Research Article |
Corresponding author: Huda Ghassan Hameed ( huda.ghassan@gau.edu.iq ) Academic editor: Alexandrina Mateeva
© 2024 Huda Ghassan Hameed, Hayder B. Sahib, Zahraa Sabbar Omran.
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:
Ghassan Hameed H, Sahib HB, Sabbar Omran Z (2024) Investigating the anti-carcinogenic potential action of 1,2,3 triazole core compounds: impact of introducing an aldehyde or Nitro group, integrating cell line studies, and in silico ADME and protein target prediction. Pharmacia 71: 1-9. https://doi.org/10.3897/pharmacia.71.e123794
|
The development of novel chemotherapeutic drugs begins with the suppression of cancer and angiogenesis. Ringed compounds with one or more heteroatoms are known as heterocyclic compounds. In organic chemistry and the pharmaceutical sector, heterocyclic compounds containing nitrogen atoms are valuable. In pharmaceutical chemistry, molecules containing a 1,2,3-triazole skeleton are particularly favored. They have great stability, making it simple to bind them to biomolecular targets. In this work, two 1,2,3-triazole scaffolds containing new chemical entities were assessed using the MTT assay against two malignant (MCF-7 and HCT116) and one normal (HUVECs) cell lines with the goal of creating a new leading prodrug for cancer treatment. The ligands were well characterized by FTIR and 1HNMR. In silico ADMET studies show acceptable pharmacokinetic properties. With the aid of the ligands’ SWISS target protein prediction, the in silico binding to target proteins was examined. The two compounds exhibited a dose-dependent cytotoxic effect, with the H4 compound demonstrating a favorable selectivity index against MCF-7 breast cancer, indicating its potential as a leading compound for anticancer prodrugs.
cancer, 1, 2, 3 triazole scaffolds, ADME (absorption, distribution, metabolism, and excretion), selectivity index
Globally, infectious illnesses and cancer represent a significant health burden on humanity (Kamal et al. 2010). The WHO predicts that in the upcoming years, the death rate from cancer will double (
Herbs have long been used to treat cancer; in fact, the use of herbs is the primary source of many traditional medicines used to treat a wide range of ailments. Even though plant-derived molecules, such as those from Phoenix Dactylifera seeds (Husseina et al. 2018), Cuminum cyminum seeds extract confirmed by Mahmood A and Sahib H (2017), Zizyphus spinachristi leaf extracts (
The development of drugs has greatly benefited from the use of heterocycles to diminish side effects and overcome drug resistance. Many previous studies have proved that the flexible scaffold 1,2,3-triazole demonstrates potential pharmacological activity and has received growing attention as a source of both antimicrobial and anticancer drugs. It is present in many bioactive compounds, including antiviral (
In the field of in silico prediction, various methods are employed to predict off-target hits in drug discovery, like the similarity ensemble approach (SEA), which has the advantages of utilizing ligand-based similarity to predict off-target interactions and can identify unreported targets of FDA-approved and investigational drugs. While the disadvantages are illustrated by the fact that the SEA approach relies on known ligand-target interactions, limiting its applicability to novel compounds, it also may not capture structural features critical for target binding (
Machine learning models are other methods for in silico prediction. It comes with the advantages of the capability to analyze large datasets, identify complex patterns, and predict off-target interactions based on diverse data sources. Disadvantages include: performance may be limited by incomplete or irrelevant training data; and challenges in predicting outside the training set due to chemical space and target diversity (
Within the FDA-approved target coverage, the Swiss Target Prediction has a bigger pharmacological space and has demonstrated a strong capacity to forecast the “old” targets for novel medications. Moreover, broad chemical space coverage with high computational efficiency is limited by the limitations of predictive accuracy and incomplete target coverage.
Each method has its strengths and limitations, and the choice of method depends on the specific research question and available data. Integrating multiple approaches can enhance the robustness of predictions and provide a more comprehensive understanding of off-target interactions in drug discovery (
This study aims to determine the effects of 1,2,3 triazole-related compounds as anti-cancer and antiangiogenic using three different cell lines, along with the ADME (absorption, distribution, metabolism, and excretion) prediction for the pharmacokinetic profile and targeted protein prediction for a possible explanation of the results.
This study was carried out at Al-Nahrain University, College of Medicine, Department of Pharmacology, during the period from the 1st of October 2022 to the 1st of April 2023. The study was given approval by the institutional Scientific and Ethical Committees. The new chemical entities will be referred to in the article as H1 and H4. Both compounds were generously provided by the Department of Pharmaceutical Chemistry, College of Pharmacy, University of Al-Kafeel, Najaf, Iraq. The chemical structures of H1 are (3-benzamido-2-(4-((4-formylphenoxy)methyl)-1H-1,2,3-triazol-1-yl)-3-phenylpropanoic acid) (Fig.
Analytical and spectral data for each compound, as presented by the supplier, are:
For H1 compound: FT-IR (KBr, cm-1) : 1215-1237 ((N-N=N) stretching vibration), 1732 ((C=O) stretching vibration of carboxylic acid), 1656 ((C=O) stretching vibration of amide), 3024 ((C-H) aromatic stretching vibration), 3410 (Broad (O-H) stretching of carboxylic acid), 3352 (N-H stretching vibration of secondary amide) , 1030 ((C-O) stretching vibration). 1H-NMR (300 MHz, DMSO-d6) δ 9.87 (s, 1H, -CHO), 8.75 (s, 1H, triazole ring), 7.96–7.09 (aromatic protons), 5.94 (d, J = 4.0 Hz, 1H, CH-triazole ring), 5.45 (d, J = 8.1, 3.9 Hz, 1H, Ph- CH-), 3.52 (s, 3H, -OCH3).
For H4 compound: FT-IR (KBr, cm-1): 1246 ((N-N=N) stretching vibration), 1739 ((C=O) stretching vibration of carboxylic acid), 1643 ((C=O) stretching vibration of amide), 3032 ((C-H) aromatic stretching vibration), 3363 (Broad (O-H) stretching of carboxylic acid), 3255 (N-H stretching vibration of secondary amide) ,1180 ((C-O-C) stretching vibration). 1H-NMR (300 MHz, DMSO-d6) δ 9.27 (s, 1H, -CHO), 7.53 (d, 1H, triazole ring), 7.88–7.26 (aromatic protons), 4.99 (m, 1H, CH-N), 8.22 (m, 2H, C-NO2), 5.26 (s, 2H, -O-CH2), 9.27 (s, 1H, CHO).
The cytotoxic effect of both compounds (H1 and H4) on cell viability and proliferation was assessed using a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, according to the Mosmann method (Mosmann 1983). The American Type Culture Collection provided the human umbilical ventricular endothelial cell line (HUVEC), breast cancer cell line (MCF-7), and colon cancer cell line (HCT116). Every cell line was kept alive in its own medium using RPMI-1640. Additionally, 10% heat-inactivated fetal calf serum (HIFCS) was bought and used. To create the full growth medium specified in the growth medium sheet included with the cell line, 1% penicillin or streptomycin was added to those mediums. Before the trials, each prepared medium had a full growth medium. at a concentration of 1 × 104 /mL were planted into 96-well culture plates with 200 μl of complete growth medium and incubated with various concentrations (6.25, 12.5, 25, 50, 100, and 200 µg/mL) of the tested compounds (H1 and H4) each alone for 24 h. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay was used to assess the test chemicals’ effects on human cancer cell viability, which was expressed as the percentage of cell survival.
Cell growth viability and inhibition were calculated using the following formula:
Cell viability (%) = (A0/A) × 100
Cell inhibition (%) = 1- cell viability (%)
Where: A0 = absorbance of the samples A = absorbance of the negative control.
Using a microplate reader, the absorbance (A) was measured at 570 nm and the reference at 650 nm. Inhibitory concentration (IC50) values were calculated by using linear and logarithmic equations.
The SI value can be calculated using the following equation (
SI = IC50 (non-target cell line)/IC50 (target cell line)
Selectivity Index = IC50 for normal cells and IC50 for cancer cells.
The absorption, distribution, metabolism, and excretion (ADME) pharmacokinetic profiles of the potential compounds were computed using Swiss ADME (
A two-sample, two-tailed t-test was used for testing the significant differences in the inhibitory effects of H1 and H4. P-values of < 0.05 are the lowest limit of significance. The Excel 2010 program was used for computing the data (Microsoft Cooperation, Redmond, Washington, U.S.). Figs
The concentration of a plant extract, drug, or chemical entity (µg/mL) at which 50% of tested animals or cells in cell lines die is known as IC50%. The IC50 value was calculated for three cell lines by MTT assay: two cancerous ones (MCF-7 and HCT-116) and one normal cell line (HUVECs), as illustrated in Table
IC50 (µg\mL) | |||
---|---|---|---|
Cell type | Cell line | H1 | H4 |
Breast cancer | MCF-7 | 24.62585028 | 15.45714282 |
Human colon cancer | HCT116 | 47.14285701 | 26.84353733 |
Human umbilical vein endothelial cells | HUVEC | 14.89455781 | 59.75272103 |
Inhibition activity of H1 and H4 compounds on human umbilical vein endothelial cells (HUVECs).
Dose (µg\mL) | Percentage of inhibition of H1 | Percentage of inhibition of H4 |
---|---|---|
200 | 13 | 73 |
100 | 16 | 63 |
50 | 22 | 55 |
25 | 16 | 52 |
12.5 | 7 | 52 |
6.25 | 12 | 53 |
Dose (µg\ml) | Percentage of inhibition of H1 | Percentage of inhibition of H4 |
---|---|---|
200 | 46 | 21 |
100 | 20 | 21 |
50 | 20 | 6 |
25 | 14 | 21 |
12.5 | 17 | 12 |
6.25 | 17 | 3 |
Dose (µg\ml) | Percentage of inhibition of H1 | Percentage of inhibition of H4 |
---|---|---|
200 | 57 | 42 |
100 | 43 | 29 |
50 | 43 | 26 |
25 | 29 | 23 |
12.5 | 19 | 14 |
6.25 | 10 | 10 |
The cytotoxic extracts’ level of selectivity was quantified as SI = IC50 in normal cells/IC50 in tumor cells. The results shown in Table
Both compounds showed low skin permeability, moderate water solubility, no BBB permeation, and similar effects on the five major isoforms of cytochromes P450 as inhibitors for CYP2C19, CYP2C9, and CYP3A4. H1 compounds show higher GI absorption and better bioavailability than H4, and they serve as substrates for the P-gp efflux pump. All pharmacokinetic results are illustrated in Table
The pharmacokinetic characteristics of the potential candidate compounds predicted by Swiss ADME.
Name | MW g/mol | Rotatable bonds | #H-bond ACC | #H-bond Don | TPSA (A2) | LOGP. (o/w) | LOGS (ESOL) | Water sol. | GI abs. | BBB perm. | P-gp substrate | CYP1A2 inh. | CYP2C19 inh. | CYP2C9 inh. | CYP2D6 inh. | CYP3A4 inh. | Log Kp cm/s | Lipinski | Bioavailability |
---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
H1 | 470.48 | 11 | 7 | 2 | 123.41 | 2.63 | -4.33 | Moderate | High | No | Yes | No | Yes | Yes | No | Yes | -7.13 | 0 | 0.56 |
H4 | 487.46 | 11 | 8 | 2 | 152.16 | 2.11 | -4.66 | Moderate | low | No | No | No | Yes | Yes | No | Yes | -6.97 | 1 | 0.11 |
Pharmacodynamics of the identified candidate compounds by Swiss Target Prediction (the common targets between the two compounds were colored the same).
Protein Target | Target Class | CHEMBL ID | |
---|---|---|---|
H1 | Integrin alpha-4/ beta-1 | Membrane receptor | CHEMBL1907599 |
Integrin alpha-4/ beta-7 | Membrane receptor | CHEMBL2095184 | |
Integrin alpha-V/ beta-3 | Membrane receptor | CHEMBL1907598 | |
Peroxisome proliferator-activated receptor gamma | Nuclear receptor | CHEMBL235 | |
Peroxisome proliferator-activated receptor alpha | Nuclear receptor | CHEMBL239 | |
Matrix metalloproteinase 12 | Protease | CHEMBL4393 | |
5-lipoxygenase activating protein | cytosolic protein | CHEMBL4550 | |
G protein-coupled receptor 44 | Family A G protein coupled receptor | CHEMBL5071 | |
Caspase-1 | Protease | CHEMBL4801 | |
H4 | Endothelin receptor ET-A | Family A G protein coupled receptor | CHEMBL252 |
Matrix metalloproteinase 3 | Protease | CHEMBL283 | |
Integrin alpha-4/ beta- | Membrane receptor | CHEMBL2095184 | |
Peroxisome proliferator-activated receptor gamma | Nuclear receptor | CHEMBL235 | |
Peroxisome proliferator-activated receptor alpha | Nuclear receptor | CHEMBL239 | |
Angiotensin converting enzyme | Protease | CHEMBL1808 | |
Caspase-1 | Protease | CHEMBL4801 | |
G protein-coupled receptor 44 | Family A G protein coupled receptor | CHEMBL5071 |
In our study, we offer a workflow for in silico target prediction that can predict and suggest new molecular targets for new chemical entities. The results revealed the polypharmacology of both H1 and H4 compounds as follows:
Regarding the anti-cancer effects, both compounds could serve as a lead to developing multitarget inhibitors directed against cancer. Molecules containing a 1,2,3-triazole skeleton are exceedingly favored in medicinal chemistry. They bind to biomolecular targets with ease and have a stable nature (
Swiss Target Prediction is a web server that uses strategies for assessing chemical similarity through the use of molecular fingerprints, often referred to as 2D similarity. Compounds that are highly similar according to these metrics typically exhibit an increased propensity to interact with similar molecular targets (
With the help of the SWISS target prediction shown in Table
Table
The ratio of the toxic concentration of a plant or drug sample against its effective bioactive concentration is known as the selectivity index (
The use of chemical side groups in prodrug synthesis in medicine is expanding on a daily basis, and the variety of their analogues offers a significant and feasible avenue for the discovery of pharmaceuticals with a range of biological uses. When it comes to creating structurally varied heterocyclic compounds with uses in combinatorial chemistry, diversity-oriented synthesis, bioconjugation chemistry, and drug discovery, the method of creating triazole derivatives may have important implications and may be considered a new hope for curing cancer.
We gratefully acknowledge all the individuals who supported us throughout the completion of this study.