Next Article in Journal
Ponatinib and other CML Tyrosine Kinase Inhibitors in Thrombosis
Next Article in Special Issue
Spermidine Attenuates Oxidative Stress-Induced Apoptosis via Blocking Ca2+ Overload in Retinal Pigment Epithelial Cells Independently of ROS
Previous Article in Journal
The Antihypertensive Effect of Quercetin in Young Spontaneously Hypertensive Rats; Role of Arachidonic Acid Metabolism
Previous Article in Special Issue
Enhanced Yield of Bioactivities from Onion (Allium cepa L.) Skin and Their Antioxidant and Anti-α-Amylase Activities
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Chemopreventive Effect of Dietary Anthocyanins against Gastrointestinal Cancers: A Review of Recent Advances and Perspectives

by
K.V. Surangi Dharmawansa
1,
David W. Hoskin
2,3 and
H. P. Vasantha Rupasinghe
1,2,*
1
Department of Plant, Food, and Environmental Sciences, Faculty of Agriculture, Dalhousie University, Truro, NS B2N 5E3, Canada
2
Department of Pathology, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada
3
Department of Microbiology and Immunology, and Department of Surgery, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2020, 21(18), 6555; https://doi.org/10.3390/ijms21186555
Submission received: 12 August 2020 / Revised: 28 August 2020 / Accepted: 2 September 2020 / Published: 8 September 2020

Abstract

:
Anthocyanins are a group of dietary polyphenols, abundant mainly in fruits and their products. Dietary interventions of anthocyanins are being studied extensively related to the prevention of gastrointestinal (GI) cancer, among many other chronic disorders. This review summarizes the hereditary and non-hereditary characteristics of GI cancers, chemistry, and bioavailability of anthocyanins, and the most recent findings of anthocyanin in GI cancer prevention through modulating cellular signaling pathways. GI cancer-preventive attributes of anthocyanins are primarily due to their antioxidative, anti-inflammatory, and anti-proliferative properties, and their ability to regulate gene expression and metabolic pathways, as well as induce the apoptosis of cancer cells.

Graphical Abstract

1. Introduction

The term “cancer” is described as a sequence of complex processes involving the accumulation of altered genetic material in cells, unlimited cell proliferation, and the formation of malignant tumors, cells from which can then migrate to and invade distant sites of the body [1]. According to the World Health Organization (WHO), cancer is responsible for one in six deaths worldwide, causing about 30% of all premature deaths in adults aged 30–69 years. Despite improvements in therapeutic strategies and screening programs, in 2018, 18.1 million people had cancer worldwide and WHO forecasts doubling of cancer cases by 2040 [2]. Among all types of cancer, gastrointestinal (GI) cancers, which include cancers of the colon and rectum (colorectal), esophagus and stomach (gastroesophageal), liver, gallbladder, pancreas, small intestine, appendix, and anus, collectively represent one of the greatest public health problems in the world, accounting for more than 35% of cancer-related deaths [2]. GI cancers have common risk factors; however, GI cancers are different in etiological, epidemiological, and clinical management profiles [3]. Colorectal cancer has become the third most common cancer in the world, and all other GI cancers still add a burden to the global incidence of cancer due to the limited number of biomarkers available for cancer screening, diagnosis, and prognosis [4]. Each year, approximately 4.1 million people are diagnosed with GI cancers, and about 3 million cancer-related deaths are due to late detection of the disease [5].
The carcinogenesis of GI cancers is linked to several molecular abnormalities, which include and are not limited to epigenetic modifications such as DNA methylation [6], and inactivation of tumor suppressor genes, i.e., TP53, which results in irregular cell cycle replication processes [7], and activation of oncogenes and various telomerases [8]. Moreover, the imbalance between cell proliferation and apoptosis leads to the pathogenesis of GI cancers [7]. Internal factors, such as chronic inflammation, which is influenced by the intestinal microbial imbalance, promote the malignant transformation of healthy cells into cancerous cells [9]. However, a third of all cancers are due to unsatisfactory lifestyles and dietary practices [10]. Alcohol consumption and exposure to environmental pollutants promote GI cancer, while regular consumption of plant-based foods containing dietary fiber reduces the risk of GI cancer [11].
Flavonoids, a group of C15 polyphenols, have been the subject of extensive research for their potential in chemoprevention and chemotherapy [12]. Flavonoids are abundant in berries (blueberry, raspberry, haskap berry, blackberry, and elderberry), vegetables (broccoli, kale, lettuce, and celery), tea, coffee, and red wine [13,14,15]. Flavonoids have gained attention as anticancer agents due to their structural diversity, relative abundance, limited toxicity, and cancer-preventive efficacy [16]. Among the major sub-groups of flavonoids, anthocyanins are widely found in plant-based food, including more than 1000 water-soluble compounds responsible for the vivid blue, purple, and red nuances of fruits, vegetables, colored grains, and beans [16,17,18].
Cancer chemoprevention refers to the use of agents for the inhibition, delay, or reversal of carcinogenesis before the local invasion of tissues occurs [19]. The results of epidemiological studies suggest that anthocyanins inhibit the initiation and progression of GI cancers [20]. The underlying molecular mechanism of anthocyanins and their colonic microorganism-generated metabolites in chemoprevention has been attributed to their antioxidant potential, anti-inflammatory activity, anti-proliferation effect, induction of apoptosis and suppression of matrix metalloproteinases in cancer cells [21]. In addition, anthocyanins are capable of stimulating the expression of tumor suppressor genes and downregulating pro-oncogenic signals [22]. The present review summarizes the latest findings on the potential of anthocyanin in the prevention of GI cancers, as well as their underlying molecular mechanisms of action, as evidenced by in vitro, in vivo, pre-clinical, and clinical studies.

2. GI Carcinogenesis

GI cancers account for more than 20% of cancers worldwide. Even countries with a high standard of living, education, and health experience a high incidence of GI cancers and associated morbidity and mortality [23]. GI cancer is a heterogeneous cancer that tends to occur in the more common sporadic forms rather than the rare inherited forms. The process of initiation and formation of neoplastic cells in the GI tract can be classified into four main mechanisms: (i) inherited transmission of mutations; (ii) exposure to different carcinogens; (iii) chronic inflammatory conditions/microbial dysbiosis; and (iv) sporadic mutations and epigenetic changes [24].

2.1. Hereditary GI Cancers

Hereditary GI cancers represent a phenotypically diverse group of diseases involving malignant tumors of the digestive tract, extra-GI cancers, and benign abnormalities characterized by inherited genetic mutations transmitted from parent to child. However, no more than 3%–5% of GI cancers have shown a clear hereditary basis [25]. The esophagus, stomach, colon, small intestine, and pancreas have been identified as the organs most likely to inherit germline mutations [24]. The best known inherited malignant tumors are associated with the GI tract, representing monogenic hereditary diseases that result from mutations in a single gene [26]. Despite the specific differences in the genes involved, inherited GI cancers share a common set of characteristics: (i) the majority of GI cancers are detectable in the early stages of life; (ii) these cancers follow an autosomal dominant inheritance mechanism in which the neoplasm occurs in 1st degree relatives; and (iii) the formation of multiple tumors [26,27]. In the hereditary form of GI cancers, the first genetic mutation in one of the alleles of a predisposition gene is acquired at the time of conception, and the somatic mutation of the second allele is then acquired via environmental insult, lifestyle practices or other exogenous factors (Figure 1). Once the two alleles of a specific predisposition gene are mutated, gene function is completely inactivated, leading to carcinogenesis. Compared to the sporadic form of GI cancer, which requires two somatic events during the inactivation of the predisposition gene, hereditary cancers present a higher risk because they need only one somatic mutation event, which explains the early onset of hereditary cancers [28]. In parallel with the advancement of DNA technologies, the genetic mutations responsible for hereditary GI cancers have been widely documented (Table 1). These hereditary GI cancers include Cowden syndrome, MUTYH-associated polyposis, hereditary pancreatic cancer, Lynch syndrome, Peutz-Jeghers syndrome, familial adenomatous polyposis (FAP), attenuated FAP, serrated polyposis syndrome, and hereditary gastric cancer. Cancer-causing mutations can be initiated in three main classes of predisposition genes, oncogenes, tumor suppressor genes, and DNA repair genes, which are involved in establishing genetic stability.

