Next Article in Journal
Theoretical Insights into the Electron Capture Behavior of H2SO4···N2O Complex: A DFT and Molecular Dynamics Study
Next Article in Special Issue
Anti-Hyperuricemic Effect of 2-Hydroxy-4-methoxy-benzophenone-5-sulfonic Acid in Hyperuricemic Mice through XOD
Previous Article in Journal
Microfluidic-Based Single-Cell Study: Current Status and Future Perspective
Previous Article in Special Issue
Novel Group of AChE Reactivators—Synthesis, In Vitro Reactivation and Molecular Docking Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

QSAR Study of N-Myristoyltransferase Inhibitors of Antimalarial Agents

by
Letícia Santos-Garcia
1,
Marco Antônio De Mecenas Filho
1,
Kamil Musilek
2,3,
Kamil Kuca
2,*,
Teodorico Castro Ramalho
1,4 and
Elaine Fontes Ferreira Da Cunha
1,*
1
Departamento de Química, Universidade Federal de Lavras, Lavras 37200-000, Brazil
2
Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove 50005, Czech Republic
3
Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove 50005, Czech Republic
4
Center for Basic and Applied Research, University Hradec Kralove, Hradec Kralove 50005, Czech Republic
*
Authors to whom correspondence should be addressed.
Molecules 2018, 23(9), 2348; https://doi.org/10.3390/molecules23092348
Submission received: 12 August 2018 / Revised: 6 September 2018 / Accepted: 12 September 2018 / Published: 13 September 2018
(This article belongs to the Special Issue QSAR and QSPR: Recent Developments and Applications)

Abstract

:
Malaria is a disease caused by protozoan parasites of the genus Plasmodium that affects millions of people worldwide. In recent years there have been parasite resistances to several drugs, including the first-line antimalarial treatment. With the aim of proposing new drugs candidates for the treatment of disease, Quantitative Structure–Activity Relationship (QSAR) methodology was applied to 83 N-myristoyltransferase inhibitors, synthesized by Leatherbarrow et al. The QSAR models were developed using 63 compounds, the training set, and externally validated using 20 compounds, the test set. Ten different alignments for the two test sets were tested and the models were generated by the technique that combines genetic algorithms and partial least squares. The best model shows r2 = 0.757, q2adjusted = 0.634, R2pred = 0.746, R2m = 0.716, ∆R2m = 0.133, R2p = 0.609, and R2r = 0.110. This work suggested a good correlation with the experimental results and allows the design of new potent N-myristoyltransferase inhibitors.

Graphical Abstract

1. Introduction

Malaria is a mosquito-borne protozoal infection caused by five parasites of the genus Plasmodium: P. falciparum, P. ovale, P. malariae, P. vivax, and P. knowlesi [1]. Among these, Plasmodium falciparum is the most prevalent and lethal [2]. Over the past 50 years, the parasite resistance to chloroquine and sulphadoxine-pyrimethamine in endemic countries has been noted [3,4]. In addition, signs of resistance to artemisinin-based combination therapies (ACTs) have been detected. Actually, ACTs are the first-line treatment for malaria and thus, new drugs are constantly required [5,6,7,8]. Researchers have identified the N-myristoyltransferase (NMT) enzyme as an important target for a generation of drugs to be used for the treatment of malaria. NMT catalyzes the transfer of the myristoyl group from a myristoyl coenzyme A (CoA) to the N-terminal glycine residue after the targeted protein [9]. N-terminal myristoylation (MYR) by NMT occurs through the Bi-Bi mechanism, where MYR-CoA binds to the apo-enzyme, inducing a conformational change that allows the NMT substrate to bind [5,10]. P. falciparum has a single NMT isoform and mRNA is expressed in asexual blood-stage forms [11]. The first reported NMT inhibitors were obtained by mimicking the structure of peptide substrates (Figure 1A) [12] or by designing non hydrolysable, methylene-bridged analogue of myristoyl coenzyme A (Figure 1B) [13]. After that, inhibitors based on a quinolone scaffold and furan core were reported (Figure 1C,D) [14]. In this work, we used dimensional quantitative structure-activity relationship analysis of 83 NMT inhibitors based on a phenyl scaffold [15] seeking to propose new candidates for NMT inhibitors. Furthermore, a physicochemical properties evaluation was performed in order to find the most appropriate compound predicted.

2. Results

The GA-PLS analysis using grid cells of 1.0 A generated several models or equations. The statistical parameters of ten alignments studied for Test Set I (compounds 1, 3, 5, 6, 12, 16, 20, 30, 33, 39, 40, 50, 56, 57, 61, 65, 66, 69, 76, and 80) and Test Set II (compounds 3, 6, 9, 13, 20, 21, 27, 28, 31, 32, 40, 56, 57, 58, 64, 70, 73, 76, 78, and 82) are shown in Table 1 and Table 2, respectively. All tested alignments showed q2 values higher than 0.5. This reveals that the model can be a useful tool for predicting affinities of new compounds based on these structures; r2 greater than 0.7 indicates that the model is correlated and may be considered to represent the training set in the same manner [16]. Alignments 6B and 7B were eliminated from the analysis because it presented a low r2 value (<0.7).
Evaluating the predictive ability in terms of R2pred (means of an external validation), the alignment 5A was eliminated (R2pred < 0.5). All R2m values were greater than 0.60, and values over 0.5 are acceptable. Analyzing the R2p values, alignments B1, B2, B4, B6, and B7 were excluded because this parameter value was less than 0.5 [17].
Alignment 3 from Test Set II (B3) provides the best 4D-QSAR models as judged by the highest q2adj, in addition to presenting fewer descriptors. Among the alignments with only seven descriptors, this still has the highest values of r2, q2adj, R2p, and the lowest value of R2r. According to these results, we selected Model B3 as the best alignment. We will only present the analysis of the best model derived from B3.
The statistical measures, including the values of r2, q2, q2adj, LSE, LOF, RMSEC, RMSECV, RMSEP, Y-Rand, R2pred, R2m, R2p, and R2r are presented below. Each GCOD (grid cell occupancy descriptors) is labeled as “x, y, z, IPE” which represent the cartesian coordinate positions of the selected grid cell (x, y, z) and the respective atom type (interaction pharmacophore elements, IPE): (i) any type (any); (ii) nonpolar (np); (iii) polar-positive charge density (p+); (iv) polar-negative charge density (p−); (v) hydrogen bond acceptor (hba); (vi) hydrogen bond donor (hbd); and (vii) aromatic systems (ar).

