VIRTUAL DESIGN OF NOVEL OF ORALLY BIOAVAILABLE PIPERAZINE INHIBITORS OF ENOYL-ACYL CARRIER PROTEIN REDUCTASE OF MYCOBACTERIUM TUBERCULOSIS WITH FAVORABLE PHARMACOKINETIC PROFILES

Koffi Charles Kouman1image, Affiba Florance Kouassi1imageYves Kily Hervé Fagnidi1,2image

Issouf Fofana1image, Koffi N’Guessan Placide Gabin Allangba1,3,4,5image, Mélalie Kéïtaépouse Guego1image

Eugene Megnassan1,6,7,8,9image

1Applied Fundamental Physics Laboratory (LPFA), Nangui Abrogoua University, Ivory Coast. 2Science and Technology Training and Research Unit, Alassane Ouattara University, Ivory Coast. 3Laboratoryof Environmental Sciences and technologies, Jean Lorougnon Guédé University, Ivory Coast. 4Laboratory of Biophysics and Nuclear Medicine (LBNM), Félix Houphouët-Boigny University, Ivory Coast. 5Department of Medical Physics, University of Trieste and International Centre for Theoretical Physics (ICTP), Trieste, Italy. 6Laboratory of Structural and Theoretical Organic Chemistry, Félix Houphouët-Boigny University, Ivory Coast. 7International Center for Theoretical Physics, ICTP-UNESCO, Coastal Road 11, I-34151 Trieste, Italy. 8International center for applied research and sustainable technology, SK-84104 Bratislava. 9Laboratory of Crystallography-Molecular Physics, Félix Houphouët-Boigny University, Ivory Coast.

 

Abstract

Background: During the previous decade, Anti-tuberculosis therapies were confronted with drug-resistant strains. We report here virtual design and evaluation of novel piperazine (PPZ) as inhibitors of InhA-Mt endowed with favorable predicted pharmacokinetic profiles.

Method: Three-dimensional (3D) models of InhA-PPZx complexes were prepared by in situ modifications of the crystal structure of InhA-PPZ1 (Protein Data Bank (PDB) entry code: 1P44), the reference compound of a training set of 12 PPZs with known experimental inhibitory potencies (IC50exp). First, in the search for active conformation of the PPZ 1-12, we built a gas phase quantitative structure-activity relationships (QSAR) model, linearly correlating the computed enthalpy of the InhA-PPZ complex formation and the pIC50exp. Finally, the VCL filtered by Lipinski’s rule-of-five was screened by the PH4 model and the potencies of the new PPZ analogs obtained were evaluated with the initial QSAR and their pharmacokinetic profile was evaluated.

Results: The VCL of 310,500 PPZs s was filtered down to 19,044 analogs by the Lipinski’s rule. The five-point PH4 screening retained 50 new potent PPZs with predicted inhibitory potencies IC50pre up to 100-times better than that of PPZ1 (IC50exp=160 nM). Predicted pharmacokinetic profile of the new analogs showed enhanced cell membrane permeability, side effect and high human oral absorption compared to current anti-tuberculosis candidates

Conclusions: Combining molecular modeling and PH4 in virtual screening of the VL resulted in the proposed novel potent antituberculotic agent candidates with favorable pharmacokinetic profiles.

Keywords: ADME, InhA inhibitors, Piperazine, pharmacophore, QSAR, tuberculosis, virtual screening.  

 

 

INTRODUCTION

 

Tuberculosis is as old as humanity itself. It has afflicted kings and queens, poets and politicians, revolutionaries and writers, activists and actors. Most of its victims, however, are poor, marginalised or malnourished, and the out-of-pocket costs associated with treating TB expose them to financial hardship or drive them further into poverty. TB is the definitive disease of deprivation1. Turning the tide on TB means screening and treatment for those it strikes, and preventing it by addressing its drivers and developing a new vaccine. Only by working together can we turn the tide against this ancient killer" is the position of the Dr. Tedros Adhanom Ghebreyesus, Director-General of World Health Organization according the 2024 WHO report2.

