US20200237688A1 - Vesicular monoamine transporter-2 ligands and their use in the treatment of psychostimulant abuse - Google Patents

Vesicular monoamine transporter-2 ligands and their use in the treatment of psychostimulant abuse Download PDF

Info

Publication number
US20200237688A1
US20200237688A1 US16/846,989 US202016846989A US2020237688A1 US 20200237688 A1 US20200237688 A1 US 20200237688A1 US 202016846989 A US202016846989 A US 202016846989A US 2020237688 A1 US2020237688 A1 US 2020237688A1
Authority
US
United States
Prior art keywords
pharmaceutically acceptable
methyl
group
racemate
enantiomer
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
US16/846,989
Inventor
Linda P. Dwoskin
Peter Anthony Crooks
Guangrong Zheng
Justin R. NICKELL
Zheng CAO
Na-ra Lee
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of Kentucky Research Foundation
Original Assignee
University of Kentucky Research Foundation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by University of Kentucky Research Foundation filed Critical University of Kentucky Research Foundation
Priority to US16/846,989 priority Critical patent/US20200237688A1/en
Publication of US20200237688A1 publication Critical patent/US20200237688A1/en
Assigned to UNIVERSITY OF KENTUCKY RESEARCH FOUNDATION reassignment UNIVERSITY OF KENTUCKY RESEARCH FOUNDATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CAO, ZHENG, LEE, NA-RA, Nickell, Justin R., ZHENG, GUANGRONG, DWOSKIN, LINDA P., CROOKS, PETER ANTHONY
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • A61K31/137Arylalkylamines, e.g. amphetamine, epinephrine, salbutamol, ephedrine or methadone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • A61K31/138Aryloxyalkylamines, e.g. propranolol, tamoxifen, phenoxybenzamine