2.2. Non-Hereditary GI Cancers

Accumulations of sporadic mutations can occur due to factors such as exposure to carcinogens [24], a westernized diet [42,43], diets rich in salt [44,45], obesity [46,47], chronic alcohol consumption [48,49], and chronic inflammation [50]. The relationships between carcinogens, diet, inflammation, and GI cancers are multiple and complex. Exposure to carcinogens can initiate cancer development via somatic mutations that include point mutations, deletions, additions, and modified methylation of DNA [51]. There are several cellular mechanisms to protect DNA from carcinogen-induced mutations and to identify and correct these mutations before they give rise to malignancy. In spite of these protective mechanisms, the GI tract is continuously exposed to chemical and biological carcinogens, often due to diets that act as carriers of preformed carcinogens [24]. Among known carcinogens, tobacco smoke hydrocarbons are one of the most potent, being comprised of more than 60 mutagens and cancer-causing chemicals directly linked to esophageal [51], pancreatic [52], and gastric cancers [53,54]. In addition, exposure to airborne occupational carcinogens such as cement dust, quartz dust, and diesel exhaust fumes increases the risk of gastric cancer [55]. Nitrosamines, which are produced from the chemical reaction between nitrates or, in reduced form, nitrites with amines present in meat products during the meat preservation process, are another group of potent carcinogens associated with the increased risk of malignancy in the liver and GI tract due to DNA alkylation and DNA adduct formation [56]. Among biological carcinogens, aflatoxin B1 is one of the most influential hepatocarcinogens produced by the Aspergillus flavis fungi. Due to its lipophilic nature, aflatoxin B1 is readily absorbed from the GI tract. Aflatoxin B1, upon its metabolism by cytochrome P450 in the liver, induces irreversible mutations in the p53 gene of hepatocytes [57,58]. Fumonisin B1 is another mycotoxin that can cause hepatic and esophageal cancers [59]. Fumonisin B1 acts in part by upregulating the production of inflammatory cytokines by gastric and colon epithelial cells [60].
GI tract carcinogenesis is attributed to chronic inflammatory conditions that occur due to microbial, viral, or disease conditions such as inflammatory bowel disease (IBD). Regarding microbial inflammation, Helicobacter pylori infection is well documented as a trigger for GI cancers. Exposure to H. pylori initiates active chronic gastritis by increasing the infiltration of inflammatory cells, which leads to the formation of intestinal adenocarcinoma and other malignant tumors of the GI tract [61]. In addition, hepatitis B and C virus infection, gastroesophageal reflux, enzyme damage, autoimmune diseases such as ulcerative colitis, and systemic stress conditions are responsible for chronic GI inflammation [24]. Clinical studies show that patients with IBD have a significantly higher risk of developing colorectal cancer, especially 8 to 10 years after the diagnosis of IBD [62]. The mechanisms underlining the link between inflammation and GI cancers are varied and include the production of high levels of reactive oxygen species (ROS) and reactive nitrogen intermediates (RNI) [63]. The macrophages that dominate the chronic inflammatory microenvironment produce increased levels of ROS and RNI, which in turn interact with the DNA of proliferating epithelial cells and generate permanent genetic mutations leading to the malignant transformation. Excessive ROS/RNI production during the process of oxidative metabolism has been reported to promote the synthesis and secretion of inflammation-promoting cytokines such as tumor necrosis factor (TNF)-α, interferon-gamma (IFN-γ), and interleukin (IL)-6 [64,65]. Moreover, other inflammatory mediators such as chemokines, growth factors, and eicosanoids in tumor microenvironments contribute to inflammation-triggered tumor progression and metastasis via modulating the immune response, inhibiting apoptosis, inducing cell proliferation, and promoting the accumulation of oncogenic mutations [66].
Diet, personal lifestyle, and the environment are all linked to the development of GI cancers. Chronic excessive caloric intake and physical inactivity leading to overweight and obesity-derived metabolic dysfunction are essential risk factors of GI carcinogenesis [46,47]. Energy imbalance causes alterations in glycemic control, insulin signaling, and upregulation of adipose tissue-derived inflammatory pathways that prolong carcinogenesis-promoting conditions [67]. Chronic alcohol consumption also increases susceptibility to GI cancers [48]. Acetaldehyde, the primary metabolite of alcohol, has recently been targeted for its involvement in ethanol-linked oxidative stress and the inhibition of DNA methylation by interfering with the metabolism of B vitamins, reducing the activity of methionine synthase and glutathione levels [68], and disrupting retinoid metabolism [48]. Depletion of systemic and tissue-specific retinoic acid levels are associated with possible malignant transformation; thus, chronic alcohol consumption reduces hepatic vitamin A and retinoic acid levels, which are strongly related to the later development of hepatocellular carcinoma (HCC) via decreasing mitogen-activated protein kinase (MAPK) and increasing levels of phosphorylated c-Jun N-terminal kinases (JNKs) [48]. Moreover, in vivo evidence reveals that high-fat and high-salt diets alter the permeability and growth of the colonic mucus layer, which in turn, leads to intestinal microbial dysbiosis that is linked with an increased incidence of GI cancer [69]. Colonic microbial imbalance resulting from prolonged consumption of westernized diets enhances the breakdown and metabolism of specific glycans in the mucus layer, leading to GI carcinogenesis [70].
Among the modifiable risk factors of GI tract cancers, diet has been identified as one of the most significant in cancer control. Extensive studies have shown the chemopreventive effect of dietary polyphenols [71,72]. These natural antioxidants can prevent the onset of GI tract cancers, thus enhancing human well-being [73]. Polyphenols are potent scavengers of ROS and other free radicals that cause DNA damage and neoplastic transformation [74]. Chemopreventive effects of polyphenols extend to the prevention of pro-carcinogen activation, downregulation of inflammation, and inhibition of cell proliferation by interfering with the cell cycle activities of cancer cells [75]. Anthocyanins, a sub-class of flavonoids categorized under the group of polyphenols, have also become prominent dietary antioxidants in GI cancer prevention owing to their strong electron donor ability [76]. According to cohort studies, frequent consumption of fruits and vegetables of vivid blue, purple and violet colors, the richest sources of anthocyanins, have been associated with a reduction in the incidence of colorectal cancer [77], bladder cancer [78], and gastric cancer [79].

3. Chemistry, Dietary Sources, Bioavailability, and Toxicology of Anthocyanin

Anthocyanins, which are a glycosidic form of anthocyanidins, possess a basic structure of C6-C3-C6 composed of two aromatic rings (A and C) and one heterocyclic ring (B) [80] (Figure 2). Anthocyanins are differentiated on the basis of the number of hydroxyl groups, the number and type of sugar moieties, and the presence or absence of acyl groups [16]. Out of over 600 anthocyanins identified in nature, six main anthocyanin classes are well distributed in fruits and vegetables (Figure 2). Cyanidin-3-O-glucoside (C3G) is highly abundant among anthocyanins, and more than 90% of anthocyanins are conjugated with glucose [81]. Families of Vitaceae (grape), Rosaceae (cherry, plum, raspberry, strawberry, blackberry, apple, and peach), Solanaceae (tamarillo and eggplant), Saxifragaceae (red and black currant), Caprifoliaceae (haskap), Cruciferae (red cabbage) and Ericaceae (blueberry and cranberry) are primary sources of dietary anthocyanin [20,82]. Due to their anionic nature, once consumed, anthocyanins undergo pH and physiological temperature-dependent transformations that have a significant impact on their biological activities, improving their capacity to mediate cancer chemoprevention [73]. Despite the beneficial properties and relative abundance of anthocyanins, their effectiveness in the prevention of cancers depends on their bioavailability. Intact forms of anthocyanins that are absorbed from the stomach, as well as the intestine via an active transport mechanism, are then subject to hepatic Phase 2 metabolism. The resulting anthocyanin metabolites enter the systemic circulation. Unabsorbed anthocyanins reach the large intestine and undergo microbial biotransformation into decomposed products that contribute to cancer-chemoprevention [83]. Gastric digestion does not significantly affect anthocyanin composition; however, approximately 42–76% of total anthocyanins and 29% of their antioxidative activity are lost during passage through the intestines [84,85]. A 13C traceability study that utilized eight healthy male participants revealed 12% relative bioavailability of C3G after receiving a 500 mg oral dose of anthocyanin [86]. In contrast, a recent human intervention study showed that only 0.02% of ingested bilberry anthocyanin is detectable in plasma over 8 h after ingestion [87]. These controversial findings indicate that further investigations of bioavailability, absorption, and excretion of anthocyanins are warranted. The maximum plasma concentration is attained within 0.5–2 h after the consumption of anthocyanin-rich foods [83]. Around 20–25% of the ingested anthocyanin is absorbed by the gastric mucosa, although this varies according to the structure of the anthocyanin [86,87,88,89]. The majority of glycosidic forms, anthocyanin monoglucosides, and non-acylated compounds are well absorbed [90,91]. Glucose transporters are not involved in gastric absorption of anthocyanin; hence, absorption is facilitated by bilitranslocase, an organic anion membrane carrier [92]. Unabsorbed anthocyanin is then metabolized into glucuronidate, sulfate, or methyl derivatives in the small intestine; the greatest amount is absorbed in the jejunum and the lowest is absorbed by duodenal tissue [93]. Anthocyanins that pass down to the large intestine are subjected to spontaneous or microbial bioconversion [94]. In vitro studies prove that upon bacterial metabolism, cleavage of glycosidic linkage and breakdown of the anthocyanidin heterocycle is possible while producing 4-hydroxybenzoic acid, protocatechuic acid (PCA), gallic acid, vanillic acid, and syringic acid as the major microbial metabolites [95]. Incubation of a mixture of anthocyanins with fecal bacteria results in the formation of gallic, syringic, and p-coumaric acids [96]. The metabolism of C3G and cyanidin-3-O-rutinoside by rat gut microflora gives rise to protocatechuic, vanillic, p-coumaric acids, and 2,4,6-trihydroxybenzaldehyde. Gallic acid, syringic acid, and 2,4,6-trihydroxybenzaldehyde are the primary metabolites of delphinidin-3-O-rutinoside [97]. Therefore, microbial metabolism of anthocyanins may contribute to their pronounced chemopreventive properties, as the microbiome enhances anthocyanin metabolite concentrations [98].
Consumption of anthocyanins has been generally considered as safe in humans and anthocyanin consumption has been increased in line with educational level and degree of physical activity of populations [99]. As far as we are aware, there are no adverse health issues reported concerning anthocyanin in reported human intervention studies. Usually, the doses used in dietary supplementations of anthocyanin are higher than the regular dietary intakes.

4. Mechanisms of Anthocyanin-Mediated Chemoprevention of GI Cancers

The mechanisms by which anthocyanins prevent GI cancers are not well understood. However, anthocyanins have emerged as promising chemopreventive compounds for GI cancers, most likely because of their antioxidant, anti-inflammatory, anti-cell proliferative, and apoptosis-inducing properties [21]. A recent study demonstrates that anthocyanins reduce carcinogen-induced DNA damage in cultured human lung epithelial cells [100], pepsin-induced DNA damage in human airway epithelial cells [101], and benzo-[a,1]-pyrene dihydrodiol (DBP-diol)-induced DNA adducts and DBP-diol and DBP-diolepoxide (DBPDE)-induced mutagenesis in lacI rat oral fibroblast cells and human oral leukoplakia cells [102]. Extensive investigations have been performed to determine the molecular mechanisms underlying the chemopreventive properties of anthocyanins. The results indicate that anthocyanins inhibit several signaling pathways involved in DNA damage, cancer initiation, cancer cell proliferation, and tumor growth [20]. Potential molecular mechanisms of anthocyanin-mediated GI cancer prevention are summarized in Figure 3.

