ACS Publications. Most Trusted. Most Cited. Most Read
My Activity
CONTENT TYPES

Figure 1Loading Img
RETURN TO ISSUEPREVResearch ArticleNEXT

Knowledge-Based Design of Long-Chain Arylpiperazine Derivatives Targeting Multiple Serotonin Receptors as Potential Candidates for Treatment of Autism Spectrum Disorder

  • Enza Lacivita*
    Enza Lacivita
    Dipartimento di Farmacia−Scienze del Farmaco, Università degli Studi di Bari Aldo Moro, via Orabona, 4, 70125 Bari, Italy
    *Email: [email protected]
  • Mauro Niso
    Mauro Niso
    Dipartimento di Farmacia−Scienze del Farmaco, Università degli Studi di Bari Aldo Moro, via Orabona, 4, 70125 Bari, Italy
    More by Mauro Niso
  • Margherita Mastromarino
    Margherita Mastromarino
    Dipartimento di Farmacia−Scienze del Farmaco, Università degli Studi di Bari Aldo Moro, via Orabona, 4, 70125 Bari, Italy
  • Andrea Garcia Silva
    Andrea Garcia Silva
    Center for Research in Molecular Medicine and Chronic Diseases (CIMUS). Universidade de Santiago de Compostela. Avda. de Barcelona, s/n, 15782 Santiago de Compostela, Spain
  • Cibell Resch
    Cibell Resch
    Cellular Neurophysiology, Hannover Medical School, 30625 Hannover, Germany
    More by Cibell Resch
  • Andre Zeug
    Andre Zeug
    Cellular Neurophysiology, Hannover Medical School, 30625 Hannover, Germany
    More by Andre Zeug
  • María I. Loza
    María I. Loza
    Center for Research in Molecular Medicine and Chronic Diseases (CIMUS). Universidade de Santiago de Compostela. Avda. de Barcelona, s/n, 15782 Santiago de Compostela, Spain
  • Marián Castro
    Marián Castro
    Center for Research in Molecular Medicine and Chronic Diseases (CIMUS). Universidade de Santiago de Compostela. Avda. de Barcelona, s/n, 15782 Santiago de Compostela, Spain
  • Evgeni Ponimaskin
    Evgeni Ponimaskin
    Cellular Neurophysiology, Hannover Medical School, 30625 Hannover, Germany
  • , and 
  • Marcello Leopoldo*
    Marcello Leopoldo
    Dipartimento di Farmacia−Scienze del Farmaco, Università degli Studi di Bari Aldo Moro, via Orabona, 4, 70125 Bari, Italy
    *Email: [email protected]
Cite this: ACS Chem. Neurosci. 2021, 12, 8, 1313–1327
Publication Date (Web):April 1, 2021
https://doi.org/10.1021/acschemneuro.0c00647

Copyright © 2022 The Authors. Published by American Chemical Society. This publication is licensed under

CC-BY-NC-ND 4.0.
  • Open Access

Article Views

1705

Altmetric

-

Citations

LEARN ABOUT THESE METRICS
PDF (4 MB)
Supporting Info (1)»

Abstract

Autism spectrum disorder (ASD) includes a group of neurodevelopmental disorders characterized by core symptoms such as impaired social interaction and communication, repetitive and stereotyped behaviors, and restricted interests. To date, there are no effective treatments for these core symptoms. Several studies have shown that the brain serotonin (5-HT) neurotransmission system is altered in both ASD patients and animal models of the disease. Multiple pieces of evidence suggest that targeting 5-HT receptors may treat the core symptoms of ASD and associated intellectual disabilities. In fact, stimulation of the 5-HT1A receptor reduces repetitive and restricted behaviors; blockade of the 5-HT2A receptor reduces both learning deficits and repetitive behavior, and activation of the 5-HT7 receptor improves cognitive performances and reduces repetitive behavior. On such a basis, we have designed novel arylpiperazine derivatives pursuing unprecedently reported activity profiles: dual 5-HT7/5-HT1A receptor agonist properties and mixed 5-HT7 agonist/5-HT1A agonist/5-HT2A antagonist properties. Seventeen new compounds were synthesized and tested in radioligand binding assay at the target receptors. We have identified the dual 5-HT1AR/5-HT7R agonists 8c and 29 and the mixed 5-HT1AR agonist/5-HT7R agonist/5-HT2AR antagonist 20b. These compounds are metabolically stable in vitro and have suitable central nervous system druglike properties.

This publication is licensed under

CC-BY-NC-ND 4.0.
  • cc licence
  • by licence
  • nc licence
  • nd licence

Introduction

ARTICLE SECTIONS
Jump To

Autism spectrum disorder (ASD) includes a group of brain developmental disorders characterized by core symptoms such as impaired social interaction and communication, repetitive and stereotyped behaviors, and restricted interests. (1) In addition, ASD patients often present a variety of additional impairments, including intellectual disability. The frequency of ASD is increasing, with present rates of about 1 in 100 children in Europe and 1 in 54 in the United States. (2) To date, the only drugs approved to treat ASD-related symptoms are the atypical antipsychotics aripiprazole and risperidone (Table 1) which are efficacious to treat irritability, hyperactivity, and aggression but not the core symptoms. Thus, ASD has no cure and its treatment represents a largely unmet clinical need. (3) Several mechanisms have been implicated in ASD, such as synaptic dysfunction (alterations in dendritic spine morphology, excitatory/inhibitory imbalance), neuroinflammation, and altered neurotransmitter systems. (3) Serotonin (5-hydroxytryptamine, 5-HT) exerts a very complex modulatory role in the central nervous system (CNS) involving at least 14 diverse membrane 5-HT receptors (5-HTRs), grouped in seven families (5-HT1–7). 5-HT plays a crucial role in shaping neuronal circuits during prenatal and postnatal development by promoting neurogenesis, neuronal differentiation, axon myelination, neuropil formation, and synaptogenesis. (4,5) Accumulating findings indicate that the brain 5-HT neurotransmission system is altered in ASD patients (6) and in rodent models of ASD. (7−9) Selective serotonin reuptake inhibitors (SSRIs) have been often prescribed to ASD patients to treat repetitive and stereotyped behaviors as these drugs are efficacious to treat obsessive-compulsive disorder. However, clinical studies in ASD patients treated with SSRIs did not give consistent results. (10) In fact, some studies reported positive effects on stereotypy and compulsions, whereas others reported that SSRIs worsen stereotypy. In addition, side-effects were highly prevalent probably due to the generalized elevation of 5-HT levels that could lead to not therapeutically valid stimulation of multiple 5-HTRs. Various studies have evidenced that targeting certain 5-HTRs has the potential to treat the core symptoms of ASD and associated intellectual disabilities. A clinical trial in young children with ASD demonstrated that the 5-HT1AR partial agonist buspirone reduces repetitive and restricted behaviors. (11) Studies in rodent models of ASD have provided further support in this direction. In fact, buspirone and the 5-HT1AR full agonist 8-OH-DPAT enhance social interactions. (12,13) The selective 5-HT2AR antagonist M100907 alleviates both learning deficits and repetitive behavior. (14,15) The selective 5-HT6R antagonist SLV reverses the social engagement deficit. (16) The selective 5-HT7R agonist LP-211 (Table 1) corrects behavioral alterations in mouse models of rare neurodevelopmental disorders. In particular, administration of LP-211 (i) improved novel object recognition performance and reduced stereotyped behavior in a mouse model of Fragile-X syndrome (an X-linked disease with autistic features); (17−19) (ii) improved anxiety-related profiles, the exploratory behavior and memory in a mouse model of Rett syndrome (a disease characterized by intellectual disability); (20−22) and (iii) normalized the prepulse inhibition defects observed in a mouse model of CDKL5 deficiency disorder (a disease characterized by severe neurodevelopmental delay impacting cognitive, speech, and visual function). (23) Altogether, the above studies provide support to the search of novel compounds with an appropriate pharmacological profile at 5-HTRs in the prospect to ameliorate the core symptoms of ASD. At present, various examples of drugs or investigational compounds targeting multiple 5-HTRs exist. The most notable examples are aripiprazole (dopamine D2R/5-HT2AR/5-HT7R antagonist, 5-HT1AR partial agonist) and risperidone (dopamine D2R/5-HT2AR/5-HT7R/5-HT1AR antagonist) (Table 1). (24) However, aripiprazole and risperidone are not effective on the core symptoms of ASD, suggesting that such particular activity profile is not suitable to treat the core symptoms of ASD. (3) Vortioxetine, a multimodal antidepressant that inhibits 5-HT transporter and activates 5-HT1A and 5-HT1B receptors, suppresses restrictive–repetitive behaviors in a rodent model of ASD but has less efficacy as a sociability enhancer. (25) These studies strongly suggest the design of new compounds characterized by an activity profile resulting from combinations of activities at 5-HTRs that are predicted to produce effects on the core symptoms of ASD. The first attempt in this direction was reported in 2015 by Canal et al., who developed the compound (+)-5-FPT which was initially described as a high-affinity 5-HT7R and 5-HT1AR partial agonist (Table 1). The (+)-5-FPT potently attenuated stereotypy in three heterogeneous models of stereotypy in a mouse strain characterized by repetitive behavior. (26) In 2020, the same research group reported that (+)-5-FPT was actually a 5-HT1AR/5-HT2CR agonist and 5-HT7R antagonist. (27) This compound was then studied in a mouse model of Fragile X syndrome and found to reduce repetitive behavior through 5-HT1AR partial agonism and to attenuate audiogenic seizure through 5-HT2CR agonism. Thus, the potential of concomitant activation of 5-HT1AR and 5-HT7R was not actually explored.
Table 1. Structural Formulas and Affinity Profiles of the Reference Compounds
a

Not Available.

Therefore, on the basis of the data discussed above, we aimed at identifying completely new modulators of 5-HTRs for studies in the context of ASD. In particular, we searched for (a) a dual 5-HT7R/5-HT1AR agonist, which is predicted to increase social interaction through the activation of 5-HT1AR and to reduce stereotypy and improve cognition through activation of 5-HT7R, and (b) a compound acting as 5-HT7R/5-HT1AR agonist and 5-HT2AR antagonist, which is predicted to improve social behavior through activation of 5-HT1AR, to reduce or eliminate stereotyped behavior by blocking 5-HT2AR, and to improve cognition through activation of 5-HT7R.

Results and Discussion

ARTICLE SECTIONS
Jump To

Study Design

Long-chain arylpiperazine derivatives are known to bind monoamine receptors, including 5-HTRs. The general formula is Ar–piperazine–linker–terminal fragment, and it is known that suitable modifications of Ar, linker, or terminal fragment can lead to selective or nonselective compounds. Two examples of selective arylpiperazine ligands are the 5-HT1AR antagonist WAY-100635 and the 5-HT7R agonist LP-211 (28) (Table 1). Instead, compounds 1 (29) and 2 (30) (Table 1) are dual 5-HT7R/5-HT1AR ligands, and compound 3 (31) is a mixed 5-HT7R/5-HT1AR/5-HT2AR ligand (Table 1). Thus, long-chain arylpiperazines are a versatile framework to identify compounds with a diversified receptor affinity profile. On the other hand, it appears much less trivial to identify arylpiperazines showing the activity profile that we are pursuing (i.e., dual 5-HT7R/5-HT1AR agonist, mixed 5-HT7R agonist/5-HT1AR agonist/5-HT2AR antagonist). To fulfill our aim, we have followed a knowledge-based design (i.e., the collection and reuse of expert knowledge) by selecting Ar groups and terminal fragments present in arylpiperazine-based 5-HTR agonists or antagonists. First, we focused on the Ar group and selected the 1-(biphenyl)piperazine moiety that is the key structural determinant for the agonism at 5-HT7R of LP-211, TP-22, (32) and BA-10 (18) (Table 1). Consequently, the majority of the target compounds are 1-[2-(4-methoxyphenyl)phenyl]piperazine derivatives (see Table 2). In addition, in order to identify dual 5-HT7R/5-HT1AR ligands, the compounds 9 and 30 (Table 2), featuring the 2-acetylphenyl group present in the dual 5-HT7R/5-HT1AR ligand 1 (Table 1), were designed. It is worth noting that, even if LP-211, TP-22, and BA-10 are 5-HT7R-preferring agonists, they still show a measurable affinity for 5-HT1AR (Table 1). Interestingly, LP-211 itself shows measurable 5-HT2AR affinity (Table 1). (28) Thus, the 1-(biphenyl)piperazine framework appears to be compatible with 5-HT1AR and 5-HT2AR affinity, and therefore, suitable variations of the terminal fragments and linkers might increase 5-HT1AR and 5-HT2AR affinity. In the case of 5-HT1AR, the terminal fragment also has a role on the functional activity of the ligand. With this respect, notable examples are the 2-methoxyphenyl piperazine derivatives WAY-100635, (33)4, (34) and MMP. (35) In fact, while WAY-100635 is a 5-HT1AR antagonist, compounds 4 and MMP are agonists. Consequently, we have selected the terminal fragments present in the high-affinity 5-HT1AR agonists MMP (which also shows 5-HT7R affinity in the nanomolar range) and UCM-2550 (36) (Table 1). As for 5-HT2AR, we selected the terminal fragments of the antagonists risperidone, brilaroxazine, (37) and 5 (38) (Table 1). Based on the structural similarity with these antagonists, our newly designed arylpiperazine derivatives were expected to act as 5-HT2AR antagonist, as they display a completely different structural motif as compared to that of 5-HT2AR agonists. (39) The fine-tuning of the affinities at the target receptors was accomplished by incorporating 2–5 atom linkers.
Table 2. CNS MultiParameter Optimization (CNS-MPO), Binding Affinities, Ki Ratios, and Microsomal Stability of the Target Compounds
a

MS: microsomal stability (% of recovery of the parent compound after 30 min incubation with rat microsomes).

b

Full displacement of the specific binding was not achieved at maximum concentration assayed (100 μM); Ki value extrapolated from the analysis might not be accurately estimated;

c

Data from ref (51).

In addition, the drug-likeness CNS multiparameter optimization (CNS MPO) algorithm (40) was applied to the designed compounds. As shown in Table 2, 7 out of 17 compounds present a CNS MPO desirability score higher than 4, predictive of alignment to ADME attributes, ability to cross the blood–brain barrier, and low safety risk. As for the remaining compounds, 8 out 10 show CNS MPO desirability score in the range of 3 to 4 which might still include compounds with desirable properties.
In order to evaluate the liability of the target compounds to metabolic degradation by first-pass oxidative metabolism, the main cause of metabolic degradation in vivo, we screened the metabolic stability in vitro by using rat liver microsomes. (41) First, the metabolic stability was assessed as the percentage of the parent compound recovered after 30 min of incubation with microsomes in the presence of an NADPH-regenerating system. (32) Subsequently, selected compounds showing recovery higher than 20% were further characterized by evaluating half-life (t1/2) and intrinsic clearance (CLint), which are predictive of in vivo hepatic clearance.

Chemistry

The target compounds 8ac and 9 bearing the 4-methyl-1,2,4-triazine-3,5(2H,4H)-dione moiety as the terminal fragment were prepared according to Scheme 1. 4-Methyl-1,2,4-triazine-3,5(2H,4H)-dione (6) (42) was alkylated with the appropriate haloalkylbromide to give the alkyl halides 7ac, which after nucleophilic substitution with the appropriate arylpiperazine, afforded the desired compounds 8ac and 9.

Scheme 1

Scheme 1. Synthesis of Target Compounds 8ac and 9a

aReagents and conditions: (A) NaH, Br–(CH2)n–X, anhydrous DMF, rt, 12 h, 40–70% yield; (B) 1-arylpiperazine; K2CO3, acetonitrile, reflux overnight, 20–50% yield.