Model B3

pIC50 = 3.997 + 4.942(0,−3,−1, hba) + 2.345(0,−5,−1, any) + 2.100(0,−1,0, any) + 1.692(0,3,−3, any) + 1.191(−1,−4,−3, any) ‒ 8.269(−1,−4,−4, np)
n = 63, GCODs = 7, r2 = 0.757, q2 = 0.702, q2adj = 0.634, LSE = 0.233, LOF = 0.418, RMSEC = 0.472, RMSECV = 0.527, RMSEP = 0.515, RMSEcy-rand = 1.055, R2pred = 0.746, R2m = 0.716, R2p = 0.609, and R2r = 0.110.
Another different variant of R2m metrics was calculated from Model B3 to assess the predictive ability of the test set, ∆R2m. The value of ∆R2m found was 0.133. It has been suggested that to be considered a predictive model, this value should be less than 0.2 [18]. Model B3 generated seven descriptors, where GCODs (−1,−4,−3, any), (0−0, any), (0,6,2, any), (0,−5,−1, any), (0,3,−3, any), and (0,−3, −1, hba) present positive coefficients (Equation (3)) and correspond to favorable interactions between the molecule substituent and amino acid residues in the active site of NMT. Therefore, substituents in these positions increase the effectiveness of the compounds. The GCOD (−1,−4,−4, np) has negative coefficient and correspond to unfavorable interactions between the molecule substituent and amino acid residues in the active site of NMT. Therefore, the occupation of GCOD (−1,−4,−4, np) decreases the compound potency.

3. Discussion

GCODs are related to the coordinates of IPE mapped in a common grid. A graphic representation of the descriptors of Model B3 is shown in Figure 2 using Compound 81 as a reference. Light and dark spheres represent GCODs with positive and negative coefficients, respectively, in accordance with Model B3. GCOD-1 (0,−3,−1, hba) (Figure 3) is the descriptor that most contributes to the increased effectiveness of compounds and presents a coefficient of 4.942. This grid cell represents an acceptor hydrogen bond atom type (IPE) and shows high frequency of occupation for compounds 42, 48, 65, 68, and 69. It is located close to the nitrogen atom of the oxadiazole ring and indicates an amino acid donor hydrogen bond in N-myristoyltransferase.
The oxadiazole ring in the ortho position allows the nitrogen atom to occupy this grid cell, exemplified by compound 42 (Figure 3). However, the most active molecules of the training set, 81 and 83, do not have this descriptor that contributes most to the increase in the potency of the compounds. Once in this position there is a methyl group and the oxadiazole group that are displaced. In fact, the oxadiazole ring in these compounds does not occupy this grid cell.
GCOD-2 (−1,−4,−4, np) (Figure 4) contributes to decrease compound potency and presents a coefficient of −8.269. This grid cell corresponds to a nonpolar IPE and shows high occupation frequency for Compounds 42, 48, and 55. These molecules present non-polar groups in this local, such as ethyl and methyl groups. Thus, the occupation of this cell is drastically reduced when this position is non-polar substituted, that decrease the activity of these compounds. Meanwhile, if the polar groups, such as OH or N in 81 or 83, respectively, are localized in this grid cell, the GCOD-2 descriptor yields less negative value which does not decrease the predicted pIC50. We can see that compound 42 occupies the descriptor that decreases the activity. However, it also presents occupancy for GCOD-1, which increases the activity. As expected, this compound exhibits an intermediate power.
This suggests that the occupation of this cell by acceptor hydrogen bond should be favorable in contrast to nonpolar atoms that are unable to perform hydrogen bond interactions.
GCOD-3 (0,−5,−1, any) is present as a non-specific IPE. The atom type “any” is used when more than one specific atom [IPE] type across the training set is found to satisfy the interaction being captured by a particular GCOD [19]. GCOD (0,−5,−1, any) has a positive coefficient of 2.345 which increases the potency of the compounds (Figure 5). This grid cell is close to methyl and ethyl groups, toward the left side, and shows greater occupation frequency for compounds 26, 30, and 33. On the other hand, compounds 42 and 45 showed no occupancy for this descriptor, because during the molecular dynamics simulation, they assumed a different conformation, facing right. On the other hand, GCOD (0,−5,−1, any) also provides occupation frequency in the benzene ring of molecules 59, 60, 61, and 62.
GCOD-4 (0,−1,0, any) have a positive coefficient and, thus, also greatly influence the increase in inhibitor potency (Figure 6). It is located near the methyl group in benzofuran, 2,3-dihidro-3-methyl and represents a non-specific IPE. It shows the highest occupation frequency for the most active compounds in this series, compounds 1, 81, and 83. The GCOD (0,−3,−1, hba) reflects the importance of occupation of this receptor region for the effectiveness of the inhibitors.
GCOD-5 (0,3,−3, any) have a positive coefficient, and so, improves the effectiveness of inhibitors (Figure 7). The GCOD (0,3,−3, any) is situated near the piperidine ring and has occupancy for a large majority of molecules, such as compounds 65 and 79. The change of position of the piperidine ring in compounds 15, 16, and 30 is not favorable, so these do not have this GCOD.
GCOD-6 (−1,−4,−3, any) represents a non-specific IPE and also has a positive coefficient, indicating an increase in potency of compounds which have high occupation frequency for this descriptor (Figure 8). The molecules 14, 19, and 22 have a high occupation frequency for this GCOD, located near the benzene group. This grid cell suggests a hydrophobic region in the receptor close to the benzene ring, which should be making a π–π staking interaction between the aromatic ring of an inhibitor and one aromatic amino acid residue.
Lastly, GCOD-7 (0,6,2, any) (Figure 9) has a positive coefficient and shows a non-specific class. Molecules 5963 have a high occupation frequency for this descriptor. The presence of naphthalene group in this grid cell increase the activity. In fact, it shows that the aromatic substituents in this position should be preferred.
In order to find new active structures, the information of the descriptors obtained by the Model B3 was used. Modifications to the structure of compounds 60, 65, and 81 are suggested and compounds AE were proposed.
The proposed compounds CE, exhibited predicted pIC50 higher than 81 (the best experimental compound). The structure of the five compounds and their predicted pIC50 values are shown in Table 3. The ADME (absorption, distribution, metabolism, and excretion) of a drug is an important property that can determine the utilization of molecules proposed in the therapeutic usage. For the evaluation of pharmacokinetic parameters for molecules AE we used the Lipinski’s Rule of Five, where molecular properties are closely related to the oral bioavailability of a drug [20], wherein compounds should not violate more than one rule. In this rule, the compounds should present logP no more than 5, molecular weight of 500 Daltons, number of hydrogen bond acceptors (nON) of 10, number of hydrogen bond donors (nOHNH) of 5, and number of rotatable bonds (nrotb) no more than 10. The proposed molecules have been designed in the Molinspiration Online Property Calculation Software Toolkit [21] to evaluate the criteria discussed above.
The Molinspiration Online Property Calculation Software Toolkit [21] was used to analyze drug likeness (Lipinski’s Rule of Five) and the results are shown in Table 4. According to the data in Table 4, no compound violated the Lipinski’s Rule of Five.

