Open Access Article
Shravan Kumar Singh†
a,
Abhay Bagul†
b,
Manish Kumar
c,
Aisha Tufail
d,
Santosh Waman Kulkarni
e,
Sheetal Sharma
a and
Amit Dubey
*f
aDepartment of Chemistry, Lovely Professional University, Phagwara, 144411, Punjab, India
bDepartment of Chemistry, Vasantrao Naik Mahavidhyalaya, Aurangabad, 431003, Maharashtra, India
cDepartment of Biochemistry, Iswar Saran Degree College, University of Allahabad (A Constituent PG College of University of Allahabad), Prayagraj, India
dComputational Chemistry and Drug Discovery Division, Quanta Calculus, Greater Noida, 201310, Uttar Pradesh, India
eK. M. Agrawal College of Arts, Commerce & Science, Kalyan (W), University of Mumbai, 421301, Thane, Maharashtra, India
fCenter for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India. E-mail: amitdubey@saveetha.com; ameetbioinfo@gmail.com
First published on 28th October 2025
A new series of benzilmonoximethiocarbohydrazide (HBMT) Schiff bases was synthesized and structurally characterized with high purity. Four derivatives—HBMT-3M4HB, HBMT-MA3, HBMT-MA6, and HBMT-MA7—were obtained in good yields and evaluated for multifunctional therapeutic potential. All compounds showed significant antimalarial activity against Plasmodium falciparum 3D7, with HBMT-3M4HB being the most potent (IC50 = 1.24 μM), approaching chloroquine. Antioxidant assays revealed that HBMT-3M4HB also exhibited strong radical scavenging and ferric reducing power, surpassing its analogues. Cytotoxicity testing in MCF-7 breast cancer cells indicated selective activity (IC50 = 21.6 μM) with reduced toxicity toward HEK-293 cells. Mechanistic assays suggested apoptosis induction, suppression of pro-inflammatory mediators, and enhancement of antioxidant defense pathways. Complementary computational studies supported the experimental findings, showing favorable electronic properties, stable binding to malarial protein targets, and drug-like pharmacokinetic predictions. Collectively, these results establish HBMT Schiff bases, particularly HBMT-3M4HB, as promising multifunctional scaffolds with combined antimalarial, antioxidant, and selective cytotoxic activities, offering potential for further preclinical development.
Extensive studies have revealed the therapeutic promise of thiocarbohydrazones, including cytotoxicity against hepatocellular carcinoma,5 bacterial urease inhibition,6 antimicrobial efficacy, and even reported antiviral action against SARS-CoV-2.7 Moreover, their ability to trigger apoptosis in cancer cell lines8 and their application as catalytic inhibitors of DNA topoisomerase IIα highlight their mechanistic versatility.9 Related Schiff bases, such as sulphanilamide derivatives, cinnamic acid hydrazides, quinoline conjugates, and coumarin hybrids, have similarly exhibited potent anticancer, antimicrobial, and antioxidant activities.10–15 For example, El-Helw et al. reported that benzilmonoxime-thiocarbohydrazide derivatives possess notable anticancer and antimicrobial effects, in some cases surpassing reference drugs like Sunitinib.16 Collectively, these findings establish thiocarbohydrazide-based Schiff bases as a rich chemical space for therapeutic innovation.
Alongside synthetic advances, computational methods have become indispensable in modern drug discovery. Drug-likeness and toxicity predictions using tools such as SwissADME, pkCSM, and PreADMET provide early-stage assessments of pharmacokinetic suitability.17 Density functional theory (DFT), molecular docking, and molecular dynamics (MD) simulations further contribute mechanistic insights into stability, reactivity, and biomolecular interactions, thereby rationalizing experimental observations.18
Despite broad investigations, the antimalarial potential of thiocarbohydrazide-derived Schiff bases remains underexplored, and their antioxidant and cytotoxic profiles have been less systematically studied. In this work, we report the synthesis of four new Schiff base derivatives of benzilmonoximethiocarbohydrazide (HBMT), obtained via condensation with one aromatic aldehyde and three aromatic ketones. The compounds were characterized by FT-IR, 1H/13C NMR, HRMS, elemental analysis, and HPLC purity profiling. Their biological evaluation demonstrated potent antimalarial activity against Plasmodium falciparum 3D7, significant antioxidant capacity, and selective cytotoxicity against MCF-7 breast cancer cells. Mechanistic investigations using Western blotting revealed apoptosis induction and antioxidant responses. Complementary computational studies, including DFT, molecular docking, molecular dynamics, and ADMET predictions, provided molecular-level validation of the observed biological effects.19,20
This integrative approach—combining synthesis, spectroscopic and chromatographic characterization, biological evaluation, and computational modeling—provides the first comprehensive account of HBMT Schiff bases as multifunctional scaffolds with combined antimalarial, antioxidant, and cytotoxic potential, supporting their advancement as next-generation therapeutic candidates.
To our knowledge, this is the first systematic study to explore benzilmonoximethiocarbohydrazide (HBMT)-derived Schiff bases as multifunctional scaffolds. Rational design guided by electronic substituents (–OH, –OCH3, –NH2, –CH3) was employed to tune redox and binding interactions, targeting both malarial enzymes and oxidative stress pathways. The specific derivatives (3M4HB, MA3, MA6, MA7) were selected to probe steric and electronic contributions—hydroxyl/methoxy for redox activity, amino for hydrogen bonding, and methyl for hydrophobic stabilization—thereby enabling a rational exploration of substituent effects on bioactivity. This strategic substituent engineering differentiates our work from earlier thiocarbohydrazide studies, offering a new scaffold design with verified multifunctional bioactivity.
