Home J Young Pharm. Vol 16/issue 2/2024 Study, Docking, in silico ADME and Predicted Acute Toxicity of Novel Hetero-Aromatic Imidazolidine Analogues as Potential Anti-Epileptic Agents

Study, Docking, in silico ADME and Predicted Acute Toxicity of Novel Hetero-Aromatic Imidazolidine Analogues as Potential Anti-Epileptic Agents

by admin

1Department of Pharmaceutical Chemistry, Pravara Rural College of Pharmacy Pravaranagar, Ahmednagar, INDIA

2Department of Pharmaceutics, Vishal Institute of Pharmaceutical Education and Research, Alephata, INDIA

3Department of Pharmacognosy, Pravara Rural College of Pharmacy Pravaranagar, Ahmednagar, INDIA

4Department of Pharmacology, R. C. Patel Institution of Pharmaceutical Education and Research, Shirpur, INDIA

5Department of Pharmacognosy, Pravara Rural College of Pharmacy Pravaranagar, Ahmednagar, INDIA

Corresponding author.

Correspondence: Dr. Rohit Jaysing Bhor Department of Pharmaceutical Chemistry, Pravara Rural College of Pharmacy Pravaranagar, Ahmednagar, Maharashtra, INDIA. Email: [email protected]
Received January 16, 2024; Revised February 16, 2024; Accepted March 01, 2024.
Copyright ©2024 Phcog.Net
This is an open access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 License, which allows others to remix, tweak, and build upon the work non-commercially, as long as the author is credited and the new creations are licensed under the identical terms.

Citation

1.Bhor RJ, Gaikwad MS, Londhe OA, Wakchaure TP, Patil SA, Ingle PS, et al. Study, Docking, in silico ADME and Predicted Acute Toxicity of Novel Hetero-Aromatic Imidazolidine Analogues as Potential Anti-Epileptic Agents. Journal of Young Pharmacists [Internet]. 2024 Jun 6;16(2):236–43. Available from: http://dx.doi.org/10.5530/jyp.2024.16.31
Copy to clipboard
Published in: Journal of Young Pharmacists, 2024; 16(2): 236-243.Published online: 03 June 2024DOI: 10.5530/jyp.2024.16.31

ABSTRACT

Background

In the present work, molecular docking analysis was conducted on proposed protein derivatives from the protein data bank using the Auto Dock for Docking programme. study on protein binding affinity using 1-acetyl-3-(2-aminophenyl)-5,5-diphenylimidazolidine- 2,4-dione derivatives and the GABA receptor. Even if a number of innovative anticonvulsants have been created in India, some types of seizures are still not adequately controlled by smaller side effects even after the use of modern and revolutionary treatments.

Materials and Methods

Target-oriented virtual screening of ligands under investigation with adaptable molecular docking techniques. We can choose the most promising compounds for further study of various derivatives by evaluating the ligands’ affinity to the GABAA active sites. Via the inhibitor GABAergic route, a number of 5,5-diphenylimidazolidine derivatives were designed and investigated in silico.

Results

Using the AutoDock 4.2 programme, a number of molecules associated with 5,5-diphenylimidazolidine were investigated for molecular docking, acute prediction, and ADMET analysis. According to docking research, these molecules have at least one hydrogen bond stabilising them. The proposed composites were all investigated for Gamma Aminobutyric Acid (GABA) inhibitory exertion and all shown good in silico ADME and molecular docking results.

Conclusion

One of the essential enzymes for biosynthesis in many natural ecosystems, Gamma Aminobutyric Acid (GABA) is present in both humans and animals. By meticulously replicating the prior pharmacological experiment’s circumstances, we are able to compare the outcomes and talk about certain commonalities in how molecule fragments affect anticonvulsant action. Docking score, ADMET analysis, acute toxicity prediction, and structural location of ligands in the active GABAA site enzyme were found to positively correlate, supporting the viability of target-based virtual screening as a way to expedite pharmacological screening.