4. Materials and Methods

4.1. Biological Data

In order to build QSAR models, 83 Plasmodium falciparum inhibitors were retrieved from Leatherbarrow et al. [15]. Twenty compounds (25%) were randomly selected to compose the test set (external validation). Two test groups were chosen. The first (Test Set I) has the following molecules: 1, 3, 5, 6, 12, 16, 20, 30, 33, 39, 40, 50, 56, 57, 61, 65, 66, 69, 76, and 80; Test Set II has the following molecules: 3, 6, 9, 13, 20, 21, 27, 28, 31, 32, 40, 56, 57, 58, 64, 70, 73, 76, 78, and 82 (Table 5).
The biological activities of these compounds were reported as the negative logarithm of concentration capable of inhibiting 50% of the enzyme activity (IC50), measured using an adapted version of the sensitive fluorescence-based assay based on detection of CoA by 7-diethylamino-3-(4-maleimido-phenyl)-4-methylcoumarin [15]. Furthermore, in an effort to eliminate the potential noise that might have been introduced by the pooling of data sets from different sources, all pharmacological data are obtained from the same laboratory. The IC50 (µM) values were converted into molar units and then expressed in negative logarithmic units (pIC50), and are depicted in Table 4. The range of pIC50 values for the training and test set spans at least three orders of magnitude (4.00 to 7.30), and the biological activity values show a regular distribution over the whole range.

4.2. Molecular Dynamic Simulation (MDS)

The three-dimensional structure from 83 analogues (Table 5) were optimized in vacuum, without any restriction, and the partial atomic charges assigned using RM1 semiempirical Hamiltonian [22]. The MDS was carried out at 300 K, close to the temperature assays, with a simulation sampling time of 100 ps, and intervals of 0.001 ps. Thus, a total sample of 100,000 conformations of each compound was produced. MDS have been performed using the GROMACS 5.1 package [23].

4.3. Alignment Definition

As the compounds are structural analogs, we will assume that all molecules bind to the receptor in a similar mode. In general, the alignments are chosen to span the common framework of the molecules in the training and test sets [24,25,26]. In this work, ten alignments were performed using atoms of the (common) benzene ring. Three-ordered atom trial alignments were selected: (1) a-b-c, (2) a-b-d, (3) b-c-d, (4) c-d-f, (5) b-c-a, (6) b-a-c, (7) a-c-b, (8) c-b-a, (9) d-a-f, and (10) a-b-e (Figure 10). The order of the three ordered-atoms is important: the first atom specified for a molecule might be expected to occupy a similar location in space as the first atom specified for the second molecule. The conformational ensemble profile (CEP) for each compound obtained after the MDS step was overlaid onto a cubic lattice with grid cell size of 1A.