We can prevent and curate tuberculosis disease. However in 2023, TB probably returned to being the world’s leading cause of death from a single infectious agent, following 3 years in which it was replaced by coronavirus disease (COVID-19), and caused almost twice as many deaths as HIV/AIDS. More than 10 million people continue to fall ill with TB every year and the number has been rising since 20218. All member states of the United Nations (UN) and World Health Organization (WHO) adopted the urgency action to end the global tuberculosis epidemic by 20304. Despite the increasing worldwide incidence of TB and the threat for the public health, no novel antituberculotic drugs have been introduced into clinical practice over the past decades. Bedaquiline, a new antimycobacterial, approved at the end of 20122 inhibits adenosine 5′-triphosphate (ATP)-synthase of MTb with good clinical efficacy against multiple resistant strains. However, this drug has cardiovascular side effects3. It is therefore imperative to diversify the mycobacterial drug targets in order to fight the increasing incidence of drug-resistant strains of MTb. The oxidoreductase activity of enoyl-acyl carrier protein reductase (InhA or ENR) plays a key role in the type-II fatty-acid synthesis (FAS-II system) of MTb. This essential enzyme catalyzes the elongation cycle of biosynthesis of mycolic acid, which is a vital component of the mycobacterial cell wall4InhA represents a validated drug target of antituberculotic agents and has been indicated in the Special Programme for Research & Training in Tropical Diseases of WHO (TDR) targets database as an attractive pharmacological target for design of new drug candidates5,6. “The term totally resistant (TDR-TB) has emerged to mean infection with a strain resistant to all first- and second-line drugs”[i]Isoniazid prodrug (INH), is a first-line medication used for prevention and treatment of TB. The INH is a prodrug that must be activated by the bacterial catalase-peroxidase enzyme (KatG) which couples the isonicotinic acyl with the reduced form of nicotinamide adenine dinucleotide (NADH) to form an isonicotinic acyl-NADH complex. The complex binds tightly to InhA, blocks the natural substrate and hinders the action of fatty acid synthase, which hampers the synthesis of mycolicacid7. Compounds that can therefore directly inhibit InhA without the need for activation by KatG are of major interest in the fight against multidrug-resistant tuberculosis (MDR-TB), extensively drug-resistant tuberculosis (XDR-TB) and drug-resistant total tuberculosis (TDR-TB)”7. Several research groups were working on the discovery of InhA inhibitors not requiring KatG activation which were based on various scaffolds: triclosan9, diphenyl ether10,11, pyrrolidinecarboxamide11, arylamide deriva-tives11  benzamide derivatives with Tyr158 'out' conformation and interaction with the Phe41 and Arg43 pocket instead of the stacking with Phe977, thiadiazole-based InhA inhibitors9 and Piperazine derivatives with Tyr158 'out' conformation and interaction with NADH cofactor8. All displaying intermediate inhibitory potencies. These studies indicated that a potent InhA inhibitor should be a relatively long molecule, which binds to the InhA next to the NADH cofactor binding site. This inhibitor should also contain a bulky group that selectively fits into a hydrophobic pocket of InhA constituted by residues Met155, Pro193, Ile215, Leu217, Leu218 and Trp222 that is located near to a larger solvent accessible cavity15The main aim of this work was to design novel potent 1-(9H-fluoren-9-yl)-piperazine (PPZs) based on a series of 12 (training set) and 3 (validation set) nanomolar inhibitors with observed inhibitory potencies as low as IC50exp=160 nM16. Starting with in situ modification of the crystal structure of InhA-PPZ1 complex (PDB: 1P44) we have elaborated a QSAR model which correlated Gibbs free energies of InhA-PPZx complex formation with the potencies IC50exp and determined the active conformation of PPZs bound at the active site of InhA of Mt (MM-PB complexation approach). Based on this active conformation we have formulated 3D QSAR pharmacophore of InhA inhibition (PH4). Large virtual library of compounds sharing the PPZ scaffold has been generated and in silico screened with the PH4. The screening yielded virtual hits that exhibited predicted inhibitory potencies IC50pre more than 100 times lower than the most active training set compound PPZ1. Finally, the hits underwent complexation simulations for evaluation of the predicted inhibitory activity for the best analogues and for calculation of their ADMET profile. 

 

MATERIAL AND METHODS

 

Structural studies and bioassays (IC50exp) of our studied piperazines derivatives (PPZ) InhA inhibitors were taken from literature17. The efficacy range of inhibitory concentrations (160≤ IC50exp ≤ 51000 nM), allows us to realize QSAR models. The whole series of 15PPZs were divided into a training (TS) and a validation (VS) sets of 12 and 3PPZs respectively18.

Model building

The whole complex (E:I), with free InhA (E) and inhibitor (I) was resolved to a reliability factor of 1.7 Å containing the Genz-10850  (PPZ1) bound to InhA (Whose crystallographic data entry code 1P4412,13 from Discovery Studio 2.5 software8. Virtual design plan to result in new PPZ analogs with higher predicted activity is presented in scheme1. The structures (E and E:I complexes) were at the neutral pH=7 and neutral N- and C-terminal residues, all protonizable and ionizableaminoacids being charged, without any crystallographic water molecules. The inhibitors were built into the 1P44 structure by in situ replacing derivative groups of the PPZ1 moiety followed by systematic conformational search of the replacing group coupled with a careful energy minimization of the modified inhibitor and surrounding InhA active site residues20-35.

Molecular mechanics

Modeling of inhibitors, InhA, and E-I complexes was carried out by molecular mechanics using CFF97 force field27 as described earlier28.

Conformational Search

For conformational research we recommend reading the following articles18,21.

Solvation Gibbs free energies

As for the free solvation energy has been described perfectly by the following articles24.

Calculation of Binding Affinity and QSAR Model

The calculation of binding affinity expressed as complexation GFE has been described fully earlier25.

Interaction energy

For interaction energy refer to the full description we reported formerly28.

Pharmacophore generation

Bound conformations of inhibitors taken from the models of E-I complexes were used for constructing of 3D-QSAR pharmacophore (PH4) by means of Catalyst HypoGen algorithm31 implemented in Discovery Studio32 as described earlier26.

Virtual library generation

The virtual library generation was performed as described earlier18.

ADME properties

The drug-likeness selection criterion served to focus the initial virtual library as described earlier17.

Pharmacophore-based library searching

The pharmacophore model (PH4) described in Section “Pharmacophore generation” and derived from the bound conformations of PPZs at the active site of InhA served as library searching tool as described earlier22.