Keywords: Gamma aminobutyric acid, Molecular Docking, Structure-Based Drug Design, 3D QSAR

INTRODUCTION

The World Health Organisation (WHO) estimates that 65 million people worldwide suffer from epilepsy. Eighty percent of them are said to live in low- and middle-income countries. Nevertheless, epilepsy is an uncontrollable, neurological seizure.13 It occurs seven times more advanced than the typical state of all chronic illnesses.47 An earlier investigation found that the US spends around $15.5 billion a year on medical expenses, including seizures.

Due to the high incidence of epilepsy in the US and India, more effective and safer anticonvulsants must be developed in order to lower the cost of treating epilepsy. Several different countries have put a lot of effort into finding novel, safe, and effective epilepsy medications. Epilepsy can take many different forms, such as absence seizures, clonic seizures, focal or generalised seizures, etc. Modern research has yielded novel anticonvulsants that are used to treat focal seizures. Current and newer medicines cannot cure all forms of epilepsy.812 Uncontrolled storms and an overabundance of transitory neuronal discharges were the causes of epilepsy.13,14 League Against Epilepsy (LAE), can be listed as two unprovoked seizures occurring during the following decade, followed by single unprovoked (or reflex) seizure and a chance of further seizures proportional to the overall recurrence risk (at least 60%). Potassium bromide, the first anticonvulsant medication, was initially described in the middle of the 1800s. Since then, a number of Antiepileptic medications (AEDs) have been authorised by medical professionals and scientists and are accessible as the first line of treatment for individuals with epilepsy.15,16 A significant part of the medication development process for a more recent class of drugs with anticonvulsant action involves these derivatives or group. Roughly forty derivatives have been demonstrated to possess anticonvulsant characteristics.1719 Presynaptic GABAergic neurons synthesise GABA (Gamma-Aminobutyric Acid) from glutamate. GABA is a key neurotransmitter in the brain that regulates the balance between excitement and inhibition. These presynaptic neurons depolarize, which causes the release of GABA for neurotransmission. GABA can bind to either of the two main GABA receptors on postsynaptic neurons-GABAA or GABAB receptors-when it is released into the synaptic cleft. The principal chloride ion channel in GABAA receptors opens as a result of GABA binding allosterically to those receptors. This ultimately causes the neuronal membrane to become hyperpolarized, which lowers cell excitability and increases neural inhibition. As metabotropic receptors, GABAB receptors are activated by GABA, which causes the corresponding potassium channels to open through G-protein coupled receptors. Hyperpolarization and neural inhibition follow, which are comparable to the actions of GABAA receptors.

MATERIALS AND METHODS

Receptors and Ligands Collections

Wang and his fellow researchers’ efforts produced a collection of trustworthy experimental anti-epilepsy data sets and their characteristics, comprising nine strong chemicals. It was demonstrated in the literature that these particular experimental AEDs had superior activity to co-compounds. Chemdraw software was utilized to create 2D structural forms from the dependable data sets, which were then stored in cdx format. The Spartan 14 software’s DFT (density functional theory) method was used to geometrically optimise the drawn structures in order to make the atoms of the compounds energetically stable and seem realistic.

Molecular Docking Simulation

In this investigation, docking simulations were carried out using the Pyrx multifunctional simulation software’s Autoduck vina docking tool. Adequate preparation was done for both the receptor and the ligands, which were then saved in PDF format using the Discovery Studio programme, uploaded into Pyrx, and converted to PSDT format. The autoduck vina feature of the programme was activated when the GABA receptor and the ligands (Table 1) were moved to the X, Y, and Z grid of the Pyrx software interface. The docked posture with the best orientation binding energy was chosen, examined, and recorded following several minutes of selective ligand poses on the Gamma-Aminobutyric Acid (GABA)A receptor binding site.