Definitions

  • the present invention relates to methods treatment of a disease or pathology of the central nervous system, an eating disorder, or substance use disorder, drug dependence/abuse and withdrawal therefrom comprising administering N-phenylalkyl amphetamine derivatives and pharmaceutical compositions containing these compounds to an individual in need thereof.
  • DA dopamine
  • NE norepinephrine
  • 5-HT serotonin
  • CNS central nervous system
  • Most neurotransmitters are stored in synaptic vesicles, which are prominent features of nerve terminals. Sequestration into vesicles appears to be responsible for maintaining a ready supply of neurotransmitter molecules available for neuronal exocytotic release into the synaptic cleft. Vesicles also serve the role of protecting the neurotransmitter molecules from metabolic breakdown.
  • VMAT2 vesicular monoamine transporter-2
  • the neurotransmitter Once the neurotransmitter is released from the terminal into the synaptic space, it interacts with postsynaptic receptors and subsequently is taken back up into the terminal via the plasma membrane transporter (e.g., DAT and/or the serotonin transporter [SERT]).
  • the plasma membrane transporter e.g., DAT and/or the serotonin transporter [SERT]
  • transporter proteins modify the concentration of neurotransmitters in the cytosolic and vesicular storage pools, and thereby have the ability to alter subsequent neurotransmission.
  • the present invention provides a compound of formula (I):
  • n is an integer in the range from 1 to 3;
  • n is zero or an integer in the range from 1 to 5;
  • R 1 is an aryl group which may be substituted by one or more substituents
  • R 2 is an aryl group which may be substituted by one or more substituents; wherein substituents on R 1 and R 2 are independently selected from the group consisting of methyl; deuteromethyl (CD 3 ); tritiomethyl (CT 3 ); ethyl; propyl; isopropyl; C 4 -C 7 straight chain or branched alkyl; C 3 -C 6 cycloalkyl; C 4 -C 7 alkenyl (including cis and trans geometrical forms); benzyl; phenylethyl; amino; N-methylamino; N,N-dimethylamino; carboxylate; methylcarboxylate; ethylcarboxylate; propylcarboxylate; isopropylcarboxylate; carboxaldehyde; acetoxy; propionyloxy; isopropionyloxy; cyano; aminomethyl; N-methylaminomethyl; N,N-dimethylaminomethyl
  • R 3 is a methyl, ethyl, propyl, isopropyl, hydroxymethyl, 2-hydroxyethyl, 1-hyhydroxyethyl, methoxymethyl, 2-methoxyethyl, 1-methoxyethyl, aminomethyl, 2-aminoethyl, 1-aminoethyl, N-methylaminomethyl, 2-N-methylaminoethyl, 1-N-methylaminoethyl, N,N-dimethylaminomethyl, 2-N,N-dimethylaminoethyl, or 1-N,N-dimethylaminoethyl group; and
  • R 4 is a hydrogen atom, a methyl, ethyl, propyl, or isopropyl group.
  • the compound of formula (I) may form pharmaceutically acceptable salts with a variety of acids.
  • the present invention provides a compound of formula (I):
  • n is an integer in the range from 1 to 3;
  • n is zero or an integer from 1 to 5;
  • R 1 and R 2 are each independently an aryl group
  • R 1 and R 2 are each independently unsubstituted or substituted by one or more substituents selected from the group consisting of methyl; deuteromethyl (CD 3 ); tritiomethyl (CT 3 ); ethyl; propyl; isopropyl; C 4 -C 7 straight chain or branched alkyl; C 3 -C 6 cycloalkyl; C 4 -C 7 alkenyl; benzyl; phenylethyl; amino; N-methylamino; N,N-dimethylamino; carboxylate; methylcarboxylate; ethylcarboxylate; propylcarboxylate; isopropylcarboxylate; carboxaldehyde; acetoxy; propionyloxy; isopropionyloxy; cyano; aminomethyl; N-methylaminomethyl; N,N-dimethylaminomethyl; carboxamide; N-methylcarboxamide; N,N-dimethyl
  • R 3 is methyl, ethyl, propyl, isopropyl, hydroxymethyl, 2-hydroxyethyl, 1-hyhydroxyethyl, methoxymethyl, 2-methoxyethyl, 1-methoxyethyl, aminomethyl, 2-aminoethyl, 1-aminoethyl, N-methylaminomethyl, 2-N-methylaminoethyl, 1-N-methylaminoethyl, N,N-dimethylaminomethyl, 2-N,N-dimethylaminoethyl, or 1-N,N-dimethylaminoethyl group; and
  • R 4 is a hydrogen atom or a methyl, ethyl, propyl, or isopropyl group
  • a further embodiment of the invention is a compound of formula (I), wherein: m is 1 or 2; and n is an integer from 1 to 5; or an enantiomer; racemate; or pharmaceutically acceptable salt thereof.
  • An further embodiment of the invention is a compound of formula (I), wherein: m is 1 or 2; and n is an integer from 1 to 5; R 3 is methyl, ethyl, propyl, or isopropyl; and R 4 is a hydrogen atom or a methyl, ethyl, propyl, or isopropyl group; or an enantiomer; racemate; or pharmaceutically acceptable salt thereof.
  • a further embodiment of the invention is a compound of formula (I), wherein: m is 1 or 2; and n is an integer from 1 to 5; wherein R 3 is methyl; and R 4 is a hydrogen atom or a methyl group; or an enantiomer; racemate; or pharmaceutically acceptable salt thereof.
  • Another embodiment of the invention is a compound of formula (I), wherein m is 1; n is 1; R 1 and R 2 are each independently a phenyl group, wherein R 1 and R 2 are each independently unsubstituted or substituted by one or more substituents selected from the group consisting of benzyl; amino; hydroxyl; methoxy; fluoro; bromo; and nitroso; R 3 is methyl; and R 4 is a hydrogen atom or a methyl group; or an enantiomer; racemate; or pharmaceutically acceptable salt thereof.
  • Another embodiment of the invention is a compound of formula (I), wherein m is 1; n is 2; R 1 and R 2 are each independently a phenyl group, wherein R 1 and R 2 are each independently unsubstituted or substituted by one or more substituents selected from the group consisting of benzyl; amino; hydroxyl; methoxy; fluoro; bromo; and nitroso; R 3 is methyl; and R 4 is a hydrogen atom or a methyl group; or an enantiomer; racemate; or pharmaceutically acceptable salt thereof.
  • Another embodiment of the invention is a compound of formula (I), wherein m is 2; n is 1; R 1 and R 2 are each independently a phenyl group, wherein R 1 and R 2 are each independently unsubstituted or substituted by one or more substituents selected from the group consisting of methoxy and bromo; R 3 is methyl; and R 4 is a hydrogen atom; or an enantiomer; racemate; or pharmaceutically acceptable salt thereof.
  • Yet another embodiment of the invention is a compound of formula (I), wherein m is 2; n is 2; R 1 and R 2 are each independently a phenyl group, wherein R 1 and R 2 are each independently unsubstituted or substituted by one or more methoxy groups; R 3 is methyl; and R 4 is a hydrogen atom or a methyl group; or an enantiomer; racemate; or pharmaceutically acceptable salt thereof.
  • Yet another embodiment of the invention is a compound of formula (I), wherein m is 1; n is an integer from 3 to 5; R 1 and R 2 are each independently a phenyl group, wherein R 1 and R 2 are each independently unsubstituted or substituted by one or more substituents selected from the group consisting of methoxy and bromo; R 3 is methyl; and R 4 is a hydrogen atom; or an enantiomer; racemate; or pharmaceutically acceptable salt thereof.
  • Yet another embodiment of the invention is a compound of formula (I), wherein m is 1; n is 2; R 1 is an unsubstituted phenyl group; R 2 is a phenyl group substituted with a methoxy group; R 3 is methyl; and R 4 is a hydrogen atom; or an enantiomer; racemate; or pharmaceutically acceptable salt thereof.
  • aryl refers to aromatic groups having 6 to 24 carbon atoms
  • substituted aryl refers to aryl groups further bearing one or more substituents.
  • alkyl refers to straight or branched chain alkyl radicals having 1 to 19 carbon atoms
  • substituted alkyl refers to alkyl radicals further bearing one or more substituents.
  • lower alkyl refers to straight or branched chain alkyl radicals having in the range of 1 to 4 carbon atoms.
  • cycloalkyl refers to cyclic ring-containing moieties containing 3 to 8 carbon atoms
  • substituted cycloalkyl refers to cycloalkyl moieties further bearing one or more substituents.
  • alkenyl refers to straight or branched chain hydrocarbyl groups having at least one carbon-carbon double bond and having 2 to 19 carbon atoms
  • substituted alkenyl refers to alkenyl groups further bearing one or more substituents.
  • heterocyclic refers to cyclic moieties containing one or more heteroatoms as part of the ring structure and having 3 to 24 carbon atoms
  • substituted heterocyclic refers to heterocyclic moieties further bearing one or more substituents.
  • salts include Cl ⁇ , Br ⁇ , I ⁇ , NO 2 ⁇ , HSO 4 ⁇ , SO 4 ⁇ , HPO 4 ⁇ , PO 4 2 ⁇ , ethanesulfonate, trifluromethane sulfate, p-toluenesulfonate, benzenesulfonate, salicylate, proprionate, ascorbate, aspartate, fumarate, galactarate, maleate, citrate, glutamate, glycolate, lactate, malate, maleate, tartrate, oxalate, succinate, or similar acid addition salts.
  • the above salt forms may be in some cases hydrates or solvates with alcohols and other solvents.
  • compositions comprising a compound of formula (I) and pharmaceutically acceptable excipients.
  • the pharmaceutical composition may include a pharmaceutically acceptable additive, such as a stabilizer, buffer, salt, preservative, filler, flavor enhancer and the like, as known to those skilled in the art.
  • a pharmaceutically acceptable additive such as a stabilizer, buffer, salt, preservative, filler, flavor enhancer and the like, as known to those skilled in the art.
  • Representative buffers include phosphates, carbonates, and citrates.
  • Exemplary preservatives include EDTA, EGTA, BHA, and BHT.
  • the pharmaceutical composition disclosed herein may be administered by inhalation (i.e., intranasally as an aerosol or nasal formulation); topically (i.e., in the form of an ointment, cream or lotion); orally (i.e., in solid or liquid form (tablet, capsule, gel cap, time release capsule, powder, solution, or suspension in aqueous or non-aqueous liquid); intravenously as an infusion or injection (i.e., as a solution, suspension or emulsion in a pharmaceutically acceptable carrier); or subcutaneously as an infusion or injection (i.e., as a solution, suspension or emulsion in a pharmaceutically acceptable carrier) or as a depot formulation; transdermally (e.g., via a transdermal patch), or rectally (e.g., as a suppository).
  • inhalation i.e., intranasally as an aerosol or nasal formulation
  • topically i.e., in the form of an ointment
  • the composition may be administered in accordance with specific form of the preparation, age, sex and the other conditions of a patient, severity of disease, etc.
  • the composition is orally administered.
  • the composition is intravenously administered alone or in a mixture with conventional replacement fluid such as glucose and amino acids, and if necessary, and the preparation alone may be also administered intramuscularly, intracutaneously, subcutaneously or interperitoneally.
  • a compound of formula (I) or a pharmaceutically acceptable salt thereof can be administered subcutaneously, intramuscularly, intravenously, transdermally, orally, intranasally, intrapulmonary, or rectally.
  • the dose of the pharmaceutical composition of the present invention is appropriately selected in accordance with dosage regimen, age, sex and the other conditions of a patient.
