyl having from 2-8 carbons, or pyridyl.

14. The method of claim 9, wherein

A=CHR4;
n=1;
R1 and R2=independently represent hydrogen, halogen, or alkoxy;
R3=OH or halogen;
R4=H; and
R5=indole or fluorenyl, and R5 is optionally substituted with alkoxy.

15. The method of claim 9, wherein

A=CHR4;
n=1;
R1 and R2=Cl;
R3=F;
R4=H; and
R5=indole or fluorenyl, and R5 is optionally substituted with methoxy.

16. The method of claim 9, wherein

A=CHR4;
n=1;
R1=H;
R2=methoxy;
R3=OH;
R4=H; and
R5=indole substituted with methoxy.

17. A method of selectively imaging dopamine D2 family binding sites of the central nervous system to detect or monitor a disease resulting from abnormal distribution and/or density of dopamine D3 receptor in the central nervous system of a patient comprising:

contacting central nervous system tissue with a compound of the following chemical formula:
wherein: A=CHR4 or trans CH═CH; n=0 or 1; R1 and R2=independently represent hydrogen, halogen, or alkoxy; R3 and R4=H, OH, OAc, alkoxy, halogen, amino, nitro, alkyl having from 2-8 carbons, or pyridyl; R5=phenyl, indole, thiophene, benzofuran, fluorenyl, or 2-pyridylphenyl; and R5 is optionally substituted with one or more of hydrogen, halogen, amino, nitro, hydroxyl, alkoxy, alkyl, and pyridyl, substitution may occur at any of the ortho, meta, or para positions; including all enantiomers and pharmaceutical salts thereof, wherein alkyl is a branched or unbranched, saturated or unsaturated, monovalent hydrocarbon radical having from 1-8 carbons, optionally substituted with one or more C1 to C3 alkyl, halogen, hydroxyl, amino, alkoxyl, or mercapto groups, and wherein alkoxy is —OR group, wherein R is a C1 to C3 alkyl optionally substituted with one or more C1 to C3 alkyl, halogen, hydroxyl, amino, alkoxyl, or mercapto groups; wherein when A=trans CH═CH, then R3=OH, OAc, alkoxy, halogen, amino, nitro, alkyl, or pyridyl; and wherein when A=CHR4, then one of R3 or R4=OH, OAc, alkoxy, halogen, amino, nitro, alkyl, or pyridyl; and detecting the binding of the compound to the central nervous system tissue.

18. The method of claim 17, further comprising:

determining the distribution and/or density of the dopamine D3 receptor in the central nervous system tissue;
comparing the distribution and/or density obtained with the distribution and/or density of dopamine D3 receptor in a corresponding normal tissue; and
diagnosing a disease state by a difference in the distribution and/or density between the normal tissue and the subject tissue.
Patent History
Publication number: 20140296249
Type: Application
Filed: Apr 30, 2014
Publication Date: Oct 2, 2014
Applicants: The United States of America, as Represented by the Secretary, Department of Health and Human Serv (Bethesda, MD), THE UNIVERSITY OF NORTH TEXAS HEALTH SCIENCE AT FORT WORTH (Fort Worth, TX)
Inventors: Amy Hauck Newman (Pheonix, MD), Peter Grundt (Duluth, MN), George C. Cyriac (Severna Park, MD), Robert Luedtke (Fort Worth, TX), Jianjing Cao (Ellicott City, MD)
Application Number: 14/266,076