The toxicity profiles of phenylpiperazinyl derivatives A1, A2, and A3, along with ten reference antimalarial drugs, were evaluated using in-silico methods (Protox II). This analysis predicted low risks of hepatotoxicity, carcinogenicity, immunotoxicity, mutagenicity, and cytotoxicity for A1-A3, unlike nine of the ten reference drugs. Mefloquine, the only exception, showed no toxicity violations. Further analysis of drug-likeness and ADME properties revealed that A1-A3 complied with the Rule of Five, displaying favorable oral bioavailability and gastrointestinal absorption.
4.1 Toxicity Prediction of Designed Drug-Candidate A1-3
The toxicity profiles (hepatoxicity, carcinogenicity, immunogenicity, mutagenicity, and cytotoxicity) [12] of the test compounds A1, A2 and A3 and also the ten standard reference drugs; artesunate, doxycycline, tafenoquine, amodiaquine, artemeter, lumefantrine, primaquine, piperaquine, mefloquine, including chloroquine were obtained through in-silico investigations using Protox II and are as outlined in Table 1 (a-b). Interestingly this analysis reveals that while all the hypothetical compounds A1-3 exhibited non-toxicity, nine out of the ten reference drugs active compounds exhibited one or more violations. The only exception being Mefloquine (Table 1a – b). Therefore, mefloquine was adopted for further virtual studies alongside the hypothetical lead compounds A1-3.
4.2 In-silico Drug-likeness and ADME Predictions
The results presented in Table 2a and Fig. 4 indicate that the hypothetical compounds A1-A3 exhibit characteristics in compliance with the Rule of Five (RO5), and showing zero violation to drug-likeness rules [13]. Additionally, compounds A1-3 presented suitable number of hydrogen bond donors (0–2 for nitrogen-hydrogen and oxygen-hydrogen bonds), and for hydrogen bond acceptors (4–7 for nitrogen or oxygen atoms). These characteristics fall within the recommended ranges (< 5 and < 10, respectively). Their molecular weights were within range and therefore align with the guideline of 150 to 500 g/mol.
The observed topological polar surface area (TPSA) values range from 40.62 to 46.94 which conform to the acceptable range of 20 to 130 Å2. Additionally, the number of rotatable bonds in compounds A1-3 does not exceed 9 (Fig. 4). Moreover, the negative log Kp values (-6.46 to -7.19) (Fig. 5) predicts that compounds A1, A2, A3 have appropriate lower permeability through human cells [19].
Interestingly, all the three designed compounds A1, A2, and A3 exhibit excellent gastrointestinal (GI) absorption rates and were permeable to blood-brain barrier (BBB) intercellular movement. These characteristics conform to potential drug leads for both gastrointestinal and nervous system interventions (Table 2b). These occurrence could be attributed to specific structural features, such as the presence of the oxygen atoms of the carbonyl and the hydroxy groups in addition to the nitrogen atoms of the central piperazinyl moiety. Furthermore, while compound A1 is a non-substrate, A2 and A3 as well as the Mefloquine reference drug were identified as substrates for P-glycoprotein (P-gp) based on previous studies [20; 19]. All three lead compounds A1-3 demonstrated good oral bioavailability, with a value of 0.55 comparable with the standard drugs, with no violation [13]. The inhibition of cytochrome P450 (CYP) isoenzymes has been recognised as a major factor contributing to pharmacokinetics-related drug-drug interactions [21]. These cytochromes play crucial roles in the metabolism and elimination of approximately 25% of clinically utilized drugs, involving the addition or removal of specific functional groups through hydroxylation, demethylation and dealkylation processes [22].
Compounds A1-3 act as inhibitors of CYP2D6 while serving as substrates for CYP1A2, CYP2C9 and CYP3A4 as presented in Table 2b. We observe in Fig. 6 the compliance of compounds A1–3 with physicochemical space for oral bioavailability where LIPO (lipophilicity): -0.7 < XLOG3 < + 5.0; SIZE: 150 g/mol < MV < 500 g/mol; POLAR (polarity): 20 A2 < TPSA < 130 A2; INSOLU (insolubility): -6 < Log S (ESOL) < 0; INSATU (insaturation): 0.25 < Fraction Csp3 < 1; FLEX (flexibility): 0 < No. of rotatable bonds < 9.