4.1. Downregulation of Pro-Inflammation and Oxidative Stress Associated with DNA Damage

4.1.1. Pro-Inflammation

Chronic inflammation is a prolonged immune response that contributes to the pathogenesis of GI cancers [103]. Under chronic inflammatory conditions, intestinal barrier function is impaired by the loss of the mucosal epithelial layer integrity layer due to decreased production and assembly of the TJ proteins and translocation of invasive microbial species and microbial products to the internal tissue environment [104]. In various systems, anthocyanins improve the intestinal TJ barrier integrity by promoting the expression of crucial barrier-forming TJ proteins such as occludin, claudin-5 and, zonnula occuldin-1 via upregulation of glucagon-like peptide (GLP)-2 intestinal hormone levels [105,106]. In addition, anthocyanins tend to improve barrier function by regulating TJ and epithelial cell permeability [107]. Anthocyanins also down-regulate the expression of major pro-inflammatory biomarkers such as TNF-α, IL-6, IL-1β, IFN-γ, prostaglandin E2 (PGE2), monocyte chemoattractant protein (MCP)-1, cyclooxygenase (COX) -2, and nuclear factor kappa B (NF-κB) [108,109,110,111]. For example, a combination of lycopene and anthocyanin inhibits expression of the cytokine IL-8, whereas, anthocyanin-rich wild blueberry extract reduces the activity of NF-κB in Caco-2 cells [111,112]. Anthocyanins extracted from red clover [113], and black rice [114], inhibit the translocation of NF-κB into the nucleus of lipopolysaccharide (LPS)-activated RAW264.7 macrophages. Furthermore, the production of nitric oxide (NO), expression of COX-2 and secretion of TNF-α and IL-6 were also diminished by black rice extracts [114]. Overexpression of the pro-inflammatory enzyme, inducible nitric oxide synthase (iNOS), is another general feature of epithelial tissue inflammation and carcinoma development [115]. In this regard, Peng et al. [116] report that the long term consumption of anthocyanin from Lycium ruthenicum Murray reduces inflammation of the colon by reducing the expression of iNos, Cox-2, TNF-α, IL-6, IL-1β, and IFN-γ mRNAs in C57BL/6 male mice. Additionally, cocoplum extract, which is rich in the anthocyanins delphinidin, cyanidin, petunidin, and peonidin, downregulates IL-1β, IL-6, and NF-κB expression in HT-29 colorectal adenocarcinoma cells while decreasing TNF-α-induced intracellular ROS-production [117]. Moreover, anthocyanins from various sources, for example, fruits of L. ruthenicum Murray [109], red raspberry [118], black rice [119], and strawberry [120], are able to attenuate dextran sulfate sodium (DSS)-induced gut inflammation in mouse models of IBD. Thus, suppression of inflammation by anthocyanins may protect against GI cancer occurrence or its progression.

4.1.2. Oxidative Stress Associated with DNA Damage

During chronic inflammation, excessive production of ROS and RNI leads to a disruption of redox homeostasis, producing oxidative stress. The redox homeostasis imbalance causes direct cellular damage by oxidation of macromolecules, including oxidative DNA damage resulting in DNA mutations [121]. Moreover, oxidative stress contributes to cancer progression by continuously creating DNA mutations in cancerous cell populations. Modulation of intracellular oxidative stress by scavenging the ROS/RNI is, therefore, beneficial in preventing cancer initiation and progression [122]. Interestingly, anthocyanins are potent inhibitors of redox dysregulation due to their ability to increase the oxygen radical-absorbing capacity of intestinal cells [123], stimulate phase II detoxification enzymes [124], reduce the formation of oxidative DNA adducts, and decrease lipid peroxidation [125]. For example, redox homeostasis in Caco-2 and HT-29 cells is restored by bilberry extract, which reduces intracellular ROS production and oxidative DNA damage, as well as increasing cellular glutathione-s-transferase (GSH) levels [126]. The antioxidant potential of anthocyanins has also been investigated in artificial alimentary tract models, including models of the stomach, small intestine, and colon. A digested form of anthocyanin extracted from purple carrot was effective in reducing oxidative DNA damage in colon mucosa and inhibited intracellular ROS while modulating the oxidative imbalance in rat liver induced by cadmium exposure [127,128]. In another study, digested products of wild raspberry, including primarily esculin, kaempferol hexoside, and pelargonidin hexoside, displayed a more pronounced effect against acrylamide-induced cytotoxicity in Caco-2 cells in comparison to the non-digested extract, which was related to reduced ROS generation and GSH depletion [129]. The antioxidative and anti-inflammatory effects of anthocyanins in intestinal ischemia-reperfusion (IIR) injury have also been reported [130,131]. Dietary supplementation with chokeberry and bilberry alone or together with probiotics inhibits oxidative stress and tissue injuries in mouse models of IIR [130]. However, anthocyanin supplements are unable to bring about a significant reduction in oxidative stress markers and pathology associated with DSS-induced colitis in Balb/c mice [132]. These occasionally inconsistent and somewhat variable results indicate that validation is required for the role of anthocyanins in modulating inflammation and oxidative stress. Nevertheless, anthocyanin-mediated prevention of hepatocarcinogenesis via activation of the Nrf-2/ARE pathway is well documented. For example, blackcurrant anthocyanins protect against diethylnitrosamine (DENA)-initiated hepatocarcinogenesis in rats by elevating the expression of protein and mRNA related to the Nrf-2 pathway [133]. These examples further support the notion that dietary anthocyanins play a significant role in the chemoprevention of colitis-associated GI cancer.

4.2. Inhibition of Cancer Cell Proliferation/Induction of Cell Cycle Arrest

The cell cycle consists of a programmed sequence of events beginning with cell size increase (G1 phase), DNA replication (S phase), cell preparation (G2 phase), and finally, cell division (M phase), which are coupled with G1-S, S, and G2-M checkpoints [134]. Under normal physiological conditions, cell cycle progression is governed by the activation/inactivation of cyclins and cyclin-dependent kinases (CDKs) [135]. The G1-S and S phases of the cell cycle are mainly regulated by CDK4-cyclin D, CDK6-cyclin D, CDK2-cyclin E, and CDK2-cyclin A sequential complexes while G2/M is controlled by CDK1-cyclin A/B [136]. Upon the segregation of DNA mutations, CDK inhibitors (CDKIs) such as p21 (cip1/waf1/cap20/sdi1/pic1), p27 (kip1), p57 (kip2) specific for CDK2 and CDK4 cyclin complexes and p16INK4, p15INK4B, p18INK4C and, p19INK4D specific for CDK4 and CDK6 cyclin complexes bind to and inactivate their respective CDK-cyclin complexes, thereby blocking cell cycle progression [137]. Hence, dysregulation of the cell cycle often leads to aberrant cell proliferation, which results in malignant cell growth during which loss of control of cell cycle checkpoints results in genetic instability [138]. Concerning GI cancers, anthocyanins prevent cancer by initiating cell cycle arrest at various stages and inducing anti-proliferative activity in a dose-dependent manner [115,139,140]. Anthocyanins are capable of upregulating CDKIs and downregulating cyclin proteins [141]. An anthocyanin-rich extract of chokeberry shows anti-proliferative effects resulting from cell cycle arrest at both G1/G0 and G2/M phases in HT-29 human colon cancer cells due to upregulation of p21, p27, and downregulation of cyclin A and B [142]. Consistently, anthocyanin metabolites, gallic acid, 3-O-methyl gallic acid, and 2,4,6-tri-hydro benzaldehyde, show the ability to block the proliferation of Caco-2 cells at G0/G1 phase [22]. Anthocyanins are also potent inducers of cell cycle blockage at the G2/M phase in oral cancer KB cells by down-regulating p53 methylation [143]. In addition to stimulating the expression of p21 and p27 CDKIs, an anthocyanin/anthocyanidin-rich extract from purple shoot tea reduced cyclin E and cyclin D1 expression in HT-29 colorectal carcinoma cells, resulting in cell cycle arrest at G0/G1 phase [144]. Similarly, delphinidin prevents HCT-116 cell proliferation by blocking the G2/M phase due to underexpression of cyclin B1 and overexpression of p53, a tumor suppressor protein, and p21WAF1/cip1 [145]. However, different anthocyanins have different effects related to cell cycle control. For example, chokeberry anthocyanins that consist mainly of cyanidin derivatives are more potent inhibitors of HT-29 cell proliferation than grape or bilberry anthocyanins, which are rich in delphinidin [146]. Similarly, malvidin and pelargonidin (100 to 200 µL/mL) effectively suppress stomach and colon cancer cell proliferation, which is not affected by cyanidin and delphinidin [147]. Both peonidin-3-glucoside and C3G interfere with CDK-1,2 and cyclin B1 expression in AGS-gastric adenocarcinoma and SKHep-1, Huh-7 hepatocellular carcinoma cells. However, the activity of cyclin E expression is only inhibited by peonidin-3-glucoside, whereas only C3G inhibits cyclin E1 expression [148].

