Available online on 15.11.2022 at http://jddtonline.info
Journal of Drug Delivery and Therapeutics
Open Access to Pharmaceutical and Medical Research
Copyright © 2022 The Author(s): This is an open-access article distributed under the terms of the CC BY-NC 4.0 which permits unrestricted use, distribution, and reproduction in any medium for non-commercial use provided the original author and source are credited
Open Access Full Text Article Research Article
ADMETox-informatics of Plant Derived Octadecanoic Acid (Stearic Acid) from Ethyl Acetate Fraction of Moringa oleifera Leaf Extract as a Natural Lead for Next Generation Drug Design, Development and Therapeutics
Murugan M.1, Kalaimathi RV.1, Krishnaveni K.2, Basha AN.1, Gilles A Pallan.1, Kandeepan C.1, Senthilkumar N.3, Mathialagan B.4, Ramya S.4, Jayakumararaj R.5*, Loganathan T.6, Pandiarajan G.7, Kaliraj P.8, Sutha S.9, Kandavel D.10, Grace Lydial Pushpalatha G11, Abraham GC.12 Ram Chand Dhakar13
1 PG & Research Department of Zoology, Arulmigu Palaniandavar College of Arts & Culture, Palani – 624601, TN, India
2 Department of Zoology, GTN Arts & Science College, Dindigul - 624005, TN, India
3Institute of Forest Genetics & Tree Breeding (IFGTB), ICFRE, Coimbatore – 641002, TN, India
4 PG Department of Zoology, Yadava College (Men), Thiruppalai - 625014, Madurai, TN, India
5 Department of Botany, Government Arts College, Melur – 625106, Madurai District, TN, India
6 Department of Plant Biology & Plant Biotechnology, LN Government College (A), Ponneri, TN, India
7 Department of Botany, Sri S Ramasamy Naidu Memorial College (A), Sattur - 626203 TN, India
8 Department of Zoology, Sri S Ramasamy Naidu Memorial College (A), Sattur - 626203 TN, India
9 Department of Medicinal Botany, Govt. Siddha Medical College, Palayamkottai, Tamil Nadu, India
10 Government Arts College for Men (Autonomous), Nandanam, Chennai – 600 035, Tamil Nadu, India
11 PG Department of Botany, Sri Meenakshi Government Arts College, Madurai – 625002, TN, India
12 PG & Research Department of Botany, The American College, Madurai – 625002, TamilNadu, India
13 Hospital Pharmacy, SRG Hospital & Medical College Jhalawar-326001, Rajasthan, India
|
Article Info: _______________________________________________ Article History: Received 26 Sep 2022 Reviewed 30 Oct 2022 Accepted 09 Nov 2022 Published 15 Nov 2022 _______________________________________________ Cite this article as: Murugan M, Kalaimathi RV, Krishnaveni K, Basha AN, Gilles A Pallan, Kandeepan C, Senthilkumar N, Mathialagan B, Ramya S, Jayakumararaj R, Loganathan T, Pandiarajan G, Kaliraj P, Sutha S, Kandavel D, Grace Lydial Pushpalatha G, Abraham GC, Dhakar RC, ADMETox-informatics of Plant Derived Octadecanoic Acid (Stearic Acid) from Ethyl Acetate Fraction of Moringa oleifera Leaf Extract as a Natural Lead for Next Generation Drug Design, Development and Therapeutics, Journal of Drug Delivery and Therapeutics. 2022; 12(6):129-141 DOI: http://dx.doi.org/10.22270/jddt.v12i6.5677 _______________________________________________*Address for Correspondence: Dr R Jayakumararaj, Department of Botany, Government Arts College, Melur – 625106, Madurai District, TN, India |
Abstract ___________________________________________________________________________________________________________________ In-silico Computer-Aided Drug Design (CADD) significantly relies on cybernetic screening of Plant Based Natural Products (PBNPs) as a prime source of bioactive compounds/ drug leads due to their unique chemical structural scaffolds and distinct functional characteristic features amenable to drug design and development. In the Post-COVID-Era a large number of publications have focused on PBNPs. Moreover, PBNPs still remain as an ideal source of novel therapeutic agents of GRAS standard. However, a well-structured, in-depth ADME/Tox profile with deeper dimensions of PBNPs has been lacking for many of natural pharma lead molecules that hamper successful exploitation of PBNPs. In the present study, ADMET-informatics of Octadecanoic Acid (Stearic Acid - SA) from ethyl acetate fraction of Moringa oleifera leaves has been envisaged to predict ADMET and pharmacokinetics (DMPK) outcomes. This work contributes to the deeper understanding of SA as major source of drug lead from Moringa oleifera with immense therapeutic potential. The data generated herein could be useful for the development of SA as plant based natural product lead (PBNPL) for drug development programs. Keywords: Moringa oleifera; Bioactive Substances; Octadecanoic Acid; Stearic Acid; ADME/Tox; Natural Product Based Drug Lead; PBNPs
|
INTRODUCTION
Chemically, Octadecanoic Acid/ Stearophanic Acid (Stearic Acid) is one of the most common long-chain fatty acids, found in combined form in animal and vegetable fats. Commercial “Stearic Acid” contains equal amounts of Stearic Acid (SA) and Palmitic Acid (PA) and small amounts of Oleic Acid (OA)1. It is one of the common saturated fatty acid naturally obtained from plant sources with the molecular formula C18H36O22,3. Stearic Acid is widely used as the major component in the production of washing detergents, soaps, and personal care products (PCP) such as cosmetics, shampoos and shaving creams. However, it must be noted that the detergent soaps are not directly made from SA, but through saponification of triglycerides of Stearic Acid Esters (SAE)2,3. SAE with ethylene glycol, glycol stearate, and glycol di-stearate are used in the preparation of shampoos, soaps, and other cosmetic products to impart a pearly effect. They are added to the product in the molten form and allowed to crystallize under specific conditions so as to impart desirable effect in the products. Best available detergents in the market are obtained from amides/ quaternary alkyl-ammonium derivatives of SAE2,3.
High fatty acid content in Moringa oleifera seed oil (MOSO) has rarely been exploited by Fast-Moving Consumer Goods (FMCG) industries for the production of Food Grade Consumable Products (FGCP) due to low melting point/ lack of plasticity. Dollah et al.4 pointed out that enzymatic inter-esterification (EIE) of MOSO with palm stearin (PS) added to palm kernel oil (PKO) could yield fat molecules with better and harder biochemical frame-works that may contain desirable food grade nutritional and physical properties.4
So far 13 species have been reported from genus Moringa. MO is native to India, however, cultivated all over the world. MO is deciduous, with brittle stem, whitish-gray bark; leaves - pale green in color, bipinnate/ tri-pinnate with opposite, ovate leaflets. All plant parts of MO are endowed with nutraceutical/ pharmaceutical properties. MO has been traditionally used in various indigenous traditional systems of medicine (ITSM) as it is endowed with antioxidants, anti-diabetic, anti-bacterial, anti-fungal, anti-carcinogenic properties however, without side effects. Recently, MO has been considered for the development of Ready to Eat Functional Food Products, Food Grade Nutraceutical Products and therapeutic agents as like other medicinal plants5-31.
Pharmacological studies indicate that extracts obtained from MO have significant medicinal properties in relation to health and disease, but there isn't enough information on SA. ADMETox information on effects of SA is parsimoniously available, therefore, in the present study ADMETox profile of SA from MO has been carried out. Furthermore, DMPK properties of MO have been “fine-tuned” in order to expand the chances of making SA an apt candidate for clinical trials and biomedical applications.