Label Structure
Sa1
Sa2
Sa3
Sa4
Sa5
Table 1:
Ligand used for Docking.

Preparation of Ligands

MarvinSketch 18.23 was used to obtain structures, which were then saved in mol format. stored as pdb files and optimised by Chem3D using the molecular-mechanical MM2 method. Protein preparation The crystal was treated with Discovery Studio Visualizer 2017 / R2 to exclude ligand molecules and water. PDB files were used to store protein structures. Native ligands have grid boxes placed on them. For docking, AutoDock Vina was employed. The acquired docking result was visualised and evaluated using Discovery Studio V17.2.0.16349 (Table 1).

RESULTS

To evaluate the physiochemical properties of each synthesised molecule (sb1-sb5), in silico evaluations were performed using Lipinski’s rule (Table 2). The Lipinski rule states that an epileptic drug’s physicochemical properties, directly correlate with how well it is absorbed. Every functional variation possesses hydrogen bond donor and acceptor domains, as per Lipinski’s criterion. The superior lipophilicity of the generated compounds leads to an increase in bioavailability in the end. The aforementioned statement was supported by an analysis of the VDss is similar to that of traditional medications. The estimated maximum total clearance values for each compound were comparable, according to the total clearance values (Table 3).

Compound Codes Lipinski rule of five Veber’s rule
M Log P Mol. Wt. (g/mol) HBA HBD Violations Total polar surface area (Å2) No. of rotatable bonds
Sa1 1.93 458.47 5 3 0 130.83 8
Sa2 2.33 459.45 6 2 0 128.03 8
Sa3 3.23 492.53 4 3 0 113.76 8
Sa4 2.41 537.52 6 3 2 159.58 9
Sa5 3.39 521.52 6 2 1 128.03 9
Table 2:
Lipinski rule of five.
Compound codes Pharmacokinetics Drug-likeness
Gl abs. BBB pen. P-gp sub. CYP1A2 CYP2C19 CYP2C9 CYP2D6 CYP3A4 Log Kp (skin permeation, cm/s) Ghose Egan Muegge Bioavailability Score
inhibitors
Sa1 High No Yes No No Yes No No -7.47 Yes Yes Yes 0.55
Sa2 High No No No Yes Yes No Yes -7.06 Yes Yes Yes 0.55
Sa3 High No No No Yes Yes No Yes -6.00 Yes Yes Yes 0.55
Sa4 Low No No No Yes Yes No Yes -6.00 No No No 0.17
Sa5 High No No No Yes Yes No No -6.27 No Yes Yes 0.55
Table 3:
Pharmacokinetics and drug-likeness properties of compounds.

Predicted Acute Toxicity of Molecules

The goal of acute toxicity studies is to identify the amount that, whether administered once or over a few administrations, will result in death or major toxicological consequences. They also function as a source of knowledge on dosages that ought to be used in later research. These investigations offer an additional chance to ascertain compound-induced effects as shown by clinical chemistry, morphology, or other assessments. Additionally, acute investigations may provide an early indicator of the potential target organ or organs. This is a section of the acute toxicity studies. The median Lethal Dose (LD50) is defined as the dosage at which 50% of exposed animals die. This discovery has been useful in comparing the acute toxicity of different substances and served as the basis for the environmental and industrial toxicant categorization. Pharmaceutical development used to begin with a formal LD50 evaluation. Experience showed that the LD50 (Table 4) used much too many animals and provided little useful information for the creation of pharmacological products.

Compound codes Parameters
LD50 (mg/ kg) Toxicity class Prediction accuracy (%) Hepatotoxicity Carcinogenicity Immunotoxicity Mutagenicity Cytotoxicity
(Probability)
Sa1 1190 4 100 A (0.69) I (0.62) A (0.96) I (0.97) I (0.93)
Sa2 5000 5 67.38 A (0.62) I (0.56) I (0.79) I (0.73) I (0.71)
Sa3 5000 5 67.38 A (0.52) I (0.51) A (0.69) I (0.72) I (0.69)
Sa4 5000 5 54.26 A (0.56) A (0.55) A (0.95) A (0.76) I (0.62)
Sa5 5000 5 54.26 A (0.64) I (0.52) A (0.82) I (0.83) I (0.70)
Table 4:
Predicted acute toxicity of molecules.