4.3. Induction of Apoptosis

Cells with damaged or mutation-containing DNA are normally eliminated by a form of cell death known as apoptosis [149]. Two distinct but interacting pathways mediate apoptosis; the extrinsic (death receptor-mediated) pathway, which activates caspase-8, and the intrinsic (mitochondrial membrane-permeabilizing) pathway, which activates caspase 9. Caspases are aspartate-specific cysteine proteins that stimulate nuclear membrane degradation, chromatin condensation, DNA fragmentation, and the formation of apoptotic bodies [150]. However, cancerous cells often fail to undergo apoptosis and, therefore, survive to form a tumor. In some cases, cancer cells may resist apoptosis by increasing or decreasing expression of anti- or pro-apoptotic genes, respectively. Moreover, cancer cells may also prevent apoptosis by changing the functions of anti- or pro-apoptotic proteins through post-translational modifications, such as phosphorylation [151].
Anthocyanins can activate both extrinsic and intrinsic pathways of apoptosis. Mechanisms of action include upregulating the expression of pro-apoptotic proteins such as B-cell lymphoma-2-like protein 4 (Bax) while downregulating the expression of anti-apoptotic proteins such as B-cell lymphoma-2 (Bcl-2), X-linked inhibitor of apoptosis protein (XIAP), caspase-recruitment domains like apoptotic proteins (CIAP)-1,2 and survivin [151,152]. For example, anthocyanins reduce the expression of anti-apoptotic proteins, survivin, CIAP-2, and XIAP in HT-29 and HCT-116 human colon carcinoma cells [153]. Human hepatoma Hep3B cells treated with an anthocyanin-rich extract from meoru (Vitis coignetiae Pulliat) exhibited significantly reduced Bcl-2, XIAP, and CIAP 1,2 protein expression [154]. Furthermore, anthocyanins increase DNA fragmentation, as indicated by the number of cells in the sub-G1 fraction, in a dose-dependent manner, which is closely related to mitochondrial dysfunction. The mitochondrial pathway of apoptosis is characterized by a profound reduction in mitochondrial membrane potential (ΔΨm). The collapse of ΔΨm leads to the opening of mitochondrial permeability transition pores in the mitochondrial membrane, thus allowing the release of cytochrome C into the cytosol, which in turn triggers caspase-9 activation and the ensuing irreversible events of the apoptosis cascade [155]. Interestingly, anthocyanins induce ΔΨm loss in GI carcinoma cells [156]. In gastric cancer cells, malvidin promotes an increase in Bax/Bcl-2 ratio, caspase-3 activation, and p38 kinase expression while reducing the ΔΨm and inducing cell cycle blockage at the G0/G1 stage [157]. Similarly, Yun et al. [145] report the cleavage of poly(ADP)-ribose polymerase (PARP), activation of caspases-3, -8, and -9, reduction of Bcl-2, and increased Bax protein expression in HCT-116 human colon cancer cells treated with anthocyanins. Although changes in Bcl-2 mRNA expression are not certain in HT-29 cells treated with bilberry extract, the pro-apoptosis marker, Bax, was increased 1.3-fold in cloudberry and bilberry treated cells [158]. The degree of cell growth inhibition followed the sequence bilberry > black currant > cloudberry > lingonberry > raspberry > strawberry, emphasizing the effect of divergence in anthocyanin source on potential GI cancer prevention. This may be due to variations in the anthocyanin profile of these fruits.
Topoisomerase inhibitors are efficient inducers of apoptosis [159]. Topoisomerase I and II enzymes play a vital role in DNA replication, facilitating the unwinding of supercoiled DNA. Inhibition of topoisomerase activity, therefore, prevents DNA replication, leading to apoptosis [160]. Anthocyanins are naturally occurring topoisomerase inhibitors [161]. Topoisomerase relaxation activity is inhibited by anthocyanin from blackberry extract at a concentration > 50 µM in the colon tissue of male Wistar rats [162]. In a similar study, berry extract at > 50 µM suppressed the activity of topoisomerase I in HT-29 cells while diminishing the activity of topoisomerase II at concentrations ≥ 1 µM [163]. However, concentrations up to 50 µM failed to induce DNA strand breaks. In contrast, C3G-rich blackberry extract suppressed camptothecin (CPT)- or doxorubicin (DOX)-induced stabilization of the covalent DNA-topoisomerase intermediate in HT-29 colon carcinoma cells [164]. These results, taken together, suggest that anthocyanins induce apoptosis in GI carcinoma cells in a dose-dependent manner via activation of extrinsic and intrinsic pathways of apoptosis, as well as by interfering with topoisomerase activity.

4.4. Regulation of Microbial Dysbiosis

Accumulating evidence indicates both a negative and positive association between gut microbiota and GI cancers. Healthy gut bacteria or probiotics are involved in activating anti-tumor immunity and boosting the efficacy of immunotherapy, whereas harmful bacteria induce inflammation-driven DNA alterations [165]. Anthocyanins and gut microbiota exhibit a two-way interaction that impacts host physiology. There is a broad agreement that dietary anthocyanins are involved in the modulation of gut microbiota, increasing the ratio of healthy/unhealthy bacteria [104]. For example, oral administration of 5 g/kg body weight of black raspberry to the diet for six weeks resulted in an increase in the abundance of healthy microbial species such as Akkermansia and Disulfovibrio (known to have anti-inflammatory effects) in F-344 rats [166]. C57BL/6J mice with colitis that were fed malvidin-3-glucoside at a dose of 24 g/kg body weight also showed a decrease in the number of pathogenic bacteria such as Ruminococcus gnavus, thereby restoring the gut microbial balance [167]. In a recent study, oral gavage of malvidin-3-galactoside (40–80 mg/kg body weight) increased butyric-producing bacteria and reduced the abundance of pathogenic bacteria in C57BL/6J mice with liver carcinogenesis [168]. As pathogenic microbes are involved in intestinal inflammation, regulation of gut microbial composition by anthocyanin is directly linked with the reduction of inflammation, hence preventing the onset of GI carcinogenesis. However, as the evidence of anthocyanins in microbial modulation is limited, additional research should be carried out to fully elucidate these interactions.

5. Anti-GI Cancer Effect of Common Dietary Anthocyanins

Although there is regular global consumption of a wide source of dietary anthocyanins, the scope of this review is limited to the chemopreventive effects of anthocyanin-rich fruits and cereals against cancers of the GI tract (Table 2). Selected major GI-cancers will be discussed in relation to the effect of anthocyanins based on in vitro and pre-clinical studies. Epidemiological studies will be discussed separately.

5.1. Oral Cancer

Malignancies that arise on the lips, tongue, gingiva, mouth floor, parotid, and salivary glands are defined as squamous cell carcinomas (SCC) or oral cancers [200]. Chemoprevention with anthocyanins may be useful for oral carcinomas as clinicians and patients can directly monitor the premalignant lesions, and medications can be applied directly to the affected area. Anthocyanins have been incorporated into bio-adhesive gels for the purpose of oral cancer prevention by inhibiting the malignant transformation of dysplastic oral lesions [143]. Intraoral bioactivation of anthocyanin occurs through the activities of oral microflora and salivary enzymes via β-glucosidase activity [152]. The anti-cancer activity of anthocyanins towards SCC is based on several factors: (1) Individual variations in anthocyanins uptake and intraoral metabolism; (2) pH dependency of the mucoadhesive gel on the penetrability of the anthocyanins, and (3) sustainability of anthocyanins at the target site [201]. As an example, berry gels prepared using 5% and 10% w/w freeze-dried black raspberry powder are absorbed readily into human oral mucosa tissue within five minutes and show more significant penetrability at pH 6.5 [202]. Studies with human oral epidermal KB and SCC131 cells show that anthocyanins induce significant apoptosis and cell cycle arrest at G2/M and G1/S phases, respectively [143]. Anthocyanins have also been shown to suppress the metastasis of human tongue epithelial CAL 27 cells and oral SCC cells [170,171]. In contrast, anthocyanin-rich cranberry extract is less able to suppress the growth of SCC of tongue and KB cells compared to the positive control drug Adriamycin [169]. As there is limited evidence on the anti-cancer effects of anthocyanins on oral cell carcinoma, additional studies are needed to determine more precisely the chemopreventive effects of these natural source compounds.

5.2. Esophageal Cancer

Due to its aggressive nature and poor survivability, esophageal cancer is the 10th most common malignancy and the 8th leading cause of cancer-related deaths worldwide [203]. Alcohol, tobacco, hot beverage, and red meat consumption are implicated as risk factors for the two prominent types of esophageal cancers, esophageal squamous cell carcinoma (ESCC), and adenocarcinoma [174,204]. A recently published meta-analysis of epidemiological studies reveals a positive correlation between anthocyanin intake and reduced esophageal cancer risk [203]. Anthocyanins are capable of reducing markers of inflammation and angiogenesis and inhibiting the migration and proliferation of human esophageal microvascular endothelial cells isolated from donor-discarded esophagus [174]. Reduced COX-2 and iNOS expression is observed in RE-149DHD and RE-149 rat esophageal cancer cell lines treated with freeze-dried black raspberries [175]. Fisher 344 (F344) rats are used extensively to model esophageal cancer induced by the nitrosamine carcinogen, N-nitroso methylbenzylamine (NMBA) [177]. C3G and cyanidin-3-rutinoside are potent inhibitors of the initiation events of esophageal cancer due to their ability to impact the metabolic activation and detoxification of NMBA [205]. Moreover, F344 rats with a diet supplemented with lyophilized anthocyanins (5–10% g/kg) show reduced formation of NMBA-induced O6-methylguanine adducts in esophageal DNA, providing evidence that consumption of berries influences the metabolism of NMBA, leading to reduced DNA damage [206]. Bio-fractionate studies reveal that diets containing approximately the same quantity of anthocyanins, regardless of their source, have a similar anti-carcinogenesis effect on esophageal cancers [178]. In line with these observations, feeding of anthocyanins extracted from seven types of berries is capable of inhibiting the initiation and progression of NMBA-induced tumors in F344 rats [179]. Furthermore, diets supplemented with 6.1% black raspberry powder, an anthocyanin-rich fraction of black raspberries (0.8 mg/g), or 500 μg/mL PCA had similar effects on cytokines produced in the esophagus and circulating in the plasma of NMBA-treated rats; relative to the NMBA-only control, proinflammatory IL-1β expression was decreased while IL-10 and IL-12 expression increased [180]. However, a crude black raspberry-supplemented diet was more effective in reducing inflammation and NMBA-induced carcinogenesis in F344 rats than the anthocyanin metabolite, PCA [207], suggesting additive or synergistic effects by the components of the crude extract. Recently, synthetic analogs of anthocyanins have been produced and tested. Dracorhodin perchlorate (DP), a synthetic analog of the anthocyanin red pigment dracorhodin, exerts various pharmacological effects, including anti-cancer activity in human ESCC cells due to G2/M phase cell cycle arrest through upregulation of p21 and p27, and downregulation of cyclin B1 and Cdc2 [208]. Importantly, anthocyanins are superior to the combination of celecoxib, a selective COX-2 inhibitor, and S,S’-1,4-phenylene-bis(1,2-ethanediyl)bis-isothiourea (PBIT), a selective iNOS inhibitor, in suppressing carcinogen-induced ESCC in rats [209].

5.3. Gastric Cancer

Gastric cancer is a heterogeneous malignancy that is mostly induced by H. pylori infection and is ranked as the 4th highest cause of cancer-related deaths [210]. Evidence that anthocyanins are effective in gastric cancer prevention is limited. A recent meta-analysis of cohort and case-control studies revealed that there is no significant association between anthocyanin intake and gastric cancer risk, nor is there any dose-dependent relationship [211]. In contrast, a case-control study of 334 gastric cancer patients showed a positive correlation between consumption of anthocyanins and reduced incidence of gastric cancer; the positive effects were predominantly seen in women [212]. Interestingly, anthocyanins are potent inhibitors of the biogenesis of H. pylori virulence proteins [213], suggesting a possible suppressive effect on H. pylori infections. Furthermore, anthocyanins extracted from black soybean inhibited H. pylori-induced inflammation in gastric cells (AGS) while reducing ROS, iNOS, and COX-2 expression, as well as proinflammatory IL-8 production [181]. Similarly, mulberry anthocyanins suppress the proliferation of SGC-7901 gastric cancer cells and upregulate their expression of caspase-8 and beclin-1, as well as increasing the Bax/Bcl-2 ratio [214]. However, anthocyanidins are more effective than anthocyanins in reducing in vitro growth of gastric cancer cells. For example, malvidin induces apoptosis of AGS cells by causing G0/G1 phase cell cycle arrest more effectively than its glycosidic form [157]. Anthocyanins also enhance the anti-cancer effects of chemotherapeutic drugs. In this regard, Lu et al. [215] have demonstrated an additive anti-cancer effect of anthocyanins in combination with cisplatin. Nevertheless, evidence of the role of anthocyanins in gastric cancer prevention is limited, indicating the need for further investigation.

5.4. Liver Cancer

Liver cancers are primarily comprised of hepatocellular carcinoma (HCC) and cholangiocarcinomas mixed liver carcinoma, of which HCC is the most common [216]. The major risk factors for HCC are chronic hepatitis B and C virus infections, cirrhosis, and metabolic liver disease [217]. Dietary interventions for the prevention of hepatic carcinogenesis have been studied for decades, and a possible role of anthocyanins in liver cancer prevention has been investigated. Anthocyanins from haskap berry (Lonicera caerulea) cv. Beilei are beneficial in adjusting the redox balance of human SMMC-7721 HHC cells in vitro, as well as promoting anti-tumor immune responses in mice bearing H22 hepatoma tumors [218]. These anthocyanins are also potent blockers of the cell cycle in the G2/M phase in hepatocellular carcinoma while at the same time decreasing the level of lipid peroxidation [218]. Malvidin-3-galactoside extracted from blueberry has anti-proliferative effects and induces apoptosis in human HepG2 cells via dose-dependent regulation of cyclin D1, cyclin B, cyclin E, caspase 3, cleaved caspase-3, Bax, and p38 MAPK expression [182]. The anti-invasive properties of anthocyanin exerted on human hepatoma Hep3B cells are the result of downregulating the expression of matrix metalloproteinase (MMP)-2 and MMP-9 [183], suggesting possible anti-metastatic activity. In tert-butyl hydroperoxide (TBHP)-treated human hepatoma cells, pre-treatment with delphinidin, cyanidin, and their glycoside and rutinoside derivatives attenuated DNA single-strand break formation, lipid peroxidation, and redox state alterations [184]. The anti-proliferative effects of anthocyanidins on hepatic carcinoma cells is more pronounced than that of anthocyanins. For example, cyanidin-3-rutinoside showed a prominent inhibitory effect on the growth of HepG2 cells that was not equaled by delphinidin and cyanidin [187]. In a diethylnitrosamine-induced hepatic carcinogenesis rat model, an anthocyanins-rich black currant extract exerted anti-inflammatory effects by increasing the hepatic expression of heat shock proteins and COX-2 in a dose-dependent manner [219]. However, in an aflatoxin-induced hepatic carcinogenesis model, anthocyanins from purple rice bran failed to affect micronucleus formation or xenobiotic-metabolizing enzymes in rat liver [220]. Anthocyanins from different sources may, therefore, not be equally effective against liver cancers.

5.5. Colorectal Cancer

Colorectal cancer (CRC) is characterized by the formation of polyps on the inner lining of the colon or rectum and is the 3rd most common cause of cancer-related deaths [221]. The five-year survivability of CRC in the United States is 64%. However, due to a lack of screening programs in many countries, only about 39% of colorectal cancers are diagnosed at an early stage, leading the majority being detected at a later stage, probably after metastasis has occurred [222]. Since about 80% of CRCs have a sporadic origin, it may be possible that adopting beneficial dietary and lifestyle practices could prevent CRC [223]. A recent meta-analysis of seven different studies revealed an inverse association between total anthocyanin consumption and CRC risk, although a dose-response relationship was not found [76]. Similarly, a recent systemic review elaborated on the positive linkage between the anthocyanin intake and reduced CRC risk via interference with CRC cell signaling and proliferation, as well as the ability to induce apoptosis by effects on several molecular pathways [224]. The protective effects of anthocyanins, crude berry extracts, and fruits with vivid purple and blue shades upon CRC have been well documented by in vitro and in vivo studies of colon cancer and inflammation models [140]. In many cases, anthocyanins have been administered in pure forms or as part of the whole fruit following processing by freeze-drying. Many of the chemopreventive properties observed seem to occur through inhibition of signaling pathways known to be important in the pathogenesis of CRC. For example, treatment of Colo 320DM cells with purple-shoot tea extracts resulted in reduced cell proliferation due to the blockade of cell cycle progression during the G0/G1 phase, as well as the induction of apoptotic death [144]. Aberrant expression of micro RNA, a class of small, endogenous, non-coding, single-stranded RNAs that bind to the 3′-untranslated region (3′-UTR) complementary sequences of their target mRNA, plays a critical role in the initiation, promotion, and progression of CRC [225]. Numerous studies have shown that anthocyanins prevent the development of CRC by improving miRNA regulation [226,227]. Exposure to black raspberry anthocyanins results in the overexpression of miR-24-1-5P in the colon tissue of carcinogen-treated mice, leading to significant suppression of β-catenin that in turn reduced CRC cell proliferation and migration, and enhanced survival [226]. In another study, black raspberry anthocyanins decreased miR-483-3p expression, which is oncogenic in a mouse model of CRC [227]. Colon-available raspberry extract (an extract that mimics the composition present in the colon) has been used to assess its chemopreventive properties in cultures of Caco-2, HT-29 and, HT 115 CRC cells [228]. Once consumed, anthocyanins are gradually digested during passage through the GI tract; thus, the composition of extracts that are available at the colon do not always mimic the composition of the original extract. However, a colon-available extract of raspberry anthocyanin, characterized by increasing amounts of polyphenols and polyphenol breakdown products but less anthocyanin than in the original, was potent in reducing H2O2-induced DNA damage in HT-29 cells and the proliferation of HT-115 CRC cells but did not affect the membrane integrity of Caco-2 cells [228]. Cancer stem cells are responsible for the initiation and progression of colorectal tumors [229]. It is, therefore, important to note that anthocyanin-containing baked purple-fleshed potato extracts suppressed the proliferation of colon cancer stem cells and increased their death by apoptosis in a p53-independent manner [230]. Furthermore, anthocyanins were found to reduce the levels of the Wnt pathway effector β-catenin, a critical regulator of cancer stem cell proliferation and epithelial-to-mesenchymal transition. Topoisomerase I and II activity in HT-29 CRC cells is also preferentially diminished in the presence of berry anthocyanin via its action as a topoisomerase-inhibiting catalyst [163]. Anthocyanins also modulate TJ proteins; anthocyanin from Vitis coignetiae Pulliat, a Korean fruit, increases the transepithelial electrical resistance of HCT 116 cells and suppresses MMP-2 and MMP-9 expression in a dose-dependent manner [231]. These findings indicate that anthocyanins may be able to maintain the integrity of epithelial barriers.
In a bio-fractionate study, anthocyanins exhibited greater anti-proliferative activity (>50%) in HT-29 and Caco-2 cell cultures than non-anthocyanin polyphenols such as flavonol, tannin, and phenolic acid fractions [189]. Moreover, structure-function relationships of anthocyanins from various sources reveal that non-acylated monoglycosylated form of anthocyanin is a more potent inhibitor of HT-29 CRC cell proliferation [232]. The varying compositions and degrees of growth inhibition suggest that the chemical structure of anthocyanins may play an essential role in their cell growth inhibitory activity. In this regard, there is a distinguishable difference between the anti-proliferative activity of anthocyanins extracted from leaf versus those extracted from the tuber of purple sweet potato in cultures of HCT-116 cells [233]. The leaf contains more cyanidin than the roots. The glycosylated form of cyanidin also suppresses the growth of tumor xenografts by targeting T-LAK-cell originated protein kinase, which plays a role in cell cycle regulation and mitotic progression [234]. Compared to delphinidin-3-O-glucoside, C3G is better able to activate the immune response in the tumor microenvironment by inhibiting the action of immune cell checkpoints [235]. However, orally administered C3G did not protect against DNA damage in a vitamin E-deficient rat model, although C3G did protect against DNA damage in human colonocytes, decreasing DNA strand breakage by 39% [236].
A significant amount of data derived from in vivo work demonstrates a potential set of benefits from dietary anthocyanins in terms of CRC prevention. As chronic inflammation is an important event leading to colon cancer, a number of pre-clinical trials have investigated the relationship between anthocyanins and acute or chronic colitis with the DSS-induced colitis model being the most common (Table 3). For example, orally administered anthocyanin-rich blueberry extract attenuates the development of DSS-induced experimental colitis in mice by reducing the accumulation of myeloperoxidase and malondialdehyde in the colon and prostaglandin E2 levels in serum while increasing the levels of SOD and catalase compared to untreated mice with colitis [237]. In addition, a diet supplemented with red raspberries resulted in a reduction in the disease activity index, histological damage, and expression of inflammatory mediators while facilitating repair of the epithelium in animals with DSS-induced colitis [238]. Azoxymethane (AOM) is a potent colon carcinogen that is used with/without DSS to induce colitis-associated carcinogenesis or non-colitis-associated carcinogenesis, respectively, in rodents [239]. In AOM-induced carcinogenesis in F344 rats, a diet that was supplemented with lyophilized black raspberries resulted in a dose-dependent reduction in aberrant crypt foci multiplicity [190]. Dietary supplementation with lyophilized strawberries also exerts an anti-cancer effect against inflammation-mediated colon carcinogenesis in mice by reducing the expression of pro-inflammatory mediators, suppressing nitrosative stress, and decreasing phosphorylation of phosphatidylinositol 3-kinase, Akt, extracellular signal-regulated kinase and NF-κB [240]. In addition, a significant decrease in adenoma number was attributed to the consumption of anthocyanin-rich sweet potato by the APCMIN+/− mice [191]. In the same animal model, oral supplementation with freeze-dried black raspberries reduced the number and size of intestinal and colonic polyps [192]. The berry supplement also significantly reversed the production of 23 APC-regulated metabolites, including 13 colonic mucosa, eight liver, and two fecal metabolites that are involved in amino acid, glutathione, lipid, and nucleotide metabolism. These results suggest the metabolic modulatory effects of anthocyanins in APCMIN+/− mice may contribute to the suppression of CRC.

