Next Article in Journal
Pharmacological Mechanism and Drug Research Prospects of Ginsenoside Rb1 as an Antidepressant
Previous Article in Journal
Naringin Suppresses CoCl2-Induced Ferroptosis in ARPE-19 Cells
Previous Article in Special Issue
In Vitro Protective Effects of a Standardized Extract of Opuntia ficus-indica (L.) Mill. Cladodes and Olea europaea L. Leaves Against Indomethacin-Induced Intestinal Epithelial Cell Injury
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Comprehensive Review of the Antitumor Activity of Olive Compounds: The Case of Olive Oil, Pomace, and Leaf Extracts, Phenolic Alcohols, Secoiridoids, and Triterpenes

by
Diana Melo Ferreira
,
Maria Beatriz P. P. Oliveira
and
Rita Carneiro Alves
*
REQUIMTE/LAQV (Rede de Química e Tecnologia/Laboratório Associado para a Química Verde), Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Street of Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
*
Author to whom correspondence should be addressed.
Antioxidants 2025, 14(2), 237; https://doi.org/10.3390/antiox14020237
Submission received: 23 December 2024 / Revised: 24 January 2025 / Accepted: 11 February 2025 / Published: 18 February 2025

Abstract

:
Olive oil is widely recognized for its cancer-prevention properties, and its by-products, such as pomace and leaves, offer an opportunity for compound extraction. This study comprehensively reviews the antitumor activities of olive extracts and compounds in both in vitro and in vivo studies. Key compounds, including hydroxytyrosol (HT), oleuropein (OL), oleocanthal (OC), and maslinic acid (MA), demonstrated significant antiproliferative, apoptotic, antimigratory, and anti-invasive effects, along with selective cytotoxicity, particularly against breast and colorectal cancer. HT, OC, and MA showed anti-angiogenic effects, while HT and OC showed antimetastatic effects. Moreover, HT, OL, and OC also presented synergistic effects when combined with anticancer drugs, improving their efficacy. Additionally, HT, OL, and MA exhibited protective effects against several side effects of chemotherapy. These compounds are able to modulate important signaling pathways such as the mammalian target of rapamycin, regulate oxidative stress through reactive oxygen species production, modulate angiogenic factors, and induce autophagy. Interestingly, the synergistic effects of the compounds within olive extracts appear to be stronger than their individual action. There is a need for dose optimization, further mechanistic studies to clarify the precise mechanisms of action, and future studies using olive pomace extracts with animal models.

1. Introduction

Cancer is a group of diseases characterized by uncontrolled abnormal cell proliferation, leading to tumor formation. It is the second leading cause of death globally, responsible for 9.6 million deaths in 2018, with lung, prostate, and colorectal cancers being the most common among men and breast, colorectal, and lung cancers being the most prevalent among women. Cancer imposes a substantial physical, emotional, and financial burden, especially in low- and middle-income countries where diagnosis and treatment options are limited [1].
Discovering new selective drugs is imperative as cancer incidence continues to escalate worldwide and current chemotherapeutic drugs cause significant toxicity on healthy tissues, with severe side effects such as nephrotoxicity (e.g., acute kidney injury), hepatotoxicity (e.g., steatosis), neurotoxicity (e.g., sensory neuropathy), and cardiotoxicity (e.g., heart failure) [2].
In addition, identifying natural compounds that can serve as anticancer agents may uncover potential therapeutic targets and minimize the side effects of chemotherapy. Plant polyphenols, known for their anti-aging, anti-inflammatory, antimicrobial, and antioxidant effects, target all phases of chemoprevention: (a) primary prevention by inhibiting cancer initiation, (b) secondary prevention by inhibiting cancer promotion, and (c) therapeutic intervention by inhibiting cancer progression [3,4].
The Mediterranean diet is widely recognized for its protective role against cancer due to the polyphenols found in olives (Olea europaea L.) and extra virgin olive oil (EVOO). Building on this premise, exploring the antitumor ability of potent natural extracts obtained from olive matrices such as olive by-products could benefit cancer prevention and treatment strategies [5,6,7]. This perspective becomes even more relevant if we consider that olive oil production generates thousands of tons of by-products which pose environmental concerns. In the 2023/24 harvest season, global olive oil production was approximately 2,564,000 tons. Within the European Union, Portugal’s production rose by 28%, totalizing 160,900 tons [8].
Olive pomace (OP) is the largest example of olive by-products. The ratio of OP to olive oil typically varies between 3 and 5 tons of OP. Consequently, in 2023/24, its global production is estimated to be between 7,692,000 and 12,820,000 tons [8]. Although OP is phytotoxic due to the limited transfer of polyphenols to olive oil, these compounds, if properly extracted, can serve as active ingredients in the development of functional foods, nutraceuticals, or anticancer drug formulations [9]. Similarly, olive leaves can offer additional value due to their promising pharmacological properties [10].
Olive leaves primarily result from tree pruning and olive harvesting, constituting approximately 10% of the total weight of harvested olives [11]. Assuming an average oil yield of 20% from olives, this production would require about 12,820,000 tons of olives. Consequently, olive leaves would have a weight of approximately 1,282,000 tons worldwide [8,12], depending on olive variety, cultivation practices, and regional differences [10,11].
Therefore, combining cancer treatment with olive compounds and extracts may offer synergistic protection against cancer progression due to their bioactivities [3,4]. For example, a functional ingredient obtained from OP demonstrated antimicrobial activity against Staphylococcus aureus and Escherichia coli, with the best minimal inhibitory concentration observed with higher hydroxytyrosol (HT) contents [13]. Other studies demonstrated the neuroprotective effects of olive extracts in preventing oxidative stress (OS) by reducing Aβ and tau protein aggregation in vivo [14,15].
This work aims to comprehensively review the studies available in the current literature regarding the antitumor activity of olive pomace extracts (OPEs). Due to the limited number of articles on OPE, studies involving olive oil extracts (OOEs) and olive leaf extracts (OLEs) were also included. Studies which used individual or multiple olive compounds were also included to provide a more complete overview, as these are the most available and extensively studied in the literature.

2. Methodology

The literature search was performed using PubMed, Scopus, ScienceDirect, Web of Science, and Google Scholar electronic databases and restricted to the years 2004 to 2024 to focus on data from the last 20 years. The search included the following keywords: “olive pomace”, “olive by-product”, “olive”, “olive oil”, “olive leaf”, “olive extract”, “phenolic compound”, “phenolic”, “polyphenol”, “tumor”, “antitumoral”, “antitumor”, “antitumorigenic”, “cancer”, “anticancer”, “anticarcinogenic”, “cytotoxicity”, “cytotoxic effect”, “apoptosis”, “apoptotic effect”, “antiproliferative effect”, “cancer cell line”, and “bioactivity”. Duplicated references were always deleted. The inclusion criteria were in vitro studies, which used cell lines, and in vivo studies, which used animal models. The exclusion criteria were human clinical trials. As a result of this search and criteria, 85 relevant articles were considered for elaboration for this review.

3. Antitumor Activity of Olive Compounds

Cancer develops due to aberrations in cellular signaling pathways, involving numerous molecular and key factors that drive uncontrolled growth [16]. Most studies discussed below have focused on these pathways, aiming to target them as potential cancer therapy. In fact, olive compounds have been reported to exert several antitumor effects (Figure 1).
Inflammation (Figure 2) also contributes to tumor development as it induces angiogenesis [17]. Angiogenesis is tightly regulated by hypoxia inducible factor (HIF); vascular endothelial growth factor (VEGF); hepatocyte growth factor (HGF); matrix metalloproteinase (MMP); transforming growth factor (TGF); and tumor necrosis factor (TNF), which promote the proliferation of endothelial cells, leading to the formation of new blood vessels to provide nutrients and oxygen to cancer cells [18].
Apoptosis (Figure 3), a programmed cell death mechanism marked by structural changes and DNA fragmentation, is often evaded by cancer cells [16]. It can occur via the intrinsic pathway, which involves mitochondrial release of pro-apoptotic factors, or the extrinsic pathway, triggered by death receptors [20].
Table 1 summarizes the antitumor activity of extracts from olive matrices. Olive oil, pomace, and leaf extracts are valuable products derived from the olive tree, each offering unique bioactive properties due to their richness in phenolic compounds [9,10,13].
Hydroxytyrosol (HT) and tyrosol (TYR) are key olive phenolic alcohols (Table 2). HT consists of a benzene ring with two hydroxyl groups (positions 3 and 4) and an ethanol side chain (position 2), while TYR lacks the hydroxyl group at position 3 [42].
Another important class is olive secoiridoids (Table 3), including oleuropein (OL), oleocanthal (OC), p-HPEA-EDA (dialdehydic form of decarboxymethyl ligstroside aglycone), oleacein, OL aglycone, and peracetylated oleuropein (Ac-OL). Structurally, these compounds are characterized by a secoiridoid skeleton, which includes a cyclopentane ring fused to a tetrahydrofuran ring. These molecules often possess hydroxyl and methoxy groups, which contribute to their potent antioxidant properties [56].
Additionally, olive pentacyclic triterpenes (Table 4) such as maslinic acid (MA), erythrodiol, oleanolic acid, and uvaol are characterized by a structure of five interconnected rings which form a rigid backbone. The structure allows for various functional groups to attach, leading to a diverse array of compounds with distinct biological activities [87].
Several compounds obtained from olive extracts exhibited strong antiproliferative, apoptotic, and cytotoxic effects against breast cancer models (Table 1). In particular, OL aglycone, isolated from EVOO, demonstrated superior efficacy in reducing cell viability compared to HT, TYR, and elenolic acid. Remarkably, the first compound also enhanced trastuzumab sensitivity in resistant cells [28].
In addition, polyphenols extracted from EVOO were as effective as lapatinib in suppressing FAS expression and inducing HER2 oncogene degradation. This is significant because it disrupts the lipogenic gain as FAS overexpression is linked to HER2 in breast cancer cells [30]. Similarly, OC combined with lapatinib synergistically inhibited the growth of HER2-positive cancer cells and suppressed the activation of HER2, EGFR, and c-MET pathways [64]. Additionally, HT also showed the ability to downregulate FAS in liver cancer cells [54].
OPEs also showed strong antiproliferative activity against breast cancer cells [5,9]. For example, a dry olive mill residue water extract demonstrated stronger antiproliferative effects than an OP water extract. However, HT, the major compound in both extracts, exhibited the highest antiproliferative potential, even stronger than 5-fluorouracil, a cytostatic agent. Interestingly, combining 5-fluorouracil with dry olive mill residue extract or HT enhanced its effects, with HT + 5-fluorouracil showing the strongest inhibition, emphasizing that HT could be the major contributor of the extracts’ effects [5].
The previous results are supported by other studies using individual compounds: HT had synergistic effects with paclitaxel in breast cancer models. One important aspect of the study was the assessment of the impact on oxidative status. Interestingly, the combined treatment resulted in lower DNA damage in lymphocytes, increased plasma antioxidant capacity, and reduced protein carbonyls [45].
Additionally, HT with cetuximab enhanced apoptosis through autophagic pathways in colorectal cancer cells [50], whereas OL achieved a synergistic effect with doxorubicin, sharply reducing tumor volume in vivo by inducing apoptosis via the mitochondrial pathway in breast cancer [59]. Moreover, OC may also enhance sensitivity to tamoxifen [62], and an OL-enriched extract enhanced the cytotoxicity of dacarbazine and everolimus in melanoma cells, outperforming OL used alone [76].
Overall, all of these studies highlight the synergistic potential of combining several olive compounds with standard chemotherapy drugs.
Regarding leaf extracts, an OLE decreased the viability of several breast cancer cell lines in a dose-dependent manner [31]. Similarly, other OLEs demonstrated antiproliferative effects on cancer cell lines (MCF-7 and T-24) and on endothelial cells (BBCE). This was also verified for luteolin and its glucosides. Additionally, mixtures of these compounds, mimicking the proportions found in the extracts, demonstrated similar antiproliferative activity. This is noteworthy because it suggests that the identified compounds are likely the main contributors of the extracts’ effects [32]. Both studies identified the extracts’ compounds but failed to apply a mechanistic approach to the results and lacked the use of animal models.
Recently, specialized drug delivery systems such as nanoparticles incorporating OLE have been developed to enhance the antitumor action of olive leaf compounds [94,95].
OOEs also showed significant antiproliferative effects in melanoma and skin cancer models. OC and oleacein inhibited cell migration and induced apoptosis in A431 cells through the suppression of the B-Raf-ERK pathway, although OC was the strongest compound [34]. Fogli et al. (2016) also demonstrated OC’s apoptotic effect in melanoma cells by inhibiting ERK1/2 [77].
Oral administration of an OLE and its main compound (OL) was able to protect against UVB-induced skin damage in vivo with epidermis and extracellular matrix preservation as confirmed histologically. They also prevented increases in MMP, VEGF, COX-2, Ki-67, and CD31 [35]. Additionally, an OOE exhibited stronger antimelanoma activity compared to OC and oleacein. Remarkably, post-treatment with EVOO’s extracts enhanced the toxic effect of higher doses of dacarbazine in A375 but not in HaCaT cells, demonstrating selective cytotoxicity [36]. This study is important because it validates the hypothesis that the entire extract’s action will be stronger than the use of the individual compounds.
According to the literature in Table 2, HT evidenced complex dose-dependent effects under hypoxia in breast cancer cells [43,46]. However, precise dose optimization remains a critical consideration, as excessive concentrations upregulate VEGF expression, promoting angiogenesis [46]. In colorectal cancer models, HT showed anti-inflammatory properties by inhibiting the expression of PGE2, COX-2, and VEGF. Moreover, its impact on HIF-1α demonstrated HT’s potential to disrupt the hypoxic tumor microenvironment [48]. HT’s capacity to modulate tumor progression via interaction with HIF-1α was also reported in breast cancer cells [46]. Additionally, HT induced a cell cycle block at the G2/M phase, mediated through the inhibition of ERK1/2 phosphorylation and downregulation of cyclin D1 expression [47].
HT has shown selective cytotoxicity against prostate cancer cells compared to normal prostate cells by targeting the androgen receptor, which is essential for prostate cancer growth and progression, being a strength of this study. HT also inhibited Akt, STAT3, and NF-κB [53]. However, prostate cancer cells exhibit higher resistance to HT’s antiproliferative effects than breast and colorectal cancer cells, which has been linked to lower H2O2 accumulation. In addition, HT dose- and time-dependently reduced H2O2-induced DNA damage in peripheral blood mononuclear cells. A noteworthy aspect of this work is that, at high doses, HT showed pro-oxidant activity and increased DNA damage [52]. This reinforces the need for dose optimization.
Regarding secoiridoids (Table 3), the combination of OL and 2-methoxyestradiol was stronger than individual treatments in inhibiting cell proliferation and migration in osteosarcoma cells, using autophagy induction as a mechanism [85]. Other studies confirmed that the probable mechanistic action of HT and OL in breast cancer cells was through induction of autophagy, although these cells were more sensitive to HT than OL [96,97]. OL aglycone also induced autophagy in neuroblastoma cells through the Ca2+-CaMKKβ-AMPK-mTOR pathway, as confirmed in cell and mice models [84]. Similarly, OC decreased viability and inhibited migration of breast cancer cells by inducing Ca2+ entry through TRPC6 channels [63]. OC effectively binds and inhibits mTOR phosphorylation, showing potent antiproliferative effects against breast cancer cells, but comparatively it demonstrated poorer activity against colorectal and cervical cancer cells [69]. Another study demonstrated that OL has antiproliferative and cytostatic effects on breast cancer cells, partially mediated by inhibition of PTP1B enzymatic activity, depending on dosage, as shown by molecular docking [60]. OC was shown to bind directly to the ERα ligand-binding domain, having reduced ERα levels in BT-474 cells in silico [62].
Additionally, HT reduced the self-renewal capacity of breast cancer stem cells, inhibited the Wnt/β-catenin pathway, and suppressed EMT by modulating transcription factors like SLUG, SNAIL, and ZEB1. HT’s ability to suppress EMT underlines its potential to reduce migration, invasion, and metastatic potential, particularly in TNBC cells [44]. Similarly, OC reduced tumor growth recurrence, stabilized E-cadherin, decreased vimentin levels, and suppressed HER2 and c-MET receptor activation, potentially via EMT inhibition in mice [65]. Moreover, OC inhibited hepatocellular carcinoma growth and spread by reducing EMT markers and suppressing MMP-2, cyclin D1, and Bcl-2 genes [75].
Notably, a number of studies reported the significant in vivo antitumor efficacy of OC by preventing tumor formation and/or decreasing tumor growth in female nude mice with cancer cell xenografts. Importantly, OC did not cause systemic toxicity in the mice in none of the studies, which is an upside [61,62,63,64,65,66,67]. OC also plays a role in suppressing EMT and inhibiting the activation of important targets in breast cancer therapy such as EGFR, HER2, and c-MET [61,62,63,64,65,66]. OC suppressed HGF-induced proliferation and migration in MDA-MB-231 cells by potentially inhibiting the Brk/paxillin/Rac1 pathway and induced apoptosis via caspases activation [61]. In fact, OC’s potential as a c-MET kinase inhibitor was confirmed in breast and prostate cancer cells. OC also downregulated CD31, a cell adhesion molecule, in endothelial cells [68]. Molecular docking also confirmed the potential of TYR sinapate as a c-MET kinase phosphorylation inhibitor in MDA-MB-231 cells [98]. Additionally, HT and TYR have shown synergic effects in promoting EGFR degradation in vivo, probably through the proteasomal pathway [49].
Wei et al. (2019) demonstrated that MA exerts anticancer effects in colorectal cancer cells by inhibiting cell migration, regulating EMT markers, and activating AMPK while inhibiting the mTOR pathway. Additionally, MA reduced colon carcinogenesis in AOM/DSS mice by attenuating inflammation, tumor growth, and cytokine dysregulation. These findings underscore MA’s potential as a chemopreventive agent for colorectal cancer, primarily through modulation of the AMPK-mTOR pathway [90]. The modulation of this pathway was also verified for OL aglycone in neuroblastoma models [84] and OOEs rich in OC, ligstroside aglycone, elenolic acid, and OL aglycone in liver cancer cells [39].
Both OL and OC demonstrated antiproliferative and apoptotic activities by altering gene expression profiles associated with TNBC. However, OC had a slightly stronger effect than OL [57]. Interestingly, OL was also able to downregulate microRNA expression, exerting antimigratory, apoptotic, and cytotoxic effects in a dose-dependent manner [58].
In colorectal cancer, EVOO’s phenolics caused the upregulation of CB1 (cannabinoid receptor) by reducing DNA methylation at the CNR1 promotor, leading to decreased proliferation in Caco-2 cells. Interestingly, this was confirmed by EVOO supplementation in vivo [33].
In another study, a hydroethanolic OPE extracted using ohmic heating-assisted extraction was the most effective at reducing Caco-2 colorectal cancer cell viability. The study suggests that OPE induces cell death via the mitochondrial pathway with p53 and caspase-3 activation [4]. Another OPE exhibited different activities against colorectal cancer cells. It was cytotoxic, decreased proliferation, and increased OS in HT-29 cells, while it decreased proliferation and cell growth but upregulated VEGF in Caco-2 cells, evidencing the complex response of different cancer cell lines even within the same type of cancer [9]. Different extracts obtained from various olive matrices also showed dose-dependent cytotoxicity against HCT-116 colorectal cancer cells [7].
In leukemia models, an OPE, HT, and verbascoside provided DNA protection against H2O2-induced genotoxicity while inducing apoptosis via caspase-3 activation [38]. Similar results were observed with an OLE, which inhibited DNA damage, reduced mutation frequencies, and demonstrated dose-dependent cytotoxicity in HL60 cells. Notably, LUT had the highest apoptotic effect [37]. Both studies distinguish themselves from the others by having used the somatic mutation and recombination test with Drosophila melanogaster.
The relationship between ROS and cancer is complex. Cancer cells, being highly metabolically active, produce increased ROS levels, which can damage DNA, causing OS which promotes cancer angiogenesis, invasiveness, and metastasis. Low ROS levels regulate hypoxia adaptation and pro-inflammatory cytokine production, while high levels can induce apoptosis and autophagy [21].
In fact, HT showed apoptotic effects by activating the caspase cascade and OS-modulating activities since it increased the activity of catalase, superoxide dismutase, and glutathione peroxidase and reduced thiobarbituric acid reactive substances in colorectal cancer cells [51]. Sun et al. (2014) emphasized the role of ROS generation in HT’s cytotoxic effects. It also highlighted the modulation of the Akt/FOXO3a pathway and the differential expression of antioxidant defense enzymes: superoxide dismutase enhanced HT’s effects while catalase reduced them [22].
In another study, erythrodiol induced apoptosis through ROS production and DNA damage, while oleanolic acid and uvaol induced it by cell cycle arrest. The triterpenes, except for high-dose erythrodiol, protected cells from H2O2-induced oxidative and DNA damage. The findings highlighted dose-dependent behaviors and emphasized the need for further research on mechanisms and dose optimization [26]. Moreover, OL increased ROS levels in breast cancer cells, showing apoptotic activity [23].
Increased ROS production, higher malondialdehyde and protein carbonyls levels, and decreased glutathione and catalase levels were also observed in colorectal and prostate cancer cells treated with an OLE, composed mostly of chlorogenic acid, showing antimigratory and apoptotic effects. However, all effects were less noticeable in the PC3 cells [27]. Ferreira et al. (2024) also found an increase in malondialdehyde levels in HT-29 colorectal cancer cells treated with an OPE, showing effects on lipid peroxidation [9]. However, all of these studies lack the use of animal models to validate the in vitro results.
In studies with olive secoiridoids, OL alleviated clinical symptoms and reduced colon inflammation, dysplastic lesions, and tumor formation in a mouse model of AOM/DSS-induced colorectal cancer [71]. Similarly, OL-enriched diets prevented preneoplastic lesions across all colon segments, reduced tumor development, and minimized AOM-induced DNA damage in leukocytes [72]. Both studies complement each other effectively, highlighting the protective effects of OL against colorectal cancer. Interestingly, OL also showed a protective effect in mice against cisplatin-induced kidney injury by suppressing OS, inflammation, and apoptosis [86]. In addition, p-HPEA-EDA inhibited tumorigenicity in a chicken embryo chorioallantoic membrane assay, possibly through AMPK activation [73].
Concerning triterpenes, MA did not affect breast cancer and lymphoma cells [26] but interfered with DNA integrity and triggered the apoptotic cascade in colorectal cancer cells [20]. Similarly, erythrodiol showed antiproliferative and apoptotic activities depending on dosage [91]. In addition, MA treatment altered β-actin, moesin and villin 1 expression, indicating cytoskeleton reorganization, which may contribute to its effects on cell cycle arrest, motility, and metastatic potential [88]. MA also inhibited intestinal polyp formation in mice. MA downregulated key components of the Wnt/β-catenin pathway [89]. OL also blocked the Wnt/β-catenin pathway in mice with colorectal cancer [71].
Additionally, MA suppressed the p38 MAPK pathway, causing apoptosis in bladder cancer cells, as evidenced by chromatin condensation and nuclear fragmentation [92]. MA decreased capillary tube formation and VEGF in HUVECs in renal cell carcinoma; this anti-angiogenic effect is significant because this is a highly vascularized tumor [93].
Since most studies focused on apoptotic routes, there is a lack of studies focusing on the anti-invasive, anti-angiogenic, and anti-inflammatory activities of MA and other triterpenes.
HT and OL inhibited cell proliferation and induced apoptosis, likely through the modulation of HIF-1α and p53 expression in colorectal cancer cells. Additionally, HT increased peroxisome proliferator-activated receptor gamma (PPARγ) expression, which may contribute to its tumor-reducing effects [99].
Hazas et al. (2017) investigated the effects of HT and its colonic metabolites, having found that HT and catechol were the only compounds which were extensively metabolized by colorectal cancer cells [100]. In another work, the dose-dependent antiproliferative effects of HT and its lipophilic fractions on a colorectal cancer cell line were dependent on the binding of these compounds to ER-β. Interestingly, HT oleate exhibited a greater antiproliferative effect in comparison to the others due to the higher lipophilicity, enabling easier passage through the cell membrane [101].
In liver cancer, HT inhibited critical oncogenic pathways, including Akt and NF-κB [54]. Zhao et al. (2014) further validated HT’s in vivo efficacy, showing reduced angiogenesis, increased apoptosis, and decreased tumor volume in orthotopic hepatocellular carcinoma models by inhibiting Akt and NF-κB [55]. MMPs are critical in liver cancer angiogenesis, playing roles in extracellular matrix remodeling and affecting tumor progression [102]. OL combined with cisplatin enhanced the inhibitory effects on MMP-7 in HepG2 cells, reducing invasion and metastasis, while increasing caspase-3 expression [74].
Regarding hepatocellular and colorectal cancer, OL inhibited cell growth and induced apoptosis through PI3K/Akt pathway suppression and ROS generation, marked by caspase cleavage, PARP fragmentation, DNA damage, and mitochondrial membrane depolarization [24]. Similar results were obtained in another study in which OC outperformed ibuprofen in inhibiting cell proliferation [25].
In lung cancer, OL induced cell cycle arrest in G2/M phases and apoptosis via the mitochondrial pathway (cytochrome c release) and activated the p38 MAPK pathway in H1299 cells [79]; also, OC inhibited c-MET activation and COX-2 activity, suppressed lung cancer progression, and prevented metastasis in vivo, demonstrating stronger effects in the presence of HGF [80]. Gu et al. (2017) also showed inhibitory effects on lung metastasis in a mice model treated with OC, having decreased Mcl-1, MMP, VEGF and Ki-67 [78].
Other olive secoiridoids also presented interesting bioactivities against neuroblastoma cells: OL induced apoptosis and cell cycle arrest and inhibited migration of SH-SY5Y cells [81]. Oleacein also reduced proliferation through cell cycle arrest, induced apoptosis via Bax and p53 upregulation, Bcl-2 downregulation, and STAT3 inhibition, and impaired cell adhesion and migration in SH-SY5Y cells [83]. OC also exhibited apoptotic and cytotoxic effects, selectively targeting cancer cells by increasing OS markers and inducing apoptosis more effectively in NB2a than BMDN cells [82].
In thyroid cancer cells, OL and Ac-OL inhibited the phosphorylation of ERK and Akt proteins, showing potential as therapeutic agents for thyroid cancers with genetic alterations in the MAPK and Akt pathways. Ac-OL showed higher efficacy than OL, likely due to increased cellular permeability [19].
OPEs inhibited proliferation across various cancer types, including pancreatic, ovarian, lung, prostate, neuroblastoma, and glioblastoma. Interestingly, at lower doses, OPEs improved normal cell viability, suggesting that they could act as protective agents for pancreatic cells [40]. Similarly, OL showed a protective effect towards normal pancreatic cells while promoting caspase-dependent apoptosis in cancer cells (MIA PaCa-2) [103].
Only one study tested olive oil’s phenolics in duos, having found that oleomissional, although less effective on its own, showed strong synergy with OL aglycone, significantly reducing cell numbers across various cancer cell lines. Oleacein, when combined with OL aglycone or ligstroside aglycone, also demonstrated significant synergistic effects in stomach, pancreatic, and lung cancer cell lines. Additionally, breast cancer and melanoma cells showed high responsiveness to these combinations, with substantial reductions in viability. The study also found that olive oils containing higher quantities of oleocanthal exhibited stronger cytotoxic effects compared to those without [41].
This is in accordance with Goren et al. (2019), who showed that EVOOs with higher OC content were more toxic to cancer cells, whereas EVOOs with low or no OC had little effect on cell viability [70]. Additionally, OC treatment resulted in a rapid loss of breast and prostate cancer cell viability while having minimal effect on normal cells. OC induced necrotic cell death in cancer cells, mainly through lysosomal membrane permeabilization, leading to the release of cathepsins into the cytosol, causing rapid cell death [70].
As shown in Table 5, oleic acid and TYR have the potential to inhibit inflammatory responses and angiogenic activities in glioblastoma cells by inhibiting COX-2, ERK and JNK and decreasing PGE2. Both compounds were also able to inhibit endothelial cells’ migration [104]. Indeed, olive matrices are a great source of oleic acid and dietary consumption of this fatty acid has been linked with a reduced incidence of pancreatic cancer due to a reduction in hyperinsulinaemia and consequent DNA mutations which can induce tumor growth [105].

4. Conclusions

To sum up, the olive compounds that appear to be the most effective against cancer are those extensively studied in both in vitro and in vivo studies, namely HT, OL, OC, and MA.
In terms of phenolic alcohols, HT presented antiproliferative, apoptotic, anti-angiogenic, antimigratory, anti-invasive, and antimetastatic effects, especially on breast and colorectal cancer, by modulating key pathways and molecules involved in cancer progression, such as EMT, EGFR, PI3K/Akt/mTOR, NF-κB, STAT3, ERK1/2, HIF-1α, PGE2, COX-2, VEGF, ROS, PARP1, FAS, and PPARγ.
Regarding secoiridoids, OL exhibited antiproliferative, apoptotic, antimigratory, and anti-invasive effects, predominantly on breast and colorectal cancer, being able to modulate gene expression, COX-2, NF-κB, ROS, and PTP1B. OC demonstrated antiproliferative, apoptotic, anti-angiogenic, antimigratory, anti-invasive, and antimetastatic effects, mainly on breast cancer, by targeting ERα, HER2, EGFR, and c-MET.
Concerning triterpenes, MA showed antiproliferative, apoptotic, anti-angiogenic, and antimigratory effects, especially on colorectal cancer, mostly through the apoptotic intrinsic pathway.
All four compounds showed selective cytotoxicity against cancer cells. Additionally, HT, OL, and OC showed synergistic effects when combined with anticancer drugs, being able to improve their efficacy. Furthermore, HT, OL and MA showed protective effects against several side effects of chemotherapy.
Most of these articles emphasize the importance of compound/extract dose optimization, because very low doses are sometimes ineffective, whereas high doses can counteract the beneficial effects, for example, being pro-oxidant.
Overall, these findings underscore the potential of olive compounds as therapeutic agents in nutraceuticals, functional foods, and anticancer drug formulations. However, further research is needed to clarify the exact mechanism of action of each compound, since the articles target different signaling pathways and molecules, hindering cross-comparison between studies.
To the best of our knowledge, there are still no studies using OPEs with mice models, which seems to be an opportunity for future research.

Author Contributions

Conceptualization, D.M.F., M.B.P.P.O., and R.C.A.; methodology, D.M.F.; validation, D.M.F.; formal analysis, D.M.F.; investigation, D.M.F.; resources, M.B.P.P.O. and R.C.A.; data curation, D.M.F.; writing—original draft preparation, D.M.F.; writing—review and editing, M.B.P.P.O. and R.C.A.; visualization, D.M.F.; supervision, M.B.P.P.O. and R.C.A.; project administration, M.B.P.P.O.; funding acquisition, M.B.P.P.O. All authors have read and agreed to the published version of the manuscript.

Funding

This work received financial support from PT national funds (FCT/MCTES, Fundação para a Ciência e Tecnologia and Ministério da Ciência, Tecnologia e Ensino Superior) through the project UID/50006-Laboratório Associado para a Química Verde—Tecnologias e Processos Limpos.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within the article.

Acknowledgments

D.M.F. thanks FCT/MCTES and ESF (European Social Fund) through NORTE 2020 (Programa Operacional Região Norte) for the PhD grant (ref. 2022.13375.BD). R.C.A. (ref. CEECIND/01120/2017; DOI 10.54499/CEECIND/01120/2017/CP1427/CT0001) thanks FCT for funding through the Scientific Employment Stimulus—Individual Call.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

Ac-OL (peracetylated derivative of oleuropein), Akt (protein kinase B), AOM/DSS (azoxymethane/dextran sulfate sodium), AMPK (adenosine monophosphate-activated protein kinase), AP-1 (activating protein 1), CaMKKβ (Ca2+/calmodulin-dependent protein kinase kinase beta), c-MET (mesenchymal–epithelial transition factor), COX (cyclooxygenase), DHPP (dihydroxyphenylpropionic acid), EC50 (half maximal effective concentration), EGF (epidermal growth factor), EGFR (epidermal growth factor receptor), EMT (epithelial–mesenchymal transition), ER (estrogen receptor), ERK (extracellular signal-regulated kinase), FAS (fatty acid synthase), GLUT (glucose transporter), HCC (hepatocellular carcinoma), H2O2 (hydrogen peroxide), HGF (hepatocyte growth factor), HIF (hypoxia inducible factor), HUVECs (human umbilical vein endothelial cells), IL (interleukin), JNK (c-Jun N-terminal kinase), LUT (luteolin), MA (maslinic acid), MAPK (mitogen-activated protein kinase), MMP (matrix metalloproteinase), MMP (matrix metalloproteinase), mTOR (mammalian target of rapamycin), NF-κB (nuclear factor-kappa B), OC (oleocanthal), OL (oleuropein), OLE (olive leaf extract), OOE (olive oil extract), OP (olive pomace), OPE (olive pomace extract), OS (oxidative stress), PARP (poly (ADP-ribose) polymerase), PBMCs (peripheral blood mononuclear cells), PGE2 (prostaglandin E2), PNET (pancreatic neuroendocrine tumor), PI3K (phosphatidylinositol-3-kinase), PPAR (peroxisome proliferator-activated receptor), PP (phenylpropionic acid), RNS (reactive nitrogen species), ROS (reactive oxygen species), STAT (signal transducer and activator of transcription), TGF (transforming growth factor), TNF (tumor necrosis factor), TNBC (triple negative breast cancer), TYR (tyrosol), VEGF (vascular endothelial growth factor).

References

  1. World Health Organization. Cancer. Available online: https://www.who.int/health-topics/cancer#tab=tab_1 (accessed on 6 May 2024).
  2. van den Boogaard, W.M.C.; Komninos, D.S.J.; Vermeij, W.P. Chemotherapy Side-Effects: Not All DNA Damage Is Equal. Cancers 2022, 14, 627. [Google Scholar] [CrossRef]
  3. Torić, J.; Marković, A.K.; Brala, C.J.; Barbarić, M. Anticancer effects of olive oil polyphenols and their combinations with anticancer drugs. Acta Pharm. 2019, 69, 461–482. [Google Scholar] [CrossRef]
  4. Quero, J.; Ballesteros, L.F.; Ferreira-Santos, P.; Velderrain-Rodriguez, G.R.; Rocha, C.M.R.; Pereira, R.N.; Teixeira, J.A.; Martin-Belloso, O.; Osada, J.; Rodríguez-Yoldi, M.J. Unveiling the Antioxidant Therapeutic Functionality of Sustainable Olive Pomace Active Ingredients. Antioxidants 2022, 11, 828. [Google Scholar] [CrossRef]
  5. Ramos, P.; Santos, S.A.O.; Guerra, Â.R.; Guerreiro, O.; Felício, L.; Jerónimo, E.; Silvestre, A.J.D.; Neto, C.P.; Duarte, M. Valorization of olive mill residues: Antioxidant and breast cancer antiproliferative activities of hydroxytyrosol-rich extracts derived from olive oil by-products. Ind. Crops Prod. 2013, 46, 359–368. [Google Scholar] [CrossRef]
  6. Calabriso, N.; Massaro, M.; Scoditti, E.; D’Amore, S.; Gnoni, A.; Pellegrino, M.; Storelli, C.; De Caterina, R.; Palasciano, G.; Carluccio, M.A. Extra virgin olive oil rich in polyphenols modulates VEGF-induced angiogenic responses by preventing NADPH oxidase activity and expression. J. Nutr. Biochem. 2016, 28, 19–29. [Google Scholar] [CrossRef]
  7. Xie, P.; Cecchi, L.; Bellumori, M.; Balli, D.; Giovannelli, L.; Huang, L.; Mulinacci, N. Phenolic compounds and triterpenes in different olive tissues and olive oil by-products, and cytotoxicity on human colorectal cancer cells: The case of frantoio, moraiolo and leccino cultivars (Olea europaea L.). Foods 2021, 10, 2823. [Google Scholar] [CrossRef]
  8. International Olive Council. World Market of Olive Oil and Table Olives—Data from December 2024. Available online: https://www.internationaloliveoil.org/world-market-of-olive-oil-and-table-olives-data-from-december-2024/?utm_source=chatgpt.com (accessed on 7 January 2025).
  9. Ferreira, D.M.; Barreto-Peixoto, J.; Andrade, N.; Machado, S.; Silva, C.; Lobo, J.C.; Nunes, M.A.; Álvarez-Rivera, G.; Ibáñez, E.; Cifuentes, A.; et al. Comprehensive analysis of the phytochemical composition and antitumoral activity of an olive pomace extract obtained by mechanical pressing. Food Biosci. 2024, 61, 104759. [Google Scholar] [CrossRef]
  10. Ferreira, D.M.; de Oliveira, N.M.; Chéu, M.H.; Meireles, D.; Lopes, L.; Oliveira, M.B.; Machado, J. Updated Organic Composition and Potential Therapeutic Properties of Different Varieties of Olive Leaves from Olea europaea. Plants 2023, 12, 688. [Google Scholar] [CrossRef] [PubMed]
  11. Palmeri, R.; Siracusa, L.; Carrubba, M.; Parafati, L.; Proetto, I.; Pesce, F.; Fallico, B. Olive Leaves, a Promising Byproduct of Olive Oil Industry: Assessment of Metabolic Profiles and Antioxidant Capacity as a Function of Cultivar and Seasonal Change. Agronomy 2022, 12, 2007. [Google Scholar] [CrossRef]
  12. Natac. Revolutionising the Olive Industry: OLEAF4VALUE Unveils Market-Ready, Bio-Based Products from Olive Leaf Valorisation. Available online: https://natacgroup.com/news/revolutionising-the-olive-industry-oleaf4value-unveils-market-ready-bio-based-products-from-olive-leaf-valorisation/?utm_source=chatgpt.com (accessed on 7 January 2025).
  13. Nunes, M.A.; Palmeira, J.D.; Melo, D.; Machado, S.; Lobo, J.C.; Costa, A.S.G.; Alves, R.C.; Ferreira, H.; Oliveira, M.B.P.P. Chemical composition and antimicrobial activity of a new olive pomace functional ingredient. Pharmaceuticals 2021, 14, 913. [Google Scholar] [CrossRef]
  14. Romero-Márquez, J.M.; Navarro-Hortal, M.D.; Jiménez-Trigo, V.; Muñoz-Ollero, P.; Forbes-Hernández, T.Y.; Esteban-Muñoz, A.; Giampieri, F.; Delgado Noya, I.; Bullón, P.; Vera-Ramírez, L.; et al. An Olive-Derived Extract 20% Rich in Hydroxytyrosol Prevents β-Amyloid Aggregation and Oxidative Stress, Two Features of Alzheimer Disease, via SKN-1/NRF2 and HSP-16.2 in Caenorhabditis elegans. Antioxidants 2022, 11, 629. [Google Scholar] [CrossRef]
  15. Romero-Márquez, J.M.; Navarro-Hortal, M.D.; Jiménez-Trigo, V.; Vera-Ramírez, L.; Forbes-Hernández, T.J.; Esteban-Muñoz, A.; Giampieri, F.; Bullón, P.; Battino, M.; Sánchez-González, C.; et al. An oleuropein rich-olive (Olea europaea L.) leaf extract reduces β-amyloid and tau proteotoxicity through regulation of oxidative- and heat shock-stress responses in Caenorhabditis elegans. Food Chem. Toxicol. 2022, 162, 112914. [Google Scholar] [CrossRef]
  16. Alam, M.; Alam, S.; Shamsi, A.; Adnan, M.; Elasbali, A.M.; Al-Soud, W.A.; Alreshidi, M.; Hawsawi, Y.M.; Tippana, A.; Pasupuleti, V.R. Bax/Bcl-2 cascade is regulated by the EGFR pathway: Therapeutic targeting of non-small cell lung cancer. Front. Oncol. 2022, 12, 869672. [Google Scholar] [CrossRef]
  17. Wen, Y.; Zhu, Y.; Zhang, C.; Yang, X.; Gao, Y.; Li, M.; Yang, H.; Liu, T.; Tang, H. Chronic inflammation, cancer development and immunotherapy. Front. Pharmacol. 2022, 13, 1040163. [Google Scholar] [CrossRef]
  18. Teleanu, R.I.; Chircov, C.; Grumezescu, A.M.; Teleanu, D.M. Tumor Angiogenesis and Anti-Angiogenic Strategies for Cancer Treatment. J. Clin. Med. 2020, 9, 84. [Google Scholar] [CrossRef]
  19. Bulotta, S.; Corradino, R.; Celano, M.; Maiuolo, J.; D’Agostino, M.; Oliverio, M.; Procopio, A.; Filetti, S.; Russo, D. Antioxidant and antigrowth action of peracetylated oleuropein in thyroid cancer cells. J. Mol. Endocrinol. 2013, 51, 181–189. [Google Scholar] [CrossRef] [PubMed]
  20. Reyes-Zurita, F.J.; Pachón-Peña, G.; Lizárraga, D.; Rufino-Palomares, E.E.; Cascante, M.; Lupiáñez, J.A. The natural triterpene maslinic acid induces apoptosis in HT29 colon cancer cells by a JNK-p53-dependent mechanism. BMC Cancer 2011, 11, 154. [Google Scholar] [CrossRef]
  21. Arfin, S.; Jha, N.K.; Jha, S.K.; Kesari, K.K.; Ruokolainen, J.; Roychoudhury, S.; Rathi, B.; Kumar, D. Oxidative Stress in Cancer Cell Metabolism. Antioxidants 2021, 10, 642. [Google Scholar] [CrossRef]
  22. Sun, L.; Luo, C.; Liu, J. Hydroxytyrosol induces apoptosis in human colon cancer cells through ROS generation. Food Funct. 2014, 5, 1909–1914. [Google Scholar] [CrossRef] [PubMed]
  23. Liu, L.; Ahn, K.S.; Shanmugam, M.K.; Wang, H.; Shen, H.; Arfuso, F.; Chinnathambi, A.; Alharbi, S.A.; Chang, Y.; Sethi, G. Oleuropein induces apoptosis via abrogating NF-κB activation cascade in estrogen receptor–negative breast cancer cells. J. Cell. Biochem. 2019, 120, 4504–4513. [Google Scholar] [CrossRef]
  24. Yan, C.M.; Chai, E.Q.; Cai, H.Y.; Miao, G.Y.; Ma, W. Oleuropein induces apoptosis via activation of caspases and suppression of phosphatidylinositol 3-kinase/protein kinase B pathway in HepG2 human hepatoma cell line. Mol. Med. Rep. 2015, 11, 4617–4624. [Google Scholar] [CrossRef] [PubMed]
  25. Cusimano, A.; Balasus, D.; Azzolina, A.; Augello, G.; Emma, M.R.; Di Sano, C.; Gramignoli, R.; Strom, S.C.; McCubrey, J.A.; Montalto, G. Oleocanthal exerts antitumor effects on human liver and colon cancer cells through ROS generation. Int. J. Oncol. 2017, 51, 533–544. [Google Scholar] [CrossRef] [PubMed]
  26. Allouche, Y.; Warleta, F.; Campos, M.; Sánchez-Quesada, C.; Uceda, M.; Beltrán, G.; Gaforio, J.J. Antioxidant, Antiproliferative, and Pro-apoptotic Capacities of Pentacyclic Triterpenes Found in the Skin of Olives on MCF-7 Human Breast Cancer Cells and Their Effects on DNA Damage. J. Agric. Food Chem. 2011, 59, 121–130. [Google Scholar] [CrossRef]
  27. Albogami, S.; Hassan, A.M. Assessment of the Efficacy of Olive Leaf (Olea europaea L.) Extracts in the Treatment of Colorectal Cancer and Prostate Cancer Using In Vitro Cell Models. Molecules 2021, 26, 4069. [Google Scholar] [CrossRef]
  28. Menendez, J.A.; Vazquez-Martin, A.; Colomer, R.; Brunet, J.; Carrasco-Pancorbo, A.; Garcia-Villalba, R.; Fernandez-Gutierrez, A.; Segura-Carretero, A. Olive oil’s bitter principle reverses acquired autoresistance to trastuzumab (Herceptin™) in HER2-overexpressing breast cancer cells. BMC Cancer 2007, 7, 80. [Google Scholar] [CrossRef]
  29. Menendez, J.A.; Vazquez-Martin, A.; Garcia-Villalba, R.; Carrasco-Pancorbo, A.; Oliveras-Ferraros, C.; Fernandez-Gutierrez, A.; Segura-Carretero, A. tabAnti-HER2 (erbB-2) oncogene effects of phenolic compounds directly isolated from commercial Extra-Virgin Olive Oil (EVOO). BMC Cancer 2008, 8, 377. [Google Scholar] [CrossRef] [PubMed]
  30. Menendez, J.A.; Vazquez-Martin, A.; Oliveras-Ferraros, C.; Garcia-Villalba, R.; Carrasco-Pancorbo, A.; Fernandez-Gutierrez, A.; Segura-Carretero, A. Analyzing effects of extra-virgin olive oil polyphenols on breast cancer-associated fatty acid synthase protein expression using reverse-phase protein microarrays. Int. J. Mol. Med. 2008, 22, 433–439. [Google Scholar] [CrossRef]
  31. Fu, S.; Arráez-Roman, D.; Segura-Carretero, A.; Menéndez, J.A.; Menéndez-Gutiérrez, M.P.; Micol, V.; Fernández-Gutiérrez, A. Qualitative screening of phenolic compounds in olive leaf extracts by hyphenated liquid chromatography and preliminary evaluation of cytotoxic activity against human breast cancer cells. Anal. Bioanal. Chem. 2010, 397, 643–654. [Google Scholar] [CrossRef]
  32. Goulas, V.; Exarchou, V.; Troganis, A.N.; Psomiadou, E.; Fotsis, T.; Briasoulis, E.; Gerothanassis, I.P. Phytochemicals in olive-leaf extracts and their antiproliferative activity against cancer and endothelial cells. Mol. Nutr. Food Res. 2009, 53, 600–608. [Google Scholar] [CrossRef]
  33. Di Francesco, A.; Falconi, A.; Di Germanio, C.; Micioni Di Bonaventura, M.V.; Costa, A.; Caramuta, S.; Del Carlo, M.; Compagnone, D.; Dainese, E.; Cifani, C.; et al. Extravirgin olive oil up-regulates CB1 tumor suppressor gene in human colon cancer cells and in rat colon via epigenetic mechanisms. J. Nutr. Biochem. 2015, 26, 250–258. [Google Scholar] [CrossRef] [PubMed]
  34. Polini, B.; Digiacomo, M.; Carpi, S.; Bertini, S.; Gado, F.; Saccomanni, G.; Macchia, M.; Nieri, P.; Manera, C.; Fogli, S. Oleocanthal and oleacein contribute to the In Vitro therapeutic potential of extra virgin oil-derived extracts in non-melanoma skin cancer. Toxicol. In Vitro 2018, 52, 243–250. [Google Scholar] [CrossRef] [PubMed]
  35. Kimura, Y.; Sumiyoshi, M. Olive Leaf Extract and Its Main Component Oleuropein Prevent Chronic Ultraviolet B Radiation-Induced Skin Damage and Carcinogenesis in Hairless Mice. J. Nutr. 2009, 139, 2079–2086. [Google Scholar] [CrossRef] [PubMed]
  36. Kugić, A.; Dabelić, S.; Brala, C.J.; Dabelić, N.; Barbarić, M. Extra Virgin Olive Oil Secoiridoids Modulate the Metabolic Activity of Dacarbazine Pre-Treated and Treatment-Naive Melanoma Cells. Molecules 2022, 27, 3310. [Google Scholar] [CrossRef]
  37. Anter, J.; Fernández-Bedmar, Z.; Villatoro-Pulido, M.; Demyda-Peyras, S.; Moreno-Millán, M.; Alonso-Moraga, Á.; Muñoz-Serrano, A.; Luque de Castro, M.D. A pilot study on the DNA-protective, cytotoxic, and apoptosis-inducing properties of olive-leaf extracts. Mutat. Res. Genet. Toxicol. Environ. Mutagen. 2011, 723, 165–170. [Google Scholar] [CrossRef] [PubMed]
  38. Anter, J.; Tasset, I.; Demyda-Peyrás, S.; Ranchal, I.; Moreno-Millán, M.; Romero-Jimenez, M.; Muntané, J.; Luque de Castro, M.D.; Muñoz-Serrano, A.; Alonso-Moraga, Á. Evaluation of potential antigenotoxic, cytotoxic and proapoptotic effects of the olive oil by-product “alperujo”, hydroxytyrosol, tyrosol and verbascoside. Mutat. Res. Genet. Toxicol. Environ. Mutagen. 2014, 772, 25–33. [Google Scholar] [CrossRef]
  39. De Stefanis, D.; Scimè, S.; Accomazzo, S.; Catti, A.; Occhipinti, A.; Bertea, C.M.; Costelli, P. Anti-Proliferative Effects of an Extra-Virgin Olive Oil Extract Enriched in Ligstroside Aglycone and Oleocanthal on Human Liver Cancer Cell Lines. Cancers 2019, 11, 1640. [Google Scholar] [CrossRef] [PubMed]
  40. Goldsmith, C.; Sakoff, J.; Stathopoulos, C.; Roach, P.; Scarlett, C. Cytotoxicity of Methanol and Aqueous Olive Pomace Extracts Towards Pancreatic Cancer Cells In Vitro. J. Nat. Prod. Cancer Chemo Prev. Agents 2018, 2, 1–6. Available online: https://www.unitedscientificgroup.org/journals/ets/articles/v1n1/jnpcpt-02-chloe-desiree-goldsmith.pdf (accessed on 22 December 2024).
  41. Papakonstantinou, A.; Koumarianou, P.; Diamantakos, P.; Melliou, E.; Magiatis, P.; Boleti, H. A Systematic Ex-Vivo Study of the Anti-Proliferative/Cytotoxic Bioactivity of Major Olive Secoiridoids’ Double Combinations and of Total Olive Oil Phenolic Extracts on Multiple Cell-Culture Based Cancer Models Highlights Synergistic Effects. Nutrients 2023, 15, 2538. [Google Scholar] [CrossRef]
  42. Sakavitsi, M.E.; Breynaert, A.; Nikou, T.; Lauwers, S.; Pieters, L.; Hermans, N.; Halabalaki, M. Availability and Metabolic Fate of Olive Phenolic Alcohols Hydroxytyrosol and Tyrosol in the Human GI Tract Simulated by the In Vitro GIDM-Colon Model. Metabolites 2022, 12, 391. [Google Scholar] [CrossRef]
  43. Calahorra, J.; Martínez-Lara, E.; De Dios, C.; Siles, E. Hypoxia modulates the antioxidant effect of hydroxytyrosol in MCF-7 breast cancer cells. PLoS ONE 2018, 13, e0203892. [Google Scholar] [CrossRef]
  44. Cruz-Lozano, M.; González-González, A.; Marchal, J.A.; Muñoz-Muela, E.; Molina, M.P.; Cara, F.E.; Brown, A.M.; García-Rivas, G.; Hernández-Brenes, C.; Lorente, J.A.; et al. Hydroxytyrosol inhibits cancer stem cells and the metastatic capacity of triple-negative breast cancer cell lines by the simultaneous targeting of epithelial-to-mesenchymal transition, Wnt/β-catenin and TGFβ signaling pathways. Eur. J. Nutr. 2019, 58, 3207–3219. [Google Scholar] [CrossRef] [PubMed]
  45. El-azem, N.; Pulido-Moran, M.; Ramirez-Tortosa, C.L.; Quiles, J.L.; Cara, F.E.; Sanchez-Rovira, P.; Granados-Principal, S.; Ramirez-Tortosa, M. Modulation by hydroxytyrosol of oxidative stress and antitumor activities of paclitaxel in breast cancer. Eur. J. Nutr. 2019, 58, 1203–1211. [Google Scholar] [CrossRef]
  46. Calahorra, J.; Martínez-Lara, E.; Granadino-Roldán, J.M.; Martí, J.M.; Cañuelo, A.; Blanco, S.; Oliver, F.J.; Siles, E. Crosstalk between hydroxytyrosol, a major olive oil phenol, and HIF-1 in MCF-7 breast cancer cells. Sci. Rep. 2020, 10, 6361. [Google Scholar] [CrossRef]
  47. Corona, G.; Deiana, M.; Incani, A.; Vauzour, D.; Dessì, M.A.; Spencer, J.P. Hydroxytyrosol inhibits the proliferation of human colon adenocarcinoma cells through inhibition of ERK1/2 and cyclin D1. Mol. Nutr. Food Res. 2009, 53, 897–903. [Google Scholar] [CrossRef] [PubMed]
  48. Terzuoli, E.; Donnini, S.; Giachetti, A.; Iñiguez, M.A.; Fresno, M.; Melillo, G.; Ziche, M. Inhibition of hypoxia inducible factor-1alpha by dihydroxyphenylethanol, a product from olive oil, blocks microsomal prostaglandin-E synthase-1/vascular endothelial growth factor expression and reduces tumor angiogenesis. Clin. Cancer Res. 2010, 16, 4207–4216. [Google Scholar] [CrossRef]
  49. Terzuoli, E.; Giachetti, A.; Ziche, M.; Donnini, S. Hydroxytyrosol, a product from olive oil, reduces colon cancer growth by enhancing epidermal growth factor receptor degradation. Mol. Nutr. Food Res. 2016, 60, 519. [Google Scholar] [CrossRef]
  50. Terzuoli, E.; Nannelli, G.; Frosini, M.; Giachetti, A.; Ziche, M.; Donnini, S. Inhibition of cell cycle progression by the hydroxytyrosol-cetuximab combination yields enhanced chemotherapeutic efficacy in colon cancer cells. Oncotarget 2017, 8, 83207–83224. [Google Scholar] [CrossRef]
  51. Hormozi, M.; Salehi Marzijerani, A.; Baharvand, P. Effects of Hydroxytyrosol on Expression of Apoptotic Genes and Activity of Antioxidant Enzymes in LS180 Cells. Cancer Manag. Res. 2020, 12, 7913–7919. [Google Scholar] [CrossRef]
  52. Rosignoli, P.; Fuccelli, R.; Sepporta, M.V.; Fabiani, R. In Vitro chemopreventive activities of hydroxytyrosol: The main phenolic compound present in extra-virgin olive oil. Food Funct. 2016, 7, 301. [Google Scholar] [CrossRef] [PubMed]
  53. Zubair, H.; Bhardwaj, A.; Ahmad, A.; Srivastava, S.K.; Khan, M.A.; Patel, G.K.; Singh, S.; Singh, A.P. Hydroxytyrosol Induces Apoptosis and Cell Cycle Arrest and Suppresses Multiple Oncogenic Signaling Pathways in Prostate Cancer Cells. Nutr. Cancer 2017, 69, 932–942. [Google Scholar] [CrossRef] [PubMed]
  54. Tutino, V.; Caruso, M.G.; Messa, C.; Perri, E.; Notarnicola, M. Antiproliferative, antioxidant and anti-inflammatory effects of hydroxytyrosol on human hepatoma HepG2 and Hep3B cell lines. Anticancer Res. 2012, 32, 5371–5377. [Google Scholar]
  55. Zhao, B.; Ma, Y.; Xu, Z.; Wang, J.; Wang, F.; Wang, D.; Pan, S.; Wu, Y.; Pan, H.; Xu, D.; et al. Hydroxytyrosol, a natural molecule from olive oil, suppresses the growth of human hepatocellular carcinoma cells via inactivating AKT and nuclear factor-kappa B pathways. Cancer Lett. 2014, 347, 79–87. [Google Scholar] [CrossRef]
  56. Bongiorno, D.; Di Stefano, V.; Indelicato, S.; Avellone, G.; Ceraulo, L. Bio-phenols determination in olive oils: Recent mass spectrometry approaches. Mass Spectrom. Rev. 2023, 42, 1462–1502. [Google Scholar] [CrossRef]
  57. Karousi, P.; Kontos, C.K.; Papakotsi, P.; Kostakis, I.K.; Skaltsounis, A.-L.; Scorilas, A. Next-generation sequencing reveals altered gene expression and enriched pathways in triple-negative breast cancer cells treated with oleuropein and oleocanthal. Funct. Integr. Genom. 2023, 23, 299. [Google Scholar] [CrossRef] [PubMed]
  58. Abtin, M.; Alivand, M.R.; Khaniani, M.S.; Bastami, M.; Zaeifizadeh, M.; Derakhshan, S.M. Simultaneous downregulation of miR-21 and miR-155 through oleuropein for breast cancer prevention and therapy. J. Cell. Biochem. 2018, 119, 7151–7165. [Google Scholar] [CrossRef]
  59. Elamin, M.H.; Elmahi, A.B.; Daghestani, M.H.; Al-Olayan, E.M.; Al-Ajmi, R.A.; Alkhuriji, A.F.; Hamed, S.S.; Elkhadragy, M.F. Synergistic anti-breast-cancer effects of combined treatment with oleuropein and doxorubicin in vivo. Altern. Ther. Health Med. 2019, 25, 17–24. [Google Scholar]
  60. Przychodzen, P.; Kuban-Jankowska, A.; Wyszkowska, R.; Barone, G.; Bosco, G.L.; Celso, F.L.; Kamm, A.; Daca, A.; Kostrzewa, T.; Gorska-Ponikowska, M. PTP1B phosphatase as a novel target of oleuropein activity in MCF-7 breast cancer model. Toxicol. In Vitro 2019, 61, 104624. [Google Scholar] [CrossRef]
  61. Akl, M.R.; Ayoub, N.M.; Mohyeldin, M.M.; Busnena, B.A.; Foudah, A.I.; Liu, Y.-Y.; Sayed, K.A.E. Olive phenolics as c-Met inhibitors:(-)-Oleocanthal attenuates cell proliferation, invasiveness, and tumor growth in breast cancer models. PLoS ONE 2014, 9, e97622. [Google Scholar] [CrossRef] [PubMed]
  62. Ayoub, N.M.; Siddique, A.B.; Ebrahim, H.Y.; Mohyeldin, M.M.; El Sayed, K.A. The olive oil phenolic (-)-oleocanthal modulates estrogen receptor expression in luminal breast cancer in vitro and in vivo and synergizes with tamoxifen treatment. Eur. J. Pharmacol. 2017, 810, 100–111. [Google Scholar] [CrossRef]
  63. Diez-Bello, R.; Jardin, I.; Lopez, J.J.; El Haouari, M.; Ortega-Vidal, J.; Altarejos, J.; Salido, G.M.; Salido, S.; Rosado, J.A. (−)-Oleocanthal inhibits proliferation and migration by modulating Ca2+ entry through TRPC6 in breast cancer cells. BBA Mol. Cell Res. 2019, 1866, 474–485. [Google Scholar] [CrossRef]
  64. Siddique, A.B.; Ebrahim, H.Y.; Akl, M.R.; Ayoub, N.M.; Goda, A.A.; Mohyeldin, M.M.; Nagumalli, S.K.; Hananeh, W.M.; Liu, Y.-Y.; Meyer, S.A.; et al. (−)-Oleocanthal Combined with Lapatinib Treatment Synergized against HER-2 Positive Breast Cancer In Vitro and In Vivo. Nutrients 2019, 11, 412. [Google Scholar] [CrossRef]
  65. Siddique, A.B.; Ayoub, N.M.; Tajmim, A.; Meyer, S.A.; Hill, R.A.; El Sayed, K.A. (−)-Oleocanthal Prevents Breast Cancer Locoregional Recurrence After Primary Tumor Surgical Excision and Neoadjuvant Targeted Therapy in Orthotopic Nude Mouse Models. Cancers 2019, 11, 637. [Google Scholar] [CrossRef] [PubMed]
  66. Qusa, M.H.; Siddique, A.B.; Nazzal, S.; El Sayed, K.A. Novel olive oil phenolic (−)-oleocanthal (+)-xylitol-based solid dispersion formulations with potent oral anti-breast cancer activities. Int. J. Pharm. 2019, 569, 118596. [Google Scholar] [CrossRef]
  67. Siddique, A.B.; Ebrahim, H.; Mohyeldin, M.; Qusa, M.; Batarseh, Y.; Fayyad, A.; Tajmim, A.; Nazzal, S.; Kaddoumi, A.; El Sayed, K. Novel liquid-liquid extraction and self-emulsion methods for simplified isolation of extra-virgin olive oil phenolics with emphasis on (-)-oleocanthal and its oral anti-breast cancer activity. PLoS ONE 2019, 14, e0214798. [Google Scholar] [CrossRef]
  68. Elnagar, A.; Sylvester, P.; El Sayed, K. Oleocanthal as a c-Met inhibitor for the control of metastatic breast and prostate cancer. Planta Medica 2011, 77, 1013–1019. [Google Scholar] [CrossRef]
  69. Khanfar, M.A.; Bardaweel, S.K.; Akl, M.R.; El Sayed, K.A. Olive oil-derived oleocanthal as potent inhibitor of mammalian target of rapamycin: Biological evaluation and molecular modeling studies. Phytother. Res. 2015, 29, 1776–1782. [Google Scholar] [CrossRef]
  70. Goren, L.; Zhang, G.; Kaushik, S.; Breslin, P.A.; Du, Y.-C.N.; Foster, D.A. (-)-Oleocanthal and (-)-oleocanthal-rich olive oils induce lysosomal membrane permeabilization in cancer cells. PLoS ONE 2019, 14, e0216024. [Google Scholar] [CrossRef]
  71. Giner, E.; Recio, M.C.; Ríos, J.L.; Cerdá-Nicolás, J.M.; Giner, R.M. Chemopreventive effect of oleuropein in colitis-associated colorectal cancer in c57bl/6 mice. Mol. Nutr. Food Res. 2016, 60, 242–255. [Google Scholar] [CrossRef]
  72. Sepporta, M.V.; Fuccelli, R.; Rosignoli, P.; Ricci, G.; Servili, M.; Fabiani, R. Oleuropein Prevents Azoxymethane-Induced Colon Crypt Dysplasia and Leukocytes DNA Damage in A/J Mice. J. Med. Food 2016, 19, 983–989. [Google Scholar] [CrossRef]
  73. Khanal, P.; Oh, W.-K.; Yun, H.J.; Namgoong, G.M.; Ahn, S.-G.; Kwon, S.-M.; Choi, H.-K.; Choi, H.S. p-HPEA-EDA, a phenolic compound of virgin olive oil, activates AMP-activated protein kinase to inhibit carcinogenesis. Carcinogenesis 2011, 32, 545–553. [Google Scholar] [CrossRef] [PubMed]
  74. Sherif, I.O.; Al-Gayyar, M.M.H. Oleuropein potentiates anti-tumor activity of cisplatin against HepG2 through affecting proNGF/NGF balance. Life Sci. 2018, 198, 87–93. [Google Scholar] [CrossRef]
  75. Pei, T.; Meng, Q.; Han, J.; Sun, H.; Li, L.; Song, R.; Sun, B.; Pan, S.; Liang, D.; Liu, L. (−)-Oleocanthal inhibits growth and metastasis by blocking activation of STAT3 in human hepatocellular carcinoma. Oncotarget 2016, 7, 43475. [Google Scholar] [CrossRef]
  76. Ruzzolini, J.; Peppicelli, S.; Andreucci, E.; Bianchini, F.; Scardigli, A.; Romani, A.; La Marca, G.; Nediani, C.; Calorini, L. Oleuropein, the Main Polyphenol of Olea europaea Leaf Extract, Has an Anti-Cancer Effect on Human BRAF Melanoma Cells and Potentiates the Cytotoxicity of Current Chemotherapies. Nutrients 2018, 10, 1950. [Google Scholar] [CrossRef] [PubMed]
  77. Fogli, S.; Arena, C.; Carpi, S.; Polini, B.; Bertini, S.; Digiacomo, M.; Gado, F.; Saba, A.; Saccomanni, G.; Breschi, M.C. Cytotoxic activity of oleocanthal isolated from virgin olive oil on human melanoma cells. Nutr. Cancer 2016, 68, 873–877. [Google Scholar] [CrossRef] [PubMed]
  78. Gu, Y.; Wang, J.; Peng, L. (-)-Oleocanthal exerts anti-melanoma activities and inhibits STAT3 signaling pathway. Oncol. Rep. 2017, 37, 483–491. [Google Scholar] [CrossRef]
  79. Wang, W.; Wu, J.; Zhang, Q.; Li, X.; Zhu, X.; Wang, Q.; Cao, S.; Du, L. Mitochondria-mediated apoptosis was induced by oleuropein in H1299 cells involving activation of p38 MAP kinase. J. Cell. Biochem. 2019, 120, 5480–5494. [Google Scholar] [CrossRef]
  80. Siddique, A.B.; Kilgore, P.C.S.R.; Tajmim, A.; Singh, S.S.; Meyer, S.A.; Jois, S.D.; Cvek, U.; Trutschl, M.; Sayed, K.A.E. (−)-Oleocanthal as a Dual c-MET-COX2 Inhibitor for the Control of Lung Cancer. Nutrients 2020, 12, 1749. [Google Scholar] [CrossRef]
  81. Seçme, M.; Eroğlu, C.; Dodurga, Y.; Bağcı, G. Investigation of anticancer mechanism of oleuropein via cell cycle and apoptotic pathways in SH-SY5Y neuroblastoma cells. Gene 2016, 585, 93–99. [Google Scholar] [CrossRef]
  82. Ünsal, Ü.Ü.; Mete, M.; Aydemir, I.; Duransoy, Y.K.; Umur, A.Ş.; Tuglu, M.I. Inhibiting effect of oleocanthal on neuroblastoma cancer cell proliferation in culture. Biotech. Histochem. 2020, 95, 233–241. [Google Scholar] [CrossRef] [PubMed]
  83. Cirmi, S.; Celano, M.; Lombardo, G.E.; Maggisano, V.; Procopio, A.; Russo, D.; Navarra, M. Oleacein inhibits STAT3, activates the apoptotic machinery, and exerts anti-metastatic effects in the SH-SY5Y human neuroblastoma cells. Food Funct. 2020, 11, 3271–3279. [Google Scholar] [CrossRef] [PubMed]
  84. Rigacci, S.; Miceli, C.; Nediani, C.; Berti, A.; Cascella, R.; Pantano, D.; Nardiello, P.; Luccarini, I.; Casamenti, F.; Stefani, M. Oleuropein aglycone induces autophagy via the AMPK/mTOR signalling pathway: A mechanistic insight. Oncotarget 2015, 6, 35344. [Google Scholar] [CrossRef]
  85. Przychodzen, P.; Wyszkowska, R.; Gorzynik-Debicka, M.; Kostrzewa, T.; Kuban-Jankowska, A.; Gorska-Ponikowska, M. Anticancer potential of oleuropein, the polyphenol of olive oil, with 2-methoxyestradiol, separately or in combination, in human osteosarcoma cells. Anticancer Res. 2019, 39, 1243–1251. [Google Scholar] [CrossRef]
  86. Potočnjak, I.; Škoda, M.; Pernjak-Pugel, E.; Peršić, M.P.; Domitrović, R. Oral administration of oleuropein attenuates cisplatin-induced acute renal injury in mice through inhibition of ERK signaling. Mol. Nutr. Food Res. 2016, 60, 530–541. [Google Scholar] [CrossRef]
  87. Salvador, J.A.R.; Leal, A.S.; Alho, D.P.S.; Gonçalves, B.M.F.; Valdeira, A.S.; Mendes, V.I.S.; Jing, Y. Chapter 2—Highlights of Pentacyclic Triterpenoids in the Cancer Settings. In Studies in Natural Products Chemistry; Atta-ur, R., Ed.; Elsevier: Amsterdam, The Netherlands, 2014; Volume 41, pp. 33–73. [Google Scholar]
  88. Rufino-Palomares, E.E.; Reyes-Zurita, F.J.; García-Salguero, L.; Mokhtari, K.; Medina, P.P.; Lupiáñez, J.A.; Peragón, J. Maslinic acid, a triterpenic anti-tumoural agent, interferes with cytoskeleton protein expression in HT29 human coloncancer cells. J. Proteom. 2013, 83, 15. [Google Scholar] [CrossRef]
  89. Sánchez-Tena, S.; Reyes-Zurita, F.J.; Díaz-Moralli, S.; Vinardell, M.P.; Reed, M.; García-García, F.; Dopazo, J.; Lupiáñez, J.A.; Günther, U.; Cascante, M. Maslinic acid-enriched diet decreases intestinal tumorigenesis in ApcMin/+ mice through transcriptomic and metabolomic reprogramming. PLoS ONE 2013, 8, e59392. [Google Scholar] [CrossRef]
  90. Wei, Q.; Zhang, B.; Li, P.; Wen, X.; Yang, J. Maslinic Acid Inhibits Colon Tumorigenesis by the AMPK–mTOR Signaling Pathway. J. Agric. Food Chem. 2019, 67, 4259–4272. [Google Scholar] [CrossRef]
  91. Juan, M.E.; Wenzel, U.; Daniel, H.; Planas, J. Erythrodiol, a natural triterpenoid from olives, has antiproliferative and apoptotic activity in HT-29 human adenocarcinoma cells. Mol. Nutr. Food Res. 2008, 52, 595–599. [Google Scholar] [CrossRef] [PubMed]
  92. Zhang, S.; Ding, D.; Zhang, X.; Shan, L.; Liu, Z. Maslinic acid induced apoptosis in bladder cancer cells through activating p38 MAPK signaling pathway. Mol. Cell. Biochem. 2014, 392, 281. [Google Scholar] [CrossRef]
  93. Thakor, P.; Song, W.; Subramanian, R.B.; Thakkar, V.R.; Vesey, D.A.; Gobe, G.C. Maslinic acid inhibits proliferation of renal cell carcinoma cell lines and suppresses angiogenesis of endothelial cells. J. Kidney Cancer VHL 2017, 4, 16. [Google Scholar] [CrossRef] [PubMed]
  94. Alowaiesh, B.F.; Alhaithloul, H.A.S.; Saad, A.M.; Hassanin, A.A. Green Biogenic of Silver Nanoparticles Using Polyphenolic Extract of Olive Leaf Wastes with Focus on Their Anticancer and Antimicrobial Activities. Plants 2023, 12, 1410. [Google Scholar] [CrossRef] [PubMed]
  95. Feng, Y.; Yu, H. Olive leaf extract green-formulated manganese nanoparticles: Characterization and antioxidant, cytotoxicity and anti-breast carcinoma properties. Inorg. Chem. Commun. 2024, 163, 112376. [Google Scholar] [CrossRef]
  96. Lu, H.-Y.; Zhu, J.-S.; Zhang, Z.; Shen, W.-J.; Jiang, S.; Long, Y.-F.; Wu, B.; Ding, T.; Huan, F.; Wang, S.-L. Hydroxytyrosol and Oleuropein Inhibit Migration and Invasion of MDA-MB-231 Triple-Negative Breast Cancer Cell via Induction of Autophagy. Anti Cancer Agents Med. Chem. 2019, 19, 1983–1990. [Google Scholar] [CrossRef]
  97. Lu, H.-Y.; Zhu, J.-S.; Xie, J.; Zhang, Z.; Zhu, J.; Jiang, S.; Shen, W.-J.; Wu, B.; Ding, T.; Wang, S.-L. Hydroxytyrosol and Oleuropein Inhibit Migration and Invasion via Induction of Autophagy in ER-Positive Breast Cancer Cell Lines (MCF7 and T47D). Nutr. Cancer 2021, 73, 350–360. [Google Scholar] [CrossRef]
  98. Busnena, B.A.; Foudah, A.I.; Melancon, T.; El Sayed, K.A. Olive secoiridoids and semisynthetic bioisostere analogues for the control of metastatic breast cancer. Bioorg. Med. Chem. 2013, 21, 2117–2127. [Google Scholar] [CrossRef]
  99. Cárdeno, A.; Sánchez-Hidalgo, M.; Rosillo, M.A.; Lastra, C.A.d.l. Oleuropein, a Secoiridoid Derived from Olive Tree, Inhibits the Proliferation of Human Colorectal Cancer Cell Through Downregulation of HIF-1α. Nutr. Cancer 2013, 65, 147–156. [Google Scholar] [CrossRef]
  100. Hazas, M.-C.L.d.l.; Piñol, C.; Macià, A.; Motilva, M.-J. Hydroxytyrosol and the Colonic Metabolites Derived from Virgin Olive Oil Intake Induce Cell Cycle Arrest and Apoptosis in Colon Cancer Cells. J. Agric. Food Chem. 2017, 65, 6467–6476. [Google Scholar] [CrossRef] [PubMed]
  101. Bernini, R.; Carastro, I.; Palmini, G.; Tanini, A.; Zonefrati, R.; Pinelli, P.; Brandi, M.L.; Romani, A. Lipophilization of Hydroxytyrosol-Enriched Fractions from Olea europaea L. Byproducts and Evaluation of the In Vitro Effects on a Model of Colorectal Cancer Cells. J. Agric. Food Chem. 2017, 65, 6506–6512. [Google Scholar] [CrossRef]
  102. Quintero-Fabián, S.; Arreola, R.; Becerril-Villanueva, E.; Torres-Romero, J.C.; Arana-Argáez, V.; Lara-Riegos, J.; Ramírez-Camacho, M.A.; Alvarez-Sánchez, M.E. Role of matrix metalloproteinases in angiogenesis and cancer. Front. Oncol. 2019, 9, 1370. [Google Scholar] [CrossRef]
  103. Goldsmith, C.D.; Bond, D.R.; Jankowski, H.; Weidenhofer, J.; Stathopoulos, C.E.; Roach, P.D.; Scarlett, C.J. The Olive Biophenols Oleuropein and Hydroxytyrosol Selectively Reduce Proliferation, Influence the Cell Cycle, and Induce Apoptosis in Pancreatic Cancer Cells. Int. J. Mol. Sci. 2018, 19, 1937. [Google Scholar] [CrossRef] [PubMed]
  104. Lamy, S.; Ben Saad, A.; Zgheib, A.; Annabi, B. Olive oil compounds inhibit the paracrine regulation of TNF-α-induced endothelial cell migration through reduced glioblastoma cell cyclooxygenase-2 expression. J. Nutr. Biochem. 2016, 27, 136–145. [Google Scholar] [CrossRef] [PubMed]
  105. Banim, P.J.R.; Luben, R.; Khaw, K.-T.; Hart, A.R. Dietary oleic acid is inversely associated with pancreatic cancer—Data from food diaries in a cohort study. Pancreatology 2018, 18, 655–660. [Google Scholar] [CrossRef]
Figure 1. Cancer signaling pathways, adapted from Alam et al. (2022). Epidermal growth factor (EGF) and transforming growth factor (TGF) bind to EGF receptor (EGFR) in the cell membrane, triggering downstream cascades, leading to the activation of phosphatidylinositol-3-kinase (PI3K) and protein kinase B (Akt). The mammalian target of the rapamycin (mTOR) pathway regulates cell growth. Nuclear factor kappa-B (NF-κB) regulates inflammation, apoptosis, and cell cycle-related genes. Signal transducer and activator of transcription 3 (STAT3) drives the transcription of genes involved in cell survival, proliferation, and angiogenesis. EGFR also activates the RAS/RAF/MEK/ERK cascade, which leads to the activation of extracellular signal-regulated kinase 1/2 (ERK1/2) and the activating protein 1 (AP-1) [16]. Hydroxytyrosol (HT), oleuropein (OL), oleocanthal (OC), maslinic acid (MA), peracetylated oleuropein (Ac-OL).
Figure 1. Cancer signaling pathways, adapted from Alam et al. (2022). Epidermal growth factor (EGF) and transforming growth factor (TGF) bind to EGF receptor (EGFR) in the cell membrane, triggering downstream cascades, leading to the activation of phosphatidylinositol-3-kinase (PI3K) and protein kinase B (Akt). The mammalian target of the rapamycin (mTOR) pathway regulates cell growth. Nuclear factor kappa-B (NF-κB) regulates inflammation, apoptosis, and cell cycle-related genes. Signal transducer and activator of transcription 3 (STAT3) drives the transcription of genes involved in cell survival, proliferation, and angiogenesis. EGFR also activates the RAS/RAF/MEK/ERK cascade, which leads to the activation of extracellular signal-regulated kinase 1/2 (ERK1/2) and the activating protein 1 (AP-1) [16]. Hydroxytyrosol (HT), oleuropein (OL), oleocanthal (OC), maslinic acid (MA), peracetylated oleuropein (Ac-OL).
Antioxidants 14 00237 g001
Figure 2. The role of inflammation in cancer, adapted from Wen et al. (2022). Inflammation triggers the release of inflammatory factors such as tumor necrosis factor (TNFα), transforming growth factor (TGFβ), and interleukins (ILs). Reactive oxygen species (ROS) and reactive nitrogen species (RNS), produced by macrophages, fibroblasts, and T lymphocytes, activate AID (activation-induced cytidine deaminase) and mutate P53 and MYC genes. Inflammation suppresses mismatch repair (MMR), leading to the accumulation of genetic alterations. Inflammation promotes the proliferation and survival of tumor progenitor cells by inducing the expression of growth factors and cytokines (NF-κB, STAT3, AP-1). RAS, MYC, and VHL genes can also promote tumor progression through the recruitment of pro-inflammatory cells to secrete inflammatory mediators, sustaining the inflammatory microenvironment [17]. A decrease in ROS production to prevent oxidative stress was observed in the studies of Quero et al. (2022) [4] and Bulotta et al. (2013) [19].
Figure 2. The role of inflammation in cancer, adapted from Wen et al. (2022). Inflammation triggers the release of inflammatory factors such as tumor necrosis factor (TNFα), transforming growth factor (TGFβ), and interleukins (ILs). Reactive oxygen species (ROS) and reactive nitrogen species (RNS), produced by macrophages, fibroblasts, and T lymphocytes, activate AID (activation-induced cytidine deaminase) and mutate P53 and MYC genes. Inflammation suppresses mismatch repair (MMR), leading to the accumulation of genetic alterations. Inflammation promotes the proliferation and survival of tumor progenitor cells by inducing the expression of growth factors and cytokines (NF-κB, STAT3, AP-1). RAS, MYC, and VHL genes can also promote tumor progression through the recruitment of pro-inflammatory cells to secrete inflammatory mediators, sustaining the inflammatory microenvironment [17]. A decrease in ROS production to prevent oxidative stress was observed in the studies of Quero et al. (2022) [4] and Bulotta et al. (2013) [19].
Antioxidants 14 00237 g002
Figure 3. Role of reactive oxygen species (ROS) in apoptosis, adapted from Arfin et al. (2021). Mitochondria are a primary ROS source, regulated by nitric oxide (NO), Ca2+, and tumor necrosis factor (TNFα). Low ROS levels regulate hypoxia adaptation and pro-inflammatory cytokine production, while high ROS levels can be toxic and damage the mitochondrial membrane, releasing cytochrome c to the cytoplasm which induces caspases cleavage, resulting in apoptosis. TNFα ligand binds to TNFR1 death receptor, triggering the activation of caspase-8 and leading to the cleavage of caspase-3 [21]. An increase in ROS production in cancer cells was observed by Sun et al. (2014) [22], Liu et al. (2019) [23], Yan et al. (2015) [24], Cusimano et al. (2017) [25], Allouche et al. (2011) [26], and Albogami et al. (2021) [27].
Figure 3. Role of reactive oxygen species (ROS) in apoptosis, adapted from Arfin et al. (2021). Mitochondria are a primary ROS source, regulated by nitric oxide (NO), Ca2+, and tumor necrosis factor (TNFα). Low ROS levels regulate hypoxia adaptation and pro-inflammatory cytokine production, while high ROS levels can be toxic and damage the mitochondrial membrane, releasing cytochrome c to the cytoplasm which induces caspases cleavage, resulting in apoptosis. TNFα ligand binds to TNFR1 death receptor, triggering the activation of caspase-8 and leading to the cleavage of caspase-3 [21]. An increase in ROS production in cancer cells was observed by Sun et al. (2014) [22], Liu et al. (2019) [23], Yan et al. (2015) [24], Cusimano et al. (2017) [25], Allouche et al. (2011) [26], and Albogami et al. (2021) [27].
Antioxidants 14 00237 g003
Table 1. Antitumor activity of olive extracts.
Table 1. Antitumor activity of olive extracts.
Comp.CancerEffectsStudyModelDoseRef.
Elenolic acid, HT, OL aglycone, TYRBreastAntiproliferative, apoptotic, OL aglycone synergy w/trastuzumabIn vitroMCF-7, SKBR3, SKBR3/Tzb1006.25–100 μM, 1–5 d[28]
Elenolic acid, HT, OL derivatives, TYR, etc.Antiproliferative, apoptotic, cytotoxicMCF-7, SKBR325–100 μM, 6 h–4 d[29]
FAS inhibition50 μM, 48 h[30]
HT, HT glucoside, OL, OL derivatives, VERAntiproliferative, HT synergy w/5-fluorouracilMDA-MB-231Extracts (0.1–20 μg/mL), HT (162–2595 μM), 48 h[5]
OL, OL derivatives, oleanolic acid, etc.CytotoxicJIMT-1, MCF-7, SKBR310–1000 μg/mL, 72 h[31]
HT, HT acetate, LUT, LUT derivatives, OLBreast, bladder, endothelialAntiproliferativeBBCE, MCF-7, T-240–50 μM, 2 d[32]
EVOO’s compounds, HTColorectalAntiproliferativeIn vitro, in vivoCaco-2, NCM460; Sprague Dawley rats50 μM, 24–48 h; 250 μL/300 g of EVOO: single intake at day 10 or daily for 10 d[33]
Apigenin, caffeic acid, HT, OL, quercetin, TYR, etc.Antiproliferative, apoptotic, selective cytotoxicity, protectiveIn vitroCaco-21.5 mg/mL, 72 h[4]
Erythrodiol, HT, MA, OC, OL aglycone, oleacein, etc.CytotoxicHCT-11688.25–875.5 μg/mL, 72 h[7]
Gallic acid, catechin, caffeic acid, kaempferol, etc.Colorectal, prostateCytotoxic, antimigratory, apoptoticHT-29, PC3198.6–535.3 μg/mL, 12–72 h[27]
OOE’s compounds, HT, OC, oleacein, TYRSkinAntiproliferative, antimigratory, apoptoticA431, HaCat1–200 μM, 72–144 h[34]
OLE’s compounds, OLAnticarcinogenic, anti-inflammatoryIn vivoAlbino hairless mice10–1000 mg/kg, 2x/d, 30 wk, oral[35]
EVOO’s compounds, OC, oleaceinMelanomaAntiproliferative, selective cytotoxicityIn vitroA375, A375M, HaCaT0.025–0.4% (v/v), 24–48 h[36]
OLE’s compounds, OL, LUTLeukemiaAntigenotoxic, antimutagenic, apoptotic, cytotoxic, protectiveIn vitro, in vivoHL60; D. melanogaster3.75–160 μL/mL, 2–640 μM, 72 h[37]
OPE’s compounds, HT, TYR, VERAntigenotoxic, antimutagenic, apoptotic, antiproliferative, protective3.75–320 μL/mL, 6.25–480 μM, 72 h[38]
Elenolic acid, ligstroside aglycone, OC, OL aglyconeLiverAntiproliferative, apoptotic, cytotoxic, autophagy activationIn vitroHep3B, HepG2, Huh74.81–9.62 μg/mL, 24–72 h[39]
OPE’s compoundsBreast, colon, glioblastoma, lung, neuroblastoma, ovarian, pancreatic, prostate, skinAntiproliferative, selective cytotoxicityMIA PaCa-2, HT-29, A2780, H460, A431, Du145, BE2-C, MCF-7, U-87, SJ-G2, SMA, MCF-10A, HPDE0–200 μg/mL, 72 h[40]
OC, oleacein, ligstroside aglycone, OL aglycone, oleomissionalBreast, cervix, colon, gastric, liver, lung, melanoma, pancreasAntiproliferative, selective cytotoxicityMDA-MB-231, SK-BR-3, MCF-7, SK-MEL-28, A2058, HT-29, AGS, HepG2, PANC-1, H1299, Hela, HaCaT, MCF-10AHalf of the EC50, 72 h[41]
Comselogoside, elenolic acid, HT, LUT, MA, VER, etc.Breast, pancreatic, colorectalAntiproliferative, cytotoxic, anti-angiogenicMCF-7, AsPC-1, HT-29, Caco-2100 mg/mL, 24 h[9]
EC50 (half maximal effective concentration), EVOO (extra virgin olive oil), FAS (fatty acid synthase), HT (hydroxytyrosol), LUT (luteolin), MA (maslinic acid), OC (oleocanthal), OL (oleuropein), OLE (olive leaf extract), OOE (olive oil extract), OPE (olive pomace extract), TYR (tyrosol), VER (verbascoside).
Table 2. Antitumor activity of olive phenolic alcohols.
Table 2. Antitumor activity of olive phenolic alcohols.
Comp.CancerEffectsStudyModelDoseRef.
HTBreastAntiproliferative, cytotoxicIn vitroMCF-75–600 μM, 16 h[43]
Anti-invasive, antimigratory, antiproliferative, antimetastaticBT549, Hs578T, MDA-MB-231, SBE-HEK293, SUM159PT0.5–100 μM, 72 h[44]
Antiproliferative, antitumorigenic, protective, synergy w/paclitaxelIn vitro, in vivoMCF-7, MDA-MB-231; Sprague Dawley rats10–100 μM, 72 h; 0.5–2 mg/kg/d, 6 wk, oral[45]