MATERIALS AND METHODS
In silico Drug-Likeliness and Bioactivity Prediction
Drug likeliness and bioactivity of selected molecule was analyzed using Molinspiration server (http://www.molinspiration.com). Molinspiration tool is cheminformatics software that provides molecular properties as well as bioactivity prediction of compounds. In Molinspiration-based drug-likeness analysis, there are two important factors, including lipophilicity level (log P) and polar surface area (PSA) directly associated with pharmacokinetic properties (PK) of the compounds32. In Molinspiration-based bioactivity analysis, calculation of bioactivity score of compounds toward GPCR ligands, ion channel modulators, kinase inhibitors, nuclear receptor ligands, protease inhibitors, and other enzyme targets were analyzed by Bayesian statistics33. The analysis was carried out for G protein-coupled receptors (GPCR)34, ion channels, kinases, nuclear hormone receptors, proteases, and other enzymes as major drug targets of SA
In silico ADMET Analysis
SwissADME is a Web tool that gives free access to a pool of fast yet robust predictive models for physicochemical properties, pharmacokinetics, druglikeness and medicinal chemistry friendliness, among which in-house proficient methods such as iLOGP (a physics-based model for lipophilicity) or BOILED-Egg.35 PK properties, such as Absorption, Distribution, Metabolism, Excretion, and Toxicity (ADMET), of SA was predicted using admerSAR v2.0 server (http://lmmd.ecust.edu.cn/admetsar2/). admerSAR server is an open-source computational tool for prediction of ADMET properties of compounds, which makes it a practical platform for drug discovery and other pharmacological research. In ADMET analysis, absorption (A) of good drugs depends on factors such as membrane permeability [colon cancer cell line (Caco-2)]36, human intestinal absorption (HIA)37, and status of either P-glycoprotein substrate/ inhibitor38. Distribution (D) of drugs mainly depends on the ability to cross blood-brain barrier (BBB) 39. Metabolism (M) of drugs is calculated by the CYP, MATE1, and OATP1B1-OATP1B3 models40. Excretion (E) of drugs is estimated based on renal OCT substrate. Toxicity (T) of drugs is predicted on human ether-a-Go-Go related gene inhibition, carcinogenic status, mutagenic status, and acute oral toxicity41,42.
vNN model building and analysis
vNN method was used to calculate the similarity distance between molecules in terms of their structure, and uses a distance threshold to define a domain of applicability to ensures that the predictions generated are reliable43. vNN models can be built keeping quantitative structure–activity relationship (QSAR) models up-to-date to maintain their performance levels44. Performance characteristics of the models are comparable, and often superior to those of other more elaborate model.15-18 One of the most widely used measures of similarity distance between two small molecules is Tanimoto distance, d, which is defined as:
where n(P∩Q) is number of features common to molecules p and q, and n(P) and n(Q) are the total numbers of features for molecules p and q, respectively. The predicted biological activity y is given by a weighted across structurally similar neighbours:
where di denotes Tanimoto distance between a query molecule for which a prediction is made and a molecule i of the training set; d0 is a Tanimoto-distance threshold, beyond which two molecules are no longer considered to be sufficiently similar to be included in the average; yi is the experimentally measured activity of molecule i; v denotes the total number of molecules in the training set that satisfies the condition di≤d0; and h is a smoothing factor, which dampens the distance penalty. Values of h and d0 are determined from cross-validation studies. To identify structurally similar compounds, Accelrys extended-connectivity fingerprints with a diameter of four chemical bonds (ECFP4) was used.
Model Validation
A 10-fold cross-validation (CV) procedure was used to validate new models and to determine the values of smoothing factor h and Tanimoto distance d0. In this procedure, data was randomly divided into 10 sets, and used 9 to develop the model and 10th to validate it, this process was repeated 10 times, leaving each set of molecules out once.
Performance Measures
Following metrics were used to assess model performance. (1) sensitivity measures a model’s ability to correctly detect true positives, (2) specificity measures a model’s ability to detect true negatives, (3) accuracy measures a model’s ability to make correct predictions and (4) kappa compares the probability of correct predictions to the probability of correct predictions by chance (its value ranges from +1 (perfect agreement between model prediction and experiment) to –1 (complete disagreement), with 0 indicating no agreement beyond that expected by chance).
where TP, TN, FP, and FN denote the numbers of true positives, true negatives, false positives, and false negatives, respectively. Kappa is a metric for assessing the quality of binary classifiers. Pr (e) is an estimate of the probability of a correct prediction by chance. It is calculated as:
The coverage is the proportion of test molecules with at least one nearest neighbour that meets the similarity criterion. The coverage is a measure of how many test compounds are within the applicability domain of a prediction model.
RESULTS AND DISCUSSION
SA is a saturated long-chain fatty acid with an 18-carbon backbone. SA is a major component of cocoa butter and shea butter. SA is a white solid with a mild odour, floats on water. SA is a saturated fatty acid present in animal and vegetable fats and oils. It is a waxy solid.
|
Chemical Kingdom |
: |
Organic Compounds |
|
Super Class |
: |
Lipids and Lipid-like Molecules |
|
Class |
: |
Fatty Acyls |
|
Subclass |
: |
Fatty Acids and Conjugates |
|
IUPAC Name |
: |
Octadecanoic Acid |
|
Common Name |
|
Stearic Acid (SA) |
|
Synonym |
: |
Ethyl Palmitate;(-)Hydroxycitric Acid;(-) |
|
Compound CID |
: |
|
|
PubChem Identifier |
: |
12366 |
|
ChEBI Identifier |
: |
28842 |
|
CAS Identifier |
: |
5281 |
|
Molecular Formula |
: |
|
|
Molecular Weight |
: |
284.5g/mol |
|
Canonical SMILES |
: |
CCCCCCCCCCCCCCCCCC(=O)O |
|
InChIKey |
: |
QIQXTHQIDYTFRH-UHFFFAOYSA-N |
Drug-likeness properties of SA
Score from cLogP: 0.358 (cLogP = 5.581); Score from logS: 0.763 (logS = -3.826); Score from molecular weight: 0.956 (molecular weight 242.0); Score from drug-likeness: 0.0 (drug-likeness = 35.364); No Risk of Mutagenicity Score = 1.0; No Risk of Tumorigenicity Score = 1.0; No Risk of Irritating Effects Score = 1.0; No Risk of Reproductive Effects Score = 1.0 respectively were predicted and the overall predicted drug score for compound 3 was calculated as 0.293.
Bio-molecular properties of SA
Calculated value for molecular properties of compound 1 were (values given in parenthesis) - miLogP (5.35); TPSA (26.30); Natoms (15); MW (214.35); nON (2); nOHNH (0); Nviolations (1); Nrotb (11); volume (214.74) respectively; and the calculated bioactivity scores for biological properties were - GPCR ligand34 (-0.41); Ion channel modulator (-0.13); Kinase inhibitor (-0.73); Nuclear receptor ligand (-0.43); Protease inhibitor (-0.46); Enzyme inhibitor (-0.11) respectively (Table 1).
Physiochemical Properties of SA
Molecular Formula of SA = C18H36O2; Molecular weight of SA = 284.48 g/mol; Number of heavy atoms in SA = 20; Number of aromatic. heavy atoms = 0; Fraction Csp3 in SA = 0.94; Number of rotatable bonds in SA = 16; Number of H-bond acceptors in SA = 2; Number of H-bond donors in SA = 1; Molar Refractivity of SA = 90.41; TPSA of SA = 37.30 Ų; Lipophilicity properties of SA = -; Log Po/w (iLOGP) = 4.30; Log Po/w (XLOGP3) = 8.23; Log Po/w (WLOGP) = 6.33; Log Po/w (MLOGP) = 4.67; Log Po/w (SILICOS-IT) = 6.13; Consensus Log Po/w = 5.93; Water Solubility properties of SA - Log S (ESOL) = -5.73; Solubility = 5.26e-04 mg/ml; 1.85e-06 mol/l; Class = Moderately soluble; Log S (Ali) = -8.87; Solubility = 3.80e-07 mg/ml; 1.33e-09 mol/l; Class = Poorly soluble; Log S (SILICOS-IT) = -6.11; Solubility = 2.19e-04 mg/ml; 7.71e-07 mol/l; Class = Poorly soluble; Pharmacokinetics properties of SA - GI absorption of SA is High BBB permeant = No; P-gp substrate = No; CYP1A2 inhibitor = Yes; CYP2C19 inhibitor = No; CYP2C9 inhibitor = No; CYP2D6 inhibitor = No; CYP3A4 inhibitor = No; Log Kp (skin permeation) = -2.19 cm/s; Druglikeness properties of SA – Lipinski’s Rule for SA is Yes; (1 violation: MLOGP>4.15); Ghose’s Rule for SA is No; (1 violation: WLOGP>5.6); Veber’s Rule for SA is No; (1 violation: Rotors>10); Egan Rule for SA is No; (1 violation: WLOGP>5.88); Muegge’s Rule for SA is No;(2 violations: XLOGP3>5, Rotors>15); Bioavailability Score for SA = 0.85 Fig. 1; Medicinal Chemistry properties of SA - PAINS for SA is 0; Brenk’s for SA is No; Leadlikeness for SA is No; (2 violations: Rotors>7, XLOGP3>3.5); Accessibility for SA = 2.54. Percentage distribution of function targets for SA using Swiss Target Prediction is given in Fig. 2; Table 2. Predicted ADMET Properties of SA is given in Table 3 and the summative physicochemical, druggable, ADMET properties of SA have been provided in Table 4.
vNN model based ADMET analysis of SA
Implemented Absorption, Distribution, Metabolism, Excretion and Toxicity (ADMET) prediction models, including their performance measures has been carried out. Model covers diverse set of ADMET endpoints for Maximum Recommended Therapeutic Dose (MRTD), mutagenicity, human liver microsomal (HLM), Pgp inhibitor/substrates (Table 5).
Liver Toxicity
Drug-induced liver injury (DILI) has been one of the most common reasons for drug withdrawal from market. This application predicts whether a compound could cause DILI. A dataset of 1,431 compounds was obtained from online sources. Dataset contained both pharmaceuticals and non-pharmaceuticals; a compound was classified as causing DILI if it was associated with a high risk of DILI and not if there was no such risk45 that includes SA (Table 5).
Cytotoxicity (HepG2)
Cytotoxicity is the degree to which a chemical causes damage to cells. A cytotoxicity prediction model was developed using in vitro data on toxicity against HepG2 cells for 6,000 structurally diverse compounds, including SA were collected from ChEMBL. In developing the model, the compounds with an IC50 ≤ 10 μM were considered in the in vitro assay as cytotoxic (Table 5).
Metabolism - HLM
Human Liver Microsomal (HLM) stability assay is commonly used to identify and exclude compounds that are too rapidly metabolized. For a drug to achieve effective therapeutic concentrations in the body, it cannot be metabolized too rapidly by the liver. Compounds with a half-life of 30 min or longer in an HLM assay were considered as stable; otherwise unstable. HLM data was retrieved from ChEMBL database, manually curated and classified compounds as stable or unstable based on reported half-life (T1/2 > 30 min was considered stable, and T1/2 < 30 min unstable. The final dataset contained 3,654 compounds. Of these, as much as 2,313 including SA were classified as stable and 1,341 as unstable (Table 5).
Cytochrome P450 enzyme (CYP) inhibition
CYPs play an important role in metabolism and detoxification of xenobiotics. In vitro data derived from five main drug-metabolizing CYPs-1A2, 3A4, 2D6, 2C9 and 2C19 was used to develop CYP inhibition models. CYP inhibitors were retrieved from PubChem and classified a compound with an IC50 ≤ 10 μM for an enzyme as an inhibitor of the enzyme. Predictions for the following enzymes: CYP1A2, CYP3A4, CYP2D6, CYP2C9, and CYP2C19 have been provided for SA in Table 5.
Membrane Transporters - Blood-Brain Barrier (BBB)
BBB is a highly selective barrier that separates the circulating blood from the central nervous system46. VNN-based BBB model has been developed, using 352 compounds whose BBB permeability values (logBB) were obtained from the literature Compounds with log BBB values of less than –0.3 and greater than +0.3 were classified as BBB non-permeable and permeable. Calculated BBB value of SA is -0.195 based on WLOGP vs TPSA using BOILED-Egg Fig. 3; Table 5.
Pgp Substrates/ Inhibitors
P-glycoprotein (Pgp) is an essential cell membrane protein that extracts many foreign substances from the cell. Cancer cells often overexpress Pgp, which increases the efflux of chemotherapeutic agents from the cell and prevents treatment by reducing the effective intracellular concentrations known as multidrug resistance. For this reason, identifying compounds that can either be transported out of the cell by Pgp (substrates) or impair Pgp function (inhibitors) is required. Models to predict both Pgp substrates and Pgp inhibitors were developed. Pgp substrate dataset consists of measurements of 422 substrates and 400 non-substrates. To generate a large Pgp inhibitor dataset, both the datasets were combined and duplicates were removed to form a combined dataset consisting of a training set of 1,319 inhibitors and 937 non-inhibitors. Analysis indicates that SA is neither a P-glycoprotein substrate nor P-glycoprotein I/II inhibitor as indicated47 (Table 5).
hERG (Cardiotoxicity):
human ether-à-go-go-related gene (hERG) codes for a potassium ion channel involved in the normal cardiac repolarization activity of the heart. Drug-induced blockade of hERG function can cause long QT syndrome, which may result in arrhythmia and death48. As much as 282 known hERG blockers from the literature were retrieved and classified compounds with an IC50 cut-off value of 10 μM or less as blockers. A set of 404 compounds with IC50 values greater than 10 μM were classified as non-blockers. Prediction indicated SA as hERG I non - inhibitor and hERG II as non - inhibitor (Table 5).
MMP (Mitochondrial Toxicity)
Fundamental role of mitochondria in cellular energetics and oxidative stress, mitochondrial dysfunction has been implicated in cancer, diabetes, neurodegenerative disorders, and cardiovascular diseases. A largest dataset of chemical-induced changes in mitochondrial membrane potential (MMP), was used based on the assumption that a compound that causes mitochondrial dysfunction is also likely to reduce the MMP. vNN-based MMP prediction model was developed using 6,261 compounds collected from a previous study that screened a library of more than 10,000 compounds (~8,300 unique chemicals) at 15 concentrations, each in triplicate, to measure changes in the MMP in HepG2 cells. The study found that 913 compounds decreased the MMP, whereas 5,395 compounds had no effect (Table 5). SA was predicted to be Non-carcinogens with a calculated value of 0.575.
Mutagenicity (Ames test)
Mutagens are chemicals that cause abnormal genetic mutations leading to cancer. A common way to assess a chemical’s mutagenicity is the Ames test. A prediction model was developed using a literature dataset of 6,512 compounds, of which 3,503 were Ames-positive. Prediction indicated SA as Non AMES toxic with a calculated value of 0.963 (Table 5).
Maximum Recommended Therapeutic Dose (MRTD)
MRTD is an estimated upper daily dose that is considered to be safe. A prediction model was developed based on a dataset of MRTD values disclosed by the FDA, mostly of single-day oral doses for an average adult with a body weight of 60 kg, for 1,220 compounds (small organic drugs). Organometallics, high-molecular weight polymers were excluded (>5,000 Da), nonorganic chemicals, mixtures of chemicals, and very small molecules (<100 Da). An external test set of 160 compounds collected by FDA was used for validation. The total dataset for the model contained 1,185 compounds49. The predicted MRTD value is reported in mg/day unit based upon an average adult weighing 60 kg. MRTD for SA was calculated as -0.791 (Table 5).
CONCLUSION
ADMET-informatics of Octadecanoic Acid (Stearic Acid) from ethyl acetate fraction of Moringa oleifera leaves has been envisaged to predict drug metabolism and pharmacokinetics (DMPK) outcomes. ADMET informatics contributes to the deeper understanding of SA as a major source of drug lead from Moringa oleifera with immense therapeutic potential. Results indicate that SA is of GRAS standard drug with predicted values within the range suitable for human consumption. Data generated herein could be useful for the development of SA as PBNPL for next generation drug design, development and therapies.
REFERENCES
1. Acid L. Final report on the safety assessment of oleic acid, laurie acid, palmitic acid, myristic acid, and stearic acid. J. Am. Coll. Toxicol. 1987; 6:321-401. https://doi.org/10.3109/10915818709098563
2. Meyer F, Bloch K. Metabolism of stearolic acid in yeast. Journal of Biological Chemistry. 1963 Aug 1; 238(8):2654-9. https://doi.org/10.1016/S0021-9258(18)67881-0
3. Warra AA, Jonathan BL, Ibrahim BD, Adedara AO. GC-MS Analysis of Hexane Extracts of Two Varieties of Sesame (Sesamun indicum L.) Seed Oil. IJCPT 2016; 1(1):1-9
4. Dollah S, Abdulkarim SM, Ahmad SH, Khoramnia A, Mohd Ghazali H. Physico‐chemical properties of Moringa oleifera seed oil enzymatically inter-esterified with palm stearin and palm kernel oil and its potential application in food. Journal of the Science of Food and Agriculture. 2016 Aug; 96(10):3321-33. https://doi.org/10.1002/jsfa.7510
5. Ramya S, Neethirajan K, Jayakumararaj R. Profile of bioactive compounds in Syzygium cumini-a review. Journal of Pharmacy Research. 2012; 5(8):4548-53.
6. Kandeepan C, Suganandam K, Jeevalatha A, Kavitha N, Senthilkumar N, Sutha S, Syed MA, Gandhi S, Ramya S, Grace Lydial Pushpalatha G, Abraham GC, King Immanuel J, Jayakumararaj R, ADMETE valuation, Pharmacokinetics, Drug-likeness and Medicinal Chemistry of GCMS Identified Bioactive Compounds of Moringa oleifera Natural-Ripened Dried Methanolic Pod Extract (MOMPE) as a Potential Source of Natural Drug Frontrunner for Next Generation Drug Design, Development and Therapeutics, Journal of Drug Delivery and Therapeutics. 2022; 12(6):65-85 https://doi.org/10.22270/jddt.v12i5-S.5657
7. Ramya S., Chandran M., King IJ., Jayakumararaj R., Loganathan T., Pandiarajan G., Kaliraj P., Grace Lydial Pushpalatha G, Abraham GC., Vijaya V., Aruna D, Sutha S, Dhakar RC, Phytochemical Screening, GCMS and FTIR Profile of Bioactive Compounds in Solanum lycopersicum Wild Fruits collected from Palani Hill Ranges of the Western Ghats, Journal of Drug Delivery and Therapeutics. 2022; 12(6):56-64 https://doi.org/10.22270/jddt.v12i2-S.5280
8. Chandran M, Priyanka R, Kavipriya D, Ramya S, Jayakumararaj R, Loganathan T, Pandiarajan G, Kaliraj P, Pushpalatha GG, Abraham GC, Dhakar RC. Reformulation and Scientific Evaluation of CUSOCO: A Traditional Toothpaste Formula from Classical Tamil Literature towards treatment of Halitosis. Journal of Drug Delivery and Therapeutics. 2022; 12(5):127-31. https://doi.org/10.22270/jddt.v12i5.5604
9. Kalaimathi RV, Krishnaveni K, Murugan M, Basha AN, Gilles AP, Kandeepan C, Senthilkumar N, Mathialagan B, Ramya S, Ramanathan L, Jayakumararaj R. ADMET informatics of Tetradecanoic acid (Myristic Acid) from ethyl acetate fraction of Moringa oleifera leaves. Journal of Drug Delivery and Therapeutics. 2022; 12(4-S):101-11. https://doi.org/10.22270/jddt.v12i4-S.5533
10. Nazar S, Jeyaseelan M, Jayakumararaj R. Local Health Traditions, Cultural Reflections and Ethno-taxonomical Information on Wild Edible Fruit Yielding Medicinal Plants in Melur Region of Madurai District, TamilNadu, India. Journal of Drug Delivery and Therapeutics. 2022; 12(3):138-57. https://doi.org/10.22270/jddt.v12i3.5405
11. Murugan M, Krishnaveni K, Sabitha M, Kandeepan C, Senthilkumar N, Loganathan T, Pushpalatha GL, Pandiarajan G, Ramya S, Jayakumararaj R. In silico Target Class Prediction and Probabilities for Plant Derived Omega 3 Fatty Acid from Ethyl Acetate Fraction of Moringa oleifera Leaf Extract. Journal of Drug Delivery and Therapeutics. 2022; 12(3):124-37. https://doi.org/10.22270/jddt.v12i3.5352
12. Parvathi K, Kandeepan C, Sabitha M, Senthilkumar N, Ramya S, Boopathi NM, Ramanathan L, Jayakumararaj R. In-silico Absorption, Distribution, Metabolism, Elimination and Toxicity profile of 9, 12, 15-Octadecatrienoic acid (ODA) from Moringa oleifera. Journal of Drug Delivery and Therapeutics. 2022; 12(2-S):142-50. https://doi.org/10.22270/jddt.v12i2-S.5289
13. Ramya S, Loganathan T, Chandran M, Priyanka R, Kavipriya K, Pushpalatha GG, Aruna D, Ramanathan L, Jayakumararaj R, Saluja V. Phytochemical Screening, GCMS, FTIR profile of Bioactive Natural Products in the methanolic extracts of Cuminum cyminum seeds and oil. Journal of Drug Delivery and Therapeutics. 2022; 12(2-S):110-8. https://doi.org/10.22270/jddt.v12i2-S.5280
14. Kandeepan C, Sabitha M, Parvathi K, Senthilkumar N, Ramya S, Boopathi NM, Jayakumararaj R. Phytochemical Screening, GCMS Profile, and In-silico properties of Bioactive Compounds in Methanolic Leaf Extracts of Moringa oleifera. Journal of Drug Delivery and Therapeutics. 2022; 12(2):87-99. https://doi.org/10.22270/jddt.v12i2.5250
15. Krishnaveni K, Sabitha M, Murugan M, Kandeepan C, Ramya S, Loganathan T, Jayakumararaj R. vNN model cross validation towards Accuracy, Sensitivity, Specificity and kappa performance measures of β-caryophyllene using a restricted-unrestricted applicability domain on Artificial Intelligence & Machine Learning approach based in-silico prediction. Journal of Drug Delivery and Therapeutics. 2022; 12(1-S):123-31. https://doi.org/10.22270/jddt.v12i1-S.5222
16. Jeevalatha A, Kalaimathi RV, Basha AN, Kandeepan C, Ramya S, Loganathan T, Jayakumararaj R. Profile of bioactive compounds in Rosmarinus officinalis. Journal of Drug Delivery and Therapeutics. 2022; 12(1):114-22. https://doi.org/10.22270/jddt.v12i1.5189
17. Kalaimathi RV, Jeevalatha A, Basha AN, Kandeepan C, Ramya S, Loganathan T, Jayakumararaj R. In-silico Absorption, Distribution, Metabolism, Elimination and Toxicity profile of Isopulegol from Rosmarinus officinalis. Journal of Drug Delivery and Therapeutics. 2022; 12(1):102-8. https://doi.org/10.22270/jddt.v12i1.5188
18. Ramya S, Loganathan T, Chandran M, Priyanka R, Kavipriya K, Pushpalatha GL, Aruna D, Abraham GC, Jayakumararaj R. ADME-Tox profile of Cuminaldehyde (4-Isopropylbenzaldehyde) from Cuminum cyminum seeds for potential biomedical applications. Journal of Drug Delivery and Therapeutics. 2022; 12(2-S):127-41. https://doi.org/10.22270/jddt.v12i2-S.5286
19. Ramya S, Murugan M, Krishnaveni K, Sabitha M, Kandeepan C, Jayakumararaj R. In-silico ADMET profile of Ellagic Acid from Syzygium cumini: A Natural Biaryl Polyphenol with Therapeutic Potential to Overcome Diabetic Associated Vascular Complications. Journal of Drug Delivery and Therapeutics. 2022; 12(1):91-101. https://doi.org/10.22270/jddt.v12i1.5179
20. Ramya S, Soorya C, Pushpalatha GG, Aruna D, Loganathan T, Balamurugan S, Abraham GC, Ponrathy T, Kandeepan C, Jayakumararaj R. Artificial Intelligence and Machine Learning approach based in-silico ADME-Tox and Pharmacokinetic Profile of α-Linolenic acid from Catharanthus roseus (L.) G. Don. Journal of Drug Delivery and Therapeutics. 2022; 12(2-S):96-109. https://doi.org/10.22270/jddt.v12i2-S.5274
21. Ramya S, Sutha S, Chandran M. Priyanka R, Loganathan T, Pandiarajan G, Kaliraj P, Grace Lydial Pushpalatha G, Abraham GC Jayakumararaj R, ADMET-informatics, Pharmacokinetics, Drug-likeness and Medicinal Chemistry of Bioactive Compounds of Physalis minima Ethanolic Leaf Extract (PMELE) as a Potential Source of Natural Lead Molecules for Next Generation Drug Design, Development and Therapies , Journal of Drug Delivery and Therapeutics. 2022; 12(5):188-200 https://doi.org/10.22270/jddt.v12i5.5654
22. Suganandam K, Jeevalatha A, Kandeepan C, Kavitha N, Senthilkumar N, Sutha S, Syed MA, Gandhi S, Ramya S, Grace Lydial Pushpalatha G, Abraham GC, Jayakumararaj R, Profile of Phytochemicals and GCMS Analysis of Bioactive Compounds in Natural Dried-Seed Removed Ripened Pods Methanolic Extracts of Moringa oleifera, Journal of Drug Delivery and Therapeutics. 2022; 12(5-S):133-141 https://doi.org/10.22270/jddt.v12i5-S.5657
23. Kandeepan C, Kalaimathi RV, Jeevalatha A, Basha AN, Ramya S, Jayakumararaj R. In-silico ADMET Pharmacoinformatics of Geraniol (3, 7-dimethylocta-trans-2, 6-dien-1-ol)-acyclic monoterpene alcohol drug from Leaf Essential Oil of Cymbopogon martinii from Sirumalai Hills (Eastern Ghats), INDIA. Journal of Drug Delivery and Therapeutics. 2021; 11(4-S):109-18. https://doi.org/10.22270/jddt.v11i4-S.4965
24. Loganathan T, Barathinivas A, Soorya C, Balamurugan S, Nagajothi TG, Ramya S, Jayakumararaj R. Physicochemical, Druggable, ADMET Pharmacoinformatics and Therapeutic Potentials of Azadirachtin-a Prenol Lipid (Triterpenoid) from Seed Oil Extracts of Azadirachta indica A. Juss. Journal of Drug Delivery and Therapeutics. 2021; 11(5):33-46. https://doi.org/10.22270/jddt.v11i5.4981
25. Sabitha M, Krishnaveni K, Murugan M, Basha AN, Pallan GA, Kandeepan C, Ramya S, Jayakumararaj R. In-silico ADMET predicated Pharmacoinformatics of Quercetin-3-Galactoside, polyphenolic compound from Azadirachta indica, a sacred tree from Hill Temple in Alagarkovil Reserve Forest, Eastern Ghats, INDIA. Journal of Drug Delivery and Therapeutics. 2021; 11(5-S):77-84. https://doi.org/10.22270/jddt.v11i5-S.5026
26. Soorya C, Balamurugan S, Basha AN, Kandeepan C, Ramya S, Jayakumararaj R. Profile of Bioactive Phyto-compounds in Essential Oil of Cymbopogon martinii from Palani Hills, Western Ghats, INDIA. Journal of Drug Delivery and Therapeutics. 2021; 11(4):60-5. https://doi.org/10.22270/jddt.v11i4.4887
27. Soorya C, Balamurugan S, Ramya S, Neethirajan K, Kandeepan C, Jayakumararaj R. Physicochemical, ADMET and Druggable properties of Myricetin: A Key Flavonoid in Syzygium cumini that regulates metabolic inflammations. Journal of Drug Delivery and Therapeutics. 2021; 11(4):66-73. https://doi.org/10.22270/jddt.v11i4.4890
28. Shanmugam S, Sundari A, Muneeswaran S, Vasanth C, Jayakumararaj R, Rajendran K. Ethnobotanical Indices on medicinal plants used to treat poisonous bites in Thiruppuvanam region of Sivagangai district in Tamil Nadu, India. Journal of Drug Delivery and Therapeutics. 2020; 10(6-s):31-6. https://doi.org/10.22270/jddt.v10i6-s.4432