6. Epidemiological Studies

Only a limited number of clinical studies have investigated the effect of anthocyanins in GI cancer prevention; however, a positive relationship between anthocyanin intake and reduced risk of GI cancers has been revealed. The consumption of a wide range of anthocyanins and a reduction in the incidence of GI cancer malignancy is associated with the mechanisms involved in (i) improving the intestinal TJ barrier integrity via AMPK activation; (ii) down-regulating pro-inflammatory molecules; (iii) inhibiting redox dysregulation; (iv) inhibiting cell proliferation by initiating cell cycle arrest; and (v) activating apoptotic pathways. More specifically, black raspberry has been extensively studied for its potential to reduce oral intraepithelial neoplasia (OIN) lesions. In a placebo-controlled study, topical application of a mucoadhesive gel containing 10% w/w freeze-dried black raspberry powder four-times daily for six weeks to OIN lesions significantly decreased lesion size, the severity of oral dysplasia, and loss of heterozygosity indices [248,249]. Elevated levels of COX-2 and iNOS are correlated with the malignant transformation of OIN. Treatment with a black raspberry powder-containing gel uniformly suppressed the expression of genes associated with RNA processing, growth factor signaling, and inhibition of apoptosis in human premalignant oral lesions [250]. Furthermore, the gel application reduced the expression of COX-2 and iNOS, which correlates with malignant transformation of oral intraepithelial neoplasia, while reducing vascular densities in the superficial connective tissues and inducing genes associated with keratinocyte terminal differentiation. In another study, daily intake of black raspberry slurry for six months was assessed in 77 individuals with Barrett’s esophagus; however, the severity of Barrett’s esophagus was not affected [251]. As the transition time of black raspberry anthocyanins through the esophagus is short, lesions may have failed to absorb a sufficient amount of anthocyanins. It is, therefore, essential to prepare anthocyanins in formulas that have an enhanced absorption into the esophageal tissues. Similarly, a cohort study of 469,008 participants was carried out to determine the association between flavonoid (including anthocyanin) intake and esophageal, head and neck, and gastric carcinoma risk by analyzing the flavonoid intake in each food item using the 2015 USDA Expanded Flavonoid Database for the Assessment of Dietary Intakes [252]. Based on the reported data, flavonoid intake has no relationship with the incidence of esophageal or gastric cancer but showed an inverse relationship with head and neck cancer. Clinical studies on humans have provided additional evidence for the use of black raspberry in CRC prevention. Black raspberry supplementation modifies energy-generating pathways by regulating multiple metabolites, which, in turn, aid in CRC prevention [253]. Among 20 patients who received a freeze-dried black raspberry supplement of 1062 mg total anthocyanins/individual/day for nine weeks, the demethylation of tumor suppressor genes was increased [253]. Methylation of tumor suppressor genes causes their silencing and can induce mutational events, which plays a fundamental role in precipitating the development of a large and diverse number of human GI cancers [254]. Based on epidemiological studies, the value of anthocyanins in GI cancer prevention remains controversial and therefore requires additional investigation.
Circulating cytokines are one of the key indicators of risk and stage of CRC; expression of IL-6, IL-8, TNF-α is upregulated, and IL-2 is downregulated in CRC development [255]. Despite the positive results reported in cell-based studies, anthocyanin intake has not always been shown effective in altering the cytokine profile in favor of CRC reduction. For example, a slurry of freeze-dried black raspberry 354 mg/day in 100 mL of drinking water was not effective in modulating the plasma concentrations of cytokines in 24 CRC patients and, indeed, increased the plasma concentrations of granulocyte-macrophage colony-stimulating factor (GM-CSF), which promotes tumorigenesis by stimulating the epithelial cell release of vascular endothelial growth factor (VEGF) that enhances tumor survivability. Those findings reveal controversy surrounding the effectiveness of anthocyanins in CRC prevention [256]. However, on the other hand, DNA methylation, methyltransferase I protein expression and p16 promoter methylation were significantly reduced in 14 FAP patients who received black raspberry powder for nine months by oral administration (1787 mg/individual/day) and rectal insertion (595 mg/individual/day) of two suppositories [257]. Although the tumor burden was reduced, raspberry supplementation did not reduce the number of tumors. Black raspberry supplements are reasonably well tolerated by cancer patients, showing no adverse effects. However, the anti-cancer effect of black raspberry anthocyanin supplementation might be impacted by variables such as the microbiome. In another clinical study, oral supplementation with commercially available black currant extract powder (672 mg/day) altered gut microbial composition in 30 healthy adult male and female subjects by increasing the relative abundance of beneficial bacteria (Lactobacillus and Bifidobacteria) while reducing Clostridium and Bacteroides numbers and inhibiting β-glucuronidase [258]. Anthocyanins are, therefore, potent modulators of gut microbial dysbiosis in CRC.

7. Conclusions and Future Directions

GI cancers remain the most common reason for cancer-related deaths worldwide. The sporadic nature of the disease provides a rationale for diet-related cancer prevention, as has been supported by considerable evidence generated from in vitro and in vivo studies and clinical trials. In this review, the diverse beneficial effects of anthocyanins in the chemoprevention of GI cancers have been discussed. Anthocyanin-rich extracts and isolated individual anthocyanins in GI cancer prevention have been investigated during the past two decades. Most of the investigated anthocyanin-rich extracts also contain other flavonoids and polyphenols, ascorbic acid, and sugars. Therefore, the chemopreventive properties of anthocyanin-rich extracts are attributed to the respective health-promoting effects of combinations of compounds; however, the synergistic effect of anthocyanins in phytocomplexes needs to be studied. Although the molecular mechanisms of cancer prevention by anthocyanins are not well elucidated, the involvement of anthocyanins in the modulation of MAPK, NF-κB, AMPK, and Wnt/β-catenin pathways of normal and cancer cells are well documented. Dietary anthocyanins contribute to the prevention of GI cancer initiation via their antioxidative properties. Findings over the past decade reveal anthocyanin-mediated direct scavenging of ROS, the elevation of oxygen radical absorbing capacity of normal cells, stimulation of the expression of phase II detoxification enzymes, reduction in the formation of oxidative DNA adducts, and inhibition of mutagenesis by environmental toxins and carcinogens. As a sub-class of flavonoids, anthocyanins may transition from antioxidants to prooxidants depending on the concentration and its micro-environment, such as the presence of transition metal ions. However, we have not come across any report on the prooxidant effect of anthocyanin related to GI cancers. Further, anthocyanins have the potential to reduce microbial dysbiosis and GI tract inflammation by improving intestinal TJ barrier integrity by promoting the mRNA expression of key barrier-forming TJ proteins such as occludin, claudin-5, and zonnula occuldin-1 via upregulating the GLP-2 intestinal hormone levels. Anthocyanins are also potent inhibitors of GI cancer cell growth due to their ability to increase the levels of cyclin-dependent kinase inhibitor proteins and cell cycle regulatory proteins such as p53, p21, and p27, arrest the cell cycle and induce GI cancer cell apoptosis by facilitating the release of mitochondrial cytochrome c, activation of caspase-releasing enzymes and increasing the Bax:Bcl-2 ratio. These factors all contribute to the prevention of GI cancer development. Anthocyanins also inhibit GI cancer progression via inhibiting metastasis by downregulation of MMP-2 and MMP-9 activity, which maintains the integrity of the epithelial barrier. However, it is important to note that many of the documented beneficial effects of anthocyanins are based on cell-based and experimental animal model-based studies. The concentration of anthocyanins with antiproliferative efficacy ranges from 25 to 200 µM in cell cultures, while the low systemic bioavailability of anthocyanins significantly diminishes their in vivo chemopreventive properties. Additional investigation is also required to develop methods of enhancing the bioavailability of anthocyanins. Novel food technologies, such as micro-encapsulation and nano-encapsulation of anthocyanins that might enhance anthocyanin delivery to targeted sites of the GI tract need further study. Identifying the most effective anthocyanin metabolites in terms of chemoprevention will facilitate the design of novel therapeutics for GI cancer prevention and treatment. Given that mixtures of different anthocyanins may be more effective than single compounds in managing the GI cancers, identification of optimal synergistic combinations of anthocyanins, as well as their formulation with other bioactives in GI cancer prevention, is a logical approach. However, as current knowledge regarding anthocyanins in GI cancer prevention is limited, future investigations are necessary to validate laboratory findings using properly designed human dietary intervention studies.

Author Contributions

Conceptualization: K.V.S.D., D.W.H. and H.P.V.R.; writing—original draft preparation: K.V.S.D.; writing—review and editing: D.W.H. and H.P.V.R.; visualization: K.V.S.D.; supervision: H.P.V.R.; funding acquisition: H.P.V.R. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Killam Chair funds (H.P.V.R.).

Acknowledgments

The authors wish to acknowledge the cancer research knowledge sharing programs offered by Beatrice Hunter Cancer Research Institute (BHCRI) of Halifax, NS, Canada.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ACFAberrant crypt foci
AOMAzoxymethane
AREAntioxidant response element
BaxB-cell lymphoma-2-like protein 4
Bcl-2B-cell lymphoma-2
C3GCyanidin-3-O-glucoside
CDKsCyclin-dependent kinases
COXCyclooxygenase
CRCColorectal cancer
DNADeoxyribose nucleic acid
DSSDextran sulfate sodium
ESCCEsophageal squamous cell carcinoma
FAPFamilial adenomatous polyposis
GIGastrointestinal
GSHGlutathione-s-transferase
HCCHepatocellular carcinoma
IFN-γInterferon-gamma
IIRIntestinal ischemia-reperfusion
ILInterleukin
iNOSInducible nitric oxide synthase
MAPKMitogen-activated protein kinase
MMPMatrix metalloproteinase
NF-κBNuclear factor-kappa B
NONitrogen oxide
Nrf-2Nuclear factor-E2-related factor-2
OINOral intraepithelial neoplasia
PCAprotocatechuic acid
RNIReactive nitrogen intermediates
RNSReactive nitrogen species
ROSReactive oxygen species
SCCSquamous cell carcinoma
TJTight junction
TNF -αTumor necrosis factor-alpha
ΔΨmMitochondrial membrane potential