Scheme 2 illustrates the synthesis of target compounds 11a,b. Compound 11a was obtained by reacting 3-(2-chloroethyl)-2-methyl-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidin-4-one (10) (43) with 1-[2-(4-methoxyphenyl)phenyl]piperazine. (44) Compound 11b was obtained through a convergent synthesis that required intermediates 13 and 16 (Scheme 2). Amine 13 was synthesized starting from alkyl chloride 10 by sequential reaction with cyanide ion to give the nitrile 12 and reduction of the latter with Raney nickel under hydrogen pressure. Bromide 16 was prepared from 2-(4-methoxyphenyl)aniline (14) (44) and 2-bromoethanol in the presence of CaCO3 to afford the bis alkylated product 15, which was converted into 16 with PBr3. Condensation of the amine 13 and the bromide 16 yielded the target compound 11b.

Scheme 2

Scheme 2. Synthesis of Target Compounds 11a,ba

aReagents and conditions: (A) 1-[2-(4-methoxyphenyl)phenyl]piperazine; K2CO3, acetonitrile, reflux overnight, 61% yield; (B) NaCN, anhydrous DMF, rt, 5 h, quantitative yield; (C) Raney-nickel, H2 (4 atm), MeOH, 50% yield; (D) 2-bromoethanol, CaCO3; reflux, 7 h, 37% yield; (E) PBr3, anhydrous toluene, reflux, 3 h, 65% yield; (F) K2CO3, acetonitrile, reflux overnight, 60% yield.

The target compounds featuring the coumarin nucleus as the terminal fragment were prepared as described in Scheme 3. In order to synthesize compound 20a from compound 19a, several unsuccessful attempts were made to alkylate 7-hydroxy-4-methylcoumarin (18) (45) with 1-bromo-2-chloroethane. Then, compound 19a was instead synthesized starting from 3-(2-bromoethoxy)phenol (17), (46) which was condensed with ethyl acetoacetate under Pechmann conditions. Nucleophilic substitution of 19a by 1-[2-(4-methoxyphenyl)phenyl]piperazine afforded the desired compound 20a. The analogues 20b,c were prepared starting from phenol 18 which was alkylated with the appropriate bromoalkylchloride to afford derivatives 19b,c. These latter compounds were condensed with 1-[2-(4-methoxyphenyl)phenyl]piperazine to give the desired compounds 20b,c, respectively.

Scheme 3

Scheme 3. Preparation of Target Compounds Featuring the Coumarin Nucleus as the Terminal Fragmenta

aReagents and conditions: (A) ethyl acetoacetate; conc. H2SO4, rt, 4 h, 34% yield; (B) 1-[2-(4-methoxyphenyl)phenyl]piperazine; K2CO3, acetonitrile, reflux overnight, 21% yield; (C) NaH, Br–(CH2)n–X, anhydrous DMF, rt, 12 h, 40–60% yield.

The final compounds 26a,b, 29, and 30 featuring the benzoxazinone terminal fragment were prepared following two distinct synthetic routes depending on the nature of the linker (Schemes 4 and 5). In fact, the synthesis of the key intermediate 25a was initially pursued through alkylation of 6-hydroxy-2-methyl-2H-benzo[b][1,4]oxazin-3(4H)-one (27) with 1-bromo-2-chloroethane, but it was unsuccessful. Thus, an alternative synthetic pathway was envisaged (Scheme 4): 4-(benzyloxy)-2-nitrophenol (21) (47) was alkylated with methyl 2-bromopropanoate to afford derivative 22, which was deprotected under basic conditions to give phenol 23. The latter was alkylated with 1,2-dibromoethane to give 24. Reduction of the nitro group of 24 by iron dust in acidic conditions resulted in the cyclization of the intermediate amine to give the key intermediate 25a, which was condensed with 1-[2-(4-methoxyphenyl)phenyl]piperazine to give the target compound 26a. The other target compound 26b bearing the benzoxazinone nucleus as terminal fragment was prepared according to Scheme 5. Alkylation of phenol 27 (48) with 1-bromo-3-chloropropane gave the chloroderivative 25b, which underwent nucleophilic substitution with 1-[2-(4-methoxyphenyl)phenyl]piperazine to give the final compound 26b. The target compounds 29 and 30 were prepared starting from phenol 27 which was alkylated with (R)-glycidylnosilate to give oxirane 28. Ring opening of 28 with the appropriate 1-arylpiperazine gave 29 and 30.

Scheme 4

Scheme 4. Synthetic Route to Obtain Final Compounds 26aa

aReagents and conditions: (A) methyl 2-bromopropionate, K2CO3, acetone, reflux, 16 h, 54% yield; (B) CH3ONa, MeOH, rt, 3 h, 89% yield; (C) 1,2-dibromoethane, K2CO3, anhydrous DMF, 85 °C, 6 h, 70% yield; (D) Fe dust, AcOH, 80 °C, 1 h, 60% yield; (E) 1-[2-(4-methoxyphenyl)phenyl]piperazine; K2CO3, acetonitrile, reflux overnight, 38% yield.

Scheme 5

Scheme 5. Synthetic Route to Obtain Final Compounds 26b, 29, and 30a

aReagents and conditions: (A) NaH, Br–(CH2)3–Cl, anhydrous DMF, rt, 24 h, 22–42% yield; (B) 1-[2-(4-methoxyphenyl)phenyl]piperazine; K2CO3, acetonitrile, reflux overnight, 20–65% yield. (C) NaH, (R)-glycidyl nosilate, anhydrous DMF, rt, overnight, 41% yield; (D) 1-arylpiperazine; EtOH, reflux, 4 h, 30–54% yield.

The target compounds bearing the tetrahydro-1H-pyrrolo[1,2-c]imidazole-1,3(2H)-dione as terminal fragment were prepared according to Scheme 6. The N-alkylation of hydantoin derivative 31 (49) with the appropriate bromoalkylchloride gave the chloroalkyl intermediates 32a,b, which were condensed with 1-[2-(4-methoxyphenyl)phenyl]piperazine to afford the desired compounds 33a,b, respectively.

Scheme 6

Scheme 6. Formation of Target Compounds Bearing the Tetrahydro-1H-pyrrolo[1,2-c]imidazole-1,3(2H)-dione as Terminal Fragmenta

aReagents and conditions: (A) NaH, Br–(CH2)n–Cl, anhydrous DMF, rt, 12 h, 55–75% yield; (B) 1-[2-(4-methoxyphenyl)phenyl]piperazine; K2CO3, acetonitrile, reflux overnight, 43–74% yield.

Binding Affinities to 5-HT7, 5-HT1A, 5-HT2A, and Dopamine D2 Receptors

All the final compounds were tested in radioligand binding assays to determine their affinity for 5-HT7R, 5-HT1AR, and 5-HT2AR (Table 2). In addition, the compounds were counter screened at dopamine D2 receptors, because blockade of this receptor may cause motor dysfunctions. (50) The assays were performed via the displacement of the specific binding of [3H]-5-CT (for 5-HT7R), [3H]-8-OH-DPAT (for 5-HT1AR), [3H]ketanserin (for 5-HT2AR), and [3H]spiperone (for dopamine D2 receptor), at the cloned human receptors stably expressed in HEK293 cells (5-HT7R, 5-HT1AR) or CHO-K1 cells (5-HT2AR, D2 receptor).
The Ki values at 5-HT7R of the target 1-[2-(4-methoxyphenyl)phenyl]piperazine derivatives 8a,b, 11a,b, 20ac, 26ac, 29, and 33a,b are in the range 6.69–91.7 nM, indicating that the structural variations introduced to the terminal fragment or linker of LP-211 were well tolerated. In fact, such variations translate into small changes in affinity, the largest variation being in the case of 33a and 33b (7-fold). Of note, the 5-HT7R affinity values of the reference compounds brilaroxazine (vs 26ac, 29), UCM-2550 (vs 33a,b), risperidone (vs 11a,b), and MMP (vs 8ac) fell in the same range. Instead, the 1-(2-acetylphenyl)piperazine derivatives 9 and 30 were 50- and 11-fold less potent than the reference compound 1 at 5-HT7R.
As for the 5-HT1AR affinities, the 1-(2-acetylphenyl)piperazine derivatives 9 and 30 were equipotent potent to the reference compound 1, whereas the target 1-[2-(4-methoxyphenyl)phenyl]piperazine derivatives 8a,b, 11a,b, 20ac, 26ac, 29, and 33a,b showed Ki values distributed in a wide range (3.77–1802 nM), as the result of a more pronounced impact of the linker length. In fact, the 4-methyl-1,2,4-triazine-3,5(2H,4H)-dione derivative 8c displays Ki = 3.77 nM, whereas its shorter homologue 8a shows a 456-fold lower 5-HT1AR affinity. Similarly, the benzoxazinone derivative 26b displays Ki = 23.2 nM, whereas its shorter homologue 26a shows 78-fold lower affinity. The comparison of the close analogs 26ac, 29, and 34 provides an interesting example of how the affinity for 5-HT7R and 5-HT1AR can be fine-tuned by the nature of the terminal fragment and the linker. It can be noted that the target compounds 26ac, 29 (vs brilaroxazine), 33a,b (vs UCM-2550), 8ac (vs MMP), and 20a–c (vs 5) display lower 5-HT1AR affinity than that of the reference compounds featuring the corresponding terminal fragment. Nonetheless, the proposed structural changes led to the identification of six compounds (8c, 11a, 20b, 26b,c, and 33b) with 5-HT1AR affinity higher than that of LP-211.
Considering the affinity at 5-HT2AR, all the target 1-[2-(4-methoxyphenyl)phenyl]piperazine derivatives show affinity higher than that of LP-211, except 8a and 33a, confirming the validity of the selection of terminal fragments present in the reference compounds. Compounds 8a and 33a represent an unfavorable combination of linker length and terminal fragment. The most pronounced increase was in the case of compound 11b in which the terminal fragment is the same as in risperidone. The 5-HT2AR affinity of the final 1-(2-acetylphenyl)piperazine derivatives 9 and 30 was in the micromolar range, which was similar to the affinity value of the corresponding reference compound 1.
The D2 receptor affinities of the target 1-[2-(4-methoxyphenyl)phenyl]piperazine derivatives were all lower than that of LP-211, but 26c that displayed a Ki value lower than 100 nM. Most importantly, the reference compounds risperidone (vs 11a,b), 5 (vs 20ac), brilaroxazine (vs 26ac, 29), and UCM-2550 (vs 33a,b) show higher D2 receptor affinity than that of the target compounds featuring the corresponding terminal fragment. Thus, the affinity of the target compounds at D2 receptors seems to be determined by the 1-[2-(4-methoxyphenyl)phenyl]piperazine moiety rather than the linker length and the terminal fragment. As for the 1-(2-acetylphenyl)piperazine derivatives 9 and 30, both compounds showed high D2 receptor affinity, being the most potent D2 receptor ligands among the newly synthesized compounds.
Next, in order to select dual 5-HT7R/5-HT1AR or mixed 5-HT7R/5-HT1AR/5-HT2AR ligands, we analyzed the 5-HT1AR/5-HT7R, 5-HT2AR/5-HT7R, and 5-HT2AR/5-HT1AR Ki ratios (Table 2). As reference values, we selected the Ki ratios of compound 26c which, according to our recent study, behaved as a 5-HT7R-preferring agonist in vitro and in vivo. (51) Consequently, to select the dual 5-HT1AR/5-HT7R ligands, we considered compounds showing a 5-HT1AR/5-HT7R Ki ratio lower than 9 (and greater than 0.11, i.e., the reciprocal of 9). Compounds 8c, 29, and 30 displayed such characteristic. These compounds were also selective over 5-HT2AR. The most balanced 5-HT1AR/5-HT7R ligands of this set were 8c and 29, as they showed the 5-HT7R/5-HT1AR Ki ratios closest to 1. On the other hand, compounds 20b, 20c, 26b, and 34 displayed mixed 5-HT7R/5-HT1AR/5-HT2AR affinity, with 20b being the compound with the most balanced affinity profile (all three Ki ratios close to 1).

In Vitro Metabolic Stability

The aim of this study is to provide the scientific community with molecules suitable for studies in vivo. In order to predict the extent of first-pass oxidative metabolism, the target compounds were incubated with rat liver microsomes in the presence of an NADPH regenerating system. (41) In the initial screening phase, we assessed the percentage of the parent compound recovered after 30 min of incubation. The percentages of recovery of LP-211 and TP-22 were 20 and 27%, respectively, (32) which represented the reference values to compare the new compounds. As it can be seen in Table 1, the majority of the new compounds show in vitro stability higher than LP-211, as 11 out of 17 compounds display a percentage of recovery >20%. Then, taking into account the affinity, selectivity, and metabolic stability data, we assessed the half-life and the intrinsic clearance in vitro of compounds 8c, 20b,c, 26a,b, and 29 (Table 3). The data indicate that all the selected compounds showed higher stability than that of LP-211, with intrinsic clearance values lower up to 5-fold as in the case of compound 29. Thus, the compounds listed in Table 3 are predicted to be low-clearance compounds and suitable for studies in vivo. (52) These results provide further support to the strategy of using structural motifs featured by druglike compounds to obtain metabolically stable compounds. (40,51)
Table 3. Half-Life (t1/2) and Intrinsic Clearance (CLint) of Selected Compounds
compoundt1/2 (min)CLint (μL/mg/min)
LP-211 (32)1545.9
TP-22 (32)4516.1
8c4116.9
20b3917.7
20c2330
26a4914.1
26b6011.5
26c (51)6311
29749.4
34 (51)5812
The metabolic stability and the affinity data supported the selection of the dual 5-HT7R/5-HT1AR ligands 8c and 29 and the mixed 5-HT7R/5-HT1AR/5-HT2AR ligand 20b for further evaluations. These compounds distinguished themselves from the reference compounds for the binding profile at 5-HT7, 5-HT1A, 5-HT2A, and D2 receptors. As shown in Table 1, most of the reference compounds display an affinity for α1A adrenoceptor, and therefore, it was not unexpected that compounds 8c, 20b, and 29 showed α1A adrenoceptor affinity (Ki= 23.5, 66.1, and 55.8 nM, respectively, see Supporting Information). In fact, the search for arylpiperazine derivatives with affinity for multiple 5-HTRs might imply that the compounds have an affinity for other monoamine receptors (see Supporting Information for off-target affinities of compounds 8c, 20b, and 29). Considering that α1A adrenoceptor activity might cause cardiovascular side-effect, this particular off-target activity is a safety warning for future developments of this class of compounds.

Functional Activities at 5-HT7R, 5-HT1AR, and 5-HT2AR of Compounds 8c, 20b, and 29

To provide a functional analysis at 5-HT7R, the cAMP response mediated by 8c, 20b, 29 and the reference 5-HT7R agonists 5-CT and LP-211 (29,53) was analyzed. To this end, 5-HT7R-mCherry was coexpressed with the FRET-based cAMP biosensor CEPAC. (54) This biosensor includes the cAMP-binding domain of the EPAC protein cloned between mCerulean (FRET donor) and Citrine (FRET acceptor). Upon cAMP binding, conformational changes of the sensor occur, leading to a decrease in the FRET signal (Figure 1A). The cAMP responses were recorded at the single-cell level by monitoring the CEPAC fluorescence intensity ratio of the acceptor to the donor (A/D ratio). The strength and speed of serotonergic signaling was determined from the amplitude and time dependence of the CEPAC fluorescence intensity ratio.

Figure 1

Figure 1. Compounds 8c, 20b, and 29 stimulate 5-HT7R-mediated cAMP production. (A) N1E cells were transfected with cAMP FRET-based biosensor CEPAC and 5-HT7R-mCherry. Cells were stimulated with the compounds, as indicated. Mean values of the cAMP-biosensor response upon stimulation with 8c 20b, and 29 are shown. Stimulation with LP-211 and 5-CT was used as a control. (B) Quantification of the response amplitude and (C) response time shown as the mean ± SEM (3 < N < 6, in each experiment at least 20 cells were analyzed).

In the absence of 5-HT7R, no cAMP response was observed upon treatment with the ligands (data not shown). In contrast, in cells expressing 5-HT7R, all the compounds (10 μM) were able to increase the intracellular cAMP level, although with different efficiencies. Statistical analysis by fitting the experimental data to the single exponential revealed that 8c and 29 elicited the largest cAMP response amplitude compared with that of 5-CT, followed by 20b (Figure 1A). Of note, response amplitude for all compounds tested was higher than that measured for highly selective 5-HT7R agonist LP-211 (Figure 1B). The mean response times for all compounds were higher (i.e., slower response kinetics) than that obtained for 5-CT and comparable with the values obtained for LP-211 (Figure 1C).
We next analyzed whether 8c, 20b, and 29 would affect the 5-HT1AR function toward cAMP inhibition. The 5-HT1AR agonist 5-CT was used as a positive control, (55) while LP-211 (selective 5-HT7R agonist) was used as a negative control. To this end, 5-HT1AR-mCherry was coexpressed with the CEPAC biosensor in N1E cells. We subsequently analyzed the ability of 5-HT1AR-mediated signaling via Gi to inhibit the forskolin (FSK)-induced cAMP accumulation following receptor stimulation with above-mentioned compounds (10 μM). Except for LP-211 treatment, an increase of the A/D ratio of CEPAC was observed in all the cases, that indicated the 5-HT1AR-mediated downregulation of intracellular concentration of cAMP (Figure 2). Statistical evaluation of amplitudes of the cAMP decay for 8c, 20b, and 29 revealed that, although these compounds were still able to evoke receptor-mediated decrease of the concentration of cAMP, they were less effective in activation of 5-HT1AR when compared with 5-CT (Figure 2B).

Figure 2

Figure 2. Compounds 8c, 20b, and 29 behave as 5-HT1AR agonists in the receptor-mediated cAMP inhibition. (A) N1E cells were transfected with cAMP FRET-based biosensor CEPAC and 5-HT1AR-mCherry. After pretreatment with 1 μM forskolin and 25 μM IBMX, cells were stimulated with the indicated compounds. Each trace shows cAMP response at the single cell. (B) Graphs show changes of cAMP response amplitude relative to pretreatment (mean ± SEM, 3 < N < 6, in each experiment at least 20 cells were analyzed).

Finally, compounds 8c, 20b, and 29 were investigated in functional assays of 5-HT2AR-mediated inositol phosphate (IP) signaling in CHO-K1 cells expressing the cloned human receptor. The three compounds, in concentration–response curves from 1 nM to 100 μM, fully antagonized in a concentration-dependent manner the stimulation of IP production elicited by 1 μM 5-HT (Figure 3). Consistently, no agonist activity was observed for 20b at the same concentrations in these assays (data not shown). IC50 values were similar for the three compounds and in the nanomolar range, consistent with their affinities at 5-HT2AR (IC50 = 595, 666, and 491 nM for 8c, 20b, and 29, respectively). The reference 5-HT2AR antagonist risperidone (0.01 nM–10 μM) showed a IC50 value of 0.81 nM (Figure 3).

Figure 3

Figure 3. Functional assays of inositol phosphate (IP) signaling at cloned human 5-HT2ARs. Concentration–response inhibition curves of 8c, 20b, 29, and risperidone (as reference 5-HT2AR antagonist) on IP production stimulated by 1 μM 5-HT in CHO-K1 cells expressing human 5-HT2ARs. The graph shows data (mean ± SEM) from one experiment performed in duplicate.

Collectively, the functional activities of the selected compounds supported the validity of the design approach. In fact, compounds 8c, 20b, and 29 behave as 5-HT7R agonists in the same way as LP-211–confirming that the 1-(biphenyl)piperazine moiety is a key structural determinant for 5-HT7R agonism and as 5-HT2AR antagonists as various other arylpiperazine derivatives. As for 5-HT1AR, the comparison of the functional activities of 8c, 20b, and 29 with LP-211 suggest that the 1-(biphenyl)piperazine moiety alone is not responsible for the functional activity at 5-HT1AR, which, instead, is determined also by the nature of the terminal fragment and linker length, as already noted in the Study Design paragraph.

Conclusions

ARTICLE SECTIONS
Jump To

5-HT neurotransmission system is an active area of investigation in ASD research. Several in vitro and in vivo studies with selective 5-HTR agonists or antagonists have suggested that targeting a subpopulation of the 5-HTRs might alleviate the core symptoms of ASD. Based on the current knowledge, we aimed at identifying a dual 5-HT7R/5-HT1AR agonist and a mixed 5-HT7R/5-HT1AR agonist/5-HT2AR antagonist. A set of novel arylpiperazine derivatives were designed by exploiting structural motifs that might drive the functional activity of the target compounds toward the desired profile (knowledge-based design). The design strategy succeeded as we identified compounds 8c and 29 that are 5-HT7R and 5-HT1AR preferring agonists and compound 20b, a mixed 5-HT7R/5-HT1AR agonist/5-HT2AR antagonist with almost identical affinity for the three receptors. The knowledge-based design strategy had a favorable influence on the in vitro pharmacokinetic properties of most of the newly designed compounds. In fact, 8c, 20b, and 29 are metabolically stable in vitro and also have suitable CNS druglike properties. Considering the complex mechanisms underlying ASD, we believe that a polypharmacology approach might be more suited than a single target approach. We hope that pharmacological tools such as 8c, 20b, and 29 will contribute to the progress of the discovery of drugs for ASD.

Methods

ARTICLE SECTIONS
Jump To

Chemistry

Chemicals were purchased from Sigma-Aldrich, Alfa Aesar, TCI Chemicals. Unless otherwise stated, all chemicals were used without further purification. Thin layer chromatography (TLC) was performed using plates from Merck (silica gel 60 F254). Column chromatography was performed with 1:30 Merck silica gel 60 Å (63–200 μm) as the stationary phase. Flash chromatographic separations were performed on a Biotage SP1 purification system using flash cartridges prepacked with KP-Sil 32–63 μm, 60 Å silica. 1H NMR spectra were recorded on a Varian Mercury-VX spectrometer (300 MHz) or on a 500-vnmrs500 Agilent spectrometer (500 MHz). All chemical shift values are reported in parts per million (ppm, δ). Splitting patterns are designated as follows: app (apparent), br (broad), s (singlet), d (doublet), t (triplet), q (quartet), dd (doublet of doublets), td (triplet of doublets). For target compounds, NMR spectra were recorded on free bases. Recording of mass spectra was done on an HP6890-5973 MSD gas chromatograph/mass spectrometer; only significant m/z peaks, with their percentage of relative intensity in parentheses, are reported. ESI-MS/MS analyses were performed with an Agilent 1100 Series LC-MSD trap System VL workstation, mass range 50–800 m/z, electrospray ion source in positive or negative ion mode. All spectra were in accordance with the assigned structures. Elemental analysis (C,H,N) of the target compounds as hydrochloride salts were performed on a Eurovector Euro EA 3000 analyzer. Analyses indicated by the symbols of the elements were within ±0.4% of the theoretical values. The purity of the target compounds listed in Table 2 was assessed by RP-HPLC and combustion analysis. All compounds showed ≥95% purity. RP-HPLC analysis was performed on an Agilent 1260 Infinity Binary LC System equipped with a diode array detector using a Phenomenex Gemini C-18 column (250 mm × 4.6 mm, 5 μm particle size). All target compounds (Table 2) were eluted with CH3CN/ammonium formate 50 mM pH 5, 8:2 (v/v) at a flow rate of 1 mL/min. All compounds showed ≥95% purity.
The following compounds were prepared as described in the literature: 1-[2-(4-methoxyphenyl)phenyl]piperazine; (46) 1-(2-(piperazin-1-yl)phenyl)ethanone; (56) 4-methyl-1,2,4-triazine-3,5(2H,4H)-dione (6); (43) 2-(4-chlorobutyl)-4-methyl-1,2,4-triazine-3,5(2H,4H)-dione (7c); (43) 3-(2-chloroethyl)-2-methyl-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidin-4-one (10); (44) 4′-methoxy-[1,1′-biphenyl]-2-amine (14); (46) 3-(2-bromoethoxy)phenol (17); (45) 7-hydroxy-4-methyl-chromen-2-one (18); (47) 4-(benzyloxy)-2-nitrophenol (21); (48) 6-[3-[4-[2-(4-methoxyphenyl)phenyl]piperazin-1-yl]butoxy]-2-methyl-2H-benzo[b][1,4]oxazin-3(4H)-one (26c); (51) 6-hydroxy-2-methyl-2H-benzo[b][1,4]oxazin-3(4H)-one (27); (49) tetrahydro-1H-pyrrolo[1,2-c]imidazole-1,3(2H)-dione (31); (50) (R)-1-(4-chloro-2-fluorophenoxy)-3-[4-[2-(4-methoxyphenyl)phenyl]piperazin-1-yl]propan-2-ol (34). (51) Complete synthetic procedures and intermediated spectroscopic data are fully reported in the Supporting Information.

General Procedure for the Preparation of Target Compounds 8a–c, 9, 11a,b, 20ac, 26a,b, and 33a,b

A stirred mixture of the appropriate alkylating agent (0.7 mmol), 1-[2-(4-methoxyphenyl)phenyl]piperazine or 1(2-acetylphenyl)piperazine (0.84 mmol), and K2CO3 (0.1 g, 0.7 mmol) in acetonitrile (20 mL) was refluxed overnight. After cooling, the mixture was evaporated to dryness, and H2O (20 mL) was added to the residue. The aqueous phase was extracted with AcOEt (2 × 30 mL). The collected organic layers were dried over Na2SO4 and evaporated under reduced pressure. The crude residue was purified by chromatographic column as detailed below to afford pure target compound.

2-{4-[4-[2-(4-Methoxyphenyl)phenyl]piperazin-1-yl]ethyl}-4-methyl-1,2,4-triazine-3,5(2H,4H)-dione (8a)

Eluted with CHCl3/MeOH, 98:2. Yellow oil, 73% yield. 1H NMR (CDCl3): δ 2.43 (br s, 4H), 2.67 (t, 2H, J = 6.6 Hz), 2.80 (br s, 4H), 3.32 (s, 3H), 3.85 (s, 3H), 4.08 (t, 2H, J = 6.6 Hz), 6.91–6.93 (m, 2H), 7.00 (d, 1H, J = 7.8 Hz), 7.04 (td, 1H, J = 1.1 and 7.6 Hz), 7.21 (dd, 1H, J = 1.5 and 7.3 Hz), 7.26 (m, 1H), 7.37 (s, 1H), 7.55–7.57 (m, 2H). GC/MS m/z 422 (M+, 4), 421 (M+, 16), 281 (100). Anal. (C23H27N5O3·HCl·H2O) C, H, N.

2-{3-[4-[2-(4-Methoxyphenyl)phenyl]piperazin-1-yl]propyl}-4-methyl-1,2,4-triazine-3,5(2H,4H)-dione (8b)

Eluted with CHCl3/EtOAc, 1:1. Brown oil, 40% yield. 1H NMR (CDCl3): δ 1.88–1.92 (m, 2H), 2.34 (br s, 4H), 2.40 (t, 2H, J = 7.0 Hz), 2.80 (br s, 4H), 3.32 (s, 3H), 3.85 (s, 3H), 4.03 (t, 2H, J = 7.0 Hz), 6.92 (d, 2H, J = 8.8 Hz), 6.98–7.05 (m, 2H), 7.02–7.07 (m, 1H), 7.19–7.22 (m, 1H), 7.36 (s, 1H), 7.56 (d, 2H, J = 8.8 Hz). GC/MS m/z 436 (M+ + 1, 20), 435 (M+, 100), 281 (70), 212 (57), 167 (34). Anal. (C24H29N5O3·HCl·H2O) C, H, N.

2-{2-[4-[2-(4-Methoxyphenyl)phenylpiperazin-1-yl]butyl}-4-methyl-1,2,4-triazine-3,5(2H,4H)-dione (8c)

Eluted with CHCl3/MeOH, 95:5. Pale yellow oil, 30% yield. 1H NMR (CDCl3): δ 1.47 (m, 2H), 1.71–1.80 (m, 2H), 2.34 (app t, 6H), 2.85 (app t, 4H), 3.33 (s, 3H), 3.98 (t, 2H, J = 7.0 Hz), 6.91–6.95 (m, 2H), 7.00–7.07 (m, 2H), 7.29–7.24 (m, 2H), 7.37 (s, 1H), 7.55–7.59 (m, 2H). 13C NMR (500 MHz, CDCl3): δ 158.7; 155.9; 148.9; 147.7; 134.9; 134.3; 132.7; 131.5; 129.9; 128.36; 124.3; 118.9; 113.8; 124.2; 118.7; 113.8; 55.3; 52.4; 50.5; 47.7; 27.1. ESI-MS m/z 472 (M + Na)+. ESI-MS/MS m/z 323 (100). Anal. (C25H31N5O3·HCl) C, H, N.

2-[4-[4-(2-Acetylphenyl)piperazin-1-yl]butyl]-4-methyl-1,2,4-triazine-3,5(2H,4H)-dione (9)

Eluted with CHCl3/MeOH, 98:2. Yellow oil, 20% yield. 1H NMR (CDCl3): δ 1.57 (m, 2H), 1.81 (m, 2H), 2.44 (t, 2H, J = 7.6 Hz), 2.60 (br s, 4H), 2.65 (s, 3H), 3.02 (app. t, 4H), 3.35 (s, 3H), 4.02 (t, 2H, J = 7.1 Hz), 7.04–7.08 (m, 2H), 7.39–7.42 (m, 3H). GC/MS m/z 386 (M+ + 1, 10), 385 (M+, 30), 237 (90), 207 (100), 161 (95). Anal. (C20H27N5O3·2HCl) C, H, N.

3-{2-[4-[2-(4-Methoxyphenyl)phenyl]piperazin-1-yl]ethyl}-2-methyl-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidin-4-one (11a)

Eluted with CHCl3/MeOH, 98:2. Brown solid, 60% yield. 1H NMR (CDCl3): δ 1.84–1.89 (m, 2H), 1.92–1.97 (m, 2H), 2.27 (s, 3H), 2.45–2.50 (m, 6H), 2.69–2.72 (br t, 2H), 2.85 (t, 2H, J = 6.8 Hz), 2.89 (br s, 4H), 3.85 (s, 3H), 3.91 (t, 2H, J = 6.1 Hz), 6.90–6.93 (m, 2H), 7.03–7.06 (m, 2H), 7.21 (dd, 1H, J = 1.5 and 7.3 Hz), 7.25 (td, 1H, J = 1.5 and 7.8 Hz), 7.55–7.58 (m, 2H). GC/MS m/z 458 (M+, 2), 281 (100). Anal. (C28H34N4O2·2HCl) C, H, N.

3-{3-[4-[2-(4-Methoxyphenyl)phenyl]piperazin-1-yl]propyl}-2-methyl-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidin-4-one (11b)

Eluted with CHCl3/EtOAc, 1:1. Brown oil, 60% yield. 1H NMR (CDCl3): δ 1.51–1.53 (m, 2H), 1.84–1.89 (m, 2H), 1.93–1.98 (m, 2H), 2.27 (s, 3H), 2.45–2.50 (m, 6H), 2.69–2.73 (br t, 2H), 2.85 (t, 2H, J = 6.8 Hz), 2.85 (br s, 4H), 3.85 (s, 3H), 3.91 (t, 2H, J = 6.1 Hz), 6.90–6.93 (m, 2H), 7.03–7.06 (m, 2H), 7.21 (dd, 1H, J = 1.5 and 7.3 Hz), 7.25 (td, 1H, J = 1.5 and 7.8 Hz), 7.55–7.58 (m, 2H). 13C NMR (500 MHz, CDCl3): δ 162.6; 158.4; 155.6; 134.5; 133.5; 131.3; 129.8; 129.7; 127.9; 122.6; 118.2; 113.5; 58.13; 55.2; 53.2; 50.6; 42.7; 31.4; 24.1; 22.0; 21.21; 19.25. GC/MS m/z 473 (M+ + 1, 2), 472 (M+, 9), 281 (42), 234 (100), 205 (51). Anal. (C29H36N4O2·2HCl·H2O) C, H, N.

7-[2-[4-[2-(4-Methoxyphenyl)phenyl]piperazin-1-yl]ethoxy]-4-methyl-2H-chromen-2-one (20a)

Eluted with CHCl3/EtOAc, 1:1. Pale yellow oil, 71% yield. 1H NMR (CDCl3): δ 2.39 (s, 3H), 2.54 (br s, 4H), 2.84 (br t, 2H), 2.91 (br s, 4H), 3.85 (s, 3H), 4.15 (br t, 2H), 6.13 (d, 1H, J = 1.5 Hz), 6.80 (d, 1H, J = 2.5 Hz), 6.86 (dd, 1H, J = 2.5 and 8.8 Hz), 6.92–6.95 (m, 2H), 7.03–7.06 (m, 2H), 7.22 (dd, 1H, J = 1.5 and 7.3 Hz), 7.24–7.27 (m, 1H), 7.48 (d, 1H, J = 8.8 Hz), 7.56–7.59 (m, 2H). ESI-MS m/z 493 (M + Na)+. ESI-MS/MS m/z 493 (89), 295 (100). Anal. (C29H30N2O4·HCl) C, H, N.

6-[3-[4-[2-(4-Methoxyphenyl)phenyl]piperazin-1-yl]propoxy]-4-methyl-2H-chromen-2-one (20b)

Eluted with CH2Cl2/EtOAc, 1:1. Transparent oil, 15% yield. 1H NMR (CDCl3): δ 1.94–1.99 (m, 2H), 2.39 (br s, 7H), 2.50–2.52 (br t, 2H), 2.86 (br s, 4H), 3.85 (s, 3H), 4.03–4.07 (br t, 2H), 6.12 (s, 1H), 6.80–6.85 (m, 2H), 6.91–6.94 (m, 2H), 7.01–7.27 (m, 2H), 7.20–7.27 (m, 3H), 7.47 (d, 1H, J = 8.2 Hz), 7.56–7.59 (m, 2H). 13C NMR (500 MHz, CDCl3): δ 162.2; 161.5; 158.5; 155.4; 152.7; 134.7; 133.7; 131.5; 130.0; 128.1; 125.6; 122.8; 118.3; 113.7; 112.7; 112.0; 101.6; 66.9; 55.4; 55.1; 53.6; 50.9; 26.6; 18.8. ESI-MS m/z 507 (M+Na)+. ESI-MS/MS m/z 507 (100), 309 (63). Anal. (C30H32N2O4·HCl) C, H, N.

7-[4-[4-[2-(4-Methoxyphenyl)phenyl]piperazin-1-yl]butoxy]-4-methyl-2H-1-chromen-2-one (20c)

Eluted with CHCl3/MeOH, 98:2. Pale yellow oil, 36% yield. 1H NMR (CDCl3): δ 1.65–1.72 (m, 2H), 1.85–1.94 (m, 2H,) 2.38–2.43 (m + d, 9H, J = 1.1 Hz), 2.85 (app t, 4H), 3.86 (s, 3H), 4.14 (t, 2H, J = 6.0 Hz), 6.17 (d, 1H, J = 1.1 Hz), 6.87–6.95 (m, 3H), 7.00–7.08 (m, 2H), 7.20–7.23 (m, 3H), 7.43 (d, 1H, J = 8.8 Hz), 7.55–7.60 (m, 2H). ESI-MS m/z 497 (M + H)+. ESI-MS/MS m/z 497 (69), 335 (100). Anal. (C31H34N2O4·HCl·H2O) C, H, N.

6-[2-[4-[2-(4-Methoxyphenyl)phenyl]piperazin-1-yl]ethoxy]-2-methyl-2H-benzo[b][1,4]oxazin-3(4H)-one (26a)

Eluted with CHCl3/MeOH, 98:2. Transparent oil, 38% yield. 1H NMR (CDCl3): δ 1.55 (d, 3H, J = 6.9 Hz), 2.53 (br s, 4H), 2.79–2.81 (m, 2H), 2.89 (br t, 4 H), 3.86 (s, 3H), 4.04 (app t, 2H), 4.57 (q, 1H, J = 6.9 Hz), 6.36 (d, 1H, J = 2.9 Hz), 6.49 (dd, 1H, J = 2.9 and 8.8 Hz), 6.87 (d, 1H, J = 8.8 Hz), 6.92–6.95 (m, 2H), 7.01–7.07 (m, 2H), 7.21–7.27 (m, 2H), 7.55–7.58 (m, 2H), 8.27 (s, 1H, D2O exchanged). ESI-MS m/z 474 (M + H)+. ESI-MS/MS m/z 474 (76), 226 (100). Anal. (C28H31N3O4·2HCl) C, H, N.

6-[3-[4-[2-(4-Methoxyphenyl)phenyl]piperazin-1-yl]propoxy]-2-methyl-2H-benzo[b][1,4]oxazin-3(4H)-one (26b)

Eluted with CHCl3/MeOH, 98:2. Transparent oil, 17% yield. 1H NMR (CDCl3): δ 1.55 (d, 3H, J = 2.5 Hz), 1.92–1.95 (m, 2H), 2.43 (br s, 4H), 2.51 (app t, 2H), 2.88 (br s, 4 H), 3.85 (s, 3H), 3.93 (t, 2H, J = 6.4 Hz), 4.57 (q, 1H, J = 6.9 Hz), 6.34 (d, 1H, J = 2.5 Hz), 6.49 (dd, 1H, J = 2.5 and 8.8 Hz), 6.86 (d, 1H, J = 8.8 Hz), 6.92–6.94 (m, 2H), 7.01–7.07 (m, 2H), 7.21–7.27 (m, 2H), 7.55–7.56 (m, 2H), 8.13 (s, 1H, D2O exchanged). GC/MS m/z 488 (M+ + 1, 5), 487 (M+, 20), 281 (30), 194 (100), 165 (33), 91 (34). Anal. (C29H33N3O4·2HCl) C, H, N.

2-[2-[4-[2-(4-Methoxyphenyl)phenyl]piperazin-1-yl]ethyl]tetrahydro-1H-pyrrolo[1,2-c]imidazole-1,3(2H)-dione (33a)

Eluted with CHCl3/EtOAc, 1:1. Transparent oil, 74% yield. 1H NMR (CDCl3): δ 1.64–1.74 (m, 1H), 1.98–2.07 (m, 2H), 2.17–2.25 (m, 1H), 2.40 (br s, 4H), 2.49–2.58 (m, 2H), 2.78 (br s, 4H), 3.19–3.24 (m, 1H), 3.53–3.59 (m, 2H), 3.64–3.69 (m, 1H), 3.85 (s, 3H), 4.05 (t, 1H, J = 8.3 Hz), 6.90–6.93 (m, 2H), 7.04 (td, 1H, J = 1.0 and 7.4 Hz), 7.19–7.25 (m, 2H), 7.54–7.56 (m, 2H). 13C NMR (500 MHz, CDCl3): δ 174.2; 161.1; 158.5; 150.3; 134.7; 133.7; 131.4; 129.9; 127.9; 122.7; 118.3; 63.4; 55.7; 54.9; 53.6; 53.2; 51.1; 45.9; 36.2; 27.8; 27.0. GC/MS m/z 435 (M+ + 1, 3), 434 (M+, 13), 281 (100). Anal. (C25H30N4O3·HCl·H2O) C, H, N.

2-[3-[4-[2-(4-Methoxyphenyl)phenyl]piperazin-1-yl]propyl]tetrahydro-1H-pyrrolo[1,2-c]imidazole-1,3(2H)-dione (33b)

Eluted with CHCl3/EtOAc, 1:1. Brown oil, 43% yield. 1H NMR (CDCl3): δ 1.64–1.71 (m, 2H), 1.72–1.82 (m, 2H), 2.02–2.11 (m, 2H), 2.18–2.28 (m, 2H), 2.34–2.38 (m, 5H), 2.85 (br s, 4H), 3.19–3.27 (m, 1H), 3.48–3.52 (m, 2H), 3.62–3.70 (m, 2H), 3.85 (s, 3H), 4.02–4.07 (m, 1H), 6.92 (d, 2H, J = 8.88 Hz), 6.99–7.07 (m, 2H), 7.19–7.27 (m, 2H), 7.55 (d, 2H, J = 8.8 Hz). GC/MS m/z 449 (M+ + 1, 20), 448 (M+, 80), 281 (100), 210 (50), 70 (31). Anal. (C26H32N4O3·HCl·H2O) C, H, N.

General Procedure for the Preparation of Compounds 29 and 30

A mixture of 1-[2-(4-methoxyphenyl)phenyl]piperazine or 1(2-acetylphenyl)piperazine (1.2 mmol) and the oxirane 28 (1.0 mmol) in ethanol (20 mL) was refluxed for 5 h. After it was cooled, the solvent was removed in vacuo, and the crude residue was chromatographed as detailed below to give desired pure compound.

6-{(2R)-2-Hydroxy-3-[4-[2-(4-methoxyphenyl)phenyl]piperazin-1-yl]propoxy}-2-methyl-2H-benzo[b][1,4]oxazin-3(4H)-one (29)

Eluted with CHCl3/AcOEt, 1:1. White semisolid, 30% yield. 1H NMR (CDCl3): δ 1.62 (br s, 1H, D2O exchanged), 1.47 (d, 3H, J = 6.8 Hz), 2.30–2.32 (m, 2H,), 2.39–2.45 (m, 2H), 2.48–2.54 (m, 2H), 2.79–2.83 (m, 4H), 3.78 (s, 3H), 3.82 (d, 2H, J = 4.9 Hz), 3.94–3.97 (m, 1H), 4.49 (q, 1H, J = 6.8 Hz), 6.79 (d, 1H, J = 8.8 Hz), 6.84–6.87 (m, 2H), 6.94 (dd, 1H, J = 1.1 and 8.3 Hz), 6.99 (td, 1H, J = 1.5 and 7.3 Hz), 7.14–7.20 (m, 4H), 7.48–7.51 (m, 2H), 8.35 (br, 1H, D2O exchanged). 13C NMR (500 MHz, CDCl3): δ 168.0; 167.9; 158.7; 153.4; 147.8; 137.6; 137.5; 134.9; 132.6; 131.5; 129.8; 128.2; 127.5; 124.2; 118.7; 117.2; 114.0; 109.9; 102.6; 73.2; 70.4; 64.6; 61.0; 55.4; 54.7; 53.4; 47.8; 47.7; 16.0. ESI-MS m/z 526 (M+Na)+. ESI-MS/MS m/z 526 (100), 347 (11). Anal. (C29H33N3O5·HCl) C, H, N.

6-[3-[4-(2-Acetylphenyl)piperazin-1-yl]-(2R)-2-hydroxypropoxy]-2-methyl-2H-benzo[b][1,4]oxazin-3(4H)-one (30)

Eluted with CHCl3/MeOH, 95:5. Brown oil, 54% yield. 1H NMR (CDCl3): δ 1.50 (d, 3H, J = 6.9 Hz), 1.81 (br s, 1H, D2O exchanged), 2.56–2.64 (m, 4H), 2.65 (s, 3H), 2.78–2.83 (m, 2H), 3.05 (br, 4 H), 3.94–3.96 (m, 2H), 4.09–4.11 (m, 1H), 4.57 (q, 1H, 6.9 Hz), 6.44 (s, 1H), 6.52 (dd, 1H, J = 2.4 and 8.8 Hz), 6.87 (d, 1H, J = 8.8 Hz), 7.05–7.08 (m, 2H), 7.39–7.42 (m, 2H), 8.49 (br s, 1H, D2O exchanged). ESI-MS m/z 462 (M + Na)+. ESI-MS/MS m/z 462 (100). Anal. (C24H29N3O5·2HCl) C, H, N.

Radioligand Binding Assays

Materials

Cell culture reagents were purchased from EuroClone (Milan, Italy). G418 (Geneticin), 5-HT, and NAN-190 were obtained from Sigma-Aldrich (Milano, Italy). 5-CT was purchased from Tocris Bioscience (Bristol, UK). [3H]-5-CT and [3H]-8-OH-DPAT were obtained from PerkinElmer Life and Analytical Sciences (Boston, MA, USA). MultiScreen plates with Glass fiber filters was purchased from Merck Millipore (Billerica, MA, USA). pcDNA3.1(+) vector containing the target 5-HT1A DNA sequence was purchased from cDNA Resource Center (Bloomsberg, PA, USA), and FuGENE HD Transfection Reagent was obtained from Promega (Madison, Wisconsin, USA).

Cell Culture

HEK-293 cell line was grown in DMEM high glucose supplemented with 10% fetal bovine serum, 2 mM glutamine, 100 U/mL penicillin, and 100 μg/mL streptomycin, in a humidified incubator at 37 °C with a 5% CO2 atmosphere. HEK-293-5-HT7A and HEK-293-5-HT1A transfected cell lines were grown in DMEM high glucose supplemented with 10% fetal bovine serum, 2 mM glutamine, 100 U/mL penicillin, 100 μg/mL streptomycin, and 0.8 μg/mL G418, in a humidified incubator at 37 °C with a 5% CO2 atmosphere.

Radioligand Binding at Human Cloned 5-HT7Rs

5-HT7R binding was carried out as previously reported. (57) The experiment was performed in MultiScreen plates (Merck Millipore) with Glass fiber filters (GF/C), presoaked in 0.3% PEI for 20 min. After this time, 130 μg of HEK-293-5-HT7A membranes, 1 nM [3H]-5-CT, and the test compounds were suspended in 0.25 mL of incubation buffer (50 mM Tris–HCl, pH 7.4, 4 mM MgCl2, 0.1% ascorbic acid, 10 μM pargyline hydrochloride). The samples were incubated for 60 min at 37 °C. The incubation was stopped by rapid filtration, and the filters were washed with 3 × 0.25 mL of ice-cold buffer (50 mM TRIS-HCl, pH 7.4). Nonspecific binding was determined in the presence of 10 μM 5-CT. Approximately 90% of specific binding was determined under these conditions. Concentrations required to inhibit 50% of radioligand specific binding (IC50) were determined by using six to nine different concentrations of the drug studied in two or three experiments with samples in duplicate. Apparent inhibition constants (Ki) values were determined by nonlinear curve fitting, using the Prism, version 5.0, GraphPad software.

Radioligand Binding at Human Cloned 5-HT1AR

5-HT1AR binding was carried out as already reported. (57) The experiment was performed in MultiScreen plates (Merck Millipore) with Glass fiber filters (GF/C), presoaked in 0.3% PEI for 20 min. After this time, 100 μg of HEK-293-5-HT1A membranes, 1.5 nM [3H]-8-OH-DPAT, and the test compound were suspended in a 0.25 mL of incubation buffer (50 mM Tris–HCl pH 7.4, 4 mM MgCl2, 0.1% ascorbic acid, 0.1 nM EDTA, 10 μM pargyline hydrochloride). The samples were incubated for 60 min at 25 °C. The incubation was stopped by rapid filtration, and the filters were washed with 3 × 0.25 mL of ice-cold buffer (50 mM TRIS-HCl, pH 7.4). Nonspecific binding was determined in the presence of 10 μM NAN-190. Approximately 90% of specific binding was determined under these conditions. Concentrations required to inhibit 50% of radioligand specific binding (IC50) were determined by using six to nine different concentrations of the test compound in two or three experiments with samples in duplicate. Apparent inhibition constants (Ki) values were determined by nonlinear curve fitting, using the Prism, version 5.0, GraphPad software.

Radioligand Binding at Human Cloned Dopamine D2 and Serotonin 5-HT2A Receptors

The affinity of the compounds for dopamine D2 and serotonin 5-HT2A receptors was evaluated in membrane preparations from CHO-K1 cells stably expressing the human cloned D2S receptor or the human cloned 5-HT2A receptor, following previously described procedures. (58) Competition binding experiments were performed using [3H]spiperone (0.2 nM; D2 receptor) or [3H]ketanserin (1 nM; 5-HT2AR) as radioligands. Nonspecific binding was assessed in the presence of 10 μM sulpiride (D2 receptor) or 1 μM methysergide (5-HT2AR). Haloperidol (D2 receptor) and risperidone (5-HT2AR) were included in the assays as reference compounds. Competition binding curves constructed with 6 different concentrations of the compounds were fitted to a one-site competition model using Prism 6 software (GraphPad, San Diego, CA, USA), and equilibrium dissociation constant (Ki) of the compounds was calculated according to the Cheng–Prusoff equation.

Analysis of the cAMP Response Using FRET-Based Biosensor CEPAC

Mouse neuroblastoma N1E115 cells (American Type Culture Collection, Manassas, USA) were seeded onto 18 mm glass coverslips and grown in DMEM containing 10% fetal bovine serum and 1% penicillin/streptomycin at 37 °C in a humidified atmosphere with 5% CO2. Expression of the cAMP-biosensor CEPAC, 5-HT7R, or 5-HT1AR was ensured by transfection (plasmid DNA to biosensor and receptor ratio of 7:3) using Lipofectamine 2000 (Life Technologies). One day after transfection, changes in cAMP levels upon perfusion with 10 μM 8c, 20b, 29, LP-211, or 5-CT 5-HT7R in Tyrode’s buffer (150 mM NaCl; 5 mM KCl; 1 mM MgCl2; 2 mM CaCl2; 10 mM HEPES; pH 7.4; adjusted osmolarity) were monitored in real-time under a Zeiss LSM 780 confocal laser-scanning microscope. A 61 cycle time series with a 10 s interval was recorded in online fingerprinting mode of the ZEN acquisition software with the following settings: image dimension = 1024 pixels × 1024 pixels, resolution = 0.346 μm × 0.346 μm, excitation = 440 nm diode and 561 nm DPSS laser lines, filters = MBS 445, MBS 458/561, objective = C-Apochromat 40×/1.2W Corr. Corresponding reference spectra for the online fingerprinting mode were obtained from separate measurements with a single fluorophore transfection. The semiautomatic biosensor data analysis relied on custom-written MATLAB scripts comprising data import, preprocessing, shift correction in the xy-plane for each time point, the exclusion of saturated pixels from evaluation, background correction, and faint data blurring with a kernel size of 0.5 according to Pawley. (59) The pixel-based ratio was calculated for selected regions of interest (ROIs) for evaluation. Traces were normalized according to their mean ratio before stimulation.

Functional Assays at 5-HT2A Receptor

The efficacy of compounds 8c, 20b, and 29 at 5-HT2A receptor was investigated in assays of inositol phosphate (IP) production in the CHO-K1 cell line stably expressing the cloned human 5-HT2A receptor employed in radioligand binding assays. Cellular IP levels were quantified by using the homogeneous time-resolved fluorescence (HTRF)-based inositol monophosphate kit IP-One Gq kit (Cisbio, Bioassays, Codolet, France) following the manufacturer protocol. Cells were seeded in 96-well plates in culture medium DMEM (Gibco, ThermoFisher Scientific, Madrid, Spain) supplemented with 10% (v/v) dialyzed fetal bovine serum (Sigma-Aldrich, Madrid, Spain), 100 U/mL penicillin/0.1 mg/mL streptomycin (Sigma-Aldrich, Madrid, Spain), and 2 mM l-glutamine (Sigma-Aldrich, Madrid, Spain)) and maintained during 24 h at 37 °C in a 5% CO2 humidified atmosphere. Prior to the assay, cell supernatant was removed and for assessment of possible agonist effect, and cells were incubated with the compounds (0.1 nM–100 μM) or 5-HT (0.1 nM–100 μM) as control agonist in stimulation buffer for 20 min at 37 °C. After this time, IP levels were quantified. For assessment of possible antagonist effect, the compounds (0.1 nM–100 μM) were added to the cells 10 min prior to the addition of 1 μM 5-HT, and assays were subsequently carried out as described above. Risperidone (0.1 nM–100 μM) was used as control antagonist in these assays. In all cases, basal IP levels were determined in control wells in the absence of compound and agonist. Antagonist concentration–response curves were fitted to a sigmoidal dose–response (inhibition) model (Hill slope (nH) = 1, with best fit in comparison to sigmoidal dose–response (variable slope) model, P < 0.05, extra sum-of-squares F test) using Prism 6 software (GraphPad, San Diego, CA) to retrieve pIC50 (−log IC50) values.

Stability Assays in Rat Liver Microsomes

Test compounds were preincubated at 37 °C with rat liver microsomes (Tebu-Bio, Milan, Italy) (1.0 mg/mL microsomal protein) at 10 μM final concentration in 100 mM potassium phosphate buffer (pH 7.4) for 10 min. Metabolic reactions were initiated by the addition of the NADPH regenerating system (containing 10 mM NADP, 50 mM glucose-6-phosphate, and 10 unit/mL glucose-6-phosphate dehydrogenase, final glucose-6-phosphate dehydrogenase concentration, 1 unit/mL). Aliquots were removed at specific time end points and immediately mixed with an equal volume of cold acetonitrile containing the internal standard. To assess in vitro in vitro half-life (t1/2) the aliquots were removed at 0, 5, 15, 30, 60, and 120 min. Test compound incubated with microsomes without NADPH regenerating system was included. Quenched samples were centrifuged at 4500 rpm for 15 min, and the supernatants were injected for quantification analysis. Samples (100 μL) were analyzed by using an Agilent 1260 Infinity Binary LC System equipped with a diode array detector (Open Lab software was used to analyze the chromatographic data) and a Phenomenex Gemini C-18 column (250 mm × 4.6 mm, 5 μm particle size). The samples were eluted using CH3CN/20 mM ammonium formate pH 5.5 (70:30, v/v) as eluent (1 mL/min). Concentrations were quantified by measuring the area under the peak. The percentage of the parent compound remaining after a 30 min incubation has been calculated according to the equation
where Cparent is ligand concentration after incubation with microsome fraction and NADPH regenerating system and Ccontrol is ligand concentration after incubation with microsome fraction only.
The in vitro half-life (t1/2) was calculated using the expression t1/2 = 0.693/b, where b is the slope found in the linear fit of the natural logarithm of the fraction remaining of the parent compound vs incubation time. (41) In vitro half-life was then used to calculate the intrinsic plasma clearance (CLint) according to the following equation:
Internal positive controls were aripiprazole (Cint = 6.93 mL/mg/min, t1/2 = 100 min) and LP-211 (Cint= 45.9 mL/mg/min, t1/2 = 45.9 min).

Supporting Information

ARTICLE SECTIONS
Jump To

The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acschemneuro.0c00647.

  • General procedures and spectroscopic data of intermediates 7a,b, 12, 13, 15, 16, 19ac, 22, 23, 24, 25a,b, 26b, 28, and 32a,b; formula, molecular weight, and monoisotopic mass of the synthesized compounds; elemental analysis of target compounds; off-target affinities of selected target compounds; and 1H NMR spectra of target compounds 8ac, 9, 14a,b, 20a–c, 26a–c, 33a,b, 29, and 30 (PDF)

Terms & Conditions

Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.

Author Information

ARTICLE SECTIONS
Jump To

  • Corresponding Authors
  • Authors
    • Mauro Niso - Dipartimento di Farmacia−Scienze del Farmaco, Università degli Studi di Bari Aldo Moro, via Orabona, 4, 70125 Bari, Italy
    • Margherita Mastromarino - Dipartimento di Farmacia−Scienze del Farmaco, Università degli Studi di Bari Aldo Moro, via Orabona, 4, 70125 Bari, Italy
    • Andrea Garcia Silva - Center for Research in Molecular Medicine and Chronic Diseases (CIMUS). Universidade de Santiago de Compostela. Avda. de Barcelona, s/n, 15782 Santiago de Compostela, Spain
    • Cibell Resch - Cellular Neurophysiology, Hannover Medical School, 30625 Hannover, Germany
    • Andre Zeug - Cellular Neurophysiology, Hannover Medical School, 30625 Hannover, Germany
    • María I. Loza - Center for Research in Molecular Medicine and Chronic Diseases (CIMUS). Universidade de Santiago de Compostela. Avda. de Barcelona, s/n, 15782 Santiago de Compostela, Spain
    • Marián Castro - Center for Research in Molecular Medicine and Chronic Diseases (CIMUS). Universidade de Santiago de Compostela. Avda. de Barcelona, s/n, 15782 Santiago de Compostela, Spain
    • Evgeni Ponimaskin - Cellular Neurophysiology, Hannover Medical School, 30625 Hannover, Germany
  • Funding

    The present work was partially supported by Telethon Foundation Grant GGP13145 (M.L.) and by German Research Foundation (DFG) (grant number PO732 to E.P. and grant number ZE994/2 to A.Z.). A.G.S., M.I.L., and M.C. acknowledge support from the Spanish Ministry of Economy and Competitiveness (MINECO) (grant number SAF2014-57138-C2–1-R), Xunta de Galicia (Centro singular de investigación de Galicia accreditation 2019–2022, grant number ED431G 2019/02), and the European Union (European Regional Development Fund - ERDF). COST Action CA 18133 “European Research Network on Signal Transduction – ERNEST” is gratefully acknowledged. Funding for open access charge: COST Action CA18133 (ERNEST).

  • Notes
    The authors declare no competing financial interest.

Abbreviations

ARTICLE SECTIONS
Jump To

5-HT

serotonin

5-HT1AR

serotonin 1A receptor

5-HT2AR

serotonin 2A receptor

5-HT7R

serotonin 7 receptor

8-OH-DPAT

8-hydroxy-2-dipropylaminotetralin

(+)-5-FTP

(+)-5-(2′-fluorophenyl)-N,N-dimethyl-1,2,3,4-tetrahidronaphthalen-2-amine

ADME

absorption, distribution, metabolism, and excretion

ASD

autism spectrum disorder

cAMP

cyclic adenosine monophosphate

CLint app

apparent intrinsic clearance

CNS

central nervous system

FRET

fluorescence resonance energy transfer

SAR

structure–activity relationship

SSRI

selective serotonin reuptake inhibitor

References

ARTICLE SECTIONS
Jump To

This article references 59 other publications.

  1. 1
    American Psychiatric Association. (2013) Diagnostic and Statistical Manual of Mental Disorders, 5th ed., American Psychiatric Publishing, Washington DC.
  2. 2
    https://www.cdc.gov/ncbddd/autism/addm-community-report/executive-summary.html (accessed on March 2021).
  3. 3
    Lacivita, E., Perrone, R., Margari, L., and Leopoldo, M. (2017) Targets for Drug Therapy for Autism Spectrum Disorder: Challenges and Future Directions. J. Med. Chem. 60, 91149141,  DOI: 10.1021/acs.jmedchem.7b00965
  4. 4
    Whitaker-Azmitia, P. (2001) Serotonin and brain development: role in human developmental diseases. Brain Res. Bull. 56, 479485,  DOI: 10.1016/S0361-9230(01)00615-3
  5. 5
    Wirth, A., Holst, K., and Ponimaskin, E. (2017) How serotonin receptors regulate morphogenic signalling in neurons. Prog. Neurobiol. 151, 3556,  DOI: 10.1016/j.pneurobio.2016.03.007
  6. 6
    Muller, C. L., Anacker, A. M. J., and Veenstra-VanderWeele, J. (2016) The serotonin system in autism spectrum disorder: From biomarker to animal models. Neuroscience 321, 2441,  DOI: 10.1016/j.neuroscience.2015.11.010
  7. 7
    Chaliha, D., Albrecht, M., Vaccarezza, M., Takechi, R., Lam, V., Al-Salami, H., and Mamo, J. A. (2020) Systematic review of the valproic-acid-induced rodent model of autism. Dev. Neurosci. 42, 1248,  DOI: 10.1159/000509109
  8. 8
    Kinast, K., Peeters, D., Kolk, S. M., Schubert, D., and Homberg, J. R. (2013) Genetic and pharmacological manipulations of the serotonergic system in early life: neurodevelopmental underpinnings of autism-related behavior. Front. Cell. Neurosci. 7, 72
  9. 9
    Guo, Y.-P. and Commons, K. G. (2017) Serotonin neurons abnormalities in the BTBR mouse model of autism. Autism Res. 10, 6677,  DOI: 10.1002/aur.1665
  10. 10
    Brandenburg, C. and Blatt, G. J. (2019) Differential serotonin transporter (5-HTT) and 5-HT2 receptor density in limbic and neocortical areas of adults and children with autism spectrum disorders: implications for selective serotonin reuptake inhibitor efficacy. J. Neurochem. 151, 642655,  DOI: 10.1111/jnc.14832
  11. 11
    Chugani, D. C., Chugani, H. T., Wiznitzer, M., Parikh, S., Evans, P. A., Hansen, R. L., Nass, R., Janisse, J. J., Dixon-Thomas, P., Behen, M. (2016) Efficacy of Low-Dose Buspirone for Restricted and Repetitive Behavior in Young Children with Autism Spectrum Disorder: A Randomized Trial. J. Pediatr. 170, 4553,  DOI: 10.1016/j.jpeds.2015.11.033
  12. 12
    Gould, G. G., Hensler, J. G., Burke, T. F., Benno, R. H., Onaivi, E. S., and Daws, L. C. (2011) Density and function of central serotonin (5-HT) transporters, 5-HT1A and 5-HT2A receptors, and effects of their targeting on BTBR T+tf/J mouse social behavior. J. Neurochem. 116, 291303,  DOI: 10.1111/j.1471-4159.2010.07104.x
  13. 13
    Wang, C.-C., Lin, H.-C., Chan, Y.-H., Gean, P.-W., Yang, Y. K., and Chen, P. S. (2013) 5-HT1A-receptor agonist modified amygdala activity and amygdala-associated social behavior in a valproate-induced rat autism model. Int. J. Neuropsychopharmacol. 16, 20272039,  DOI: 10.1017/S1461145713000473
  14. 14
    Amodeo, D. A., Rivera, E., Dunn, J. T., and Ragozzino, M. E. (2016) M100907 attenuates elevated grooming behavior in the BTBR mouse. Behav. Brain Res. 313, 6770,  DOI: 10.1016/j.bbr.2016.06.064
  15. 15
    Amodeo, D. A., Rivera, E., Cook, E. H., Sweeney, J. A., and Ragozzino, M. E. (2017) 5-HT2A receptor blockade in dorsomedial striatum reduces repetitive behaviors in BTBR mice. Genes Brain. Behav. 16, 342351,  DOI: 10.1111/gbb.12343
  16. 16
    de Bruin, N. M. W. J., van Loevezijn, A., Wicke, K. M., de Haan, M., Venhorst, J., Lange, J. H. M., de Groote, L., van der Neut, M. A. W., Prickaerts, J., Andriambeloson, E. (2016) G. The selective 5-HT6 receptor antagonist SLV has putative cognitive- and social interaction enhancing properties in rodent models of cognitive impairment. Neurobiol. Learn. Mem. 133, 100117,  DOI: 10.1016/j.nlm.2016.06.020
  17. 17
    Costa, L., Spatuzza, M., D’Antoni, S., Bonaccorso, C. M., Trovato, C., Musumeci, S. A., Leopoldo, M., Lacivita, E., Catania, M. V., and Ciranna, L. (2012) Activation of 5-HT7 serotonin receptors reverses metabotropic glutamate receptor-mediated synaptic plasticity in wild-type and Fmr1 knockout mice, a model of Fragile X syndrome. Biol. Psychiatry 72, 924933,  DOI: 10.1016/j.biopsych.2012.06.008
  18. 18
    Costa, L., Sardone, L. M., Lacivita, E., Leopoldo, M., and Ciranna, L. (2015) Novel agonists for serotonin 5-HT7 receptors reverse metabotropic glutamate receptor-mediated long-term depression in the hippocampus of wild-type and Fmr1 KO mice, a model of Fragile X Syndrome. Front. Behav. Neurosci. 9, 65
  19. 19
    Costa, L., Sardone, L. M., Bonaccorso, C. M., D’Antoni, S., Spatuzza, M., Gulisano, W., Tropea, M. R., Puzzo, D., Leopoldo, M., Lacivita, E., Catania, M. V., Ciranna, L. (2018) Activation of serotonin 5-HT7 receptors modulates hippocampal synaptic plasticity by stimulation of adenylate cyclases and rescues learning and behavior in a mouse model of fragile X syndrome. Front. Mol. Neurosci. 11, 353,  DOI: 10.3389/fnmol.2018.00353
  20. 20
    De Filippis, B., Nativio, P., Fabbri, A., Ricceri, L., Adriani, W., Lacivita, E., Leopoldo, M., Passarelli, F., Fuso, A., and Laviola, G. (2014) Pharmacological stimulation of the brain serotonin receptor 7 as a novel therapeutic approach for Rett syndrome. Neuropsychopharmacology 39, 25062518,  DOI: 10.1038/npp.2014.105
  21. 21
    De Filippis, B., Chiodi, V., Adriani, W., Lacivita, E., Mallozzi, C., Leopoldo, M., Domenici, M. R., Fuso, A., and Laviola, G. (2015) Long-lasting beneficial effects of central serotonin receptor 7 stimulation in female mice modeling Rett syndrome. Front. Behav. Neurosci. 9, 86
  22. 22
    Valenti, D., de Bari, L., Vigli, D., Lacivita, E., Leopoldo, M., Laviola, G., Vacca, R. A., and De Filippis, B. (2017) Stimulation of the brain serotonin receptor 7 rescues mitochondrial dysfunction in female mice from two models of Rett syndrome. Neuropharmacology 121, 7988,  DOI: 10.1016/j.neuropharm.2017.04.024
  23. 23
    Vigli, D., Rusconi, L., Valenti, D., La Montanara, P., Cosentino, L., Lacivita, E., Leopoldo, M., Amendola, E., Gross, C., Landsberger, N. (2019) Rescue of prepulse inhibition deficit and brain mitochondrial dysfunction by pharmacological stimulation of the central serotonin receptor 7 in a mouse model of CDKL5 Deficiency Disorder. Neuropharmacology 144, 104114,  DOI: 10.1016/j.neuropharm.2018.10.018
  24. 24
    Kroeze, W. K., Hufeisen, S. J., Popadak, B. A., Renock, S. M., Steinberg, S., Ernsberger, P., Jayathilake, K., Meltzer, H. Y., and Roth, B. L. (2003) H1-histamine receptor affinity predicts short-term weight gain for typical and atypical antipsychotic drugs. Neuropsychopharmacology 28, 519526,  DOI: 10.1038/sj.npp.1300027
  25. 25
    Witt, N. A., Lee, B., Ghent, K., Zhang, W. Q., Pehrson, A. L., Sánchez, C., and Gould, G. G. (2019) Vortioxetine reduces marble burying but only transiently enhances social interaction preference in adult male BTBR T+Itpr3tf/J mice. ACS Chem. Neurosci. 10, 43194327,  DOI: 10.1021/acschemneuro.9b00386
  26. 26
    Canal, C. E., Felsing, D. E., Liu, Y., Zhu, W., Wood, J. T., Perry, C. K., Vemula, R., and Booth, R. G. (2015) An orally active phenylaminotetralin-chemotype serotonin 5-HT7 and 5-HT1A receptor partial agonist that corrects motor stereotypy in mouse models. ACS Chem. Neurosci. 6, 12591270,  DOI: 10.1021/acschemneuro.5b00099
  27. 27
    Armstrong, J. L., Casey, A. B., Saraf, T. S., Mukherjee, M., Booth, R. G., and Canal, C. E. (2020) (S)-5-(2′-Fluorophenyl)-N,N-dimethyl-1,2,3,4-tetrahydronaphthalen-2-amine, a serotonin receptor modulator, possesses anticonvulsant, prosocial, and anxiolytic-like properties in an Fmr1 knockout mouse model of fragile X syndrome and autism spectrum disorder. ACS Pharmacol. Transl. Sci. 3, 509523,  DOI: 10.1021/acsptsci.9b00101
  28. 28
    Hedlund, P. B., Leopoldo, M., Caccia, S., Sarkisyan, G., Fracasso, C., Martelli, G., Lacivita, E., Berardi, F., and Perrone, R. (2010) LP-211 is a brain penetrant selective agonist for the serotonin 5-HT7 receptor. Neurosci. Lett. 481, 1216,  DOI: 10.1016/j.neulet.2010.06.036
  29. 29
    Leopoldo, M., Lacivita, E., Contino, M., Colabufo, N. A., Berardi, F., and Perrone, R. (2007) Structure-activity relationship study on N-(1,2,3,4-tetrahydronaphthalen-1-yl)-4-aryl-1-piperazinehexanamides, a class of 5-HT7 receptor agents. 2. J. Med. Chem. 50, 42144221,  DOI: 10.1021/jm070487n
  30. 30
    Salerno, L., Pittalà, V., Modica, M. N., Siracusa, M. A., Intagliata, S., Cagnotto, A., Salmona, M., Kurczab, R., Bojarski, A. J., and Romeo, G. (2014) Structure-activity relationships and molecular modeling studies of novel arylpiperazinylalkyl 2-benzoxazolones and 2-benzothiazolones as 5-HT7 and 5-HT1A receptor ligands. Eur. J. Med. Chem. 85, 716726,  DOI: 10.1016/j.ejmech.2014.08.023
  31. 31
    Zajdel, P., Kos, T., Marciniec, K., Satała, G., Canale, V., Kamiński, K., Hołuj, M., Lenda, T., Koralewski, R., Bednarski, M. (2018) Novel multi-target azinesulfonamides of cyclic amine derivatives as potential antipsychotics with pro-social and pro-cognitive effects. Eur. J. Med. Chem. 145, 790804,  DOI: 10.1016/j.ejmech.2018.01.002
  32. 32
    Lacivita, E., Podlewska, S., Speranza, L., Niso, M., Satała, G., Perrone, R., Perrone-Capano, C., Bojarski, A. J., and Leopoldo, M. (2016) Structural modifications of the serotonin 5-HT7 receptor agonist N-(4-cyanophenylmethyl)-4-(2-biphenyl)-1-piperazinehexanamide (LP-211) to improve in vitro microsomal stability: a case study. Eur. J. Med. Chem. 120, 363379,  DOI: 10.1016/j.ejmech.2016.05.005
  33. 33
    Forster, E. A., Cliffe, I. A., Bill, D. J., Dover, G. M., Jones, D., Reilly, Y., and Fletcher, A. (1995) A pharmacological profile of the selective silent 5-HT1A receptor antagonist, WAY-100635. Eur. J. Pharmacol. 281, 8188,  DOI: 10.1016/0014-2999(95)00234-C
  34. 34
    Bojarski, A. J., Paluchowska, M. H., Duszyńska, B., Bugno, R., Kłodzińska, A., Tatarczyńska, E., and Chojnacka-Wójcik, E. (2006) Structure-intrinsic activity relationship studies in the group of 1-imido/amido substituted 4-(4-arylpiperazin-1-yl)cyclohexane derivatives; new, potent 5-HT1A receptor agents with anxiolytic- like activity. Bioorg. Med. Chem. 14, 13911402,  DOI: 10.1016/j.bmc.2005.09.060
  35. 35
    Kumar, J. S., Prabhakaran, J., Majo, V. J., Milak, M. S., Hsiung, S. C., Tamir, H., Simpson, N. R., Van Heertum, R. L., Mann, J. J., and Parsey, R. V. (2007) Synthesis and in vivo evaluation of a novel 5-HT1A receptor agonist radioligand (O-methyl- 11C]2-(4-(4-(2-methoxyphenyl)piperazin-1-yl)butyl)-4-methyl-1,2,4-triazine-3,5(2H,4H)dione in nonhuman primates. Eur. J. Nucl. Med. Mol. Imaging 34, 10501060,  DOI: 10.1007/s00259-006-0324-y
  36. 36
    López-Rodríguez, M. L., Morcillo, M. J., Fernández, E., Benhamú, B., Tejada, I., Ayala, D., Viso, A., Campillo, M., Pardo, L., Delgado, M. (2005) Synthesis and structure-activity relationships of a new model of arylpiperazines. 8. Computational simulation of ligand-receptor interaction of 5-HT1AR agonists with selectivity over alpha1-adrenoceptors. J. Med. Chem. 48, 25482558,  DOI: 10.1021/jm048999e
  37. 37
    Cantillon, M., Prakash, A., Alexander, A., Ings, R., Sweitzer, D., and Bhat, L. (2017) Dopamine serotonin stabilizer RP5063: a randomized, double-blind, placebo-controlled multicenter trial of safety and efficacy in exacerbation of schizophrenia or schizoaffective disorder. Schizophr. Res. 189, 126133,  DOI: 10.1016/j.schres.2017.01.043
  38. 38
    Chen, Y., Wang, S., Xu, X., Liu, X., Yu, M., Zhao, S., Liu, S., Qiu, Y., Zhang, T., Liu, B. F. (2013) Synthesis and biological investigation of coumarin piperazine (piperidine) derivatives as potential multireceptor atypical antipsychotics. J. Med. Chem. 56, 46714690,  DOI: 10.1021/jm400408r
  39. 39
    Bojarski, A. J. (2006) Pharmacophore models for metabotropic 5-HT receptor ligands. Curr. Top. Med. Chem. 6, 20052026,  DOI: 10.2174/156802606778522186
  40. 40
    Wager, T. T., Hou, X., Verhoest, P. R., and Villalobos, A. (2016) Central Nervous System Multiparameter Optimization Desirability: application in drug discovery. ACS Chem. Neurosci. 7, 767775,  DOI: 10.1021/acschemneuro.6b00029
  41. 41
    Obach, R. S., Baxter, J. G., Liston, T. E., Silber, B. M., Jones, B. C., MacIntyre, F., Rance, D. J., and Wastall, P. (1997) The prediction of human pharmacokinetic parameters from preclinical and in vitro metabolism data. J. Pharmacol. Exp. Ther. 283, 4658
  42. 42
    Kumar, J. S., Majo, V. J., Hsiung, S. C., Millak, M. S., Liu, K. P., Tamir, H., Prabhakaran, J., Simpson, N. R., Van Heertum, R. L., Mann, J. J. (2006) Synthesis and in vivo validation of [O-methyl-11C]2-{4-[4-(7-methoxynaphthalen-1-yl)piperazin-1-yl]butyl}-4-methyl-2H-[1,2,4]triazine-3,5-dione: a novel 5-HT1A receptor agonist positron emission tomography ligand. J. Med. Chem. 49, 125134,  DOI: 10.1021/jm050725j
  43. 43
    Srinivasa, R. G., Prasanna, K. B. N., Manjunatha, S. G., and Kulkarni, A. K. (2005) Process for the preparation of risperidone, WO2005030772.
  44. 44
    Lacivita, E., Patarnello, D., Stroth, N., Caroli, A., Niso, M., Contino, M., De Giorgio, P., Di Pilato, P., Colabufo, N. A., Berardi, F. (2012) Investigations on the 1-(2-biphenyl)piperazine motif: identification of new potent and selective ligands for the serotonin7 (5-HT7) receptor with agonist or antagonist action in vitro or ex vivo. J. Med. Chem. 55, 63756380,  DOI: 10.1021/jm3003679
  45. 45
    Elgogary, S. R., Hashem, N. M., and Khodeir, M. N. (2015) Synthesis and photooxygenation of linear and angular furocoumarin derivatives as a hydroxyl radical source: psoralen, pseudopsoralen, isopseudopsoralen, and allopsoralen. J. Heteroc. Chem. 52, 506512,  DOI: 10.1002/jhet.2084
  46. 46
    Holloway, B. R., Howe, R., Rao, B. S., and Stribling, D. (1990) Amide derivatives, US4927836.
  47. 47
    Pendin, D., Norante, R., De Nadai, A., Gherardi, G., Vajente, N., Basso, E., Kaludercic, N., Mammucari, C., Paradisi, C., Pozzan, T. (2019) A synthetic fluorescent mitochondria-targeted sensor for ratiometric imaging of calcium in live cells. Angew. Chem., Int. Ed. 58, 99179922,  DOI: 10.1002/anie.201902272
  48. 48
    Brown, J. W., Gangloff, A. R., Jennings, A. J., and Vu, P. H. (2010) Poly (ADP-Ribose) Polymerase (PARP) inhibitors, WO2010111626.
  49. 49
    Kolyasnikova, K. N., Vichuzhanin, M. V., Konstantinopol’skii, M. A., Trofimov, S. S., and Gudasheva, T. A. (2012) Synthesis and pharmacological activity of analogs of the endogenous neuropeptide cycloprolylglycine. Pharm. Chem. J. 46, 96102,  DOI: 10.1007/s11094-012-0741-0
  50. 50
    Kaar, S. J., Natesan, S., McCutcheon, R., and Howes, O. D. (2020) Antipsychotics: mechanisms underlying clinical response and side-effects and novel treatment approaches based on pathophysiology. Neuropharmacology 172, 107704,  DOI: 10.1016/j.neuropharm.2019.107704
  51. 51
    Lacivita, E., Niso, M., Stama, M. L., Arzuaga, A., Altamura, C., Costa, L., Desaphy, J. F., Ragozzino, M. E., Ciranna, L., and Leopoldo, M. (2020) Privileged scaffold-based design to identify a novel drug-like 5-HT7 receptor-preferring agonist to target fragile X syndrome. Eur. J. Med. Chem. 199, 112395,  DOI: 10.1016/j.ejmech.2020.112395
  52. 52
    Di, L., Kerns, E. H., Ma, X. J., Huang, Y., and Carter, G. T. (2008) Applications of high throughput microsomal stability assay in drug discovery. Comb. Chem. High Throughput Screening 11, 469476,  DOI: 10.2174/138620708784911429
  53. 53
    Guscott, M. R., Egan, E., Cook, G. P., Stanton, J. A., Beer, M. S., Rosahl, T. W., Hartmann, S., Kulagowski, J., McAllister, G., Fone, K. F. C. (2003) The hypothermic effect of 5-CT in mice is mediated through the 5-HT7 receptor. Neuropharmacology 44, 10311037,  DOI: 10.1016/S0028-3908(03)00117-5
  54. 54
    Salonikidis, P. S., Zeug, A., Kobe, F., Ponimaskin, E., and Richter, D. W. (2008) Quantitative measurement of cAMP concentration using an exchange protein directly activated by a cAMP- based FRET-sensor. Biophys. J. 95, 54125423,  DOI: 10.1529/biophysj.107.125666
  55. 55
    Prasad, S., Ponimaskin, E., and Zeug, A. (2019) Serotonin receptor oligomerization regulates cAMP-based signaling. J. Cell. Sci. 132, 1,  DOI: 10.1242/jcs.230334
  56. 56
    Tran, J. A., Pontillo, J., Arellano, M., White, N. S., Fleck, B. A., Marinkovic, D., Tucci, F. C., Lanier, M., Nelson, J., Saunders, J., Foster, A. C., and Chen, C. (2005) Identification of agonists and antagonists of the human melanocortin-4 receptor from piperazinebenzylamines. Bioorg. Med. Chem. Lett. 15, 833837,  DOI: 10.1016/j.bmcl.2004.10.096
  57. 57
    Lacivita, E., Niso, M., Hansen, H. D., Di Pilato, P., Herth, M. M., Lehel, S., Ettrup, A., Montenegro, L., Perrone, R., Berardi, F. (2014) Design, synthesis, radiolabeling and in vivo evaluation of potential positron emission tomography (PET) radioligands for brain imaging of the 5-HT7 receptor. Bioorg. Med. Chem. 22, 17361750,  DOI: 10.1016/j.bmc.2014.01.016
  58. 58
    Kaczor, A. A., Silva, A. G., Loza, M. I., Kolb, P., Castro, M., and Poso, A. (2016) Structure-based virtual screening for dopamine D2 receptor ligands as potential antipsychotics. ChemMedChem 11, 718729,  DOI: 10.1002/cmdc.201500599
  59. 59
    Pawley, J. B. (2006) Handbook of Biological Confocal Microscopy, 3rd ed., Springer-Verlag, US, Boston.

Cited By

This article is cited by 3 publications.

  1. Margherita Mastromarino, Maria Favia, Igor A. Schepetkin, Lylia N. Kirpotina, Ewa Trojan, Mauro Niso, Antonio Carrieri, Monika Leśkiewicz, Magdalena Regulska, Massimiliano Darida, Francesco Rossignolo, Stefano Fontana, Mark T. Quinn, Agnieszka Basta-Kaim, Marcello Leopoldo, Enza Lacivita. Design, Synthesis, Biological Evaluation, and Computational Studies of Novel Ureidopropanamides as Formyl Peptide Receptor 2 (FPR2) Agonists to Target the Resolution of Inflammation in Central Nervous System Disorders. Journal of Medicinal Chemistry 2022, 65 (6) , 5004-5028. https://doi.org/10.1021/acs.jmedchem.1c02203
  2. Clinton E. Canal, Marcello Leopoldo. The Need To Improve Reporting of the Pharmacological Action of New Molecules. ACS Chemical Neuroscience 2022, 13 (2) , 185-186. https://doi.org/10.1021/acschemneuro.1c00829
  3. Xiawei Dang, Sidney B. Williams, Sriram Devanathan, Antonietta Franco, Lijun Fu, Peter R. Bernstein, Daniel Walters, Gerald W. Dorn, II. Pharmacophore-Based Design of Phenyl-[hydroxycyclohexyl] Cycloalkyl-Carboxamide Mitofusin Activators with Improved Neuronal Activity. Journal of Medicinal Chemistry 2021, 64 (17) , 12506-12524. https://doi.org/10.1021/acs.jmedchem.1c00163
  • Abstract

    Scheme 1

    Scheme 1. Synthesis of Target Compounds 8ac and 9a

    aReagents and conditions: (A) NaH, Br–(CH2)n–X, anhydrous DMF, rt, 12 h, 40–70% yield; (B) 1-arylpiperazine; K2CO3, acetonitrile, reflux overnight, 20–50% yield.

    Scheme 2

    Scheme 2. Synthesis of Target Compounds 11a,ba

    aReagents and conditions: (A) 1-[2-(4-methoxyphenyl)phenyl]piperazine; K2CO3, acetonitrile, reflux overnight, 61% yield; (B) NaCN, anhydrous DMF, rt, 5 h, quantitative yield; (C) Raney-nickel, H2 (4 atm), MeOH, 50% yield; (D) 2-bromoethanol, CaCO3; reflux, 7 h, 37% yield; (E) PBr3, anhydrous toluene, reflux, 3 h, 65% yield; (F) K2CO3, acetonitrile, reflux overnight, 60% yield.

    Scheme 3

    Scheme 3. Preparation of Target Compounds Featuring the Coumarin Nucleus as the Terminal Fragmenta

    aReagents and conditions: (A) ethyl acetoacetate; conc. H2SO4, rt, 4 h, 34% yield; (B) 1-[2-(4-methoxyphenyl)phenyl]piperazine; K2CO3, acetonitrile, reflux overnight, 21% yield; (C) NaH, Br–(CH2)n–X, anhydrous DMF, rt, 12 h, 40–60% yield.

    Scheme 4

    Scheme 4. Synthetic Route to Obtain Final Compounds 26aa

    aReagents and conditions: (A) methyl 2-bromopropionate, K2CO3, acetone, reflux, 16 h, 54% yield; (B) CH3ONa, MeOH, rt, 3 h, 89% yield; (C) 1,2-dibromoethane, K2CO3, anhydrous DMF, 85 °C, 6 h, 70% yield; (D) Fe dust, AcOH, 80 °C, 1 h, 60% yield; (E) 1-[2-(4-methoxyphenyl)phenyl]piperazine; K2CO3, acetonitrile, reflux overnight, 38% yield.

    Scheme 5

    Scheme 5. Synthetic Route to Obtain Final Compounds 26b, 29, and 30a

    aReagents and conditions: (A) NaH, Br–(CH2)3–Cl, anhydrous DMF, rt, 24 h, 22–42% yield; (B) 1-[2-(4-methoxyphenyl)phenyl]piperazine; K2CO3, acetonitrile, reflux overnight, 20–65% yield. (C) NaH, (R)-glycidyl nosilate, anhydrous DMF, rt, overnight, 41% yield; (D) 1-arylpiperazine; EtOH, reflux, 4 h, 30–54% yield.

    Scheme 6

    Scheme 6. Formation of Target Compounds Bearing the Tetrahydro-1H-pyrrolo[1,2-c]imidazole-1,3(2H)-dione as Terminal Fragmenta

    aReagents and conditions: (A) NaH, Br–(CH2)n–Cl, anhydrous DMF, rt, 12 h, 55–75% yield; (B) 1-[2-(4-methoxyphenyl)phenyl]piperazine; K2CO3, acetonitrile, reflux overnight, 43–74% yield.

    Figure 1

    Figure 1. Compounds 8c, 20b, and 29 stimulate 5-HT7R-mediated cAMP production. (A) N1E cells were transfected with cAMP FRET-based biosensor CEPAC and 5-HT7R-mCherry. Cells were stimulated with the compounds, as indicated. Mean values of the cAMP-biosensor response upon stimulation with 8c 20b, and 29 are shown. Stimulation with LP-211 and 5-CT was used as a control. (B) Quantification of the response amplitude and (C) response time shown as the mean ± SEM (3 < N < 6, in each experiment at least 20 cells were analyzed).

    Figure 2

    Figure 2. Compounds 8c, 20b, and 29 behave as 5-HT1AR agonists in the receptor-mediated cAMP inhibition. (A) N1E cells were transfected with cAMP FRET-based biosensor CEPAC and 5-HT1AR-mCherry. After pretreatment with 1 μM forskolin and 25 μM IBMX, cells were stimulated with the indicated compounds. Each trace shows cAMP response at the single cell. (B) Graphs show changes of cAMP response amplitude relative to pretreatment (mean ± SEM, 3 < N < 6, in each experiment at least 20 cells were analyzed).

    Figure 3

    Figure 3. Functional assays of inositol phosphate (IP) signaling at cloned human 5-HT2ARs. Concentration–response inhibition curves of 8c, 20b, 29, and risperidone (as reference 5-HT2AR antagonist) on IP production stimulated by 1 μM 5-HT in CHO-K1 cells expressing human 5-HT2ARs. The graph shows data (mean ± SEM) from one experiment performed in duplicate.

  • References

    ARTICLE SECTIONS
    Jump To

    This article references 59 other publications.

    1. 1
      American Psychiatric Association. (2013) Diagnostic and Statistical Manual of Mental Disorders, 5th ed., American Psychiatric Publishing, Washington DC.
    2. 2
      https://www.cdc.gov/ncbddd/autism/addm-community-report/executive-summary.html (accessed on March 2021).
    3. 3
      Lacivita, E., Perrone, R., Margari, L., and Leopoldo, M. (2017) Targets for Drug Therapy for Autism Spectrum Disorder: Challenges and Future Directions. J. Med. Chem. 60, 91149141,  DOI: 10.1021/acs.jmedchem.7b00965
    4. 4
      Whitaker-Azmitia, P. (2001) Serotonin and brain development: role in human developmental diseases. Brain Res. Bull. 56, 479485,  DOI: 10.1016/S0361-9230(01)00615-3
    5. 5
      Wirth, A., Holst, K., and Ponimaskin, E. (2017) How serotonin receptors regulate morphogenic signalling in neurons. Prog. Neurobiol. 151, 3556,  DOI: 10.1016/j.pneurobio.2016.03.007
    6. 6
      Muller, C. L., Anacker, A. M. J., and Veenstra-VanderWeele, J. (2016) The serotonin system in autism spectrum disorder: From biomarker to animal models. Neuroscience 321, 2441,  DOI: 10.1016/j.neuroscience.2015.11.010
    7. 7
      Chaliha, D., Albrecht, M., Vaccarezza, M., Takechi, R., Lam, V., Al-Salami, H., and Mamo, J. A. (2020) Systematic review of the valproic-acid-induced rodent model of autism. Dev. Neurosci. 42, 1248,  DOI: 10.1159/000509109
    8. 8
      Kinast, K., Peeters, D., Kolk, S. M., Schubert, D., and Homberg, J. R. (2013) Genetic and pharmacological manipulations of the serotonergic system in early life: neurodevelopmental underpinnings of autism-related behavior. Front. Cell. Neurosci. 7, 72
    9. 9
      Guo, Y.-P. and Commons, K. G. (2017) Serotonin neurons abnormalities in the BTBR mouse model of autism. Autism Res. 10, 6677,  DOI: 10.1002/aur.1665
    10. 10
      Brandenburg, C. and Blatt, G. J. (2019) Differential serotonin transporter (5-HTT) and 5-HT2 receptor density in limbic and neocortical areas of adults and children with autism spectrum disorders: implications for selective serotonin reuptake inhibitor efficacy. J. Neurochem. 151, 642655,  DOI: 10.1111/jnc.14832
    11. 11
      Chugani, D. C., Chugani, H. T., Wiznitzer, M., Parikh, S., Evans, P. A., Hansen, R. L., Nass, R., Janisse, J. J., Dixon-Thomas, P., Behen, M. (2016) Efficacy of Low-Dose Buspirone for Restricted and Repetitive Behavior in Young Children with Autism Spectrum Disorder: A Randomized Trial. J. Pediatr. 170, 4553,  DOI: 10.1016/j.jpeds.2015.11.033
    12. 12
      Gould, G. G., Hensler, J. G., Burke, T. F., Benno, R. H., Onaivi, E. S., and Daws, L. C. (2011) Density and function of central serotonin (5-HT) transporters, 5-HT1A and 5-HT2A receptors, and effects of their targeting on BTBR T+tf/J mouse social behavior. J. Neurochem. 116, 291303,  DOI: 10.1111/j.1471-4159.2010.07104.x
    13. 13
      Wang, C.-C., Lin, H.-C., Chan, Y.-H., Gean, P.-W., Yang, Y. K., and Chen, P. S. (2013) 5-HT1A-receptor agonist modified amygdala activity and amygdala-associated social behavior in a valproate-induced rat autism model. Int. J. Neuropsychopharmacol. 16, 20272039,  DOI: 10.1017/S1461145713000473
    14. 14
      Amodeo, D. A., Rivera, E., Dunn, J. T., and Ragozzino, M. E. (2016) M100907 attenuates elevated grooming behavior in the BTBR mouse. Behav. Brain Res. 313, 6770,  DOI: 10.1016/j.bbr.2016.06.064
    15. 15
      Amodeo, D. A., Rivera, E., Cook, E. H., Sweeney, J. A., and Ragozzino, M. E. (2017) 5-HT2A receptor blockade in dorsomedial striatum reduces repetitive behaviors in BTBR mice. Genes Brain. Behav. 16, 342351,  DOI: 10.1111/gbb.12343
    16. 16
      de Bruin, N. M. W. J., van Loevezijn, A., Wicke, K. M., de Haan, M., Venhorst, J., Lange, J. H. M., de Groote, L., van der Neut, M. A. W., Prickaerts, J., Andriambeloson, E. (2016) G. The selective 5-HT6 receptor antagonist SLV has putative cognitive- and social interaction enhancing properties in rodent models of cognitive impairment. Neurobiol. Learn. Mem. 133, 100117,  DOI: 10.1016/j.nlm.2016.06.020
    17. 17
      Costa, L., Spatuzza, M., D’Antoni, S., Bonaccorso, C. M., Trovato, C., Musumeci, S. A., Leopoldo, M., Lacivita, E., Catania, M. V., and Ciranna, L. (2012) Activation of 5-HT7 serotonin receptors reverses metabotropic glutamate receptor-mediated synaptic plasticity in wild-type and Fmr1 knockout mice, a model of Fragile X syndrome. Biol. Psychiatry 72, 924933,  DOI: 10.1016/j.biopsych.2012.06.008
    18. 18
      Costa, L., Sardone, L. M., Lacivita, E., Leopoldo, M., and Ciranna, L. (2015) Novel agonists for serotonin 5-HT7 receptors reverse metabotropic glutamate receptor-mediated long-term depression in the hippocampus of wild-type and Fmr1 KO mice, a model of Fragile X Syndrome. Front. Behav. Neurosci. 9, 65
    19. 19
      Costa, L., Sardone, L. M., Bonaccorso, C. M., D’Antoni, S., Spatuzza, M., Gulisano, W., Tropea, M. R., Puzzo, D., Leopoldo, M., Lacivita, E., Catania, M. V., Ciranna, L. (2018) Activation of serotonin 5-HT7 receptors modulates hippocampal synaptic plasticity by stimulation of adenylate cyclases and rescues learning and behavior in a mouse model of fragile X syndrome. Front. Mol. Neurosci. 11, 353,  DOI: 10.3389/fnmol.2018.00353
    20. 20
      De Filippis, B., Nativio, P., Fabbri, A., Ricceri, L., Adriani, W., Lacivita, E., Leopoldo, M., Passarelli, F., Fuso, A., and Laviola, G. (2014) Pharmacological stimulation of the brain serotonin receptor 7 as a novel therapeutic approach for Rett syndrome. Neuropsychopharmacology 39, 25062518,  DOI: 10.1038/npp.2014.105
    21. 21
      De Filippis, B., Chiodi, V., Adriani, W., Lacivita, E., Mallozzi, C., Leopoldo, M., Domenici, M. R., Fuso, A., and Laviola, G. (2015) Long-lasting beneficial effects of central serotonin receptor 7 stimulation in female mice modeling Rett syndrome. Front. Behav. Neurosci. 9, 86
    22. 22
      Valenti, D., de Bari, L., Vigli, D., Lacivita, E., Leopoldo, M., Laviola, G., Vacca, R. A., and De Filippis, B. (2017) Stimulation of the brain serotonin receptor 7 rescues mitochondrial dysfunction in female mice from two models of Rett syndrome. Neuropharmacology 121, 7988,  DOI: 10.1016/j.neuropharm.2017.04.024
    23. 23
      Vigli, D., Rusconi, L., Valenti, D., La Montanara, P., Cosentino, L., Lacivita, E., Leopoldo, M., Amendola, E., Gross, C., Landsberger, N. (2019) Rescue of prepulse inhibition deficit and brain mitochondrial dysfunction by pharmacological stimulation of the central serotonin receptor 7 in a mouse model of CDKL5 Deficiency Disorder. Neuropharmacology 144, 104114,  DOI: 10.1016/j.neuropharm.2018.10.018
    24. 24
      Kroeze, W. K., Hufeisen, S. J., Popadak, B. A., Renock, S. M., Steinberg, S., Ernsberger, P., Jayathilake, K., Meltzer, H. Y., and Roth, B. L. (2003) H1-histamine receptor affinity predicts short-term weight gain for typical and atypical antipsychotic drugs. Neuropsychopharmacology 28, 519526,  DOI: 10.1038/sj.npp.1300027
    25. 25
      Witt, N. A., Lee, B., Ghent, K., Zhang, W. Q., Pehrson, A. L., Sánchez, C., and Gould, G. G. (2019) Vortioxetine reduces marble burying but only transiently enhances social interaction preference in adult male BTBR T+Itpr3tf/J mice. ACS Chem. Neurosci. 10, 43194327,  DOI: 10.1021/acschemneuro.9b00386
    26. 26
      Canal, C. E., Felsing, D. E., Liu, Y., Zhu, W., Wood, J. T., Perry, C. K., Vemula, R., and Booth, R. G. (2015) An orally active phenylaminotetralin-chemotype serotonin 5-HT7 and 5-HT1A receptor partial agonist that corrects motor stereotypy in mouse models. ACS Chem. Neurosci. 6, 12591270,  DOI: 10.1021/acschemneuro.5b00099
    27. 27
      Armstrong, J. L., Casey, A. B., Saraf, T. S., Mukherjee, M., Booth, R. G., and Canal, C. E. (2020) (S)-5-(2′-Fluorophenyl)-N,N-dimethyl-1,2,3,4-tetrahydronaphthalen-2-amine, a serotonin receptor modulator, possesses anticonvulsant, prosocial, and anxiolytic-like properties in an Fmr1 knockout mouse model of fragile X syndrome and autism spectrum disorder. ACS Pharmacol. Transl. Sci. 3, 509523,  DOI: 10.1021/acsptsci.9b00101
    28. 28
      Hedlund, P. B., Leopoldo, M., Caccia, S., Sarkisyan, G., Fracasso, C., Martelli, G., Lacivita, E., Berardi, F., and Perrone, R. (2010) LP-211 is a brain penetrant selective agonist for the serotonin 5-HT7 receptor. Neurosci. Lett. 481, 1216,  DOI: 10.1016/j.neulet.2010.06.036
    29. 29
      Leopoldo, M., Lacivita, E., Contino, M., Colabufo, N. A., Berardi, F., and Perrone, R. (2007) Structure-activity relationship study on N-(1,2,3,4-tetrahydronaphthalen-1-yl)-4-aryl-1-piperazinehexanamides, a class of 5-HT7 receptor agents. 2. J. Med. Chem. 50, 42144221,  DOI: 10.1021/jm070487n
    30. 30
      Salerno, L., Pittalà, V., Modica, M. N., Siracusa, M. A., Intagliata, S., Cagnotto, A., Salmona, M., Kurczab, R., Bojarski, A. J., and Romeo, G. (2014) Structure-activity relationships and molecular modeling studies of novel arylpiperazinylalkyl 2-benzoxazolones and 2-benzothiazolones as 5-HT7 and 5-HT1A receptor ligands. Eur. J. Med. Chem. 85, 716726,  DOI: 10.1016/j.ejmech.2014.08.023
    31. 31
      Zajdel, P., Kos, T., Marciniec, K., Satała, G., Canale, V., Kamiński, K., Hołuj, M., Lenda, T., Koralewski, R., Bednarski, M. (2018) Novel multi-target azinesulfonamides of cyclic amine derivatives as potential antipsychotics with pro-social and pro-cognitive effects. Eur. J. Med. Chem. 145, 790804,  DOI: 10.1016/j.ejmech.2018.01.002
    32. 32
      Lacivita, E., Podlewska, S., Speranza, L., Niso, M., Satała, G., Perrone, R., Perrone-Capano, C., Bojarski, A. J., and Leopoldo, M. (2016) Structural modifications of the serotonin 5-HT7 receptor agonist N-(4-cyanophenylmethyl)-4-(2-biphenyl)-1-piperazinehexanamide (LP-211) to improve in vitro microsomal stability: a case study. Eur. J. Med. Chem. 120, 363379,  DOI: 10.1016/j.ejmech.2016.05.005
    33. 33
      Forster, E. A., Cliffe, I. A., Bill, D. J., Dover, G. M., Jones, D., Reilly, Y., and Fletcher, A. (1995) A pharmacological profile of the selective silent 5-HT1A receptor antagonist, WAY-100635. Eur. J. Pharmacol. 281, 8188,  DOI: 10.1016/0014-2999(95)00234-C
    34. 34
      Bojarski, A. J., Paluchowska, M. H., Duszyńska, B., Bugno, R., Kłodzińska, A., Tatarczyńska, E., and Chojnacka-Wójcik, E. (2006) Structure-intrinsic activity relationship studies in the group of 1-imido/amido substituted 4-(4-arylpiperazin-1-yl)cyclohexane derivatives; new, potent 5-HT1A receptor agents with anxiolytic- like activity. Bioorg. Med. Chem. 14, 13911402,  DOI: 10.1016/j.bmc.2005.09.060
    35. 35
      Kumar, J. S., Prabhakaran, J., Majo, V. J., Milak, M. S., Hsiung, S. C., Tamir, H., Simpson, N. R., Van Heertum, R. L., Mann, J. J., and Parsey, R. V. (2007) Synthesis and in vivo evaluation of a novel 5-HT1A receptor agonist radioligand (O-methyl- 11C]2-(4-(4-(2-methoxyphenyl)piperazin-1-yl)butyl)-4-methyl-1,2,4-triazine-3,5(2H,4H)dione in nonhuman primates. Eur. J. Nucl. Med. Mol. Imaging 34, 10501060,  DOI: 10.1007/s00259-006-0324-y
    36. 36
      López-Rodríguez, M. L., Morcillo, M. J., Fernández, E., Benhamú, B., Tejada, I., Ayala, D., Viso, A., Campillo, M., Pardo, L., Delgado, M. (2005) Synthesis and structure-activity relationships of a new model of arylpiperazines. 8. Computational simulation of ligand-receptor interaction of 5-HT1AR agonists with selectivity over alpha1-adrenoceptors. J. Med. Chem. 48, 25482558,  DOI: 10.1021/jm048999e
    37. 37
      Cantillon, M., Prakash, A., Alexander, A., Ings, R., Sweitzer, D., and Bhat, L. (2017) Dopamine serotonin stabilizer RP5063: a randomized, double-blind, placebo-controlled multicenter trial of safety and efficacy in exacerbation of schizophrenia or schizoaffective disorder. Schizophr. Res. 189, 126133,  DOI: 10.1016/j.schres.2017.01.043
    38. 38
      Chen, Y., Wang, S., Xu, X., Liu, X., Yu, M., Zhao, S., Liu, S., Qiu, Y., Zhang, T., Liu, B. F. (2013) Synthesis and biological investigation of coumarin piperazine (piperidine) derivatives as potential multireceptor atypical antipsychotics. J. Med. Chem. 56, 46714690,  DOI: 10.1021/jm400408r
    39. 39
      Bojarski, A. J. (2006) Pharmacophore models for metabotropic 5-HT receptor ligands. Curr. Top. Med. Chem. 6, 20052026,  DOI: 10.2174/156802606778522186
    40. 40
      Wager, T. T., Hou, X., Verhoest, P. R., and Villalobos, A. (2016) Central Nervous System Multiparameter Optimization Desirability: application in drug discovery. ACS Chem. Neurosci. 7, 767775,  DOI: 10.1021/acschemneuro.6b00029
    41. 41
      Obach, R. S., Baxter, J. G., Liston, T. E., Silber, B. M., Jones, B. C., MacIntyre, F., Rance, D. J., and Wastall, P. (1997) The prediction of human pharmacokinetic parameters from preclinical and in vitro metabolism data. J. Pharmacol. Exp. Ther. 283, 4658
    42. 42
      Kumar, J. S., Majo, V. J., Hsiung, S. C., Millak, M. S., Liu, K. P., Tamir, H., Prabhakaran, J., Simpson, N. R., Van Heertum, R. L., Mann, J. J. (2006) Synthesis and in vivo validation of [O-methyl-11C]2-{4-[4-(7-methoxynaphthalen-1-yl)piperazin-1-yl]butyl}-4-methyl-2H-[1,2,4]triazine-3,5-dione: a novel 5-HT1A receptor agonist positron emission tomography ligand. J. Med. Chem. 49, 125134,  DOI: 10.1021/jm050725j
    43. 43
      Srinivasa, R. G., Prasanna, K. B. N., Manjunatha, S. G., and Kulkarni, A. K. (2005) Process for the preparation of risperidone, WO2005030772.
    44. 44
      Lacivita, E., Patarnello, D., Stroth, N., Caroli, A., Niso, M., Contino, M., De Giorgio, P., Di Pilato, P., Colabufo, N. A., Berardi, F. (2012) Investigations on the 1-(2-biphenyl)piperazine motif: identification of new potent and selective ligands for the serotonin7 (5-HT7) receptor with agonist or antagonist action in vitro or ex vivo. J. Med. Chem. 55, 63756380,  DOI: 10.1021/jm3003679
    45. 45
      Elgogary, S. R., Hashem, N. M., and Khodeir, M. N. (2015) Synthesis and photooxygenation of linear and angular furocoumarin derivatives as a hydroxyl radical source: psoralen, pseudopsoralen, isopseudopsoralen, and allopsoralen. J. Heteroc. Chem. 52, 506512,  DOI: 10.1002/jhet.2084
    46. 46
      Holloway, B. R., Howe, R., Rao, B. S., and Stribling, D. (1990) Amide derivatives, US4927836.
    47. 47
      Pendin, D., Norante, R., De Nadai, A., Gherardi, G., Vajente, N., Basso, E., Kaludercic, N., Mammucari, C., Paradisi, C., Pozzan, T. (2019) A synthetic fluorescent mitochondria-targeted sensor for ratiometric imaging of calcium in live cells. Angew. Chem., Int. Ed. 58, 99179922,  DOI: 10.1002/anie.201902272
    48. 48
      Brown, J. W., Gangloff, A. R., Jennings, A. J., and Vu, P. H. (2010) Poly (ADP-Ribose) Polymerase (PARP) inhibitors, WO2010111626.
    49. 49
      Kolyasnikova, K. N., Vichuzhanin, M. V., Konstantinopol’skii, M. A., Trofimov, S. S., and Gudasheva, T. A. (2012) Synthesis and pharmacological activity of analogs of the endogenous neuropeptide cycloprolylglycine. Pharm. Chem. J. 46, 96102,  DOI: 10.1007/s11094-012-0741-0
    50. 50
      Kaar, S. J., Natesan, S., McCutcheon, R., and Howes, O. D. (2020) Antipsychotics: mechanisms underlying clinical response and side-effects and novel treatment approaches based on pathophysiology. Neuropharmacology 172, 107704,  DOI: 10.1016/j.neuropharm.2019.107704
    51. 51
      Lacivita, E., Niso, M., Stama, M. L., Arzuaga, A., Altamura, C., Costa, L., Desaphy, J. F., Ragozzino, M. E., Ciranna, L., and Leopoldo, M. (2020) Privileged scaffold-based design to identify a novel drug-like 5-HT7 receptor-preferring agonist to target fragile X syndrome. Eur. J. Med. Chem. 199, 112395,  DOI: 10.1016/j.ejmech.2020.112395
    52. 52
      Di, L., Kerns, E. H., Ma, X. J., Huang, Y., and Carter, G. T. (2008) Applications of high throughput microsomal stability assay in drug discovery. Comb. Chem. High Throughput Screening 11, 469476,  DOI: 10.2174/138620708784911429
    53. 53
      Guscott, M. R., Egan, E., Cook, G. P., Stanton, J. A., Beer, M. S., Rosahl, T. W., Hartmann, S., Kulagowski, J., McAllister, G., Fone, K. F. C. (2003) The hypothermic effect of 5-CT in mice is mediated through the 5-HT7 receptor. Neuropharmacology 44, 10311037,  DOI: 10.1016/S0028-3908(03)00117-5
    54. 54
      Salonikidis, P. S., Zeug, A., Kobe, F., Ponimaskin, E., and Richter, D. W. (2008) Quantitative measurement of cAMP concentration using an exchange protein directly activated by a cAMP- based FRET-sensor. Biophys. J. 95, 54125423,  DOI: 10.1529/biophysj.107.125666
    55. 55
      Prasad, S., Ponimaskin, E., and Zeug, A. (2019) Serotonin receptor oligomerization regulates cAMP-based signaling. J. Cell. Sci. 132, 1,  DOI: 10.1242/jcs.230334
    56. 56
      Tran, J. A., Pontillo, J., Arellano, M., White, N. S., Fleck, B. A., Marinkovic, D., Tucci, F. C., Lanier, M., Nelson, J., Saunders, J., Foster, A. C., and Chen, C. (2005) Identification of agonists and antagonists of the human melanocortin-4 receptor from piperazinebenzylamines. Bioorg. Med. Chem. Lett. 15, 833837,  DOI: 10.1016/j.bmcl.2004.10.096
    57. 57
      Lacivita, E., Niso, M., Hansen, H. D., Di Pilato, P., Herth, M. M., Lehel, S., Ettrup, A., Montenegro, L., Perrone, R., Berardi, F. (2014) Design, synthesis, radiolabeling and in vivo evaluation of potential positron emission tomography (PET) radioligands for brain imaging of the 5-HT7 receptor. Bioorg. Med. Chem. 22, 17361750,  DOI: 10.1016/j.bmc.2014.01.016
    58. 58
      Kaczor, A. A., Silva, A. G., Loza, M. I., Kolb, P., Castro, M., and Poso, A. (2016) Structure-based virtual screening for dopamine D2 receptor ligands as potential antipsychotics. ChemMedChem 11, 718729,  DOI: 10.1002/cmdc.201500599
    59. 59
      Pawley, J. B. (2006) Handbook of Biological Confocal Microscopy, 3rd ed., Springer-Verlag, US, Boston.
  • Supporting Information

    Supporting Information

    ARTICLE SECTIONS
    Jump To

    The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acschemneuro.0c00647.

    • General procedures and spectroscopic data of intermediates 7a,b, 12, 13, 15, 16, 19ac, 22, 23, 24, 25a,b, 26b, 28, and 32a,b; formula, molecular weight, and monoisotopic mass of the synthesized compounds; elemental analysis of target compounds; off-target affinities of selected target compounds; and 1H NMR spectra of target compounds 8ac, 9, 14a,b, 20a–c, 26a–c, 33a,b, 29, and 30 (PDF)


    Terms & Conditions

    Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.

Pair your accounts.

Export articles to Mendeley

Get article recommendations from ACS based on references in your Mendeley library.

Pair your accounts.

Export articles to Mendeley

Get article recommendations from ACS based on references in your Mendeley library.

You’ve supercharged your research process with ACS and Mendeley!

STEP 1:
Click to create an ACS ID

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

MENDELEY PAIRING EXPIRED
Your Mendeley pairing has expired. Please reconnect