  • the amount to be administered depends to some extent on the lipophilicity of the specific compound selected, since it is expected that this property of the compound will cause it to partition into fat deposits of the subject.
  • the precise amount to be administered can be determined by the skilled practitioner in view of desired dosages, side effects and medical history of the patient and the like.
  • the pharmaceutical composition may comprise a compound of formula (I), or an enantiomer or racemate thereof, or pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable additive or a pharmaceutically acceptable excipient.
  • VMAT2 is considered as a valid target for the development of treatments for the abuse of drugs, including, for example, methamphetamine (METH), amphetamine, cocaine, methylphenidate, and opiate abuse.
  • METH methamphetamine
  • amphetamine cocaine
  • methylphenidate opiate abuse.
  • METH can be considered as a structural fragment of lobelane/nor-lobelane, and introducing bulky substituents onto the N-atom of METH afforded compounds with increased inhibitory potency at VMAT2, reduced the psychostimulant effects of METH, diminishing its abuse potential. Importantly, these new analogs did not increase locomotor activity in rats, indicating they do not act as psychostimulants.
  • VMAT2 is a target for the development of treatments for a disease or pathology of the central nervous system, including, for example, cognitive disorders, brain trauma, memory loss, psychosis, sleep disorders, obsessive compulsive disorders, panic disorders, myasthenia gravis, Parkinson's disease, Alzheimer's disease, schizophrenia, Tourette's syndrome, Huntington's disease, attention deficit hyperactivity disorder, hyperkinetic syndrome, chronic nervous exhaustion, narcolepsy, pain, motion sickness, and/or depression.
  • cognitive disorders including, for example, cognitive disorders, brain trauma, memory loss, psychosis, sleep disorders, obsessive compulsive disorders, panic disorders, myasthenia gravis, Parkinson's disease, Alzheimer's disease, schizophrenia, Tourette's syndrome, Huntington's disease, attention deficit hyperactivity disorder, hyperkinetic syndrome, chronic nervous exhaustion, narcolepsy, pain, motion sickness, and/or depression.
  • VMAT2 is also a target for the development of treatments for eating disorders, including, for example, obesity. Compulsive eating is linked to addiction-like neuroadaptive responses in brain reward circuits. Johnson, P. M. et al., “Dopamine D2 receptors in addiction-like reward dysfunction and compulsive eating in obese rats,” Nature Neuroscience, 2010, 13, 635-641. Modulation of VMAT2 may lead to reduced reward responses, diminishing overeating.
  • a more preferred embodiment of the invention is a compound of formula (I), wherein the compound is 3-(4-methoxyphenyl)-N-(1-phenylpropan-2-yl)propan-1-amine or an enantiomer; racemate; or pharmaceutically acceptable salt thereof.
  • An even more preferred embodiment of the invention is a compound of formula (I), wherein the compound is (S)-3-(4-methoxyphenyl)-N-(1-phenylpropan-2-yl)propan-1-amine hydrochloride.
  • a more preferred embodiment of the invention is a pharmaceutical composition comprising 3-(4-methoxyphenyl)-N-(1-phenylpropan-2-yl)propan-1-amine or an enantiomer; racemate; or pharmaceutically acceptable salt thereof and a pharmaceutically acceptable additive or a pharmaceutically acceptable excipient.
  • An even more preferred embodiment of the invention is a pharmaceutical composition comprising (S)-3-(4-methoxyphenyl)-N-(1-phenylpropan-2-yl)propan-1-amine hydrochloride and a pharmaceutically acceptable additive or a pharmaceutically acceptable excipient.
  • Another preferred embodiment of the invention is a method of treating a disease or pathology of the central nervous system or an eating disorder in an individual in need thereof, wherein the method comprises the step of administering to the individual a compound of formula (I) or a pharmaceutically acceptable salt thereof.
  • the compounds of the present invention may be used in a method of treating a disease or pathology of the central nervous system, wherein the disease may be cognitive disorders, brain trauma, memory loss, psychosis, sleep disorders, obsessive compulsive disorders, panic disorders, myasthenia gravis, Parkinson's disease, Alzheimer's disease, schizophrenia, Tourette's syndrome, Huntington's disease, attention deficit hyperactivity disorder, hyperkinetic syndrome, chronic nervous exhaustion, narcolepsy, pain, motion sickness, and/or depression.
  • the disease may be cognitive disorders, brain trauma, memory loss, psychosis, sleep disorders, obsessive compulsive disorders, panic disorders, myasthenia gravis, Parkinson's disease, Alzheimer's disease, schizophrenia, Tourette's syndrome, Huntington's disease, attention deficit hyperactivity disorder, hyperkinetic syndrome, chronic nervous exhaustion, narcolepsy, pain, motion sickness, and/or depression.
  • the compounds of the present invention may also be used in a method of treating an eating disorder.
  • the eating disorder may be obesity.
  • the compounds of the present invention may be used in a method of treating substance use disorder, drug dependence/abuse or withdrawal from drug dependence/abuse in an individual in need thereof.
  • Substance use disorder and drug dependence/abuse includes dependence/abuse of psychostimulants or drugs that release dopamine.
  • the drug which the individual is using and/or dependent upon may be amphetamine, methamphetamine, and other drugs that release dopamine.
  • VMAT2 inhibitors were synthesized and evaluated for their activity at VMAT2, dopamine transporters (DAT), and serotonin transporters (SERT) using rat striatum, hERG channels expressed by HEK-293 cells, and for their effect on METH-stimulated locomotor activity in rats.
  • Compounds of the invention exhibited affinity at VMAT2 as well as selectivity at VMAT2 over hERG, DAT, and SERT. Further, a significant reduction of METH-stimulated locomotor activity was observed after administration of the compounds.
  • the resulting light yellow oil was mixed with sodium azide (5.95 g, 91.5 mmol) in DMF (40 mL) and heated at 55° C. for 3 hrs.
  • the reaction mixture was diluted with diethyl ether (150 mL) and washed with water (2 ⁇ 100 mL) and brine (100 mL).
  • the organic layer was dried over anhydrous Na 2 SO 4 , filtered and concentrated under reduced pressure.
  • the crude product was chromatographed on silica (hexanes/ethyl acetate 50:1 to 20:1) to afford (S)-(2-azidopropyl)benzene (4.38 g) as a colorless oil.
  • Synaptic vesicles were prepared from rat brain using a modification of a previously described procedure (Teng et al., 1998). Briefly, fresh whole brain (excluding cerebellum) was homogenized using a Teflon pestle (clearance 0.003 inches) with 7 vertical strokes at 800 rpm in 20 vol of ice-cold 0.32 M sucrose and centrifuged at 1000 g for 12 min at 4° C. The resulting supernatant (S 1 ) was then centrifuged at 22,000 g for 10 min at 4° C. The synaptosomal pellets (P 2 ) were homogenized in 18 mL of ice-cold Milli-Q water and exposed for 5 min for lysing synaptosomes.
  • [ 3 H]Dofetilide binding assays were conducted using commercially available HEK-293 cell membranes which stably express the hERG channel. Membranes were suspended in assay buffer (50 mM Tris, 10 mM KCl, 1 mM MgCl 2 , pH 7.4) prior to the experiment. Assays were performed in duplicate in a total volume of 250 ⁇ L. Aliquots of the HEK-293 cell membrane suspension which contained 5 ⁇ g membrane protein were added to tubes containing assay buffer, 5 nM [ 3 H]dofetilide and a range of concentrations of analog (10 nM-100 ⁇ M). Nonspecific binding was determined in the presence of amitriptyline (1 mM).
  • [ 3 H]DA and [ 3 H]5-HT uptake into striatal synaptosomes was determined to evaluate compound inhibition of the dopamine transporter (DAT) and the serotonin transporter (SERT), respectively. Striata from individual rats were homogenized in ice-cold sucrose solution containing 5 mM NaHCO 3 (pH 7.4), with 16 up-and-down strokes of a Teflon pestle homogenizer (clearance ⁇ 0.003′′). Homogenates were centrifuged at 2000 g for 10 min at 4° C., and resulting supernatants were centrifuged at 20,000 g for 17 min at 4° C.
  • DAT dopamine transporter
  • SERT serotonin transporter
  • Pellets were resuspended in 2.4 mL (for DAT assays) or 1.5 mL (for SERT assays) of assay buffer (125 mM NaCl, 5 mM KCl, 1.5 mM MgSO 4 , 1.25 mM CaCl 2 , 1.5 mM KH 2 PO 4 , 10 mM alpha-D-glucose, 25 mM HEPES, 0.1 mM EDTA, 0.1 mM pargyline, 0.1 mM ascorbic acid, saturated with 95% O 2 /5% CO 2 , pH 7.4).
  • assay buffer 125 mM NaCl, 5 mM KCl, 1.5 mM MgSO 4 , 1.25 mM CaCl 2 , 1.5 mM KH 2 PO 4 , 10 mM alpha-D-glucose, 25 mM HEPES, 0.1 mM EDTA, 0.1 mM pargyline, 0.1
  • Assays were performed in duplicate in a total volume of 500 ⁇ L (for DAT assays) or 250 ⁇ L (for SERT assays). Aliquots of the synaptosomal suspension (25 ⁇ L for DAT, 50 ⁇ L for SERT) were added to tubes containing assay buffer and various concentrations of analog (1 nM-100 ⁇ M), and incubated at 34° C. for 5 min. Nonspecific uptake was determined in the presence of nomifensine (10 ⁇ M) for DAT assays or fluoxetine (10 ⁇ M) for SERT assays.
  • GBR-12935 (100 nM) was included in the assay buffer for the SERT assay to maximally inhibit [ 3 H]5-HT uptake through DAT and isolate uptake to SERT.
  • Samples were placed on ice, and 50 ⁇ L of 0.1 ⁇ M [ 3 H]DA (for DAT assays) or 25 ⁇ L of 0.1 ⁇ M [ 3 H]5-HT (for SERT assays) was added to each tube, and incubated for 10 min at 34° C.
  • Exemplary compounds 1-53 were tested in [ 3 H]dihydrotetrabenazine ([ 3 H]DTBZ) binding assay according to Example 2 and the [ 3 H]dopamine ([ 3 H]DA) uptake assay according to Example 3. The results of these assays are set forth in Table 1.
  • VMAT2 Compound Structure
  • VMAT2 ( ⁇ M) ( ⁇ M) 1 7.70 ⁇ 1.25 0.063 ⁇ 0.005 2 7.53 ⁇ 2.16 0.51 ⁇ 0.10 3 1.23 ⁇ 0.13 0.033 ⁇ 0.007 4 1.13 ⁇ 0.31 0.007 ⁇ 0.002 5 0.91 ⁇ 0.59 0.006 ⁇ 0.001 6 0.75 ⁇ 0.14 0.065 ⁇ 0.004 7 1.56 ⁇ 0.47 0.096 ⁇ 0.010 8 2.2 ⁇ 0.11 0.014 ⁇ 0.003 9 — 0.0087 ⁇ 0.0065 10 — 0.026 ⁇ 0.0036 11 0.70 ⁇ 0.063 0.008 ⁇ 0.001 12 — 0.006 ⁇ 0.001 13 — 0.032 ⁇ 0.004 14 0.081 ⁇ 0.016 0.003 ⁇ 0.0003 15 0.19 ⁇ 0.020 0.003 ⁇ 0.002 16
  • compounds of the invention exhibited high affinity at VMAT2 as well as selectivity at VMAT2 over hERG, DAT, and SERT.
  • Compound 9 exhibited a K i value of 8.71 ⁇ 3.65 at VMAT2
  • Compound 10 exhibited a K i value of 25.5 ⁇ 3.57 nM at VMAT2.
  • both Compound 9 and Compound 10 exhibited a >30-fold selectivity at VMAT2 over hERG, DAT, and SERT.
  • the compounds of the invention were also evaluated for their effect on METH-stimulated locomotor activity was evaluated in rats.
  • the compounds were observed to exhibit a significant reduction of METH-stimulated locomotor activity after administration.
  • a significant reduction of METH-stimulated locomotor activity was observed after administration of Compound 9 (3 mg/kg, s.c.) and Compound 10 (17 mg/kg, s.c.).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Emergency Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

