ABSTRACT
Monoterpenes like thymol and carvacrol are recognized for their anti‐inflammatory and anticancer properties, predominantly found in the Lamiaceae family, particularly in Thymus species, but also present in Verbenaceae, Scrophulariaceae, Ranunculaceae, and Apiaceae families. This review explores their anticancer potential, molecular mechanisms, and synergism with other cancer therapies. The data was collected by using several keywords and MeSH terms, and data from Google Scholar, PubMed, Scopus, and Web of Science indicate that thymol and carvacrol modulate key signaling pathways, including MAPK/ERK, PI3K/AKT, Wnt/β‐catenin, JAK/STAT, HH/GLI, and NF‐κB. They upregulate pro‐apoptotic genes (Bax, Bak, Bid, p53, and SIVA) while downregulating anti‐apoptotic genes (Bcl‐2, Bcl‐xL, XIAP, and cIAP1), leading to apoptosis and cell cycle arrest at G0/G1 and G2/M phases. Thymol derivatives, such as 1,2,3‐triazoles and carvacrol, effectively target breast cancer (BC) through PI3K/AKT/mTOR and NOTCH pathways and inhibit PIK3CA expression. In lung cancer (LC), they act as SphK1 inhibitors in NSCLC H1299 and A549 cell lines. Additionally, thymol exhibits anti‐EGFR activity, while carvacrol modulates the HIF‐1α/VEGF pathway, making them potential candidates for colorectal cancer (CRC) management. In vitro studies confirm their efficacy against multiple cancer cell lines (MCF‐7, HT‐29, HeLa, PC‐3, HepG2, HL‐60), while in vivo animal models highlight their antiproliferative and antitumor effects. Their synergistic potential with chemotherapy, radiotherapy, and other bioactive compounds strengthens their therapeutic promise. However, challenges such as stability, bioavailability, and the need for clinical trials hinder their clinical application. This review is the first to comprehensively report thymol and carvacrol in a single study, offering new insights into their anticancer potential.
Keywords: anticancer, apoptosis, carvacrol, cell cycle arrest, PI3K/AKT, thymol, Wnt/ß‐catenin
Monoterpenes like thymol and carvacrol are recognized for their anti‐inflammatory and anticancer properties, predominantly found in the Lamiaceae family, particularly in Thymus species, but also present in Verbenaceae, Scrophulariaceae, Ranunculaceae, and Apiaceae families. This review explores their anticancer potential, molecular mechanisms, and synergism with other cancer therapies.

Abbreviations
- ↑
upward arrow
- ↓
downward arrow
- ADHD
attention deficit hyperactivity disorder
- ATG5
autophagy‐related protein 5
- CDK4
cyclin‐dependent kinase 4
- CDK6
cyclin‐dependent kinase 6
- CDKs
cyclin‐dependent kinases
- CIC
circulating immune complex
- CIMP
CpG island methylator phenotype
- CIN
chromosomal instability
- COX‐2
cyclooxygenase‐2
- CPK
creatine phosphokinase
- EMT
epithelial‐mesenchymal transition
- ESR
erythrocyte sedimentation rate
- HH/GLI
hedgehog/GLI
- HIF‐1α/VEGF
hypoxia‐inducible factor 1, alpha/vascular endothelial growth factor
- IGF‐1
insulin‐like growth factor 1
- JAK/STAT
janus kinase‐signal transducer/activator of transcription
- JNK
c‐Jun N‐terminal kinase
- LDH
lactate dehydrogenase
- MAPK/ERK
mitogen‐activated protein kinase/extracellular signal‐regulated kinase
- MDA
malondialdehyde
- MeSH
medical subject headings
- MSI
microsatellite instability
- mTOR
mechanistic target of rapamycin
- NF‐κB
nuclear factor kappa B
- NOTCH
neurogenic locus notch homolog protein
- Nrf2/HO‐1
nuclear factor erythroid 2‐related factor 2/transcription factor/heme oxygenase‐1
- PI3K/AKT
phosphatidylinositol 3‐kinase/Protein kinase B
- STAT3
signal transducer and activator of transcription 3
- TGF‐β
transforming growth factor‐beta
- TRPM7
transient receptor potential cation channel subfamily M member 7
- Wnt/β‐catenin
wingless‐related integration site/catenin beta‐1
1. Introduction
Cancer is the leading cause of morbidities and fatalities, affecting ~20 million people in 2022. The overproduction or imbalance of reactive oxygen species (ROS), which are highly unstable and lead to oxidative stress (OS), inflammation, DNA damage, and genetic and epigenetic alterations, which are major risk factors for cancer development. The activation of pro‐inflammatory markers (IL‐6, IL‐1β, TNF‐α, MDA), suppression of tumor suppressor genes (TSGs) like p53, APC, CDKN2A, BRCA1, and BRCA2, and activation of oncogenes (ERBB2, BCR/ABL1, K‐Ras, PIK3CA, NMYC) are significant events in oncogenesis (Greten and Grivennikov 2019). Medicinal herbs and bioactive compounds are highly admired worldwide due to their pharmacological properties, such as anti‐inflammation and anticancer, and in the last few decades, their demand has increased (Chopra et al. 2022). The anticancer effects of these herbs are due to versatile bioactive compounds, that is, terpenes, anthocyanins, carotenoids, saponins, and alkaloids (Dubey et al. 2022).
Thymol, also known as 2‐isopropyl‐5‐methylphenol or 5‐methyl‐2‐isopropylphenol (C10H14O), is a monoterpene, colorless, crystalline, with a distinctive odor, and the active ingredient of T. serpyllum oil. Besides T. serpyllum , it is also present in other varieties of the genus thyme, like T. vulgaris , T. zygis, T. praecox , and oregano of the genus Origanum. Thymol is highly soluble in organic solvents but slightly soluble in water at neutral pH (Escobar et al. 2020). Medicinal properties of thymol, such as antioxidant (Yildiz et al. 2021), anti‐inflammatory (Islam et al. 2024), anticancer (Taibi et al. 2024), antidiabetic (Sachan et al. 2022), and hepato‐renal protection (Jamshidi and Taheri 2021; Özmen et al. 2021), have been reported.
Carvacrol, 2‐Methyl‐5‐(propane‐2‐yl) phenol, an isomer of thymol, a monoterpene also referred to as cymophenol with the chemical formula (C6H3(CH3) (OH)C3H7). It is insoluble in water while highly soluble in ethanol, CCl4, and diethyl ether. Major sources of carvacrol are Lavandula multifida, Nigella sativa, Origanum vulgare, Lippia graveolens, and Thymus glandulosus (Imran et al. 2022). The biological properties of carvacrol include antioxidant (Ridaoui et al. 2024), anti‐inflammatory (De Souza et al. 2023), anticancer (Abed et al. 2024), antidiabetes (Hoca et al. 2023), antihypertension (Khazdair et al. 2024), cardioprotective (Joshi et al. 2023), neuroprotective (Forqani et al. 2023) and hepato‐nephroprotective (Cerrah et al. 2023). The chemical structures of both compounds, thymol (a) and carvacrol (b) are illustrated in Figure 1.
FIGURE 1.

Chemical structures of Thymol (a) and carvacrol (b).
The current review covers several aspects of thymol, carvacrol, and cancer management. The review focuses on the anticancer potential of thymol and carvacrol and their possible mechanisms. This review also illuminates mechanisms of anticancer activity like apoptosis induction, cell cycle arrest, and molecular pathways. Moreover, the detailed in vitro and in vivo studies and the synergetic role of thymol and carvacrol with other bioactive compounds, chemotherapeutic drugs, and radiotherapy are the limelight of this review. Lastly, the limitations and challenges, such as the bioavailability of both compounds and strategies to enhance therapeutic activities and clinical trials, are also significant features of this review.
2. Search Strategy
The methodology segment was designed to review the updated anticancer properties of carvacrol and thymol (Lo 2022). Relevant peer‐reviewed articles were retrieved from databases and search engines such as Google Scholar, PubMed, Scopus, and Web of Science, along with keywords and MeSH terms like “Thymol” and “Carvacrol” (e.g., “Thymol and Carvacrol,” “Monoterpenes”), “Anticancer Effects” (e.g., “Antineoplastic Molecules and Agents,” “Cancer Therapy”) and definite cancer types modalities. Moreover, Boolean operators such as AND and OR were used to enhance the search. Inclusion criteria: original research articles inspecting carvacrol and thymol's effects on cancer cells or murine cancer models, studies that explicated molecular mechanisms of carvacrol and thymol's action, and clinical trials evaluating carvacrol and thymol's efficiency. Exclusion criteria: non‐English articles, studies using homeopathic substances, and research including other compounds besides thymol and carvacrol. Data mining focused on the study model, type of cancer, thymol and carvacrol dosage and route of administration, results on cancer cell propagation, apoptosis, cell cycle arrest, and effects on main signaling pathways. The findings were systematically analyzed to evaluate the therapeutic potential of carvacrol and thymol in cancer management. The PRISMA flow diagram of the literature search process is shown in Figure 2.
FIGURE 2.

PRISMA flow diagram of the literature search process.
As shown in the PRISMA flow diagram (Figure 2), 377 studies were found. After removing duplicates and excluding irrelevant papers based on titles and abstracts, 277 articles were retained for full‐text review. Forty articles were excluded after title and abstract assessment, and 50 were excluded after full‐text review, with the reasons for their removal detailed in the PRISMA flow diagram. Finally, 187 studies were included in the review.
3. Mechanisms of Anticancer Activity
Studies have demonstrated that anticancer agents act on multiple targets, and their anticancer activity includes several mechanisms of action, such as apoptosis induction, cell cycle arrest, modulation of OS, anti‐metastasis, inhibition of angiogenesis, immune‐modulatory impact, modulation of different pathways, and enzymatic activity (Tang et al. 2022).
3.1. Induction of Apoptosis
Apoptosis is a critical factor in programmed cell death, vital for the developmental process and maintaining cellular homeostasis. It is a Greek word that refers to the fall of the leaves during autumn, achieved through external and internal factors (Singh et al. 2022). Studies on thymol have demonstrated its potential role in apoptosis through 2D and 3D techniques. Concerning this, a 2D and 3D (spheroids) murine mammary tumor cancer model, including monolayer 4 T1 cells and L929 as a control, was used to investigate thymol‐rich essential oil apoptotic activity. The results showed that the 4 T1 tumor line was inhibited without substantially affecting normal L929 cells. The apoptosis was due to ROS production, variations in the mitochondrial membrane, caspase‐3 activation, and DNA damage (Jamali et al. 2020). Thymol derivatives showed better antiproliferative activity than common chemotherapeutic drugs like 5‐fluorouracil against BC cells (MCF‐7 and MDA‐MB‐231). Thymol derivative, known as compound 10 (IC50 6.17 μM) exhibited 3.2‐fold more inhibition than 5‐fluorouracil (IC50 20.09 μM) against MCF‐7 cell lines, while (IC50 10.52 μM) of compound 10 proved 1.42 and 2.4 times better than tamoxifen (IC50 15.01 μM) and 5‐fluorouracil (IC50 25.31 μM) respectively, in MDA‐MB‐231 cell lines (Alam et al. 2021). Conobea scoparioides essential oil (EO) containing thymol methyl ether (62%) and thymol (16%) proved effective in in vitro and in vivo studies. The 40 and 80 mg/kg EO reduced tumor mass to 36.7% and 55.8%, respectively, whereas in vitro analysis showed that EO induced apoptosis in HepG2, HCT116, and MCF‐7 cell lines (de Lima et al. 2020).
The association between Hedgehog/GLI (HH/GLI) signaling pathway and oncogenesis has been established in studies. This pathway is involved in cell growth and stem cell maintenance, primarily active in the embryonic stage, wound healing, and tissue repair. The irregularities in this pathway result in brain, lung, pancreas, cervical, and prostate cancer development (Trocchianesi et al. 2023). Carvacrol has been reported to have the potential to induce apoptosis and inhibit cell multiplication through modulation of the HH/GLI pathway in C33A cervical cancer (CC) cells. Carvacrol (25, 50, 75, and 90 μM) enhanced the expression of Bax, Bad, Fas‐L, and cytochrome c, activated caspase‐9/3 and caspase‐8, induced cell cycle at G0/G1, improved the expression of proteins (p21, cyclin D1, CDK4), and downregulated the SMO and GLI1 proteins expression in CC (Ahmad et al. 2023). The p53 gene regulates the transcription of Bcl‐2 and Bax genes and modulates Bax/Bcl‐2 expression. However, microRNA‐21 (miR‐21) and TTDA protein inhibit the expression of p53, resulting in alterations in Bcl‐2/Bax expression (Luo et al. 2020). Carvacrol (305 μM) induced apoptosis in MCF‐7 bc cell lines via modulating p53/Bax/Bcl‐2 axis. The findings demonstrated that carvacrol improved the expression of p53/Bax, while downregulated Bcl‐2 (Moradipour et al. 2022). Similarly, carvacrol (0–250 μmol/L) initiates apoptosis and cell cycle arrest against MCF‐7 cancer cell lines by modulating the PI3K/AKT pathway. The results revealed that carvacrol (200 μmol/L) at 24 and 48 h significantly inhibited cell proliferation, triggered apoptosis by inhibiting p‐Rb, CDK4, CDK6, and cyclin D1, decreased Bcl‐2, and improved Bax expressions (Mari et al. 2021).
3.2. Cell Cycle Arrest
Cancer is characterized by dysregulation and abnormality in the cell cycle regulation. The cell cycle is divided into two phases: the interphase and the mitosis (M) phase, involving the division of cell components and conversion of the cell into two identical daughter cells (Matthews et al. 2022). The disrupted cell cycle leads to cell proliferation and cancer, and irregular cell cycle checkpoints increase the risk of genomic instability. Studies on thymol have reported its role in cell cycle arrest at different stages through several mechanisms. Balan et al. (2021) reported that thymol (112 μg/mL) caused cell cycle arrest at the G0/G1 phase and triggered apoptosis in A549 cells. They concluded that upregulation of Bax, suppression of Bcl‐2 expression, increased ROS production, elevated lipid peroxides, and mitochondrial membrane depolarization are substantial mechanisms responsible for cell death. Thymol extracted from Thymus vulgaris exhibited in vivo and in vitro anticancer potential against HCT116 and Lovo cell lines. Thymol (75 and 150 mg/kg) reduced tumor volume, induced cell death, and modulated BAX/Bcl‐2 and Wnt/β‐catenin pathways in an in vivo analysis. In vitro study showed that thymol (0, 10, 20, 40, 80, and 120 μg/mL) enhanced apoptosis and induced cell cycle arrest in cell lines (Zeng et al. 2020). Current studies have confirmed that thymol can improve the potential of chemotherapeutic drugs to obstruct cell proliferation and cancer progression. In an in vitro study, the synergetic impact of thymol on 5‐Fluorouracil (5‐FU) against KYSE‐30 esophageal cancer (EC) cells has been reported. Thymol enhanced the cytotoxicity of 5‐FU via augmented ROS generation, improved p53 and Bax expression, decreased Bcl‐2 expression, and declined MMP‐2 activity. Moreover, combined therapy induced cell cycle arrest at the G2/M phase (Pouyamanesh et al. 2024). Thymol induced cell cycle arrest at the G0/G1 phase, enhanced ROS production, caused apoptosis, and activated caspase‐9/3 in CRC cell lines (Anvarbatcha et al. 2023).
Carvacrol is a less studied monoterpene compared to thymol concerning cell cycle arrest. Previous studies have reported its impact and potential in cell cycle arrest. Notch signaling is closely linked with prostate cancer (PC), and carvacrol has been proven effective in inhibiting PC progression. Carvacrol induced apoptosis and cell cycle arrest at the G0/G1 phase via ROS production, BAX/Bcl‐2 expression modulation, and cyclin D1 and CDK4 inhibition. In addition, carvacrol modulated Notch signaling in PC by downregulating Notch‐1 and Jagged‐1 expression (Khan et al. 2019). Drug‐resistant cancer cells could be the major problem in cancer treatment. Drug resistance occurs due to molecular changes in cancerous cells; moreover, genetic mutations in cancer cells that affect genes targeted by drugs and genetic modifications in genes that are responsible for repairing DNA damage are other aspects that are leading to drug resistance (Garg et al. 2024). Carvacrol induced apoptosis in doxorubicin‐resistant A549 lung cancer cells, proved by higher expression of Bax, cytochrome c, and caspase 3/9. Moreover, cell cycle arrest was noticed due to reduced CDK2, CDK4, cyclin D1, and enhanced p21 protein expression and declined autophagy marker ATG5 (Khan et al. 2018). In another study, carvacrol induced cell cycle arrest at the G0/G1 phase by ROS production against DU145 PC cell lines, which were reported, confirmed by caspase‐3 activation and mitochondrial membrane potential disruption (Khan et al. 2017).
4. Modulation of Signaling Pathways
The MAPK/ERK pathway is a crucial signaling pathway involved in cell division and differentiation. An extracellular stimulus, such as growth factors, binds to receptors on the cell surface, thus triggering the pathway, and dysregulation of this pathway promotes cancer development (Hendrikse et al. 2023). The PI3K/AKT signaling pathway is vital in cell proliferation, and its impairment is commonly observed in several cancers, making it a substantial target for anticancer remedies (Glaviano et al. 2023). Like others, Wnt/β‐catenin, JAK/STAT, and NF‐κB pathways are other significant signaling pathways in cell multiplication, inflammatory response, and cytokine signal transmission (Xue et al. 2023; Zhang et al. 2021).
The studies on thymol's anticancer potential showed that it can modulate all these pathways. Thymol can hinder the phosphorylation of vital components in the pathway (MEK1/2 and ERK1/2), leading to decreased cell multiplication and augmented apoptosis across different cancers. Moreover, thymol also inhibits MPPs and other invasion proteins, thus contributing to the modulation of the MAPK/ERK pathway (Guo et al. 2020). Multiple concentrations of thymol (0, 25, 50, 100, and 150 μM) were evaluated against bladder cancer cell lines (T24 and J82). It was found that thymol induced cell cycle arrest at the G2/M phase via caspase‐3/9 stimulation, downregulating Bcl‐2 family proteins and ROS production. Moreover, thymol activated JNK and p38 MAPK while impeding the ERK pathway, indicating that JNK and p38 are critical factors for apoptosis (Li et al. 2017). Thymol (50, 100 mg/kg) remarkably protected against pulmonary fibrosis by regulating PI3K/AKT signaling in mice via reduced TNF‐α, IL‐6, NF‐kB, and TGF‐β expression (Hussein et al. 2023). Thymol reduced cell proliferation and augmented apoptosis in lung carcinoma by reducing AKT phosphorylation, thus exerting an anticancer effect via PI3K/AKT pathway modulation (Sampaio et al. 2021). Another study reported that thymol decreased HT29 cell migration, inhibited MMP‐2/9 activity, and downregulated PI3K/AKT and ERK pathways (Lv and Chen 2017). Khan et al. (2024) stated that thymol inhibited STAT3 phosphorylation and downregulated JAK/STAT signaling, thus showing antiproliferative properties. The STAT3 is interlinked with HIF‐1α, as both are transcription factors and can cooperate to regulate gene expression, especially in response to hypoxia. STAT3 can increase the stability and activity of HIF‐1α, leading to augmented expression of HIF‐1α target genes and angiogenesis (Dinarello et al. 2023). Thymol (30 mg/kg/day), along with thymoquinone (10 mg/kg/day), alleviated MSG‐induced ADHD in rats via modulating Wnt/β‐Catenin, Nrf2/HO‐1, and TLR4/NF‐κB/NLRP3/caspase‐1 pathways (Abu‐Elfotuh et al. 2022).
Besides thymol, carvacrol has also been reported to inhibit MAPK or ERK pathways in previous studies. Carvacrol (25, 50, and 100 mg/kg) for 6 weeks modulated the MAPK signaling pathway and inhibited TRPM7 expression in liver fibrotic C57BL/6J mice (Cai et al. 2021). Carvacrol (25, 50 and 100 mg/kg) modulated the ERK pathway, upregulated the AKT/eNOS pathway while imposing no impact on p38MAPK and JNK in myocardial ischemia/reperfusion injury, proving its cardioprotective potential (Chen et al. 2017). The PI3K/AKT is connected with TRPM7, a channel kinase that activates the PI3K/AKT pathway. Carvacrol (500 μM) inhibited cell proliferation in PC‐3 and DU145 PC cancer cells by inhibiting MMP‐2 and TRPM7 channels and suppressing PI3K/AKT signaling pathways (Luo et al. 2016). Carvacrol (1, 3, and 10 mg/kg body weight) for 48 h ameliorated cisplatin‐induced renal injury in BALB/cN mice. The study demonstrated that carvacrol alleviated renal injury by modulating p‐NF‐κB, PI3K/AKT, and ERK pathways (Potočnjak and Domitrović 2016). Carvacrol (15 mg/kg) combined with sorafenib modulated HIF‐1α/STAT3/FGL1 pathway in a hepatocellular carcinoma (HCC) murine model. The findings showed that carvacrol increased CD8+ T cells and reduced animal fibrinogen‐like protein 1 (FGL1) expression. In a xenograft animal model, carvacrol proved effective against osteosarcoma by regulating the Wnt/β‐catenin pathway. Moreover, the study revealed that carvacrol treatment enhanced Bax while decreasing Bcl‐2 and MMP‐9 expression (Zhang et al. 2022). The modulating impact of thymol and carvacrol on different cancer pathways is illustrated in Figure 3.
FIGURE 3.

Modulation of MAPK/ERK, PI3K/AKT, Wnt/β‐catenin, and JAK/STAT by thymol and carvacrol via cell cycle arrest, apoptosis induction, inhibited phosphorylation of MEK1/2, ERK1/2, and STAT3, caspase‐3/9, JNK, and p38 MAPK activation, downregulation of Bcl‐2 proteins, reducing TNF‐α, IL‐1β, IL‐6, and TGF‐β expression, regulation of Nrf2/HO‐1, TLR4/NF‐κB/NLRP3/caspase‐1, and HIF‐1α/STAT3/FGL1 pathways, upregulation of AKT/eNOS pathway, and inhibition of MMP‐2 and TRPM7.
5. Anticancer Perspective of Thymol and Carvacrol
Cancer is affecting millions of people worldwide; however, numbers are increasing in middle‐ and low‐income countries due to poor and unhygienic dietary choices, polluted environmental conditions, and prevailing pathogenic diseases. Thymol and carvacrol, monoterpenes from essential oils of herbs, exhibit anticancer potential through apoptosis induction, OS reduction, tumor growth inhibition, and modulation of TSGs and oncogenes. The anticancer potential of thymol and carvacrol and possible mechanisms are mentioned in Table 1.
TABLE 1.
Anticancer studies of thymol and carvacrol through possible mechanisms.
| Compound | Cancer | Study | Mechanism/outcome | References |
|---|---|---|---|---|
| Thymol | Colorectal cancer | In vitro | ↑Apoptosis, ↓EGFR genes expression | Keshavarz et al. (2024) |
| ↑ROS, cell cycle arrest at G0/G1 phase, ↑ p53 expression, ↓ Bcl‐xL | Anvarbatcha et al. (2023) | |||
| Breast cancer | Network pharmacology | Modulated PIK3CA, ↓proliferation | Laamari et al. (2025) | |
| In vitro | ↓COX‐2 and cell proliferation, ↑apoptosis | Taibi et al. (2024) | ||
| Lung cancer | In vitro | ↑ROS, ↑SphK1 | Shakeel et al. (2024) | |
| ↑Apoptosis | dos Santos et al. (2021) | |||
| Liver cancer | In vitro | ↓VEGFR and VEGF genes expression | Hussein (2024) | |
| Molecular docking | Bind and inhibit P38 Protein | Tabassum and Ahmad (2020) | ||
| Esophageal cancer | In vitro | ↑ROS, p53 and Bax, ↓ Bcl‐2 and MMP‐2, cell cycle arrest at G2/M phase | Pouyamanesh et al. (2024) | |
| Ovarian cancer | In vitro | ↓ Bcl‐2, ↑caspase‐3/8/9 and Bax | Seçme and İlhan (2025) | |
| Apoptosis induction | Elbe, Yigitturk, Cavusoglu, Baygar, et al. (2020) | |||
| Bladder cancer | In vitro | Cell cycle arrest at the G2/M phase, ↓ Bcl‐2, caspase‐3/9 activation | Li et al. (2017) | |
| Prostate cancer | In vitro | Induced apoptosis, ↓cell viability | Yeh et al. (2017) | |
| Induced apoptosis, caspase‐3 activation | Singhal et al. (2021) | |||
| Cervical cancer | In vitro | ↑Apoptosis | Osarieme Imade et al. (2023) | |
| Skin cancer | In vitro | Modulation of VEGF and VEGFR genes expression | Feyzmohamadi Khoramabadi et al. (2024) | |
| Carvacrol | Colorectal cancer | In vitro | ↓Cell proliferation and migration | Abed et al. (2024) |
| Breast cancer | In vitro | ↓ Jagged‐1 and cyclin D1, modulated NOTCH pathway | Pandey et al. (2024) | |
| Breast cancer | In vitro and in vivo | Modulated PI3K/AKT/mTOR, ↑OS and PTEN expression | Srinivasan and Namasivayam (2024) | |
| In vitro | Cell cycle arrest at S and G2/M phase, ↓ Bcl‐2 and PI3K expression | Chen et al. (2024) | ||
| Cell cycle arrest at G2/M phase, apoptosis induction | McClements (2024) | |||
| Lung cancer | In vitro | Cell cycle arrest at G2/M phase and apoptosis induction | Bansal et al. (2022) | |
| In vitro and molecular docking | Modulated EGFR and BRAF proteins | Çakır et al. (2025) | ||
| Gastric cancer | In vitro | ↓ Bcl‐2, ↑ Bax | Elahi et al. (2022) | |
| Prostate cancer | In vitro | ↑JNK and p53 proteins | Razack et al. (2025) | |
| Cervical cancer | In vitro | Cell cycle arrest at G0/G1 phase, ↓ E6 and E7 oncogenes, ↑ p53expression | Ahmad et al. (2025) | |
| ↓Cell proliferation | Akhlaq et al. (2023) | |||
| Liver cancer | In vitro | Improved Topotecan cytotoxicity | Bayoumi et al. (2021) | |
| Pancreatic Cancer | In vitro | Induced apoptosis, ↓CDH1, TIMP1, TIMP3, and ZEB1expression | Gunes et al. (2022) | |
| Plasma cell cancer (Myeloma) | In vitro | ↓MMP level, Bcl‐2, ↑ Bax, caspase‐3/9, ROS, apoptosis | Zhang et al. (2023) | |
| Skin cancer | In vitro | Induced apoptosis, ↓metastasis | Osanloo et al. (2023) |
5.1. Breast Cancer
Breast cancer (BC) is the 2nd leading cancer among different cancers and ranked 1st among women concerning prevalence worldwide, and according to WHO, ~2.3 million women were identified with BC in 2022 (Nardin et al. 2020). Hormonal replacement therapy (HRT) is a key risk factor for BC. However, risk can be limited if HRT is used after 60 years (Smolarz et al. 2022). Genetic mutations are only counted in 5 to 10% of cases of BC development, and BRCA1 and BRCA2 are TSGs positioned on chromosomes 17 and 13, respectively, involved in genomic stability, encrypting nuclear protein, and repairing double DNA strand breaks. Some other genes like CHEK2, ATM, PALB2, and BRIP1 show a modest tendency to BC. However, patients with these gene mutations have 2 to 3 times greater risk of developing malignant tumors (Chamseddine et al. 2022).
Phosphatidylinositol 3‐kinase (PI3K) is a key factor responsible for cell growth, proliferation, and motility and works with mTOR and AKT to regulate all these cellular mechanisms. The dysregulation of this PI3K/AKT/mTOR pathway is involved in multiple cancer developments, including BC (Bertucci et al. 2023). However, thymol derivative 1,2,3‐triazoles have been proven to be a potent anticancer agent. In this context, Laamari et al. (2025) investigated 1,2,3‐triazoles against breast cancer by targeting PIK3CA and concluded that 1,2,3‐triazoles modulate PIK3CA to regulate cell growth and proliferation. Thymol (0–1000 μM) concentrations from N. sativa were evaluated against MCF‐7 bc cells, and it has been found that thymol substantially inhibited 50% of cell growth at 200 μM concentration. Moreover, it downregulated cyclin D1 and PCNA gene expression to inhibit cell invasion (Vahitha et al. 2024). Recent novel strategies such as nanoparticles (NPs) have emerged as promising anticancer therapies due to their effectiveness in releasing drugs/compounds at the targeted locations. Thymol‐iron oxide NPs were applied against MCF‐7 cancer cell lines to study their cytotoxic impact on cancer cells. The study showed that 90.4 μg/mL of NPs effectively inhibited 50% of cell growth and improved the BAX and CASP8 gene expression (Fekri Kohan et al. 2023). Similarly, Alam et al. (2021) examined thymol 1,2,3‐triazole derivatives against MCF‐7 cell lines. They concluded that 1,2,3‐triazole (IC50 6.17 μM) proved more potent against MCF‐7 cells compared with 5‐FU (IC50 20.09 μM). Furthermore, compound 10 showed (IC50 10.52 μM) against MDA‐MB‐231 cells compared with 5‐FU (IC50 25.31 μM) and tamoxifen (IC50 15.01 μM). Consequently, thymol derivatives induced cell cycle arrest at the G2/M phase via inhibiting thymidylate synthase.
Along with thymol, carvacrol has been reported to have remarkable potential to ameliorate BC. Carvacrol (0–65 μM) significantly reduced MDA‐MB‐231 cell proliferation via downregulated Jagged‐1 and cyclin D1 gene expression and modulation of the NOTCH pathway (Pandey et al. 2024). Chemotherapeutic drug resistance could be a major obstacle to attenuating cancer progression. Chen et al. (2024) reported that carvacrol (0, 100, 150, and 200 μM) inhibited cell development and induced apoptosis in Doxorubicin (Dox) resistant MDA‐MB‐231 breast cancer cells via cell cycle arrest at S and G2/M phases, enhanced Bax expression, while reduced Bcl‐2, PI3K, and P‐AKT expression. Carvacrol‐loaded selenium NPs (8.3 μg/mL) proved effective in inducing apoptosis in MCF‐7 bc cell lines by increasing OS markers (8‐OHDG, IL‐1β, NO, and LPO) and downregulated PCNA expression (Othman et al. 2023). Moradipour et al. (2022) stated that carvacrol (305 μM) inhibited MCF‐7 cell lines via enhancing Bax expression, suppressing Bcl‐2 expression, and induced apoptosis through upregulation of p53 expression. The anticancer mechanism of thymol and carvacrol in breast cancer is illuminated in Figure 4.
FIGURE 4.

Anticancer role of thymol and carvacrol in breast cancer via cell cycle arrest, apoptosis induction, oxidative stress production, enhancing NO, LPO, IL‐1β, and 8‐OHDG, downregulating PCNA, cyclin D1, BCL‐2, and PI3K, upregulating p53, BAX, and CASP8 expression, and modulating PI3KCA.
5.2. Lung Cancer
Lung cancer (LC) is the leading cancer among all cancers, including small cell lung cancer (SCLC) and non‐small cell lung cancer (NSCLC). Several factors are involved in LC, and smoking is the leading risk factor responsible for almost 85% of cases. Smoking can lead to dysplasia of lung epithelium, which may cause genetic alterations and affect protein synthesis upon continuing. Genetic mutation in MYC, Bcl‐2, and p53 genes is responsible for causing NSCLC (Kenzerki et al. 2023). Moreover, the dysfunction of the RB family (p107, p130), tumor suppressor PTEN, chromatin regulator CREBBP, and NOTCH receptors are other factors contributing to cancer pathogenesis. PI3K/AKT/mTOR pathway activation is also related to SCLC progression (Lázaro et al. 2022).
Sphingosine‐1‐phosphate (S1P), activated through catalytic actions of sphingosine kinase 1 (SphK1), is a key factor responsible for cell growth, survival, migration, and angiogenesis and is involved in neoplasm (Bravo et al. 2022). A comparative study of thymol and thymoquinone used as SphK1 inhibitors against NSCLC H1299 and A549 cell lines showed that both compounds remarkably inhibited SphK1 with the IC50 values of 35.52 μM and 53.68 μM, respectively. However, thymoquinone exhibited a better cytotoxic effect than thymol, and the IC50 values were 27.96 μM for H1299 and 54.43 μM for A549 lines (Shakeel et al. 2024). Thymol (80, 120, and 180 μM) extracted from Thymbra spicata was evaluated against the NSCLC cell line A549, and it was found that thymol induced a cytotoxic effect on the cell line via activation of NOX2, ROS production, increased Cai2+, and Bax/Bcl‐2 ratio (Moayeri et al. 2021). Similarly, carvacrol from ethanolic Origanum vulgare extract showed cytotoxic activity against A549 LC cell lines. The results revealed that carvacrol was the major compound in the extract, and the cellular uptake of carvacrol by cells was 56 μM. Moreover, (0–250 μg/mL) Origanum vulgare ethanolic extract reduced cell proliferation and induced cytotoxicity (Coccimiglio et al. 2016). The AXL receptor tyrosine kinase (RTK) is linked with cell proliferation, migration, and neoplasm and also promotes epithelial‐mesenchymal transition (EMT), a significant factor in drug‐resistance metastasis (Yan et al. 2021). Carvacrol has been reported to reduce AXL overexpression and suppress cell propagation and migration in NSCLC (Jung et al. 2018).
5.3. Colorectal Cancer
Colorectal cancer (CRC) is the 3rd leading cancer and 2nd in casualties globally. CRC develops from the growth of tiny cell groups (polyps) inside the colon, and somehow with age, these polyps transform into tumors in the next 5–10 years. Inherited disorders, genetic alterations, inflammatory bowel disease (IBD), and other malignancies are risk factors for CRC progression (Al‐Muswie et al. 2023). The epigenetics and genetics of CRC involve microRNAs (miRNAs) that induce cancer‐associated pathway disturbances at the post‐transcriptional level. The disturbances result in alterations in oncogenes and TSGs, metastasis, and dysplastic epithelium in the adenoma‐carcinoma process, eventually leading to CRC development (Fischer et al. 2019).
Numerous studies have discussed the association between growth factors and neoplasm. Epidermal growth factor receptor (EGFR) is a tyrosine kinase receptor that plays a significant role in CRC by stimulating several pathways like RAF, MAPK, RAS, and MEK. EGF receptor inhibition has been reported as a practical approach to treating CRC (Janani et al. 2022). Keshavarz et al. (2024) investigated the anti‐EGFR activity of thymol‐based nanoliposome in SW84 and SW111 CRC cell lines and found that thymol‐nanoliposome (IC50 14.2 and 6.4 μg/mL) inhibited SW48 and SW1116 cells growth respectively. Moreover, a significant reduction in EGFR gene expression and enhanced apoptosis was noticed. Thymol (100–1000 μM) showed an antiproliferative effect against HCT‐8 CRC cell lines via cell cycle arrest at G0/G1 phase, upregulating pro‐apoptotic p53 expression, suppressing anti‐apoptotic Bcl‐xL expression, activating caspase‐9/3, and OS‐induced apoptosis (Anvarbatcha et al. 2023). Thymol derivatives with admirable anticancer properties have been studied against HT‐29 and HCT‐116 cell lines. The findings of the study demonstrated that acetic acid thymol ester (IC50 0.08 μg/mL) increased ROS production, induced apoptosis, and subdued cell proliferation in cancerous cells, compared with standard thymol (IC50 ~ 60 μg/mL) (Blažíčková et al. 2022). The activation of Wnt/β‐catenin signaling is noticed in 90% of CRC cases, thus playing a main role in CRC progression. The mechanism is a series of complex steps involving WNT‐protein ligands binding with LRP5/6 membrane receptors, translocation of β‐catenin, and activation of c‐Myc and cyclin D1, subsequently leading to cell proliferation, migration, and oncogenesis (He and Gan 2023). Previously, Zeng et al. (2020) reported that thymol (75 and 150 mg/kg) from T. vulgaris reduced tumor volume and cell metastasis by activating the Bax/Bcl‐2 pathway, suppressing EMT, and regulating the Wnt/β‐catenin pathway.
The studies have established that VEGF promotes matrix metalloproteinases (MMPs) expression, degrades the extracellular matrix, and mediates HIF‐1α. Thus, HIF‐1α contributes significantly to cell migration, proliferation, and induced EMT in malignant tissue. Several chemotherapeutic drugs have been applied to modulate the HIF‐1α/VEGF signaling pathway, but their hostile outcomes, like anemia and appetite loss, are alarming (Chen et al. 2022). Carvacrol has been investigated in a chemical‐induced hypoxic CRC cell model. It was found that carvacrol (6.25, 12.5, 25, and 50 μg/mL) inhibited cell migration in SW480 cell lines. Moreover, carvacrol at higher concentrations suppressed the HIF‐1α/VEGF pathway to inhibit cell proliferation. Another study showed that carvacrol reduced HCT116 and LoVo cell lines invasion via cell cycle arrest at the G2/M phase, diminishing MMP‐2/9, Bcl‐2, and cyclin B1 expression, upregulating Bax and c‐Jun N‐terminal kinase expression, and modulating MAPK and PI3K/AKT signaling pathways (Fan et al. 2015). Figure 5 demonstrates the anticancer mechanism of thymol and carvacrol against CRC.
FIGURE 5.

Anticancer mechanism of thymol and carvacrol against CRC via cell cycle arrest, apoptosis induction, Wnt/β‐catenin, HIF‐1α/VEGF, and PI3K/AKT pathways modulation, downregulating MMP‐2/9, Bcl‐2, cyclin B1, EGFR, and upregulating Bax, p53, and c‐Jun N‐terminal kinase expression.
5.4. Liver Cancer
Liver cancer or hepatocellular carcinoma (HCC) is the 6th most common cancer, affecting approximately 41,630 people globally. Liver cirrhosis, fatty liver disease (FLD), alcohol consumption, exposure to toxins and chemicals, hepatitis, and congenital disorders are major risk factors contributing to HCC development (Huang et al. 2021). Viral hepatitis, a significant risk factor of HCC, mainly alters genes such as TERT, PDGFR β, and MAPK1. Moreover, the virus alters other proteins like HBx, which can mutate Ras, JNK, Raf, MAPK, and ERK gene expression (Wang and Deng 2023).
Studies have shown that VEGF is often overexpressed in HCC compared to normal liver tissue, especially VEGF‐A, which is the isoform responsible for angiogenesis and vascular remodeling, and its receptors (VEGFRs) are critical for HCC growth and development (Jia et al. 2021). Moreover, hypoxia is a key player in VEGF activation, angiogenesis, and carcinogenesis. In this regard, thymol nanoemulsions have been proven effective against liver cancer via modulating VEGF gene expression (Hussein 2024). Thymol (11 μM) induced apoptosis and genotoxicity in HepG2 cell lines but did not alter the expressions of Bax, Bcl‐2, and caspase‐3. Furthermore, several studies on thymol's protective effect against liver injury via regulating inflammatory markers and pathways have demonstrated that it has the potential to inhibit inflammation and genetic modifications, eventually protecting from tumor growth and cell invasion (Yousef et al. 2023). Molecular docking of thymoquinone and thymol from Nigella sativa L. inhibited P38 protein, showing that it could be the probable remedy for HCC (Tabassum and Ahmad 2020).
Studies on carvacrol also proved its potential to induce apoptosis in cancer cells and modulate signaling pathways to reduce cell proliferation. Carvacrol (15 mg/kg) by intragastric tube for 6 weeks daily reduced HIF‐1α and downregulated STAT3, JAK2, and FGL1, thus inhibiting cell division and improving immunity via enhancing CD8+ T cells (Yousef et al. 2024). Carvacrol (15 mg/kg/day) enhanced antitumor activity combined with sorafenib (10 mg/KG/day) orally in the HCC rats' model. The findings showed that combined therapy induced apoptosis in cancer cells by reducing cyclin D1 and Bcl‐2 expression while improving Bax and caspase‐3 expression. Additionally, the downregulation of ABCG2, NOTCH1, SALL4, and CD133 was observed in the study results (Yousef et al. 2023). An in vitro analysis unveiled that carvacrol (45 μg/mL) and essential oil (0.08 μg/mL) of Origanum onites altered the expression of 48 genes out of 84 in HepG2 cells. These 48 genes are associated with HCC and inflammatory pathways, thus proving them a promising anticancer and hepatoprotective agent (Tomsuk et al. 2024). Previously, Yin et al. (2022) reported that carvacrol inhibited cell proliferation and invasion in HepG2 cell lines by targeting 40 genes. They concluded that carvacrol affected SLC6A3 and SCN4A, and the downregulation of SLC6A3 inhibited cell viability.
5.5. Blood Cancer
Blood cancer is a generic term encompassing different types of cancer, like leukemia, lymphoma, and myeloma. Leukemia is one of the most prevalent blood cancers, affecting 474,519 people in 2020, thus making it the 12th most common cancer worldwide (Huang et al. 2022). Acute myeloid leukemia (AML) is the most common type of leukemia, featured by the overproduction of abnormal blood cells, specifically immature white blood cells called myeloblasts. It mainly affects the blood and bone marrow (Wachter and Pikman 2024).
An in vitro study conducted on human AML cell lines (K562, KG1, and HL60) showed that both thymol (25, 50, 75, and 100 μM) and carvacrol (100, 200, 300, and 400 μM) induced caspase‐dependent apoptosis in HL60, while inducing caspase‐independent apoptosis in KG1 and K562 (Bouhtit et al. 2021). Studies on terpenes have proved their effectiveness against leukemia by targeting various factors. Cymbopogon flexuosus volatile oil (CFVO) containing terpenes, including thymol and carvacrol, suppressed NF‐κB‐dependent proteins like anti‐apoptotic (cFLIP, Bcl‐xL and XIAP) proliferative (Cyclin E1, Cyclin D) and invasion (ICAM‐1) proteins. Moreover, CFVO also reduced TNF‐α, thus establishing its anti‐leukemic potential against KBM‐5 cell lines (Sajid et al. 2025). Another study reported that thymol‐induced apoptosis modulates signaling cascades to inhibit cell invasion in leukemia cell lines (Logesh et al. 2024).
6. In Vitro and In Vivo Anticancer Activity of Carvacrol
Carvacrol exhibits significant anticancer properties in both in vitro and in vivo studies, demonstrating apoptosis induction, cell cycle arrest, and antiproliferative effects against various cancer types, highlighting its therapeutic potential. Carvacrol (25–200 μM) inhibited CRC by reducing HT‐29 cell line proliferation via modulating CDK4, Cyclin D1, Bax, and Bcl‐2 expression (Pakdemirli et al. 2020). Likewise, carvacrol (25–500 μM) inhibited MCF‐7 bc cell lines by enhancing p53, Bax, caspase 3/6/7 expression, downregulating Bcl‐2 expression, and modulating the PI3K/p‐AKT pathway (Jamali et al. 2018). The SiHa cell lines of cervical cancer revealed apoptosis via increased expression of Bax and diminished expression of Bcl‐2 upon being treated with carvacrol (25–500 μM) (Al‐Fatlawi 2018). Carvacrol (200–1000 μM) modulated caspase‐3 and Bcl‐2 expression in K562 leukemia cell lines (Bouhtit et al. 2021). The anticancer activity of carvacrol against different cancerous cell lines and in the in vivo model is listed in Table 2.
TABLE 2.
In vitro and in vivo anticancer potential of carvacrol.
| Cancer | Cell lines/animal type | Concentration/dose/route/duration | Molecular targets | References |
|---|---|---|---|---|
| Breast cancer | MDA‐MB‐231 | 1–10,000 μM | Cyt c, Bax, CDK4 | Baranauskaite et al. (2017) |
| MCF‐7 | 25–500 μM | p53, Bax, caspase 3/6/7, Bcl‐2 PI3K/p‐AKT | Jamali et al. (2018); Al‐Fatlawi and Ahmad (2014) | |
| BT‐483, BT‐474 | 25–500 μM | cyclin B, CDK4 | Li et al. (2021) | |
| Cervical cancer | HeLa | 25–800 μM | Bax, Bcl‐2, p53, caspase‐3/6/9 | Mehdi et al. (2011) |
| SiHa | 25–500 μM | Bax, Bcl‐2, p53, caspase‐3/6/9 | Al‐Fatlawi (2018) | |
| Choriocarcinoma | JAR, JEG3 | 50–300 μM | PI3K/AKT, p‐ERK1/2 p‐p38, MMP | Lim et al. (2019) |
| Lung cancer | A549 | 100–1000 μM | p38, NF‐κB, AXL, TNFα, GSK‐3b, Beclin1 | Jung et al. (2018) |
| H460 | 30–300 μM | AXL | Coccimiglio et al. (2016) | |
| H1299 | 25–1800 μM | Cell membrane and DNA damage | Ozkan and Erdogan (2012) | |
| Colorectal cancer | HT‐29 | 25–200 μM | CDK4, Cyclin D1, Bax, Bcl‐2 | Pakdemirli et al. (2020) |
| Caco‐2 | 100–2500 μM | DNA damage | Llana‐Ruiz‐Cabello et al. (2014) | |
| LoVo | 100–900 μmol/L | Bcl‐2, Bax, MMP‐2/9, Cyclin B1 | Fan et al. (2015) | |
| HCT 116 | 100–900 μmol/L | PI3K/p‐AKT | Fan et al. (2015) | |
| PROState cancer | DU145 | 10–500 μM | Bax, Bcl‐2, Notch1, Jagged‐1, MMP2 | Tayarani‐Najaran et al. (2019) |
| PC‐3 | 25–800 μM | PI3K/AKT, TRPM7, p‐STAT3, p‐ERK1/2 | Luo et al. (2016) | |
| Melanoma | A375 | 3.906–1000 μg/mL | Bcl‐2, cell cycle arrest | Govindaraju and Arulselvi (2018) |
| Gastric adenocarcinoma | AGS | 100–600 μM | Bax, Bcl‐2, Caspase‐3/9 | Günes‐Bayir et al. (2018) |
| Glioblastoma | U87 | 1–10,000 μM | PI3K/AKT, MAPK, TRPM7 | Chen et al. (2015) |
| DBTRG‐05MG | 200–1000 μM | Caspase‐3, ROS production | Liang and Lu (2012) | |
| Neuroblastoma | SH‐SY5Y | 12.5‐50 μM | Bax, Bcl‐2 | Çalibaşi Koçal and Pakdemirli (2020) |
| N2a | 10–400 mg/L | TNF‐α, MYCN | Aydın et al. (2014) | |
| Oral cancer | Tca‐8113 | 10–80 μM | CCND1 CDK4, p21, Bcl‐2/Bax, COX‐2 | Dai et al. (2016) |
| SCC‐25 | 167 μg/mL | MMP‐2/9 COX‐2, p21 | Dai et al. (2016) | |
| OC2 cells | 200–1000 μM | Caspase‐3, ROS generation | Liang et al. (2013) | |
| Leukemia | HL‐60 | 10–400 μM | MMP, Bcl‐2 | Bhakkiyalakshmi et al. (2016) |
| K562 | 200–1000 μM | Caspase‐3, Bcl‐2 | Bouhtit et al. (2021) | |
| KG1 | 100–400 μM | pp38 | Bouhtit et al. (2021) | |
| CEM | 0.05–1.25 μM | Cell cycle interruption | Jaafari et al. (2012) | |
| P‐815 | 0.05–1.25 μM | Cell cycle arrest | Jaafari et al. (2012) | |
| Liver cancer | Hep3B | 1–1000 μM | Bcl‐2, pp38 | Fitsiou et al. (2016) |
| HepG2 | 100–1000 μM | MAPK p‐ERK 1/2, Caspase‐3 | Elshafie et al. (2017) | |
| Ovarian cancer | SKOV‐3 | 100–600 μM | GSK‐3β, NF‐κB | Elbe, Yigitturk, Cavusoglu, Uyanikgil, and Ozturk (2020) |
| Breast cancer | Holtzman mice | 50, 100, 200 mg/kg for 14 weeks (orally) | Antioxidant activity | Rojas‐Armas et al. (2020) |
| Colon cancer | Wistar rats | 20, 40, 80 mg/kg/day for 16 weeks (i.p.) | ↑GPx, GSH, SOD, CAT | Sivaranjani et al. (2016) |
| Liver Cancer | Wistar rats | 15 mg/kg for 16 weeks (orally) | Improved AST, ALT, ALP, cGT | Jayakumar et al. (2012) |
| 15 mg/kg for 15 weeks (orally) | ↓AFP, VEGF, AFU, and GGT PARP | Ahmed et al. (2013) | ||
| 15 mg/kg carvacrol for 16 weeks (orally) | ↓MMP‐2/9, AgNORs, PCNA | Subramaniyan et al. (2014) | ||
| C57BL/6 mice | 20 weeks (i.g.) | Modulation of DAPK1 and PPP2R2A | Li et al. (2019) |
7. In Vitro and In Vivo Anticancer Studies of Thymol
In vitro and in vivo anticancer studies of thymol show its potential in inhibiting cancer cell growth, inducing apoptosis, and reducing tumor size, highlighting its promise as a natural anticancer agent. Qoorchi Moheb Seraj et al. (2022) reported that thymol (230 μM) reduced AKT and enhanced p38 expression in U‐87 glioblastoma cell lines, thus effectively decreasing oncogenesis. In another study, 46.74 μg/mL and 41.46 μg/mL thymol inhibited HCT116 and LoVo cell line invasion via downregulating the Wnt/β‐catenin pathway and reducing c‐Myc and Cyclin D1 expression (Zeng et al. 2020). Thymol (15.6 to 2000 μM) induced apoptosis in KYSE‐30 esophageal cancer cell lines through improved ROS production, enhanced p53, and Bax expression, and declined Bcl‐2 expression (Pouyamanesh et al. 2024). Thymol (4.3 mM) increased ROS production and induced apoptosis against oral cancer in athymic nude mice (Zeng et al. 2020). The in vitro and in vivo studies of thymol against various cancers are highlighted in Table 3.
TABLE 3.
In vitro and in vivo anticancer studies of thymol.
| Cancer | Cell lines/animal type | Concentration/dose | Molecular targets | References |
|---|---|---|---|---|
| Glioblastoma | U‐87 | 230 μM | ↓ AKT ↑p38 | Qoorchi Moheb Seraj et al. (2022) |
| Colorectal cancer | HCT116 | 46.74 μg/mL | ↓Wnt/β‐catenin | Zeng et al. (2020) |
| LoVo | 41.46 μg/mL | c‐Myc, Cyclin D1 | ||
| Prostate cancer | PC‐3, DU145 | 100, 200, 400, 600, 800 μM | Bax, Bcl‐2 | Elbe, Yigitturk, Cavusoglu, Uyanikgil, and Ozturk (2020) |
| Breast cancer | MDA‐MB‐231 | |||
| Lung cancer | KLN205 | |||
| Colon cancer | HT29 | 4 mM | PI3K/AKT, ↓ ERK | Lv and Chen (2017) |
| Leukemia | KG1 | 100 mM | ↑Apoptosis, OS, autophagy | Bouhtit et al. (2021) |
| HL60 | ||||
| K562 | ||||
| Esophageal cancer | KYSE‐30 | 15.6 to 2000 μM | ↑ROS production, p53, Bax, ↓Bcl‐2 | Pouyamanesh et al. (2024) |
| Ovarian cancer | OVCAR‐3 | 145.683 μg/mL and 388.53 μg/mL | ↓ Bcl‐2, ↑caspase‐3/8/9 and Bax | Seçme and İlhan (2025) |
| Prostate cancer | LNCaP | 50, 100, 150, 200, and 250 μM | ↑Apoptosis | Singhal et al. (2021) |
| Cervical cancer | HeLa | 10–1000 μg/mL | ↑Apoptosis | Osarieme Imade et al. (2023) |
| Skin cancer | A2058 | 62.5–500 μg/mL | VEGF and VEGFR genes | Feyzmohamadi Khoramabadi et al. (2024) |
| Colon cancer | Wistar rats | 20 mg/kg/day | ↓NF‐κB, ↑Caspase‐3 | Hassan et al. (2021) |
| Oral cancer | Athymic nude mice | 4.3 mM | ROS production and apoptosis | De La Chapa et al. (2018) |
| Colorectal cancer | BALB/c nude mice | 75–150 mg/kg once/day | Wnt/β‐catenin | Zeng et al. (2020) |
| Breast cancer | BALB/c mice, Sprague–Dawley rats | 0.1% and 1% | Bax, CD44, ALDH1A1, and VEGFR‐2 | Kubatka et al. (2019) |
8. Anticancer Synergistic Role With Other Phytochemicals, Chemotherapy, and Radiotherapy
Several studies have proved that thymol and carvacrol enhance anticancer effects by synergizing with bioactive compounds, chemotherapy, and radiotherapy, improving efficacy, inducing apoptosis, oxidative stress, and modulating inflammation in cancer treatment. Bouhtit and colleagues reported that thymol and carvacrol found in the essential oil of Ptychotis verticillate induced cell apoptosis in KG1, HL60, and K562 leukemia cell lines (Bouhtit et al. 2021). The combined therapy of Zataria multiflora extract (ZME) comprising thymol, carvacrol, and doxorubicin proved efficacious against lymphoblastic leukemia (ALL) Nalm‐6 cells and induced apoptosis via suppressing anti‐apoptotic proteins (Lashkari et al. 2022). Thymol enhanced the cytotoxicity and antiproliferative activity of 5‐FU against BC and CRC cell lines. In addition, thymol attenuated 5‐FU‐induced intestinal mucositis in the murine model (Badr et al. 2022). Another investigation reported that thymol protected and ameliorated 5‐FU‐induced intestinal mucositis via suppressing the TGF‐β/p38/p‐JNK signaling and inhibiting NF‐κB and TNF‐α (Al‐Khrashi et al. 2022).
The chitosan nanoparticles of thymol oil extract and bee pollen were examined against HepG2 and MCF‐7 cancer cells. The findings showed that the combination promoted apoptosis, disrupted the cell cycle via improved caspase‐3 expression, and downregulated caspase‐9 and P53 (Alshehri and Abdella 2023). Thymol and carvacrol in nanoemulsions and nanocapsules of O. glandulosum Desf. oil exhibited cytotoxic effects on Hep G‐2 and THLE2 cell lines. It was observed that nanocapsules (54.93 μg/mL) revealed better anticancer potential compared with nanoemulsions (131.6 μg/mL) and standard drug 5‐FU (5–400 μg/mL) (Ali et al. 2020). Carvacrol, thymol, γ‐terpinene, methyl carvacrol ether, and p‐cymene are the key components of Zataria multiflora essential oil (ZEO) (Torabiardekani et al. 2023). The combination of ZEO and doxorubicin was investigated in a study against PC3 cell lines, and it was found that ZEO enhanced doxorubicin anticancer activity by ROS production and induced apoptosis in PC3 cell lines, showing the synergic role of thymol and carvacrol with chemotherapy (Zare et al. 2021).
Radiotherapy is a vital treatment in the fight against neoplasm, as it exploits DNA damage to eradicate tumors efficiently. However, this treatment can harm healthy cells (Barazzuol et al. 2020). In this context, carvacrol and thymol were evaluated to observe the protective effect on rat ovaries against the side effects of radiotherapy. The study elaborated that both compounds improved ovarian follicle development and recovered hormone levels in ovaries. Furthermore, these compounds counteracted OS, improved IGF‐1 levels, and reduced TNF‐α production in signaling pathways (Mahran et al. 2019). Thymol, along with hesperidin, prevented submandibular gland damage caused by radiation therapy. Both compounds decreased the injury by lessening oxidants and improving levels of antioxidant enzymes; however, thymol displayed better protective activity than hesperidin, indicating its ability to reduce the damage caused by radiotherapy (Sakat et al. 2023). Similarly, chemotherapy also imposes an adverse impact on healthy tissues and organs. The studies on carvacrol have proved that it alleviated cisplatin and doxorubicin‐induced cardiotoxicity in rats via apoptosis induction and modulating PI3K/AKT, Notch/Hes1 pathways (Akaras et al. 2024; Retnosari et al. 2024).
9. Carvacrol‐ and Thymol‐Based Hybrids With Anticancer Activity
Carvacrol‐ and thymol‐based hybrid compounds with anticancer potential have opened new avenues in drug delivery systems. Their biocompatibility and promising therapeutic strategies have garnered significant attention in research. Demirbolat et al. (2022) developed carvacrol hybrid molecules and validated their cytotoxic impact on PC‐3, SH‐SY5Y, MCF‐7, NIH/3 T3, K562, and A549 cell lines. The study showed compound 3 with a benzene ring exhibited promising cytotoxic activity against MCF‐7 compared with doxorubicin, with the IC50 of 12.8 μM and 49.05 μM, respectively. However, the thymol hybrid derivative (its isomeric partner) showed an IC50 of 5.96 μM against PC‐3 lines. Another study showed that p‐methoxy thymol pyrazole hybrids had an anticancer effect against four cancer cell lines. The findings showed that p‐methoxy hybrids displayed better cytotoxic effects than other thymol pyrazole hybrids due to the ether group on the thymol moiety. Thus, 5a and 5b hybrids were prominent as the most potent compounds against A‐549 and HT‐1080 cell lines (Laamari et al. 2024). Likewise, thymol hybrids 6a–m (1.8 μM) and 6b–f (1.4 μM) revealed greater antiproliferative properties than doxorubicin and 5‐FU (18.74 μM) against HCT‐116, MCF‐7, and HepG2 cell lines, indicating their better anticancer potential in comparison with drugs (Almalki et al. 2021). Thymol–ciprofloxacin hybrids were investigated against four cancer and one normal cell line. The compound 7a–b demonstrated a promising anticancer effect against the cancer cells with IC50 values (> 52 μM), compared to doxorubicin, and no substantial cytotoxic result was noticeable on the normal cell lines (Szostek et al. 2022).
Carvacrol hybrids were synthesized by Sisto et al. (2020) and evaluated against gastric adenocarcinoma cell lines. All the hybrids displayed poor activity compared to 5‐FU; however, the introduction of the benzyl moiety, specifically with 3‐CH3, 4‐SO2CH3, and 4‐SOCH3 on the meta‐ and para‐positions, upgraded the pharmacological activity of hybrid compounds. In another investigation, compound 8 of carvacrol hybrids was an effective anticancer compound with IC50 values of 0.47 and 0.75 μM against MCF‐12A and MCF‐7 lines, respectively. The better anticancer activity was credited to integrating artesunate by an ester link into the carvacrol moiety (Mbese et al. 2022). The derivatives/hybrids of thymol and carvacrol with anticancer potential are described in Table 4.
TABLE 4.
Anticancer activity of thymol and carvacrol hybrids against different cancer cell lines.
| Compound | Hybrids | Study type/cell lines | IC50/concentration | References |
|---|---|---|---|---|
| Thymol | 3 | PC‐3, MCF‐7, K562, and SH‐SY5Y | 12.8 μM and 5.96 μM | Demirbolat et al. (2022) |
| 4 | MCF‐7, A‐549, MDA‐MB‐231, and HT‐1080 | 7.10–19 μM | Laamari et al. (2023) | |
| 5a and 5b | A‐549 and HT‐1080 | 22.17–62.72 μM | Laamari et al. (2024) | |
| 6a–m, 6b–f | HCT‐116, MCF‐7, and HepG2 | 1.8 μM and 1.4 μM | Almalki et al. (2021) | |
| 7a–b | SW480, HCT116, HepG2, HaCaT | > 52 μM | Szostek et al. (2022) | |
| 11a | MCF‐7, PC‐3, and HT‐29 | 2.48 μM | Zengin Kurt et al. (2023) | |
| 12a–c | PC3 and DLD‐1 | 7.67 μM and 12.39 μM | Sahin et al. (2023) | |
| 14a–c | HepG2, A549, MCF‐7, and HeLa | 6.24–11.96 μM | Yu et al. (2020) | |
| Carvacrol | 16, 21, 35, 38 | Gastric adenocarcinoma | 6.5 μM | Sisto et al. (2020) |
| 8 | MCF‐7, MCF‐12A | 0.47 and 0.75 μM | Mbese et al. (2022) | |
| 9a–d | Artemia salina nauplii (Brine shrimp method) | 50.39 μg/mL | Valverde Sancho et al. (2023) | |
| 10a–c | SH‐SY5Y and HEK‐293 | 9.79–64.72 μM | Vasconcelos et al. (2024) | |
| 11b | PC3, HT‐29, and MCF‐7 | 9.10, 9.40, and 12.01 μM | Zengin Kurt et al. (2023) | |
| 15a–c | MCF‐7, MD‐MBA‐231, and HeLa | 51.05–64.75 μg/mL | Khwaza et al. (2023) |
10. Clinical Trials and Strategies to Increase the Therapeutic Efficacy
Clinical trials are research studies that evaluate new treatments, drugs, or medical interventions in humans to assess safety, efficacy, and side effects before regulatory approval for widespread medical use. Despite thymol and carvacrol's prime importance in cancer therapy, the clinical trials are lacking in anticancer perspectives, thus hindering their medicinal and clinical significance. However, some trials on other diseases are available, illuminating their therapeutic role in disease management. In a trial with 30 participants fed on thymol (25 mg/kg/day) orally for 30 days and gel application along with low‐level laser therapy, reduced cytokines (IL‐1β, TNF‐α), MDA, H2O2, triglycerides, and total cholesterol in patients with type 2 diabetes mellitus and dermatitis (Martirosyan et al. 2022). In a randomized, cross‐over study, 12 individuals were provided with two different dental varnishes, including 10% fluoride (F‐) varnish and 1% chlorhexidine (CHX) and 1% thymol varnish. The outcomes showed that both varnishes exhibited a better remineralization effect than control (Bizhang et al. 2015). The safety and tolerability of carvacrol were evaluated in healthy participants receiving 1 and 2 mg/kg/day for 1 month. The ESR, calcium, MCV, Hb, and HCT levels were decreased, while CPK, LDH, MCH, MCHC, and triglycerides increased in the 1 mg/kg/day group. The 2 mg/kg/day group depicted significant reductions in HDL, total bilirubin, RBC, and HCT. The outcomes of this phase I study concerning carvacrol's impact on healthy subjects displayed clinical safety and tolerability (Ghorani, Alavinezhad, Rajabi, and Boskabady 2021). A randomized, placebo‐double‐blind clinical trial involving 33 participants was divided into two groups: placebo and carvacrol group (1.2 mg/kg/day, n = 17). The carvacrol group was the asthmatic patients' group, which received prepared capsules for 2 months and 3 times/day with routine medications. The results showed that respiratory symptoms and OS markers substantially declined after a 2‐month treatment with carvacrol. The anti‐inflammatory and antioxidant effects of carvacrol make it a promising therapeutic agent for asthma (Ghorani, Alavinezhad, Rajabi, Mohammadpour, and Boskabady 2021). The sta bility and bioavailability of compounds are critical to achieving their maximum functional efficiency in different fields. They rapidly lose stability in unfavorable conditions and are mostly sensitive to light, temperature, oxygen, and pH. Likewise, thymol and carvacrol are volatile, and their stability is influenced by these factors (temperature, light, oxygen, and pH). Both compounds are stable under moderate circumstances, but prolonged exposure to heat, UV light, or air can lead to degradation (Ates and Yildiz 2025). The stability and bioavailability of compounds can be increased via nanoencapsulation/nanocarriers (Niaz et al. 2021).
Novel techniques, like liposomes or polymer‐based carrier encapsulation, are often applied to improve their stability in formulations. Moreover, maintaining neutral to slightly acidic pH levels can preserve their activity, making them effective in food preservation and pharmaceutical applications (Gong et al. 2021). Regarding this, Ates and Yildiz (2025) determined the stability of carvacrol in β‐cyclodext rin metal–organic frameworks (β‐CD‐MOFs). They concluded that the thermal stability of the carvacrol‐β‐CD‐MOFs complex improved by alterations in temperatures. Furthermore, the antioxidant capacity and solubility of the complex also improved, as demonstrated by the controlled release behavior of carvacrol. Additionally, nanoencapsulation of carvacrol with Eudragit and chia mucilage was evaluated, and it was found that carvacrol and Eudragit NPs preserved their nanometric dimensions for 180 days, while carvacrol and chia mucilage complex sustained this property for only 30 days (Tópor Nunes et al. 2024).
Protein‐based nanocarriers are an effective way to enhance thymol's stability and application performance. Rao et al. (2024) studied the synergistic role of succinylation amendment and ε‐polylysine in thymol‐loaded succinylated ovalbumin/ε‐polylysine nanogel. They reported that succinylation elevated the negative charges of OVA, and ε‐PL complexation enhanced electrostatic interactions and hydrogen bonding of the nanogel. The study's findings indicated that the nanogel showed exceptional thermal and pH stability. Another study reported that thymol‐lauric acid‐loaded casein NPs (ThyLA‐NPs) revealed better thermal and storage stability than simple thymol‐casein NPs. The better performance of ThyLA‐NPs is due to the high hydrophobic nature of the eutectic solvents (Ge et al. 2023).
Ultrasonication is an effective technique to enhance the stability of thymol and other bioactive compounds. The ultrasonication relies on the efficacy of the cavitation mechanism, which includes sinusoidal pressure fluctuations within the fluid, thus causing cavitation bubbles. Moreover, due to their hydrophilic nature, polysaccharide nano‐emulsions can improve stability and solubility (Mehta et al. 2022). Recently, Phyo et al. (2024) used ultrasonication to enhance the stability and physicochemical properties of thymol and cinnamaldehyde‐loaded chitosan NPs. The study findings showed that 400 watts of power elevated the performance of NC‐CH formulations, and NC‐CH‐400 displayed increased solubility. Moreover, the NC‐CH‐400 formulation exhibited maximum thermal stability, verified by the melting points, indicating superior storage durability. The nano‐capsules also displayed a smooth microstructure without surface cracking.
11. Limitations
Thymol and carvacrol face significant limitations in clinical application due to poor stability and low bioavailability. Their volatility and susceptibility to degradation reduce efficacy during storage and processing. In vivo, both compounds are rapidly metabolized and eliminated, limiting their systemic availability. Moreover, their hydrophobic nature hinders effective absorption. Clinical trials evaluating thymol and carvacrol remain limited, with most evidence stemming from in vitro or animal studies. This lack of robust human trials regarding cancer hampers conclusive therapeutic validation. Overcoming these limitations requires the development of improved delivery systems and more comprehensive clinical research to establish the efficacy and safety profiles.
12. Conclusion and Future Perspectives
Thymol and carvacrol exhibit substantial anticancer potential through multiple molecular mechanisms, including apoptosis induction, oxidative stress modulation, and anti‐inflammatory effects. Their ability to enhance chemotherapy and radiotherapy efficacy while reducing toxicity makes them promising adjuncts in cancer treatment. Additionally, their synergy with other bioactive compounds further amplifies their therapeutic benefits. As natural, biocompatible agents, thymol and carvacrol offer a novel approach to cancer management, potentially improving treatment outcomes. Moreover, drug‐resistant cancer cells limit treatment efficacy, but thymol and carvacrol offer solutions by modulating resistance mechanisms, enhancing drug sensitivity, and improving therapeutic outcomes. Future research should optimize their delivery, bioavailability, and clinical applications to harness their full potential in integrative oncology. Advanced drug delivery systems, such as nano‐formulations and encapsulation techniques, can improve solubility, controlled release, and targeted action. Further clinical trials are required regarding cancer to validate their efficacy, safety, and synergistic potential with existing therapies. Understanding their pharmacokinetics and mechanisms of action will facilitate their transition into clinical applications. These efforts could establish thymol and carvacrol as effective anticancer agents, paving the way for novel, natural‐based therapeutics.
Author Contributions
Ahmad Mujtaba Noman: conceptualization (equal), writing – original draft (equal). Muhammad Tauseef Sultan: conceptualization (equal), writing – original draft (equal). Shehnshah Zafar: resources (equal), writing – original draft (equal). Muhammad Maaz: data curation (equal), writing – review and editing (equal). Aimen Mazhar: writing – original draft (equal). Muzzamal Hussain: investigation (equal), writing – review and editing (equal). Muhammad Imran: data curation (equal), resources (equal). Ahmed Mujtaba: validation (equal), visualization (equal). Muhammad Tajammal Hussain: data curation (equal), investigation (equal), writing – review and editing (equal). Suliman A. Alsagaby: visualization (equal), writing – review and editing (equal). Waleed Al Abdulmonem: data curation (equal), writing – review and editing (equal). Muhammad Asif Khan: data curation (equal), investigation (equal), methodology (equal). Entessar Al Jbawi: data curation (equal), investigation (equal), supervision (equal).
Ethics Statement
The authors confirm that this manuscript adheres to all ethical standards.
Conflicts of Interest
The authors declare no conflicts of interest.
Noman, A. M. , Sultan M. T., Zafar S., et al. 2025. “Thymol and Carvacrol: Molecular Mechanisms, Therapeutic Potential, and Synergy With Conventional Therapies in Cancer Management.” Food Science & Nutrition 13, no. 9: e70936. 10.1002/fsn3.70936.
Funding: The authors received no specific funding for this work.
Contributor Information
Muzzamal Hussain, Email: muzzamalhussain24@gcuf.edu.pk.
Entessar Al Jbawi, Email: dr.entessara@gmail.com.
Data Availability Statement
The data that support the findings of this study are available on request from the corresponding author.
References
- Abed, A. T. , AlMudhafar A. M., and Hadi N. R.. 2024. “Anti‐Cancer Study of Carvacrol in Hypoxic‐Induced Colorectal Cancer Cell.” Journal of Angiotherapy 8, no. 1: 9441. 10.25163/angiotherapy.819441. [DOI] [Google Scholar]
- Abu‐Elfotuh, K. , Abdel‐Sattar S. A., Abbas A. N., et al. 2022. “The Protective Effect of Thymoquinone or/and Thymol Against Monosodium Glutamate‐Induced Attention‐Deficit/Hyperactivity Disorder (ADHD)‐like Behavior in Rats: Modulation of Nrf2/HO‐1, TLR4/NF‐κB/NLRP3/Caspase‐1 and Wnt/β‐Catenin Signaling Pathways in Rat Model.” Biomedicine and Pharmacotherapy 155: 113799. 10.1016/j.biopha.2022.113799. [DOI] [PubMed] [Google Scholar]
- Ahmad, A. , Tiwari R. K., and Ansari I. A.. 2025. “Targeting the Interplay Between Human Papillomavirus Oncoproteins and Hedgehog Signaling: Assessment of Chemopreventive Potential of Carvacrol in Cervical Cancer.” 3 Biotech 15, no. 4: 1–19. 10.1007/s13205-025-04250-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ahmad, A. , Tiwari R. K., Saeed M., et al. 2023. “Carvacrol Instigates Intrinsic and Extrinsic Apoptosis With Abrogation of Cell Cycle Progression in Cervical Cancer Cells: Inhibition of Hedgehog/Gli Signaling Cascade.” Frontiers in Chemistry 10: 1064191. 10.3389/fchem.2022.1064191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ahmed, H. H. , Shousha W. G., El‐Mezayen H. A., Ismaiel N. N., and Mahmoud N. S.. 2013. “In Vivo Antitumor Potential of Carvacerol Against Hepatocellular Carcinoma in Rat Model.” World Journal of Pharmacy and Pharmaceutical Sciences 2, no. 5: 2367–2396. [Google Scholar]
- Akaras, N. , Kandemir Ö., and Şimşek H.. 2024. “Carvacrol Ameliorates Cisplatin‐Induced Cardiotoxicity by Regulating Notch/Hes1 Signaling Pathway, Oxidative Stress and Cell Death in Rat Cardiac Tissue.” Türk Doğa Ve Fen Dergisi 13, no. 4: 180–191. 10.46810/tdfd.1564047. [DOI] [Google Scholar]
- Akhlaq, A. , Ashraf M., Omer M. O., and Altaf I.. 2023. “Carvacrol‐Fabricated Chitosan Nanoparticle Synergistic Potential With Topoisomerase Inhibitors on Breast and Cervical Cancer Cells.” ACS Omega 8, no. 35: 31826–31838. 10.1021/acsomega.3c03337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alam, M. M. , Malebari A. M., Syed N., et al. 2021. “Design, Synthesis and Molecular Docking Studies of Thymol Based 1, 2, 3‐TriazoleHhybrids as Thymidylate Synthase Inhibitors and Apoptosis Inducers Against Breast Cancer Cells.” Bioorganic & Medicinal Chemistry 38: 116136. 10.1016/j.bmc.2021.116136. [DOI] [PubMed] [Google Scholar]
- Al‐Fatlawi, A. A. , and Ahmad A.. 2014. “Cytotoxicity and Pro‐Apoptotic Activity of Carvacrol on Human Breast Cancer Cell Line MCF‐7.” World Journal of Pharmaceutical Sciences 2, no. 10: 1218–1223. [Google Scholar]
- Al‐Fatlawi, A. A. Y. 2018. “Anti‐Proliferative and Pro‐Apoptotic Activity of Carvacrol on Human Cancer Cells.” International Journal of Pharmaceutical Research 10: 174–180. [Google Scholar]
- Ali, H. , Al‐Khalifa A. R., Aouf A., Boukhebti H., and Farouk A.. 2020. “Effect of Nanoencapsulation on Volatile Constituents, and Antioxidant and Anticancer Activities of Algerian Origanum Glandulosum Desf. Essential Oil.” Scientific Reports 10, no. 1: 2812. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Al‐Khrashi, L. A. , Badr A. M., AL‐Amin M. A., and Mahran Y. F.. 2022. “Thymol Ameliorates 5‐Fluorouracil‐Induced Intestinal Mucositis: Evidence of Down‐Regulatory Effect on Tgf‐β/Mapk Pathways Through Nf‐κB.” Journal of Biochemical and Molecular Toxicology 36, no. 1: e22932. 10.1002/jbt.22932. [DOI] [PubMed] [Google Scholar]
- Almalki, A. S. , Nazreen S., Malebari A. M., et al. 2021. “Synthesis and Biological Evaluation of 1, 2, 3‐Triazole Tethered Thymol‐1, 3, 4‐Oxadiazole Derivatives as Anticancer and Antimicrobial Agents.” Pharmaceuticals 14, no. 9: 866. 10.3390/ph14090866. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Al‐Muswie, R. T. , Abbas A. M., and Chayan M. A.. 2023. “Colon Cancer: Risk Factors and Therapeutic Approaches.” Central Asian Journal of Medical and Natural Science 4, no. 5: 908–918. [Google Scholar]
- Alshehri, K. M. , and Abdella E. M.. 2023. “Development of Ternary Nanoformulation Comprising Bee Pollen‐Thymol Oil Extracts and Chitosan Nanoparticles for Anti‐Inflammatory and Anticancer Applications.” International Journal of Biological Macromolecules 242: 124584. 10.1016/j.ijbiomac.2023.124584. [DOI] [PubMed] [Google Scholar]
- Anvarbatcha, R. , Kunnathodi F., and Islam M.. 2023. “Induction of G0/G1 Phase Cell Cycle Arrest and Apoptosis by Thymol Through Ros Generation and Caspase‐9/‐3 Activation in Breast and Colorectal Cancer Cell Lines.” Journal of Cancer Research and Therapeutics 19, no. 7: 1915–1924. 10.4103/jcrt.jcrt_308_22. [DOI] [PubMed] [Google Scholar]
- Ates, K. , and Yildiz Z. I.. 2025. “Encapsulation of Carvacrol in Β‐Cyclodextrin Metal‐Organic Frameworks: Improved Solubility, Stability, Antioxidant Capacity and Controlled Release of Carvacrol.” Journal of Food Engineering 391: 112445. 10.1016/j.jfoodeng.2024.112445. [DOI] [Google Scholar]
- Aydın, E. , Türkez H., and Keleş M. S.. 2014. “The Effect of Carvacrol on Healthy Neurons and N2a Cancer Cells: Some Biochemical, Anticancerogenicity and Genotoxicity Studies.” Cytotechnology 66: 149–157. 10.1007/s10616-013-9547-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Badr, A. M. , Alkharashi L. A., Sherif I. O., Alanteet A. A., Alotaibi H. N., and Mahran Y. F.. 2022. “Il‐17/Notch1/Stat3 Pathway Contributes to 5‐Fluorouracil‐Induced Intestinal Mucositis in Rats: Amelioration by Thymol Treatment.” Pharmaceuticals 15, no. 11: 1412. 10.3390/ph15111412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Balan, D. J. , Rajavel T., Das M., Sathya S., Jeyakumar M., and Devi K. P.. 2021. “Thymol Induces Mitochondrial Pathway‐Mediated Apoptosis via Ros Generation, Macromolecular Damage and SOD Diminution in A549 Cells.” Pharmacological Reports 73: 240–254. 10.1007/s43440-020-00171-6. [DOI] [PubMed] [Google Scholar]
- Bansal, A. , Saleh‐E‐In M. M., Kar P., Roy A., and Sharma N. R.. 2022. “Synthesis of Carvacrol Derivatives as Potential New Anticancer Agent Against Lung Cancer.” Molecules 27, no. 14: 4597. 10.3390/molecules27144597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baranauskaite, J. , Kubiliene A., Marksa M., et al. 2017. “The Influence of Different Oregano Species on the Antioxidant Activity Determined Using Hplc Postcolumn Dpph Method and Anticancer Activity of Carvacrol and Rosmarinic Acid.” BioMed Research International 2017, no. 1: 1681392. 10.1155/2017/1681392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Barazzuol, L. , Coppes R. P., and van Luijk P.. 2020. “Prevention and Treatment of Radiotherapy‐Induced Side Effects.” Molecular Oncology 14, no. 7: 1538–1554. 10.1002/1878-0261.12750. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bayoumi, H. M. , Alkhatib M. H., and Al‐Seeni M. N.. 2021. “Carvacrol Effect on Topotecan Cytotoxicity in Various Human Cancer Cells In Vitro.” Pharmacia 68, no. 2: 353–363. 10.3897/pharmacia.68.e65878. [DOI] [Google Scholar]
- Bertucci, A. , Bertucci F., and Gonçalves A.. 2023. “Phosphoinositide 3‐Kinase (PI3K) Inhibitors and Breast Cancer: An Overview of Current Achievements.” Cancers 15, no. 5: 1416. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bhakkiyalakshmi, E. , Suganya N., Sireesh D., et al. 2016. “Carvacrol Induces Mitochondria‐Mediated Apoptosis in hl‐60 Promyelocytic and Jurkat T Lymphoma Cells.” European Journal of Pharmacology 772: 92–98. 10.1016/j.ejphar.2015.12.046. [DOI] [PubMed] [Google Scholar]
- Bizhang, M. , Kaleta‐Kragt S., Singh‐Huesgen P., Altenburger M. J., and Zimmer S.. 2015. “Effect of 10% Fluoride on the Remineralization of Dentin In Situ.” Journal of Applied Oral Science 23, no. 6: 562–570. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Blažíčková, M. , Blaško J., Kubinec R., and Kozics K.. 2022. “Newly Synthesized Thymol Derivative and Its Effect on Colorectal Cancer Cells.” Molecules 27, no. 9: 2622. 10.3390/molecules27092622. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bouhtit, F. , Najar M., Moussa Agha D., et al. 2021. “New Anti‐Leukemic Effect of Carvacrol and Thymol Combination Through Synergistic Induction of Different Cell Death Pathways.” Molecules 26, no. 2: 410. 10.3390/molecules26020410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bravo, G. Á. , Cedeño R. R., Casadevall M. P., and Ramió‐Torrentà L.. 2022. “Sphingosine‐1‐Phosphate (S1p) and S1p Signaling Pathway Modulators, From Current Insights to Future Perspectives.” Cells 11, no. 13: 2058. 10.3390/cells11132058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cai, S. , Wu L., Yuan S., et al. 2021. “Carvacrol Alleviates Liver Fibrosis by Inhibiting Trpm7 and Modulating the Mapk Signaling Pathway.” European Journal of Pharmacology 898: 173982. 10.1016/j.ejphar.2021.173982. [DOI] [PubMed] [Google Scholar]
- Çakır, F. , Ateşoğlu S., Müderrisoğlu Z. R., et al. 2025. “Targeting Lung Cancer With Carvacrol‐Triazole‐Arylidenehydrazide Hybrids: In Vitro and In Silico Cytotoxicity Assessments.” Chemistry & Biodiversity 22, no. 6: e202402963. 10.1002/cbdv.202402963. [DOI] [PubMed] [Google Scholar]
- Çalibaşi Koçal, G. İ. Z. E. M. , and Pakdemirli A.. 2020. “Antiproliferative Effects of Carvacrol on Neuroblastoma Cells.” İzmir Dr. Behçet Uz Çocuk Hastanesi Dergisi 10, no. 1: 61–64. 10.5222/buchd.2020.59251. [DOI] [Google Scholar]
- Cerrah, S. , Ozcicek F., Gundogdu B., et al. 2023. “Carvacrol Prevents Acrylamide‐Induced Oxidative and Inflammatory Liver Damage and Dysfunction in Rats.” Frontiers in Pharmacology 14: 1161448. 10.3389/fphar.2023.1161448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chamseddine, R. S. , Wang C., Yin K., et al. 2022. “Penetrance of Male Breast Cancer Susceptibility Genes: A Systematic Review.” Breast Cancer Research and Treatment 191, no. 1: 31–38. 10.1007/s10549-021-06413-2. [DOI] [PubMed] [Google Scholar]
- Chen, H. H. , Zhou J. H., and Fong S. Y.. 2024. “Carvacrol Induces Doxorubicin‐Resistant Breast Cancer Cell Apoptosis via Inhibition of the PI3K/AKT Pathway.” Chinese Journal of Integrative Medicine 27, no. 9: 680–687. [Google Scholar]
- Chen, W. L. , Barszczyk A., Turlova E., et al. 2015. “Inhibition of TRPM7 by Carvacrol Suppresses Glioblastoma Cell Proliferation, Migration and Invasion.” Oncotarget 6, no. 18: 16321. 10.18632/oncotarget.3872. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen, Y. , Ba L., Huang W., et al. 2017. “Role of Carvacrol in Cardioprotection Against Myocardial Ischemia/Reperfusion Injury in Rats Through Activation of Mapk/Erk and Akt/Enos Signaling Pathways.” European Journal of Pharmacology 796: 90–100. [DOI] [PubMed] [Google Scholar]
- Chen, Y. H. , Xu N. Z., Hong C., et al. 2022. “Myo1b Promotes Tumor Progression and Angiogenesis by Inhibiting Autophagic Degradation of Hif‐1α in Colorectal Cancer.” Cell Death and Disease 13, no. 11: 939. 10.1038/s41419-022-05397-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chopra, A. S. , Lordan R., Horbańczuk O. K., et al. 2022. “The Current Use and Evolving Landscape of Nutraceuticals.” Pharmacological Research 175: 106001. [DOI] [PubMed] [Google Scholar]
- Coccimiglio, J. , Alipour M., Jiang Z. H., Gottardo C., and Suntres Z.. 2016. “Antioxidant, Antibacterial, and Cytotoxic Activities of the Ethanolic Origanum vulgare Extract and Its Major Constituents.” Oxidative Medicine and Cellular Longevity 2016: 1404505. 10.1155/2016/1404505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dai, W. , Sun C., Huang S., and Zhou Q.. 2016. “Carvacrol Suppresses Proliferation and Invasion in Human Oral Squamous Cell Carcinoma.” Oncotargets and Therapy 9: 2297–2304. 10.2147/OTT.S98875. [DOI] [PMC free article] [PubMed] [Google Scholar]
- De La Chapa, J. J. , Singha P. K., Lee D. R., and Gonzales C. B.. 2018. “Thymol Inhibits Oral Squamous Cell Carcinoma Growth via Mitochondria‐Mediated Apoptosis.” Journal of Oral Pathology and Medicine 47, no. 7: 674–682. 10.1111/jop.12735. [DOI] [PMC free article] [PubMed] [Google Scholar]
- de Lima, E. J. , Fontes S. S., Nogueira M. L., et al. 2020. “Essential Oil From Leaves of Conobea Scoparioides (Cham. & Schltdl.) Benth.(Plantaginaceae) Causes Cell Death in HepG2 Cells and Inhibits Tumor Development in a Xenograft Model.” Biomedicine & Pharmacotherapy 129: 110402. 10.1016/j.biopha.2020.110402. [DOI] [PubMed] [Google Scholar]
- De Souza, R. L. , de Oliveira Melo C., Opretzka L. C. F., Wândega E. L., Villarreal C. F., and Oliveira E. E.. 2023. “Nanoemulsion Improves the Anti‐Inflammatory Activity of Carvacrol Upon Oral Administration.” Revista Brasileira de Farmacognosia 33, no. 1: 164–172. 10.1007/s43450-022-00355-6. [DOI] [Google Scholar]
- Demirbolat, İ. , Kulabaş N., Gürboğa M., et al. 2022. “Synthesis and Evaluation of Antiproliferative and mPGES‐1 Inhibitory Activities of Novel Carvacrol‐Triazole Conjugates.” Organic Communications 15, no. 4: 356–377. 10.25135/acg.oc.142.2212.2651. [DOI] [Google Scholar]
- Dinarello, A. , Betto R. M., Diamante L., et al. 2023. “STAT3 and HIF1α Cooperatively Mediate the Transcriptional and Physiological Responses to Hypoxia.” Cell Death Discovery 9, no. 1: 226. 10.1038/s41420-023-01507-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- dos Santos, J. G. , Biavatti M., Mansani G. C., Cruz L. S., and Kanunfre C. C.. 2021. “Effect of Thymol Vapor Phase on Lung Cancer Cells Exposed to Radiation.” Brazilian Journal of Development 7, no. 7: 65323–65334. 10.34117/bjdv7n7-009. [DOI] [Google Scholar]
- Dubey, A. , Ghosh N. S., Agnihotri N., Kumar A., Pandey M., and Nishad S.. 2022. “Herbs Derived Bioactive Compounds and Their Potential for the Treatment of Neurological Disorders.” Clinical Schizophrenia and Related Psychoses 16, no. 2: 1–11. 10.3371/CSRP.DANG.081922. [DOI] [Google Scholar]
- Elahi, M. G. , Hekmati M., Esmaeili D., Ziarati P., and Yousefi M.. 2022. “Evaluation and Efficacy Modified Carvacrol and Anti‐Cancer Peptide Against Cell Line Gastric AGS.” International Journal of Peptide Research and Therapeutics 28, no. 4: 125. 10.1007/s10989-022-10426-w. [DOI] [Google Scholar]
- Elbe, H. , Yigitturk G., Cavusoglu T., Baygar T., Ozgul Onal M., and Ozturk F.. 2020. “Comparison of Ultrastructural Changes and the Anticarcinogenic Effects of Thymol and Carvacrol on Ovarian Cancer Cells: Which Is More Effective?” Ultrastructural Pathology 44, no. 2: 193–202. 10.1080/01913123.2020.1740366. [DOI] [PubMed] [Google Scholar]
- Elbe, H. , Yigitturk G., Cavusoglu T., Uyanikgil Y., and Ozturk F.. 2020. “Apoptotic Effects of Thymol, a Novel Monoterpene Phenol, on Different Types of Cancer.” Bratislava Medical Journal/Bratislavske Lekarske Listy 121, no. 2: 122–128. [DOI] [PubMed] [Google Scholar]
- Elshafie, H. S. , Armentano M. F., Carmosino M., Bufo S. A., De Feo V., and Camele I.. 2017. “Cytotoxic Activity of Origanum vulgare L. on Hepatocellular Carcinoma Cell Line HEPG2 and Evaluation of Its Biological Activity.” Molecules 22, no. 9: 1435. 10.3390/molecules22091435. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Escobar, A. , Perez M., Romanelli G., and Blustein G.. 2020. “Thymol Bioactivity: A Review Focusing on Practical Applications.” Arabian Journal of Chemistry 13, no. 12: 9243–9269. [Google Scholar]
- Fan, K. , Li X., Cao Y., et al. 2015. “Carvacrol Inhibits Proliferation and Induces Apoptosis in Human Colon Cancer Cells.” Anti‐Cancer Drugs 26, no. 8: 813–823. [DOI] [PubMed] [Google Scholar]
- Fekri Kohan, S. , Zamani H., and Salehzadeh A.. 2023. “Antibacterial Potential and Cytotoxic Activity of Iron Oxide Nanoparticles Conjugated With Thymol (Fe3O4@ Glu‐Thymol) on Breast Cancer Cells and Investigating the Expression of BAX, CASP8, and BCL‐2 Genes.” Biometals 36, no. 6: 1273–1284. 10.1007/s10534-023-00516-7. [DOI] [PubMed] [Google Scholar]
- Feyzmohamadi Khoramabadi, M. , Nemati A., and Homayouni Tabrizi M.. 2024. “Evaluation of the Toxicity and Anti‐Angiogenic Effects of Beta‐Cyclodextrin Complex Loaded With Thymol in Melanoma Skin Cancer Cells (A2058).” Feyz Medical Sciences Journal 28, no. 3: 261–271. [Google Scholar]
- Fischer, J. , Walker L. C., Robinson B. A., Frizelle F. A., Church J. M., and Eglinton T. W.. 2019. “Clinical Implications of the Genetics of Sporadic Colorectal Cancer.” ANZ Journal of Surgery 89, no. 10: 1224–1229. [DOI] [PubMed] [Google Scholar]
- Fitsiou, E. , Anestopoulos I., Chlichlia K., et al. 2016. “Antioxidant and Antiproliferative Properties of the Essential Oils of Satureja Thymbra and Satureja Parnassica and Their Major Constituents.” Anticancer Research 36, no. 11: 5757–5763. [DOI] [PubMed] [Google Scholar]
- Forqani, M. A. , Akbarian M., Amirahmadi S., Soukhtanloo M., Hosseini M., and Forouzanfar F.. 2023. “Carvacrol Improved Learning and Memory and Attenuated the Brain Tissue Oxidative Damage in Aged Male Rats.” International Journal of Neuroscience 134: 1242–1249. 10.1080/00207454.2023.2257877. [DOI] [PubMed] [Google Scholar]
- Garg, P. , Malhotra J., Kulkarni P., Horne D., Salgia R., and Singhal S. S.. 2024. “Emerging Therapeutic Strategies to Overcome Drug Resistance in Cancer Cells.” Cancers 16, no. 13: 2478. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ge, Y. , Fang X., Zhang Y., et al. 2023. “Improving the Stability and Anti‐l. Monocytogenes Activity of Thymol Casein Nanoparticles by Forming Eutectic Solvent With Lauric Acid.” LWT 189: 115493. 10.1016/j.lwt.2023.115493. [DOI] [Google Scholar]
- Ghorani, V. , Alavinezhad A., Rajabi O., and Boskabady M. H.. 2021. “Carvacrol Improves Pulmonary Function Tests, Oxidant/Antioxidant Parameters and Cytokine Levels in Asthmatic Patients: A Randomized, Double‐Blind, Clinical Trial.” Phytomedicine 85: 153539. 10.1016/j.phymed.2021.153539. [DOI] [PubMed] [Google Scholar]
- Ghorani, V. , Alavinezhad A., Rajabi O., Mohammadpour A. H., and Boskabady M. H.. 2021. “Safety and Tolerability of Carvacrol in Healthy Subjects: A Phase I Clinical Study.” Drug and Chemical Toxicology 44, no. 2: 177–189. [DOI] [PubMed] [Google Scholar]
- Glaviano, A. , Foo A. S., Lam H. Y., et al. 2023. “PI3K/AKT/mTOR Signaling Transduction Pathway and Targeted Therapies in Cancer.” Molecular Cancer 22, no. 1: 138. 10.1186/s12943-023-01827-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gong, S. , Wang D., Tao S., et al. 2021. “Facile Encapsulation of Thymol Within Deamidated Zein Nanoparticles for Enhanced Stability and Antibacterial Properties.” Colloids and Surfaces A: Physicochemical and Engineering Aspects 626: 126940. 10.1016/j.colsurfa.2021.126940. [DOI] [Google Scholar]
- Govindaraju, S. , and Arulselvi P. I.. 2018. “Characterization of Coleus Aromaticus Essential Oil and Its Major Constituent Carvacrol for In Vitro Antidiabetic and Antiproliferative Activities.” Journal of Herbs, Spices & Medicinal Plants 24, no. 1: 37–51. [Google Scholar]
- Greten, F. R. , and Grivennikov S. I.. 2019. “Inflammation and Cancer: Triggers, Mechanisms, and Consequences.” Immunity 51, no. 1: 27–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gunes, C. E. , Secme M., Kurar E., and Donmez H.. 2022. “Apoptotic and Anti‐Metastatic Effect of Carvacrol in PANC‐1 Human Pancreatic Cancer Cells.” Natural Products and Biotechnology 2, no. 1: 42–50. [Google Scholar]
- Günes‐Bayir, A. , Kocyigit A., Güler E. M., Bilgin M. G., Ergün I. S., and Dadak A.. 2018. “Effects of Carvacrol on Human Fibroblast (WS‐1) and Gastric Adenocarcinoma (AGS) Cells In Vitro and on Wistar Rats In Vivo.” Molecular and Cellular Biochemistry 448: 237–249. 10.1007/s11010-018-3329-5. [DOI] [PubMed] [Google Scholar]
- Guo, Y. J. , Pan W. W., Liu S. B., Shen Z. F., Xu Y., and Hu L. L.. 2020. “ERK/MAPK Signalling Pathway and Tumorigenesis.” Experimental and Therapeutic Medicine 19, no. 3: 1997–2007. 10.3892/etm.2020.8454. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hassan, H. F. H. , Mansour A. M., Salama S. A., and El‐Sayed E. S. M.. 2021. “The Chemopreventive Effect of Thymol Against Dimethylhydrazine and/or High Fat Diet‐Induced Colon Cancer in Rats: Relevance to NF‐κB.” Life Sciences 274: 119335. 10.1016/j.lfs.2021.119335. [DOI] [PubMed] [Google Scholar]
- He, K. , and Gan W. J.. 2023. “Wnt/β‐Catenin Signaling Pathway in the Development and Progression of Colorectal Cancer.” Cancer Management and Research 15: 435–448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hendrikse, C. S. E. , Theelen P. M. M., van der Ploeg P., et al. 2023. “The Potential of RAS/RAF/MEK/ERK (MAPK) Signaling Pathway Inhibitors in Ovarian Cancer: A Systematic Review and Meta‐Analysis.” Gynecologic Oncology 171: 83–94. 10.1016/j.ygyno.2023.01.038. [DOI] [PubMed] [Google Scholar]
- Hoca, M. , Becer E., and Vatansever H. S.. 2023. “Carvacrol Is Potential Molecule for Diabetes Treatment.” Archives of Physiology and Biochemistry 130: 1–8. 10.1080/13813455.2023.2288537. [DOI] [PubMed] [Google Scholar]
- Huang, J. , Chan S. C., Ngai C. H., et al. 2022. “Disease Burden, Risk Factors, and Trends of Leukaemia: A Global Analysis.” Frontiers in Oncology 12: 904292. 10.3389/fonc.2022.904292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang, J. , Lok V., Ngai C. H., et al. 2021. “Disease Burden, Risk Factors, and Recent Trends of Liver Cancer: A Global Country‐Level Analysis.” Liver Cancer 10, no. 4: 330–345. 10.1159/000515304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hussein, A. H. 2024. “Studying the Effect of Thymol‐Containing Nano Emulsion Extracted From Thyme on Angiogenesis Controlling Genes VEGF and VEGFR in a Human Liver Cancer Cell Line.” Baghdad Science Journal 22: 8. [Google Scholar]
- Hussein, R. M. , Arafa E. S. A., Raheem S. A., and Mohamed W. R.. 2023. “Thymol Protects Against Bleomycin‐Induced Pulmonary Fibrosis via Abrogation of Oxidative Stress, Inflammation, and Modulation of miR‐29a/TGF‐β and PI3K/AKT Signaling in Mice.” Life Sciences 314: 121256. 10.1016/j.lfs.2022.121256. [DOI] [PubMed] [Google Scholar]
- Imran, M. , Aslam M., Alsagaby S. A., et al. 2022. “Therapeutic Application of Carvacrol: A Comprehensive Review.” Food Science and Nutrition 10, no. 11: 3544–3561. 10.1002/fsn3.2994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Islam, M. T. , Bappi M. H., Bhuia M. S., et al. 2024. “Anti‐Inflammatory Effects of Thymol: An Emphasis on the Molecular Interactions Through In Vivo Approach and Molecular Dynamic Simulations.” Frontiers in Chemistry 12: 1376783. 10.3389/fchem.2024.1376783. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jaafari, A. , Tilaoui M., Mouse H. A., et al. 2012. “Comparative Study of the Antitumor Effect of Natural Monoterpenes: Relationship to Cell Cycle Analysis.” Revista Brasileira de Farmacognosia 22: 534–540. 10.1590/S0102-695X2012005000021. [DOI] [Google Scholar]
- Jamali, T. , Kavoosi G., and Ardestani S. K.. 2020. “In‐Vitro and In‐Vivo Anti‐Breast Cancer Activity of Oeo (Oliveria Decumbens Vent Essential Oil) Through Promoting the Apoptosis and Immunomodulatory Effects.” Journal of Ethnopharmacology 248: 112313. 10.1016/j.jep.2019.112313. [DOI] [PubMed] [Google Scholar]
- Jamali, T. , Kavoosi G., Safavi M., and Ardestani S. K.. 2018. “In‐Vitro Evaluation of Apoptotic Effect of Oeo and Thymol in 2d and 3d Cell Cultures and the Study of Their Interaction Mode With DNA.” Scientific Reports 8, no. 1: 15787. 10.1038/s41598-018-34055-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jamshidi, H. R. , and Taheri F.. 2021. “The Effect of Thymol on Renal Toxicity Induced by Mercury Chloride in Rats.” International Journal of Medical Laboratory 8, no. 3: 223–232. [Google Scholar]
- Janani, B. , Vijayakumar M., Priya K., et al. 2022. “EGFR‐Based Targeted Therapy for Colorectal Cancer—Promises and Challenges.” Vaccine 10, no. 4: 499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jayakumar, S. , Madankumar A., Asokkumar S., et al. 2012. “Potential Preventive Effect of Carvacrol Against Diethylnitrosamine‐Induced Hepatocellular Carcinoma in Rats.” Molecular and Cellular Biochemistry 360: 51–60. 10.1007/s11010-011-1043-7. [DOI] [PubMed] [Google Scholar]
- Jia, Y. , Xing Y., and Yang M.. 2021. “Efficacy of Sorafenib Combined With Interventional Therapy on Primary Liver Cancer Patients and Its Effect on Serum AFP, VEGF, and GGT.” Journal of Oncology 2021, no. 1: 9120265. 10.1155/2021/9120265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Joshi, S. , Kundu S., Priya V. V., Kulhari U., Mugale M. N., and Sahu B. D.. 2023. “Anti‐Inflammatory Activity of Carvacrol Protects the Heart From Lipopolysaccharide‐Induced Cardiac Dysfunction by Inhibiting Pyroptosis via NLRP3/Caspase1/Gasdermin D Signaling Axis.” Life Sciences 324: 121743. 10.1016/j.lfs.2023.121743. [DOI] [PubMed] [Google Scholar]
- Jung, C. Y. , Kim S. Y., and Lee C.. 2018. “Carvacrol Targets AXL to Inhibit Cell Proliferation and Migration in Non‐Small Cell Lung Cancer Cells.” Anticancer Research 38, no. 1: 279–286. [DOI] [PubMed] [Google Scholar]
- Kenzerki, M. E. , Ahmadi M., Mousavi P., and Ghafouri‐Fard S.. 2023. “MYC and Non‐Small Cell Lung Cancer: A Comprehensive Review.” Human Genetics 37: 201185. 10.1016/j.humgen.2023.201185. [DOI] [Google Scholar]
- Keshavarz, F. , Soltanshahi M., Khosravani F., et al. 2024. “Thymol‐Loaded Liposomes Effectively Induced Apoptosis and Decreased EGFR Expression in Colorectal Cancer Cells.” Naunyn‐Schmiedeberg's Archives of Pharmacology 397, no. 7: 5157–5165. 10.1007/s00210-024-02945-8. [DOI] [PubMed] [Google Scholar]
- Khan, F. , Khan I., Farooqui A., and Ansari I. A.. 2017. “Carvacrol Induces Reactive Oxygen Species (ROS)‐Mediated Apoptosis Along With Cell Cycle Arrest at G0/G1 in Human Prostate Cancer Cells.” Nutrition and Cancer 69, no. 7: 1075–1087. 10.1080/01635581.2017.1359321. [DOI] [PubMed] [Google Scholar]
- Khan, F. , Pandey P., Verma M., and Upadhyay T. K.. 2024. “Terpenoid‐Mediated Targeting of STAT3 Signaling in Cancer: An Overview of Preclinical Studies.” Biomolecules 14, no. 2: 200. 10.3390/biom14020200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Khan, F. , Singh V. K., Saeed M., Kausar M. A., and Ansari I. A.. 2019. “Carvacrol Induced Program Cell Death and Cell Cycle Arrest in Androgen‐Independent Human Prostate Cancer Cells via Inhibition of Notch Signaling.” Anti‐Cancer Agents in Medicinal Chemistry 19, no. 13: 1588–1608. [DOI] [PubMed] [Google Scholar]
- Khan, I. , Bahuguna A., Bhardwaj M., Khaket T. P., and Kang S. C.. 2018. “Carvacrol Nanoemulsion Evokes Cell Cycle Arrest, Apoptosis Induction and Autophagy Inhibition in Doxorubicin Resistant‐A549 Cell Line.” Artificial Cells, Nanomedicine, and Biotechnology 46, no. 1: 664–675. 10.1080/21691401.2018.1434187. [DOI] [PubMed] [Google Scholar]
- Khazdair, M. R. , Moshtagh M., Anaeigoudari A., Jafari S., and Kazemi T.. 2024. “Protective Effects of Carvacrol on Lipid Profiles, Oxidative Stress, Hypertension, and Cardiac Dysfunction–A Comprehensive Review.” Food Science and Nutrition 12, no. 5: 3137–3149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Khwaza, V. , Oyedeji O. O., Oselusi S. O., et al. 2023. “Synthesis of Ester‐Linked Ursolic Acid‐Based Hybrid Compounds: Potential Antibacterial and Anticancer Agents.” Chemistry and Biodiversity 20, no. 4: e202300034. 10.1002/cbdv.202300034. [DOI] [PubMed] [Google Scholar]
- Kubatka, P. , Uramova S., Kello M., et al. 2019. “Anticancer Activities of Thymus vulgaris L. in Experimental Breast Carcinoma In Vivo and In Vitro.” International Journal of Molecular Sciences 20, no. 7: 1749. 10.3390/ijms20071749. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Laamari, Y. , Bimoussa A., Chagaleti B. K., et al. 2025. “Thymol‐1, 2, 3‐Triazole Derivatives: Network Pharmacology, Molecular Simulations and Synthesis Targeting Breast Cancer.” Journal of Molecular Structure 1321: 140060. 10.1016/j.molstruc.2024.140060. [DOI] [Google Scholar]
- Laamari, Y. , Bimoussa A., Fawzi M., et al. 2023. “Synthesis, Crystal Structure and Evaluation of Anticancer Activities of Some Novel Heterocyclic Compounds Based on Thymol.” Journal of Molecular Structure 1278: 134906. 10.1016/j.molstruc.2023.134906. [DOI] [Google Scholar]
- Laamari, Y. , Fawzi M., Hachim M. E., et al. 2024. “Synthesis, Characterization and Cytotoxic Activity of Pyrazole Derivatives Based on Thymol.” Journal of Molecular Structure 1297: 136864. 10.1016/j.molstruc.2023.136864. [DOI] [Google Scholar]
- Lashkari, M. , Fatemi A., Valandani H. M., and Khalilabadi R. M.. 2022. “Promising Anti‐Leukemic Effect of Zataria Multiflora Extract in Combination With Doxorubicin to Combat Acute Lymphoblastic Leukemia Cells (Nalm‐6) (In Vitro and In Silico).” Scientific Reports 12, no. 1: 12657. 10.1038/s41598-022-16943-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lázaro, S. , Lorz C., Enguita A. B., Seller I., Paramio J. M., and Santos M.. 2022. “Pten and P53 Loss in the Mouse Lung Causes Adenocarcinoma and Sarcomatoid Carcinoma.” Cancers 14, no. 15: 3671. 10.3390/cancers14153671. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li, L. , He L., Wu Y., and Zhang Y.. 2021. “Carvacrol Affects Breast Cancer Cells Through TRPM7 Mediated Cell Cycle Regulation.” Life Sciences 266: 118894. 10.1016/j.lfs.2020.118894. [DOI] [PubMed] [Google Scholar]
- Li, S. , Zhao H., and Bao L.. 2019. “Preliminary Study on the Mechanism of Carvacrol Regulating Hepatocellular Carcinoma Based on Network Pharmacology.” Letters in Drug Design and Discovery 16, no. 11: 1286–1295. 10.2174/1570180816666190516105906. [DOI] [Google Scholar]
- Li, Y. , Wen J. M., Du C. J., et al. 2017. “Thymol Inhibits Bladder Cancer Cell Proliferation via Inducing Cell Cycle Arrest and Apoptosis.” Biochemical and Biophysical Research Communications 491, no. 2: 530–536. 10.1016/j.bbrc.2017.04.009. [DOI] [PubMed] [Google Scholar]
- Liang, W. Z. , Chou C. T., Lu T., et al. 2013. “The Mechanism of Carvacrol‐Evoked [Ca2+] i Rises and Non‐Ca2+‐Triggered Cell Death in OC2 Human Oral Cancer Cells.” Toxicology 303: 152–161. 10.1016/j.tox.2012.10.026. [DOI] [PubMed] [Google Scholar]
- Liang, W. Z. , and Lu C. H.. 2012. “Carvacrol‐Induced [Ca2+] i Rise and Apoptosis in Human Glioblastoma Cells.” Life Sciences 90, no. 17–18: 703–711. [DOI] [PubMed] [Google Scholar]
- Lim, W. , Ham J., Bazer F. W., and Song G.. 2019. “Carvacrol Induces Mitochondria‐Mediated Apoptosis via Disruption of Calcium Homeostasis in Human Choriocarcinoma Cells.” Journal of Cellular Physiology 234, no. 2: 1803–1815. [DOI] [PubMed] [Google Scholar]
- Llana‐Ruiz‐Cabello, M. , Gutiérrez‐Praena D., Pichardo S., et al. 2014. “Cytotoxicity and Morphological Effects Induced by Carvacrol and Thymol on the Human Cell Line Caco‐2.” Food and Chemical Toxicology 64: 281–290. [DOI] [PubMed] [Google Scholar]
- Lo, Y. M. 2022. “The ART and LOGIC of Scholarly Communication: Effective Skills for Publication and Beyond.” Food Science and Nutrition 10, no. 4: 981–984. 10.1002/fsn3.2766. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Logesh, R. , Alam W., Alsharif K. F., Albrakati A., Akkol E. K., and Khan H.. 2024. “Role of Terpenes in Regulating Leukemia: Mechanistic and Clinical Updates.” Minerva Biotechnology and Biomolecular Research 36, no. 3: 130–143. [Google Scholar]
- Luo, Y. , Fu X., Ru R., et al. 2020. “CpG Oligodeoxynucleotides Induces Apoptosis of Human Bladder Cancer Cells via Caspase‐3‐Bax/Bcl‐2‐p53 Axis.” Archives of Medical Research 51, no. 3: 233–244. [DOI] [PubMed] [Google Scholar]
- Luo, Y. , Wu J. Y., Lu M. H., Shi Z., Na N., and Di J. M.. 2016. “Carvacrol Alleviates Prostate Cancer Cell Proliferation, Migration, and Invasion Through Regulation of PI3K/AKT and MAPK Signaling Pathways.” Oxidative Medicine and Cellular Longevity 2016: 1469693. 10.1155/2016/1469693. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lv, R. , and Chen Z.. 2017. “Thymol Inhibits Cell Migration and Invasion by Downregulating the Activation of PI3K/AKT and ERK Pathways in Human Colon Cancer Cells.” Tropical Journal of Pharmaceutical Research 16, no. 12: 2895–2901. 10.4314/tjpr.v16i12.13. [DOI] [Google Scholar]
- Mahran, Y. F. , Badr A. M., Aldosari A., Bin‐Zaid R., and Alotaibi H. N.. 2019. “Carvacrol and Thymol Modulate the CROSs‐Talk Between TNF‐α and IGF‐1 Signaling in Radiotherapy‐Induced Ovarian Failure.” Oxidative Medicine and Cellular Longevity 2019: 3173745. 10.1155/2019/3173745. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mari, A. , Mani G., Nagabhishek S. N., et al. 2021. “Carvacrol Promotes Cell Cycle Arrest and Apoptosis Through PI3K/AKT Signaling Pathway in MCF‐7 Breast Cancer Cells.” Chinese Journal of Integrative Medicine 27: 680–687. 10.1007/s11655-020-3193-5. [DOI] [PubMed] [Google Scholar]
- Martirosyan, D. , Jahanbakhshi F., Ashoori M. R., et al. 2022. “Effect of Oral Administration and Topical Gel Application of Thymol and Low‐Level Laser Therapy on Oxidative Stress, Inflammatory Biomarkers and Dermatitis in Patients With Type 2 Diabetes Mellitus.” Bioactive Compounds in Health and Disease 5, no. 4: 93–105. [Google Scholar]
- Matthews, H. K. , Bertoli C., and de Bruin R. A.. 2022. “Cell Cycle Control in Cancer.” Nature Reviews Molecular Cell Biology 23, no. 1: 74–88. [DOI] [PubMed] [Google Scholar]
- Mbese, Z. , Nell M., Fonkui Y. T., Ndinteh D. T., Steenkamp V., and Aderibigbe B. A.. 2022. “Hybrid Compounds Containing Carvacrol Scaffold: In Vitro Antibacterial and Cytotoxicity Evaluation.” Recent Advances in Anti‐Infective Drug Discovery 17, no. 1: 54–68. [DOI] [PubMed] [Google Scholar]
- McClements, D. J. 2024. “Surface Modified Carvacrol‐Rich Satureja Khuzestanica Essential Oil Nanoemulsion: A Novel Paclitaxel Formulation Induced Apoptosis on Paclitaxel‐Resistant Breast Cancer Cells.” Pharmaceutical Sciences 30: 85–97. 10.34172/PS.2023.22. [DOI] [Google Scholar]
- Mehdi, S. J. , Ahmad A., Irshad M., Manzoor N., and Rizvi M. M. A.. 2011. “Cytotoxic Effect of Carvacrol on Human Cervical Cancer Cells.” Biology and Medicine 3, no. 2: 307–312. [Google Scholar]
- Mehta, N. , Kumar P., Verma A. K., et al. 2022. “Ultrasound‐Assisted Extraction and the Encapsulation of Bioactive Components for Food Applications.” Food 11, no. 19: 2973. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moayeri, A. , Mohammadpour S., Aidy A., Karimi E., and Ghaneialvar H.. 2021. “The Effect of Thymbra Spicata Extract and Its Bioactive Component Thymol on Non‐Small‐Cells Lung Cancer Cell Line A549.” Archives of Medical Science. 10.5114/aoms/136485. [DOI] [Google Scholar]
- Moradipour, A. , Dariushnejad H., Ahmadizadeh C., and Lashgarian H. E.. 2022. “Dietary Flavonoid Carvacrol Triggers the Apoptosis of Human Breast Cancer MCF‐7 Cells via the p53/Bax/Bcl‐2 Axis.” Medical Oncology 40, no. 1: 46. [DOI] [PubMed] [Google Scholar]
- Nardin, S. , Mora E., Varughese F. M., et al. 2020. “Breast Cancer Survivorship, Quality of Life, and Late Toxicities.” Frontiers in Oncology 10: 864. 10.3389/fonc.2020.00864. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Niaz, T. , Imran M., and Mackie A.. 2021. “Improving Carvacrol Bioaccessibility Using Core–Shell Carrier‐Systems Under Simulated Gastrointestinal Digestion.” Food Chemistry 353: 129505. [DOI] [PubMed] [Google Scholar]
- Osanloo, M. , Alipanah H., Farjam M., Taheri A., and Zarenezhad E.. 2023. “Anticancer Activity of Chitosan Nanoparticles Containing Satureja Khuzistanica Essential Oil, and Carvacrol Against Human Melanoma and Breast Cancer.” Russian Journal of Bioorganic Chemistry 49, no. 3: 594–601. [Google Scholar]
- Osarieme Imade, R. , Adesina Ayinde B., and Alam A.. 2023. “GC‐MS Analysis and In Vitro Cytotoxic Effects of Ocimum gratissimum (Lamiaceae) Volatile Oil and Thymol on Cancer Cells.” Pharmaceutical and Biomedical Research 9, no. 2: 115–124. 10.32598/PBR.9.2.1110.1. [DOI] [Google Scholar]
- Othman, M. S. , Aboelnaga S. M., Habotta O. A., Moneim A. E. A., and Hussein M. M.. 2023. “The Potential Therapeutic Role of GreenSynthesized Selenium Nanoparticles Using Carvacrol in Human Breast Cancer MCF‐7 Cells.” Applied Sciences 13, no. 12: 7039. [Google Scholar]
- Ozkan, A. , and Erdogan A.. 2012. “A Comparative Study of the Antioxidant/Prooxidant Effects of Carvacrol and Thymol at Various Concentrations on Membrane and DNA of Parental and Drug Resistant H1299 Cells.” Natural Product Communications 7, no. 12: 1557–1560. [PubMed] [Google Scholar]
- Özmen, H. K. , Akdemir F. N. E., Tanyeli A., et al. 2021. “Thymol May Be an Effective Agent in the Treatment of Liver and Kidney Damages Caused by Ionizing Radiation.” Turkish Journal of Oncology 36, no. 1: 71–78. 10.5505/tjo.2020.2556. [DOI] [Google Scholar]
- Pakdemirli, A. , Karaca C., Sever T., et al. 2020. “Carvacrol Alters Soluble Factors in HCT‐116 and HT‐29 Cell Lines.” Turkish Journal of Medical Sciences 50, no. 1: 271–276. 10.3906/sag-1907-173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pandey, P. , Ramniwas S., Verma M., et al. 2024. “A Study to Investigate the Anticancer Potential of Carvacrol via Targeting Notch Signaling in Breast Cancer.” Open Chemistry 22, no. 1: 20240008. 10.1515/chem-2024-0008. [DOI] [Google Scholar]
- Phyo, H. M. , Al‐Maqtari Q. A., Othman N., et al. 2024. “Ultrasonication‐Enhanced Nanoencapsulation of Thymol and Cinnamaldehyde With Chitosan and Hydroxypropyl Methylcellulose: A Novel Approach to Improving Physicochemical Properties, Stability, and Bioactive Content.” Food and Bioprocess Technology 18, no. 3: 2925–2948. 10.1016/j.cej.2024.151430. [DOI] [Google Scholar]
- Potočnjak, I. , and Domitrović R.. 2016. “Carvacrol Attenuates Acute Kidney Injury Induced by Cisplatin Through Suppression of ERK and PI3K/AKT Activation.” Food and Chemical Toxicology 98: 251–261. 10.1016/j.fct.2016.11.004. [DOI] [PubMed] [Google Scholar]
- Pouyamanesh, G. , Ameli N., Metanat Y., et al. 2024. “Thymol Enhances 5‐Fluorouracil Cytotoxicity by Reducing Migration and Increasing Apoptosis and Cell Cycle Arrest in Esophageal Cancer Cells: An In‐Vitro Study.” Indian Journal of Clinical Biochemistry 40: 1–12. 10.1007/s12291-024-01219-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Qoorchi Moheb Seraj, F. , Heravi‐Faz N., Soltani A., et al. 2022. “Thymol Has Anticancer Effects in U‐87 Human Malignant Glioblastoma Cells.” Molecular Biology Reports 49, no. 10: 9623–9632. 10.1007/s11033-022-07867-3. [DOI] [PubMed] [Google Scholar]
- Rao, S. Q. , Zhang R. Y., Gao X. R., et al. 2024. “Formation Mechanism, Environmental Stability and Antibacterial Activity of Succinylated Ovalbumin/Ε‐Polylysine Nanogel Loaded With Thymol: The Synergistic Roles of Succinylation Modification and Ε‐Polylysine.” Industrial Crops and Products 220: 119192. 10.1016/j.indcrop.2024.119192. [DOI] [Google Scholar]
- Razack, S. A. , Kim Y. E., and Kang H. W.. 2025. “κ‐Carrageenan–Gelatin Hydrogel Embedding Carvacrol Loaded Gold Nanobipyramids for Treating Prostate Cancer via Fractionated Photothermal‐Chemotherapy.” International Journal of Biological Macromolecules 291: 138974. 10.1016/j.ijbiomac.2024.138974. [DOI] [PubMed] [Google Scholar]
- Retnosari, R. , Abdul Ghani M. A., Alkharji M. M., et al. 2024. “The Protective Effects of Carvacrol Against Doxorubicin‐Induced Cardiotoxicity In Vitro and In Vivo.” Cardiovascular Toxicology 25: 1–15. 10.1007/s12012-024-09940-8. [DOI] [PubMed] [Google Scholar]
- Ridaoui, K. , Ziyadi S., Abdou A., et al. 2024. “Purification, Chemical Characterization and Evaluation of the Antioxidant Potential of Carvacrol From Thymus vulgaris .” Journal of Essential Oil Bearing Plants 27, no. 1: 251–262. [Google Scholar]
- Rojas‐Armas, J. P. , Arroyo‐Acevedo J. L., Palomino‐Pacheco M., et al. 2020. “The Essential Oil of Cymbopogon citratus Stapt and Carvacrol: An Approach of the Antitumor Effect on 7, 12‐Dimethylbenz‐[Α]‐Anthracene (Dmba)‐Induced Breast Cancer in Female Rats.” Molecules 25, no. 14: 3284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sachan, N. , Saraswat N., Chandra P., Khalid M., and Kabra A.. 2022. “Research Article Isolation of Thymol From Trachyspermum ammi Fruits for Treatment of Diabetes and Diabetic Neuropathy in STZ‐Induced Rats.” 2022: 8263999. 10.1155/2022/8263999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sahin, D. , Kepekci R. A., Türkmenoğlu B., and Akkoc S.. 2023. “Biological Evaluations and Computational Studies of Newly Synthesized Thymol‐Based Schiff Bases as Anticancer, Antimicrobial and Antioxidant Agents.” Journal of Biomolecular Structure and Dynamics 43: 3375–3389. 10.1080/07391102.2023.2222833. [DOI] [PubMed] [Google Scholar]
- Sajid, A. , Manzoor Q., Sajid A., et al. 2025. “In Vitro and In Silico Studies of Terpenes Extracted From Cymbopogon Flexuous Leaves Against Human Myeloid Leukemia as an Inhibitor of Nf‐Κb Activation Signaling Pathway.” Journal of Molecular Structure 1321: 139675. 10.1007/s12672-024-01716-4. [DOI] [Google Scholar]
- Sakat, M. S. , Kılıç K., Sahin A., Kiziltunc Ozmen H., Yıldırım S., and Egilmez E.. 2023. “The Protective Efficacy of Hesperidin and Thymol on Radiation‐Induced Submandibular Gland Damage.” Laryngoscope 133, no. 8: 1885–1892. 10.1002/lary.30405. [DOI] [PubMed] [Google Scholar]
- Sampaio, L. A. , Pina L. T. S., Serafini M. R., Tavares D. D. S., and Guimaraes A. G.. 2021. “Antitumor Effects of Carvacrol and Thymol: A Systematic Review.” Frontiers in Pharmacology 12: 702487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Seçme, M. , and İlhan H.. 2025. “Synthesis and Characterization of Thymol Carbon Nanodot Functionalized Silver Nanoparticles (ThCND‐AgNPs) and Evaluation of Their Antiproliferative, Anti‐Invasive, and Apoptotic Effects on OVCAR‐3 Ovarian Cancer Cells.” Microscopy Research and Technique 88, no. 3: 668–677. 10.1002/jemt.24724. [DOI] [PubMed] [Google Scholar]
- Shakeel, I. , Haider S., Khan S., et al. 2024. “Thymoquinone, Artemisinin, and Thymol Attenuate Proliferation of Lung Cancer Cells as Sphingosine Kinase 1 Inhibitors.” Biomedicine and Pharmacotherapy 177: 117123. 10.1016/j.biopha.2024.117123. [DOI] [PubMed] [Google Scholar]
- Singh, V. , Khurana A., Navik U., Allawadhi P., Bharani K. K., and Weiskirchen R.. 2022. “Apoptosis and Pharmacological Therapies for Targeting Thereof for Cancer Therapeutics.” Sciences 4, no. 2: 15. [Google Scholar]
- Singhal, B. , Pandey P., Khan F., Singh S. K., and Kumar T.. 2021. “In Vitro Elucidation of Antiproliferative and Apoptotic Effects of Thymol Against Prostate Cancer LNCAP Cells.” Biointerface Research in Applied Chemistry 12: 1279–1289. [Google Scholar]
- Sisto, F. , Carradori S., Guglielmi P., et al. 2020. “Synthesis and Bological Evaluation of Carvacrol‐Based Derivatives as Dual Inhibitors of H. pylori Strains and AGS Cell Proliferation.” Pharmaceuticals 13, no. 11: 405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sivaranjani, A. , Sivagami G., and Nalini N.. 2016. “Chemopreventive Effect of Carvacrol on 1, 2‐Dimethylhydrazine Induced Experimental Colon Carcinogenesis.” Journal of Cancer Research and Therapeutics 12, no. 2: 755–762. [DOI] [PubMed] [Google Scholar]
- Smolarz, B. , Nowak A. Z., and Romanowicz H.. 2022. “Breast Cancer—Epidemiology, Classification, Pathogenesis and Treatment (Review of Literature).” Cancers 14, no. 10: 2569. 10.3390/cancers14102569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Srinivasan, M. K. , and Namasivayam N.. 2024. “Evaluating the In Vitro and In Vivo Effects of Carvacrol Zinc Oxide Quantum Dots in Breast Cancer.” Journal of Biomaterials Science Polymer Edition 36: 1–20. [DOI] [PubMed] [Google Scholar]
- Subramaniyan, J. , Krishnan G., Balan R., et al. 2014. “Carvacrol Modulates Instability of Xenobiotic Metabolizing Enzymes and Downregulates the Expressions of PCNA, MMP‐2, and MMP‐9 During DiethylnitROSamine‐Induced Hepatocarcinogenesis in Rats.” Molecular and Cellular Biochemistry 395: 65–76. 10.1007/s11010-014-2112-5. [DOI] [PubMed] [Google Scholar]
- Szostek, T. , Szulczyk D., Szymańska‐Majchrzak J., et al. 2022. “Design and Synthesis of Menthol and Thymol Derived Ciprofloxacin: Influence of Structural Modifications on the Antibacterial Activity and Anticancer Properties.” International Journal of Molecular Sciences 23, no. 12: 6600. 10.3390/ijms23126600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tabassum, H. , and Ahmad I. Z.. 2020. “Molecular Docking and Dynamics Simulation Analysis of Thymoquinone and Thymol Compounds From Nigella sativa L. That Inhibits P38 Protein: Probable Remedies for Hepatocellular Carcinoma.” Medicinal Chemistry 16, no. 3: 350–357. 10.2174/1573406415666190416165732. [DOI] [PubMed] [Google Scholar]
- Taibi, M. , Elbouzidi A., Haddou M., et al. 2024. “Evaluation of the Interaction Between Carvacrol and Thymol, Major Compounds of Ptychotis Verticillata Essential Oil: Antioxidant, Anti‐Inflammatory and Anticancer Activities Against Breast Cancer Lines.” Life 14, no. 8: 1037. 10.3390/life14081037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tang, Z. Y. , Li Y., Tang Y. T., Ma X. D., and Tang Z. Y.. 2022. “Anticancer Activity of Oleanolic Acid and Its Derivatives: Recent Advances in Evidence, Target Profiling and Mechanisms of Action.” Biomedicine and Pharmacotherapy 145: 112397. 10.1016/j.biopha.2021.112397. [DOI] [PubMed] [Google Scholar]
- Tayarani‐Najaran, Z. , Akaberi M., Hassanzadeh B., et al. 2019. “Analysis of the Essential Oils of Five Artemisia Species and Evaluation of Their Cytotoxic and Pro‐Apoptotic Effects.” Mini‐Reviews in Medicinal Chemistry 19, no. 11: 902–912. 10.2174/1389557519666190311155021. [DOI] [PubMed] [Google Scholar]
- Tomsuk, Ö. , Kuete V., Sivas H., and Kürkçüoğlu M.. 2024. “Effects of Essential Oil of Origanum Onites and Its Major Component Carvacrol on the Expression of Toxicity Pathway Genes in HepG2 Cells.” BMC Complementary Medicine and Therapies 24, no. 1: 265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tópor Nunes, A. A. , Veras F. F., Cacciatore F. A., Silveira R. D., MalheiROS P. D. S., and Welke J. E.. 2024. “Nanoencapsulation With Eudragit and Chia Mucilage Increases the Stability and Antifungal Efficacy of Carvacrol Against Aspergillus spp.” Food Additives and Contaminants. Part A, Chemistry, Analysis, Control, Exposure & Risk Assessment 42, no. 1: 75–91. 10.1080/19440049.2024.2427670. [DOI] [PubMed] [Google Scholar]
- Torabiardekani, N. , Karami F., Khorram M., et al. 2023. “Encapsulation of Zataria Multiflora Essential Oil in Polyvinyl Alcohol/Chitosan/Gelatin Thermo‐Responsive Hydrogel: Synthesis, Physico‐Chemical Properties, and Biological Investigations.” International Journal of Biological Macromolecules 243: 125073. 10.1016/j.ijbiomac.2023.125073. [DOI] [PubMed] [Google Scholar]
- Trocchianesi, S. , Po A., Citarella A., et al. 2023. “Molecular Mechanisms of the Tyrosine Kinase Inhibitor Pralsetinib Activity in In‐Vitro Models of Medullary Thyroid Carcinoma: Aberrant Activation of the HH‐Gli Signaling Pathway in Acquired Resistance.” Biomedicine and Pharmacotherapy 164: 114995. 10.1016/j.biopha.2023.114995. [DOI] [PubMed] [Google Scholar]
- Vahitha, V. , Lali G., Prasad S., Karuppiah P., Karunakaran G., and AlSalhi M. S.. 2024. “Unveiling the Therapeutic Potential of Thymol From Nigella sativa L. Seed: Selective Anticancer Action Against Human Breast Cancer Cells (MCF‐7) Through Down‐Regulation of Cyclin D1 and Proliferative Cell Nuclear Antigen (PCNA) Expressions.” Molecular Biology Reports 51, no. 1: 61. 10.1007/s11033-023-09032-w. [DOI] [PubMed] [Google Scholar]
- Valverde Sancho, J. , Carreño Amate C., Caparrós Pérez M. D. M., Santana Méridas O., and Julio L. F.. 2023. “Biological Activity of Hybrid Molecules Based on Major Constituents of Cinnammomun Verum and Thymus vulgaris Essential Oils.” Life 13, no. 2: 499. 10.3390/life13020499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vasconcelos, A. P. , Xavier F. J., Castro A., et al. 2024. “Synthesis and Analysis of Carvacrol‐Derived Morita‐Baylis‐Hillman Adducts as Potential Anticancer Agents.” Journal of the Brazilian Chemical Society 35, no. 8: e20240022. [Google Scholar]
- Wachter, F. , and Pikman Y.. 2024. “Pathophysiology of Acute Myeloid Leukemia.” Acta Haematologica 147, no. 2: 229–246. [DOI] [PubMed] [Google Scholar]
- Wang, Y. , and Deng B.. 2023. “Hepatocellular Carcinoma: Molecular Mechanism, Targeted Therapy, and Biomarkers.” Cancer and Metastasis Reviews 42, no. 3: 629–652. [DOI] [PubMed] [Google Scholar]
- Xue, C. , Yao Q., Gu X., et al. 2023. “Evolving Cognition of the JAK‐STAT Signaling Pathway: Autoimmune Disorders and Cancer.” Signal Transduction and Targeted Therapy 8, no. 1: 204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yan, D. , Earp H. S., DeRyckere D., and Graham D. K.. 2021. “Targeting MERTK and AXL in EGFR Mutant Non‐Small Cell Lung Cancer.” Cancers 13, no. 22: 5639. 10.3390/cancers13225639. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yeh, J. H. , Chou C. T., Chen I. S., et al. 2017. “Effect of Thymol on Ca2+ Homeostasis and Viability in PC3 Human Prostate Cancer Cells.” Chinese Journal of Physiology 60, no. 1: 32–40. [DOI] [PubMed] [Google Scholar]
- Yildiz, S. , Turan S., Kiralan M., and Ramadan M. F.. 2021. “Antioxidant Properties of Thymol, Carvacrol, and Thymoquinone and Its Efficiencies on the Stabilization of Refined and Stripped Corn Oils.” Journal of Food Measurement and Characterization 15, no. 1: 621–632. 10.1007/s11694-020-00665-0. [DOI] [Google Scholar]
- Yin, X. , Chen H., Chen S., and Zhang S.. 2022. “Screening and Validation of a Carvacrol‐Targeting Viability‐Regulating Protein, SLC6A3, in Liver Hepatocellular Carcinoma.” Disease Markers 2022, no. 1: 3736104. 10.1155/2022/3736104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yousef, E. H. , El Gayar A. M., and Abo El‐Magd N. F.. 2024. “Carvacrol Potentiates Immunity and Sorafenib Anti‐Cancer Efficacy by Targeting HIF‐1α/STAT3/FGL1 Pathway: In Silico and In Vivo Study.” Naunyn‐Schmiedeberg's Archives of Pharmacology 398: 4335–4353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yousef, E. H. , El‐Magd N. F. A., and El Gayar A. M.. 2023. “Carvacrol Enhances Anti‐Tumor Activity and Mitigates Cardiotoxicity of Sorafenib in Thioacetamide‐Induced Hepatocellular Carcinoma Model Through Inhibiting TRPM7.” Life Sciences 324: 121735. [DOI] [PubMed] [Google Scholar]
- Yu, Y. , Liu Y., Shi R., Zhang D., Li C., and Shi J.. 2020. “New Thymol and Isothymol Derivatives From Eupatorium Fortunei and Their Cytotoxic Effects.” Bioorganic Chemistry 98: 103644. [DOI] [PubMed] [Google Scholar]
- Zare, E. , Jamali T., Ardestani S. K., and Kavoosi G.. 2021. “Synergistic Effect of Zataria Multiflora Essential Oil on Doxorubicin‐Induced Growth Inhibition of PC3 Cancer Cells and Apoptosis.” Complementary Therapies in Clinical Practice 42: 101286. [DOI] [PubMed] [Google Scholar]
- Zeng, Q. , Che Y., Zhang Y., Chen M., Guo Q., and Zhang W.. 2020. “Thymol Isolated From Thymus vulgaris L. Inhibits Colorectal Cancer Cell Growth and Metastasis by Suppressing the Wnt/β‐Catenin Pathway.” Drug Design, Development and Therapy 14: 2535–2547. 10.2147/DDDT.S254218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zengin Kurt, B. , Celebi G., Ozturk Civelek D., et al. 2023. “Tail‐Approach‐Based Design and Synthesis of Coumarin‐Monoterpenes as Carbonic Anhydrase Inhibitors and Anticancer Agents.” ACS Omega 8, no. 6: 5787–5807. 10.1021/acsomega.2c07459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang, H. , Zhao J., Chinnathambi A., Meganathan V., and Gu X.. 2023. “Anti‐Cancer Potential of Selenium‐Chitosan‐Polyethylene Glycol‐Carvacrol Nanocomposites in Multiple Myeloma U266 Cells.” Journal of Biochemical and Molecular Toxicology 37, no. 10: e23424. [DOI] [PubMed] [Google Scholar]
- Zhang, S. , He L., Shang J., et al. 2022. “Carvacrol Suppresses Human Osteosarcoma Cells via the Wnt/β‐Catenin Signaling Pathway.” Anti‐Cancer Agents in Medicinal Chemistry 22, no. 9: 1714–1722. 10.2174/1871520621666210901111932. [DOI] [PubMed] [Google Scholar]
- Zhang, T. , Ma C., Zhang Z., Zhang H., and Hu H.. 2021. “NF‐κB Signaling in Inflammation and Cancer.” MedComm 2, no. 4: 618–653. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
The data that support the findings of this study are available on request from the corresponding author.
