Next Article in Journal
Novel Isolongifolenone-Based Caprolactam Derivatives as Potential Anticancer Agents via the p53/mTOR/Autophagy Pathway
Previous Article in Journal
Synthesis of New Brassinosteroid Analogs with Androstane Skeleton and Heterocyclic Acyl Side Chains: Preliminary Molecular Docking Studies
Previous Article in Special Issue
Cytotoxic Activity of Essential Oils from Middle Eastern Medicinal Plants on Malignant Keratinocytes
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

The Therapeutic Potential of Laurus nobilis L. Leaves Ethanolic Extract in Cancer Therapy

1
Department of Pharmaceutical Sciences, Faculty of Pharmacy, Zarqa University, Zarqa 13110, Jordan
2
Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmacy, Yarmouk University, Irbid 21163, Jordan
3
Department of Pharmacology and Biomedical Sciences, Faculty of Pharmacy and Medical Sciences, University of Petra, Amman 11196, Jordan
4
Cell Therapy Center, The University of Jordan, Amman 11942, Jordan
5
Department of Pharmaceutical Technology and Pharmaceutics, Faculty of Pharmacy, Yarmouk University, Irbid 21163, Jordan
*
Author to whom correspondence should be addressed.
Molecules 2025, 30(19), 4012; https://doi.org/10.3390/molecules30194012
Submission received: 8 August 2025 / Revised: 16 September 2025 / Accepted: 22 September 2025 / Published: 7 October 2025
(This article belongs to the Special Issue Advances in Plant-Sourced Natural Compounds as Anticancer Agents)

Abstract

This study explores the anticancer, antioxidant, and phytochemical activities of Laurus nobilis L. ethanolic leaf extract. The extract demonstrated selective cytotoxicity against four human cancer cell lines, showing strong cytotoxic effect against ovarian (ES2), head and neck (SAS), and colorectal (HT-29) cancer cells, with IC50 values ranging from 3.8 ± 0.3 to 4.4 ± 0.6 µg/mL. Notably, it exhibited only moderate inhibition of the MDA-MB-231 breast cancer cell line (IC50 = 18.5 ± 0.8 µg/mL), possibly reflecting intrinsic differences in cell line sensitivity. Importantly, the extract showed low toxicity toward normal human fibroblasts (HDF), with an IC50 value exceeding 100 µg/mL, indicating a favorable selectivity profile. The flow cytometry analysis showed that the extract caused cell death and stopped the cell cycle in both SAS and ES2 cancer cell lines. In SAS cells, extract treatment significantly increased apoptotic cells (21.1% ± 0.3%) compared to the control (6.3% ± 0.4%), along with G2 phase accumulation, indicating G2 arrest. Similarly, in ES2 cells, apoptosis increased (16.2% ± 1.3% vs. control 8.1% ± 1.0%), and a significant cell accumulation in the S phase was observed, suggesting disruption of cell cycle progression. Antioxidant screenings showed impressive dose-dependent DPPH radical scavenging activity (25–2000 µg/mL), although less potent than ascorbic acid (2.6 µg/mL). UPLC-QTOF/MS phytochemical analysis revealed various phenolic constituents, such as flavonoids and phenolic acids, and an inferred association with the recorded bioactivities. This preliminary work indicates that L. nobilis extracts may act as natural anticancer and antioxidant agents; however, it was limited to in vitro testing with non-standardized samples, underscoring the need for further research to validate and extend these findings for future applications.

Graphical Abstract

1. Introduction

Cancer is the second-leading cause of death worldwide, and the global cancer burden is predicted to grow over the next two decades [1]. Conventional cancer treatments, such as chemotherapy, can impose significant physical and psychological burdens, prompting increased interest in plant-based remedies as safer, adjunctive options [2]. Throughout history, people have turned to nature as a source of medicine, a tradition that persists in modern healthcare. The global popularity of herbal medicine has grown significantly, particularly as complementary and alternative therapies gain attention for their potential role in managing diseases, including cancer [3,4].
Laurus nobilis (family Lauraceae), commonly known as bay leaf, is a well-known medicinal plant widely used in Mediterranean regions, including Jordan. Its leaves and fruits are valued for their aromatic properties and have traditionally been used to treat gastrointestinal issues such as bloating and indigestion [5]. The essential oil extracted from the leaves has been shown to have antibacterial, antioxidant, and anti-inflammatory properties. Additional leaves of L. nobilis have been used to cure rheumatism, neuralgia, and scabies [6]. The studies suggest that L. nobilis possesses phenolic compounds, including flavonoids, phenolic acids, and tannins (proanthocyanidins) [7], with antioxidant properties, potentially contributing to its anticancer effects [5]. Because oxidative stress is a well-documented phenomenon in cancer, it is reasonable that antioxidants can greatly reduce cancer incidence and development [8]. However, further research is required to validate its therapeutic applications and understand its mechanisms of action. In Jordan, breast cancer is the most common cancer among women and is often diagnosed at advanced stages and younger ages than in Western countries [9]. Ovarian cancer ranks as the sixth leading cause of cancer-related deaths among women globally [10], while colorectal cancer is the most prevalent cancer in Jordanian men and the second most common in women [8]. Additionally, head and neck cancers, the seventh most common malignancies worldwide, contribute significantly to male cancer mortality [11]. Despite the growing global interest in herbal medicine and preliminary reports highlighting the antioxidant and potential anticancer properties of L. nobilis, evidence for its efficacy against specific cancer types remains limited, particularly those prevalent in Jordan. This study is novel in demonstrating its selective activity against multiple cancer cell lines, including SAS head and neck cancer cells, while confirming safety on normal fibroblasts and revealing mechanisms such as apoptosis and cell cycle arrest. Therefore, the present work aims to evaluate the anticancer and antioxidant activities of the ethanolic leaf extracts of L. nobilis and to identify their main phytochemical constituents using UPLC-QTOF/MS. The study is preliminary and seeks to establish the potential use of L. nobilis extract components in the future.

2. Results and Discussion

2.1. Cytotoxic Activity

The MTT assay was performed to evaluate the anticancer activity of the extract against MDA-MB-231, SAS, HT-29, and ES2 cell lines. As shown in Figure 1, treatment with increasing concentrations of the extract (0.03–100 µg/mL) resulted in a dose-dependent decrease in cell viability, indicating its potential cytotoxic effect across all tested cell lines.
To quantify this effect, IC50 values were determined from dose–response curves, including normal human fibroblasts, to evaluate the extract’s selectivity toward cancer cells. As summarized in Table 1, the extract exhibited potent inhibition on ES2 (IC50 = 4.2 ± 0.2 µg/mL), SAS (IC50 = 3.8 ± 0.2 µg/mL), and HT-29 (IC50 = 4.4 ± 0.6 µg/mL) cell lines, while demonstrating moderate inhibition against MDA-MB-231 cells (IC50 = 18.5 ± 0.8 µg/mL). The noticeably higher IC50 value observed in MDA-MB-231 cells (18.5 µg/mL) compared to the values obtained for ES2, HT-29, and SAS cells (3.8–4.4 µg/mL) may reflect intrinsic differences in cell line sensitivity. MDA-MB-231 is a triple-negative breast cancer cell line known for its aggressive phenotype, high metastatic potential, and resistance to a wide range of chemotherapeutic agents, which may contribute to the reduced sensitivity observed in our assay [12]. This highlights the potential cell type-specific activity of the extract, which may be more effective against certain tumor types. Notably, the extract displayed low toxicity toward normal fibroblast cells (IC50 > 100 µg/mL), indicating selectivity for cancer cells.
To benchmark the anticancer efficacy of L. nobilis ethanolic extract, its IC50 values were compared with those of cisplatin, a widely used chemotherapeutic agent. As shown in Table 1, the extract exhibited comparable cytotoxicity to cisplatin in ES2 and SAS cells, with IC50 values of 4.2 ± 0.2 and 3.8 ± 0.2 µg/mL, respectively, versus 3.6 ± 0.3 and 2.9 ± 0.5 µg/mL for cisplatin. Notably, the extract demonstrated superior cytotoxicity in HT-29 colorectal cancer cells (4.4 ± 0.6 µg/mL) compared to cisplatin (10.7 ± 2.1 µg/mL), underscoring its potential therapeutic relevance in colorectal cancer. For MDA-MB-231, a triple-negative breast cancer cell line known for its chemoresistance, both agents showed relatively higher IC50 values (18.5 ± 0.8 µg/mL for the extract vs. 16.7 ± 1.1 µg/mL for cisplatin), reflecting lower susceptibility of this phenotype.
These findings suggest a broad-spectrum anticancer activity of the extract, as it effectively inhibited the growth of multiple cancer cell lines from different tissue origins, including head and neck, colorectal, ovarian, and breast cancer [13]. The significant cytotoxic effects observed in ES2, SAS, and HT-29 cells, along with the moderate effect on MDA-MB-231, indicate that the extract may interfere with key oncogenic pathways shared across various cancer types. However, further investigation is needed to identify the specific molecular targets and mechanisms underlying its anticancer effects, which could provide valuable insights for future therapeutic development. Additionally, its low toxicity toward normal HDF highlights its potential selectivity for malignant cells, making it a promising candidate for further preclinical development as an anticancer agent [14]. This selective cytotoxicity may be explained by inherent differences between cancerous and normal cells. The selective cytotoxicity of the L. nobilis ethanolic extract toward malignant cells over normal human fibroblasts may be attributed to several cancer-specific biological characteristics. One key factor is the significantly higher proliferation rate of cancer cells compared to normal fibroblasts [15]. Rapidly dividing cells are more susceptible to agents that interfere with DNA replication, cell cycle progression, and oxidative stress—all of which are known mechanisms of action for phenolic compounds such as quercetin, kaempferol, and gallic acid identified in our extract [16]. Moreover, cancer cells typically exhibit elevated basal levels of reactive oxygen species and a disrupted redox balance [17], rendering them more vulnerable to additional oxidative stress induced by phenolic compounds [18].

2.2. Apoptosis Evaluation and Cell Cycle Analysis

To further investigate the mechanisms underlying the reduced cell viability, the induction of apoptosis in SAS and ES2 cells following extract treatment was assessed using Annexin V/PI double staining, as described in Section 3 [19]. The results, presented in Figure 2A,B, reveal a significant increase in Annexin V-positive apoptotic cells after treatment with the extract (SAS: control 6.3% ± 0.4% vs. extract 21.1% ± 0.3%, n = 3) and (ES2: control 8.1% ± 1.0% vs. extract 16.2% ± 1.3%, n = 3). These findings suggest that the extract may promote cell death through apoptosis, consistent with previous observations of plant extract-induced apoptosis in cancer cells [20]. However, further investigations are required to confirm this apoptotic effect by examining caspase activation, mitochondrial membrane potential, and ROS generation, which are key markers of intrinsic apoptotic pathways [21].
To further investigate the mechanism of action of the L. nobilis extract, we examined its impact on cell cycle progression in SAS and ES2 cells. As shown in Figure 2C, treatment with the extract resulted in a significant accumulation of cells in the G2 phase (SAS: control 42.5% ± 6.1% vs. extract 77.9% ± 1.7%, n = 3). This was accompanied by a marked reduction in the proportion of cells in the G1 and S phases, indicating G2 phase arrest (G1 phase: control 52.9% ± 6.5% vs. extract 20.7% ± 1.9%, n = 3; S phase: control 4.6% ± 1.4% vs. extract 1.4% ± 0.2%, n = 3). In contrast, ES2 cells exhibited a distinct response, with extract treatment leading to a marked accumulation in the S phase (control 7.4% ± 0.3% vs. extract 51.2% ± 1.9%, n = 3) and a substantial reduction in the G2 phase population (control 43.9% ± 0.7% vs. extract 20.7% ± 1.9%). These findings indicate that L. nobilis extract induces cell cycle arrest in a cell type-dependent manner, consistent with the phase-specific responses seen in many plant-derived compounds [22,23]. The observed disruption in normal cell cycle progression, particularly G2 arrest in SAS and S phase accumulation in ES2, may prevent cancer cells from entering mitosis, thus inhibiting their proliferation and contributing to the extract’s anticancer potential [24]. This finding indicates that the extract may exert its anticancer effects by inducing cell cycle arrest at a critical checkpoint, thereby inhibiting tumor cell proliferation [25]. Further studies are needed to elucidate the molecular mechanisms underlying this effect, including the potential involvement of key cell cycle regulators such as CDK1, Cyclin B1, and WEE1 kinase. Cell cycle arrest, notably at the G2 and S phases, is often associated with the activation of apoptotic pathways, especially when cells are unable to repair damage or resume progression [26]. Here, the extract’s ability to halt cell cycle progression may also trigger apoptosis, thereby enhancing its anticancer potential. These findings suggest a dual mechanism of action, involving both cell cycle disruption and induction of programmed cell death [27].
Moreover, previous studies have evaluated various L. nobilis extracts for their cytotoxic potential across different cancer cell lines. Essential oils derived from L. nobilis leaves and seeds demonstrated weak to moderate cytotoxicity, with IC50 values exceeding 500 µg/mL against SH-SY5Y neuroblastoma cells [28] and 75–95 µg/mL against K562 leukemia cells [29]. In stark contrast, our phenolic-rich ethanolic leaf extract exhibited markedly stronger anticancer activity, with IC50 values ranging from 3.8 to 18.5 µg/mL against the investigated cell lines. This represents more than a 20-fold increase in potency compared to essential oil extracts. These findings underscore the importance of non-volatile, polyphenolic constituents—such as quercetin, kaempferol, and gallic acid—identified in our extract via UPLC-QTOF/MS and known for their pro-apoptotic and antiproliferative effects [30].
Similarly, chloroform extracts and their fractions showed lower cytotoxic activity against SK-N-BE (2)-C and SH-SY5Y neuroblastoma cell lines, with IC50 values ranging from 19 to 250 µg/mL [23]. A previous ethanolic extract also exhibited an IC50 of 24.49 µg/mL against the MCF-7 breast cancer cell line [31] This aligns with our findings against the MDA-MB-231 breast cancer cell line (IC50 = 18.5 ± 0.8 µg/mL), although our extract demonstrated even greater potency against SAS, ES2, and HT-29 cells. Collectively, these comparisons highlight the potential of phenolic-rich ethanolic extracts as promising sources for the development of selective anticancer agents from L. nobilis.
Although the L. nobilis extract is slightly less potent than standard chemotherapeutic agents such as cisplatin, which typically exhibit IC50 values of 1.6–4 µg/mL against the investigated cell lines [32], its cytotoxic activity remains remarkable for a crude plant extract. Notably, these values are substantially lower than those reported for many other botanical extracts [33].

2.3. Antioxidant Activity Extracts by DPPH Assay

The antioxidant activity of the L. nobilis leaves extract was determined by DPPH radical scavenging assay. The scavenging of DPPH radicals caused by hydrogen donation from antioxidants result in decrease DPPH radical absorbance at 517 nm [34]. As shown in Table 2, the extract showed significant concentration-dependent free radical scavenging activity from (25–2000 µg/mL), with a IC50 at a concentration of 713 µg/mL as compared with the standard ascorbic acid (2.6 µg/mL). Low IC50 values correspond to high antioxidant activity [34].
The results could hint to considerable quenching actions of phenolic compounds like flavonoids found in laurel leaves, which have been detected by LC-MS/MS, against DPPH radicals. As excessive free radicals contribute to many chronic clinical conditions, the extract may help to minimize the radicals damage [34]. This antioxidant capability is especially important in the context of oxidative stress and disease development. Antioxidants are widely characterized as “substances that delay, prevent, or remove oxidative damage to specific target molecules by reacting with oxidants,” such as ROS. Antioxidants and oxidants in equilibrium maintain redox homeostasis, while an imbalance in favor of oxidants can cause oxidative stress, which has been associated with cancer etiology. Cancer tissues are thought to produce more ROS because to altered metabolism, inflammation, a hypoxic environment, and oncogene-induced activation of ROS-generating enzymes, highlighting the role of antioxidants in buffering excess ROS and potentially slowing cancer growth [35]. Besides from redox regulation, antioxidants may also induce intrinsic cellular responses that contribute to cancer cell death. one of the most potent cytotoxic processes activated by these natural antioxidant is apoptosis. This approach can cause planned cell death and work alongside with necrosis. Because there are distinct indicators for determining which apoptotic pathway has happened, the action mechanism of each medication may be studied more precisely in future investigations [36].

2.4. Bioactive Compounds in L. nobilis Leaves Ethanolic Extract

In order to provide insight into the polyphenolic composition of the L. nobilis leaves ethanolic extract, UPLC-MS/MS analysis was performed. A number of phenolic compounds, including flavonoids, and phenolic acids, were identified in the extract Table 3.
Phenolic compounds have attracted great attention on the strength of their capacity to enhance the anticancer activity of conventional chemotherapeutic agents. There is growing evidence that suggests such bioactive compounds have the potential to exert synergistic anticancer effects in various cancer cell lines and therefore represent potential adjuncts to anticancer therapy [37].
Flavonoids are promising cytotoxic anticancer compounds due to their biological potential and minimal side effects. Flavonoids exert their anticancer effect by various mechanisms like ROS scavenging activity, suppress cancer cells proliferation, and participating in arresting the cell cycle [38]. Several key flavonoids have been identified in the L. nobilis leaves ethanolic extract (Table 3) like quercetin, kaempferol, naringenin, and rutin where previous studies have shown their potential role against various cancer cell lines [39] and therefore they contribute to the observed cytotoxicity of L. nobilis leaves extract against ES2, SAS, and HT-29 cancer cells.
Naringin, a flavonoid polyphenol found primarily in grapefruit and certain citrus fruits, has been reported to exhibit chemosensitizing activity, particularly in combination with paclitaxel. Naringin has been shown by research to enhance the cytotoxicity of paclitaxel in both androgen-independent and androgen-dependent human prostate cancer cells [40]. The enhancement procedure was performed through the mediation of intrinsic apoptosis pathway activation and the G1 cell cycle arrest. Naringin also had the ability to enhance PTEN expression, a negative regulator of the PI3K/Akt signaling cascade. In parallel, it prevents cancer cell invasion and migration through the downregulation of NF-κB, Snail, Twist, and c-Myc mRNA levels, all of which are implicated in metastasis and cancer development [41].
Similarly, quercetin, a well-studied phenolic compound, was also identified to be a potentiator of temozolomide, an oral alkylating agent employed in glioblastoma treatment. In vitro testing on U87 and U251 glioblastoma cell lines revealed that quercetin sensitize the cells to temozolomide by blocking heat-shock proteins, which are typically at the center of chemoresistance mechanisms [41].
Furthermore, in a phase I clinical trial, resveratrol dramatically decreased the levels of circulating cancer biomarkers such as Insulin-like growth factor 1 and Insulin-like growth factor-binding protein 3 [42]. Another phase I, double-blind, randomized clinical investigation found that resveratrol raised the levels of cleaved caspase-3 in malignant liver tissue [43].
Phenolic acids, a large group of non-flavonoid phenolics of plant origin, exist either in the free form (aglycones) or as glycosides. They are distributed extensively in all food sources like oilseeds, cereals, legumes, fruits and vegetables, herbs, and an enormous variety of beverages. These phenolic acids have attracted increasing attention due to their versatile biological activities, with particular interest in their potential anticancer activity [44]. The UPLC-MS/MS analysis of the L. nobilis leaves ethanolic extract indicated the presence of biologically important phenolic acids like gallic acid, caffeic acid, cinnamic acid, and vanillic acid. It was previously reported that phenolic acids show their anticancer effect through antioxidant activity, cell cycle arrest, and induction of apoptosis [45] which may explain the observed antioxidant and cytotoxicity of L. nobilis leaves extract.
Gallic acid, which is well-characterized phenolic acid, showed potential anticancer activity in vitro. Its activity was recently studied by Ko et al. against the A549 NSCLC cell line. Cell viability was studied after 48 h through a dose-dependent cell proliferation inhibition assay, suggesting its potential as a cytotoxic agent against NSCLC cells [46]. Similarly, caffeic acid has shown activity against estrogen receptor-positive (ER+) and estrogen receptor-negative (ER−) breast cancer cell lines. Rosendahl et al. established that caffeic acid is an anti-estrogenic mimic and disrupts significant signaling pathways, including the IGF-IR/pAkt and ER/cyclin D1 axes. Disruption results in cell cycle arrest and reduced proliferation, and the findings suggest that caffeic acid sensitizes tumor cells to endocrine treatment such as tamoxifen, enhancing the treatment response [47]. Vanillic acid has also been reported to have anticancer properties. In an experiment that tested its effect on breast and prostate cancer cell lines, it was determined that vanillic acid level is associated with reduced viability of cells. These results suggest that vanillic acid may be among the major compounds responsible for the antiproliferative effects in these cancer models, thus a suitable candidate as a bioactive compound to be involved in dietary cancer prevention [48].
The present study highlights a key strength compared to previous studies in that it evaluated the selective anticancer activity of the crude ethanolic extract of L. nobilis against different cancer cell lines, including some (e.g., SAS head and neck cancer cells) that have not been studied before, while simultaneously assessing its safety on normal fibroblasts. Unlike many earlier studies that focused primarily on general antioxidant or cytotoxic properties, this work offers evidence of both selectivity and underlying mechanisms, including apoptosis induction and cell cycle arrest. Collectively, these findings underscore the therapeutic promise of phenolic compounds in overcoming drug resistance and enhancing the anticancer efficacy of standard chemotherapeutic agents. Though the current study is limited to in vitro assays and crude extract analysis, and the samples examined were not standardized—making the reproducibility of the findings dependent on the stability of the extract’s composition—which highlights the need for further in vivo and mechanistic studies to validate and extend these results.

3. Materials and Methods

3.1. Plant Material and Extraction

The leaves of L. nobilis were purchased from the local traditional herbal market in Amman, Jordan. The plant was taxonomically identified with the assistance of Dr. Al-Gharaibeh, Faculty of Agriculture, Jordan Department of Plant Production, University of Science and Technology. The voucher specimens (No. Bay-2023) were deposited at the Department of Pharmaceutical Sciences, Faculty of Pharmacy, Zarqa University, Zaqa, Jordan (Pharmacognosy Lab). The ground leaves of L. nobilis leaves (50 g) were extracted with 70% ethanol at 60 °C in a Soxhlet apparatus until the refluxed solvent became colorless. The extract was separately evaporated to dryness in a rotary vacuum evaporator, then weighted. The ethanolic extract was stored at 4 °C until subjected to analysis and in vitro tests.

3.2. Cell Culture

Prof. Stephan Feller, Institute of Molecular Medicine, Martin Luther University, Germany, kindly provided the human cancer cell lines MDA-MB-231 (breast cancer cells), SAS (head and neck cancer cells), HT-29 (colorectal cancer cells), and ES2 (ovarian cancer cells). The human dermal fibroblast (HDF) cell line was obtained as a kind gift from Prof. Lina Dahabiyeh (Jordan University, Amman, Jordan). The choice of cancer cell lines was based on their clinical and epidemiological significance, given that breast, colorectal, ovarian, and head and neck cancers rank among the most prevalent malignancies in Jordan. HDFs were included as a non-malignant control to evaluate the extract’s selectivity toward cancer cells.
Cancer cell lines were cultured in Dulbecco’s Modified Eagle’s Medium (DMEM) (Sigma-Aldrich, Darmstadt, Germany) at 37 °C in a humidified incubator with 5% CO2. Human dermal fibroblasts were maintained under the same conditions using Iscove’s Modified Dulbecco’s Medium (IMDM (Sigma-Aldrich, Darmstadt, Germany). The media were supplemented with 10% fetal bovine serum (FBS), 100 µg/mL streptomycin, 100 U/mL penicillin, and 1% non-essential amino acids. Cell cultures were grown to confluence, with the medium replaced twice weekly.

3.3. In Vitro Cytotoxicity Assay

The MTT (3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl tetrazolium bromide) assay was used to examine cell viability and to calculate the half-maximal inhibitory concentration (IC50) values of the L. nobilis extract against various cancer cells and the HDF cell line. A stock solution of the extract was prepared at 100 mg/mL in DMSO, where it dissolved completely. Cells were seeded in 96-well plates at a density of 6000 cells/well for MDA-MB-231, HT-29, and HDF cells and 4000 cells/well for SAS and ES2 cells. After 24 h of incubation, cells were treated with serial concentrations of the extract or cisplatin (100, 30, 10, 3, 1, 0.3, 0.1, and 0.03 µg/mL) for 72 h. At the end of the treatment period, the supernatant was aspirated, and 50 µL of fresh DMEM containing MTT (0.5 mg/mL) was added to each well. Cells were then incubated at 37 °C for 2–4 h. The medium was then completely removed, and 50 µL of DMSO was added to dissolve the precipitated formazan crystals formed by viable cells [49].
The Synergy HTX Multimode Reader (BioTek, Shoreline, WA, USA) was used to measure cell viability, as represented by the absorbance of the formazan solution, at 570 nm. For each concentration, the relative cell viability was calculated as follows:
Relative cell viability = (mean treatment absorbance/mean control absorbance) × 100%.
Assays were performed in duplicate on three independent experiments. IC50 values were determined utilizing the four-parameter logistic function within the Sigmaplot 12.5 software and are expressed as the mean ± SD derived from three independent experiments.

3.4. Cell Cycle Analysis

SAS and ES2 cells were seeded in 6-well plates at a density of 1 × 105 cells/well and treated for 72 h with the IC50 concentration of the extract or DMSO as a vehicle control. Following treatment, both the floating dead cells and attached cells were collected and fixed in cold ethanol at −20 °C for 30 min. Cells were then stained with a propidium iodide (PI) working solution (1 µg/mL PI and 10 µg/mL RNase A in PBS) for 30 min at 37 °C in the dark, followed by PBS washing [50]. Fluorescence was analyzed using a FACS Accuri C6 Plus flow cytometer (BD Biosciences, San Jose, CA, USA). Histograms representing cell counts versus fluorescence intensity were generated, and cell cycle distribution was determined based on mean fluorescence intensity values. A total of 10,000 events were measured in each experiment and analyzed using FlowJo software (version 10). All flow cytometry experiments were performed in three independent biological replicates. Results are reported as mean ± standard deviation (SD) to reflect reproducibility across independent experiments.

3.5. Apoptosis Evaluation by Annexin V-FITC/PI Staining

SAS and ES2 cells were seeded in 6-well plates and treated with the extract or DMSO as previously described. Following treatment, both floating dead cells and attached cells were collected, washed with PBS, and resuspended in 100 μL of Annexin V solution for 30 min, following the manufacturer’s instructions (Miltenyi Biotec, Bergisch Gladbach, Germany) #130-092-052) [51]. After Annexin V staining, cells were briefly stained with PI to exclude dead cells. Apoptosis quantification was performed using a FACS Accuri C6 Plus flow cytometer (BD Biosciences, San Jose, CA, USA), with 10,000 events recorded and analyzed using FlowJo software (version 10).

3.6. Antioxidant Assay

The free radical scavenging activity of L. nobilis leaves extract was determined by measuring the decrease in absorbance of DPPH at 517 nm as described by Gulcin et al. (2003) [52]. Briefly, 1 mL of 0.1 mM DPPH in methanol was added to 3 mL of bay extract solution in methanol at various concentrations (25–2000 µg/mL). The resulting mixture was incubated in the dark for 30 min, and then absorbance was measured at 517 nm. The control sample was prepared by adding 1 mL of DPPH to 3 mL of methanol. Vitamin C was used as a reference standard.
The ability of the extract to scavenge DPPH radical was calculated according to the following equation:
DPPH Scavenging Activity = [(Ao − A1/Ao) × 100]
where Ao is the absorbance of the control reaction and A1 is the absorbance in the presence of the sample of L. nobilis extract. The results were expressed as mean value ± standard deviation (SD).

3.7. Identification of Bioactive Compounds in L. nobilis Leaves Ethanolic Extract

Ultra-performance liquid chromatography (UPLC) equipped with quadrupole time-of-flight mass spectrometry (Q-TOF/MS) was utilized to characterize bioactive chemicals. The UPLC chromatographic conditions are detailed in Table 4. The Apollo II ion funnel electrospray source was employed for ionization in both positive and negative modes. The mass spectrometry conditions were as follows:
  • Capillary voltage: 2500 volts (VT);
  • Nebulizer gas pressure: 2.0 bar;
  • Dry gas (nitrogen) flow rate: 8 L/min;
  • Dry gas temperature: 200 °C.
The instrument provided a mass resolution of 50,000 full sensitivity resolution (FSR) and a mass accuracy of 1 ppm. The time-of-flight (TOF) repetition rate reached up to 20 kHz. Data analysis was conducted using the Data Analysis 4.2 software (Bruker Daltonics, Bremen, Germany). Compound identification was carried out by LC-MS/MS using two complementary approaches: (i) direct comparison with authentic standards when available, and (ii) tentative assignment based on accurate mass and fragmentation patterns with spectral library matching.

4. Conclusions

L. nobilis leaf extract exhibited selective anticancer activity against ovarian (ES2), head and neck (SAS), and colorectal (HT-29) cancer cell lines, while showing minimal cytotoxicity toward normal fibroblasts. Its effects included the induction of apoptosis and interruption of cancer cell proliferation by causing G2 phase arrest in SAS cells and S phase accumulation in ES2 cells. The extract was also found to have antioxidant activity, though less than ascorbic acid. These effects can be attributed to the known phenolic compounds such as flavonoids and phenolic acids.
Notably, the crude ethanolic extract (10% yield from dried leaves) was directly assessed without additional purification, which may pose challenges in pinpointing the exact bioactive molecules. Nevertheless, the LC-MS chromatogram of the crude extract supports the presence of diverse phenolic constituents, likely contributing synergistically to the observed effects. Future studies involving fractionation, purification, and detailed profiling are warranted to improve reproducibility and to detect the compounds most responsible for the anticancer and antioxidant activities.
Based on this study, L. nobilis may be deemed to possess excellent potential as a source of natural development of selective anticancer and antioxidant drugs and deserves further research.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/molecules30194012/s1, Table S1: Some of previously reported anticancer activities of identified phenolic compounds in Laurus nobilis ethanolic leaf extract; Figure S1: Chromatograms of compounds identified in L. noblis extract, see [53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111].

Author Contributions

Conceptualization, F.A.-M. and A.M.A.Q.; methodology, F.A.-M., A.M.A.Q. and A.H.; validation, investigation and data curation F.A.-M., A.M.A.Q., D.A.-T., D.A. and A.H.; writing—review and editing, F.A.-M., D.A.-T. and A.H.; supervision, F.A.-M.; project administration, F.A.-M. All authors have read and agreed to the published version of the manuscript.

Funding

This research is partially funded by Zarqa University-Jordan.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The original contributions presented in this study are included in the article. Further inquiries can be directed to the corresponding author.

Acknowledgments

The authors express their sincere gratitude to Stephan Feller, Institute of Molecular Medicine, Martin Luther University, Germany, for generously offering the human cancer cell lines used in the current study and for Lina Dahabiyeh for generously providing the Human Dermal Fibroblast (HDF) cell line. They also extend their sincere appreciation to Naba Hikma Industrial and Testing Services for their technical assistance and support. Appreciation is expressed to Al-Gharaibeh, Faculty of Agriculture, Department of Plant Production, Jordan University of Science and Technology, for providing assistance in the taxonomic identification of Laurus nobilis.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
DMSODimethyl Sulfoxide
DMEMDulbecco’s Modified Eagle’s Medium
FBSFetal Bovine Serum
FSRFull Sensitivity Resolution
IC50Half Maximal Inhibitory Concentration
HDFHuman Dermal Fibroblast
IGF-IRInsulin-like Growth Factor-1 Receptor
IMDMIscove’s Modified Medium
NSCLCNon-Small Cell Lung Cancer
NF-ĸBNuclear Factor KappaB
PTENPhosphate and TENsin Homolog
PBS Phosphate-Buffer Saline
PI3KPhosphoinositide 3-Kinases
PIPropidium Iodide
Q-TOF/MSQuadrupole Time-of-Flight Mass Spectrometry
ROSReactive Oxygen Species
TOFTime-of-Flight
UPLCUltra Performance Liquid Chromatography

References

  1. Kocarnik, J.M.; Compton, K.; Dean, F.E.; Fu, W.; Gaw, B.L.; Harvey, J.D.; Henrikson, H.J.; Lu, D.; Pennini, A.; Xu, R.; et al. Cancer incidence, mortality, years of life lost, years lived with disability, and disability-adjusted life years for 29 cancer groups from 2010 to 2019: A systematic analysis for the Global Burden of Disease Study 2019. JAMA Oncol. 2022, 8, 420–444. [Google Scholar] [CrossRef] [PubMed]
  2. Wani, M.; Parakh, S.; Dehghan, M.; Polshettiwar, S.; Chopade, V.; Chepurwar, S. Herbal medicine and its standardization. Pharm. Rev. 2007, 5, 411038–411049. [Google Scholar]
  3. Ochwang’i, D.O.; Kimwele, C.N.; Oduma, J.A.; Gathumbi, P.K.; Mbaria, J.M.; Kiama, S.G. Medicinal plants used in treatment and management of cancer in Kakamega County, Kenya. J. Ethnopharmacol. 2014, 151, 1040–1055. [Google Scholar] [CrossRef] [PubMed]
  4. Greenwell, M.; Rahman, P. Medicinal plants: Their use in anticancer treatment. Int. J. Pharm. Sci. Res. 2015, 6, 4103–4112. [Google Scholar] [CrossRef]
  5. Abu-Dahab, R.; Kasabri, V.; Afifi, F.U. Evaluation of the Volatile Oil Composition and Antiproliferative Activity of Laurus nobilis L. (Lauraceae) on Breast Cancer Cell Line Models. Rec. Nat. Prod. 2014, 8, 136–147. [Google Scholar]
  6. Mohammed, R.R.; Omer, A.K.; Yener, Z.; Uyar, A.; Ahmed, A.K. Biomedical effects of Laurus nobilis L. leaf extract on vital organs in streptozotocin-induced diabetic rats: Experimental research. Ann. Med. Surg. 2021, 61, 188–197. [Google Scholar] [CrossRef]
  7. Khodja, Y.K.; Bachir-Bey, M.; Belmouhoub, M.; Ladjouzi, R.; Dahmoune, F.; Khettal, B. The botanical study, phytochemical composition, and biological activities of Laurus nobilis L. leaves: A review. Int. J. Second. Metab. 2023, 10, 269–296. [Google Scholar] [CrossRef]
  8. Luo, M.; Zhou, L.; Huang, Z.; Li, B.; Nice, E.C.; Xu, J.; Huang, C. Antioxidant therapy in cancer: Rationale and progress. Antioxidants 2022, 11, 1128. [Google Scholar] [CrossRef]
  9. Abdel-Razeq, H.; Mansour, A.; Jaddan, D. Breast cancer care in Jordan. JCO Glob. Oncol. 2020, 6, 260–268. [Google Scholar] [CrossRef]
  10. Azar, J.; Kaddoura, T.; Timonian, M.A.; Karam, E.S.; Abou-Kheir, W.; Daoud, G. Ovarian cancer in the Arab world: An updated review. Gene Rep. 2024, 35, 102025. [Google Scholar] [CrossRef]
  11. Alotaibi, T.N.; Alsheshani, T.M.; Mohammed, K. Systematic Review of Head and Neck Cancer in Jordan. Int. J. Med. Sci. Clin. Res. Rev. 2024, 7, 1010–1020. [Google Scholar]
  12. Błaszczak, E.; Miziak, P.; Odrzywolski, A.; Baran, M.; Gumbarewicz, E.; Stepulak, A. Triple-Negative Breast Cancer Progression and Drug Resistance in the Context of Epithelial–Mesenchymal Transition. Cancers 2025, 17, 228. [Google Scholar] [CrossRef] [PubMed]
  13. Chandra, S.; Gahlot, M.; Choudhary, A.N.; Palai, S.; de Almeida, R.S.; de Vasconcelos, J.E.L.; dos Santos, F.A.V.; de Farias, P.A.M.; Coutinho, H.D.M. Scientific evidences of anticancer potential of medicinal plants. Food Chem. Adv. 2023, 2, 100239. [Google Scholar] [CrossRef]
  14. Nalkiran, I.; Sevim Nalkiran, H. Phytochemical Profile and Anticancer Potential of Helichrysum arenarium Extracts on Glioblastoma, Bladder Cancer, and Breast Cancer Cells. Pharmaceuticals 2025, 18, 144. [Google Scholar] [CrossRef]
  15. Hu, D.; Zhuo, W.; Gong, P.; Ji, F.; Zhang, X.; Chen, Y.; Mao, M.; Ju, S.; Pan, Y.; Shen, J. Biological differences between normal and cancer-associated fibroblasts in breast cancer. Heliyon 2023, 9, e19140. [Google Scholar] [CrossRef]
  16. Yang, P.; Li, X.; Wen, Q.; Zhao, X. Quercetin attenuates the proliferation of arsenic-related lung cancer cells via a caspase-dependent DNA damage signaling. Mol. Carcinog. 2022, 61, 655–663. [Google Scholar] [CrossRef]
  17. Attique, I.; Haider, Z.; Khan, M.; Hassan, S.; Soliman, M.M.; Ibrahim, W.N.; Anjum, S. Reactive Oxygen Species: From Tumorigenesis to Therapeutic Strategies in Cancer. Cancer Med. 2025, 14, e70947. [Google Scholar] [CrossRef]
  18. Li, L.; Jin, P.; Guan, Y.; Luo, M.; Wang, Y.; He, B.; Li, B.; He, K.; Cao, J.; Huang, C.; et al. Exploiting Polyphenol-Mediated Redox Reorientation in Cancer Therapy. Pharmaceuticals 2022, 15, 1540. [Google Scholar] [CrossRef]
  19. Hmedat, A.N.; Morejón, M.C.; Rivera, D.G.; Pantelić, N.Đ.; Wessjohann, L.A.; Kaluđerović, G.N. Cyclic Lipopeptides as Selective Anticancer Agents: In Vitro Efficacy on B16F10 Mouse Melanoma Cells. Anti-Cancer Agents Med. Chem. 2025, 25, 873–882. [Google Scholar] [CrossRef]
  20. Alaouna, M.; Molefi, T.; Khanyile, R.; Chauke-Malinga, N.; Chatziioannou, A.; Luvhengo, T.E.; Raletsena, M.; Penny, C.; Hull, R.; Dlamini, Z. The Potential of the South African Plant Tulbaghia violacea Harv for the Treatment of Triple-Negative Breast Cancer. Sci. Rep. 2025, 15, 5737. [Google Scholar] [CrossRef]
  21. Wani, A.K.; Akhtar, N.; Mir, T.u.G.; Singh, R.; Jha, P.K.; Mallik, S.K.; Sinha, S.; Tripathi, S.K.; Jain, A.; Jha, A.; et al. Targeting Apoptotic Pathway of Cancer Cells with Phytochemicals and Plant-Based Nanomaterials. Biomolecules 2023, 13, 194. [Google Scholar] [CrossRef]
  22. Bian, Y.; Yang, L.; Sheng, W.; Li, Z.; Xu, Y.; Li, W.; Zeng, L. Ligustrazine Induces the Colorectal Cancer Cells Apoptosis via p53-Dependent Mitochondrial Pathway and Cell Cycle Arrest at the G0/G1 Phase. Ann. Palliat. Med. 2021, 10, 1578–1588. [Google Scholar] [CrossRef]
  23. Li, W.M.; Liu, H.T.; Li, X.Y.; Wu, J.Y.; Xu, G.; Teng, Y.Z.; Ding, S.T.; Yu, C. The Effect of Tetramethylpyrazine on Hydrogen Peroxide-Induced Oxidative Damage in Human Umbilical Vein Endothelial Cells. Basic Clin. Pharmacol. Toxicol. 2010, 106, 45–52. [Google Scholar] [CrossRef]
  24. Pacifico, S.; Gallicchio, M.; Lorenz, P.; Potenza, N.; Galasso, S.; Marciano, S.; Fiorentino, A.; Stintzing, F.C.; Monaco, P. Apolar Laurus nobilis Leaf Extracts Induce Cytotoxicity and Apoptosis towards Three Nervous System Cell Lines. Food Chem. Toxicol. 2013, 62, 628–637. [Google Scholar] [CrossRef] [PubMed]
  25. El-Huneidi, W.; Shehab, N.G.; Bajbouj, K.; Vinod, A.; El-Serafi, A.; Shafarin, J.; Bou Malhab, L.J.; Abdel-Rahman, W.M.; Abu-Gharbieh, E. Micromeria fruticosa Induces Cell Cycle Arrest and Apoptosis in Breast and Colorectal Cancer Cells. Pharmaceuticals 2020, 13, 115. [Google Scholar] [CrossRef] [PubMed]
  26. Rattanapornsompong, K.; Khattiya, J.; Phannasil, P.; Phaonakrop, N.; Roytrakul, S.; Jitrapakdee, S.; Akekawatchai, C. Impaired G2/M Cell Cycle Arrest Induces Apoptosis in Pyruvate Carboxylase Knockdown MDA-MB-231 Cells. Biochem. Biophys. Rep. 2021, 25, 100903. [Google Scholar] [CrossRef] [PubMed]
  27. Chaudhry, G.-e.-S.; Md Akim, A.; Sung, Y.Y.; Sifzizul, T.M.T. Cancer and apoptosis: The apoptotic activity of plant and marine natural products and their potential as targeted cancer therapeutics. Front. Pharmacol. 2022, 13, 842376. [Google Scholar] [CrossRef]
  28. Caputo, L.; Nazzaro, F.; Souza, L.F.; Aliberti, L.; De Martino, L.; Fratianni, F.; Coppola, R.; De Feo, V. Laurus nobilis: Composition of essential oil and its biological activities. Molecules 2017, 22, 930. [Google Scholar] [CrossRef]
  29. Saab, A.M.; Tundis, R.; Loizzo, M.R.; Lampronti, I.; Borgatti, M.; Gambari, R.; Menichini, F.; Esseily, F.; Menichini, F. Antioxidant and antiproliferative activity of Laurus nobilis L. (Lauraceae) leaves and seeds essential oils against K562 human chronic myelogenous leukaemia cells. Nat. Prod. Res. 2012, 26, 1741–1745. [Google Scholar] [CrossRef]
  30. Nguyen, L.T.; Lee, Y.H.; Sharma, A.R.; Park, J.B.; Jagga, S.; Sharma, G.; Lee, S.S.; Nam, J.S. Quercetin induces apoptosis and cell cycle arrest in triple-negative breast cancer cells through modulation of Foxo3a activity. Korean J. Physiol. Pharmacol. 2017, 21, 205–213. [Google Scholar] [CrossRef]
  31. Al-Kalaldeh, J.Z.; Abu-Dahab, R.; Afifi, F.U. Volatile oil composition and antiproliferative activity of Laurus nobilis, Origanum syriacum, Origanum vulgare, and Salvia triloba against human breast adenocarcinoma cells. Nutr. Res. 2010, 30, 271–278. [Google Scholar] [CrossRef]
  32. Ota, T.; Jono, H.; Ota, K.; Shinriki, S.; Ueda, M.; Sueyoshi, T.; Nakatani, K.; Hiraishi, Y.; Wada, T.; Fujita, S.; et al. Downregulation of midkine induces cisplatin resistance in human oral squamous cell carcinoma. Oncol. Rep. 2012, 27, 1674–1680. [Google Scholar] [CrossRef] [PubMed]
  33. Bagheri, E.; Hajiaghaalipour, F.; Nyamathulla, S.; Salehen, N.A. The apoptotic effects of Brucea javanica fruit extract against HT29 cells associated with p53 upregulation and inhibition of NF-κB translocation. Drug Des. Devel. Ther. 2018, 12, 657–671. [Google Scholar] [CrossRef] [PubMed]
  34. Elmastaş, M.; Gülçin, İ.; Işildak, Ö.; Küfrevioğlu, Ö.; İbaoğlu, K.; Aboul-Enein, H. Radical scavenging activity and antioxidant capacity of bay leaf extracts. J. Iran. Chem. Soc. 2006, 3, 258–266. [Google Scholar] [CrossRef]
  35. Hecht, F.; Zocchi, M.; Alimohammadi, F.; Harris, I.S. Regulation of antioxidants in cancer. Mol. Cell 2024, 84, 23–33. [Google Scholar] [CrossRef]
  36. Oliveira, C.; Nicolau, A.; Teixeira, J.A.; Domingues, L. Cytotoxic effects of native and recombinant frutalin, a plant galactose-binding lectin, on HeLa cervical cancer cells. J. Biomed. Biotechnol. 2011, 2011, 568932. [Google Scholar] [CrossRef]
  37. Lewandowska, U.; Gorlach, S.; Owczarek, K.; Hrabec, E.; Szewczyk, K. Synergistic Interactions Between Anticancer Chemotherapeutics and Phenolic Compounds and Anticancer Synergy Between Polyphenols. Postepy Hig. Med. Dosw. 2014, 68, 528–540. [Google Scholar] [CrossRef]
  38. de Luna, F.C.F.; Ferreira, W.A.S.; Casseb, S.M.M.; de Oliveira, E.H.C. Anticancer potential of flavonoids: An overview with an emphasis on tangeretin. Pharmaceuticals 2023, 16, 1229. [Google Scholar] [CrossRef]
  39. Kopustinskiene, D.M.; Jakstas, V.; Savickas, A.; Bernatoniene, J. Flavonoids as anticancer agents. Nutrients 2020, 12, 457. [Google Scholar] [CrossRef]
  40. Erdogan, S.; Doganlar, O.; Doganlar, Z.B.; Turkekul, K. Naringin sensitizes human prostate cancer cells to paclitaxel therapy. Prostate Int. 2018, 6, 126–135. [Google Scholar] [CrossRef]
  41. Kikuchi, H.; Yuan, B.; Hu, X.; Okazaki, M. Chemopreventive and anticancer activity of flavonoids and their potential for clinical use with conventional chemotherapeutic agents. Am. J. Cancer Res. 2019, 9, 1517–1535. [Google Scholar] [PubMed]
  42. Brown, V.A.; Patel, K.R.; Viskaduraki, M.; Crowell, J.A.; Perloff, M.; Booth, T.D.; Vasilinin, G.; Sen, A.; Schinas, A.M.; Piccirilli, G.; et al. Repeat dose study of the cancer chemopreventive agent resveratrol in healthy volunteers: Safety, pharmacokinetics, and effect on the insulin-like growth factor axis. Cancer Res. 2010, 70, 9003–9011. [Google Scholar] [CrossRef] [PubMed]
  43. Howells, L.M.; Berry, D.P.; Elliott, P.J.; Jacobson, E.W.; Hoffmann, E.; Hegarty, B.; Brown, K.; Steward, W.P.; Gescher, A.J. Phase I randomized, double-blind pilot study of micronized resveratrol (SRT501) in patients with hepatic metastases: Safety, pharmacokinetics, and pharmacodynamics. Cancer Prev. Res. 2011, 4, 1419–1425. [Google Scholar] [CrossRef] [PubMed]
  44. Bento, C.; Gonçalves, A.C.; Jesus, F.; Simões, M.; Silva, L.R. Phenolic compounds: Sources, properties and applications. In Bioactive Compounds: Sources, Properties and Applications; Porter, R., Parker, N., Eds.; Nova Science Publishers, Inc.: Hauppauge, NY, USA, 2017; pp. 271–299. [Google Scholar]
  45. Abotaleb, M.; Liskova, A.; Kubatka, P.; Büsselberg, D. Therapeutic potential of plant phenolic acids in the treatment of cancer. Biomolecules 2020, 10, 221. [Google Scholar] [CrossRef] [PubMed]
  46. Ko, E.B.; Jang, Y.G.; Kim, C.W.; Go, R.E.; Lee, H.K.; Choi, K.C. Gallic acid hinders lung cancer progression by inducing cell cycle arrest and apoptosis in A549 lung cancer cells via PI3K/Akt pathway. Biomol. Ther. 2022, 30, 151–161. [Google Scholar] [CrossRef]
  47. Rosendahl, A.H.; Perks, C.M.; Zeng, L.; Markkula, A.; Simonsson, M.; Rose, C.; Ingvar, C.; Holly, J.M.; Jernström, H. Caffeine and caffeic acid inhibit growth and modify estrogen receptor and insulin-like growth factor I receptor levels in human breast cancer. Clin. Cancer Res. 2015, 21, 1877–1887. [Google Scholar] [CrossRef]
  48. Kumar, P.; Ammani, K.; Mahammad, A.; Gosala, J. Vanillic acid induces oxidative stress and apoptosis in non-small cell lung cancer cell line. Int. J. Recent Sci. Res. 2013, 4, 1077–1083. [Google Scholar]
  49. Hmedat, A.N.; Morejón, M.C.; Rivera, D.G.; Pantelić, N.Đ.; Wessjohann, L.A.; Kaluđerović, G.N. In vitro anti-cancer studies of a small library of cyclic lipopeptides against the human cervix adenocarcinoma HeLa cells. J. Serbian Chem. Soc. 2024, 89, 471–484. [Google Scholar] [CrossRef]
  50. Rahmé, R. Assaying cell cycle status using flow cytometry. In Cell Cycle Checkpoints: Methods and Protocols; Humana: New York, NY, USA, 2021; pp. 165–179. [Google Scholar] [CrossRef]
  51. Lakshmanan, I.; Batra, S.K. Protocol for apoptosis assay by flow cytometry using annexin V staining method. Bio-Protocol 2013, 3, e374. [Google Scholar] [CrossRef]
  52. Gulcin, I.; Oktay, M.; Kirecci, E.; Kufrevioglu, I.O. Screening of antioxidant and antimicrobial activities of anise (Pimpinella anisum L.) seed extracts. Food Chem. 2003, 83, 371–382. [Google Scholar] [CrossRef]
  53. Zhang, T.; Ma, L.; Wu, P.; Li, W.; Li, T.; Gu, R.; Dan, X.; Li, Z.; Fan, X.; Xiao, Z. Gallic acid has anticancer activity and enhances the anticancer effects of cisplatin in non-small cell lung cancer A549 cells via the JAK/STAT3 signaling pathway. Oncol. Rep. 2019, 41, 1779–1788. [Google Scholar] [CrossRef]
  54. Jiang, Y.; Pei, J.; Zheng, Y.; Miao, Y.-J.; Duan, B.-Z.; Huang, L.-F. Gallic acid: A potential anti-cancer agent. Chin. J. Integr. Med. 2022, 28, 661–671. [Google Scholar] [CrossRef]
  55. Aborehab, N.M.; Osama, N. Effect of Gallic acid in potentiating chemotherapeutic effect of Paclitaxel in HeLa cervical cancer cells. Cancer Cell Int. 2019, 19, 154. [Google Scholar] [CrossRef]
  56. Tang, S.-M.; Deng, X.-T.; Zhou, J.; Li, Q.-P.; Ge, X.-X.; Miao, L. Pharmacological basis and new insights of quercetin action in respect to its anti-cancer effects. Biomed. Pharmacother. 2020, 121, 109604. [Google Scholar] [CrossRef]
  57. Reyes-Farias, M.; Carrasco-Pozo, C. The anti-cancer effect of quercetin: Molecular implications in cancer metabolism. Int. J. Mol. Sci. 2019, 20, 3177. [Google Scholar] [CrossRef] [PubMed]
  58. Hashemzaei, M.; Delarami Far, A.; Yari, A.; Heravi, R.E.; Tabrizian, K.; Taghdisi, S.M.; Sadegh, S.E.; Tsarouhas, K.; Kouretas, D.; Tzanakakis, G.; et al. Anticancer and apoptosis-inducing effects of quercetin in vitro and in vivo. Oncol. Rep. 2017, 38, 819–828. [Google Scholar] [CrossRef] [PubMed]
  59. Bhatiya, M.; Pathak, S.; Jothimani, G.; Duttaroy, A.K.; Banerjee, A. A comprehensive study on the anti-cancer effects of quercetin and its epigenetic modifications in arresting progression of colon cancer cell proliferation. Arch. Immunol. Ther. Exp. 2023, 71, 6. [Google Scholar] [CrossRef] [PubMed]
  60. Biswas, P.; Kaium, M.A.; Tareq, M.M.I.; Tauhida, S.J.; Hossain, M.R.; Siam, L.S.; Parvez, A.; Bibi, S.; Hasan, M.H.; Rahman, M.M.; et al. The experimental significance of isorhamnetin as an effective therapeutic option for cancer: A comprehensive analysis. Biomed. Pharmacother. 2024, 176, 116860. [Google Scholar] [CrossRef] [PubMed]
  61. Mei, C.; Liu, Y.; Lyu, X.; Jiang, Z.; Liu, Z.; Zhi, Y.; Xu, X.; Wang, H. Advances in Isorhamnetin Treatment of Malignant Tumors: Mechanisms and Applications. Nutrients 2025, 17, 1853. [Google Scholar] [CrossRef]
  62. Khaled, R. Biological activities of isorhamnetin: A review. Plantae Sci. 2020, 3, 78–81. [Google Scholar] [CrossRef]
  63. Park, C.; Cha, H.-J.; Choi, E.O.; Lee, H.; Hwang-Bo, H.; Ji, S.Y.; Kim, M.Y.; Kim, S.Y.; Hong, S.H.; Cheong, J.; et al. Isorhamnetin induces cell cycle arrest and apoptosis via reactive oxygen species-mediated AMP-activated protein kinase signaling pathway activation in human bladder cancer cells. Cancers 2019, 11, 1494. [Google Scholar] [CrossRef]
  64. Wang, X.; Yang, Y.; An, Y.; Fang, G. The mechanism of anticancer action and potential clinical use of kaempferol in the treatment of breast cancer. Biomed. Pharmacother. 2019, 117, 109086. [Google Scholar] [CrossRef] [PubMed]
  65. Imran, M.; Salehi, B.; Sharifi-Rad, J.; Gondal, T.A.; Saeed, F.; Imran, A.; Shahbaz, M.; Fokou, P.V.T.; Arshad, M.U.; Khan, H.; et al. Kaempferol: A key emphasis to its anticancer potential. Molecules 2019, 24, 2277. [Google Scholar] [CrossRef] [PubMed]
  66. Kaur, S.; Mendonca, P.; Soliman, K.F. The Anticancer Effects and Therapeutic Potential of Kaempferol in Triple-Negative Breast Cancer. Nutrients 2024, 16, 2392. [Google Scholar] [CrossRef]
  67. Alyami, N.M.; Alyami, H.M.; Almeer, R. Using green biosynthesized kaempferol-coated sliver nanoparticles to inhibit cancer cells growth: An in vitro study using hepatocellular carcinoma (HepG2). Cancer Nanotechnol. 2022, 13, 26. [Google Scholar] [CrossRef]
  68. Huminiecki, L.; Horbańczuk, J. The functional genomic studies of resveratrol in respect to its anti-cancer effects. Biotechnol. Adv. 2018, 36, 1699–1708. [Google Scholar] [CrossRef]
  69. Varoni, E.M.; Lo Faro, A.F.; Sharifi-Rad, J.; Iriti, M. Anticancer molecular mechanisms of resveratrol. Front. Nutr. 2016, 3, 8. [Google Scholar] [CrossRef]
  70. Öner, Ç.; Soyergin, D.; Özyurt, A.; Çolak, E. 4-Hydroxycoumarin effects on both cellular and genetic characteristics of hepatocellular carcinoma cells. Cytol. Genet. 2022, 56, 292–300. [Google Scholar] [CrossRef]
  71. Dimić, D.S.; Kaluđerović, G.N.; Avdović, E.H.; Milenković, D.A.; Živanović, M.N.; Potočňák, I.; Samoľová, E.; Dimitrijević, M.S.; Saso, L.; Marković, Z.S.; et al. Synthesis, Crystallographic, quantum chemical, antitumor, and molecular docking/dynamic studies of 4-hydroxycoumarin-neurotransmitter derivatives. Int. J. Mol. Sci. 2022, 23, 1001. [Google Scholar]
  72. Czarnecka, M.; Świtalska, M.; Wietrzyk, J.; Maciejewska, G.; Gliszczyńska, A. Synthesis, characterization, and in vitro cancer cell growth inhibition evaluation of novel phosphatidylcholines with anisic and veratric acids. Molecules 2018, 23, 2022. [Google Scholar] [CrossRef]
  73. Palko-Łabuz, A.; Gliszczyńska, A.; Skonieczna, M.; Poła, A.; Wesołowska, O.; Środa-Pomianek, K. Conjugation with phospholipids as a modification increasing anticancer activity of phenolic acids in metastatic melanoma—In vitro and in silico studies. Int. J. Mol. Sci. 2021, 22, 8397. [Google Scholar] [CrossRef]
  74. Kanimozhi, G.; Prasad, N. Anticancer effect of caffeic acid on human cervical cancer cells. In Coffee in Health and Disease Prevention; Elsevier: Amsterdam, The Netherlands, 2015; pp. 655–661. [Google Scholar]
  75. Espíndola, K.M.M.; Ferreira, R.G.; Narvaez, L.E.M.; Rosario, A.C.R.S.; da Silva, A.H.M.; Silva, A.G.B.; Vieira, A.P.O.; Monteiro, M.C. Chemical and pharmacological aspects of caffeic acid and its activity in hepatocarcinoma. Front. Oncol. 2019, 9, 541. [Google Scholar] [CrossRef] [PubMed]
  76. Alam, M.; Ahmed, S.; Elasbali, A.M.; Adnan, M.; Alam, S.; Hassan, I.; Pasupuleti, V.R. Therapeutic implications of caffeic acid in cancer and neurological diseases. Front. Oncol. 2022, 12, 860508. [Google Scholar] [CrossRef] [PubMed]
  77. Pelinson, L.P.; Assmann, C.E.; Palma, T.V.; da Cruz, I.; Pillat, M.M.; Mânica, A.; Stefanello, N.; Weis, G.C.C.; de Oliveira Alves, A.; De Andrade, C.M.; et al. Antiproliferative and apoptotic effects of caffeic acid on SK-Mel-28 human melanoma cancer cells. Mol. Biol. Rep. 2019, 46, 2085–2092. [Google Scholar] [CrossRef] [PubMed]
  78. Huang, S.; Wang, L.-L.; Xue, N.-N.; Li, C.; Guo, H.-H.; Ren, T.-K.; Zhan, Y.; Li, W.-B.; Zhang, J.; Chen, X.-G.; et al. Chlorogenic acid effectively treats cancers through induction of cancer cell differentiation. Theranostics 2019, 9, 6745. [Google Scholar] [CrossRef]
  79. Yamagata, K.; Izawa, Y.; Onodera, D.; Tagami, M. Chlorogenic acid regulates apoptosis and stem cell marker-related gene expression in A549 human lung cancer cells. Mol. Cell. Biochem. 2018, 441, 9–19. [Google Scholar] [CrossRef]
  80. Bender, O.; Atalay, A. Polyphenol chlorogenic acid, antioxidant profile, and breast cancer. In Cancer; Elsevier: Amsterdam, The Netherlands, 2021; pp. 311–321. [Google Scholar]
  81. Wang, L.; Pan, X.; Jiang, L.; Chu, Y.; Gao, S.; Jiang, X.; Zhang, Y.; Chen, Y.; Luo, S.; Peng, C. The biological activity mechanism of chlorogenic acid and its applications in food industry: A review. Front. Nutr. 2022, 9, 943911. [Google Scholar] [CrossRef]
  82. Mutlu, D. Cytotoxic activity, anti-migration and in silico study of o-coumaric acid on H1975 non-small cell lung cancer cells. Cumhur. Sci. J. 2025, 46, 35–40. [Google Scholar] [CrossRef]
  83. Mercado, Y.G.; Díaz, J.M.; Hernández, D.O.; Gonzalez, F.L.; Zaldivar, E.R.; Sapiens, M.H.; Pinedo, U.G.; Estrada, R.; Carballo, M.M.; Aguirre, A.C. Ortho-coumaric acid derivatives with therapeutic potential in a three-dimensional culture of the immortalised U-138 MG glioblastoma multiforme cell line. Neurol. Perspect. 2022, 2, S19–S30. [Google Scholar] [CrossRef]
  84. Burhanoğlu, T.; Halbutoğulları, Z.S.; Turhal, G.; Demiroglu-Zergeroglu, A. Evaluation of the anticancer effects of hydroxycinnamic acid isomers on breast cancer stem cells. Med. Oncol. 2025, 42, 73. [Google Scholar] [CrossRef]
  85. Cheng, Z.; Zhang, Z.; Han, Y.; Wang, J.; Wang, Y.; Chen, X.; Shao, Y.; Cheng, Y.; Zhou, W.; Lu, X.; et al. A review on anti-cancer effect of green tea catechins. J. Funct. Foods 2020, 74, 104172. [Google Scholar] [CrossRef]
  86. Sun, H.; Yin, M.; Hao, D.; Shen, Y. Anti-cancer activity of catechin against A549 lung carcinoma cells by induction of cyclin kinase inhibitor P21 and suppression of cyclin E1 and P–AKT. Appl. Sci. 2020, 10, 2065. [Google Scholar] [CrossRef]
  87. Silva, C.; Correia-Branco, A.; Andrade, N.; Ferreira, A.C.; Soares, M.L.; Sonveaux, P.; Stephenne, J.; Martel, F. Selective pro-apoptotic and antimigratory effects of polyphenol complex catechin: Lysine 1: 2 in breast, pancreatic and colorectal cancer cell lines. Eur. J. Pharmacol. 2019, 859, 172533. [Google Scholar] [CrossRef]
  88. Stabrauskiene, J.; Kopustinskiene, D.M.; Lazauskas, R.; Bernatoniene, J. Naringin and naringenin: Their mechanisms of action and the potential anticancer activities. Biomedicines 2022, 10, 1686. [Google Scholar] [CrossRef] [PubMed]
  89. Rauf, A.; Shariati, M.A.; Imran, M.; Bashir, K.; Khan, S.A.; Mitra, S.; Bin Emran, T.; Badalova, K.; Uddin, S.; Mubarak, M.S.; et al. Comprehensive review on naringenin and naringin polyphenols as a potent anticancer agent. Environ. Sci. Pollut. Res. 2022, 29, 31025–31041. [Google Scholar] [CrossRef] [PubMed]
  90. Choi, J.; Lee, D.-H.; Jang, H.; Park, S.-Y.; Seol, J.-W. Naringenin exerts anticancer effects by inducing tumor cell death and inhibiting angiogenesis in malignant melanoma. Int. J. Med. Sci. 2020, 17, 3049. [Google Scholar] [CrossRef]
  91. Venkidasamy, B.; Subramanian, U.; Almoallim, H.S.; Alharbi, S.A.; Lakshmikumar, R.R.C.; Thiruvengadam, M. Vanillic Acid Nanocomposite: Synthesis, Characterization Analysis, Antimicrobial, and Anticancer Potentials. Molecules 2024, 29, 3098. [Google Scholar] [CrossRef] [PubMed]
  92. Gong, J.; Zhou, S.; Yang, S. Vanillic acid suppresses HIF-1α expression via inhibition of mTOR/p70S6K/4E-BP1 and Raf/MEK/ERK pathways in human colon cancer HCT116 cells. Int. J. Mol. Sci. 2019, 20, 465. [Google Scholar] [CrossRef]
  93. Kaur, J.; Gulati, M.; Gowthamarajan, K.; Vishwas, S.; Chellappan, D.K.; Gupta, G.; Dua, K.; Pandey, N.K.; Kumar, B.; Singh, S.K. Combination therapy of vanillic acid and oxaliplatin co-loaded in polysaccharide based functionalized polymeric micelles could offer effective treatment for colon cancer: A hypothesis. Med. Hypotheses 2021, 156, 110679. [Google Scholar] [CrossRef]
  94. Velli, S.K.; Sundaram, J.; Murugan, M.; Balaraman, G.; Thiruvengadam, D. Protective effect of vanillic acid against benzo (a) pyrene induced lung cancer in Swiss albino mice. J. Biochem. Mol. Toxicol. 2019, 33, e22382. [Google Scholar] [CrossRef]
  95. Tian, Q.; Wang, L.; Sun, X.; Zeng, F.; Pan, Q.; Xue, M. Scopoletin exerts anticancer effects on human cervical cancer cell lines by triggering apoptosis, cell cycle arrest, inhibition of cell invasion and PI3K/AKT signalling pathway. J BUON 2019, 24, 997–1002. [Google Scholar] [PubMed]
  96. Yuan, C.; Wang, M.-H.; Wang, F.; Chen, P.-Y.; Ke, X.-G.; Yu, B.; Yang, Y.-F.; You, P.-T.; Wu, H.-Z. Network pharmacology and molecular docking reveal the mechanism of Scopoletin against non-small cell lung cancer. Life Sci. 2021, 270, 119105. [Google Scholar] [CrossRef] [PubMed]
  97. Shi, Z.; Chen, L.; Sun, J. Novel scopoletin derivatives kill cancer cells by inducing mitochondrial depolarization and apoptosis. Anti-Cancer Agents Med. Chem. (Former. Curr. Med. Chem.-Anti-Cancer Agents) 2021, 21, 1774–1782. [Google Scholar] [CrossRef] [PubMed]
  98. Liu, X.; Jiang, Q.; Liu, H.; Luo, S. Vitexin induces apoptosis through mitochondrial pathway and PI3K/Akt/mTOR signaling in human non-small cell lung cancer A549 cells. Biol. Res. 2019, 52, 7. [Google Scholar] [CrossRef]
  99. Wang, Q.; Zhang, J.; Ye, J.; Guo, J. Vitexin exerts anti-tumor and anti-angiogensis effects on cervical cancer through VEGFA/VEGFR2 pathway. Eur. J. Gynaecol. Oncol. 2022, 43, 86. [Google Scholar]
  100. Ghazy, E.; Taghi, H.S. The autophagy-inducing mechanisms of vitexin, cinobufacini, and physalis alkekengi hydroalcoholic extract against breast cancer in vitro and in vivo. J. Gastrointest. Cancer 2022, 53, 592–596. [Google Scholar] [CrossRef]
  101. Caparica, R.; Júlio, A.; Araújo, M.E.M.; Baby, A.R.; Fonte, P.; Costa, J.G.; Santos de Almeida, T. Anticancer activity of rutin and its combination with ionic liquids on renal cells. Biomolecules 2020, 10, 233. [Google Scholar] [CrossRef]
  102. Paudel, K.R.; Wadhwa, R.; Tew, X.N.; Lau, N.J.X.; Madheswaran, T.; Panneerselvam, J.; Zeeshan, F.; Kumar, P.; Gupta, G.; Anand, K.; et al. Rutin loaded liquid crystalline nanoparticles inhibit non-small cell lung cancer proliferation and migration in vitro. Life Sci. 2021, 276, 119436. [Google Scholar] [CrossRef]
  103. Kong, Y.; Sun, W.; Wu, P. Hyperoside exerts potent anticancer activity in skin cancer. Front. Biosci.-Landmark 2020, 25, 463–479. [Google Scholar]
  104. Qiu, J.; Zhang, T.; Zhu, X.; Yang, C.; Wang, Y.; Zhou, N.; Ju, B.; Zhou, T.; Deng, G.; Qiu, C. Hyperoside induces breast cancer cells apoptosis via ROS-mediated NF-κB signaling pathway. Int. J. Mol. Sciences 2019, 21, 131. [Google Scholar] [CrossRef]
  105. Hu, Z.; Zhao, P.; Xu, H. Hyperoside exhibits anticancer activity in non-small cell lung cancer cells with T790M mutations by upregulating FoxO1 via CCAT1. Oncol. Rep. 2020, 43, 617–624. [Google Scholar] [CrossRef]
  106. Hunke, M.; Martinez, W.; Kashyap, A.; Bokoskie, T.; Pattabiraman, M.; Chandra, S. Antineoplastic actions of cinnamic acids and their dimers in breast cancer cells: A comparative study. Anticancer. Res. 2018, 38, 4469–4474. [Google Scholar] [CrossRef]
  107. Yenigül, M.; Akçok, İ.; Gencer Akçok, E.B. Ethacrynic acid and cinnamic acid combination exhibits selective anticancer effects on K562 chronic myeloid leukemia cells. Mol. Biol. Rep. 2022, 49, 7521–7530. [Google Scholar] [CrossRef]
  108. Imai, M.; Yokoe, H.; Tsubuki, M.; Takahashi, N. Growth inhibition of human breast and prostate cancer cells by cinnamic acid derivatives and their mechanism of action. Biol. Pharm. Bull. 2019, 42, 1134–1139. [Google Scholar] [CrossRef] [PubMed]
  109. Zhang, R.; Yu, Q.; Lu, W.; Shen, J.; Zhou, D.; Wang, Y.; Gao, S.; Wang, Z. Grape seed procyanidin B2 promotes the autophagy and apoptosis in colorectal cancer cells via regulating PI3K/Akt signaling pathway. OncoTargets Ther. 2019, 12, 4109–4118. [Google Scholar] [CrossRef] [PubMed]
  110. Li, Y.; Lu, X.; Tian, P.; Wang, K.; Shi, J. Procyanidin B2 induces apoptosis and autophagy in gastric cancer cells by inhibiting Akt/mTOR signaling pathway. BMC Complement. Med. Ther. 2021, 21, 76. [Google Scholar] [CrossRef] [PubMed]
  111. Xue, H.-K.; Tan, J.-Q.; Li, Q.; Tang, J.-T. Procyanidin B2-induced apoptosis in MCF-7 cells and its mechanism. Food Sci. 2021, 42, 91–99. [Google Scholar]
Figure 1. Cytotoxic activity of L. nobilis leaves ethanolic extract, as determined by the MTT assay. Dose–response curves show a concentration-dependent decrease in cell viability. Data are presented as mean ± SD from three independent experiments.
Figure 1. Cytotoxic activity of L. nobilis leaves ethanolic extract, as determined by the MTT assay. Dose–response curves show a concentration-dependent decrease in cell viability. Data are presented as mean ± SD from three independent experiments.
Molecules 30 04012 g001
Figure 2. The extract induces apoptosis and cell cycle arrest in SAS and ES2 cells. Cells were treated with DMSO (CT) or the L. nobilis leaves ethanolic extract (EX) for 72 h. (A) Representative fluorescence images of different treatment groups. The x-axis represents Annexin V fluorescence intensity, while the y-axis indicates PI fluorescence intensity. (B) Numerical representation of Annexin +ve cells. (C) Cell cycle distribution of SAS and ES2 cells analyzed by flow cytometry following PI staining. The proportions of cells in G1, S, and G2/M phases are shown as percentages of the total cell population. Results in (B,C) are expressed as means and standard deviation taken from 3 independent experiments.
Figure 2. The extract induces apoptosis and cell cycle arrest in SAS and ES2 cells. Cells were treated with DMSO (CT) or the L. nobilis leaves ethanolic extract (EX) for 72 h. (A) Representative fluorescence images of different treatment groups. The x-axis represents Annexin V fluorescence intensity, while the y-axis indicates PI fluorescence intensity. (B) Numerical representation of Annexin +ve cells. (C) Cell cycle distribution of SAS and ES2 cells analyzed by flow cytometry following PI staining. The proportions of cells in G1, S, and G2/M phases are shown as percentages of the total cell population. Results in (B,C) are expressed as means and standard deviation taken from 3 independent experiments.
Molecules 30 04012 g002
Table 1. IC50 values of the L. nobilis extract and cisplatin against cancer cell lines and normal fibroblasts (µg/mL).
Table 1. IC50 values of the L. nobilis extract and cisplatin against cancer cell lines and normal fibroblasts (µg/mL).
Cell LineL. nobilis ExtractCisplatin
ES24.2 ± 0.23.6 ± 0.3
SAS3.8 ± 0.32.9 ± 0.5
HT-294.4 ± 0.610.7 ± 2.1
MDA-MB-23118.5 ± 0.816.7 ± 1.1
HDF>100>100
Table 2. DPPH scavenging activity of L. nobilis leaves ethanolic extract.
Table 2. DPPH scavenging activity of L. nobilis leaves ethanolic extract.
Concentration (µg/mL)DPPH Scavenging Activity %
254.2 ± 0.002
508.9 ± 0.001
10015.7 ± 0.0006
20027.2 ± 0.002
50041.9 ± 0.002
100055.5 ± 0.0006
200077.5 ± 0.0006
Table 3. Phenolic compounds and their phytochemical class identification in L. nobilis leaves ethanolic extract using the LC-MS/MS.
Table 3. Phenolic compounds and their phytochemical class identification in L. nobilis leaves ethanolic extract using the LC-MS/MS.
NRT (min)Identified Compound NamePhytochemical ClassChemical FormulaMolecular Weight (Da)m/zIon Mode
13.39Gallic acidPhenolic acidC7H6O5170.0216171.0289[M+H]+
25.29QuercetinFlavonoidC15H10O7302.04226303.04954[M+H]+
35.35IsorhamnetinFlavonoidC16H12O7316.05796317.06524[M+H]+
45.8KaempferolFlavonoidC15H10O6286.04752287.0548[M+H]+
56.24ResveratrolStilbenoidC14H12O3228.07855229.08582[M+H]+
611.154-hydroxycoumarinPhenolic acidC9H6O3162.0317163.03894[M+H]+
71.52Anisic acidPhenolic acidC8H8O3152.0474153.05472[M+H]+
81.89GallocatechinFlavonoidC15H14O7306.07196307.07923[M+H]+
93.07Caffeic AcidPhenolic acidC9H8O4180.0399181.04718[M+H]+
103.17Chlorogenic acidPhenolic acidC16H18O9354.09439355.10166[M+H]+
113.98o-Coumaric acidPhenolic acidC9H8O3164.04732165.0546[M+H]+
124.24CatechinFlavonoidC15H14O6290.07941291.08652[M+H]+
134.25NaringeninFlavonoidC15H12O5272.06789273.07516[M+H]+
144.51Vanillic acidPhenolic acidC8H8O4168.04219169.04947[M+H]+
154.74ScopoletinCoumarinC10H8O4192.04185193.04913[M+H]+
164.82VitexinFlavonoidC21H20O10432.10515433.11243[M+H]+
174.91RutinFlavonoidC27H30O16610.15234611.1598[M+H]+
184.96HyperosideFlavonoidC21H20O12464.0949465.10217[M+H]+
195.28SaponarinFlavonoidC27H30O15594.1577595.16512[M+H]+
206.51Cinnamic acidPhenolic acidC9H8O2148.05233149.05961[M+H]+
213.82ProcyanidinB2FlavonoidC30H26O12578.14289577.13561[M−H]
The identified chemicals were compared with some existing literature on anticancer mechanisms. The findings are elaborated in Supplementary Table S1.
Table 4. Summary of chromatographic conditions.
Table 4. Summary of chromatographic conditions.
UPLC
Conditions
Injection VolumeFlow RateAutosampler TemperatureColumn Oven TemperatureTotal Run Time
3 µL0.51 mL/min8 °C40 °C35 min.
ChromatographyMobile phase
Solvents:
(A) Water with 0.1% methanol

(B) Methanol
0–3 min, 5% B;
3–23 min, 60% B;
23–28 min, 95% B;
28–31 min, 95% B;
31.0–31.1 min, 5% B;
31.1–35 min 5% B;
Column typeBruker Daltonik (Berlin, Germany) C-18 column (100 × 2.1 mm × 2 µm) (120 Aº)
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Al-Mammori, F.; Qasem, A.M.A.; Al-Tawalbeh, D.; Abuarqoub, D.; Hmedat, A. The Therapeutic Potential of Laurus nobilis L. Leaves Ethanolic Extract in Cancer Therapy. Molecules 2025, 30, 4012. https://doi.org/10.3390/molecules30194012

AMA Style

Al-Mammori F, Qasem AMA, Al-Tawalbeh D, Abuarqoub D, Hmedat A. The Therapeutic Potential of Laurus nobilis L. Leaves Ethanolic Extract in Cancer Therapy. Molecules. 2025; 30(19):4012. https://doi.org/10.3390/molecules30194012

Chicago/Turabian Style

Al-Mammori, Farah, Ashraf M. A. Qasem, Deniz Al-Tawalbeh, Duaa Abuarqoub, and Ali Hmedat. 2025. "The Therapeutic Potential of Laurus nobilis L. Leaves Ethanolic Extract in Cancer Therapy" Molecules 30, no. 19: 4012. https://doi.org/10.3390/molecules30194012

APA Style

Al-Mammori, F., Qasem, A. M. A., Al-Tawalbeh, D., Abuarqoub, D., & Hmedat, A. (2025). The Therapeutic Potential of Laurus nobilis L. Leaves Ethanolic Extract in Cancer Therapy. Molecules, 30(19), 4012. https://doi.org/10.3390/molecules30194012

Article Metrics

Back to TopTop