Home Physical Sciences FMS-like tyrosine kinase 3 inhibitory potentials of some phytochemicals from anti-leukemic plants using computational chemical methodologies
Article Open Access

FMS-like tyrosine kinase 3 inhibitory potentials of some phytochemicals from anti-leukemic plants using computational chemical methodologies

  • Haruna Isiyaku Umar EMAIL logo , Zainab Ashimiyu-Abdusalam , Marwa Alaqarbeh , Wanche Ernest Magani , Omoboyede Victor , Rukayat Yetunde Omotosho-Sanni , Ridwan Opeyemi Bello , Yousef A. Bin Jardan , Samir Ibenmoussa , Mohammed Bourhia , Gezahign Fentahun Wondmie EMAIL logo and Mohammed Bouachrine
Published/Copyright: June 18, 2024

Abstract

Acute myeloid leukemia (AML) takes center stage as a highly prevalent and aggressive clonal disorder affecting hematological stem cells. FMS-like tyrosine kinase 3 (FLT3) mutations were prevalent in nearly 30% of the AML cases. However, efforts have led to the development of anti-mutant FLT3 drugs, such as midostaurin, gilteritinib, and quizartinib, to improve treatments. Currently, we are exploring the ability of compounds from anti-leukemic plants to be used in AML therapies, focusing on mutant FLT3 inhibition. Employing computational techniques such as drug-likeness assessment, molecular docking, pharmacokinetics properties profiling, molecular dynamics simulations (MDS), and free energy calculations, we identified 43 out of 57 compounds with oral drug potential. Notably, 7 out of 43 compounds, including flavopiridol, sanggenol Q, norwogonin, oblongixanthones A, oblongixanthones B, apigenin, and luteolin exhibited strong binding affinities ranging from −9.0 to −9.8 kcal/mol, surpassing the control drug gilteritinib (−6.3 kcal/mol). Notably, flavopiridol and norwogonin displayed highly favorable pharmacokinetics and low toxicity profiles. MDS confirmed the stability of their binding through parameters such as root mean square deviation, root mean square fluctuation, and radius of gyration (R g) over 100 ns simulations. Flavopiridol and norwogonin emerge as promising candidates for the development of mutant FLT3 inhibitors. Therefore, experimental studies are warranted to validate their therapeutic potential.

1 Introduction

Acute myeloid leukemia (AML) is a rapidly progressing hematologic malevolence that affects the bone marrow, blood cells, and other tissues [1]. It is typified by the abnormal growth, abnormal differentiation, proliferation, and poor differentiation of immature white blood cells that cause interference with the production of normal blood cells, leading to anemia, infections, and bleeding disorders [2].

The Cancer Society outfit (American Cancer Society, ACS) in the United States forecasted that approximately 60,650 new cases of leukemia alone would surface in 2022, with AML being the most disastrous subdivision and responsible for 48% of the leukemia-associated mortalities [3].

Despite significant advances in cancer therapy, the prognosis for AML remains deprived, with a 5-year survival frequency of less than 30% [46]. In older patients, the median survival outcome is between 5 and 10 months, including individuals with comorbidities whose body systems cannot endure penetrative chemotherapy [2]. One of the significant challenges in AML treatment is the development of drug resistance, which limits the effectiveness of conventional therapies such as chemotherapy and radiotherapy. Therefore, there is a need for novel and effective therapies that target specific molecular pathways involved in AML.

FMS-like tyrosine kinase 3 (FLT3) is a tyrosine kinase receptor that plays a critical role in normal hematopoiesis, the process by which blood cells are molded. It is overexpressed in up to 70% of the AML cases, and its activation has been linked to pathogenesis and progression of the disease. FLT3 mutations, particularly internal tandem duplications, and tyrosine kinase domain mutations, are frequent in AML and are connected with humble prognosis and drug resistance. Therefore, FLT3 has emerged as a viable therapeutic target in AML [7]. Several FLT3 inhibitors have been developed for AML treatment, including midostaurin, gilteritinib, and quizartinib. However, these drugs have shown limited efficacy and significant toxicities, and drug resistance remains a major challenge. Therefore, there is a necessity for the development of innovative and more effectual FLT3 inhibitors that can overcome drug resistance and reduce toxicity [8].

Plants have long been the go-to material in old-style medicine for the treatment of several diseases, cancer inclusive. Many associated plant compounds have been established to possess anti-cancer properties, and some have shown promise in AML treatment. For example, resveratrol, a polyphenol found in grapes and red wine, has been exposed to counter FLT3 activity and prompt apoptosis in AML cells [9]. Curcumin, a compound found in turmeric, has also been found to have anti-cancer properties and inhibits FLT3 signaling in AML cells [10].

Recently, there has been a growing interest in using natural plant products as a source of novel anti-cancer agents, particularly for treating AML. Natural products have several advantages over synthetic compounds, including their structural diversity, potential for multi-targeted activity, and low toxicity [11]. Furthermore, the use of natural products in cancer therapy has a long history of successful outcomes, including the discovery of taxanes, vinca alkaloids, and anthracyclines [12].

In recent years, computational drug discovery has arisen as an influential tool for the identification and optimization of unique drug candidates. Computer-based approaches that comprise molecular docking, virtual screening, and molecular dynamics simulations (MDS) [1316] have been expanded to forecast the binding affinity and efficacy of potential FLT3 inhibitors. It is interesting to note that this technique can aid in the optimization of potential drugs, leading to their approvals being granted. Instances of some licensed drugs that were optimized using CADD are captopril, dorzolamide, oseltamivir, aliskiren, and nolatrexed [13,17,18].

The aim of this research is to investigate the potential of selected compounds from anti-leukemic plants as an anti-cancer therapy for AML, targeting the FLT3 protein using computer-enabled techniques such as drug-likeness, molecular docking, in silico pharmacokinetics profiling, MDS, and free energy calculations. The ultimate goal of the research is to identify novel and effective natural plant-derived compounds for treating AML that can overcome drug resistance and reduce toxicity.

2 Materials and methods

2.1 Ligand data sets

Fifty-seven bioactive compounds from 29 plants reported to possess anti-leukemic potential were chosen from the literature [1922]. The control drug chosen for this study is gilteritinib, which is an approved medication in the treatment of degenerated/obstinate (R/R) AML with an FLT3 mutation. Table S1 illustrates the list of bioactive compounds, source plants, their respective PubChem IDs, and canonical smiles.

2.2 Selection and preparation of the protein target

The three-dimensional (3D) structure of FLT3 co-crystallized with gilteritinib (PDB ID: 6JQR) at 2.20 Å resolution [23] was accessed through the Protein Database (PDB) (http://www.pdb.org/pdb). This structure was selected based on the fact that it was co-crystalized with a known approved drug for AML, low resolution, and recently submitted to the protein database. The structure of the target protein is shown in Figure 1. The preparation for docking and minimization of the protein was achieved through the dock prep module of UCSF-Chimera software. The proteins were liberated from all heteroatoms, such as co-crystallized ligands and water molecules. Additionally, charges and hydrogen atoms using gasteiger charge were added. Finally, minimization was achieved by employing the amber force field 94 fs (Amberff94fs) [24].

Figure 1 
                  3D structure of FLT3 bound to gilteritinib. FLT3 is presented in a cartoon with its α-helices (red), loops (green), and β-sheets (yellow). Gilteritinib is shown in purple, occupying the binding pocket.
Figure 1

3D structure of FLT3 bound to gilteritinib. FLT3 is presented in a cartoon with its α-helices (red), loops (green), and β-sheets (yellow). Gilteritinib is shown in purple, occupying the binding pocket.

2.3 Screening for drug-like potentials

Fifty-seven bioactive compounds were sieved for druglikeness employing three rules, viz: Lipinski’s [25], Veber’s [26], and Egan’s [27], and bioavailability score via the SwissAdme website [28]. Thus, compounds with no more than one violation of the three rules and a bioavailability score ≥0.55 are deemed fit for molecular docking against FLT3.

2.4 Retrieval and preparation of ligands for molecular docking against FLT3

The 3D coordinates in the structure data file of gilteritinib (control drug) and 43 [42] compounds were retrieved from the chemical virtual store accommodated in the National Centre for Biotechnological Information called PubChem (PubChem (nih.gov)), which is a global leading collection of freely obtainable cheminformatics records. Likewise, the acquired compound structures were transformed into the best energetic and steady conformations choosing the Merck molecular force field (MMFF94) [29]; in addition, the optimization’s system, conjugate gradient, by the use of the Open babel icon in Python Prescription suite (version 0.8).

2.5 Computer-assisted docking against FLT3

The docking steps were executed with the aid of AutoDock Vina, accommodated in open-source Python Prescription 0.8 [30] to secure probable binding geometries and binding energies (BEs) of compounds at the designated binding pocket of FLT3. The pocket was enclosed by adjusting the grid box with magnitudes (16.7026 × 16.8947 × 20.4031) Å, and the center was adjusted in line with the site of gilteritinib binding in the FLT3 binding cavity. The cavity comprises Leu818, Phe830, Ala642, Gly697, Tyr693, Leu616, Cys694, Glu692, and Asp829 [23]. After the docking simulation run, docking score (BE) of compounds below −9.0 and gilteritinib were submitted for molecular visualization procedures in order to explore their binding positions (3D) and molecular contact patterns (2D) via PyMOL© Molecular Graphics (version 2.4, 2016, Schrödinger LLC) [31] and Maestro 11.1’s Ligand interaction option (Schrödinger 2017 ver.), respectively.

2.6 Prediction of in silico pharmacokinetics profiling

Absorption, distribution, metabolism, excretion, and toxicity (ADMET) is imperative during the penultimate phase of drug discovery and design conduit to scrutinize the pharmacokinetics and pharmacodynamics of the projected compounds with the potential of becoming a drug. Here, we employed ADMETSar server (http://lmmd.ecust.edu.cn/) to forecast the ADMET properties of the compounds with the best hits post-molecular docking analysis [32,33]. The web server was served with the simplified molecular-input line-entry system (SMILE) strings of the compounds from PubChem (https://pubchem.ncbi.nlm.nih.gov/compound/) through the search space and executed for ADMET features prediction.

2.7 Molecular dynamics simulations

The firmness of the protein–ligand (FLT3 with flavopiridol, norwogonin, and sanggenol Q) interactions was evaluated by performing MD calculations on the optimal docking postures. GROMACS 2020.2 software was deployed for the equilibration and production run stages of all MD simulations. The CHARMM36 forcefield was deployed to simulate the protein–ligand combination. The system was subsequently subjected to a constant number of particles, volume, and temperature and a constant number of particles, pressure, and temperature (NPT) ensembles to stabilize its temperature and pressure. It was simulated for a duration of 125 ps at a temperature of 300.15 K, with positional restrictions of 400 and 40 kJ/mol nm2 for the backbone and side chains, respectively. The complex is ultimately exposed to a production simulation lasting 100 ns, conducted within an NPT ensemble at a temperature of 300.15 K and a pressure of 1 bar.

2.8 Trajectory analysis

The GROMACS program was employed to perform MD simulations. The root mean square deviation (RMSD) of atom positions was determined for both the ligands and FLT3 protein after fitting the FLT3 backbone using the gmx_rms subprogram. The RMSF was computed using the gmx_rmsf subprogram, based on the FLT3 C-alpha atoms. The R g of all FLT3 atoms was calculated using the gmx_gyrate subprogram. The gmx_hbond subprogram was utilized to evaluate hydrogen bonds within the protein–ligand interface. The mass distance between FLT3 and the ligands was quantified during the simulation using the gmx_distance subprogram. Finally, the visual molecular dynamics (VMD) molecular graphics program was utilized for trajectory visualization and protein–ligand contact frequency analysis.

2.9 Molecular mechanics Poisson–Boltzmann surface area (MMPBSA) calculations

The systems selected for further research underwent MMPBSA calculations using g_mmpbsa, a computational tool within the GROMACS software package, which is employed to determine the binding affinity. In a broad context, the thermodynamic quantity known as the binding free energy, which characterizes the strength of interaction between the FLT3 and its ligand in a solvent, can be mathematically represented as

Δ G bindin g = Δ G complex ( Δ G protein + Δ G ligand ) .

G complex is the total free energy of the protein–ligand complex, and ∆G protein is the total free energy of the isolated FLT3 and ligands in a solvent. By individually calculating the energy values for each residue and then adding them together, it was possible to determine the BE contribution of each residue. Given the limited compatibility of g_mmpbsa with specific GROMACS versions, it was necessary to recreate the binary run input file (.tpr) using GROMACS 5.1.4 to perform MMPBSA calculations with g_mmpbsa. The generation of the binary run input file required the utilization of three essential files: the molecular structure file (.gro), the topology file (.top), and the MD-parameter file (.mdp). These files were obtained via the MD process.

3 Results and discussions

AML is a form of leukemia that has high morbidity and mortality among adults and continues to be a major source of concern due to its poor prognosis and as a result of the limited efficacy of the regimens that are used in its management and treatment [5]. Consequently, the search for newer and alternative classes of anti-AML drugs remains unfaltering. To this end, this study aimed to identify potential inhibitors of FLT3 from plants with reported anti-leukemic activities.

In total, 57 bioactive compounds from 29 medicinal plants with anti-leukemic activities were identified and retrieved from the PubChem database. These compounds included alkaloids, ansamycins, flavonoids, terpenes, and phytosterols; following their retrieval, they were subjected to druglikeness evaluation aimed at identifying their fitness for use as oral drugs based on the rule of five (Ro5), Veber, and Egan. The Ro5 evaluates the fitness of small molecules to serve as drugs based on parameters, including the molecular weight <500 Da, number of hydrogen bond donors <10, number of hydrogen acceptors <5, and octanol–water partition coefficient <5. The violation of more than one of the previously mentioned parameters may render small molecules unfit for use as oral drugs. Similarly, Veber and Egan’s rules are considered a subset of the Ro5 due to overlapping parameters of the rules.

The screening identified 43 compounds out of the 57 compounds as having oral drug potential (Table S2). Following subjecting the compounds to the filtering criteria, the compounds with oral drug potentials were identified and subsequently subjected to a preparation pipeline aimed at making them fit for molecular docking. These included the addition of explicit hydrogens and energy minimization among others. The energy minimization of compounds is an essential step in the preparatory pipeline that compounds are subjected to prior molecular docking, which involves the optimization of the compound structures by adjusting bond lengths, angles, and torsion angles to reach a stable conformation [34,35]. This optimization enhances the accuracy of molecular docking predictions by providing more reliable starting structures for the docking simulations. It is worth noting that there exist several approaches to compound minimization, which include the molecular mechanics and the quantum mechanical approaches that utilize methods, including the semi-empirical method, density functional theory (DFT) method, and the ab initio methods. The molecular mechanics methods are commonly the go-to approach in the screening of compound libraries due to their balance between accuracy, computational power, and time. Similarly, the semi-empirical methods also provide a desirable balance between accuracy and required computational power, and they are commonly used in quantitative structure–activity relationship modeling and other drug discovery approach [36,37]. Compared to the DFT method, which requires high computing power, semi-empirical methods have been reported to produce DFT-level accuracy and exhibit excellent correlation with the experimental results [38,39]. It is worth noting that the molecular mechanics approach was used in this study, and they are widely used in molecular modeling for drug discovery due to their accuracy and time effectiveness [40,41]. Similarly, the structure of the FLT3 protein was also retrieved and prepared using a previously mentioned approach.

MDS of the compounds against the protein was performed to identify the hit compounds against the protein with gilteritinib as the control drug. Interestingly, the compounds exhibited high binding affinities for the protein, as revealed by their docking scores that ranged from −5.1 to −9.8 kcal/mol (Table S3). Notably, these compounds exhibited affinities that were much higher compared to that of gilteritinib, with compounds including sanggenol Q and flavopiridol exhibiting docking scores as high as −9.8 and −9.6 kcal/mol, respectively (Table 1).

Table 1

Druglikeness and molecular docking outcomes of drug-like compounds from plants with anti-leukemic potentials against FLT3 protein

S/No. Compounds PubChem ID MW Lipinski #violations Veber #violations Egan #violations Bioavailability Score BE (kcal/mol)
1. Flavopiridol 5287969 401.84 0 0 0 0.55 −9.6
2. Sanggenol Q 11796489 422.47 0 0 0 0.55 −9.8
3 Norwogonin 5281674 270.24 0 0 0 0.55 −9.6
4. Oblongixanthones A 25209069 326.3 0 0 0 0.55 −9.3
5. Oblongixanthones B 25209201 478.58 0 0 1 0.55 −9.7
6. Apigenin 5280443 270.24 0 0 0 0.55 −9.0
7. Luteolin 5280445 286.24 0 0 0 0.55 −9.0
8. Gilteritinib 49803313 552.71 2 3 0 0.17 −6.3

To identify the potential drug candidates, the pharmacokinetic properties and toxicity profiles of the top seven hit compounds were studied (Table 2, Table S4). Notably, poor pharmacokinetics and toxicity profiles are the major reasons why most promising compounds fail in the later stage of clinical trials; hence, studying these properties has emerged as a viable means to circumvent these. Human intestinal absorption (HIA) is the process by which drugs that are orally administered are taken up from the gastrointestinal tract to the bloodstream prior to their distribution to target sites. All the selected hit compounds were predicted to be capable of being taken up into the bloodstream, and they also possessed a minimum bioavailability score of 55%, signifying their ability to be transported to the target site in high concentration upon absorption. Of all the hit compounds, only apigenin, luteolin, and oblongixanthones A were predicted to be Caco-2- permeable. While both the Caco-2 and HIA tests aim to give insights into the oral bioavailability of drugs, the HIA tests can be said to be the more accurate test due to their modalities that are based on humans as opposed to in vitro models. The P-glycoprotein serves as an efflux protein; it is known to reduce the efficacy of drugs that serve as its substrates, while its inhibition also could lead to potential drug–drug interactions on co-administration of the inhibitor with some drugs. Among the selected hit compounds in this study, flavopiridol and sanggenol Q were found to be substrates of P-glycoprotein, which indicates the potential of the compounds to be effluxed out of the cell rapidly when administered as a drug. However, the effect of this phenomenon on their efficacy can be alleviated by the administration of a higher dosage of the drug, with consideration to potential dose-induced toxicity. Similarly, oblongixanthones B and sanggenol Q were found to be potential inhibitors of P-glycoprotein. The administration of both compounds as drugs could be limited in some clinical usage due to their potential to induce drug toxicity as a result of the inhibition of the transport of some toxic drug metabolites out of the cell by this protein.

Table 2

ADMET profiling of hit plant compounds from different plants with anti-leukemic potential after molecular docking against FLT3 protein

ADMET profile Apigenin Flavopiridol Luteolin Norwoginin Oblongixanthones A Oblongixanthones B Gilteritinib Sanggenol Q
Ames mutagenesis + + +
Toxicity (class) III III II II III III III III
Blood–brain barrier + + +
Caco-2 + + +
Carcinogenicity
CYP1A2 inhibition + + + + + +
CYP2C19 inhibition + +
CYP2C9 inhibition + + + +
CYP2C9 substrate
CYP2D6 inhibition
CYP2D6 substrate
CYP3A4 inhibition + + + +
CYP3A4 substrate + + + + +
CYP inhibitory promiscuity + + + + +
Hepatotoxicity + + + + +
hERG inhibition + + + +
HIA + + + + + + + +
HOB +
Nephrotoxicity
Acute oral toxicity 1.484 2.658 1.998 2.037 2.215 2.557 2.361 1.688
P-gp inhibitor + + +
P-gp substrate + + +
PP binding 1.083 1.028 1.066 1.048 0.807 0.918 0.710 0.937
Subcellular localization Mitochondria Lysosomes Mitochondria Mitochondria Mitochondria Mitochondria Lysosomes Mitochondria
UGT catalyzed + + + + +
Water solubility −2.777 −3.299 −2.999 −2.999 −3.189 −4.524 −3.200 −4.077

The metabolism profiles of the hit compounds based on their potential to be metabolized in the phase I reaction mediated by cytochrome P450 (CYP450) enzymes were studied. The compounds were found to be metabolizable by at least one of the CYP450 isozymes involved in drug metabolism. However, some of the compounds were found to be inhibitors of some of the isozymes; hence, there must be selective administration of the compounds as drugs. Evaluation of the toxicity profiles of the hit compounds revealed oblongixanthones A and oblongixanthones B to be Ames-mutagenic; hence, they could be capable of causing potential alterations to nucleotide sequences and cause DNA damage. However, further studies revealed them non-carcinogenic; hence, the alteration caused did not give rise to molecular events that resulted in cancer development. Also, the other compounds were found to be non-carcinogenic. The potential of the compounds to serve as inhibitors of the human ether-a-go-go related gene (hERG), which codes for the potassium ion channel of the heart, was examined. Flavopiridol, oblongixanthones B, and sanggenol Q were found to be inhibitors of the hERG. This implies that the administration of the drug could lead to a condition referred to as cardiac arrhythmia. Apigenin, oblongixanthones A, oblongixanthones B, and sanggenol Q were found to be hepatotoxic; hence, their usage is dose-dependent (Figure 2).

Figure 2 
               Molecular docking poses of hit compounds and gilteritinib against FLT3 in 3D rendition. (a) Cartoon and (b) molecular surface representations of binding poses of hit compounds (in sticks) in the binding pocket of FLT3. Apigenin (green), flavopiridol (yellow), gilteritinib (blue), luteolin (cyan), norwogonin (pink), obongixanthones A (pale pink), oblongixanthones B (gray), and sanggenol Q (light green) were observed to occupy similar portions of the binding pocket.
Figure 2

Molecular docking poses of hit compounds and gilteritinib against FLT3 in 3D rendition. (a) Cartoon and (b) molecular surface representations of binding poses of hit compounds (in sticks) in the binding pocket of FLT3. Apigenin (green), flavopiridol (yellow), gilteritinib (blue), luteolin (cyan), norwogonin (pink), obongixanthones A (pale pink), oblongixanthones B (gray), and sanggenol Q (light green) were observed to occupy similar portions of the binding pocket.

After conducting molecular docking analysis, the subsequent molecular interaction assessments, facilitated through PyMOL and the ligand interaction module of Schrodinger Maestro 11.1, unveiled the robust binding of our investigated compounds within the active sites of the mutant FLT3 (Figure 3). Notably, all the identified hit compounds displayed similar interaction profiles to those of gilteritinib, the control drug. These interactions encompassed the formation of essential hydrogen bonds with Cys694, Leu616, and Gly697, as well as prominent hydrophobic interactions with Cys694, Tyr693, Phe691, Ala642, Leu818, Val675, Phe830, Val624, Leu616, and Ile62. Additionally, negative charge interactions were observed with Asp829, while positive interactions were noted with Lys644. An exception to this pattern was oblongixanthones B, which exhibited a distinctive polar interaction with Asn701. It is noteworthy that the binding pocket of the mutant FLT3 primarily comprises hydrophobic regions, reinforcing the compounds’ affinity for the active site.

Figure 3 
               Molecular docking 2D interaction analyses of hit compounds and gilteritinib against FLT3. (a) Apigenin, (b) flavopiridol, (c) gilteritinib, (d) luteolin, (e) norwogonin, (f) oblongixanthones b, and (g) sanggenol q and (h) oblongixanthones A were observed to interact with amino acid residues found along the binding pocket of FLT3 via hydrogen bonding and hydrophobic interactions.
Figure 3

Molecular docking 2D interaction analyses of hit compounds and gilteritinib against FLT3. (a) Apigenin, (b) flavopiridol, (c) gilteritinib, (d) luteolin, (e) norwogonin, (f) oblongixanthones b, and (g) sanggenol q and (h) oblongixanthones A were observed to interact with amino acid residues found along the binding pocket of FLT3 via hydrogen bonding and hydrophobic interactions.

To assess the binding stability [4245] of FLT3 protein–ligand complexes, including flavopiridol, norwogonin, and sanggenol Q, we conducted MDS at room temperature over a 100 ns duration. MDS were run for 100 ns at room temperature to evaluate the binding stability of FLT3–ligand complexes (flavopiridol, norwogonin, and sanggenol Q). After the simulation run, the trajectory data analysis showed that all ligands stayed bound to the ligand-binding groove inside the FLT3 pocket. The stability of each structure was evaluated by performing RMSD, RMSF, Rg, hydrogen bonding, average center of mass (COM) distance calculations between FLT3 and ligand, and MMPBSA calculations.

Figure 4a depicts the structure’s complex RMSD during 100 ns simulations. After a simulation time of 20 ns, it was observed that all complex RMSD curves exhibited consistently low and steady values. The complex flavopiridol exhibits slight variations and fluctuations, followed by norwogonin and sanggenol Q. Figure 5b shows all complexes’ RMSD and their corresponding curves. The RMSD curve of the flavopiridol ligand exhibits minimal variations during the 100 ns simulation.

Figure 4 
               (a) RMSD, (b) RMSF, (c) radius of gyration, and (d) hydrogen bonding of the complexes during 100 ns MDS.
Figure 4

(a) RMSD, (b) RMSF, (c) radius of gyration, and (d) hydrogen bonding of the complexes during 100 ns MDS.

Figure 5 
               (a) Ligand RMSD and (b) the average distance between ligand and the protein of the complexes during 100 ns MDS.
Figure 5

(a) Ligand RMSD and (b) the average distance between ligand and the protein of the complexes during 100 ns MDS.

Additional experimental analyses, on the other hand, confirmed that the ligand norwogonin had the lowest binding affinity among the three ligands and the highest degree of variability in the ligand RMSD curve. The radius of gyration (Figure 4c) matches the results of the RMSD analysis for the complexes. All compounds show small changes (less than 0.5 Å) during the simulation. This suggests that the protein–ligand systems maintain a compact and stable conformation throughout the simulation. Although the ligand sanggenol Q remains stable within its binding site, R g analysis indicates a gradual increase in contrast to flavopiridol and norwogonin. This suggests a protein conformation change, with the R g value ranging from 19.5 to 20.1 Å. In order to determine the RMSF of the protein complex, we analyzed the positional deviations of the “C-alpha” atoms using GROMACS software. The compound usually experiences fluctuation intensities below 2.0 Å, except for specific residues corresponding to protein regions characterized by loops or turns (Figure 4b).

Figure 4d illustrates the cumulative count of hydrogen bonds established between the ligand and protein during a 100 ns simulation. Throughout the simulation, it was observed that all ligands consistently exhibited an average of one hydrogen bond.

The mean center-of-mass distance between the ligand and protein for a 100 ns simulation period is depicted in Figure 5b. The observed data indicate that the ligands remained at their respective binding sites, as evidenced by the minimum variability of the COM distance across all systems (less than 1.5 Å). Notably, high RMSD values indicate the inability of the ligand to maximize the interaction with the binding pocket of the protein being investigated; hence, the significant instability [46,47]. In order to conduct a more comprehensive assessment of the interaction between the protein and the ligands under investigation, a contact frequency (CF) analysis was carried out. This analysis utilized the contactFreq.tcl module in VMD, employing a cutoff distance of 4 Å. Among the three, ligand sanggenol Q has the most significant quantity and proportion of ligand interactions. The residues exhibiting the most significant carbon footprint percentages are depicted in Figure 6. The residues that exhibited the most significant CF throughout all simulations were Leu616, Val624, Ala642, Glu692, Tyr693, Cys694, Gly697, Leu818, Cys828, and Phe830.

Figure 6 
               CF analysis of the complexes.
Figure 6

CF analysis of the complexes.

The MMPBSA method was chosen to re-evaluate the complexes due to its effectiveness as a force field-based approach for calculating binding free energy. This approach is particularly advantageous compared to other methods, such as free energy perturbation or thermodynamic integration, as it offers a faster solution. The g-mmpbsa program was utilized to carry out the MM/PBSA calculation. The results of the binding free energies are detailed in Table 3.

Table 3

Calculated binding free energies of the tested compounds (kJ/mol)

Complex G van der Waals energy Electrostatic energy Polar solvation energy SASA energy
Flavopiridol −137.603 ± 13.791 −189.311 ± 6.884 −59.704 ± 13.073 131.023 ± 7.764 −19.610 ± 0.827
Norwogonin −29.364 ± 17.416 −133.805 ± 7.506 −73.167 ± 30.426 191.326 ± 22.176 −13.718 ± 0.671
Sanggenol Q −135.809 ± 25.042 −211.697 ± 13.615 −51.223 ± 22.348 149.476 ± 8.593 −22.364 ± 0.457

4 Conclusions

This study investigated the potentials of 57 compounds sourced from 29 plants with reported anti-leukemic activities to serve as inhibitors of FLT3 using molecular modeling methods. Initially, the compound library was filtered based on three druglikeness rules: a process that led to the identification of 43 compounds with the potential to serve as oral drugs. Subsequent molecular docking simulation revealed flavopiridol, sanggenol Q, norwogonin, oblongixanthones A, oblongixanthones B, apigenin, and luteolin as the hit compounds based on their high affinities, as evident from their docking scores that ranged from −9.0 to −9.8 kcal/mol, while further screening revealed flavopiridol, sanggenol Q, and norwogonin as compounds with admirable ADMET properties. Also, MDS-based stability assessment of the compounds’ interactions with the residues that constitute the binding pocket of the target revealed their stable interaction as evident from the RMSD values, which is less than >2.5 Å. In conclusion, the results of this study revealed flavopiridol, sanggenol Q, and norwogonin worthy of exploration in further computational and experimental studies aimed at developing therapeutic regimens aimed at combating AML via mutant FLT3 inhibition.

Acknowledgements

The authors would like to extendtheir sincere appreciation to their respective institutions for the conducive environment in which to conduct thisresearch. The authors also would like to extend their sincere appreciation to the Researchers Supporting Project , King Saud University, Riyadh, Saudi Arabia for funding this work through the project number (RSP2024R457).

  1. Funding information: This work is financially supported by the Researchers Supporting Project (RSP2024R457). King Saud University, Riyadh, Saudi Arabia.

  2. Author contributions: All authors collaboratively executed the study in this manuscript. ZAA and HIU: conceptualized the study; MA, ROB, OV, RYO, MBouachrine, MB, SI, YAB, and HIU: prepared the first draft of the manuscript; HIU, ZAA, SI, YAB, WEM, and RYO: substantively verified the methodology, validated the in silico procedure; MB, WEM, MA, OV, GFW, ROB, and MBouachrine: revised the manuscript. All the authors read and approved the final manuscript.

  3. Conflict of interest: The authors declare no conflict of interest.

  4. Ethical approval: The conducted research is not related to either human or animals use.

  5. Data availability statement: The datasets generated during and/or analyzed during the current study are available from the corresponding author on reasonable request.

References

[1] Wang R, Yang X, Liu J, Zhong F, Zhang C, Chen Y, et al. Gut microbiota regulates acute myeloid leukaemia via alteration of intestinal barrier function mediated by butyrate. Nat Commun. 2022 May 9;13(1):2522.10.1038/s41467-022-30240-8Search in Google Scholar PubMed PubMed Central

[2] Döhner H, Weisdorf DJ, Bloomfield CD. Acute myeloid leukemia. N Engl J Med. 2015 Sep;373(12):1136–52.10.1056/NEJMra1406184Search in Google Scholar PubMed

[3] Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA: A Cancer J Clin. 2022 Jan;72(1):7–33.10.3322/caac.21708Search in Google Scholar PubMed

[4] Döhner H, Estey EH, Amadori S, Appelbaum FR, Büchner T, Burnett AK, et al. Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet. Blood. 2010 Jan;115(3):453–74.10.1182/blood-2009-07-235358Search in Google Scholar PubMed

[5] Döhner H, Estey E, Grimwade D, Amadori S, Appelbaum FR, Büchner T, et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood. 2017 Jan;129(4):424–7.10.1182/blood-2016-08-733196Search in Google Scholar PubMed PubMed Central

[6] Rowe JM. Perspectives on current survival and new developments in AML. Best Pract & Res Clin Haematology. 2021 Mar;34(1):101248.10.1016/j.beha.2021.101248Search in Google Scholar PubMed

[7] Bystrom R, Levis MJ. An update on FLT3 in Acute myeloid leukemia: pathophysiology and therapeutic landscape. Curr Oncol Rep. 2023 Apr;25(4):369–78.10.1007/s11912-023-01389-2Search in Google Scholar PubMed

[8] Majothi S, Adams D, Loke J, Stevens SP, Wheatley K, Wilson JS. FLT3 inhibitors in acute myeloid leukaemia: assessment of clinical effectiveness, adverse events and future research – a systematic review and meta-analysis. Syst Rev. 2020 Dec;9(1):285.10.1186/s13643-020-01540-1Search in Google Scholar PubMed PubMed Central

[9] Ersöz NŞ, Adan A. Resveratrol triggers anti-proliferative and apoptotic effects in FLT3-ITD-positive acute myeloid leukemia cells via inhibiting ceramide catabolism enzymes. Med Oncol. 2022 Mar;39(3):35.10.1007/s12032-021-01627-2Search in Google Scholar PubMed

[10] Zoi V, Galani V, Lianos GD, Voulgaris S, Kyritsis AP, Alexiou GA. The role of curcumin in cancer treatment. Biomedicines. 2021 Aug;9(9):1086.10.3390/biomedicines9091086Search in Google Scholar PubMed PubMed Central

[11] Aung T, Qu Z, Kortschak R, Adelson D. Understanding the effectiveness of natural compound mixtures in cancer through their molecular mode of action. IJMS. 2017 Mar;18(3):656.10.3390/ijms18030656Search in Google Scholar PubMed PubMed Central

[12] Cragg GM, Pezzuto JM. Natural products as a vital source for the discovery of cancer chemotherapeutic and chemopreventive agents. Med Princ Pract. 2016;25(Suppl. 2):41–59.10.1159/000443404Search in Google Scholar PubMed PubMed Central

[13] Cruz-Vicente P, Passarinha LA, Silvestre S, Gallardo E. Recent developments in new therapeutic agents against alzheimer and parkinson diseases: in-silico approaches. Molecules. 2021 Apr;26(8):2193.10.3390/molecules26082193Search in Google Scholar PubMed PubMed Central

[14] Ece A. Computer-aided drug design. BMC Chem. 2023 Mar;17(1):26.10.1186/s13065-023-00939-wSearch in Google Scholar PubMed PubMed Central

[15] Ishiki HM, Filho JMB, Da Silva MS, Scotti MT, Scotti L. Computer-aided drug design applied to parkinson targets. CN. 2018 Jun;16(6):865–80.10.2174/1570159X15666171128145423Search in Google Scholar PubMed PubMed Central

[16] Kulkarni AM, Rampogu S, Lee KW. Computer-aided drug discovery identifies alkaloid inhibitors of parkinson’s disease associated protein, prolyl oligopeptidase. In: Shanak S, editor. Evidence-Based Complementary and Alternative Medicine. 2021, 2021 Apr. p. 1–10.10.1155/2021/6687572Search in Google Scholar PubMed PubMed Central

[17] Aldewachi H, Al-Zidan RN, Conner MT, Salman MM. High-throughput screening platforms in the discovery of novel drugs for neurodegenerative diseases. Bioengineering. 2021 Feb;8(2):30.10.3390/bioengineering8020030Search in Google Scholar PubMed PubMed Central

[18] Salman MM, Al-Obaidi Z, Kitchen P, Loreto A, Bill RM, Wade-Martins R. Advances in applying computer-aided drug design for neurodegenerative diseases. IJMS. 2021 Apr;22(9):4688.10.3390/ijms22094688Search in Google Scholar PubMed PubMed Central

[19] Khan T, Ali M, Khan A, Nisar P, Jan SA, Afridi S, et al. Anti-cancer plants: a review of the active phytochemicals, applications in animal models, and regulatory aspects. Biomolecules. 2019 Dec;10(1):47.10.3390/biom10010047Search in Google Scholar PubMed PubMed Central

[20] Ren Y, De Blanco EJC, Fuchs JR, Soejarto DD, Burdette JE, Swanson SM, et al. Potential anti-cancer agents characterized from selected tropical plants. J Nat Prod. 2019 Mar;82(3):657–79.10.1021/acs.jnatprod.9b00018Search in Google Scholar PubMed PubMed Central

[21] Salehi B, Upadhyay S, Erdogan Orhan I, Kumar Jugran A, Jayaweera LD, SA, Dias D, et al. Therapeutic potential of α- and β-pinene: a miracle gift of nature. Biomolecules. 2019 Nov;9(11):738.10.3390/biom9110738Search in Google Scholar

[22] Yuan M, Zhang G, Bai W, Han X, Li C, Bian S. The role of bioactive compounds in natural products extracted from plants in cancer treatment and their mechanisms related to anti-cancer effects. In: Qin S, editor. Oxidative Medicine and Cellular Longevity; 2022 Feb2022. 1–1910.1155/2022/1429869Search in Google Scholar

[23] Kawase T, Nakazawa T, Eguchi T, Tsuzuki H. Effect of Fms-like tyrosine kinase 3 ( FLT3) ligand ( FL) on antitumor activity of gilteritinib, a FLT3 inhibitor, in mice xenografted with FL-overexpressing cells. Oncotarget. 2019;10(58):6111–23.10.18632/oncotarget.27222Search in Google Scholar

[24] Pettersen EF, Goddard TD, Huang CC, Couch GS, Greenblatt DM, Meng EC, et al. UCSF chimera-A visualization system for exploratory research and analysis. J Comput Chem. 2004;25(13):1605–12.10.1002/jcc.20084Search in Google Scholar

[25] Lipinski CA, Lombardo F, Dominy BW, Feeney PJ. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv Drug Delivery Rev. 2001;46:3–26.10.1016/S0169-409X(00)00129-0Search in Google Scholar

[26] Veber DF, Johnson SR, Cheng HY, Smith BR, Ward KW, Kopple KD. Molecular properties that influence the oral bioavailability of drug candidates. J Med Chem. 2002;45:2615–23.10.1021/jm020017nSearch in Google Scholar

[27] Egan WJ, Merz KM, Baldwin JJ. Prediction of Drug Absorption Using Multivariate Statistics. J Med Chem. 2000;43:3867–77.10.1021/jm000292eSearch in Google Scholar

[28] Daina A, Michielin O, Zoete V. SwissADME: a free web tool to evaluate pharmacokinetics, drug- likeness and medicinal chemistry friendliness of small molecules. Sci Rep. 2017;7:1–13.10.1038/srep42717Search in Google Scholar

[29] Halgren TA. merck Molecular force field. I. basis, form, scope, parameterization, and performance of MMFF94. J Comput Chem. 1996;17(5):490–519.10.1002/(SICI)1096-987X(199604)17:5/6<490::AID-JCC1>3.0.CO;2-PSearch in Google Scholar

[30] Trott O, Olson, AJ. AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization and multithreading. J Comput Chem. 2010;31(2):455–61.10.1002/jcc.21334Search in Google Scholar

[31] Seeliger D, Groot BLDe. Ligand docking and binding site analysis with PyMOL and Autodock/Vina. J Comput Mol Des. 2010;24:417–22.10.1007/s10822-010-9352-6Search in Google Scholar PubMed PubMed Central

[32] Cheng F, Li W, Zhou Y, Jie S, Wu Z, Liu G, et al. admetSAR: A comprehensive source and free tool for assessment of chemical ADMET properties. J Chem Inf Model. 2012;52:3099–105.10.1021/ci300367aSearch in Google Scholar PubMed

[33] Yang H, Lou C, Sun L, Li J, Cai Y, Li W, et al. admetSAR 2. 0: web-service for prediction and optimization of chemical ADMET properties. 2018 Aug:2016–7.10.1093/bioinformatics/bty707Search in Google Scholar PubMed

[34] Cetin A. Some flavolignans as potent Sars-Cov-2 inhibitors via molecular docking,molecular dynamic simulations and ADME analysis. CAD. 2022 Aug;18(5):337–46.10.2174/1573409918666220816113516Search in Google Scholar PubMed

[35] Olukunle OF, Olowosoke CB, Khalid A, Oke GA, Omoboyede V, Umar HI, et al. Identification of a 1, 8-naphthyridine-containing compound endowed with the inhibition of p53-MDM2/X interaction signaling: a computational perspective. Mol Diversity. 2023 Apr. [cited 2023 Aug 5] 10.1007/s11030-023-10637-3 Search in Google Scholar PubMed

[36] Bondarchuk SV. Prediction of aquatic toxicity of energetic materials using genetic function approximation. FirePhysChem. 2023 Mar;3(1):23–8.10.1016/j.fpc.2022.07.001Search in Google Scholar

[37] Omoboyede V, Onile OS, Oyeyemi BF, Aruleba RT, Fadahunsi AI, Oke GA, et al. Unravelling the anti-inflammatory mechanism of Allium cepa: an integration of network pharmacology and molecular docking approaches. Mol Diversity. 2023 Mar. [cited 2023 May 16] 10.1007/s11030-023-10614-w Search in Google Scholar PubMed

[38] Kříž K, Řezáč J. Benchmarking of semi-empirical quantum-mechanical methods on systems relevant to computer-aided drug design. J Chem Inf Model. 2020 Mar;60(3):1453–60.10.1021/acs.jcim.9b01171Search in Google Scholar PubMed

[39] Pecina A, Fanfrlík J, Lepšík M, Řezáč J. SQM2.20: Semi-empirical quantum-mechanical scoring function yields DFT-quality protein–ligand binding affinity predictions in minutes. Nat Commun. 2024 Feb;15(1):1127.10.1038/s41467-024-45431-8Search in Google Scholar PubMed PubMed Central

[40] Chukwuemeka PO, Umar HI, Iwaloye I, Oretade OM, Olowosoke CB, Elabiyi MO, et al. Targeting p53-MDM2 interactions to identify small molecule inhibitors for cancer therapy: beyond “Failure to rescue. J Biomol Struct Dyn. 202110.1080/07391102.2021.1924267Search in Google Scholar PubMed

[41] Umar HI, Siraj B, Ajayi A, Jimoh TO, Chukwuemeka PO. Molecular docking studies of some selected gallic acid derivatives against five non-structural proteins of novel coronavirus. J Genet Eng Biotechnol. 2021 Dec;19(1):16.10.1186/s43141-021-00120-7Search in Google Scholar PubMed PubMed Central

[42] Cavasotto CN, Aucar MG, Adler NS. Computational chemistry in drug lead discovery and design. Int J Quantum Chem. 2019 Jan 15;119(2):e25678.10.1002/qua.25678Search in Google Scholar

[43] Du X, Li Y, Xia YL, Ai SM, Liang J, Sang P, et al. Insights into protein–ligand interactions: mechanisms, models, and methods. IJMS. 2016 Jan;17(2):144.10.3390/ijms17020144Search in Google Scholar PubMed PubMed Central

[44] Gelpi J, Hospital A, Goñi R, Orozco M. Molecular dynamics simulations: advances and applications. AABC. 2015 Nov;37.10.2147/AABC.S70333Search in Google Scholar PubMed PubMed Central

[45] Saurabh S, Sivakumar PM, Perumal V, Khosravi A, Sugumaran A, Prabhawathi V. Molecular Dynamics simulations in drug discovery and drug delivery. In: Krishnan A, Chuturgoon A, editors. Integrative Nanomedicine for New Therapies [Internet]. Cham: Springer International Publishing; 2020. [cited 2024 May 18]. p. 275–301. (Engineering Materials). 10.1007/978-3-030-36260-7_10.Search in Google Scholar

[46] Al-Karmalawy AA, Dahab MA, Metwaly AM, Elhady SS, Elkaeed EB, Eissa IH, et al. Molecular docking and dynamics simulation revealed the potential inhibitory activity of ACEIs against SARS-CoV-2 targeting the hACE2 receptor. Front Chem. 2021;9:661230.10.3389/fchem.2021.661230Search in Google Scholar PubMed PubMed Central

[47] Liu K, Kokubo H. Exploring the stability of ligand binding modes to proteins by molecular dynamics simulations: a cross-docking study. J Chem Inf Model. 2017 Oct;57(10):2514–22.10.1021/acs.jcim.7b00412Search in Google Scholar PubMed

Received: 2024-01-25
Revised: 2024-04-28
Accepted: 2024-05-09
Published Online: 2024-06-18

© 2024 the author(s), published by De Gruyter

This work is licensed under the Creative Commons Attribution 4.0 International License.

Articles in the same Issue

  1. Regular Articles
  2. Porous silicon nanostructures: Synthesis, characterization, and their antifungal activity
  3. Biochar from de-oiled Chlorella vulgaris and its adsorption on antibiotics
  4. Phytochemicals profiling, in vitro and in vivo antidiabetic activity, and in silico studies on Ajuga iva (L.) Schreb.: A comprehensive approach
  5. Synthesis, characterization, in silico and in vitro studies of novel glycoconjugates as potential antibacterial, antifungal, and antileishmanial agents
  6. Sonochemical synthesis of gold nanoparticles mediated by potato starch: Its performance in the treatment of esophageal cancer
  7. Computational study of ADME-Tox prediction of selected phytochemicals from Punica granatum peels
  8. Phytochemical analysis, in vitro antioxidant and antifungal activities of extracts and essential oil derived from Artemisia herba-alba Asso
  9. Two triazole-based coordination polymers: Synthesis and crystal structure characterization
  10. Phytochemical and physicochemical studies of different apple varieties grown in Morocco
  11. Synthesis of multi-template molecularly imprinted polymers (MT-MIPs) for isolating ethyl para-methoxycinnamate and ethyl cinnamate from Kaempferia galanga L., extract with methacrylic acid as functional monomer
  12. Nutraceutical potential of Mesembryanthemum forsskaolii Hochst. ex Bioss.: Insights into its nutritional composition, phytochemical contents, and antioxidant activity
  13. Evaluation of influence of Butea monosperma floral extract on inflammatory biomarkers
  14. Cannabis sativa L. essential oil: Chemical composition, anti-oxidant, anti-microbial properties, and acute toxicity: In vitro, in vivo, and in silico study
  15. The effect of gamma radiation on 5-hydroxymethylfurfural conversion in water and dimethyl sulfoxide
  16. Hollow mushroom nanomaterials for potentiometric sensing of Pb2+ ions in water via the intercalation of iodide ions into the polypyrrole matrix
  17. Determination of essential oil and chemical composition of St. John’s Wort
  18. Computational design and in vitro assay of lantadene-based novel inhibitors of NS3 protease of dengue virus
  19. Anti-parasitic activity and computational studies on a novel labdane diterpene from the roots of Vachellia nilotica
  20. Microbial dynamics and dehydrogenase activity in tomato (Lycopersicon esculentum Mill.) rhizospheres: Impacts on growth and soil health across different soil types
  21. Correlation between in vitro anti-urease activity and in silico molecular modeling approach of novel imidazopyridine–oxadiazole hybrids derivatives
  22. Spatial mapping of indoor air quality in a light metro system using the geographic information system method
  23. Iron indices and hemogram in renal anemia and the improvement with Tribulus terrestris green-formulated silver nanoparticles applied on rat model
  24. Integrated track of nano-informatics coupling with the enrichment concept in developing a novel nanoparticle targeting ERK protein in Naegleria fowleri
  25. Cytotoxic and phytochemical screening of Solanum lycopersicum–Daucus carota hydro-ethanolic extract and in silico evaluation of its lycopene content as anticancer agent
  26. Protective activities of silver nanoparticles containing Panax japonicus on apoptotic, inflammatory, and oxidative alterations in isoproterenol-induced cardiotoxicity
  27. pH-based colorimetric detection of monofunctional aldehydes in liquid and gas phases
  28. Investigating the effect of resveratrol on apoptosis and regulation of gene expression of Caco-2 cells: Unravelling potential implications for colorectal cancer treatment
  29. Metformin inhibits knee osteoarthritis induced by type 2 diabetes mellitus in rats: S100A8/9 and S100A12 as players and therapeutic targets
  30. Effect of silver nanoparticles formulated by Silybum marianum on menopausal urinary incontinence in ovariectomized rats
  31. Synthesis of new analogs of N-substituted(benzoylamino)-1,2,3,6-tetrahydropyridines
  32. Response of yield and quality of Japonica rice to different gradients of moisture deficit at grain-filling stage in cold regions
  33. Preparation of an inclusion complex of nickel-based β-cyclodextrin: Characterization and accelerating the osteoarthritis articular cartilage repair
  34. Empagliflozin-loaded nanomicelles responsive to reactive oxygen species for renal ischemia/reperfusion injury protection
  35. Preparation and pharmacodynamic evaluation of sodium aescinate solid lipid nanoparticles
  36. Assessment of potentially toxic elements and health risks of agricultural soil in Southwest Riyadh, Saudi Arabia
  37. Theoretical investigation of hydrogen-rich fuel production through ammonia decomposition
  38. Biosynthesis and screening of cobalt nanoparticles using citrus species for antimicrobial activity
  39. Investigating the interplay of genetic variations, MCP-1 polymorphism, and docking with phytochemical inhibitors for combatting dengue virus pathogenicity through in silico analysis
  40. Ultrasound induced biosynthesis of silver nanoparticles embedded into chitosan polymers: Investigation of its anti-cutaneous squamous cell carcinoma effects
  41. Copper oxide nanoparticles-mediated Heliotropium bacciferum leaf extract: Antifungal activity and molecular docking assays against strawberry pathogens
  42. Sprouted wheat flour for improving physical, chemical, rheological, microbial load, and quality properties of fino bread
  43. Comparative toxicity assessment of fisetin-aided artificial intelligence-assisted drug design targeting epibulbar dermoid through phytochemicals
  44. Acute toxicity and anti-inflammatory activity of bis-thiourea derivatives
  45. Anti-diabetic activity-guided isolation of α-amylase and α-glucosidase inhibitory terpenes from Capsella bursa-pastoris Linn.
  46. GC–MS analysis of Lactobacillus plantarum YW11 metabolites and its computational analysis on familial pulmonary fibrosis hub genes
  47. Green formulation of copper nanoparticles by Pistacia khinjuk leaf aqueous extract: Introducing a novel chemotherapeutic drug for the treatment of prostate cancer
  48. Improved photocatalytic properties of WO3 nanoparticles for Malachite green dye degradation under visible light irradiation: An effect of La doping
  49. One-pot synthesis of a network of Mn2O3–MnO2–poly(m-methylaniline) composite nanorods on a polypyrrole film presents a promising and efficient optoelectronic and solar cell device
  50. Groundwater quality and health risk assessment of nitrate and fluoride in Al Qaseem area, Saudi Arabia
  51. A comparative study of the antifungal efficacy and phytochemical composition of date palm leaflet extracts
  52. Processing of alcohol pomelo beverage (Citrus grandis (L.) Osbeck) using saccharomyces yeast: Optimization, physicochemical quality, and sensory characteristics
  53. Specialized compounds of four Cameroonian spices: Isolation, characterization, and in silico evaluation as prospective SARS-CoV-2 inhibitors
  54. Identification of a novel drug target in Porphyromonas gingivalis by a computational genome analysis approach
  55. Physico-chemical properties and durability of a fly-ash-based geopolymer
  56. FMS-like tyrosine kinase 3 inhibitory potentials of some phytochemicals from anti-leukemic plants using computational chemical methodologies
  57. Wild Thymus zygis L. ssp. gracilis and Eucalyptus camaldulensis Dehnh.: Chemical composition, antioxidant and antibacterial activities of essential oils
  58. 3D-QSAR, molecular docking, ADMET, simulation dynamic, and retrosynthesis studies on new styrylquinolines derivatives against breast cancer
  59. Deciphering the influenza neuraminidase inhibitory potential of naturally occurring biflavonoids: An in silico approach
  60. Determination of heavy elements in agricultural regions, Saudi Arabia
  61. Synthesis and characterization of antioxidant-enriched Moringa oil-based edible oleogel
  62. Ameliorative effects of thistle and thyme honeys on cyclophosphamide-induced toxicity in mice
  63. Study of phytochemical compound and antipyretic activity of Chenopodium ambrosioides L. fractions
  64. Investigating the adsorption mechanism of zinc chloride-modified porous carbon for sulfadiazine removal from water
  65. Performance repair of building materials using alumina and silica composite nanomaterials with electrodynamic properties
  66. Effects of nanoparticles on the activity and resistance genes of anaerobic digestion enzymes in livestock and poultry manure containing the antibiotic tetracycline
  67. Effect of copper nanoparticles green-synthesized using Ocimum basilicum against Pseudomonas aeruginosa in mice lung infection model
  68. Cardioprotective effects of nanoparticles green formulated by Spinacia oleracea extract on isoproterenol-induced myocardial infarction in mice by the determination of PPAR-γ/NF-κB pathway
  69. Anti-OTC antibody-conjugated fluorescent magnetic/silica and fluorescent hybrid silica nanoparticles for oxytetracycline detection
  70. Curcumin conjugated zinc nanoparticles for the treatment of myocardial infarction
  71. Identification and in silico screening of natural phloroglucinols as potential PI3Kα inhibitors: A computational approach for drug discovery
  72. Exploring the phytochemical profile and antioxidant evaluation: Molecular docking and ADMET analysis of main compounds from three Solanum species in Saudi Arabia
  73. Unveiling the molecular composition and biological properties of essential oil derived from the leaves of wild Mentha aquatica L.: A comprehensive in vitro and in silico exploration
  74. Analysis of bioactive compounds present in Boerhavia elegans seeds by GC-MS
  75. Homology modeling and molecular docking study of corticotrophin-releasing hormone: An approach to treat stress-related diseases
  76. LncRNA MIR17HG alleviates heart failure via targeting MIR17HG/miR-153-3p/SIRT1 axis in in vitro model
  77. Development and validation of a stability indicating UPLC-DAD method coupled with MS-TQD for ramipril and thymoquinone in bioactive SNEDDS with in silico toxicity analysis of ramipril degradation products
  78. Biosynthesis of Ag/Cu nanocomposite mediated by Curcuma longa: Evaluation of its antibacterial properties against oral pathogens
  79. Development of AMBER-compliant transferable force field parameters for polytetrafluoroethylene
  80. Treatment of gestational diabetes by Acroptilon repens leaf aqueous extract green-formulated iron nanoparticles in rats
  81. Development and characterization of new ecological adsorbents based on cardoon wastes: Application to brilliant green adsorption
  82. A fast, sensitive, greener, and stability-indicating HPLC method for the standardization and quantitative determination of chlorhexidine acetate in commercial products
  83. Assessment of Se, As, Cd, Cr, Hg, and Pb content status in Ankang tea plantations of China
  84. Effect of transition metal chloride (ZnCl2) on low-temperature pyrolysis of high ash bituminous coal
  85. Evaluating polyphenol and ascorbic acid contents, tannin removal ability, and physical properties during hydrolysis and convective hot-air drying of cashew apple powder
  86. Development and characterization of functional low-fat frozen dairy dessert enhanced with dried lemongrass powder
  87. Scrutinizing the effect of additive and synergistic antibiotics against carbapenem-resistant Pseudomonas aeruginosa
  88. Preparation, characterization, and determination of the therapeutic effects of copper nanoparticles green-formulated by Pistacia atlantica in diabetes-induced cardiac dysfunction in rat
  89. Antioxidant and antidiabetic potentials of methoxy-substituted Schiff bases using in vitro, in vivo, and molecular simulation approaches
  90. Anti-melanoma cancer activity and chemical profile of the essential oil of Seseli yunnanense Franch
  91. Molecular docking analysis of subtilisin-like alkaline serine protease (SLASP) and laccase with natural biopolymers
  92. Overcoming methicillin resistance by methicillin-resistant Staphylococcus aureus: Computational evaluation of napthyridine and oxadiazoles compounds for potential dual inhibition of PBP-2a and FemA proteins
  93. Exploring novel antitubercular agents: Innovative design of 2,3-diaryl-quinoxalines targeting DprE1 for effective tuberculosis treatment
  94. Drimia maritima flowers as a source of biologically potent components: Optimization of bioactive compound extractions, isolation, UPLC–ESI–MS/MS, and pharmacological properties
  95. Estimating molecular properties, drug-likeness, cardiotoxic risk, liability profile, and molecular docking study to characterize binding process of key phyto-compounds against serotonin 5-HT2A receptor
  96. Fabrication of β-cyclodextrin-based microgels for enhancing solubility of Terbinafine: An in-vitro and in-vivo toxicological evaluation
  97. Phyto-mediated synthesis of ZnO nanoparticles and their sunlight-driven photocatalytic degradation of cationic and anionic dyes
  98. Monosodium glutamate induces hypothalamic–pituitary–adrenal axis hyperactivation, glucocorticoid receptors down-regulation, and systemic inflammatory response in young male rats: Impact on miR-155 and miR-218
  99. Quality control analyses of selected honey samples from Serbia based on their mineral and flavonoid profiles, and the invertase activity
  100. Eco-friendly synthesis of silver nanoparticles using Phyllanthus niruri leaf extract: Assessment of antimicrobial activity, effectiveness on tropical neglected mosquito vector control, and biocompatibility using a fibroblast cell line model
  101. Green synthesis of silver nanoparticles containing Cichorium intybus to treat the sepsis-induced DNA damage in the liver of Wistar albino rats
  102. Quality changes of durian pulp (Durio ziberhinus Murr.) in cold storage
  103. Study on recrystallization process of nitroguanidine by directly adding cold water to control temperature
  104. Determination of heavy metals and health risk assessment in drinking water in Bukayriyah City, Saudi Arabia
  105. Larvicidal properties of essential oils of three Artemisia species against the chemically insecticide-resistant Nile fever vector Culex pipiens (L.) (Diptera: Culicidae): In vitro and in silico studies
  106. Design, synthesis, characterization, and theoretical calculations, along with in silico and in vitro antimicrobial proprieties of new isoxazole-amide conjugates
  107. The impact of drying and extraction methods on total lipid, fatty acid profile, and cytotoxicity of Tenebrio molitor larvae
  108. A zinc oxide–tin oxide–nerolidol hybrid nanomaterial: Efficacy against esophageal squamous cell carcinoma
  109. Research on technological process for production of muskmelon juice (Cucumis melo L.)
  110. Physicochemical components, antioxidant activity, and predictive models for quality of soursop tea (Annona muricata L.) during heat pump drying
  111. Characterization and application of Fe1−xCoxFe2O4 nanoparticles in Direct Red 79 adsorption
  112. Torilis arvensis ethanolic extract: Phytochemical analysis, antifungal efficacy, and cytotoxicity properties
  113. Magnetite–poly-1H pyrrole dendritic nanocomposite seeded on poly-1H pyrrole: A promising photocathode for green hydrogen generation from sanitation water without using external sacrificing agent
  114. HPLC and GC–MS analyses of phytochemical compounds in Haloxylon salicornicum extract: Antibacterial and antifungal activity assessment of phytopathogens
  115. Efficient and stable to coking catalysts of ethanol steam reforming comprised of Ni + Ru loaded on MgAl2O4 + LnFe0.7Ni0.3O3 (Ln = La, Pr) nanocomposites prepared via cost-effective procedure with Pluronic P123 copolymer
  116. Nitrogen and boron co-doped carbon dots probe for selectively detecting Hg2+ in water samples and the detection mechanism
  117. Heavy metals in road dust from typical old industrial areas of Wuhan: Seasonal distribution and bioaccessibility-based health risk assessment
  118. Phytochemical profiling and bioactivity evaluation of CBD- and THC-enriched Cannabis sativa extracts: In vitro and in silico investigation of antioxidant and anti-inflammatory effects
  119. Investigating dye adsorption: The role of surface-modified montmorillonite nanoclay in kinetics, isotherms, and thermodynamics
  120. Antimicrobial activity, induction of ROS generation in HepG2 liver cancer cells, and chemical composition of Pterospermum heterophyllum
  121. Study on the performance of nanoparticle-modified PVDF membrane in delaying membrane aging
  122. Impact of cholesterol in encapsulated vitamin E acetate within cocoliposomes
  123. Review Articles
  124. Structural aspects of Pt(η3-X1N1X2)(PL) (X1,2 = O, C, or Se) and Pt(η3-N1N2X1)(PL) (X1 = C, S, or Se) derivatives
  125. Biosurfactants in biocorrosion and corrosion mitigation of metals: An overview
  126. Stimulus-responsive MOF–hydrogel composites: Classification, preparation, characterization, and their advancement in medical treatments
  127. Electrochemical dissolution of titanium under alternating current polarization to obtain its dioxide
  128. Special Issue on Recent Trends in Green Chemistry
  129. Phytochemical screening and antioxidant activity of Vitex agnus-castus L.
  130. Phytochemical study, antioxidant activity, and dermoprotective activity of Chenopodium ambrosioides (L.)
  131. Exploitation of mangliculous marine fungi, Amarenographium solium, for the green synthesis of silver nanoparticles and their activity against multiple drug-resistant bacteria
  132. Study of the phytotoxicity of margines on Pistia stratiotes L.
  133. Special Issue on Advanced Nanomaterials for Energy, Environmental and Biological Applications - Part III
  134. Impact of biogenic zinc oxide nanoparticles on growth, development, and antioxidant system of high protein content crop (Lablab purpureus L.) sweet
  135. Green synthesis, characterization, and application of iron and molybdenum nanoparticles and their composites for enhancing the growth of Solanum lycopersicum
  136. Green synthesis of silver nanoparticles from Olea europaea L. extracted polysaccharides, characterization, and its assessment as an antimicrobial agent against multiple pathogenic microbes
  137. Photocatalytic treatment of organic dyes using metal oxides and nanocomposites: A quantitative study
  138. Antifungal, antioxidant, and photocatalytic activities of greenly synthesized iron oxide nanoparticles
  139. Special Issue on Phytochemical and Pharmacological Scrutinization of Medicinal Plants
  140. Hepatoprotective effects of safranal on acetaminophen-induced hepatotoxicity in rats
  141. Chemical composition and biological properties of Thymus capitatus plants from Algerian high plains: A comparative and analytical study
  142. Chemical composition and bioactivities of the methanol root extracts of Saussurea costus
  143. In vivo protective effects of vitamin C against cyto-genotoxicity induced by Dysphania ambrosioides aqueous extract
  144. Insights about the deleterious impact of a carbamate pesticide on some metabolic immune and antioxidant functions and a focus on the protective ability of a Saharan shrub and its anti-edematous property
  145. A comprehensive review uncovering the anticancerous potential of genkwanin (plant-derived compound) in several human carcinomas
  146. A study to investigate the anticancer potential of carvacrol via targeting Notch signaling in breast cancer
  147. Assessment of anti-diabetic properties of Ziziphus oenopolia (L.) wild edible fruit extract: In vitro and in silico investigations through molecular docking analysis
  148. Optimization of polyphenol extraction, phenolic profile by LC-ESI-MS/MS, antioxidant, anti-enzymatic, and cytotoxic activities of Physalis acutifolia
  149. Phytochemical screening, antioxidant properties, and photo-protective activities of Salvia balansae de Noé ex Coss
  150. Antihyperglycemic, antiglycation, anti-hypercholesteremic, and toxicity evaluation with gas chromatography mass spectrometry profiling for Aloe armatissima leaves
  151. Phyto-fabrication and characterization of gold nanoparticles by using Timur (Zanthoxylum armatum DC) and their effect on wound healing
  152. Does Erodium trifolium (Cav.) Guitt exhibit medicinal properties? Response elements from phytochemical profiling, enzyme-inhibiting, and antioxidant and antimicrobial activities
  153. Integrative in silico evaluation of the antiviral potential of terpenoids and its metal complexes derived from Homalomena aromatica based on main protease of SARS-CoV-2
  154. 6-Methoxyflavone improves anxiety, depression, and memory by increasing monoamines in mice brain: HPLC analysis and in silico studies
  155. Simultaneous extraction and quantification of hydrophilic and lipophilic antioxidants in Solanum lycopersicum L. varieties marketed in Saudi Arabia
  156. Biological evaluation of CH3OH and C2H5OH of Berberis vulgaris for in vivo antileishmanial potential against Leishmania tropica in murine models
Downloaded on 6.12.2025 from https://www.degruyterbrill.com/document/doi/10.1515/chem-2024-0045/html
Scroll to top button