In silico Docking Studies

Molecular docking study was done to identify the many binding interactions of synthetic molecules within the target pocket of well-known antiepileptic medications, such as GABA receptors. The development of the anticonvulsant activity of the investigated medications was also aided by the comparative investigation of interactions with different targets. Every synthetic molecule having antiepileptic efficacy in vivo has been selected for molecular docking investigations using a variety of targets in silico (Table 5). The binding energies of the conventional medications (phenytoin and phenobarbital) and active compounds (sa4, sa3, and sa5) with selected targets are listed in Table 5. The binding energies of all active molecules range from -11.7 to -9.7 kcal/mol, which is quite similar to the binding energies of phenytoin (-5.48 kcal/mol) of common pharmaceuticals.

Active Amino acid Bond length Bond Type Bond Category Docking score
Compound: Sa1
SER331 2.1288 Conventional Hydrogen Bond Hydrogen Bond -10.5
ILE328 3.02982 Conventional Hydrogen Bond Hydrogen Bond
SER332 1.83542 Conventional Hydrogen Bond Hydrogen Bond
UNL1 3.12931 Pi-Donor Hydrogen Bond Hydrogen Bond
PHE297 4.35718 Pi-Pi Stacked Hydrophobic
PHE297 3.86223 Pi-Pi Stacked Hydrophobic
PHE255 5.22747 Pi-Pi T-shaped Hydrophobic
ILE328 5.40136 Pi-Alkyl Hydrophobic
ARG335 5.19041 Pi-Alkyl Hydrophobic
Compound: Sa2
SER547 3.02173 Conventional Hydrogen Bond Hydrogen Bond -9.7
SER547 3.3004 Conventional Hydrogen Bond Hydrogen Bond
LEU551 2.83608 Conventional Hydrogen Bond Hydrogen Bond
UNL1 2.65348 Conventional Hydrogen Bond Hydrogen Bond
ILE475 3.45533 Pi-Sigma Hydrophobic
TYR563 3.41608 Pi-Sigma Hydrophobic
TYR532 4.85189 Pi-Pi Stacked Hydrophobic
ILE475 4.43218 Pi-Alkyl Hydrophobic
ARG554 4.37848 Pi-Alkyl Hydrophobic
ALA472 4.9063 Pi-Alkyl Hydrophobic
ALA528 4.4155 Pi-Alkyl Hydrophobic
Compound: Sa3
SER331 3.16048 Conventional Hydrogen Bond Hydrogen Bond -10.9
ARG335 3.01627 Conventional Hydrogen Bond Hydrogen Bond
SER331 2.51564 Conventional Hydrogen Bond Hydrogen Bond
UNL1 1.90316 Conventional Hydrogen Bond Hydrogen Bond
GLN321 2.45132 Conventional Hydrogen Bond Hydrogen Bond
LEU270 3.34915 Carbon Hydrogen Bond Hydrogen Bond
ILE285 3.64701 Pi-Sigma Hydrophobic
MET299 4.93138 Pi-Sulfur Other
PHE297 5.96706 Pi-Pi T-shaped Hydrophobic
LYS322 4.28553 Pi-Alkyl Hydrophobic
ARG335 4.28831 Pi-Alkyl Hydrophobic
ILE257 4.55796 Pi-Alkyl Hydrophobic
LEU270 5.1494 Pi-Alkyl Hydrophobic
Compound: Sa4
SER624 3.299 Conventional Hydrogen Bond Hydrogen Bond -11.7
ARG643 2.8825 Conventional Hydrogen Bond Hydrogen Bond
SER547 2.80776 Carbon Hydrogen Bond Hydrogen Bond
GLU567 3.75048 Pi-Anion Electrostatic
TYR532 3.83066 Pi-Donor Hydrogen Bond Hydrogen Bond
SER547 3.35935 Pi-Donor Hydrogen Bond Hydrogen Bond
ILE475 3.47636 Pi-Sigma Hydrophobic
ILE475 3.60702 Pi-Sigma Hydrophobic
TYR532 4.62253 Pi-Pi Stacked Hydrophobic
LEU551 4.62429 Pi-Alkyl Hydrophobic
ARG554 5.25732 Pi-Alkyl Hydrophobic
ALA472 4.60765 Pi-Alkyl Hydrophobic
ALA528 4.43503 Pi-Alkyl Hydrophobic
VAL544 5.1625 Pi-Alkyl Hydrophobic
Compound: Sa5
ARG268 3.03078 Conventional Hydrogen Bond Hydrogen Bond -10.7
SER331 2.82898 Conventional Hydrogen Bond Hydrogen Bond
SER33 2.04784 Conventional Hydrogen Bond Hydrogen Bond
UNL1 2.16101 Conventional Hydrogen Bond Hydrogen Bond
ILE257 3.63488 Carbon Hydrogen Bond Hydrogen Bond
GLY271 3.4741 Carbon Hydrogen Bond Hydrogen Bond
GLY271 4.02217 Pi-Donor Hydrogen Bond Hydrogen Bond
ILE274 3.57086 Pi-Sigma Hydrophobic
ILE257 5.36084 Pi-Alkyl Hydrophobic
ARG335 3.9779 Pi-Alkyl Hydrophobic
LEU270 4.69216 Pi-Alkyl Hydrophobic
VAL262 5.49931 Pi-Alkyl Hydrophobic
Table 5:
The active amino residues, bond length, bond category, bond type, ligand energies, and docking scores.