29. Sundari A, Jayakumararaj R. Herbal remedies used to treat skin disorders in Arasankulam region of Thoothukudi District in Tamil Nadu, India. Journal of Drug Delivery and Therapeutics. 2020; 10(5):33-8. https://doi.org/10.22270/jddt.v10i5.4277
30. Sundari A, Jayakumararaj R. Medicinal plants used to cure cuts and wounds in Athur region of Thoothukudi district in Tamil Nadu, India. Journal of Drug Delivery and Therapeutics. 2020; 10(6-s):26-30. https://doi.org/10.22270/jddt.v10i6-s.4429
31. Meena R, Prajapati SK, Nagar R, Porwal O, Nagar T, Tilak VK, Jayakumararaj R, Arya RK, Dhakar RC. Application of Moringa oleifera in Dentistry. Asian Journal of Dental and Health Sciences. 2021; 1(1):10-3. https://doi.org/10.22270/ijmspr.v6i1.25
32. Beetge E, du Plessis J, Müller DG, Goosen C, van Rensburg FJ. The influence of the physicochemical characteristics and pharmacokinetic properties of selected NSAID's on their transdermal absorption. International journal of Pharmaceutics. 2000; 193(2):261-4. https://doi.org/10.1016/S0378-5173(99)00340-3
33. Mabkhot YN, Alatibi F, El-Sayed NN, Al-Showiman S, Kheder NA, Wadood A, Rauf A, Bawazeer S, Hadda TB. Antimicrobial activity of some novel armed thiophene derivatives and petra/osiris/molinspiration (POM) analyses. Molecules. 2016; 21(2):222. https://doi.org/10.3390/molecules21020222
34. Hauser AS, Attwood MM, Rask-Andersen M, Schiöth HB, Gloriam DE. Trends in GPCR drug discovery: new agents, targets and indications. Nature reviews Drug discovery. 2017 Dec; 16(12):829-42. https://doi.org/10.1038/nrd.2017.178
35. Daina A, Michielin O, Zoete V. SwissADME: a free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Scientific reports. 2017; 7(1):1-3 https://doi.org/10.1038/srep42717
36. Hubatsch I, Ragnarsson EG, Artursson P. Determination of drug permeability and prediction of drug absorption in Caco-2 monolayers. Nature protocols. 2007; 2(9):2111-9. https://doi.org/10.1038/nprot.2007.303
37. Radchenko EV, Dyabina AS, Palyulin VA, Zefirov NS. Prediction of human intestinal absorption of drug compounds. Russian Chemical Bulletin. 2016; 65(2):576-80. https://doi.org/10.1007/s11172-016-1340-0
38. Alam A, Kowal J, Broude E, Roninson I, Locher KP. Structural insight into substrate and inhibitor discrimination by human Pglycoprotein. Science. 2019; 363(6428):753-6. https://doi.org/10.1126/science.aav7102
39. Daina A, Zoete V. A boiled‐egg to predict gastrointestinal absorption and brain penetration of small molecules. ChemMedChem. 2016; 11(11):1117-21. https://doi.org/10.1002/cmdc.201600182
40. Liao M, Jaw-Tsai S, Beltman J, Simmons AD, Harding TC, Xiao JJ. Evaluation of in vitro absorption, distribution, metabolism, and excretion and assessment of drug-drug interaction of rucaparib, an orally potent poly (ADP-ribose) polymerase inhibitor. Xenobiotica. 2020; 50(9):1032-42. https://doi.org/10.1080/00498254.2020.1737759
41. Jia CY, Li JY, Hao GF, Yang GF. A drug-likeness toolbox facilitates ADMET study in drug discovery. Drug Discovery Today. 2020; 25(1):248-58. https://doi.org/10.1016/j.drudis.2019.10.014
42. Cheng F, Li W, Zhou Y, Shen J, Wu Z, Liu G, Lee PW, Tang Y. admetSAR: a comprehensive source and free tool for assessment of chemical ADMET properties.
43. Schyman, P., R. Liu, V. Desai, and A. Wallqvist. vNN web server for ADMET predictions. Frontiers in Pharmacology. 2017 December 4; 8:889. https://doi.org/10.3389/fphar.2017.00889
44. Liu, R., P. Schyman, and A. Wallqvist. Critically assessing the predictive power of QSAR models for human liver microsomal stability. Journal of Chemical Information and Modeling. 2015; 55(8):1566-1575. https://doi.org/10.1021/acs.jcim.5b00255
45. Xu, Y., Z. Dai, F. Chen, S. Gao, J. Pei, and L. Lai. Deep learning for drug induced liver injury. 2015, 55(10):2085-2093. https://doi.org/10.1021/acs.jcim.5b00238
46. Muehlbacher, M., G. Spitzer, K. Liedl, J. Kornhuber. Qualitative prediction of blood-brain barrier permeability on a large and refined dataset. Journal of Computer-Aided Molecular Design. 2011; 25:1095. https://doi.org/10.1007/s10822-011-9478-1
47. Schyman, P., R. Liu, and A. Wallqvist. Using the variable-nearest neighbor method to identify P-glycoprotein substrates and inhibitors. ACS Omega. 2016; 1(5):923-929 https://doi.org/10.1021/acsomega.6b00247
48. Schyman, P., R. Liu, and A. Wallqvist. General purpose 2D and 3D similarity approach to identify hERG blockers. Journal of Chemical Information and Modeling. 2016; 56(1):213-222 https://doi.org/10.1021/acs.jcim.5b00616
49. Liu, R., G. Tawa, and A. Wallqvist. Locally weighted learning methods for predicting dose-dependent toxicity with application to the human maximum recommended daily dose. Chemical Research in Toxicology. 2012; 25(10):2216-2226. https://doi.org/10.1021/tx300279f
Figure 1: Schematic diagram of Bioavailability Radar for Drug likeness of SA (lipophilicity: XLOGP3 between-0.7 and+5.0, size: MW between 150 and 500 g/mol, polarity: TPSA between 20 and 130 A2, solubility: log S not higher than 6, saturation: fraction of carbons in the sp3 hybridization not less than 0.25, and flexibility: no more than 9 rotatable bonds
Figure 2: Percentage distribution of function targets for SA using SwissTargetPrediction
Figure 3: Schematic representation of perceptive evaluation of passive gastrointestinal absorption (HIA) and Brain penetration (BBB) of SA with WLOGP-versus-TPSA using BOILED-Egg
Table 1: In silico Drug-Likeliness and Bioactivity Prediction
|
|
Molecular Properties |
Calculated Values |
|
8.07 |
||
|
37.30 |
||
|
Natoms |
20 |
|
|
MW |
284.48 |
|
|
nON |
2 |
|
|
nOHNH |
1 |
|
|
Nviolations |
1 |
|
|
Nrotb |
16 |
|
|
325.03 |
||
|
Biological Properties |
Bioactivity Scores |
|
|
GPCR ligand |
0.11 |
|
|
Ion channel modulator |
0.05 |
|
|
Kinase inhibitor |
-0.20 |
|
|
Nuclear receptor ligand |
0.17 |
|
|
Protease inhibitor |
0.06 |
|
|
Enzyme inhibitor |
0.20 |
Table 2: Predicted Target/ Target Class and Functional Probabilities of SA
|
TARGET |
COMMON.NAME |
UNIPROT.ID |
TARGET CLASS |
PROBABILITY* |
|
Peroxisome proliferator-activated receptor alpha |
Nuclear receptor |
0.929299883958 |
||
|
Peroxisome proliferator-activated receptor delta |
Nuclear receptor |
0.929299883958 |
||
|
Fatty acid binding protein adipocyte |
Fatty acid BPF |
0.714850037542 |
||
|
Fatty acid binding protein epidermal |
Fatty acid BPF |
0.714850037542 |
||
|
Fatty acid binding protein muscle |
Fatty acid BPF |
0.526361274524 |
||
|
Fatty acid binding protein intestinal |
Fatty acid BPF |
0.526361274524 |
||
|
Free fatty acid receptor 1 |
Family A GPCR |
0.370888463379 |
||
|
Solute carrier family 22 member 6 |
Electrochemical transporter |
0.207053973629 |
||
|
Dual specificity phosphatase Cdc25A |
Phosphatase |
0.17427075329 |
||
|
Aldo-keto reductase family 1 member B10 |
Enzyme |
0.149732593856 |
||
|
11-beta-hydroxysteroid dehydrogenase 1 |
Enzyme |
0.149732593856 |
||
|
Bile acid receptor FXR |
Nuclear receptor |
0.125142648574 |
||
|
UDP-glucuronosyltransferase 2B7 |
Enzyme |
0.125142648574 |
||
|
Prostanoid EP2 receptor |
Family A GPCR |
0.125142648574 |
||
|
DNA polymerase beta |
Enzyme |
0.125142648574 |
||
|
Cytochrome P450 19A1 |
Cytochrome P450 |
0.116965063224 |
||
|
Corticosteroid binding globulin |
Secreted protein |
0.116965063224 |
||
|
Testis-specific androgen-binding protein |
Secreted protein |
0.116965063224 |
||
|
Estradiol 17-beta-dehydrogenase 3 |
Enzyme |
0.116965063224 |
||
|
Glucose-6-phosphate 1-dehydrogenase |
Enzyme |
0.116965063224 |
||
|
GABA-B receptor |
Family C GPCR |
0.116965063224 |
||
|
G-protein coupled bile acid receptor 1 |
Family A GPCR |
0.108770969359 |
||
|
Niemann-Pick C1-like protein 1 |
Other membrane protein |
0.108770969359 |
||
|
GABA A receptor alpha-2/beta-2/gamma-2 |
Ligand-gated ion channel |
0.108770969359 |
||
|
Lysine-specific demethylase 2A |
Eraser |
0.108770969359 |
||
|
Lysine-specific demethylase 5C |
Eraser |
0.108770969359 |
||
|
Vitamin D receptor |
Nuclear receptor |
0.108770969359 |
||
|
Androgen Receptor |
Nuclear receptor |
0.100578902067 |
||
|
Protein farnesyltransferase |
Enzyme |
0.100578902067 |
||
|
Histone lysine demethylase PHF8 |
Eraser |
0.100578902067 |
||
|
Plasminogen |
Protease |
0.100578902067 |
||
|
Glutathione S-transferase kappa 1 |
Enzyme |
0.100578902067 |
||
|
Protein-tyrosine phosphatase 1B |
Phosphatase |
0.100578902067 |
||
|
Anandamide amidohydrolase |
Enzyme |
0.100578902067 |
||
|
Peroxisome proliferator-activated receptor gamma |
Nuclear receptor |
0.100578902067 |
||
|
Telomerase reverse transcriptase |
Enzyme |
0.100578902067 |
||
|
Fatty acid-binding protein, liver |
Fatty acid BPF |
0.100578902067 |
||
|
Retinoic acid receptor gamma |
Nuclear receptor |
0.100578902067 |
||
|
Retinoic acid receptor beta |
Nuclear receptor |
0.100578902067 |
||
|
Retinoic acid receptor alpha |
Nuclear receptor |
0.100578902067 |
||
|
Glycine receptor subunit alpha-1 |
Ligand-gated ion channel |
0.100578902067 |
||
|
11-beta-hydroxysteroid dehydrogenase 2 |
Enzyme |
0.100578902067 |
||
|
Prostanoid FP receptor |
Family A GPCR |
0.100578902067 |
||
|
CDC45-related protein |
Other nuclear protein |
0.100578902067 |
||
|
Leukocyte common antigen |
Enzyme |
0.100578902067 |
||
|
Hydroxyacid oxidase 1 |
Enzyme |
0.100578902067 |
||
|
Nuclear receptor subfamily 0 group B member 2 |
Nuclear receptor |
0.100578902067 |
||
|
Cytochrome P450 26B1 |
Cytochrome P450 |
0.100578902067 |
||
|
Cytochrome P450 26A1 |
Cytochrome P450 |
0.100578902067 |
||
|
Acyl-CoA desaturase |
Enzyme |
0.100578902067 |
||
|
Retinoid X receptor beta |
Nuclear receptor |
0.100578902067 |
||
|
Retinoid X receptor gamma |
Nuclear receptor |
0.100578902067 |
||
|
Retinoid X receptor alpha |
Nuclear receptor |
0.100578902067 |
||
|
Voltage-gated calcium channel alpha2/delta subunit 1 |
Calcium channel |
0.100578902067 |
||
|
HMG-CoA reductase |
Oxidoreductase |
0.100578902067 |
||
|
Prostanoid EP4 receptor |
Family A GPCR |
0.100578902067 |
||
|
Neuronal acetylcholine receptor protein alpha-7 subunit |
Ligand-gated ion channel |
0.100578902067 |
||
|
Carbonic anhydrase II |
Lyase |
0.100578902067 |
||
|
Carbonic anhydrase I |
Lyase |
0.100578902067 |
||
|
Glucagon |
Unclassified protein |
0.100578902067 |
||
|
SUMO-activating enzyme |
Enzyme |
0.100578902067 |
||
|
Metabotropic glutamate receptor 5 |
Family C GPCR |
0.100578902067 |
||
|
Phosphodiesterase 4A |
Phosphodiesterase |
0.100578902067 |
||
|
Phosphodiesterase 4B |
Phosphodiesterase |
0.100578902067 |
Table 3: Predicted ADMET Properties of SA
|
Property |
Model Name |
Predicted Value |
Unit |
|
Absorption |
Water solubility |
-5.973 |
Numeric (log mol/L) |
|
Absorption |
Caco2 permeability |
1.556 |
Numeric (log Papp in 10-6 cm/s) |
|
Absorption |
Intestinal absorption (human) |
91.317 |
Numeric (% Absorbed) |
|
Absorption |
Skin Permeability |
-2.726 |
Numeric (log Kp) |
|
Absorption |
P-glycoprotein substrate |
No |
Categorical (Yes/No) |
|
Absorption |
P-glycoprotein I inhibitor |
No |
Categorical (Yes/No) |
|
Absorption |
P-glycoprotein II inhibitor |
No |
Categorical (Yes/No) |
|
Distribution |
VDss (human) |
-0.528 |
Numeric (log L/kg) |
|
Distribution |
Fraction unbound (human) |
0.051 |
Numeric (Fu) |
|
Distribution |
BBB permeability |
-0.195 |
Numeric (log BB) |
|
Distribution |
CNS permeability |
-1.707 |
Numeric (log PS) |
|
Metabolism |
CYP2D6 substrate |
No |
Categorical (Yes/No) |
|
Metabolism |
CYP3A4 substrate |
Yes |
Categorical (Yes/No) |
|
Metabolism |
CYP1A2 inhibitior |
Yes |
Categorical (Yes/No) |
|
Metabolism |
CYP2C19 inhibitior |
No |
Categorical (Yes/No) |
|
Metabolism |
CYP2C9 inhibitior |
No |
Categorical (Yes/No) |
|
Metabolism |
CYP2D6 inhibitior |
No |
Categorical (Yes/No) |
|
Metabolism |
CYP3A4 inhibitior |
No |
Categorical (Yes/No) |
|
Excretion |
Total Clearance |
1.832 |
Numeric (log ml/min/kg) |
|
Excretion |
Renal OCT2 substrate |
No |
Categorical (Yes/No) |
|
Toxicity |
AMES toxicity |
No |
Categorical (Yes/No) |
|
Toxicity |
Max. tolerated dose (human) |
-0.791 |
Numeric (log mg/kg/day) |
|
Toxicity |
hERG I inhibitor |
No |
Categorical (Yes/No) |
|
Toxicity |
hERG II inhibitor |
No |
Categorical (Yes/No) |
|
Toxicity |
Oral Rat Acute Toxicity (LD50) |
1.406 |
Numeric (mol/kg) |
|
Toxicity |
Oral Rat Chronic Toxicity (LOAEL) |
3.33 |
Numeric (log mg/kg_bw/day) |
|
Toxicity |
Hepatotoxicity |
No |
Categorical (Yes/No) |
|
Toxicity |
Skin Sensitisation |
Yes |
Categorical (Yes/No) |
|
Toxicity |
T.Pyriformis toxicity |
0.65 |
Numeric (log ug/L) |
|
Toxicity |
Minnow toxicity |
-1.565 |
Numeric (log mM) |
Table 4: Summative Physicochemical, Druggable, ADMET Properties of SA
Property |
Value |
|
|
Molecular weight |
284.48 g/mol |
|
|
LogP |
6.03 |
|
|
LogD |
6.76 |
|
|
LogSw |
-5.51 |
|
|
Number of stereocenters |
0 |
|
|
Stereochemical complexity |
0.000 |
|
|
Fsp3 |
0.944 |
|
|
Topological polar surface area |
26.30 Å2 |
|
|
Number of hydrogen bond donors |
0 |
|
|
Number of hydrogen bond acceptors |
2 |
|
|
Number of smallest set of smallest rings (SSSR) |
0 |
|
|
Size of the biggest system ring |
0 |
|
|
Number of rotatable bonds |
15 |
|
|
Number of rigid bonds |
1 |
|
|
Number of charged groups |
0 |
|
|
Total charge of the compound |
0 |
|
|
Number of carbon atoms |
18 |
|
|
Number of heteroatoms |
2 |
|
|
Number of heavy atoms |
20 |
|
|
Ratio between the number of non-carbon atoms and the number of carbon atoms |
0.11 |
|
|
Druggability Properties |
|
|
|
Lipinski's rule of 5 violations |
1 |
|
|
Veber rule |
Good |
|
|
Egan rule |
Good |
|
|
Oral PhysChem score (Traffic Lights) |
4 |
|
|
GSK's 4/400 score |
Good |
|
|
Pfizer's 3/75 score |
Bad |
|
|
Weighted quantitative estimate of drug-likeness (QEDw) score |
0.213 |
|
|
Solubility |
1148.54 |
|
|
Solubility Forecast Index |
Reduced |
|
|
ADMET Properties Property |
Value |
|
|
Human Intestinal Absorption |
HIA+ |
0.994 |
|
Blood Brain Barrier |
BBB+ |
0.986 |
|
Caco-2 permeable |
Caco2+ |
0.801 |
|
P-glycoprotein substrate |
Non-substrate |
0.708 |
|
P-glycoprotein inhibitor I |
Non-inhibitor |
0.913 |
|
P-glycoprotein inhibitor II |
Non-inhibitor |
0.889 |
|
CYP450 2C9 substrate |
Non-substrate |
0.870 |
|
CYP450 2D6 substrate |
Non-substrate |
0.892 |
|
CYP450 3A4 substrate |
Non-substrate |
0.643 |
|
CYP450 1A2 inhibitor |
Inhibitor |
0.500 |
|
CYP450 2C9 inhibitor |
Non-inhibitor |
0.928 |
|
CYP450 2D6 inhibitor |
Non-inhibitor |
0.923 |
|
CYP450 2C19 inhibitor |
Non-inhibitor |
0.939 |
|
CYP450 3A4 inhibitor |
Non-inhibitor |
0.951 |
|
CYP450 inhibitory promiscuity |
Low CYP Inhibitory Promiscuity |
0.852 |
|
Ames test |
Non AMES toxic |
0.963 |
|
Carcinogenicity |
Non-carcinogens |
0.575 |
|
Biodegradation |
Ready biodegradable |
0.937 |
|
Rat acute toxicity |
1.328 LD50, mol/kg |
NA |
|
hERG inhibition (predictor I) |
Weak inhibitor |
0.929 |
|
hERG inhibition (predictor II) |
Non-inhibitor |
0.849 |
Table 5 Performance measures of vNN models in 10-fold cross validation using a restricted or unrestricted applicability domain
|
Model |
Dataa |
d0b |
hc |
Accuracy |
Sensitivity |
Specificity |
kappa |
Rd |
Coverage |
|
DILI |
1427 |
0.60 |
0.50 |
0.71 |
0.70 |
0.73 |
0.42 |
0.66 |
|
|
1.00 |
0.20 |
0.67 |
0.62 |
0.72 |
0.34 |
1.00 |
|||
|
Cytotox (hep2g) |
6097 |
0.40 |
0.20 |
0.84 |
0.88 |
0.76 |
0.64 |
0.89 |
|
|
1.00 |
0.20 |
0.84 |
0.73 |
0.89 |
0.62 |
1.00 |
|||
|
HLM |
3219 |
0.40 |
0.20 |
0.81 |
0.72 |
0.87 |
0.59 |
0.91 |
|
|
1.00 |
0.20 |
0.81 |
0.70 |
0.87 |
0.57 |
1.00 |
|||
|
CYP1A2 |
7558 |
0.50 |
0.20 |
0.90 |
0.70 |
0.95 |
0.66 |
0.75 |
|
|
1.00 |
0.20 |
0.89 |
0.61 |
0.95 |
0.60 |
1.00 |
|||
|
CYP2C9 |
8072 |
0.50 |
0.20 |
0.91 |
0.55 |
0.96 |
0.54 |
0.76 |
|
|
1.00 |
0.20 |
0.90 |
0.44 |
0.96 |
0.46 |
1.00 |
|||
|
CYP2C19 |
8155 |
0.55 |
0.20 |
0.87 |
0.64 |
0.93 |
0.58 |
0.76 |
|
|
1.00 |
0.20 |
0.86 |
0.52 |
0.94 |
0.50 |
1.00 |
|||
|
CYP2D6 |
7805 |
0.50 |
0.20 |
0.89 |
0.61 |
0.94 |
0.57 |
0.75 |
|
|
1.00 |
0.20 |
0.88 |
0.52 |
0.95 |
0.51 |
1.00 |
|||
|
CYP3A4 |
10373 |
0.50 |
0.20 |
0.88 |
0.76 |
0.92 |
0.68 |
0.78 |
|
|
1.00 |
0.20 |
0.88 |
0.69 |
0.93 |
0.64 |
1.00 |
|||
|
BBB |
353 |
0.60 |
0.20 |
0.90 |
0.94 |
0.86 |
0.80 |
0.61 |
|
|
1.00 |
0.10 |
0.82 |
0.88 |
0.75 |
0.64 |
1.00 |
|||
|
Pgp Substrate |
822 |
0.60 |
0.20 |
0.79 |
0.80 |
0.79 |
0.58 |
0.66 |
|
|
1.00 |
0.20 |
0.73 |
0.73 |
0.74 |
0.47 |
1.00 |
|||
|
Pgp Inhibitor |
2304 |
0.50 |
0.20 |
0.85 |
0.91 |
0.73 |
0.66 |
0.76 |
|
|
1.00 |
0.10 |
0.81 |
0.86 |
0.74 |
0.61 |
1.00 |
|||
|
hERG |
685 |
0.70 |
0.70 |
0.84 |
0.84 |
0.83 |
0.68 |
0.80 |
|
|
1.00 |
0.20 |
0.82 |
0.82 |
0.83 |
0.64 |
1.00 |
|||
|
MMP |
6261 |
0.50 |
0.40 |
0.89 |
0.64 |
0.94 |
0.61 |
0.69 |
|
|
1.00 |
0.20 |
0.87 |
0.52 |
0.94 |
0.50 |
1.00 |
|||
|
AMES |
6512 |
0.50 |
0.40 |
0.82 |
0.86 |
0.75 |
0.62 |
0.79 |
|
|
1.00 |
0.20 |
0.79 |
0.82 |
0.75 |
0.57 |
1.00 |
|||
|
MRTDe |
1184 |
0.60 |
0.20 |
-0.79 |
0.69 |
||||
|
1.00 |
0.20 |
-0.74 |
1.00 |