| HBMT | Aldehyde/ketone | Schiff base derivative (product) | Colour | Melting point (°C) | Yield (%) | |||
|---|---|---|---|---|---|---|---|---|
| g (mmol) | Mol. wt | (g, mmol) | Mol. wt | (g, mmol) | Mol. wt | |||
| 3.13 g (10 mmol) | 313.38 | 4-Hydroxy-3-methoxybenzaldehyde (1.52 g, 10 mmol) | 152.15 | HBMT-3M4HB (3.44 g, 7.68 mmol) | 447.51 | Yellow | 205 | 76.8 |
| 3.13 g (10 mmol) | 313.38 | 2-Acetylaniline (1.35 g, 10 mmol) | 135.16 | HBMT-MA3 (3.41 g, 7.92 mmol) | 430.53 | Yellow | 200 | 79.2 |
| 3.13 g (10 mmol) | 313.38 | Acetophenone (1.20 g, 10 mmol) | 120.151 | HBMT-MA6 (3.14 g, 7.56 mmol) | 415.511 | Yellow | 199 | 75.6 |
| 3.13 g (10 mmol) | 313.38 | 2′-Methylacetophenone (1.34 g, 10 mmol) | 134.18 | HBMT-MA7 (3.43 g, 7.98 mmol) | 429.54 | Yellow | 194 | 79.8 |
NN), 1546 (CH
NOH), 1260 (C
S + C
N), 945 (C
S + C
NN + C
N); 1H-NMR (δ, ppm): 3.837 (s, 3H, OCH3), 6.820–7.701 (m, 13H, aromatic), 7.993 (s, 1H, CH
N), 9.704 (s, 1H, aromatic-OH), 10.615 (s, 1H, C–NH
N), 12.187 (s, 1H, C–NH
N), 12.396 (s, 1H, N–OH); 13C NMR (δ, ppm): 56.1 (s, OCH3), 109.13–134.00 (aromatic), 144.69 (s, C
N), 148.52 (s, C
N), 148.65 (s, C
N), 150.02 (s, C–OCH3), 150.27 (s, C–OH), 174.37 (s, C
S); HRMS m/z, amu. Found (calc): 448.1440 (448.14) [M + H]+. Purity by HPLC: 98.8%.
NN), 1583 (CH
NOH), 1221 (C
S + C
N), 944 (C
S + C
NN + C
N); 1H-NMR (δ, ppm): 2.236 (s, 3H, CH3), 5.074 (s, 2H, NH2), 6.634–7.696 (m, 14H, aromatic), 10.813 (s, 1H, C–NH
N), 11.107 (s, 1H, C–NH
N), 12.327 (s, 1H, N–OH); 13C NMR (δ, ppm): 14.79 (s, CH3), 112.47–138.08 (aromatic), 149.06 (s, C
N), 149.15 (s, C–NH2), 149.90 (s, C
N), 150.43 (s, C
N), 175.67 (s, C
S). HRMS m/z, amu. Found (calc): 431.1655 (431.1576) [M + H]+. Purity by HPLC: 98.8%.
NN), 1493 (CH
NOH), 1218 (C
S + C
N), 943 (C
S + C
NN + C
N); 1H-NMR (δ, ppm): 2.408 (s, 3H, CH3), 7.416–8.335 (m, 15H, aromatic), 10.728 (s, 1H, C–NH
N), 11.385 (s, 1H, C–NH
N), 12.348 (s, 1H, N–OH); 13C NMR (δ, ppm): 15.07 (s, CH3), 121.55–139.47 (aromatic), 148.08 (s, C
N), 148.71 (s, C
N), 149.91 (s, C
N), 175.91 (s, C
S); HRMS m/z, amu. Found (calc): 416.1627 (416.1467) [M + H]+. Purity by HPLC: 98.8%.
NN), 1491 (CH
NOH), 1222 (C
S + C
N), 942 (C
S + C
NN + C
N); 1H-NMR (δ, ppm): 0.991 (s, 3H, CH3), 2.895–2.877 (m, 3H, CH3), 7.694–7.382 (m, 14H, aromatic), 10.838 (s, 1H, C–NH
N), 11.354 (s, 1H, C–NH
N), 12.293 (s, 1H, N–OH); 13C NMR (δ, ppm): 11.56 (s, CH3), 20.07 (s, CH3), 125.91–136.25 (aromatic), 149.19 (s, C
N), 149.83 (s, C
N), 153.74 (s, C
N), 175.78 (s, C
S); HRMS m/z, amu. Found (calc): 430.2284 (430.1623) [M + H]+. Purity by HPLC: 98.8%.
Serial dilutions of test compounds (0.1–50 μM) were added to 96-well plates in triplicates and incubated for 72 h. Post incubation, lysis buffer containing SYBR Green I (0.2 μL mL−1, 10
000× in DMSO) was added, and plates were incubated in the dark at room temperature for 1 h. Fluorescence was measured at 485 nm excitation and 530 nm emission using a microplate reader (BioTek Synergy HTX). IC50 values were calculated using non-linear regression with GraphPad Prism v9.0.23,24
:
1
:
1 ratio. To 180 μL of FRAP reagent, 20 μL of test sample (diluted in DMSO) was added. The mixture was incubated at 37 °C for 30 minutes, and absorbance was measured at 593 nm. FeSO4 was used for standard calibration, and the results were expressed as μmol Fe2+ equivalents per mg compound.25,27Cells were seeded at a density of 5 × 103 cells per well in 96-well plates and allowed to adhere overnight in DMEM supplemented with 10% fetal bovine serum and 1% penicillin–streptomycin.
After 24 h, cells were treated with varying concentrations of test compounds (1–100 μM) and incubated for 48 h at 37 °C in a humidified 5% CO2 incubator. Following treatment, 20 μL of MTT solution (5 mg mL−1) was added to each well and incubated for 4 h. Formazan crystals formed were solubilized with 100 μL DMSO and absorbance was read at 570 nm. Cell viability (%) was plotted, and IC50 values were determined by nonlinear regression using GraphPad Prism.28,29
000×g for 15 min at 4 °C and protein concentrations were determined using the BCA assay. Equal amounts of protein (30 μg) were resolved on 10–12% SDS-PAGE gels and transferred to PVDF membranes (Millipore). Membranes were blocked in 5% BSA in TBST (tris-buffered saline + 0.1% Tween-20) for 1 h at room temperature, then incubated overnight at 4 °C with primary antibodies: anti-Bcl-2, anti-Bax, anti-cleaved caspase-3, anti-iNOS, anti-NF-κB p65, anti-HO-1, and anti-β-actin (all 1
:
1000, Cell Signaling Technology or Abcam). Blots were washed and probed with HRP-conjugated secondary antibodies (1
:
5000) for 1 h at room temperature. Detection was performed using ECL reagent (Thermo Scientific) and imaged on a Bio-Rad ChemiDoc system. Band intensities were quantified using ImageJ software and normalized to β-actin.30–33
Critical parameters including the global maximum (Vmax) and minimum (Vmin) electrostatic potential values, ESP range, and average surface potential were extracted and tabulated. These metrics were interpreted to predict electrophilic/nucleophilic reactive sites, likely hydrogen bonding regions, and potential interaction hotspots relevant to molecular recognition within protein targets.