BOILED-Egg plot has proven straightforward interpretation and efficient translation to molecular design in a variety of drug discovery initiatives. This graphical output (Fig. 7) is an intuitive method to simultaneously predict two key ADME parameters, i.e. the passive gastrointestinal absorption (HIA) and brain access (BBB) relying on WLOGP and TPSA physicochemical descriptors for lipophilicity and apparent polarity [23]. The egg-shaped classification plot includes the yolk (physicochemical space for highly probable BBB permeation) and the white albumen (physicochemical space for highly probable HIA absorption). The outside grey region stands for molecules with properties implying predicted low absorption and limited brain penetration. Furthermore, it provides information on active efflux from the CNS or to the gastrointestinal lumen (an important active efflux mechanism involved in those biological barriers) by colour-coding in which the blue dots signifies a P-gp substrates (PGP+) while red dots for P-gp non-substrate (PGP−) [24]. BOILED-Egg plot (Fig. 7) of test compounds releaved compounds A1 (Molecule 1) to be P-gp non-substrate (PGP−) with active influx/efflux from the CNS, while compounds A2 and A3 (Molecule 2 and 3) were both P-gp substrates (PGP+) with active CNS efflux/influx mechanism.
4.3 Bioactivity Score for Phenylpiperazinyl derivatives A1 – A3
The candidacy of drug leads can be assessed by evaluating their bioactivity scores. All the compounds A1-3 (Fig. 5) were found to exhibit high or moderate bioactivity across various parameters. Bioactivity scores for organic molecules are interpreted as active (when the bioactivity score > 0), moderately active (when the bioactivity score lies between − 5.0 and 0.0), and inactive (when the bioactivity score < − 5.0). Specifically, compound A1 displays high activity in one out of the six parameters, with bioactivity scores 0.03 glycoprotein receptors (GPCR) while A2 and A3 only exhibited moderate activities. All the compounds A1-3 demonstrated moderate bioactivity as kinase inhibitors (KI) (with scores ranging from − 0.03 to -0.08), which suggests their potential to inhibit cancer cells [24]. They also exhibited moderate bioactivity as protease inhibitor (PI) (with scores of -0.03 to -0.04) suggesting their potential to impede the maturation of new HIV cells [25].
Additionally, we observed these ligan compounds A1-3 having potentials as moderate nuclear receptors (with scores of 0.18 and 0.26, respectively) and ion channel modulators (ICM) (scores ranging between − 0.08 and − 0.21) showing their ability to interact with hydrophobic molecules such as fatty acids, cholesterol, and lipophilic hormones [26–27]. Furthermore, A1-3 can regulate moderately metabolic enzymes and promoter proteins [28]. We observed their propensity to serve as enzyme inhibitor (with a score of -0.05 to -0.18), implying capability to bind to additional sites on the enzyme [10]. Among the hypothetical ligand compounds, A1 exhibited overall best activity especially as GCPR, ICM, KI, PI and EI, while A2 exhibits the least bioactivity score especially as ion channel modulators (ICM) and nuclear reactor ligand.
4.4 Structural Analysis of Reactivity of Compounds A1-3 Using DFT Studies
The chemical structure of a candidate drug compound influences its effectivenesss. Therefore, the chemical reactivity of the test compounds A1-3 were investigated using DFT analysis. The optimized structures and the calculated infrared spectra (IR) of A1-3 by DFT method using B3LYP functional and 6-31G** basis set are shown in Figs. 9 and 10. Also the obtained calculated parameters are displayed in Table 3. The results revealed that compound A1 has the highest electron affinity (EA) whereas A2 indicated the lowest ionization potential (IP). Ionization potential (IP) and electron affinity (EA) are the eigenvalues of HOMO and LUMO energy levels [29]. Consequently, the obtained IP and EA for the test compounds are the negative numerical values of EH and EL respectively (Table 3). The high EA of compound A1 suggest it has strong affinity to accept electron from other species, whereas compound A2 will readily lose its electron [30] relative to others owing to its lower IP value. This observation points to the fact that the binding energy trend of each compounds A1-3 is a function of EA energy of the molecules.
Furthermore, compound A3 relatively has the highest value of the global chemical hardness (η), chemical electrophilicity (ω) and electronic chemical potential (µ) whereas compound A2 displayed the highest chemical softness S value. This shows that compound A3 is less reactive unlike compound A2 which displayed the lowest chemical hardness and higest softness values. Additionally, the high µ value observed for A3 is suggestive of its good electron acceptor potential comparatively. Generally, a reagent with a high electronic chemical potential µ is a good electron acceptor, whereas a reagent with small electronic chemical potential µ is a good electron donor [31]. The µ values of compounds A1-3 confirms the binding energy trend of the test molecules with (P. falciparum dihydrofolate reductase-thymidylate synthase (PDB ID: 3UM8) receptor for which compound A1 exhibits the higest binding energy.
The electron acceptor ability of test molecules A1-3 were illustrated through conventional hydrogen bonding with the target. For instance, in compound A1, one of the carbonyl groups form hydrogen bonding with Ala16 of the receptor 3UM8 by accepting electrons from the amino group on the residue (Fig. 11). However, compound A3 also shows a non-conventional hydrogen bonding by donating electrons to the carbonyl on Ile164 residue (Fig. 14). The electrophilicity ω of the studied scaffolds also support this explanation with A1 having the highest ω value of 2.393 eV.
The molecular electrostatic potential (MEP) surfaces mapped over the total density of the geometrically optimized molecules A1-3 are presented in Fig. 9a-c. The MEP maps revealed the charge distribution around the molecules and describes enhanced electrophilicity or nucleophilicity of the studied molecules [32]. The colour map ranging from the region of red to blue depicts increase in nucleophilicity of the surface. The red regions are areas of nucleophilic attack on the molecular surface while blue regions are available for electrophilic attack. The green regions show areas of neutral charge. The hydrogen bonding shown by A1 and A3 with the target molecule 3UM8 was due to its electrophilic attack on the receptor at residues Ala16 and Ile164 respectively. The optimised structures of A1–3 and their calculated infrared spectra are also presented in Figs. 10a-c.
4.5 Molecular docking study
Dihydrofolate reductase (DHFR) is a crucial enzyme in folate metabolism, responsible for reducing dihydrofolate to tetrahydrofolate, a necessary cofactor for the synthesis of purines, thymidylate, and certain amino acids. Inhibition of DHFR disrupts DNA synthesis and cell division, making it an important target for antimalarial drugs. Drugs like methotrexate, pyrimethamine, and trimethoprim have historically been used to inhibit DHFR (Pal et al., 2023). However, drug resistance has driven the need to discover new inhibitors, often through computational methods and in silico docking studies like those represented in Fig. 11–15.
The findings from the docking simulations of ligands A1-3 and reference drugs active compounds against P. falciparum dihydrofolate reductase-thymidylate synthase are summarized in Table 4. The binding energies for compound A1 (-9.20 kcal/mol) is effectively comparable with the best reference drug, Artesunate (-9.20 kcal/mol). Furthermore, the reduced derivative A2 though performing below three of the reference drugs Artesunate, Doxycycline and Tafenoquine, nevertheless it performs better than seven other reference drugs active compounds.
The extracted 3D structures reveal interactions of the best ligand and reference drug with P. falciparum dihydrofolate reductase (PDB ID: 3UM8) are presented in Fig. 11. This reveals the binding of specific amino acid residues responsible for the reduction of DHFR to THFR for protein’s translation, normal mutations and growth (Alanine to Valine), for reproduction (Phenylalanine and Leucine), for resistance, invasion and survival (Ileucine and Asparagine) and reproduction (mutations of Threonine to Serine /Asparagine). Table 5 and Fig. 11 revealed unique interactions within the A1-3UM8 and Artesunate-3UM8 complexes exhibiting inhibition of similar amino acid residues Ala16, Leu40 and Ser108. Other variant residues are Leu46 and Val195 for A1-3UM8, and Ile164 and Phe58 for Artesunate-3UM8 interactions. Interactions shown by derivative A2-3 and the non-toxic reference drug, Mefolquine are also presented in Fig. 12.
The A1 derivative forms multiple interactions with DHFR (Fig. 11), as seen in the bond length, residues, hydrophobic surface, and solvent accessibility interactions. Hydrophobic residues like leucine (Leu) and alanine (Ala) play a central role in stabilizing the interaction, with Leu 40 and Ala 16 being particularly important. Serine (Ser) at position 195 is also highlighted, suggesting a hydrogen-bonding interaction, which contributes to the overall stability of the complex. The bond lengths reflect proximity to key active site residues, indicating effective binding within the enzyme’s active pocket. The hydrophobic surface shown around the ligand suggests that the interaction is predominantly non-polar, which is beneficial for binding affinity and inhibition.
Artesunate, a well-known antimalarial drug, forms specific interactions with DHFR (Fig. 12), as indicated by the bond length and residue interactions. Leucine (Leu 195) and isoleucine (Ile 46) seem to contribute to hydrophobic interactions that stabilize the binding. The image indicates that artesunate’s interaction with DHFR involves both hydrophobic regions and solvent-accessible areas. The solvent accessibility surface interaction, particularly the exposed hydrophilic areas, suggests that artesunate could form additional interactions with surrounding water molecules or other active-site residues, enhancing the drug's binding affinity.
For the A2 derivative (Fig. 13), there are significant binding interactions with residues such as Leu 40 and Ile 46. The positioning of these residues, coupled with their hydrophobic properties, suggests that the binding of A2 is largely driven by van der Waals forces and hydrophobic effects. The relatively close bond lengths imply a strong interaction between the drug and the DHFR active site. This could enhance A2’s ability to inhibit DHFR by blocking the active site, preventing the binding of natural substrates necessary for folate metabolism.
The A3 derivative appears to engage in more complex interactions compared to A1 and A2. In addition to hydrophobic residues like Leu 40 and Ile 46, this derivative shows enhanced solvent area accessibility interactions, as seen in the visualization (Fig. 14). This suggests that A3 may form water-mediated hydrogen bonds or interact with nearby polar residues in the enzyme, potentially leading to greater specificity and binding strength. The hydrophobicity of A3 is balanced by its interactions with solvent-accessible regions, which may contribute to improved solubility and bioavailability.
Mefloquine, another well-established antimalarial drug, interacts primarily with hydrophobic residues such as Leu 195 and Ile 46 as shown in Fig. 15. The bond length measurements indicate close interactions, which could contribute to the strong binding affinity observed with DHFR. The solvent accessibility surface interaction reveals that mefloquine has portions exposed to the surrounding solvent, which may aid in its transport and diffusion within the biological system. The hydrophobicity of the interaction suggests that the drug remains well-anchored in the DHFR active site, preventing substrate access and inhibiting the enzyme's activity.
4.6 Significance of A1 – A3 bond lengths, residues, hydrophobicity and solvent interactions
The bond lengths between drug derivatives and DHFR active site residues are crucial in determining the strength of the interaction. Shorter bond lengths typically indicate stronger interactions, which can translate to better inhibition of DHFR. Residues such as leucine, isoleucine, alanine, and serine frequently participate in these interactions, either through van der Waals forces or hydrogen bonding. Hydrophobic residues like Leu and Ile are particularly significant in stabilizing the drug-enzyme complex by creating a favorable non-polar environment within the binding pocket. Hydrogen bonds involving serine residues, on the other hand, contribute to specificity and binding strength.
Hydrophobic interactions dominate the binding of these derivatives to DHFR, as evidenced by the brown areas surrounding the ligands in the images. Hydrophobicity plays a key role in drug binding since it often correlates with higher affinity and reduced off-target interactions. The balance between hydrophobicity and solvent-accessibility is crucial for determining a drug's bioavailability and solubility. Compounds that interact strongly with hydrophobic residues but still maintain some degree of solvent interaction, like A3 and mefloquine, have better pharmacokinetic properties, such as absorption and distribution within the body.