DISCUSSION

Molecular docking experiments have been utilized to determine how conventional medicines interact with ligands on proteins. The goal of the molecular docking study was to access and comprehend the interaction binding energy dynamics of a few strong anti-epilepsy drugs that were produced experimentally on the Gamma-Aminobutyric Acid (GABA) A receptor. Table 2 revealed that the docking scores of the most powerful AED- selected anti-epilepsy medication candidates ranged from -5.1 to -6.8 kcal/mol. Consequently, the effective mechanism of 1-acetyl- 3-(2-aminophenyl)-5,5-diphenylimidazolidine-2,4-dione might be through the inhibition of GABA (Gamma-Aminobutyric Acid) A receptor activity on the binding site. This is due to the fact that the strength of the docking scores strongly correlates with the ability to start ligand-protein interactions. A molecular docking analysis was done on each and every synthesised chemical. They were discovered to entirely occupy the active regions in the target protein, therefore potently inhibiting the Gamma-Aminobutyric Acid (GABA)A receptor. Compared to the typical anticonvulsant medication, some of the compounds had higher docking scores. Compound SA4, which had the highest docking score of (-6.33), was determined to be the most powerful of all the synthesised compounds. Due to hydrogen bonding, compound SA5’s binding mode takes a favourable orientation (-0.85) as compared to the reference (-4.03 as Dock Score). Additionally, compounds SA4 and SA5 had strong docking scores of (-6.32) and H bonds (-0.85), respectively, which are higher than those of the commonly used medication. The studied chemicals’ interactions with other targets, however, are not as significant. The most active compounds (sb4, sb3, and sb5) and reference medication (phenytoin) in 2D interaction images. Compound SA3’s methoxy group joins forces with residues at SER A:678 and THR A:516 to create a standard hydrogen bond. Compound SB4’s nitro group joins SER A:678 in a typical hydrogen bond. Compound SA5’s hydroxyl group joins GLN A:485 in a typical hydrogen bond. The data shown above suggests that the tested drugs and targets interact well (Table 6). The facts presented above clearly demonstrated the good prospective antiepileptic properties of the synthesised compounds, which acted mainly through the mechanism of action of conventional medications like phenytoin. Additionally, it appears that the synthetic chemicals show strong anticonvulsant effects upon docking with the voltage-gated calcium channel receptor.

