Research Article | | Peer-Reviewed

Computer-aided Design of Coumarin Inhibitors of Quinone Oxidoreductase-1 (NQO1) with a Favorable Pharmacokinetic Profile

Received: 30 June 2025     Accepted: 14 July 2025     Published: 4 August 2025
Views:       Downloads:
Abstract

Over the last ten years, cancer therapies have struggled with drug resistance. In this report, we explore new coumarin (COU) compounds designed to inhibit the enzyme NQO1, which shows potential for effective treatment due to their favorable predicted drug properties. Three-dimensional (3D) models of NQO1-COUx complexes were generated through in situ modifications of the crystal structure of NQO1-COU12 (PDB entry code: 3JSX), which served as the reference compound for a training set of of 22 and a validation set of 6 VCOUs with known experimental inhibitory potencies. To identify the active conformation of COU1-22, we developed a gas-phase quantitative structure-activity relationship (QSAR) model that established a linear correlation between the calculated enthalpy of NQO1-COU complex formation and the values of experimental activities. Subsequently, we screened the Virtual Compound Library (VCL) using Lipinski's Rule of Five and the PH4 model, then assessed the potency of the new COU analogues using the retained QSAR model. The pharmacokinetic profile of the analogues obtained was also evaluated using the linear correlation equation derived from the QSAR model. The coefficient of determination (R²), the Leave One Out (LOO) cross-validated Squared and the Standard error of regression σ for this equation are 0.91, 0.94 and 0.14, respectively, thus revealing the high predictive power of this model. Similarly, the PH4 model, with a correlation coefficient of 0.91, demonstrated robust predictive power. A comprehensive screening of the COU virtual analogue library yielded a total of 63 drug candidates with oral bioavailability, among which the most promising compounds exhibited a predicted potency of 12.22 and a favorable pharmacokinetic profile. The integration of Quantitative Structure-Activity Relationship (QSAR) techniques and in silico screening, based on the PH4 model, has enabled us to propose potent anticancer candidates with optimal pharmacokinetic profiles.

Published in Science Journal of Chemistry (Volume 13, Issue 4)
DOI 10.11648/j.sjc.20251304.12
Page(s) 102-121
Creative Commons

This is an Open Access article, distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution and reproduction in any medium or format, provided the original work is properly cited.

Copyright

Copyright © The Author(s), 2025. Published by Science Publishing Group

Keywords

Coumarins, NQO1, Drug Resistance, Anticancer Therapy, Virtual Screening, Pharmacokinetics

1. Introduction
NAD(P)H Quinone Oxidoreductase-1 (NQO1) is an important enzyme involved in cellular redox homeostasis, playing a dual role in both detoxification of quinones and activation of certain anticancer drugs . However, its overexpression in various cancers, including pancreatic and colorectal carcinomas, has made it a potential therapeutic target . Inhibiting NQO1 could disrupt tumor cell metabolism and sensitize them to oxidative stress, paving the way for novel anticancer strategies .
Coumarin-based molecules, known for their bioactive properties, have shown promising inhibitory activity against NQO1 . However, the development of selective and potent inhibitors requires a rational design approach to optimize binding affinity, pharmacokinetics, and toxicity profiles . In this context, computer-aided drug design (CADD) plays a crucial role in accelerating the identification and optimization of lead compounds .
This study explores the computer-aided design of coumarin-based NQO1 inhibitors, integrating molecular docking, quantitative structure-activity relationship (QSAR) models, and molecular dynamics simulations . In silico methods enable the identification of the most promising candidate compounds prior to synthesis, thereby minimizing experimental costs and significantly improving the efficiency of the drug discovery process . Ultimately, this research aims to develop optimized therapeutic candidates with improved selectivity and bioavailability, offering new perspectives for cancer treatment .
Structural studies have shown that NQO1 exists as a homodimer, with each subunit containing a molecule of flavin adenine dinucleotide (FAD) that is non-covalently bound to the protein . The FAD is positioned such that its isoalloxazine ring forms the floor of the active site cavity. The two active sites are located at opposite ends of the dimer and contain residues from both monomers . The FAD pocket, a buried cavity, is surrounded by the residues Pro68, Trp105, Phe106, Tyr126, Tyr128 and Phe178. The more exposed access pocket, bounded by Tyr128, Met131, Gly149, Gly150, Met154, His161, His194, Phe232 and Phe236 . The catalytic cycle of NQO1 operates through a "ping-pong" mechanism, which occurs in two distinct steps: the transfer of hydride from NAD(P)H to the FAD cofactor, followed by the release of NAD(P)+ and the transfer of hydride from the reduced cofactor to the substrate. Compound 1 is a potent competitive inhibitor of NQO1, competing with NAD(P)H for binding to NQO1, thereby preventing electron transfer to FAD . Compound 16 (old identification) or COU12 is frequently used to study the functional consequences of NQO1 inactivity in cells, and recently, much of the research involving 16 has focused on its ability to suppress the malignant phenotype of pancreatic cancer cells both in vitro and in vivo . NQO1 expression is upregulated in pancreatic cancer, and the underlying mechanism by which 16 acts is believed to interfere with NQO1’s ability to protect cells against oxidative damage . However, it is known that 16 has a variety of other biochemical effects, such as mitochondrial uncoupling, which may complicate the interpretation of NQO1's cellular properties .
Starting with the in-situ modification of the crystal structure of the NQO1-COU12 complex (PDB: 3JSX), we developed a quantitative structure-activity relationship (QSAR) model that correlated the Gibbs free energies of NQO1-COUx complex formation with the PIC50exp determined from experimental IC50exp values.
This allowed us to identify the active conformation of COUs at the active site of NQO1 from cancer using a molecular mechanics-Poisson-Boltzmann (MM-PB) complexation approach. Utilizing this active conformation, we created a 3D QSAR pharmacophore for NQO1 inhibition (designated PH4). A large virtual library of compounds sharing the COU scaffold was generated and screened in silico against the PH4 pharmacophore. The screening results revealed virtual hits with predicted inhibitory potencies (pIC50pre) larger or IC50pre lower than that of the most potent training compound, COU1. Subsequently, these hits underwent complexation simulations to evaluate their predicted inhibitory activities, and their ADMET profiles were calculated for further assessment.
2. Methods
Training and validation sets
The training and validation set for the coumarin analog inhibitors of human NQO1 used in this study were obtained from the literature . The experimental values (IC50exp) exhibit a broad range, spanning from 0.18 to 1221 nM, covering over four orders of magnitude. This wide distribution makes them well-suited for developing a robust QSAR model. Among the 28 compounds considered, 22 were allocated to the training set (TS), while 6 compounds were selected for the validation set (VS).
Model building
Three-dimensional (3D) models of the free inhibitors (I), the free NQO1 enzyme (E), and the enzyme-inhibitor complexes (E) were created based on the high-resolution crystal structure (2.45 Å) of a reference complex with the inhibitor COU12 (PDB code: 3JSX) . These models were generated using the graphical interfaces of the molecular modeling tools Insight-II and Discovery Studio 2.5 .
Molecular mechanics
The modeling of the COU and the PL ligand complexes was carried out using molecular mechanics, applying the CFF force field , as outlined in previous studies . This process involved optimizing the geometry of the ligand-enzyme interactions by minimizing energy, allowing for a more accurate representation of the binding and structural characteristics of the complexes.
Conformational research
The conformations of the free inhibitors were derived from their bound conformations within the protein-ligand (PL) complexes. This was realized through the implementation of a stepwise relaxation process, thereby enabling the structures to undergo gradual adjustment until they attained the nearest local energy minimum, in accordance with the procedure delineated in preceding studies .
Gibbs Free Energies Solvation
Ligand-receptor interactions take place in a solvent environment, which affects the binding process through hydrogen bonding and solvation effects. To account for these influences, the electrostatic component of the Gibbs free energy (GFE) was calculated by solving the nonlinear Poisson-Boltzmann equation , incorporating ionic strength. This computation was performed using the Delphi module implemented in Discovery Studio 2.5 , according with the approach detailed in previous studies .
Interaction energy
The interaction energy (Eint) between the enzyme residues and the inhibitor was determined using the CFF force field, following the methodology previously reported .
Generation of pharmacophores
The 3D-QSAR (PH4) pharmacophore generation protocol in Discovery Studio , based on the Catalyst HypoGen algorithm , was used to develop the PH4 model for NQO1 inhibition, following the approach previously described .
ADME properties
The pharmacokinetic profile of COUs was assessed using the QikProp program , which predicts key ADME (Absorption, Distribution, Metabolism, and Excretion) properties. This analysis was conducted by the methodology previously described .
Virtual library generation
The virtual library was constructed with the protocol previously established, involving the systematic generation and screening of molecular structures based on predefined chemical criteria and computational modeling techniques .
ADME based library
The orientation of the virtual library was carried out based on multiple selection criteria, including molecular alignment, binding interactions, and geometric constraints, in accordance with the approach previously described .
Pharmacophore-based library search
The pharmacophore model (PH4), derived from the bound conformations of COUs at the NQO1 active site, was used as a tool for searching the library, as previously described .
Inhibitory power prediction
For each group within the targeted library subset, the conformer that exhibited the best alignment with the PH4 pharmacophore was selected for in silico screening using the complexation QSAR model. The ∆∆Gcom value for each newly selected analog was calculated to estimate its NQO1 inhibitory potency (pIC50pre). This prediction was integrated into the target-specific scoring function, as defined in equation (1), which was parameterized using the COU inhibitor training set within the complexation QSAR model .
pIC50exp=-log10IC50exp=a. ΔΔGcom+b(1)
3. Results
Training and validation sets:
A total of 28 COUs (Table 1) were selected from a series of compounds with experimentally determined properties, all originating from the same laboratory . Their measured inhibitory activities (0.18 nM ≤ IC50exp ≤ 1221 nM) cover a sufficiently broad concentration range to support the development of a robust QSAR model. The ratio between the training and validation set sizes is essential for accurate classification but is limited by the availability of homologous compound datasets in the literature . In this study, a training set of 22 COUs and a validation set of 6 VCOUs were constructed using the appropriate module in Discovery Studio 2.5 .
Table 1. NQO1 inhibitors (compounds from 1 to 15) used in the development of the QSAR inhibitor binding model. Group R is numbered in the table column as follows: R5, R6, R7, R8 and group X index .

Compound identification

R5

R6

R7

R8

X

IC50exp (nM)

1

H

H

H

H

H

2,6

2

H

CH3

H

H

H

14,00

3

OCH3

H

H

H

H

2,80

4

H

OCH3

H

H

H

11,00

5

H

H

OCH3

H

H

6,00

6

H

F

H

H

H

4,50

7

H

H

F

H

H

3,80

8

H

Cl

H

H

H

9,00

9

H

Br

H

Br

H

4,80

10

H

CH3

CH3

H

H

0,41

12

H

H

C4H4

C4H4

H

0,18

13

H

H

CH3

CH3

H

0,42

14

H

H

H

H

CO2Et

1221,00

15

H

H

H

H

C3H7

588,00

Table 2. NQO1inhibitors (compounds from 16 to 29) used in the development of the QSAR inhibitor binding model. Group R is numbered in the table column as follows: R5, R6, R7, R8 and group X index .

Compound identification

R5

R6

R7

R8

X

IC50exp (nM)

16

H

CH3

CH3

H

1-naphthyl

7,70

17

H

CH3

CH3

H

2-naphthyl

2,50

18

H

CH3

CH3

H

phenyl

39,00

19

H

H

C4H4

C4H4

1-naphthyl

6,30

20

H

H

C4H4

C4H4

2-naphthyl

2,20

21

H

H

C4H4

C4H4

phenyl

35,00

22

C4H4

C4H4

H

H

1-naphthyl

6,30

23

C4H4

C4H4

H

H

2-naphthyl

6,00

24

H

C4H4

H

H

phenyl

15,00

25

H

H

H

H

1-naphthyl

24,00

26

H

H

H

H

2-naphthyl

14,00

27

H

H

H

H

phenyl

144,00

28

H

H

H

H

3,4-diméthyl phenyl

31,00

29

H

CH3

CH3

H

3,4-diméthyl phenyl

9,90

3.1. QSAR Model
One-descriptor QSAR model: Each of the 22-training set (TS) and 6 validations set (VS) complexes was generated through in situ modification of the crystal structure of the refined NQO1 complex (PDB code: 3JSX) , following the procedure detailed in the Methods section. The relative Gibbs free energy (GFE) of NQO1 complex formation Gcom was computed for each of the 28 optimized enzyme-inhibitor complexes. Table 3 shows the calculated values (∆∆Gcom) for the molecules used for the training set (TS) . Table 4 shows the molecules used in the validation set (VS) .
To establish a quantitative structure-activity relationship (QSAR), the experimental inhibitory activities (IC50exp) of COUs were converted to (pIC50exp), then correlated with the computed ∆∆Gcom using linear regression (Table 3). The resulting strong correlation confirmed the active bound conformation of COUs within the NQO1 binding site, leading to the definition of the NQO1 inhibition pharmacophore (PH4).
Table 3. Gibbs free energy (binding affinity) and its components for the training set of NQO1 inhibitors COU1-22 .

Training Set

MW [g.mol-1]

∆∆HMM [kca.mol-1]

∆∆Gsol [kca.mol-1]

∆∆TSvib [kca.mol-1]

∆∆Gcom [kca.mol-1]

pIC50exp

New id.

Old id.

4]

IC50exp nM

COU1

12

0.18

436

0.00

0.00

0.00

0.00

9.74

COU2

1

2.60

336

3.32

1.29

0.40

4.21

8.59

COU3

2

14

364

8.76

-2.21

1.34

5.22

7.85

COU4

3

2.80

396

2.28

1.10

1.11

2.27

8.55

COU5

4

11

396

4.41

0.74

0.68

4.47

7.96

COU6

6

4.50

372

2.48

-0.59

-2.15

4.03

8.35

COU7

7

3.80

372

6.35

-3.52

0.44

2.39

8.42

COU8

10

0.41

392

0.01

1.75

3.91

-2.14

9.39

COU9

13

0.42

392

0.15

-0.44

0.26

-0.55

9.38

COU10

14

1221

408

14.24

-4.37

-0.80

10.66

5.91

COU11

15

588

378

13.79

-1.46

2.54

9.78

6.23

COU12

16

7.70

318

9.48

-2.02

3.93

3.53

8.11

COU13

17

2.50

318

6.73

-1.35

3.55

1.83

8.60

COU14

18

39

280

10.58

-1.59

2.74

6.24

7.41

COU15

20

2.20

352

2.56

0.66

2.36

0.86

8.66

COU16

21

35

302

11.6

-2.63

2.04

6.39

7.46

COU17

24

15

302

6.79

-1.86

1.66

3.27

7.82

COU18

25

24

302

6.47

0.91

3.08

4.30

7.62

COU19

26

14

302

7.89

-0.67

2.61

4.61

7.85

COU20

27

144

252

12.15

-1.84

2.12

8.19

6.84

COU21

28

31

280

9.92

-1.15

3.58

5.20

7.51

COU22

29

9.90

308

8.44

-1.02

4.51

2.91

8.00

To further elucidate COUs' binding affinity to NQO1, the gas-phase complexation enthalpy (∆∆HMM) was initially computed and analyzed by correlating it with pIC50exp. The statistical significance of this linear relationship (see Table 5, equation A) underscored the crucial role of inhibitor-enzyme interactions (∆∆HMM), particularly when solvent effects and entropy loss upon binding to NQO1 were neglected. This correlation explained approximately 85% of the variation in pIC50exp values, highlighting the enthalpic contribution to ligand binding affinity.
Additionally, a more comprehensive descriptor, the GFE of NQO1 complex formation, incorporating all key components ∆∆HMM, ∆∆TSvib, and ∆∆Gsol was evaluated (see Table 5, equation B). The relatively high regression coefficient (R²) demonstrated that structural insights derived from 3D models of NQO1 complexes could reliably predict the NQO1 inhibitory potency of novel COU analogs (with a similar binding mode) using the QSAR B model, as shown in Table 3.
Binding mode of COUs: The inhibitors (COUs) investigated in this study belong to a newly synthesized series of Coumarin derivatives, as detailed in reference . Coumarins are well known for their metal ion-chelating properties, with their acidic functional group playing a crucial role in NQO1 inhibition. The active site was analyzed using X-ray crystallographic data of NQO1 (PDB code: 3JSX) complexed with one of the most potent inhibitors identified in this study.
Table 4. Gibbs free energy (binding affinity) and its components for the validation set inhibitors VCOU1-6 .

Validation Set

MW [g.mol-1]

∆∆HMM [kca.mol-1]

∆∆Gsol [kca.mol-1]

∆∆TSvib [kca.mol-1]

∆∆Gcom [kca.mol-1]

pIC50pre

pIC50pre/ pIc50exp 

New id.

Old id.

IC50exp (nM)

pIC50exp

VCOU1

5

6.00

8.22

396

2.47

0.68

0.32

2.83

8.34

1.01

VCOU2

8

9.00

8.05

405

2.74

-0.41

-2.97

5.30

7.64

0.95

VCOU3

9

4.80

8.32

652

-2.34

-3.13

-8.78

3.30

8.21

0.99

VCOU4

19

6.30

8.20

352

6.86

-0.56

2.67

3.63

8.11

0.99

VCOU5

22

6.30

8.20

352

6.05

-0.91

2.04

3.11

8.26

1.01

VCOU6

23

6.00

8.22

352

7.65

-2.06

1.36

4.24

7.94

0.97

a for the chemical structures of the training set of inhibitors see Table 1; b Mw is the molar mass of inhibitors; c ∆∆HMM is the relative enthalpic contribution to the GFE change related to E-I complex formation derived by MM; ∆∆HMM ≈[EMM{E-Ix} − EMM{Ix}] − [EMM{E-Iref} − EMM{Iref}], Iref is the reference inhibitor COU12; d∆∆Gsol is the relative solvent effect contribution to the GFE change of E-I complex formation: ∆∆Gsol = [Gsol{E-Ix} − Gsol{Ix}] − [Gsol{E-Iref} − Gsol{Iref}]; e −∆∆TSvib is the relative entropic contribution of inhibitor to the GFE of E-Ix complex formation: ∆∆TSvib = [TSvib{Ix}E − TSvib{Ix}] − [TSvib{Iref}E − TSvib{Iref}]; f∆∆Gcom is the overall relative GFE change of E-Ix complex formation: ∆∆Gcom ≈∆∆HMM + ∆∆Gsol − ∆∆TSvib; g Kiexp is the experimental inhibitory concentration of NQO1 obtained from ref. ; h ratio of predicted and experimental half-maximal inhibition concentrations pIC50pre/pIC50exp (pIC50pre = −log10IC50pre) was predicted from computed ∆∆Gcom using the regression equation for NQO1 shown in Table 3 B.
Table 5. Analysis of computed binding affinities Gcom, its enthalpic component ΔΔHMM, and experimental inhibitory concentrations pIC50exp=-log10IC50exp of COUs towards NQO1 .

Statistical Data of Linear Regression

A

B

pIC50exp = - 0,1981×∆∆HMM + 9,3435 A

pIC50exp = - 0,2867×∆∆Gcom + 9,1546 B

Number of compound n

22

22

Squared correlation coefficient of

0.85

0.91

LOO cross-validated Squared

0.86

0.94

Standard error of regression σ

0,23

0.14

Statistical significance of regression.

224.1

584.3

Level of statistical significance α

> 95

> 95

Range of activities IC50exp [nM]

0.18 –1221

Interaction Energy: Key structural insights were obtained from the interaction energy (IE, ∆Eint) diagram generated for each inhibitor in the training set. The decomposition of IE into contributions from specific NQO1 active site residues provided valuable guidance for selecting relevant R groups at sites to enhance the binding affinity of COU analogs, thereby improving ligand potency. A comparative analysis of the IE values calculated for COUs, classified into three levels of activity (highest, moderate and lowest), identified residues for which it would be possible to increase the contributions to binding affinity. However, the results indicated that the IE contributions from active site residues remained consistent across all three classes of inhibitors, and no viable substitutions were identified to enhance binding affinity, unlike previous findings for thymine-like inhibitors of NQO1.
The statistical data depicted in Table 5 confirmed the validity of the correlation Equations (A) and (B). Likewise, the ratio pIC50prep/IC50exp close to one (1) for the validation set, where the pIC50pre values were estimated with correlation Equation (B), exhibits the substantial predictive power of the complexation QSAR model as showed in Table 3. Thus, the regression Equation (B) and the computed ∆∆Gcom GFEs can be used for the prediction of the inhibitory potencies pIC50pre against NQO1 for novel COU analogs, provided they share the same binding mode as the training set COU1-22.
Figure 1. (Left) plot of correlation equation between pIC50exp and relative enthalpic contribution to the GFE (∆∆HMM [kcal.mol-1]). (Right) similar plot for relative complexation Gibbs free energies of the NQO1-COU complex formation ∆∆Gcom [kcal.mol-1] of the training set . The validation set data points are shown in red color.
Figure 2. (Left) 2D schematic interaction diagram of the most potent inhibitor COU1 at the active site of NQO1 and (Right) 3D schematic interaction of COU1 at the enzyme active site.
Figure 3. Molecular Mechanics intermolecular interaction energy Eint breakdown to residue contributions in [kcal.mol-1]: (A: Top) the most active inhibitors, (B: Middle) moderately active inhibitors, (C: Bottom) less active inhibitors, Table 2 .
3.2. 3D-QSAR Pharmacophore Model
Generation and Validation of 3D-QSAR Pharmacophore: A 3D-QSAR pharmacophore model for NQO1 inhibition was developed using the active conformations of the 22 inhibitors originating from the trainings set and validated with the 6 inhibitors of the validation set. These inhibitors spanned a broad range of experimental activities (0,18–1221 nM), covering nearly three orders of magnitude. The pharmacophore generation process followed three key steps: (i) the constructive step, (ii) the subtractive step, and (iii) the optimization step , as previously described .
Hypotheses were evaluated based on errors in activity estimates from regression and complexity using a simulated annealing approach. The top 10 unique pharmacophore hypotheses were retained, all exhibiting five-point features, with the corresponding data listed in Table 6. Their selection was based on significant statistical parameters, including a high correlation coefficient, low total cost, and low RMSD. The cost difference (Δ = 986.56) was calculated as follows: null cost (1038.78) – fixed cost (52.14), indicating a high probability (>90%) that the model represents a true correlation .
The evaluation of hypothesis 1 (Hypo1) involves mapping the most active compound from the training set, COU1 (Figure 4D), illustrating the geometry of the Hypo1 pharmacophore for NQO1 inhibition. The following regression equation correlating pIC50exp as a function of pIC50pre for Hypo1 is pIC50exp=0.9092 × pIC50pre+0.7386. This relationship is plotted in Figure 4E (n = 22, R² = 0.91, R²xv = 0.90, F-test = 191, σ = 0.30, α > 95%). These results indicate that the PH4 model is highly reliable and could be efficiently used to identify new COU analogs.
Figure 4. (A) Features coordinates of centers, (B) mapping of pharmacophore of NQO1 inhibitor with the most potent molecule COU1, (C) Distances between centers, (D) angles between centers of pharmacophoric features. Feature legend: HYD = Hydrophobic (cyan), HBA = Hydrogen bond Acceptor (green), HBD = Hydrogen bond Donor (pink), Excluded volume. (E) Correlation plot of experimental vs. predicted inhibitory activity.
Table 6. Parameters of 10 generated PH4 pharmacophoric hypotheses for NQO1 inhibitor after Cat-Scramble validation procedure (49 scrambled runs for each hypothesis at the selected level of confidence of 98%).

Hypothesis

RSMDa

R2 b

Total Costc

Costs Differenced

Closest Randome

Featuresf

Hypo 1

2.896

0.95

145.5

893.2

192.1

HBD, HYD-Ar, HYD, HYD, R-Ar

Hypo 2

3.314

0.94

173.0

865.7

314.6

HBD, HYD-Ar, HYD, HYD, R-Ar

Hypo 3

3.416

0.93

180.5

858.2

351.2

HBD, HYD-Ar, HYD, HYD, R-Ar

Hypo 4

3.618

0.93

196.2

842.5

388.4

HBD, HYD-Ar, HYD-Ar, HYD, R-Ar

Hypo 5

3.634

0.92

197.4

841.3

425.7

HBD, HYD, HYD, HYD, R-Ar

Hypo 6

3.937

0.91

222.7

816.0

426.5

HBD, HYD, HYD, HYD, R-Ar

Hypo 7

4.004

0.90

228.5

810.2

439.1

HBD, HYD-Ar, HYD, HYD, R-Ar

Hypo 8

4.069

0.91

234.2

804.5

459.5

HBD, HYD-Ar, HYD, HYD, R-Ar

Hypo 9

4.569

0.88

282.2

756.5

479.4

HBD, HYD-Ar, HYD-Ar, HYD, HYD

Hypo 10

4.659

0.87

291.7

747.0

520.3

HBD, HYD-Ar, HYD-Ar, HYD

Fixed Cost

0

0

52.14

Null Cost

9.53

0

1038.78

Configuration cost = 12.09.
a root mean squared deviation; b squared correlation coefficient; c overall cost parameter of PH4 pharmacophore; d cost difference between Null cost and hypothesis total cost; e lowest cost from 49 scrambled runs at a selected level of confidence of 98%. f HBA (hydrogen-bond acceptor); HYD (hydrophobic); HYD-Ar (hydrophobic aromatic)
3.3. Virtual Screening
Virtual screening of combinatorial libraries has emerged as a powerful approach for identifying potential hit compounds. Our previous studies on inhibitor design have demonstrated its effectiveness in selecting promising candidates by exploring vast chemical spaces and predicting key molecular interactions.
Virtual Library
A preliminary virtual library (VL) was generated by modifying the COU scaffold at positions R1, R2, R3, and R4, as outlined in Table 7. During the enumeration process, the R-groups listed in Table 7 were systematically attached to in positions R1 to R4 of the COU scaffold to form a combinatorial library of the size: R1×R2×R3×R4=16×16×16×16= 65.536 analogs. All generated analogs maintained the substitution pattern of the most potent inhibitor, COU1.
To construct this library, fragments were sourced from chemical databases of available compounds . To enhance drug-like properties and reduce the library size, we applied a series of selection filters. These included the Lipinski rule-of-five, which, for example, excluded compounds with molecular weights above 500 g/mol , This refinement resulted in a more focused library, better suited for subsequent in silico screening.
3.4. In Silico Screening of Library of COUs
The focused library of 65.536 analogs was further screened for molecular structures that align with the 3D-PH4 pharmacophore model Hypo1 for NQO1 inhibition. A total of 63 COU analogs matched at least four features of the pharmacophore. These top-ranking analogs (PH4 hits) were then subjected to complexation QSAR model screening. The computed Gibbs free energy (GFE) of NQO1-COU complex formation, along with its components and the predicted half-maximal inhibitory concentrations (pIC50pre), derived from correlation Equation B (Table 3), are summarized in Table 8.
Figure 5. Best COU Analogs with scaffold of NQO1, the name is concatenation.
Figure 6. (A) Close up of virtual hit F8-F6-F1-F8, the most active designed COU analog (pIC50pre=12.22) at the active site of NQO1. (B) Mapping of the COU F8-F6-F1-F8 to NQO1 inhibition pharmacophore. (C) 2D schematic interaction diagram of the best active designed COU analog F8-F6-F1-F8 at the active site of NQO1. (D) Surface of the active site of NQO1 with bound best active designed COU analog. The binding site surface is colored according to residue hydrophobicity: red = hydrophobic, blue = hydrophilic, and white = intermediate.
Table 7. R1, R2, R3 and R4-groups (fragments, building blocks, substituents) used in the design of the initial diversity virtual combinatorial library.

New analogues

C1F

3-[(7-amino-4-hydroxy-2-oxo-8-vinyl-chromen-3-yl)methyl]-8-ethyl-4-hydroxy-7-vinyl-chromen-2-one

C2F

3-[(8-ethyl-4,7-dihydroxy-2-oxo-chromen-3-yl)methyl]-4-hydroxy-7-methyl-8-vinyl-chromen-2-one

C3F

3-[(8-ethyl-4-hydroxy-7-nitroso-2-oxo-chromen-3-yl)methyl]-4-methoxy-7-methyl-8-vinyl-chromen-2-one

C4F

3-[(7-acetyl-4-hydroxy-8-methyl-2-oxo-chromen-3-yl)methyl]-7-ethyl-4-hydroxy-8-methyl-chromen-2-one

C5F

3-[(8-ethyl-4-hydroxy-2-oxo-7-vinyl-chromen-3-yl)methyl]-4-methoxy-8-methyl-7-vinyl-chromen-2-one

C7F

8-ethyl-4-hydroxy-3-[[4-methoxy-8-(methylamino)-2-oxo-7-vinyl-chromen-3-yl]methyl]-7-vinyl-chromen-2-one

C9F

3-[(8-ethyl-4-hydroxy-7-nitroso-2-oxo-chromen-3-yl)methyl]-4-methoxy-7-methyl-8-vinyl-chromen-2-one

C10F

3-[(7-amino-8-ethyl-4-hydroxy-2-oxo-chromen-3-yl)methyl]-4-hydroxy-8-phosphanyl-7-propyl-chromen-2-one

C13F

3-[(8-acetyl-4-hydroxy-2-oxo-7-vinyl-chromen-3-yl)methyl]-4-hydroxy-8-methyl-7-propyl-chromen-2-one

C14F

4-hydroxy-3-[(7-hydroxy-4-methoxy-2-oxo-8-vinyl-chromen-3-yl)methyl]-2-oxo-8-propyl-chromene-7-carboxylic acid

C15F

3-[(4,7-dihydroxy-8-methyl-2-oxo-chromen-3-yl)methyl]-4-hydroxy-2-oxo-7-vinyl-chromene-8-carboxylic acid; ethane

C17F

3-[(8-ethyl-4-hydroxy-2-oxo-7-vinyl-chromen-3-yl)methyl]-4-methoxy-8-methyl-2-oxo-chromene-7-carbaldehyde

C18F

4-hydroxy-3-[(4-hydroxy-7-methyl-2-oxo-8-propyl-chromen-3-yl)methyl]-2-oxo-8-vinyl-chromene-7-carbaldehyde

C19F

4-hydroxy-3-[(4-hydroxy-8-methyl-2-oxo-7-phosphanyl-chromen-3-yl)methyl]-2-oxo-7-propyl-chromene-8-carboxamide

C20F

3-[(8-acetyl-4-hydroxy-2-oxo-7-vinyl-chromen-3-yl)methyl]-4-methoxy-8-methyl-7-vinyl-chromen-2-one

C23F

7-acetyl-4-hydroxy-3-[(7-hydroxy-4-methoxy-2-oxo-8-phosphanyl-chromen-3-yl)methyl]-8-propyl-chromen-2-one

C25F

4-hydroxy-3-[(4-hydroxy-8-methyl-7-nitroso-2-oxo-chromen-3-yl)methyl]-7-methyl-8-sulfanyl-chromen-2-one

C26F

8-ethyl-4-hydroxy-3-[(4-methoxy-7,8-dimethyl-2-oxo-chromen-3-yl)methyl]-2-oxo-chromene-7-carbaldehyde

C27F

4-hydroxy-3-[(4-hydroxy-2-oxo-8-phosphanyl-7-propyl-chromen-3-yl)methyl]-7-methyl-8-vinyl-chromen-2-one

C28F

7-ethyl-4-hydroxy-3-[(4-hydroxy-2-oxo-8-phosphanyl-7-propyl-chromen-3-yl)methyl]-2-oxo-chromene-8-carboxamide

C29F

8-(1-aminovinyl)-4-hydroxy-3-[(4-hydroxy-2-oxo-7,8-divinyl-chromen-3-yl)methyl]-7-methyl-chromen-2-one; hydrate

C30F

3-[(7-ethyl-4-hydroxy-2-oxo-8-vinyl-chromen-3-yl)methyl]-4-hydroxy-2-oxo-7-phosphanyl-chromene-8-carboxylic acid

C31F

8-ethyl-3-[(7-hydrosulfinyl-4-hydroxy-8-isopropenyl-2-oxo-chromen-3-yl)methyl]-4-hydroxy-7-sulfanyl-chromen-2-one

C33F

4,7-dihydroxy-3-[(4-hydroxy-2-oxo-7-phosphanyl-8-propyl-chromen-3-yl)methyl]-8-vinyl-chromen-2-one

C34F

3-[(7-amino-8-hydrosulfinyl-4-hydroxy-2-oxo-chromen-3-yl)methyl]-4-hydroxy-7-methyl-8-vinyl-chromen-2-one

C35F

4,7-dihydroxy-3-[(4-hydroxy-2-oxo-8-propyl-7-sulfanyl-chromen-3-yl)methyl]-8-phosphanyl-chromen-2-one

C36F

8-acetyl-3-[(8-amino-4-hydroxy-2-oxo-7-vinyl-chromen-3-yl)methyl]-7-ethyl-4-hydroxy-chromen-2-one

C37F

7-ethyl-3-[(7-formyl-4-hydroxy-2-oxo-8-vinyl-chromen-3-yl)methyl]-4-hydroxy-2-oxo-chromene-8-carboxylic acid

C38F

4-hydroxy-3-[(4-hydroxy-2-oxo-8-phosphanyl-7-propyl-chromen-3-yl)methyl]-2-oxo-7-sulfanyl-chromene-8-carboxylic acid

C39F

ammonia; 3-[(8-ethyl-4-hydroxy-2-oxo-7-vinyl-chromen-3-yl)methyl]-4-methoxy-7-methyl-8-vinyl-chromen-2-one

C40F

4,7-dihydroxy-3-[(4-hydroxy-8-nitroso-2-oxo-7-sulfanyl-chromen-3-yl)methyl]-8-vinyl-chromen-2-one

C41F

8-ethyl-4-hydroxy-3-[(7-hydroxy-4-methoxy-2-oxo-8-vinyl-chromen-3-yl)methyl]-7-nitroso-chromen-2-one

C42F

3-[(7-amino-4-hydroxy-2-oxo-8-phosphanyl-chromen-3-yl)methyl]-4-hydroxy-2-oxo-7-vinyl-chromene-8-carbaldehyde

C43F

3-[(7-acetyl-4-hydroxy-2-oxo-8-vinyl-chromen-3-yl)methyl]-4-hydroxy-8-nitroso-7-propyl-chromen-2-one

C45F

8-formyl-3-[(4-hydroxy-8-methyl-2-oxo-7-vinyl-chromen-3-yl)methyl]-4-methoxy-2-oxo-chromene-7-carboxamide

C46F

3-[(4,7-dihydroxy-2-oxo-8-vinyl-chromen-3-yl)methyl]-8-hydrosulfinyl-4-hydroxy-7-propyl-chromen-2-one

C51F

3-[(8-ethyl-4-hydroxy-7-nitroso-2-oxo-chromen-3-yl)methyl]-4-methoxy-7-methyl-8-vinyl-chromen-2-one

C52F

4-hydroxy-3-[(4-hydroxy-2-oxo-8-phosphanyl-7-sulfanyl-chromen-3-yl)methyl]-2-oxo-8-vinyl-chromene-7-carbaldehyde

C53F

3-[(7-ethyl-4-hydroxy-2-oxo-8-sulfanyl-chromen-3-yl)methyl]-4-hydroxy-2-oxo-8-vinyl-chromene-7-carboxylic

C54F

7-ethyl-3-[(7-formyl-4-hydroxy-2-oxo-8-vinyl-chromen-3-yl)methyl]-4-hydroxy-2-oxo-chromene-8-carboxylic

C55F

3-[(7-amino-4-hydroxy-2-oxo-8-vinyl-chromen-3-yl)methyl]-4-hydroxy-7-methyl-8-vinyl-chromen-2-one

C56F

4-hydroxy-3-[(4-hydroxy-2-oxo-8-sulfanyl-7-vinyl-chromen-3-yl)methyl]-2-oxo-chromene-7,8-dicarbaldehyde

C57F

3-[(4,7-dihydroxy-8-methyl-2-oxo-chromen-3-yl)methyl]-4,7-dihydroxy-2-oxo-chromene-8-carbaldehyde

C587

3-[(4,8-dihydroxy-2-oxo-7-vinyl-chromen-3-yl)methyl]-4-hydroxy-8-phosphanyl-7-propyl-chromen-2-one

C60F

3-[(4,8-dihydroxy-2-oxo-7-phosphanyl-chromen-3-yl)methyl]-4-hydroxy-8-phosphanyl-7-propyl-chromen-2-one

C61F

3-[(8-formyl-4-hydroxy-2-oxo-7-phosphanyl-chromen-3-yl)methyl]-4-hydroxy-2-oxo-8-phosphanyl-chromene-7-carbaldehyde

C63F

3-[(4,8-dihydroxy-7-methyl-2-oxo-chromen-3-yl)methyl]-4-hydroxy-7-methyl-8-vinyl-chromen-2-one

C64F

3-[(4,8-dihydroxy-2-oxo-7-phosphanyl-chromen-3-yl)methyl]-4-hydroxy-8-nitroso-7-phosphanyl-chromen-2-one

C73F

3-[(7-formyl-4-hydroxy-2-oxo-8-sulfanyl-chromen-3-yl)methyl]-4,8-dihydroxy-2-oxo-chromene-7-carbaldehyde

C76F

4-hydroxy-3-[(4-hydroxy-2-oxo-8-phosphanyl-7-propyl-chromen-3-yl)methyl]-7-methyl-8-vinyl-chromen-2-one

C78F

7-ethyl-4-hydroxy-3-[(4-hydroxy-2-oxo-8-phosphanyl-7-propyl-chromen-3-yl)methyl]-2-oxo-chromene-8-carboxamide

C81F

3-[(8-ethyl-4,7-dihydroxy-2-oxo-chromen-3-yl)methyl]-4-hydroxy-8-nitroso-7-vinyl-chromen-2-one

C82F

7-ethyl-4-hydroxy-3-[(4-hydroxy-8-nitroso-2-oxo-7-phosphanyl-chromen-3-yl)methyl]-8-vinyl-chromen-2-one

C83F

4-hydroxy-3-[(4-hydroxy-8-methyl-2-oxo-7-propyl-chromen-3-yl)methyl]-2-oxo-8-phosphanyl-chromene-7-carboxylic

C85F

3-[(4,7-dihydroxy-2-oxo-8-vinyl-chromen-3-yl)methyl]-7-ethyl-4-hydroxy-8-nitroso-chromen-2-one

C89F

3-[(4,7-dihydroxy-8-nitroso-2-oxo-chromen-3-yl)methyl]-4-hydroxy-8-phosphanyl-7-propyl-chromen-2-one

C91F

4-hydroxy-3-[(4-hydroxy-2-oxo-7-propyl-8-sulfanyl-chromen-3-yl)methyl]-2-oxo-8-phosphanyl-chromene-7-carboxylic

C92F

3-[(8-acetyl-4-hydroxy-7-nitroso-2-oxo-chromen-3-yl)methyl]-4-methoxy-7-methyl-8-phosphanyl-chromen-2-one

C93F

4-hydroxy-3-[(4-hydroxy-2-oxo-8-phosphanyl-7-propyl-chromen-3-yl)methyl]-2-oxo-7-sulfanyl-chromene-8-carboxylic

C96F

3-[(4,7-dihydroxy-2-oxo-8-vinyl-chromen-3-yl)methyl]-4-hydroxy-7-propyl-8-sulfanyl-chromen-2-one

C98F

7-ethyl-4-hydroxy-3-[(4-hydroxy-2-oxo-7-phosphanyl-8-sulfanyl-chromen-3-yl)methyl]-8-vinyl-chromen-2-one

C100

8-acetyl-4-hydroxy-3-[(4-hydroxy-2-oxo-7-propyl-8-sulfanyl-chromen-3-yl)methyl]-7-methyl-chromen-2-one

C108F

3-[(7-amino-8-hydrosulfinyl-4-hydroxy-2-oxo-chromen-3-yl)methyl]-4-hydroxy-7-propyl-8-sulfanyl-chromen-2-one

Table 8. GFE and their components for the top scoring 63 virtual COU analogs. The analog numbering concatenates the index of each substituent R1, R2, R3, and R4 with the substituent numbers taken from Table 7.

N

Analogs

MW [g.mol-1]

∆∆HMM [kca.mol-1]

∆∆Gsol [kca.mol-1]

∆∆TSvib [kca.mol-1]

∆∆Gcom [kca.mol-1]

pIC50pre

C1

F1-F8-F8-F6

431

3.55

-0.75

5.14

-2.35

9.83

C2

F1- F10- F8- F2

420

-0.08

1.97

2.41

-0.53

9.31

C3

F1-F13-F8-F10

435

4.79

-0,43

-0.37

4.73

7.80

C4

F2-F1-F2-F4

434

-1,36

2.51

5.85

-4.70

10.50

C5

F1-F8-F2-F9

432

4.18

-0.75

0.43

2.99

8.30

C7

F1-F8-F6-F8

458

3.23

-0.55

4.54

-1.85

9.69

C9

F1-F13-F8-F6

434

2.69

-0.47

2.12

0.11

9.12

C10

F1-F6-F14-F3

453

-3.41

3.17

8.74

-8.99

11.73

C13

F2-F3-F4-F8

460

-2.64

3.53

3.59

-2.70

9.93

C14

F3-F7-F8-F10

464

3.06

1.62

1.31

3.37

8.19

C15

F2-F10-F7-F8

436

-0.27

2.13

-1.29

3.16

8.25

C17

F2-F9-F1-F8

432

4.17

-0.74

0.40

3.04

8.28

C18

F3-F2-F8-F9

446

3.51

2,19

3.46

2.24

8.51

C19

F2-F14-F5-F3

467

-2.68

3.38

0.09

0.60

8.98

C20

F2-F8-F4-F8

444

2.11

-0.68

2.82

-1.39

9.55

C23

F3-F4-F14-F10

468

3.10

0.49

5.27

-1.68

9.64

C25

F2-F13-F16-F2

425

0.66

2.25

-1.33

4.24

7.94

C26

F2-F6-F1-F9

421

2.71

-0.19

0.38

2.14

8.54

C27

F8-F2-F14-F3

450

-2.84

2.99

2.06

-1.91

9.70

C28

F5-F1-F14-F3

481

-1.90

1.93

3.91

-3.87

10.26

C29

F5-F2-F8-F8

445

-0.96

3.26

3.19

-0.89

9.41

C30

F8-F1-F7-F14

466

-2.33

2.71

-17.93

18.32

3.90

C31

F4-F15-F1-F16

486

0.81

2.52

-0.36

3.69

8.10

C33

F3-F14-F8-F10

452

4.48

2.52

4.58

2.41

8.46

C34

F8-F2-F15-F6

439

1.81

1.64

1.55

1.89

8.61

C35

F3-F16-F14-F10

458

4.02

2.77

2.55

4.24

7.94

C36

F4-F1-F6-F8

447

-3.22

2.72

2.92

-3.43

10.14

C37

F7-F1-F8-F9

462

-2.04

2.19

-0.61

0.76

8.94

C38

F7-F16-F14-F3

486

3.59

3.14

1.47

-1.92

9.70

C39

F8-F6-F1-F8

431

-5.25

-0.37

5.06

-10.68

12.22

C40

F8-F10-F13-F16

439

0.75

3.00

1.07

2.68

8.39

C41

F8-F10-F1-F13

435

4.79

-0.42

-0.35

4.72

7.80

C42

F9-F8-F14-F6

437

-0.53

2.29

-0.58

2.34

8.48

C43

F8-F4-F13-F3

475

-2.49

2,68

-0.16

0.34

9.06

C45

F9-F5-F2-F8

447

3.32

-0.70

-1.13

3.75

8.08

C46

F8-F10-F15-F3

468

0.55

3.69

1.95

2.28

8.50

C51

F8-F6-F1-F13

434

2.69

1.45

2.12

2.03

8,57

C52

F8-F9-F14-F16

454

1.02

1.98

-1.12

4,12

7.97

C53

F8-F7-F16-F1

466

-1.55

2.67

0.26

0.86

8.91

C54

F8-F9-F7-F1

462

-2.02

2.21

-1.21

1.40

8.75

C55

F8-F6-F8-F2

417

-0.35

2.65

1.46

0.84

8.91

C56

F9-F9-F16-F8

450

-0.03

2.42

-0.76

3.16

8.25

C57

F9-F10-F2-F10

410

3.25

2.14

-1.35

6.74

7.22

C58

F10-F8-F14-F3

452

-3.50

2.99

4.86

-5,37

10.69

C60

F10-F14-F14-F3

458

-3.54

3.19

5.10

-5.45

10.72

C61

F9-F14-F14-F9

456

-1.07

2,20

-1.61

2.74

8.37

C63

F10-F2-F8-F2

406

0.25

2.52

2.30

0.47

9.02

C64

F10-F14-F13-F14

445

-1.86

3.03

-0.26

1,43

8.74

C73

F10-F9-F16-F9

440

-0.49

4.20

-1.74

5.46

7.59

C76

F14-F3-F8-F2

450

-3.41

2.86

2.41

-2.95

10.00

C78

F14-F3-F5-F1

481

-5.56

4.72

2.80

-3.64

10.20

C81

F13-F8-F1-F10

435

-0.14

2.13

1.83

0.16

9.11

C82

F13-F14-F8-F1

451

-2.46

3.27

2.00

-1,18

9.49

C83

F14-F7-F2-F3

468

-3.17

2.78

1.53

-1.91

9.70

C85

F13-F1-F8-F10

435

1.70

2.50

0,77

3.43

8.17

C89

F13-F10-F14-F3

455

-0.90

2,94

3.79

-1.74

9.65

C91

F14-F7-F16-F3

486

-2.64

2.74

0.06

0.03

9.15

C92

F14-F16-F4-F13

471

1.48

0.00

0.16

1.32

8.77

C93

F14-F3-F7-F16

486

-3.41

3.12

1.43

-1.71

9.65

C96

F16-F3-F8-F10

452

-1,15

2.77

1.99

-0.36

9.26

C98

F16-F14-F8-F1

454

-0.67

2.44

2.55

-0.78

9.38

C100

F16-F3-F4-F2

466

-1.67

3.14

3.77

-2.31

9.82

C108

F15-F6-F16-F3

473

-0.48

2.42

1.42

0.52

9.01

Ref

COU1

436

0

0

0

0

pIC50exp=9.74g

a Mw is molar mass of inhibitor; b ∆∆HMM is the relative enthalpic contribution to the GFE change of the NQO1-COU complex formation ∆∆Gcom (for details see footnote of Table 2); c ∆∆Gsol is the relative solvation GFE contribution to ∆∆Gcom; d ∆∆TSvib is the relative (vibrational) entropic contribution to ∆∆Gcom; e ∆∆Gcom is the relative Gibbs free energy change related to the enzyme–inhibitor NQO1-COU complex formation ∆∆Gcom ≡ ∆∆HMM + ∆∆Gsol − ∆∆TSvib; f pIC50pre is the predicted inhibition potency towards NQO1 calculated from ∆∆Gcom using correlation Equation B, Table 3; g pIC50exp is given for the reference inhibitor COU1.
Figure 7. Histograms of frequency of occurrence of individual R-groups in the 63 best selected analogs mapping to four features of the PH4 pharmacophore hypothesis Hypo1.
Table 9. ADME-related properties of the best designed analogs and known anticancer agents either in clinical use or currently undergoing clinical testing computed by QikProp .

Analoguesa

#starsb

Mwc

Smold

Smolhfoe

Vmolf

#rotBg

HBdonh

F1-F8-F8-F6

0

431

725.8

250.2

1299.1

8

3.5

F2-F1-F2-F4

0

434

726.5

358.7

1306.2

6

2

F1-F6-F14-F3

1

454

747.5

293.5

1345.1

9

5.5

F2-F3-F4-F8

0

460

768.5

367.3

1390.2

8

2

F5-F1-F14-F3

1

481

751.3

290.4

1378.3

9

6

F4-F1-F6-F8

0

447

702.3

273.6

1293.5

8

3.5

F8-F6-F1-F8

0

431

718.7

250.1

12978

8

3.5

F10-F8-F14-F3

1

452

730

241.1

1308

9

5

F10-F14-F14-F3

2

458

724

177

1289.2

9

7

F14-F3-F8-F2

1

450

714.4

301.6

1311.2

8

4

F14-F3-F5-F1

1

481

752.2

291.1

1379.3

9

6

F16-F3-F4-F2

0

466

734.5

322.6

1331.1

8

2.8

COU1

0

436

666.6

167.9

1201

7

4

Finastéride

0

372.550

605.996

431.904

1152.042

3

2.000

Alfuzosine

1

389.453

919.272

417.508

1140.426

3

3.000

Téraz (R)

0

387.438

621.508

422.153

1137.212

9

2.000

Téraz (S)

0

387.438

603.495

405.495

1131.925

5

2.000

Analoguesa

HBacci

LogPo/wj

LogSwatk

LogKhsal

LogB/Bm

BIPcacon

#metabo

PIC50prep

HOAq

%HOAr

F1-F8-F8-F6

7.5

2.845

-6.032

0.235

-2.063

119.2

5

9.83

3

80.8

F2-F1-F2-F4

8.5

2.651

-5.822

0.230

-1.870

135.2

6

10.50

3

80.6

F1-F6-F14-F3

7.5

2.878

-6.241

0.138

-1.993

117.2

7

11.73

2

80.8

F2-F3-F4-F8

8.5

3.290

-6.337

0.349

-1.928

173.6

5

9.93

3

86.3

F5-F1-F14-F3

9

2.330

-6.208

-0.007

-2.137

79.6

7

10.26

2

74.6

F4-F1-F6-F8

9.5

2.047

-5.621

-0.057

-1.936

119.9

6

10.14

3

76.1

F8-F6-F1-F8

7.5

2.841

-6.032

0.239

-2.018

120.8

5

12.22

3

80.8

F10-F8-F14-F3

7.3

3.015

-6.365

0.133

-1.812

158.5

6

10.69

3

84

F10-F14-F14-F3

7.3

2.639

-6.227

-0.106

-1.779

117

6

10.72

3

79.4

F14-F3-F8-F2

6.5

3.830

-6.706

0.396

-1.162

472.2

6

10.00

3

100

F14-F3-F5-F1

9

2.344

-6.208

-0.005

-2.129

81.2

7

10.20

2

74.9

F16-F3-F4-F2

9

2.827

-6.219

0.107

-1.687

190.1

6

9.82

3

84.3

COU1

9.3

1.641

-5.464

-0.364

-2.453

8.3

5

2

53.0

Finastéride

5.000

3.553

-4.91

0.549

-0.75

777.322

1

3

100

Alfuzosine

8.700

1.322

-3.639

-0.601

-1.076

347.334

5

3

80.158

Téraz (R)

9.200

1.273

-3.425

-0.452

-0.646

470.273

4

3

82.229

Téraz (S)

9.200

1.262

-3.425

-0.440

-0.596

467.119

4

3

82.113

a designed COU analogs and known anticancer agents, Table 6; b drug likeness, number of property descriptors (24 out of the full list of 49 descriptors of QikProp, ver. 3.7, release 14) that fall outside of the range of values for 95% of known drugs; c molar mass in [g.mol-1] (range for 95% of drugs: 300–500 g.mol-1) ; d total solvent-accessible molecular surface, in [Å2] (probe radius 1.4 Å) (range for 95% of drugs: 300–1000 Å2); e hydrophobic portion of the solvent-accessible molecular surface, in [Å2] (probe radius 1.4 Å) (range for 95% of drugs: 0–750 Å2); f total volume of molecule enclosed by solvent-accessible molecular surface, in [Å3] (probe radius 1.4 Å) (range for 95% of drugs: 500–2000 Å3); g number of non-trivial (not CX3), non-hindered (not alkene, amide, small ring) rotatable bonds (range for 95% of drugs: 0–15); h estimated number of hydrogen bonds that would be donated by the solute to water molecules in an aqueous solution. Values are averages taken over a number of configurations, so they can assume non-integer values (range for 95% of drugs: 0.0–6.0); i estimated number of hydrogen bonds that would be accepted by the solute from water molecules in an aqueous solution. Values are averages taken over a number of configurations, so they can assume non-integer values (range for 95% of drugs: 2.0–20.0); j logarithm of partitioning coefficient between n-octanol and water phases (range for 95% of drugs: -2 to 6.5); k logarithm of predicted aqueous solubility, log S in [mol.dm–3] is the concentration of the solute in a saturated solution that is in equilibrium with the crystalline solid (range for 95% of drugs: -6.0 to 0.5); l logarithm of predicted binding constant to human serum albumin (range for 95% of drugs: -1.5 to 1.5); m logarithm of predicted brain/blood partition coefficient (range for 95% of drugs: -3.0 to 1.2); n predicted apparent Caco-2 cell membrane permeability in Boehringer-Ingelheim scale in [nm s-1] (range for 95% of drugs: < 25 poor, > 500 nm s -1 great); o number of likely metabolic reactions (range for 95% of drugs: 1–8); p predicted inhibition constants Kipre. Kiprewas predicted from computed ∆∆Gcom using the regression Equation B shown in Table 3; q human oral absorption (1 = low, 2 = medium, 3 = high); r percentage of human oral absorption in gastrointestinal tract (80% = high); * star in any column indicates that the property descriptor value of the compound falls outside the range of values for 95% of known drugs
3.5. Analysis of Novel COUs Analogs
The virtual library of novel COU analogs was designed based on structural information obtained from the active conformation of COUs. This information guided the selection of suitable substituents at positions R1, R2, R3, and R4. To identify which substituents, contribute to the highest predicted potency against NQO1, we analyzed the frequency of occurrence of R1, R2, R3, and R4 among the top 63 PH4 hits. The results are presented as histograms in Figure 7. These histograms show that the substituent (F8) is the most frequent at position R1, with an occurrence frequency of 13. The fragments at position R2 i.e., fragments (F10) and (F14) are the most repetitive and are followed by the substituent (F1, F3, F6 and F8) with occurrence frequencies of 7 and 6 respectively. As for the substituents at position R3, the substituents with the highest occurrence frequency are the substituents (F8) and (F14), with frequencies of 16 and 12 respectively. Concerning the fragments at position R4, the relative histogram reveals that the substituent (F3) is the most important with an occurrence rate of 14. Analysis of the frequencies of occurrence of substituents at R1, R2, R3 and R4 highlights recurring combinations among the most active analogues, including R1-F8, R2-F10, R3-F8 and R4-F3. These fragments appear to play a key role in enhancing biological activity and provide a solid basis for the design of new optimized derivatives.
The optimization of R1 is particularly crucial, with fragment F8 being the most frequent (13 occurrences), suggesting that it significantly improves biological activity. In R2, fragments F10 and F14 are predominant, followed by F1, F3, F6, and F8 (frequencies of 7 and 6 occurrences). In R3, fragments F8 and F14 dominate (16 and 12 occurrences, respectively), indicating that this position plays a key role in affinity optimization. Finally, substituent F3 in R4 is the most recurrent (14 occurrences), suggesting that it is essential for interaction with the target and influences selectivity. Statistical analysis of recurring substituents provides a rational basis for optimizing future COU analogs. These trends provide valuable information to guide the future design of analogs, thus optimizing their efficacy and selectivity.
Figure 8. Molecular mechanics inter-molecular interaction energy Eint break-down to active site residue contributions in [kcal.mol-1]: the best four novel designed COU analogs (the color coding refers to ligands given in the legend).
ADME profiles of designed
The pharmacokinetic profile of NQO1 inhibitors still requires further research. As presented in Table 9, the ADME properties of our novel analogs were previously analyzed using the QikProp program, based on Jorgensen’s method. The fundamental principles of this approach have been described earlier. Our top-performing analogs are compared with commercially available drugs used for cancer treatment (Table 9).
4. Discussion
Beyond the robustness of the QSAR model, the analysis of interactions between COU and the active site residues revealed the key interactions responsible for COUs affinity with NQO1, including hydrogen bonds, van der Waals interactions, and hydrophobic contacts.
Additional insights into the interaction of ligands from the training set are provided by the NQO1−COUx interaction energy diagram, which illustrates the individual energetic contributions of each active site residue in NQO1. The decomposition of interaction energy into individual contributions from NQO1's active site residues is crucial for identifying substituents that can enhance the binding affinity of COU analogs with NQO1 and, consequently, improve their inhibitory capacity. The individual contributions of interaction energies are categorized into three groups based on ligand activity levels within the training set: most active ligands, moderately active ligands and least active ligands. A comparative analysis of the interaction energy contributions across these groups (Figure 3) reveals that the binding affinity of the most active ligands is the strongest, highlighting key residues that play a significant role in ligand stabilization and potency.
The top 4 best hit fit from the PH4 result were evaluated with the QSAR complexation model, equation B from Table 3, in order to predict their inhibitory activity (pIC50pre): F10-F8-F14-F3: (pIC50pre =10.69); F10-F14-F14-F3: (pIC50pre =10.72); F1-F6-F14-F3: (pIC50pre =11.73); F8-F6-F1-F8: (pIC50pre = 12.22).
5. Conclusion
Structural information from the crystallography of the NQO1-COU12 complex enabled the development of a reliable QSAR model for NQO1 inhibition by coumarin inhibitors, establishing a correlation between the calculated Gibbs free energies of complexation and the observed inhibitory potencies. Furthermore, a PH4 pharmacophore model was developed for these inhibitors using a training set of 22 coumarin derivatives. This model was validated with 6 coumarin derivatives for which their inhibitory. The analysis of interactions between NQO1 and coumarins within the enzyme's active site guided the design of an initial virtual combinatorial library of new coumarin analogues featuring various substitutions on the coumarin core. A refined selection, based on ADME-related descriptors and screening for alignment with the PH4 pharmacophore, allowed the identification of a subset of coumarins with potential oral bioavailability. This group, consisting of the 63 best virtual candidates, was then evaluated for predicted inhibitory activities using the QSAR complexation model, revealing analogues with estimated activities in the picomolar concentration range. The best designed COU analogues F10-F8-F14-F3: (IC50pre=20.3 pM; pIC50pre=10,69); F10-F14-F14-F3: (IC50pre=19.2 pM; pIC50pre=10,72); F1-F6-F14-F3: (IC50pre=1.9 pM; pIC50pre=11,73); F8-F6-F1-F8: (IC50pre=0.6 pM; pIC50pre=12,22), Table 6, are recommended for synthesis and further evaluation of activity in NQO1 inhibition assays and could lead to the discovery of new potent anticancer agents, whether synthetic or natural, with good bioavailability.
Abbreviations

2D

Two-dimensional

3D

Three-dimensional

ADME

Absorption, Distribution, Metabolism, and Excretion

COU

Coumarin Inhibitors

COUx

Coumarin Inhibitors of the Training Set

VCOUs

Coumarin Inhibitors of the Validation Set

∆∆Gcom

Relative Complexation Gibbs Free Energy

GFE

Gibbs Free Energy

∆∆Gsol

Relative Solvation Gibbs Free Energy

HBA

Hydrogen Bond Acceptor

HBD

Hydrogen Bond Donor

HMM

Enthalpy Component of Gibbs Free Energy

HOA

Human Oral Absorption

HYD

Hydrophobic

HYDA

Hydrophobic Aliphatic

Ki

Inhibitory Concentration

MM

Molecular Mechanics

MM-PB

Molecular Mechanics–poisson–boltzmann

NQO1

NAD(P)H Quinone Oxydoréductase 1

PDB

Protein Data Bank

PH4

Pharmacophore

QSAR

Quantitative Structure–activity Relationships

RMSD

Root-mean Square Deviation

SAR

Structure–activity Relationships

TS

Training Set

VS

Validation Set

Conflicts of Interest
The authors declare no conflicts of interest.
References
[1] Xie Y H, Chen Y X, Fang J Y. Comprehensive review of targeted therapy for colorectal cancer. Signal transduction and targeted therapy, 2020, 5(1), 22.
[2] Kolotyeva N A, Groshkov A A, Rozanova N A, Berdnikov A K, Novikova, S V, Komleva, Y K, Piradov, M A. Pathobiochemistry of Aging and Neurodegeneration: Deregulation of NAD+ Metabolism in Brain Cells. Biomolecules, 2024, 14(12), 1556.
[3] Nolan K A, Doncaster J R, Dunstan M S, Scott K A, Frenkel A D, Siegel D, Bryce R A. Synthesis and biological evaluation of coumarin-based inhibitors of NAD (P) H: quinone oxidoreductase-1 (NQO1). Journal of medicinal chemistry, 2009, 52(22), 7142-7156.
[4] Verma S, Pathak R K. Discovery and optimization of lead molecules in drug designing. In Bioinformatics. Academic Press, 2022, 253-267.
[5] Niazi S K, and Mariam Z. Computer-aided drug design and drug discovery: a prospective analysis. Pharmaceuticals, 2023, 17(1), 22.
[6] Araújo R S, Silva-Junior E F, Aquino T M, Scotti M T, Ishiki H M, Scotti L, Mendonça-Junior F J B. Computer-aided drug design applied to secondary metabolites as anticancer agents. Current Topics in Medicinal Chemistry, 2020, 20(19), 1677-1703.
[7] Lionta E, Spyrou G, Vassilatis D K, Cournia Z. Structure-based virtual screening for drug discovery: principles, applications and recent advances. Current Topics in Medicinal Chemistry, (2014), 14(16): 1923–1938.
[8] Zhang T, Li X, Kumar R, Chen Y. Future perspective on computer-aided drug design for oncology therapeutics. Pharmacological Innovations, 2024, 32(3): 55-69.
[9] Li R, Bianchet M A, Talalay P, Amzel L M. The three-dimensional structure of NAD (P) H: quinone reductase, a flavoprotein involved in cancer chemoprotection and chemotherapy: mechanism of the two-electron reduction. Proceedings of the National academy of sciences, (1995), 92(19): 8846-8850.
[10] Yang C, Hu Q. Roles of NAD(P)H: quinone oxidoreductase 1 (NQO1) in diverse diseases. Life, 2021, 11(12): 1301.
[11] Silva V L, Silva-Reis R, Moreira-Pais A, Ferreira T, Oliveira P A, Ferreira R, Grozea I. Dicoumarol: From chemistry to antitumor benefits. Chinese Medicine, 2022, 17(1), 145.
[12] Kang S Y, Lee J H, Park E J. IP‑DNQ induces mitochondrial dysfunction and ROS‑mediated apoptosis in NQO1‑overexpressing pancreatic cancer cells. Antioxidants & Redox Signaling, 2023.
[13] Berridge M V, Herst P M, Prata C. Cellular reductive stress: Is plasma membrane electron transport an evolutionarily-conserved safety valve? Redox Biochem. Chem., 2023, 5(6): 1-9.
[14] Insight-II and Discover Molecular Modeling and Simulation Package, version 2005; Accelrys: San Diego, CA, USA, 2005.
[15] Discovery Studio molecular modeling and simulation program, Version 2.5, Accelrys, Inc., San Diego, CA, 2009.
[16] Wang X, Xu Y, Zheng H, Yu K. An Extendible, Graph-Neural-Network-Based Approach for Accurate Force Field Development of Large Flexible Organic Molecules. arXiv preprint arXiv: 2106.00927, 2021.
[17] Niaré A., Alex, A. Y. Z., Bernard, D. A. M., Marius, K. S., Stéphane, D. S., Moise, K. A., Guy-Richard, K. M., Fagnidi, Y. K. H., & Doh, S. (2025). Study by Molecular Docking of the Interactions Between Dihydroorotate Dehydrogenase and a Series of Inhibitors of Pyrrole Derivatives for the Treatment of Malaria. Asian Journal of Chemical Sciences, 15(1): 92-110.
[18] Soro I, N’Guessan H, Abou A, N’Guessan R K, Megnassan E. Conformational study of molecules in a biological environment, design of inhibitors of human aminopeptidase m1 implicated in cancer therapy. Universal Journal of Pharmaceutical Research. in our previous studies on the design of inhibitors. Universal J. of Pharm Research, 2023, 8(5): 71-86.
[19] Gilson, Michael K. and Barry H. The inclusion of electrostatic hydration energies in molecular mechanics calculations, J. Comput. Aided Mol. Des., 1991, 5(1), 5-20.
[20] Owono Owono L C, Ntie‐Kang F, Keita M, Megnassan E, Frecer V, Miertus S. Virtually Designed Triclosan‐Based Inhibitors of Enoyl‐Acyl Carrier Protein Reductase of Mycobacterium tuberculosis and of Plasmodium falciparum. Molecular Informatics, 2015, 34(5), 292-307.
[21] QikProp; Version 3.7, Release 14, X Schrödinger; LLC: New York, NY, USA, 2014.
[22] Herbert J M. Dielectric continuum methods for quantum chemistry. WIREs Computational Molecular Science, 2022, 12(1): 1519.
[23] Barber C, Heghes C, Johnston L. A framework to support the application of the OECD guidance documents on (Q) SAR model validation and prediction assessment for regulatory decisions. Computational Toxicology, 2024, 30: 100305.
[24] N’Guessan H, Soro I, Keita M, Megnassan E. Design and In silico Screening of Combinatorial Library of New Herbicidal Analogs of Cycloalka [d] quinazoline 2, 4 dione− Benzoxazinones Inhibiting Protoporphyrinogen IX Oxidase. J Pharm Res Int, 2022, 34(56): 42-61.
[25] Niaré, A., N’guessan, A.-B., Djako, B. A. M., Dembélé, G. S., Koné, M. G.-R., Yéo, Y. (2024). QSAR, pharmacophore, and molecular docking studies for the design of novel arylamide-derived inhibitors of Mycobacterium tuberculosis. Chemical Science International Journal, 33(6), 212–224.
[26] Keita M, Yaya Y, Yvon B M B, Esmel A E, Dali B, N’Guessan, H. Molecular and Thermodynamic Modeling of the Protein-Ligand Interaction. Application to Computer-Assisted Design of Anti-Competitive Inhibi-tors of Human Histone Deacetylase, 2021, 2: 606-630.
[27] Kone M, N’Guessan H, N'gouan A J, Megnassan E. Computer-aided design of new hydroxamic acid derivatives targeting the Plasmodium falciparum M17 Metallo-aminopeptidase with Favorable Pharmacokinetic Profile. Int. J. Pharm. Sci. Drug Res., 2023: 356-375.
[28] Niaré A, N’Guessan H, Dali B, Megnassan E. Computer-assisted design of hydroxamic acid derivatives inhibitors of M1 Metallo Aminopeptidase of Plasmodium falciparum with favorable pharmacokinetic profile. J Pharm Res Int, 2022: 21-44.
[29] Bléhoué I C, Koné M, Esmel E A, Fofana I, Kéïta M, Megnassan E. Molecular modelling and virtual screening application to the computer-aided design of anticancer inhibitors with a favorable pharmacokinetic profile against e6 papillomavirus type 16. Universal Journal of Pharmaceutical Research. design of inhibitors. 2024.
[30] Revillo Imbernon J, Jacquemard C, Bret G, Marcou G, Kellenberger E. Comprehensive analysis of commercial fragment libraries. RSC Medicinal Chemistry, 2022, 13(3): 300–310.
[31] Bernard Djako A M, Adama N, Issouf F, Mathias M L, Mélalie K et Eugène M. Computer-assisted Design of Novel Polyketide Synthase 13 of Mycobacterium tuberculosis Inhibitors Using Molecular Modeling and Virtual Screening. Journal of Advances in Medical and Pharmaceutical Sciences, 2025, 27(6): 1-18.
[32] Davis J M, Smith L R. Advances in computational approaches for estimating passive permeability in drug discovery. Membranes, 2023, 13(11) 851.
Cite This Article
  • APA Style

    Yao, H. K., Abou, A., Djandé, A., Adama, N., Eugène, M., et al. (2025). Computer-aided Design of Coumarin Inhibitors of Quinone Oxidoreductase-1 (NQO1) with a Favorable Pharmacokinetic Profile. Science Journal of Chemistry, 13(4), 102-121. https://doi.org/10.11648/j.sjc.20251304.12

    Copy | Download

    ACS Style

    Yao, H. K.; Abou, A.; Djandé, A.; Adama, N.; Eugène, M., et al. Computer-aided Design of Coumarin Inhibitors of Quinone Oxidoreductase-1 (NQO1) with a Favorable Pharmacokinetic Profile. Sci. J. Chem. 2025, 13(4), 102-121. doi: 10.11648/j.sjc.20251304.12

    Copy | Download

    AMA Style

    Yao HK, Abou A, Djandé A, Adama N, Eugène M, et al. Computer-aided Design of Coumarin Inhibitors of Quinone Oxidoreductase-1 (NQO1) with a Favorable Pharmacokinetic Profile. Sci J Chem. 2025;13(4):102-121. doi: 10.11648/j.sjc.20251304.12

    Copy | Download

  • @article{10.11648/j.sjc.20251304.12,
      author = {Honoré Kouadio Yao and Akoun Abou and Abdoulaye Djandé and Niaré Adama and Megnassan Eugène and Issouf Soro},
      title = {Computer-aided Design of Coumarin Inhibitors of Quinone Oxidoreductase-1 (NQO1) with a Favorable Pharmacokinetic Profile
    },
      journal = {Science Journal of Chemistry},
      volume = {13},
      number = {4},
      pages = {102-121},
      doi = {10.11648/j.sjc.20251304.12},
      url = {https://doi.org/10.11648/j.sjc.20251304.12},
      eprint = {https://article.sciencepublishinggroup.com/pdf/10.11648.j.sjc.20251304.12},
      abstract = {Over the last ten years, cancer therapies have struggled with drug resistance. In this report, we explore new coumarin (COU) compounds designed to inhibit the enzyme NQO1, which shows potential for effective treatment due to their favorable predicted drug properties. Three-dimensional (3D) models of NQO1-COUx complexes were generated through in situ modifications of the crystal structure of NQO1-COU12 (PDB entry code: 3JSX), which served as the reference compound for a training set of of 22 and a validation set of 6 VCOUs with known experimental inhibitory potencies. To identify the active conformation of COU1-22, we developed a gas-phase quantitative structure-activity relationship (QSAR) model that established a linear correlation between the calculated enthalpy of NQO1-COU complex formation and the values of experimental activities. Subsequently, we screened the Virtual Compound Library (VCL) using Lipinski's Rule of Five and the PH4 model, then assessed the potency of the new COU analogues using the retained QSAR model. The pharmacokinetic profile of the analogues obtained was also evaluated using the linear correlation equation derived from the QSAR model. The coefficient of determination (R²), the Leave One Out (LOO) cross-validated Squared and the Standard error of regression σ for this equation are 0.91, 0.94 and 0.14, respectively, thus revealing the high predictive power of this model. Similarly, the PH4 model, with a correlation coefficient of 0.91, demonstrated robust predictive power. A comprehensive screening of the COU virtual analogue library yielded a total of 63 drug candidates with oral bioavailability, among which the most promising compounds exhibited a predicted potency of 12.22 and a favorable pharmacokinetic profile. The integration of Quantitative Structure-Activity Relationship (QSAR) techniques and in silico screening, based on the PH4 model, has enabled us to propose potent anticancer candidates with optimal pharmacokinetic profiles.},
     year = {2025}
    }
    

    Copy | Download

  • TY  - JOUR
    T1  - Computer-aided Design of Coumarin Inhibitors of Quinone Oxidoreductase-1 (NQO1) with a Favorable Pharmacokinetic Profile
    
    AU  - Honoré Kouadio Yao
    AU  - Akoun Abou
    AU  - Abdoulaye Djandé
    AU  - Niaré Adama
    AU  - Megnassan Eugène
    AU  - Issouf Soro
    Y1  - 2025/08/04
    PY  - 2025
    N1  - https://doi.org/10.11648/j.sjc.20251304.12
    DO  - 10.11648/j.sjc.20251304.12
    T2  - Science Journal of Chemistry
    JF  - Science Journal of Chemistry
    JO  - Science Journal of Chemistry
    SP  - 102
    EP  - 121
    PB  - Science Publishing Group
    SN  - 2330-099X
    UR  - https://doi.org/10.11648/j.sjc.20251304.12
    AB  - Over the last ten years, cancer therapies have struggled with drug resistance. In this report, we explore new coumarin (COU) compounds designed to inhibit the enzyme NQO1, which shows potential for effective treatment due to their favorable predicted drug properties. Three-dimensional (3D) models of NQO1-COUx complexes were generated through in situ modifications of the crystal structure of NQO1-COU12 (PDB entry code: 3JSX), which served as the reference compound for a training set of of 22 and a validation set of 6 VCOUs with known experimental inhibitory potencies. To identify the active conformation of COU1-22, we developed a gas-phase quantitative structure-activity relationship (QSAR) model that established a linear correlation between the calculated enthalpy of NQO1-COU complex formation and the values of experimental activities. Subsequently, we screened the Virtual Compound Library (VCL) using Lipinski's Rule of Five and the PH4 model, then assessed the potency of the new COU analogues using the retained QSAR model. The pharmacokinetic profile of the analogues obtained was also evaluated using the linear correlation equation derived from the QSAR model. The coefficient of determination (R²), the Leave One Out (LOO) cross-validated Squared and the Standard error of regression σ for this equation are 0.91, 0.94 and 0.14, respectively, thus revealing the high predictive power of this model. Similarly, the PH4 model, with a correlation coefficient of 0.91, demonstrated robust predictive power. A comprehensive screening of the COU virtual analogue library yielded a total of 63 drug candidates with oral bioavailability, among which the most promising compounds exhibited a predicted potency of 12.22 and a favorable pharmacokinetic profile. The integration of Quantitative Structure-Activity Relationship (QSAR) techniques and in silico screening, based on the PH4 model, has enabled us to propose potent anticancer candidates with optimal pharmacokinetic profiles.
    VL  - 13
    IS  - 4
    ER  - 

    Copy | Download

Author Information
  • Joint Research and Innovation Unit for Engineering Sciences and Techniques, (UMRI STI), Félix Houphouet-Boigny National Polytechnic Institute, Yamoussoukro, Côte d’Ivoire

  • Joint Research and Innovation Unit for Engineering Sciences and Techniques, (UMRI STI), Félix Houphouet-Boigny National Polytechnic Institute, Yamoussoukro, Côte d’Ivoire

  • Laboratory of Molecular Chemistry and Materials, University Joseph KI-ZERBO, Ouagadougou, Burkina Faso

  • Laboratory of Fundamental and Applied Physics, University of Nangui Abrogoua, Abidjan, Côte d’Ivoire

  • Laboratory of Fundamental and Applied Physics, University of Nangui Abrogoua, Abidjan, Côte d’Ivoire. Laboratory of Crystallography and Molecular Physics, University of Felix Houphouët-BOIGNY, Abidjan, Côte d’Ivoire. ICTP-UNESCO, QLS, Strada Costiera, Trieste, Italy

  • Laboratory of Fundamental and Applied Physics, University of Nangui Abrogoua, Abidjan, Côte d’Ivoire