AntitumorigenicIn vitro, in silicoMCF-75–400 μM, 16 h[46]
ColorectalAntiproliferativeIn vitroCaco-2HT (5–162.5 µM, 15 m-96 h)[47]
Anti-angiogenic, anti-inflammatory, antitumorigenicIn vitro, in vivoHT-29, WiDr; mice w/HT-29 xenograft50–100 μM, 3–48 h; 10 mg/kg/d, 14 d[48]
Apoptotic, selective cytotoxicityIn vitroCRL1807, DLD150–200 μM, 24, 48 h[22]
HT, TYRSelective cytotoxicity, antiproliferative, antitumorigenicIn vitro, in vivoCaco-2, CCD-18Co, HT-29, WiDr; mice w/HT-29 xenograft1–300 μM, 2–48 h; 10 mg/kg/d, 14 d, intraperitoneal[49]
HTAntiproliferative, selective cytotoxicity, synergy w/cetuximab, activated autophagyIn vitroCaco-2, CCD-18Co, HaCaT, HT-29, WiDr1–300 μM, 8–48 h[50]
ApoptoticLS18050–150 μM, 24 h[51]
Breast, colorectal, prostateAntiproliferative, protectiveHCT, LNCAP, MCF-7, MDA, PBMC, PC3, SW48010–100 μM, 0.5–24 h[52]
ProstateAntiproliferative, apoptotic, selective cytotoxicityC4-2, LNCaP, RWPE1, RWPE210–400 μM, 24–72 h[53]
LiverAnti-inflammatory, antiproliferativeHep3B, HepG230–200 μM, 48–72 h[54]
Anti-angiogenic, antiproliferative, apoptotic, selective cytotoxicity, antitumorigenicIn vitro, in vivoHep3B, HepG2, HL-7702, Huh-7, SK-HEP-1; HCC mice100–400 μM, 48–72 h; 10–20 mg/kg/d, 3 wk, intraperitoneal[55]
HCC (hepatocellular carcinoma), HT (hydroxytyrosol), TYR (tyrosol).
Table 3. Antitumor activity of olive secoiridoids.
Table 3. Antitumor activity of olive secoiridoids.
Comp.CancerEffectsStudyModelDoseRef.
OC, OLBreastAntiproliferative, apoptoticIn vitroMDA-MB-231, MDA-MB-468OC: 250 μM, OL: 500 μM, 12–48 h[57]
OLAntimigratory, antiproliferative, apoptotic, cytotoxicMCF-7150–2400 μg/mL, 24–72 h[58]
Antiproliferative, apoptotic, selective cytotoxicity, antitumorigenic, synergy w/doxorubicinIn vitro, in vivoMCF-10A, MDA-MB-231; mice injected w/MDA-MB-23150 mg/kg, 72 h; 50 mg/kg, 4 wk, 1X/wk, intraperitoneal[59]
Anti-invasive, antimigratory, apoptoticIn vitroMCF-7, MDA-MB-23112.5–100 μM, 24–72 h[23]
Antiproliferative, cytostaticIn vitro, in silicoMCF-70.98–250 μM, 24–48 h[60]
OCAnti-invasive, antimigratory, antiproliferative, apoptotic, selective cytotoxicityIn vitro, in vivoBT-474, MCF-7, MCF-10A, MDA-MB-231, mice w/MDA-MB-231 xenograft5–15 μM, 24–72 h; 5 mg/kg, 3X/wk, intraperitoneal[61]
Antiproliferative, antitumorigenic, selective toxicity, synergy w/tamoxifenIn vitro, in vivo, in silicoBT-474, MCF-7, T-47D, mice w/BT-474 xenograft5–60 μM, 24–48 h; 5–10 mg/kg, 3X/wk, 43 d, intraperitoneal[62]
Antimigratory, antiproliferative, selective cytotoxicityIn vitroMCF-7, MCF-10A, MDA-MB-23110–20 μM, 24–72 h[63]
Anti-invasive, antimigratory, antiproliferative, selective cytotoxicity, antitumorigenic, synergy w/lapatinibIn vitro, in vivoBT-474, MCF-12A, SKBR3, mice w/BT-474 xenograft5–80 μM, 24–48 h, 10 mg/kg, 3X/wk, intraperitoneal[64]
Antitumorigenic, selective toxicityIn vivoMice w/BT-474 or MDA-MB-231 xenograft10 mg/kg/d, 40 d, oral[65]
In vivo, in silicoMice w/MDA-MB-231 xenograft10 mg/kg/d, 15–70 d, oral[66]
Antiproliferative, selective cytotoxicity, antitumorigenicIn vitro, in vivoMDA-MB-231, MDA-MB-468, BT-474, MCF-7, MCF10A; Foxn1nu/Foxn1+ mice2.5–80 μM; 10 mg/kg, 7X/wk, 40 d, oral[67]
OCBreast, prostateAnti-angiogenic, anti-invasive, antimigratory, antiproliferativeIn vitro, in silicoMCF-7, MDA-MB-231, PC-32–100 μM, 24 h[68]
Breast, cervical, colorectalAntiproliferativeCaco-2, HeLa, MCF-7, MDA-MB-231, T47D0.1–50 μM, 24–72 h[69]
Breast, pancreas, prostateAntiproliferative, selective cytotoxicity, antitumorigenicIn vitro, in vivoBJ-hTert, MCF-7, MCF-10A, MDA-MB-231, HEK-293T, PC3, PNET mice1–100 μM, 1–24 h; 5 mg/kg/d, 5 wk, intraperitoneal[70]
OLColorectalPreventive against colitisIn vivoC57BL/6 mice50–100 mg/kg, 63 d, oral[71]
Preventive against carcinogen (azoxymethane)A/J mice w/azoxymethane-induced tumors125 mg/kg, 6 wk, 1X/wk, intraperitoneal[72]
p-HPEA-EDAApoptotic, antitumorigenic, inhibited colony formationIn vitro, in vivoHCT-116, HT-29, JB6Cl41, SKBR3; chicken embryo chorioallantoic membrane0.1–10 μg/mL, 12–48 h; 50 μg/mL, 3 d[73]
OLColorectal, liverAntiproliferative, apoptoticIn vitroHepG2, Huh7, RKO10–80 μM, 24 h[24]
OCAntiproliferative, apoptotic, selective cytotoxicityHep3B, HepG2, HT-29, Huh7, PLC/PRF/5, SW4800.78–100 μM, 24–72 h[25]
OLLiverSynergy w/cisplatin, cytotoxicHepG2100–400 μM, 24–48 h[74]
OCAnti-invasive, antimigratory, antiproliferative, apoptotic, selective cytotoxicity, antitumorigenic, antimetastaticIn vitro, in vivoHCCLM3, HepG2, Huh-7, LO2; HCC mice10–80 μM, 24–72 h; 5–10 mg/kg/d, 5 wk, intraperitoneal[75]
OLMelanomaAntiproliferative, apoptotic, cytotoxic, synergy w/dacarbazine and everolimusIn vitroA375, M21, WM266–4250–800 μM, 24–72 h[76]
OCAntiproliferative, apoptotic, selective cytotoxicityA375, 501Mel, HDFa0.01–50 μM, 72 h[77]
OCMelanoma, lungAnti-angiogenic, anti-invasive, antimigratory, antiproliferative, apoptotic, antitumorigenic, antimetastaticIn vitro, in vivoA2058, A375, HaCaT, HUVEC; mice w/A375 xenograft; lung metastasis model5–60 μM, 24–48 h; 10 mg/kg/d, 3 wk, intraperitoneal; 15 mg/kg/d, 6 wk[78]
OLLungAntiproliferative, apoptoticIn vitroH129950–200 μM, 24 h[79]
OCAntimigratory, antiproliferative, selective cytotoxicity, antitumorigenic, antimetastaticIn vitro, in vivoA549, HMVEC, NCI-H322M; Foxn1nu/Foxn1+ mice injected w/A5491–60 μM, 24–72 h; 10 mg/kg/d, 8 wk, oral[80]
OLNeuroblastomaAnti-invasive, antimigratory, antiproliferative, apoptoticIn vitroSH-SY5Y25–800 μM, 24–72 h[81]
OCApoptotic, selective cytotoxicityBMDN, NB2a0.1–1000 μM, 24 h[82]
OleaceinAntimigratory, antiproliferative, apoptotic, selective cytotoxicitySH-SY5Y, WI-381–50 μM, 24–72 h[83]
OL aglyconeAutophagy inductionIn vitro, in vivoSH-SY5Y; TgCRND8 mice50 μM, 5 m-5 h; 50 mg/kg of diet, 8 wk, oral[84]
OLOsteosarcomaAntimigratory, antiproliferative, autophagy induction, synergy w/2-methoxyestradiolIn vitro143B1–250 μM, 24–60 h[85]
OL-Protective effect against acute renal injury caused by cisplatinIn vivoBALB/cN mice20 mg/kg, oral gavage[86]
OL, Ac-OLThyroidAntiproliferative, selective cytotoxicityIn vitroBCPAP, TAD-2, TPC-110–100 μM, 24–48 h[19]
Ac-OL (peracetylated derivative of oleuropein), HCC (hepatocellular carcinoma), HUVEC (human umbilical vein endothelial cells), OC (oleocanthal), OL (oleuropein), p-HPEA-EDA (dialdehydic form of decarboxymethyl ligstroside aglycone), PNET (pancreatic neuroendocrine tumor).
Table 4. Antitumor activity of olive pentacyclic triterpenes.
Table 4. Antitumor activity of olive pentacyclic triterpenes.
Comp.CancerEffectsStudyModelDoseRef.
MAColorectalApoptotic, clastogenic, cytotoxicIn vitroHT-2961 μM, 12–72 h[20]
Antiproliferative, cytotoxic3.75–30 μM, 3–72 h[88]
Antitumorigenic, chemopreventive, protective, apoptoticIn vivoApcMin/+ mice100 mg/kg of feed, 6 wk, oral[89]
Antimigratory, antiproliferative, apoptoticIn vitro, in vivoHCT116, SW480; AOM/DSS mice; BALB/c mice w/HCT116 xenograft5–20 μM, 12–36 h; 10–30 mg/kg every 2 d, 17 d, oral gavage[90]
ErythrodiolAntiproliferative, apoptoticIn vitroHT-2910–150 μM, 24–72 h[91]
MABladderApoptotic, selective cytotoxicity, antitumorigenicIn vitro, in vivo253J, L-02, MRC-5, RT4, T24, TCCSUP, PBC-1, PBC-2; BALB/c mice xenograft model1–1000 μM, 48–96 h; 5–20 mg/kg every other day, intraperitoneal[92]
KidneyAnti-angiogenic, antiproliferative, selective cytotoxicityIn vitroACHN, Caki-1, HUVEC, PTEC, SN12K147.11–97.04 μM, 24 h[93]
Erythrodiol, MA, oleanolic acid, uvaolBreast, lymphomaAntiproliferative, apoptotic, cytotoxicMCF-7, MDA-MB-231, U93712.5–100 μM, 24–120 h[26]
AOM/DSS (azoxymethane/dextran sulfate sodium), HUVEC (human umbilical vein endothelial cells), MA (maslinic acid).
Table 5. Antitumor activity of olive compounds from different classes.
Table 5. Antitumor activity of olive compounds from different classes.
Comp.CancerEffectsStudyModelDoseRef.
OC, ligstroside aglycone, TYR derivativesBreastAnti-invasive, antimigratory, antiproliferative, selective cytotoxicityIn vitro, in silicoMDA-MB-231, MCF-10A10–100 μM, 24–48 h[98]
HT, OLAnti-invasive, antimigratory, antiproliferativeIn vitroMDA-MB-2310–150 μM, 24–48 h[96]
MCF-7, T47D[97]
ColorectalAntiproliferative, apoptoticHT-29200–800 μM, 24–72 h[99]
HT, HT butanoate, octanoate and oleateAntiproliferativeHCT8-β8n5–50 μM, 20–80 h[101]
HT, catechol, PA, PP, HPP, DHPPAntiproliferative, apoptotic, cytotoxicCaco-2, HT-29100–200 μM, 8–48 h[100]
HT, TYR, OL, oleic acidGlioblastomaAnti-angiogenic, anti-inflammatory, antimigratoryHBMEC, U-87100 μM, 24 h[104]
HT, OLPancreaticAntiproliferative, apoptotic, selective cytotoxicityASPC-1, BxPC-3, CFPAC-1, HPDE, MIA PaCa-20–300 μM, 24–48 h[103]
DHPP (dihydroxyphenylpropionic acid), HPP (hydroxyphenylpropionic acid), HT (hydroxytyrosol), OC (oleocanthal), OL (oleuropein), PA (phenylacetic acid), PP (phenylpropionic acid), TYR (tyrosol).
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Melo Ferreira, D.; Oliveira, M.B.P.P.; Alves, R.C. A Comprehensive Review of the Antitumor Activity of Olive Compounds: The Case of Olive Oil, Pomace, and Leaf Extracts, Phenolic Alcohols, Secoiridoids, and Triterpenes. Antioxidants 2025, 14, 237. https://doi.org/10.3390/antiox14020237

AMA Style

Melo Ferreira D, Oliveira MBPP, Alves RC. A Comprehensive Review of the Antitumor Activity of Olive Compounds: The Case of Olive Oil, Pomace, and Leaf Extracts, Phenolic Alcohols, Secoiridoids, and Triterpenes. Antioxidants. 2025; 14(2):237. https://doi.org/10.3390/antiox14020237

Chicago/Turabian Style

Melo Ferreira, Diana, Maria Beatriz P. P. Oliveira, and Rita Carneiro Alves. 2025. "A Comprehensive Review of the Antitumor Activity of Olive Compounds: The Case of Olive Oil, Pomace, and Leaf Extracts, Phenolic Alcohols, Secoiridoids, and Triterpenes" Antioxidants 14, no. 2: 237. https://doi.org/10.3390/antiox14020237

APA Style

Melo Ferreira, D., Oliveira, M. B. P. P., & Alves, R. C. (2025). A Comprehensive Review of the Antitumor Activity of Olive Compounds: The Case of Olive Oil, Pomace, and Leaf Extracts, Phenolic Alcohols, Secoiridoids, and Triterpenes. Antioxidants, 14(2), 237. https://doi.org/10.3390/antiox14020237

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop