Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
AZETIDINE COMPOUNDS, PHARMACEUTICAL COMPOSITIONS THEREOF, PREPARATION METHODS AND THEIR USES IN THE TREATMENT OF CNS DISORDERS
Document Type and Number:
WIPO Patent Application WO/2023/214119
Kind Code:
A1
Abstract:
The present disclosure relates to compounds of formula (I) and stereoisomers and pharmaceutically acceptable salts thereof, wherein R1 to R9 are as defined in the claims. Further is disclosed compounds of formula (I) for use in the treatment or prevention of drug addiction or CNS related diseases or conditions. The invention also relates to pharmaceutical compositions, and to methods for the preparation of the aforementioned compounds.

Inventors:
TAKIO VILLE EMIL (FI)
HEIKKINEN JENNI (FI)
Application Number:
PCT/FI2023/050241
Publication Date:
November 09, 2023
Filing Date:
May 03, 2023
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
EQUINORM LTD (FI)
International Classes:
C07D205/04; A61K31/397; A61P25/00
Domestic Patent References:
WO2019234115A12019-12-12
WO2010058020A12010-05-27
WO2006134487A12006-12-21
WO2005049605A12005-06-02
WO2016193241A12016-12-08
WO2016177849A12016-11-10
Foreign References:
GB1236078A1971-06-16
EA202092955A12021-04-08
US20120232026A12012-09-13
Other References:
HE ZHI-TAO ET AL: "Palladium-catalyzed [alpha]-arylation for the addition of small rings to aromatic compounds", NATURE COMMUNICATIONS, vol. 10, no. 1, 9 September 2019 (2019-09-09), XP093056488, Retrieved from the Internet DOI: 10.1038/s41467-019-12090-z
DATABASE REGISTRY [online] CHEMICAL ABSTRACT SERVICE, COLUMBUS, OHIO, US; 29 December 2021 (2021-12-29), COMPOUNDS FROM CHEMCATS CATALOG: "azetidine", XP093056691, Database accession no. 2751306-39-3
"Protective Groups in Organic Synthesis", 2007, JOHN WILEY & SONS, INC
CHEN ET AL.: "Chemoselective direct reductive trifluoromethylation of amides: a flexible access to functionalized α- trifluoromethylamines", ORG. CHEM. FRONT, vol. 5, 2018, pages 943 - 947
KRASOVSKIY, AKNOCHEL, P: "Angew. Chem. Int. Ed", vol. 43, 2004, pages: 3333
"Re-mington's Pharmaceutical Sciences", 2000, MACK PUBLISHING COMPANY
HEIDBREDER, C.A ET AL., ADDICT BIOL, vol. 12, no. 1, March 2007 (2007-03-01), pages 35 - 50
OKUDA, K. ET AL., BIOL PHARM BULL., vol. 29, no. 7, July 2006 (2006-07-01), pages 1401 - 1403
ALHOSSARY A. ET AL.: "Fast, Accurate, and Reliable Molecular Docking with QuickVina 2", BIOINFORMATICS, vol. 31, no. 13, 2015, pages 2214 - 2216
O. TROTTA. J. OLSON: "AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization and multithreading", JOURNAL OF COMPUTATIONAL CHEMISTRY, vol. 31, 2010, pages 455 - 461
FEINSTEIN WPBRYLINSKI M: "Calculating an optimal box size for ligand docking and virtual screening against experimental and predicted binding pockets", J. CHEMINFORM, vol. 7, no. 1, 2015, pages 18
TETKO, I. V ET AL.: "Virtual computational chemistry laboratory - design and description", J. COMPUT. AID. MOL. DES, vol. 19, 2005, pages 453 - 63, XP019248187
"VCCLAB, Virtual", COMPUTATIONAL CHEMISTRY LABORATORY, Retrieved from the Internet
PANDY-SZEKERES GMUNK CTSONKOV TMMORDALSKI SHARPSØE KHAUSER ASBOJARSKI AJGLORIAM DE: "GPCRdb in 2018: adding GPCR structure models and ligands", NUCLEIC ACIDS RES, 16 November 2017 (2017-11-16), Retrieved from the Internet
Attorney, Agent or Firm:
PAPULA OY (FI)
Download PDF:
Claims:
CLAIMS 1. A compound of formula (I) , wherein R1, R2, R3, R4, and R5 are each independently selected from the group consisting of H, halogen, C1-4- alkyl, C1-3-(per)haloalkyl, C1-3-alkoxy, C1-3-(per)haloal- koxy, and OH; or R1, R2, and R5 are each independently selected from the group consisting of H, halogen, C1-4-alkyl, C1- 3-(per)haloalkyl, C1-3-alkoxy, C1-3-(per)haloalkoxy, and OH, and R3 and R4 together form a group selected from OC(R’)2O; or R1, R4, and R5 are each independently selected from the group consisting of H, halogen, C1-4-alkyl, C1- 3-(per)haloalkyl, C1-3-alkoxy, C1-3-(per)haloalkoxy, and OH, and R2 and R3 together form a group selected from OC(R’)2O; R6 and R7 are each independently selected from the group consisting of H, methyl, and ethyl; R8 is selected from the group consisting of H, methyl, ethyl, CH2Ph(o-OMe), CH2Ph(o-OH), C(O)CH2NH2, C(O)CH(NH2)(CH2)4NH2, C(O)CH(NH2)(CH2)3 N(H)C(NH)(NH2), 5- (1,2-dithiolan-3-yl)valeryl, and 2-cyano-1-phenylethyl; R9 is selected from the group consisting of CO2R10, and COSR10; R10 is selected from the group consisting of H, methyl, and ethyl; and each R’ is independently selected from the group consisting of H, and F; or a stereoisomer or a pharmaceutically ac- ceptable salt thereof.

2. The compound as claimed in claim 1, wherein R9 is selected from the group consisting of CO2R10; and R10 is as defined in claim 1; or a stereoisomer or a pharmaceutically ac- ceptable salt thereof. 3. The compound as claimed in any of the preceding claims, wherein R1, R2 and R5 are H; R3 and R4 are each independently selected from the group consisting of H, halogen, C1-4-alkyl; R9 is CO2R10; and R10 is as defined in claim 1; or a stereoisomer or a pharmaceutically ac- ceptable salt thereof. 4. The compound as claimed in any of the pre- ceding claims, wherein at least one of R6, R7, and R8 is each inde- pendently selected from the group consisting of methyl, and ethyl, and the other ones of R6, R7, and R8 are each independently selected from the group consisting of H, methyl, and ethyl; or a stereoisomer or a pharmaceutically ac- ceptable salt thereof. 5. The compound as claimed in any of the pre- ceding claims, wherein at least one of R6, R7, and R8 is each inde- pendently selected from the group consisting of methyl, and ethyl, and the other ones of R6, R7, and R8 are each independently selected from the group consisting of H, methyl, and ethyl; R9 is CO2R10; and R10 is selected from the group consisting of methyl, and ethyl; or a stereoisomer or a pharmaceutically ac- ceptable salt thereof.

6. The compound as claimed in any of the pre- ceding claims, wherein R6 and R7 are each independently selected from the group consisting of methyl, and ethyl, or one of R6 and R7 is selected from the group consisting of methyl, and ethyl, and the other one of R6 and R7 is H; R8 is selected from the group consisting of H, methyl, and ethyl; R9 is CO2R10; and R10 is selected from the group consisting of methyl, and ethyl; or a stereoisomer or a pharmaceutically ac- ceptable salt thereof. 7. The compound as claimed in any of claims 1 – 5, wherein R6 and R7 are both H; R9 is CO2R10; and R10 is selected from the group consisting of methyl, and ethyl; or a stereoisomer or a pharmaceutically ac- ceptable salt thereof. 8. The compound as claimed in any of claims 1 – 2 or claims 4 - 7, wherein R1, R2 and R5 are H; R3 and R4 are each independently selected from the group consisting of H, halogen, C1-4-alkyl, C1-3- (per)haloalkyl, C1-3-alkoxy, C1-3-(per)haloalkoxy, and OH, with the provision that if one of R3 or R4 is H then at least one of R6, and R7 is each independently selected from the group consisting of methyl, and ethyl, and the other one of R6, and R7 is selected from the group con- sisting of H, methyl, and ethyl, or R6, and R7 are both H, and R8 is selected from the group consisting of me- thyl, ethyl, CH2Ph(o-OMe), CH2Ph(o-OH), C(O)CH2NH2, C(O)CH(NH2)(CH2)4NH2, C(O)CH(NH2)(CH2)3 N(H)C(NH)(NH2), 5- (1,2-dithiolan-3-yl)valeryl, and 2-cyano-1-phenylethyl; or a stereoisomer or a pharmaceutically ac- ceptable salt thereof. 9. The compound as claimed in any of the pre- ceding claims, wherein R1, R2 and R5 are H; one of R3 and R4 is selected from the group consisting of F, Cl, methyl, and ethyl, and the other one of R3 and R4 is selected from the group consisting of H, F, methyl, and ethyl; R9 is CO2R10; and R10 is selected from the group consisting of methyl, and ethyl; or a stereoisomer or a pharmaceutically ac- ceptable salt thereof. 10. The compound as claimed in any of claims 1 - 8, wherein R1, R2 and R5 are H; and R3 and R4 are both halogen; or one of R3 and R4 is halogen or methyl, and the other one of R3 and R4 is H provided that R8 is selected from the group consisting of methyl, ethyl, CH2Ph(o-OMe), CH2Ph(o-OH), C(O)CH2NH2, C(O)CH(NH2)(CH2)4NH2, C(O)CH(NH2)(CH2)3 N(H)C(NH)(NH2), 5- (1,2-dithiolan-3-yl)valeryl, and 2-cyano-1-phenylethyl, or provided that at least one of R6, and R7 is each independently selected from the group consisting of me- thyl, and ethyl, and the other one of R6, and R7 is selected from the group consisting of H, methyl, and ethyl; or a stereoisomer or a pharmaceutically ac- ceptable salt thereof. 11. The compound as claimed in any of claims 1 – 5 or claims 7 – 10, wherein R1, R2 and R5 are H; R3 and R4 are both F, or one of R3 and R4 is F and the other one of R3 and R4 is H; R6 and R7 are each independently selected from the group consisting of H, and methyl; R9 is CO2R10; and R10 is selected from the group consisting of methyl, and ethyl; or a stereoisomer or a pharmaceutically ac- ceptable salt thereof. 12. The compound as claimed in any of claims 1 – 5 or claims 7 – 11, wherein R1, R2 and R5 are H; R3 and R4 are both F; R6 and R7 are each independently selected from the group consisting of H, and methyl; R9 is CO2R10; and R10 is selected from the group consisting of methyl, and ethyl; or a stereoisomer or a pharmaceutically ac- ceptable salt thereof. 13. The compound as claimed in any of claims 1 – 10, wherein R1, R2 and R5 are H; one of R3 and R4 is methyl and the other one of R3 and R4 is H; R9 is CO2R10; and R10 is selected from the group consisting of methyl, and ethyl; or a stereoisomer or a pharmaceutically ac- ceptable salt thereof. 14. The compound as claimed in any of claims 1 – 12, wherein R1, R2 and R5 are H; R3 and R4 are both F; R8 is selected from the group consisting of methyl, and ethyl; R9 is CO2R10; and R10 is selected from the group consisting of methyl, and ethyl; or a stereoisomer or a pharmaceutically ac- ceptable salt thereof. 15. The compound as claimed in any of the pre- ceding claims, wherein the compound is selected from the group consisting of: ethyl 3-(3,4-difluorophenyl)-1,2,4-trime- thylazetidine-3-carboxylate (1); ethyl 3-(3,4-difluorophenyl)-1-ethylazet- idine-3-carboxylate (2); ethyl 3-(3,4-difluorophenyl)-1-methylazet- idine-3-carboxylate (3); ethyl 3-(3,4-difluorophenyl)azetidine-3-car- boxylate (4); ethyl 3-(3-fluorophenyl)-2,4-dimethylazet- idine-3-carboxylate (5); ethyl 3-(3-fluorophenyl)-2-methylazetidine-3- carboxylate (6); ethyl 3-(4-methylphenyl)azetidine-3-carbox- ylate (7); methyl 2-methyl-3-(3-methylphenyl)azetidine- 3-carboxylate (8); methyl 3-(3,4-difluorophenyl)-1-ethyl-2- methylazetidine-3-carboxylate (9); methyl 3-(3,4-difluorophenyl)-2-methylazet- idine-3-carboxylate (10); ethyl 2-methyl-3-(3-methylphenyl)azetidine-3- carboxylate (11); ethyl 1-ethyl-3-(4-methylphenyl)azetidine-3- carboxylate (12); ethyl 3-(3,4-difluorophenyl)-2,4-dime- thylazetidine-3-carboxylate (13); methyl 3-(3,4-difluorophenyl)-1-ethyl-2,4-di- methylazetidine-3-carboxylate (14); methyl 1-ethyl-2-methyl-3-(3- methylphenyl)azetidine-3-carboxylate (15); ethyl 3-(3-methylphenyl)azetidine-3-carbox- ylate (16); ethyl 3-(3-fluorophenyl)-1,2,4-trimethylazet- idine-3-carboxylate (17); ethyl 1-ethyl-2,4-dimethyl-3-(3- methylphenyl)azetidine-3-carboxylate (18); ethyl 3-(3,4-difluorophenyl)-2-methylazet- idine-3-carboxylate (19); ethyl 3-(4-methoxyphenyl)azetidine-3-carbox- ylate (20); ethyl 1-ethyl-3-(3-methylphenyl)azetidine-3- carboxylate (21); methyl 1-ethyl-3-(3-methoxyphenyl)-2,4-dime- thylazetidine-3-carboxylate (22); ethyl 1-ethyl-2,4-dimethyl-3-(4- methylphenyl)azetidine-3-carboxylate (23); ethyl 2,4-dimethyl-3-(3-methylphenyl)azet- idine-3-carboxylate (24); methyl 3-(3,4-difluorophenyl)-2,4-dime- thylazetidine-3-carboxylate (25); ethyl 1,2-dimethyl-3-(3-methylphenyl)azet- idine-3-carboxylate (26); methyl 1-ethyl-2,4-dimethyl-3-(3- methylphenyl)azetidine-3-carboxylate (27); methyl 1-ethyl-2-methyl-3-(4- methylphenyl)azetidine-3-carboxylate (28); methyl 1-ethyl-3-(4-methoxyphenyl)-2,4-dime- thylazetidine-3-carboxylate (29); methyl 1-ethyl-3-(4-methoxyphenyl)-2- methylazetidine-3-carboxylate (30); methyl 2,4-dimethyl-3-(4-methylphenyl)azet- idine-3-carboxylate (31); ethyl 3-(3,4-difluorophenyl)-1-ethyl-2- methylazetidine-3-carboxylate (32); methyl 2-methyl-3-(4-methylphenyl)azetidine- 3-carboxylate (33); methyl 3-(4-methylphenyl)-1,2-dimethylazet- idine-3-carboxylate (34); ethyl 3-(3-fluorophenyl)azetidine-3-carbox- ylate (35); ethyl 1,2-dimethyl-3-(4-methylphenyl)azet- idine-3-carboxylate (36); and methyl 1-ethyl-3-(3-fluorophenyl)-2,4-dime- thylazetidine-3-carboxylate (37); or a stereoisomer or a pharmaceutically accepta- ble salt thereof. 16. A pharmaceutical composition comprising an effective amount of one or more compounds, or a stere- oisomer or a pharmaceutically acceptable salt thereof, as defined in any of the preceding claims, together with one or more pharmaceutically acceptable excipient(s). 17. The pharmaceutical composition as claimed in claim 16, in combination with one or more pharmaceu- tically acceptable carrier(s). 18. The pharmaceutical composition as claimed in any of claims 16 – 17, in combination with one or more other active ingredient(s). 19. A compound, or a stereoisomer or pharmaceu- tically acceptable salt thereof, as defined in any of claims 1 – 15, for use as a medicament. 20. A compound, or a stereoisomer or pharmaceu- tically acceptable salt thereof, as defined in any of claims 1 – 15, for use in the treatment or prevention of drug addiction or a CNS related disease or condition. 21. A compound, or a stereoisomer or pharmaceu- tically acceptable salt thereof, as defined in any of claims 1 – 15, for the use in the treatment or prevention of drug addiction or a CNS related disease or condition as claimed in claim 17, wherein the drug addiction or the CNS related disease or condition is selected from the group consisting of stimulant addiction, ADHD, ADD, sluggish cognitive tempo, concentration deficit disor- der, motivational or reward system dysfunction, autism spectrum disorder, disruptive, impulse control, and con- duct disorders, anxiety disorders, eating disorders, depression, dysthymia, Alzheimer’s disease, Parkinson’s disease, hyperactivity, narcolepsy, and alcoholism. 22. A method for the preparation of a compound of formula (I), or pharmaceutically acceptable salt or a stereoisomer thereof, as defined in any one of claims 1 – 15, comprising: (i) providing a compound of formula (II) , wherein R1, R2, R3, R4, R5, R6, and R7 are as defined in claim 1; and R8 is methyl, ethyl, CH2Ph(o-OMe), or a protecting group; (ii) reacting the compound of formula (II) with one or more compounds each independently selected from the group consisting of acids, bases, and water; or reacting the compound of formula (II) with methanol or ethanol, and a Lewis catalyst; (iii) optionally performing an esterifica- tion with a compound of formula (III) R11-R’’ (III), wherein R11 is methyl or ethyl; and R’’ is selected from the group consisting of halogen, SR’’’, OR’’’, and a first activating group, wherein R’’’ is H, or a second activating group, optionally in the presence of one or more activating group reactant(s) and/or one or more activating agent(s); (iv) optionally performing one or more first deprotection reaction(s); (v) optionally performing a transesteri- fication with a compound of formula (III), wherein R11 is methyl or ethyl; and R’’ is selected from the group consisting of SH, and OH, optionally in the presence of one or more activating group reactant(s) and/or one or more activating agent(s); (vi) optionally performing an amidation with a compound of formula (IV) R12-R8’ (IV), wherein R12 is HO, R13O, or R13, wherein R13 is an activating group; and R8’ is C(O)CH2NHR14, C(O)CH(NHR14)(CH2)4NHR14, C(O)CH(NHR14)(CH2)3N(R14)C(NR14)(NHR14), or 5-(1,2-dithio- lan-3-yl)valeryl, wherein each R14 is independently se- lected from the group consisting of H, and protecting groups, optionally in the presence of one or more acti- vating group reactant(s) and/or one or more activating agent(s); (vii) optionally performing a reductive al- kylation with a compound of formula (V) R8’’-CHO (V), wherein R8’’ is H, or CH3; and a reducing agent; (viii) optionally performing an N-alkylation with a compound of formula (VI) R8’’’-R15 (VI), wherein R8’’’ is methyl, ethyl, (o-OMe)PhCH2, or (Ph)CH(CH2CN); and R15 is selected from the group consisting of halogen, SR16, and OR16, wherein R16 is an activating group, op- tionally in the presence of one or more activating group reactant(s) and/or one or more activating agent(s); (ix) optionally performing one or more second deprotection reaction(s); to obtain the compound of formula (I), wherein R1, R2, R3, R4, R5, R6, R7, R8, R9 and R10 are as defined in claim 1; and (x) optionally converting the compound of formula (I) to a pharmaceutically acceptable salt thereof.

Description:
NOVEL HETEROCYCLES, PREPARATION METHODS AND USES THEREOF TECHNICAL FIELD The present invention relates to novel compounds derived from 3,3-disubstituted azetidines and pharmaceutical compositions comprising said compounds. The invention also relates to said compounds for use in the treatment or prevention of drug addiction or a CNS related disease or condition, in addition to methods for the preparation of said compounds. BACKGROUND Azacyclobutane (azetidine) is a heterocyclic compound containing three carbon atoms and one nitrogen atom. Azetidine and its derivatives are relatively rare structural motifs in natural products. They are a component of mugineic acids and penaresidins. An azetidine containing natural product is azetidine-2- carboxylic acid, which is a toxic mimic of proline. EA202092955A1 discloses certain 3,3- disubstituted azetidines as LPAl receptor modulators in the treatment of fibrosis. US2012232026A1 discloses as antiparasitic agents certain 3,3-disubstituted azetidines that are spirocyclic isoxazoline derivatives. WO2005049605A1 discloses antibacterial aminoquinazolidinedione derivatives, wherein some of the derivatives contain a 3,3-disubstituted azetidine- 1-yl group. WO2016193241A1 discloses certain 3,3- disubstituted azetidines having muscarinic receptor antagonist and beta2 adrenergic receptor agonist activity. WO2016177849A1 discloses certain compounds containing a 3,3-disubstituted azetidine-1-yl group, wherein the compounds are both phosphodiesterase 4 (PDE4) enzyme inhibitors and muscarinic M3 receptor antagonists for use in the prevention and/or treatment asthma or COPD. TAAR1 (trace amine associated receptor 1) is a protein that in humans is encoded by the TAAR1 gene. TAAR1 plays a role in regulating neurotransmission in dopamine, norepinephrine, and serotonin neurons in the CNS and it affects immune system and neuroimmune system function through different mechanisms. Among endogenous ligands of the human TAAR1 (hTAAR1) receptor are tyramine, β-phenethylamine (PEA), dopamine, and octopamine. TAAR1 is a high-affinity receptor for amphetamine, methamphetamine, and trace amines which mediates some of their cellular effects in monoamine neurons within the central nervous system. Low PEA concentration in the brain is associated with major depressive disorder, and high concentrations are associated with schizophrenia. In addition, low PEA levels and under-activation of TAAR1 seems to be associated with attention deficit hyperactivity disorder (ADHD). TAAR1-selective ligands have been proposed to have therapeutic potential for treating psychostimulant addictions (e.g., cocaine, amphetamine, methamphetamine). The dopamine transporter (DAT) is a membrane- spanning protein that pumps the neurotransmitter dopamine out of the synaptic cleft back into cytosol. Therefore, DAT is implicated in a number of dopamine- related disorders, including ADHD, bipolar disorder, alcoholism, and substance use disorder (drug addiction, alcoholism). In addition, increased activity of DAT is linked to clinical depression. Also dopamine receptors are involved in many neurological disorders, diseases, and conditions, including ADHD, Parkinson's disease (PD), schizophrenia, neuroleptic malignant syndrome, drug addiction, alcoholism, social phobia, and Tourette's syndrome. Dopamine receptors bind dopamine; while psychostimulants are typically dopamine receptors’ indirect agonists, dopamine receptors antagonists often work as antipsychotics. As dopamine receptors, also adrenergic receptors are a class of G protein-coupled receptors (GPCRs). Adrenergic receptors are targets of catecholamines like norepinephrine and epinephrine. FDA-approved drugs that bind to adrenergic receptors are used to treat e.g. hypertension, and ADHD, and agonists to β2-adrenergic receptors have been found to be effective in the treatment of PD. Serotonin receptors are also GPCRs and are ligand-gated ion channels that are activated by the endogenous neurotransmitter serotonin. Serotonin receptors are found in the central and peripheral nervous systems. Various biological and neurological processes that serotonin receptors influence are e.g. aggression, anxiety, appetite, cognition, learning, memory, mood, nausea, sleep, and thermoregulation. Therefore, serotonin receptors are the target of drugs, such as antidepressants, antipsychotics, anorectics, antiemetics, gastroprokinetic agents, antimigraine agents, hallucinogens, and entactogens. The NMDA receptor (N-methyl-D-aspartate receptor) is a glutamate receptor and ion channel found in neurons. The NMDA receptor may be important for controlling synaptic plasticity and mediating learning and memory functions. NMDA receptor inhibitors and antagonists are used for the treatment of Alzheimer's disease and depression, and antagonists are used as anasthetics. Cholinergic receptors are receptors and integral membrane proteins that are activated by the neurotransmitter acetylcholine. Among cholinergic receptors are the nicotinic acetylcholine (nAChR) and muscarinic acetylcholine (mAChR) receptors. Monoamine oxidase A (MAOA) is an enzyme that in humans is encoded by the MAOA gene. Diseases, disorders, and conditions associated with MAOA include Alzheimer's disease (AD), aggression, panic disorder, bipolar affective disorder, depression, ADHD, and antisocial behaviour. Catechol-O-methyltransferase (COMT) is one of several enzymes that inactivates catecholamines such as the neurotransmitters dopamine, epinephrine, and norepinephrine, and catecholestrogens, all having a catechol structure, by introducing a methyl group to the catechol structure. COMT inhibitors are used for the treatment of PD. Dopamine beta-hydroxylase (DBH) is an enzyme that catalyzes the conversion of dopamine to norepinephrine. DBH has been suggested as the target of drugs in diseases, conditions, and disorders such as drug addiction, alcoholism, ADHD, schizophrenia, and AD. The μ-opioid receptors (MORs) are inhibitory GPCPs that have a high affinity for the endogenous peptides enkephalins and β-endorphin. MOR agonists are used for treating or preventing pain. SUMMARY This Summary is provided to introduce a selection of concepts in a simplified form that are further described below in the Detailed Description. This Summary is not intended to identify key features or essential features of the claimed subject matter, nor is it intended to be used to limit the scope of the claimed subject matter. An object of the present invention is to provide compounds useful in treating or preventing drug addiction or a CNS related disease or condition. It has now surprisingly been found that compounds derived from 3,3-disubstituted azetidines show binding affinities to proteins associated to drug addiction or a CNS related disease or condition. Disclosed are compounds of formula (I) , wherein R 1 , R 2 , R 3 , R 4 , and R 5 are each independently selected from the group consisting of H, halogen, C 1-4 - alkyl, C 1-3 -(per)haloalkyl, C 1-3 -alkoxy, C 1-3 -(per)haloal- koxy, and OH; or R 1 , R 2 , and R 5 are each independently selected from the group consisting of H, halogen, C 1-4 -alkyl, C 1- 3 -(per)haloalkyl, C 1-3 -alkoxy, C 1-3 -(per)haloalkoxy, and OH, and R 3 and R 4 together form a group selected from OC(R’) 2 O; or R 1 , R 4 , and R 5 are each independently selected from the group consisting of H, halogen, C 1-4 -alkyl, C 1- 3 -(per)haloalkyl, C 1-3 -alkoxy, C 1-3 -(per)haloalkoxy, and OH, and R 2 and R 3 together form a group selected from OC(R’) 2 O; R 6 and R 7 are each independently selected from the group consisting of H, methyl, and ethyl; R 8 is selected from the group consisting of H, methyl, ethyl, CH 2 Ph(o-OMe), CH 2 Ph(o-OH), C(O)CH 2 NH 2 , C(O)CH(NH 2 )(CH 2 ) 4 NH 2 , C(O)CH(NH 2 )(CH 2 ) 3 N(H)C(NH)(NH 2 ), 5- (1,2-dithiolan-3-yl)valeryl, and 2-cyano-1-phenylethyl; R 9 is selected from the group consisting of CO 2 R 10 , and COSR 10 ; R 10 is selected from the group consisting of H, methyl, and ethyl; and each R’ is independently selected from the group consisting of H, and F; or a stereoisomer or a pharmaceutically ac- ceptable salt thereof. The present disclosure also provides pharmaceutical compositions comprising an effective amount of one or more compounds, or a stereoisomer or a pharmaceutically acceptable salt thereof, as defined in the present disclosure, together with one or more pharmaceutically acceptable excipient(s). The present disclosure also provides a compound of formula (I), or a stereoisomer or pharmaceutically acceptable salt thereof, for use as a medicament. The present disclosure also provides a compound of formula (I), or a stereoisomer or pharmaceutically acceptable salt thereof, for use in the treatment or prevention of drug addiction or a CNS related disease or condition. Further, the present disclosure provides a compound of formula (I), or a stereoisomer or pharmaceutically acceptable salt thereof, for use in the treatment or prevention of drug addiction or a CNS related disease or condition, wherein the drug addiction or the CNS related disease or condition is selected from the group consisting of stimulant addiction, ADHD, ADD, sluggish cognitive tempo, concentration deficit disorder, motivational or reward system dysfunction, autism spectrum disorder, disruptive, impulse control, and conduct disorders, anxiety disorders, eating disorders, depression, dysthymia, Alzheimer’s disease, Parkinson’s disease, hyperactivity, narcolepsy, and alcoholism. Further, the present disclosure provides methods for the preparation of compounds of formula (I), or pharmaceutically acceptable salts or a stereoisomers thereof. The objects of the invention are achieved by compounds and methods for the preparation thereof, and said compounds for use as a medicament that are characterized by what is stated in the independent claims. The preferred embodiments of the invention are disclosed in the dependent claims. DETAILED DESCRIPTION Compounds as disclosed herein and hereafter are derivatives of 3,3-disubstituted azetidines that provide the inventive properties of the compounds of the present invention. Specific substitution patterns of the compounds are beneficial for binding affinity for proteins that are associated with drug addiction or a CNS related disease or condition. “Comprises” or “comprising” denotes that the subsequently described feature(s) or act(s) may but need not include other feature(s) or act(s). It will further be understood that reference to 'an' item refers to one or more of those items. “Optional” or “optionally” denotes that the subsequently described event or circumstance may but need not occur, and that the description includes instances where the event or circumstance occurs and instances in which it does not. The term “halogen” as used herein and hereafter by itself or as part of other groups refers to the Group VIIa elements and includes F, Cl, Br and I groups. The term “alkyl” as used herein and hereafter is an aliphatic linear, branched or cyclic, especially linear or branched, hydrocarbon group having the indicated number of carbon atoms, for example C 1-6 -alkyl has 1 to 6 carbon atoms in the alkyl moiety and thus, for example, C 1-4 -alkyl includes methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl and C 1-6 -alkyl additionally includes, but is not limited to, branched and straight chain pentyl and hexyl. Other examples of alkyl are benzyl (may be a protecting group), 2-methoxybenzyl (CH 2 Ph(o-OMe)), 2-hydroxybenzyl (CH 2 Ph(o-OH)), and 2-cyano-1-phenylethyl. The term “C 1-3 -(per)haloalkyl” as used herein and hereafter refers to any of the above alkyl groups where one or more hydrogen atoms are replaced by halogen(s): in particular I, Br, F or Cl. Examples of haloalkyl groups include without limitation chloromethyl, fluoromethyl and -CH 2 CF 3 . The term “perhaloalkyl” is understood to refer to an alkyl group, in which all the hydrogen atoms are replaced by halogen atoms. Examples of perhaloalkyls include, but are not limited to, trifluoromethyl (-CF 3 ) and trichloromethyl (-CCl 3 ). The term “C 1-3 -alkoxy” as used herein and hereafter refers to a –O-(C 1-3 -alkyl) group where the “C 1-3 -alkyl” has the above-defined meaning. Examples of alkoxy groups include, but are not limited to, methoxy, ethoxy, and iso-propyloxy. The term “C 1-3 -(per)haloalkoxy” as used herein and hereafter refers to a –O-(C 1-3 -(per)haloalkyl) group where the C 1-3 -(per)haloalkyl has the above-defined meaning. Examples of (per)haloalkoxy groups include, but are not limited to, trifluoromethoxy, 2,2,2- trichloromethoxy, and 1,1,1,3,3,3-hexafluoro- isopropoxy. The term “stereoisomer” as used herein and hereafter refers to stereoisomers of compounds. Examples of stereoisomers include, but are not limited to, enantiomers, diastereomers, cis–trans-isomers, and E-Z- isomers. The term “pharmaceutically acceptable salt” as used herein and hereafter refers to salts which are known to be non-toxic and are commonly used in the pharmaceutical literature. Typically, these are acid addition salts or base addition salts of the referred compounds as disclosed herein and hereafter. The expression “acid addition salt” includes any non-toxic organic and inorganic acid addition salts that that the compounds of formula (I) can form. Illustrative inorganic acids, which form suitable acid addition salts, include, but are not limited to, hydrogen chloride, hydrogen bromide, sulphuric and phosphoric acids. Illustrative organic acids, which form suitable acid addition salts, include, but are not limited to, acetic acid, lactic acid, malonic acid, succinic acid, glutaric acid, fumaric acid, malic acid, tartaric acid, citric acid, ascorbic acid, maleic acid, benzoic acid, phenylacetic acid, cinnamic acid, methane sulfonic acid, salicylic acid, and the like. The term “acid addition salt” as used herein also comprises solvates which the compounds and salts thereof are able to form, such as, for example, hydrates, alcoholates, and the like. These salts also include salts useful for the chiral resolution of racemates. The expression “base addition salt” includes any non-toxic base addition salts that the compounds of formula (I) can form. Suitable base addition salts include, but are not limited to, those derived from inorganic bases such as aluminum, ammonium, calcium, copper, iron, lithium, magnesium, manganese, potassium, sodium, and zinc salts, in particular sodium and ammonium salts. Further examples of organic base addition salts include salts of trialkylamines, such as triethyl amine and trimethyl amine, other salts of organic amines such as methylamine, dimethylamine, trimethylamine, ethylamine, triethylamine, morpholine, and the like, and choline salts. The term “base addition salt” as used herein also comprises solvates which the compounds and salts thereof are able to form, such as, for example, hydrates, alcoholates, and the like. These salts also include salts useful for the chiral resolution of racemates. Pharmaceutical compositions of the present invention may be administered in an effective amount within the dosage range of about 0.1 µg/kg to about 300 mg/kg, preferably between 1.0 µg/kg to 10 mg/kg body weight. Compounds of the present invention may be administered in a single daily dose, or the total daily dosage may be administered in divided doses of two, three, four, five or six times daily. The term “effective amount” refers to an amount of a composition or a pharmaceutical composition that confers a therapeutic effect on the treated subject. The therapeutic effect may be objective (i.e. measurable by some test or marker) or subjective (i.e. subject gives an indication of or feels an effect). Such treatment need not necessarily completely ameliorate the disease or condition. Further, such treatment or prevention can be used in conjunction with other traditional treatments for treating the disease or condition known to those skilled in the art. The effective amount will typically be determined by a physician, and depend on the disease or condition to be treated, the chosen route of administration, the actual compound administered, the age, gender, weight, and response of the individual patient, the severity of the patient's symptoms, and the like. Skilled artisans possess the knowledge and skill in the art to enable them to select suitable pharmaceutically acceptable excipients in appropriate amounts for use in the invention. In addition, there are a number of resources that are available to the skilled artisan which describe pharmaceutically acceptable excipients and may be useful in selecting suitable pharmaceutically acceptable excipients. Suitable pharmaceutically acceptable excipients include, but are not limited to, the following types of excipients: diluents (for example starches, mannitol), fillers (for example lactose, microcrystalline cellulose or calcium hydrogen phosphate), binders (for example pre-gelatised corn starch, polyvinylpyrrolidone or methylcellulose), additives (for example magnesium stearate, talc, silica), disintegrants (for example potato starch), lubricants (for example sodium lauryl sulphate), glidants (for example fumed silica, talc, magnesium carbonate), granulating agents (for example water, ethanol), coating agents (for example hydroxypropyl methylcellulose, gelatin, waxes, shellac, plastics, plant fibers), wetting agents (for example sorbitan monopalmitate, poloxamer 407), solvents (for example water), co-solvents (for example ethanol, propylene glycol), suspending agents (for example sorbitol, cellulose derivatives, edible hydrogenated fats), emulsifiers (for example lecithin or acacia), sweeteners (for example sucrose), flavoring agents (for example cherry, lime), flavor masking agents (for example vanilla, citrus), coloring agents (for example titanium oxide), anti-caking agents (for example silicon dioxide), humectants (for example glycerine, sorbitol), chelating agents (for example EDTA salts, histidine, aspartic acid), plasticizers (for example tributyl citrate, diethyl phthalate), viscosity increasing agents (for example methylcellulose), antioxidants (for example (ascorbic acid, cysteine), preservatives (for example methyl or propyl p-hydroxybenzoates, sorbic acid or ascorbic acid), stabilizers (for example polysorbate 20 & 80, poloxamer 407), surfactants (for example polyethylene glycol, polysorbate 80), and buffering agents (for example sodium and potassium phosphates, citrate, acetate, carbonate or glycine buffers de- pending on the targeted pH-range). Excipients and/or auxiliaries may facilitate processing of the active agent(s) into preparations that can be used pharmaceutically. The skilled artisan will appreciate that certain pharmaceutically acceptable excipients may serve more than one function and may serve alternative functions depending on how much of the excipient is present in the pharmaceutical composition and what other ingredients are present in the pharmaceutical composition. Pharmaceutical compositions of the invention are most preferably used alone or in combination i.e. administered simultaneously, separately, or sequentially with other active ingredients, e.g. pharmaceutically active compounds or biologic products. The amounts of the pharmaceutical composition(s) of the invention, particularly a pharmaceutical composition comprising a compound of formula (I), or stereoisomers or pharmaceutically acceptable salts thereof, and the other active ingredient(s) and the relative timings of administration will be selected in order to achieve the desired combined therapeutic effect. Pharmaceutical compositions of the invention may be administered by various routes, for example, parenteral, subcutaneous, intravenous, intra-articular, intrathecal, intramuscular, intraperitoneal, topical, and by intradermal injections, and via transdermal, rectal, buccal, oromucosal, nasal, ocular routes and via inhalation and via implant. Pharmaceutical compositions may be formulated into suitable pharmaceutical formulations; suitable pharmaceutical formulations include, for example, solutions, dispersions, suspensions, sterile aqueous or non-aqueous solvents, emulsions, powders, capsules, tablets, pills, controlled release capsules, controlled release tablets and controlled release pills. In addition, or alternatively, to pharmaceutically acceptable excipient(s) and/or other active ingredients(s), the pharmaceutical compositions may be combined with one or more pharmaceutically acceptable carrier(s). The term “pharmaceutically acceptable carrier(s)” as used herein and hereafter refers to substrates comprised in pharmaceutical compositions for drug delivery, which serves to improve the selectivity, effectiveness, and/or safety of drug administration. Examples of pharmaceutically acceptable carriers include, but are not limited to, pharmaceutically acceptable excipients, liposomes, (polymeric) micelles, microspheres, nanoparticles, and protein-drug conjugates. The pharmaceutical compositions of the invention are prepared using techniques and methods known to those skilled in the art. Pharmaceutical formulations of the pharmaceutical compositions as disclosed herein and hereafter for parenteral or topical use may include, but are not limited to, sterile aqueous and/or non-aqueous solvents, suspensions, and emulsions. Examples of non-aqueous solvents of pharmaceutical formulations are propylene glycol, polyethylene glycol, vegetable oil, fish oil, and injectable organic esters. Aqueous carriers include, but are not limited to, water, water-alcohol solutions, including saline and buffered medial parenteral vehicles including sodium chloride solution, Ringer’s dextrose solution, dextrose plus sodium chloride solution, Ringer’s solution containing lactose, or fixed oils. Intravenous vehicles include, but are not limited to, fluid and nutrient replenishers, electrolyte replenishers, such as those based on Ringer’s dextrose and the like. Aqueous pharmaceutical compositions according to the invention may comprise suitable buffer agents, such as sodium and potassium phosphates, citrate, acetate, carbonate or glycine buffers depending on the targeted pH-range. The use of sodium chloride as a tonicity adjuster is also useful. Pharmaceutical compositions may include other excipients, such as stabilizing agents or preservatives. Useful stabilizing excipients include surfactants (polysorbate 20 & 80, poloxamer 407), polymers (polyethylene glycols, povidones), carbohydrates (sucrose, mannitol, glucose, lactose), alcohols (sorbitol, glycerol propylene glycol, ethylene glycol), suitable proteins (albumin), suitable amino acids (glycine, glutamic acid), fatty acids (ethanolamine), antioxidants (ascorbic acid, cysteine etc.), chelating agents (EDTA salts, histidine, aspartic acid) or metal ions (Ca, Ni, Mg, Mn). Among useful preservative agents are benzyl alcohol, chlorobutanol, benzalkonium chloride and possibly parabens. The pharmaceutical composition according to the present invention may be provided in concentrated form or in form of a powder to be reconstituted on demand. In such cases formulations of powder for solution for injection/infusion excipients mentioned above may be used. In case of lyophilizing, certain cryoprotectants are preferred, including polymers (povidones, polyethylene glycol, dextran), sugars (sucrose, glucose, lactose), amino acids (glycine, arginine, glutamic acid) and albumin. If solution for reconstitution is added to the packaging, it may consist e.g., of pure water for injection or sodium chloride solution or dextrose or glucose solutions. The terms “treatment or prevention” as used herein and hereafter includes prophylaxis, or prevention of, as well as lowering the individual's risk of falling ill with the named disorder or condition, or alleviation, amelioration, elimination, or cure of the said disorder or condition once it has been established. The terms “administering” or “administered” to a subject or patient includes dispensing, delivering or applying a compound as disclosed herein and hereafter, or a stereoisomer or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition as disclosed herein and hereafter to the subject or patient by any suitable route for delivery of the composition or pharmaceutical composition to a site in the body where desired. Compounds of formula (I) of the present disclosure may be useful in therapy, especially in the treatment or prevention of drug addiction or a CNS related disease or condition in animals, in particular mammals, and humans. In particular, compounds of formula (I) possess pharmacological properties for the treatment and/or prophylaxis of drug addiction or a CNS related disease or condition that include, but are not limited to, stimulant addiction, ADHD, ADD, sluggish cognitive tempo, concentration deficit disorder, motivational or reward system dysfunction, autism spectrum disorder, disruptive, impulse control, and conduct disorders, anxiety disorders, eating disorders, depression, dysthymia, Alzheimer’s disease, Parkinson’s disease, anxiety, hyperactivity, narcolepsy, and alcoholism. The compounds of formula (I) of the present disclosure may bind to one or more protein involved in drug addiction or a CNS related disease or condition, wherein examples of the proteins include, but are not limited to, trace amine receptors (trace amine- associated receptors (TAARs), such as TAAR1), dopamine- and serotonin receptors, dopamine- and serotonin transporters, acyl and methyl transferases, norpinephrine transporter, monoamino-oxidases such as MAO-A, catecholine-O-methylransferase (COMT), adrenergic receptors, tyrosine hydroxylase, histamine receptors such as H1 histamine receptor, orexin receptors, NMDA-receptors, sigma-1 receptor, muscarinic and nicotinic acetylcholine receptors such as M1, M2, M3, and M4 muscarinic acetylcholine receptors, opioid receptors such as μ-opioid receptors (MOR), neuropeptide receptors such as Neuropeptide Y2 receptor, melanocortin receptors (excluding MC3R), neurokinin receptors such as NKR2 and NK3R, corticotropin-releasing factor receptor 1, acetylcholinesterase, 1B melatonin receptor, cholinergic receptors, and dopamine beta- hydroxylase (dopamine beta-monooxygenase, DBH), to name a few. The compounds of formula (I) as disclosed herein and hereafter may bind to and inhibit a protein including, but not limited to, HIV-1 reverse transcriptase, oxidosqualene cyclase, aldose reductase, and hepatitis C virus NS5B RNA-dependent RNA polymerase. The term “protecting group” as used herein and hereafter refers to a chemical group, which is covalently attached to an atom or a functional group, which has been modified by the protecting group. Said protecting group may enable chemoselectivity in a reaction, therefore, the protecting group may protect an atom or a functional group from, at least partially, reacting in a reaction. It is to be understood that the protecting group protects the atom or functional group fully or partly, i.e., the atom or the functional group protected with a protecting group may or may not react partly in a reaction. Skilled artisans possess the knowledge and skill in the art to enable them to select suitable protecting groups for the atoms or functional groups to be protected. In addition, there are a number of resources that are available to the skilled artisan which describe protecting groups and may be useful in selecting suitable protecting groups for the atoms or functional groups to be protected. For suitable protecting groups and methods to protect compounds with suitable protecting groups, see, for example, Protective Groups in Organic Synthesis, 4th Edition, 2007, John Wiley & Sons, Inc., Hoboken, New Jersey. Examples of atoms and functional groups that may be protected with protecting groups include, but are not limited to, O, S, N, NH, NH 2 , OH, SH, carbonyls such as aldehydes and ketones, ethers, esters, and amides. Examples of protecting groups include, but are not limited to, carbobenzyloxy (Cbz), p-methoxybenzyl carbonyl, BOC, Fmoc, acetyl, benzoyl, phenyl, benzyl, trityl, sulfonyls such as phenylsulfonyl, tosyl (Ts), mesyl, and trifyl; tosylate, silyl ethers such as trimethylsilyl (TMS), tert-butyldimethylsilyl (TBDMS), tri-iso- propylsilyloxymethyl (TOM), and triisopropylsilyl (TIPS) ethers; tetrahydropyranyl (THP), p-methoxyphenyl ether (PMP), p-methoxybenzyl ether (PMB), β- methoxyethoxymethyl ether (MEM), pivaloyl, thioethers, acetals, ketals, dithianes, benzyl esters, tert-butyl esters, orthoesters, and photolabile groups. The term “activating group” as used herein and hereafter refers to a functional group of a chemical compound that promotes a reaction to occur and/or has a positive influence of the overall reaction rate and/or have a directing effect on positional isomer of the products that are formed. Said activating group may or may not be part of the formed product, i.e. it is to be understood that the activating group may be present in the product, or the activating group may be a leaving group, or part of the leaving group, in e.g. S N 2, S N 1 and addition-elimination reactions. A compound disclosed herein may have one or more activating group(s) that may be the same or different. Examples of activating groups include, but are not limited to, sulfonyls such as phenylsulfonyl, tosyl (Ts), mesyl, and trifyl; halogens such as Cl, Br, I, and F; (substituted) amino groups such as 1-imidazolyl, ion pair of intermediates formed from the reaction between PPh 3 , azodicarboxylates, such as DEAD and DIAD, and carboxylic acid, amides, esters such as O-acylisourea of DCC and carboxyl acid, hydroxy, alkoxy such as 1H-1,2,3- benzotriazol-1-olate, anion of N-hydroxysuccinimide, acyloxy, phenyliodine(III) diacetate [PhI(OAc) 2 , (PIDA)], metals and complexes thereof such as Ni, Ru, Os, Ir, Rh, Pd, Fe-PNP; thiols, alkyls, (per)haloalkoxy, (per)haloalkyl, and photolabile groups. The term “activating group reactant” as used herein and hereafter refers to a chemical compound, comprising an activating group, wherein the chemical compound reacts with another chemical compound in a reaction to form a chemical compound or an intermediate comprising said activating group. It is to be understand that there may be different activating group reactants comprising the same activating group. A reaction may comprise one or more activating group reactant(s) that may be the same or different. Examples of activating group reactants include, but are not limited to, 1,1’- carbonyldiimidazole (CDI), diethyl azodicarboxylate (DEAD), diisopropyl azodicarboxylate (DIAD), N,N′- dicyclohexylcarbodiimide (DCC), triphenylphosphine (PPh 3 ), phenyliodine(III) diacetate [PhI(OAc) 2 , (PIDA)], sulfinic acids such as phenylsulfinic acid and p- toluenesulfinic acid; mesyl halides such as methanesulfonyl chloride; trifyl azide, trifluoromethanesulfonyl chloride, halogens such as Br 2 ; metal halides such as NaI; trifluoroacetyl chloride, and trifluoroacetic anhydride. The term “activating agent” as used herein and hereafter refers to a substance or chemical compound added to a reaction to facilitate a chemical reaction. Said activating agent may or may not be a catalyst. It is to be understood that said activating agent may or may not be consumed in the reaction. Examples of activating agents include, but are not limited to, acids such as HCl, and AcOH; bases such as NaOH and NaH; water, 4-dimethylaminopyridine (DMAP), triphenylphosphine (PPh 3 ), metal halides such as NaI; Lewis acids such as TiCl 4 , boron trifluoride, and boron trifluoride diethyl etherate. The terms “performing one or more reactions” as used herein and hereafter refers to one or more method steps, i.e., reacting a compound of a method for the preparation of a compound of formula (I) in an indicated reaction. It is to be understood that if e.g. two reactions are performed a first reaction may be performed forming one or more first reaction products, wherein at least one of the first reaction products may be used in the subsequent reactions forming one or more second reaction products that may or may not be the compound of formula (I). Further, the one or more (first, second, third, fourth, and/or fifth) reaction products may or may not be separated and/or purified after the (first, second, third, fourth, and/or fifth) reaction, i.e., the reactions product may or may not be used directly in subsequent reaction. In addition, one or more reactions may be performed simultaneously as long as the reactions conditions allows it. The term “esterification” as used herein and hereafter refers to a reaction wherein an ester or a thioester is formed from a carboxylic acid as disclosed herein and hereafter. The term “transesterification” as used herein and hereafter refers to a reaction wherein an alkoxy group of an ester or a thiolate group of a thioester is converted to another alkoxy group or thiolate group, i.e., an ester group or thioester group of a compound disclosed herein and hereafter is changed to another ester group or thioester group. Skilled artisans possess the knowledge and skill in the art to enable them to select suitable reactants and reagents for the transesterification. Examples of transesterification reactions include, but are not limited to, combining HCl, methanol or ethanol or methanethiol or ethanethiol, and a compound of a method for the preparation of a compound of formula (I) to form a first composition, and optionally heating the formed first composition. The term “amidation” as used herein and hereafter refers to a reaction, wherein an ester group, a carboxylic acid group, and/or an amine group of a compound of a method for the preparation of a compound of formula (I) is converted to an amide group. I.e., the carboxylic group, the ester group, and/or the amine group of a compound of a method for the preparation of a compound of formula (I) is reacting forming an amide group of said compound. Preferably, the amine group of an azetidine as disclosed herein and hereafter is converted to an amide group. Examples of amidation reactions include, but are not limited to, combining a carboxylic acid and a compound of a method for the preparation of a compound of formula (I), preferably an azetidine, optionally in the presence of one or more activating group reactant(s) and/or one or more activating agent(s), to form a first composition, and optionally heating the formed first composition to form an amide. The term “reductive alkylation” as used herein and hereafter refers to reductive amination, wherein a carbonyl group of a chemical compound is converted to an amine via an intermediate imine. Examples of reductive alkylation reactions include, but are not limited to, combining methanal or ethanal with a compound comprising a secondary amine group of a method for the preparation of a compound of formula (I) (i.e., for example a compound of formula (I), wherein R 8 is H, and R 10 is methyl or ethyl), optionally in the presence of one or more water removing compounds or apparatus arranged to remove water, forming an imine that is combined with a reducing agent, such as sodium borohydride, to form a tertiary amine. The term “N-alkylation” as used herein and hereafter refers to a reaction, wherein an amine group is alkylated. Examples of N-alkylation reactions include, but are not limited to, combining a haloalkane such as methylbromide, ethylbromide, methyliodide, and ethyliodide, and a secondary amine group comprising compound of a method for the preparation of a compound of formula (I), optionally in the presence of one or more activating group reactant(s) and/or one or more activating agent(s) such as sodium iodide, to form a N- alkylated compound. The term “reduction” as used herein and hereafter refers to a reaction, wherein a functional group of a chemical compound is reduced, i.e., the oxidation state of a first functional group is reduced thereby forming a second functional group with a lower oxidation state compared to the oxidation state of the first functional group. The term “hydrolysis” as used herein and hereafter refers to a chemical reaction in which a molecule of water breaks one or more chemical bonds. Typically, hydrolysis is performed on a carboxylic ester forming the respective carboxylic acid, or on an amide forming the respective carboxylic acid. The term “O-alkylation” as used herein and hereafter refers to a reaction, wherein an OH group is alkylated. Examples of O-alkylation reactions include, but are not limited to, combining a haloalkane such as methylbromide, ethylbromide, methyliodide, and ethyliodide, and a hydroxy group comprising compound of a method for the preparation of a compound of formula (I), optionally in the presence of one or more bases, one or more activating group reactant(s) and/or one or more activating agent(s) such as sodium iodide, to form a O-alkylated compound. The term “reductive trifluoromethylation” as used herein and hereafter refers to a chemical reaction that entails the introduction of one or more CF3 into a compound by also reducing the oxidation state of said compound. An example of reductive trifluoromethylation include, but is not limited to, the transformation of a C(O)NHMe group of a compound to a CH(CF 3 )NHMe group of the compound, see, for example, Chen et al., Chemoselective direct reductive trifluoromethylation of amides: a flexible access to functionalized α- trifluoromethylamines, Org. Chem. Front., 2018,5, 943- 947. Particularly, a compound having one or more CF 3 groups has enhanced metabolic stability, lipophilicity, and bioavailability. The term “fluorination” as used herein and hereafter refers to a chemical reaction that entails the introduction of one or more F into a compound. The terms “performing one or more deprotection reaction(s)” as used herein and hereafter refers to performing one or more reaction(s), wherein a protecting group is removed from a compound. Skilled artisans possess the knowledge and skill in the art to enable them to select suitable reactants or reagents for the deprotection reaction(s). In addition, there are a number of resources that are available to the skilled artisan which describe suitable reagents and reactants and may be useful in selecting suitable reagents and reactants for the protecting groups to be deprotected. For suitable deprotecting reactions, reactants and reagents, see, for example, Protective Groups in Organic Synthesis, 4th Edition, 2007, John Wiley & Sons, Inc., Hoboken, New Jersey. Examples of reactants and reagents usable in the deprotection reaction(s) (with examples of the protecting group to be removed in parenthesis) include, but are not limited to, tetra-n-butylammonium fluoride (TMS), H 2 (benzyl), bases such as NaOH (acetyl), acids such as pyridinium p-toluenesulfonate and EtOH (THP), and 2,3-dichloro-5,6-dicyano-p- benzoquinone (PMB). In one aspect is disclosed a compound of formula (I) , wherein R 1 , R 2 , R 3 , R 4 , and R 5 are each independently selected from the group consisting of H, halogen, C 1-4 - alkyl, C 1-3 -(per)haloalkyl, C 1-3 -alkoxy, C 1-3 -(per)haloal- koxy, and OH; or R 1 , R 2 , and R 5 are each independently selected from the group consisting of H, halogen, C 1-4 -alkyl, C 1- 3 -(per)haloalkyl, C 1-3 -alkoxy, C 1-3 -(per)haloalkoxy, and OH, and R 3 and R 4 together form a group selected from OC(R’) 2 O; or R 1 , R 4 , and R 5 are each independently selected from the group consisting of H, halogen, C 1-4 -alkyl, C 1- 3 -(per)haloalkyl, C 1-3 -alkoxy, C 1-3 -(per)haloalkoxy, and OH, and R 2 and R 3 together form a group selected from OC(R’) 2 O; R 6 and R 7 are each independently selected from the group consisting of H, methyl, and ethyl; R 8 is selected from the group consisting of H, methyl, ethyl, CH 2 Ph(o-OMe), CH 2 Ph(o-OH), C(O)CH 2 NH 2 , C(O)CH(NH 2 )(CH 2 ) 4 NH 2 , C(O)CH(NH 2 )(CH 2 ) 3 N(H)C(NH)(NH 2 ), 5- (1,2-dithiolan-3-yl)valeryl, and 2-cyano-1-phenylethyl; R 9 is selected from the group consisting of CO 2 R 10 , and COSR 10 ; R 10 is selected from the group consisting of H, methyl, and ethyl; and each R’ is independently selected from the group consisting of H, and F; or a stereoisomer or a pharmaceutically acceptable salt thereof. These compounds may have a particular strong binding affinity to one or more proteins that are associated with drug addiction or a CNS related disease or condition. Additionally, or alternatively, in embodiments R 9 is selected from the group consisting of CO 2 R 10 , COSR 10 , CH 2 OR 10 ’, C(O)CH 2 R 10 ’, CH(CF 3 )NHR 10 ’, C(O)NFR 10 ’, and C(O)N(R 10 ’) 2 ; R 10 is selected from the group consisting of H, methyl, and ethyl; and each R 10 ’ is independently selected from the group consisting of H, methyl, and ethyl; or a stereoisomer or a pharmaceutically ac- ceptable salt thereof. Additionally, or alternatively, in embodiments R 9 is selected from the group consisting of CO 2 R 10 ; and R 10 is as defined in the present disclosure; or a stereoisomer or a pharmaceutically acceptable salt thereof. These compounds may have a particular strong binding affinity to proteins that are associated with drug addiction or a CNS related disease or condition. Additionally, or alternatively, in embodiments R 1 , R 2 and R 5 are H; R 3 and R 4 are each independently selected from the group consisting of H, halogen, C1-4- alkyl; R 9 is CO 2 R 10 ; and R 10 is as defined in the present disclosure; or a stereoisomer or a pharmaceutically acceptable salt thereof. These compounds may have a particular strong binding affinity to proteins that are associated with drug addiction or a CNS related disease or condition. Additionally, or alternatively, in embodiments at least one of R 6 , R 7 , and R 8 is each independently selected from the group consisting of methyl, and ethyl, and the other ones of R 6 , R 7 , and R 8 are each independently selected from the group consisting of H, methyl, and ethyl; or a stereoisomer or a pharmaceutically acceptable salt thereof. These compounds may have a particular strong binding affinity to proteins that are associated with drug addiction or a CNS related disease or condition. Additionally, or alternatively, in embodiments R 1 , R 2 and R 5 are H; and R 3 and R 4 are both halogen, preferably F; or one of R 3 and R 4 is halogen or methyl, preferably F, and the other one of R 3 and R 4 is H provided that R 8 is selected from the group consisting of methyl, ethyl, CH 2 Ph(o-OMe), CH 2 Ph(o-OH), C(O)CH 2 NH 2 , C(O)CH(NH 2 )(CH 2 ) 4 NH 2 , C(O)CH(NH 2 )(CH 2 ) 3 N(H)C(NH)(NH 2 ), 5- (1,2-dithiolan-3-yl)valeryl, and 2-cyano-1-phenylethyl, or provided that at least one of R 6 , and R 7 is each independently selected from the group consisting of methyl, and ethyl, and the other one of R 6 , and R 7 is selected from the group consisting of H, methyl, and ethyl; or a stereoisomer or a pharmaceutically acceptable salt thereof. These compounds may have a particular strong binding affinity to proteins that are associated with drug addiction or a CNS related disease or condition. Additionally, or alternatively, in embodiments R 1 , R 2 and R 5 are H; and R 3 and R 4 are both halogen, preferably F; or one of R 3 and R 4 is halogen or methyl, preferably F, and the other one of R 3 and R 4 is H provided that R 8 is selected from the group consisting of methyl, ethyl, CH 2 Ph(o-OMe), CH 2 Ph(o-OH), C(O)CH 2 NH 2 , C(O)CH(NH 2 )(CH 2 ) 4 NH 2 , C(O)CH(NH 2 )(CH 2 ) 3 N(H)C(NH)(NH 2 ), 5- (1,2-dithiolan-3-yl)valeryl, and 2-cyano-1-phenylethyl, preferably selected from the group consisting of methyl, and ethyl; or a stereoisomer or a pharmaceutically acceptable salt thereof. These compounds may have a particular strong binding affinity to proteins that are associated with drug addiction or a CNS related disease or condition. Additionally, or alternatively, in embodiments R 1 , R 2 and R 5 are H; and R 3 and R 4 are both halogen, preferably F; or one of R 3 and R 4 is halogen or methyl, preferably F, and the other one of R 3 and R 4 is H provided that at least one of R 6 , and R 7 is each independently selected from the group consisting of methyl, and ethyl, and the other one of R 6 , and R 7 is selected from the group consisting of H, methyl, and ethyl; or a stereoisomer or a pharmaceutically acceptable salt thereof. These compounds may have a particular strong binding affinity to proteins that are associated with drug addiction or a CNS related disease or condition. Additionally, or alternatively, in embodiments R 1 , R 2 and R 5 are H; and R 8 is selected from the group consisting of methyl, ethyl, CH 2 Ph(o-OMe), CH 2 Ph(o-OH), C(O)CH 2 NH 2 , C(O)CH(NH 2 )(CH 2 ) 4 NH 2 , C(O)CH(NH 2 )(CH 2 ) 3 N(H)C(NH)(NH 2 ), 5-(1,2-dithiolan-3-yl)valeryl, and 2- cyano-1-phenylethyl; or a stereoisomer or a pharmaceutically acceptable salt thereof. These compounds may have a particular strong binding affinity to proteins that are associated with drug addiction or a CNS related disease or condition. Additionally, or alternatively, in embodiments R 1 , R 2 and R 5 are H; at least one of R 3 and R 4 is each independently selected from the group consisting of halogen, C 1-4 -alkyl, C 1-3 -(per)haloalkyl, C 1-3 -alkoxy, C 1- 3 -(per)haloalkoxy, and OH, preferably selected from the group consisting of F, Cl, methyl, and ethyl, and the other one of R 3 , and R 4 is selected from the group consisting of H, halogen, C 1-4 -alkyl, C 1-3 - (per)haloalkyl, C 1-3 -alkoxy, C 1-3 -(per)haloalkoxy, and OH, preferably selected from the group consisting of H, F, Cl, methyl, and ethyl; and R 8 is selected from the group consisting of H, methyl, ethyl, CH 2 Ph(o-OMe), CH 2 Ph(o-OH), C(O)CH 2 NH 2 , C(O)CH(NH 2 )(CH 2 ) 4 NH 2 , C(O)CH(NH 2 )(CH 2 ) 3 N(H)C(NH)(NH 2 ), 5-(1,2-dithiolan-3- yl)valeryl, and 2-cyano-1-phenylethyl, preferably selected from the group consisting of H, methyl, and ethyl; or a stereoisomer or a pharmaceutically acceptable salt thereof. These compounds may have a particular strong binding affinity to proteins that are associated with drug addiction or a CNS related disease or condition. Additionally, or alternatively, in embodiments R 8 is selected from the group consisting of H, methyl, ethyl, CH 2 Ph(o-OMe), CH 2 Ph(o-OH), C(O)CH 2 NH 2 , C(O)CH(NH 2 )(CH 2 ) 4 NH 2 , C(O)CH(NH 2 )(CH 2 ) 3 N(H)C(NH)(NH 2 ), 5- (1,2-dithiolan-3-yl)valeryl, and 2-cyano-1-phenylethyl, provided that if R 8 is H then at least one of R 6 , and R 7 is each independently selected from the group consisting of methyl, and ethyl, and the other one of R 6 , and R 7 is selected from the group consisting of methyl, and ethyl; or a stereoisomer or a pharmaceutically acceptable salt thereof. These compounds may have a particular strong binding affinity to proteins that are associated with drug addiction or a CNS related disease or condition. Additionally, or alternatively, in embodiments R 1 , R 2 and R 5 are H; and R 3 and R 4 are each independently selected from the group consisting of H, halogen, C 1-4 - alkyl, C 1-3 -(per)haloalkyl, C 1-3 -alkoxy, C 1-3 -(per)haloal- koxy, and OH, with the provision that if one of R 3 or R 4 is H then at least one of R 6 , and R 7 is each independently selected from the group consisting of methyl, and ethyl, and the other one of R 6 , and R 7 is selected from the group consisting of H, methyl, and ethyl, or R 6 , and R 7 are both H, and R 8 is selected from the group consisting of methyl, ethyl, CH 2 Ph(o-OMe), CH 2 Ph(o-OH), C(O)CH 2 NH 2 , C(O)CH(NH 2 )(CH 2 ) 4 NH 2 , C(O)CH(NH 2 )(CH 2 ) 3 N(H)C(NH)(NH 2 ), 5- (1,2-dithiolan-3-yl)valeryl, and 2-cyano-1-phenylethyl, preferably selected from the group consisting of methyl, and ethyl; or a stereoisomer or a pharmaceutically ac- ceptable salt thereof. These compounds may have a par- ticular strong binding affinity to proteins that are associated with drug addiction or a CNS related disease or condition. Additionally, or alternatively, in embodiments at least one of R 6 , R 7 , and R 8 is each independently selected from the group consisting of methyl, and ethyl, and the other ones of R 6 , R 7 , and R 8 are each independently selected from the group consisting of H, methyl, and ethyl; R 9 is CO 2 R 10 ; and R 10 is selected from the group consisting of methyl, and ethyl; or a stereoisomer or a pharmaceutically acceptable salt thereof. These compounds may have a particular strong binding affinity to one or more proteins that are associated with drug addiction or a CNS related disease or condition, such as trace amine receptor 1 (TAAR1), norepinephrine transporter (NET), 5-HT2A receptor, 5- HT2B receptor, OX1 orexin receptor, Mu-opioid receptor, Neurokinin 1 receptor (NK1R), Melanocortin 3 receptor (MC3R), dopamine transporter (DAT), phenylethanolamine N-methyltransferase (PNMT), dopamine 4 (D4) receptor, N-acetylserotonin methyltransferase, 1A Melatonin receptor, Melanocortin 1 receptor (MSHR, MC1R), Alpha- 2A adrenergic receptor, Prolactin receptor, dopamine 3 (D3) receptor, Kappa-opioid receptor, serotonin transporter (SERT), 5-HT1A receptor, 5-HT1B receptor, β1 adrenergic receptor, 5-HT2C receptor, Alpha-1B adrenergic receptor, Alpha-2B adrenergic receptor, Melanocortin 2 receptor (ACTHR, MC2R), Melanocortin 4 receptor (MC4R), 5-HT3 receptor, COMT, Alpha-1A adrenergic receptor, Nociceptin / orphanin receptor, β2 adrenergic receptor, Phenylalanine hydroxylase, Tyrosine hydroxylase, H1 histamine receptor, H3 histamine receptor, Neurokinin 2 receptor (NK2R), OX2 orexin receptor, NMDA receptor GluN1, NMDA receptor Glu2B, M2 muscarinic acetylcholine receptor, neuropeptide Y1 receptor, Delta-opioid receptor, M1 muscarinic acetylcholine receptor, M3 Muscarinic acetylcholine receptor, M4 muscarinic acetylcholine, Acetylcholinesterase, Melanocortin 5 receptor (MC5R), Neuropeptide Y2 receptor, Neurokinin 3 receptor (NK3R), Oxytocin receptor (OXYR), and Corticotropin-releasing factor receptor 1 (CRF1R). Additionally, or alternatively, in embodiments R 6 and R 7 are each independently selected from the group consisting of methyl, and ethyl, or one of R 6 and R 7 is selected from the group consisting of methyl, and ethyl, and the other one of R 6 and R 7 is H; R 8 is selected from the group consisting of H, methyl, and ethyl; R 9 is CO2R 10 ; and R 10 is selected from the group consisting of methyl, and ethyl; or a stereoisomer or a pharmaceuti- cally acceptable salt thereof. Additionally, or alternatively, in embodiments R 6 and R 7 are both H; R 9 is CO 2 R 10 ; and R 10 is selected from the group consisting of methyl, and ethyl; or a stereoisomer or a pharmaceutically acceptable salt thereof. These compounds may have a particular strong binding affinity to one or more proteins that are asso- ciated with drug addiction or a CNS related disease or condition, such as dopamine transporter (DAT), MAO-B, dopamine 2 (D2) receptor, MAO-A, Alpha-2C adrenergic receptor, α4β2 nicotinic receptor, CHK1 kinase, seroto- nin transporter (SERT), serotonin N-acetyltransferase, H1 histamine receptor, Neuropeptide Y2 receptor, and Oxytocin receptor (OXYR). Additionally, in embodiments R 3 is methyl or methoxy; and R 1 , R 2 , R 3 , R 4 , R 5 , and R 8 is H. These compounds may have a particular strong bind- ing affinity to one or more of dopamine 2 (D2) receptor, MAO-A, Alpha-2C adrenergic receptor, α4β2 nicotinic re- ceptor, CHK1 kinase, and serotonin transporter (SERT). Additionally, or alternatively, in embodiments R 1 , R 2 and R 5 are H; one of R 3 and R 4 is methoxy, and the other one of R 3 and R 4 is H; R 9 is CO 2 R 10 ; and R 10 is selected from the group consisting of methyl, and ethyl; or a stereoisomer or a pharmaceutically acceptable salt thereof. These compounds may have a particular strong binding affinity to one or more of serotonin transporter (SERT), 5-HT1B receptor, β1 adrenergic receptor, Phe- nylalanine hydroxylase, and Tyrosine hydroxylase. Addi- tionally, in embodiments at least one of R 6 and R 7 is each independently selected from the group consisting of methyl, and ethyl, preferably methyl; and the other one of R 6 and R 7 is H, methyl, or ethyl, preferably H, or methyl; R 8 is selected from the group consisting of methyl, and ethyl, preferably ethyl; R 9 is CO 2 R 10 ; and R 10 is selected from the group consisting of methyl, and ethyl, preferably methyl. These compounds may have a particular strong binding affinity to one or more of 5- HT1B receptor, β1 adrenergic receptor, Phenylalanine hydroxylase, and Tyrosine hydroxylase. Additionally, or alternatively, in embodiments R 1 and R 2 are H; at least one of R 3 , R 4 , and R 5 is F, and the other ones of R 3 , R 4 , and R 5 are H; R 6 and R 7 are each independently selected from the group consisting of H, methyl, and ethyl; R 9 is CO 2 R 10 ; and R 10 is selected from the group consisting of methyl, and ethyl; or a stere- oisomer or a pharmaceutically acceptable salt thereof. In embodiments, R 1 and R 2 are H; one of R 3 , R 4 , and R 5 is F, and the other ones of R 3 , R 4 , and R 5 are H; R 6 and R 7 are each independently selected from the group consist- ing of H, methyl, and ethyl; R 9 is CO 2 R 10 ; and R 10 is selected from the group consisting of methyl, and ethyl. Additionally, or alternatively, in embodiments R 1 , R 2 and R 5 are H; one of R 3 and R 4 is selected from the group consisting of F, Cl, methyl, and ethyl, and the other one of R 3 and R 4 is selected from the group consisting of H, F, methyl, and ethyl; R 9 is CO 2 R 10 ; and R 10 is selected from the group consisting of methyl, and ethyl; or a stereoisomer or a pharmaceutically acceptable salt thereof. Additionally, or alternatively, in embodiments R 1 , R 2 and R 5 are H; R 3 and R 4 are both F, or one of R 3 and R 4 is F and the other one of R 3 and R 4 is H; R 6 and R 7 are each independently selected from the group consisting of H, and methyl; R 9 is CO 2 R 10 ; and R 10 is selected from the group consisting of methyl, and ethyl; or a stereoisomer or a pharmaceutically acceptable salt thereof. These compounds may have a particular strong binding affinity to one or more proteins that are associated with drug addiction or a CNS related disease or condition, such as trace amine receptor 1 (TAAR1), norepinephrine transporter (NET), 5-HT2A receptor, 5- HT2B receptor, OX1 orexin receptor, Mu-opioid receptor, Neurokinin 1 receptor (NK1R), Melanocortin 3 receptor (MC3R), dopamine transporter (DAT), MAO-B, phenylethanolamine N-methyltransferase (PNMT), N- acetylserotonin methyltransferase, 1A Melatonin receptor, Melanocortin 1 receptor (MSHR, MC1R), Alpha- 2A adrenergic receptor, and Prolactin receptor, dopamine 3 (D3) receptor, Kappa-opioid receptor, 5-HT3 receptor, OX2 orexin receptor, NMDA receptor GluN1, NMDA receptor Glu2B, M2 muscarinic acetylcholine receptor, neuropeptide Y1 receptor, Delta-opioid receptor, M1 muscarinic acetylcholine receptor, M3 Muscarinic acetylcholine receptor, M4 muscarinic acetylcholine receptor, and Corticotropin-releasing factor receptor 1 (CRF1R). Additionally, or alternatively, in embodiments R 1 , R 2 and R 5 are H; R 3 and R 4 are both F; R 6 and R 7 are each independently selected from the group consisting of H, and methyl; R 9 is CO 2 R 10 ; and R 10 is selected from the group consisting of methyl, and ethyl; or a stere- oisomer or a pharmaceutically acceptable salt thereof. These compounds may have a particular strong binding affinity to one or more proteins that are associated with drug addiction or a CNS related disease or condi- tion, such as trace amine receptor 1 (TAAR1), norepi- nephrine transporter (NET), 5-HT2A receptor, 5-HT2B re- ceptor, OX1 orexin receptor, Mu-opioid receptor, Neuro- kinin 1 receptor (NK1R), Melanocortin 3 receptor (MC3R), dopamine transporter (DAT), MAO-B, N-acetylserotonin methyltransferase, 1A Melatonin receptor, Melanocortin 1 receptor (MSHR, MC1R), Alpha-2A adrenergic receptor, Prolactin receptor, serotonin transporter (SERT), 5- HT1A receptor, COMT, NMDA receptor GluN1, Delta-opioid receptor, M1 muscarinic acetylcholine receptor, M3 Mus- carinic acetylcholine receptor, M4 muscarinic acetyl- choline, Acetylcholinesterase, and Melanocortin 5 re- ceptor (MC5R). Additionally, or alternatively, in embodiments R 1 , R 2 and R 5 are H; one of R 3 and R 4 is selected from the group consisting of methyl, and ethyl, and the other one of R 3 and R 4 is H; R 9 is CO 2 R 10 ; and R 10 is selected from the group consisting of methyl, and ethyl; or a stereoisomer or a pharmaceutically acceptable salt thereof. Additionally, or alternatively, in embodiments R 1 , R 2 and R 5 are H; one of R 3 and R 4 is methyl and the other one of R 3 and R 4 is H; R 9 is CO 2 R 10 ; and R 10 is selected from the group consisting of methyl, and ethyl; or a stereoisomer or a pharmaceutically acceptable salt thereof. These compounds may have a particular strong binding affinity to one or more proteins that are asso- ciated with drug addiction or a CNS related disease or condition, such as dopamine 2 (D2) receptor, MAO-A, Al- pha-2C adrenergic receptor, α4β2 nicotinic receptor, CHK1 kinase, dopamine 4 (D4) receptor, dopamine 1 (D1) receptor, Sigma-1 receptor, 1B Melatonin receptor, ser- otonin N-acetyltransferase, 5-HT1B receptor, β1 adren- ergic receptor, 5-HT2C receptor, Alpha-1B adrenergic receptor, Alpha-2B adrenergic receptor, Melanocortin 2 receptor (ACTHR, MC2R), Melanocortin 4 receptor (MC4R), COMT, Alpha-1A adrenergic receptor, Nociceptin / or- phanin receptor, β2 adrenergic receptor, Tyrosine hy- droxylase, H1 histamine receptor, NMDA receptor GluN1, Acetylcholinesterase, Melanocortin 5 receptor (MC5R), Neuropeptide Y2 receptor, Neuropeptide Y5 receptor, Neu- rokinin 3 receptor (NK3R), and Corticotropin-releasing factor receptor 1 (CRF1R). Additionally, or alternatively, in embodiments R 1 , R 2 and R 5 are H; R 3 and R 4 are both F; R 8 is selected from the group consisting of methyl, and ethyl; R 9 is CO 2 R 10 ; and R 10 is selected from the group consisting of methyl, and ethyl; or a stereoisomer or a pharmaceutically acceptable salt thereof. These compounds may have a particular strong binding affinity to one or more proteins that are associated with drug addiction or a CNS related disease or condition, such as trace amine receptor 1 (TAAR1), norepinephrine transporter (NET), 5-HT2A receptor, 5-HT2B receptor, OX1 orexin receptor, Mu-opioid receptor, Neurokinin 1 receptor (NK1R), Melanocortin 3 receptor (MC3R), dopamine transporter (DAT), N-acetylserotonin methyltransferase, 1A Melatonin receptor, Melanocortin 1 receptor (MSHR, MC1R), Alpha-2A adrenergic receptor, Prolactin receptor, OX2 orexin receptor, Delta-opioid receptor, M1 muscarinic acetylcholine receptor, M3 Muscarinic acetylcholine receptor, and M4 muscarinic acetylcholine receptor. Additionally, or alternatively, in embodiments the compound is selected from the group consisting of: ethyl 3-(3,4-difluorophenyl)-1,2,4-trime- thylazetidine-3-carboxylate (1); ethyl 3-(3,4-difluorophenyl)-1-ethylazet- idine-3-carboxylate (2); ethyl 3-(3,4-difluorophenyl)-1-methylazet- idine-3-carboxylate (3); ethyl 3-(3,4-difluorophenyl)azetidine-3-car- boxylate (4); ethyl 3-(3-fluorophenyl)-2,4-dimethylazet- idine-3-carboxylate (5); ethyl 3-(3-fluorophenyl)-2-methylazetidine-3- carboxylate (6); ethyl 3-(4-methylphenyl)azetidine-3-carbox- ylate (7); methyl 2-methyl-3-(3-methylphenyl)azetidine- 3-carboxylate (8); methyl 3-(3,4-difluorophenyl)-1-ethyl-2- methylazetidine-3-carboxylate (9); methyl 3-(3,4-difluorophenyl)-2-methylazet- idine-3-carboxylate (10); ethyl 2-methyl-3-(3-methylphenyl)azetidine-3- carboxylate (11); ethyl 1-ethyl-3-(4-methylphenyl)azetidine-3- carboxylate (12); ethyl 3-(3,4-difluorophenyl)-2,4-dime- thylazetidine-3-carboxylate (13); methyl 3-(3,4-difluorophenyl)-1-ethyl-2,4-di- methylazetidine-3-carboxylate (14); methyl 1-ethyl-2-methyl-3-(3- methylphenyl)azetidine-3-carboxylate (15); ethyl 3-(3-methylphenyl)azetidine-3-carbox- ylate (16); ethyl 3-(3-fluorophenyl)-1,2,4-trimethylazet- idine-3-carboxylate (17); ethyl 1-ethyl-2,4-dimethyl-3-(3- methylphenyl)azetidine-3-carboxylate (18); ethyl 3-(3,4-difluorophenyl)-2-methylazet- idine-3-carboxylate (19); ethyl 3-(4-methoxyphenyl)azetidine-3-carbox- ylate (20); ethyl 1-ethyl-3-(3-methylphenyl)azetidine-3- carboxylate (21); methyl 1-ethyl-3-(3-methoxyphenyl)-2,4-dime- thylazetidine-3-carboxylate (22); ethyl 1-ethyl-2,4-dimethyl-3-(4- methylphenyl)azetidine-3-carboxylate (23); ethyl 2,4-dimethyl-3-(3-methylphenyl)azet- idine-3-carboxylate (24); methyl 3-(3,4-difluorophenyl)-2,4-dime- thylazetidine-3-carboxylate (25); ethyl 1,2-dimethyl-3-(3-methylphenyl)azet- idine-3-carboxylate (26); methyl 1-ethyl-2,4-dimethyl-3-(3- methylphenyl)azetidine-3-carboxylate (27); methyl 1-ethyl-2-methyl-3-(4- methylphenyl)azetidine-3-carboxylate (28); methyl 1-ethyl-3-(4-methoxyphenyl)-2,4-dime- thylazetidine-3-carboxylate (29); methyl 1-ethyl-3-(4-methoxyphenyl)-2- methylazetidine-3-carboxylate (30); methyl 2,4-dimethyl-3-(4-methylphenyl)azet- idine-3-carboxylate (31); ethyl 3-(3,4-difluorophenyl)-1-ethyl-2- methylazetidine-3-carboxylate (32); methyl 2-methyl-3-(4-methylphenyl)azetidine- 3-carboxylate (33); methyl 3-(4-methylphenyl)-1,2-dimethylazet- idine-3-carboxylate (34); ethyl 3-(3-fluorophenyl)azetidine-3-carbox- ylate (35); ethyl 1,2-dimethyl-3-(4-methylphenyl)azet- idine-3-carboxylate (36); and methyl 1-ethyl-3-(3-fluorophenyl)-2,4-dime- thylazetidine-3-carboxylate (37); or a stereoisomer or a pharmaceutically ac- ceptable salt thereof. Additionally, or alternatively, the compound of formula (I) is dextrorotatory. In one aspect is disclosed a pharmaceutical composition comprising an effective amount of one or more compounds, or a stereoisomer or a pharmaceutically acceptable salt thereof, as defined in the present disclosure, together with one or more pharmaceutically acceptable excipi- ent(s). It is to be understood that the one or more compounds, or a stereoisomer or a pharmaceutically ac- ceptable salt thereof, as defined in the present dis- closure refers to compounds of formula (I), or a stere- oisomer or a pharmaceutically acceptable salt thereof, as defined in the present disclosure. Additionally, or alternatively, in embodiments a pharmaceutical composition as defined in the present disclosure, in combination with one or more pharmaceu- tically acceptable carrier(s). Therefore, in embodi- ments, pharmaceutical compositions comprise an effec- tive amount of one or more compounds of formula (I), or a stereoisomer or a pharmaceutically acceptable salt thereof, together with one or more pharmaceutically ac- ceptable excipient(s) and/or one or more pharmaceuti- cally acceptable carrier(s), or any combination thereof. Preferably, pharmaceutical compositions comprise one compound of formula (I), or a stereoisomer or a pharma- ceutically acceptable salt thereof, preferably a phar- maceutically acceptable salt thereof, in combination with one or more pharmaceutically acceptable excipi- ent(s) and one pharmaceutically acceptable carrier. Additionally, or alternatively, in embodiments the pharmaceutical composition as defined in the present disclosure, in combination with one or more other active ingredient(s). Therefore, additionally, or alterna- tively, to pharmaceutically acceptable excipient(s) and/or pharmaceutically acceptable carrier(s), pharma- ceutical compositions of the present disclosure may com- prise an effective amount of one or more compounds of formula (I), or a stereoisomer or a pharmaceutically acceptable salt thereof, in combination with one or more other active ingredient(s). Therefore, in embodiments, pharmaceutical compositions comprise an effective amount of one or more compounds of formula (I), or a stereoisomer or a pharmaceutically acceptable salt thereof, together with one or more pharmaceutically ac- ceptable excipient(s) and/or one or more pharmaceuti- cally acceptable carrier(s) and/or one or more other active ingredient(s), or any combination thereof. Additionally, or alternatively, the pharmaceutical compositions consist an effective amount of one or more compounds of formula (I), or a stereoisomer or a pharmaceutically acceptable salt thereof, preferably a pharmaceutically acceptable salt thereof, together with one or more pharmaceutically acceptable excipient(s), preferably 1, 2, or 3 pharmaceutically acceptable excipient(s), more preferably 1 pharmaceutically acceptable excipient. Additionally, or alternatively, the pharmaceutical compositions consist an effective amount of one or more compounds of formula (I), preferably an effective amount of 1 or 2 compounds of formula (I), more preferably 1 compound of formula (I), or a stereoisomer or a pharmaceutically acceptable salt thereof, preferably a pharmaceutically acceptable salt thereof, together with one or more pharmaceutically acceptable excipient(s), preferably 1, 2, or 3 pharmaceutically acceptable excipient(s), and/or one or more pharmaceutically acceptable carrier(s), preferably 1, 2, or 3 pharmaceutically acceptable carrier(s), more preferably 1 pharmaceutically acceptable carrier. Additionally, or alternatively, the pharmaceutical compositions consist an effective amount of one or more compounds of formula (I), preferably an effective amount of 1 or 2 compounds of formula (I), more preferably 1 compound of formula (I), or a stereoisomer or a pharmaceutically acceptable salt thereof, preferably a pharmaceutically acceptable salt thereof, together with one or more pharmaceutically acceptable excipient(s), preferably 1, 2, or 3 pharmaceutically acceptable excipient(s), and/or one or more pharmaceutically acceptable carrier(s), preferably 1, 2, or 3 pharmaceutically acceptable carrier(s), more preferably 1 pharmaceutically acceptable carrier, and/or one or more other active ingredient(s), preferably one other active ingredient. In one aspect is disclosed a compound, or a stereoisomer or pharmaceutically acceptable salt thereof, as defined in the present disclosure, for use as a medicament. In one aspect is disclosed a compound, or a stereoisomer or pharmaceutically acceptable salt thereof, as defined in the present disclosure, for use in the treatment or prevention of drug addiction or a CNS related disease or condition. Additionally, or alternatively, in embodiments the drug addiction or the CNS related disease or condi- tion is selected from the group consisting of stimulant addiction, ADHD, ADD, sluggish cognitive tempo, concen- tration deficit disorder, motivational or reward system dysfunction, autism spectrum disorder, disruptive, im- pulse control, and conduct disorders, anxiety disorders, eating disorders, depression, dysthymia, Alzheimer’s disease, Parkinson’s disease, hyperactivity, narco- lepsy, and alcoholism. Additionally, or alternatively, the stimulant is selected from the group consisting of cocaine, amphetamine, and methylphenidate. Additionally, or alternatively, the attention deficit hyperactivity disorder (ADHD) is selected from the group consisting of attention deficit hyperactivity disorder predominantly inattentive (ADHD-PI or ADHD-I), attention deficit hyperactivity disorder predominantly hyperactive-impulsive (ADHD-PH or ADHD-HI), and attention deficit hyperactivity disorder combined type (ADHD-C). Additionally, or alternatively, the motivational or reward system dysfunction is selected from the group consisting of anhedonia, apathy, akinetic mutism, abulia, avolition, psychomotor retardation, slowing, and anergia. Additionally, or alternatively, the disruptive, impulse control, and conduct disorders is selected from the group consisting of oppositional defiant disorder, and obsessive-compulsive disorder. Additionally, or alternatively, the anxiety disorders are selected from the group consisting of generalized anxiety disorder (GAD), phobias, panic disorder, agoraphobia, social anxiety disorder, post- traumatic stress disorder, separation anxiety disorder, obsessive–compulsive disorder, and selective mutism. Additionally, or alternatively, the eating disorders are selected from the group consisting of obesity, binge-eating disorder, bulimia nervosa, other specified feeding or eating disorder (OSFED), anorexia nervosa, bulimia nervosa, and binge-purge subtype of anorexia nervosa. Further, a compound, or a stereoisomer or pharmaceutically acceptable salt thereof, as disclosed herein and herafter may also be beneficial for use in the treatment or prevention of a disease or condition that is associated with HIV-1 reverse transcriptase, oxidosqualene cyclase, aldose reductase, or hepatitis C virus NS5B RNA-dependent RNA polymerase. Additionally, or alternatively, the disease or condition that is associated with HIV-1 reverse transcriptase, oxidosqualene cyclase, aldose reductase, or hepatitis C virus NS5B RNA-dependent RNA polymerase is selected from the group consisting of acquired immunodeficiency syndrome (AIDS), hypercholesterolemia, Chagas' disease, diabetes mellitus, hepatitis C. In one aspect is provided use of a compound of formula (I), or a stereoisomer or pharmaceutically acceptable salt thereof, as a nootropic compound. Substitution, especially fluorination, at the position of metabolic attack – mainly at positions corresponding to substituents R 2 , R 3 , and R 4 of compounds having formula (I) may be used to alter the route and rate of metabolic degradation. Furthermore, substitution, especially alkylation and/or halogenation, in particular methyl and/or F, mainly at positions corresponding to substituents R 2 , R 3 , and/or R 4 may alter the binding affinity to a protein and/or lipophilicity, conformation, electrostatic potential, dipoles, and pKa of the compound of formula (I). Substitution, in particular fluorination and/or alkylation, may also alter the tissue distribution, pharmacodynamics, and toxicology of the compound. It can be generalized that replacing hydrogen with fluorine at positions corresponding to substituents R 2 , R 3 , and/or R 4 may cause minimal steric effects at the receptor. The compounds as disclosed herein and hereafter may be actively transported over the blood brain barrier by organic cation transporters and may be concentrated in the brain. Compounds as disclosed herein and hereafter may not be oxidized to form epoxides thereby making them less prone to cause oxidative stress. The compounds as disclosed herein and hereafter may be associated with neuroprotective and neuroregenerative properties instead of neurotoxicity or neuro- degeneration. The compounds as disclosed herein and hereafter may be used, in addition to in treatment or prevention of e.g. Alzheimer’s disease and Parkinson’s disease, in treatment or prevention of addictions in general. Number of positive pharmacological responses can be achieved simultaneously. While decreasing the tendency to relapse and likelihood of developing an addiction, these compounds may act as general mood stabilizers and general neuroprotectants possessing remarkable antiparkinsonian and antiepileptic character. Without being limited to any particular theory or mechanism, it is believed that the mechanism of action of the disclosed compounds involve proteins in the mesocorticolimbic dopamine pathway and/or the locus coeruleus-noradrenergic system. The invention is based on the realization that the compounds of the invention bind to and/or affect the activity of proteins that are associated with drug addiction or a CNS related disease or condition. Also disclosed is a method of treating or preventing a disease or condition selected from the group consisting of drug addiction or a CNS related disease or condition is selected from the group consisting of stimulant addiction, ADHD, ADD, sluggish cognitive tempo, concentration deficit disorder, motivational or reward system dysfunction, autism spectrum disorder, disruptive, impulse control, and conduct disorders, anxiety disorders, eating disorders, depression, dysthymia, Alzheimer’s disease, Parkinson’s disease, hyperactivity, narcolepsy, and alcoholism in a patient in need thereof, comprising administering a compound as disclosed herein and hereafter, or a stereoisomer or a pharmaceutically acceptable salt thereof, to the patient. Additionally, or alternatively, the compound, or the stereoisomer or pharmaceutically acceptable salt thereof, as disclosed herein and hereafter for use in treatment or prevention of drug addiction or a CNS related disease or condition requiring the inhibition or activation of at least one protein selected from the group consisting of trace amine receptors (trace amine- associated receptors (TAARs), such as TAAR1), dopamine- and serotonin receptors, dopamine- and serotonin transporters, acyl and methyl transferases, norpinephrine transporter, monoamino-oxidases such as MAO-A, catecholine-O-methylransferase (COMT), adrenergic receptors, tyrosine hydroxylase, histamine receptors such as H1 histamine receptor, orexin receptors, NMDA-receptors, sigma-1 receptor, muscarinic and nicotinic acetylcholine receptors such as M1, M3, and M4 muscarinic acetylcholine receptors, opioid receptors such as μ-opioid receptors (MOR), neuropeptide receptors such as Neuropeptide Y2 receptor, melanocortin receptors (excluding MC3R), neurokinin receptors such as NKR2 and NK3R, corticotropin-releasing factor receptor 1, acetylcholinesterase, 1B melatonin receptor, cholinergic receptors, and dopamine beta- hydroxylase (dopamine beta-monooxygenase, DBH). In one aspect is disclosed a method for the preparation of a compound of formula (I), or pharmaceutically ac- ceptable salt or a stereoisomer thereof, as defined in the present disclosure, comprising: (i) providing a compound of formula (II) , wherein R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , and R 7 are as defined in the present disclosure; and R 8 is methyl, ethyl, CH 2 Ph(o- OMe), or a protecting group; (ii) reacting the compound of formula (II) with one or more compounds each independently selected from the group consisting of acids, bases, and water; or reacting the compound of formula (II) with methanol or ethanol, and a Lewis catalyst; (iii) optionally performing an esterifica- tion with a compound of formula (III) R 11 -R’’ (III), wherein R 11 is methyl or ethyl; and R’’ is selected from the group consisting of halogen, SR’’’, OR’’’, and a first activating group, wherein R’’’ is H, or a second activating group, optionally in the presence of one or more activating group reactant(s) and/or one or more activating agent(s); (iv) optionally performing one or more first deprotection reaction(s); (v) optionally performing a transesteri- fication with a compound of formula (III), wherein R 11 is methyl or ethyl; and R’’ is selected from the group consisting of SH, and OH, optionally in the presence of one or more activating group reactant(s) and/or one or more activating agent(s); (vi) optionally performing an amidation with a compound of formula (IV) R 12 -R 8’ (IV), wherein R 12 is HO, R 13 O, or R 13 , wherein R 13 is an activating group; and R 8’ is C(O)CH 2 NHR 14 , C(O)CH(NHR 14 )(CH 2 ) 4 NHR 14 , C(O)CH(NHR 14 )(CH 2 ) 3 N(R 14 )C(NR 14 )(NHR 14 ), or 5-(1,2-dithio- lan-3-yl)valeryl, wherein each R 14 is independently se- lected from the group consisting of H, and protecting groups, optionally in the presence of one or more acti- vating group reactant(s) and/or one or more activating agent(s); (vii) optionally performing a reductive al- kylation with a compound of formula (V) R 8’’ -CHO (V), wherein R 8’’ is H, or CH 3 ; and a reducing agent; (viii) optionally performing an N-alkylation with a compound of formula (VI) R 8’’’ -R 15 (VI), wherein R 8’’’ is methyl, ethyl, (o-OMe)PhCH 2 , or (Ph)CH(CH 2 CN); and R 15 is selected from the group consisting of halogen, SR 16 , and OR 16 , wherein R 16 is an activating group, op- tionally in the presence of one or more activating group reactant(s) and/or one or more activating agent(s); (ix) optionally performing one or more second deprotection reaction(s); to obtain the compound of formula (I), wherein R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 and R 10 are as defined in the present disclosure; and (x) optionally converting the compound of formula (I) to a pharmaceutically acceptable salt thereof. Additionally, or alternatively, in (i), R 8 is methyl, ethyl, CH 2 Ph(o-OMe), or a protecting group, preferably the protecting group is benzyl. Additionally, or alternatively, in (ii), reacting the compound of formula (II) with one or more compounds each independently selected from the group consisting of acids, bases, and water, preferably the one or more compounds are HCl and water, preferably the one or more compounds are two compounds, wherein the two compounds are HCl and water. A compound of formula (I), wherein R 9 is CO 2 H; R 8 is as defined for the compound of formula (II) as defined in the present disclosure; and R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , and R 7 are as defined in the present disclosure, is formed. Additionally, or alternatively, in ii), reacting the compound of formula (II) with methanol or ethanol, and a Lewis catalyst, preferably the Lewis catalyst is trimethylsilyl triflate. A compound of formula (I), wherein R 9 is CO 2 R 10 , R 10 is methyl or ethyl, R 8 is as defined for the compound of formula (II) as defined in the present disclosure; and R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , and R 7 are as defined in the present disclosure, is formed. Additionally, or alternatively, the method comprises (iii) performing an esterification with a com- pound of formula (III), R 11 -R’’ (III), wherein R 11 is methyl or ethyl; and R’’ is selected from the group consisting of halogen, SR’’’, OR’’’, and a first activating group, wherein R’’’ is H, or a second activating group, optionally in the presence of one or more activating group reactant(s) and/or one or more activating agent(s), preferably the halogen is Cl, Br, or I; the first activating group is selected from the group consisting of a sulfonate, preferably phenylsulfonate, tosylate, mesylate, or triflate; a halogen, preferably Cl, Br, or I; an amino group, preferably 1-imidazolyl; an alkoxy, preferably 1H-1,2,3-benzotriazol-1-olate; R’’’ is H, or a second activating group, preferably the second activating group is selected from the group consisting of a sulfonyl, preferably tosyl (Ts), mesyl, or trifyl; and an amino group, preferably benzotriazolyl, preferably in the presence of one activating group reactant and/or one or more activating agent(s), wherein the one activating group reactant is 1,1’-carbonyldiimidazole (CDI), diethyl azodicarboxylate (DEAD), diisopropyl azodicarboxylate (DIAD), N,N′-dicyclohexylcarbodiimide (DCC), triphenylphosphine (PPh 3 ), a sulfinic acid, preferably phenylsulfinic acid or p-toluenesulfinic acid; a mesyl halide, preferably methanesulfonyl chloride; trifyl azide, trifluoromethanesulfonyl chloride, a metal halide, preferably NaI; trifluoroacetyl chloride, and trifluoroacetic anhydride; and the one or more activating agent(s) is one activating agent, preferably selected from an acid, preferably HCl, H 2 SO 4 or AcOH; a base, preferably NaOH or NaH; water, 4- dimethylaminopyridine (DMAP), triphenylphosphine (PPh 3 ), a metal halide, preferably NaI; a Lewis acid, preferably TiCl 4 , boron trifluoride, and boron trifluoride diethyl etherate. More preferably the compound of formula (III) is methanol or ethanol, in the presence of HCl, or H 2 SO 4 . A compound of formula (I), wherein R 9 is CO 2 R 10 or COSR 10 , preferably CO 2 R 10 ; R 10 is methyl or ethyl; R 8 is as defined for the compound of formula (II) as defined in the present disclosure; and R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , and R 7 are as defined in the present disclosure, is formed from a compound of formula (I), wherein R 9 is CO 2 H; R 8 is as defined for the compound of formula (II) as defined in the present disclosure; and R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , and R 7 are as defined in the present disclosure. Additionally, or alternatively, the method comprises (iv) performing one or more first deprotection reaction(s), preferably one first deprotection reaction, more preferably wherein the one or more first deprotection reaction(s) are performed with a reactant or reagent selected from tetra-n-butylammonium fluoride, BBr 3, H 2 , preferably H 2 in the presence of Pd/C, and 2,3-dichloro-5,6-dicyano-p-benzoquinone. One or more protecting group(s) may be removed from a compound of formula (I), wherein R 9 is CO2R 10 or COSR 10 , preferably CO 2 R 10 , R 10 is H, provided that R 9 is CO 2 R 10 ; methyl or ethyl; R 8 is CH 2 Ph(o-OMe), or a protecting group; and R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , and R 7 and are as defined in the present disclosure. It is to be understood that the one or more protecting group(s) that may be removed may be the R 8 group (i.e., the R 8 group being a protecting group) or one or more protecting group(s) of the R 8 group (e.g., one or more of a methyl, TMS, or benzyl), preferably the one or more protecting group(s) is one protecting group, wherein the one protecting group is benzyl or methyl, more preferably the one protecting group is the methyl of CH 2 Ph(o-OMe) of R 8 or the one protecting group is R 8 , wherein R 8 is benzyl, even more preferably the one protecting group is R 8 , wherein R 8 is benzyl. A compound of formula (I), wherein R 9 is CO 2 R 10 or COSR 10 ; R 10 is H, provided that R 9 is CO 2 R 10 ; methyl or ethyl; R 8 is H, methyl, ethyl, or CH 2 Ph(o-OH); and R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , and R 7 are as defined in the present disclosure, is formed. Additionally, or alternatively, the method comprises (v) performing a transesterification with a compound of formula (III), wherein R 11 is methyl or ethyl; and R’’ is selected from the group consisting of SH, and OH, optionally in the presence of one or more activating group reactant(s) and/or one or more activating agent(s), preferably in the presence of one activating agent, more preferably in the presence of one activating agent selected from the group consisting of an acid, preferably HCl, H 2 SO 4 or AcOH, even more preferably the compound of formula (III) is methanol or ethanol, in the presence of HCl, or H 2 SO 4 . A compound of formula (I), wherein R 9 is CO 2 R 10 or COSR 10 , preferably R 9 is CO 2 R 10 ; R 10 is methyl or ethyl; R 8 is as defined for the compound of formula (II) as defined in the present disclosure; and R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , and R 7 are as defined in the present disclosure, is formed from a compound of formula (I), wherein R 9 is CO2R 10 or COSR 10 , preferably R 9 is CO 2 R 10 ; R 10 is methyl or ethyl; R 8 is as defined for the compound of formula (II) as defined in the present disclosure; and R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , and R 7 are as defined in the present disclosure. Additionally, or alternatively, the method comprises (vi) performing an amidation with a compound of formula (IV) R 12 -R 8’ (IV), wherein R 12 is HO, R 13 O, or R 13 , wherein R 13 is an activating group; and R 8’ is C(O)CH 2 NHR 14 , C(O)CH(NHR 14 )(CH 2 ) 4 NHR 14 , C(O)CH(NHR 14 )(CH 2 ) 3 N(R 14 )C(NR 14 )(NHR 14 ), or 5-(1,2-dithio- lan-3-yl)valeryl, wherein each R 14 is independently se- lected from the group consisting of H, and protecting groups, optionally in the presence of one or more acti- vating group reactant(s) and/or one or more activating agent(s), preferably the activating group is selected from Cl, Br, and I; a (substituted) amino group, pref- erably 1-imidazolyl; an alkoxy, preferably 1H-1,2,3- benzotriazol-1-olate; each R 14 is independently a pro- tecting group each independently selected from Fmoc and t-Boc; and in the presence of one activating group re- actant and/or one activating agent, more preferably in the presence of one activating group reactant and one activating agent, even more preferably the activating group reactant(s) is selected from 1,1’-carbonyldiimid- azole (CDI), diethyl azodicarboxylate (DEAD), diisopro- pyl azodicarboxylate (DIAD), N,N′-dicyclohexylcar- bodiimide (DCC); and the activating agent(s) is selected from 1-hydroxy-7-azabenzotriazole (HOAt), hydroxyben- zotriazole (HOBt), and 4-dimethylaminopyridine (DMAP). A compound of formula (I), wherein R 9 is CO 2 R 10 or COSR 10 , preferably R 9 is CO 2 R 10 ; R 10 is methyl or ethyl; R 8 is C(O)CH 2 NHR 14 , C(O)CH(NHR 14 )(CH 2 ) 4 NHR 14 , C(O)CH(NHR 14 )(CH 2 ) 3 N(R 14 )C(NR 14 )(NHR 14 ), or 5-(1,2-dithio- lan-3-yl)valeryl; and R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , and R 7 are as defined in the present disclosure, is formed from a compound of formula (I), wherein R 9 is CO 2 R 10 or COSR 10 , preferably R 9 is CO 2 R 10 ; R 10 is methyl or ethyl; R 8 is H; and R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , and R 7 are as defined in the present disclosure. It is to be understood that in a method for the preparation of a compound of formula (I), wherein R 9 is CO 2 H or COSH , preferably R 9 is CO 2 H; R 8 is C(O)CH 2 NHR 14 , C(O)CH(NHR 14 )(CH 2 ) 4 NHR 14 , C(O)CH(NHR 14 )(CH 2 ) 3 N(R 14 )C(NR 14 )(NHR 14 ), or 5-(1,2-dithio- lan-3-yl)valeryl; and R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , and R 7 are as defined in the present disclosure, the ester group of R 9 (CO 2 R 10 or COSR 10 , preferably CO 2 R 10 ; R 10 is methyl or ethyl) may be hydrolysed with a method known to those skilled in the art (e.g., using KOH or LiOH). Additionally, or alternatively, the method com- prises (vii) performing a reductive alkylation with a compound of formula (V) R 8’’ -CHO (V), wherein R 8’’ is H, or CH 3 ; and a reducing agent. Preferably the reducing agent is sodium borohydride. In embodiments the compound of formula (V) is first reacted with a compound of formula (I), wherein R 9 is CO 2 R 10 or COSR 10 , preferably R 9 is CO 2 R 10 ; R 10 is methyl or ethyl; R 8 is H; and R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , and R 7 are as defined in the present disclosure, to form an intermediate compound that is reduced with the reducing agent, preferably sodium bo- rohydride. A compound of formula (I), wherein R 9 is CO 2 R10 or COSR 10 , preferably R 9 is CO 2 R 10 ; R 10 is methyl or ethyl; R 8 is methyl or ethyl; and R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , and R 7 are as defined in the present disclosure, is formed from a compound of formula (I), wherein R 9 is CO 2 R 10 or COSR 10 , preferably R 9 is CO 2 R 10 ; R 10 is methyl or ethyl; R 8 is H; and R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , and R 7 are as defined in the present disclosure. It is understood that the formed compound may be further hydrolysed with a method known to those skilled in the art (e.g., using KOH or LiOH) to form a compound of formula (I), wherein R 9 is CO 2 H or COSH, preferably R 9 is CO 2 H. Additionally, or alternatively, the method com- prises (viii) performing an N-alkylation with a compound of formula (VI) R 8’’’ -R 15 (VI), wherein R 8’’’ is methyl, ethyl, (o-OMe)PhCH 2 , or (Ph)CH(CH 2 CN); and R 15 is selected from the group consisting of halogen, SR 16 , and OR 16 , wherein R 16 is an activating group, op- tionally in the presence of one or more activating group reactant(s) and/or one or more activating agent(s). In embodiments R 15 is Cl, Br, I, SR 16 , or OR 16 , wherein R 16 is an activating group; preferably the activating group is selected from a sulfonyl, preferably tosyl (Ts), me- syl, or trifyl. In embodiments (viii) comprises one or more activating group reactant(s) and/or one or more activating agent(s), preferably the one or more acti- vating group reactant(s) and/or one or more activating agent(s)is one activating agent selected from a metal halide, preferably NaI. A compound of formula (I), wherein R 9 is CO 2 R 10 or COSR 10 , preferably R 9 is CO 2 R 10 ; R 10 is methyl or ethyl; R 8 is methyl, ethyl, (o-OMe)PhCH 2 , or (Ph)CH(CH 2 CN); and R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , and R 7 are as defined in the present disclosure, is formed from a compound of formula (I), wherein R 9 is CO 2 R 10 or COSR 10 , preferably R 9 is CO 2 R 10 ; R 10 is methyl or ethyl; R 8 is H; and R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , and R 7 are as defined in the present disclosure. It is understood that the formed compound may be further hydrolysed with a method known to those skilled in the art (e.g., using KOH or LiOH) to form a compound of formula (I), wherein R 9 is CO 2 H or COSH, preferably R 9 is CO 2 H. Additionally, or alternatively, the method com- prises (iX) performing one or more second deprotection reaction(s), preferably one second deprotection reac- tion, more preferably wherein the one or more second deprotection reaction(s) are performed with a reactant or reagent selected from bases, preferably NaOH or KOH; tetra-n-butylammonium fluoride; acids, preferably tri- fluoroacetic acid or HCl; BBr 3, H 2 , preferably H 2 in the presence of Pd/C, and 2,3-dichloro-5,6-dicyano-p-benzo- quinone. One or more protecting group(s) may be removed from a compound of formula (I), wherein R 9 is CO 2 R 10 or COSR 10 , preferably CO 2 R 10 , R 10 is H, provided that R 9 is CO 2 R 10 ; methyl or ethyl; R 8 is C(O)CH 2 NHR 14 , C(O)CH(NHR 14 )(CH 2 ) 4 NHR 14 , C(O)CH(NHR 14 )(CH 2 ) 3 N(R 14 )C(NR 14 )(NHR 14 ), CH 2 Ph(o-OMe), or a protecting group, wherein each R 14 is independently se- lected from the group consisting of H, and protecting groups; and R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , and R 7 and are as defined in the present disclosure to form a compound of formula (I), wherein R 9 is CO 2 R 10 or COSR 10 , preferably CO 2 R 10 ; R 10 is H, methyl, or ethyl; R 8 is H, CH 2 Ph(o-OH), C(O)CH 2 NH 2 , C(O)CH(NH 2 )(CH 2 ) 4 NH 2 , or C(O)CH(NH 2 )(CH 2 ) 3 N(H)C(NH)(NH 2 ); and R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , and R 7 are as defined in the present disclosure. It is to be under- stood that any of said protecting group(s) that may be removed may be the R 8 group, preferably R 8 is benzyl; or one or more protecting group(s) of the R 8 group, pref- erably the methyl of CH 2 Ph(o-OMe) of R 8 ; or one or more protecting groups of R 14 of R 8 , preferably R 14 is t-Boc or Fmoc, or combinations thereof. Additionally, or alternatively, the method com- prises (x) converting the compound of formula (I) to a pharmaceutically acceptable salt thereof. Additionally, or alternatively, to converting the compound of formula (I) to a pharmaceutically acceptable salt thereof, the method for the preparation of a compound of formula (I), or pharmaceutically acceptable salt or a stereoisomer thereof, further comprises isolating an isomer of the compound of formula (I), preferably isolating an isomer of the compound of formula (I), which is dextrorotatory, wherein said step is performed before or after the step of optionally converting the compound of formula (I) to a pharmaceutically acceptable salt thereof, preferably before the step of optionally converting the compound of formula (I) to a pharmaceutically acceptable salt thereof. Additionally, or alternatively, to converting the compound of formula (I) to a pharmaceutically acceptable salt thereof, the method for the preparation of a compound of formula (I), or pharmaceutically acceptable salt or a stereoisomer thereof, further comprises converting the compound of formula (I) to another stereoisomer thereof, wherein said step is performed before or after the step of optionally converting the compound of formula (I) to a pharmaceutically acceptable salt thereof. Additionally, or alternatively, the method further comprises one or more steps of separating and/or purifying one or more compound(s) of formula (I) and/or formula (II). The compounds as disclosed herein and hereafter can be synthesized using well documented reactions and commercially available starting materials. These novel compounds may be used to achieve desirable pharmacological responses. Furthermore, compounds of formula (I) may be used as synthesis intermediates for the preparation of other compounds or other pharmaceutically active compositions, which are obtainable from compounds of formula (I) and, for example by introduction of substituents or modification of functional moieties. The compounds and pharmaceutical compositions as defined in the present disclosure may also be useful in medical devices and medical kits. GENERAL PREPARATION METHODS The compounds according to the present invention may be prepared by processes known per se as follows. The following non-limiting examples illustrate the preparation of compounds of formula (I). Not all steps or features of the reactions are discussed in detail, as many of the steps or features will be obvious for the person skilled in the art. General procedure A for the preparation of compounds of formula (I) Step 1: was prepared as follows (Krasovskiy, A.; Knochel, P. Angew. Chem. Int. Ed. 2004, 43, 3333): a dry and argon flushed flask, equipped with a magnetic stirrer and a septum, was charged with i-PrMgCl·LiCl in THF, (1.05-1.2 equiv.) and kept under 15 °C. Aryl halide (such as 4-chloro-1,2- difluorobenzene, 1 equiv.) was dropwise added to the i- PrMgCl·LiCl at 0 °C - 15 °C, the formed reaction mixture was stirred at a temperature under 15 °C, and the hal- ogen/Mg exchange was checked by TLC. The formed crude Grignard reagent was used in a subsequent reaction step without purification; step 2: a reaction mixture of optionally substituted 3-azet- idinone (1 equiv.), alkylhalide (such as methyl bromide or benzylchloride) (1.05-1.2 equiv.), K 2 CO 3 (1.05-1.3 equiv.) and MeOH/isopropylalcohol (10-20 mL/ 10 mmol of 3-azetidinone) was refluxed to form crude optionally substituted 1-alkyl-3-azetidinone (such as 1-benzyl-3- azetidinone or 1-methyl-3-azetidinone) that was used in a subsequent reaction step without purification; step 3: the formed Grignard reagent from step 1 of general pro- cedure A (in THF) was cooled to -10 °C and the formed optionally substituted 1-alkyl-3-azetidinone (1.1-1.2 equiv.) from step 2 of general procedure A was dropwise added. The mixture was stirred for 1-2 h (until comple- tion of the reaction) at an ice bath and was quenched with sat. aqueous NH 4 Cl solution. The aqueous phase was extracted with DCM, dried with Na 2 SO 4 , and concentrated in vacuo. The formed crude optionally substituted 1- alkyl-3-phenyl-3-azetidinol was used in a subsequent reaction step without purification; step 4: optionally substituted 1-alkyl-3-phenyl-3-azetidinol (1 equiv.) from step 3 of general procedure A was dissolved into ether (20 mL per gram of optionally substituted 1- alkyl-3-phenyl-3-azetidinol) at 0 °C under stirring on an ice bath. PBr 3 (0.41 equiv.) was slowly added to the formed solution to maintain the temperature and stirring was continued further for 2 h. The reaction mixture was poured into basified water, such as saturated NaHCO 3 solution, (6.4 mL per gram of optionally substituted 1- alkyl-3-phenyl-3-azetidinol), the organic phase was separated, and the aqueous phase was extracted twice with diethyl ether (2 ml per gram of optionally substi- tuted 1-alkyl-3-phenyl-3-azetidinol). The ether phases were combined and washed twice with water, dried with MgSO 4 , and concentrated in vacuo. The formed crude op- tionally substituted 3-bromo-1-alkyl-3-phenylazetidine was used in a subsequent reaction step without further purification; step 5: optionally substituted 3-bromo-1-alkyl-3-phenylazet- idine (1 equiv.) from step 4 of general procedure A was dissolved into DMF (20 mL / gram) under stirring at 0 °C, then KCN (2.1 equiv.) was added to the solution and the ice-bath was removed. After 1.5 hours the mixture was poured into water (40 mL / gram of optionally sub- stituted 3-bromo-1-alkyl-3-phenylazetidine) and ex- tracted twice with hexane (2 mL / 1 g of the optionally substituted 3-bromo-1-alkyl-3-phenylazetidine). The or- ganic phases were combined, washed twice with water (2 mL / 1g of the optionally substituted 3-bromo-1-alkyl- 3-phenylazetidine), dried with MgSO 4 and concentrated in vacuo. The formed crude optionally substituted 1-alkyl- 3-phenyl-3-azetidinecarbonitrile was recrystallized from petroleum ether, or used in a subsequent reaction step without further purification; in the case of preparation of a product of formula (I), wherein R 8 = alkyl (such as methyl, ethyl, or protecting group such as benzyl), and R 9 = CO 2 H, otherwise skip to step 6 (alternative 1b): a suspension of optionally substituted 1-alkyl-3-phe- nyl-3-azetidinecarbonitrile from step 5 of general pro- cedure A in an aqueous solution of HCl (pH being 4-5) and toluene was refluxed for 1-2 h followed by an aze- otropic distillation, yielding optionally substituted 1-alkyl-3-phenyl-3-azetidinecarboxylic acid that was used in a subsequent reaction step without further pu- rification or optionally purified by flash chromatog- raphy; or step 6 (alternative 1b): in the case of preparation of a product of formula (I), wherein R 8 = H, and R 9 = CO 2 H, otherwise skip to Step 6 (alternative 2): step 6 (alternative 1a) of general procedure A was fol- lowed using 1-alkyl-3-phenyl-3-azetidinecarbonitrile (alkyl = protecting group, such as benzyl) from step 5 of general procedure A, and the crude residue was puri- fied by flash chromatography or used in a subsequent reaction step without purification. The synthesis pro- ceeds with deprotection of optional protecting group (i.e. R 8 = protecting group, such as benzyl) of the nitrogen according to procedures well known in the art, e.g., with catalytic hydrogenation using H 2 and plati- num(IV) oxide (PtO 2 ), Raney nickel, palladium on carbon (Pd/C) and/or Platinum on carbon (Pt/C). The crude res- idue was optionally purified by flash chromatography yielding optionally substituted 3-phenyl-3-azetidine- carboxylic acid; or step 6 (alternative 2): in the case of preparation of a product of formula (I), wherein R 8 = alkyl (such as methyl, ethyl, or 2-cyano- 1-phenylethyl), and R 9 = CO2R 10 (R 10 = alkyl such as methyl or ethyl) from the corresponding nitrile, otherwise skip to optional step 7 (alternative 1): to a suspension of optionally substituted 1-alkyl-3- phenyl-3-azetidinecarbonitrile (3 equiv., from step 5) and methanol or ethanol (1 equiv.), and optionally ni- trobenzene (1 equiv.) was added a Lewis catalyst such as trimethylsilyl triflate (TMSOTf, 2 equiv.), and the mixture is stirred at rt for 65 h. H 2 O (25 mL) and brine (25 mL) are added, and the mixture is extracted with EtOAc (3 × 30 mL). The combined organic layers are dried (Na 2 SO 4 ) and concentrated. The crude product is purified by flash column chromatography (silica gel) yielding optionally substituted alkyl 1-alkyl-3-phenyl-3-azet- idinecarboxylate (the alkyls may be the same or differ- ent alkyls). In the case of protecting group(s) at R 8 , the protecting group(s) was/were removed according to procedures well known in the art, e.g., in the case the protecting group = TIPS, 2.2 mol eq. (TBAF) (1 M solution in THF) was used. The crude product is purified by flash column chromatography (silica gel) yielding the unpro- tected optionally substituted alkyl 1-alkyl-3-phenyl-3- azetidinecarboxylate (the alkyls may be the same or dif- ferent alkyls). Optional step 7 (alternative 1): in the case of preparation of a product of formula (I), wherein R 8 = alkyl (such as methyl, ethyl, or 2-cyano- 1-phenylethyl), and R 9 = CO2R 10 (R 10 = alkyl such as methyl or ethyl) from a carboxylic acid, otherwise skip to optional step 7 (alternative 2): a suspension of optionally substituted 1-alkyl-3-phe- nyl-3-azetidinecarboxylic acid from step 6 (alternative 1a), excess alcohol (methanol or ethanol) (10 mL/ 1 g of the optionally substituted 1-alkyl-3-phenyl-3-azet- idinecarboxylic acid), and a catalytic amount of H 2 SO 4 was refluxed for 18 h. EtOAc (2 mL/ 1 g of the optionally substituted 1-alkyl-3-phenyl-3-azetidinecarboxylic acid) and a saturated solution of NaHCO 3 (1 mL/ 1 g of the optionally substituted 1-alkyl-3-phenyl-3-azet- idinecarboxylic acid) is added, and the mixture is washed with a saturated solution of NaHCO 3 (2 × 1 mL/ 1 g of the optionally substituted 1-alkyl-3-phenyl-3- azetidinecarboxylic acid). The organic layer is dried (Na 2 SO 4 ) and concentrated. The crude product is purified by flash column chromatography (silica gel) yielding optionally substituted alkyl 1-alkyl-3-phenyl-3-azet- idinecarboxylate (the alkyls may be the same or differ- ent alkyls). In the case of protecting group(s) at R 8 , the protecting group(s) may be removed according to pro- cedures well known in the art, e.g., in the case the protecting group = TIPS, 2.2 mol eq. (TBAF) (1 M solution in THF) was used. The crude product is purified by flash column chromatography (silica gel) yielding the unpro- tected optionally substituted alkyl 1-alkyl-3-phenyl-3- azetidinecarboxylate (the alkyls may be the same or dif- ferent alkyls); or optional step 7 (alternative 2): in the case of preparation of a product of formula (I), wherein R 8 = H, and R 9 = CO 2 R 10 (R 10 = alkyl such as methyl or ethyl) from a protected carboxylic acid (i.e., R 8 = protecting group), otherwise skip to optional step 7 (alternative 3): a suspension of optionally substituted 1-alkyl-3-phe- nyl-3-azetidinecarboxylic acid from step 6 (alternative 1a), excess alcohol (methanol or ethanol) (10 mL/ 1 g of the optionally substituted 1-alkyl-3-phenyl-3-azet- idinecarboxylic acid), and a catalytic amount of H 2 SO 4 was refluxed for 18 h. EtOAc (2 mL/ 1 g of the optionally substituted 1-alkyl-3-phenyl-3-azetidinecarboxylic acid) and a saturated solution of NaHCO 3 (1 mL/ 1 g of the optionally substituted 1-alkyl-3-phenyl-3-azet- idinecarboxylic acid) is added, and the mixture is washed with a saturated solution of NaHCO 3 (2 × 1 mL/ 1 g of the optionally substituted 1-alkyl-3-phenyl-3- azetidinecarboxylic acid). The organic layer is dried (Na 2 SO 4 ) and concentrated. The crude product is purified by flash column chromatography (silica gel) yielding optionally substituted alkyl 1-alkyl-3-phenyl-3-azet- idinecarboxylate (the alkyls may be the same or differ- ent alkyls). In the case of protecting group(s) present, the protecting group(s) was removed according to proce- dures well known in the art, e.g., in the case the R 8 is a protecting group, which is benzyl, with catalytic hydrogenation using H2 and platinum(IV) oxide (PtO2), Raney nickel, palladium on carbon (Pd/C) and/or Platinum on carbon (Pt/C). The crude product was optionally pu- rified by flash column chromatography (silica gel) yielding the unprotected optionally substituted alkyl 3-phenyl-3-azetidinecarboxylate; or optional step 7 (alternative 3): in the case of preparation of a product of formula (I), wherein R 8 = H, and R 9 = CO 2 R 10 (R 10 = alkyl such as methyl or ethyl) from a protected carboxylic ester (i.e., R 8 = protecting group), otherwise skip to optional step 7 (alternative 4): deprotection of the possible protecting group of the nitrogen of the optionally substituted alkyl 1-alkyl-3- phenyl-3-azetidinecarboxylate (i.e., R 8 = protecting group such as benzyl, e.g. the benzyl of methyl 1-ben- zyl-3-phenyl-3-azetidinecarboxylate) from step 6 (al- ternative 2) or optional step 7 (alternative 1) was performed according to procedures well known in the art, e.g., with catalytic hydrogenation using H 2 and plati- num(IV) oxide (PtO 2 ), Raney nickel, palladium on carbon (Pd/C) and/or Platinum on carbon (Pt/C). The crude res- idue was optionally purified by flash chromatography yielding optionally substituted alkyl 3-phenyl-3-azet- idinecarboxylate; or optional step 7 (alternative 4): in the case of preparation of a product of formula (I), wherein R 8 = methyl, ethyl, CH 2 Ph(o-OMe), or CH 2 Ph(o- OH), and R 9 = CO2R 10 (R 10 = alkyl such as methyl or ethyl) from an optionally substituted alkyl 3-phenyl-3-azet- idinecarboxylate, otherwise skip to step 7 (alternative 5): a reductive alkylation of the optionally substituted alkyl 3-phenyl-3-azetidinecarboxylate from optional step 7 (alternative 2 or 3) was performed according to procedures well known in the art, e.g., by combining methanal, ethanal, o-[tris(isopropyl)siloxy]benzalde- hyde, or 2-methoxybenzaldehyde with the alkyl 3-phenyl- 3-azetidinecarboxylate from optional step 7 (alterna- tive 2 or 3), optionally in the presence of one or more water removing compounds or apparatus arranged to remove water, forming the imine. The formed imine was reduced with a reducing agent, such as sodium borohydride. The crude residue was purified by flash chromatography yielding optionally substituted alkyl 1-alkyl-3-phenyl- 3-azetidinecarboxylate (the alkyls may be the same or different alkyls). In the case of protecting group(s) at R 8 , the protecting group(s) was/were removed accord- ing to procedures well known in the art, e.g., in the case the protecting group = TIPS, 2.2 mol eq. (TBAF) (1 M solution in THF) was used. The crude product was pu- rified by flash column chromatography (silica gel) yielding the unprotected optionally substituted alkyl 1-alkyl-3-phenyl-3-azetidinecarboxylate (the alkyls may be the same or different alkyls); or an N-alkylation of the optionally substituted alkyl 3- phenyl-3-azetidinecarboxylate from optional step 7 (al- ternative 2 or 3) was performed according to procedures well known in the art, e.g., combining a haloalkane such as methylbromide, ethylbromide, methyliodide, ethylio- dide, [o-(bromomethyl)phenoxy]tris(isopropyl)silane, or o-(bromomethyl)methoxybenzene, with the alkyl 3-phenyl- 3-azetidinecarboxylate from optional step 7 (alterna- tive 2 or 3), optionally in the presence of one or more activating group reactant(s) and/or one or more acti- vating agent(s) such as sodium iodide. The crude residue was optionally purified by flash chromatography yielding optionally substituted alkyl 1-alkyl-3-phenyl-3-azet- idinecarboxylate (the alkyls may be the same or differ- ent alkyls). In the case of protecting group(s) at R 8 , the protecting group(s) was/were removed according to procedures well known in the art, e.g., in the case the protecting group = TIPS, 2.2 mol eq. (TBAF) (1 M solution in THF) was used. The crude product is optionally puri- fied by flash column chromatography (silica gel) yield- ing the unprotected optionally substituted alkyl 1-al- kyl-3-phenyl-3-azetidinecarboxylate (the alkyls may be the same or different alkyls); or optional step 7 (alternative 5): in the case of preparation of a product of formula (I), wherein R 8 = C(O)CH2NH2, C(O)CH(NH2)(CH2)4NH2, C(O)CH(NH 2 )(CH 2 ) 3 NHCNH(NH 2 ), and R 9 = CO 2 R 10 (R 10 = alkyl such as methyl or ethyl) from a carboxylic ester, oth- erwise skip to optional step 7 (alternative 6): an amidation of the optionally substituted alkyl 3-phe- nyl-3-azetidinecarboxylate from optional step 7 (alter- native 2 or 3) was performed according to procedures well known in the art, e.g., combining an optionally protected amino acid (such as N-(tert-butoxycar- bonyl)glycine) with the optionally substituted alkyl 3- phenyl-3-azetidinecarboxylate from optional step 7 (al- ternative 2 or 3) with an activating group reactant (such as a carbodiimide such as diethyl azodicarboxylate (DEAD), optionally in the presence of an activating agent (such as 1-hydroxy-7-azabenzotriazole (HOAt). The crude formed residue was purified by flash chromatog- raphy or used in an optional subsequent reaction step without purification. In the case of protecting group(s) at R 8 , the protecting group(s) was/were removed accord- ing to procedures well known in the art, e.g., in the case of t-Boc protected compounds by treating with tri- fluoroacetic acid in dichloromethane, or with HCl in methanol. The formed crude product was purified by flash chromatography or used in an optional subsequent reac- tion step without purification; or optional step 7 (alternative 6): in the case of preparation of a product of formula (I), wherein R 8 = methyl, ethyl, CH 2 Ph(o-OMe), CH 2 Ph(o-OH), C(O)CH 2 NH 2 , C(O)CH(NH 2 )(CH 2 ) 4 NH 2 , C(O)CH(NH 2 )(CH 2 ) 3 N(H)C(NH)(NH 2 ), 5-(1,2-dithiolan-3-yl)valeryl, and 2- cyano-1-phenylethyl, and R 9 = CO 2 H from a carboxylic ester: hydrolysis of the carboxylic ester from step 6 (alter- native 2), optional step 7 (alternative 1, 4, or 5 was performed according to procedures well known in the art, e.g., reacting the carboxylic ester with a solution com- prising NaHCO 3 and water or a solution comprising di- luted aqueous NaOH at room temperature. The crude res- idue was acidified, extracted with EtOAc, dried with MgSO 4 , and concentrated in vacuo, and optionally puri- fied by flash chromatography yielding the optionally substituted carboxylic acid. The pharmaceutically acceptable salts of the compounds of formula (I) may be prepared by conventional processes well-known to the person skilled in the art. For the preparation of pharmaceutical compositions and dosage forms as well as the carriers, diluents and ex- pedients used in the preparation, see, for example, Re- mington's Pharmaceutical Sciences, 20th Edition, 2000, Mack Publishing Company, Easton, Pennsylvania. The pharmacological activity of the compounds of the invention may be verified by methods known in the art. For example, the reducing effect on alcohol seeking behavior can be verified using the procedure described by Heidbreder, C.A., et al., Addict Biol. 2007 Mar;12(1):35-50. The Parkinsonism-preventing activity can be shown, for example, as described by Okuda, K., et al. Biol Pharm Bull. 2006 Jul;29(7):1401-1403. Reference will now be made in detail to some embodiments. The following specific non-limiting examples will further identify the compounds as disclosed herein and hereafter. Not all steps or features of the embodiments are discussed in detail, as many of the steps or features will be obvious for the person skilled in the art based on this specification. COMPOUND 1 Ethyl 3-(3,4-difluorophenyl)-1,2,4-trimethylazetidine- 3-carboxylate The general procedure A (step 1) was followed starting from 4-chloro-1,2-difluorobenzene. 2,4-Dimethyl-3- azetidinone and methyl iodide were used in step 2 of the general procedure A. Subsequently, steps 3-5, step 6 (alternative 1a), and optional step 7 (alternative 1, wherein ethanol was used), of the general procedure A The general procedure A (step 1) was followed starting from 4-chloro-1,2-difluorobenzene. 3-Azetidinone and ethyl iodide were used in step 2 of the general procedure A. Subsequently, steps 3-5, step 6 (alternative 1a), and The general procedure A (step 1) was followed starting from 4-chloro-1,2-difluorobenzene. 3- Azetidinone and methyl iodide were used in step 2 of the general procedure A. Subsequently, steps 3-5, step 6 (alternative 1a), and optional step 7 (alternative 1, wherein ethanol was used), of the general procedure A were followed. 1 H NMR (500 MHz, DMSO-d6) δ 7.29 - 7.14 (m, 3H), 4.23 (q, J = 6.1 Hz, 2H), 3.95 (d, J = 11.5 Hz, 2H), 3.53 (dd, J = 11.5, 7.1 Hz, 2H), 2.34 (s, 3H), 1.22 starting from p-chlorotoluene. 3-Azetidinone and benzyl bromide were used in step 2 of the general procedure A. Subsequently, steps 3-5, step 6 (alternative 1a), and optional step 7 (alternative 2, wherein ethanol was used, and removal of the benzyl protecting group using from m-chlorofluorobenzene. 2,4-Dimethyl-3-azetidinone and benzyl bromide were used in step 2 of the general procedure A. Subsequently, steps 3-5, step 6 (alternative 1a), and optional step 7 (alternative 2, wherein ethanol was used, and removal of the benzyl protecting group using H 2 and Pd/C), of the general procedure A were followed. 1 H NMR (500 MHz, DMSO-d6) δ 7.34 (dddd, J = 7.9, 7.4, 5.0, 0.5 Hz, 1H), 7.30 - 7.19 (m, 2H), 7.10 (dddd, J = 10.1, 7.3, 2.2, 1.3 Hz, 1H), The general procedure A (step 1) was followed starting from m-chlorofluorobenzene. 2-Methyl-3-azetidinone and benzyl bromide were used in step 2 of the general procedure A. Subsequently, steps 3-5, step 6 Ethyl 3-(4-methylphenyl)azetidine-3-carboxylate The general procedure A (step 1) was followed starting from p-chloromethylbenzene. 3-Azetidinone and benzyl bromide were used in step 2 of the general procedure A. Subsequently, steps 3-5, step 6 (alternative 1a), and optional step 7 (alternative 2, wherein ethanol was used, and removal of the benzyl protecting group using H 2 and Pd/C), of the general procedure A were followed. 1H NMR (500 MHz, DMSO-d6) δ 7.33 - 7.27 (m, 2H), 7.10 (dp, J = 8.3, 0.6 Hz, 2H), 4.23 (q, J = 6.0 Hz, 2H), 3.97 (dd, J = 13.0, 3.5 Hz, 2H), 3.66 (dd, J = 13.1, 3.5 Hz, 2H), 3.59 - 3.52 (m, 1H), 2.30 (d, J = 1.3 Hz, 3H), 1.22 (t, J = 6.0 Hz, 3H). COMPOUND 8 Methyl 2-methyl-3-(3-methylphenyl)azetidine-3- carboxylate The general procedure A (step 1) was followed starting from m-chlorotoluene. 2-Methyl-3-azetidinone and benzyl bromide were used in step 2 of the general procedure A. Subsequently, steps 3-5, step 6 (alternative 1a), and 7 (alternative 2, wherein methanol was of the benzyl protecting group using of the general procedure A were followed. DMSO-d6) δ 7.30 (ddd, J = 7.7, 2.2, 1.3 - 7.19 (m, 2H), 7.11 (dddd, J = 7.3, 2.1, 1H), 4.18 (dqd, J = 5.8, 4.8, 1.0 Hz, 1H), = 13.0, 3.6 Hz, 1H), 3.73 (ddd, J = 12.9, 4H), 3.20 (dt, J = 5.9, 3.6 Hz, 1H), 2.31 Hz, 3H), 1.10 (d, J = 4.8 Hz, 3H). 3-(3,4-difluorophenyl)-1-ethyl-2- 3-carboxylate procedure A (step 1) was followed starting 1,2-difluorobenzene. 2-Methyl-3- azetidinone and benzyl bromide were used in step 2 of the general procedure A. Subsequently, steps 3-5, step 6 (alternative 1a), optional step 7 (alternative 2, wherein methanol was used, and removal of the benzyl protecting group), and optional step 7 (alternative 4, reductive alkylation using ethanal and sodium borohydride) of the general procedure A were followed. the general procedure A. Subsequently, steps 3-5, step 6 (alternative 1a), and optional step 7 (alternative 2, wherein methanol was used, and removal of the benzyl protecting group using H 2 and Pd/C) of the general procedure A were followed. 1 H NMR (500 MHz, DMSO-d6) δ 7.30 - 7.19 (m, 2H), 7.16 (dddd, J = 9.2, 3.5, 2.1, 0.3 Hz, 1H), 4.20 (dqd, J = 5.8, 4.8, 1.0 Hz, 1H), 3.95 (dd, J = 13.0, 3.6 Hz, 1H), 3.75 (ddd, J = 13.0, 3.6, 1.0 Hz, 4H), 3.21 (dt, J = 5.9, 3.6 Hz, 1H), 1.10 (d, J = 4.8 Hz, 3H). EXPERIMENTS RELATING TO BINDING AFFINITIES OF THE COMPOUNDS OF THE INVENTION Calculations of binding affinities of the compounds of the invention were performed using Qvina2 using the value 5061982 for explicit random seed. Exhaustiveness was 100. The method of performing binding affinity -calculations of the compounds of the invention is well known in the art and also described in Alhossary A. et al. Fast, Accurate, and Reliable Molecular Docking with QuickVina 2 Bioinformatics (2015) 31 (13) 2214- 2216, O. Trott, A. J. Olson, AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization and multithreading, Journal of Computational Chemistry 31 (2010) 455-461, Feinstein WP, Brylinski M. Calculating an optimal box size for ligand docking and virtual screening against experimental and predicted binding pockets. J. Cheminform. (2015), 7 (1): 18, Tetko, I. V. et al. Virtual computational chemistry laboratory - design and description, J. Comput. Aid. Mol. Des., 2005, 19, 453- 63, VCCLAB, Virtual Computational Chemistry Laboratory, http://www.vcclab.org, 2005. The search space (X,Y,Z)- coordinates of the centre for the calculations of binding affinities can be obtained with methods well known in the art. E.g., the search space (X,Y,Z)- coordinates for PDB ID 1bp3 and protein homolog ClassA_5ht1a_human_Active_6G79_2018-07-10_GPCRDB were (13.730, 30.880, 13.170) and (91.84457623, 54.99481, 63.31365), respectively. The PDB IDs and the protein homolog codes of proteins used in the calculations of the compounds of the invention are listed in Table 1 and some of the results of the calculations are presented in Table 2. Table 1. The PDB IDs and the protein homolog codes of proteins used in the calculations of binding affinities of compounds disclosed herein and hereafter. 1 The PDB structures were obtained from the Protein Data Bank: http://www.rcsb.org/. 2 Protein homologs were obtained from GPCRdb (http://docs.gpcrdb.org/index.html , Pándy-Szekeres G, Munk C, Tsonkov TM, Mordalski S, Harpsøe K, Hauser AS, Bojarski AJ, Gloriam DE. GPCRdb in 2018: adding GPCR structure models and ligands. 2017, Nucleic Acids Res., Nov 16. http://dx.doi.org/10.1093/nar/gkx1109). Table 2. Results of binding affinity calculations of compounds of the invention performed on proteins, logP and logS values. 1 The number of the docked protein corresponds to the entry number found in Table 1. The compound with strongest binding affinity for trace amine receptor 1 (TAAR1), norepinephrine transporter (NET), 5-HT2A receptor, 5-HT2B receptor, OX1 orexin receptor, Mu-opioid receptor, Neurokinin 1 receptor (NK1R), and Melanocortin 3 receptor (MC3R) is ethyl 3- (3,4-difluorophenyl)-1,2,4-trimethylazetidine-3- carboxylate (1). The compound with strongest binding affinity for dopamine transporter (DAT) is ethyl 3-(3,4- difluorophenyl)-1-methylazetidine-3-carboxylate (3). The compound with strongest binding affinity for dopamine 1 (D1) receptor, Sigma-1 receptor, and 1B Melatonin receptor is methyl 1-ethyl-2-methyl-3-(3- methylphenyl)azetidine-3-carboxylate (15): The compound with strongest binding affinity for dopamine 2 (D2) receptor, MAO-A, Alpha-2C adrenergic receptor, α4β2 nicotinic receptor, and CHK1 kinase is ethyl 3-(4-methylphenyl)azetidine-3-carboxylate (7). The compound with strongest binding affinity for dopamine 3 (D3) receptor, and Kappa-opioid receptor is ethyl 3-(3,4-difluorophenyl)-2-methylazetidine-3- carboxylate (19): The compound with strongest binding affinity for dopamine 4 (D4) receptor is methyl 2-methyl-3-(3- methylphenyl)azetidine-3-carboxylate (8). The compound with strongest binding affinity for phenylethanolamine N-methyltransferase (PNMT) is ethyl 3-(3-fluorophenyl)-2,4-dimethylazetidine-3-carboxylate (5). The compound with strongest binding affinity for serotonin transporter (SERT) is ethyl 3-(4- 3-carboxylate (20): The compound with strongest binding affinity for serotonin N-acetyltransferase is ethyl 1-ethyl-3-(3- methylphenyl)azetidine-3-carboxylate (21): The compound with strongest binding affinity for N- acetylserotonin methyltransferase, 1A Melatonin receptor, and Melanocortin 1 receptor (MSHR, MC1R) is methyl 3-(3,4-difluorophenyl)-1-ethyl-2- methylazetidine-3-carboxylate (9). The compound with strongest binding affinity for 5-HT1A receptor is methyl 1-ethyl-3-(3-methoxyphenyl)-2,4- dimethylazetidine-3-carboxylate (22): (22). The compound with strongest binding affinity for 5-HT1B receptor, and β1 adrenergic receptor is ethyl 1-ethyl- 2,4-dimethyl-3-(4-methylphenyl)azetidine-3-carboxylate The compound with strongest binding affinity for 5-HT2C receptor, Alpha-1B adrenergic receptor, Alpha-2B adrenergic receptor, Melanocortin 2 receptor (ACTHR, MC2R), and Melanocortin 4 receptor (MC4R) is ethyl 2,4- dimethyl-3-(3-methylphenyl)azetidine-3-carboxylate (24). The compound with strongest binding affinity for 5-HT3 receptor is methyl 3-(3,4-difluorophenyl)-2,4- dimethylazetidine-3-carboxylate (25): The compound with strongest binding affinity for MAO-B is ethyl 3-(3,4-difluorophenyl)azetidine-3-carboxylate (4). The compound with strongest binding affinity for COMT is ethyl 1,2-dimethyl-3-(3-methylphenyl)azetidine-3- carboxylate (26): The compound with strongest binding affinity for Alpha- 1A adrenergic receptor, and Nociceptin / orphanin receptor is methyl 1-ethyl-2,4-dimethyl-3-(3- methylphenyl)azetidine-3-carboxylate (27): The compound with strongest binding affinity for Alpha- 2A adrenergic receptor, and Prolactin receptor is methyl 3-(3,4-difluorophenyl)-2-methylazetidine-3-carboxylate (10). The compound with strongest binding affinity for β2 adrenergic receptor methyl 1-ethyl-2-methyl-3-(4- methylphenyl)azetidine-3-carboxylate (28): (28). The compound with strongest binding affinity for Phenylalanine hydroxylase is methyl 1-ethyl-3-(4- methoxyphenyl)-2,4-dimethylazetidine-3-carboxylate The compounds with strongest binding affinity for Tyrosine hydroxylase are methyl 1-ethyl-3-(4- methoxyphenyl)-2-methylazetidine-3-carboxylate (30): The compound with strongest binding affinity for H1 histamine receptor is ethyl 1-ethyl-3-(4- methylphenyl)azetidine-3-carboxylate (12): (12). The compound with strongest binding affinity for H3 histamine receptor, and Neurokinin 2 receptor (NK2R) is ethyl 3-(3-fluorophenyl)-1,2,4-trimethylazetidine-3- carboxylate (17): (17). The compound with strongest binding affinity for OX2 orexin receptor is ethyl 3-(3,4-difluorophenyl)-1- ethyl-2-methylazetidine-3-carboxylate (32): (32). The compound with strongest binding affinity for NMDA receptor GluN1 is methyl 2-methyl-3-(4- methylphenyl)azetidine-3-carboxylate (33): The compound with strongest binding affinity for NMDA receptor Glu2B, M2 muscarinic acetylcholine receptor, and neuropeptide Y1 receptor is ethyl 3-(3,4- difluorophenyl)-2,4-dimethylazetidine-3-carboxylate The compound with strongest binding affinity for Delta- opioid receptor, M1 muscarinic acetylcholine receptor, M3 Muscarinic acetylcholine receptor, and M4 muscarinic acetylcholine receptor is methyl 3-(3,4- difluorophenyl)-1-ethyl-2,4-dimethylazetidine-3- carboxylate (14): The compound with strongest binding affinity for Acetylcholinesterase, and Melanocortin 5 receptor (MC5R) is ethyl 2-methyl-3-(3-methylphenyl)azetidine-3- carboxylate (11): The compound with strongest binding affinity for Neuropeptide Y2 receptor is ethyl 3-(3- methylphenyl)azetidine-3-carboxylate (16): The compound with strongest binding affinity for Neuropeptide Y5 receptor is methyl 3-(4-methylphenyl)- 1,2-dimethylazetidine-3-carboxylate (34): The compound with strongest binding affinity for Neurokinin 3 receptor (NK3R) is ethyl 1-ethyl-2,4- dimethyl-3-(3-methylphenyl)azetidine-3-carboxylate (18). The compound with strongest binding affinity for Oxytocin receptor (OXYR) is ethyl 3-(3- fluorophenyl)azetidine-3-carboxylate (35): The compound with strongest binding affinity for Corticotropin-releasing factor receptor 1 (CRF1R) is ethyl 1,2-dimethyl-3-(4-methylphenyl)azetidine-3- carboxylate (36): It will be obvious to a person skilled in the art that, as the technology advances, the inventive concept can be implemented in various ways. The invention and its embodiments are not limited to the examples described above but may vary within the scope of the claims. The embodiments described hereinbefore may be used in any combination with each other. Several of the embodiments may be combined together to form a further embodiment. A product, a system, a method, or a use, disclosed herein, may comprise at least one of the embodiments described hereinbefore. It will be understood that the benefits and advantages described above may relate to one embodiment or may relate to several embodiments. The embodiments are not limited to those that solve any or all of the stated problems or those that have any or all of the stated benefits and advantages.