The present invention relates to methods of treatment of a disease or pathology of the central nervous system, an eating disorder, or substance use disorder, drug dependence/abuse and withdrawal therefrom comprising administering at least one N-phenylalkyl amphetamine derivative and pharmaceutical compositions comprising at least one N-phenylalkyl amphetamine derivative to an individual in need thereof.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • The present application is a continuation of U.S. application Ser. No. 15/493,836, filed Apr. 21, 2017, which claims the benefit of U.S. Provisional Application No. 62/325,875, filed Apr. 21, 2016, both of which are herein incorporated by reference in their entireties.
  • FIELD OF THE INVENTION
  • The present invention relates to methods treatment of a disease or pathology of the central nervous system, an eating disorder, or substance use disorder, drug dependence/abuse and withdrawal therefrom comprising administering N-phenylalkyl amphetamine derivatives and pharmaceutical compositions containing these compounds to an individual in need thereof.
  • BACKGROUND OF THE INVENTION
  • The action of many therapeutic neuropharmacological agents involve the modulation of dopamine (DA), norepinephrine (NE) and serotonin (5-HT) release, uptake and storage within their respective terminals in the central nervous system (CNS). Most neurotransmitters are stored in synaptic vesicles, which are prominent features of nerve terminals. Sequestration into vesicles appears to be responsible for maintaining a ready supply of neurotransmitter molecules available for neuronal exocytotic release into the synaptic cleft. Vesicles also serve the role of protecting the neurotransmitter molecules from metabolic breakdown. One transport site on the vesicle membrane is the vesicular monoamine transporter-2 (VMAT2), whose role is to transport transmitters from the cytosol into the synaptic vesicle. Once the neurotransmitter is released from the terminal into the synaptic space, it interacts with postsynaptic receptors and subsequently is taken back up into the terminal via the plasma membrane transporter (e.g., DAT and/or the serotonin transporter [SERT]). Thus, transporter proteins modify the concentration of neurotransmitters in the cytosolic and vesicular storage pools, and thereby have the ability to alter subsequent neurotransmission.
  • SUMMARY OF THE INVENTION
  • The present invention provides a compound of formula (I):
  • Figure US20200237688A1-20200730-C00001
  • wherein
  • m is an integer in the range from 1 to 3;
  • n is zero or an integer in the range from 1 to 5;
  • R1 is an aryl group which may be substituted by one or more substituents;
  • R2 is an aryl group which may be substituted by one or more substituents; wherein substituents on R1 and R2 are independently selected from the group consisting of methyl; deuteromethyl (CD3); tritiomethyl (CT3); ethyl; propyl; isopropyl; C4-C7 straight chain or branched alkyl; C3-C6 cycloalkyl; C4-C7 alkenyl (including cis and trans geometrical forms); benzyl; phenylethyl; amino; N-methylamino; N,N-dimethylamino; carboxylate; methylcarboxylate; ethylcarboxylate; propylcarboxylate; isopropylcarboxylate; carboxaldehyde; acetoxy; propionyloxy; isopropionyloxy; cyano; aminomethyl; N-methylaminomethyl; N,N-dimethylaminomethyl; carboxamide; N-methylcarboxamide; N,N-dimethylcarboxamide; acetyl; propionyl; formyl; benzoyl sulfate; phenyl; methylsulfate; hydroxyl; methoxy; ethoxy; propoxy; isopropoxy; thiol; methylthio; ethylthio; propiothiol; fluoro; chloro; bromo; iodo; trifluoromethyl; vinyl; allyl; propargyl; nitro; carbamoyl; ureido; azido; isocyanate; thioisocyanate; hydroxylamino; nitroso; a saturated or unsaturated hydrocarbon ring; a nitrogen containing heterocyclic moiety; an oxygen containing heterocyclic moiety; a sulfur containing heterocyclic moiety; a selenium containing heterocyclic moiety; a mixed heterocyclic moiety containing at least two atoms selected from the group consisting of nitrogen, oxygen and sulfur; and ortho, meta or para-substituted benzene;
  • R3 is a methyl, ethyl, propyl, isopropyl, hydroxymethyl, 2-hydroxyethyl, 1-hyhydroxyethyl, methoxymethyl, 2-methoxyethyl, 1-methoxyethyl, aminomethyl, 2-aminoethyl, 1-aminoethyl, N-methylaminomethyl, 2-N-methylaminoethyl, 1-N-methylaminoethyl, N,N-dimethylaminomethyl, 2-N,N-dimethylaminoethyl, or 1-N,N-dimethylaminoethyl group; and
  • R4 is a hydrogen atom, a methyl, ethyl, propyl, or isopropyl group.
  • The compound of formula (I) may form pharmaceutically acceptable salts with a variety of acids.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention provides a compound of formula (I):
  • Figure US20200237688A1-20200730-C00002
  • wherein
  • m is an integer in the range from 1 to 3;
  • n is zero or an integer from 1 to 5;
  • R1 and R2 are each independently an aryl group,
  • wherein R1 and R2 are each independently unsubstituted or substituted by one or more substituents selected from the group consisting of methyl; deuteromethyl (CD3); tritiomethyl (CT3); ethyl; propyl; isopropyl; C4-C7 straight chain or branched alkyl; C3-C6 cycloalkyl; C4-C7 alkenyl; benzyl; phenylethyl; amino; N-methylamino; N,N-dimethylamino; carboxylate; methylcarboxylate; ethylcarboxylate; propylcarboxylate; isopropylcarboxylate; carboxaldehyde; acetoxy; propionyloxy; isopropionyloxy; cyano; aminomethyl; N-methylaminomethyl; N,N-dimethylaminomethyl; carboxamide; N-methylcarboxamide; N,N-dimethylcarboxamide; acetyl; propionyl; formyl; benzoyl sulfate; phenyl; methylsulfate; hydroxyl; methoxy; ethoxy; propoxy; isopropoxy; thiol; methylthio; ethylthio; propiothiol; fluoro; chloro; bromo; iodo; trifluoromethyl; vinyl; allyl; propargyl; nitro; carbamoyl; ureido; azido; isocyanate; thioisocyanate; hydroxylamino; nitroso; a saturated or unsaturated hydrocarbon ring; a nitrogen-containing heterocyclic moiety; an oxygen-containing heterocyclic moiety; a sulfur-containing heterocyclic moiety; a selenium-containing heterocyclic moiety; a mixed heterocyclic moiety containing at least two atoms selected from the group consisting of nitrogen, oxygen and sulfur; and ortho, meta or para-substituted benzene;
  • R3 is methyl, ethyl, propyl, isopropyl, hydroxymethyl, 2-hydroxyethyl, 1-hyhydroxyethyl, methoxymethyl, 2-methoxyethyl, 1-methoxyethyl, aminomethyl, 2-aminoethyl, 1-aminoethyl, N-methylaminomethyl, 2-N-methylaminoethyl, 1-N-methylaminoethyl, N,N-dimethylaminomethyl, 2-N,N-dimethylaminoethyl, or 1-N,N-dimethylaminoethyl group; and
  • R4 is a hydrogen atom or a methyl, ethyl, propyl, or isopropyl group; or
  • an enantiomer; racemate; or pharmaceutically acceptable salt thereof.
  • A further embodiment of the invention is a compound of formula (I), wherein: m is 1 or 2; and n is an integer from 1 to 5; or an enantiomer; racemate; or pharmaceutically acceptable salt thereof.
  • An further embodiment of the invention is a compound of formula (I), wherein: m is 1 or 2; and n is an integer from 1 to 5; R3 is methyl, ethyl, propyl, or isopropyl; and R4 is a hydrogen atom or a methyl, ethyl, propyl, or isopropyl group; or an enantiomer; racemate; or pharmaceutically acceptable salt thereof.
  • A further embodiment of the invention is a compound of formula (I), wherein: m is 1 or 2; and n is an integer from 1 to 5; wherein R3 is methyl; and R4 is a hydrogen atom or a methyl group; or an enantiomer; racemate; or pharmaceutically acceptable salt thereof.
  • Another embodiment of the invention is a compound of formula (I), wherein m is 1; n is 1; R1 and R2 are each independently a phenyl group, wherein R1 and R2 are each independently unsubstituted or substituted by one or more substituents selected from the group consisting of benzyl; amino; hydroxyl; methoxy; fluoro; bromo; and nitroso; R3 is methyl; and R4 is a hydrogen atom or a methyl group; or an enantiomer; racemate; or pharmaceutically acceptable salt thereof.
  • Another embodiment of the invention is a compound of formula (I), wherein m is 1; n is 2; R1 and R2 are each independently a phenyl group, wherein R1 and R2 are each independently unsubstituted or substituted by one or more substituents selected from the group consisting of benzyl; amino; hydroxyl; methoxy; fluoro; bromo; and nitroso; R3 is methyl; and R4 is a hydrogen atom or a methyl group; or an enantiomer; racemate; or pharmaceutically acceptable salt thereof.
  • Another embodiment of the invention is a compound of formula (I), wherein m is 2; n is 1; R1 and R2 are each independently a phenyl group, wherein R1 and R2 are each independently unsubstituted or substituted by one or more substituents selected from the group consisting of methoxy and bromo; R3 is methyl; and R4 is a hydrogen atom; or an enantiomer; racemate; or pharmaceutically acceptable salt thereof.
  • Yet another embodiment of the invention is a compound of formula (I), wherein m is 2; n is 2; R1 and R2 are each independently a phenyl group, wherein R1 and R2 are each independently unsubstituted or substituted by one or more methoxy groups; R3 is methyl; and R4 is a hydrogen atom or a methyl group; or an enantiomer; racemate; or pharmaceutically acceptable salt thereof.
  • Yet another embodiment of the invention is a compound of formula (I), wherein m is 1; n is an integer from 3 to 5; R1 and R2 are each independently a phenyl group, wherein R1 and R2 are each independently unsubstituted or substituted by one or more substituents selected from the group consisting of methoxy and bromo; R3 is methyl; and R4 is a hydrogen atom; or an enantiomer; racemate; or pharmaceutically acceptable salt thereof.
  • Yet another embodiment of the invention is a compound of formula (I), wherein m is 1; n is 2; R1 is an unsubstituted phenyl group; R2 is a phenyl group substituted with a methoxy group; R3 is methyl; and R4 is a hydrogen atom; or an enantiomer; racemate; or pharmaceutically acceptable salt thereof.
  • Compounds of formula (I) include the following compounds:
  • 1. m=1, n=1, R1=Ph, R2=Ph, R3=Me, R4═H
    2. m=1, n=1, R1=Ph, R2=Ph, R3=Me, R4═CH3
    3. m=1, n=1, R1=4-BrPh, R2=Ph, R3=Me, R4═H
    4. m=1, n=2, R1=Ph, R2=Ph, R3=Me, R4═H
    5. R-, m=1, n=2, R1=Ph, R2=Ph, R3=Me, R4═H
    6. S-, m=1, n=2, R1=Ph, R2=Ph, R3=Me, R4═H
    7. m=1, n=2, R1=Ph, R2=Ph, R3=Me, R4═CH3
    8. m=1, n=2, R1=Ph, R2=4-MeOPh, R3=Me, R4═H
    9. R-, m=1, n=2, R1=Ph, R2=4-MeOPh, R3=Me, R4═H
    10. S-, m=1, n=2, R1=Ph, R2=4-MeOPh, R3=Me, R4═H
    11. m=1, n=2, R1=4-BrPh, R2=Ph, R3=Me, R4═H
    12. R-, m=1, n=2, R1=4-BrPh, R2=Ph, R3=Me, R4═H
    13. S-, m=1, n=2, R1=4-BrPh, R2=Ph, R3=Me, R4═H
    14. m=1, n=3, R1=4-BrPh, R2=Ph, R3=Me, R4═H
    15. m=1, n=3, R1=Ph, R2=Ph, R3=Me, R4═H
    16. m=1, n=2, R1=4-BrPh, R2=4-MeOPh, R3=Me, R4═H
    17. m=1, n=4, R1=Ph, R2=Ph, R3=Me, R4═H
    18. m=1, n=5, R1=Ph, R2=Ph, R2=Me, R4═H
    19. m=1 n=1, R1=Ph, R2=PhO, R3=Me, R4═H
    20. m=1, n=1, R1=4-BrPh, R2=PhO, R3=Me, R4═H
    21. m=1, n=1, R1=Ph, R2=4-OHPh, R3=Me, R4═H
    22. m=1, n=1, R1=Ph, R2=3,4-diBnOPh, R3=Me, R4═H
    23. m=1, n=1, R1=Ph, R2=3,4-diOHPh, R3=Me, R4═H
    24. m=1, n=1, R1=4-MeOPh, R2=Ph, R3=Me, R4═H
    25. m=1, n=1, R1=4-MeOPh, R2=Ph, R3=Me, R4═CH3
    26. m=1, n=2, R1=4-MeOPh, R2=Ph, R3=Me, R4═H
    27. R-, m=1, n=2, R1=4-MeOPh, R2=Ph, R3=Me, R4═H
    28. S-, m=1, n=2, R1=4-MeOPh, R2=Ph, R3=Me, R4═H
    29. m=1, n=2, R1=4-MeOPh, R2=4-MeOPh, R3=Me, R4═H
    30. m=1, n=2, R1=4-MeOPh, R2=Ph, R3=Me, R4═CH3
    31. m=2, n=1, R1=4-MeOPh, R2=Ph, R3=Me, R4═H
    32. m=2, n=2, R1=4-MeOPh, R2=Ph, R3=Me, R4═H
    33. m=2, n=1, R1=4-MeOPh, R2=4-MeOPh, R3=Me, R4═H
    34. m=2, n=1, R1=4-MeOPh, R2=4-BrPh, R3=Me, R4═H
    35. m=1, n=1, R1=4-OHPh, R2=Ph, R3=Me, R4═H
    36. m=1, n=2, R1=4-OHPh, R2=4-MeOPh, R3=Me, R4═H
    37. m=1, n=2, R1=4-OHPh, R2=Ph, R3=Me, R4═H
    38. m=1, n=1, R1=4-FPh, R2=Ph, R3=Me, R4═H
    39. m=1, n=2, R1=4-FPh, R2=Ph, R3=Me, R4═H
    40. m=1, n=2, R1=4-FPh, R2=4-MeOPh, R3=Me, R4═H
    41. m=1, n=1, R1=4-NO2Ph, R2=Ph, R3=Me, R4═H
    42. m=1, n=2, R1=4-NO2Ph, R2=Ph, R3=Me, R4═H
    43. m=1, n=2, R1=4-NO2Ph, R2=4-MeOPh, R3=Me, R4═H
    44. m=1, n=1, R1=4-NH2Ph, R2=Ph, R3=Me, R4═H
    45. m=1, n=2, R1=4-NH2Ph, R2=Ph, R3=Me, R4═H
    46. m=1, n=2, R1=3,4-diMeOPh, R2=Ph, R3=Me, R4═H
    47. m=1, n=3, R1=3,4-diMeOPh, R2=Ph, R3=Me, R4═H
    48. m=2, n=1, R1=Ph, R2=Ph, R3=Me, R4═H
    49. m=2, n=1, R1=Ph, R2=Ph, R3=Me, R4═H
    50. m=2, n=2, R1=Ph, R2=Ph, R3=Me, R4═H
    51. m=2, n=2, R1=Ph, R2=Ph, R3=Me, R4═CH3
    52. m=2, n=2, R1=Ph, R2=4-MeOPh, R3=Me, R4═H
    53. m=2, n=2, R1=4-MeOPh, R2=4-MeOPh, R3=Me, R4═H.
  • Definitions
  • The term “aryl” refers to aromatic groups having 6 to 24 carbon atoms, and “substituted aryl” refers to aryl groups further bearing one or more substituents.
  • The term “alkyl” refers to straight or branched chain alkyl radicals having 1 to 19 carbon atoms, and “substituted alkyl” refers to alkyl radicals further bearing one or more substituents.
  • The term “lower alkyl” refers to straight or branched chain alkyl radicals having in the range of 1 to 4 carbon atoms.