3D-docking poses 2D-docking poses
Compound: Sa1
Compound: Sa2
Compound: Sa3
Compound: Sa4
Compound: Sa5
Table 6:
Docking Poses 2D and 3D.

CONCLUSION

Promising anticonvulsants, 1-acetyl-3-(2-aminophenyl)- 5,5-diphenylimidazolidine-2,4-dione derivatives was synthesised as a structural homologue of phenytoin. These ligands were chosen for pharmacological screening based on how well they docked onto the GABAA receptor’s active sites when compared to other substances. Acute toxicity and molecular docking ADMET analysis were seen in just 5 drugs. We might suggest the docking technology that has been given as a means of expediting screening because of the shown correlation between the active GABAA receptor sites and screening results.

Cite this article:

Bhor RJ, Gaikwad MS, Londhe OA, Wakchaure TP, Patil SA, Ingle PS, etal. Study, Docking, in silico ADME and Predicted Acute Toxicity of Novel Hetero-Aromatic Imidazolidine Analogues as Potential Anti-Epileptic Agents. J Young Pharm. 2024;16(2):236-43.

ACKNOWLEDGEMENT

The authors are thankful to Dr. S.B. Bhawar, Pravara Rural College of Pharmacy, Pravaranagar.

ABBREVIATIONS

mg/kg Milligram/kilograms
sec Seconds
kcal Kilocalorie
Mol. Wt Molecular Weight
g Gram
LEU Leucine
THR Threonine
ALA Alanine
MET Methionine
PHE Phenylalanine
WHO World health association
Log P Partition coefficient

References

  1. Anger T, Madge DJ, Mulla M, Riddall D. Medicinal Chemistry of Neuronal Voltage-Gated Sodium Channel Blockers. J Med Chem. 2001;44:115-37. [Google Scholar]
  2. Ragavendran JV, Sriram D, Kotapati S, Stables J, Yogeeswari P. Newer GABA Derivatives for the Treatment of Epilepsy Including Febrile Seizures: A Bioisosteric Approach. Eur J Med Chem. 2008;43:2650-5. [Google Scholar]
  3. Wu T, Ohgoh M, Hanada T. Mode of Seizure Inhibition by Sodium Channel Blockers, an SV2A Ligand, and an AMPA Receptor Antagonist in a Rat Amygdala Kindling Model. Epilepsy Research. 2019;154:42-9. [Google Scholar]
  4. Alexander SM, Harkless J, Butcher JR, Scott KR, Jackson-Ayotunde LP. An Examination of Some Ethyl Ester Enaminone Derivatives as Anticonvulsant Agents. Bioorganic & Medicinal Chemistry. 2013;21:3272-9. [Google Scholar]
  5. Heinbockel T, Wang Z, Jackson-Ayotunde PL. Allosteric Modulation of GABAA Receptors by an Anilino Enaminone in an Olfactory Center of the Mouse Brain. Pharmaceuticals. 2014;7:1069-90. [Google Scholar]
  6. Wang Z, Sun L, Jackson PL, Scott RK, Heinbockel T. A Substituted Anilino Enaminone Acts as a Novel Positive Allosteric Modulator of GABAA Re-ceptors in the Mouse Brain. Journal of Pharmacology and Experimental Therapeutics. 2011;336 [Google Scholar]
  7. Jackson PL, Hanson CD, Farrell AK, Raymond BJ, Stables JP, Eddington ND, Scott KR, et al. An Explanation of Anticonvulsant Activity and Toxicity Per Linus Pauling’s Clathrate Hypothesis. Array. 2012;51:42-51. [Google Scholar]
  8. Anderson AJ, Nicholson JM, Bakare O, Butcher RJ, Wilson TL, Scott KR, et al. Further Studies on the Anticonvulsant Activity and Potential Type IV Phosphodiesterase Inhibitory Activity of Substituted Vinylic Benzamides. Bioorganic & Medicinal Chemistry. 2006;14:997-1006. [Google Scholar]
  9. Jackson PL, Scott KR, Southerland WM, Fang Y. CoMFA and CoMSIA Studies on Some Anticonvulsant Enaminones. Bioorganic & Medicinal Chemistry. 2009;17:133-40. [Google Scholar]
  10. Edafiogho IO, Kombian SB, Anathalaskshmi KVV, Salama NN, Eddington ND, Wilson TL, et al. Exploring Additional Therapeutic Activities. Journal of Pharmaceutical Sciences. 2007;96:2509-31. [Google Scholar]
  11. Amaye IJ, Heinbockel T, Woods J, Wang Z, Martin-Caraballo M, Jackson-Ayotunde P, et al. 6 Hz Active Anticonvulsant Fluorinated N-Benzamide Enaminones and Their Inhibitory Neuronal Activity. International Journal of Environmental Research and Public Health. 2018;15:1784 [Google Scholar]
  12. Sula A, Booker J, Ng LC, Naylor CE, DeCaen PG, Wallace BA, et al. The Complete Structure of an Activated Open Sodium Channel. Nature Communications. 2012;8:14205-14. [Google Scholar]
  13. Temme L, Boergel F, Schepmann D, Robaa D, Sippl W, Daniliuc C, et al. Impact of hydroxy moieties at the benzo annulene ring system of GluN2B ligands: Design, synthesis and biological evaluation. Bioorganic and Medicinal Chemistry. 2019;27(23) [Google Scholar]
  14. Yadagiri B, Holagunda UD, Bantu R, Nagarapu L, Guguloth V, Polepally S, et al. Rational design, synthesis and anti-proliferative evaluation of novel benzosuberone tethered with hydrazide-hydrazones. Bioorganic and medicinal chemistry letters. 2014;24(21):5041-4. [Google Scholar]
  15. Althagafi I, Farghaly TA, Abbas EM, Harras MF. Benzosuberone as Precursor for Synthesis of Antimicrobial Agents: Synthesis, Antimicrobial Activity, and Molecular Docking. Polycyclic Aromatic Compounds. 2021;41(8):1646-66. [Google Scholar]
  16. Grimaldi TB, Back DF, Zeni G. Potassium tert-butoxide promoted annulation of 2-alkynylphenyl propargyl ethers: Selective synthesis of benzofuran and 12 H-benzoannulene derivatives. The Journal of organic chemistry. 2013;78(21):11017-31. [Google Scholar]
  17. Ahmed SK, Haese NN, Cowan JT, Pathak V, Moukha-Chafiq O, Smith VJ, et al. Targeting chikungunya virus replication by benzoannulene inhibitors. Journal of Medicinal Chemistry. 2021;64(8):4762-86. [Google Scholar]
  18. Oh CH, Kim JH, Oh BK, Park JR, Lee JH. Gold-Catalyzed Sequential Activation of Propargylic Carboxylates: A Facile Route to Benzoannulenes. Chemistry-A European Journal. 2013;19(8):2592-6. [Google Scholar]
  19. Lazzari P, Distinto R, Manca I, Baillie G, Murineddu G, Pira M, et al. A critical review of both the synthesis approach and the receptor profile of the 8-chloro-1-(2’, 4’-dichlorophenyl)-N-piperidin-1-yl-1, 4, 5, 6-tetra hydro benzo [6, 7] cyclohepta [1, 2-c] pyrazole-3-carboxamide and analogue derivatives. European Journal of Medicinal Chemistry. 2016;121:194-208. [Google Scholar]