Research Article |
|
Corresponding author: Ali Jabbar Radhi ( alijebar56@alkafeel.edu.iq ) Academic editor: Ivan Dimitrov
© 2025 Ali Jabbar Radhi, Mahmood M. Fahad, Ihsan Alrubaie, Nusrat Shafiq, Kinza Afzal.
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:
Radhi AJ, Fahad MM, Alrubaie I, Shafiq N, Afzal K (2025) In silico and in vitro studies of gemcitabine derivatives as anticancer agents. Pharmacia 72: 1-16. https://doi.org/10.3897/pharmacia.72.e153719
|
Cancer remains one of the leading causes of death globally and is expected to increase in incidence. The present investigation aimed to evaluate the anticancer efficacy of gemcitabine derivatives on MCF7, A549, and PC3 cell lines. Compounds Com. 10 and Com. 16 were the most potent, with IC₅₀ values of 9.45, 6.93, and 12.09 µM (Com. 10) and 12.23, 6.60, and 21.57 µM (Com. 16) for the three cell lines, showing a better response compared to the reference drug gemcitabine. Com. 9 and Com. 14 were also moderately active, preferentially on A549 and PC3. Com. 16 showed strong binding with the active residues as determined by molecular docking. Com. 16 bound to key cancer targets Vav1 (6NEW), ERK2 (4ZXT), and CYP3A4 (7LXL), with docking scores of −191.72, −187.66, and −221.08 kcal/mol, respectively. These interactions were electrostatic, hydrophobic, and π-type, similar to those induced by gemcitabine. Several other compounds also demonstrated good docking scores (−160 to −180 kcal/mol), and eight protein–ligand complexes emerged as leads. Moreover, ADME and physicochemical analyses of the top six active compounds indicated good drug-likeness and a favorable PK profile compared to gemcitabine.
MCF7, PC3, A549, gemcitabine, molecular docking, anticancer activity
Cancer ranks third as a cause of death worldwide, resulting in an estimated 10 million deaths in 2020 (
There is a correlation between low gemcitabine cytotoxicity and low dCK levels (
The cytotoxic activity of gemcitabine (obtained from Sigma) and its derivatives (Com. 1–Com. 16), which were synthesized in our previous studies as reported in the literature (Ali Jabbar
% Cell inhibition = 100 – [(At – Ab) / (Ac – Ab)] × 100
Where, At = absorbance of test compound, Ab = absorbance of blank, and Ac = absorbance of control.
The gemcitabine is symbolized in blue, and its functional groups are emphasized in red. These colors are for illustrative purposes only.
Physicochemical property calculation was performed using ChemBio3D in the ChemBioOffice Ultra 18.0 software package (PerkinElmer, USA, https://www.perkinelmer.com). In silico absorption, distribution, metabolism, excretion, and toxicity (ADMET) predictions were performed using two online programs: SwissADME and ADMETlab 2.0. The pharmacokinetic/drug-like features profile screener SwissADME (http://www.swissadme.ch) offers basic pharmacokinetic and drug-like profiles, while ADMETlab 2.0 uses machine learning algorithms, including random forest (RF), support vector machine (SVM), and k-nearest neighbor (k-NN), trained on curated and validated chemical datasets. These models were developed based on structure–activity relationship (QSAR) analysis to predict important descriptors in five pharmacokinetic and toxicological domains, with classification (e.g., toxic/non-toxic) and regression (e.g., logP, half-life) capabilities. In this QSAR, molecular structure in SMILES format was given as input, and molecular- and physicochemical descriptor-based predictions were made (
Three protein-based targets one for each cancer cell line (MCF7, A549, and PC3) were chosen for molecular docking studies in the present work. The docking accuracy was verified by re-docking the native ligands of 4ZXT and 7LXL, with the resulting RMSD at 1.83 Å and 1.17 Å, respectively. Overfitting was estimated by re-docking one of the ligands (Com. 16) already tested, and an RMSD of 1.24 Å was obtained for 6NEW, which had no native ligand, thus confirming the validation of our docking methodology. The criteria for selection were based on relevance to cancer biology, structural availability in the Protein Data Bank (PDB), and the importance of each target in critical oncogenic signaling pathways. The nature of each target is described in Table
Structural and functional profiling of tumor suppressor proteins: a mosaic of PDB sum contents and cancer-associated context.
| Target protein (PDB ID) | Proteome source | Protein Function | Cancer relevance | Justification for selection | Resolution | Organism |
|---|---|---|---|---|---|---|
| 6NEW (Vav1) | Human (MCF7) | Rho/Rac GTPase exchange factor for guanine nucleotide. | Modulator of cell survival and migration; involved in breast cancer | Reservoirs of oncogenic signaling and cytoskeletal regulation | 2.50 Å | Homo sapiens |
| 4ZXT (ERK2 Complex) | Human (A549) | MAPK/ERK signaling kinase | Overexpressed in lung cancer; promotes growth and survival | Targeted by MAPK inhibitors; implications for NSCLC treatment | 2.00 Å | Homo sapiens |
| 7LXL (CYP3A4) | Human (PC3) | Cytochrome P450 responsible for metabolism of drugs | Related to drug resistance of prostate cancer | Affects gemcitabine bioactivation and elimination | 2.75 Å | Homo sapiens |
Vav1 (PDB ID: 6NEW): Vav1 belongs to the Vav family of guanine nucleotide exchange factors (GEFs), which selectively activate Rho family GTPases, including Rac1 and Cdc42. These small GTPases are involved in various cellular functions, including cytoskeletal remodeling, transcription, cell proliferation, and survival. Vav1 is an important hematopoietic cell-mediated transducer, but its expression is dysregulated in several solid tumors, such as breast cancer. Its oncogenic function correlates with the induction of cell migration, invasion, and metastasis via the activation of actin cytoskeleton reorganization and regulation of downstream signaling pathways, including NF-κB and MAPK. High levels of Vav1 are associated with poor prognosis for breast cancer patients and hence become attractive targets for therapy (
Primarily, chemical structures were carefully illustrated using ChemDraw (version 19.0.1.8, PerkinElmer) and subsequently brought into Chem3D Ultra (version 19.0.1.8, PerkinElmer). Optimization was conducted using MM2 and MMFF9 force fields, safeguarding the refinement of three-dimensional geometries for precise structural analysis, and saved in Mol2 format.
The 3D protein structures of target proteins corresponding to the three cancer cell lines (MCF7, PC3, and A549) were retrieved from the RCSB PDB Protein Data Bank (www.rcsb.org) and prepared within Discovery Studio 2021 in accordance with conventional crystallographic principles. This preparation consisted of loop building, completion of missing atoms in unresolved residues, and removal of water and heteroatoms. The processed protein structures were subsequently loaded into Molegro Virtual Docker (MVD) 6.0 for docking (
For in silico studies, Molegro Virtual Docker (MVD) was used for docking simulations, followed by post-docking modeling and visualization in Discovery Studio. First, the prepared protein was imported into MVD, then the desired ligand was introduced. Docking parameters were kept at default. The docking exercise was directed to generate a bioactive binding pose at active sites. The lead pose was chosen based on the MolDock score, representing the binding potential between the protein and ligand.
Afterward, a 3D image of the docking results was analyzed. Field interactions were examined to explain hydrogen bonding in the workspace. The docked elements were transferred to Discovery Studio for ligand interaction and hydrophobicity assessment. The hydrophobicity of a protein–ligand complex was computed on a scale of 3 to −3, with positive values indicating an increase and negative values indicating a decrease in hydrophobicity. These hydrophobic values were used to quantify the energy contributions of hydrophobic interactions within the protein–ligand complex. A two-dimensional image was created to detect interactions between the ligand and its locale. The image was annotated with hydrogen bonds, covalent bonds, π, and π–alkyl interactions. However, unfavorable steric bumps, donor/acceptor clashes, and charge repulsion were purposely disregarded. Additionally, amino acid labels with three-letter codes were added for clarity.
The gemcitabine compounds (Com. 1–Com. 16) produced in earlier studies (Ali Jabbar
Anticancer (IC50, µM) activities of prepared compounds (Com. 1–Com. 16).
| References | WI-38 | PC3 | A549 | MCF7 | Sr. no. |
|---|---|---|---|---|---|
| ( |
- | 51.93 ± 1.78 | 86.12 ± 2.17 | 106.4 ± 1.14 | Com.1 |
| ( |
- | 124.4 ± 1.24 | 128.5 ± 1.67 | 130.1 ± 1.01 | Com.2 |
| ( |
- | 102.9 ± 1.09 | 45.32 ± 1.17 | 103.5 ± 1.08 | Com.3 |
| ( |
- | 113.8 ± 2.45 | 114.9 ± 0.89 | 45.48 ± 0.54 | Com.4 |
| ( |
- | 71.06 ± 1.68 | 190.3 ± 2.07 | 269.1 ± 0.91 | Com.5 |
| ( |
- | 69.39 ± 1.49 | 135.7 ± 1.27 | 146.7 ± 1.51 | Com.6 |
| ( |
- | 44.56 ± 1.65 | 43.96 ± 0.57 | 135.2 ± 2.04 | Com.7 |
| ( |
- | 53.11 ± 1.42 | 70.72 ± 1.78 | 628.6 ± 1.35 | Com.8 |
| (Ali et al. 2021) | 38.78 ± 0.79 | 39.39 ± 0.86 | 29.42 ± 1.68 | 91.12 ± 1.98 | Com.9 |
| (Ali et al. 2021) | 27.14 ± 1.08 | 21.57 ± 2.34 | 6.599 ± 1.08 | 12.23 ± 1.38 | Com.10 |
| ( |
- | 45.95 ± 2.17 | 106.2 ± 0.97 | 81.58 ± 0.87 | Com.11 |
| ( |
- | 85.15 ± 1.08 | 89 ± 2.52 | 113.7 ± 1.09 | Com.12 |
| ( |
- | 89.71 ± 1.37 | 52.37 ± 1.38 | 114.2 ± 1.68 | Com.13 |
| ( |
74.25 ± 1.12 | 36.29 ± 1.54 | 58.94 ± 1.82 | 88.4 ± 1.47 | Com.14 |
| ( |
- | 40.81 ± 1.33 | 41.05 ± 1.68 | 70.34 ± 0.98 | Com.15 |
| ( |
27.18 ± 1.27 | 12.09 ± 1.03 | 6.931 ± 0.76 | 9.454 ± 1.78 | Com.16 |
| 51.18 ± 1.05 | 24.09 ± 2.04 | 19.35 ± 0.99 | 15.34 ± 1.07 | Re. |
In a series of gemcitabine derivatives (Com. 1–Com. 16) used as anticancer agents, good to moderate activity (IC₅₀ = 6.599–45.48 µM) or weak activity (IC₅₀ > 50 µM) was observed toward MCF7, A549, and PC3 cell lines. The extent of cell viability after treatment with the gemcitabine derivatives used in this study was visualized through microscopic images obtained using the MTT method (Figs
Compounds Com. 10 and Com. 16 showed significant anticancer activity against MCF7 cell lines (IC₅₀ = 12.23 and 9.454 µM, respectively) when compared to gemcitabine (IC₅₀ = 15.34 µM). On the other hand, compound Com. 4 had moderate anticancer activity against MCF7 cell lines (IC₅₀ = 45.48 µM), and other gemcitabine derivatives had weak anticancer activity (IC₅₀ > 50.00 µM), as shown in Fig.
Compounds (Com9, Com10, and Com16) showed good anticancer activity against the human lung cancer A549 cell lines with IC50 values at (29.42, 6.599, and 6.931 µM), respectively, in comparison with the control drug gemcitabine (IC50 = 19.35 µM), but compounds (Com.3, Com.7, and Com.15) exhibited moderate anticancer activity against A549 cell lines (IC50 = 45.32, 43.96, and 41.05 µM, respectively). The remaining drugs have poor anticancer action against the A549 cell lines, with IC50 values greater than 50 µM. (Fig.
All gemcitabine derivatives were also tested for anticancer activity against the human prostate cancer PC3 cell lines. Compounds Com. 10, Com. 14, and Com. 16 showed good anticancer activity against PC3 cell lines, with IC₅₀ values of 21.57, 36.29, and 12.09 µM, respectively, compared with the control drug gemcitabine (IC₅₀ = 24.09 µM). Some gemcitabine compounds (Com. 7, Com. 9, Com. 11, and Com. 15) exhibited moderate anticancer activity against PC3 cell lines (IC₅₀ = 44.56, 39.39, 45.95, and 40.81 µM, respectively). The other gemcitabine derivatives showed weak anticancer activity against PC3 cell lines, with IC₅₀ values greater than 50.00 µM (Fig.
On the other hand, some compounds exhibited good anticancer activity against only one cell line, such as compound Com. 9 against A549 and compound Com. 14 against PC3 cell lines, whereas compounds Com. 10 and Com. 16 showed good anticancer activity across all cell lines employed in this investigation.
Here, gemcitabine was taken as a reference, by which the in vitro cytotoxicity of the top potent compounds Com. 9, Com. 10, Com. 14, and Com. 16 was tested against the human lung normal cell line WI-38. The synthetic compounds Com. 9, Com. 10, Com. 14, and Com. 16 had IC₅₀ values of 38.78, 24.14, 74.25, and 27.18 µM in non-cancerous WI-38 cells, respectively (Table
The selectivity index (SI) is an important endpoint in evaluating how well a compound is able to selectively kill cancer cells relative to normal cells (Equation 1), and it serves as a quantitative indicator of the therapeutic window for these compounds (Zhou et al. 2020;
Cytotoxicity and SI determination of the tested gemcitabine derivatives showed that Com. 16 possessed the most attractive profile in terms of potent anticancer activity against MCF7, A549, and PC3 (IC₅₀: 9.45, 6.93, 12.09 µM) with high selectivity against cancer cells over normal cells (SI: 2.88, 3.92, and 2.25, respectively) and thus could be considered the worthiest candidate for future investigation. Com. 10 also exerted significant cytotoxicity, especially for MCF7, PC3, and A549, and showed moderate SI values (2.22, 1.26, and 4.11), indicating possible cancer selectivity. Com. 14 showed some selectivity, predominantly for PC3 (SI = 2.05), but it was less potent. In contrast, Com. 9 presented low SI values for all lines (0.43–1.32), signifying poor discrimination between cancerous and non-cancerous cells, which would limit its use as a therapy. These results also emphasize the need for normal cell line toxicity testing to determine compounds with a favorable therapeutic index early in drug development. All results are summarized in Table
| Com. | PC3 SI (Prostate) | A549 SI (Lung) | MCF7 SI (Breast) |
|---|---|---|---|
| Com.9 | 0.98 ± 0.02 | 1.32 ± 0.07 | 0.43 ± 0.01 |
| Com.10 | 1.26 ± 0.08 | 4.11 ± 0.67 | 2.22 ± 0.20 |
| Com.14 | 2.05 ± 0.05 | 1.26 ± 0.04 | 0.84 ± 0.01 |
| Com.16 | 2.25 ± 0.10 | 3.92 ± 0.43 | 2.88 ± 0.54 |
| Re. | 2.12 ± 0.09 | 2.64 ± 0.14 | 3.34 ± 0.23 |
The threshold for docking scores was established by comparing the docking scores of the test compounds (Com. 1–Com. 16) with the reference drug gemcitabine. Compounds with more negative scores than gemcitabine were considered to have better binding affinities. Favorable interactions with the major active site residues were also visualized by binding mode inspection (
The molecular docking scores and interaction of ligands Com.10 and Com.16 with MCF7.
The molecular docking scores and interaction of ligands Com.10 and Com.16 with A549.
The molecular docking scores and interaction of ligands Com.1- Com.16 with PC3.
| St. NO | M. W. | H-B-A | H-B-D | TPSA | ILOGP | H-A | R-B | Bio-S | GI | BBB |
|---|---|---|---|---|---|---|---|---|---|---|
| Com.7 | 468.44 | 9 | 3 | 150.54 | 1.19 | 34 | 6 | 0.55 | Low | No |
| Com.9 | 834.7 | 21 | 6 | 366.09 | 1.69 | 59 | 12 | 0.17 | Low | No |
| Com.10 | 882.75 | 21 | 6 | 366.09 | 1.24 | 63 | 12 | 0.17 | Low | No |
| Com.14 | 546.49 | 13 | 4 | 234.53 | 1.77 | 38 | 7 | 0.17 | Low | No |
| Com.15 | 804.72 | 18 | 6 | 308.4 | 2.73 | 58 | 9 | 0.17 | Low | No |
| Com.16 | 840.73 | 20 | 6 | 350.92 | 2.1 | 59 | 10 | 0.17 | Low | No |
Inspecting cell line A549, the protein 4ZXT among all compounds, Com. 7, Com. 9, Com. 10, and Com. 16 stood out significantly, displaying MolDock scores of −163.136, −160.115, −170.009, and −187.661, respectively, suggesting their potential. Firstly, Com. 7 displays 19 contact points with LYS114, MET108, THR110, GLU33, GLY34, GLU33, ILE31, ALA35, ALA35, TYR36, LYS54, and LYS114. They were engaged in hydrogen bonding. In contrast, LEU156, VAL39, LYS54, ALA35, LYS54, LYS54, and ILE56 are hydrophobic contacts (Fig.
Examination of cell line PC3, the protein 7LXL, displays notable interactions with Com. 2, Com. 8, Com. 16, and Com. 17 with MolDock scores of −161.155, −170.469, −167.632, −163.718, and −221.078, respectively. Initially, Com. 1 showed five contact points with ALA370, ASP76, ASP76, PHE57, and ILE223. The first three are hydrogen bonds, and the last two are hydrophobic in nature (Fig.
Docking simulations indicated that gemcitabine had stable binding to the main ERK2 residues (LYS54, GLU71, ASP111) in the ATP binding pocket (RMSD 1.83 Å), which could imply the inhibition of kinase activity (Yoshimi Nakajima et al. 2012;
During the drug discovery process, it is imperative to investigate the variables of absorption, distribution, metabolism, and excretion (ADME) to minimize the risk of pharmacokinetics-related clinical failure. With their molecular weights more than 500 (MW > 500), except for Com. 7 (MW = 468.44), IlogP values ranging from 1.19 to 2.73 (IlogP < 5), number of hydrogen bond acceptors (H-B-As) ranging from 9 to 21 (HBA ≥ 10), and number of hydrogen bond donors (HBDs) ranging from 3 to 6 (HBDs ≥ 5) (
In summary, sixteen gemcitabine derivatives, including three heterocyclic moieties, are 1,2,3-triazoline, five compounds of 1,2,3-triazole, and eight tetrazole compounds. All these compounds were prepared from previous studies using gemcitabine as a starting material. In this study, we evaluated them for their anticancer activities. In addition, compounds Com. 10 and Com. 16 showed potent anticancer activity against all tested cell lines. The compound Com. 16 exhibited potent anticancer activity with IC₅₀ = 12.23, 6.599, and 21.57 µM against all MCF7, A549, and PC3 compared with the reference drug gemcitabine. Whereas, Com. 10 exhibited potent anticancer activity with IC₅₀ = 9.454, 6.931, and 12.09 µM against all MCF7, A549, and PC3 compared with the reference drug gemcitabine. Moreover, other compounds, Com. 9 and Com. 14, also exhibited potent anticancer activity against some tested cell lines; Com. 9 showed potent anticancer activity with IC₅₀ = 29.42 µM against the A549 cell line. On the other hand, Com. 9 and Com. 14 showed potent anticancer activity with IC₅₀ = 39.39 and 36.29 µM against the PC3 cell line, respectively. Molecular docking showed that the most suitable targets for anticancer efficacy are dihydrofolate reductase protein from cell lines, vascular endothelial growth factor receptor, and histone deacetylase. Compound Com. 16, the most effective anticancer, showed good interactions against 6NEW, 4ZXT, and 4LXT with affinities of −191.721, −187.661, and −221.078 kcal/mol, respectively. Especially, this compound showed electrostatic carbon–hydrogen bonds with halogen and hydrophobic interactions, hydrophobic being π–sigma, π–sulfur, π–lone pair, π–π T-shaped, and π–alkyl type, that resemble the co-crystallization ligand and reference drugs. Other compounds showed good docking results; the threshold is set to be −160 to −180. Only eight protein–ligand complexes emerged as lead based on their score. The ADME profile was evaluated for the six most active compounds in comparison to gemcitabine as a reference drug.
The authors are greatly thankful to the Al-Kafeel Center for Medical and Pharmaceutical Sciences staff for carrying out the anticancer activity tests and for the assistance provided in completing this study.
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.
Use of AI
No use of AI was reported.
Funding
This work was supported by AlKafeel Center for Medical and Pharmaceutical Sciences.
Author contributions
All authors have contributed equally.
Author ORCIDs
Ali Jabbar Radhi https://orcid.org/0000-0002-7578-1716
Data availability
All of the data that support the findings of this study are available in the main text or Supplementary Information.
Supplamantary data
Data type: docx