1. Introduction
Ocimum sanctum L. (also known as Tulsi) has been used for thousands of years in Ayurveda for its diverse healing properties. Tulsi, the Queen of herbs, the legendary ‘Incomparable one’ of India, is one of the holiest and most cherished of the many healing and healthy giving herbs of the orient (Priyabrata Pattanayak
et al., 2010) [
1].
Ocimum sanctum L. also known as “
Tulsi,” is an aromatic plant in the basil family Lamiaceae (tribe ocimeae), which is native throughout the eastern world tropics. It is an erect, much branched subshrub, 30–60 cm tall with hairy stems and simple, opposite, green leaves that are strongly scented. Leaves have petioles and are ovate up-to 5 cm long, usually slightly toothed (Felix Bast
et al., 2014) [
2].
Tulsi has been used traditionally in Ayurveda and Siddha systems of medicine for prevention and cure of common cold, headache, cough, influenza, earache, fever, colic pain, sore throat, bronchitis, asthma, hepatic diseases, malarial fever, as an antidote for snake bite and scorpion sting, flatulence, migraine headaches, fatigue, skin diseases, wound, insomnia, arthritis, digestive disorders, night blindness and diarrhoea (Rakesh Kumar Joshi
etal., 2017)[
3]. It is one of the holiest and most sacred herbs grown widely in India. It is a herb that is bestowed with enormous antimicrobial substances and is used to treat a variety of illnesses ranging from diabetes mellitus, arthritis, bronchitis, skin diseases, etc. (Prakash P
et al., 2005 & Viyoch J
et al., 2006 & Magesh V
et al., 2009) [
4,
5,
6].
The Plant has many medicinal properties like Antimalarial, Insecticidal activity, Treating Hepatic disorders, Antiemetic and Anti helminthic action, Antidiabetic, Animal bite antidote, Antiulcer activity, Heart tonic activity, Antifertility effect, Anti stress activity and other Pharmacological Activity (Harish Chandra Andola
et al., 2011 & Amit Kumar
et al., 2013 & Nipun Mahajan
et al., 2013) [
7,
8,
9]. One such website that supports drug development is Swiss ADME. It allows individuals to compute physicochemical characteristics as well as predict ADME parameters, pharmacokinetic features, drug-like nature, and medicinal chemistry compatibility of one or more small compounds. The aim of the present study was to investigate the individual ADME behaviour of the bioactive substances found in Ocimum sanctum and interpret the findings using the Swiss ADME website (
www.swissadme.ch).
2. Materials and Methods
2.1. Swiss ADME
The Swiss Institute of Bioinformatics' Swiss ADME software (
www.swissadme.ch) was accessed using a web server that shows the Swiss ADME submission page in Google to estimate the individual ADME behaviours of the constituents from Ocimum sanctum. The findings have been presented for each input molecule in tables, graphs, and an excel spreadsheet. The list was created to handle one input molecule per line that contains several inputs, described by the simplified molecular input line entry system (SMILES).
2.2. Physicochemical properties
The two-dimensional chemical structure was described in the first section using canonical SMILES. A preliminary assessment of the pharmacological similarity of the compounds of interest is given by the bioavailability radar, which considers six physicochemical parameters: LIPO (Lipophilicity), SIZE, POLAR (Polarity), INSOLU (Insolubility), INSATU (Insaturation), and FLEX (Flexibility). These sections include clean molecular and physicochemical properties like molar refractivity, TPSA, number of rotatable bonds, number of H-bond acceptors, number of H-bond donors, number of heavy atoms, number of aromatic heavy atoms, and fraction csp3.
2.3. Solubility
The solvent used, along with the temperature and pressure of the surrounding environment, have a significant impact on a compound's solubility. The range of solubility is defined as the saturation concentration, at which the concentration of the solute in the solution does not rise as more solute is added (Lachman
et al., 1986) [
10]. When the maximum dose strength of the drug dissolves in 250 mL or less of aqueous media with a pH range of 1 to 7.5, that drug is considered to be extremely soluble. Two topological approaches included in Swiss ADME to predict water solubility, the first one is the application of ESOL model (Solubility class: Log S Scale: Insoluble<-10 poorly<-6, moderately<-4 soluble<-2, very<0<highly) and the second one is (Solubility class: Log S Scale: Insoluble<-10 poorly<-6, moderately<-4 soluble<-2, very<0<highly). Both differ from the fundamental general solubility equation (Yalkowsky
et al.,1980) [
11]. Since they avoid the melting point parameter but the linear correlation between predicted and experimental values were strong (R2=0.69 and 0.81 respectively). The third predictor of Swiss ADME was developed by SILICOS-IT (Solubility class: Log S Scale: Insoluble<-10 poorly<-6, moderately<-4 soluble<-2 very<0<highly) where the linear coefficient is corrected by molecular weight (R2=0.75). The fractional logarithm of the molar solubility in water (log S) represents all expected values. Along with qualitative solubility classes, Swiss ADME also offers solubility in mol/l and mg/ml.
2.4. Lipophilicity
Lipophilicity is a crucial molecular characteristic that frequently has a positive correlation with the bioactivity of substances (Melanie Kah
et al., 2008) [
12]. It is demonstrated experimentally using either distribution coefficients (log D) or partition coefficients (log P). The representation of P illustrates the partition equilibrium of a unionized solute between water and an immiscible organic solvent. One of the key physicochemical characteristics, lipophilicity has connections to both pharmacodynamic and pharmacokinetic properties. It establishes how medications are absorbed, metabolized, distributed, excreted, and toxic (ADMET) (Lungu
et al., 2019 & Giaginis
et al., 2018 & Kamel M.S
et al., 2022 & Erckes V
et al., 2022) [
13,
14,
15,
16].
A crucial element in the transport of molecules through membranes is lipophilicity. Additionally, it affects how they bind to receptors at the drug's action site and to plasma proteins. Lipophilicity is therefore regarded as a reference measure for the estimation of a drug's biological activity. In terms of biology, lipophillicity is the logarithm of a chemical's n-octanol to water partition (logP). This parameter has been used in studies on the quantitative relationship between the structure and the activity (QSAR) (Ginex T
et al., 2019 & Kempinska D
et al., 2019 & Dolowy M
et al., 2021) [
17,
18,
19]. Swiss ADME presents five accessible models, namely XLOGP3, WLOGP, MLOGP, SILICOS-IT, and iLOGP, for analysing the lipophilicity character in a molecule. XLOGP3, an atomistic accost including corrective factors and knowledge based library (Cheng T
et al., 2007) [
20]; WLOGP, application of purely atomistic method stationed on fragmental system (Wildman SA
et al., 1999) [
21]; MLOGP, 13 molecular descriptors were implemented in an archetype of topological technique indicated on a linear relationship (Moriguchi
et al., 1994) [
22]; SILICOS-IT, 7 topological descriptors and 27 fragments are used in a mongrel technique; iLOGP, a physics-based technique that relies on the generalized-born and solvent accessible surface area (GB/SA) model to compute the free energies of solvation in n-Octanol and water; An arithmetic average of the values predicted by the five suggested approaches is called consensus log P o/w. (Daina Antoine
et al.,2017) [
23].