  • The term “cycloalkyl” refers to cyclic ring-containing moieties containing 3 to 8 carbon atoms, and “substituted cycloalkyl” refers to cycloalkyl moieties further bearing one or more substituents.
  • The term “alkenyl” refers to straight or branched chain hydrocarbyl groups having at least one carbon-carbon double bond and having 2 to 19 carbon atoms, and “substituted alkenyl” refers to alkenyl groups further bearing one or more substituents.
  • The term “heterocyclic” refers to cyclic moieties containing one or more heteroatoms as part of the ring structure and having 3 to 24 carbon atoms, and “substituted heterocyclic” refers to heterocyclic moieties further bearing one or more substituents.
  • Pharmaceutically acceptable salts include Cl, Br, I, NO2 , HSO4 , SO4 , HPO4 , PO4 2−, ethanesulfonate, trifluromethane sulfate, p-toluenesulfonate, benzenesulfonate, salicylate, proprionate, ascorbate, aspartate, fumarate, galactarate, maleate, citrate, glutamate, glycolate, lactate, malate, maleate, tartrate, oxalate, succinate, or similar acid addition salts. The above salt forms may be in some cases hydrates or solvates with alcohols and other solvents.
  • Pharmaceutical Compositions
  • Also disclosed herein are pharmaceutical compositions comprising a compound of formula (I) and pharmaceutically acceptable excipients. For example, the pharmaceutical composition may include a pharmaceutically acceptable additive, such as a stabilizer, buffer, salt, preservative, filler, flavor enhancer and the like, as known to those skilled in the art. Representative buffers include phosphates, carbonates, and citrates. Exemplary preservatives include EDTA, EGTA, BHA, and BHT.
  • The pharmaceutical composition disclosed herein may be administered by inhalation (i.e., intranasally as an aerosol or nasal formulation); topically (i.e., in the form of an ointment, cream or lotion); orally (i.e., in solid or liquid form (tablet, capsule, gel cap, time release capsule, powder, solution, or suspension in aqueous or non-aqueous liquid); intravenously as an infusion or injection (i.e., as a solution, suspension or emulsion in a pharmaceutically acceptable carrier); or subcutaneously as an infusion or injection (i.e., as a solution, suspension or emulsion in a pharmaceutically acceptable carrier) or as a depot formulation; transdermally (e.g., via a transdermal patch), or rectally (e.g., as a suppository).
  • There is no limitation in the route of administration or dosage form, and the composition may be administered in accordance with specific form of the preparation, age, sex and the other conditions of a patient, severity of disease, etc. For example, in the case of tablet, pill, solution, suspension, emulsion, granule and capsule, the composition is orally administered. In the case of injection, the composition is intravenously administered alone or in a mixture with conventional replacement fluid such as glucose and amino acids, and if necessary, and the preparation alone may be also administered intramuscularly, intracutaneously, subcutaneously or interperitoneally.
  • The compounds disclosed herein can be administered alone, combined with a pharmaceutically acceptable excipient, or co-administered with a second drug. Co-administration may provide a similar or synergistic effect. A compound of formula (I) or a pharmaceutically acceptable salt thereof can be administered subcutaneously, intramuscularly, intravenously, transdermally, orally, intranasally, intrapulmonary, or rectally.
  • The dose of the pharmaceutical composition of the present invention is appropriately selected in accordance with dosage regimen, age, sex and the other conditions of a patient. The amount to be administered depends to some extent on the lipophilicity of the specific compound selected, since it is expected that this property of the compound will cause it to partition into fat deposits of the subject. The precise amount to be administered can be determined by the skilled practitioner in view of desired dosages, side effects and medical history of the patient and the like.
  • The pharmaceutical composition may comprise a compound of formula (I), or an enantiomer or racemate thereof, or pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable additive or a pharmaceutically acceptable excipient.
  • Treatment of Drug Abuse
  • VMAT2 is considered as a valid target for the development of treatments for the abuse of drugs, including, for example, methamphetamine (METH), amphetamine, cocaine, methylphenidate, and opiate abuse. For example, METH decreases vesicular DA sequestration by inhibiting vesicular uptake through VMAT2 (IC50=14.9 μM) and by diffusing across the vesicular membranes to decrease the pH gradient, resulting in the loss of free energy needed for monoamine transport.
  • Lobelane competitively inhibits [3H]DA uptake into rat brain vesicles via interaction with VMAT2 (Ki=45 nM), decreases METH-evoked DA overflow from rat striatal slices, and decreases METH self-administration, but does not act as a psychostimulant, suggesting that it has potential as a novel treatment for METH abuse. Nor-Lobelane (Ki=44 nM) is equipotent with lobelane at VMAT2 in inhibiting [3H]DA uptake.
  • METH can be considered as a structural fragment of lobelane/nor-lobelane, and introducing bulky substituents onto the N-atom of METH afforded compounds with increased inhibitory potency at VMAT2, reduced the psychostimulant effects of METH, diminishing its abuse potential. Importantly, these new analogs did not increase locomotor activity in rats, indicating they do not act as psychostimulants. Lee, N.-R et al., Drug and Alcohol Dependence, “Enantiomers of (±)GZ-888 potently and selectively inhibit vesicular monoamine transporter-2 function and methamphetamine-stimulated locomotor activity.” (2016).
  • Figure US20200237688A1-20200730-C00003
  • Treatment of a Disease or Pathology of the Central Nervous System or an Eating Disorder
  • Modulation of VMAT2 has potential as a therapeutic for central nervous system diseases or pathologies. Thus, VMAT2 is a target for the development of treatments for a disease or pathology of the central nervous system, including, for example, cognitive disorders, brain trauma, memory loss, psychosis, sleep disorders, obsessive compulsive disorders, panic disorders, myasthenia gravis, Parkinson's disease, Alzheimer's disease, schizophrenia, Tourette's syndrome, Huntington's disease, attention deficit hyperactivity disorder, hyperkinetic syndrome, chronic nervous exhaustion, narcolepsy, pain, motion sickness, and/or depression.
  • VMAT2 is also a target for the development of treatments for eating disorders, including, for example, obesity. Compulsive eating is linked to addiction-like neuroadaptive responses in brain reward circuits. Johnson, P. M. et al., “Dopamine D2 receptors in addiction-like reward dysfunction and compulsive eating in obese rats,” Nature Neuroscience, 2010, 13, 635-641. Modulation of VMAT2 may lead to reduced reward responses, diminishing overeating.
  • A more preferred embodiment of the invention is a compound of formula (I), wherein the compound is 3-(4-methoxyphenyl)-N-(1-phenylpropan-2-yl)propan-1-amine or an enantiomer; racemate; or pharmaceutically acceptable salt thereof.
  • An even more preferred embodiment of the invention is a compound of formula (I), wherein the compound is (S)-3-(4-methoxyphenyl)-N-(1-phenylpropan-2-yl)propan-1-amine hydrochloride.
  • A more preferred embodiment of the invention is a pharmaceutical composition comprising 3-(4-methoxyphenyl)-N-(1-phenylpropan-2-yl)propan-1-amine or an enantiomer; racemate; or pharmaceutically acceptable salt thereof and a pharmaceutically acceptable additive or a pharmaceutically acceptable excipient. An even more preferred embodiment of the invention is a pharmaceutical composition comprising (S)-3-(4-methoxyphenyl)-N-(1-phenylpropan-2-yl)propan-1-amine hydrochloride and a pharmaceutically acceptable additive or a pharmaceutically acceptable excipient.
  • Another preferred embodiment of the invention is a method of treating a disease or pathology of the central nervous system or an eating disorder in an individual in need thereof, wherein the method comprises the step of administering to the individual a compound of formula (I) or a pharmaceutically acceptable salt thereof.
  • The compounds of the present invention may be used in a method of treating a disease or pathology of the central nervous system, wherein the disease may be cognitive disorders, brain trauma, memory loss, psychosis, sleep disorders, obsessive compulsive disorders, panic disorders, myasthenia gravis, Parkinson's disease, Alzheimer's disease, schizophrenia, Tourette's syndrome, Huntington's disease, attention deficit hyperactivity disorder, hyperkinetic syndrome, chronic nervous exhaustion, narcolepsy, pain, motion sickness, and/or depression.
  • The compounds of the present invention may also be used in a method of treating an eating disorder. The eating disorder may be obesity.
  • The compounds of the present invention may be used in a method of treating substance use disorder, drug dependence/abuse or withdrawal from drug dependence/abuse in an individual in need thereof. Substance use disorder and drug dependence/abuse includes dependence/abuse of psychostimulants or drugs that release dopamine. For example, the drug which the individual is using and/or dependent upon may be amphetamine, methamphetamine, and other drugs that release dopamine.
  • EXAMPLES
  • A series of VMAT2 inhibitors were synthesized and evaluated for their activity at VMAT2, dopamine transporters (DAT), and serotonin transporters (SERT) using rat striatum, hERG channels expressed by HEK-293 cells, and for their effect on METH-stimulated locomotor activity in rats. Compounds of the invention exhibited affinity at VMAT2 as well as selectivity at VMAT2 over hERG, DAT, and SERT. Further, a significant reduction of METH-stimulated locomotor activity was observed after administration of the compounds.
  • Example 1 Synthesis of Compound 10. (S)-3-(4-methoxyphenyl)-N-(1-phenylpropan-2-yl)propan-1-amine
  • To a solution of phenyllithium (50 mL, 1.8 M in dibutyl ether) in THF (50 mL) was added (R)-propylene oxide (5 g) dropwise at −78° C. The resulting mixture was stirred at the same temperature for 1 hr before warmed to room temperature. After stirred at room temperature overnight, the reaction was quenched by adding saturated NH4Cl aqueous solution. The aqueous phase was extracted with ethyl acetate (3×50 mL) and the combined organic layers were washed with brine (100 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The crude product was chromatographed on silica (hexanes/ethyl acetate 10:1 to 3:1) to afford (R)-1-phenylpropan-2-ol (10.4 g) as a colorless oil.
  • To a solution of (R)-1-phenylpropan-2-ol (4.16 g, 30.50 mmol) and triethylamine (7.72 g, 10.64 mL, 76.27 mmol) in dichloromethane (100 mL) was added methanesulfonyl chloride (4.54 gm, 3.07 mL, 39.65 mmol) dropwise at 0° C. The resulting mixture was stirred at 0° C. for 15 min. Dichloromethane (100 mL) was added to the mixture and then washed with water (2×150 mL). The organic layer was dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The resulting light yellow oil was mixed with sodium azide (5.95 g, 91.5 mmol) in DMF (40 mL) and heated at 55° C. for 3 hrs. The reaction mixture was diluted with diethyl ether (150 mL) and washed with water (2×100 mL) and brine (100 mL). The organic layer was dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The crude product was chromatographed on silica (hexanes/ethyl acetate 50:1 to 20:1) to afford (S)-(2-azidopropyl)benzene (4.38 g) as a colorless oil.
  • To a solution of (S)-(2-azidopropyl)benzene (4.0 g, 24.81 mmol) in THF (90 mL) and water (10 mL) was added triphenylphosphine (9.11 g, 34.74 mmol) at room temperature. The resulting mixture was stirred for 18 hrs and water (50 mL) was added. The resulting mixture was treated with HCl (1.0 M) to pH ˜1 and the aqueous phase was extracted with diethyl ether (3×100 mL) and dichloromethane (2×100 mL). NaOH (15%) was added dropwise to the aqueous phase to adjust the pH to about 11 and extracted with dichloromethane (5×60 mL). The combined organic layers were dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to afford (S)-1-phenylpropan-2-amine as a colorless oil. The crude amino product (3.03 g, 22.41 mmol) was mixed with 3-(4-methoxyphenyl)propanoic acid (4.44 g, 24.65 mmol), and HOBt (3.63 g, 26.89 mmol) in dichloromethane (60 mL) at room temperature. Triethylamine (5.67 g, 56.03 mmol) was added followed by EDCI (5.15 g, 26.89 mmol). The resulting mixture was stirred overnight. The reaction mixture was diluted with dichloromethane (100 mL) and washed with water (3×50 mL), and brine (50 mL). The organic layer was dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The crude product was chromatographed on silica (dichloromethane/ethyl acetate 10:1) to afford (S)-3-(4-methoxyphenyl)-N-(1-phenylpropan-2-yl)propanamide (6.18 g) as a white solid. (S)-3-(4-Methoxyphenyl)-N-(1-phenylpropan-2-yl)propanamide (5.0 g, 16.81 mmol) in THF was cooled to 0° C. LAH (60 mL, 1.0 M in THF) was added dropwise and the resulted reaction mixture was heated at reflux for 6 hrs. After cooled to 0° C., water (2.28 mL) was carefully added, followed by NaOH (15%, 2.28 mL) and water (6.84 mL). The resulted mixture was warmed to room temperature and stirred for 2 hrs. Filtered over celite and washed with ethyl acetate. The filtrate was concentrated under reduced pressure and the crude product was chromatographed on silica (dichloromethane/methanol 30:1 to 10:1) to afford compound 10 (4.3 g) as a white solid.
  • Example 2 [3H]Dihydrotetrabenazine ([3H]DTBZ) Binding Assay, Vesicular Preparation
  • Synaptic vesicles were prepared from rat brain using a modification of a previously described procedure (Teng et al., 1998). Briefly, fresh whole brain (excluding cerebellum) was homogenized using a Teflon pestle (clearance 0.003 inches) with 7 vertical strokes at 800 rpm in 20 vol of ice-cold 0.32 M sucrose and centrifuged at 1000 g for 12 min at 4° C. The resulting supernatant (S1) was then centrifuged at 22,000 g for 10 min at 4° C. The synaptosomal pellets (P2) were homogenized in 18 mL of ice-cold Milli-Q water and exposed for 5 min for lysing synaptosomes. Osmolarity was restored by addition of 2 mL of 25 mM HEPES with 100 mM dipotassium tartrate (pH 7.5). Samples were centrifuged at 20,000 g for 20 min at 4° C. to remove lysed synaptosomal membranes. MgSO4 (1 mM) was added to the supernatant (S3), and was centrifuged at 100,000 g for 45 min at 4° C. The final vesicular pellets (P4) were resuspended in ice-cold assay buffer (see below) providing ˜15 μg protein/100 μL, determined by the method of Bradford (1976) using bovine serum albumin as a the standard. Aliquot parts (100 μL) of suspension of vesicle membrane protein were incubated in assay buffer (25 mM HEPES, 100 mM dipotassium tartrate, 5 mM MgSO4, 0.1 mM EDTA and 0.05 mM EGTA, pH 7.5, at 25° C.) in the presence of 3 nM [3H]DTBZ and at least 7 concentrations (1 nM-1 mM) of compound for 1 hr at room temperature. Nonspecific binding was determined in the presence of 20 μM tetrabenazine, a standard compound. Assays were performed in duplicate using a 96-well plate format. Reactions were terminated by filtration of samples on a Unifilter-96 GF/B filter plates (presoaked in 0.5% polyethylenimine), using a FilterMate harvester (Packard BioScience Co., Meriden, Conn.). After washing 5 times with 350 μL of the ice-cold wash buffer (25 mM HEPES, 100 mM dipotassium tartrate, 5 mM MgSO4 and 10 mM NaCl, pH 7.5), filter plates were dried, sealed and each well filled with 40 μL Packard's MicroScint 20 cocktail. Bound [3H]DTBZ was measured using a Packard TopCount NXT scintillation counter with a Packard Windows NT based operating system.
  • Example 3 [3H]Dopamine ([3H]DA) Uptake Assay, Vesicular Preparation
  • Inhibition of [3H]DA uptake was conducted using isolated synaptic vesicle preparations (Teng et al., 1997). Briefly, rat striata were homogenized with 10 up-and-down strokes of a Teflon pestle homogenizer (clearance ˜0.003″) in 14 ml of 0.32 M sucrose solution. Homogenates were centrifuged (2,000 g for 10 min at 4° C.), and then the supernatants were centrifuged (10,000 g for 30 min at 4° C.). Pellets were resuspended in 2 ml of 0.32 M sucrose solution and subjected to osmotic shock by adding 7 ml of ice-cold MilliQ water to the preparation. After 1 min, osmolarity was restored by adding 900 μl of 0.25 M HEPES buffer and 900 μl of 1.0 M potassium tartrate solution. Samples were centrifuged (20,000 g for 20 min at 4° C.), and the supernatants were centrifuged (55,000 g for 1 hr at 4° C.), followed by addition of 100 μl of 10 mM MgSO4, 100 μl of 0.25 M HEPES and 100 μl of 1.0 M potassium tartrate solution prior to the final centrifugation (100,000 g for 45 min at 4° C.). Final pellets were resuspended in 2.4 ml of assay buffer (25 mM HEPES, 100 mM potassium tartrate, 50 μM EGTA, 100 μM EDTA, 1.7 mM ascorbic acid, 2 mM ATP-Mg2+, pH 7.4). Aliquots of the vesicular suspension (100 μl) were added to tubes containing assay buffer, various concentrations of compound (0.1 nM-10 mM) and 0.1 μM [3H]DA in a final volume of 500 μl, and incubated at 37° C. for 8 min. Nonspecific uptake was determined in the presence of the standard compound, Ro4-1284 (10 M). Reactions were terminated by filtration, and radioactivity retained by the filters was determined by liquid scintillation spectrometry (Tri-Carb 2100TR liquid scintillation analyzer; PerkinElmer Life and Analytical Sciences, Boston, Mass.).
  • Example 4 [3H]Dofetilide Binding Assay, HEK-293 Cell Membrane Preparation
  • [3H]Dofetilide binding assays were conducted using commercially available HEK-293 cell membranes which stably express the hERG channel. Membranes were suspended in assay buffer (50 mM Tris, 10 mM KCl, 1 mM MgCl2, pH 7.4) prior to the experiment. Assays were performed in duplicate in a total volume of 250 μL. Aliquots of the HEK-293 cell membrane suspension which contained 5 μg membrane protein were added to tubes containing assay buffer, 5 nM [3H]dofetilide and a range of concentrations of analog (10 nM-100 μM). Nonspecific binding was determined in the presence of amitriptyline (1 mM). Samples were incubated for 1 hr. at 24° C., followed by rapid filtration. Radioactivity retained by the filters was determined by liquid scintillation spectrometry as described above for the [3H]DA uptake assay. The affinity for the [3H]dofetilide binding site on the hERG channel expressed in the HEK-293 cellular membrane was determined from the analog concentration response curves.
  • Example 5 [3H]DA and [3H]5-HT Uptake Assay, Synaptosomal Preparation
  • [3H]DA and [3H]5-HT uptake into striatal synaptosomes was determined to evaluate compound inhibition of the dopamine transporter (DAT) and the serotonin transporter (SERT), respectively. Striata from individual rats were homogenized in ice-cold sucrose solution containing 5 mM NaHCO3 (pH 7.4), with 16 up-and-down strokes of a Teflon pestle homogenizer (clearance≈0.003″). Homogenates were centrifuged at 2000 g for 10 min at 4° C., and resulting supernatants were centrifuged at 20,000 g for 17 min at 4° C. Pellets were resuspended in 2.4 mL (for DAT assays) or 1.5 mL (for SERT assays) of assay buffer (125 mM NaCl, 5 mM KCl, 1.5 mM MgSO4, 1.25 mM CaCl2, 1.5 mM KH2PO4, 10 mM alpha-D-glucose, 25 mM HEPES, 0.1 mM EDTA, 0.1 mM pargyline, 0.1 mM ascorbic acid, saturated with 95% O2/5% CO2, pH 7.4). Assays were performed in duplicate in a total volume of 500 μL (for DAT assays) or 250 μL (for SERT assays). Aliquots of the synaptosomal suspension (25 μL for DAT, 50 μL for SERT) were added to tubes containing assay buffer and various concentrations of analog (1 nM-100 μM), and incubated at 34° C. for 5 min. Nonspecific uptake was determined in the presence of nomifensine (10 μM) for DAT assays or fluoxetine (10 μM) for SERT assays. GBR-12935 (100 nM) was included in the assay buffer for the SERT assay to maximally inhibit [3H]5-HT uptake through DAT and isolate uptake to SERT. Samples were placed on ice, and 50 μL of 0.1 μM [3H]DA (for DAT assays) or 25 μL of 0.1 μM [3H]5-HT (for SERT assays) was added to each tube, and incubated for 10 min at 34° C. Reactions were terminated by addition of 3 mL of ice-cold assay buffer and subsequent filtration and radioactivity retained by the filters was determined by liquid scintillation spectrometry (Tri-Carb 2100TR liquid scintillation analyzer; PerkinElmer Life and Analytical Sciences, Boston, Mass.).
  • Exemplary compounds 1-53 were tested in [3H]dihydrotetrabenazine ([3H]DTBZ) binding assay according to Example 2 and the [3H]dopamine ([3H]DA) uptake assay according to Example 3. The results of these assays are set forth in Table 1.
  • TABLE 1
    [3H]DA uptake
    [3H]DTBZ binding (Ki) VMAT2
    Compound Structure (Ki) VMAT2 (μM) (μM)
     1
    Figure US20200237688A1-20200730-C00004
    7.70 ± 1.25 0.063 ± 0.005
     2
    Figure US20200237688A1-20200730-C00005
    7.53 ± 2.16 0.51 ± 0.10
     3
    Figure US20200237688A1-20200730-C00006
    1.23 ± 0.13 0.033 ± 0.007
     4
    Figure US20200237688A1-20200730-C00007
    1.13 ± 0.31 0.007 ± 0.002
     5
    Figure US20200237688A1-20200730-C00008
    0.91 ± 0.59 0.006 ± 0.001
     6
    Figure US20200237688A1-20200730-C00009
    0.75 ± 0.14 0.065 ± 0.004
     7
    Figure US20200237688A1-20200730-C00010
    1.56 ± 0.47 0.096 ± 0.010
     8
    Figure US20200237688A1-20200730-C00011
     2.2 ± 0.11 0.014 ± 0.003
     9
    Figure US20200237688A1-20200730-C00012
    0.0087 ± 0.0065
    10
    Figure US20200237688A1-20200730-C00013
     0.026 ± 0.0036
    11
    Figure US20200237688A1-20200730-C00014
     0.70 ± 0.063 0.008 ± 0.001
    12
    Figure US20200237688A1-20200730-C00015
    0.006 ± 0.001
    13
    Figure US20200237688A1-20200730-C00016
    0.032 ± 0.004
    14
    Figure US20200237688A1-20200730-C00017
    0.081 ± 0.016  0.003 ± 0.0003
    15
    Figure US20200237688A1-20200730-C00018
     0.19 ± 0.020 0.003 ± 0.002
    16
    Figure US20200237688A1-20200730-C00019
    0.46 ± 0.08 0.012 ± 0.003
    17
    Figure US20200237688A1-20200730-C00020
    0.63 0.014 ± 0.001
    18
    Figure US20200237688A1-20200730-C00021
    0.91 ± 0.59 0.009 ± 0.002
    19
    Figure US20200237688A1-20200730-C00022
    4.1 ± 0.3 0.059 ± 0.007
    20
    Figure US20200237688A1-20200730-C00023
    2.0 ± 0.1 0.013 ± 0.009
    21
    Figure US20200237688A1-20200730-C00024
    1.5 ± 0.2 0.12 ± 0.01
    22
    Figure US20200237688A1-20200730-C00025
    2.0 ± 0.1  0.12 ± 0.001
    23
    Figure US20200237688A1-20200730-C00026
    3.4 ± 0.6 0.092 ± 0.017
    24
    Figure US20200237688A1-20200730-C00027
    3.77 ± 0.99 0.074 ± 0.002
    25
    Figure US20200237688A1-20200730-C00028
    11.9 ± 1.84  0.46 ± 0.075
    26
    Figure US20200237688A1-20200730-C00029
    1.27 ± 0.09 0.022 ± 0.003
    27
    Figure US20200237688A1-20200730-C00030
    0.045 ± 0.004
    28
    Figure US20200237688A1-20200730-C00031
    0.020 ± 0.001
    29
    Figure US20200237688A1-20200730-C00032
    0.41 ± 0.14 0.011 ± 0.002
    30
    Figure US20200237688A1-20200730-C00033
    7.11 ± 0.82  0.27 ± 0.038
    31
    Figure US20200237688A1-20200730-C00034
    2.4 ± 0.1 0.062 ± 0.029
    32
    Figure US20200237688A1-20200730-C00035
    4.7 ± 0.4 0.030 ± 0.002
    33
    Figure US20200237688A1-20200730-C00036
    1.5 ± 0.2 0.060 ± 0.007
    34
    Figure US20200237688A1-20200730-C00037
    0.87 ± 0.03 0.010 ± 0.004
    35
    Figure US20200237688A1-20200730-C00038
    2.6 ± 0.4 0.047 ± 0.006
    36
    Figure US20200237688A1-20200730-C00039
    0.23 ± 0.06 0.072 ± 0.030
    37
    Figure US20200237688A1-20200730-C00040
    0.82 ± 0.27 0.033 ± 0.008
    38
    Figure US20200237688A1-20200730-C00041
    2.5 ± 1.6 0.069 ± 0.009
    39
    Figure US20200237688A1-20200730-C00042
    1.3 ± 0.3  0.010 ± 0.0002
    40
    Figure US20200237688A1-20200730-C00043
    1.7 ± 0.4 0.017 ± 0.003
    41
    Figure US20200237688A1-20200730-C00044
    5.1 ± 1.2 0.060 ± 0.007
    42
    Figure US20200237688A1-20200730-C00045
    2.0 ± 0.3 0.013 ± 0.004
    43
    Figure US20200237688A1-20200730-C00046
    1.4 ± 0.4 0.008 ± 0.002
    44
    Figure US20200237688A1-20200730-C00047
     17 ± 6.6 0.21 ± 0.08
    45
    Figure US20200237688A1-20200730-C00048
    2.8 ± 0.7 0.036 ± 0.003
    46
    Figure US20200237688A1-20200730-C00049
    3.8 ± 0.1  0.14 ± 0.013
    47
    Figure US20200237688A1-20200730-C00050
    1.1 ± 0.1 0.029 ± 0.011
    48
    Figure US20200237688A1-20200730-C00051
    1.60 ± 0.33 0.050 ± 0.004
    49
    Figure US20200237688A1-20200730-C00052
    29.8 ± 11.2 0.084 ± 0.004
    50
    Figure US20200237688A1-20200730-C00053
    4.81 ± 1.13 0.050 ± 0.012
    51
    Figure US20200237688A1-20200730-C00054
    17.2 ± 6.33  0.36 ± 0.034
    52
    Figure US20200237688A1-20200730-C00055
    3.2 ± 0.1 0.026 ± 0.002
    53
    Figure US20200237688A1-20200730-C00056
    1.5 ± 0.7 0.024 ± 0.006
  • As illustrated by Table 1, compounds of the invention exhibited high affinity at VMAT2 as well as selectivity at VMAT2 over hERG, DAT, and SERT. For example, Compound 9 exhibited a Ki value of 8.71±3.65 at VMAT2, and Compound 10 exhibited a Ki value of 25.5±3.57 nM at VMAT2. Further, both Compound 9 and Compound 10 exhibited a >30-fold selectivity at VMAT2 over hERG, DAT, and SERT.
  • The compounds of the invention were also evaluated for their effect on METH-stimulated locomotor activity was evaluated in rats. The compounds were observed to exhibit a significant reduction of METH-stimulated locomotor activity after administration. For example, a significant reduction of METH-stimulated locomotor activity was observed after administration of Compound 9 (3 mg/kg, s.c.) and Compound 10 (17 mg/kg, s.c.).
  • The foregoing description and examples have been set forth merely to illustrate the invention and are not meant to be limiting. Since modifications of the described embodiments incorporating the spirit and the substance of the invention may occur to persons skilled in the art, the invention should be construed broadly to include all variations within the scope of the claims and equivalents thereof.

Claims (17)

1. A method of treating a disease or pathology of the central nervous system or an eating disorder in an individual in need thereof, wherein the method comprises the step of administering to the individual a compound of formula (I):
Figure US20200237688A1-20200730-C00057
wherein
m is an integer in the range from 1 to 3;
n is zero or an integer from 1 to 5;
R1 and R2 are each independently an aryl group,
wherein R1 and R2 are each independently unsubstituted or substituted by one or more substituents selected from the group consisting of methyl; deuteromethyl (CD3); tritiomethyl (CT3); ethyl; propyl; isopropyl; C4-C7 straight chain or branched alkyl; C3-C6 cycloalkyl; C4-C7 alkenyl; benzyl; phenylethyl; amino; N-methylamino; N,N-dimethylamino; carboxylate; methylcarboxylate; ethylcarboxylate; propylcarboxylate; isopropylcarboxylate; carboxaldehyde; acetoxy; propionyloxy; isopropionyloxy; cyano; aminomethyl; N-methylaminomethyl; N,N-dimethylaminomethyl; carboxamide; N-methylcarboxamide; N,N-dimethylcarboxamide; acetyl; propionyl; formyl; benzoyl sulfate; phenyl; methylsulfate; hydroxyl; methoxy; ethoxy; propoxy; isopropoxy; thiol; methylthio; ethylthio; propiothiol; fluoro; chloro; bromo; iodo; trifluoromethyl; vinyl; allyl; propargyl; nitro; carbamoyl; ureido; azido; isocyanate; thioisocyanate; hydroxylamino; nitroso; a saturated or unsaturated hydrocarbon ring; a nitrogen-containing heterocyclic moiety; an oxygen-containing heterocyclic moiety; a sulfur-containing heterocyclic moiety; a selenium-containing heterocyclic moiety; a mixed heterocyclic moiety containing at least two atoms selected from the group consisting of nitrogen, oxygen and sulfur; and ortho, meta or para-substituted benzene;
R3 is methyl, ethyl, propyl, isopropyl, hydroxymethyl, 2-hydroxyethyl, 1-hyhydroxyethyl, methoxymethyl, 2-methoxyethyl, 1-methoxyethyl, aminomethyl, 2-aminoethyl, 1-aminoethyl, N-methylaminomethyl, 2-N-methylaminoethyl, 1-N-methylaminoethyl, N,N-dimethylaminomethyl, 2-N,N-dimethylaminoethyl, or 1-N,N-dimethylaminoethyl group; and
R4 is a hydrogen atom or a methyl, ethyl, propyl, or isopropyl group; or
an enantiomer; racemate; or pharmaceutically acceptable salt thereof.
2. The method of claim 1, wherein:
m is 1 or 2; and
n is an integer from 1 to 5; or
an enantiomer; racemate; or pharmaceutically acceptable salt thereof.
3. The method of claim 2, wherein:
R3 is methyl, ethyl, propyl, or isopropyl; and
R4 is a hydrogen atom or a methyl, ethyl, propyl, or isopropyl group; or
an enantiomer; racemate; or pharmaceutically acceptable salt thereof.
4. The method of claim 1, wherein the compound of formula (I) is 3-(4-methoxyphenyl)-N-(1-phenylpropan-2-yl)propan-1-amine or an enantiomer; racemate; or pharmaceutically acceptable salt thereof.
5. The method of claim 1, wherein the compound of formula (I) is (S)-3-(4-methoxyphenyl)-N-(1-phenylpropan-2-yl)propan-1-amine hydrochloride.
6. The method of claim 1, wherein the disease or pathology of the central nervous system is selected from the group consisting of cognitive disorders, brain trauma, memory loss, psychosis, sleep disorders, obsessive compulsive disorders, panic disorders, myasthenia gravis, Parkinson's disease, Alzheimer's disease, schizophrenia, Tourette's syndrome, Huntington's disease, attention deficit hyperactivity disorder, hyperkinetic syndrome, chronic nervous exhaustion, narcolepsy, pain, motion sickness, and depression.
7. The method of claim 1, wherein the eating disorder is obesity.
8. The method of claim 1, wherein the compound of formula (I), enantiomer, racemate, or pharmaceutically acceptable salt thereof is present in a pharmaceutical composition further comprising a pharmaceutically acceptable excipient.
9. The method of claim 8, wherein the pharmaceutical composition is administered to the individual by inhalation; topically; orally; intravenously as an infusion or injection; or subcutaneously as an infusion, injection, or depot formulation; transdermally; or rectally.
10. A method of treating a substance use disorder, drug dependence/abuse or withdrawal from drug dependence/abuse in an individual in need thereof, wherein the method comprises the step of administering to the individual a compound of formula (I):
Figure US20200237688A1-20200730-C00058
wherein
m is an integer in the range from 1 to 3;
n is zero or an integer from 1 to 5;
R1 and R2 are each independently an aryl group,
wherein R1 and R2 are each independently unsubstituted or substituted by one or more substituents selected from the group consisting of methyl; deuteromethyl (CD3); tritiomethyl (CT3); ethyl; propyl; isopropyl; C4-C7 straight chain or branched alkyl; C3-C6 cycloalkyl; C4-C7 alkenyl; benzyl; phenylethyl; amino; N-methylamino; N,N-dimethylamino; carboxylate; methylcarboxylate; ethylcarboxylate; propylcarboxylate; isopropylcarboxylate; carboxaldehyde; acetoxy; propionyloxy; isopropionyloxy; cyano; aminomethyl; N-methylaminomethyl; N,N-dimethylaminomethyl; carboxamide; N-methylcarboxamide; N,N-dimethylcarboxamide; acetyl; propionyl; formyl; benzoyl sulfate; phenyl; methylsulfate; hydroxyl; methoxy; ethoxy; propoxy; isopropoxy; thiol; methylthio; ethylthio; propiothiol; fluoro; chloro; bromo; iodo; trifluoromethyl; vinyl; allyl; propargyl; nitro; carbamoyl; ureido; azido; isocyanate; thioisocyanate; hydroxylamino; nitroso; a saturated or unsaturated hydrocarbon ring; a nitrogen-containing heterocyclic moiety; an oxygen-containing heterocyclic moiety; a sulfur-containing heterocyclic moiety; a selenium-containing heterocyclic moiety; a mixed heterocyclic moiety containing at least two atoms selected from the group consisting of nitrogen, oxygen and sulfur; and ortho, meta or para-substituted benzene;
R3 is methyl, ethyl, propyl, isopropyl, hydroxymethyl, 2-hydroxyethyl, 1-hyhydroxyethyl, methoxymethyl, 2-methoxyethyl, 1-methoxyethyl, aminomethyl, 2-aminoethyl, 1-aminoethyl, N-methylaminomethyl, 2-N-methylaminoethyl, 1-N-methylaminoethyl, N,N-dimethylaminomethyl, 2-N,N-dimethylaminoethyl, or 1-N,N-dimethylaminoethyl group; and
R4 is a hydrogen atom or a methyl, ethyl, propyl, or isopropyl group; or
an enantiomer; racemate; or pharmaceutically acceptable salt thereof.
11. The method of claim 10, wherein:
m is 1 or 2; and
n is an integer from 1 to 5; or
an enantiomer; racemate; or pharmaceutically acceptable salt thereof.
12. The method of claim 11, wherein:
R3 is methyl, ethyl, propyl, or isopropyl; and
R4 is a hydrogen atom or a methyl, ethyl, propyl, or isopropyl group; or
an enantiomer; racemate; or pharmaceutically acceptable salt thereof.
13. The method of claim 10, wherein the compound of formula (I) is 3-(4-methoxyphenyl)-N-(1-phenylpropan-2-yl)propan-1-amine or an enantiomer; racemate; or pharmaceutically acceptable salt thereof.
14. The method of claim 10, wherein the compound of formula (I) is (S)-3-(4-methoxyphenyl)-N-(1-phenylpropan-2-yl)propan-1-amine hydrochloride.
15. The method of claim 10, wherein the drug is methamphetamine.
16. The method of claim 10, wherein the compound of formula (I), enantiomer, racemate, or pharmaceutically acceptable salt thereof is present in a pharmaceutical composition further comprising a pharmaceutically acceptable excipient.
17. The method of claim 16, wherein the pharmaceutical composition is administered to the individual by inhalation; topically; orally; intravenously as an infusion or injection; or subcutaneously as an infusion, injection, or depot formulation; transdermally; or rectally.
US16/846,989 2016-04-21 2020-04-13 Vesicular monoamine transporter-2 ligands and their use in the treatment of psychostimulant abuse Pending US20200237688A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US16/846,989 US20200237688A1 (en) 2016-04-21 2020-04-13 Vesicular monoamine transporter-2 ligands and their use in the treatment of psychostimulant abuse

