Hostname: page-component-848d4c4894-xfwgj Total loading time: 0 Render date: 2024-06-25T07:51:25.613Z Has data issue: false hasContentIssue false

Potential of olive oil and its phenolic compounds as therapeutic intervention against colorectal cancer: a comprehensive review

Published online by Cambridge University Press:  02 August 2021

Arindam Sain
Affiliation:
Department of Biotechnology, Maulana Abul Kalam Azad University of Technology, West Bengal, Haringhata, West Bengal, India
Sakshi Sahu
Affiliation:
Department of Biotechnology, Maulana Abul Kalam Azad University of Technology, West Bengal, Haringhata, West Bengal, India
Debdut Naskar*
Affiliation:
Department of Biotechnology, Maulana Abul Kalam Azad University of Technology, West Bengal, Haringhata, West Bengal, India
*
*Corresponding author: Debdut Naskar, email debdut.naskar@makautwb.ac.in
Rights & Permissions [Opens in a new window]

Abstract

Colorectal cancer (CRC) is one of the major causes of death across the world and incidence rate of CRC increasing alarmingly each passing year. Diet, genomic anomalies, inflammation and deregulated signalling pathways are among the major causes of CRC. Because of numerous side effects of CRC therapies available now, researchers all over the world looking for alternative treatment/preventive strategy with lesser/no side effects. Olive oil which is part of Mediterranean diet contains numerous phenolic compounds that fight against free radicals and inflammation and also well-known for protective role against CRC. The current review focused on the recent evidences where olive oil and its phenolic compounds such as hydroxytyrosol, oleuropein and oleocanthal showed activities against CRC as well to analyse the cellular and molecular signalling mechanism through which these compounds act on. These compounds shown to combat CRC by reducing proliferation, migration, invasion and angiogenesis through regulation of numerous signalling pathways including MAPK pathway, PI3K-Akt pathway and Wnt/β-catenin pathway and at the same time, induce apoptosis in different CRC model. However, further research is an absolute necessity to establish these compounds as nutritional supplements and develop therapeutic strategy in CRC.

Type
Research Article
Copyright
© The Author(s), 2021. Published by Cambridge University Press on behalf of The Nutrition Society

Colorectal cancer (CRC) is the third leading cause of cancer-related death globally(Reference Rawla, Sunkara and Barsouk1). Approximately, 1 200 000 new cases and 609 000 deaths occur across the globe in each year. At the same time, CRC accounts for about 10 % of all cancers in men and women worldwide(Reference Sümbül and Akkız2). Alarmingly the global burden of CRC is presumed to rise by 60 % in the coming years exceeding 2·2 million new cases and 1·1 million cancer deaths by 2030(Reference Bray, Ferlay and Soerjomataram3). The situation is becoming frightening as the CRC incidence rate increased by 1·6 % in adults aged below 50 years during the period of 2000–2013. Mortality is also increased by 13 % in the same period(Reference Siegel, Miller and Fedewa4). CRC originates from the epithelial cell lining of the colon or rectum in the gastrointestinal tract under the influence of genetic and environmental factors along with other factors like diet, lifestyle, genomic mutation, inflammatory bowel disease and an imbalance in gut microbiota(Reference Wang, Cui and Pan5). However, more than 70 % of the cases are still considered sporadic with no family history or genetic predisposition(Reference Yamagishi, Kuroda and Imai6). Inflammatory bowel disorder is one of those reasons and deemed as the third-highest risk factor for CRC only after the familial adenomatous polyposis and hereditary non-polyposis CRC(Reference Kim and Chang7). The stage of diagnosis is one of the principal determinants of the outcome of any cancer, including CRC. Therefore, the search for early diagnosis is ever demanding in clinical set-up and regarded as the principal determinant of fruitful outcome post-treatment.

Currently, CRC is clinically treated by surgery and subsequent chemotherapy. Unfortunately, chemotherapeutics are always associated with unavoidable toxicity which worsens the quality of an individual’s life(Reference Nurgali, Jagoe and Abalo8). Adverse side effects arise as chemotherapeutic agents can leave their mark on the fast-dividing non-malignant cells like hair follicle cells or digestive tract cells, along with the tumour cell(Reference Redondo-Blanco, Fernández and Gutiérrez-Del-Río9). The first line of CRC chemotherapy is based on 5-fluorouracil which can cause adverse side effects like nausea, vomiting, diarrhoea, mucosal and submucosal tissue damage, inhibition of the haematopoietic function of the bone marrow, leukopenia, etc.(Reference Gillis and Eminger10). Therefore, search for an alternative treatment strategy with minimal side effect to treat or prevent CRC is always on. In this context, olive oil and its phenolic compounds find their place as one of the alternative strategies used by the different research groups as this is a part of the natural diet, various ethnic groups all the world, especially Mediterranean people follow. A voluminous literature focused on the activity of different biologically active compounds present in the diet, resisting different cancers. Accumulating evidence suggests regular intake of olive oil may protect against developing CRC. The main aims of this review are to accumulate and critically asses the chemopreventive activities of olive oil as well as some of the phytochemicals originating from it, including hydroxytyrosol, tyrosol, oleuropein, oleocanthal (OC), apigenin, luteolin, etc. and the mechanisms behind the protection.

An extensive search in the PubMed, Google Scholar and Medline databases carried out using relevant keywords, for example, ‘colorectal cancer’ or ‘colon cancer’ combined with other terms including ‘olive’, ‘olive oil’, ‘virgin olive oil’, ‘extra virgin olive oil’, ‘hydroxytyrosol’, ‘oleuropein’, ‘oleocanthal’, ‘apigenin’, ‘luteolin’ and ‘olive phenolic extract’; we filtered our search within the literature by sticking to the time frame January 2010–April 2021.

Molecular insight of colorectal cancer

Genomic instability is a major driving force behind CRC(Reference Grady and Carethers11) and major molecular events include chromosomal instability (CIN), microsatellite instability and CpG island methylation that may lead to genomic instability(Reference Mármol, Sánchez-de-Diego and Pradilla Dieste12). In ˜85 % of CRC cases, CIN prominently presents either in the form of loss of tumour suppressor genes (TSG) or activation of oncogenes(Reference Tsang, Cheng and Wong13,Reference Damilakis, Mavroudis and Sfakianaki14) . TSG like adenomatous polyposis coli (APC), TP53 and SMAD4 are either physically lost from the genome or mutated in CRC. CIN can also drive the activation of different oncogenes like KRAS (Kirsten Rat Sarcoma viral oncogene homolog), BRAF (v-raf murine sarcoma viral oncogene homolog B1) and PIK3CA (phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha) by accumulating mutations (Fig. 1)(Reference Wang, Cui and Pan5,Reference Nguyen and Duong15) . Microsatellite instability presents in 15–20 % of sporadic CRC and more than 95 % in hereditary non-polyposis colon cancer(Reference Nguyen and Duong15). Mismatch repair genes are also affected by microsatellite instability which include silencing of MutL homolog 1 in HNPPC patients, already having a higher risk of developing CRC(Reference Ponz de Leon and Percesepe16,Reference Turano, Delrio and Rega17) . Inactivation of DNA mismatch repair results in the alteration of different key regulatory genes like TSG (e.g. TGFBR2, TCF4) and apoptosis-pathway related genes (e.g. BAX, caspase 5) (Fig. 1)(Reference Turano, Delrio and Rega17,Reference Mori, Yin and Rashid18) . Another important driving factor of CRC tumourigenesis is CpG island methylator phenotype, an epigenetic alteration that causes aberrant methylation of CpG islands and present in around 20–30 % of all CRC (Fig. 1)(Reference De Palma, D’Argenio and Pol19,Reference Advani, Advani and Brown20) . It is characterised by DNA hypermethylation at promoter-associated CpG islands of TSG results in transcription inhibition of the particular TSG(Reference Curtin, Slattery and Samowitz21).

Fig. 1. Schematic representation of key molecular events that drive colorectal carcinoma. APC mutation acts as the stepping stone in the process of transforming normal colorectal epithelium to adenoma, whereas the adenoma–carcinoma sequential progression is supported by alteration in three crucial events: CIN, microsatellite instability and CpG island methylator phenotype. Once the oncogenesis initiated, further accumulation of genetic changes by mutations of regulatory genes, such as DNA repair genes drive the progression. Finally, modifications of the genes related to epithelial–mesenchymal transition, basement membrane disruption, cell motility and angiogenesis contribute to metastasis.

New factors coming to the mix include microRNA (miRNA) and long non-coding RNA (lncRNA), which are thought to play a significant role in the carcinogenesis process of CRC as the expression of both miRNA and lncRNA altered in CRC(Reference To, Tong and Wu22,Reference Yao, Wang and Chen23) . Aberrant expressions of miRNA (e.g. miR-106a, miR-143) and lncRNA (e.g. HOTAIR, MALAT1) can lead carcinogenesis by altering the expression of different key regulatory genes (e.g. RB1 (retinoblastoma), BCL2 (B-cell lymphoma 2), KRAS, etc.)(Reference Lin, Chuang and Zuo24,Reference Galamb, Barták and Kalmár25) . Recently, it has been shown that the miR-200 family including miR-141, miR-200a, miR-200b, miR-200c and miR-429 is down-regulated in CRC and linked to epithelial-to-mesenchymal transition of cancer cells(Reference Ranković, Zidar and Žlajpah26). Similarly, lncRNA such as H19 or MALAT1 can promote metastasis and invasion in CRC(Reference Bermúdez, Aguilar-Medina and Lizárraga-Verdugo27). More fascinating connections are coming through the rank as microRNA, along with lncRNA, shown to have a role in the acquisition of post-treatment drug resistance(Reference Corrà, Agnoletto and Minotti28).

Alterations of key signalling pathways: driving force behind colorectal cancer development

It is a well-known fact that impairment of cell signalling pathways help tumour cells to survive within the microenvironment(Reference Wood, Parsons and Jones29,Reference Pennel, Park and McMillan30) . Some of the key signalling pathways documented to be involved in CRC include Wnt/β-catenin pathway, epidermal growth factor receptor (EGFR)/MAPK pathway, PI3K pathway, NF-κβ pathway, TGFβ signalling pathway and JAK/STAT pathway(Reference Wang, Cui and Pan5,Reference Koveitypour, Panahi and Vakilian31) . All the more, these intracellular pathways do not work in an isolated manner within the cancer milieu rather their crosstalk with each other fuel the progression and invasiveness of CRC and responsible for increased drug resistance(Reference Koveitypour, Panahi and Vakilian31Reference Yuan, Tao and Zhang33).

Wnt/β-catenin signalling serves as the central organiser of epithelial stem cell identity and crypt maintenance(Reference Koch34) and highly interlinked with several other signalling pathways (e.g. Notch, Hedgehog, BMP). The combinatorial signalling events shape the homoeostasis of the intestinal epithelium and responsible for tissue regeneration (Fig. 2)(Reference Vanuytsel, Senger and Fasano35,Reference Kaemmerer, Jeon and Berndt36) from the stem cells reside at the lower crypt of the intestine. β-catenin-mediated canonical Wnt signalling drives proliferation at the lower crypts(Reference Komiya and Habas37) whereas the non-canonical Wnt signalling (β-catenin independent) operates predominantly in the upper crypt area, where the proliferation comes to a halt and differentiation becomes essential. β-catenin gets accumulated and stabilised as a result of the Wnt activation. Subsequently, β-catenin-dependent transcription of several target genes controls the proliferation of intestinal stem cells(Reference Martini, Krug and Siegmund38). This pathway is one of the most significant pathways as APC gene is the most often mutated in CRC and linked to both sporadic and hereditary carcinogenesis(Reference Kaemmerer, Jeon and Berndt36,Reference Armaghany, Wilson and Chu39,Reference Kirsanov, Fetisov and Lesovaya40) . Mutation at APC is one of the main factors in the development of familial adenomatous polyposis syndrome as well as found around 80 % in sporadic CRCs(Reference Cheng, Xu and Chen41). APC acts as an integral member of the β-catenin destruction complex and thereby prevents β-catenin accumulation in cytoplasm(Reference Hankey, Frankel and Groden42). So, in the absence of APC or in case of mutated APC condition, β-catenin accumulates to a higher level and translocates into the nucleus. In the nucleus, β-catenin binds to DNA and activates the transcription of different proto-oncogenes linked to CRC, like c-myc, cyclin D1 and matrix metalloproteinase-7(Reference Cheng, Xu and Chen41). Recently, Yaegar et al. (Reference Yaeger, Chatila and Lipsyc43) observed several alterations in the core Wnt regulator genes within a set of 400 genes and identified oncogenic Wnt activation in 96 % of human CRCs. Similarly, Wnt signalling in tumour microenvironment linked to tumour immunomodulation and immune suppression(Reference Goldsberry, Londoño and Randall44). So, it is quite evident that targeting Wnt/β-catenin is always a major focus of the CRC research.

Fig. 2. Schematic depiction of colonic epithelium structure and components. The colonic crypt can be subdivided into three zones depending on the presence of different types of cells: stem cell zone, transit-amplifying (TA) cell zone and the differentiated zone. Reg4+ (regenerating islet-derived family member 4) deep crypt secretory cells (DCS) reside at the bottom of the colonic crypt and provide necessary support to the Lgr5+ (leucine-rich repeat-containing G-protein coupled receptor 5) stem cells, similar to the Paneth cells present in the small intestines. Quiescent stem cells or label-retaining cells (LRC) are located at the +4 position of the stem cell zone. TA cells are rapidly dividing and eventually differentiate into functional cells. The presence of Wnt, Notch, BMP, BMP antagonists, and Hedgehog and their respective concentration gradient in different zones is indicated by upward and downward triangles.

Apart from the Wnt/β-catenin pathway, several other signalling pathways like EGFR/MAPK signalling pathway, phosphatidylinositol-3-kinase (PI3K) signalling pathway and NF-κβ pathway also contribute to the development and progression of CRC. Different key players of the EGFR/MAPK signalling pathway (e.g. KRAS, BRAF, etc.) are mutated in CRC(Reference Krasinskas45) which limits the efficacy of EGFR inhibitors like cetuximab in metastatic CRC(Reference Zhang, Roberts and Shivdasani46). On the other hand, PI3K pathway influences the initiation and progression of CRC. Mutations in PIK3CA and PIK3CB (phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit beta) gene and loss of function of TSG PTEN (phosphatase and tensin homolog) can accord the process of benign to malignant transformation(Reference Papadatos-Pastos, Rabbie and Ross47). Akt, which acts as the downstream effector of the PI3K pathway, also involved in the proliferation as well as apoptosis inhibition in CRC(Reference Koveitypour, Panahi and Vakilian31). Under the influence of Akt, further downstream effector mTOR supports angiogenesis, protein translation, growth and metabolism(Reference Koveitypour, Panahi and Vakilian31). PIK3CA mutation even confers resistance to first-line chemotherapy (FOLFOX regimen) in CRC as survival and proliferation of CRC stem cells are up-regulated by PI3K/Akt signalling(Reference Wang, Shi and Zhou48). Different types of inhibitors like pan PI3K inhibitors, Akt inhibitors and PI3K/mTOR dual inhibitors are being tested in clinics to restrain PI3K/Akt/mTOR axis. On the other hand, the NF-κβ signalling pathway serves as a major regulator of inflammation and activated NF-κβ is linked to DNA damage, carcinogenic mutations and redox imbalance. All these can lead to CIN, aneuploidy and epigenetic changes related to tumourigenesis(Reference Yang, Dang and Ji49). Along with the STAT3 signalling pathway, NF-κβ plays an integral role in the transformation of inflammation into CRC by regulating cellular signal transduction(Reference Yang, Dang and Ji49). NF-κB action also promotes the proliferation and invasion and metastasis by regulating signalling pathways including epithelial-to-mesenchymal transition(Reference Li, Lin and Chen50).

Olive oil and its phenols: is it worthy of use in colorectal cancer?

Natural products including phytochemicals are gradually coming to the mix in search for inhibitors of aberrant cellular signalling networks and dietary modification could hold the key to prevent CRC by regulating cell signalling. Owing to the drug resistance and unanticipated side effects of chemotherapy, a voluminous quantity of research focused on the activity of different biologically active compounds present in the diet as an alternative strategy in CRC. The biologically active compounds from plants are defined as phytochemicals which include polyphenols, flavonoids, phytoalexins, phenolic acids, etc.(Reference Dariya, Rajitha and Alam51). Olive oil, the principal culinary fat in the traditional Mediterranean diet, is a bountiful source of phenolic compounds(Reference Papanikolaou, Melliou and Magiatis52). A number of phytochemicals isolated from olive oil polyphenols (OOP) have been shown to exert anti-inflammatory as well as anti-cancer properties(Reference Bassani, Rossi and De Stefano53) and the health claim of hydroxytyrosol (a phenolic compound present in olive oil) already approved by the EFSA (European Food Safety Authority) in 2017 (https://www.efsa.europa.eu/en/efsajournal/pub/4728, accessed on 6th July 2021). Over the years, it has been learned from in vitro and in vivo models and that OOP may evolve as a novel therapeutic strategy to avert and treat disease with minimal side effects. Additionally, the amalgamation of chemotherapeutic drugs and phenolic compounds present in olive oil could synergistically augment positive treatment outcome in cancer by reducing the undesirable side effects of conventional anticancer drugs(Reference Terzuoli, Nannelli and Frosini54,Reference Torić, Marković and Brala55) . In the past few decades, OOP been exploited effectively as preventive and therapeutic agents in a spectrum of diseases including CVD(Reference Pelucchi, Bosetti and Negri56) obesity(Reference Soriguer, Almaraz and Ruiz-de-Adana57), diabetes mellitus(Reference Schwingshackl, Lampousi and Portillo58), Alzheimer’s disease(Reference Román, Jackson and Reis59) and different cancers like breast, liver, lungs and CRC(Reference Borzì, Biondi and Basile60). Thus, it is worthy to analyse the current developments of olive oil effects on CRC and future strategies to include olive oil components in the treatment protocol.

Protective role of different forms of olive oil against colorectal cancer

Olive oil is regarded as the plentiful source of phenolic compounds. Among all the phenolic compounds present in olive oil, tyrosol (Tyr), hydroxytyrosol (HTyr) (the concentration of total tyrosol and hydroxytyrosol is 100–400 mg/kg oil)(Reference Romero and Brenes61), oleuropein (Ole) (3·8 mg/kg oil)(Reference Medeiros de Azevedo, Ferreira Ribeiro de Oliveira and Alves Alcântara62) and its aglycone (222·62–537·83 mg/kg)(Reference Ouni, Taamalli and Gómez-Caravaca63) are well characterised and most studied (Fig. 3).

Fig. 3. Structure of major phenolic compounds present in olive oil. Major phenolic compounds present in olive oil and their structure are shown here, which include simple phenol like hydroxytyrosol, secoiridoid like oleuropein and flavone like apigenin.

Dietary habit is related to cancer and accumulating pieces of evidence hint at a link between the consumption of red meat and CRC risk(Reference Aykan64). Consumption of red meat may lead to an increased level of secondary bile salt in the gut(Reference Trefflich, Marschall and Giuseppe65), that may in turn inhibit the action of diamine oxidase, an enzyme present in a high level at ileal mucosa and colon. All these actions can lead to mucosal proliferation as well as carcinoma(Reference Imran, Nadeem and Gilani66). Stoneham et al.(Reference Stoneham, Goldacre and Seagroatt67) first demonstrated that olive oil consumption could protect against CRC development by influencing polyamine metabolism in the colon through altering secondary bile acid patterns. However, the missing link between consumptions of olive oils and effect of its constituents on normal healthy cells’ metabolism yet to be documented which requires extensive research as polyamines are vital for normal cell growth as well. Similarly, in human colon adenocarcinoma cell line (Caco-2), extra virgin olive oil (EVOO) polyphenols protect against inflammation induced by oxysterol (present in cholesterol containing food items) by reducing the NF-κB pathway(Reference Serra, Incani and Serreli68). Hence, diet containing olive oil could protect gut epithelium from potentially harmful components present in food like oxysterols and help in maintaining gut homoeostasis. Protective role of EVOO against intestinal inflammation is well documented as EVOO protects against intestinal inflammation induced by 5 % (w/v) of dextran sodium sulphate in drinking water for 10 d in mice by reducing the expression of pro-inflammatory genes (e.g. IL-1β, TGFβ, IL-6)(Reference Cariello, Contursi and Gadaleta69). Further olive oil may bring about cancer cell death by inducing apoptosis in CRC cells in vitro by virtue of its antioxidant properties(Reference Centrone, D’Agostino and Difonzo70) and interfere in colorectal carcinogenesis by reducing COX-2 (cyclooxygenase-2) and Bcl-2 level(Reference Llor, Pons and Roca71). It is also shown to interfere all the three stages of CRC development including initiation, promotion and metastasis(Reference Costea, Hudiţă and Ciolac72). Another aspect of protective role of olive oil against colon carcinogenesis is possible through improving barrier function, reducing DNA damage and decreasing invasiveness as shown in in vitro (HT-29, HT-119 and Caco-2)(Reference Gill, Boyd and McDermott73) as well as in colon carcinoma in vivo rat model. In rat model, olive oil potentially acts on arachidonic acid metabolism and PGE2 synthesis to protect against colon carcinogenesis(Reference Bartolí, Fernández-Bañares and Navarro74). In a very recent study, it has been further solicited that EVOO-rich diet is capable to prevent colorectal carcinogenesis virtue of its ability to modify gut microbiota in mice(Reference Rodríguez-García, Sánchez-Quesada and Algarra75). Involvement of olive oil containing diet on gut barrier health should be explored critically as leaky gut and altered gut microbiome are proved to be critical in colon carcinogenesis. Not only the VOO alone, but the metabolites generated from VOO by gut microbiota such as HTyr and phenylacetic and hydroxyphenylpropionic acids also help cell cycle arrest and promote apoptosis(Reference López de Las Hazas, Piñol and Macià76). Therefore, next line of research should be focused on the olive oil metabolites as well. Both virgin olive oil and OVP (virgin olive oil phenolics extract) have shown the anti-invasive properties in vitro (HT-115)(Reference Hashim, Rowland and McGlynn77) and in vivo (SCID BALB/c mice) model by reducing different integrin protein expression to control metastasis(Reference Hashim, Worthington and Allsopp78). Pampaloni et al.(Reference Pampaloni, Mavilia and Fabbri79) revealed that EVOO inhibits CRC cell growth by acting on oestrogen receptor-β. However, the precise role of the individual phenolic compounds on oestrogen receptors is yet to be discovered. Effectiveness of olive oil is shown against a common environmental toxicant, benzo[a]pyrene (B[a]P)-induced colon carcinogenesis in mouse model where it accelerates B[a]P detoxification in the liver and thereby decreases oxidative damage caused by otherwise harmful metabolites generated via B[a]P biotransformation(Reference Banks, Amoah and Niaz80). In this context, effect of olive oil on phase II metabolism of carcinogens should be studied in great detail to establish olive oil-based diet as a preventive strategy against common carcinogens. Similarly, EVOO-enriched diet could have a preventive role in ulcerative colitis-associated colon carcinogenesis(Reference Sánchez-Fidalgo, Villegas and Cárdeno81). Epigenetic modifications of key regulatory genes by changing the DNA methylation are also quite possible as evidenced in pre-clinical DMH (1, 2-dimethylhydrazine) treated colon cancer in rat model where olive oil treatment inhibited the NF-κB inflammatory pathway and restored apoptotic pathways by altering miRNA and methylation pattern(Reference Nanda, Mahmood and Bhatia82). Thus, epigenetic therapy based on olive oil components could be a reality in coming years although the effect of olive oil on methylation pattern of regulatory genes essential for normal cell function should be characterised in great detail. In another remarkable study, EVOO polyphenols alone shown to inhibit the colon cancer cell (SW480) growth, but in combination with anticancer drugs such as carboplatin, cisplatin, 5-fluorouracil and irinotecan enhance the metabolic activity and survival of cancer cell which imply cautious intake of olive oil in patients under chemotherapy(Reference Torić, Brozovic and Baus Lončar83). Therefore, to better evaluate the efficacy of olive oil in CRC, more clinical research should be designed to evaluate the role of individual components of olive oil and their metabolites. Table 1 summarises the outcomes of several studies where various form of olive oil used as intervention.

Table 1. Summary of studies involving different forms of olive oil as intervention in CRC

Olive oil polyphenols against colorectal cancer

Hydroxytyrosol and tyrosol

Hydroxytyrosol and tyrosol are two phenolic compounds which are found abundantly in olive oil and both are known for their antioxidant attributes(Reference Karković Marković, Torić and Barbarić84). Different studies have weighed on their possible effect in different cancers(Reference Toteda, Lupinacci and Vizza85,Reference Chimento, Casaburi and Rosano86) . Tyrosol can potentially curb intestinal inflammation by attenuating IL-8 secretion as shown in human colon adenocarcinoma cells, WiDr(Reference Ye, Chang and Tseng87). Anti-invasive property of tyrosol explored in HT-115 colon carcinoma cells, where tyrosol reduced invasion by ˜30–70 %(Reference Hashim, Rowland and McGlynn77). However, the effect of tyrosol as sole intervention is not studied in great detail in animal model of CRC which could be important to assess the potential of OOP as possible preventive measures. On the other hand, HTyr can induce apoptosis in human colon cancer cells (DLD1) possibly by generating reactive oxygen species and destabilising the intrinsic redox status of cancer cell through PI3K/Akt signalling pathway(Reference Sun, Luo and Liu88). It also shown to stimulate apoptotic cell death of CRC cells (HT-29) in a p53-dependent way(Reference Cárdeno, Sánchez-Hidalgo and Rosillo89). Olive oil polyphenolic extract containing both Tyr and HTyr along with Ole led to cell cycle arrest in colon adenocarcinoma cells as these phenols have a strong negative effect on CRC cell proliferation by blocking the cell cycle at the G2/M phase(Reference Corona, Deiana and Incani90). Authors further suggested that interference in the cell cycle is due to obstructive COX-2 expression through inhibition of p38 and transcription factor, CREB (cAMP response element-binding protein)(Reference Corona, Deiana and Incani90). Another study by the same group pointed out that HTyr is able to reduce the level of cyclin D1 through inhibition of extracellular signal-regulated kinase (ERK)1/2 phosphorylation and therefore CRC cell proliferation(Reference Corona, Deiana and Incani91). G1 phase blockade of human colon cancer cells (Caco-2 and HT-29) was possible with HTyr and it instigated caspase-dependent apoptosis in CRC cells(Reference López de Las Hazas, Piñol and Macià76). It seems that HTyr has both anti-proliferative and pro-apoptotic properties against CRC cells, but the effect of HTyr on cell survival pathways like autophagy should be studied at the same time to evaluate possible resistance against HTyr by the cancer cells. Another feature of HTyr protection against CRC may be through its anti-metabolic properties as it can influence the activity of a major anabolic enzyme fatty acid synthase, an important regulator of the AMPK/mTOR pathway in human colon cancer cells(Reference Notarnicola, Pisanti and Tutino92). Fatty acid synthase plays a critical role during cancer cell growth transformation, that is, from two-dimensional to three-dimensional growth(Reference Bueno, Jimenez-Renard and Samino93). In a different mechanism proposed by Di Francesco et al.(Reference Di Francesco, Falconi and Di Germanio94) HTyr alters the function of TSG CNR1 that codes for type 1 cannabinoid receptor (CB1) by reducing the level of DNA methylation at the promoter region of CNR1 gene which subsequently leads to the increased CB1 expression (up to 4-fold) in colon of Sprague–Dawley rats. HTyr also increased the CNR1 expression through reduction of the CNR1 targeting miRNA (e.g. miR23a and miR-301a). This is the initial hint of epigenetic modification of regulatory genes by HTyr. Although epigenetic modifications of other oncogenes or TSG by HTyr not documented yet, HTyr can also exert its action on cancer cells through cell surface receptors or intracellular receptors. It reduces CRC cell proliferation via intracellular oestrogen receptors as lyophilised extracts containing HTyr minimised human colon cancer cell proliferation, through oestrogen receptor-β (Reference Bernini, Carastro and Palmini95). In a recent study, it is unveiled that HTyr can hinder the activity of cell surface receptor EGFR which is strongly associated with CRC progression. Treatment with HTyr in colonic adenocarcinoma cells (CaCo2, HT-29 and WiDr) resulted in a decrease in EGFR expression through lysosomal and proteasomal machinery and subsequent halt in cell proliferation. HTyr further directs EGFR degradation by inducing ubiquitination of EGFR through phosphorylation of the docking site of Cbl (E3 ubiquitin-protein ligase), pY1045. Inhibition of EGFR and subsequent decrease in tumour growth by HTyr have been shown in animal model (HT-29 xenografts) as well(Reference Terzuoli, Giachetti and Ziche96). HTyr even capable of mounting cetuximab (EGFR inhibitor) action against CRC cells. The combination of HTyr and cetuximab showed stronger cytotoxicity against CRC adenocarcinoma cells (WiDr and HT-29) compared with cetuximab alone. This combinational treatment resulted in the cell cycle blockade at G2/M phase by down-regulating various cell cycle regulators such as cyclins B, D1 and E, and cyclin-dependent kinase (CDK)2, CDK4 and CDK6. Enhanced apoptosis (caspase-independent) and autophagy were also observed in colon cancer cells after the combination treatment. Remarkably, normal colon cells or human keratinocytes were least affected from this combinational therapy(Reference Terzuoli, Nannelli and Frosini54) which indicates diet containing HTyr during cetuximab therapy might protect healthy cells, for example, skin or haematopoietic cells from severe side effects of cetuximab in CRC patients receiving cetuximab. So, there is a possibility that hydroxytyrosol supplement to the patients receiving cetuximab therapy might improve the quality of patients’ life in the clinic. On this backdrop, it should be noted that HTyr action depends on its concentration being used in the experimental set-up as it may act as both anti- and pro-oxidant within the physiological system. When given at a higher dose (100 µM), HTyr showed pro-oxidant effects in CRC cells (SW480 and HCT116) and generated H2O2 to kill cancer cells(Reference Fabiani, Sepporta and Rosignoli97). On the other hand, at low doses (10µM), it is potent to counteract the DNA damage in peripheral blood mononuclear cells induced by external H2O2 treatment(Reference Rosignoli, Fuccelli and Sepporta98). It is also possible that the sensitivity of different cancer cells to HTyr treatment is inversely proportional to the ability of the different cells to remove hydrogen peroxide from the cell culture medium(Reference Rosignoli, Fuccelli and Sepporta98). However, different scientific communities disagreed with this hypothesis and they argued that sodium bicarbonate which is commonly present in cell culture media is responsible for pro-oxidant behaviour of HTyr at higher concentrations(Reference Odiatou, Skaltsounis and Constantinou99). Therefore, dosing of HTyr should be determined by considering the fact in mind that HTyr may act as either antioxidant or pro-oxidant depending on the concentration.

Not only HTyr but also metabolites generated by HTyr are also shown to act as antioxidants to protect intestinal cells (Caco-2 monolayers) from the oxidising action of oxidised cholesterol in in vitro culture conditions(Reference Atzeri, Lucas and Incani100). Especially, glucuronide and sulphate metabolites of Tyr and HTyr are capable to protect intestinal cells against pathological overproduction of nitric oxides(Reference Serreli, Melis and Corona101). The anti-cancerous effect of hydroxytyrosol acetate (HTyr-Ac) in human CRC cells (Caco-2/TC7) further demonstrated by another group of scientists. HTyr-Ac impeded the cell cycle by increasing p21 and CCNG2 (encodes Cyclin-G2) and down-regulating the CCNB1 (encodes Cyclin B1) gene expression. HTyr-Ac action is not only limited to cell cycle blockade in CRC cells as it can modify transcription of programmed cell death associated genes (BNIP3, BNIP3L, PDCD4 and ATF3) and can activate caspase-3. Carcinogen detoxification could be enhanced upon HTyr-Ac exposure, as it enhances UGT1A10 and CYP1A1, known xenobiotic-metabolising enzymes(Reference Mateos, Pereira-Caro and Bacon102). Thus, the secondary metabolites of HTyr especially HTyr-Ac should be characterised in humans to rule out any possibility of their negative effect on cell cycle or cell death in other parts of the body except the tumour site.

Apart from olive oil, olive mill wastewater could be a cheap source of HTyr as the purified olive mill wastewater shown to have chemopreventive properties in both human (HCT116 and HT-29) and murine (CT-26) CRC cells. In animal model, olive mill wastewater shown to suppress IL-8 and vascular endothelial growth factor expression and reduce tumour growth(Reference Bassani, Rossi and De Stefano53). Key findings from different studies using HTyr as intervention are summarised in Table 2.

Table 2. Summary of studies employed hydroxytyrosol as intervention in CRC

Oleuropein

Oleuropein, another important phenolic compound present in high concentration in olive oil and leaves(Reference Caponio, Alloggio and Gomes103) has gained scientific attention recently due to the accounted health benefits(Reference Sun, Frost and Liu104). Oleuropein can reduce CRC cell proliferation as well as invasion as shown in LoVo, a human colon cancer cell line(Reference Hamdi and Castellon105). Metabolic inhibition in cancer cell with oleuropein also documented in human colon cancer cells (HCT116). Inhibition of glycolysis and reduced cell viability was seen under the influence of oleuropein in tumour cells(Reference Ruzzolini, Peppicelli and Bianchini106). It could be an alternative approach to target cancer cells specifically via glycolysis inhibition as cancer cells are known for their high glycolytic activity. Studies in animal model also indicated the efficacy of oleuropein against colorectal carcinogenesis as it protected C57BL/6 mice from azoxymethane (AOM)/dextran sodium sulphate/) induced colitis through down-regulation of signalling pathways including Wnt/β-catenin, P3IK/Akt, NF-κB and STAT3. Oleuropein reduced the pro-inflammatory mediators such as IL-6, TNF-α, IFN-γ and IL-17A in mice group treated with AOM/dextran sodium sulphate by influencing the signalling cascades(Reference Giner, Recio and Ríos107). Oleuropein treatment also decreased the level of COX-2, Bax and PCNA (proliferating cell nuclear antigen protein) expression. Therefore, it could be a possibility that a diet containing oleuropein might prevent the chronification of intestinal inflammation and might be useful in colitis patients. In another in vivo study, oleuropein supplementation (125 mg/kg) reduced the formation of preneoplastic lesions in different segments of colon in AOM-treated A/J mice(Reference Sepporta, Fuccelli and Rosignoli108). AOM is known for inducing inflammation-driven CRC. Oleuropein action was specific to the tumour cells as it reduced AOM-driven tumour incidence from 57 % to 14 % in the medial segment of the colon and at the same time shown to protect peripheral leukocytes from AOM-induced DNA damage in the A/J mice(Reference Sepporta, Fuccelli and Rosignoli108). Pro-apoptotic effect of oleuropein in colon cancer cells also explored as oleuropein limits CRC cells’ growth by stimulating p53-dependent apoptosis(Reference Cárdeno, Sánchez-Hidalgo and Rosillo89). Hence, oleuropein could be effective against CRC by virtue of its anti-inflammatory properties and through regulating cellular signalling pathways. Significant findings of various studies with oleuropein are highlighted in Table 3.

Table 3. Summary of studies involving oleuropein as intervention in CRC

Oleocanthal

OC is a phenolic secoiridoid present in abundance in olive oil(Reference Cicerale, Conlan and Sinclair109). Mounting scientific evidences suggest that OC can be effective in different cancers like lung or breast cancer(Reference Elnagar, Sylvester and El Sayed110,Reference Siddique, Kilgore and Tajmim111) . In various type of cancer, inflammation plays crucial role in cancer development and progression. Therefore, tumour-associated inflammation has been a target for cancer therapy for decades. In this context, OC could play an important role in strategies to combat CRC development and progression as OC documented to have ibuprofen-like anti-inflammatory actions(Reference Beauchamp, Keast and Morel112). In in vitro study, OC shown to be more effective than ibuprofen (a non-steroidal anti-inflammatory drug) as an anti-inflammatory agent to inhibit COX-1 and COX-2, most common targets for anti-inflammatory drugs(Reference Parkinson and Cicerale113). In an interesting study conducted by Cusimano et al.(Reference Cusimano, Balasus and Azzolina114) OC was shown to be more effective than commonly used COX inhibitors such as nimesulide, indomethacin to reduce inflammation via COX suppression. Same study also reported that OC is capable of inducing apoptosis by inducing PARP cleavage as well by activating of caspases 3/7. However, anti-cancerous activities of OC in CRC cells might be independent of COX inhibition as OC is able to inhibit the cancer cell growth of both COX-2 positive (HT-29) and COX-2 negative (SW480) colonic adenocarcinoma cells with equal efficiency(Reference Cusimano, Balasus and Azzolina114).

A few studies also shed light on the anti-cancerous activities of OC in CRC through a wide variety of mechanisms. Exposure to a lower concentration of OC (2–5 µg/ml) induced apoptosis in HT-29 colon cancer cells by reducing anti-apoptotic protein Bcl-2. Cleavage of the poly-adenosine diphosphate-ribose polymerase (PARP) as well as caspase-3 related to the apoptosis cell death pathway observed under the influence of OC in HT-29 cells which consequently led to DNA fragmentation. The same study also shed light on the ability of OC to induce apoptosis and reduce cell viability through a different mechanism by means of suppressing COX-2 expression and activation of AMPK (adenosine monophosphate-activated protein kinase)(Reference Khanal, Oh and Yun115). On the flip side, higher concentration of OC (50 µM) induced apoptosis in CRC cells in a completely different mechanism by increasing intracellular reactive oxygen species level. Increased reactive oxygen species caused DNA damage and impairment of mitochondrial membrane integrity but fascinatingly normal cells remained unharmed after long-term exposure with even higher dosage of OC (100 µM)(Reference Cusimano, Balasus and Azzolina114). The crucial findings from the studies employed with OC are summarised in Table 4.

Table 4. Summary of oleocanthal mediated anti-CRC activities

Apigenin and luteolin

Apigenin and luteolin, two most important phenolic compounds belong to flavonoids group, have shown therapeutic potential in different cancers like melanoma and cervical cancer(Reference Tuorkey116). Apart from olive oil, these two compounds present at varying concentration in different other sources (pepper, carrot, celery, thyme, rosemary, oregano, etc.)(Reference Wang, Chen and Zhu117). Apigenin has been shown to reduce proliferation, migration and invasion of different CRC cells in a dose-dependent manner through down-regulation Wnt/β-catenin pathway. In particular, apigenin inhibited β-catenin activation and its nuclear entry, thereby downstream Wnt gene expression(Reference Xu, Wang and Song118). Wnt/β-catenin is particularly important in intestinal stem cell renewal during homoeostasis as well as played a significant role in intestinal diseases like CRC. Inhibitory action of apigenin on Wnt/β-catenin signalling further confirmed in CRC organoid model as in presence of apigenin intestinal organoid growth was significantly suppressed(Reference Xu, Wang and Song118). Apigenin possesses anti-metastatic properties as well, shown in BALB/c-nu mice where apigenin protected from metastasis in liver and lung(Reference Chunhua, Donglan and Xiuqiong119). Moreover, apigenin can accomplish pro-apoptotic role in CRC cells by boosting FADD (Fas-associated protein with death domain) expression and phosphorylation of FADD(Reference Wang, Yao and Wen120). It could synergistically augment the chemotherapeutic action of 5-Fluorouracil (5-FU), in a liposome formulation containing both apigenin and 5-FU. The combination therapy showed better efficacy than the drug alone in tumour xenograft model in nude mice(Reference Sen, Banerjee and Mandal121). It is also shown to regulate a range of cellular functions to combat CRC like NF-κB/Snail pathway(Reference Tong, Shen and Zhang122), PI3K/Akt/mTOR pathway, autophagy(Reference Chen, Xu and Yu123), STAT3 signalling(Reference Maeda, Takahashi and Nakai124), glycolysis(Reference Shan, Shi and Yang125) and gut microbiome(Reference Bian, Wan and Liao126). Apart from several health benefits of apigenin, at high concentration it may also act as a sedative(Reference Gazola, Costa and Castellanos127). So, there is a long road ahead before integrating apigenin in treatment protocol for CRC patients.

Another flavonoid, luteolin can inhibit colorectal carcinogenesis by activating Nrf2/ARE pathway through epigenetic modifications(Reference Zuo, Wu and Xiao128). It suppresses the expression of DNA methyltransferases whereas activated the expression of DNA demethylases to increase the Nrf2 expression. Nrf2 may then interact with p53 to direct CRC cell death via apoptosis(Reference Kang, Piao and Hyun129). Anti-tumour activities of luteolin may also depend on ERK1/2 as it ameliorated epithelial-to-mesenchymal transition in metastatic colon cancer cells, SW620 through activation of ERK1/2 and FOXO3a(Reference Potočnjak, Šimić and Gobin130). Luteolin can interfere in the cell cycle as well and can block cell cycle at the G2/M phase and induce apoptosis subsequently(Reference Chen, Zhang and Gao131). Furthermore, it is also shown to suppress CRC metastasis by regulating micro-RNA (miR-384) or CREB1 expression(Reference Yao, Rao and Zheng132,Reference Liu, Lang and Jin133) and also potent to reduce colon carcinogenesis by suppressing the matrix metalloproteinases in animal model(Reference Pandurangan, Dharmalingam and Sadagopan134). Significant studies with apigenin or luteolin in CRC are featured in Table 5.

Table 5. Summary of studies utilised apigenin and luteolin as intervention

Pharmacokinetics and toxicity profile of olive phenols

EVOO has several health benefits due to the presence of phenolic compounds. In this section, we have included a brief overview on the pharmacokinetics of the principal phenolics present in olive oil. Phenolic compounds are absorbed in a dose-dependent manner in the gut and go through intestinal/hepatic first-pass metabolism(Reference Visioli, Galli and Bornet135). Olive oil phenols are readily absorbed in the small intestine and colon by passive transport, though it depends on the vehicle employed(Reference Vissers, Zock and Roodenburg136). In that case, EVOO is considered as the best matrix for HTyr for its oily nature(Reference Alemán-Jiménez, Domínguez-Perles and Medina137). HTyr reaches maximum plasma concentration quickly (˜7 min) after intake. HTyr and its derivatives are well distributed in different tissues like muscle, liver, testis, brain and kidney(Reference Robles-Almazan, Pulido-Moran and Moreno-Fernandez138) and converted into both oxidised and methylated derivatives (like O-methylated derivative of HTyr, glucuronides of HTyr) revealed by HPLC analysis(Reference Mateos, Goya and Bravo139). Metabolites from HTyr and its derivatives are primarily excreted by the kidneys with a complete elimination time of approximately 6 h(Reference Rodríguez-Morató, Boronat and Kotronoulas140). However, one problem of hydroxytyrosol is its poor bioavailability as Covas et al.(Reference Covas, de la Torre and Farré-Albaladejo141) demonstrated that the maximum level of HTyr achieved in plasma was ˜15 µM when given diet of 40 ml of olive oil to healthy human volunteer (366 mg/kg). The reason behind this almost undetectable level (0·1–1 %) of free form of HTyr in body fluids is probably due to extensive first pass metabolism in both gut and liver(Reference de la Torre142,Reference Pastor, Rodríguez-Morató and Olesti143) . Hence, critical measurement of free HTyr in plasma possibly by novel methodologies would help to understand its dose-effect better.

On the other hand, the metabolism of oleuropein goes through the rapid degradation by colonic microflora to form HTyr, which significantly increases the amount of free HTyr. So, it should come into consideration while consuming crude extract containing both oleuropein and hydroxytyrosol, could increase the free HTyr level in plasma. Sulphated and glucuronidated metabolites of HTyr are the primary metabolites of oleuropein in plasma and urine in humans(Reference de Bock, Thorstensen and Derraik144).

In the case of OC, it is believed that passive diffusion of OC in small intestine is possible(Reference Lozano-Castellón, López-Yerena and Rinaldi de Alvarenga145) and it is rapidly hydrolysed through the gastrointestinal tract(Reference Mehmood, Usman and Patil146). OC is mainly metabolised by phase I reactions (hydration, hydrogenation and hydroxylation) and mainly happens in the liver and small intestine. The hydrogenated and hydrated metabolites of OC are further glucuronidated through phase II reactions(Reference López-Yerena, Vallverdú-Queralt and Mols147). However, oral bioavailability of OC is compromised due to the high intestinal metabolism. Despite the current surge of research with OC due to its anti-inflammatory properties, its absorption, distribution, metabolism and excretion properties are not well characterised. Therefore, extensive in vivo analysis with OC is crucial to develop it as a therapeutic intervention.

Comparatively, flavones (apigenin and luteolin) are less absorbed with < 1 µmol/l plasma concentration in human compared with other polyphenols(Reference Hostetler, Ralston and Schwartz148). Apigenin is also well distributed into the tissues after administration in rat or mice(Reference Wang, Firrman and Liu149). After absorption, apigenin remains in blood circulation or tissues in the form of glucuronide, sulphate conjugates or luteolin as these are the major metabolites of apigenin(Reference Tang, Chen and Huang150). Apigenin has a slow elimination rate and possibly accumulates in the body(Reference Gradolatto, Basly and Berges151). Despite the numerous favourable effects of apigenin, in vivo studies involving animal model as well as human studies are considerably less in number which is probably because of apigenin’s low water solubility (1·35 μg/ml) and high permeability(Reference Salehi, Venditti and Sharifi-Rad152). Therefore, different methodologies such as liposome, nanosuspension and micelle have been explored by different groups to improve the solubility and bioavailability of apigenin(Reference Ding, Chen and Wang153,Reference Zhai, Guo and Liu154) . On the other hand, glucuronidation and methylation are major metabolic pathways of luteolin in humans which are mediated by UDP-glucuronosyltransferases and catechol-O-methyltransferases, respectively (Reference Wang, Chen and Zhu117). Monoglucuronide form of luteolin is predominant in human serum(Reference Shimoi, Okada and Furugori155). Apigenin and luteolin are mainly excreted in bile, urine or faeces(Reference Tang, Chen and Huang150,Reference Simons, Renouf and Murphy156) .

Toxicological studies along with the in-vitro genotoxicity studies revealed HTyr as a non-mutagenic, non-genotoxic compound and advocate for its long-term consumption(Reference Bertelli, Kiani and Paolacci157). Even at very high dose (500 mg/kg/d), HTyr exerts no adverse effects in rats(Reference Auñon-Calles, Canut and Visioli158). Since 2011, European Food Safety Authority authorised health claim on olive oil containing at least 250 mg/kg of hydroxytyrosol and its derivatives(Reference López-Huertas, Lozano-Sánchez and Segura-Carretero159). Ames test results ascertain that neither apigenin nor luteolin is mutagenic or toxic(Reference Czeczot, Tudek and Kusztelak160). Overall, olive oil phenolics are considered safe(Reference Romani, Ieri and Urciuoli161,Reference Ashrafizadeh, Bakhoda and Bahmanpour162) although recently Kouka et al.(Reference Kouka, Tekos and Papoutsaki163) revealed that protective action of olive oil may be tissue specific and it can act as both antioxidant (in brain or muscle tissues) and pro-oxidants in tissues such as spleen or pancreas as shown in male Wistar rats(Reference Kouka, Tekos and Papoutsaki163). Therefore, effect of olive oil on different human organs should be exploited critically before developing the dosing protocol.

The complete metabolic profile of OOP is yet to be elucidated. To develop OOP as clinical intervention, biological relevance of phenolic metabolites should be characterised. Further efforts are needed to increase the bioavailability of HTyr or apigenin possibly by changing the solubility. Novel formulation strategies are crucial in this sense for better absorption of phenolic compounds, especially for flavonoids.

Discussion

Olive oil is full of beneficial components which may turn useful for the prevention and possible therapeutic intervention in CRC. Mounting evidence advocates the chemotherapeutic potentiality of olive oil phenolic compounds, particularly in CRC. The phenolic components of olive oil can act on different stages of carcinogenesis process, such as oxidative stress, inflammation, cell cycle, immune regulation, apoptosis as well as an epigenetic alteration. Waste products produced during olive oil extraction may also be used as a cheap alternative of olive oil to develop food supplement to combat CRC. Altering the gut microbiome could hold the key to amend several intestinal disorders including CRC. On that background, a few studies have already provided evidences to link imbalance of the intestinal microbiota and occurrence of CRC. On the other hand, EVOO is capable of altering the gut microbial population by stimulating the growth of beneficial bacteria, for example, lactic acid bacteria(Reference Luisi, Lucarini and Biffi164) and at the same time reducing the abundance of pathogenic bacteria (e.g. Enterococcus, Staphylococcus)(Reference Rodríguez-García, Sánchez-Quesada and Algarra75). EVOO also possesses anti-inflammatory effects in the gut by producing SCFA(Reference Millman, Okamoto and Teruya165). Because of the significant role played by gut microbiome for maintaining cellular integrity and protecting against pathogenic organisms, any changes in the gut microbial community can exert adverse effects. For example, during the intestinal dysbiosis, disruption of the homoeostasis between the host and the intestinal microbiota occurs(Reference Zhang, Li and Gan166,Reference Iebba, Totino and Gagliardi167) , which turns out to be one of the major causes of inflammatory bowel disease(Reference Schippa and Conte168,Reference Nagao-Kitamoto, Kitamoto and Kuffa169) and eventual progression to CRC(Reference Wu, Yang and Zhang170,Reference Sheflin, Whitney and Weir171) . Therefore, maintenance or restoration of homoeostasis of intestinal microbiota could be a substantial treatment or prevention strategy against the CRC. In this context, olive oil and its phenolic compounds could be useful to restore/modify gut microbiome for good and prevent carcinogenesis.

Conclusion and future direction

Most of the potential benefits olive oil discussed in the current review have emerged mainly from in vitro studies and animal studies. Therefore, additional efforts are need of the hour to mechanistically characterise biological activities of EVOO or individual phenolic components in human. Pharmacokinetics and pharmacodynamics must be studied extensively to develop the effective dose of these compounds. The relation between the structure and activity of these olive oil phenolics should be deciphered to engineer new drugs based on the molecular scaffold of these olive oil components. Further, clinical trial with hydroxytyrosol or oleuropein or the combination of different components from olive oil must be started immediately to develop a chemopreventive strategy or therapeutic intervention. This current review critically assessed the potential of olive oil phenolic constitutes as a preventive or possible therapeutic agent in CRC by studying the molecular mechanism of the each of the olive oil phenolic compounds and the olive oil phenolic extract as a whole (summarised in Fig. 4). As the exploration to find the novel and cheap therapeutic strategy against CRC lingers, interventions by means of various olive oil-derived phenolic compounds may ultimately turn out to be a precise management system to control or prevent CRC.

Fig. 4. Summary of the key anti-colorectal cancer activities of olive oil and its phenolic compounds. The major activities of each phenolic compound are shown here. For each activity, molecular pathways/signalling molecules targeted by olive oil phenolic compounds demonstrated here by indicating upward arrowhead (↑ = up-regulating), downward arrowhead (↓ = down-regulating) and cross sign (× = blocking).

Acknowledgements

The authors greatly acknowledge the help of authority of MAKAUT, WB for providing the necessary support.

This study was supported by DST Inspire faculty research grant (DST/INSPIRE/04/2017/000675; India) and MAKAUT, WB research seed grant to D. N. A. S. was supported by Junior research fellowship from Council of Scientific and Industrial research (Award no: 09/1213(0002)/2019-EMR-I; India). S. S. was supported by the scholarship from Department of Biotechnology, Govt. of India.

All authors participated in data curation, developing the methodology and writing the original draft of the paper. A. S. and D. N. were responsible for conceptualisation, formal analysis, investigation and reviewing and editing subsequent versions of the paper. A. S. was solely responsible for visualisation, while D. N. was solely responsible for funding acquisition, project administration, handling of resources, and supervision and validation of the work

There are no conflicts of interest.

References

Rawla, P, Sunkara, T & Barsouk, A (2019) Epidemiology of colorectal cancer: incidence, mortality, survival, and risk factors. Prz Gastroenterol 14, 89103.Google ScholarPubMed
Sümbül, HE & Akkız, H (2019) Importance of autophagy in colorectal cancer: a cross-sectional study. J Surg Med 3, 246249.Google Scholar
Bray, F, Ferlay, J, Soerjomataram, I, et al. (2018) Global Cancer Statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 68, 394424.CrossRefGoogle ScholarPubMed
Siegel, RL, Miller, KD, Fedewa, SA, et al. (2017) Colorectal cancer statistics, 2017. CA Cancer J Clin 67, 177193.CrossRefGoogle ScholarPubMed
Wang, ST, Cui, WQ, Pan, D, et al. (2020) Tea polyphenols and their chemopreventive and therapeutic effects on colorectal cancer. World J Gastroenterol 26, 562597.CrossRefGoogle ScholarPubMed
Yamagishi, H, Kuroda, H, Imai, Y, et al. (2016) Molecular pathogenesis of sporadic colorectal cancers. Chin J Cancer 35, 4.CrossRefGoogle ScholarPubMed
Kim, ER & Chang, DK (2014) Colorectal cancer in inflammatory bowel disease: the risk, pathogenesis, prevention and diagnosis. World J Gastroenterol 20, 98729881.CrossRefGoogle ScholarPubMed
Nurgali, K, Jagoe, RT & Abalo, R (2018) Editorial: adverse effects of cancer chemotherapy: anything new to improve tolerance and reduce sequelae? Front Pharmacol 9, 245.CrossRefGoogle ScholarPubMed
Redondo-Blanco, S, Fernández, J, Gutiérrez-Del-Río, I, et al. New insights toward colorectal cancer chemotherapy using natural bioactive compounds. Front Pharmacol 8, 109.Google Scholar
Gillis, A & Eminger, L (2020) Hypogeusia and hyposmia with topical 5-fluorouracil treatment. JAAD Case Rep 6, 650651.CrossRefGoogle ScholarPubMed
Grady, WM & Carethers, JM (2008) Genomic and epigenetic instability in colorectal cancer pathogenesis. Gastroenterology 135, 10791099.CrossRefGoogle ScholarPubMed
Mármol, I, Sánchez-de-Diego, C, Pradilla Dieste, A, et al. (2017) Colorectal carcinoma: a general overview and future perspectives in colorectal cancer. Int J Mol Sci 18, 197.CrossRefGoogle ScholarPubMed
Tsang, AHF, Cheng, KH, Wong, ASP, et al. (2014) Current and future molecular diagnostics in colorectal cancer and colorectal adenoma. World J Gastroenterol 20, 38473857.CrossRefGoogle ScholarPubMed
Damilakis, E, Mavroudis, D, Sfakianaki, M, et al. (2020) Immunotherapy in metastatic colorectal cancer: could the latest developments hold the key to improving patient survival? Cancers 12, 889.CrossRefGoogle ScholarPubMed
Nguyen, H & Duong, H (2018) The molecular characteristics of colorectal cancer: implications for diagnosis and therapy. Oncol Lett 16, 918.Google ScholarPubMed
Ponz de Leon, M & Percesepe, A (2000) Pathogenesis of colorectal cancer. Dig Liver Dis 32, 807821.CrossRefGoogle ScholarPubMed
Turano, M, Delrio, P, Rega, D, et al. (2019) Promising colorectal cancer biomarkers for precision prevention and therapy. Cancers 11, 1932.CrossRefGoogle ScholarPubMed
Mori, Y, Yin, J, Rashid, A, et al. (2001) Instabilotyping: comprehensive identification of frameshift mutations caused by coding region microsatellite instability. Cancer Res 61, 60466049.Google ScholarPubMed
De Palma, FDE, D’Argenio, V, Pol, J, et al. (2019) The molecular hallmarks of the serrated pathway in colorectal cancer. Cancers 11, 1017.CrossRefGoogle ScholarPubMed
Advani, SM, Advani, PS, Brown, DW, et al. (2019) global differences in the prevalence of the CpG island methylator phenotype of colorectal cancer. BMC Cancer 19, 964.CrossRefGoogle ScholarPubMed
Curtin, K, Slattery, ML & Samowitz, WS (2011) CpG island methylation in colorectal cancer: past, present and future. Patholog Res Int 2011, 902674.Google ScholarPubMed
To, KKW, Tong, CWS, Wu, M, et al. (2018) MicroRNAs in the prognosis and therapy of colorectal cancer: from bench to bedside. World J Gastroenterol 24, 29492973.CrossRefGoogle ScholarPubMed
Yao, RW, Wang, Y & Chen, LL (2019) Cellular functions of long noncoding RNAs. Nat Cell Biol 21, 542551.CrossRefGoogle ScholarPubMed
Lin, J, Chuang, CC & Zuo, L (2017) Potential roles of MicroRNAs and ROS in colorectal cancer: diagnostic biomarkers and therapeutic targets. Oncotarget 8, 1732817346.CrossRefGoogle ScholarPubMed
Galamb, O, Barták, BK, Kalmár, A, et al. (2019) Diagnostic and prognostic potential of tissue and circulating long non-coding RNAs in colorectal tumors. World J Gastroenterol 25, 50265048.CrossRefGoogle ScholarPubMed
Ranković, B, Zidar, N, Žlajpah, M, et al. (2019) Epithelial-mesenchymal transition-related MicroRNAs and their target genes in colorectal cancerogenesis. J Clin Med 8, 1603.CrossRefGoogle ScholarPubMed
Bermúdez, M, Aguilar-Medina, M, Lizárraga-Verdugo, E, et al. (2019) LncRNAs as regulators of autophagy and drug resistance in colorectal cancer. Front Oncol 9, 1008.CrossRefGoogle ScholarPubMed
Corrà, F, Agnoletto, C, Minotti, L, et al. (2018) The network of non-coding RNAs in cancer drug resistance. Front Oncol 8, 327.CrossRefGoogle Scholar
Wood, LD, Parsons, DW, Jones, S, et al. (2007) The genomic landscapes of human breast and colorectal cancers. Science 318, 11081113.CrossRefGoogle ScholarPubMed
Pennel, KAF, Park, JH, McMillan, DC, et al. (2019) Signal interaction between the tumour and inflammatory cells in patients with gastrointestinal cancer: implications for treatment. Cell Signal 54, 8190.CrossRefGoogle ScholarPubMed
Koveitypour, Z, Panahi, F, Vakilian, M, et al. (2019) Signaling pathways involved in colorectal cancer progression. Cell Biosci 9, 97.CrossRefGoogle ScholarPubMed
He, L, Zhu, H, Zhou, S, et al. (2018) Wnt pathway is involved in 5-FU drug resistance of colorectal cancer cells. Exp Mol Med 50, 112.CrossRefGoogle ScholarPubMed
Yuan, S, Tao, F, Zhang, X, et al. (2020) Role of Wnt/β-catenin signaling in the chemoresistance modulation of colorectal cancer. Biomed Res Int 2020, 9390878.CrossRefGoogle ScholarPubMed
Koch, S (2017) Extrinsic control of Wnt signaling in the intestine. Differ 97, 18.CrossRefGoogle ScholarPubMed
Vanuytsel, T, Senger, S, Fasano, A, et al. (2013) Major signaling pathways in intestinal stem cells. Biochim Biophys Acta 1830, 24102426.CrossRefGoogle ScholarPubMed
Kaemmerer, E, Jeon, MK, Berndt, A, et al. (2019) Targeting Wnt signaling via notch in intestinal carcinogenesis. Cancers 11, 555.CrossRefGoogle ScholarPubMed
Komiya, Y & Habas, R (2008) Wnt signal transduction pathways. Organogenesis 4, 6875.CrossRefGoogle ScholarPubMed
Martini, E, Krug, SM, Siegmund, B, et al. (2017) Mend your fences: the epithelial barrier and its relationship with mucosal immunity in inflammatory bowel disease. Cell Mol Gastroenterol Hepatol 4, 3346.CrossRefGoogle ScholarPubMed
Armaghany, T, Wilson, JD, Chu, Q, et al. (2012) Genetic alterations in colorectal cancer. Gastrointest Cancer Res 5, 1927.Google ScholarPubMed
Kirsanov, K, Fetisov, T, Lesovaya, EA, et al. (2020) Prevention of colorectal carcinogenesis by DNA-binding small-molecule curaxin CBL0137 involves suppression of Wnt signaling. Cancer Prev Res 13, 5364.CrossRefGoogle ScholarPubMed
Cheng, X, Xu, X, Chen, D, et al. (2019) Therapeutic potential of targeting the Wnt/β-catenin signaling pathway in colorectal cancer. Biomed Pharmacother 110, 473481.CrossRefGoogle ScholarPubMed
Hankey, W, Frankel, WL & Groden, J (2018) Functions of the APC tumor suppressor protein dependent and independent of canonical WNT signaling: implications for therapeutic targeting. Cancer Metastasis Rev 37, 159172.CrossRefGoogle ScholarPubMed
Yaeger, R, Chatila, WK, Lipsyc, MD, et al. (2018) Clinical sequencing defines the genomic landscape of metastatic colorectal cancer. Cancer Cell 33, 125136.e3.CrossRefGoogle ScholarPubMed
Goldsberry, WN, Londoño, A, Randall, TD, et al. (2019) A review of the role of Wnt in cancer immunomodulation. Cancers 11, 771.CrossRefGoogle ScholarPubMed
Krasinskas, AM (2011) EGFR signaling in colorectal carcinoma. Patholog Res Int 2011, 932932.Google ScholarPubMed
Zhang, J, Roberts, TM & Shivdasani, RA (2011) Targeting PI3K signaling as a therapeutic approach for colorectal cancer. Gastroenterology 141, 5061.CrossRefGoogle ScholarPubMed
Papadatos-Pastos, D, Rabbie, R, Ross, P, et al. (2015) The role of the PI3K pathway in colorectal cancer. Crit Rev Oncol Hematol 94, 1830.CrossRefGoogle ScholarPubMed
Wang, Q, Shi, YL, Zhou, K, et al. (2018) PIK3CA mutations confer resistance to first-line chemotherapy in colorectal cancer. Cell Death Dis 9, 111.Google ScholarPubMed
Yang, ZH, Dang, YQ & Ji, G (2019) Role of epigenetics in transformation of inflammation into colorectal cancer. World J Gastroenterol 25, 28632877.CrossRefGoogle Scholar
Li, Y, Lin, Z, Chen, B, et al. (2017) Ezrin/NF-KB activation regulates epithelial- mesenchymal transition induced by egf and promotes metastasis of colorectal cancer. Biomed Pharmacother 92, 140148.CrossRefGoogle ScholarPubMed
Dariya, B, Rajitha, B, Alam, A, et al. (2020) Therapeutic Role of Phytochemicals in Colorectal Cancer. Theranostics Approaches to Gastric and Colon Cancer. Singapore: Springer.Google Scholar
Papanikolaou, C, Melliou, E & Magiatis, P (2019) Olive Oil Phenols. Functional Foods. London: IntechOpen.Google Scholar
Bassani, B, Rossi, T, De Stefano, D, et al. (2016) Potential chemopreventive activities of a polyphenol rich purified extract from olive mill wastewater on colon cancer cells. J Funct Foods 27, 236248.CrossRefGoogle Scholar
Terzuoli, E, Nannelli, G, Frosini, M, et al. (2017) Inhibition of cell cycle progression by the hydroxytyrosol-cetuximab combination yields enhanced chemotherapeutic efficacy in colon cancer cells. Oncotarget 8, 8320783224.CrossRefGoogle ScholarPubMed
Torić, J, Marković, AK, Brala, CJ, et al. (2019) Anticancer effects of olive oil polyphenols and their combinations with anticancer drugs. Acta Pharm 69, 461482.CrossRefGoogle ScholarPubMed
Pelucchi, C, Bosetti, C, Negri, E, et al. (2011) Olive oil and cancer risk: an update of epidemiological findings through 2010. Curr Pharm 17, 805812.CrossRefGoogle ScholarPubMed
Soriguer, F, Almaraz, MC, Ruiz-de-Adana, MS, et al. (2009) Incidence of obesity is lower in persons who consume olive oil. Eur J Clin Nutr 63, 13711374.CrossRefGoogle ScholarPubMed
Schwingshackl, L, Lampousi, AM, Portillo, MP, et al. (2017) Olive oil in the prevention and management of type 2 diabetes mellitus: a systematic review and meta-analysis of cohort studies and intervention trials. Nutr Diabetes 7, e262e262.CrossRefGoogle ScholarPubMed
Román, GC, Jackson, RE, Reis, J, et al. (2019) Extra-virgin olive oil for potential prevention of Alzheimer disease. Rev Neurol 175, 705723.CrossRefGoogle ScholarPubMed
Borzì, AM, Biondi, A, Basile, F, et al. Olive oil effects on colorectal cancer. Nutrients 11, 32.CrossRefGoogle Scholar
Romero, C & Brenes, M (2012) Analysis of total contents of hydroxytyrosol and tyrosol in olive oils. J Agric Food Chem 60, 90179022.CrossRefGoogle ScholarPubMed
Medeiros de Azevedo, W, Ferreira Ribeiro de Oliveira, L, Alves Alcântara, M, et al. (2020) Physicochemical characterization, fatty acid profile, antioxidant activity and antibacterial potential of cacay oil, coconut oil and cacay butter. PLoS One 15, e0232224.CrossRefGoogle ScholarPubMed
Ouni, Y, Taamalli, A, Gómez-Caravaca, AM, et al. (2011) Characterisation and quantification of phenolic compounds of extra-virgin olive oils according to their geographical origin by a rapid and resolutive LC-ESI-TOF MS method. Food Chem 127, 12631267.CrossRefGoogle ScholarPubMed
Aykan, NF (2015) Red meat and colorectal cancer. Oncol Rev 9, 288.Google ScholarPubMed
Trefflich, I, Marschall, HU, Giuseppe, RD, et al. (2019) Associations between dietary patterns and bile acids-results from a cross-sectional study in vegans and omnivores. Nutrients 12, 47.CrossRefGoogle ScholarPubMed
Imran, M, Nadeem, M, Gilani, SA, et al. (2018) Antitumor perspectives of oleuropein and its metabolite hydroxytyrosol: recent updates. J Food Sci 83, 17811791.CrossRefGoogle ScholarPubMed
Stoneham, M, Goldacre, M, Seagroatt, V, et al. (2000) Olive oil, diet and colorectal cancer: an ecological study and a hypothesis. J Epidemiol Community Health 54, 756760.CrossRefGoogle ScholarPubMed
Serra, G, Incani, A, Serreli, G, et al. (2018) Olive oil polyphenols reduce oxysterols -induced redox imbalance and pro-inflammatory response in intestinal cells. Redox Biol 17, 348354.CrossRefGoogle ScholarPubMed
Cariello, M, Contursi, A, Gadaleta, RM, et al. (2020) Extra-virgin olive oil from Apulian cultivars and intestinal inflammation. Nutrients 12, 1084.CrossRefGoogle ScholarPubMed
Centrone, M, D’Agostino, M, Difonzo, G, et al. (2020) Antioxidant efficacy of olive by-product extracts in human colon HCT8 cells. Foods 10, 11.CrossRefGoogle ScholarPubMed
Llor, X, Pons, E, Roca, A, et al. (2003) The effects of fish oil, olive oil, oleic acid and linoleic acid on colorectal neoplastic processes. Clin Nutr 22, 7179.CrossRefGoogle ScholarPubMed
Costea, T, Hudiţă, A, Ciolac, OA, et al. (2018) Chemoprevention of colorectal cancer by dietary compounds. Int J Mol Sci 19, 3787.CrossRefGoogle ScholarPubMed
Gill, CIR, Boyd, A, McDermott, E, et al. (2005) Potential anti-cancer effects of virgin olive oil phenols on colorectal carcinogenesis models in vitro . Int J Cancer 117, 17.CrossRefGoogle ScholarPubMed
Bartolí, R, Fernández-Bañares, F, Navarro, E, et al. (2000) Effect of olive oil on early and late events of colon carcinogenesis in rats: modulation of arachidonic acid metabolism and local prostaglandin E(2) synthesis. Gut 46, 191199.CrossRefGoogle ScholarPubMed
Rodríguez-García, C, Sánchez-Quesada, C, Algarra, I, et al. (2020) The high-fat diet based on extra-virgin olive oil causes dysbiosis linked to colorectal cancer prevention. Nutrients 12, 1705.CrossRefGoogle ScholarPubMed
López de Las Hazas, MC, Piñol, C, Macià, A, et al. (2017) 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 65, 64676476.CrossRefGoogle ScholarPubMed
Hashim, YZHY, Rowland, IR, McGlynn, H, et al. (2008) Inhibitory effects of olive oil phenolics on invasion in human colon adenocarcinoma cells in vitro . Int J Cancer 122, 495500.CrossRefGoogle ScholarPubMed
Hashim, YZHY, Worthington, J, Allsopp, P, et al. (2014) Virgin olive oil phenolics extract inhibit invasion of HT115 human colon cancer cells in vitro and in vivo . Food Funct 5, 15131519.CrossRefGoogle ScholarPubMed
Pampaloni, B, Mavilia, C, Fabbri, S, et al. (2014) In vitro effects of extracts of extra virgin olive oil on human colon cancer cells. Nutr Cancer 66, 12281236.CrossRefGoogle ScholarPubMed
Banks, LD, Amoah, P, Niaz, MS, et al. (2016) Olive oil prevents Benzo(a)Pyrene [B(a)P]-induced colon carcinogenesis through altered B(a)P metabolism and decreased oxidative damage in ApcMin mouse model. J Nutr Biochem 28, 3750.CrossRefGoogle Scholar
Sánchez-Fidalgo, S, Villegas, I, Cárdeno, A, et al. (2010) Extra-virgin olive oil-enriched diet modulates DSS-Colitis-Associated colon carcinogenesis in mice. Clin Nutr 29, 663673.CrossRefGoogle ScholarPubMed
Nanda, N, Mahmood, S, Bhatia, A, et al. (2019) Chemopreventive role of olive oil in colon carcinogenesis by targeting noncoding RNAs and methylation machinery: chemopreventive role of olive oil in colon carcinogenesis. Int J Cancer 144, 11801194.CrossRefGoogle ScholarPubMed
Torić, J, Brozovic, A, Baus Lončar, M, et al. (2020) Biological activity of phenolic compounds in extra virgin olive oils through their phenolic profile and their combination with anticancer drugs observed in human cervical carcinoma and colon adenocarcinoma cells. Antioxidants 9, 453.CrossRefGoogle ScholarPubMed
Karković Marković, A, Torić, J, Barbarić, M, et al. (2019) Hydroxytyrosol, tyrosol and derivatives and their potential effects on human health. Molecules 24, 2001.CrossRefGoogle ScholarPubMed
Toteda, G, Lupinacci, S, Vizza, D, et al. (2017) High doses of hydroxytyrosol induce apoptosis in papillary and follicular thyroid cancer cells. J Endocrinol Invest 40, 153162.CrossRefGoogle ScholarPubMed
Chimento, A, Casaburi, I, Rosano, C, et al. (2014) Oleuropein and hydroxytyrosol activate GPER/GPR30-dependent pathways leading to apoptosis of ER-negative SKBR3 breast cancer cells. Mol Nutr Food Res 58, 478489.CrossRefGoogle ScholarPubMed
Ye, YL, Chang, HS, Tseng, YF, et al. (2017) Suppression of IL-8 release by sweet olive ethanolic extract and compounds in WiDr colon adenocarcinoma cells. J Food Sci 82, 17921798.CrossRefGoogle ScholarPubMed
Sun, L, Luo, C & Liu, J (2014) Hydroxytyrosol induces apoptosis in human colon cancer cells through ROS generation. Food Funct 5, 19091914.CrossRefGoogle ScholarPubMed
Cárdeno, A, Sánchez-Hidalgo, M, Rosillo, MA, et al. (2013) Oleuropein, a secoiridoid derived from olive tree, inhibits the proliferation of human colorectal cancer cell through downregulation of HIF-1α . Nutr Cancer 65, 147156.CrossRefGoogle ScholarPubMed
Corona, G, Deiana, M, Incani, A, et al. (2007) Inhibition of P38/CREB phosphorylation and cox-2 expression by olive oil polyphenols underlies their anti-proliferative effects. Biochem Biophys Res Commun 362, 606611.CrossRefGoogle ScholarPubMed
Corona, G, Deiana, M, Incani, A, et al. (2009) Hydroxytyrosol inhibits the proliferation of human colon adenocarcinoma cells through inhibition of ERK1/2 and Cyclin D1. Mol Nutr Food Res 53, 897903.CrossRefGoogle ScholarPubMed
Notarnicola, M, Pisanti, S, Tutino, V, et al. (2011) Effects of olive oil polyphenols on fatty acid synthase gene expression and activity in human colorectal cancer cells. Genes Nutr 6, 6369.CrossRefGoogle ScholarPubMed
Bueno, MJ, Jimenez-Renard, V, Samino, S, et al. (2019) Essentiality of fatty acid synthase in the 2D to anchorage-independent growth transition in transforming cells. Nat Commun 10, 5011.CrossRefGoogle ScholarPubMed
Di Francesco, A, Falconi, A, Di Germanio, C, et al. (2015) Extravirgin olive oil up-regulates CB1 tumor suppressor gene in human colon cancer cells and in rat colon via epigenetic mechanisms. J Nutr Biochem 26, 250258.CrossRefGoogle ScholarPubMed
Bernini, R, Carastro, I, Palmini, G, et al. (2017) 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 65, 65066512.CrossRefGoogle Scholar
Terzuoli, E, Giachetti, A, Ziche, M, et al. (2016) Hydroxytyrosol, a product from olive oil, reduces colon cancer growth by enhancing epidermal growth factor receptor degradation. Mol Nutr Food Res 60, 519529.CrossRefGoogle ScholarPubMed
Fabiani, R, Sepporta, MV, Rosignoli, P, et al. (2012) Anti-proliferative and pro-apoptotic activities of hydroxytyrosol on different tumour cells: the role of extracellular production of hydrogen peroxide. Eur J Nutr 51, 455464.CrossRefGoogle ScholarPubMed
Rosignoli, P, Fuccelli, R, Sepporta, MV, et al. (2016) In vitro chemo-preventive activities of hydroxytyrosol: the main phenolic compound present in extra-virgin olive oil. Food Funct 7, 301307.CrossRefGoogle ScholarPubMed
Odiatou, EM, Skaltsounis, AL & Constantinou, AI (2013) Identification of the factors responsible for the in vitro pro-oxidant and cytotoxic activities of the olive polyphenols oleuropein and hydroxytyrosol. Cancer Lett 330, 113121.CrossRefGoogle ScholarPubMed
Atzeri, A, Lucas, R, Incani, A, et al. (2016) Hydroxytyrosol and tyrosol sulfate metabolites protect against the oxidized cholesterol pro-oxidant effect in Caco-2 human enterocyte-like cells. Food Funct 7, 337346.CrossRefGoogle ScholarPubMed
Serreli, G, Melis, MP, Corona, G, et al. (2019) Modulation of LPS-Induced nitric oxide production in intestinal cells by hydroxytyrosol and tyrosol metabolites: insight into the mechanism of action. Food Chem Toxicol 125, 520527.CrossRefGoogle ScholarPubMed
Mateos, R, Pereira-Caro, G, Bacon, JR, et al. (2013) Anticancer activity of olive oil hydroxytyrosyl acetate in human adenocarcinoma Caco-2 Cells. J Agric Food Chem 61, 32643269.CrossRefGoogle ScholarPubMed
Caponio, F, Alloggio, V & Gomes, T (1999) Phenolic compounds of virgin olive oil: influence of paste preparation techniques. Food Chem 64, 203209.CrossRefGoogle Scholar
Sun, W, Frost, B & Liu, J (2017) Oleuropein, unexpected benefits! Oncotarget 8, 17409.CrossRefGoogle ScholarPubMed
Hamdi, HK & Castellon, R (2005) Oleuropein, a non-toxic olive iridoid, is an anti-tumor agent and cytoskeleton disruptor. Biochem Biophys Res Commun 334, 769778.CrossRefGoogle ScholarPubMed
Ruzzolini, J, Peppicelli, S, Bianchini, F, et al. (2020) Cancer glycolytic dependence as a new target of olive leaf extract. Cancers 12, 317.CrossRefGoogle ScholarPubMed
Giner, E, Recio, MC, Ríos, JL, et al. (2016) Chemopreventive effect of oleuropein in colitis-associated colorectal cancer in C57bl/6 mice. Mol Nutr Food Res 60, 242255.CrossRefGoogle ScholarPubMed
Sepporta, MV, Fuccelli, R, Rosignoli, P, et al. (2016) Oleuropein prevents azoxymethane-induced colon crypt dysplasia and leukocytes DNA damage in A/J mice. J Med Food 19, 983989.CrossRefGoogle ScholarPubMed
Cicerale, S, Conlan, XA, Sinclair, AJ, et al. (2008) Chemistry and health of olive oil phenolics. Crit Rev Food Sci Nutr 49, 218236.CrossRefGoogle Scholar
Elnagar, AY, Sylvester, PW & El Sayed, K (2011) (-)-Oleocanthal as a c-Met inhibitor for the control of metastatic breast and prostate cancers. Planta Med 77, 10131019.CrossRefGoogle ScholarPubMed
Siddique, AB, Kilgore, PCSR, Tajmim, A, et al. (2020) (-)-Oleocanthal as a dual c-MET-COX2 inhibitor for the control of lung cancer. Nutrients 12, 1749.CrossRefGoogle ScholarPubMed
Beauchamp, GK, Keast, RSJ, Morel, D, et al. (2005) Phytochemistry: ibuprofen-like activity in extra-virgin olive oil: phytochemistry. Nature 437, 4546.CrossRefGoogle ScholarPubMed
Parkinson, L & Cicerale, S (2016) The health benefiting mechanisms of virgin olive oil phenolic compounds. Molecules 21, 1734.CrossRefGoogle ScholarPubMed
Cusimano, A, Balasus, D, Azzolina, A, et al. (2017) Oleocanthal exerts antitumor effects on human liver and colon cancer cells through ROS generation. Int J Oncol 51, 533544.CrossRefGoogle ScholarPubMed
Khanal, P, Oh, WK, Yun, HJ, et al. (2011) P-HPEA-EDA, a phenolic compound of virgin olive oil, activates amp-activated protein kinase to inhibit carcinogenesis. Carcinog 32, 545553.CrossRefGoogle ScholarPubMed
Tuorkey, MJ (2016) Molecular targets of luteolin in cancer. Eur J Cancer Prev 25, 6576.CrossRefGoogle ScholarPubMed
Wang, L, Chen, Q, Zhu, L, et al. (2017) Metabolic disposition of luteolin is mediated by the interplay of udp-glucuronosyltransferases and Catechol-O-Methyltransferases in rats. Drug Metab Dispos 45, 306315.CrossRefGoogle ScholarPubMed
Xu, M, Wang, S, Song, YU, et al. (2016) Apigenin suppresses colorectal cancer cell proliferation, migration and invasion via inhibition of the Wnt/β-Catenin signaling pathway. Oncol Lett 11, 30753080.CrossRefGoogle ScholarPubMed
Chunhua, L, Donglan, L, Xiuqiong, F, et al. (2013) Apigenin up-regulates transgelin and inhibits invasion and migration of colorectal cancer through decreased phosphorylation of AKT. J Nutr Biochem 24, 17661775.CrossRefGoogle ScholarPubMed
Wang, QR, Yao, XQ, Wen, G, et al. (2011) Apigenin suppresses the growth of colorectal cancer xenografts via phosphorylation and up-regulated FADD expression. Oncol Lett 2, 4347.CrossRefGoogle ScholarPubMed
Sen, K, Banerjee, S & Mandal, M (2019) Dual drug loaded liposome bearing apigenin and 5-fluorouracil for synergistic therapeutic efficacy in colorectal cancer. Colloids Surf B Biointerfaces 180, 922.CrossRefGoogle ScholarPubMed
Tong, J, Shen, Y, Zhang, Z, et al. (2019) Apigenin inhibits epithelial-mesenchymal transition of human colon cancer cells through NF-KB/snail signaling pathway. Biosci Rep 39, BSR20190452.CrossRefGoogle ScholarPubMed
Chen, X, Xu, H, Yu, X, et al. (2019) Apigenin inhibits in vitro and in vivo tumorigenesis in cisplatin-resistant colon cancer cells by inducing autophagy, programmed cell death and targeting m-TOR/PI3K/Akt signalling pathway. J Buon 24, 488493.Google ScholarPubMed
Maeda, Y, Takahashi, H, Nakai, N, et al. (2018) Apigenin induces apoptosis by suppressing Bcl-Xl and Mcl-1 simultaneously via signal transducer and activator of transcription 3 signaling in colon cancer. Int J Oncol 52, 16611673.Google ScholarPubMed
Shan, S, Shi, J, Yang, P, et al. (2017) Apigenin restrains colon cancer cell proliferation via targeted blocking of pyruvate kinase M2-Dependent glycolysis. J Agric Food Chem 65, 81368144.CrossRefGoogle ScholarPubMed
Bian, S, Wan, H, Liao, X, et al. (2020) Inhibitory effects of apigenin on tumor carcinogenesis by altering the gut microbiota. Mediators Inflamm 2020, 7141970.CrossRefGoogle ScholarPubMed
Gazola, AC, Costa, GM, Castellanos, L, et al. (2015) Involvement of GABAergic pathway in the sedative activity of apigenin, the main flavonoid from Passiflora quadrangularis pericarp. Rev Bras Farmacogn 25, 158163.CrossRefGoogle Scholar
Zuo, Q, Wu, R, Xiao, X, et al. (2018) The dietary flavone luteolin epigenetically activates the nrf2 pathway and blocks cell transformation in human colorectal cancer HCT116 Cells. J Cell Biochem 119, 95739582.CrossRefGoogle ScholarPubMed
Kang, KA, Piao, MJ, Hyun, YJ, et al. (2019) Luteolin promotes apoptotic cell death via upregulation of Nrf2 expression by DNA demethylase and the interaction of Nrf2 with P53 in human colon cancer cells. Exp Mol Med 51, 114.Google ScholarPubMed
Potočnjak, I, Šimić, L, Gobin, I, et al. (2020) Antitumor activity of luteolin in human colon cancer SW620 cells is mediated by the ERK/FOXO3a signaling pathway. Toxicol 66, 104852.Google ScholarPubMed
Chen, Z, Zhang, B, Gao, F, et al. (2017) Modulation of G2/M cell cycle arrest and apoptosis by luteolin in human colon cancer cells and xenografts. Oncol Lett 15, 15591565.Google Scholar
Yao, Y, Rao, C, Zheng, G, et al. (2019) Luteolin suppresses colorectal cancer cell metastasis via regulation of the MiR-384/pleiotrophin axis. Oncol Rep 42, 131141.Google ScholarPubMed
Liu, Y, Lang, T, Jin, B, et al. (2017) Luteolin inhibits colorectal cancer cell epithelial-to-mesenchymal transition by suppressing CREB1 expression revealed by comparative proteomics study. J Proteomics 161, 110.CrossRefGoogle ScholarPubMed
Pandurangan, AK, Dharmalingam, P, Sadagopan, SKA, et al. (2014) Luteolin inhibits matrix metalloproteinase 9 and 2 in azoxymethane-induced colon carcinogenesis. Hum Exp Toxicol 33, 11761185.CrossRefGoogle ScholarPubMed
Visioli, F, Galli, C, Bornet, F, et al. (2000) Olive oil phenolics are dose-dependently absorbed in humans. FEBS Lett 468, 159160.CrossRefGoogle ScholarPubMed
Vissers, MN, Zock, PL, Roodenburg, AJC, et al. (2002) Olive oil phenols are absorbed in humans. J Nutr 132, 409417.CrossRefGoogle ScholarPubMed
Alemán-Jiménez, C, Domínguez-Perles, R, Medina, S, et al. (2021) Pharmacokinetics and bioavailability of hydroxytyrosol are dependent on the food matrix in humans. Eur J Nutr 60, 905915.CrossRefGoogle ScholarPubMed
Robles-Almazan, M, Pulido-Moran, M, Moreno-Fernandez, J, et al. (2018) Hydroxytyrosol: bioavailability, toxicity, and clinical applications. Food Res Int 105, 654667.CrossRefGoogle ScholarPubMed
Mateos, R, Goya, L & Bravo, L (2005) Metabolism of the olive oil phenols hydroxytyrosol, tyrosol, and hydroxytyrosyl acetate by human hepatoma HepG2 cells. J Agric Food Chem 53, 98979905.CrossRefGoogle ScholarPubMed
Rodríguez-Morató, J, Boronat, A, Kotronoulas, A, et al. (2016) metabolic disposition and biological significance of simple phenols of dietary origin: hydroxytyrosol and tyrosol. Drug Metab Rev 48, 218236.CrossRefGoogle ScholarPubMed
Covas, MI, de la Torre, K, Farré-Albaladejo, M, et al. (2006) Postprandial LDL phenolic content and LDL oxidation are modulated by olive oil phenolic compounds in humans. Free Radic Biol Med 40, 608616.CrossRefGoogle ScholarPubMed
de la Torre, R (2008) Bioavailability of olive oil phenolic compounds in humans. Inflammopharmacology 16, 245247.CrossRefGoogle ScholarPubMed
Pastor, A, Rodríguez-Morató, J, Olesti, E, et al. (2016) Analysis of free hydroxytyrosol in human plasma following the administration of olive oil. J Chromatogr A 1437, 183190.CrossRefGoogle ScholarPubMed
de Bock, M, Thorstensen, EB, Derraik, JGB, et al. (2013) Human absorption and metabolism of oleuropein and hydroxytyrosol ingested as olive (Olea Europaea L.) leaf extract. Mol Nutr Food Res 57, 20792085.CrossRefGoogle ScholarPubMed
Lozano-Castellón, J, López-Yerena, A, Rinaldi de Alvarenga, JF, et al. (2020) Health-promoting properties of oleocanthal and oleacein: two secoiridoids from extra-virgin olive oil. Crit Rev Food Sci Nutr 60, 25322548.CrossRefGoogle ScholarPubMed
Mehmood, A, Usman, M, Patil, P, et al. (2020) A review on management of cardiovascular diseases by olive polyphenols. Food Sci Nutr 8, 46394655.CrossRefGoogle ScholarPubMed
López-Yerena, A, Vallverdú-Queralt, A, Mols, R, et al. (2020) Absorption and intestinal metabolic profile of oleocanthal in rats. Pharmaceutics 12, 134.CrossRefGoogle ScholarPubMed
Hostetler, GL, Ralston, RA & Schwartz, SJ (2017) Flavones: food sources, bioavailability, metabolism, and bioactivity. Adv Nutr 8, 423435.CrossRefGoogle ScholarPubMed
Wang, M, Firrman, J, Liu, L, et al. (2019) A Review on flavonoid apigenin: dietary intake, ADME, antimicrobial effects, and interactions with human gut microbiota. Biomed Res Int 2019, 7010467.Google ScholarPubMed
Tang, D, Chen, K, Huang, L, et al. (2017) Pharmacokinetic properties and drug interactions of apigenin, a natural flavone. Expert Opin Drug Metab Toxicol 13, 323330.CrossRefGoogle ScholarPubMed
Gradolatto, A, Basly, JP, Berges, R, et al. (2005) Pharmacokinetics and metabolism of apigenin in female and male rats after a single oral administration. Drug Metab Dispos 33, 4954.CrossRefGoogle ScholarPubMed
Salehi, B, Venditti, A, Sharifi-Rad, M, et al. (2019) The therapeutic potential of apigenin. Int J Mol Sci 20, 1305.CrossRefGoogle ScholarPubMed
Ding, B, Chen, H, Wang, C, et al. (2013) Preparation and in vitro evaluation of apigenin loaded lipid nanocapsules. J Nanosci Nanotechnol 13, 65466552.CrossRefGoogle ScholarPubMed
Zhai, Y, Guo, S, Liu, C, et al. (2013) Preparation and in vitro evaluation of apigenin-loaded polymeric micelles. Colloids Surf A Physicochem Eng Asp 429, 2430.CrossRefGoogle Scholar
Shimoi, K, Okada, H, Furugori, M, et al. (1998) Intestinal absorption of luteolin and luteolin 7-O-β-glucoside in rats and humans. FEBS Lett 438, 220224.CrossRefGoogle ScholarPubMed
Simons, AL, Renouf, M, Murphy, PA, et al. (2010) greater apparent absorption of flavonoids is associated with lesser human fecal flavonoid disappearance rates. J Agric Food Chem 58, 141147.CrossRefGoogle ScholarPubMed
Bertelli, M, Kiani, AK, Paolacci, S, et al. (2020) Hydroxytyrosol: a natural compound with promising pharmacological activities. J Biotechnol 309, 2933.CrossRefGoogle ScholarPubMed
Auñon-Calles, D, Canut, L & Visioli, F (2013) Toxicological evaluation of pure hydroxytyrosol. Food Chem Toxicol 55, 498504.CrossRefGoogle ScholarPubMed
López-Huertas, E, Lozano-Sánchez, J & Segura-Carretero, A (2021) Olive oil varieties and ripening stages containing the antioxidants hydroxytyrosol and derivatives in compliance with EFSA health claim. Food Chem 342, 128291.CrossRefGoogle ScholarPubMed
Czeczot, H, Tudek, B, Kusztelak, J, et al. (1990) Isolation and studies of the mutagenic activity in the Ames test of flavonoids naturally occurring in medical herbs. Mutat Res 240, 209216.CrossRefGoogle ScholarPubMed
Romani, A, Ieri, F, Urciuoli, S, et al. (2019) Health effects of phenolic compounds found in extra-virgin olive oil, by-products, and leaf of Olea Europaea L. Nutrients 11, 1776.CrossRefGoogle ScholarPubMed
Ashrafizadeh, M, Bakhoda, MR, Bahmanpour, Z, et al. (2020) Apigenin as tumor suppressor in cancers: biotherapeutic activity, nanodelivery, and mechanisms with emphasis on pancreatic cancer. Front Chem 8, 829.CrossRefGoogle ScholarPubMed
Kouka, P, Tekos, F, Papoutsaki, Z, et al. Olive oil with high polyphenolic content induces both beneficial and harmful alterations on rat redox status depending on the tissue. Toxicol Rep 7, 421432.CrossRefGoogle Scholar
Luisi, M, Lucarini, L, Biffi, B, et al. (2019) Effect of Mediterranean Diet enriched in high quality extra virgin olive oil on oxidative stress, inflammation and gut microbiota in obese and normal weight adult subjects. Front Pharmacol 10, 1366.CrossRefGoogle ScholarPubMed
Millman, JF, Okamoto, S, Teruya, T, et al. (2021) Extra-virgin olive oil and the gut-brain axis: influence on gut microbiota, mucosal immunity, and cardiometabolic and cognitive health. Nutr Rev nuaa148. Online ahead of print. https://doi.org/10.1093/nutrit/nuaa148 Google ScholarPubMed
Zhang, YJ, Li, S, Gan, RY, et al. (2015) Impacts of gut bacteria on human health and diseases. Int J Mol Sci 16, 74937519.CrossRefGoogle ScholarPubMed
Iebba, V, Totino, V, Gagliardi, A, et al. (2016) Eubiosis and dysbiosis: the two sides of the microbiota. New Microbiol 39, 112.Google ScholarPubMed
Schippa, S & Conte, MP (2014) Dysbiotic events in gut microbiota: impact on human health. Nutrients 6, 57865805.CrossRefGoogle ScholarPubMed
Nagao-Kitamoto, H, Kitamoto, S, Kuffa, P, et al. (2016) Pathogenic role of the gut microbiota in gastrointestinal diseases. Intest Res 14, 127138.CrossRefGoogle ScholarPubMed
Wu, N, Yang, X, Zhang, R, et al. (2013) Dysbiosis signature of fecal microbiota in colorectal cancer patients. Microb Ecol 66, 462470.CrossRefGoogle ScholarPubMed
Sheflin, AM, Whitney, AK & Weir, TL (2014) Cancer-promoting effects of microbial dysbiosis. Curr Oncol Rep 16, 406.CrossRefGoogle ScholarPubMed
Figure 0

Fig. 1. Schematic representation of key molecular events that drive colorectal carcinoma. APC mutation acts as the stepping stone in the process of transforming normal colorectal epithelium to adenoma, whereas the adenoma–carcinoma sequential progression is supported by alteration in three crucial events: CIN, microsatellite instability and CpG island methylator phenotype. Once the oncogenesis initiated, further accumulation of genetic changes by mutations of regulatory genes, such as DNA repair genes drive the progression. Finally, modifications of the genes related to epithelial–mesenchymal transition, basement membrane disruption, cell motility and angiogenesis contribute to metastasis.

Figure 1

Fig. 2. Schematic depiction of colonic epithelium structure and components. The colonic crypt can be subdivided into three zones depending on the presence of different types of cells: stem cell zone, transit-amplifying (TA) cell zone and the differentiated zone. Reg4+ (regenerating islet-derived family member 4) deep crypt secretory cells (DCS) reside at the bottom of the colonic crypt and provide necessary support to the Lgr5+ (leucine-rich repeat-containing G-protein coupled receptor 5) stem cells, similar to the Paneth cells present in the small intestines. Quiescent stem cells or label-retaining cells (LRC) are located at the +4 position of the stem cell zone. TA cells are rapidly dividing and eventually differentiate into functional cells. The presence of Wnt, Notch, BMP, BMP antagonists, and Hedgehog and their respective concentration gradient in different zones is indicated by upward and downward triangles.

Figure 2

Fig. 3. Structure of major phenolic compounds present in olive oil. Major phenolic compounds present in olive oil and their structure are shown here, which include simple phenol like hydroxytyrosol, secoiridoid like oleuropein and flavone like apigenin.

Figure 3

Table 1. Summary of studies involving different forms of olive oil as intervention in CRC

Figure 4

Table 2. Summary of studies employed hydroxytyrosol as intervention in CRC

Figure 5

Table 3. Summary of studies involving oleuropein as intervention in CRC

Figure 6

Table 4. Summary of oleocanthal mediated anti-CRC activities

Figure 7

Table 5. Summary of studies utilised apigenin and luteolin as intervention

Figure 8

Fig. 4. Summary of the key anti-colorectal cancer activities of olive oil and its phenolic compounds. The major activities of each phenolic compound are shown here. For each activity, molecular pathways/signalling molecules targeted by olive oil phenolic compounds demonstrated here by indicating upward arrowhead (↑ = up-regulating), downward arrowhead (↓ = down-regulating) and cross sign (× = blocking).