2.5. Drug likeness
Swiss ADME filters chemical libraries using five different rule-based filters from major pharmaceutical companies in an attempt to enhance the quality of proprietary chemical groups by eliminating molecules with characteristics that are not compatible with an acceptable pharmacokinetics profile (Daina Antoine
et al.,2017) [
23].
The Muegge filter (also known as the Bayer filter) distinguishes molecules that are identical to drugs from substances that are different. These models illustrate molecules as drugs, such as those with molecular weights of 200 to 600 Da, XLOGP values within -2 and 5, TPSA values of 150, etc. Number of rings is 7, number of carbon atoms is greater than 4, number of heteroatoms is greater than 1, number of rotatable bonds is 15, and H-bond acceptor and donor are 10 and 5 respectively.
Egan filter (Pharmacia filter) predicts that mechanisms involved in a small molecule's membrane permeability will influence drug absorption. These models illustrate a drug's molecule assuming it were given a WLOGP value of 5.88 and a TPSA value of 131.6, respectively. Because it takes into consideration active transport and efflux mechanisms, the Egan computational model for human passive intestinal absorption (HIA) of small molecules is accurate in predicting drug absorption (Egan WJ
et al., 2000) [
24].
According to physicochemical properties, the presence of functional groups, and substructures, the Ghose filter (Amgen) determines small molecules. The qualifying range for small molecules is between 20 and 70 atoms, whereas the qualifying range for large molecules is between 160 and 480 Da, WlogP is between -0.4 and 5.6, and molar refractivity (MR) is between 40 and 130 (Ghose AK
et al., 1998) [
25].
Veber filter (GSK filter) model classifies compounds as drug-like if they have 12 or less H-bond donors and acceptors, a TPSA of 140 or less, and fewer than 10 rotatable bonds. Reduced TPSA correlates with increased permeation rate, increased rotatable bond counts correlate to lower permeation rate, and compounds having these characteristics will have good oral bioavailability (Veber DF
et al., 2002) [
26].
Molecular weight (MW) less than 500, MLOGP ≤ 4.15, N or O ≤ 10, NH or OH ≤ 5, and the Lipinski filter (Pfizer) are the pioneer rules of five that describe tiny compounds based on physicochemical property profiles. All nitrogen and oxygen are strictly considered by Lipinski as H-bond acceptors, while all nitrogen and oxygen that contain at least one hydrogen are regarded as H-bond donors. In addition, neither nitrogen nor aliphatic fluorine are donors or acceptors (Lipinski CA
et al., 2001) [
27].
The Abbott bioavailability score is intended to predict the probability that a substance will have at least 10% oral bioavailability in rodents or measurable Caco-2 permeability, which predicts the probability that a compound would have F>10% based on the predominant charge at biological pH in a rat model. It involves rapid chemical libraries screening to identify the optimum compounds for synthesis (Martin YC, 2005) [
28].
2.6. Medicinal Chemistry
These areas of study are intended to assist medicinal chemistry investigators in their ongoing seek for new medications. In particular, these substances have been shown to be active in a number of tests, which could be considered as prospective beginning points for additional research. Frequent hits or promiscuous compounds, often known as PAINS (Pan Assay Interference Compounds), are chemicals that exhibit a robust reaction in assays regardless of the protein Targets. SwissADME returns warnings if such moieties are found in the molecule under evaluation (Baell
et al., 2010) [
29].
To allow leads through high throughput screening (HTS) with exceptional affinity the chance to be utilized for more interactions throughout the lead optimization phase, the concept of a lead likeness was developed. Leads are exposed to chemical changes that will most likely cause them to shrink and become more lipophilic in nature which is less hydrophobic than molecules similar to drugs molecules.
Lead optimization has been done by rule based method consisting of molecules with molecular weight in between 100 and 350 Da, ClogP between 1 and 3.0 and are greatly considered as superior to those of drug like compounds (Teague.S
et al.,1999 & Hann MM
et al.,2012) [
30,
31].
2.7. Pharmacokinetics
A plot between WLOGP and TPSA is drawn to indicate the estimated amounts for gastrointestinal absorption and brain penetration of compounds. The Egan egg, which is used to evaluate the predictive efficacy of the model for GI passive absorption and prediction for brain access by passive diffusion to ultimately lay the BOILED-Egg (Brain Or IntestinaL EstimateD permeation model), is an elliptical region that is most populated by easily absorbed molecules.
For the purpose of drug discovery and research, the BOILED-Egg model generates a rapid, spontaneously accurately mimicked, yet lively way to forecast passive GI absorption (Brito Sanchez
et al., 2015 & Di LP et al., 2012) [
32,
33].
The molecules in the white region are those that are more likely to get absorbed by the GI tract, while the molecules in the yellow region (the yolk) are those that are most likely to reach the brain. The knowledge about compounds being substrate or non-substrate of the permeability glycoprotein (P-gp, suggested the most important member among ATP-binding cassette transporters or ABC-transporters) is key to appraise active efflux through biological membranes, for instance from the gastrointestinal wall to the lumen or from the brain (Montanari et al.,2015) [
34]. Through metabolic biotransformation, Cytochromes P450 (CYP), a superfamily of isoenzymes, is an essential component of drug elimination (Testa
et al., 2007) [
35].
One can estimate that 50 to 90% (depending on the authors) of therapeutic molecules are substrate of five major isoforms (CYP1A2, CYP2C19, CYP2C9, CYP2D6, CYP3A4) (Di, L.,2014) [
36]. SwissADME provides it possible to determine whether a compound is a P-gp substrate or an inhibitor of the most significant CYP isoenzymes. On thoroughly cleaned, large databases of known substrates/non-substrates or inhibitors/non-inhibitors, we employ the support vector machine algorithm (SVM) (Cortes et al.,1995) [
37].
SVM was found to perform better than other machine-learning algorithms for binary classification. The models return “Yes” or “No” if the molecule under investigation has higher probability to be substrate or non-substrate of P-gp (respectively inhibitor or non-inhibitor of a given CYP) (Mishra
et al.,2010) [
38].
3. Results
Table 1.
General Characteristics of Phytoconstituents of Ocimum sanctum.
Table 1.
General Characteristics of Phytoconstituents of Ocimum sanctum.