4.4. Interaction Pharmacophore Elements

According to the 4D-QSAR methodology, atoms of each compound are defined by seven types of interaction pharmacophore elements (IPEs). IPEs correspond to the interactions that may occur between ligand and the active site: (i) any type (any); (ii) nonpolar (np); (iii) polar-positive charge density (p+); 9iv) polar-negative charge density (p−); (v) hydrogen bond acceptor (hba); (vi) hydrogen bond donor (hbd); and (vii) aromatic systems (ar). The occupancy of the grid cells by each IPE type are recorded over the conformational assembly profile, and forms the set of grid cell occupancy descriptors (GCOD) to be utilized as the pool of trial descriptors in the model building and optimization process [16]. The idea underlying a 4D-QSAR analysis is that variations in biological responses were related to differences in the Boltzmann average spatial distribution of molecular shape with respect to the IPE. Thus, the normalized grid cell absolute occupancy, defined as the number of times that a cell was occupied by an atom type during the MDS, divided by the size of the CEP (1000 conformations), was used to define the GCODs.
4D-QSAR model calculation. In order to exclude data noise from databases generated by the alignments, partial least-squares (PLS) regression analysis was performed as a data reduction fit between the observed dependent variable measures and the corresponding set of GCOD values. Additionally, PLS identifies the most highly weighted GCODs from data set of local grid cells [16].
The two hundred GCODs with the highest weight from the data reduction were chosen to form the trial descriptor basis sets for model optimization by genetic function approximation (GFA) analysis [16]. Optimizations were initiated using 100 randomly generated models and 10,000–100,000 crossover operations. Mutation probability over the crossover optimization cycle was set at 10–30%. The smoothing factor, the variable that specifies the number of descriptors in the QSAR models, was varied between 1.0 and 3.0, in order to determine equations with no more than twelve terms. Each alignment was evaluated using the procedure described above.
The best models, resulting from the 4D-QSAR study were based on different criteria [18,26,27,28]:
(1)
Coefficient of determination (r2): is a measure of how well the regression line represents the data.
(2)
Adjusted cross-validated squared correlation coefficient (q2adj): allows the comparison between models with different number of variables.
(3)
Correlation coefficient of external validation set (R2pred): reflects the degree of correlation between the observed (YExp(test))and predicted (YPred(test)) activity data of the test set:
  R Pred 2 = 1 1 n ( Y Exp ( test ) Y Pred ( test ) ) 2 1 n ( Y Exp ( test ) Y ¯ Training ) 2  
where Y ¯ Training is average value for the dependent variable for the training set.
(4)
Modified r2 (r2m(test)) equation determining the proximity between the observed and predicted values with the zero axis intersection:
  r m ( test ) 2 = r 2 ( 1 | r 2 r 0 2 | )  
(5)
Y-randomization (R2r) consists of the random exchange of the independent variable values. Thus, the R2r value must be less than the correlation coefficient of the non-randomized models.
(6)
R2p penalizes the model R2 for the difference between the squared mean correlation coefficient (R2r) of randomized models and the square correlation coefficient (r2) of the non-randomized model:
  R p 2 = r 2 * r 2 R r 2  

4.5. Conformational Selection

In the 4D-QSAR method, the conformation of each compound can be postulated as the lowest-energy conformer state from the set sampled for each compound, which predicted the maximum activity using the optimum 4D-QSAR model [16,29,30,31,32].

5. Conclusions

In summary, 4D-QSAR models for NMT inhibitors were built and evaluated. Two test groups were evaluated for the ten tested alignments. The best model was obtained from Alignment B3, and generated an equation with seven descriptors, six of which have positive coefficients and one a negative coefficient. Model B3 showed a satisfactory statistical quality and predictive abilities as shown by r2 = 0.757, q2 = 0.702, q2adjusted = 0.634, R2pred = 0.746, R2m = 0.716, and R2p = 0.609. Furthermore, it showed low values of R2r = 0.110, and ∆R2m(test) = 0.133. 4D-QSAR analysis indicated an important role of acceptor hydrogen bonding groups and aromatic groups, allowing to propose five structures. These have proved more active than compound 81, in addition to being assessed by the Lipinski’s Rule. Accordingly, these molecules may be considered promising prototypes against malaria.

Author Contributions

Conceptualization E.F.F.d.C., K.K. and K.M.; L.S.-G. and M.A.d.M.F. did the computational experiments; T.C.R. did data interpretation; E.F.F.d.C. and L.S.-G. Writing-Original Draft Preparation; E.F.F.d.C., and K.M. Writing-Review & Editing.

Funding

This research was funded by Conselho Nacional de Desenvolvimento Científico e Tecnológico (307837/2014-9).

Acknowledgments

We thank the Brazilian agencies “Conselho Nacional de Desenvolvimento Científico e Tecnológico” (CNPq, Brazil), “Coordenação de Aperfeiçoamento de Pessoal de Nível Superior” (CAPES) and “Fundação de Amparo à Pesquisa do Estado de Minas Gerais” (FAPEMIG, Brazil) for their support. We thank A. J. Hopfinger who kindly supplied the 4D-QSAR program for academic use. This work was also supported by Excellence Project FIM, Long-Term Development Plan UHK, and the Ministry of Health (Czech Republic) project NV16-34390A.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Njoroge, M.; Njuguna, N.M.; Mutai, P.; Ongarora, D.S.B.; Smith, P.W.; Chibale, K. Recent Approaches to Chemical Discovery and Development against Malaria and the Neglected Tropical Diseases Human African Trypanosomiasis and Schistosomiasis. Chem. Rev. 2014, 114, 11138–11163. [Google Scholar] [CrossRef] [PubMed]
  2. Liu, W.; Li, Y.; Learn, G.H.; Rudicell, R.S. Origin of the human malaria parasite Plasmodium falciparum in gorillas. Nature 2010, 467, 420–467. [Google Scholar] [CrossRef] [PubMed]
  3. Krungkrai, S.R.; Krungkrai, J. Insights into the pyrimidine biosynthetic pathway of human malaria parasite Plasmodium falciparum as chemotherapeutic target. Asian Pac. J. Trop. Med. 2016, 9, 525–534. [Google Scholar] [CrossRef] [PubMed]
  4. Ridley, R.G. Medical need, scientific opportunity and the drive for antimalarial drugs. Nature 2002, 415, 686–693. [Google Scholar] [CrossRef] [PubMed]
  5. Tate, E.W.; Bell, A.S.; Rackham, M.D.; Wright, A.H. N-yristoyltransferase as a potential drug target in malaria and leishmaniasis. Parasitology 2014, 141, 37–49. [Google Scholar] [CrossRef] [PubMed]
  6. Fidock, D.A.; Nomura, T.; Talley, A.K.; Cooper, R.A.; Dzekunov, S.M.; Ferdig, M.T.; Ursos, L.M.B.; Sidhu, A.B.S.; Naude, B.; Deitsch, K.W. Mutations in the P-falciparum digestive vacuole transmembrane protein PfCRT and evidence for their role in chloroquine resistance. Mol. Cell. 2000, 6, 861–871. [Google Scholar] [CrossRef]
  7. Dondorp, A.M.; Nosten, F.; Yi, P.; Das, D.; Phyo, A.P.; Tarning, J.; Lwin, K.M.; Ariey, F.; Hanpithakpong, W.; Lee, S.J. Artemisinin Resistance in Plasmodium falciparum Malaria. N. Engl. J. Med. 2009, 361, 455–467. [Google Scholar] [CrossRef] [PubMed]
  8. Okombo, J.; Chibale, K. Antiplasmodial drug targets: A patent review (2000–2013). Expert.Opin. Ther. Pat. 2016, 26, 107–130. [Google Scholar] [CrossRef] [PubMed]
  9. Goldston, A.M.; Sharma, A.I.; Paul, K.S.; Engman, D.M. Acylation in trypanosomatids: An essential process and potential drug target. Trends Parasitol. 2014, 30, 350–360. [Google Scholar] [CrossRef] [PubMed]
  10. Wright, M.H.; Heal, W.P.; Mann, D.J.; Tate, E.W. Protein myristoylation in health and disease. J. Chem. Biol. 2010, 3, 19–35. [Google Scholar] [CrossRef] [PubMed]
  11. Gunaratne, R.S.; Sajid, M.; Ling, I.T.; Tripathi, R.; Pachebat, J.A.; Holder, A.A. Characterization of N-myristoyltransferase from Plasmodium falciparum. Biochem. J. 2000, 348, 459–463. [Google Scholar] [CrossRef] [PubMed]
  12. Devadas, B.; ZUpec, M.E.; Freeman, S.K.; Brown, D.L.; Nagarajan, S.; Sikorski, J.A.; McWherter, C.A.; Getman, D.P.; Gordon, J.I. Design and Syntheses of Potent and Selective Dipeptide Inhibitors of Candida albicans Myristoyl-CoA: Protein N-Myristoyltransferase. Med. Chem. 1995, 38, 1837–1840. [Google Scholar] [CrossRef]
  13. Paige, L.A.; Zheng, G.; DeFrees, S.A.; Cassady, J.M.; Geahlen, R.L. S-(2-Oxopentadecyl)-CoA, a Nonhydrolyzable Analogue of Myristoyl-CoA, Is a Potent Inhibitor of Myristoyl-CoA: Protein N-Myristoyltransferase. J. Med. Chem. 1989, 32, 1665–1667. [Google Scholar] [CrossRef] [PubMed]
  14. Gonçalves, V.; Brannigan, J.A.; Laporte, A.; Bell, A.S.; Robert, S.M.; Wilkinson, A.J.; Leatherbarrow, R.J.; Tate, E.W. Structure-guided optimization of quinoline inhibitors of Plasmodium N-myristoyltransferase. Med. Chem. Comm. 2016, 8, 191–197. [Google Scholar] [CrossRef] [PubMed]
  15. Leatherbarrow, R.; Tate, E.; Yu, Z.; Racklam, M. Novel Compounds and Their Use in Therapy. London Patent WO 2013/083991, 13 June 2013. [Google Scholar]
  16. Hopfinger, A.J.; Wang, S.; Tokarski, J.S.; Jin, B.Q.; Albuquerque, M.G.; Madhav, P.J.; Duraiswami, C. Construction of 3D-QSAR models using the 4D-QSAR analysis formalism. J. Am. Chem. Soc. 1997, 119, 10509–10524. [Google Scholar] [CrossRef]
  17. Roy, P.P.; Paul, S.; Mitra, I.; Roy, K. On Two Novel Parameters for Validation of Predictive QSAR Models. Molecules 2009, 14, 1660–1701. [Google Scholar] [CrossRef] [Green Version]
  18. Roy, K.; Chakraborty, P.; Mitra, I.; Ojha, P.K.; Kar, S.; Das, R.N. Some case studies on application of “rm2” metrics for judging quality of quantitative structure-activity relationship predictions: Emphasis on scaling of response data. J. Comput. Chem. 2013, 34, 1071–1082. [Google Scholar] [CrossRef] [PubMed]
  19. Pan, D.H.; Liu, J.Z.; Senese, C.; Hopfinger, A.J.; Tseng, Y. Characterization of a ligand-receptor binding event using receptor-dependent four-dimensional quantitative structure-activity relationship analysis. J. Med. Chem. 2004, 47, 3075–3088. [Google Scholar] [CrossRef] [PubMed]
  20. Patel, H.M.; Sing, B.; Bhardwaj, V.; Palkar, M.; Shaikh, M.S.; Rane, R.; Alwan, W.S.; Gadad, A.K.; Noolvi, M.N.; Karpoormath, R. Design, synthesis and evaluation of small molecule imidazo [2,1-b][1,3,4] thiadiazoles as inhibitors of transforming growth factor-beta type-I receptor kinase (ALK5). Eur. J. Med. Chem. 2015, 93, 599–613. [Google Scholar] [CrossRef] [PubMed]
  21. Molinspiration Cheminformatics (Nova ulica, Slovensky Grob, Slovak Republic). Available online: http://www.molinspiration.com/ (accessed on 13 September 2018).
  22. Rocha, G.B.; Freire, R.O.; Simas, A.M.; Stewart, J.J.P. RM1: A reparameterization of AM1 for H, C, N, O, P, S, F, Cl, Br, and I. J. Comput. Chem. 2006, 27, 1101–1111. [Google Scholar] [CrossRef] [PubMed]
  23. Páll, S.; Abraham, M.J.; Kutzner, C.; Hess, B.; Lindahl, E. Tackling Exascale Software Challenges in Molecular Dynamics Simulations with GROMACS. In International Conference on Exascale Applications and Software; Springer International Publishing: Cham, Switzerland, 2015; pp. 3–27. [Google Scholar]
  24. Oliveira, A.A.; Ramalho, T.C.; da Cunha, E.F.F. QSAR Study of Androstenedione Analogs as Aromatase Inhibitors. Lett. Drug Des. Discov. 2009, 6, 554–562. [Google Scholar] [CrossRef]
  25. da Cunha, E.F.F.; Albuquerque, M.G.; Antunes, O.A.C.; de Alencastro, R.B. 4D-QSAR models of HOE/BAY-793 analogues as HIV-1 protease inhibitors. QSAR Comb. Sci. 2015, 24, 240–253. [Google Scholar] [CrossRef]
  26. Caldas, G.B.; Ramalho, T.C.; da Cunha, E.F.F. Application of 4D-QSAR studies to a series of benzothiophene analogs. J. Mol. Mod. 2014, 20, 2420. [Google Scholar] [CrossRef] [PubMed]
  27. Kiralj, R.; Ferreira, M.M.C. Basic Validation Procedures for Regression Models in QSAR and QSPR Studies: Theory and Application. J. Braz. Chem. Soc. 2009, 20, 770–787. [Google Scholar] [CrossRef]
  28. Roy, K.; Paul, S. Exploring 2D and 3D QSARs of 2,4-Diphenyl-1,3-oxazolines for Ovicidal Activity against Tetranychus urticae. QSAR Comb. Sci. 2009, 28, 406–425. [Google Scholar] [CrossRef]
  29. Veerasamy, R.; Rajak, H.; Jain, A.; Sivadasan, S.; Varghese, C.P.; Agrawal, R.K. Validation of QSAR Models—Strategies and Importance. Int. J. Drug Des. Discov. 2011, 3, 511–519. [Google Scholar]
  30. Cormanich, R.A.; Moreira, M.A.; Freitas, M.P.; Ramalho, T.C.; Anconi, C.P.A.; Rittner, R.; Contreras, R.H.; Tormena, C.F. 1hJFH coupling in 2-fluorophenol revisited: Is intramolecular hydrogen bond responsible for this long-range coupling? Magn. Reson. Chem. 2011, 49, 763–767. [Google Scholar] [CrossRef] [PubMed]
  31. Caetano, M.S.; Ramalho, T.C.; Botrel, D.F.; da Cunha, E.F.F.; de Mello, W.C. Understanding the inactivation process of organophosphorus herbicides: A DFT study of glyphosate metallic complexes with Zn2+, Ca2+, Mg2+, Cu2+, Co3+, Fe3+, Cr3+, and Al3+. Int. J. Quantum Chem. 2012, 112, 2752–2762. [Google Scholar] [CrossRef]
  32. De Castro, A.A.; Prandi, I.G.; Kuca, K.; Ramalho, T.C. Organophosphorus degrading enzymes: Molecular basis and perspectives for enzymatic bioremediation of agrochemicals. Cienc. Agrotecnol. 2017, 41, 471–482. [Google Scholar]
Sample Availability: Samples of all compounds are available from the authors.
Figure 1. (A) Inhibitor mimicking the structure of substrates (Ki = 5.0, 8.0, and 35.0 μM for S. cerevisiae, C. albicans, and human NMT) [12]; (B) inhibitor methylene-bridged analogue of myristoyl coenzyme A (Ki = 24.0 nM) [13]; (C) inhibitor based on a quinolone scaffold (Ki = 4.7, and >100 μM for Plasmodium vivax and Plasmodium falciparum NMT, respectively); and (D) inhibitor based on a furan core RO-09-4609 (IC50 = 0.1 and >540 μM for C. albicans, and human NMT) [14].
Figure 1. (A) Inhibitor mimicking the structure of substrates (Ki = 5.0, 8.0, and 35.0 μM for S. cerevisiae, C. albicans, and human NMT) [12]; (B) inhibitor methylene-bridged analogue of myristoyl coenzyme A (Ki = 24.0 nM) [13]; (C) inhibitor based on a quinolone scaffold (Ki = 4.7, and >100 μM for Plasmodium vivax and Plasmodium falciparum NMT, respectively); and (D) inhibitor based on a furan core RO-09-4609 (IC50 = 0.1 and >540 μM for C. albicans, and human NMT) [14].
Molecules 23 02348 g001
Figure 2. Graphic representation of Compound 81 according to the 4D-QSAR Model B3. GCODs occupancy represented by white spheres contributes to increasing the potency of compounds, and black spheres to decrease the potency of the compounds. The GCODs described are: (1) (0,−3,−1, hba), (2) (−1,−4,−4, np), (3) (0,−5,−1, any) (4) (0,−1,0, any), (5) (0,3,−3, any) (6) (−1,−4,−3, any), and (7) (0,6,2, any). The gray and red representations are carbons and oxygen atoms.
Figure 2. Graphic representation of Compound 81 according to the 4D-QSAR Model B3. GCODs occupancy represented by white spheres contributes to increasing the potency of compounds, and black spheres to decrease the potency of the compounds. The GCODs described are: (1) (0,−3,−1, hba), (2) (−1,−4,−4, np), (3) (0,−5,−1, any) (4) (0,−1,0, any), (5) (0,3,−3, any) (6) (−1,−4,−3, any), and (7) (0,6,2, any). The gray and red representations are carbons and oxygen atoms.
Molecules 23 02348 g002
Figure 3. (A) Representation of compound 42 and GCOD-1 (0,−3,−1, hba) (white sphere), and (B) compound 81. GCODs occupancy represented by white spheres contributes to increasing the potency of compounds. The gray and red representations are carbons and oxygen atoms.
Figure 3. (A) Representation of compound 42 and GCOD-1 (0,−3,−1, hba) (white sphere), and (B) compound 81. GCODs occupancy represented by white spheres contributes to increasing the potency of compounds. The gray and red representations are carbons and oxygen atoms.
Molecules 23 02348 g003
Figure 4. Representation of GCOD-2 (−1,−4,−4, np) (black sphere) and (A) compound 42, (B) compound 81, and (C) compound 83. CODs occupancy represented by black spheres to decrease the potency of the compounds. The gray and red representations are carbons and oxygen atoms.
Figure 4. Representation of GCOD-2 (−1,−4,−4, np) (black sphere) and (A) compound 42, (B) compound 81, and (C) compound 83. CODs occupancy represented by black spheres to decrease the potency of the compounds. The gray and red representations are carbons and oxygen atoms.
Molecules 23 02348 g004
Figure 5. Representation of GCOD-3 (0,−5,−1, any) (white sphere) and (A) compound 30, (B) compound 42, and (C) compound 60. CODs occupancy represented by white spheres contributes to increasing the potency of compounds. The gray and red representations are carbons and oxygen atoms.
Figure 5. Representation of GCOD-3 (0,−5,−1, any) (white sphere) and (A) compound 30, (B) compound 42, and (C) compound 60. CODs occupancy represented by white spheres contributes to increasing the potency of compounds. The gray and red representations are carbons and oxygen atoms.
Molecules 23 02348 g005
Figure 6. Representation of GCOD-4 (0,−1,0, any) (white sphere) and (A) compound 81, and (B) compound 83. CODs occupancy represented by white spheres contributes to increasing the potency of compounds, and the gray and red representations are carbons and oxygen atoms.
Figure 6. Representation of GCOD-4 (0,−1,0, any) (white sphere) and (A) compound 81, and (B) compound 83. CODs occupancy represented by white spheres contributes to increasing the potency of compounds, and the gray and red representations are carbons and oxygen atoms.
Molecules 23 02348 g006
Figure 7. Representation of GCOD-5 (0,3,−3, any) (white sphere) and (A) compound 79, and (B) compound 30. CODs occupancy represented by white spheres contributes to increasing the potency of compounds, and the gray and red representations are carbons and oxygen atoms.
Figure 7. Representation of GCOD-5 (0,3,−3, any) (white sphere) and (A) compound 79, and (B) compound 30. CODs occupancy represented by white spheres contributes to increasing the potency of compounds, and the gray and red representations are carbons and oxygen atoms.
Molecules 23 02348 g007
Figure 8. Representation of GCOD-6 (−1,−4,−3, any) (white sphere) and (A) compound 14, (B) compound 19, and (C) compound 22. CODs occupancy represented by white spheres contributes to increasing the potency of compounds, and the gray and red representations are carbons and oxygen atoms.
Figure 8. Representation of GCOD-6 (−1,−4,−3, any) (white sphere) and (A) compound 14, (B) compound 19, and (C) compound 22. CODs occupancy represented by white spheres contributes to increasing the potency of compounds, and the gray and red representations are carbons and oxygen atoms.
Molecules 23 02348 g008
Figure 9. Representation of GCOD-7 (0,6,2, any) (white sphere) and (A) compound 62, and (B) compound 59, and the gray and red representations are carbons and oxygen atoms.
Figure 9. Representation of GCOD-7 (0,6,2, any) (white sphere) and (A) compound 62, and (B) compound 59, and the gray and red representations are carbons and oxygen atoms.
Molecules 23 02348 g009
Figure 10. Ordered atom letter codes (a–f) used in the 4D-QSAR analysis defines the three trial alignments. Compound 81 (pIC50 = 7.301) is used to define the atom letter code.
Figure 10. Ordered atom letter codes (a–f) used in the 4D-QSAR analysis defines the three trial alignments. Compound 81 (pIC50 = 7.301) is used to define the atom letter code.
Molecules 23 02348 g010
Table 1. Statistical parameters evaluated in the 4D-QSAR analysis for the ten performed alignments of Test I.
Table 1. Statistical parameters evaluated in the 4D-QSAR analysis for the ten performed alignments of Test I.
Alignmentr2RMSECq2adjRMSECVR2PredRMSEPr2mR2rR2p
A10.7460.4810.6070.5490.5320.650.710.3120.82
A20.7440.4780.6080.5480.5480.6630.6920.3430.799
A30.7610.4690.6090.5460.5080.7020.7350.1820.994
A40.7080.5080.5760.5790.5880.5950.6450.2870.825
A50.7360.5110.5890.5660.4770.6980.7660.2450.895
A60.7390.4770.5820.5630.5670.6370.670.2860.83
A70.7220.5030.5840.5710.5550.6560.6830.2910.831
A80.7460.4450.6050.5510.620.590.6060.2160.891
A90.7340.4910.5780.5660.5470.6840.6930.250.861
A100.7230.5190.5830.5720.5030.6760.740.3110.816
r2: Coefficient of determination; RMSEc: root mean square deviation of calibration; q2adj: adjusted cross-validated squared correlation coefficient; RMSEcv: root mean square deviation of cross validation; R2pred: correlation coefficient of external validation; RMSEp: root mean square deviation of prediction; r2m(test): Equation (3) (Materials and Methods); R2r: Y-randomization; R2p: Equation (2) (Materials and Methods).
Table 2. Statistical parameters evaluated in the 4D-QSAR analysis for the ten performed alignments of Test II.
Table 2. Statistical parameters evaluated in the 4D-QSAR analysis for the ten performed alignments of Test II.
Alignmentr2RMSECq2adjRMSECVR2PredRMSEPR2mR2rR2p
B10.7280.5040.6170.5440.7280.5320.6880.3010.476
B20.7280.5150.6070.5530.7630.4960.7490.2890.482
B30.7570.4720.6340.5270.7460.5150.7160.110.609
B40.7040.5490.5850.5730.7820.4760.7650.2530.473
B50.7250.50.6010.550.7060.5530.6920.1980.526
B60.6920.5590.5760.5810.7710.4890.7550.2720.448
B70.690.5560.5810.5770.7510.5090.7350.2890.437
B80.730.5140.60.550.770.4890.750.2090.527
B90.7230.5280.6050.5550.7860.4720.7730.2290.508
B100.7440.5010.6190.5420.7790.480.7440.2890.502
r2: coefficient of determination; RMSEc: root mean square deviation of calibration; q2adj: adjusted cross-validated squared correlation coefficient; RMSEcv: root mean square deviation of cross validation; R2pred: correlation coefficient of external validation; RMSEp: root mean square deviation of prediction; r2m(test): Equation (3) (Materials and Methods); R2r: Y-randomization; R2p: Equation (2) (Materials and Methods).
Table 3. Structures of the compounds proposed and the predicted pIC50 values based on Model B3.
Table 3. Structures of the compounds proposed and the predicted pIC50 values based on Model B3.
No.StructurepIC50No.StructurepIC50
A Molecules 23 02348 i0017.014B Molecules 23 02348 i0027.171
C Molecules 23 02348 i0037.622D Molecules 23 02348 i0048.161
E Molecules 23 02348 i0057.894
Table 4. Calculated parameters of the Lipinski rule of five for the proposed molecules.
Table 4. Calculated parameters of the Lipinski rule of five for the proposed molecules.
MoleculemiLogPMWnONnOHNHnnviolations
A3.13514.9710181
B2.74430.898170
C3.13415.887160
D2.41356.816140
E3.52415.887160
Table 5. Chemical structures and experimental pIC50Exp (M) values of Plasmodium falciparum inhibitors. Test Set I compound numbers are marked with an asterisk. Test Set II compound numbers are underlined.
Table 5. Chemical structures and experimental pIC50Exp (M) values of Plasmodium falciparum inhibitors. Test Set I compound numbers are marked with an asterisk. Test Set II compound numbers are underlined.
No.StructurepIC50No.StructurepIC50
1 * Molecules 23 02348 i0066.1552 Molecules 23 02348 i0074.000
3 * Molecules 23 02348 i0084.0004 Molecules 23 02348 i0094.000
5 * Molecules 23 02348 i0104.0006 * Molecules 23 02348 i0114.000
7 Molecules 23 02348 i0124.0008 Molecules 23 02348 i0134.000
9 Molecules 23 02348 i0144.00010 Molecules 23 02348 i0154.000
11 Molecules 23 02348 i0164.00012 * Molecules 23 02348 i0175.721
13 Molecules 23 02348 i0184.78514 Molecules 23 02348 i0195.113
15 Molecules 23 02348 i0204.00016 * Molecules 23 02348 i0214.000
17 Molecules 23 02348 i0224.00018 Molecules 23 02348 i0234.745
19 Molecules 23 02348 i0245.21520 * Molecules 23 02348 i0254.366
21 Molecules 23 02348 i0264.00022 Molecules 23 02348 i0274.000
23 Molecules 23 02348 i0284.00024 Molecules 23 02348 i0294.000
25 Molecules 23 02348 i0304.00026 Molecules 23 02348 i0315.699
27 Molecules 23 02348 i0326.40028 Molecules 23 02348 i0336.102
29 Molecules 23 02348 i0345.78030 * Molecules 23 02348 i0356.398
31 Molecules 23 02348 i0365.79632 Molecules 23 02348 i0375.420
33 * Molecules 23 02348 i0385.29234 Molecules 23 02348 i0396.456
35 Molecules 23 02348 i0406.67836 Molecules 23 02348 i0416.468
37 Molecules 23 02348 i0425.13138 Molecules 23 02348 i0435.585
39 * Molecules 23 02348 i0444.73040 * Molecules 23 02348 i0455.585
41 Molecules 23 02348 i0465.88642 Molecules 23 02348 i0475.284
43 Molecules 23 02348 i0486.00044 Molecules 23 02348 i0495.602
45 Molecules 23 02348 i0504.87646 Molecules 23 02348 i0516.319
47 Molecules 23 02348 i0526.21548 Molecules 23 02348 i0536.051
49 Molecules 23 02348 i0544.44550 * Molecules 23 02348 i0554.958
51 Molecules 23 02348 i0564.08652 Molecules 23 02348 i0574.217
53 Molecules 23 02348 i0586.39854 Molecules 23 02348 i0595.569
55 Molecules 23 02348 i0604.66356 * Molecules 23 02348 i0616.229
57 * Molecules 23 02348 i0624.18258 Molecules 23 02348 i0635.056
59 Molecules 23 02348 i0645.00960 Molecules 23 02348 i0655.149
61 * Molecules 23 02348 i0665.88662 Molecules 23 02348 i0675.886
63 Molecules 23 02348 i0684.80164 Molecules 23 02348 i0695.201
65 * Molecules 23 02348 i0706.95966 * Molecules 23 02348 i0715.538
67 Molecules 23 02348 i0726.48268 Molecules 23 02348 i0735.921
69* Molecules 23 02348 i0746.76970 Molecules 23 02348 i0755.569
71 Molecules 23 02348 i0766.82472 Molecules 23 02348 i0776.051
73 Molecules 23 02348 i0787.22274 Molecules 23 02348 i0796.000
75 Molecules 23 02348 i0806.62076 * Molecules 23 02348 i0816.638
77 Molecules 23 02348 i0825.49578 Molecules 23 02348 i0835.959
79 Molecules 23 02348 i0846.18180 * Molecules 23 02348 i0855.187
81 Molecules 23 02348 i0867.30182 Molecules 23 02348 i0876.921
83 Molecules 23 02348 i0886.201
* Test set group of compounds.

Share and Cite

MDPI and ACS Style

Santos-Garcia, L.; De Mecenas Filho, M.A.; Musilek, K.; Kuca, K.; Ramalho, T.C.; Da Cunha, E.F.F. QSAR Study of N-Myristoyltransferase Inhibitors of Antimalarial Agents. Molecules 2018, 23, 2348. https://doi.org/10.3390/molecules23092348

AMA Style

Santos-Garcia L, De Mecenas Filho MA, Musilek K, Kuca K, Ramalho TC, Da Cunha EFF. QSAR Study of N-Myristoyltransferase Inhibitors of Antimalarial Agents. Molecules. 2018; 23(9):2348. https://doi.org/10.3390/molecules23092348

Chicago/Turabian Style

Santos-Garcia, Letícia, Marco Antônio De Mecenas Filho, Kamil Musilek, Kamil Kuca, Teodorico Castro Ramalho, and Elaine Fontes Ferreira Da Cunha. 2018. "QSAR Study of N-Myristoyltransferase Inhibitors of Antimalarial Agents" Molecules 23, no. 9: 2348. https://doi.org/10.3390/molecules23092348

Article Metrics

Back to TopTop