Research Article |
Corresponding author: Nety Kurniaty ( nety.kurniaty@unisba.ac.id ) Academic editor: Irini Doytchinova
© 2025 Nety Kurniaty, Taufik Muhammad Fakih, Rani Maharani, Unang Supratman, Ace Tatang Hidayat.
This is an open access article distributed under the terms of the Creative Commons Attribution License (CC BY 4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Citation:
Kurniaty N, Fakih TM, Maharani R, Supratman U, Hidayat AT (2025) Molecular docking, density functional theory, and molecular dynamics study of pipecolisporin derivatives: Unveiling the antimalarial potential of novel cyclic peptides. Pharmacia 72: 1-21. https://doi.org/10.3897/pharmacia.72.e142361
|
Malaria remains a persistent global health issue, with escalating resistance to existing antimalarial treatments driving the urgent need for novel therapeutic agents. This study aimed to evaluate the in silico antimalarial potential of pipecolisporin analogs by investigating their binding affinity to key Plasmodium proteins and assessing their pharmacokinetic and toxicity profiles. We employed molecular docking and molecular dynamics (MD) simulations to investigate the interactions between pipecolisporin analogs and three key Plasmodium proteins: dihydrofolate reductase (2BL9), plasmepsin V (4ZL4), and lactate dehydrogenase (1CET). The pharmacokinetic (ADME) properties and toxicity of the analogs were predicted using cheminformatics tools to assess their potential bioavailability and safety. Among the tested compounds, analog-3 demonstrated the highest binding affinity with 2BL9 (−13.02 kcal/mol) and 4ZL4 (−8.07 kcal/mol). MD simulations confirmed the stability of the analog-3-protein complexes, reinforcing its potential as an effective enzyme inhibitor. ADME predictions showed that all analogs had low gastrointestinal absorption and poor ability to cross the blood-brain barrier. Toxicity assessments indicated the presence of neurotoxic and respiratory risks across all analogs. Despite pharmacokinetic limitations and toxicity concerns, pipecolisporin analogs, particularly analog-3, exhibit strong inhibitory potential against key Plasmodium proteins. With further structural optimization to improve bioavailability and reduce toxicity, these compounds hold promise as novel antimalarial agents.
Plasmodium protein, pipecolisporin analog, molecular docking study, molecular dynamics simulation, pharmacokinetic properties, antimalarial candidate
In 2022, the global malaria burden rose to 249 million cases, up from 244 million in 2021, with 608,000 deaths, predominantly in sub-Saharan Africa. Children under five remain the most vulnerable, accounting for 78% of malaria-related fatalities (
Previous research has shown that traditional medicine, particularly in malaria-endemic regions, has been a source of many antimalarial compounds derived from plants. Flavonoids and other plant-based compounds have demonstrated significant antimalarial activity, forming a foundation for modern drug discovery (
The development of antimalarial drugs has increasingly relied on advanced computational tools such as computer-aided drug design (CADD) to expedite the drug discovery process. CADD includes various techniques like molecular docking, which allows for the simulation of interactions between drug candidates and target proteins, providing valuable insights into their binding affinity and potential efficacy (
In recent years, the isolation and synthesis of peptides with antimalarial properties have garnered increasing attention as a viable approach in combating drug-resistant malaria. One promising class of compounds includes various cyclic peptides, known for their ability to inhibit key Plasmodium proteins that are essential for the parasite’s survival (
In this study, we aim to evaluate the biological activity of pipecolisporin analogs through molecular docking and molecular dynamics (MD) simulations to assess their interactions with key Plasmodium proteins. In addition to computational analyses, we will investigate the pharmacokinetic properties of these analogs, including their absorption, distribution, metabolism, and excretion (ADME) profiles. Toxicity predictions will also be conducted to evaluate the safety of these compounds. Given the growing resistance to existing antimalarial treatments, the identification of new compounds with favorable binding properties and minimal toxicity is critical. By targeting essential proteins in the Plasmodium life cycle, pipecolisporin analogs could provide a new class of therapeutic agents for malaria treatment. The results of this study will contribute to the ongoing search for novel antimalarial drugs and provide insights into the potential of pipecolisporin analogs as enzyme inhibitors. Through this research, we hope to identify promising compounds that can be further developed into effective treatments for malaria.
The computational analysis in this study was performed using both Windows 10 and Linux Ubuntu 18.10 operating systems. The software utilized includes BIOVIA Discovery Studio 2024 Client 24.1, MGLTools 1.5.7 with AutoDock 4.2.6, PatchDock, PEP-FOLD 3.5, Orca 6.0, Avogadro 1.2.0, Chemcraft, Chimera 1.14, PyMOL 2.5.8, VMD 1.9.2, and GROMACS 2016.3 with the g_mmpbsa package for molecular dynamics and binding free energy calculations. Additionally, web-based platforms such as I-TASSER, SwissADME, pKCSM, ProTox-3.0, and Molinspiration were employed for structure prediction, pharmacokinetic analysis, and toxicity assessment. The computational hardware used in this study consisted of a desktop computer with an Intel Core i5-8500 CPU @ 4.30 GHz (6 cores), 16 GB RAM, a 2 TB HDD, a 120 GB SSD, and an NVIDIA GeForce GTX 1080 Ti GPU. These specifications provided sufficient computational power for molecular docking, density functional theory (DFT) calculations, molecular dynamics simulations, and free energy assessments.
The crystallographic data for essential receptors involved with Plasmodium vivax (identifiers 2BL9 (https://www.rcsb.org/structure/2BL9) (
The pipecolisporin derivatives (analog-1 through analog-6) were sourced in SDF file format from PubChem (available at https://pubchem.ncbi.nlm.nih.gov/) (Fig.
Density Functional Theory (DFT), grounded in quantum mechanics, offers an exceptionally precise representation of electron distribution within molecules, enabling the computation of multiple properties such as molecular energies, geometries, and electronic characteristics. The Orca 6.0 software package was employed to calculate these quantum mechanical properties (
The experimental ligands were accommodated within a uniformly sized grid box measuring 64 × 60 × 60 Å for all structures, ensuring coverage of the entire active site. The coordinates for the grid boxes were set based on the natural ligand’s previous positions, providing additional space for ligand flexibility within the docking process: 2BL9 (89.726, 13.626, 34.293), 4ZL4 (−3.040, 99.279, 42.328), and 1CET (36.211, 10.539, 19.830). The Lamarckian genetic algorithm was utilized in conjunction with a rigid receptor framework to identify optimal ligand conformations. Each structure underwent ten separate docking trials using MGLTools 1.5.7 and AutoDockTools (ADT) 4.2.6, from which the most consistent energy and pose across replicates were determined as the definitive outcomes (
Molecular dynamics simulations were performed to evaluate the binding stability, conformational behavior, and interaction modes between the compounds (analog-1 to analog-6) and the receptors (1CET, 2BL9, and 4ZL4). The ligand–receptor complex files were analyzed using GROMACS 2016.3 software for these simulations (
The outcomes of molecular dynamics simulations were employed to determine the binding free energies between the protein-ligand complexes using the MM-PBSA method (
This equation is further expanded as ∆Gbind = ∆GMM + ∆GPB + ∆GSA − T∆S. In this formula, ∆GMM represents the molecular mechanics interactions, which are the sum of electrostatic and van der Waals forces. ∆GPB and ∆GSA denote polar and non-polar solvation energies, respectively, while T∆S accounts for the entropic contribution to the binding free energy.
The ProTox-3.0 (Prediction of TOXicity of chemicals) (https://tox.charite.de/protox3/) server was employed to forecast the toxicity of the selected compounds, with the input provided in the form of canonical SMILES notation (
Frontier molecular orbital (FMO) analysis is a quantum chemistry computational method used to examine the energy levels, configuration, and electron distribution of a molecule’s highest occupied molecular orbital (HOMO) and lowest unoccupied molecular orbital (LUMO). HOMO-LUMO analysis is performed to determine the reactivity of molecular orbitals in organic compounds (
The global reactivity descriptors (in eV) for four selected compounds were calculated using the DFT B3LYP/6-31G method, based on the energy levels of the HOMO and LUMO orbitals.
Properties | Analog-1 | Analog-2 | Analog-3 | Analog-4 | Analog-5 | Analog-6 |
---|---|---|---|---|---|---|
Electronic energy (Eh) | −2264.156 | −2319.183464 | −2431.659809 | −2428.09052 | −2319.194922 | −2319.173796 |
Dipole moment (D) | 4.2718 | 9.7271 | 9.9702 | 7.6997 | 5.0153 | 6.4377 |
EHOMO (eV) | −0.19810 | −0.19965 | −0.20167 | −0.19296 | −0.18243 | −0.19921 |
ELUMO (eV) | −0.00608 | −0.01142 | −0.01071 | −0.00731 | −0.00058 | −0.01193 |
∆Egap (eV) | −0.19202 | −0.18823 | −0.19096 | −0.18565 | −0.18185 | −0.18728 |
Ionization potential (eV) | 0.1981 | 0.19965 | 0.20167 | 0.19296 | 0.18243 | 0.19921 |
Electron affinity (eV) | 0.00608 | 0.01142 | 0.01071 | 0.00731 | 0.00058 | 0.01193 |
Electronegativity (eV) | 0.896109 | 0.913784 | 0.909338 | 0.924093 | 0.914044 | 0.910748 |
Chemical potential (eV) | 0.10209 | 0.105535 | 0.10619 | 0.100135 | 0.091505 | 0.10557 |
Hardness (eV) | 0.09601 | 0.094115 | 0.09548 | 0.092825 | 0.090925 | 0.09364 |
Softness (eV−1) | 5.207790855 | 5.312649418 | 5.236698785 | 5.386479935 | 5.499037668 | 5.339598462 |
Electronic potential (eV) | −0.10209 | −0.105535 | −0.10619 | −0.100135 | −0.091505 | −0.10557 |
Electrophilicity (eV) | 0.054277513 | 0.059170357 | 0.059050671 | 0.054010332 | 0.04604435 | 0.059509958 |
The EHOMO values spanned from −0.18243 to −0.20167 eV, with analog-3 showing the lowest and analog-5 reaching the highest. ELUMO values ranged between −0.00058 and −0.01193 eV, where analog-6 exhibited the lowest and analog-5 the highest readings. The energy gap (∆Egap) extended from −0.18185 to −0.19202 eV, with analog-1 demonstrating the broadest gap. Ionization potential fluctuated from 0.18243 eV to 0.20167 eV. Electronegativity varied from 0.896109 to 0.924093, with analog-4 registering the highest. Chemical potential values were found between 0.091505 and 0.10619 eV. Hardness ranged from 0.090925 to 0.09601 eV, with analog-1 having the greatest hardness. Softness spanned from 5.207790855 to 5.499037668 eV−1. Electronic potential oscillated between −0.091505 and −0.10557 eV. Electrophilicity ranged from 0.04604435 to 0.059509958 eV.
The key compounds display distinct electronic characteristics when compared with each other. Notably, analog-3 features a significantly lower EHOMO and a notably tighter energy gap, suggesting it behaves differently in terms of electronic structure relative to the others. Analog-6, with the lowest ELUMO, also demonstrates higher values for electronegativity and softness, highlighting unique patterns in electron arrangement and reactivity. Analog-5, on the other hand, has the most elevated EHOMO and ELUMO values, alongside a higher ionization potential, pointing to a unique ionization profile among the group. Furthermore, their variances in electronegativity and electrophilicity signal distinct chemical reactivity and interaction potentials, particularly in contrast with the reference molecule, analog-1.
Molecular docking analysis was conducted to evaluate the interaction potential between pipecolisporin derivative compounds and specific target proteins. The docking process was carried out using MGLTools 1.5.7 and AutoDockTools (ADT) 4.2.6 to determine the binding energy values, which serve as indicators of molecular affinity. The selected target proteins, 2BL9, 4ZL4, and 1CET, are crucial in malaria treatment and possess well-defined binding sites within their crystalline structures, as documented in the .pdb file. The docking simulation was performed using a grid dimension of 64 × 60 × 60 Å. The results demonstrated that all compounds exhibited binding energies below −6.06 kcal/mol (Table
Receptor Target | Pipecolisporin Compound | Molecular Affinity | Inhibitory Value | Residue Interaction | Classification | Type of Interaction |
---|---|---|---|---|---|---|
2BL9 | Analog-1 | −10.67 kcal/mol | 15.03 nM (nanomolar) | A:LEU45 | Hydrogen Bond | Conventional Hydrogen Bond |
A:ASP53 | Hydrogen Bond | Carbon Hydrogen Bond | ||||
A:ASP53 | Electrostatic | Pi-Anion | ||||
A:LYS48 | Hydrophobic | Alkyl | ||||
A:CYS49 | Hydrophobic | Alkyl | ||||
A:MET54 | Hydrophobic | Alkyl | ||||
A:PRO122 | Hydrophobic | Alkyl | ||||
A:LEU45 | Hydrophobic | Alkyl | ||||
A:ILE121 | Hydrophobic | Alkyl | ||||
A:ALA15 | Hydrophobic | Pi-Alkyl | ||||
A:ALA15 | Hydrophobic | Pi-Alkyl | ||||
A:LEU45 | Hydrophobic | Pi-Alkyl | ||||
Analog-2 | −11.05 kcal/mol | 7.95 nM (nanomolar) | A:TRP47 | Hydrogen Bond | Conventional Hydrogen Bond | |
A:ASP53 | Hydrogen Bond | Conventional Hydrogen Bond | ||||
A:ASP53 | Hydrogen Bond | Conventional Hydrogen Bond | ||||
A:THR44 | Hydrogen Bond | Pi-Donor Hydrogen Bond | ||||
A:LEU45 A:TYR125 | Hydrogen Bond | Pi-Donor Hydrogen Bond | ||||
A:ALA15 | Hydrophobic | Pi-Sigma | ||||
A:LEU45 | Hydrophobic | Alkyl | ||||
A:MET54 | Hydrophobic | Alkyl | ||||
A:LEU45 | Hydrophobic | Alkyl | ||||
A:LEU128 | Hydrophobic | Alkyl | ||||
A:PRO129 | Hydrophobic | Alkyl | ||||
A:TRP47 | Hydrophobic | Alkyl | ||||
A:LEU45 | Hydrophobic | Pi-Alkyl | ||||
A:LEU45 | Hydrophobic | Pi-Alkyl | ||||
Hydrophobic | Pi-Alkyl | |||||
Analog-3 | −13.02 kcal/mol | 284.71 pM (picomolar) | A:SER120 | Hydrogen Bond | Conventional Hydrogen Bond | |
A:ASP53 | Hydrogen Bond | Conventional Hydrogen Bond | ||||
A:MET54 | Hydrogen Bond | Carbon Hydrogen Bond | ||||
A:ARG131 | Hydrogen Bond | Pi-Cation | ||||
A:ARG131 | Electrostatic | Pi-Donor Hydrogen Bond | ||||
A:LEU45 | Hydrogen Bond | Pi-Donor Hydrogen Bond | ||||
A:THR44 | Hydrophobic | Pi-Sigma | ||||
A:LEU45 | Hydrophobic | Alkyl | ||||
A:MET54 A:ILE13 | Hydrophobic | Alkyl | ||||
A:TRP47 | Hydrophobic | Alkyl | ||||
A:PHE57 A:LEU45 | Hydrophobic | Pi-Alkyl | ||||
A:LEU45 | Hydrophobic | Pi-Alkyl | ||||
A:LEU128 | Hydrophobic | Pi-Alkyl | ||||
Hydrophobic | Pi-Alkyl | |||||
Hydrophobic | Pi-Alkyl | |||||
Analog-4 | −10.36 kcal/mol | 25.40 nM (nanomolar) | A:ASP53 | Hydrogen Bond | Salt Bridge | |
A:ASP53 | Electrostatic | Attractive Charge | ||||
A:ASP53 | Hydrogen Bond | Conventional Hydrogen Bond | ||||
A:ASP53 | Hydrogen Bond | Conventional Hydrogen Bond | ||||
A:CYS49 | Hydrogen Bond | Conventional Hydrogen Bond | ||||
A:TYR125 | Hydrogen Bond | Pi-Donor Hydrogen Bond | ||||
A:LEU128 | Hydrophobic | Alkyl | ||||
A:ILE121 | Hydrophobic | Alkyl | ||||
A:CYS49 | Hydrophobic | Pi-Alkyl | ||||
A:MET54 | Hydrophobic | Pi-Alkyl | ||||
Analog-5 | −10.78 kcal/mol | 12.57 nM (nanomolar) | A:CYS49 A:TYR125 A:ASP53 A:ASP53 | Hydrogen Bond | Conventional Hydrogen Bond | |
A:ARG131 | Hydrogen Bond | Conventional Hydrogen Bond | ||||
A:TYR125 | Hydrogen Bond | Conventional Hydrogen Bond | ||||
A:MET54 | Hydrogen Bond | Conventional Hydrogen Bond | ||||
A:LEU45 | Electrostatic | Pi-Cation | ||||
A:MET54 | Hydrophobic | Pi-Sigma | ||||
A:TRP47 A:TYR125 A:LEU128 | Hydrophobic | Alkyl | ||||
A:PRO129 | Hydrophobic | Alkyl | ||||
Hydrophobic | Alkyl | |||||
Hydrophobic | Pi-Alkyl | |||||
Hydrophobic | Pi-Alkyl | |||||
Hydrophobic | Pi-Alkyl | |||||
Hydrophobic | Pi-Alkyl | |||||
2BL9 | Analog-6 | −10.01 kcal/mol | 46.27 nM (nanomolar) | A:SER117 | Hydrogen Bond | Conventional Hydrogen Bond |
A:SER117 | Hydrogen Bond | Conventional Hydrogen Bond | ||||
A:SER117 | Hydrogen Bond | Conventional Hydrogen Bond | ||||
A:ALA15 A:ILE121 | Hydrophobic | Alkyl | ||||
A:LEU128 | Hydrophobic | Alkyl | ||||
A:LEU45 | Hydrophobic | Alkyl | ||||
A:PHE57 A:MET54 | Hydrophobic | Alkyl | ||||
A:CYS49 | Hydrophobic | Pi-Alkyl | ||||
A:MET54 | Hydrophobic | Pi-Alkyl | ||||
Hydrophobic | Pi-Alkyl | |||||
Hydrophobic | Pi-Alkyl | |||||
4ZL4 | Analog-1 | −6.49 kcal/mol | 17.39 uM (micromolar) | A:GLU141 | Hydrogen Bond | Conventional Hydrogen Bond |
A:ASN435 | Hydrogen Bond | Conventional Hydrogen Bond | ||||
A:VAL434 | Hydrogen Bond | Pi-Donor Hydrogen Bond | ||||
A:VAL434 | Hydrophobic | Pi-Sigma | ||||
A:HIS320 | Hydrophobic | Pi-Pi T-shaped | ||||
A:VAL434 | Hydrophobic | Alkyl | ||||
A:CYS140 | Hydrophobic | Pi-Alkyl | ||||
A:CYS140 | Hydrophobic | Pi-Alkyl | ||||
A:VAL434 | Hydrophobic | Pi-Alkyl | ||||
A:LYS437 | Hydrophobic | Pi-Alkyl | ||||
A:ILE439 | Hydrophobic | Pi-Alkyl | ||||
Analog-2 | −7.22 kcal/mol | 5.13 uM (micromolar) | A:GLU431 | Hydrogen Bond | Conventional Hydrogen Bond | |
A:TYR59 A:ASP387 | Hydrogen Bond | Carbon Hydrogen Bond | ||||
A:PHE318 | Hydrogen Bond | Carbon Hydrogen Bond | ||||
A:LEU179 | Hydrophobic | Pi-Pi T-shaped | ||||
Hydrophobic | Alkyl | |||||
Analog-3 | −8.07 kcal/mol | 1.21 uM (micromolar) | A:THR317 | Hydrogen Bond | Conventional Hydrogen Bond | |
A:ASP57 A:GLU431 | Hydrogen Bond | Conventional Hydrogen Bond | ||||
A:GLU431 | Hydrogen Bond | Conventional Hydrogen Bond | ||||
A:TYR59 | Hydrogen Bond | Conventional Hydrogen Bond | ||||
A:ALA60 | Hydrogen Bond | Carbon Hydrogen Bond | ||||
A:LEU179 | Hydrophobic | Alkyl | ||||
A:PHE318 | Hydrophobic | Alkyl | ||||
A:HIS320 A:ALA60 | Hydrophobic | Pi-Alkyl | ||||
A:LEU179 | Hydrophobic | Pi-Alkyl | ||||
Hydrophobic | Pi-Alkyl | |||||
Hydrophobic | Pi-Alkyl | |||||
Analog-4 | −6.91 kcal/mol | 8.54 uM (micromolar) | A:GLU141 | Hydrogen Bond | Salt Bridge | |
A:GLU141 | Electrostatic | Attractive Charge | ||||
A:GLY315 | Hydrogen Bond | Conventional Hydrogen Bond | ||||
A:GLU141 | Hydrogen Bond | Conventional Hydrogen Bond | ||||
A:GLY315 | Hydrogen Bond | Conventional Hydrogen Bond | ||||
A:GLU431 | Hydrogen Bond | Conventional Hydrogen Bond | ||||
A:VAL434 | Hydrophobic | Pi-Sigma | ||||
A:LEU179 | Hydrophobic | Alkyl | ||||
A:VAL434 | Hydrophobic | Pi-Alkyl | ||||
Analog-5 | −6.89 kcal/mol | 8.90 uM (micromolar) | A:CYS140 | Hydrophobic | Pi-Alkyl | |
A:LEU179 | Hydrophobic | Alkyl | ||||
A:VAL434 | Hydrophobic | Pi-Alkyl | ||||
A:GLU141 | Hydrogen Bond | Conventional Hydrogen Bond | ||||
A:GLU431 | Hydrogen Bond | Conventional Hydrogen Bond | ||||
A:ALA60 | Hydrophobic | Alkyl | ||||
A:HIS320 | Hydrophobic | Pi-Pi Stacked | ||||
A:HIS320 | Hydrophobic | Pi-Pi Stacked | ||||
A:VAL434 | Hydrophobic | Alkyl | ||||
Analog-6 | −6.99 kcal/mol | 7.54 uM (micromolar) | A:THR317 | Hydrogen Bond | Conventional Hydrogen Bond | |
A:ILE56 | Hydrogen Bond | Conventional Hydrogen Bond | ||||
A:ASP57 | Hydrogen Bond | Conventional Hydrogen Bond | ||||
A:GLU43 | Hydrogen Bond | Conventional Hydrogen Bond | ||||
A:GLU58 | Hydrogen Bond | Carbon Hydrogen Bond | ||||
4ZL4 | Analog-6 | −6.99 kcal/mol | 7.54 uM (micromolar) | A:ALA60 | Hydrophobic | Pi-Sigma |
A:ALA60 | Hydrophobic | Alkyl | ||||
A:LEU179 | Hydrophobic | Alkyl | ||||
A:PHE318 | Hydrophobic | Pi-Alkyl | ||||
A:ALA60 | Hydrophobic | Pi-Alkyl | ||||
1CET | Analog-1 | −6.84 kcal/mol | 9.64 uM (micromolar) | A:GLU122 A:GLU122 | Hydrogen Bond | Conventional Hydrogen Bond |
A:ILE119 | Hydrogen Bond | Conventional Hydrogen Bond | ||||
A:TYR85 | Hydrogen Bond | Carbon Hydrogen Bond | ||||
A:TYR85 | Hydrophobic | Pi-Pi T-shaped | ||||
A:ILE54 | Hydrophobic | Pi-Pi T-shaped | ||||
A:ALA98 | Hydrophobic | Alkyl | ||||
A:LYS118 | Hydrophobic | Alkyl | ||||
A:ILE119 | Hydrophobic | Alkyl | ||||
A:ILE54 | Hydrophobic | Alkyl | ||||
A:ILE119 | Hydrophobic | Alkyl | ||||
A:ILE119 | Hydrophobic | Alkyl | ||||
A:PHE100 | Hydrophobic | Alkyl | ||||
Hydrophobic | Pi-Alkyl | |||||
Analog-2 | −6.91 kcal/mol | 8.66 uM (micromolar) | A:TYR85 | Hydrogen Bond | Conventional Hydrogen Bond | |
A:GLU122 | Hydrogen Bond | Conventional Hydrogen Bond | ||||
A:GLU122 | Hydrogen Bond | Conventional Hydrogen Bond | ||||
A:GLU122 | Hydrogen Bond | Conventional Hydrogen Bond | ||||
A:ILE54 | Hydrophobic | Pi-Sigma | ||||
A:ILE54 | Hydrophobic | Pi-Sigma | ||||
A:ILE54 A:ILE54 | Hydrophobic | Alkyl | ||||
A:ILE119 | Hydrophobic | Pi-Alkyl | ||||
A:ALA98 | Hydrophobic | Pi-Alkyl | ||||
A:ILE119 | Hydrophobic | Pi-Alkyl | ||||
Hydrophobic | Pi-Alkyl | |||||
Analog-3 | −7.28 kcal/mol | 4.60 uM (micromolar) | A:GLU122 | Hydrogen Bond | Conventional Hydrogen Bond | |
A:GLU122 A:GLU122 | Hydrogen Bond | Conventional Hydrogen Bond | ||||
A:ILE119 A:ILE54 | Hydrogen Bond | Conventional Hydrogen Bond | ||||
A:LYS118 | Hydrophobic | Alkyl | ||||
A:LYS118 | Hydrophobic | Alkyl | ||||
Hydrophobic | Pi-Alkyl | |||||
Hydrophobic | Pi-Alkyl | |||||
Analog-4 | −6.06 kcal/mol | 36.01 uM (micromolar) | A:ASP53 | Electrostatic | Attractive Charge | |
A:ASP53 | Hydrogen Bond | Conventional Hydrogen Bond | ||||
A:ASP53 | Hydrogen Bond | Conventional Hydrogen Bond | ||||
A:ILE54 | Hydrophobic | Pi-Sigma | ||||
A:ILE54 | Hydrophobic | Alkyl | ||||
A:PHE100 | Hydrophobic | Pi-Alkyl | ||||
A:ILE54 | Hydrophobic | Pi-Alkyl | ||||
A:VAL55 | Hydrophobic | Pi-Alkyl | ||||
Analog-5 | −6.64 kcal/mol | 13.63 uM (micromolar) | A:GLU122 A:GLU122 | Hydrogen Bond | Conventional Hydrogen Bond | |
A:TYR85 | Hydrogen Bond | Conventional Hydrogen Bond | ||||
A:GLU122 | Hydrogen Bond | Conventional Hydrogen Bond | ||||
A:PHE100 A:LEU115 | Hydrogen Bond | Conventional Hydrogen Bond | ||||
A:ILE119 | Hydrophobic | Pi-Pi T-shaped | ||||
A:ILE54 | Hydrophobic | Alkyl | ||||
A:PHE100 | Hydrophobic | Alkyl | ||||
Hydrophobic | Alkyl | |||||
Hydrophobic | Pi-Alkyl | |||||
Analog-6 | −6.65 kcal/mol | 13.31 uM (micromolar) | A:LYS118 A:ASP53 | Hydrogen Bond | Conventional Hydrogen Bond | |
A:LYS118 | Hydrogen Bond | Conventional Hydrogen Bond | ||||
A:LYS118 | Hydrogen Bond | Pi-Cation | ||||
A:ILE54 | Electrostatic | Pi-Donor Hydrogen Bond | ||||
A:ILE119 | Hydrophobic | Alkyl | ||||
A:PHE100 | Hydrophobic | Alkyl | ||||
A:PHE100 | Hydrophobic | Pi-Alkyl | ||||
A:LYS118 | Hydrophobic | Pi-Alkyl | ||||
A:ILE121 | Hydrophobic | Pi-Alkyl | ||||
A:LYS118 | Hydrophobic | Pi-Alkyl | ||||
A:ILE121 | Hydrophobic | Pi-Alkyl | ||||
Hydrophobic | Pi-Alkyl |
Based on previous studies conducted on the original, unmodified pipecolisporin, this compound exhibited its highest binding affinity towards the 2BL9 target, with a value of −10.26 kcal/mol and an inhibition constant of 29.90 nM, indicating strong inhibitory potential. However, for the 1CET and 4ZL4 targets, pipecolisporin showed lower binding affinities of −6.59 kcal/mol and −5.38 kcal/mol, respectively (
When compared with previously studied small molecules, analog-3 demonstrated superior binding affinity, particularly against 2BL9. Studies have shown that some small molecules exhibited binding affinities of −8.70 kcal/mol to −8.50 kcal/mol for 2BL9, which are significantly weaker than Analog-3’s binding affinity of −13.02 kcal/mol. This reinforces analog-3 as a highly potent dihydrofolate reductase inhibitor. However, against 4ZL4, certain small molecules outperformed analog-3, with reported binding affinities ranging from −9.60 kcal/mol to −8.30 kcal/mol, compared to analog-3’s −8.07 kcal/mol. Similarly, for 1CET, analog-3 (−13.02 kcal/mol) exhibited a stronger binding affinity than chloroquine (−6.30 kcal/mol) but was weaker than some other reported small molecules, which had affinities of −9.10 kcal/mol and −7.90 kcal/mol (
Fig.
In each visualization, the position of the ligand within the receptor’s binding pocket is clearly depicted, highlighting how analog-3 interacts with the surrounding amino acids. The visible hydrogen bonds help stabilize the ligand-receptor interaction, while hydrophobic interactions support the ligand’s affinity for the protein. In the complex with 2BL9, analog-3 exhibits several strong hydrophobic interactions, particularly with residues LEU45 and MET54. In the interaction with 4ZL4, hydrogen bonding is more dominant, providing a balance between stability and flexibility of the complex. The visualization for 1CET shows a combination of hydrogen and hydrophobic bonds, offering a comprehensive view of how analog-3 modulates binding in various protein environments. This visualization is crucial for understanding molecular binding mechanisms and guiding further drug development.
Following the assessment of protein-ligand interactions and the achievement of favorable results, molecular dynamics (MD) analysis was performed on the ligands exhibiting the highest binding affinities to evaluate the stability of the protein-ligand complexes (
The RMSD of the protein backbone was tracked throughout the 500 ns simulation to assess the structural stability and fluctuations (Fig.
The RMSF analysis provides insights into residue flexibility across the 500 ns simulation, highlighting key differences between analog-2 and analog-3 complexes. In the analog-2–2BL9 complex, fluctuations peaked around residue 150 at 0.5 nm, whereas analog-3–2BL9 exhibited lower fluctuations, suggesting better stability. Similarly, analog-2–4ZL4 showed high RMSF values near residues 50, 200, and 400, while analog-3–4ZL4 had a smoother fluctuation pattern. For analog-2–1CET, a notable peak around residue 250 reached nearly 0.45 nm, whereas analog-3–1CET displayed more uniform fluctuations. In all cases, analog-3 derivatives exhibited reduced fluctuations, indicating stronger binding stability. The consistently lower RMSF values in analog-3 complexes support the RMSD findings, reinforcing their structural rigidity. High RMSF peaks in analog-2 suggest flexible regions or surface-exposed loops that may impact binding efficiency. These variations in residue flexibility further emphasize differences in the dynamic behavior of the two analog series.
The radius of gyration (Rg) values were analyzed to assess the compactness and structural flexibility of the protein-ligand complexes over the simulation time (Fig.
Solvent-accessible surface area (SASA) values were also plotted over time to evaluate the exposure of the complex to the solvent (Fig.
The MM-PBSA approach was utilized to evaluate the interactions and binding energies between pipecolisporin compounds and their receptor targets. These simulations provided an estimation of how analog-2 and analog-3 bind with three receptors: 2BL9, 4ZL4, and 1CET (Table
Interaction energies and binding energies with various receptor targets.
Receptor Target | Pipecolisporin Compound | van der Waal Energy | Electrostattic Energy | Polar Solvation Energy | SASA Energy | Binding Energy |
---|---|---|---|---|---|---|
2BL9 | Analog-2 | –239.972 +/– 26.886 kJ/mol | –35.206 +/– 17.243 kJ/mol | 167.286 +/– 28.002 kJ/mol | –27.279 +/– 2.247 kJ/mol | –135.171 +/– 16.456 kJ/mol |
Analog-3 | –245.322 +/– 19.641 kJ/mol | –62.626 +/– 12.899 kJ/mol | 204.279 +/– 30.805 kJ/mol | –26.384 +/– 1.460 kJ/mol | –130.053 +/– 16.713 kJ/mol | |
4ZL4 | Analog-2 | –166.391 +/ 19.719 kJ/mol | –20.319 +/ 22.485 kJ/mol | 128.862 +/– 29.736 kJ/mol | –19.418 +/– 2.123 kJ/mol | –77.266 +/– 20.757 kJ/mol |
Analog-3 | –117.754 +/– 28.406 kJ/mol | –31.247 +/– 30.912 kJ/mol | 87.962 +/– 31.182 kJ/mol | –13.145 +/– 3.343 kJ/mol | –74.184 +/– 28.429 kJ/mol | |
1CET | Analog-2 | –138.726 +/– 32.181 kJ/mol | –33.369 +/– 19.713 kJ/mol | 100.347 +/– 42.146 kJ/mol | –16.704 +/– 3.280 kJ/mol | –88.453 +/– 30.267 kJ/mol |
Analog-3 | –105.654 +/– 31.441 kJ/mol | –26.064 +/– 22.681 kJ/mol | 85.468 +/– 42.486 kJ/mol | –13.39 +/– 3.880 kJ/mol | –59.645 +/– 36.700 kJ/mol |
Additional energy factors, such as van der Waals, electrostatic, polar solvation, and SASA energy, highlight the different interaction mechanisms. For example, the electrostatic interaction for analog-3 with 2BL9, at −62.626 kJ/mol, is notably stronger than that of analog-2 at −35.206 kJ/mol. The MM-PBSA analysis adds depth to molecular docking by offering a clearer and more precise calculation of binding energies, incorporating solvation effects and flexibility of the compounds. The binding energy calculations show that both analog-2 and analog-3 form highly stable complexes with the receptors, positioning them as promising leads for further drug development, especially considering their strong interactions across different receptor targets.
In terms of pharmacological properties, all the analogs violate Lipinski’s rule of five, with common violations related to high molecular weight (MW>500) and an excessive number of hydrogen bond donors and acceptors (Table
Parameters | Analog-1 | Analog-2 | Analog-3 | Analog-4 | Analog-5 | Analog-6 |
---|---|---|---|---|---|---|
Physiochemical parameters | ||||||
Formula | C37H53N7O6 | C37H54N8O6 | C40H52N8O6 | C37H54N10O6 | C37H54N8O6 | C37H54N8O6 |
Molecular weight | 691.86 g/mol | 706.87 g/mol | 740.89 g/mol | 734.89 g/mol | 706.87 g/mol | 706.87 g/mol |
Number heavy atoms | 50 | 51 | 54 | 53 | 51 | 51 |
Number aromatic heavy atoms | 9 | 9 | 15 | 9 | 9 | 9 |
Fraction Csp3 | 0.62 | 0.62 | 0.50 | 0.59 | 0.62 | 0.62 |
Number rotatable bonds | 6 | 8 | 8 | 9 | 8 | 8 |
Number H-bond acceptors | 6 | 7 | 7 | 7 | 7 | 7 |
Num. H-bond donors | 5 | 6 | 6 | 8 | 6 | 6 |
Molar Refractivity | 213.21 | 215.91 | 225.98 | 222.31 | 215.91 | 215.91 |
TPSA | 172.81 Ų | 198.83 Ų | 198.83 Ų | 234.71 Ų | 198.83 Ų | 198.83 Ų |
Pharmacological parameters | Analog-1 | Analog-2 | Analog-3 | Analog-4 | Analog-5 | Analog-6 |
Lipinski | No; 2 violations: MW>500, NorO>10 | No; 3 violations: MW>500, NorO>10, NHorOH>5 | No; 3 violations: MW>500, NorO>10, NHorOH>5 | No; 3 violations: MW>500, NorO>10, NHorOH>5 | No; 3 violations: MW>500, NorO>10, NHorOH>5 | No; 3 violations: MW>500, NorO>10, NHorOH>5 |
Ghose | No; 3 violations: MW>480, MR>130, #atoms>70 | No; 4 violations: MW>480, WLOGP<-0.4, MR>130, #atoms>70 | No; 4 violations: MW>480, WLOGP<-0.4, MR>130, #atoms>70 | No; 4 violations: MW>480, WLOGP<-0.4, MR>130, #atoms>70 | No; 4 violations: MW>480, WLOGP<-0.4, MR>130, #atoms>70 | No; 4 violations: MW>480, WLOGP<-0.4, MR>130, #atoms>70 |
Veber | No; 1 violation: TPSA>140 | No; 1 violation: TPSA>140 | No; 1 violation: TPSA>140 | No; 1 violation: TPSA>140 | No; 1 violation: TPSA>140 | No; 1 violation: TPSA>140 |
Egan | No; 1 violation: TPSA>131.6 | No; 1 violation: TPSA>131.6 | No; 1 violation: TPSA>131.6 | No; 1 violation: TPSA>131.6 | No; 1 violation: TPSA>131.6 | No; 1 violation: TPSA>131.6 |
Muegge | No; 2 violations: MW>600, TPSA>150 | No; 3 violations: MW>600, TPSA>150, H-don>5 | No; 3 violations: MW>600, TPSA>150, H-don>5 | No; 3 violations: MW>600, TPSA>150, H-don>5 | No; 3 violations: MW>600, TPSA>150, H-don>5 | No; 3 violations: MW>600, TPSA>150, H-don>5 |
Bioavailability Score | 0.17 | 0.17 | 0.17 | 0.17 | 0.17 | 0.17 |
PAINS | 0 alert | 0 alert | 0 alert | 0 alert | 0 alert | 0 alert |
Brenk | 0 alert | 0 alert | 0 alert | 2 alerts: imine_1, imine_2 | 0 alert | 0 alert |
Leadlikeness | No; 2 violations: MW>350, XLOGP3>3.5 | No; 2 violations: MW>350, Rotors>7 | No; 2 violations: MW>350, Rotors>7 | No; 2 violations: MW>350, Rotors>7 | No; 2 violations: MW>350, Rotors>7 | No; 2 violations: MW>350, Rotors>7 |
Synthetic accessibility | 6.55 | 6.62 | 6.56 | 6.82 | 6.52 | 6.55 |
The toxicity profiles of the pipecolisporin analogs (analog-1 through analog-6) reveal significant insights regarding their safety and potential pharmacological effects (Fig.
From the pharmacokinetic and toxicity evaluation, all analogs showed low gastrointestinal absorption, an inability to cross the blood-brain barrier, and functioned as P-gp substrates, which could affect bioavailability. Although they are not hepatotoxic or cardiotoxic, all analogs have neurotoxic potential and toxicity effects on the respiratory and immune systems. Their biological activity against targets such as GPCRs, ion channel modulators, and kinases showed low potential, although some analogs exhibited slight activity as protease inhibitors. This aligns with predictions made using Molinspiration’s Bioavailability Suite, which utilizes chemical descriptors and machine learning/QSAR models to predict bioactivity scores. Based on the data in Table
Parameters | Analog-1 | Analog-2 | Analog-3 | Analog-4 | Analog-5 | Analog-6 |
---|---|---|---|---|---|---|
ADME | ||||||
GI absorption | Low | Low | Low | Low | Low | Low |
BBB permeant | No | No | No | No | No | No |
P-gp substrate | Yes | Yes | Yes | Yes | Yes | Yes |
CYP1A2 inhibitor | No | No | No | No | No | No |
CYP2C19 inhibitor | No | No | No | No | No | No |
CYP2C9 inhibitor | No | No | No | No | No | No |
CYP2D6 inhibitor | No | No | No | No | No | No |
CYP3A4 inhibitor | Yes | Yes | Yes | Yes | Yes | Yes |
Log Kp (skin permeation) | −7.82 cm/s | −9.37 cm/s | −9.39 cm/s | −9.96 cm/s | −9.37 cm/s | −9.01 cm/s |
Consensus Log Po/w | 1.97 | 0.81 | 1.08 | 0.44 | 1.05 | 1.04 |
Log S (ESOL) | −6.26 | −4.93 | −5.38 | −4.66 | −4.93 | −5.24 |
Toxicity | Analog-1 | Analog-2 | Analog-3 | Analog-4 | Analog-5 | Analog-6 |
Hepatotoxicity | Inactive | Inactive | Inactive | Inactive | Inactive | Inactive |
Neurotoxicity | Active | Active | Active | Active | Active | Active |
Nephrotoxicity | Inactive | Inactive | Inactive | Inactive | Inactive | Inactive |
Respiratory toxicity | Active | Active | Active | Active | Active | Active |
Cardiotoxicity | Inactive | Inactive | Inactive | Inactive | Inactive | Inactive |
Carcinogenicity | Inactive | Inactive | Inactive | Inactive | Inactive | Inactive |
Immunotoxicity | Active | Active | Active | Active | Active | Active |
Mutagenicity | Inactive | Inactive | Inactive | Inactive | Inactive | Inactive |
Cytotoxicity | Inactive | Inactive | Inactive | Inactive | Inactive | Inactive |
BBB-barrier | Inactive | Inactive | Inactive | Inactive | Inactive | Inactive |
Ecotoxicity | Inactive | Inactive | Inactive | Inactive | Inactive | Inactive |
Clinical toxicity | Active | Active | Active | Active | Active | Active |
Nutritional toxicity | Active | Inactive | Inactive | Inactive | Active | Active |
Bioactivity | Analog-1 | Analog-2 | Analog-3 | Analog-4 | Analog-5 | Analog-6 |
GPCR ligand | −0.24 | −0.32 | −0.76 | −0.55 | −0.29 | −0.30 |
Ion channel modulator | −1.30 | −1.46 | −2.04 | −1.81 | −1.41 | −1.39 |
Kinase inhibitor | −0.99 | −1.10 | −1.62 | −1.51 | −1.07 | −1.05 |
Nuclear receptor ligand | −1.28 | −1.49 | −1.97 | −2.00 | −1.37 | −1.35 |
Protease inhibitor | 0.23 | 0.16 | −0.27 | 0.04 | 0.16 | 0.16 |
Enzyme inhibitor | −0.76 | −0.85 | −1.38 | −1.17 | −0.86 | −0.87 |
The chart in Fig.
Pipecolisporin analogs were evaluated for their therapeutic potential, particularly in terms of binding affinity, stability, pharmacokinetics, and toxicity. Like the ongoing global efforts to combat malaria, where new compounds are continuously sought to inhibit Plasmodium spp., these analogs offer promise as potential therapeutic agents for diseases where enzyme inhibition is critical (
Molecular dynamics simulations further confirmed that analog-3 formed highly stable complexes with the target proteins, particularly 2BL9 and 4ZL4. The root mean square deviation (RMSD) analysis showed that analog-3 complexes remained stable throughout the 500 ns simulation, while analog-2 exhibited greater fluctuations. Similarly, the root mean square fluctuation (RMSF) results indicated that analog-3 had lower residue fluctuations, suggesting stronger binding interactions and structural rigidity. The radius of gyration (Rg) analysis showed that analog-3 maintained a more compact structure, reinforcing its stability over time. In addition, solvent-accessible surface area (SASA) analysis revealed that analog-3 complexes had lower exposure to solvents, further confirming their structural integrity. These findings demonstrated that analog-3 is more stable than analog-2, which supports its potential as a more effective inhibitor. The combined docking and MD results suggest that analog-3 could be a promising candidate for enzyme inhibition applications.
The binding free energy (MM-PBSA) analysis provided further validation of the strong interactions between analog-3 and its target proteins. The results showed that analog-3 consistently exhibited lower binding energy values compared to analog-2, indicating stronger ligand-protein binding. The electrostatic interactions of analog-3–2BL9 were significantly stronger, contributing to its stability in the binding pocket. Additionally, van der Waals forces played an important role in stabilizing the ligand-protein complexes, particularly in analog-3–4ZL4 interactions. The MM-PBSA calculations confirmed that analog-3 formed more stable complexes, which correlated with the findings from docking and MD simulations. These results reinforce the hypothesis that analog-3 has a high potential for enzyme inhibition due to its strong and stable interactions with key residues. However, despite these promising findings, binding energy alone is not sufficient to determine drug-like properties (
The absorption, distribution, metabolism, and excretion (ADME) analysis indicated that all pipecolisporin analogs exhibited poor gastrointestinal (GI) absorption, which could limit their potential for oral administration. Additionally, none of the analogs were able to penetrate the blood-brain barrier (BBB), suggesting that they may not be effective for central nervous system (CNS) applications. Furthermore, all analogs were identified as P-gp substrates, which means they could be actively transported out of cells, potentially reducing their intracellular concentrations. On the other hand, the toxicity analysis showed that none of the analogs exhibited hepatotoxicity or cardiotoxicity, indicating a relatively safe profile in terms of liver and heart toxicity. However, neurotoxicity, respiratory toxicity, and immunotoxicity were detected in all analogs, which could pose challenges in drug development. These findings suggest that structural modifications will be necessary to enhance the pharmacokinetic properties and reduce potential toxicity risks (
Despite the pharmacokinetic challenges, analog-3 exhibited promising enzyme inhibition activity, particularly as a potential protease inhibitor. The biological activity assessment showed that analog-3 had weak activity as a GPCR ligand, ion channel modulator, and kinase inhibitor, but it demonstrated slight protease inhibition potential. This suggests that analog-3 could still be valuable in targeting protease-related diseases, despite its pharmacokinetic drawbacks. The results indicate that structural optimization efforts should focus on improving bioavailability while maintaining strong binding affinity. Future studies should explore chemical modifications to enhance GI absorption and minimize toxicity, which could make analog-3 a viable drug candidate. Finally, analog-3 was identified as the most promising compound, exhibiting the best binding affinity, stability, and interaction strength among all tested pipecolisporin derivatives. However, to develop it into a potential drug, further structural refinements and experimental validation are necessary to overcome pharmacokinetic and toxicity challenges.
This study highlights the potential of pipecolisporin analogs as enzyme inhibitors, particularly in their interaction with protease targets. Among the six analogs, analog-3 exhibited the most promising activity, demonstrating strong binding affinities and stable interactions in molecular dynamics simulations, especially with the 2BL9 and 4ZL4 proteins. However, the pharmacokinetic analysis revealed poor gastrointestinal absorption and limited bioavailability due to P-gp substrate activity, indicating challenges for oral administration. While the analogs showed no hepatotoxicity or cardiotoxicity, the presence of neurotoxicity and respiratory toxicity raises concerns that must be addressed. Despite these limitations, analog-3, with further optimization and toxicity reduction, possesses the necessary characteristics to be explored as a potential therapeutic agent. These findings suggest that pipecolisporin analogs, with structural improvements, could be promising candidates for the development of enzyme-targeting therapies.
Authors would like to thank the research grants of RIIM-BRIN 2023-2024 for research financial support and Universitas Islam Bandung for the APC.
Conflict of interest
The authors have declared that no competing interests exist.
Ethical statements
The authors declared that no clinical trials were used in the present study.
The authors declared that no experiments on humans or human tissues were performed for the present study.
The authors declared that no informed consent was obtained from the humans, donors or donors’ representatives participating in the study.
The authors declared that no experiments on animals were performed for the present study.
The authors declared that no commercially available immortalised human and animal cell lines were used in the present study.
Funding
This work was supported by Universitas Padjadjaran and Universitas Islam Bandung.
Author contributions
NK: performed computational experiments, collected and analyzed data, and drafted the manuscript; TMF: performed computational experiments, collected and analyzed data; RM: supervised, analyzed data, and revised the manuscript; US: supervised and revised the manuscript; ATH: supervised and revised the manuscript.
Author ORCIDs
Nety Kurniaty https://orcid.org/0000-0002-4519-8327
Taufik Muhammad Fakih https://orcid.org/0000-0001-7155-4412
Rani Maharani https://orcid.org/0000-0001-8156-9773
Data availability
All of the data that support the findings of this study are available in the main text.