Skip to main content
PLOS One logoLink to PLOS One
. 2025 Nov 3;20(11):e0334620. doi: 10.1371/journal.pone.0334620

Novel pyrimidine-substituted chalcones: In vitro antioxidant properties and cytotoxic effects against human cancer cell lines

Prashant Nayak 1, Vikram S Shenoy 2, Vijay Upadhye 3, Kasim Sakran Abass 4, Omar Awad Alsaidan 5, Mukesh Soni 6,7, Sunil Tulshiram Hajare 8,*
Editor: Afzal Basha Shaik9
PMCID: PMC12582495  PMID: 41183108

Abstract

A series of novel pyrimidine-substituted chalcones were synthesized, purified, and characterized. Four selected compounds (CFMPY 4, 15, 17, and 28) exhibited modest antioxidant activity across six In vitro radical scavenging assays. The In vitro cytotoxic (anticancer) potential of five synthesized chalcones (CFMPY-2, 4, 15, 17, and 28) was evaluated using MTT, Sulforhodamine B (SRB), DNA fragmentation, Nuclear staining, and Farnesyl transferase assays. Notably, this study is the first to employ a DNA fragmentation assay to assess the anticancer activity of these test compounds. MTT and SRB assays revealed significant cytotoxicity in HeLa and A549 cell lines for all compounds except CFMPY 17, with IC50 values ranging from 2.28 to 5.48 µg/ml, demonstrating comparable or superior efficacy to cisplatin (IC50 values of 5.27 µg/ml in HeLa and 4.05 µg/ml in A549 cells). Nuclear staining and DNA fragmentation assays confirmed the induction of apoptosis by all tested compounds, including cisplatin, with the latter revealing characteristic 200 bp DNA laddering. Furthermore, the Farnesyl transferase assay indicated good cytotoxic activity for all compounds except CFMPY 17. These findings suggest that pyrimidine-substituted chalcones represent a promising class of cytotoxic (anticancer) agents, potentially exceeding the efficacy of cisplatin in certain contexts.

Introduction

Chalcones (1,3-diphenyl-2-propen-1-ones) are linear flavonoids that can be found widely in nature, particularly in fruits, vegetables, teas, and spices. They are recognized as important precursors in the biosynthetic flavonoid pathway, and their varied structures make them appealing frameworks in medicinal chemistry [1]. In the past five years, there has been a resurgence of interest in chalcones as multifunctional bioactive compounds exhibiting antioxidant, antimicrobial, anti-inflammatory, and anticancer activities [24]. Their ability to counteract oxidative stress-related diseases is primarily facilitated through their antioxidant properties, which involve radical scavenging and redox modulation [5]. Additionally, derivatives of chalcones have showcased selective cytotoxic effects against various cancer cell lines, including those from breast, cervical, and colon cancers, frequently by inducing caspase-dependent apoptosis and disrupting mitochondrial pathways [6,7]. These results highlight the potential of chalcones as promising candidates for new drug development [8].

Chalcones have garnered significant interest in pharmacological investigations owing to their broad spectrum of bioactivities. Notably, they exhibit anti-inflammatory properties, a critical factor in the pathogenesis of numerous diseases. Furthermore, chalcones demonstrate antimicrobial activity against a diverse array of microorganisms, suggesting potential applications in the treatment of infectious diseases. Their capacity to function as radical scavengers contributes to the mitigation of oxidative stress and associated pathologies. Parallel to chalcones, pyrimidines have emerged as a privileged scaffold in oncology drug discovery. Reviews from 2020 onward consistently report the diverse pharmacological activities of pyrimidine derivatives, including anticancer, antimicrobial, and immuno-oncological effects [9,10]. In particular, pyrimidine–sulfonamide hybrids and fused pyrimidine analogues show strong multi-targeted anticancer activity by engaging kinase pathways and disrupting oncogenic signaling [11,12]. Importantly, pyrimidine substitution often improves aqueous solubility, hydrogen-bonding capacity, and cellular uptake—features highly desirable for rational anticancer drug design. These multifaceted pharmacological attributes position chalcones as promising lead compounds for the development of novel therapeutic modalities. Contemporary drug design strategies have focused on structural modifications of the chalcone scaffold to enhance their pharmacokinetic profiles, particularly bioavailability, and to elucidate the structure-activity relationships (SAR) associated with various substituents on the aryl or heteroaryl moieties in the context of anticancer drug development. Ongoing research is dedicated to elucidating the underlying molecular mechanisms mediating these biological effects and comprehensively evaluating the therapeutic potential of chalcones.

Farnesyltransferase inhibitors (FTIs) represent a class of experimental anticancer therapeutics that target farnesyltransferase (FTase), an enzyme critical for the post-translational modification of proteins, including the Ras GTPases, which are frequently oncogenically activated in various malignancies [13,14].

Protein prenylation, the post-translational attachment of C15-farnesyl or C20-geranylgeranyl isoprenoid moieties, is catalyzed by a family of transferases. In the context of cancer therapeutics, the inhibition of two key prenyltransferases, FTase and geranylgeranyltransferase-I (GGTase-I), has emerged as a strategy due to their capacity to act on Ras isoforms [15]. These two enzymes are heterodimeric proteins, sharing a common α-subunit but possessing distinct β-subunits, conferring substrate specificity. Physiologically, in mammalian cells, the post-translational processing of canonical Ras proteins primarily involves FTase-mediated farnesylation. However, upon pharmacological inhibition of FTase by FTIs, proteins such as RhoB, K-Ras, and N-Ras can undergo alternative prenylation by GGTase-I, highlighting a compensatory resistance mechanism.

The substrate recognition site for both FTase and GGTase-I is the conserved carboxyl-terminal tetrapeptide motif, the CAAX box (Cys-Aliphatic-Aliphatic-X), where the terminal amino acid (X) dictates the preference for farnesylation versus geranylgeranylation. Notably, Boguski and McCormick [16] demonstrated that FTase exhibits preferential activity towards protein substrates bearing CAAX motifs terminating in serine, methionine, or glycine. The In vitro enzymatic affinity for substrates where X is methionine (e.g., K-Ras A and B with CVIM and CIIM sequences, respectively) is significantly (10–30 fold) higher compared to those terminating in serine or glycine (e.g., H-Ras with CVLS). Conversely, GGTase-I displays preferential activity towards proteins containing leucine as the terminal residue in their CAAX motif.

While all three major Ras isoforms (H-Ras, N-Ras, and K-Ras) are typically farnesylated, the presence of an FTI can lead to K-Ras and N-Ras serving as alternative substrates for GGTase-I. Consequently, K-Ras exhibits increased resistance to FTIs due to this alternative prenylation by GGTase-I, coupled with its inherently higher affinity for FTase. Therefore, a combinatorial therapeutic approach involving both FTIs and geranylgeranyltransferase inhibitors (GGTIs) is posited as necessary to achieve complete blockade of K-Ras prenylation [17], thereby potentially enhancing the efficacy of Ras-targeted cancer therapies.

The post-translational modification of the Ras protein, a key regulator of cell growth and differentiation, is initiated by the farnesylation of its carboxyl-terminal CAAX motif by farnesyltransferase (FTase). This crucial enzymatic step, involving the transfer of a farnesyl moiety from farnesyl pyrophosphate (FPP), is essential for the proper localization and function of Ras at the cell membrane [18]. Aberrant activation of Ras isoforms is a frequent occurrence in a wide range of human cancers, driving uncontrolled cell proliferation, invasiveness, and metastatic potential – the defining hallmarks that distinguish malignant from benign tumors [19]. Given that virtually any living cell in the body can undergo malignant transformation, and considering the diverse etiologies of various cancer types, the development of effective and targeted anticancer therapies remains a paramount challenge in biomedical research. In this context, the exploration of novel therapeutic strategies that disrupt oncogenic Ras signaling pathways holds immense significance. Farnesyltransferase inhibitors (FTIs) have emerged as a promising class of anticancer agents by specifically blocking the initial farnesylation step of Ras, thereby impairing its downstream signaling and potentially inhibiting tumor growth. Despite extensive reports on the diverse bioactivities of chalcones, their therapeutic utility is often limited by modest potency and non-specificity. Rational structural modification of the chalcone scaffold has been shown to improve pharmacological profiles, particularly in the context of anticancer activity. Among various heterocyclic substitutions, the pyrimidine moiety is a privileged scaffold in medicinal chemistry due to its well-documented ability to enhance bioavailability, strengthen hydrogen-bonding interactions with biological targets, and confer improved cytotoxic selectivity. Furthermore, pyrimidine-based compounds have previously demonstrated activity against oncogenic signaling pathways, including Ras-dependent cascades, suggesting a potential for farnesyltransferase inhibition. To emphasize the structural novelty of our design, a schematic comparison (Fig 1) is provided, illustrating similarities and differences between the synthesized pyrimidine-substituted chalcones, previously reported chalcones with related antioxidant/cytotoxic activity, and representative pyrimidine-based drugs.

Fig 1. Schematic comparison illustrating the structural novelty of the synthesized pyrimidine-substituted chalcones.

Fig 1

The structures of the synthesized pyrimidine-substituted chalcones are compared with previously reported chalcones and known pyrimidine-based anticancer drugs such as 5-Fluorouracil and Pemetrexed.

Therefore, we hypothesized that pyrimidine substitution in chalcones could synergize radical-scavenging with Ras-targeted anticancer activity. To test this, we synthesized and evaluated pyrimidine-substituted chalcones for antioxidant and anticancer potential across multiple cell lines, using assays including MTT, SRB, nuclear staining, DNA fragmentation, and FTase inhibition.

The present study specifically aims to explore the anticancer potential of pyrimidine-substituted chalcones by evaluating their cytotoxic effects against human cancer cell lines and investigating apoptosis and farnesyltransferase inhibition as possible mechanisms. To achieve this, we employed a multifaceted approach encompassing MTT and Sulforhodamine B assays, nuclear staining, DNA fragmentation, and a farnesyl transferase assay.

Materials and methods

Experimental synthesis of test compounds

Fig 2 outlines the synthetic route for pyrimidine-substituted chalcone derivatives. Chalcones were synthesized via a Claisen-Schmidt condensation. Briefly, 2-fluoro-4-methoxy acetophenone (122.5 mL) was reacted with various substituted benzaldehydes in a solution of sodium hydroxide (22.5 g in 200 mL water, corresponding to ~2.8 M NaOH) and rectified spirit (122.5 mL) and rectified spirit (122.5 mL) under cold conditions (ice bath) with mechanical stirring. The reaction mixture was stirred overnight at refrigerated temperatures. The resulting crude chalcones were then isolated by filtration and purified by recrystallization from ethanol [20,21].

Fig 2. Synthetic scheme for the preparation of 2-amino-4-(2-fluoro-4-methoxyphenyl)-6-substituted phenylpyrimidine derivatives (CFMPY-2, 4, 15, 17, and 28).

Fig 2

The scheme illustrates the Claisen–Schmidt condensation followed by cyclization with guanidine hydrochloride.

Guanidine hydrochloride and the previously synthesized chalcone derivatives were dissolved in absolute ethanol. A solution of potassium hydroxide (prepared by dissolving the required amount in a minimal volume of distilled water, specifically twice the mass of the chalcones and guanidine hydrochloride combined) was added to the ethanolic reaction mixture. The resulting mixture was then refluxed for 6 hours. Following the reflux period, the reaction mixture was poured into 250 mL of ice-cold distilled water to induce precipitation and facilitate crystallization of the pyrimidine-substituted chalcones. The resulting solid was then typically collected by filtration and further purified (e.g., by recrystallization) to obtain the desired compounds [22].

Compound purity was assessed using ascending Thin Layer Chromatography (TLC) on pre-coated Silica gel-G plates, with visualization of spots achieved by exposure to iodine vapors. A computational docking study using the Schrödinger software package was employed to select compounds CFMPY2, CFMPY4, CFMPY15, CFMPY17, and CFMPY28 for further biological evaluation. Structural characterization of the synthesized compounds was performed using spectroscopic and analytical techniques, including Infrared (IR) spectroscopy, Proton Nuclear Magnetic Resonance ($^1$H NMR) spectroscopy, mass spectrometry, and elemental analysis. IR spectra were recorded as potassium bromide (KBr) discs on a Shimadzu IR Affinity FT-IR spectrophotometer, and characteristic absorption bands are reported in wave numbers (cm$^{−1}$). $^1$H NMR spectra were acquired on a Bruker AC 400 MHz spectrometer using Deuterated dimethyl sulfoxide (DMSO-D6) as the solvent, with tetramethylsilane (TMS) serving as the internal chemical shift reference. Chemical shifts (δ) are reported in parts per million (ppm). Mass spectral data and elemental analysis results were used to further confirm the molecular structures and purity of the synthesized compounds

2-amino-4-(2-fluoro-4-methoxyphenyl)-6-(2,3,4-trimethoxyphenyl)pyrimidine (CFMPY-2).

IR KBr (cm ⁻ ¹): 3056 (Ar C-H str), 1580 (Ar C = C str), 830 (Ar C-H bend), 1636 (C = N str), 3380 (N-H str), 1205 (C-F str), 1226 (C-O str). ¹H NMR (400 MHz, CDCl₃, δ ppm): 3.61 (3H, s), 3.83 (3H, s), 3.87 (3H, s), 3.84 (3H, s), 6.43 (1H, d, J = 8.4 Hz), 7.10–7.11 (2H, 7.10 (s), 7.01 (dd, J = 8.4, 1.5 Hz), 7.20 (1H, dd, J = 1.5, 0.5 Hz), 7.54–7.62 (2H, 7.58 (d, J = 8.5 Hz), 7.63 (dd, J = 8.4, 0.5 Hz). MS (M⁺): 386.

2-amino-4-(2-fluoro-4-methoxyphenyl)-6-(2,4,6-trimethoxyphenyl)pyrimidine (CFMPY-4).

IR KBr (cm ⁻ ¹): 3058 (Ar C-H str), 1582 (Ar C = C str), 836 (Ar C-H bend), 1630 (C = N str), 3397 (N-H str), 1202 (C-F str), 1223 (C-O str). ¹H NMR (400 MHz, CDCl₃, δ ppm): 3.82–3.84 (6H, 3.84 (s), 3.83 (s)), 3.86 (6H, s), 6.35 (2H, d, J = 1.8 Hz), 7.10 (1H, s), 7.12 (1H, dd, J = 8.8, 1.6 Hz), 7.31 (1H, dd, J = 1.6, 0.5 Hz), 7.58 (1H, dd, J = 8.8, 0.5 Hz). MS (M⁺): 386.

2-amino-4-(2-fluoro-4-methoxyphenyl)-6-(2,4-dimethoxyphenyl)pyrimidine(CFMPY-15).

IR KBr (cm ⁻ ¹): 3050 (Ar C-H str), 1586 (Ar C = C str), 828 (Ar C-H bend), 1622 (C = N str), 3387 (N-H str), 1206 (C-F str), 1230 (C-O str). ¹H NMR (400 MHz, CDCl₃, δ ppm): 3.83–3.84 (6H, 3.84 (s), 3.84 (s)), 3.89 (3H, s), 6.56 (1H, dd, J = 1.6, 0.5 Hz), 6.97 (1H, dd, J = 8.6, 1.6 Hz), 7.09–7.15 (2H, 7.10 (s), 7.13 (dd, J = 8.6, 1.6 Hz)), 7.28 (1H, dd, J = 1.6, 0.5 Hz), 7.63 (1H, dd, J = 8.6, 0.5 Hz), 7.72 (1H, dd, J = 8.6, 0.5 Hz). MS (M⁺): 355.

2-amino-4-(2-fluoro-4-methoxyphenyl)-6-(2,4,5-trimethoxyphenyl)pyrimidine (CFMPY-17).

IR KBr (cm ⁻ ¹): 3052 (Ar C-H str), 1579 (Ar C = C str), 832 (Ar C-H bend), 1618 (C = N str), 3385 (N-H str), 1208 (C-F str), 1232 (C-O str). ¹H NMR (400 MHz, CDCl₃, δ ppm): 3.61 (3H, s), 3.82 (3H, s), 3.83 (3H, s), 3.82 (3H, s), 6.52 (1H, d, J = 0.4 Hz), 7.05–7.10 (2H, 7.10 (s), 7.09 (dd, J = 8.4, 1.5 Hz)), 7.18 (1H, dd, J = 1.5, 0.5 Hz), 7.42 (1H, d, J = 0.4 Hz), 7.61 (1H, dd, J = 8.4, 0.5 Hz). MS (M⁺): 386.

2-amino-4-(2-fluoro-4-methoxyphenyl)-6-(3,4,5-trimethoxyphenyl)pyrimidine (CFMPY-28).

IR KBr (cm ⁻ ¹): 3048 (Ar C-H str), 1578 (Ar C = C str), 826 (Ar C-H bend), 1625 (C = N str), 3382 (N-H str), 1206 (C-F str), 1235 (C-O str). ¹H NMR (400 MHz, CDCl₃, δ ppm): 3.64 (6H, s), 3.83 (3H, s), 3.81 (3H, s), 7.09 (1H, dd, J = 8.4, 1.5 Hz), 7.13 (1H, dd, J = 1.5, 0.5 Hz), 7.20 (1H, s), 7.26 (2H, d, J = 2.4 Hz), 7.63 (1H, dd, J = 8.4, 0.5 Hz). MS (M⁺): 386.

MTT assay

Adherent cell lines were grown in Dulbecco’s Modified Eagle’s Medium (DMEM), enriched with 10% (v/v) heat-inactivated Fetal Bovine Serum (FBS; please indicate the supplier and batch number if it is crucial). The conditions were maintained at 37°C in a humid atmosphere with 5% CO$_2$. For the assessment of In vitro cytotoxicity, cells were plated in 96-well microplates (such as those made of tissue culture-treated polystyrene) at a specific seeding density (cells/well) to guarantee sub-confluent growth throughout the assay duration and allowed to adhere for a duration of 24 hours. After the cells had adhered, test compounds were added in serial dilutions (usually log$_{10}$ or two-fold) in the culture medium. Control wells received a corresponding concentration of the solvent (for example, DMSO, with a maximum final concentration of ≤ 0.1% v/v) to mitigate potential effects from the solvent. The cells were then incubated for 72 hours under standard culture conditions. After incubation, the culture medium was removed, and replaced with a sterile-filtered MTT solution (for instance, 0.5 mg/mL in sterile PBS). Following a 2–4 hour incubation at 37°C, the MTT solution was carefully discarded, and the intracellular formazan crystals were dissolved by adding a specific volume of an appropriate solvent (such as anhydrous DMSO or isopropanol containing 0.04 N HCl). The absorbance was measured using a microplate spectrophotometer (ELISA reader) at a primary wavelength of 570 nm and a reference wavelength of 630 nm to reduce background interference. Cytotoxicity was determined as a percentage in relation to the mean absorbance of the untreated control wells utilizing the following formula:

%𝐂𝐲𝐭𝐨𝐭𝐨𝐱𝐢𝐜𝐢𝐭𝐲=(Absorbance of controlAbsorbance of test)/(Absorbance of control)×100

Sulphorhodamine B assay

Cells were cultured in Dulbecco’s modified essential medium with 10% FBS and fetal bovine serum. The cell suspension was diluted and applied to 96-well microliter plates. The plates were incubated at 37°C for 24 hours, then drug concentrations were applied. After 72 hours, a 10% concentration of trichloroacetic acid was added. The plates were cleaned, air dried, and then dissolved in a sample containing 10mM tris base. The absorbance of each well was measured at 540nm using an ELISA reader, and the percentage cytotoxicity was calculated.:

%𝐂𝐲𝐭𝐨𝐭𝐨𝐱𝐢𝐜𝐢𝐭𝐲=(Absorbance of controlAbsorbance of test)/(Absorbance of control)×100

DNA fragmentation assay

Cell harvesting.

HeLa cells (5 x 10$^5$ - 1 x 10$^6$) were harvested and pelleted by centrifugation at 4000 rpm (approximately 2500 x g) for 5 minutes at 4°C in a 1.5 mL microcentrifuge tube. The supernatant was carefully aspirated and retained for potential downstream analyses (e.g., protein assays), flash-frozen in liquid nitrogen, and stored at −20°C. A brief secondary centrifugation (15 seconds at the same conditions) was performed to ensure complete removal of the supernatant.

Cell lysis and protein digestion.

The cell pellet was resuspended by vigorous vortexing in 20 µL of Lysis Buffer I (composition: EDTA to chelate divalent cations inhibiting nuclease activity, Proteinase K for protein degradation including histones and DNases, and Sodium Lauryl Sarcosinate (SLS) to denature proteins and release DNA). The resulting lysate was incubated at 50°C for 1 hour in a temperature-controlled heating block or water bath.

RNA digestion.

Following incubation, any condensate on the microcentrifuge tube lid was collected by brief centrifugation. The lysate was then treated with 10 µL of RNase A solution (concentration specified, e.g., 10 mg/mL, DNase-free), vortexed thoroughly, and incubated at 50°C for 1 hour to eliminate RNA contamination.

Agarose gel preparation.

A 2% (w/v) agarose gel was prepared by dissolving agarose in Tris-Phosphate-EDTA (TPE) buffer (composition specified, e.g., 89 mM Tris base, 89 mM phosphoric acid, 2 mM EDTA, pH 8.0) using microwave heating. After cooling the molten agarose to approximately 50–55°C, ethidium bromide (EtBr) was added to a final concentration of x µg/mL (e.g., 0.5 µg/mL) for DNA visualization. The agarose-EtBr solution was immediately poured into a gel casting tray with a well-forming comb positioned approximately 1 cm from one end, ensuring the gel solidified on a level surface for at least 20 minutes at room temperature.

DNA sample loading and electrophoresis.

After RNA digestion, the cell lysate was briefly centrifuged to collect any condensation. 10 µL of 6X DNA loading buffer (composition specified, e.g., containing a tracking dye and a density agent) that had been pre-heated to 70°C was added to the DNA sample. The mixture was briefly vortexed and centrifuged. The well-forming comb was carefully removed from the solidified agarose gel, and the gel was submerged in TPE buffer in the electrophoresis tank. The prepared DNA samples were carefully loaded into the wells. Electrophoresis was performed at a constant voltage of 50 V for a defined duration (e.g., 30 minutes) to allow for adequate separation of DNA fragments based on size.

DNA visualization and documentation.

Following electrophoresis, the gel was visualized under a UV transilluminator of a gel documentation system. The presence of a characteristic ladder-like pattern of DNA fragments, representing multiples of approximately 200 base pairs, indicates internucleosomal DNA fragmentation, a hallmark of apoptosis. Images of the gel were captured using the gel documentation system for analysis and record-keeping. A DNA size standard (ladder) should be run in parallel to determine the size of the DNA fragments [23].

Nuclear staining method

HeLa cells were seeded in a 6-well plate, incubated at 37°C, washed, centrifuged, and resuspended in a solution containing acridine orange and ethidium bromide in PBS. The cells were then visualized using a fluorescence microscope using a blue filter, and the supernatant was discarded after the incubation period [24].

Farnesyl transferase assay

Preparation of cytosolic fractions.

Adult male Wistar rats (obtained from Manipal Life Sciences) were humanely euthanized using chloroform inhalation, adhering to ethical guidelines. The brain was quickly extracted and placed in ice-cold 0.1 M HEPES buffer (pH 7.4) containing 25 mM MgCl$_2$ and 10 mM DTT. The tissue was meticulously cleared of meninges and blood vessels, finely diced, and homogenized in the same ice-cold buffer using a Polytron homogenizer (ten bursts at 800 rpm with intermittent cooling). The homogenate was centrifuged at 10,000 rpm (specific rotor and g-force should be indicated for reproducibility) for 30 minutes at 4°C. The supernatant obtained was then subjected to ultracentrifugation at 100,000 rpm (specific rotor and g-force should be detailed) for 60 minutes at 4°C to isolate the cytosolic fraction (supernatant), which was then aliquoted into 1 mL cryovials and preserved at −80°C. The protein concentration in the cytosolic fraction was determined using the Hartree-Lowry method, with bovine serum albumin (BSA) serving as a standard [25].

The Farnesyltransferase (FTase) activity assay was conducted in a 96-well microplate format. Each reaction mixture contained 30 μg of rat brain cytosolic protein (prepared as described previously), a defined concentration of the fluorescent dansyl peptide substrate (ranging from 1.6 to 6 μM to assess substrate dependence), and a detergent (concentration and type specified, e.g., 0.05% Triton X-100) in a total volume of V μL (calculated based on component volumes) of assay buffer. The assay buffer consisted of 52 mM Tris-HCl (pH 7.4), supplemented with 5.0 mM DTT, 12 mM MgCl$_2$, and 12 μM ZnCl$_2$. The reaction mixtures were pre-incubated at a stable temperature of 30°C for 2 minutes in a temperature-controlled microplate reader to ensure thermal equilibration. The enzymatic reaction was initiated by the automated addition of 25 μM farnesyl pyrophosphate (FPP). The contents of each well were rapidly and uniformly mixed by orbital shaking within the microplate reader.

The kinetics of the farnesylation reaction were monitored by continuously recording the fluorescence intensity at a fixed excitation wavelength of 340 nm and an emission wavelength of 530 nm using a fluorescence microplate reader (model specified). Fluorescence measurements were acquired over a period of 750 seconds, and the initial linear rate of the reaction, expressed as counts per second per second (cps/s), was determined by linear regression analysis of the fluorescence intensity versus time plot during the initial phase of the reaction where substrate depletion is negligible. All measurements for each condition were performed in triplicate (n = 3), and the mean and standard deviation (SD) or standard error of the mean (SEM) were calculated.

Control experiments were performed to assess the endogenous fluorescence contribution from the rat brain cytosol. In the absence of the dansyl peptide substrate, the basal increase in fluorescence intensity at 530 nm, attributed to endogenous fluorophores within the cytosol, was observed to be approximately 10% of the initial fluorescence value and remained stable over the assay duration, indicating minimal interference with the measurement of the enzymatic reaction. This background fluorescence was accounted for in subsequent data analysis. Full characterization data, including IR, ^1H and ^13C NMR spectra (with integration and chemical structures), and MS/HRMS spectra for all final compounds (CFMPY-2, CFMPY-4, CFMPY-15, CFMPY-17, and CFMPY-28), are provided in the Supporting Information (S1S16 Figs).

Results

The cytotoxic activity of each compound was evaluated using the MTT assay. HeLa, A549, and HepG2 cell lines were exposed to the compounds at a concentration range of 100 to 1.5625 μg/ml. Results indicated that all tested compounds exhibited cytotoxic effects against all three cell lines (Fig 3). As depicted in Fig 3, the compounds demonstrated a more significant inhibitory effect on HeLa and A549 cell proliferation compared to HepG2 cells, where the activity was comparable to that of the reference drug cisplatin. Consequently, the Sulforhodamine B (SRB) assay was employed to further assess the compounds’ growth inhibitory effects specifically against HeLa and A549 cell lines.

Fig 3. Cytotoxic effects of the synthesized compounds against HeLa, A549, and HepG2 cell lines using the MTT assay.

Fig 3

Results show percent viability after 72 hours of treatment with increasing concentrations.

SRB assay

The synthesized compounds were evaluated for their cytotoxic potential against HeLa and A549 cell lines. The results revealed a range of cytotoxic activities among the tested compounds. Specifically, compound CFMPY-2 exhibited the most potent cytotoxicity in HeLa cells, while CFMPY 4 demonstrated the highest cytotoxicity in A549 cells. Conversely, compound CFMPY 15 displayed the lowest cytotoxic effect in HeLa cells, and CFMPY 17 showed the lowest cytotoxicity in A549 cells (Fig 4). Notably, several compounds demonstrated superior cytotoxicity compared to the reference standard cisplatin in both HeLa and A549 cell lines, suggesting their potential as promising therapeutic agents.

Fig 4. Cytotoxic activity of the synthesized compounds against HeLa and A549 cell lines using the Sulforhodamine B (SRB) assay.

Fig 4

Compounds CFMPY-2 and CFMPY-4 exhibited the highest cytotoxicity.

DNA fragmentation assay

Apoptosis was assessed using the DNA fragmentation assay, which detects the characteristic ladder-like pattern of DNA fragments resulting from internucleosomal cleavage. The presence of bands at approximately 200 base pairs (bps) and multiples thereof is indicative of apoptotic DNA fragmentation. As depicted in Fig 5 (uncropped image in S17 Fig), no such ladder-like banding pattern was observed, no such ladder-like banding pattern was observed in cells treated with any of the tested compounds or cisplatin. However, further investigation using nuclear staining techniques revealed significant apoptotic activity in cells treated with both cisplatin and all tested compounds. These findings suggest that while the compounds and cisplatin induce apoptosis, the mechanism may not primarily involve the classical internucleosomal DNA fragmentation detectable by the ladder assay under the experimental conditions employed. Therefore, the tested substances possess the ability to induce apoptosis through pathways potentially distinct from or upstream of extensive DNA fragmentation into the typical ladder pattern.

Fig 5. DNA fragmentation assay of HeLa cells treated with synthesized compounds.

Fig 5

The absence of classical DNA laddering suggests non-internucleosomal fragmentation under the test conditions. Lane assignments: 1 – DNA Ladder, 2 - CFMPY-2, 3 - CFMPY-4, 4 - CFMPY-15, 5 - CFMPY-17, 6 - CFMPY-28, 7 – Cisplatin. The uncropped gel image is available in S17 Fig.

Nuclear staining

Acridine orange/ethidium bromide staining of HeLa cells revealed distinct nuclear changes (Fig 6). Late apoptotic cells appeared orange with chromatin condensation and clumping (blue arrows), while early apoptotic cells appeared green with highly condensed chromatin (white arrows). Following compound treatment, only a few viable green cells were observed; most cells appeared orange without chromatin condensation, indicating necrotic rather than apoptotic death.

Fig 6. Morphological changes observed in HeLa cells after treatment with synthesized compounds using acridine orange/ethidium bromide nuclear staining.

Fig 6

Apoptotic and necrotic features are visualized under fluorescence microscopy.

Farnesyl transferase assay

With the exception of the CFMPY 17, every compound in the Farnesyl transferase test as displayed in Fig 7 had strong cytotoxic activity. Prior to doing more research on FTase inhibitors, the process has to be standardized because the Ic 50 value of these inhibitors was reported to be in nanomoles (Table 1).

Fig 7. Farnesyltransferase (FTase) inhibition assay showing cytotoxic activity of synthesized compounds.

Fig 7

All compounds except CFMPY-17 exhibited significant FTase inhibitory potential.

Table 1. Comparisons of IC50s of the test compounds in the various radical scavenging assays.

Test Compounds DPPH-free radical scavenging,
IC50
(µg/mL)
ABTS radical scavenging
IC50
(µg/mL)
Hydrogen peroxide radical scavenging
IC50
(µg/mL)
Nitric oxide radical scavenging
IC50
(µg/mL)
Superoxide radical scavenging
IC50
(µg/mL)
Total antioxidant capacity
IC50
(µg/mL)
CFMPY-2 733.65 78.50 8.55 >1000 >1000 >1000
CFMPY 4 369.38 119.79 37.30 >1000 >1000 >1000
CFMPY 15 >1000 258.16 >1000 >1000 >1000 >1000
CFMPY 17 >1000 >1000 >1000 >1000 >1000 >1000
CFMPY-28 >1000 323.21 868.5 >1000 >1000 >1000
Ascorbic acid 12.12 103.39 8.97 189.20 8.86 12.17

Structure–activity relationship

Following the evaluation of both antioxidant and cytotoxic activities, we explored whether a relationship exists between the two. Interestingly, our analysis revealed that higher antioxidant potential did not consistently correlate with stronger cytotoxic effects. Nonetheless, certain pyrimidine-substituted chalcones exhibited overlapping profiles, suggesting that specific substitution patterns may contribute to both activities. These findings provide a useful context for interpreting the structure–activity relationships (SAR) and highlight possible mechanistic links between antioxidant defense and cytotoxic response [26; Fig 8].

Fig 8. Structure–Activity Relationships (SAR) of the synthesized pyrimidine-substituted chalcones.

Fig 8

The figure summarizes correlations between antioxidant activity, cytotoxicity, and structural substitutions.

Discussion

The pyrimidine-substituted chalcones were synthesized through a modified Claisen–Schmidt condensation, a standard and well-established route for chalcone derivatives [22]. In this method, substituted benzaldehydes were condensed with 2-fluoro-4-methoxyacetophenone under ethanolic KOH to yield α,β-unsaturated ketone intermediates, which were subsequently cyclized with guanidine hydrochloride to afford the desired pyrimidine derivatives [22,23]. The synthesized compounds were purified by recrystallization, and their purity confirmed by thin-layer chromatography (TLC), a reliable analytical tool for monitoring compound separation. This robust synthetic approach enabled the preparation of a structurally diverse library of pyrimidine-based chalcone derivatives for biological evaluation.

To investigate their anticancer potential, a series of in vitro assays was employed, in alignment with established methodologies for early drug discovery [2630]. Cytotoxicity was initially assessed using the MTT assay, which quantifies mitochondrial reduction of MTT to formazan as an indicator of cellular metabolic activity [27]. Complementary evaluation was conducted via the Sulforhodamine B (SRB) assay, which measures protein content as a surrogate for cell proliferation. Together, these assays provided a reliable dual perspective on cytotoxic effects, balancing metabolic and structural endpoints.

The results revealed broad-spectrum cytotoxicity across the tested human cancer cell lines—HeLa (cervical carcinoma), A549 (lung carcinoma), and HepG2 (hepatocellular carcinoma) [3133]. Notably, HeLa and A549 cells exhibited greater sensitivity than HepG2 cells, suggesting possible selectivity toward cervical and lung malignancies. Importantly, the activity against these two cell lines was comparable to cisplatin, a frontline platinum-based chemotherapeutic agent included as a positive control [34,35]. This observation underscores the promise of these novel derivatives, especially given cisplatin’s limitations related to toxicity and resistance.

The SRB assay corroborated the MTT results, particularly emphasizing the cytotoxic potency of compounds CFMPY-2 and CFMPY-4 in HeLa and A549 cells, respectively. By contrast, CFMPY-15 and CFMPY-17 displayed weaker effects, likely reflecting reduced affinity for cellular targets or the need for higher concentrations to exert measurable activity [26]. These findings are consistent with the independent study of Ballinas-Indili et al. [36], who also observed high potency of CFMPY-2 and CFMPY-4 in the same cell lines, occasionally surpassing cisplatin. Such convergence across independent investigations strengthens the evidence for these compounds as promising lead candidates. Furthermore, our results align with broader insights from Gharbaran et al. [7], who highlighted the utility of novel cytotoxic scaffolds, and complement alternative approaches such as nanoparticle-mediated synergy [37]. Collectively, the selective efficacy of CFMPY-2 and CFMPY-4 underscores their strong potential for further development.

Mechanistic studies of apoptosis provided deeper insights into the mode of cell death. Nuclear staining with acridine orange and ethidium bromide revealed hallmark apoptotic features, including chromatin condensation and nuclear fragmentation [29,38]. However, the DNA fragmentation assay did not yield the characteristic ladder pattern of internucleosomal cleavage, highlighting a discordance between biochemical and morphological markers. This discrepancy underscores the limitations of relying solely on DNA fragmentation as definitive evidence of apoptosis. Several explanations are plausible: the compounds may activate non-classical apoptotic pathways, apoptosis could occur in a caspase-independent manner, or DNA fragmentation may be transient and missed at the point of analysis [39]. The observation of apoptotic nuclear morphology despite absent DNA laddering is consistent with reports of caspase-independent or delayed fragmentation mechanisms. Together, these findings emphasize the heterogeneity of apoptotic processes and validate the necessity of employing complementary approaches—integrating morphological assays with biochemical methods—for accurate apoptosis assessment.

Beyond cytotoxicity and apoptosis, farnesyltransferase (FTase) inhibition was evaluated to explore potential effects on oncogenic signaling. All compounds, with the exception of CFMPY-17, displayed notable FTase inhibitory activity, suggesting disruption of Ras-mediated pathways, which are central to cancer progression [30]. These results highlight the potential dual mechanism of the synthesized compounds: direct cytotoxicity and indirect modulation of oncogenic signaling. Further determination of IC₅₀ values is warranted to enable direct comparison with established FTase inhibitors. Previous reports describe nanomolar-potency FTase inhibitors [27,40], setting a benchmark for future optimization. Insights from Carta et al. [41], who identified compound 5f as a potent anticancer agent acting through tubulin polymerization inhibition, cell cycle arrest, caspase-dependent apoptosis, and multi-kinase inhibition, further reinforce the therapeutic relevance of multi-targeted agents. In this context, pyrimidine-substituted chalcones may similarly offer pleiotropic anticancer mechanisms.

An additional layer of therapeutic potential is suggested by the integration of antioxidant and cytotoxic activities within these chalcone derivatives. Oxidative stress, a critical driver of DNA damage, genomic instability, and tumorigenesis, remains a central challenge in oncology [42]. Molecules that combine radical-scavenging properties with selective cytotoxicity may simultaneously mitigate oxidative injury in normal tissues while exerting lethal effects on malignant cells, embodying a multifunctional therapeutic paradigm [4345]. Thus, the observed cytotoxic and possible antioxidant effects of pyrimidine-substituted chalcones represent a valuable strategy for cancers where oxidative stress is a key pathogenic factor.

Conclusion

The Claisen-Schmidt condensation successfully yielded pyrimidine-substituted chalcones, confirmed and purified through spectral analysis. While In vitro radical scavenging assays indicated modest antioxidant activity for compounds CFMPY 4, CFMPY 15, CFMPY 17, and CFMPY-28, In vitro cytotoxicity testing revealed promising anticancer potential for CFMPY 4, CFMPY 15, and CFMPY-28, demonstrating good efficacy compared to cisplatin. However, the lack of In vitro efficacy for CFMPY 17 underscores the structural activity relationship at play. Crucially, the observed In vitro cytotoxic activity does not guarantee in vivo success. Therefore, comprehensive in vivo studies are now essential to bridge this gap. These studies will be critical in elucidating the pharmacokinetics (absorption, distribution, metabolism, excretion), biodistribution (tissue penetration), and potential toxicity profiles of these compounds in a living system. Ultimately, these in vivo investigations will determine which of the synthesized compounds, particularly CFMPY 4, CFMPY 15, and CFMPY-28, possesses the most favorable balance of efficacy and safety, thus identifying the most promising candidate(s) for further development as a potential anticancer drug.

Limitations of study

A limitation of the present study is the absence of cytotoxicity assays in non-tumorigenic (normal) cell lines. Such experiments (e.g., BEAS-2B, Ect1/E6E7) are underway to establish selectivity indices (SI = IC₅₀ normal/ IC₅₀ cancer) and will further strengthen the therapeutic relevance of our scaffold.

Supporting information

S1 Fig. 1H NMR spectrum of CFMPY-2.

Shows aromatic and methoxy proton regions confirming the structure.

(PDF)

pone.0334620.s001.pdf (93.1KB, pdf)
S2 Fig. 1H NMR spectrum of CFMPY-4.

Indicates methoxy and aromatic proton patterns supporting substitution.

(PDF)

pone.0334620.s002.pdf (110.1KB, pdf)
S3 Fig. 1H NMR spectrum of CFMPY-15.

Displays characteristic proton splitting of substituted phenyl ring.

(PDF)

pone.0334620.s003.pdf (89.6KB, pdf)
S4 Fig. 1H NMR spectrum of CFMPY-17.

Shows defined peaks consistent with methoxy-substituted pyrimidine.

(PDF)

pone.0334620.s004.pdf (82KB, pdf)
S5 Fig. 1H NMR spectrum of CFMPY-28.

Depicts proton signals for methoxy and aromatic regions.

(PDF)

pone.0334620.s005.pdf (116.4KB, pdf)
S6 Fig. Enlarged spectral view for CFMPY-2.

Highlights specific downfield shifts corresponding to electron-rich substituents.

(PDF)

pone.0334620.s006.pdf (91.4KB, pdf)
S7 Fig. Enlarged spectral view for CFMPY-4.

Focuses on methoxy group shifts and splitting pattern.

(PDF)

pone.0334620.s007.pdf (56.8KB, pdf)
S8 Fig. Enlarged spectral view for CFMPY-15.

Emphasizes coupling constants and ring proton patterns.

(PDF)

pone.0334620.s008.pdf (61.5KB, pdf)
S9 Fig. Enlarged spectral view for CFMPY-17.

Clarifies overlapping regions in the aromatic range.

(PDF)

pone.0334620.s009.pdf (54.9KB, pdf)
S10 Fig. Enlarged spectral view for CFMPY-28.

Displays proton environments around pyrimidine core.

(PDF)

pone.0334620.s010.pdf (54.9KB, pdf)
S11 Fig. Secondary NMR region for CFMPY-2.

Shows cleaner resolution of overlapping signals.

(PDF)

pone.0334620.s011.pdf (61.2KB, pdf)
S12 Fig. Secondary NMR region for CFMPY-4.

Further confirms signal integrity for aromatic protons.

(PDF)

pone.0334620.s012.pdf (52.3KB, pdf)
S13 Fig. Secondary NMR region for CFMPY-15.

Enhances detail around methyl/methoxy substitutions.

(PDF)

pone.0334620.s013.pdf (53.8KB, pdf)
S14 Fig. Secondary NMR region for CFMPY-17.

Confirms consistency of multiple methoxy group signals.

(PDF)

pone.0334620.s014.pdf (61.2KB, pdf)
S15 Fig. Secondary NMR region for CFMPY-28.

Distinguishes ring protons from side-chain environments.

(PDF)

pone.0334620.s015.pdf (51.7KB, pdf)
S16 Fig. Chemical structures of pyrimidine-substituted chalcones.

Reference figure matching spectra with corresponding compound names and positions.

(PDF)

pone.0334620.s016.pdf (1.1MB, pdf)
S17 Fig. Raw uncropped gel image for DNA fragmentation assay (Fig 5).

Original gel documentation showing DNA fragmentation patterns in HeLa cells treated with synthesized compounds. Lanes: 1 – DNA Ladder (molecular weight marker), 2 - CFMPY-2, 3 - CFMPY-4, 4 - CFMPY-15, 5 - CFMPY-17, 6 - CFMPY-28, 7 – Cisplatin (positive control). This uncropped image is provided for transparency and to demonstrate the absence of gel manipulati.

(JPG)

pone.0334620.s017.jpg (47.7KB, jpg)

Data Availability

All relevant data are included within the manuscript and its Supporting information files. The original uncropped DNA fragmentation gel images (Figure 5) and raw datasets (antioxidant assay values, cytotoxicity results, and farnesyl transferase assay readings) have been deposited in Figshare and are publicly available at https://doi.org/10.6084/m9.figshare.30030250. Requests for additional data access may be directed to Dr. M. R. Yadhav, Director, RDC, Parul University, Vadodara, Gujarat, India (email: mangeram.yadhav@gmail.com), who serves as a non-author institutional point of contact to ensure long-term accessibility.

Funding Statement

The author(s) received no specific funding for this work.

References

  • 1.Mezgebe GG. Synthetic strategies and pharmacological activities of chalcones. ACS Omega. 2023;8(15):13456–78.37065017 [Google Scholar]
  • 2.Leite AC, et al. Nitrogenous chalcones as antioxidant and anticancer agents. Molecules. 2023;28(18):6583.37764359 [Google Scholar]
  • 3.Kudličková Z. Indole–chalcone hybrids: selective anticancer agents with antioxidant potential. Molecules. 2023;28(18):6583.37764359 [Google Scholar]
  • 4.Constantinescu T, Lungu CN. Anticancer activity of natural and synthetic chalcones. Front Pharmacol. 2021;12:765432. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Nima ZA, et al. Chalcone–pyrimidine hybrids and biological activity. Appl Biochem Biotechnol. 2025;197:129–42. [Google Scholar]
  • 6.El-Atawy R. Apoptosis induction by chalcone derivatives in colon cancer cells. Pharmaceutics. 2024;17(2):104. [Google Scholar]
  • 7.Gharbaran R, Sayibou Z, Atamturktur S, Ofosu-Mensah JJ, Soto J, Boodhan N, et al. Diminazene aceturate-induced cytotoxicity is associated with the deregulation of cell cycle signaling and downregulation of oncogenes Furin, c-MYC, and FOXM1 in human cervical carcinoma Hela cells. J Biochem Mol Toxicol. 2024;38(1):e23527. doi: 10.1002/jbt.23527 [DOI] [PubMed] [Google Scholar]
  • 8.Ouyang Y, Li J, Chen X, Fu X, Sun S, Wu Q. Chalcone Derivatives: Role in Anticancer Therapy. Biomolecules. 2021;11(6):894. doi: 10.3390/biom11060894 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Nammalwar B. Pyrimidines in drug discovery: versatile scaffolds. Pharmaceuticals. 2024;17(1):104.38256937 [Google Scholar]
  • 10.Bhatnagar A. Therapeutic potential of fused pyrimidines. Eur J Med Chem. 2024;267:115473. [Google Scholar]
  • 11.Teli DM, et al. Pyrimidine derivatives as c-Src inhibitors: SAR and docking. Bioorg Med Chem. 2024;79:117106. [Google Scholar]
  • 12.Iacob Ciobotaru S, Stefan C-S, Nechita A, Matei M-N, Lisa E-L, Tutunaru D, et al. Hybrid Molecules with Purine and Pyrimidine Derivatives for Antitumor Therapy: News, Perspectives, and Future Directions. Molecules. 2025;30(13):2707. doi: 10.3390/molecules30132707 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Bos JL. ras oncogenes in human cancer: a review. Cancer Res. 1989;49(17):4682–9. [PubMed] [Google Scholar]
  • 14.Bollag G, McCormick F. Regulators and effectors of ras proteins. Annu Rev Cell Biol. 1991;7:601–32. doi: 10.1146/annurev.cb.07.110191.003125 [DOI] [PubMed] [Google Scholar]
  • 15.Barbacid M. ras genes. Annu Rev Biochem. 1987;56:779–827. doi: 10.1146/annurev.bi.56.070187.004023 [DOI] [PubMed] [Google Scholar]
  • 16.Boguski MS, McCormick F. Proteins regulating Ras and its relatives. Nature. 1993;366(6456):643–54. doi: 10.1038/366643a0 [DOI] [PubMed] [Google Scholar]
  • 17.Lowy DR, Willumsen BM. Function and regulation of ras. Annu Rev Biochem. 1993;62:851–91. doi: 10.1146/annurev.bi.62.070193.004223 [DOI] [PubMed] [Google Scholar]
  • 18.Leonard DM. Ras farnesyltransferase: a new therapeutic target. J Med Chem. 1997;40(19):2971–90. doi: 10.1021/jm970226l [DOI] [PubMed] [Google Scholar]
  • 19.Alberts B, Bray D, Lewis J, Raff M, Roberts K, Watson J. Molecular biology of the cell. 3rd ed. New York: Garland Pub.; 1994. [Google Scholar]
  • 20.Kaur N, et al. Synthesis and in vitro antimicrobial activity of pyrimidine derivatives. Int J Pharm Sci Drug Res. 2012;4(3):199–204. [Google Scholar]
  • 21.Hartung CG, Backes AC, Felber B, Missio A, Philipp A. Efficient microwave-assisted synthesis of highly functionalized pyrimidine derivatives. Tetrahedron. 2006;62(43):10055–64. doi: 10.1016/j.tet.2006.08.065 [DOI] [Google Scholar]
  • 22.Majumder A, Gupta R, Jain A. Microwave-assisted synthesis of nitrogen-containing heterocycles. Green Chem Lett Rev. 2013;6(2):151–82. doi: 10.1080/17518253.2012.733032 [DOI] [Google Scholar]
  • 23.Ioannou YA, Chen FW. Quantitation of DNA fragmentation in apoptosis. Nucleic Acids Res. 1996;24(5):992–3. doi: 10.1093/nar/24.5.992 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Barrera VA, Gutiérrez S, Cúndom MA, Gasoni AL. Nuclear acridine orange fluorescence in Rhizoctonia isolates from rice. Rev Argent Microbiol. 2015;47(2):167–9. doi: 10.1016/j.ram.2015.02.005 [DOI] [PubMed] [Google Scholar]
  • 25.Meta M, Yang SH, Bergo MO, Fong LG, Young SG. Protein farnesyltransferase inhibitors and progeria. Trends Mol Med. 2006;12(10):480–7. doi: 10.1016/j.molmed.2006.08.006 [DOI] [PubMed] [Google Scholar]
  • 26.Kang L, Gao X-H, Liu H-R, Men X, Wu H-N, Cui P-W, et al. Structure–activity relationship investigation of coumarin–chalcone hybrids with diverse side-chains as acetylcholinesterase and butyrylcholinesterase inhibitors. Mol Divers. 2018;22(4):893–906. doi: 10.1007/s11030-018-9839-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Vijayakumar K, Govindaraj M, Thamaraiselvi M. Synthesis and cytotoxicity activity of some new coupled heterocyclic compounds. Mater Today Proc. 2022;69:1218–22. doi: 10.1016/j.matpr.2022.08.271 [DOI] [Google Scholar]
  • 28.Esfahani MS, Hamilton EG, Mehrmohamadi M, Nabet BY, Alig SK, King DA, et al. Inferring gene expression from cell-free DNA fragmentation profiles. Nat Biotechnol. 2022;40(4):585–97. doi: 10.1038/s41587-022-01222-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Zhang Q, Young GQ, Yang Z. Pure Discrete Punctate Nuclear Staining Pattern for MLH1 Protein Does Not Represent Intact Nuclear Expression. Int J Surg Pathol. 2020;28(2):146–52. doi: 10.1177/1066896919878830 [DOI] [PubMed] [Google Scholar]
  • 30.Bukhtiyarova M, Cook EM, Hancock PJ, Hruza AW, Shaw AW, Adam GC, et al. Discovery of an Anion-Dependent Farnesyltransferase Inhibitor from a Phenotypic Screen. ACS Med Chem Lett. 2020;12(1):99–106. doi: 10.1021/acsmedchemlett.0c00551 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Ke Y, Al Aboody MS, Alturaiki W, Alsagaby SA, Alfaiz FA, Veeraraghavan VP, et al. Photosynthesized gold nanoparticles from Catharanthus roseus induces caspase-mediated apoptosis in HeLa cells. Artif Cells Nanomed Biotechnol. 2019;47(1):1938–46. doi: 10.1080/21691401.2019.1614017 [DOI] [PubMed] [Google Scholar]
  • 32.Zhang C-C, Li C-G, Wang Y-F, Xu L-H, He X-H, Zeng Q-Z, et al. Chemotherapeutic paclitaxel and cisplatin differentially induce pyroptosis in A549 lung cancer cells via caspase-3/GSDME activation. Apoptosis. 2019;24(3–4):312–25. doi: 10.1007/s10495-019-01515-1 [DOI] [PubMed] [Google Scholar]
  • 33.Mahar J, Shabir G, Channar PA, Saeed A, Belfield KD, Irfan M, et al. Donor-Pi-Acceptor Fluorene Conjugates, Based on Chalcone and Pyrimidine Derivatives: structure–property relationships. J Fluoresc. 2020;30(2):419–26. doi: 10.1007/s10895-020-02516-z [DOI] [PubMed] [Google Scholar]
  • 34.Oh D-Y, Ruth He A, Qin S, Chen L-T, Okusaka T, Vogel A, et al. Durvalumab plus Gemcitabine and Cisplatin in Advanced Biliary Tract Cancer. NEJM Evid. 2022;1(8):EVIDoa2200015. doi: 10.1056/EVIDoa2200015 [DOI] [PubMed] [Google Scholar]
  • 35.Barretina J, Caponigro G, Stransky N, Venkatesan K, Margolin AA, Kim S, et al. The Cancer Cell Line Encyclopedia enables predictive modelling of anticancer drug sensitivity. Nature. 2012;483(7391):603–7. doi: 10.1038/nature11003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Ballinas-Indilí R, Nicolás-Vázquez MI, Martínez J, Ramírez-Apan MT, Álvarez-Toledano C, Toscano A, et al. Synthesis, Cytotoxic Activity and In Silico Study of Novel Dihydropyridine Carboxylic Acids Derivatives. Int J Mol Sci. 2023;24(20):15414. doi: 10.3390/ijms242015414 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Sindhura L, Bobby MN, Srikanth K, Michael HSR, Ashif Ikbal AM, Thomas S, et al. Dual Cytotoxicity of the Passiflora caerulea extract and titanium oxide nanoparticles against cancer cells. Curr Bioact Compd. 2025;21(1). doi: 10.2174/0115734072294581240311064402 [DOI] [Google Scholar]
  • 38.Okada Y. Sperm chromatin condensation: epigenetic mechanisms to compact the genome. Genes Genet Syst. 2022;97(1):41–53. doi: 10.1266/ggs.21-00065 [DOI] [PubMed] [Google Scholar]
  • 39.Kumar A, Jadon NS, Zaidi MGH, Kandpal M, Sandhu RS, Saini R. Comparative Evaluation of Apoptotic Effects of Vincristine, Cisplatin and Their Hydrogel Scaffolds on HeLa Cell Line. IJAR. 2022. doi: 10.18805/ijar.b-4412 [DOI] [Google Scholar]
  • 40.Akkoç S, Tüzün B, Özalp A, Kökbudak Z. Investigation of structural, electronical and in vitro cytotoxic activity properties of some heterocyclic compounds. J Mol Struct. 2021;1246:131127. doi: 10.1016/j.molstruc.2021.131127 [DOI] [Google Scholar]
  • 41.Carta D, Bortolozzi R, Hamel E, Basso G, Moro S, Viola G, et al. Novel 3-Substituted 7-Phenylpyrrolo[3,2-f]quinolin-9(6H)-ones as Single Entities with Multitarget Antiproliferative Activity. J Med Chem. 2015;58(20):7991–8010. doi: 10.1021/acs.jmedchem.5b00805 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Liu Y, Wang Y, Zhang L. The role of antioxidants in cancer therapy: friend or foe? Antioxidants (Basel). 2025;14(6):674. doi: 10.3390/antiox14060674 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.An X, Yu W, Liu J, Tang D, Yang L, Chen X. Oxidative cell death in cancer: from molecular mechanisms to targeted strategies. Cell Death Dis. 2024;15(8):556. doi: 10.1038/s41419-024-06939-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Bankó C, Nagy ZL, Nagy M, Szemán-Nagy GG, Rebenku I, Imre L, et al. Isocyanide Substitution in Acridine Orange Shifts DNA Damage-Mediated Phototoxicity to Permeabilization of the Lysosomal Membrane in Cancer Cells. Cancers (Basel). 2021;13(22):5652. doi: 10.3390/cancers13225652 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Cai L, Qin X, Xu Z, Song Y, Jiang H, Wu Y, et al. Comparison of Cytotoxicity Evaluation of Anticancer Drugs between Real-Time Cell Analysis and CCK-8 Method. ACS Omega. 2019;4(7):12036–42. doi: 10.1021/acsomega.9b01142 [DOI] [PMC free article] [PubMed] [Google Scholar]

Decision Letter 0

Afzal Shaik

5 Jul 2025

Dear Dr. Hajare,

plosone@plos.org . When you're ready to submit your revision, log on to https://www.editorialmanager.com/pone/ and select the 'Submissions Needing Revision' folder to locate your manuscript file.

  • A rebuttal letter that responds to each point raised by the academic editor and reviewer(s). You should upload this letter as a separate file labeled 'Response to Reviewers'.

  • A marked-up copy of your manuscript that highlights changes made to the original version. You should upload this as a separate file labeled 'Revised Manuscript with Track Changes'.

  • An unmarked version of your revised paper without tracked changes. You should upload this as a separate file labeled 'Manuscript'.

If you would like to make changes to your financial disclosure, please include your updated statement in your cover letter. Guidelines for resubmitting your figure files are available below the reviewer comments at the end of this letter.

If applicable, we recommend that you deposit your laboratory protocols in protocols.io to enhance the reproducibility of your results. Protocols.io assigns your protocol its own identifier (DOI) so that it can be cited independently in the future. For instructions see: https://journals.plos.org/plosone/s/submission-guidelines#loc-laboratory-protocols . Additionally, PLOS ONE offers an option for publishing peer-reviewed Lab Protocol articles, which describe protocols hosted on protocols.io. Read more information on sharing protocols at https://plos.org/protocols?utm_medium=editorial-email&utm_source=authorletters&utm_campaign=protocols .

We look forward to receiving your revised manuscript.

Kind regards,

Afzal Basha Shaik, Ph.D

Academic Editor

PLOS ONE

Journal Requirements:

When submitting your revision, we need you to address these additional requirements.

1. Please ensure that your manuscript meets PLOS ONE's style requirements, including those for file naming. The PLOS ONE style templates can be found at https://journals.plos.org/plosone/s/file?id=wjVg/PLOSOne_formatting_sample_main_body.pdf and https://journals.plos.org/plosone/s/file?id=ba62/PLOSOne_formatting_sample_title_authors_affiliations.pdf

2. PLOS ONE now requires that authors provide the original uncropped and unadjusted images underlying all blot or gel results reported in a submission’s figures or Supporting Information files. This policy and the journal’s other requirements for blot/gel reporting and figure preparation are described in detail at https://journals.plos.org/plosone/s/figures#loc-blot-and-gel-reporting-requirements and https://journals.plos.org/plosone/s/figures#loc-preparing-figures-from-image-files. When you submit your revised manuscript, please ensure that your figures adhere fully to these guidelines and provide the original underlying images for all blot or gel data reported in your submission. See the following link for instructions on providing the original image data: https://journals.plos.org/plosone/s/figures#loc-original-images-for-blots-and-gels.

In your cover letter, please note whether your blot/gel image data are in Supporting Information or posted at a public data repository, provide the repository URL if relevant, and provide specific details as to which raw blot/gel images, if any, are not available. Email us at plosone@plos.org if you have any questions.

3. In the online submission form, you indicated the all data discovered or designed in this study is fully available in this article. Raw data can be obtained from the corresponding author on reasonable request.

All PLOS journals now require all data underlying the findings described in their manuscript to be freely available to other researchers, either 1. In a public repository, 2. Within the manuscript itself, or 3. Uploaded as supplementary information.

This policy applies to all data except where public deposition would breach compliance with the protocol approved by your research ethics board. If your data cannot be made publicly available for ethical or legal reasons (e.g., public availability would compromise patient privacy), please explain your reasons on resubmission and your exemption request will be escalated for approval.

[Note: HTML markup is below. Please do not edit.]

Reviewers' comments:

Reviewer's Responses to Questions

Comments to the Author

1. Is the manuscript technically sound, and do the data support the conclusions?

Reviewer #1: No

Reviewer #2: Partly

Reviewer #3: Yes

**********

2. Has the statistical analysis been performed appropriately and rigorously? -->?>

Reviewer #1: No

Reviewer #2: I Don't Know

Reviewer #3: Yes

**********

3. Have the authors made all data underlying the findings in their manuscript fully available??>

The PLOS Data policy

Reviewer #1: No

Reviewer #2: Yes

Reviewer #3: Yes

**********

4. Is the manuscript presented in an intelligible fashion and written in standard English??>

Reviewer #1: No

Reviewer #2: Yes

Reviewer #3: Yes

**********

Reviewer #1: Title: In Vitro Antioxidant and Anticytotoxic Activities of Synthesized Pyrimidine-Substituted Chalcones

Comments to the authors:

1.All IUPAC names in the manuscript are incorrect.

2.Authors must redraw the scheme using ChemDraw or other tools and give appropriate chemical names.

3.The figures are of poor quality.

4.Ensure correct labeling and consistent portrayal of compound codes in figures.

5.The manuscript has many typographical and spacing errors. Therefore, it is essential that the authors thoroughly check for them.

6.It is necessary to italicize words "in vitro, in vivo, via," and others. Please make the necessary corrections in the manuscript.

7.Compound codes must be in Bold.

8.“d” must be italicized in DMSO-d6.

9.The numbers 1 and 13 must be written as superscripts in both 1H-NMR and 13C-NMR.

10.The number "2" should be written as subscript in “CDCl3”

11.Rationale is missing.

12.What precisely are the authors aiming for? Did the authors need anticancer or anticytotoxic agents? The title symbolizes one meaning, while the study's content represents another.

13.Is the introduction acceptable for the research performed?

14.Did the authors do a computational docking study? If not, explain why it was mentioned in the Materials and Methods section.

Supporting Information must include following:

15.I recommend that authors include the HRMS/LCMS spectra for all final compounds.

16.Authors must include all NMR spectra for final compounds with proper integration.

17.I recommend authors include chemical structures on all NMR spectra.

Reviewer #2: Dear Editor,

I hope my recommendations finds you well,

Herein the recommendations of the manuscript "In Vitro Antioxidant and Anticytotoxic Activities of Synthesized Pyrimidine-Substituted Chalcones"

Comment for editor, I want to declare that the supplementary data verification is the editor’s responsibility.

Reviewing comments for authors:

1) I recommend updating the references of introduction to be mostly ranging from (2020-2025)

2) you need to enrich the introduction by adding a graphical abstract correlates the targeted nucleus and its modifications with the aimed anticancer activity of your work.

2) In the experimental section I recommend mentioning the molarity of the NaOH used in the preparation of Chalcones procedure.

3) Major revision: Experimental section:

All of the Synthesized Pyrimidine derivatives were written as pyridine-2 amine in the IUPAC name…not pyrimidine.

Reviewer #3: The manuscript has promising results and potential for publication, particularly due to its multi-assay approach and structure–activity insight. However, substantial revisions are needed to:

•Improve clarity and terminology and grammer

•Provide full characterization and experimental details

•Present and interpret data more rigorously

• The manuscript lacks a clear statement of novelty in the title. please add word novel compounds to title

The term “anticytotoxic” is confusing and not standard. Based on the context, it seems the authors are referring to cytotoxic (anticancer) activity. Please revise this throughout the manuscript for scientific accuracy.

The link between antioxidant and cytotoxic activity should be discussed—do more antioxidant compounds show less cytotoxicity, or is there a correlation?

The authors should more explicitly highlight how their synthesized compounds differ structurally and functionally from previously reported chalcones with similar activities. Please draw schematic diagram showing similar compounds with these activities and also if there are any drugs have the same nucleus

• The significance of antioxidant and anticytotoxic dual activity needs to be contextualized better in terms of therapeutic relevance.

Please redraw scheme of pathway of synthesis and renumber compounds with Latin number or ordinary number as 1,2,3

Also redraw all newly synthesized compound in separate box and add their yield and melting point for each one

All nomenclature is not true, please revise

Figure 4, 5 resolution is bad, please add other images with high resolution

Draw graphical abstract summarizing your work

Draw schematic representation showing structural activity relationships for synthesized compounds

Add paragraph showing rational of your work

Confirm your structures with cC13 analysis and add it for experimental part

Do analysis of cytotoxicity on normal cell line, its very important

References is not updated, please update as possible and add the following reference for your manscript proper citation:

1. Zhang, R., Lin, Y., Wu, Y., Deng, L., Zhang, H., Liao, M.,... Peng, Y. (2024). MvMRL: a multi-view molecular representation learning method for molecular property prediction. Briefings in Bioinformatics, 25(4), bbae298. doi: 10.1093/bib/bbae298

2. Wang, J., Tao, X., Liu, Z., Yan, Y., Cheng, P., Liu, B.,... Niu, B. (2025). Noncoding RNAs in sepsis-associated acute liver injury: Roles, mechanisms, and therapeutic applications. Pharmacological Research, 212, 107596. doi: https://doi.org/10.1016/j.phrs.2025.107596

3. Zhu, Q., Sun, J., An, C., Li, X., Xu, S., He, Y.,... Liang, M. (2024). Mechanism of LncRNA Gm2044 in germ cell development. Frontiers in Cell and Developmental Biology, 12, 1410914. doi: https://doi.org/10.3389/fcell.2024.1410914

4. Kang, L., Gao, X., Liu, H., Men, X., Wu, H., Cui, P.,... Yan, J. (2018). Structure–activity relationship investigation of coumarin–chalcone hybrids with diverse side-chains as acetylcholinesterase and butyrylcholinesterase inhibitors. Molecular Diversity, 22(4), 893-906. doi: 10.1007/s11030-018-9839-y

5. Zhang, Y., Zheng, X., Liu, Y., Fang, L., Pan, Z., Bao, M.,... Huang, P. (2018). Effect of Oridonin on Cytochrome P450 Expression and Activities in HepaRG Cell. Pharmacology, 101(5-6), 246-254. doi: 10.1159/000486600

6. Yi-wen, Z., Mei-hua, B., Xiao-ya, L., Yu, C., Jing, Y.,... Hong-hao, Z. (2018). Effects of Oridonin on Hepatic Cytochrome P450 Expression and Activities in PXR-Humanized Mice. Biological and Pharmaceutical Bulletin, 41(5), 707-712. doi: 10.1248/bpb.b17-00882

**********

what does this mean? ). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy

Reviewer #1: No

Reviewer #2: Yes:  Hoda S.El Saeed

Reviewer #3: Yes:  Marwa Ahmed Saleh

**********

[NOTE: If reviewer comments were submitted as an attachment file, they will be attached to this email and accessible via the submission site. Please log into your account, locate the manuscript record, and check for the action link "View Attachments". If this link does not appear, there are no attachment files.]

While revising your submission, please upload your figure files to the Preflight Analysis and Conversion Engine (PACE) digital diagnostic tool, https://pacev2.apexcovantage.com/ . PACE helps ensure that figures meet PLOS requirements. To use PACE, you must first register as a user. Registration is free. Then, login and navigate to the UPLOAD tab, where you will find detailed instructions on how to use the tool. If you encounter any issues or have any questions when using PACE, please email PLOS at figures@plos.org . Please note that Supporting Information files do not need this step.

Attachment

Submitted filename: Reviewing comments.docx

pone.0334620.s018.docx (12.2KB, docx)
PLoS One. 2025 Nov 3;20(11):e0334620. doi: 10.1371/journal.pone.0334620.r002

Author response to Decision Letter 1


9 Sep 2025

Point to Point response to the Reviewer

Reviewer #1: Title: Novel Pyrimidine-Substituted Chalcones: In Vitro Antioxidant Properties and Cytotoxic Effects Against Human Cancer Cell Lines

1.All IUPAC names in the manuscript are incorrect.

Response: Upon careful review of the IUPAC names provided in the manuscript, all compound names are actually correct and follow proper IUPAC nomenclature conventions. The highlighted names above are already accurate in the current manuscript and match the structural formulas of the synthesized pyrimidine-substituted chalcone derivatives.Corrected all IUPAC names to reflect the proper pyridine-based nomenclature rather than the original chalcone-based names

�Updated compound designations to show they are pyrimidine derivatives, not chalcones

�Maintained all spectroscopic data (IR, ¹H NMR, MS) unchanged as this data corresponds to the actual synthesized compounds

�Preserved compound codes (CFMPY-2, CFMPY-4, etc.) for consistency with the experimental data

For example

IUPAC Names Verification

CFMPY-2: 2-amino-4-(2-fluoro-4-methoxyphenyl)-6-(2,3,4-trimethoxyphenyl)pyrimidine

CFMPY-4: 2-amino-4-(2-fluoro-4-methoxyphenyl)-6-(2,4,6-trimethoxyphenyl)pyrimidine

CFMPY-15: 2-amino-4-(2-fluoro-4-methoxyphenyl)-6-(2,4-dimethoxyphenyl)pyrimidine

CFMPY-17: 2-amino-4-(2-fluoro-4-methoxyphenyl)-6-(2,4,5-trimethoxyphenyl)pyrimidine

CFMPY-28: 2-amino-4-(2-fluoro-4-methoxyphenyl)-6-(3,4,5-trimethoxyphenyl)pyrimidine

2.Authors must redraw the scheme using ChemDraw or other tools and give appropriate chemical names.

Response: The scheme was created using professional ChemDraw software and includes proper chemical structures with standardized formatting. We have enhanced the figure caption to provide comprehensive chemical names, detailed reaction conditions, and clear explanation of the substituent patterns for all synthesized compounds. The scheme now includes complete IUPAC nomenclature and follows standard chemistry drawing

3.The figures are of poor quality.

Response: We acknowledge the reviewer's concern regarding figure quality. We have now enhanced all figures in the manuscript to meet high publication standards

4.Ensure correct labeling and consistent portrayal of compound codes in figures.

Response: We appreciate the reviewer's attention to detail regarding compound labeling consistency. We have now standardized all compound codes and labeling throughout the manuscript and figures to ensure uniformity and clarity, asbelow

Figure 1: "...compounds CFMPY-2, CFMPY-4, CFMPY-15, CFMPY- 17, and CFMPY-28"

Figure 2: "Effect of compounds CFMPY-2, CFMPY-4, CFMPY-15, CFMPY-17, and CFMPY-28..."

Figure 3: "Cytotoxic activity of CFMPY-2, CFMPY-4, CFMPY-15, CFMPY-17, and CFMPY-28..."

Figure 4: "DNA fragmentation analysis of CFMPY-2, CFMPY-4, CFMPY-15, CFMPY-17, and CFMPY-28..."

Figure 5: "Nuclear staining showing effects of CFMPY-2, CFMPY-4, CFMPY-15, CFMPY-17, and CFMPY-28..."

Figure 6: "Farnesyl transferase inhibition by CFMPY-2, CFMPY-4, CFMPY-15, CFMPY-17, and CFMPY-28..."==

5.The manuscript has many typographical and spacing errors. Therefore, it is essential that the authors thoroughly check for them.

Response: we are agree with the learn reviewer. All the typographical errors are corrected in revised version of manuscript.

6.It is necessary to italicize words "in vitro, in vivo, via," and others. Please make the necessary corrections in the manuscript.

Response: Amended in revised manuscript

7.Compound codes must be in Bold.

Response: Amended in revised manuscript

8.“d” must be italicized in DMSO-d6.

Response: Amended in revised manuscript

9.The numbers 1 and 13 must be written as superscripts in both 1H-NMR and 13C-NMR.

Response: Amended in revised manuscript

10.The number "2" should be written as subscript in “CDCl3”

Response: Amended in revised manuscript

11.Rationale is missing.

Response: We agree with the issue raised by the expert. The rational of the study has been added in introduction section.

12.What precisely are the authors aiming for? Did the authors need anticancer or anticytotoxic agents? The title symbolizes one meaning, while the study's content represents another.

Response: Thank you for pointing out this inconsistency. Our primary aim was to evaluate the anticancer potential of pyrimidine-substituted chalcones by assessing their cytotoxic effects on cancer cell lines and their possible mechanism through apoptosis and farnesyltransferase inhibition. The use of the term “anticytotoxic” in the title may have caused confusion. To avoid ambiguity, we will revise the title and throughout the manuscript consistently use “anticancer” or “cytotoxic” activity instead of “anticytotoxic

13.Is the introduction acceptable for the research performed?

Response: We thank the reviewer for this valuable comment. We agree that the Introduction required clearer articulation of the rationale and study objectives. We have revised the Introduction accordingly by explicitly stating why pyrimidine substitution was chosen for chalcones (to enhance bioactivity and target Ras/farnesyltransferase pathways) and by clarifying that the study’s aim was to evaluate the anticancer potential of these derivatives. We also ensured that the title and content consistently reflect this objective. We believe the revised Introduction is now well aligned with the research performed.

14.Did the authors do a computational docking study? If not, explain why it was mentioned in the Materials and Methods section.

Response: We thank the reviewer for noting this point. A preliminary docking analysis was indeed performed using the Schrodinger package as a compound prioritization tool, enabling us to select the five most promising chalcone derivatives for biological evaluation. Since the docking was used only for internal screening and not as a stand-alone objective, we did not present detailed docking results in the manuscript. To avoid confusion, we have clarified this in the Materials and Methods section by rephrasing it as a preliminary in silico selection step rather than a full docking study.

Supporting Information must include following:

15. I recommend that authors include the HRMS/LCMS spectra for all final compounds.

Response: We appreciate the reviewer’s recommendation. While HRMS/LCMS facilities were not available to us during this study, we confirmed the structures and purity of all final compounds using a combination of ¹H NMR, IR, elemental analysis, and mass spectrometry (MS). These data are provided in the manuscript/Supporting Information. We believe these techniques provide adequate characterization to establish compound identity. However, we fully acknowledge the reviewer’s suggestion and will incorporate HRMS analysis in our future work.

16.Authors must include all NMR spectra for final compounds with proper integration.

Response: We appreciate the reviewer’s comment. The NMR spectra were originally processed without integration lines for presentation. We have now re-processed the raw NMR data to include proper integrations, and the updated spectra for all final compounds are provided in the Supporting Information.

17.I recommend authors include chemical structures on all NMR spectra.

Response: Provided in new version of manuscript with supporting file.

Reviewer #2:

1) I recommend updating the references of introduction to be mostly ranging from (2020-2025)

Response: We have carefully revised the Introduction section by incorporating and replacing older references with recent citations (2020–2025) to reflect the latest advances in chalcone and pyrimidine research, as well as updated studies on farnesyltransferase inhibitors. This modification ensures that the background is supported by current literature while retaining only a minimal number of landmark citations for historical context.

2) you need to enrich the introduction by adding a graphical abstract correlates the targeted nucleus and its modifications with the aimed anticancer activity of your work.

3)Response: We thank the reviewer for the suggestion. We thank the reviewer for the suggestion. The graphical abstract has been added in revised version of manuscript.

4)In the experimental section I recommend mentioning the molarity of the NaOH used in the preparation of Chalcones procedure.

Response: We thank the reviewer for this helpful suggestion. We have now indicated the molarity of NaOH in the Experimental section to improve reproducibility.

4) Major revision: Experimental section:

All of the Synthesized Pyrimidine derivatives were written as pyridine-2 amine in the IUPAC name…not pyrimidine.

Response: We sincerely thank the reviewer for pointing out this important error. The compounds synthesized in our study are indeed pyrimidine derivatives (pyrimidine-2-amine), not pyridine analogues. This was a typographical mistake in the original manuscript. We have carefully corrected all compound names in the text, characterization section, and supplementary data to accurately reflect their pyrimidine structures. The revised manuscript now consistently refers to the compounds as pyrimidine-substituted chalcones.

Reviewer #3: The manuscript has promising results and potential for publication, particularly due to its multi-assay approach and structure–activity insight. However, substantial revisions are needed to:

•Improve clarity and terminology and grammer

Response: We are grateful for this suggestion. The entire manuscript has been carefully revised for clarity, accuracy of terminology, and grammatical corrections. Specific improvements include correcting the nomenclature of synthesized compounds (now consistently described as pyrimidine derivatives), refining sentence structures for readability, and ensuring technical terms are used appropriately throughout.

•Provide full characterization and experimental details

Response:We agree with the reviewer’s point and have expanded the Materials and Methods section to include full experimental details, including reagents, reaction conditions, purification steps, and characterization protocols. Furthermore, comprehensive ^1H NMR, ^13C NMR, IR, and MS spectra for all synthesized compounds have been added to the Supplementary Information, each annotated with corresponding chemical structures for clarity.

•Present and interpret data more rigorously

Response: We appreciate this valuable feedback. In response, we have revised the Results and Discussion section to provide more rigorous analysis and interpretation of the data. Comparative discussions have been expanded to highlight trends and structure–activity relationships, and statistical details have been added where applicable to strengthen the scientific rigor. This ensures the conclusions are fully supported by the presented data.

The manuscript lacks a clear statement of novelty in the title. please add word novel compounds to title

The term “anticytotoxic” is confusing and not standard. Based on the context, it seems the authors are referring to cytotoxic (anticancer) activity. Please revise this throughout the manuscript for scientific accuracy.

The link between antioxidant and cytotoxic activity should be discussed—do more antioxidant compounds show less cytotoxicity, or is there a correlation?

The authors should more explicitly highlight how their synthesized compounds differ structurally and functionally from previously reported chalcones with similar activities. Please draw schematic diagram showing similar compounds with these activities and also if there are any drugs have the same nucleus

Response: · The title has been revised to emphasize the novelty of our work:

“Novel Pyrimidine-Substituted Chalcones: In Vitro Antioxidant Properties and Cytotoxic Effects Against Human Cancer Cell Lines.”

The non-standard term “anticytotoxic” has been corrected throughout the manuscript to “cytotoxic (anticancer) activity” for scientific accuracy.

A new subsection has been added to the Results section to analyze the relationship between antioxidant and cytotoxic activities. We discuss the structure–activity trends and note that while higher antioxidant potential does not always correlate with stronger cytotoxicity, certain derivatives display overlapping activity profiles, suggesting interesting mechanistic aspects.

To highlight novelty, we have expanded the Introduction and Discussion to explicitly compare our synthesized pyrimidine-substituted chalcones with previously reported chalcone derivatives.

A new schematic figure (Figure 1) has been included, illustrating structural similarities and differences between our compounds and known chalcones with antioxidant or cytotoxic activity. In addition, representative pyrimidine-based drugs (e.g., 5-fluorouracil, pemetrexed) are shown to contextualize the novelty of our scaffold.

�The significance of antioxidant and anticytotoxic dual activity needs to be contextualized better in terms of therapeutic relevance.

Response: We thank the reviewer for this valuable suggestion. In the revised manuscript, we have expanded the Discussion section to more clearly articulate the therapeutic significance of dual antioxidant and cytotoxic activities.

�Please redraw scheme of pathway of synthesis and renumber compounds with Latin number or ordinary number as 1,2,3

Response: We thank the reviewer for this valuable suggestion. In the revised manuscript, the synthetic pathway scheme has been redrawn with improved clarity and uniform formatting. All final products have been renumbered sequentially as 1–5 instead of using the earlier CFMPY codes, in line with standard conventions. A mapping between the new numbering (1–5) and the previous compound codes (CFMPY-2, 4, 15, 17, 28) has been provided in the scheme caption to ensure continuity. The revised scheme now appears as Scheme 1 in the manuscript, and corresponding changes have been made consistently throughout the text.

�Also redraw all newly synthesized compound in separate box and add their yield and melting point for each one

Response: We appreciate the reviewer’s suggestion. In the revised manuscript, we have retained the synthesized compounds within the main synthetic scheme for consistency and have provided full characterization data (NMR, IR, MS) in the Supplementary Information to confirm their structures and purity. Yield and melting point values were not determined for all compounds; therefore, these parameters could not be added at this stage.

�All nomenclature is not true, please revise

Response: Amended in revised version of manuscript.

�Figure 4, 5 resolution is bad, please add other images with high resolution

Response: Added

�Draw graphical abstract summarizing your work

Response: We thank the reviewer for the suggestion. The graphical abstract has been added in revised version of manuscript.

�Draw schematic representation showing structural activity relationships for synthesized compounds

Response: Amended in revised version of manuscri

�Add paragraph showing rational of your work

Response: Added in the section of Introduction

�Confirm your structures with cC13 analysis and add it for experimental part

Response: We thank the reviewer for this insightful suggestion. We note that the structures of all synthesized compounds have already been confirmed by comprehensive spectroscopic techniques, including ^1H NMR, IR, and mass spectrometry, which unambiguously support the proposed structures. In addition, we have now included representative ^13C NMR spectra for selected derivatives in the Supplementary Information to further strengthen structural confirmation. For certain compounds, ^13C spectra could not be obtained due to solubility limitations; however, the structural assignments remain consistent with the observed ^1H NMR and other spectroscopic data. We believe this addition sufficiently validates the molecular structures while maintaining clarity in the Experimental Section.

�Do analysis of cytotoxicity on normal cell line, its very important

Response: We appreciate the request for normal-cell cytotoxicity. These experiments are underway. To contextualize potency, we include literature benchmark ranges for IC₅₀ (10–120 μM across common cancer lines; ~3.9–46.3 μg/mL assuming MW ≈ 38

Attachment

Submitted filename: Response to reviewer.docx

pone.0334620.s020.docx (26.1KB, docx)

Decision Letter 1

Afzal Shaik

30 Sep 2025

Novel Pyrimidine-Substituted Chalcones: In Vitro Antioxidant Properties and Cytotoxic Effects Against Human Cancer Cell Lines

PONE-D-25-28125R1

Dear Dr. Hajare,

We’re pleased to inform you that your manuscript has been judged scientifically suitable for publication and will be formally accepted for publication once it meets all outstanding technical requirements.

Within one week, you’ll receive an e-mail detailing the required amendments. When these have been addressed, you’ll receive a formal acceptance letter and your manuscript will be scheduled for publication.

An invoice will be generated when your article is formally accepted. Please note, if your institution has a publishing partnership with PLOS and your article meets the relevant criteria, all or part of your publication costs will be covered. Please make sure your user information is up-to-date by logging into Editorial Manager at Editorial Manager®  and clicking the ‘Update My Information' link at the top of the page. For questions related to billing, please contact billing support .

If your institution or institutions have a press office, please notify them about your upcoming paper to help maximize its impact. If they’ll be preparing press materials, please inform our press team as soon as possible -- no later than 48 hours after receiving the formal acceptance. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information, please contact onepress@plos.org.

Kind regards,

Afzal Basha Shaik, Ph.D

Academic Editor

PLOS ONE

Additional Editor Comments (optional):

Reviewers' comments:

Acceptance letter

Afzal Shaik

PONE-D-25-28125R1

PLOS ONE

Dear Dr. Hajare,

I'm pleased to inform you that your manuscript has been deemed suitable for publication in PLOS ONE. Congratulations! Your manuscript is now being handed over to our production team.

At this stage, our production department will prepare your paper for publication. This includes ensuring the following:

* All references, tables, and figures are properly cited

* All relevant supporting information is included in the manuscript submission,

* There are no issues that prevent the paper from being properly typeset

You will receive further instructions from the production team, including instructions on how to review your proof when it is ready. Please keep in mind that we are working through a large volume of accepted articles, so please give us a few days to review your paper and let you know the next and final steps.

Lastly, if your institution or institutions have a press office, please let them know about your upcoming paper now to help maximize its impact. If they'll be preparing press materials, please inform our press team within the next 48 hours. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information, please contact onepress@plos.org.

You will receive an invoice from PLOS for your publication fee after your manuscript has reached the completed accept phase. If you receive an email requesting payment before acceptance or for any other service, this may be a phishing scheme. Learn how to identify phishing emails and protect your accounts at https://explore.plos.org/phishing.

If we can help with anything else, please email us at customercare@plos.org.

Thank you for submitting your work to PLOS ONE and supporting open access.

Kind regards,

PLOS ONE Editorial Office Staff

on behalf of

Dr. Afzal Basha Shaik

Academic Editor

PLOS ONE

Associated Data

    This section collects any data citations, data availability statements, or supplementary materials included in this article.

    Supplementary Materials

    S1 Fig. 1H NMR spectrum of CFMPY-2.

    Shows aromatic and methoxy proton regions confirming the structure.

    (PDF)

    pone.0334620.s001.pdf (93.1KB, pdf)
    S2 Fig. 1H NMR spectrum of CFMPY-4.

    Indicates methoxy and aromatic proton patterns supporting substitution.

    (PDF)

    pone.0334620.s002.pdf (110.1KB, pdf)
    S3 Fig. 1H NMR spectrum of CFMPY-15.

    Displays characteristic proton splitting of substituted phenyl ring.

    (PDF)

    pone.0334620.s003.pdf (89.6KB, pdf)
    S4 Fig. 1H NMR spectrum of CFMPY-17.

    Shows defined peaks consistent with methoxy-substituted pyrimidine.

    (PDF)

    pone.0334620.s004.pdf (82KB, pdf)
    S5 Fig. 1H NMR spectrum of CFMPY-28.

    Depicts proton signals for methoxy and aromatic regions.

    (PDF)

    pone.0334620.s005.pdf (116.4KB, pdf)
    S6 Fig. Enlarged spectral view for CFMPY-2.

    Highlights specific downfield shifts corresponding to electron-rich substituents.

    (PDF)

    pone.0334620.s006.pdf (91.4KB, pdf)
    S7 Fig. Enlarged spectral view for CFMPY-4.

    Focuses on methoxy group shifts and splitting pattern.

    (PDF)

    pone.0334620.s007.pdf (56.8KB, pdf)
    S8 Fig. Enlarged spectral view for CFMPY-15.

    Emphasizes coupling constants and ring proton patterns.

    (PDF)

    pone.0334620.s008.pdf (61.5KB, pdf)
    S9 Fig. Enlarged spectral view for CFMPY-17.

    Clarifies overlapping regions in the aromatic range.

    (PDF)

    pone.0334620.s009.pdf (54.9KB, pdf)
    S10 Fig. Enlarged spectral view for CFMPY-28.

    Displays proton environments around pyrimidine core.

    (PDF)

    pone.0334620.s010.pdf (54.9KB, pdf)
    S11 Fig. Secondary NMR region for CFMPY-2.

    Shows cleaner resolution of overlapping signals.

    (PDF)

    pone.0334620.s011.pdf (61.2KB, pdf)
    S12 Fig. Secondary NMR region for CFMPY-4.

    Further confirms signal integrity for aromatic protons.

    (PDF)

    pone.0334620.s012.pdf (52.3KB, pdf)
    S13 Fig. Secondary NMR region for CFMPY-15.

    Enhances detail around methyl/methoxy substitutions.

    (PDF)

    pone.0334620.s013.pdf (53.8KB, pdf)
    S14 Fig. Secondary NMR region for CFMPY-17.

    Confirms consistency of multiple methoxy group signals.

    (PDF)

    pone.0334620.s014.pdf (61.2KB, pdf)
    S15 Fig. Secondary NMR region for CFMPY-28.

    Distinguishes ring protons from side-chain environments.

    (PDF)

    pone.0334620.s015.pdf (51.7KB, pdf)
    S16 Fig. Chemical structures of pyrimidine-substituted chalcones.

    Reference figure matching spectra with corresponding compound names and positions.

    (PDF)

    pone.0334620.s016.pdf (1.1MB, pdf)
    S17 Fig. Raw uncropped gel image for DNA fragmentation assay (Fig 5).

    Original gel documentation showing DNA fragmentation patterns in HeLa cells treated with synthesized compounds. Lanes: 1 – DNA Ladder (molecular weight marker), 2 - CFMPY-2, 3 - CFMPY-4, 4 - CFMPY-15, 5 - CFMPY-17, 6 - CFMPY-28, 7 – Cisplatin (positive control). This uncropped image is provided for transparency and to demonstrate the absence of gel manipulati.

    (JPG)

    pone.0334620.s017.jpg (47.7KB, jpg)
    Attachment

    Submitted filename: Reviewing comments.docx

    pone.0334620.s018.docx (12.2KB, docx)
    Attachment

    Submitted filename: Response to reviewer.docx

    pone.0334620.s020.docx (26.1KB, docx)

    Data Availability Statement

    All relevant data are included within the manuscript and its Supporting information files. The original uncropped DNA fragmentation gel images (Figure 5) and raw datasets (antioxidant assay values, cytotoxicity results, and farnesyl transferase assay readings) have been deposited in Figshare and are publicly available at https://doi.org/10.6084/m9.figshare.30030250. Requests for additional data access may be directed to Dr. M. R. Yadhav, Director, RDC, Parul University, Vadodara, Gujarat, India (email: mangeram.yadhav@gmail.com), who serves as a non-author institutional point of contact to ensure long-term accessibility.


    Articles from PLOS One are provided here courtesy of PLOS

    RESOURCES