Sl.No |
Molecules |
Formula |
Molecular Weight (in g/mol) |
Canonical SMILES |
1 |
MethylEugenol |
C11H14O2
|
178.23 |
C=CCc1ccc(c(c1)OC)OC |
2 |
Eugenol |
C10H12O2
|
164.2 |
C=CCc1ccc(c(c1)OC)O |
3 |
δ-Caryophyllene |
C15H24
|
204.35 |
C/C/1=C\CCC(=C)[C@@H]2 [C@@H](CC1)C(C2)(C)C |
4 |
Caryophyllene oxide |
C17H30O |
250.42 |
C[C@]12CC[C@@]3(C)O[C@]3 (CCC[C@@]2(CC1(C)C)C)C |
5 |
β-Elemene |
C15H24
|
204.35 |
C=C[C@]1(C)CC[C@H] (C[C@H]1C(=C)C)C(=C)C |
6 |
MethylChavicol |
C10H12O |
148.2 |
COc1ccc(cc1)CC=C |
7 |
Linalool |
C10H18O |
154.25 |
C=C[C@](CCC=C(C)C)(O)C |
8 |
δ-Cardinene |
C15H24
|
204.35 |
CC1=C[C@H]2C(=C(C) CC[C@@H]2C(C)C)CC1 |
9 |
β-Bisabolene |
C15H24 |
204.35 |
CC(=CCCC(=C)[C@H] 1CCC(=CC1)C)C |
10 |
1,8-Cineole |
C10H16 |
154.25 |
CC12CCC(CC1)C(O2)(C)C |
11 |
Camphor |
C10H16O |
152.23 |
O=C1CC2C(C1(C)CC2)(C)C |
12 |
Isocaryophyllene |
C10H16 |
136.23 |
CC1=CCC2C(C1)C2(C)C |
13 |
Apigenin-7-O-glucuronide |
C21H18O11 |
446.36 |
OC(=O)[C@H]1O[C@H](Oc2cc(O)c3c(c2)oc(cc3=O) c2ccc(cc2)O)[C@@H]([C@H]([C@@H]1O)O)O |
14 |
Carvacrol |
C10H14O |
150.22 |
CC(c1ccc(c(c1)O)C)C |
15 |
Circimaritin |
C17H14O6
|
314.29 |
COc1cc2oc(cc(=O)c2c (c1OC)O)c1ccc(cc1)O |
16 |
Isothymusin |
C17H14O7
|
330.29 |
COc1c(O)c2oc(cc(=O)c 2c(c1OC)O)c1ccc(cc1)O |
17 |
Pinene |
C11H16 |
148.24 |
CC1=CCC23C1C(C)(C)C2C3 |
18 |
Molludistin |
C21H20O9 |
416.38 |
COc1cc(O)c2c(c1[C@@H]1OC[C@@H] ([C@@H](C1O)O)O)oc(cc2=O)c1ccc(cc1)O |
19 |
Rosameric acid |
C18H16O8 |
360.31 |
O=C(O[C@@H](C(=O)O)Cc1ccc(c(c1) O)O)/C=C/c1ccc(c(c1)O)O |
20 |
Orientin |
C21H22O11
|
450.39 |
OC[C@H]1O[C@@H]([C@@H]([C@H]([C@@H] 1O)O)O)c1c(O)cc(c2c1OC(=CC2O)c1ccc(c(c1)O)O)O |
21 |
Vicenin |
C27H30O15 |
594.52 |
OC[C@H]1O[C@H]([C@@H]([C@H]([C@@H]1O) O)O)c1c(O)c([C@@H]2O[C@H](CO)[C@H]([C@@H] ([C@H]2O)O)O)c(c2c1oc(cc2=O)c1ccc(cc1)O)O |
22 |
Urosolic acid |
C30H48O3 |
456.7 |
C[C@@H]1CC[C@]2([C@@H]([C@H]1C) C1=CC[C@H]3[C@@]([C@@]1(CC2)C)(C)CC [C@@H]1[C@]3(C)CC[C@@H](C1(C)C)O)C(=O)O |
23 |
Luteolin |
C15H10O6
|
286.24 |
Oc1cc(O)c2c(c1)oc(cc2=O)c1ccc(c(c1)O)O |
24 |
Luteolin-7-O-glucuronide |
C21H20O12 |
464.38 |
OC(=O)[C@H]1O[C@@H](Oc2cc3OC(=CC(c3c(c2)O) O)c2ccc(c(c2)O)O)[C@@H]([C@H]([C@@H]1O)O)O |
25 |
3-Carene |
C10H16 |
136.23 |
CC1=CC[C@@H]2[C@H](C1)C2(C)C |
26 |
Citral |
C10H16O |
152.23 |
O=C/C=C(\CCC=C(C)C)/C |
27 |
Geraniol |
C10H18O |
154.25 |
OC/C=C(/CCC=C(C)C)\C |
28 |
Methyl Cinnamate |
C11H12O2 |
176.21 |
CC(=O)OC/C=C/c1ccccc1 |
29 |
β-Ocimene |
C10H16
|
136.23 |
C=C/C(=C\CC=C(C)C)/C |
30 |
λ-Terpineol |
C10H18O |
154.25 |
CC1=CCC(CC1)C(O)(C)C |
31 |
PhenylPropanoids |
C9H11NO2 |
165.19 |
N[C@H](C(=O)O)Cc1ccccc1 |
32 |
Germacrene-D |
C15H24 |
204.35 |
C/C/1=C/CCC(=C)/C=C\[C@@H](CC1)C(C)C |
33 |
λ-Humulene |
C15H24
|
204.35 |
C/C/1=C\CC(C)(C)/C=C/C/C(=C/CC1)/C |
34 |
Camphene |
C10H16
|
136.23 |
C=C1C2CCC(C1(C)C)C2 |
35 |
Myrcene |
C10H16
|
136.23 |
C=CC(=C)CCC=C(C)C |
36 |
Thymol |
C10H14O |
150.22 |
Cc1ccc(c(c1)O)C(C)C
|
37 |
λ-Linolenic acid |
C18H30O2
|
278.43 |
CC/C=C\C/C=C\C/C=C\CCCCCCCC(=O)O |
Table 2.
Physicochemical Properties of the Phytoconstituents of Ocimum sanctum Linn.
Table 2.
Physicochemical Properties of the Phytoconstituents of Ocimum sanctum Linn.
Sl.No |
Molecules |
Heavy atoms |
Aromatic heavy atoms |
Fraction Csp3 |
Rotatable bonds |
H-bond acceptors |
H-bond donors |
Molar Refractivity |
TPSA |
1 |
MethylEugenol |
13 |
6 |
0.27 |
4 |
2 |
0 |
53.53 |
18.46 |
2 |
Eugenol |
12 |
6 |
0.2 |
3 |
2 |
1 |
49.06 |
29.46 |
3 |
δ-Caryophyllene |
15 |
0 |
0.73 |
0 |
0 |
0 |
68.78 |
0 |
4 |
Caryophyllene oxide |
18 |
0 |
1 |
0 |
1 |
0 |
77.87 |
12.53 |
5 |
β-Elemene |
15 |
0 |
0.6 |
3 |
0 |
0 |
70.42 |
0 |
6 |
MethylChavicol |
11 |
6 |
0.2 |
3 |
1 |
0 |
47.04 |
9.23 |
7 |
Linalool |
11 |
0 |
0.6 |
4 |
1 |
1 |
50.44 |
20.23 |
8 |
δ-Cardinene |
15 |
0 |
0.73 |
1 |
0 |
0 |
69.04 |
0 |
9 |
β-Bisabolene |
15 |
0 |
0.6 |
4 |
0 |
0 |
70.68 |
0 |
10 |
1,8-Cineole |
11 |
0 |
1 |
0 |
1 |
0 |
47.12 |
9.23 |
11 |
Camphor |
11 |
0 |
0.9 |
0 |
1 |
0 |
45.64 |
17.07 |
12 |
Isocaryophyllene |
10 |
0 |
0.8 |
0 |
0 |
0 |
45.22 |
0 |
13 |
Apigenin-7-O-glucuronide |
32 |
16 |
0.24 |
4 |
11 |
6 |
106.72 |
187.12 |
14 |
Carvacrol |
11 |
6 |
0.4 |
1 |
1 |
1 |
48.01 |
20.23 |
15 |
Circimaritin |
23 |
16 |
0.12 |
3 |
6 |
2 |
84.95 |
89.13 |
16 |
Isothymusin |
24 |
16 |
0.12 |
3 |
7 |
3 |
86.97 |
109.36 |
17 |
Pinene |
11 |
0 |
0.82 |
0 |
0 |
0 |
47.66 |
0 |
18 |
Molludistin |
30 |
16 |
0.29 |
3 |
9 |
5 |
105.11 |
149.82 |
19 |
Rosameric acid |
26 |
12 |
0.11 |
7 |
8 |
5 |
91.4 |
144.52 |
20 |
Orientin |
32 |
12 |
0.33 |
3 |
11 |
9 |
107.27 |
200.53 |
21 |
Vicenin |
42 |
16 |
0.44 |
5 |
15 |
11 |
139.23 |
271.2 |
22 |
Urosolic acid |
33 |
0 |
0.9 |
1 |
3 |
2 |
136.91 |
57.53 |
23 |
Luteolin |
21 |
16 |
0 |
1 |
6 |
4 |
76.01 |
111.13 |
24 |
Luteolin-7-O-glucuronide |
33 |
12 |
0.29 |
4 |
12 |
8 |
107.38 |
206.6 |
25 |
3-Carene |
10 |
0 |
0.8 |
0 |
0 |
0 |
45.22 |
0 |
26 |
Citral |
11 |
0 |
0.5 |
4 |
1 |
0 |
49.44 |
17.07 |
27 |
Geraniol |
11 |
0 |
0.6 |
4 |
1 |
1 |
50.4 |
20.23 |
28 |
Methyl Cinnamate |
13 |
6 |
0.18 |
4 |
2 |
0 |
52.24 |
26.3 |
29 |
β-Ocimene |
10 |
0 |
0.4 |
3 |
0 |
0 |
48.76 |
0 |
30 |
λ-Terpineol |
11 |
0 |
0.8 |
1 |
1 |
1 |
48.8 |
20.23 |
31 |
PhenylPropanoids |
12 |
6 |
0.22 |
3 |
3 |
2 |
45.5 |
63.32 |
32 |
Germacrene-D |
15 |
0 |
0.6 |
1 |
0 |
0 |
70.68 |
0 |
33 |
λ-Humulene |
15 |
0 |
0.6 |
0 |
0 |
0 |
70.42 |
0 |
34 |
Camphene |
10 |
0 |
0.8 |
0 |
0 |
0 |
45.22 |
0 |
35 |
Myrcene |
10 |
0 |
0.4 |
4 |
0 |
0 |
48.76 |
0 |
36 |
Thymol |
11 |
6 |
0.4 |
1 |
1 |
1 |
48.01 |
20.23 |
37 |
λ-Linolenic acid |
20 |
0 |
0.61 |
13 |
2 |
1 |
88.99 |
37.3 |
Table 3.
Solubility of the Phytoconstituents of Ocimum sanctum Linn.
Table 3.
Solubility of the Phytoconstituents of Ocimum sanctum Linn.
Table 3.
Lipophilicity of the Phytoconstituents of Ocimum sanctum Linn.
Table 3.
Lipophilicity of the Phytoconstituents of Ocimum sanctum Linn.
Sl.No |
Molecules |
iLOGP |
XLOGP3 |
WLOGP |
MLOGP |
Silicos-IT Log P |
Consensus Log P |
1 |
MethylEugenol |
2.65 |
2.52 |
2.43 |
2.3 |
3 |
2.58 |
2 |
Eugenol |
2.37 |
2.27 |
2.13 |
2.01 |
2.48 |
2.25 |
3 |
δ-Caryophyllene |
3.25 |
4.38 |
4.73 |
4.63 |
4.19 |
4.24 |
4 |
Caryophyllene oxide |
3.53 |
4.91 |
4.94 |
4.31 |
5.15 |
4.57 |
5 |
β-Elemene |
3.37 |
6.11 |
4.75 |
4.53 |
4.5 |
4.65 |
6 |
MethylChavicol |
2.47 |
3.37 |
2.42 |
2.67 |
2.96 |
2.78 |
7 |
Linalool |
2.7 |
2.97 |
2.67 |
2.59 |
2.35 |
2.66 |
8 |
δ-Cardinene |
3.41 |
3.8 |
4.73 |
4.63 |
4.12 |
4.14 |
9 |
β-Bisabolene |
3.67 |
6.43 |
5.04 |
4.53 |
4.5 |
4.83 |
10 |
1,8-Cineole |
2.58 |
2.74 |
2.74 |
2.45 |
2.86 |
2.67 |
11 |
Camphor |
2.12 |
2.19 |
2.4 |
2.3 |
2.85 |
2.37 |
12 |
Isocaryophyllene |
2.63 |
4.38 |
3 |
4.29 |
2.79 |
3.42 |
13 |
Apigenin-7-O-glucuronide |
1 |
1.46 |
0.14 |
-1.63 |
-0.1 |
0.17 |
14 |
Carvacrol |
2.24 |
3.49 |
2.82 |
2.76 |
2.79 |
2.82 |
15 |
Circimaritin |
2.56 |
3.32 |
2.89 |
0.47 |
3.07 |
2.46 |
16 |
Isothymusin |
2.58 |
2.61 |
2.59 |
-0.07 |
2.59 |
2.06 |
17 |
Pinene |
2.66 |
2.74 |
3 |
4.58 |
3.06 |
3.21 |
18 |
Molludistin |
2.21 |
0.6 |
0.71 |
-1.25 |
1.37 |
0.73 |
19 |
Rosameric acid |
1.48 |
2.36 |
1.65 |
0.9 |
1.5 |
1.58 |
20 |
Orientin |
1.02 |
-1.03 |
-1.16 |
-2.32 |
-1.21 |
-0.94 |
21 |
Vicenin |
1.73 |
-2.26 |
-3.04 |
-4.51 |
-1.8 |
-1.98 |
22 |
Urosolic acid |
3.95 |
7.34 |
7.09 |
5.82 |
5.46 |
5.93 |
23 |
Luteolin |
1.86 |
2.53 |
2.28 |
-0.03 |
2.03 |
1.73 |
24 |
Luteolin-7-O-glucuronide |
1.79 |
-0.13 |
-0.78 |
-1.94 |
-1.64 |
-0.54 |
25 |
3-Carene |
2.63 |
4.38 |
3 |
4.29 |
2.79 |
3.42 |
26 |
Citral |
2.47 |
3.03 |
2.88 |
2.49 |
2.65 |
2.71 |
27 |
Geraniol |
2.52 |
3.56 |
2.67 |
2.59 |
2.35 |
2.74 |
28 |
Methyl Cinnamate |
2.17 |
2.25 |
2.15 |
2.49 |
2.57 |
2.33 |
29 |
β-Ocimene |
2.91 |
4.26 |
3.48 |
3.56 |
2.88 |
3.42 |
30 |
λ-Terpineol |
2.51 |
3.39 |
2.5 |
2.3 |
2.17 |
2.58 |
31 |
PhenylPropanoids |
1.08 |
-1.52 |
0.64 |
-1.11 |
0.86 |
-0.01 |
32 |
Germacrene-D |
3.14 |
4.74 |
4.89 |
4.53 |
4.01 |
4.26 |
33 |
λ-Humulene |
3.29 |
4.55 |
5.04 |
4.53 |
3.91 |
4.26 |
34 |
Camphene |
2.58 |
4.22 |
3 |
4.29 |
3.08 |
3.43 |
35 |
Myrcene |
2.89 |
4.17 |
3.48 |
3.56 |
3.05 |
3.43 |
36 |
Thymol |
2.32 |
3.3 |
2.82 |
2.76 |
2.79 |
2.8 |
37 |
λ-Linolenic acid |
2.32 |
3.3 |
5.66 |
2.76 |
2.79 |
2.8 |
Table 4.
Drug likeness of the Phytoconstituents of Ocimum sanctum Linn.
Table 4.
Drug likeness of the Phytoconstituents of Ocimum sanctum Linn.
Sl.No |
Molecules |
Lipinski violations |
Ghose violations |
Veber violations |
Egan violations |
Muegge violations |
Bioavailability Score |
1 |
MethylEugenol |
0 |
0 |
0 |
0 |
1 |
0.55 |
2 |
Eugenol |
0 |
0 |
0 |
0 |
1 |
0.55 |
3 |
δ-Caryophyllene |
1 |
0 |
0 |
0 |
1 |
0.55 |
4 |
Caryophyllene oxide |
1 |
0 |
0 |
0 |
1 |
0.55 |
5 |
β-Elemene |
1 |
0 |
0 |
0 |
2 |
0.55 |
6 |
MethylChavicol |
0 |
1 |
0 |
0 |
2 |
0.55 |
7 |
Linalool |
0 |
1 |
0 |
0 |
2 |
0.55 |
8 |
δ-Cardinene |
1 |
0 |
0 |
0 |
1 |
0.55 |
9 |
β-Bisabolene |
1 |
0 |
0 |
0 |
2 |
0.55 |
10 |
1,8-Cineole |
0 |
1 |
0 |
0 |
2 |
0.55 |
11 |
Camphor |
0 |
1 |
0 |
0 |
2 |
0.55 |
12 |
Isocaryophyllene |
1 |
1 |
0 |
0 |
2 |
0.55 |
13 |
Apigenin-7-O-glucuronide |
2 |
0 |
1 |
1 |
3 |
0.11 |
14 |
Carvacrol |
0 |
1 |
0 |
0 |
2 |
0.55 |
15 |
Circimaritin |
0 |
0 |
0 |
0 |
0 |
0.55 |
16 |
Isothymusin |
0 |
0 |
0 |
0 |
0 |
0.55 |
17 |
Pinene |
1 |
1 |
0 |
0 |
2 |
0.55 |
18 |
Molludistin |
0 |
0 |
1 |
1 |
0 |
0.55 |
19 |
Rosameric acid |
0 |
0 |
1 |
1 |
0 |
0.56 |
20 |
Orientin |
2 |
1 |
1 |
1 |
3 |
0.17 |
21 |
Vicenin |
3 |
4 |
1 |
1 |
4 |
0.17 |
22 |
Urosolic acid |
1 |
3 |
0 |
1 |
1 |
0.85 |
23 |
Luteolin |
0 |
0 |
0 |
0 |
0 |
0.55 |
24 |
Luteolin-7-O-glucuronide |
2 |
1 |
1 |
1 |
3 |
0.11 |
25 |
3-Carene |
1 |
1 |
0 |
0 |
2 |
0.55 |
26 |
Citral |
0 |
1 |
0 |
0 |
2 |
0.55 |
27 |
Geraniol |
0 |
1 |
0 |
0 |
2 |
0.55 |
28 |
Methyl Cinnamate |
0 |
0 |
0 |
0 |
1 |
0.55 |
29 |
β-Ocimene |
0 |
1 |
0 |
0 |
2 |
0.55 |
30 |
λ-Terpineol |
0 |
1 |
0 |
0 |
2 |
0.55 |
31 |
PhenylPropanoids |
0 |
0 |
0 |
0 |
1 |
0.55 |
32 |
Germacrene-D |
1 |
0 |
0 |
0 |
1 |
0.55 |
33 |
λ-Humulene |
1 |
0 |
0 |
0 |
1 |
0.55 |
34 |
Camphene |
1 |
1 |
0 |
0 |
2 |
0.55 |
35 |
Myrcene |
0 |
1 |
0 |
0 |
2 |
0.55 |
36 |
Thymol |
0 |
1 |
0 |
0 |
2 |
0.55 |
37 |
λ-Linolenic acid |
0 |
1 |
0 |
0 |
2 |
0.55 |
Table 5.
Medicinal Chemistry Properties of Phytoconstituents of Ocimum sanctum Linn.
Table 5.
Medicinal Chemistry Properties of Phytoconstituents of Ocimum sanctum Linn.
Sl.No |
Molecules |
PAINS |
Brenk |
Leadlikeness |
Synthetic Accessibility |
1 |
MethylEugenol |
0 |
1 |
1 |
1.71 |
2 |
Eugenol |
0 |
1 |
1 |
1.58 |
3 |
δ-Caryophyllene |
0 |
1 |
2 |
4.51 |
4 |
Caryophyllene oxide |
0 |
1 |
1 |
4.46 |
5 |
β-Elemene |
0 |
1 |
2 |
3.63 |
6 |
MethylChavicol |
0 |
1 |
1 |
1.28 |
7 |
Linalool |
0 |
1 |
1 |
2.74 |
8 |
δ-Cardinene |
0 |
1 |
2 |
4.14 |
9 |
β-Bisabolene |
0 |
1 |
2 |
3.9 |
10 |
1,8-Cineole |
0 |
0 |
1 |
3.65 |
11 |
Camphor |
0 |
0 |
1 |
3.22 |
12 |
Isocaryophyllene |
0 |
1 |
2 |
3.84 |
13 |
Apigenin-7-O-glucuronide |
0 |
0 |
1 |
5.06 |
14 |
Carvacrol |
0 |
0 |
1 |
1 |
15 |
Circimaritin |
0 |
0 |
0 |
3.27 |
16 |
Isothymusin |
0 |
1 |
0 |
3.38 |
17 |
Pinene |
0 |
1 |
1 |
4.81 |
18 |
Molludistin |
0 |
0 |
1 |
4.91 |
19 |
Rosameric acid |
1 |
2 |
1 |
3.38 |
20 |
Orientin |
1 |
1 |
1 |
5.34 |
21 |
Vicenin |
0 |
0 |
1 |
6.4 |
22 |
Urosolic acid |
0 |
1 |
2 |
6.21 |
23 |
Luteolin |
1 |
1 |
0 |
3.02 |
24 |
Luteolin-7-O-glucuronide |
1 |
1 |
1 |
5.32 |
25 |
3-Carene |
0 |
1 |
2 |
3.84 |
26 |
Citral |
0 |
3 |
1 |
2.49 |
27 |
Geraniol |
0 |
1 |
2 |
2.58 |
28 |
Methyl Cinnamate |
0 |
0 |
1 |
1.98 |
29 |
β-Ocimene |
0 |
2 |
2 |
3.63 |
30 |
λ-Terpineol |
0 |
1 |
1 |
3.24 |
31 |
PhenylPropanoids |
0 |
0 |
1 |
1.46 |
32 |
Germacrene-D |
0 |
1 |
2 |
4.55 |
33 |
λ-Humulene |
0 |
1 |
2 |
3.66 |
34 |
Camphene |
0 |
1 |
2 |
3.5 |
35 |
Myrcene |
0 |
2 |
2 |
2.85 |
36 |
Thymol |
0 |
0 |
1 |
1 |
37 |
λ-Linolenic acid |
0 |
0 |
1 |
1 |
Table 6.
Pharmacokinetic Parameters of the Phytoconstituents of Ocimum sanctum.
Table 6.
Pharmacokinetic Parameters of the Phytoconstituents of Ocimum sanctum.
Sl.No |
Molecules |
GI absorption |
BBB permeant |
P-gp substrate |
CYP1A2 inhibitor |
CYP2C19 inhibitor |
CYP2C9 inhibitor |
CYP2D6 inhibitor |
CYP3A4 inhibitor |
log Kp (cm/s) |
1 |
MethylEugenol |
High |
Yes |
No |
Yes |
No |
No |
No |
No |
-5.6 |
2 |
Eugenol |
High |
Yes |
No |
Yes |
No |
No |
No |
No |
-5.69 |
3 |
δ-Caryophyllene |
Low |
No |
No |
No |
Yes |
Yes |
No |
No |
-4.44 |
4 |
Caryophyllene oxide |
High |
Yes |
No |
No |
No |
Yes |
No |
No |
-4.34 |
5 |
β-Elemene |
Low |
No |
No |
No |
Yes |
Yes |
No |
No |
-3.21 |
6 |
MethylChavicol |
High |
Yes |
No |
Yes |
No |
No |
No |
No |
-4.81 |
7 |
Linalool |
High |
Yes |
No |
No |
No |
No |
No |
No |
-5.13 |
8 |
δ-Cardinene |
Low |
No |
No |
No |
Yes |
Yes |
No |
No |
-4.85 |
9 |
β-Bisabolene |
Low |
No |
No |
No |
No |
Yes |
No |
No |
-2.98 |
10 |
1,8-Cineole |
High |
Yes |
No |
No |
No |
No |
No |
No |
-5.3 |
11 |
Camphor |
High |
Yes |
No |
No |
No |
No |
No |
No |
-5.67 |
12 |
Isocaryophyllene |
Low |
Yes |
No |
No |
No |
Yes |
No |
No |
-4.02 |
13 |
Apigenin-7-O-glucuronide |
Low |
No |
Yes |
No |
No |
No |
No |
No |
-7.99 |
14 |
Carvacrol |
High |
Yes |
No |
Yes |
No |
No |
No |
No |
-4.74 |
15 |
Circimaritin |
High |
No |
No |
Yes |
No |
Yes |
Yes |
Yes |
-5.86 |
16 |
Isothymusin |
High |
No |
No |
Yes |
No |
Yes |
Yes |
Yes |
-6.46 |
17 |
Pinene |
Low |
Yes |
No |
No |
No |
Yes |
No |
No |
-5.26 |
18 |
Molludistin |
Low |
No |
No |
No |
No |
No |
No |
No |
-8.41 |
19 |
Rosameric acid |
Low |
No |
No |
No |
No |
No |
No |
No |
-6.82 |
20 |
Orientin |
Low |
No |
No |
No |
No |
No |
No |
No |
-9.78 |
21 |
Vicenin |
Low |
No |
Yes |
No |
No |
No |
No |
No |
-11.53 |
22 |
Urosolic acid |
Low |
No |
No |
No |
No |
No |
No |
No |
-3.87 |
23 |
Luteolin |
High |
No |
No |
Yes |
No |
No |
Yes |
Yes |
-6.25 |
24 |
Luteolin-7-O-glucuronide |
Low |
No |
Yes |
No |
No |
No |
No |
No |
-9.22 |
25 |
3-Carene |
Low |
Yes |
No |
No |
No |
Yes |
No |
No |
-4.02 |
26 |
Citral |
High |
Yes |
No |
No |
No |
No |
No |
No |
-5.08 |
27 |
Geraniol |
High |
Yes |
No |
No |
No |
No |
No |
No |
-4.71 |
28 |
Methyl Cinnamate |
High |
Yes |
No |
No |
No |
No |
No |
No |
-5.78 |
29 |
β-Ocimene |
Low |
Yes |
No |
No |
No |
No |
No |
No |
-4.11 |
30 |
λ-Terpineol |
High |
Yes |
No |
No |
No |
No |
No |
No |
-4.83 |
31 |
PhenylPropanoids |
High |
No |
No |
No |
No |
No |
No |
No |
-8.39 |
32 |
Germacrene-D |
Low |
No |
No |
No |
No |
Yes |
No |
No |
-4.18 |
33 |
λ-Humulene |
Low |
No |
No |
No |
No |
Yes |
No |
No |
-4.32 |
34 |
Camphene |
Low |
Yes |
No |
No |
No |
Yes |
No |
No |
-4.13 |
35 |
Myrcene |
Low |
Yes |
No |
No |
No |
No |
No |
No |
-4.17 |
36 |
Thymol |
High |
Yes |
No |
Yes |
No |
No |
No |
No |
-4.87 |
37 |
λ-Linolenic acid |
High |
Yes |
No |
Yes |
No |
No |
No |
No |
-4.87 |
Figure 1.
Boiled Egg Model of the Phytoconstituents of Ocimumsanctum Linn.
Figure 1.
Boiled Egg Model of the Phytoconstituents of Ocimumsanctum Linn.
4. Discussion
For the purpose of the current study, we evaluated the ADME properties of Ocimum sanctum Linn using the free online SwissADME software application. Through the software, the phytoconstituents of the plants were identified, they are Methyl Eugenol, Eugenol, β-Caryophyllene, Caryophyllene oxide, β-Elemene, Methyl Chavicol, Linalool, δ-Cadinene, β-Bisabolene, 1,8-Cineole, Camphor, Isocaryophyllene, Apigenin-7-O-glucuronide, Carvacrol, Circimaritin, Isothymusin, Pinene, Molludistin, Rosameric acid, Orientin, Vicenin, Urosolic acid, Luteolin, Luteolin-7-O-glucuronide, 3-Carene, Citral, Geraniol, Methyl Cinnamate, β-Ocimene, α-Terpineol, PhenylPropanoids, Germacrene-D, α-Humulene, Camphene, Myrcene, Thymol, α-Linolenic acid (Roshan Kumar
et al.,2022 & Bhattacharya AK
et al.,1996 & Kothari SK
et al.,2005 & Awasthi PK
et al.,2007 & Kicel A
et al.,2005 & Khan A
et al.,2010 & Machado MIL
et al.,1999 & Pino JA
et al.,1988 & Brophy JJ
et al.,1993 & Kashyap C
et al.,2011)[
39,
40,
41,
42,
43,
44,
45,
46,
47,
48].
As a result, the phytoconstituents ADME characteristics were examined, and the results were presented in appropriate tables and figures. Researchers and scientists can also utilize the values as monographs to generate future research semi- and synthetic pharmaceuticals for a wide range of applications.
5. Conclusion
CADD (Computer Aided Drug Design) has significantly changed research and development routes in drug concept exploration as an outcome of rapid growth in biological and chemical information. In terms of application, time, and cost, the use of computational techniques in the drug discovery and development process is often acclaimed. Modern chemistry relies extensively on computers, which are essential for both drug discovery and development. CADD is used in the pharmaceutical industry to develop and improve novel, safe, and effective pharmaceuticals. In these studies, a free web-based tool called SwissADME is presented to evaluate the ADME characteristics of Phytoconstituents found in the Ocimum sanctum Linn plant. This information could be used as a starting point for a more comprehensive evaluation of the plant's biological and pharmacological properties.
References
- Pattanayak, P.; Behera, P.; Das, D.; Panda, S.K. Ocimum sanctum Linn. A reservoir plant for therapeutic applications: An overview. Pharmacogn. Rev. 2010, 4, 95–105. [Google Scholar] [CrossRef] [PubMed]
- Bast, F.; Rani, P.; Meena, D. Chloroplast DNA Phylogeography of Holy Basil (Ocimum tenuiflorum) in Indian Subcontinent. Sci. World J. 2014, 2014, 847482. [Google Scholar] [CrossRef] [PubMed]
- Joshi, R.K.; Setzer, W.N.; da Silva, J.K. Phytoconstituents, traditional medicinal uses and bioactivities of Tulsi (Ocimum sanctum Linn. ): A review. American Journal of Essential Oils and Natural Products 2017, 5, 18–21. [Google Scholar]
- Prakash, P.; Gupta, N. Therapeutic uses of Ocimum sanctum Linn (Tulsi) with a note on eugenol and its pharmacological actions: a short review. Indian J. Physiol. Pharmacol. 2005, 49, 125–131. [Google Scholar] [PubMed]
- Viyoch, J.; Pisutthanan, N.; Faikreua, A.; Nupangta, K.; Wangtorpol, K.; Ngokkuen, J. Evaluation of in vitro antimicrobial activity of Thai basil oils and their micro-emulsion formulas against Propionibacterium acnes. Int. J. Cosmet. Sci. 2006, 28, 125–133. [Google Scholar] [CrossRef] [PubMed]
- Magesh, V.; Lee, J.C.; Ahn, K.S.; Lee, H.J.; Lee, E.O.; Shim, B.S.; et al. Ocimum sanctum induces apoptosis in A549 lung cancer cells and suppresses the in vivo growth of Lewis lung carcinoma cells. Phytother Res 2009, 23, 1385–1391. [Google Scholar] [CrossRef] [PubMed]
- Kumar, V.; Andola, H.C.; Lohani, H.; Chauhan, N. Pharmacological Review on Ocimum sanctum Linnaeus: A Queen of herbs. Journal of Pharmacy Research 2011, 4, 366–368. [Google Scholar]
- Kumar, A.; Rahal, A.; Chakraborty, S.; et al. Ocimum sanctum (Tulsi): a miracle herb and boon to medical science – A Review. International Journal of Agronomy and Plant Production 2013, 4, 1580–1589. [Google Scholar]
- Mahajan, N.; Rawal, S.; et al. A phytopharmacological overview on Ocimum species with special emphasis on Ocimum sanctum. Biomedicine & Preventive Nutrition 2013, 3, 185–192. [Google Scholar]
- Lachman LH, Lieberman, Kanig JL. The Theory and Practice of Industrial Pharmacy, Lea & Febiger, 3rd edition, 1986.
- Yalkowsky, S.H.; Valvani, S.C. Solubility and partitioning I: Solubility of nonelectrolytes in water. J. Pharm. Sci. 1980, 69, 912–922. [Google Scholar] [CrossRef] [PubMed]
- Kah, M.; Brown, C.D. LogD: Lipophilicity for ionisable compounds. Chemosphere 2008, 72, 1401–1408. [Google Scholar] [CrossRef]
- Constantinescu, T.; Lungu, C.N.; Lung, I. Lipophilicity as a Central Component of Drug-Like Properties of Chalchones and Flavonoid Derivatives. Molecules 2019, 24, 1505. [Google Scholar] [CrossRef]
- Giaginis, C.; Tsopelas, F.; Tsantili-Kakoulidou, A. The Impact of Lipophilicity in Drug Discovery: Rapid Measurements by Means of Reversed-Phase HPLC. 2018, 1824, 217–228. [Google Scholar] [CrossRef] [PubMed]
- Kamel, M.S.; Belal, A.; Aboelez, M.O.; Shokr, E.K.; Abdel-Ghany, H.; Mansour, H.S.; Shawky, A.M.; El-Remaily, M.A.E.A.A.A. Microwave-Assisted Synthesis, Biological Activity Evaluation, Molecular Docking, and ADMET Studies of Some Novel Pyrrolo [2,3-b] Pyrrole Derivatives. Molecules 2022, 27, 2061. [Google Scholar] [CrossRef] [PubMed]
- Erckes, V.; Steuer, C. A story of peptides, lipophilicity and chromatography—Back and forth in time. RSC Med. Chem. 2022, 22, 676–687. [Google Scholar] [CrossRef]
- Ginex, T.; Vazquez, J.; Gilbert, E.; Herrero, E.; Luque, F.J. Lipophilicity in drug design: an overview of lipophilicity descriptors in 3D-QSAR studies. Futur. Med. Chem. 2019, 11, 1177–1193. [Google Scholar] [CrossRef] [PubMed]
- Kempińska, D.; Chmiel, T.; Kot-Wasik, A.; Mróz, A.; Mazerska, Z.; Namieśnik, J. State of the art and prospects of methods for determination of lipophilicity of chemical compounds. TrAC Trends Anal. Chem. 2019, 113, 54–73. [Google Scholar] [CrossRef]
- Dołowy, M.; Jampilek, J.; Bober-Majnusz, K. A Comparative Study of the Lipophilicity of Metformin and Phenformin. Molecules 2021, 26, 6613. [Google Scholar] [CrossRef] [PubMed]
- Cheng, T.; et al. Computation of Octanol Water Partition Coefficients by Guiding an Additive Model with Knowledge. J Chem Inf. Model. 2007, 47, 2140–2148. [Google Scholar] [CrossRef] [PubMed]
- Wildman, S.A.; Crippen, G.M. Prediction of Physicochemical Parameters by Atomic Contributions. J. Chem. Inf. Comput. Sci. 1999, 39, 868–873. [Google Scholar] [CrossRef]
- Moriguchi, I.; Hirono, S.; Nakagome, I.; Hirano, H. Comparison of Reliability of log P Values for Drugs Calculated by Several Methods. Chem. Pharm. Bull. 1994, 42, 976–978. [Google Scholar] [CrossRef]
- Antoine, D.; Michielin, O.; Zoete, V. SwissADME: a free web tool to evaluate pharmacokinetics, drug likeness and medicinal chemistry friendliness of small molecules. Nature - Scientific Reports 2017, 7, 42717.1–13. [Google Scholar]
- Egan, W.J.; Merz, K.M.; Baldwin, J.J. Prediction of Drug Absorption Using Multivariate Statistics. J. Med. Chem. 2000, 43, 3867–3877. [Google Scholar] [CrossRef] [PubMed]
- Ghose, A.K.; Viswanadhan, V.N.; Wendoloski, J.J. Prediction of Hydrophobic (Lipophilic) Properties of Small Organic Molecules Using Fragmental Methods: An Analysis of ALOGP and CLOGP Methods. J. Phys. Chem. A 1998, 102, 3762–3772. [Google Scholar] [CrossRef]
- Veber, D.F.; Johnson, S.R.; Cheng, H.Y.; Smith, B.R.; Ward, K.W.; Kopple, K.D.; et al. Molecular properties that influence the oral bioavailability of drug candidates. J Med Chem. 2002, 45, 2615–2623. [Google Scholar] [CrossRef] [PubMed]
- 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 Deliv. Rev. 1997, 23, 3–25. [Google Scholar] [CrossRef]
- Martin, Y.C. A Bioavailability Score. J. Med. Chem. 2005, 48, 3164–3170. [Google Scholar] [CrossRef] [PubMed]
- Baell, J.B.; Holloway, G.A. New Substructure Filters for Removal of Pan Assay Interference Compounds (PAINS) from Screening Libraries and for Their Exclusion in Bioassays. J. Med. Chem. 2010, 53, 2719–2740. [Google Scholar] [CrossRef] [PubMed]
- Teague, S.; Davis, A.; Leeson, P.; Oprea, T. The Design of Leadlike Combinatorial Libraries. Angew. Chem. Int. Ed. Engl. 1999, 38, 3743–3748. [Google Scholar] [CrossRef]
- Hann, M.M.; Keserü, G.M. Finding the sweet spot: the role of nature and nurture in medicinal chemistry. Nat. Rev. Drug Discov. 2012, 11, 355–365. [Google Scholar] [CrossRef] [PubMed]
- Brito Sanchez, Y.; Marrero-Ponce, Y.; Barigye, S.J.; Yaber Goenaga, I.; Morell Prez, C.; et al. Mol. Inf. 2015, 34, 308–330. [Google Scholar]
- Di, L.P.; Artursson, A.; Avdeef, G.F.; Ecker, B.; Faller, H.; Fischer, J.B.; et al. Drug Discov. Today 2012, 17, 905–912. [Google Scholar]
- Montanari, F.; Ecker, G. F. Prediction of drug-ABC-transporter interaction–Recent advances and future challenges. Adv. Drug Deliv. Rev. 2015, 86, 17–26. [Google Scholar] [CrossRef] [PubMed]
- Testa, B.; Kraemer, S. D. The Biochemistry of Drug Metabolism – An Introduction - Testa - 2007 - Chemistry & Biodiversity - Wiley Online Library. Chem. Biodivers ( 2007.
- Di, L. The role of drug metabolizing enzymes in clearance. Expert Opin. Drug Metab. Toxicol. 2014, 10, 379–393. [Google Scholar] [CrossRef] [PubMed]
- Cortes, C.; Vapnik, V. Support-vector networks. Mach. Learn. 1995, 20, 273–297. [Google Scholar] [CrossRef]
- Mishra, N.K.; Agarwal, S.; Raghava, G.P. Prediction of cytochrome P450 isoform responsible for metabolizing a drug molecule. BMC Pharmacol. 2010, 10, 8. [Google Scholar] [CrossRef] [PubMed]
- Kumar, R.; et al. A Systemic Review of Ocimum sanctum(Tulsi): Morphological Characteristics, Phytoconstituents and Therapeutic Applications. International Journal for Research in Applied Sciences and Biotechnology, Volume-9, Issue-2(March2022).
- Bhattacharya, A.K.; Kaul, P.N.; Rajeswara Rao, B.R. Essentialoils of Ocimum gratissimum L. and Ocimum tenuiflorum L. (Syn. Ocimum sanctum L.) grown in Andhra Pradesh. Indian Perfumer. 1996, 40, 73–75. [Google Scholar]
- Kothari SK, Bhattacharya AK, Ramesh S, Garg SN,Khanuja SPS. Volatile constituents in oil from different plant parts of methyl eugenol-rich Ocimum tenuiflorum L. f. (Syn. O. sanctum L.) grown in South India. Journal of Essential Oil Res. 2005, 17, 656–658.
- Awasthi, P.; Dixit, S. Chemical Compositions ofOcimum sanctumShyama and0cimum sanctumRama Oils from the Plains of Northern India. J. Essent. Oil Bear. Plants 2007, 10, 292–296. [Google Scholar] [CrossRef]
- Kicel, A.; Kurowska, A.; Kalemba, D. Composition of the essential oil of Ocimum sanctum L. grown in Poland during vegetation. Journal of Essential Oil Res. 2005, 17, 217–219. [Google Scholar] [CrossRef]
- Khan, A.; Ahmad, A.; Akhtar, F.; Yousuf, S.; Xess, I.; Khan, L.A.; Manzoor, N. Ocimum sanctum essential oil and its active principles exert their antifungal activity by disrupting ergosterol biosynthesis and membrane integrity. Res. Microbiol. 2010, 161, 816–823. [Google Scholar] [CrossRef] [PubMed]
- Machado, M.I.L.; Silva, M.G.V.; Matos, F.J.A.; Craveiro, A.A.; Alencar, J.W. Volatile constituents from leaves and inflorescence oil of Ocimum tenuiflorum L. f. (Syn. O.sanctum L.) grown in Northeastern Brazil. Journal of Essential Oil Res. 1999, 11, 324–326. [Google Scholar] [CrossRef]
- Pino, J.A.; Rosado, A.; Rodriguez, M.; Garcia, D. Composition of the essential oil of Ocimum tenuiflorum L. grown in Cuba. Journal of Essential Oil Res. 1988, 10, 437–438. [Google Scholar] [CrossRef]
- Brophy, J.J.; Goldsack, R.J.; Clarkson, J.R. The essential oil of Ocimum tenuiflorum L. (Lamiaceae) growing in Northern Australia. Journal of Essential Oil Res. 1993, 5, 459–461. [Google Scholar] [CrossRef]
- Kashyap, C.; Ranjeet, K.; Vikrant, A.; Vipin, K. Therapeutic Potency of Ocimum Kilimandscharicum Guerke-A Review. Global Journal of Pharmacology 2011, 5, 191–200. [Google Scholar]
|
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).