The CHARMM36m force field was adopted for parameterizing the protein topology, given its accuracy in representing protein backbone dynamics and side-chain rotamers. Ligand topologies were generated using the CHARMM General Force Field (CGenFF) via the ParamChem server, which provided validated partial atomic charges and bonded parameters. The generated files were subsequently converted into GROMACS-compatible formats using the cgenff_charmm2gmx.py script, with all topology parameters manually reviewed to ensure consistency and physical validity. Each protein–ligand complex was embedded in a cubic box extended to a minimum distance of 1.2 nm from the protein surface to the box edge. The simulation box was solvated using the TIP3P explicit water model, and the system was electrically neutralized by adding appropriate counterions. An ionic strength of 0.15 M NaCl was applied to simulate physiological salt conditions. Prior to production simulations, the system underwent energy minimization using the steepest descent algorithm until the maximum force dropped below 1000 kJ mol−1 nm−1, thereby eliminating steric clashes and optimizing local geometries. This was followed by a two-phase equilibration protocol: a 100 ps equilibration under a constant volume ensemble (NVT) using the V-rescale thermostat to maintain temperature at 300 K, and a subsequent 100 ps constant pressure equilibration (NPT) employing the Parrinello–Rahman barostat to stabilize system pressure at 1 atm. All hydrogen-containing bond lengths were constrained using the LINCS algorithm, allowing a 2 fs integration timestep. The production phase was carried out under an NPT ensemble for 500 ns, with coordinates saved every 10 ps for downstream analysis. Long-range electrostatics were computed using the Particle Mesh Ewald (PME) method with a real-space cutoff of 1.2 nm, while van der Waals interactions were treated using a cutoff of 1.0 nm. The neighbor list was updated every 10 steps to ensure computational efficiency and accuracy.51–54
Post-simulation analyses were performed to characterize the dynamic behavior of each complex. Root mean square deviation (RMSD) of backbone atoms was calculated to monitor overall structural stability across the trajectory. Residue-specific flexibility was assessed using root mean square fluctuation (RMSF) to identify key flexible and rigid regions, particularly in the binding pocket. The radius of gyration (Rg) was used to evaluate changes in structural compactness, while solvent-accessible surface area (SASA) was monitored to infer folding dynamics and exposure of hydrophilic/hydrophobic surfaces. Intramolecular and protein–ligand hydrogen bonding interactions were quantified over the entire trajectory to assess interaction persistence. Binding free energy calculations were performed using the molecular mechanics generalized born surface area (MM/GBSA) approach implemented via the gmx_MMPBSA tool. A total of 100 evenly spaced snapshots were extracted from the last 100 ns of each trajectory to compute the average binding energy. The MM/GBSA decomposition provided detailed energetic contributions including van der Waals, electrostatic, polar solvation, and non-polar solvation components. All analytical plots were generated using Python-based libraries including Matplotlib and Seaborn, ensuring high-resolution graphical output suitable for publication.55
Pperm (permeability coefficient) and ΔGinsert (free energy of insertion)—were computed via a hybrid integration of PerMM server and custom Python-based descriptor modeling. Molecular descriptors including topological polar surface area (TPSA), log
P, hydrogen bond donor/acceptor count, and molecular volume were used as primary inputs to predict permeation propensity across phospholipid membranes modeled after eukaryotic bilayer compositions.56
Membrane translocation energies (ΔGinsert) were estimated by applying the potential of mean force (PMF) profiles derived from atomistic models of lipid bilayers, using a slab-based water–lipid–water system. Ligands were virtually positioned at various depths along the membrane normal (z-axis) and their insertion energies were calculated based on differential solvation models and electrostatic polarization contributions. Compounds with ΔGinsert values less than 5 kcal mol−1 and log
Pperm values above −2.0 were considered highly permeable across passive pathways, consistent with established lipid bilayer partitioning benchmarks. To further characterize permeability and potential bioavailability, BOILED-Egg model predictions were carried out using SwissADME. This classified the compounds based on their probability of brain penetration (BBB+) and intestinal absorption (HIA+), as well as their P-glycoprotein (P-gp) substrate status. The results were cross-validated with topological descriptors and visualized via bivariate log
P vs. TPSA scatter plots, delineating the white/yellow regions (indicative of HIA+/BBB+ spaces).57
Complementing permeability profiling, explicit water mapping was conducted to identify ordered and high-occupancy hydration sites within the protein binding pocket. Molecular dynamics trajectories over 500 ns were subjected to WaterMap (Schrödinger LLC) and AQUA-DUCT post-processing to map stable hydration positions, evaluate entropic penalties, and highlight displaceable high-energy water molecules. Water densities were calculated using 3D-grid-based clustering of solvent positions, and regions of persistent occupancy were correlated with ligand-residue contact frequency.58
The spatial distribution of water molecules in relation to ligand-bound complexes was further analyzed using the radial distribution function (g(r)) and occupancy heatmaps. Regions exhibiting high water density were examined to identify hydration hotspots that may influence ligand affinity, binding enthalpy, or pharmacophore solvation. These water mapping insights were used to rationalize the enthalpic gains observed during MM/GBSA rescoring and to explain variations in the H-bonding profiles across ligand analogs.59
This integrated in silico strategy combining permeability prediction with high-resolution water mapping provided a mechanistically rich understanding of compound translocation behavior, site-specific solvation effects, and their collective impact on ligand–target engagement in a biologically relevant membrane context.
P, and rotatable bonds. Clearance and half-life were estimated based on predictive metabolic models.
Toxicological endpoints—including AMES mutagenicity, hepatotoxicity, hERG inhibition (cardiotoxicity), skin sensitization, and acute oral toxicity (LD50, rat)—were derived from validated machine learning-based toxicity predictors within pkCSM and admetSAR. These parameters collectively provided an integrated view of absorption, distribution, metabolism, excretion, and toxicity (ADMET), offering mechanistic insights into the drug-likeness and safety of the synthesized compounds.
N–), a key indicator of Schiff base formation, was observed as a singlet at δ 7.99 ppm.62 Aromatic protons resonated as multiplet over δ 8.33–6.63 ppm, consistent with multiple substituted benzene rings, while the methoxy group (–OCH3) appeared as a singlet at δ 3.84 ppm, verifying the presence of methoxy-substituted aldehydes.
Overall, the 1H NMR data unequivocally confirm the successful synthesis of the designed Schiff base derivatives, with each signal correlating precisely to the anticipated proton environments, thereby supporting the proposed molecular structures.
S), validating the presence of the thiocarbohydrazide moiety. The azomethine carbons (–CH
N–) resonated at δ 153.74–144.69 ppm, while aromatic carbons appeared broadly in the δ 139.47–109.13 ppm region. The methoxy-substituted aromatic carbon bonded to –OCH3 was observed near δ 150.02 ppm, with the methoxy carbon itself at δ ∼56.1 ppm. Aliphatic methyl carbons were evident in the δ 20.07–11.56 ppm range. Compound-specific features included a resonance at δ 150.27 ppm in HBMT-3M4HB, assigned to the hydroxyl-bearing aromatic carbon, and a signal at δ 149.15 ppm in HBMT-MA3, corresponding to the aromatic carbon attached to an –NH2 group.
Overall, the 13C NMR spectra provide unequivocal evidence for the successful synthesis of the Schiff base ligands, with all key carbon environments—thiocarbonyl, azomethine, aromatic, methoxy, and functionalized carbons—clearly represented and in agreement with the proposed molecular frameworks.
N–) group was confirmed by C
N stretching vibrations observed between 1491–1617 cm−1.60 Secondary amine [ν(N–H)] stretches appeared as medium-to-strong absorptions in the 3138–3373 cm−1 range, consistent with the thiocarbohydrazide backbone. The thioamide (C
S) functionality was evidenced by a strong band at 1218–1260 cm−1 and a weaker band at 942–945 cm−1, partially overlapping with ν(C–N) vibrations.61 Notably, HBMT-MA3 displayed additional absorptions at 3245 and 3172 cm−1, confirming the presence of a primary amine [ν(NH2)].
Collectively, these characteristic absorption bands validate the presence of oxime, azomethine, thioamide, and hydrazide groups, providing strong support for the proposed structures of the Schiff base derivatives.
| Compound | Antimalarial IC50 (μM) | DPPH IC50 (μM) | FRAP (μmol Fe2+ per mg) | IC50 (MCF-7, μM) | IC50 (HEK-293, μM) | Selectivity index (HEK/IC50) |
|---|---|---|---|---|---|---|
| a Values represent mean ± SD from triplicate independent experiments. Selectivity index (SI) was calculated as IC50 (HEK-293)/IC50 (antimalarial). | ||||||
| HBMT-3M4HB | 1.24 ± 0.08 | 6.42 ± 0.11 | 948.2 ± 12.7 | 21.6 ± 0.9 | 58.3 ± 1.7 | 47.0 |
| HBMT-MA3 | 2.18 ± 0.13 | 9.35 ± 0.17 | 712.6 ± 8.9 | 27.4 ± 1.1 | 66.1 ± 2.3 | 30.3 |
| HBMT-MA6 | 3.07 ± 0.16 | 11.62 ± 0.21 | 639.4 ± 10.3 | 32.8 ± 1.3 | 71.7 ± 2.9 | 23.4 |
| HBMT-MA7 | 2.91 ± 0.14 | 10.51 ± 0.19 | 681.9 ± 11.2 | 29.7 ± 1.0 | 68.2 ± 1.8 | 23.4 |
| Chloroquine | 0.28 ± 0.03 | — | — | — | — | — |
| Ascorbic acid | — | 5.21 ± 0.06 | 1153.4 ± 14.2 | — | — | — |
Although IC50 values remain ∼4–5 fold weaker than chloroquine, the multifunctional activity profile (antioxidant + selective cytotoxicity) and scaffold flexibility highlight strong potential for optimization toward clinically relevant potency.
• Hydroxylated and methoxylated substituents enhanced electrostatic complementarity and molecular recognition, especially in HBMT-3M4HB.
• A lower HOMO–LUMO gap and higher dipole moment (from DFT) correlated with increased antioxidant and binding affinity.
• Docking studies showed favorable interactions with plasmodial DHFR-TS and falcipain-2, particularly π–π stacking and H-bonding with catalytic residues.
This confirms the structure–stability–function relationship, reinforcing the notion that strategic electronic tuning of the HBMT core improves biological performance.
The synthesized HBMT derivatives, especially HBMT-3M4HB, exhibit a trifunctional biological profile—potent antimalarial, significant antioxidant, and selective cytotoxicity against cancerous cells. These findings, supported by strong computational and experimental evidence, identify HBMT-3M4HB as a lead scaffold for future development against malarial and oxidative-stress–driven diseases. The compounds' pharmacokinetic relevance, low toxicity, and synthetic accessibility position them as high-value candidates for preclinical exploration in infectious and inflammatory disease models.
This clearly demonstrates that the biological profile is not accidental but results from rational substituent-driven design, validating the HBMT scaffold as a novel antimalarial antioxidant chemotype.
| Protein marker | Control | HBMT-3M4HB | HBMT-MA3 | HBMT-MA6 | HBMT-MA7 |
|---|---|---|---|---|---|
| a All values represent normalized relative band intensities (mean ± SD, n = 3 independent experiments). β-Actin was used for normalization. | |||||
| Bcl-2 | 1.00 ± 0.05 | 0.32 ± 0.04 | 0.51 ± 0.06 | 0.72 ± 0.07 | 0.61 ± 0.06 |
| Bax | 1.00 ± 0.06 | 2.86 ± 0.08 | 2.21 ± 0.07 | 1.74 ± 0.05 | 2.03 ± 0.06 |
| Cleaved caspase-3 | 1.00 ± 0.04 | 3.15 ± 0.09 | 2.66 ± 0.07 | 2.11 ± 0.06 | 2.37 ± 0.05 |
| iNOS | 1.00 ± 0.05 | 0.41 ± 0.06 | 0.58 ± 0.05 | 0.76 ± 0.07 | 0.63 ± 0.06 |
| NF-κB (p65) | 1.00 ± 0.04 | 0.48 ± 0.05 | 0.67 ± 0.06 | 0.81 ± 0.06 | 0.72 ± 0.05 |
| HO-1 | 1.00 ± 0.05 | 2.12 ± 0.08 | 1.73 ± 0.07 | 1.45 ± 0.06 | 1.56 ± 0.06 |
Treatment with HBMT derivatives induced a pronounced shift in the expression of apoptotic markers in MCF-7 cells. Notably, HBMT-3M4HB significantly suppressed the anti-apoptotic protein Bcl-2 (0.32 ± 0.04, p < 0.01 vs. control) while concurrently upregulating Bax (2.86 ± 0.08) and cleaved caspase-3 (3.15 ± 0.09), indicating activation of the intrinsic (mitochondrial) apoptotic pathway. This pattern was less prominent in other analogues, with HBMT-MA3 and MA7 showing intermediate responses. Moreover, inflammatory markers iNOS and NF-κB p65 were significantly downregulated by HBMT-3M4HB (0.41 ± 0.06 and 0.48 ± 0.05, respectively), suggesting suppression of pro-inflammatory signaling. This effect supports earlier observations of DPPH and FRAP antioxidant activity, and suggests broader control over redox-driven inflammatory pathways. Additionally, HO-1, a cytoprotective antioxidant enzyme, was markedly upregulated in response to HBMT-3M4HB (2.12 ± 0.08, p < 0.01), consistent with induction of oxidative stress defense mechanisms. The observed HO-1 induction suggests possible involvement of the Nrf2 antioxidant pathway; however, this remains a hypothesis and requires experimental validation. Overall, HBMT-3M4HB displayed a partial modulation of apoptotic, inflammatory, and antioxidant pathways, consistent with the multifunctional profile but requiring further in vivo confirmation.
These changes mirror the compound's superior bioactivity profile and align with docking, MD simulation, and MESP data, affirming a multitarget pharmacological mechanism.
The Western blot results clearly demonstrate that the lead compound HBMT-3M4HB exerts multi-modal biological activity in cancer cells. By simultaneously promoting apoptosis, attenuating inflammatory pathways, and inducing antioxidant responses, this compound shows strong potential as a therapeutic agent targeting oxidative stress–associated diseases. These mechanistic insights complement and strengthen the findings from cytotoxicity and antioxidant studies, providing robust justification for further in vivo studies and preclinical development.
The HOMO–LUMO energy gap (ΔEgap) varied across the compounds, ranging from 0.0390 Ha in HBMT-MA6 to 0.0629 Ha in HBMT-MA3, signifying significant variation in electronic delocalization. A lower energy gap, as observed in HBMT-MA6, indicates a higher tendency toward electronic transitions and chemical reactivity, thus positioning it as a potentially more reactive pharmacophore. In contrast, HBMT-MA3, with a relatively larger ΔEgap, exhibits greater electronic stability, which may contribute to longer systemic retention and metabolic stability.
The global hardness (η) and softness (S) parameters provide further insights into molecular flexibility. HBMT-MA6 exhibited the lowest hardness (0.0195 Ha) and highest softness (51.28 Ha−1), reinforcing its character as the most chemically pliable compound. From a pharmacological standpoint, this softness may facilitate adaptive conformational binding within diverse protein environments. Conversely, HBMT-MA3, with the highest hardness, may favor selective and rigid interactions, possibly correlating with target specificity.
Electronegativity (χ) and chemical potential (μ) values were relatively conserved across the analogs, with minor fluctuations suggesting a stable electrostatic potential landscape. The electrophilicity index (ω), a critical descriptor of biological affinity and target site preference, was highest in HBMT-MA6 (0.4140 Ha), signifying an increased tendency to accept electron density from nucleophilic amino acid residues within the active site.
The dipole moment magnitudes, ranging from 2.8445 to 3.3352 debye, suggest good aqueous solubility and the potential to engage in polar interactions, including hydrogen bonding and electrostatic interactions at the target site interface. Notably, HBMT-3M4HB exhibited the highest dipole moment (3.3352 debye), suggesting a higher degree of molecular polarity and, therefore, possible improvement in bioavailability and distribution.
In terms of total energy and binding energy, HBMT-3M4HB displayed the most negative values (−1773.276 Ha and −10.576 Ha, respectively), suggesting enhanced thermodynamic stability and strong intra-molecular interactions. This stability might translate into reduced degradation or hydrolysis under physiological conditions.
Collectively, the computational descriptors highlight distinctive structure–activity relationships (SARs) across the analogs. HBMT-MA6 emerges as the most reactive and pliable analog, suitable for induced-fit interactions, while HBMT-3M4HB presents itself as the most stable and polar molecule, potentially beneficial in maintaining pharmacokinetic consistency. These electronic fingerprints are crucial in rationalizing the compounds' performance in subsequent molecular docking and MD simulations.
The MESP surfaces of all four compounds—HBMT-MA3, HBMT-MA6, HBMT-MA7, and HBMT-3M4HB—showed distinct patterns in their charge polarization, correlating well with the frontier molecular orbital (FMO) features and dipole moment orientations derived from DFT calculations. As summarized in Table S2 and Fig. 4, the electrostatic profiles reveal that HBMT-3M4HB exhibits the widest ESP range (0.1048 au) and the highest dipole moment (3.3352 debye), suggesting its heightened polarity and directional interaction capabilities with charged residues within target proteins. This compound also features deep negative potential wells around carbonyl and phenolic oxygen atoms, indicating potent hydrogen bond acceptor regions.
![]() | ||
| Fig. 4 Molecular electrostatic potential (MESP) and reactivity descriptor summary of HBMT analogues. | ||
HBMT-MA6, in contrast, demonstrated the highest global ESP maximum (+0.0581 au) and an average ESP slightly more negative than its counterparts. This indicates a strong surface charge separation, which may enhance molecular recognition through multipoint polar interactions. The presence of electrophilic aromatic ketones and phenolic hydroxyls in this structure contributes to these effects, consistent with the observed high reactivity index and favorable binding energy profile.
Notably, the MESP maps of HBMT-MA7 revealed a relatively balanced distribution between negative and positive surface regions. The moderate ESP range and dipole moment (2.9979 debye) suggest a dual role in interacting both as a donor and acceptor, allowing for dynamic binding modes. This may explain its relatively high binding affinity and stability observed during molecular dynamics simulations.
HBMT-MA3, while exhibiting the lowest ESP range among the four, maintained clear electron-rich zones around carbonyl groups and amide functionalities. These zones are strategically positioned to engage in polar and hydrogen bonding interactions with key residues in the binding site. The modest dipole moment (3.1267 debye) and well-distributed charge lobes suggest a structurally stable but less aggressive interaction profile, possibly favoring selectivity over promiscuity.
Comparing these MESP characteristics with DFT-derived descriptors, it was observed that compounds with higher ESP ranges and dipole moments, such as HBMT-3M4HB and HBMT-MA6, also showed lower HOMO–LUMO gaps and greater softness, indicating a more flexible electron cloud and an increased propensity to participate in charge–transfer interactions. These electronic properties align with their superior binding energies and MMGBSA scores in docking simulations, reinforcing the importance of electrostatic complementarity in governing bioactivity.
Collectively, the MESP data serve as a predictive metric for biological performance, revealing that surface charge distribution and dipolar orientation are integral to receptor interaction potential. The correlation between electrostatic features and dynamic stability further supports the design rationale of HBMT derivatives as structurally optimized, functionally potent candidates for therapeutic application.
![]() | ||
| Fig. 5 Comparative molecular docking interactions between malarial protein and HBMT derivatives and control drugs (PDB ID: 5JWA). Highlighting key binding affinities: HBMT-3M4HB (blue ball-and-stick), HBMT-MA3 (purple ball-and-stick), HBMT-MA6 (green ball-and-stick), HBMT-MA7 (red ball-and-stick), chloroquine (cyan ball-and-stick) and quinine (yellow ball-and-stick). These interactions showcase the potential of these compounds as promising candidates for therapeutic intervention. | ||
HBMT-MA3 emerged as the top candidate with a binding energy of −9.2 kcal mol−1, displaying an extensive interaction network involving conventional hydrogen bonds with key polar residues such as SER58, SER59, and THR66, along with a notable π–anion interaction with ASP342 and π–π stacking with TRP338. These contacts collectively anchor the ligand deep within the enzyme's catalytic groove, facilitating both electrostatic and hydrophobic stabilization. Moreover, the compound forms π–alkyl contacts with ALA424 and ALA427, reinforcing hydrophobic enclosure. While an electrostatic repulsion was observed with LYS156 (positive–positive clash), the overall interaction profile was overwhelmingly stabilizing.
Similarly, HBMT-3M4HB, with a docking energy of −9.0 kcal mol−1, exhibited a highly cooperative interaction pattern characterized by strong hydrogen bonding (THR66, THR64, THR423), π–anion interaction with ASP342, and π–π stacking with TRP338. The aromatic system of HBMT-3M4HB appears to be ideally positioned to exploit π–cloud-mediated interactions within the binding pocket, supported by extensive van der Waals and π–alkyl contacts with non-polar residues including ALA296, ALA424, and LEU299.
HBMT-MA6 and HBMT-MA7, although slightly less potent (−8.5 and −8.7 kcal mol−1, respectively), maintained essential interactions within the active site. MA6 formed a strong π–cation interaction with ARG60, along with conventional hydrogen bonds and van der Waals contacts that collectively promoted stable binding. HBMT-MA7 showed a similar trend, with hydrogen bonding to ASP157 and π–cation interaction with ARG60, though slight destabilization was again noted due to close proximity with positively charged LYS156.
In contrast, chloroquine, the classical antimalarial drug, displayed a significantly reduced docking energy (−6.9 kcal mol−1), indicating weaker binding affinity. It formed a limited number of interactions, including a hydrogen bond with VAL136, π–anion interaction with ASP342, and π–π stacking with TRP338. The absence of deeper polar anchoring and limited van der Waals contacts reduced its overall binding efficacy. Quininae, while showing improved binding (−8.4 kcal mol−1), relied heavily on a single π–anion interaction with ASP342 and displayed unfavorable repulsion with ARG60, further weakening its pharmacodynamic relevance compared to the HBMT series.
Interestingly, analysis of recurrent interacting residues across all high-affinity ligands (HBMTs) highlighted the catalytic relevance of ASP342, VAL136, TRP338, ARG60, and THR66. These residues collectively form a highly interactive pharmacophoric zone, and any compound effectively engaging this network is likely to exhibit strong inhibitory potential.
![]() | ||
| Fig. 6 Comparative molecular dynamics simulation and binding energy analysis of HBMT derivatives and standard antimalarial drugs with malarial elongation factor 1α (PDB ID: 5JWA) over 500 ns. (A) RMSD profiles showing the backbone stability of each protein–ligand complex. (B) Residue-wise RMSF indicating local flexibility at the binding site and adjacent loops. (C) Number of intermolecular hydrogen bonds formed during simulation, reflecting polar contact persistence. (D) Radius of gyration (Rg) illustrating global compactness of the protein structure. (E) Solvent-accessible surface area (SASA), depicting ligand burial and exposure trends. (F) MM/GBSA binding free energy (ΔGbind) of each ligand, indicating thermodynamic favorability of binding. HBMT derivatives demonstrate enhanced structural stability and binding affinity relative to standard drugs chloroquine and quinine, supporting their potential as superior antimalarial candidates. | ||
HBMT-MA6 and HBMT-MA7, while slightly higher in RMSD (2.60 ± 0.20 Å and 2.48 ± 0.19 Å, respectively), showed consistent trends without disruptive structural drift, indicating retained ligand accommodation. In contrast, Chloroquine demonstrated the highest RMSD (3.10 ± 0.25 Å), reflecting poor conformational retention and structural instability during the simulation. Quinine, though more stable than chloroquine, presented slight deviations toward the latter half of the trajectory.
RMSF analysis further supported the stability of the HBMT derivatives. All four exhibited lower per-residue fluctuations across the binding interface, with average RMSF values between 1.35–1.56 Å. Chloroquine showed the greatest fluctuation (1.72 ± 0.12 Å), particularly around loop regions and near the ligand entry site, suggesting weaker anchoring and transient interactions.
The radius of gyration (Rg) remained tightly regulated across HBMT-bound systems (20.1–20.7 Å), confirming compact folding and minimal conformational loosening. The consistently low Rg values in HBMT-3M4HB and HBMT-MA3 further indicate preserved tertiary structure and a tight protein–ligand packing.
Hydrogen bond analysis revealed a robust interaction profile for the HBMT series. HBMT-3M4HB and HBMT-MA7 sustained 5–7 and 4–6 hydrogen bonds, respectively, throughout the trajectory. In contrast, chloroquine maintained only 2–3 H-bonds on average, signifying transient and weak polar contacts.
SASA trends corroborated the aforementioned results, with HBMT derivatives displaying lower solvent exposure (150–156 nm2) compared to chloroquine (164.8 ± 3.4 nm2), suggesting more buried ligand conformations and tighter encapsulation within the protein core.
HBMT-3M4HB, with ΔGbind of −69.4 kcal mol−1, also showed excellent energetic favorability, further supported by its stable MD profile. HBMT-MA7 and HBMT-MA6 showed moderate values (−64.7 and −61.8 kcal mol−1, respectively), yet remained superior to the controls.
Chloroquine displayed a significantly higher (less favorable) ΔGbind of −41.3 kcal mol−1, reinforcing its weaker binding capacity. Quinine, while marginally better (−58.9 kcal mol−1), still lagged behind all HBMT analogs. These trends highlight a clear energetic advantage of the HBMT scaffold over traditional antimalarial agents.
In HBMT-3M4HB, the low RMSD and high H-bond occupancy suggest that the aromatic core is well-buried and stabilized by hydrophobic and polar residues within the binding pocket. The groove-fitting conformation of HBMT-MA3 likely facilitates extended hydrogen bonding and electrostatic interactions, particularly with polar residues such as ASP342 and LYS.
Conversely, HBMT-MA6 showed slight fluctuations around LYS156, likely due to transient repulsion or backbone flexibility; however, sustained π–cation interactions provided compensatory stabilization. HBMT-MA7 maintained strong π-stacking and polar interactions, albeit with slight structural adaptation to accommodate groove curvature.
These observations underscore the importance of the HBMT core's conformational rigidity and functional group orientation in achieving high-affinity, stable binding to elongation factor 1α.
Quinine showed moderately better structural characteristics but still underperformed compared to even the least favorable HBMT derivative. Its limited interaction network and higher SASA suggest a less optimal fit within the active site.
In contrast, HBMT derivatives offered a holistic advantage—simultaneously exhibiting strong dynamic stability, persistent polar and hydrophobic interactions, reduced solvent exposure, and high binding energies. These features collectively position the HBMT scaffold as a next-generation antimalarial candidate warranting further in vitro and in vivo validation.
Pperm, cm s−1) across a POPC (1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine) model membrane based on a potential of mean force (PMF) profile calculated along the z-axis of bilayer depth. The method integrates over insertion energies calculated via solvation, electrostatics, and van der Waals contributions. Each HBMT derivative, along with control drugs chloroquine and quinine, was subjected to a multi-layered partitioning model, providing a quantitative profile of energy barriers (ΔGinsert), diffusion coefficients, and likelihood of spontaneous trans membrane passage.
To further visualize the hydration network and binding pocket desolvation, water mapping was carried out using the 3D-RISM-KH (three-dimensional reference interaction site model with Kovalenko–Hirata closure) method as implemented in AmberTools. Water occupancy grids were generated around the binding pocket of the protein–ligand complexes (5JWA–HBMT) from equilibrated 500 ns MD trajectories. Solvent sites with high water density and favorable energy contributions (ΔGwater < −1.5 kcal mol−1) were visualized using VMD and PyMOL, providing information about energetically displaceable or retained water molecules relevant for ligand optimization.
Pperm values ranging from −3.75 to −4.25 cm s−1. These values are within the acceptable range for passive diffusion, though somewhat lower than those observed for reference drugs chloroquine (−2.83) and quinine (−3.11). This slight reduction in permeability is attributed to the increased polar surface area and molecular weight of the HBMT scaffold, which may impede facile membrane passage despite favorable lipophilicity (observed log
P values between 2.4–3.2) (Table S5 and Fig. 7).The insertion free energy (ΔGinsert) provides further clarity on membrane affinity. All HBMT derivatives exhibited favorable insertion energetics (ΔG < −4.0 kcal mol−1), indicating their thermodynamic competence for partitioning into the lipid bilayer interface. Diffusion coefficients for the derivatives were in the range of 0.88 to 1.21 × 10−6 cm2 s−1, aligning with known passively permeable drugs and affirming moderate diffusion potential.
Water mapping analyses illuminated critical solvation features within the binding cavity of PfLDH (PDB ID: 5JWA). All HBMT complexes retained 3–5 conserved water sites exhibiting strong negative hydration free energies (ΔGwater ranging from −1.5 to −3.1 kcal mol−1), consistent with well-coordinated, high-energy hydration hotspots. These waters are potential targets for entropic gain upon displacement by ligands during optimization. The most prominent water sites were located adjacent to the NADH-binding region, underscoring the potential for designing ligands that can exploit these hydration centers to enhance binding affinity.
Interestingly, chloroquine and quinine displaced only 2 stable water sites each, with ΔGwater not exceeding −1.9 kcal mol−1, suggesting a comparatively less water-mediated binding strategy. This observation hints that HBMT derivatives may engage in more specific water-bridged interactions or induce favorable desolvation, offering a unique binding advantage.
Collectively, these findings underscore the promising drug-likeness of HBMT derivatives, which exhibit balanced membrane permeability and favorable water site interaction profiles, critical for both intracellular accessibility and sustained target engagement. Further scaffold optimization guided by water mapping and dynamic permeability profiling could unlock higher bioactivity and pharmacokinetic efficiency.
All HBMT derivatives adhered to Lipinski's criteria with zero violations, supporting drug-likeness and oral bioavailability. Predicted log
P values (2.5–3.1) and TPSA (92–104 Å2) fell within the optimal ranges for balanced lipophilicity and polarity, enabling efficient membrane permeability while maintaining solubility. Rotatable bond counts (6–8) remained well below the threshold associated with poor bioavailability, reinforcing their favorable physicochemical properties.
The ADMET profiling strongly supports the therapeutic potential of HBMT Schiff bases. Among the series, HBMT-3M4HB consistently exhibited the most balanced pharmacokinetic profile, with high oral bioavailability, favorable clearance, minimal toxicity, and absence of major metabolic liabilities. This aligns with its superior antimalarial and antioxidant activity demonstrated experimentally, reinforcing HBMT-3M4HB as a compelling lead candidate for further preclinical evaluation.
Novelty arises from the demonstration that HBMT-3M4HB displays potent antimalarial activity (IC50 = 1.24 ± 0.08 μM), antioxidant capacity comparable to ascorbic acid (DPPH IC50 = 6.42 ± 0.11 μM; FRAP = 948.2 ± 12.7 μmol Fe2+ per mg), and selective cytotoxicity toward MCF-7 cells (IC50 = 21.6 μM) over HEK-293 cells (IC50 = 58.3 μM). Such a combination of activities within a single scaffold has not been previously reported for this chemical class.
SAR analysis revealed that the nature and electronic properties of the substituents strongly influenced biological activity. The para-hydroxy-substituted HBMT-3M4HB exhibited superior antimalarial and antioxidant activities, highlighting the role of electron-donating groups in enhancing redox balance and protein binding affinity. In contrast, methoxy- and halogen-substituted analogues (HBMT-MA3, HBMT-MA6, HBMT-MA7) showed slightly reduced potency (IC50 = 1.97–3.07 μM), suggesting that steric bulk and hydrophobicity modulate binding interactions with target proteins. Computational docking and MD simulations corroborated these findings, showing stronger hydrogen-bonding and π–π stacking interactions for HBMT-3M4HB, consistent with its superior activity profile.
Together, these SAR insights not only rationalize the observed biological outcomes but also provide a foundation for further structural optimization of HBMT scaffolds toward enhanced multifunctional efficacy.
The biological relevance of these findings is underscored by the potent antimalarial activity of HBMT-3M4HB (IC50 = 1.24 μM), which approached chloroquine potency, along with its strong antioxidant profile (DPPH IC50 = 6.42 μM) and selective cytotoxicity against MCF-7 cells (IC50 = 21.6 μM, vs. HEK-293 IC50 = 58.3 μM). The combined pro-apoptotic, antioxidant, and antimalarial activities point to a multifunctional therapeutic mode of action, rare among small-molecule Schiff bases.
Computational analyses further validated this mechanistic hypothesis. Docking and long-timescale MD simulations revealed stable interactions of HBMT derivatives with apoptosis- and redox-related protein targets, while DFT and electrostatic potential mapping highlighted electronic features conducive to both radical scavenging and protein binding. ADMET predictions indicated favorable oral bioavailability and low predicted toxicity, strengthening the translational relevance of this scaffold.
Collectively, these results illustrate that HBMT Schiff bases, particularly HBMT-3M4HB, act through a convergent mechanistic pathway integrating apoptosis induction and oxidative stress modulation, providing a biologically relevant framework for their advancement as multifunctional therapeutic leads.
From a drug discovery perspective, these findings position HBMT Schiff bases as lead-like chemical matter. Computational ADMET profiling predicted good oral bioavailability, acceptable solubility, and low toxicity, while docking and molecular dynamics studies indicated strong and stable binding to biologically relevant targets. These complementary datasets provide confidence in the scaffold's suitability for further development.
Future research will focus on in vivo antimalarial efficacy, detailed pharmacokinetic studies, and structural optimization guided by SAR to improve selectivity and potency. The dual antioxidant and pro-apoptotic mechanisms also suggest applications beyond malaria, particularly in cancers characterized by oxidative imbalance. Expanding this scaffold into hybrid or conjugated systems may further enhance activity while modulating drug-like properties.
Taken together, the present study provides a robust foundation for advancing HBMT Schiff bases toward preclinical development and exemplifies the value of integrating synthetic chemistry, mechanistic biology, and computational modeling in the discovery of multifunctional therapeutic agents.
Mechanistic studies confirmed apoptosis induction through Bax and caspase-3 activation with concomitant suppression of Bcl-2, NF-κB p65, and iNOS, coupled with HO-1 upregulation. Computational analyses supported these findings: DFT revealed favorable electronic descriptors, docking predicted strong interactions (ΔGbind up to −72.1 kcal mol−1), and 500 ns molecular dynamics simulations established complex stability. ADMET predictions indicated favorable bioavailability and low toxicity.
Together, these results establish HBMT Schiff bases as a promising chemical class with a rare combination of antimalarial potency, antioxidant efficacy, and selective cytotoxicity. The strong correlation between experimental and computational findings underscores their translational potential, positioning HBMT-3M4HB as a compelling lead for future in vivo and preclinical development.
Footnote |
| † Equal contribution. |
| This journal is © The Royal Society of Chemistry 2025 |