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201662325875P 2016-04-21 2016-04-21
US15/493,836 US10668030B2 (en) 2016-04-21 2017-04-21 Vesicular monoamine transporter-2 ligands and their use in the treatment of psychostimulant abuse
US16/846,989 US20200237688A1 (en) 2016-04-21 2020-04-13 Vesicular monoamine transporter-2 ligands and their use in the treatment of psychostimulant abuse

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US15/493,836 Continuation US10668030B2 (en) 2016-04-21 2017-04-21 Vesicular monoamine transporter-2 ligands and their use in the treatment of psychostimulant abuse

Publications (1)

Publication Number Publication Date
US20200237688A1 true US20200237688A1 (en) 2020-07-30

Family

ID=60089278

Family Applications (2)

Application Number Title Priority Date Filing Date
US15/493,836 Active US10668030B2 (en) 2016-04-21 2017-04-21 Vesicular monoamine transporter-2 ligands and their use in the treatment of psychostimulant abuse
US16/846,989 Pending US20200237688A1 (en) 2016-04-21 2020-04-13 Vesicular monoamine transporter-2 ligands and their use in the treatment of psychostimulant abuse

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US15/493,836 Active US10668030B2 (en) 2016-04-21 2017-04-21 Vesicular monoamine transporter-2 ligands and their use in the treatment of psychostimulant abuse

Country Status (1)

Country Link
US (2) US10668030B2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11718618B2 (en) 2021-03-22 2023-08-08 Neurocrine Biosciences, Inc. Substituted pyrido[2,1-a]isoquinolines as VMAT2 inhibitors

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20200290948A1 (en) * 2016-04-21 2020-09-17 University Of Kentucky Research Foundation Vesicular Monoamine Transporter-2 Ligands and Their Use in the Treatment of Psychostimulant Abuse
KR20230012501A (en) 2020-05-19 2023-01-26 사이빈 아이알엘 리미티드 Deuterated Tryptamine Derivatives and Methods of Use

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB2088873A (en) * 1980-12-10 1982-06-16 Thomae Gmbh Dr K Chemical Compounds
US6057371A (en) * 1989-12-28 2000-05-02 Virginia Commonwealth University Sigma receptor ligands and the use thereof

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0591426A4 (en) 1991-06-27 1996-08-21 Univ Virginia Commonwealth Sigma receptor ligands and the use thereof
WO2006017861A2 (en) * 2004-08-13 2006-02-16 Omeros Corporation Treatment for methamphetamine addiction and reduction of methamphetamine use using serotonin antagonists

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB2088873A (en) * 1980-12-10 1982-06-16 Thomae Gmbh Dr K Chemical Compounds
US6057371A (en) * 1989-12-28 2000-05-02 Virginia Commonwealth University Sigma receptor ligands and the use thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
CAS STN Database Registry No. 415970-89-7 [Entered STN: 15 May 2002]. (Year: 2002) *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11718618B2 (en) 2021-03-22 2023-08-08 Neurocrine Biosciences, Inc. Substituted pyrido[2,1-a]isoquinolines as VMAT2 inhibitors

Also Published As

Publication number Publication date
US10668030B2 (en) 2020-06-02
US20170304227A1 (en) 2017-10-26

Similar Documents

Publication Publication Date Title
US11691947B2 (en) Isoindoline compositions and methods for treating neurodegenerative disease
US20200237688A1 (en) Vesicular monoamine transporter-2 ligands and their use in the treatment of psychostimulant abuse
TW201733B (en)
US11697639B2 (en) Quaternary ammonium salt compound, preparation method therefor and use thereof
US10941138B2 (en) Quinoline and isoquinoline derivatives for treating pain and pain related conditions
US10590140B2 (en) Tetrahydropyrimidodiazepine and dihydropyridodiazepine compounds for treating pain and pain related conditions
US9359346B2 (en) Benzamide derivative and use thereof
US10633336B2 (en) Dopamine D2 receptor ligands
US20220315523A1 (en) Vesicular Monoamine Transporter-2 Ligands and Their Use in the Treatment of Psychostimulant Abuse
TWI236469B (en) Diamine compound with condensed-ring groups
EP2970290B1 (en) A crystalline form of an anxiolytic compound
US10464883B2 (en) Compounds and methods for the treatment of neurodegenerative diseases
KR20170096008A (en) Crosslinked polydiallymine copolymers for the treatment of type 2 diabetes
US20190270698A1 (en) Compounds and methods for the treatment of neurodegenerative diseases
WO2022125614A1 (en) Phosphonates as inhibitors of enpp1 and cdnp
ES2889876T3 (en) Cyclopropylmethanamines as selective 5-HT(2C) receptor agonists
US20210395254A1 (en) New tetrahydropyrimidodiazepin and tetrahydropyridodiazepin compounds for treating pain and pain related conditions
TW200916092A (en) N-oxides of venlafaxine and O-desmethylvenlafaxine as prodrugs
WO2006095187A1 (en) Benzoxazocines and their therapeutic use
US20170226072A1 (en) 1,4-Disubstituted Piperidines, 1,4-Disubstituted Piperazines, 1,4-Disubstituted Diazepines, and 1,3-Disubstituted Pyrrolidine Compounds
JP2019509321A (en) Combinations for treating pain
US20240158394A1 (en) Nek7 inhibitors
JPH09268162A (en) Isoprene derivative
EP3153168B1 (en) Phacetoperane for treating attention deficit hyperactivity disorder and synthesis method

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

AS Assignment

Owner name: UNIVERSITY OF KENTUCKY RESEARCH FOUNDATION, KENTUCKY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:DWOSKIN, LINDA P.;CROOKS, PETER ANTHONY;ZHENG, GUANGRONG;AND OTHERS;SIGNING DATES FROM 20170725 TO 20191125;REEL/FRAME:058879/0048

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED