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

Physicochemical and Pharmacokinetic Analysis of Anacardic Acid Derivatives

  • Fahmina Zafar*
    Fahmina Zafar
    Inorganic Materials Research Laboratory, Department of Chemistry, Jamia Millia Islamia, New Delhi 110025, India
    *E-mail: [email protected] (F.Z.).
    More by Fahmina Zafar
  • Anjali Gupta*
    Anjali Gupta
    Division of Chemistry, School of Basic and Applied Science, Galgotias University, Greater Noida 201310, Uttar Pradesh, India
    *E-mail: [email protected] (A.G.).
    More by Anjali Gupta
  • Karthick Thangavel
    Karthick Thangavel
    Department of Physics, School of Electrical and Electronics Engineering, SASTRA Deemed University, Thanjavur 613 401, Tamil Nadu, India
  • Kavita Khatana
    Kavita Khatana
    Division of Chemistry, School of Basic and Applied Science, Galgotias University, Greater Noida 201310, Uttar Pradesh, India
    More by Kavita Khatana
  • Ali Alhaji Sani
    Ali Alhaji Sani
    Division of Chemistry, School of Basic and Applied Science, Galgotias University, Greater Noida 201310, Uttar Pradesh, India
    More by Ali Alhaji Sani
  • Anujit Ghosal
    Anujit Ghosal
    Inorganic Materials Research Laboratory, Department of Chemistry, Jamia Millia Islamia, New Delhi 110025, India
    Division of Chemistry, School of Basic and Applied Science, Galgotias University, Greater Noida 201310, Uttar Pradesh, India
    School of Life Sciences, Beijing Institute of Technology, Beijing 100811, China
    More by Anujit Ghosal
  • Poonam Tandon
    Poonam Tandon
    Department of Physics, University of Lucknow, Lucknow 226007, India
    More by Poonam Tandon
  • , and 
  • Nahid Nishat*
    Nahid Nishat
    Inorganic Materials Research Laboratory, Department of Chemistry, Jamia Millia Islamia, New Delhi 110025, India
    *E-mail: [email protected] (N.N.).
    More by Nahid Nishat
Cite this: ACS Omega 2020, 5, 11, 6021–6030
Publication Date (Web):March 13, 2020
https://doi.org/10.1021/acsomega.9b04398

Copyright © 2020 American Chemical Society. This publication is licensed under these Terms of Use.

  • Open Access

Article Views

5991

Altmetric

-

Citations

41
LEARN ABOUT THESE METRICS
PDF (4 MB)
Supporting Info (1)»

Abstract

Anacardic acid (AA) and its derivatives are well-known for their therapeutic applications ranging from antitumor, antibacterial, antioxidant, anticancer, and so forth. However, their poor pharmacokinetic and safety properties create significant hurdles in the formulation of the final drug molecule. As a part of our endeavor to enhance the potential and exploration of the anticancer activities, a detailed study on the properties of selected AA derivatives was performed in this work. A comprehensive analysis of the drug-like properties of 100 naturally occurring AA derivatives was performed, and the results were compared with certain marketed anticancer drugs. The work focused on the understanding of the interplay among eight physicochemical properties. The relationships between the physicochemical properties, absorption, distribution, metabolism, and excretion attributes, and the in silico toxicity profile for the set of AA derivatives were established. The ligand efficacy of the finally scrutinized 17 AA derivatives on the basis of pharmacokinetic properties and toxicity parameters was further subjected to dock against the potential anticancer target cyclin-dependent kinase 2 (PDB ID: 1W98). In the docked complex, the ligand molecules (AA derivatives) selectively bind with the target residues, and a high binding affinity of the ligand molecules was ensured by the full fitness score using the SwissDock Web server. The BOILED-Egg model shows that out of 17 scrutinized molecules, 3 molecules exhibit gastrointestinal absorption capability and 14 molecules exhibit permeability through the blood–brain barrier penetration. The analysis can also provide some useful insights to chemists to modify the existing natural scaffolds in designing new anacardic anticancer drugs. The increased probability of success may lead to the identification of drug-like candidates with favorable safety profiles after further clinical evaluation.

1. Introduction

ARTICLE SECTIONS
Jump To

Cancer is one of the major concerns in the medical field as it causes increasing number of deaths day by day. (1) In the past decades, there have been numerous anticancer drugs synthesized. However, tumor recurrence and the adverse effects of the drugs because of toxic intolerance are the key problems in the treatment of cancer. Thus, there is an urgent and desperate need of finding anticancerous drug molecules with negligible side effects and high efficacy. The naturally occurring products have received special attention toward the preparation of novel cancer preventives and therapeutic agents. (2−4) Particularly, more natural products were identified with potential anticancer activity. (5,6) In the present work, virtual screening and suitability of anacardic acid (AA) derivatives that inhibit the anticancer target cyclin-dependent kinase 2 (PDB ID: 1W98) were studied. AA originates from the nutshells of Anacardium occidentale (cashew) (7) and found to act as an antitumor, antibiotic, antioxidant, and gastroprotective agent. (8) It is a combination of numerous closely related organic compounds, each consisting of salicylic acid substituted with an alkyl chain. In addition, it is used as a synthon for the production of a variety of biologically active compounds and a capping agent for the development of nanomaterials. (9) For the treatment of the most serious pathophysiological disorders like cancer, oxidative damage, inflammation, and obesity, AA and its derivatives have been strongly supported as therapeutic agents. (7) Recent studies show that AA derivatives exert their action in treating ovarian cancer, prostate cancer, breast carcinoma, and lung carcinoma through various mechanisms. (10−13) AA acts as a potential inhibitor of histone acetyltransferase activity and sensitizes tumor cells to ionizing radiation. (13) With the anticipation that AA can be eventually applied in the medicinal industry after successful preclinical trials, Xiu et al. have investigated the role of AA in cancer. (14)
However, during the clearance of the molecule under clinical trials, wastage of the drugs and human efforts is unavoidable. In this regard, computation methods were developed to predict the human pharmacokinetic properties. With different levels of complexity for the screening of large data set compounds, a variety of useful in silico ADME (absorption, distribution, metabolism, excretion) models have been established. Nowadays, the in silico tools are faster, simpler, and more cost-effective than transitional investigational procedures. (15) At present, because of the toxicity or lag of optimal pharmacokinetics properties, large attrition rates of preclinical and clinical candidates were observed in pharmaceutical industry. It is possible for medicinal chemists to control the pharmacokinetic and toxicity properties of a molecule through the structural modifications. (16) Hence, in the present work, physicochemical, ADME attributes and in silico toxicity (17) of 100 AA derivatives were evaluated, and the results were compared with well-known anticancer drugs. The study would definitely help to identify the suitable oral AA-based drug molecules based on the abovementioned properties and their binding activities with the target cyclin-dependent kinase 2. Figure 1 shows the schematic diagram of the work done in which the selection of compounds and screening methods were illustrated.

Figure 1

Figure 1. Flowchart illustrating the selection of AA derivatives, screening process, and their interactions with the target (PDB ID: 1W98).

2. Materials and Methods

ARTICLE SECTIONS
Jump To

One of the main causes of drug development termination is poor pharmacokinetic properties. Less or no toxicity, good oral bioavailability, and optimum values of physicochemical properties are the key parameters for the discovery of anticancer drugs. In the present work, 100 AA derivatives were selected from the previous literatures (8−14,18,19) for the evaluation of their pharmacokinetic properties. Among these, the derivatives AA1–AA25 were obtained by alkyl chain modifications, whereas AA26–AA50 were obtained by modifying both the alkyl chain and functional groups (Figures S1 and S2). AA51–AA100 derivatives were obtained by introducing functional groups like hydroxyl (−OH) and carboxylic (−COOH) groups (Figures S3 and S4). A complete list of AA derivatives used in this study is depicted in Table S1, whereas the name and structures of the finally scrutinized molecules are depicted in Table 1 and the physicochemical properties of these derivatives are listed in Tables 26.
Table 1. Names and Structures of the Screened Out Derivatives
Table 2. Important Computed Physicochemical Properties of the Screened Out AA Derivativesa
AA derivatives MW nRot HBA HBD TPSA M log P Ali log S MR
AA11 328.4 9 4 2 66.76 3.66 –6.79 95.4
AA12 302.32 7 5 2 75.99 2.41 –5.19 82.51
AA16 264.36 9 3 2 57.53 3.55 –6.62 78.85
AA20 274.27 4 5 4 97.99 1.91 –4.88 73.73
AA21 242.27 4 3 2 57.53 3.06 –4.76 69.68
AA22 290.27 4 6 5 118.22 1.37 –4.94 75.75
AA23 220.26 3 3 2 57.53 2.39 –5.01 62.31
AA24 238.28 7 4 3 77.76 1.93 –4.32 65.59
AA25 264.32 9 4 2 74.6 2.37 –5.12 74.24
AA33 266.33 9 4 2 66.76 2.45 –4.8 74.71
AA34 294.39 11 4 2 66.76 2.95 –5.92 84.33
AA72 426.59 5 3 1 34.15 4.21 –5.3 127.19
AA75 388.51 3 4 0 52.83 4.4 –5.08 109.46
AA95 262.34 1 3 0 35.53 3.16 –5.13 75.91
AA96 260.33 1 3 0 35.53 3.08 –4.83 76.23
AA97 276.33 1 4 0 48.06 2.31 –3.69 74.88
AA98 276.33 1 4 0 52.6 2.23 –3.54 76.11
a

Optimal range: molecular weight (MW) ≤ 600, lipophilicity log or Moriguchi octane–water partition coefficient (M log P) ≤ 5, aqueous solubility descriptor (Ali log S) ≤ 0, hydrogen-bonded acceptor (HBA) ≤ 10, hydrogen-bonded donor (HBD) ≤ 5, topological polar surface area (TPSA) ≤ 150 Å2, number of rotatable bonds (nRot) ≤ 10, and molar refractivity (MR) ≤ 155.

Table 3. Important Computed ADMET Properties of the Screened Out AA Derivativesa
AA derivatives Caco2 permeability (log Papp in 10–6 cm/s) intestinal absorption (human) (% absorbed) VDss (human) (log L/kg) fraction unbound (human) P-gp substrate (yes/no)
AA11 0.684 98.43 –1.596 0.04 no
AA12 1.07 100 –1.485 0.125 yes
AA16 1.25 95.899 –1.496 0.232 no
AA20 0.873 56.592 –0.398 0.199 yes
AA21 1.249 96.995 –1.12 0.089 yes
AA22 0.305 46.001 –0.308 0.153 yes
AA23 1.222 89.976 –0.231 0.206 no
AA24 0.888 95.688 –1.723 0.41 no
AA25 0.92 97.934 –1.641 0.29 no
AA33 1.127 91.257 –0.035 0.166 yes
AA34 1.084 90.567 0.014 0.113 yes
AA72 1.314 97.793 1.026 0.007 yes
AA75 1.568 100 –0.158 0 no
AA95 1.031 96.303 0.107 0.268 no
AA96 1.778 94.729 0.161 0.202 no
AA97 1.386 95.664 0.083 0.225 no
AA98 1.351 97.828 –0.082 0.249 no
a

Caco-2 cell permeability (log Papp in 10–6 cm/s >0.09); intestinal absorption (human), % absorbed (>30); VDss (human) (log L/kg) (low if <−0.15 and high if >0.45).

Table 4. Important Computed Physicochemical Properties of Some Marketed Anticancer Drugs
Drugs MW nRot HBA HBD TPSA M log P Ali log S MR
Abemaciclib 502.63 7 7 1 75 2.87 –5.38 154.18
Ambochlorin 293.37 4 4 0 78.29 1.71 –3.3 83.81
Anastrozole 276.21 5 6 1 74.92 2.03 –4.6 64.19
Capecitabine 317.22 3 7 1 95.23 1.43 –3.63 77.26
Erivedge (Vismodegib) 421.3 5 4 1 84.51 3.24 –5.31 107
Flutamide 499.61 11 5 2 87.55 1.71 –5.25 150.43
Nelarabine 359.35 8 8 3 122.91 0.53 –2.71 85.25
Nilutamide 167.19 0 3 2 119.28 –0.96 –2.31 43.34
Osimertinib 297.27 3 8 4 148.77 –2.12 –1.61 69.17
Purinethol 304.21 9 2 1 40.54 3.29 –2.17 81.01
Table 5. Important Computed ADMET Properties for Some Marketed Anticancer Drugs
Drugs Caco2 permeability (log Papp in 10–6 cm/s) intestinal absorption (human) (% absorbed) VDss (human) (log L/kg) fraction unbound (human) P-gp substrate (yes/no)
Abemaciclib 1.38 88.951 0.535 0.285 yes
Ambochlorin 1.069 98.691 –0.03 0.138 no
Anastrazole 0.869 88.757 –0.118 0.029 yes
Capecitabine 1.2 87.504 –0.423 0.097 no
Erivedge (Vismodegib) 1.074 94.883 –0.075 0.127 no
Flutamide 0.811 95.992 1.093 0.145 yes
Nelarabine 0.319 51.344 –0.073 0.424 no
Nilutamide 1.207 84.051 –0.421 0.636 yes
Osimertinib –0.023 48.895 –0.013 0.841 no
Purinethol 1.439 92.268 –0.165 0.117 no
Table 6. Computed Safety End Points for AA Derivatives
Der. CYP2D6 inhibitor CYP3A4 inhibitor total clearance renal OCT2 substrate AMES toxicity hERG I inhibitor oral rat acute toxicity (LD50) oral rat chronic toxicity (LOAEL) hepatotoxicity skin sensitization
AA11 no no 0.658 no no no 2.901 2.237 no no
AA12 no no 0.544 no no no 2.919 2.074 no no
AA16 no no 1.312 no no no 2.53 2.759 no no
AA20 no no 0.505 no no no 2.353 1.774 no no
AA21 no no 0.656 no no no 2.673 2.888 no no
AA22 no no 0.333 no no no 2.317 3.039 no no
AA23 no no 0.551 no no no 2.432 2.207 no no
AA24 no no 0.672 no no no 2.324 2.445 no no
AA25 no no 1.265 no no no 2.554 2.247 no no
AA33 no no 0.828 no no 1.987 2.141 no no no
AA34 no no 1.488 no no no 2.097 2.192 no no
AA72 yes no 0.725 yes no no 2.054 1.463 yes no
AA75 no no 0.67 no no no 1.829 2.357 yes no
AA95 no no 1.351 no no no 2.012 2.044 no no
AA96 no no 0.619 no no no 2.228 1.871 no no
AA97 no no 1.174 no no no 2.15 1.701 no no
AA98 no no 0.645 no no no 2.231 1.756 no no
The ADME-related physicochemical properties of 100 AA derivatives including the marketed anticancer drugs were predicted by the Swiss ADME online Web server. (20) Two-dimensional structures were drawn with the help of Cambridge software, that is, ChemDraw Pro, version 12.0. A simplified molecular input line entry system of all AA derivatives was generated with the help of an online tool, that is, Swiss ADME. The physicochemical properties, lipophilicity and solubility, of the derivatives were considered for the analysis. (21)
In addition, the BOILED-Egg model of the molecules was predicted to reveal the capability of gastrointestinal (GI) absorption and permeability of the blood–brain penetration barrier. (22) The cutoff values of the physicochemical properties were set by certain rules, that is, Lipinski’s rule of five (ROF), bioavailability score, Ghose’s, and Veber’s rules. (23−25) For drug-likeliness, the molecular parameters such as MW (molecular weight), HBD (hydrogen bond donor), HBA (hydrogen bond acceptor), log P (lipophilicity log), log S (aqueous solubility), TPSA (topological polar surface area), MW, nRot (number of rotatable bonds), and MR (molar refractivity) were evaluated. For predicting the aforesaid properties, Swiss vector machine algorithm (26) is used.
There are two more important parameters, that is, plasma proteins (Fu) and volume of distribution (VDss), which help in the determination of the distribution of derivatives. The absorption, distribution, metabolism, excretion, and toxicity (ADMET) properties of all AA derivatives were analyzed through the online available tool pkSCM (http://bleoberis.bioc.cam.ac.uk/pkcsm/). (27)
In silico data corresponding to the major human cytochrome P450 (CYP) isoforms involved in drug metabolism, such as CYP2C9, CYP2D6, and CYP3A4, were also generated. To determine the excretion routes of AA derivatives, the total clearance and renal OCT2 substrate were predicted quantitatively. In drug attrition, the safety profile of the derivatives comes under major factors. (28) As per pharmacokinetic analysis, we assess some of the major toxicity end points of all derivatives. Moreover, certain safety parameters such as LD50, hepatotoxicity, skin sensitization, cellular toxicity, and hERG liability (inhibition of dofetilide binding) were also evaluated.
After the pharmacokinetic evaluation of all the AA derivatives, 17 AA molecules were chosen based on the cutoff values set by Lipinski’s ROF, bioavailability score, Ghose’s, and Veber’s rules. Finally, the chosen molecules were docked with cyclin-dependent kinase 2 to evaluate their binding affinities and active binding residues. For this process, the SwissDock docking server (http://www.swissdock.ch) is used. The preparation of the target for docking was done by UCSF chimera. Various docking parameters such as full fitness (FF, kcal/mol), total number of clusters, cluster rank, Gibbs free energy (ΔG), hydrogen bonding, deltaGligsolvpol, and so forth were analyzed.

3. Results and Discussion

ARTICLE SECTIONS
Jump To

3.1. Structural and Physicochemical Properties

The main emphasis has been on defining the physicochemical property rules for the derivatives to reduce attrition and increase the likelihood of them at various stages of anticancer drug development. Based on the gold standards and earlier published research on various oral bioavailability rules, 100 AA derivatives were evaluated together with 10 marketed anticancer drugs against the key parameters of physicochemical properties, that is, lipophilicity, log P, MW, TPSA, HBD, HBA, nRot, and aqueous solubility (log S). (23−25) All physicochemical properties are depicted in Table S1.
The MW range of AA1–AA25 derivatives varies from 220 to 376, AA26–AA50 derivatives varies from 264 to 518, and AA51–AA80 derivatives varies from 388 to 598, whereas the MW range for AA81–AA100 is much broader and varies from 260 to 597. The M log P value of AA1–AA25 derivatives varies from 1.4 to 6, AA26–AA50 derivatives varies from 2.4 to 5.3, and AA51–AA80 derivatives varies from 3.1 to 7.5, whereas AA81–AA100 derivatives show values varying from 2.2 to 6.4. There were no significant differences in the log P values among the four groups, although the alkyl-modified derivative set AA1–AA25 had a lower span of log P values; meanwhile, each set shows predicted values of some derivatives which are higher than the cutoff value set by the Lipinski rule, RO5 ≤ 4.15. This reflects the fact that molecules with out-of-range values of log P are poorly soluble in fats, oil, lipids, and nonpolar solvents. The predicted polarity (TPSA) ranges from 56 to 157 Å2 for AA1–AA25 derivatives, 20 to 120 Å2 for AA26–AA50 derivatives, 14 to 77 Å2 for AA51–AA80 derivatives, and 35 to 115 Å2 for AA81–AA100 derivatives. According to the cutoff values set for TPSA (≤140 Å2) by Veber et al., (24) 95% of the AA derivatives reported in this study are likely to have high probability of oral bioavailability. A minimal number of HBDs ranging from 0 to 5 was found in the AA derivatives.
All the 100 derivatives of AA in this study satisfy the rule of Lipinski, HBA < 10. One important parameter for the oral bioavailability is “MR” described by the Ghose filter. (25) According to Lipinski’s rule, the value of MR should lie between 40 and 130 (23) for drug-likeness. The derivatives AA1–AA50 show that all the predicted values fall within the range as described by the Ghose filter rule, whereas the derivative set AA50–AA100 shows 30% violation.
In order to accurately access drug-likeness, Lipinski’s RO5, Ghose filter, Vebers’s, Egan’s, and Muegge’s rules are compared with certain marketed drugs. The detailed data set of all AA derivatives with the violation of rules is shown in Figure 2.

Figure 2

Figure 2. Results of AA derivatives against different violations.

The graph shown in Figure 2a reveals that most of the derivatives show one violation of each rule, and few structures viz. AA11, AA12, AA16, and AA20–AA25 did not show any violation. Hence, they can be provisionally approved for preclinical trials. The structures that show three violations of the Muegge and Ghose filters must be excluded from further preclinical processes. In Figure 2b, it is clear that the structures AA33 and AA34 did not show any violation, and most of the derivatives violate more than one rule for each filter. The Lipinski RO5 rule graph as shown in Figure 2c,d suggests that the compounds AA72, AA75, and AA95–AA98 can be subjected for further preclinical trials. Out of 100 investigated compounds, only 17 compounds fall within the allowed range of physicochemical properties and satisfy all the key parameters of physicochemical properties.

3.2. Comparison of Physicochemical Properties of AA Derivatives with Marketed Anticancer Drugs

Our analysis shows that the optimal property ranges (covering 100 AA derivatives) are 200 < MW ≤ 600, 1 < M log P ≤ 5, −6 ≤ Ali log S ≤ 0, 2 ≤ HBA ≤ 10, 1 ≤ HBD ≤ 5, 50 ≤ TPSA ≤ 150 Å2, 0 ≤ nRot ≤ 10, and 40 ≤ MR ≤ 155. They may be very helpful in the prospective design of anticancer molecules. According to the RO5 rule, the key parameters of physicochemical properties showed better results as compared to the marketed anticancer drugs. The poor biopharmaceutical properties of the molecules, viz. poor aqueous solubility and slow dissolution rate resulted in poor oral bioavailability. In general, poor aqueous solubility is connected to high lipophilicity, and hydrophilic derivatives are connected to poor permeability, which means low absorption. Hence, ionization constants, solubility, and lipophilicity measurements are helpful in the high-throughput drug discovery paradigm. The relationship between lipophilicity and pharmacokinetic properties is explained in in silico studies. (29,30) In the present work, quantitative analysis shows that the finally scrutinized AA derivatives were predicted to have a higher optimum range of 13.18 for M log P, whereas the predicted values of M log P for the marketed anticancer drugs are in the range 2.12–5.6. Tables S1 and S2 depict the physicochemical and ADMET parameters of all the selected molecules.

3.3. BOILED-Egg for Prediction of GI Absorption and Brain Penetration

In the drug development process, GI absorption and blood–brain barrier (BBB) penetration play an important role. The BOILED-Egg model helps in the computation of polarity and lipophilicity of derivatives as it gives datasets with accuracy, speed, and clear graphical outputs. (22) This model helps in drug development by filtering the chemical libraries.
In the BOILED-Egg model, the high probability of passive absorption of the GI tract is represented by the white region, whereas the yellow region (yolk) represents the high probability of the BBB penetration. In addition, the blue color indicator of the molecule shows that the molecule is actively effluxed by P-glycoprotein, represented as (PGP+), whereas the red color indicator shows the nonsubstrate of P-gp, represented as (PGP).
The analysis predicts that the molecules AA1–AA25 (Figure S5a) show high GI absorption, with nearly 40% of the molecules also exhibiting BBB penetration, and all of them are predicted as the nonsubstrates of P-gp (PGP); 90% of the molecules AA26–AA50 (Figure S5b) show GI absorption, with nearly 40% of them also exhibiting BBB penetration. The BOILED-Egg plot of all the AA derivatives is presented in Figure S5. The predicted results of the derivatives AA51–AA80 (Figure S5c) consisting of benzamide and benzyl amine are shown. All the benzamide-based AA molecules and most of the benzyl amine-substituted AA derivatives show low GI absorption, and they do not exhibit significant BBB permeability. Most of the derivatives in the molecule set AA5–AA80 are P-gp substrates. The prediction of the BOILED-Egg results of the molecule set AA81–AA100 (Figure S5d), which includes thiazole, imidazole, oxazole, sildenafil, dihydropyridine, and macrolide AA derivatives, shows low GI absorption, and they do not exhibit BBB permeation. The BOILED-Egg model prediction of GI absorption and BBB permeation of the finally scrutinized 17 AA derivatives is presented in Figure 3.

Figure 3

Figure 3. BOILED-Egg model of AA derivatives.

3.4. Prediction of ADMET and Related Properties

For the development of a drug, the derivatives should have good ADMET profile. With the help of significant advancement in high-throughput in vitro ADME, computational scientists were able to predict the potential liabilities, that is, susceptibility to efflux transporters, low permeability, and so forth, of derivatives, which are associated with new potential derivatives. For the assessment of Caco2 cell permeability, P-gp efflux liability, and human intestinal absorption of anacardic derivatives, we have evaluated the ADME properties (in silico profiling). (31) On the basis of the ADME model, we can evaluate the permeability of a molecule, whether it is low or high. The permeability, that is, log Papp (10–6 cm/s) rate, is considered to be high if log Papp > 0.9 and considered to be low if log Papp < 0.9. The Caco2 cell permeability of AA derivatives is mentioned in Table S2.
Of the AA derivatives, 80% show high log Papp values. Similar results were also analyzed in the assessment of P-gp efflux liabilities of AA derivatives. The assessment of P-gp efflux liabilities was done with the help of preADMET’s [https://preadmet.bmdrc.kr/] P-gp substrate model. Only 25% of anacardic derivatives were found as P-gp efflux substrates. The predicted ADMET parameters of AA derivatives are listed in Table S2. For the achievement of an optimal clinical drug/derivative, a derivative should have high log Papp and low P-gp efflux liability.
In addition, we have calculated the total human intestinal absorption percentage (%). For the GI tract dissolution and stability, a systematic oral dosage should have pH 1–2 in fasted state and 3–7 in fed state in stomach, including a neutral environment of small intestine, that is, pH 4.4–6.6. (31) The human intestinal absorption percentage (%) of the AA derivatives was analyzed with the help of an online server tool, pkCSM. (27) Except two AA molecules (AA99 and AA100), the rest of the other AA derivatives shows good human intestinal absorption. The predicted human intestinal absorption percentage (%) values for all the AA molecules are listed in Table S2.
In plasma, almost all drugs exist in equilibrium between the bound and unbound states with serum proteins at different affinities. Only an unbound drug can show interactions with anticipated molecular targets. (22) Therefore, the efficiency of a drug is affected by the binding efficacy of the drug with whole blood proteins. With the help of the predictive model of pkCSM, we have evaluated the fraction unbound (Fu) and steady-state volume of distribution (VDss) of 100 AA derivatives. VDss is a key parameter, which helps in suggestion for the total dose of a drug. The predicted values of Fu and VDss of all AA derivatives are listed in Table S2.
For drug designing and screening of new chemical drugs, pharmaceutical industries use in silico studies for early prediction. (32) For evaluating AA derivatives as anticancer drugs, we have evaluated the toxicity end points with the help of the pkCSM tool. The toxicity end points consist of the inhibition of cytochrome P450 (CYPs) monooxygenase enzymes. (33) AMES toxicity, LD50 (lethal rat acute toxicity), hepatotoxicity, skin sensitization, and inhibition of hERG potassium ion channel effects are determined for the evaluation of drug–drug interactions (DDIs). (34) The toxicity data sets of all AA derivatives are listed in Table S3. With the help of pkCSM, inhibitions of CYP2D6 and CYP3A4 were qualitatively predicted and the results are depicted in Table S3. Out of 100 AA derivatives, 80% AA derivatives were evaluated as noninhibitors of CYP3A4, and only two AA derivatives show inhibition against CYP2D6. After the evaluation of these parameters, drug excretion and drug metabolism were evaluated. With the prior knowledge of metabolic pathways of drugs, one can easily predict DDIs, pharmacokinetics, and toxicities. (35) Here, our primary concern is the inhibition of CYP3A4, which helps in finding out the correlation with increasing MW and log P. (36) Inhibition of CYP3A4 is related to issues with DDIs and clearance. The total clearance of 100 AA derivatives was measured, which is a combination of hepatic and renal clearance, and is listed in Table S3.
The major evaluation that emerged from these data was that the derivatives showing clog P < 3 and TPSA > 75 Å2 were 2.5 times more nontoxic, whereas derivatives showing high lipophilicity (clog P > 3) and low polar surface area (TPSA < 75 Å2) are considered to be highly toxic in short-term animal studies. Hence, we have evaluated the lipophilicity of AA derivatives. Lipophilicity with small polar functionalities has high chances of becoming toxic. From the evaluation, it is revealed that toxicity occurs when log P > 3; on the other hand, the value of TPSA shows very little or no influence on the drug toxicity. Hence, it is clear that the early prediction of log P and TPSA is very helpful in the drug designing and development process in order to avoid wastage of chemicals.
Establishment of meaningful correlations between the physicochemical properties and ADMET properties of AA derivatives might be useful for future anticancer drug discovery. Based on our analysis, the results of AA derivatives show good and accepted prediction of physicochemical properties in comparison to Lipinski’s RO5, Ghose filter, Vebers’s, Egan’s, and Muegge’s rules for oral bioavailable drugs and in comparison with the physicochemical properties of marketed anticancer drugs. The correlation of the results of the accepted physicochemical properties with the predicted ADMET results shows that only seven AA derivatives, AA11, AA22, AA24, AA25, AA95, AA97, and AA98, fall within the accepted range of the parameters of ADMET. However, the docking studies of the finally scrutinized 17 derivatives were investigated, as they acquire adequate pharmacokinetic properties for oral bioavailability.

3.5. Molecular Docking Studies

Molecular docking studies were analyzed for exploring the interaction mechanism between the receptor sites and inhibitors. (37) In the field of drug discovery, the prediction of interactions between molecules and their targets has a great importance. One can easily find out the mechanisms of selectivity by the docking of molecules with protein targets. (38−40) In this work, the activities of anacardic derivatives were screened out against the target cyclin-dependent kinase 2 (PDB ID: 1w98). Cyclin-dependent kinase 2, also known as cell-division protein kinase 2, is an enzyme usually found in humans. It is encoded as CDK2 gene. It is necessary for cell cycle progression and mostly overexpressed in cancer cells. By using the SwissDock Web server (http://www.swissdock.ch), molecular docking of all the filtered 17 AA derivatives (AA11, AA12, AA16, AA20–AA25, AA33, AA34, AA72, AA75, AA95, AA96, AA97, and AA98) was performed against human cyclins or cell division protein kinase 2 (PDB ID: 1w98). (41) For the cell cycle, cyclins are essential proteins. Perturbation of the cyclin function can cause cancer formation. The structural factors and atomic coordinates of the protein cyclin were deposited. (42) The results pointed out that the filtered AA derivatives form conventional bonds with the receptor sites. Different parameters like FF (kcal/mol), ΔG (Gibb’s free energy), ligand solvation energy, hydrogen bonding (interactions), and so forth of docking were analyzed. On the basis of FF and cluster formation, all suitable binding modes were analyzed. (43) All the output clusters were ranked in accordance with hydrogen bonding (interactions) and the FF score. A cluster rank “0” is considered to be the best FF score. A molecule is said to have more favorable binding modes with a better fit if the FF score shows greater negative value. (44) The docked conformations having the lowest binding energy were selected for discussion. (45) However, for the most favorable interaction, if a molecule has a small docking score, then it is said to have a higher binding affinity of ligands for a particular receptor. (46) The FF score, binding affinity, and active residues of docked conformations are depicted in Table 7. Out of 17 docked complexes, the derivatives AA20, AA21, and AA22 show three hydrogen-bonding interactions with the target residues, whereas the derivative AA75 shows the maximum FF score (−3054.45) with one hydrogen-bonding interaction with LEU 2.418 Å. The docked conformations of 17 AA derivatives and the target (PDB ID: 1w98) are presented in Figure 4.

Figure 4

Figure 4. Docked ligands with target-1W98: (a) AA11, (b) AA12, (c) AA16, (d) AA20, (e) AA21, (f) AA22 (g) AA23, (h) AA24, (i) AA25 (j) AA33, (k) AA34, (l) AA72 (m) AA75, (n) AA95, (o) AA96, (p) AA97, and (q) AA98.

Table 7. Results of Docking of AA Derivatives with 1W98
Der. docked with 1W98 ΔG deltaGligsolvpol FF (kcal/mol) energy (protein···ligand) sites type/binding residue/H-bonding distance
AA11 –7.2239 –9.9810 –3029.41 7.2787 N–H···O/GLN174/2.345 Å
          O···H/LYS170/2.344 Å
AA12 –7.5062 –11.9606 –3014.64 21.7154 N–H···O/GLN174/2.359 Å
          O···H/LYS170/2.155 Å
AA16 –6.3474 –7.9222 –3040.32 –9.9220 O···H/LEU113/2.618 Å
          N–H···O/LEU113/2.256 Å
AA20 –6.8278 –16.5444 –3045.13 7.2032 O···H/LEU277/1.953 Å
          O···H/TYR112/2.301 Å
          O···H/GLU109/2.646 Å
AA21 –6.5128 –8.9698 –3018.17 16.201 O···H/LYS170/2.013 Å
          O···H/THR171/2.575 Å
          N–H···O/GLN174/2.523 Å
AA22 –7.8004 –19.1000 –3041.91 5.6960 O···H/LEU277/2.19 Å
          O···H/GLU109/2.508 Å
          O···H/GLU278/2.478 Å
AA23 –6.7354 –7.7501 –3035.65 –1.1662 O···H/THR171/2.481 Å
AA24 –6.3980 –10.9386 –3038.37 –3.4394 O···H/GLU109/2.562 Å
          O···H/GLU109/2.310 Å
AA25 –6.9110 –11.2838 –3049.17 –12.0395 O···H/LYS170/2.512 Å
          N–H···O/GLN174/2.393 Å
AA33 –7.1697 –6.6352 –3034.61 –4.6578 O···H/THR171/2.150 Å
AA34 –7.3850 –7.5994 –3031.71 –6.3850 O···H/LYS170/2.586 Å
AA72 –7.8560 –7.7790 –3020.07 –4.9699 O···H/THR202/2.302 Å
AA75 –7.5994 –7.5706 –3054.45 –28.0006 N–H···N/LEU113/2.418 Å
AA95 –6.2015 –4.3915 –3006.90 16.0120 N–H···O/ARG114/2.275 Å
AA96 –6.4796 –4.7592 –3008.22 15.3224 N–H···O/ASN74/2.606 Å
AA97 –6.4855 –6.0909 –2787.35 240.0530 N–H···O/LEU113/2.580 Å
AA98 –6.4602 –7.4286 –3010.43 11.7630 N–H···O/ARG114/2.472 Å

4. Conclusions

ARTICLE SECTIONS
Jump To

For the drug designing and development process, the evaluation of pharmacokinetic and physicochemical properties is the primary task. In the present work, 100 AA derivatives were evaluated with the in silico screening procedure based on ADME parameters. The results show the optimal property range (covering 100 derivatives of AA) used to select oral bioavailable drugs. From the evaluation of in silico computational studies including ADME, the pkCSM data revealed that 80% of the selected AA derivatives in this work have shown high Caco2 permeability of log Papp <0.9 and 25% of the AA derivatives were found as P-gp efflux substrates with low-to-moderate clearance rates. The docking results of the finally scrutinized 17 derivatives which were allowed to dock with the anticancer target cyclin-dependent kinase 2 reveal that the AA derivatives are strongly bound with the targets and inhibit the target through hydrogen-bonding interactions. Better bioactivity score, drug adsorption ability, and drug-likeness of these derivatives pave a new research arena with vegetable feedstocks. Rege et al. have recently explored the dual functionality of certain AA derivatives for efficient paclitaxel delivery in breast cancer therapy. (47,48) The higher binding affinity results of the 17 AA derivatives with the anticancer target (PDB ID: 1W98) and their in silico pharmacokinetic studies would definitely provide motivation to biochemists for performing further wet lab and clinical evaluations.

Supporting Information

ARTICLE SECTIONS
Jump To

The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acsomega.9b04398.

  • Chemical structures of alkyl chain-modified AA derivatives (labeled from 1 to 29); chemical structures of alkyl chain- and functional group-modified AA derivatives labeled from 30 to 49; chemical structures of alkyl chain- and functional group-modified AA derivatives labeled from 50 to 76; chemical structures of alkyl chain- and functional group-modified AA derivatives labeled from 77 to 100; BOILED-Egg model of AA1–AA100 derivatives; important computed physicochemical properties for AA1–AA100; important computed ADMET properties for AA1–AA100 derivatives; and computed safety end points for AA1–AA100 derivatives (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
    • Fahmina Zafar - Inorganic Materials Research Laboratory, Department of Chemistry, Jamia Millia Islamia, New Delhi 110025, IndiaOrcidhttp://orcid.org/0000-0001-5454-3981 Email: [email protected]
    • Anjali Gupta - Division of Chemistry, School of Basic and Applied Science, Galgotias University, Greater Noida 201310, Uttar Pradesh, India Email: [email protected]
    • Nahid Nishat - Inorganic Materials Research Laboratory, Department of Chemistry, Jamia Millia Islamia, New Delhi 110025, India Email: [email protected]
  • Authors
    • Karthick Thangavel - Department of Physics, School of Electrical and Electronics Engineering, SASTRA Deemed University, Thanjavur 613 401, Tamil Nadu, India
    • Kavita Khatana - Division of Chemistry, School of Basic and Applied Science, Galgotias University, Greater Noida 201310, Uttar Pradesh, India
    • Ali Alhaji Sani - Division of Chemistry, School of Basic and Applied Science, Galgotias University, Greater Noida 201310, Uttar Pradesh, India
    • Anujit Ghosal - Inorganic Materials Research Laboratory, Department of Chemistry, Jamia Millia Islamia, New Delhi 110025, IndiaDivision of Chemistry, School of Basic and Applied Science, Galgotias University, Greater Noida 201310, Uttar Pradesh, IndiaSchool of Life Sciences, Beijing Institute of Technology, Beijing 100811, China
    • Poonam Tandon - Department of Physics, University of Lucknow, Lucknow 226007, IndiaOrcidhttp://orcid.org/0000-0002-8120-0498
  • Notes
    The authors declare no competing financial interest.

Acknowledgments

ARTICLE SECTIONS
Jump To

F.Z. acknowledges Department of Science and Technology, New Delhi, India, for the Women Scientist Scheme (WOS) for Research in Basic/Applied Sciences, Rf# SR/WOSA/CS-97/2016. The authors are thankful to the Head, Department of Chemistry, Jamia Millia Islamia (JMI), and Galgotias University, for providing facilities to carry out the work.

References

ARTICLE SECTIONS
Jump To

This article references 48 other publications.

  1. 1
    Siegel, R.; Ma, J.; Zou, Z.; Jemal, A. “Cancer statistics”. Ca-Cancer J. Clin. 2014, 64, 929,  DOI: 10.3322/caac.21208
  2. 2
    Newman, D. J. Natural products as leads to potential drugs: An old process or the new hope for drug discovery?. J. Med. Chem. 2008, 51, 25892599,  DOI: 10.1021/jm0704090
  3. 3
    Elmsellem, H.; El Ouadi, Y.; Mokhtari, M.; Bendaif, H.; Steli, H.; Aouniti, A.; Almehdi, A. M.; Abdel-Rahman, I.; Kusuma, H. S.; Hammouti, B. A natural antioxidant and an environmentally friendly inhibitor of mild steel corrosion: A commercial oil of basil (Ocimum Basilicum L.). J. Chem. Technol. Metall. 2019, 54, 742749
  4. 4
    Moussaoui, El. A.; Jawhari, F. Z.; Almehdi, A. M.; Elmsellem, H.; Benbrahim, K. F.; Bousta, D.; Bari, A. Antibacterial, antifungal and antioxidant activity of total polyphenols of Withania frutescens. L. Bioorg. Chem. 2019, 93, 103337,  DOI: 10.1016/j.bioorg.2019.103337
  5. 5
    Lichota, A.; Gwozdzinski, K. Anticancer activity of natural compounds from Plant and marine environment. Int. J. Mol. Sci. 2018, 19, 3533,  DOI: 10.3390/ijms19113533
  6. 6
    Rayan, A.; Raiyn, J.; Falah, M. Nature is the best source of anticancer drugs: Indexing natural products for their anticancer bioactivity. PLoS One 2017, 12, e0187925  DOI: 10.1371/journal.pone.0187925
  7. 7
    Newman, D. J.; Cragg, G. M. Natural Products As Sources of New Drugs over the 30 Years from 1981 to 2010. J. Nat. Prod. 2012, 75, 311335,  DOI: 10.1021/np200906s
  8. 8
    Hemshekhar, M.; Santhosh, M. S.; Kemparaju, K.; Girish, K. S. Emerging roles of anacardic acid and its derivatives: A pharmacological overview. Basic Clin. Pharmacol. Toxicol. 2012, 110, 122132,  DOI: 10.1111/j.1742-7843.2011.00833.x
  9. 9
    Hamad, F.; Mubofu, E. Potential biological applications of bio-based anacardic acids and their derivatives. Int. J. Mol. Sci. 2015, 16, 85698590,  DOI: 10.3390/ijms16048569
  10. 10
    Xiu, Y.-L.; Zhao, Y.; Gou, W.-F.; Chen, S.; Takano, Y.; Zheng, H.-C. Anacardic acid enhances the proliferation of human ovarian cancer cells. PLoS One 2014, 9, e99361  DOI: 10.1371/journal.pone.0099361
  11. 11
    Sung, B.; Pandey, M. K.; Ahn, K. S.; Yi, T.; Chaturvedi, M. M.; Liu, M.; Aggarwal, B. B. Anacardic acid (6-nonadecyl salicylic acid), an inhibitor of histone acetyltransferase, suppresses expression of nuclear factor-κB-regulated gene products involved in cell survival, proliferation, invasion, and inflammation through inhibition of the inhibitory subunit of nuclear factor-κBα kinase, leading to potentiation of apoptosis. Blood 2008, 111, 48804891,  DOI: 10.1182/blood-2007-10-117994
  12. 12
    Tan, J.; Chen, B.; He, L.; Tang, Y.; Jiang, Z.; Yin, G.; Wang, J.; Jiang, X. Anacardic acid (6-pentadecylsalicylic acid) induces apoptosis of prostate cancer cells through inhibition of androgen receptor and activation of p53 signaling. Chin. J. Cancer Res. 2012, 24, 275283,  DOI: 10.1007/s11670-012-0264-y
  13. 13
    Sun, Y.; Jiang, X.; Chen, S.; Price, B. D. Inhibition of histone acetyltransferase activity by anacardic acid sensitizes tumor cells to ionizing radiation. FEBS Lett. 2006, 580, 43534356,  DOI: 10.1016/j.febslet.2006.06.092
  14. 14
    Xiu, Y.-L.; Zhao, Y.; Gou, W.-F.; Chen, S.; Takano, Y.; Zheng, H.-C. Anacardic acid enhances the proliferation of human ovarian cancer cells. PLoS One 2014, 9, e99361  DOI: 10.1371/journal.pone.0099361
  15. 15
    Moda, T. L.; Torres, L. G.; Carrara, A. E.; Andricopulo, A. D. PK/DB: Database for pharmacokinetic properties and predictive in silico ADME models. Bioinformatics 2008, 24, 22702271,  DOI: 10.1093/bioinformatics/btn415
  16. 16
    van de Waterbeemd, H.; Gifford, E. ADMET in silico modelling: Towards prediction paradise?. Nat. Rev. Drug Discovery 2003, 2, 192204,  DOI: 10.1038/nrd1032
  17. 17
    Gleeson, M. P. Generation of a set of simple, interpretable ADMET rules of thumb. J. Med. Chem. 2008, 51, 817834,  DOI: 10.1021/jm701122q
  18. 18
    Green, I. R.; Tocoli, F. E.; Lee, S. H.; Nihei, K.-i.; Kubo, I. Design and evaluation of anacardic acid derivatives as anticavity agents. Eur. J. Med. Chem. 2008, 43, 13151320,  DOI: 10.1016/j.ejmech.2007.08.012
  19. 19
    Mamidyala, S. K.; Ramu, S.; Huang, J. X.; Robertson, A. A. B.; Cooper, M. A. Efficient synthesis of anacardic acid analogues and their antibacterial activities. Bioorg. Med. Chem. Lett. 2013, 23, 16671670,  DOI: 10.1016/j.bmcl.2013.01.074
  20. 20
    Zoete, V.; Daina, A.; Bovigny, C.; Michielin, O. SwissSimilarity: A Web Tool for Low to Ultra High Throughput Ligand-Based Virtual Screening. J. Chem. Inf. Model. 2016, 56, 13991404,  DOI: 10.1021/acs.jcim.6b00174
  21. 21
    Daina, A.; Michielin, O.; Zoete, V. SwissADME: A free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Sci. Rep. 2017, 7, 42717,  DOI: 10.1038/srep42717
  22. 22
    Daina, A.; Zoete, V. A BOILED-Egg To Predict Gastrointestinal Absorption and Brain Penetration of Small Molecules. ChemMedChem 2016, 11, 11171121,  DOI: 10.1002/cmdc.201600182
  23. 23
    Lipinski, C. A.; Lombardo, F.; Dominy, B. W.; Feeney, P. J. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv. Drug Delivery Rev. 2012, 64, 417,  DOI: 10.1016/j.addr.2012.09.019
  24. 24
    Veber, D. F.; Johnson, S. R.; Cheng, H.; Smith, B. R.; Ward, K. W.; Kopple, K. D. Molecular Properties That Influence the Oral Bioavailability of Drug Candidates. J. Med. Chem. 2002, 45, 26152623,  DOI: 10.1021/jm020017n
  25. 25
    Ghose, A. K.; Viswanadhan, V. N.; Wendoloski, J. J. A knowledge-based approach in designing combinatorial or medicinal chemistry libraries for drug discovery. 1. A qualitative and quantitative characterization of known drug databases. J. Comb. Chem. 1999, 1, 5568,  DOI: 10.1021/cc9800071
  26. 26
    Cortes, C.; Vapnik, V. Support-Vector Networks. Mach. Learn. 1995, 20, 273297,  DOI: 10.1023/a:1022627411411
  27. 27
    Pires, D. E. V.; Blundell, T. L.; Ascher, D. B. pkCSM: Predicting small-molecule pharmacokinetic and toxicity properties using graph-based signatures. J. Med. Chem. 2015, 58, 40664072,  DOI: 10.1021/acs.jmedchem.5b00104
  28. 28
    Pellicciari, A. Attrition in the Pharmaceutical Industry - Reasons, Implications, and Pathways Forward. Edited by Alexander Alex, C. John Harris, Dennis A. Smith. ChemMedChem 2017, 12, 10971098,  DOI: 10.1002/cmdc.201600621
  29. 29
    van deWaterbeemd, H.; Smith, D. A.; Jones, B. C. Lipophilicity in PK design: Methyl, ethyl, futile. J. Comput.-Aided Mol. Des. 2001, 15, 273286,  DOI: 10.1023/a:1008192010023
  30. 30
    Pliška, V.; Testa, B.; Waterbeemd, H. V. Lipophilicity in Drug Action and Toxicology; VCH Verlagsgesellschaft mbH: Weinheim (Federal Republic of Germany), 1996.
  31. 31
    Smith, D. A.; Di, L.; Kerns, E. H. The effect of plasma protein binding on in vivo efficacy: Misconceptions in drug discovery. Nat. Rev. Drug Discovery 2010, 9, 929939,  DOI: 10.1038/nrd3287
  32. 32
    Di, L.; Kerns, E. H. Drug-Like Properties: Concepts, Structure Design and Methods from ADME to Toxicity Optimization; Elsevier publisher, 2008.
  33. 33
    Khojasteh, S. C.; Wong, H.; Hop, C. E. C. A. Pharmacokinetics. Drug Metabolism and Pharmacokinetics Quick Guide; Springer, 2011; p 1.
  34. 34
    Li, A. P.; Jurima-Romet, M. Applications of primary human hepatocytes in the evaluation of pharmacokinetic drug-drug interactions: Evaluation of model drugs terfenadine and rifampin. Cell Biol. Toxicol. 1997, 13, 365374,  DOI: 10.1023/A:1007451911843
  35. 35
    Danielsson, B. R.; Lansdell, K.; Patmore, L.; Tomson, T. Phenytoin and phenobarbital inhibit human HERG potassium channels. Epilepsy Res. 2003, 55, 147157,  DOI: 10.1016/s0920-1211(03)00119-0
  36. 36
    Hosey, C. M.; Benet, L. Z. Predicting the extent of metabolism using in vitro permeability rate measurements and in silico permeability rate predictions. Mol. Pharm. 2015, 12, 14561466,  DOI: 10.1021/mp500783g
  37. 37
    Czechtizky, C.; Hamley, P. Small Molecule Medicinal Chemistry: Strategies and Technologies; John Wiley & Sons, 2015.
  38. 38
    Singh, U.; Gangwal, R. P.; Dhoke, G. V.; Prajapati, R.; Damre, M.; Sangamwar, A. T. 3D-QSAR and molecular docking analysis of (4-piperidinyl)-piperazines as acetyl-CoA carboxylases inhibitors. Arabian J. Chem. 2017, 10, S617S626,  DOI: 10.1016/j.arabjc.2012.10.023
  39. 39
    Bendaif, H.; Melhaoui, A.; Ramdani, M.; Elmsellem, H.; Douez, C.; El Ouadi, Y. Antibacterial activity and virtual screening by molecular docking of lycorine from Pancratium foetidum Pom (Moroccan endemic Amaryllidaceae). Microb. Pathog. 2018, 115, 138145,  DOI: 10.1016/j.micpath.2017.12.037
  40. 40
    Karthick, T.; Tandon, P. Computational approaches to find the active binding sites of biological targets against busulfan. J. Mol. Model. 2016, 22, 142,  DOI: 10.1007/s00894-016-3015-z
  41. 41
    Shaikh, S.; Shastri, S. L.; Naik, N. S.; Kulkarni, R.; Madar, J. M.; Shastri, L. A.; Joshi, S. D.; Sunagar, V. Synthesis, antitubercular and antimicrobial activity of 1,2,4-triazolidine-3-thione functionalized coumarin and phenyl derivatives and molecular docking studies. ChemistrySelect 2019, 4, 105115,  DOI: 10.1002/slct.201802395
  42. 42
    Sidhu, A.; Kukreja, S. Synthesis of novel fluorinated benzothiazol-2-yl-1,2,4-triazoles: Molecular docking, antifungal evaluation and in silico evaluation for SAR. Arabian J. Chem. 2019, 12, 2118,  DOI: 10.1016/j.arabjc.2015.01.009
  43. 43
    Kareem, A.; Nami, S. A. A.; Khan, M. S.; Bhat, S. A.; Mirza, A. U.; Nishat, N. Self-assembled transition metal dithiocarbamates of pyridine-3-carboxamide: synthesis, spectral characterization, thermal and biological studies. New J. Chem. 2019, 43, 4413,  DOI: 10.1039/c8nj03660h
  44. 44
    Kaviyarasi, N. S.; Prashantha, C. N.; Suryanarayana, V. V. S. In Silico Analysis of Inhibitor and Substrate Binding Site of Serrapeptidase from Serratia Marcescens MTCC 8708. Int. J. Pharmacol. Pharm. Sci. 2016, 8, 123128
  45. 45
    De Vita, D.; Pandolfi, F.; Ornano, L.; Feroci, M.; Chiarotto, I.; Sileno, I.; Pepi, F.; Costi, R.; Di Santo, R.; Scipione, L. New N,N-Dimethylcarbamate Inhibitors of Acetylcholinesterase: Design Synthesis and Biological Evaluation. J. Enzyme Inhib. Med. Chem. 2016, 31, 106113,  DOI: 10.1080/14756366.2016.1220377
  46. 46
    Iftikhar, H.; Batool, S.; Deep, A.; Narasimhan, B.; Sharma, P. C.; Malhotra, M. In Silico Analysis of the Inhibitory Activities of GABA Derivatives on 4-Aminobutyrate Transaminase. Arabian J. Chem. 2017, 10, S1267S1275,  DOI: 10.1016/j.arabjc.2013.03.007
  47. 47
    Chhajed, S. S.; Upasani, C. D. Synthesis, Biological Screening and Molecular Modeling Studies of Novel 3-Chloro-4-Substituted-1-(2-(1H-Benzimidazol-2-Yl)Phenyl))-Azetidin-2-Ones. J. Enzyme Inhib. Med. Chem. 2012, 27, 504,  DOI: 10.3109/14756366.2011.598867
  48. 48
    Rege, M. D.; Ghadi, R.; Katiyar, S. S.; Kushwah, V.; Jain, S. Exploring an interesting dual functionality of anacardic acid for efficient paclitaxel delivery in breast cancer therapy. Nanomedicine 2019, 14, 57,  DOI: 10.2217/nnm-2018-0138

Cited By

ARTICLE SECTIONS
Jump To

This article is cited by 41 publications.

  1. Melissa Mejia-Gutierrez, Bryan D. Vásquez-Paz, Leonardo Fierro, Julio R. Maza. In Silico Repositioning of Dopamine Modulators with Possible Application to Schizophrenia: Pharmacophore Mapping, Molecular Docking and Molecular Dynamics Analysis. ACS Omega 2021, 6 (23) , 14748-14764. https://doi.org/10.1021/acsomega.0c05984
  2. Khalifa Sunusi Aminu, Adamu Uzairu, Anshuman Chandra, Nagendra Singh, Stephen Eyije Abechi, Gideon Adamu Shallangwa, Abdullahi Bello Umar. Exploring the potential of 2-arylbenzimidazole scaffolds as novel α-amylase inhibitors: QSAR, molecular docking, simulation and pharmacokinetic studies. In Silico Pharmacology 2024, 12 (1) https://doi.org/10.1007/s40203-024-00205-4
  3. Ali Al-Samydai, Hamdi Nsairat, Maha N. Abu Hajleh, Bayan Aburas, Amal Akour, Tha’er Husam Ata, Tabarek H. Mahmood, Tabarak Riyadh Al-sammarraie, Rawan Atta, Rahaf Ali, Qasim Khalid Alazzawi, Ahmed Aburjai, Simone Carradori. Meta-analysis of nano-phytosomes: unleashing the potential of plant-derived compounds for advancing cancer therapy. Natural Product Research 2024, , 1-20. https://doi.org/10.1080/14786419.2024.2344182
  4. Kavita Khatana, Anjali Gupta, Anujit Ghosal, Pinki Dey, Fahmina Zafar, Anubha Srivastava, Priya Verma. In silico identification and validation of phenolic lipids as potential inhibitor against bacterial and viral strains. Journal of Biomolecular Structure and Dynamics 2024, 42 (5) , 2525-2538. https://doi.org/10.1080/07391102.2023.2212811
  5. Saudatu Chinade Ja’afaru, Adamu Uzairu, Muhammed Sani Sallau, George Iloegbulam Ndukwe, Muhammad Tukur Ibrahim, Ammar Usman Danazumi. Structure-Based Design of Potential Anti-schistosomiasis Agent Targeting SmHDAC8: An In Silico Approach Utilizing QSAR, MD Simulation and ADMET Prediction. Chemistry Africa 2024, 7 (2) , 725-745. https://doi.org/10.1007/s42250-023-00777-2
  6. Kavita Khatana, Anjali Gupta, Anujit Ghosal, Fahmina Zafar. Computational/In‐Silico Methods: An Influential Approach for Drug Designing and Development. Macromolecular Symposia 2024, 413 (1) https://doi.org/10.1002/masy.202300022
  7. Lalita Dahiya, Rajiv Kumar, Anurag T. K. Baidya, Sunil Kumar, Rajnish Kumar, Sandip V. Pawar, Ashok Kumar Yadav. Design, synthesis, biological evaluations and in silico studies of N-substituted 2,4-thiazolidinedione derivatives as potential a-glucosidase inhibitors. Journal of Biomolecular Structure and Dynamics 2023, 27 , 1-18. https://doi.org/10.1080/07391102.2023.2291158
  8. Sajad Ahmad Bhat, Syed Kazim Hasan, Zahoor Ahmad Parray, Zaheenul Islam Siddiqui, Shabnam Ansari, Ayesha Anwer, Saniya Khan, Fatima Amir, Mahboubeh Mehmankhah, Asimul Islam, Zarrin Minuchehr, Syed Naqui Kazim. Potential antiviral activities of chrysin against hepatitis B virus. Gut Pathogens 2023, 15 (1) https://doi.org/10.1186/s13099-023-00531-6
  9. Taye Temitope Alawode, Labunmi Lajide, Mary Olaleye, Bodunde Owolabi. Crinum jagus: antiproliferative studies of extracts on HepG2 cell line and in silico assessment of phytoconstituents as potential inhibitors of p53–mortalin interaction. Future Journal of Pharmaceutical Sciences 2023, 9 (1) https://doi.org/10.1186/s43094-023-00490-4
  10. Bichun Guo, Sainan Zhang, Shanshan Wang, Huidan Zhang, Junshun Fang, Nannan Kang, Xin Zhen, Yang Zhang, Jidong Zhou, Guijun Yan, Haixiang Sun, Lijun Ding, Chuanming Liu. Decreased HAT1 expression in granulosa cells disturbs oocyte meiosis during mouse ovarian aging. Reproductive Biology and Endocrinology 2023, 21 (1) https://doi.org/10.1186/s12958-023-01147-w
  11. Zakari Y. Ibrahim, Adamu Uzairu, Gideon A. Shallangwa, Stephen E. Abechi, Sulaiman Isyaku. Homology modeling, docking, and ADMET studies of benzoheterocyclic 4-aminoquinolines analogs as inhibitors of Plasmodium falciparum. Journal of Taibah University Medical Sciences 2023, 18 (6) , 1200-1216. https://doi.org/10.1016/j.jtumed.2023.04.005
  12. Hanadi A. Katouah. Synthesis, Antioxidant, and Cytotoxic Activities of New 1,3,4-Thiadiazoldiazenylacrylonitrile Derivatives. Polycyclic Aromatic Compounds 2023, 43 (9) , 7808-7827. https://doi.org/10.1080/10406638.2022.2140172
  13. Kotyada Suryanarayana, Kranthi Kumar Gangu, Nagaraju Kerru, Kavita Khatana, Botcha Satyanarayana, Suresh Maddila. Synthesis of a novel chloroquinoline, rhodanine encompassed 1,2,3-triazole scaffolds and molecular docking evaluation of their cytotoxicity. Journal of the Iranian Chemical Society 2023, 20 (10) , 2643-2655. https://doi.org/10.1007/s13738-023-02862-2
  14. Godfried Dougnon, Michiho Ito. Molecular Descriptors and QSAR Models for Sedative Activity of Sesquiterpenes Administered to Mice via Inhalation. Planta Medica 2023, 89 (13) , 1236-1249. https://doi.org/10.1055/a-1770-7581
  15. Md Meraj Anjum, Dulla Naveen Kumar, Sankha Bhattacharya, Krishna Kumar Patel, Mahalingam Rajamanickam Vijayakumar, Ashish Kumar Agrawal, Sanjay Singh. Topical delivery of cyclodextrin crosslinked nanosponge of anacardic acid for treatment of UV-B induced skin photoaging: Formulation, characterization and biochemical estimation. Journal of Drug Delivery Science and Technology 2023, 87 , 104840. https://doi.org/10.1016/j.jddst.2023.104840
  16. D C Vinay Kumar, B S Chethan, Darshini Gowda, K S Rangappa, N K Lokanath. Investigation of the crystal structure, supramolecular architecture and in-silico myelofibrosis inhibition of a triazole derivative: a structural and theoretical approach. Journal of Molecular Structure 2023, 1288 , 135770. https://doi.org/10.1016/j.molstruc.2023.135770
  17. Wiem Baccari, Ilyes Saidi, Insaf Filali, Mansour Znati, Moncef Tounsi, Roberta Ascrizzi, Guido Flamini, Hichem Ben Jannet. The root essential oil from the Tunisian endemic plant Ferula tunetana: Chemical composition, biological evaluation, molecular docking analysis and drug-likeness prediction. Arabian Journal of Chemistry 2023, 16 (9) , 105044. https://doi.org/10.1016/j.arabjc.2023.105044
  18. Mahboube Eslami Moghadam, Morteza Rezaeisadat, Elaheh Shahryari, Hassan Mansouri-Torshizi, Maryam Heydari. Biological interaction of Pt complex with imidazole derivative as an anticancer compound with DNA: Experimental and theoretical studies. International Journal of Biological Macromolecules 2023, 249 , 126097. https://doi.org/10.1016/j.ijbiomac.2023.126097
  19. Imad Ahmad, Haroon Khan, Goncagül Serdaroğlu. Physicochemical properties, drug likeness, ADMET, DFT studies, and in vitro antioxidant activity of oxindole derivatives. Computational Biology and Chemistry 2023, 104 , 107861. https://doi.org/10.1016/j.compbiolchem.2023.107861
  20. Mahboube Eslami Moghadam, Morteza Rezaeisadat, Hassan Mansouri-Torshizi, Soudeh Hosseinzadeh, Hesam Daneshyar. New anticancer potential Pt complex with tertamyl dithiocarbamate ligand: Synthesis, DNA targeting behavior, molecular dynamic, and biological activity. Journal of Molecular Liquids 2023, 379 , 121651. https://doi.org/10.1016/j.molliq.2023.121651
  21. Elena V. Girich, Phan Thi Hoai Trinh, Liliana E. Nesterenko, Roman S. Popov, Natalya Yu. Kim, Anton B. Rasin, Ekaterina S. Menchinskaya, Aleksandra S. Kuzmich, Ekaterina A. Chingizova, Artem S. Minin, Ngo Thi Duy Ngoc, Tran Thi Thanh Van, Ekaterina A. Yurchenko, Anton N. Yurchenko, Dmitry V. Berdyshev. Absolute Stereochemistry and Cytotoxic Effects of Vismione E from Marine Sponge-Derived Fungus Aspergillus sp. 1901NT-1.2.2. International Journal of Molecular Sciences 2023, 24 (9) , 8150. https://doi.org/10.3390/ijms24098150
  22. Njogu M. Kimani, Charles O. Ochieng, Mike Don Ogutu, Kevin Otieno Yamo, Joab Otieno Onyango, Cleydson B. R. Santos. Inhibition Kinetics and Theoretical Studies on Zanthoxylum chalybeum Engl. Dual Inhibitors of α-Glucosidase and α-Amylase. Journal of Xenobiotics 2023, 13 (1) , 102-120. https://doi.org/10.3390/jox13010009
  23. Saurav Katuwal, Siddha Raj Upadhyaya, Rishab Marahatha, Asmita Shrestha, Bishnu P. Regmi, Karan Khadayat, Saroj Basnet, Ram Chandra Basnyat, Niranjan Parajuli, . In Silico Study of Coumarins: Wedelolactone as a Potential Inhibitor of the Spike Protein of the SARS-CoV-2 Variants. Journal of Tropical Medicine 2023, 2023 , 1-19. https://doi.org/10.1155/2023/4771745
  24. Zakari Ya'u Ibrahim, Adamu Uzairu, Gideon Adamu Shallangwa, Stephen Eyije Abechi, Sulaiman Isyaku. Quantitative Structure-Activity Relationship, Structure-based Design, and ADMET studies of pyrimethamine and cycloguanil analogs inhibitors of Plasmodium falciparum dihydrofolate reductase-thymidylate synthase (PfDHFR-TS).. Chemical Physics Impact 2022, 5 , 100132. https://doi.org/10.1016/j.chphi.2022.100132
  25. Khalifa Sunusi Aminu, Adamu Uzairu, Abdullahi Bello Umar, Muhammad Tukur Ibrahim. Salicylic acid derivatives as potential α-glucosidase inhibitors: drug design, molecular docking and pharmacokinetic studies. Bulletin of the National Research Centre 2022, 46 (1) https://doi.org/10.1186/s42269-022-00853-6
  26. Demis Zelelew, Milkyas Endale, Yadessa Melaku, Taye B. Demissie, Japheth O. Ombito, Rajalakshmanan Eswaramoorthy, . ZnO Nanoparticle-Assisted Synthesis of Thiazolo[3,2-α]Pyrimidine Analogs: Antibacterial and Antioxidant Activity, In Silico Molecular Docking, and ADMET Prediction Study. Journal of Chemistry 2022, 2022 , 1-20. https://doi.org/10.1155/2022/1346856
  27. Margarita E. Neganova, Sergey G. Klochkov, Yulia R. Aleksandrova, Gjumrakch Aliev. Histone modifications in epigenetic regulation of cancer: Perspectives and achieved progress. Seminars in Cancer Biology 2022, 83 , 452-471. https://doi.org/10.1016/j.semcancer.2020.07.015
  28. Lina S. Prieto Cárdenas, Karen A. Arias Soler, Diana L. Nossa González, Wilson E. Rozo Núñez, Agobardo Cárdenas-Chaparro, Pablo R. Duchowicz, Jovanny A. Gómez Castaño. In Silico Antiprotozoal Evaluation of 1,4-Naphthoquinone Derivatives against Chagas and Leishmaniasis Diseases Using QSAR, Molecular Docking, and ADME Approaches. Pharmaceuticals 2022, 15 (6) , 687. https://doi.org/10.3390/ph15060687
  29. Chi Dang, Ogadimma Okagu, Xiaohong Sun, Chibuike C. Udenigwe. Bioinformatics analysis of adhesin-binding potential and ADME/Tox profile of anti-Helicobacter pylori peptides derived from wheat germ proteins. Heliyon 2022, 8 (6) , e09629. https://doi.org/10.1016/j.heliyon.2022.e09629
  30. Youness Moukhliss, Yassine Koubi, Marwa Alaqarbeh, Nada Alsakhen, Samer Hamzeh, Hamid Maghat, Abdelouahid Sbai, Mohammed Bouachrine, Tahar Lakhlifi. A study of drug candidates derived from pleconaril for inhibiting coxsackievirus B3 (Cvb3) by ADMET, molecular docking, molecular dynamics and retrosynthesis. New Journal of Chemistry 2022, 46 (21) , 10154-10161. https://doi.org/10.1039/D2NJ01397E
  31. Siraj Khan, Nesrin Buğday, Asim Ur Rehman, Ihsan Ul Haq, Sedat Yaşar, İsmail Özdemir. Synthesis, molecular docking, and biological evaluation of 5‐alkyl(aryl)‐2‐isobutylthiazole derivatives: As α‐amylase, α‐glucosidase, and protein kinase inhibitors. Applied Organometallic Chemistry 2022, 36 (5) https://doi.org/10.1002/aoc.6641
  32. Kotyada Suryanarayana, Suresh Maddila, Kerru Nagaraju, Sreekantha B. Jonnalagadda. Design, synthesis, docking study and biological evaluation of novel thieno[2,3-d]-pyrimidine tethered 1,2,3-triazole scaffolds. Journal of Molecular Structure 2022, 1250 , 131713. https://doi.org/10.1016/j.molstruc.2021.131713
  33. Ermias Mergia Terefe, Arabinda Ghosh. Molecular Docking, Validation, Dynamics Simulations, and Pharmacokinetic Prediction of Phytochemicals Isolated From Croton dichogamus Against the HIV-1 Reverse Transcriptase. Bioinformatics and Biology Insights 2022, 16 , 117793222211256. https://doi.org/10.1177/11779322221125605
  34. Zakari Ya'u Ibrahim, Adamu Uzairu, Gideon Adamu Shallangwa, Stephen Eyije Abechi, Sulaiman Isyaku. Quantitative Structure-Activity Relationship, Structure-Based Design, And ADMET Studies of Pyrimethamine and Cycloguanil Analogues Inhibitors of Plasmodium Falciparum Dihydrofolate Reductase-Thymidylate Synthase (Pf-DHFR-TS). SSRN Electronic Journal 2022, 2 https://doi.org/10.2139/ssrn.4153714
  35. Bohui Peng, Chang Peng, Xiaomei Luo, Shuqi Wu, Qian Mao, Huanting Zhang, Xiao Han, . JNK signaling-dependent regulation of histone acetylation are involved in anacardic acid alleviates cardiomyocyte hypertrophy induced by phenylephrine. PLOS ONE 2021, 16 (12) , e0261388. https://doi.org/10.1371/journal.pone.0261388
  36. Louis K. S. Darko, Emmanuel Broni, Dominic S. Y. Amuzu, Michael D. Wilson, Christian S. Parry, Samuel K. Kwofie. Computational Study on Potential Novel Anti-Ebola Virus Protein VP35 Natural Compounds. Biomedicines 2021, 9 (12) , 1796. https://doi.org/10.3390/biomedicines9121796
  37. Hriday Kumar Basak, Uttam Paswan, Joydeep Ghosh, Soumen Saha, Abhik Chatterjee. In silico Evaluation of Savirin Derivatives As Inhibitors of the agr Quorum Sensing System of Staphylococcus aureus. Analytical Chemistry Letters 2021, 11 (5) , 661-683. https://doi.org/10.1080/22297928.2021.1937314
  38. Shahab Shahryari, Parvin Mohammadnejad, Kambiz Akbari Noghabi. Screening of anti- Acinetobacter baumannii phytochemicals, based on the potential inhibitory effect on OmpA and OmpW functions. Royal Society Open Science 2021, 8 (8) , 201652. https://doi.org/10.1098/rsos.201652
  39. Songül Şahin, Necmi Dege. A newly synthesized small molecule: the evaluation against Alzheimer's Disease by in silico drug design and computational structure analysis methods. Journal of Molecular Structure 2021, 1236 , 130337. https://doi.org/10.1016/j.molstruc.2021.130337
  40. Alaka Sahoo, Shasank Sekhar Swain, Ayusman Behera, Gunanidhi Sahoo, Pravati Kumari Mahapatra, Sujogya Kumar Panda. Antimicrobial Peptides Derived From Insects Offer a Novel Therapeutic Option to Combat Biofilm: A Review. Frontiers in Microbiology 2021, 12 https://doi.org/10.3389/fmicb.2021.661195
  41. Chi Dang, Ogadimma Okagu, Xiaohong Sun, Chibuike C. Udenigwe. Bioinformatics Analysis of Adhesin-Binding Potential and ADME/Tox Profile of Anti-Helicobacter Pylori Peptides Derived from Wheat Germ Proteins. SSRN Electronic Journal 2021, 410 https://doi.org/10.2139/ssrn.3943475
  • Abstract

    Figure 1

    Figure 1. Flowchart illustrating the selection of AA derivatives, screening process, and their interactions with the target (PDB ID: 1W98).

    Figure 2

    Figure 2. Results of AA derivatives against different violations.

    Figure 3

    Figure 3. BOILED-Egg model of AA derivatives.

    Figure 4

    Figure 4. Docked ligands with target-1W98: (a) AA11, (b) AA12, (c) AA16, (d) AA20, (e) AA21, (f) AA22 (g) AA23, (h) AA24, (i) AA25 (j) AA33, (k) AA34, (l) AA72 (m) AA75, (n) AA95, (o) AA96, (p) AA97, and (q) AA98.

  • References

    ARTICLE SECTIONS
    Jump To

    This article references 48 other publications.

    1. 1
      Siegel, R.; Ma, J.; Zou, Z.; Jemal, A. “Cancer statistics”. Ca-Cancer J. Clin. 2014, 64, 929,  DOI: 10.3322/caac.21208
    2. 2
      Newman, D. J. Natural products as leads to potential drugs: An old process or the new hope for drug discovery?. J. Med. Chem. 2008, 51, 25892599,  DOI: 10.1021/jm0704090
    3. 3
      Elmsellem, H.; El Ouadi, Y.; Mokhtari, M.; Bendaif, H.; Steli, H.; Aouniti, A.; Almehdi, A. M.; Abdel-Rahman, I.; Kusuma, H. S.; Hammouti, B. A natural antioxidant and an environmentally friendly inhibitor of mild steel corrosion: A commercial oil of basil (Ocimum Basilicum L.). J. Chem. Technol. Metall. 2019, 54, 742749
    4. 4
      Moussaoui, El. A.; Jawhari, F. Z.; Almehdi, A. M.; Elmsellem, H.; Benbrahim, K. F.; Bousta, D.; Bari, A. Antibacterial, antifungal and antioxidant activity of total polyphenols of Withania frutescens. L. Bioorg. Chem. 2019, 93, 103337,  DOI: 10.1016/j.bioorg.2019.103337
    5. 5
      Lichota, A.; Gwozdzinski, K. Anticancer activity of natural compounds from Plant and marine environment. Int. J. Mol. Sci. 2018, 19, 3533,  DOI: 10.3390/ijms19113533
    6. 6
      Rayan, A.; Raiyn, J.; Falah, M. Nature is the best source of anticancer drugs: Indexing natural products for their anticancer bioactivity. PLoS One 2017, 12, e0187925  DOI: 10.1371/journal.pone.0187925
    7. 7
      Newman, D. J.; Cragg, G. M. Natural Products As Sources of New Drugs over the 30 Years from 1981 to 2010. J. Nat. Prod. 2012, 75, 311335,  DOI: 10.1021/np200906s
    8. 8
      Hemshekhar, M.; Santhosh, M. S.; Kemparaju, K.; Girish, K. S. Emerging roles of anacardic acid and its derivatives: A pharmacological overview. Basic Clin. Pharmacol. Toxicol. 2012, 110, 122132,  DOI: 10.1111/j.1742-7843.2011.00833.x
    9. 9
      Hamad, F.; Mubofu, E. Potential biological applications of bio-based anacardic acids and their derivatives. Int. J. Mol. Sci. 2015, 16, 85698590,  DOI: 10.3390/ijms16048569
    10. 10
      Xiu, Y.-L.; Zhao, Y.; Gou, W.-F.; Chen, S.; Takano, Y.; Zheng, H.-C. Anacardic acid enhances the proliferation of human ovarian cancer cells. PLoS One 2014, 9, e99361  DOI: 10.1371/journal.pone.0099361
    11. 11
      Sung, B.; Pandey, M. K.; Ahn, K. S.; Yi, T.; Chaturvedi, M. M.; Liu, M.; Aggarwal, B. B. Anacardic acid (6-nonadecyl salicylic acid), an inhibitor of histone acetyltransferase, suppresses expression of nuclear factor-κB-regulated gene products involved in cell survival, proliferation, invasion, and inflammation through inhibition of the inhibitory subunit of nuclear factor-κBα kinase, leading to potentiation of apoptosis. Blood 2008, 111, 48804891,  DOI: 10.1182/blood-2007-10-117994
    12. 12
      Tan, J.; Chen, B.; He, L.; Tang, Y.; Jiang, Z.; Yin, G.; Wang, J.; Jiang, X. Anacardic acid (6-pentadecylsalicylic acid) induces apoptosis of prostate cancer cells through inhibition of androgen receptor and activation of p53 signaling. Chin. J. Cancer Res. 2012, 24, 275283,  DOI: 10.1007/s11670-012-0264-y
    13. 13
      Sun, Y.; Jiang, X.; Chen, S.; Price, B. D. Inhibition of histone acetyltransferase activity by anacardic acid sensitizes tumor cells to ionizing radiation. FEBS Lett. 2006, 580, 43534356,  DOI: 10.1016/j.febslet.2006.06.092
    14. 14
      Xiu, Y.-L.; Zhao, Y.; Gou, W.-F.; Chen, S.; Takano, Y.; Zheng, H.-C. Anacardic acid enhances the proliferation of human ovarian cancer cells. PLoS One 2014, 9, e99361  DOI: 10.1371/journal.pone.0099361
    15. 15
      Moda, T. L.; Torres, L. G.; Carrara, A. E.; Andricopulo, A. D. PK/DB: Database for pharmacokinetic properties and predictive in silico ADME models. Bioinformatics 2008, 24, 22702271,  DOI: 10.1093/bioinformatics/btn415
    16. 16
      van de Waterbeemd, H.; Gifford, E. ADMET in silico modelling: Towards prediction paradise?. Nat. Rev. Drug Discovery 2003, 2, 192204,  DOI: 10.1038/nrd1032
    17. 17
      Gleeson, M. P. Generation of a set of simple, interpretable ADMET rules of thumb. J. Med. Chem. 2008, 51, 817834,  DOI: 10.1021/jm701122q
    18. 18
      Green, I. R.; Tocoli, F. E.; Lee, S. H.; Nihei, K.-i.; Kubo, I. Design and evaluation of anacardic acid derivatives as anticavity agents. Eur. J. Med. Chem. 2008, 43, 13151320,  DOI: 10.1016/j.ejmech.2007.08.012
    19. 19
      Mamidyala, S. K.; Ramu, S.; Huang, J. X.; Robertson, A. A. B.; Cooper, M. A. Efficient synthesis of anacardic acid analogues and their antibacterial activities. Bioorg. Med. Chem. Lett. 2013, 23, 16671670,  DOI: 10.1016/j.bmcl.2013.01.074
    20. 20
      Zoete, V.; Daina, A.; Bovigny, C.; Michielin, O. SwissSimilarity: A Web Tool for Low to Ultra High Throughput Ligand-Based Virtual Screening. J. Chem. Inf. Model. 2016, 56, 13991404,  DOI: 10.1021/acs.jcim.6b00174
    21. 21
      Daina, A.; Michielin, O.; Zoete, V. SwissADME: A free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Sci. Rep. 2017, 7, 42717,  DOI: 10.1038/srep42717
    22. 22
      Daina, A.; Zoete, V. A BOILED-Egg To Predict Gastrointestinal Absorption and Brain Penetration of Small Molecules. ChemMedChem 2016, 11, 11171121,  DOI: 10.1002/cmdc.201600182
    23. 23
      Lipinski, C. A.; Lombardo, F.; Dominy, B. W.; Feeney, P. J. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv. Drug Delivery Rev. 2012, 64, 417,  DOI: 10.1016/j.addr.2012.09.019
    24. 24
      Veber, D. F.; Johnson, S. R.; Cheng, H.; Smith, B. R.; Ward, K. W.; Kopple, K. D. Molecular Properties That Influence the Oral Bioavailability of Drug Candidates. J. Med. Chem. 2002, 45, 26152623,  DOI: 10.1021/jm020017n
    25. 25
      Ghose, A. K.; Viswanadhan, V. N.; Wendoloski, J. J. A knowledge-based approach in designing combinatorial or medicinal chemistry libraries for drug discovery. 1. A qualitative and quantitative characterization of known drug databases. J. Comb. Chem. 1999, 1, 5568,  DOI: 10.1021/cc9800071
    26. 26
      Cortes, C.; Vapnik, V. Support-Vector Networks. Mach. Learn. 1995, 20, 273297,  DOI: 10.1023/a:1022627411411
    27. 27
      Pires, D. E. V.; Blundell, T. L.; Ascher, D. B. pkCSM: Predicting small-molecule pharmacokinetic and toxicity properties using graph-based signatures. J. Med. Chem. 2015, 58, 40664072,  DOI: 10.1021/acs.jmedchem.5b00104
    28. 28
      Pellicciari, A. Attrition in the Pharmaceutical Industry - Reasons, Implications, and Pathways Forward. Edited by Alexander Alex, C. John Harris, Dennis A. Smith. ChemMedChem 2017, 12, 10971098,  DOI: 10.1002/cmdc.201600621
    29. 29
      van deWaterbeemd, H.; Smith, D. A.; Jones, B. C. Lipophilicity in PK design: Methyl, ethyl, futile. J. Comput.-Aided Mol. Des. 2001, 15, 273286,  DOI: 10.1023/a:1008192010023
    30. 30
      Pliška, V.; Testa, B.; Waterbeemd, H. V. Lipophilicity in Drug Action and Toxicology; VCH Verlagsgesellschaft mbH: Weinheim (Federal Republic of Germany), 1996.
    31. 31
      Smith, D. A.; Di, L.; Kerns, E. H. The effect of plasma protein binding on in vivo efficacy: Misconceptions in drug discovery. Nat. Rev. Drug Discovery 2010, 9, 929939,  DOI: 10.1038/nrd3287
    32. 32
      Di, L.; Kerns, E. H. Drug-Like Properties: Concepts, Structure Design and Methods from ADME to Toxicity Optimization; Elsevier publisher, 2008.
    33. 33
      Khojasteh, S. C.; Wong, H.; Hop, C. E. C. A. Pharmacokinetics. Drug Metabolism and Pharmacokinetics Quick Guide; Springer, 2011; p 1.
    34. 34
      Li, A. P.; Jurima-Romet, M. Applications of primary human hepatocytes in the evaluation of pharmacokinetic drug-drug interactions: Evaluation of model drugs terfenadine and rifampin. Cell Biol. Toxicol. 1997, 13, 365374,  DOI: 10.1023/A:1007451911843
    35. 35
      Danielsson, B. R.; Lansdell, K.; Patmore, L.; Tomson, T. Phenytoin and phenobarbital inhibit human HERG potassium channels. Epilepsy Res. 2003, 55, 147157,  DOI: 10.1016/s0920-1211(03)00119-0
    36. 36
      Hosey, C. M.; Benet, L. Z. Predicting the extent of metabolism using in vitro permeability rate measurements and in silico permeability rate predictions. Mol. Pharm. 2015, 12, 14561466,  DOI: 10.1021/mp500783g
    37. 37
      Czechtizky, C.; Hamley, P. Small Molecule Medicinal Chemistry: Strategies and Technologies; John Wiley & Sons, 2015.
    38. 38
      Singh, U.; Gangwal, R. P.; Dhoke, G. V.; Prajapati, R.; Damre, M.; Sangamwar, A. T. 3D-QSAR and molecular docking analysis of (4-piperidinyl)-piperazines as acetyl-CoA carboxylases inhibitors. Arabian J. Chem. 2017, 10, S617S626,  DOI: 10.1016/j.arabjc.2012.10.023
    39. 39
      Bendaif, H.; Melhaoui, A.; Ramdani, M.; Elmsellem, H.; Douez, C.; El Ouadi, Y. Antibacterial activity and virtual screening by molecular docking of lycorine from Pancratium foetidum Pom (Moroccan endemic Amaryllidaceae). Microb. Pathog. 2018, 115, 138145,  DOI: 10.1016/j.micpath.2017.12.037
    40. 40
      Karthick, T.; Tandon, P. Computational approaches to find the active binding sites of biological targets against busulfan. J. Mol. Model. 2016, 22, 142,  DOI: 10.1007/s00894-016-3015-z
    41. 41
      Shaikh, S.; Shastri, S. L.; Naik, N. S.; Kulkarni, R.; Madar, J. M.; Shastri, L. A.; Joshi, S. D.; Sunagar, V. Synthesis, antitubercular and antimicrobial activity of 1,2,4-triazolidine-3-thione functionalized coumarin and phenyl derivatives and molecular docking studies. ChemistrySelect 2019, 4, 105115,  DOI: 10.1002/slct.201802395
    42. 42
      Sidhu, A.; Kukreja, S. Synthesis of novel fluorinated benzothiazol-2-yl-1,2,4-triazoles: Molecular docking, antifungal evaluation and in silico evaluation for SAR. Arabian J. Chem. 2019, 12, 2118,  DOI: 10.1016/j.arabjc.2015.01.009
    43. 43
      Kareem, A.; Nami, S. A. A.; Khan, M. S.; Bhat, S. A.; Mirza, A. U.; Nishat, N. Self-assembled transition metal dithiocarbamates of pyridine-3-carboxamide: synthesis, spectral characterization, thermal and biological studies. New J. Chem. 2019, 43, 4413,  DOI: 10.1039/c8nj03660h
    44. 44
      Kaviyarasi, N. S.; Prashantha, C. N.; Suryanarayana, V. V. S. In Silico Analysis of Inhibitor and Substrate Binding Site of Serrapeptidase from Serratia Marcescens MTCC 8708. Int. J. Pharmacol. Pharm. Sci. 2016, 8, 123128
    45. 45
      De Vita, D.; Pandolfi, F.; Ornano, L.; Feroci, M.; Chiarotto, I.; Sileno, I.; Pepi, F.; Costi, R.; Di Santo, R.; Scipione, L. New N,N-Dimethylcarbamate Inhibitors of Acetylcholinesterase: Design Synthesis and Biological Evaluation. J. Enzyme Inhib. Med. Chem. 2016, 31, 106113,  DOI: 10.1080/14756366.2016.1220377
    46. 46
      Iftikhar, H.; Batool, S.; Deep, A.; Narasimhan, B.; Sharma, P. C.; Malhotra, M. In Silico Analysis of the Inhibitory Activities of GABA Derivatives on 4-Aminobutyrate Transaminase. Arabian J. Chem. 2017, 10, S1267S1275,  DOI: 10.1016/j.arabjc.2013.03.007
    47. 47
      Chhajed, S. S.; Upasani, C. D. Synthesis, Biological Screening and Molecular Modeling Studies of Novel 3-Chloro-4-Substituted-1-(2-(1H-Benzimidazol-2-Yl)Phenyl))-Azetidin-2-Ones. J. Enzyme Inhib. Med. Chem. 2012, 27, 504,  DOI: 10.3109/14756366.2011.598867
    48. 48
      Rege, M. D.; Ghadi, R.; Katiyar, S. S.; Kushwah, V.; Jain, S. Exploring an interesting dual functionality of anacardic acid for efficient paclitaxel delivery in breast cancer therapy. Nanomedicine 2019, 14, 57,  DOI: 10.2217/nnm-2018-0138
  • Supporting Information

    Supporting Information

    ARTICLE SECTIONS
    Jump To

    The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acsomega.9b04398.

    • Chemical structures of alkyl chain-modified AA derivatives (labeled from 1 to 29); chemical structures of alkyl chain- and functional group-modified AA derivatives labeled from 30 to 49; chemical structures of alkyl chain- and functional group-modified AA derivatives labeled from 50 to 76; chemical structures of alkyl chain- and functional group-modified AA derivatives labeled from 77 to 100; BOILED-Egg model of AA1–AA100 derivatives; important computed physicochemical properties for AA1–AA100; important computed ADMET properties for AA1–AA100 derivatives; and computed safety end points for AA1–AA100 derivatives (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