Inhibitory potency prediction

The conformer with the best mapping on the PH4 pharmacophore in each cluster of the focused library subset was used for image calculation and IC50 preestimation (virtual screening) by the complexation QSAR model as described earlier21.

 

RESULTS

 

Training and validation sets

The training set of 12 PPZs and validation set of another 3 analogs (Table 1) were selected from a heterogeneous series of InhA inhibitors with known experimentally determined inhibitory activities originating from a single laboratory23. First, 1-(9H-fluoren-9-yl) -piperazine derivatives were synthesized bearing modifications around the carbonyl hydrogen bond acceptor. Using 1-(9H-fluoren-9-yl)-piperazine (3), compounds 2 and 4 - 8 were synthesized in low to good yields. Further, direct sulfonylation was used for the synthesis of benzenesulfonyl derivatives 5a - b to obtain the desired products25. The whole series was obtained by variations at five positions of the substituents on the phenyl rings in Table 1. The experimental half-maximal inhibitory concentrations (160 ≤ IC50exp ≤ 51000 nM)28 cover a sufficiently wide concentration range for the construction of a reliable QSAR model.

One descriptor QSAR models

Each of the 12training set (TS) and 3 validation set (VS) InhA- PPZx complexes (Table 1), was prepared by in situ modification of the refined template crystal structure (PDB entry code 1P4422 of the complex InhA-PPZ1 as described in the Methods section. Further the relative Gibbs free energy of the InhA-PPZx complex formation (∆∆Gcom) was computed for each of the 15 optimized enzyme-inhibitor complexes. Table 2 lists computed values of ∆∆Gcom and its components as defined earlier25 for the TS and VS of piperazine27. The QSAR model explained variation in the PPZs experimental inhibitory potencies (pIC50exp=–log10(IC50exp)28 by correlating it with computed GFE ∆∆Gcom through linear regression (eq. B), Table 2. In search for a better insight into the binding affinity of PPZs towards MtInhA, the enthalpy of complexation in gas phase ∆∆HMM was analyzed by correlating it with the pIC50exp. The validity of this linear correlation (for statistical data of the regression see Table 3, eq. A) allowed assessment of the significance of inhibitor-enzyme interactions (∆∆HMM) without considering solvent effect and loss of entropy of the inhibitor upon binding to the enzyme. This correlation explained about 88% of the pIC50exp data variation and showed the role not negligible of the enthalpic contribution to the binding affinity of the ligand to active site. Likewise, the more advanced descriptor, namely the GFE of the InhA-PPZx complex formation containing all components: ∆∆HMM, ∆∆TSvib and ∆∆Gsol, has been evaluated (see Table 3 for statistical data, eq. B). Certainly the strong relationship between the 3D model of inhibitor binding and the observed inhibitory potencies of the PPZs was explained by the relatively high values of the regression coefficient R2, leave-one-out cross-validated regression coefficient R2xv and Fischer F-test of the correlation suggest29.Therefore, the active bound conformation of the PPZs at the InhA binding site enabled definition of the PH4 pharmacophore and structural information derived from the 3D models of InhA-PPZx complexes can be expected to lead to reliable prediction of InhA inhibitory potencies for new PPZs analogs based on the QSAR model B, Table 3.

The statistical data confirmed validity of the correlation equations (A) and (B) plotted on Figure 1. The ratio pIC50pre/pIC50exp 1 (the pIC50pre values were estimated using correlation eq. B, Table 3) calculated for the validation set PPV13-15 documents the substantial predictive power of the complexation QSAR model from Table 2. Thus, the regression equation B (Table 3) and computed ∆∆Gcom GFEs can be used for prediction of inhibitory potencies IC50pre against MtInhA for novel PPZ analogs, provided that they share the same binding mode as the training set piperazine PPZ1-12.

Binding mode of PPZs

In the crystal structure of InhA –PPZ123 the substitution at R-groups of the piperazine derivative scaffold of the inhibitor sits in a hydrophobic cavity of the active-site surrounded by side chains of predominantly nonpolar residues: Pro 99, Gly 104, Met103, Tyr158, Phe149 and Met161, Met 98, Ala 198. According to the structures of PPZ8 and PPZ5 studied by docking simulations, the presence of the methyl group at the C2 position ofthe aryl moiety causes steric hindrance due to the restricted space in the cavity formed by Met103, Tyr158, and Met161 residues of MtInhA. In contrast to the recently reported methyl thiazoles that interact with MtInhA in a “Tyr158-out” binding mode direct inhibitors such as pyrrolidine carboxamides8 and piperazine-indole derivatives7 have explored polar interactions involving a ribose hydroxyl, the Tyr158 hydroxyl and a hydrogen bond acceptor in the compounds.

Interaction Energy

Another key structural information was provided by the interaction energy (IE, ΔEint) diagram obtained for four training set inhibitor. IE breakdown to contributions from InhA active site residue is helpful for the choice of relevant R-groups which could improve the binding affinity of PPZ analogs to the MtInhA and the subsequently enhance the inhibitory potency. A comparative analysis of computed IE for training set PPZs (Figure 3) divided into three classes,(highest (PPZ1), moderate (PPZ5 and PPZ8) and lowest (PPZ12) activity) has been carried out to identify the residues for which the contribution to binding affinity could be increased. However, the comparative analysis showed about the same level of IE contributions from active site residues for all three classes of inhibitors.

Therefore, no suggestions of suitable substitutions able to improve the binding affinity as we previously reported for thymine-like inhibitors of Mt thymidine monophosphate kinase design, could be proposed37. Since specific substitutions could not be proposed we have adopted a combinatorial approach to novel PPZ analogs design and in silico screened a virtual library of 310,500PPZanalogs with help of the PH4 pharmacophore of InhA inhibition derived from the complexation QSAR model. As we can see from the IE analysis (Figure 3) the TS and VS piperazine36 do not show significant interaction energies with the residues of hydrophobic pocket surrounding aromatic ring.

3D-QSAR Pharmacophore Model

The interaction generation protocol in Discovery Studio molecular modeling program38 provides the pharmacophore features of the active site of a protein. InhA predominantly displays hydrophobic features at the active site39. InhA substrate-competitive inhibitors design often exploits the pocket flexibility because of the high mobility of the Tyr158, Phe149 side chains and the substrate-binding loop (Thr196-Gly208)40

Generation and validation of 3D-QSAR pharmacophore

InhA inhibition 3D-QSAR pharmacophore was generated from the active conformation of 12 TS PPZ1-12 and assessed by 3 VS PPV13-15 covering a large range of experimental activity (160 - 51000 nM) spanning more than two orders of magnitude. The generation process was carried out into three main steps: constructive, subtractive and optimization39 as described earlier40. During the constructive phase, PPZ1 alone was retained as the lead because only the activity of PPZ1 fulfilled the threshold criterion (IC50exp ≤ 5/4 × 160 nM) and used to generate the starting PH4 feature. In the of the threshold criterion subtractive phase, compounds for which: IC50exp > 160 × 103.5 nM =505 968 nM were considered inactive. Considering this criterion, none of the training set PPZx was inactive and no starting PH4 features were removed. Finally, during the optimization phase the score of the pharmacophoric hypotheses was improved. Hypotheses were scored according to errors in activity estimates from regression and complexity via a simulated annealing approach. At the end of the optimization, the top scoring 10 of pharmacophore hypotheses were kept, all displaying five points features.To be statistically significant a hypothesis has to be as close as possible to the fixed cost and as far as possible from the null cost. For the set of 10 hypotheses the difference ∆ ≥ 506.68, which attests high quality of the pharmacophore model. The standard indicators such as the RMSD between the hypotheses ranged from 1.75 to 2.60and the squared correlation coefficient (R2) falls to an interval from 0.98 to 0.96. The first PH4 hypothesis with the closest cost (51.9) to the fixed one (32.01), best RMSD and R2 was retained for further analysis. The statistical data for the set of hypotheses (costs, RMSD, R2) are listed in Table 4. The configuration cost (9.65 for all hypotheses) far below 17 confirms this pharmacophore as a reasonable one.The regression equation for pKiexp vs. pKipre estimated from Hypo1: image=0.9961 × image+ 0,0226 (n=12. R2=0.97. R2xv=0.96. F-test=294. σ=0.18, α > 98%) is also plotted on Figure 4 (E). Therefore, the PH4 is good potentially to choice the new PPZ analogs. These parameters are in accordance with the OECD QSAR guidelines5. To assess the predictive power of the pharmacophore model, we calculated the ratio between the activities predicted by the PH4 model and those observed experimentally (pIC50pre/pIC50exp) for the validation set (PPV13-15). The following ratios were obtained PPV13: 0.95, PPV14: 1.08 and PPV15: 1.07.

All ratios were close to one, demonstrating the strong predictive power of this regression for the optimal PH4 model. Another evaluation of hypothesis 1 is the mapping of the PH4 binding mode in the 3D QSAR (Figure 4) of the most active PPZ1. We can carry out computational design and selection of new PPZ analogs with elevated inhibitory potencies against MtInhA based on a strategy using the noticeable presence of the hydrophobic features included in the best pharmacophore model.

Virtual Screnning

In silico screening of a virtual (combinatorial) library can lead to hit identification as it was shown in our previous works on inhibitors design38.

   Virtual library

An initial virtual library (VL) was generated by substitutions small fragments (R1 to R5) on the aromatic ring of the PPZ scaffold based on structural information gathered39 (Table 5). During the virtual library enumeration all R-groups listed in Table 5 were attached to in positions R1 to R5 of the PPZ scaffold to form a combinatorial library of the size: 

R1 × R2 ×R3×R4×R5= 6×23×30×15×5=310,500 analogs.

This initial diversity library was generated from building blocks (chemicals) listed in the databases of available chemicals[i]. To design a more focused library of a reduced size and increased content of drug-like molecules, we have introduced a set of filters and penalties such as the Lipinski rule-of-five, which helped to select smaller number of suitable PPZs that could be submitted to in silico screening. This focusing has reduced the size of the initial library to 19,044 analogs, 6% of its original number size.

Virtual screening of library of PPZs

The focused library of 19044 analogs was further screened for molecular structures matching the 3D-QSAR PH4 pharmacophore model Hypo1 of InhA inhibition. 60 PPZs mapped to at least 4 pharmacophoric features, 50 of which mapped to at least 5 features of the pharmacophore. These best fitting analogs (PH4 hits) then underwent complexation QSAR model screening. 

The computed GFE of InhA-PPZx complex formation, their components and predicted half-maximal inhibitory concentrations IC50pre calculated from the correlation equation B (Table 3), are listed in Table 6.

Analysis of novel PPZs inhibitors

The design of virtual library of novel analogs was guided by structural information retrieved from the PPZx active conformation and was used for the selection of appropriate substituents at position R1, R2, R3, R4 and R5). In order to identify which substituents, lead to new inhibitor candidates with the highest predicted potencies towards the InhA of Mt, we have prepared histograms of the frequency of occurrence of R1, R2, R3, R4 and R5 among the 50 best fit PH4 hits (Figure 6). The histograms show that the R1groups 1 and 4 were represented respectively with the highest frequency of occurrence (10) and (27) among the 50 hits; the R2 groups:  12 (5); 22 (6); 20 (11); R3 groups :  7, 29, (6) and 21 (9); R4 groups : 1 (5) and 2, 7 (4) and 12, 13 (6) and 8 (7);  R5 groups : 1(12) and 2(7) and 3 (30). The top ten scoring virtual hits namely analogs are: 1-21-7-2-4 (IC50pre=2100 pM), 2-12-7-12-4 (4510 pM), 2-20-29-7-4 (360 pM), 4-1-7-9-4 (1010  pM), 4-8-8-1-4 (910 pM), 4-9-6-8-4 (700 pM), 4-8-27-12-1 (4460 pM), 4-6-29-7-4 (1130 pM), 4-15-7-6-1(6980 pM) and 4-20-9-13-4 (7810 pM). These analogues include mostly the following substituents at R1 position: 1, 2, 4; R2 position: 21, 12, 20, 18, 9, 6, 15; R3 position : 7, 29, 8, 6, 27, 7, 9; R4 groups : 2, 12, 7, 9, 1, 8, 6, 13; R5  groups :  4, 1.

Due to amino acid composition of the larger hydrophobic pocket, all the R-groups display preferences for shorter aliphatic building blocks as showed the Table 5. 

The substitutions in R1 to R5 positions of PPZs led to an overall increase of affinity of InhA binding as exemplified by the inhibitory potencies of majority of new designed analogs. The best designed benzamide PPZ 2-20-29-7-4 displays predicted half-minimal inhibitory concentration of IC50pre=360 pM that is more than 100-times lower than that of the most active compound of the TS, namely the PPZ1 with IC50exp=160 nM, Figure 5.

ADME profiles of designed PPZs

Pharmacokinetic profile obtained of InhA inhibitors still requires increased research. Presented in Table 7, the ADME of our new analogs, were described earlier by the QikProp program24 taken from the method of Jorgensen8. The fundamental principles of this method are described previously39. Our best analogs are compared with that of drugs used on the market to treat tuberculosis disease (Table 7).

 

DISCUSSION

 

A training set of 12 PPZs and validation set of 3 PPZs (Table1) were selected from a heterogeneous   series of InhA inhibitors for which experimentally determined inhibitory activities were available from a single laboratory35. Both were obtained by synthesize from the 1-(9H-fluoren-9-yl)-piperazine. The first one is from the synthesis bearing modifications around the carbonyl hydrogen bond acceptor using 1-(9H-fluoren-9-yl)-piperazine and the second group were from direct sulfonylation. The whole series was obtained by substitutions at five positions of the aromatic ring of the other side of the sulfonyl and carbonyl surrounded by hydrophobic residues constituted of residues constituted of residues Pro 99, Gly 104, Met 103, Tyr 158, Phe 149 and Met 161, Met 98, Ala 198 of Genz-10850 (PPZ1) as shown in Table1. Their experimental inhibitory concentrations IC50exp30 cover a concentration range sufficiently wide to serve well for building of a reliable QSAR model of InhA inhibition. Accord to the PPZ8 According to the structures of PPZ8 and PPZ5 studied, the presence of the methyl group at the C2 position of the aryl moiety causes steric hindrance due to the restricted space in the cavity formed by Met103, Tyr158, and Met161 residues of MtInhA41. In order to identify structural modifications of the aromatic ring leading to increased binding affinity of PPZs to InhA of MTb we have carried out detailed analysis of interactions in a series of InhA-PPZs complexes with help of the complexation QSAR model. The first step of this analysis aimed at obtaining insight into InhA active-site interactions by performing the interaction energy breakdown into contributions from individual residues filling the hydrophobic pocket displayed on Figure 2 for the most active inhibitor PPZ1, moderate active (PPZ5 and PPZ8) and the lowest (PPZ12) activity, Table 243. Figure 3 showed that there no too difference concerning the interaction Energy (IE) between the three classes of activity. To obtain the best analogs PPZ we proceed by a combinatory library. As the pocket containing the R1 groups and formed by Met103, Tyr158, and Met161is too small we put their short substituents. We noticed that the analogs dominated by small fragment like halogenure and hydroxyd have the most frequency of occurrence. The convenable size of the fragment chosed lead to obtain the best analogs considerably most actif than the most active of the training and the validation sets. The most four virtual active analogs were PPZ 2-20-29-7-4 (IC50pre=360 pM), PPZ 4-9-6-8-4(IC50pre=700 pM), PPZ 4-8-8-1-4 (IC50pre= 910 pM) and PPZ 4-1-7-9-4 (IC50pre=1010 pM) with favorable predicted pharmacokinetic profile than the older currently used drugs. We can suggest them for synthesis.

 

CONCLUSIONS AND RECOMMANDATION

 

In this work the crystallographic structure of the InhA-PPZ1 (1P44) complex and the structural properties of the Piperazine derivatives identified by MarianeRotta et al.22 as a potential antituberculosis agent and whose target is InhA enabled us to develop a QSAR complexation model capable of explaining more than 92% of the variation in the experimental inhibitory activity of Piperazine derivatives by the Gibbs free energy of formation of the InhA-PPZx complex. Following this QSAR model, we obtained a 3D-QSAR PH4 pharmacophore model using a training set of 12 PPZs and a validation set of 3 PPZs with known inhibitory activities42. The visual analysis and calculation of the interactions between InhA and PPZs in the active site of the enzyme guided us in the design of a virtual combinatorial library of new PPZ analogs with a substitution on the scaffold at the position R1 to R5 on the aromatic ring. The virtual library obtained was first focused by considering Lipinski's five rule and then screened by the 3D QSAR pharmacophore identified during chemical space exploration around R1to R5 positions novel PPZs analogsby the QSAR complexation modelwith predicted picomolarMtInhA inhibitory potencies PPZ 2-20-29-7-4 (IC50pre=360  pM), PPZ 4-9-6-8-4(IC50pre=700 pM), PPZ 4-8-8-1-4 (IC50pre=910 pM) and PPZ 4-1-7-9-4 (IC50pre=1010 pM) all display also favorable pharmacokinetic profiles compared to current antituberculotics. We believe that they are worth synthesizing and evaluating.

 

ACKNOWLEDGEMENTS

 

The authors would like to thank the Laboratory of Fundamental and Applied Physics at NANGUI ABROGOUA University, in Côte d’Ivoire, for providing the facilities necessary for this work.

 

AUTHOR’S CONTRIBUTION

 

Kouman C: performed the complexation study, PH4 pharmacophore generation, interaction energy analysis, the PH4-based VL searching and the analogues evaluation. Kouassi F: established the first preliminary complexation model in order to confirm the feasibility of this work. Fagnidi H:performed the VL generation and focusing and wrote original draft, methodology, investigation. Kéita M: editing, review. Fofana I: performed the VL generation and focusing. Allangba G: formal analysis. Megnassan E: writing, review, and editing, data curation. All authors read and approved the final manuscript for publication.

 

DATA AVAILABILITY

 

Data will be available on request to anyone from the correspondence author.

 

CONFLICT OF INTERESTS

 

The authors declared no conflict of interests

 

REFERENCES

 

  1. Global tuberculosis report 2024. Geneva: World Health Organization; 2024. Licence: CC BY-NC-SA 3.0 IGO
  2. Koul A, Arnoult E, Lounis N, Guillemont J, Andries K. The challenge of new drug discovery for tuberculosis. Nature. 2011; 469(7331):483−90.https://doi.org/10.1038/nature09657
  1. Palomino JC, Martin A. TMC207 becomes bedaquiline, a new anti-TB drug. Future Microbiol. 2013;8(9):1071−80. https://doi.org/10.2217/fmb.13.85
  2. Vilcheze, C.; Morbidoni, H.R.; Weisbrod, T.R.; et al. Inactivation of the InhA-encoded fatty acid synthase II (FASII) enoyl-acyl carrier protein reductase induces accumulation of the FASI end products and cell lysis of Mycobacterium smegmatis. J. Bacteriol. 2000, 182, 4059–4067. https://doi.org/10.1128/JB.182.14.4059-4067.2000
  3. Aguero F, Al-Lazikani B, Aslett M, et al. Genomic-scale prioritization of drug targets: The TDR Targets database. Nat Rev Drug Discov 2008, 7, 900–907.https://doi.org/10.1038/nrd2684
  1. Campbell JW, Cronan JE, Jr. Bacterial fatty acid biosynthesis: Targets for antibacterial drug discovery. Annu Rev Microbiol 2001, 55, 305–332 https://doi.org/10.1146/annurev.micro.55.1.305
  1. Udwadia ZF, Amale RA, AjbaniKK, Rodrigues C. Totally drug-resistant tuberculosis in India. Clin Infect Dis 2012;54(4):579−81.https://doi.org/10.1093/cid/cir889
  1. Timmins, G.S.; Deretic, V. Mechanisms of action of isoniazid.Mol. Microbiol. 2006, 62, 1220–1227.https://doi.org/10.1111/j.1365-2958.2006.05467.x
  1. Freundlich JS, Wang F, Vilcheze C, et al. Triclosan derivatives: towards potent inhibitors of drug-sensitive and drug resistant Mycobacterium tuberculosis. Chem Med Chem 2009;4(2):241−48. https://doi.org/10.1002/cmdc.200800261
  1. Am Ende CW, Knudson SE, Liu N, et al. Synthesis and in vitro antimycobacterial activity of B-ring modified diaryl ether InhA inhibitors. Bioorg Med Chem Lett 2008, 18, 3029–3033. https://doi.org/10.1016/j.bmcl.2008.04.038
  1. Luckner SR, Liu N, Am Ende CW, et al. A slow, tight binding inhibitor of InhA, the enoyl-acyl carrier protein reductase from Mycobacterium tuberculosis. J Biol Chem 2010; 285: 14330–14337. https://doi.org/10.1074/jbc.M109.090373
  1. He X, Alian A, Stroud R, de Montellano PR. Pyrrolidinecarboxamides as a novel class of inhibitors of enoyl acyl carrier protein reductase from Mycobacterium tuberculosis. J Med Chem 2006; 49: 6308–6323. https://doi.org/10.1021/jm060715y
  1. He X, Alian A, de Montellano P.R. Inhibition of the Mycobacterium tuberculosis enoyl acyl carrier protein reductase InhA by arylamides. Bioorg Med Chem 2007; 15: 6649–6658. https://doi.org/10.1016/j.bmc.2007.08.013
  2. Guardia A, Gulten G, Fernandez R, et al. N-Benzyl-4-((heteroaryl)methyl)benzamides: A new class of direct NADH-dependent 2-trans enoyl–acyl carrier protein reductase (InhA) inhibitors with antitubercular activity. Chem Med Chem 2016; 11: 687 – 701. https://doi.org/10.1002/cmdc.201600020
  1. Shirude PS, Madhavapeddi P, Naik M, et al. Methyl-thiazoles: A novel mode of inhibition with the potential to develop novel inhibitors targeting InhA in Mycobacterium tuberculosis. J Med Chem 2013;56(21):8533−42. https://doi.org/10.1021/jm4012033
  1. Rotta M, Timmers M, Pissinate K, et al. Piperazine derivatives: Synthesis, inhibition of the Mycobacterium tuberculosis enoyl-acyl carrier protein reductase and SAR studies. European J Med Chem 2015; 90:436-447. https://doi.org/10.1016/j.ejmech.2014.11.034
  1. Punkvang A, Kamsri P, Kumkong A, et al. The structural requirement of direct InhA inhibitors for high potency against M. tuberculosis based on computer aided molecular design, Science against microbial pathogens: communicating current research and technological advances, A. Mendez-Vilas (Ed.), Microbiology Book Series No. 3, Formatex Research Center, Badajos, Spain, 2011, pp. 160-168.
  2. Berman HM, Westbrook J, Feng Z, Gilliland G, Bhat TN, Wessig H, Shindyalov IN, Bourne PE. The protein data bank.Nucl. Acids Res 2000; 28:235-242. https://doi.org/10.1107/s0907444902003451 
  1. Discovery Studio molecular modeling and simulation program version 2.5, Accelrys, Inc., San Diego, CA, 92121, USA, 2009
  2. OwonoOwono LC, Keita M, Megnassan E, Frecer V, Miertus S. Design of thymidine analogues targeting thymidilate kinase of Mycobacterium tuberculosis. Tuberculosis Res Treat 2013;670836. https://doi.org/10.1155/2013/670 836
  1. Frecer V, Miertus S, Tossi A, Romeo D. Rational design of inhibitors for drug-resistant HIV-1 aspartic protease mutants. Drug Des Discov 1998;15(4):211-231
  2. Frecer V, Miertus S. Interactions of ligands with macromole-cules: rational design of specific inhibitors of aspartic protea-se of HIV-1. Macromol Chem Phys 2002;203:1650–1657. https://doi.org/10.1002/15213935
  3. Frecer V, Berti F, Benedetti F, Miertus S. Design of peptidomimetic inhibitors of aspartic protease of HIV-1 containing -Phe Psi Pro- core and displaying favourable ADME-related properties. J Mol Graph Model 2008;27 (3):376-387. https://doi.org/10.1016/j.jmgm.2008.06.006
  4. Dali B, Keita M, Megnassan E, Frecer V, Miertus S. Insight into selectivity of peptidomimetic inhibitors with modified statine core for plasmepsin II of Plasmodium falciparum over human cathepsin D. Chem Biol Drug Des. 2012;79 (4):411-430. https://doi.org/10.1111/j.17470285.2011.01276.x
  1. Megnassan E, Keita M, Bieri C, Esmel A, Frecer V, Miertus S. Design of novel dihydroxynaphthoic acid inhibitors of Plasmodium falciparum Lactate Dehydrogenase. Med Chem 2012, 8, 970-984. https://doi.org/10.2174/157340612802084324
  1. Keita M, Kumar A, Dali B, et al. Quantitative structure-activity relationships and design of thymine-like inhibitors of thymidine monophosphate kinase of Mycobacterium tuberculosis with favourable pharmacokinetic profiles. RSC Adv. 2014, 4, 55853-55866. https://doi.org/10.1039/c4ra06917j
  1. Owono Owono LC, Ntie-Kang F, Keita M, et al. Virtually designed triclosan-based inhibitors of enoyl-acyl carrier protein reductase of Mycobacterium tuberculosis and of Plasmodium falciparum. Mol Inform 2015, 34, 292–307. https://doi.org/10.1002/minf.201400141
  1. Kouassi AF, Kone M, Keita M, et al. Computer-aided design of orally bioavailable pyrrolidinecarboxamide inhibitors of Enoyl-Acyl carrier protein reductase of Mycobacterium tuberculosis with favorable pharmacokinetic profiles. Int J Mol Sci 2015, 16, 29744-29771. https://doi.org/10.3390/ijms161226196
  2. Allangba KNPG, Keita M, Frecer V, et al. Virtual design of novel Plasmodium falciparum cysteine protease falcipain-2 hybrid lactone-chalcone and isatin-chalcone inhibitors probing the S2 active site pocket. J Enz Inhib Med Chem 2018; 34: 547-561. https://doi.org/10.1080/14756366.2018.1564288
  1. Kouman KC, Keita M, N’Guessan KR, et al. Structure-based design andin silico screening of virtual combinatorial library of benzamides inhibiting 2-transEnoyl-acyl carrier protein reductase of Mycobacterium tuberculosis with favourable predicted pharmacokinetic profiles. Int J Mol Sci 2019; 20:4730. https://doi.org/10.3390/ijms20194 730
  1. N’Guessan H, Soro I, Keita M, Megnassan E. Design and in silico screening of combinatorial library of new herbicidalanalogs of cycloalka[d]quinazoline-2,4dione-benzoxazinones inhibiting protoporphyrino-gen ix oxidase. J Pharm Res Int 2022;34(56):42–61. https://doi.org/10.9734/jpri/2022/v34i567251
  1. Djako B, Keita M, Bisseyou Y, Esmel A, Megnassan E. Computer-assisted design of novel polyketide synthase 13 of mycobacterium tuberculosis inhibitors using molecular modeling and virtual screening. J Pharm Res Int 2022;34(56):12- 41. https://doi.org/10.9734/jpri/2022/v34i567250
  1. Bieri C, Esmel A, Keita M, et al. Structure-based design and pharmacophore-based virtual screening of combinatorial library of triclosan analogues active against enoyl-acyl carrier protein reductase of Plasmodium falciparum with favourable ADME profiles. Int J Mol Sci 2023;24,(8): 6916. https://doi.org/10.3390/ijms24086916
  1. Kone M, N’Guessan H, N’Gouan AJ, MKoblavi F, Megnassan E. Computer-aied design of new hydroxamic acid derivatives targeting the Plasmodium falciparum M17 metallo-aminopeptidase with favorable pharmcokinetic profile. Int J Pharm Sci Drug Res 2023;15(3):356-375. https://doi.org/10.25004/IJPSDR.2023.150317
  2. Ziki E, Akonan L, Kouman KC, et al. Virtual design of novel coumarinyl substituted sulfonamide inhibitors of carbonic anhydrase II as potential drugs against glaucoma. J Pharm Res Int 2023; 35(24): 10-33. https://doi.org/10.9734/JPRI/2023/v35i247424
  1. Saura J, Kettler R, Da Prada M, Richards JG. Quantitative enzyme radioautography with 3H-Ro 41-I 049 and 3H-Ro 19-6327 in vitro: Localization and abundance of MAOA and MAO-B in rat CNS, peripheral organs, and human brain. J. Neurosci 1992; 12(5):1977-1999. https://doi.org/10.1523/JNEUR OSCI.12-0501977.1992
  1. Lee J, Natalie B, Drinkwater N, et al. Novel Human Aminopeptidase N Inhibitors: Discovery and optimization of subsite binding interactions. J Med. Chem 2019; 62(15): 7185‑7209. https://doi:10.1021/acs.jmedchem.9b00757
  1. He X, Alian A, Stroud R, Ortiz de Montellano PR. Pyrrolidine carboxamides as a novel class of inhibitors of enoyl acyl carrier protein reductase from Mycobacterium tuberculosis. J Med Chem 49 (21) (2006) 6308 – 6323.  https://doi.org/10.1021/jm060715y 
  1. Kuo MR, Morbidoni HR, Alland D, et al. Targeting tuberculosis and malaria through inhibition of enoylreductase: compound activity and structural data, J Biol Chem 2003; 278 (23):20851e20859. https://doi.org/10.1074/jbc.M211968200
  1. Sullivan TJ, Truglio JJ, Boyne ME, et al. High affinity InhA inhibitors with activity against drug-resistant strains of Mycobacterium tuberculosis. ACS Chem Biol 2006; 1: 43−53. https://doi.org/10.1021/cb0500042
  2. OECD (2014), Guidance Document on the Validation of (Quantitative) Structure-Activity Relationship [(Q)SAR] Models, OECD Series on Testing and Assessment, No. 69, OECD Publishing, Paris. https://doi.org/10.1787/9789264085442-en
  1. Available Chemicals Directory, Version 95.1, MDL Information Systems, San Leandro, CA
  2. Lipinski CA, Lombardo F, Dominy BW, Feeney PJ. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv Drug Deliv Rev 2001; 46, 3–26. https://doi.org/10.1016/S0169-409X(00)00129-0
  3. Qik Prop, version 3.7, release 14; XSchrödinger, LLC: New York, NY; 2014
  4. Jorgensen WL, Duffy EM. Prediction of drug solubility from montecarlo simulations. Bioorg Med Chem Let. 2000;10:1155-1158 https://doi.org/10.1016/S0960-894X(00)00172-4