References

  1. George, V.C.; Dellaire, G.; Rupasinghe, H.P.V. Plant flavonoids in cancer chemoprevention: Role in genome stability. J. Nutr. Biochem. 2017, 45, 1–14. [Google Scholar] [CrossRef] [PubMed]
  2. Technical Report: WHO Report on Cancer: Setting Priorities, Investing Wisely and Providing Care for All; World Health Organization: Geneva, Switzerland, 2020.
  3. Arnold, M.; Abnet, C.C.; Neale, R.E.; Vignat, J.; Edward, L.; Mcglynn, K.A.; Bray, F. Global burden of 5 major types of gastrointestinal cancer. Gastroenterology 2020, 159, 335–349. [Google Scholar] [CrossRef] [PubMed]
  4. Court, C.M.; Ankeny, J.S.; Sho, S.; Tomlinson, J.S. Circulating tumor cells in gastrointestinal cancer: Current practices and future directions. Cancer Treat. Res. 2016, 168, 345–376. [Google Scholar] [PubMed]
  5. Vedeld, H.M.; Goel, A.; Lind, G.E. Epigenetic biomarkers in gastrointestinal cancers: The current state and clinical perspectives. Semin. Cancer Biol. 2018, 51, 36–49. [Google Scholar] [CrossRef]
  6. Patel, T.N.; Roy, S.; Ravi, R. Gastric cancer and related epigenetic alterations. Ecancermedicalscience 2017, 11, 714. [Google Scholar] [CrossRef]
  7. Al-Ishaq, R.K.; Overy, A.J.; Büsselberg, D. Phytochemicals and gastrointestinal cancer: Cellular mechanisms and effects to change cancer progression. Biomolecules 2020, 10, 105. [Google Scholar] [CrossRef] [Green Version]
  8. Wei, D.; Kanai, M.; Huang, S.; Xie, K. Emerging role of KLF4 in human gastrointestinal cancer. Carcinogenesis 2006, 27, 23–31. [Google Scholar] [CrossRef] [Green Version]
  9. Ida, S.; Watanabe, M.; Baba, H. Chronic inflammation and gastrointestinal cancer. J. Cancer Metastasis Treat. 2015, 1, 138. [Google Scholar]
  10. Ferlay, J.; Soerjomataram, I.; Dikshit, R.; Eser, S.; Mathers, C.; Rebelo, M.; Parkin, D.M.; Forman, D.; Bray, F. Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. Int. J. Cancer 2015, 136, 359–386. [Google Scholar] [CrossRef]
  11. Li, Y.; Zhang, T.; Chen, G.Y. Flavonoids and colorectal cancer prevention. Antioxidants 2018, 7, 187. [Google Scholar] [CrossRef] [Green Version]
  12. Rodríguez-García, C.; Sánchez-Quesada, C.; Gaforio, J.J.; Gaforio, J.J. Dietary flavonoids as cancer chemopreventive agents: An updated review of human studies. Antioxidants 2019, 8, 137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Kozłowska, A.; Szostak-Węgierek, D. Flavonoids–Food Sources, Health Benefits, and Mechanisms Involved. In Bioactive Molecules in Food; Springer International Publishing: Cham, Switzerland, 2018; pp. 1–27. [Google Scholar]
  14. Yao, L.H.; Jiang, Y.M.; Shi, J.; Tomás-Barberán, F.A.; Datta, N.; Singanusong, R.; Chen, S.S. Flavonoids in food and their health benefits. Plant Foods Hum. Nutr. 2004, 59, 113–122. [Google Scholar] [CrossRef] [PubMed]
  15. Ruiz-Cruz, S.; Chaparro-Hernández, S.; Ruiz, K.L.H.; Cira-Chávez, L.A.; Estrada-Alvarado, M.I.; Ortega, L.E.G.; Ornelas-Paz, J.J.; Mata, M.A.L. Flavonoids: Important Biocompounds in Food. In Flavonoids—From Biosynthesis to Human Health; InTech: London, UK, 2017. [Google Scholar]
  16. Rupasinghe, H.P.V.; Arumuggam, N. Health Benefits of Anthocyanins. In Food Chemistry, Function and Analysis; Royal Society of Chemistry: London, UK, 2019; pp. 123–158. [Google Scholar]
  17. Khoo, H.E.; Azlan, A.; Tang, S.T.; Lim, S.M. Anthocyanidins and anthocyanins: Colored pigments as food, pharmaceutical ingredients, and the potential health benefits. Food Nutr. Res. 2017, 61, 1361779. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Fang, J. Classification of fruits based on anthocyanin types and relevance to their health effects. Nutrition 2015, 31, 1301–1306. [Google Scholar] [CrossRef]
  19. Benetou, V.; Lagiou, A.; Lagiou, P. Chemoprevention of cancer: Current evidence and future prospects. F1000Research 2015, 4, 916. [Google Scholar] [CrossRef] [Green Version]
  20. Li, D.; Wang, P.; Luo, Y.; Zhao, M.; Chen, F. Health benefits of anthocyanins and molecular mechanisms: Update from recent decade. Crit. Rev. Food Sci. Nutr. 2017, 57, 1729–1741. [Google Scholar] [CrossRef]
  21. De Sousa Moraes, L.F.; Sun, X.F.; Peluzio, M.d.C.G.; Zhu, M.J. Anthocyanins/anthocyanidins and colorectal cancer: What is behind the scenes? Crit. Rev. Food Sci. Nutr. 2017, 1–13. [Google Scholar]
  22. Forester, S.C.; Choy, Y.Y.; Waterhouse, A.L.; Oteiza, P.I. The anthocyanin metabolites gallic acid, 3-O-methylgallic acid, and 2,4,6-trihydroxybenzaldehyde decrease human colon cancer cell viability by regulating pro-oncogenic signals. Mol. Carcinog. 2014, 53, 432–439. [Google Scholar] [CrossRef]
  23. Global Cancer Facts & Figures 4th Edition; American Cancer Society: Atlanta, GA, USA, 2018.
  24. Katona, B.W.; Lynch, J.P. Mechanisms of Gastrointestinal Malignancies. In Physiology of the Gastrointestinal Tract: Sixth Edition; Elsevier Inc: Cambridge, MN, USA, 2018; Volume 2, pp. 1615–1642. [Google Scholar]
  25. Thirumurthi, S.; Vilar, E.; Lynch, P.J. Hereditary Gastrointestinal Cancers. In Textbook of Gastrointestinal Oncology; Springer: Cham, Switzerland, 2019; pp. 595–611. [Google Scholar]
  26. Rahner, N.; Steinke, V. Hereditary cancer syndromes. Dtsch. Arztebl. 2008, 105, 706–714. [Google Scholar] [CrossRef]
  27. Coleman, W.B. Neoplasia. In Molecular Pathology: The Molecular Basis of Human Disease; Elsevier Inc.: Cambridge, MN, USA, 2018; pp. 71–97. [Google Scholar]
  28. Knudson, A.G. Mutation and cancer: Statistical study of retinoblastoma. Proc. Natl. Acad. Sci. USA 1971, 68, 820–823. [Google Scholar] [CrossRef] [Green Version]
  29. Van Nistelrooij, A.M.J.; Dinjens, W.N.M.; Wagner, A.; Spaander, M.C.W.; van Lanschot, J.J.B.; Wijnhoven, B.P.L. Hereditary factors in esophageal adenocarcinoma. Gastrointest. Tumors 2014, 1, 93–98. [Google Scholar] [CrossRef] [PubMed]
  30. Ellis, A.; Risk, J.M.; Maruthappu, T.; Kelsell, D.P. Tylosis with oesophageal cancer: Diagnosis, management and molecular mechanisms. Orphanet. J. Rare Dis. 2015, 10, 126. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Colvin, H.; Yamamoto, K.; Wada, N.; Mori, M. Hereditary gastric cancer syndromes. Surg. Oncol. Clin. N. Am. 2015, 24, 765–777. [Google Scholar] [CrossRef] [PubMed]
  32. Eguchi, H.; Kobayashi, S.; Gotoh, K.; Noda, T.; Doki, Y. Characteristics of early-onset pancreatic cancer and its association with familial pancreatic cancer and hereditary pancreatic cancer syndromes. Ann. Gastroenterol. Surg. 2020, 4, 229–233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Wells, K.; Wise, P.E. Hereditary colorectal cancer syndromes. Surg. Clin. N. Am. 2017, 97, 605–625. [Google Scholar] [CrossRef] [PubMed]
  34. Kanth, P.; Grimmett, J.; Champine, M.; Burt, R.; Samadder, J.N. Hereditary colorectal polyposis and cancer syndromes: A primer on diagnosis and management. Am. J. Gastroenterol. 2017, 112, 1509–1525. [Google Scholar] [CrossRef]
  35. Shenoy, S. Genetic risks and familial associations of small bowel carcinoma. World J. Gastrointest. Oncol. 2016, 8, 509. [Google Scholar] [CrossRef]
  36. Vanier, M.T. Niemann-Pick diseases. In Handbook of Clinical Neurology; Elsevier B.V.: Berlin/Heidelberg, Germany, 2013; Volume 113, pp. 1717–1721. [Google Scholar]
  37. Yang Chou, J.; Mansfield, B.C. Molecular genetics of type 1 glycogen storage diseases. Trends Endocrinol. Metab. 1999, 10, 104–113. [Google Scholar] [CrossRef]
  38. Villanueva, A.; Newell, P.; Hoshida, Y. Inherited hepatocellular carcinoma. Best Pract. Res. Clin. Gastroenterol. 2010, 24, 725–734. [Google Scholar] [CrossRef]
  39. Chang, I.J.; Hahn, S.H. The genetics of Wilson disease. In Handbook of Clinical Neurology; Elsevier B.V.: Berlin/Heidelberg, Germany, 2017; Volume 142, pp. 19–34. [Google Scholar]
  40. MacIas, I.; Laín, A.; Bernardo-Seisdedos, G.; Gil, D.; Gonzalez, E.; Falcon-Perez, J.M.; Millet, O. Hereditary tyrosinemia type I-associated mutations in fumarylacetoacetate hydrolase reduce the enzyme stability and increase its aggregation rate. J. Biol. Chem. 2019, 294, 13051–13060. [Google Scholar] [CrossRef] [Green Version]
  41. Gidi, A.D.G.; González-Chávez, M.A.; Villegas-Tovar, E.; Visag-Castillo, V.; Pantoja-Millan, J.P.; Vélez-Pérez, F.M.; Cano-García, F.; Contreras, A.G. An unusual type of biliar cyst: A case report. Ann. Hepatol. 2016, 15, 788–794. [Google Scholar] [PubMed]
  42. Moss, A.; Nalankilli, K. The association between diet and colorectal cancer risk: Moving beyond generalizations. Gastroenterology 2017, 152, 1821–1823. [Google Scholar] [CrossRef] [Green Version]
  43. Mehta, R.S.; Song, M.; Nishihara, R.; Drew, D.A.; Wu, K.; Qian, Z.R.; Fung, T.T.; Hamada, T.; Masugi, Y.; da Silva, A.; et al. Dietary patterns and risk of colorectal cancer: Analysis by tumor location and molecular subtypes. Gastroenterology 2017, 152, 1944–1953. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Gaddy, J.A.; Radin, J.N.; Loh, J.T.; Zhang, F.; Kay Washington, M.; Peek, R.M.; Scott Algood, H.M.; Cover, T.L. Hiharriscagh dietary salt intake exacerbates Helicobacter pylori-induced gastric carcinogenesis. Infect. Immun. 2013, 81, 2258–2267. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. D’Elia, L.; Galletti, F.; Strazzullo, P. Dietary salt intake and risk of gastric cancer. Cancer Treat. Res. 2014, 159, 83–95. [Google Scholar]
  46. Ulrich, C.M.; Himbert, C.; Holowatyj, A.N.; Hursting, S.D. Energy balance and gastrointestinal cancer: Risk, interventions, outcomes and mechanisms. Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 683–698. [Google Scholar] [CrossRef]
  47. Gunter, M.J.; Riboli, E. Obesity and gastrointestinal cancers—Where do we go from here? Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 651–652. [Google Scholar] [CrossRef]
  48. Seitz, H.K.; Stickel, F. Molecular mechanisms of alcohol-mediated carcinogenesis. Nat. Rev. Cancer 2007, 7, 599–612. [Google Scholar] [CrossRef]
  49. Baan, R.; Straif, K.; Grosse, Y.; Secretan, B.; El Ghissassi, F.; Bouvard, V.; Altieri, A.; Cogliano, V. Carcinogenicity of alcoholic beverages. Lancet Oncol. 2007, 8, 292–293. [Google Scholar] [CrossRef]
  50. Sierra, J.C.; Piazuelo, M.B.; Luis, P.B.; Barry, D.P.; Allaman, M.M.; Asim, M.; Sebrell, T.A.; Finley, J.L.; Rose, K.L.; Hill, S.; et al. Spermine oxidase mediates Helicobacter pylori-induced gastric inflammation, DNA damage, and carcinogenic signaling. Oncogene 2020, 1–10. [Google Scholar] [CrossRef]
  51. Huang, F.L.; Yu, S.J. Esophageal cancer: Risk factors, genetic association, and treatment. Asian J. Surg. 2018, 41, 210–215. [Google Scholar] [CrossRef] [PubMed]
  52. Bao, Y.; Giovannucci, E.; Fuchs, C.S.; Michaud, D.S. Passive smoking and pancreatic cancer in women: A prospective cohort study. Cancer Epidemiol. Biomarkers Prev. 2009, 18, 2292–2296. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Praud, D.; Rota, M.; Pelucchi, C.; Bertuccio, P.; Rosso, T.; Galeone, C.; Zhang, Z.F.; Matsuo, K.; Ito, H.; Hu, J.; et al. Cigarette smoking and gastric cancer in the Stomach Cancer Pooling (StoP) Project. Eur. J. Cancer Prev. 2018, 27, 124–133. [Google Scholar] [CrossRef] [PubMed]
  54. Moy, K.A.; Fan, Y.; Wang, R.; Gao, Y.T.; Yu, M.C.; Yuan, J.M. Alcohol and tobacco use in relation to gastric cancer: A prospective study of men in Shanghai, China. Cancer Epidemiol. Biomark. Prev. 2010, 19, 2287–2297. [Google Scholar] [CrossRef] [Green Version]
  55. Sjödahl, K.; Jansson, C.; Bergdahl, I.A.; Adami, J.; Boffetta, P.; Lagergren, J. Airborne exposures and risk of gastric cancer: A prospective cohort study. Int. J. Cancer. 2007, 120, 2013–2018. [Google Scholar] [CrossRef]
  56. De Oliveira, G.A.; Cheng, R.Y.S.; Ridnour, L.A.; Basudhar, D.; Somasundaram, V.; McVicar, D.W.; Monteiro, H.P.; Wink, D.A. Inducible nitric oxide synthase in the carcinogenesis of gastrointestinal cancers. Antioxid. Redox Signal. 2017, 26, 1059–1077. [Google Scholar] [CrossRef]
  57. Adam, M.A.A.; Tabana, Y.M.; Musa, K.B.; Sandai, D.A. Effects of different mycotoxins on humans, cell genome and their involvement in cancer (Review). Oncol. Rep. 2017, 37, 1321–1336. [Google Scholar] [CrossRef] [Green Version]
  58. Marchese, S.; Polo, A.; Ariano, A.; Velotto, S.; Costantini, S.; Severino, L. Aflatoxin B1 and M1: Biological properties and their involvement in cancer development. Toxins 2018, 10, 214. [Google Scholar] [CrossRef] [Green Version]
  59. Myburg, R.B.; Dutton, M.F.; Chuturgoon, A.A. Cytotoxicity of fumonisin B1, diethylnitrosamine, and catechol on the SNO esophageal cancer cell line. Environ. Health Perspect. 2002, 110, 813–815. [Google Scholar] [CrossRef]
  60. Mahmoodi, M.; Alizadeh, A.M.; Sohanaki, H.; Rezaei, N.; Amini-Najafi, F.; Khosravi, A.R.; Hosseini, S.K.; Safari, Z.; Hydarnasab, D.; Khori, V. Impact of fumonisin B1 on the production of inflammatory cytokines by gastric and colon cell lines. Iran. J. Allergy Asthma Immunol. 2012, 11, 165–173. [Google Scholar]
  61. Alfarouk, K.O.; Bashir, A.H.H.; Aljarbou, A.N.; Ramadan, A.M.; Muddathir, A.K.; AlHoufie, S.T.S.; Hifny, A.; Elhassan, G.O.; Ibrahim, M.E.; Alqahtani, S.S.; et al. The possible role of Helicobacter pylori gastric cancer and its management. Front. Oncol. 2019, 9, 75. [Google Scholar] [CrossRef] [PubMed]
  62. Axelrad, J.E.; Lichtiger, S.; Yajnik, V. Inflammatory bowel disease and cancer: The role of inflammation, immunosuppression, and cancer treatment. World J. Gastroenterol. 2016, 22, 4794–4801. [Google Scholar] [CrossRef] [PubMed]
  63. Singh, N.; Baby, D.; Rajguru, J.; Patil, P.; Thakkannavar, S.; Pujari, V. Inflammation and cancer. Ann. Afr. Med. 2019, 18, 121. [Google Scholar] [CrossRef] [PubMed]
  64. Vaidya, F.U.; Chhipa, A.S.; Sagar, N.; Pathak, C. Oxidative Stress and Inflammation Can Fuel Cancer. In Role of Oxidative Stress in Pathophysiology of Diseases; Springer: Singapore, 2020; pp. 229–258. [Google Scholar]
  65. Yu, J.H.; Kim, H. Oxidative stress and cytokines in the pathogenesis of pancreatic cancer. J. Cancer Prev. 2014, 19, 97–102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Federico, A.; Morgillo, F.; Tuccillo, C.; Ciardiello, F.; Loguercio, C. Chronic inflammation and oxidative stress in human carcinogenesis. Int. J. Cancer 2007, 121, 2381–2386. [Google Scholar] [CrossRef] [PubMed]
  67. O’Sullivan, J.; Lysaght, J.; Donohoe, C.L.; Reynolds, J.V. Obesity and gastrointestinal cancer: The interrelationship of adipose and tumour microenvironments. Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 699–714. [Google Scholar] [CrossRef] [PubMed]
  68. Stickel, F.; Schuppan, D.; Hahn, E.G.; Seitz, H.K. Cocarcinogenic effects of alcohol in hepatocarcinogenesis. Gut 2002, 51, 132–139. [Google Scholar] [CrossRef] [Green Version]
  69. Schroeder, B.O.; Birchenough, G.M.H.; Ståhlman, M.; Arike, L.; Johansson, M.E.V.; Hansson, G.C.; Bäckhed, F. Bifidobacteria or fiber protects against diet-induced microbiota-mediated colonic mucus deterioration. Cell Host Microbe 2018, 23, 27–40. [Google Scholar] [CrossRef] [Green Version]
  70. Weng, M.T.; Chiu, Y.T.; Wei, P.Y.; Chiang, C.W.; Fang, H.L.; Wei, S.C. Microbiota and gastrointestinal cancer. J. Formos. Med. Assoc. 2019, 118, 32–41. [Google Scholar] [CrossRef]
  71. Amararathna, M.; Johnston, M.R.; Rupasinghe, H.P.V. Plant polyphenols as chemopreventive agents for lung cancer. Int. J. Mol. Sci. 2016, 17, 1352. [Google Scholar] [CrossRef] [Green Version]
  72. Sayed, M.; ElHamid Mahmou, A.A. Cancer chemoprevention by dietary polyphenols. In Carcinogenesis; InTech: London, UK, 2013. [Google Scholar]
  73. Fernandes, I.; de Freitas, V.; Mateus, N. Anthocyanins and human health: How gastric absorption may influence acute human physiology. Nutr. Aging 2014, 2, 1–14. [Google Scholar] [CrossRef] [Green Version]
  74. Engwa, G.A. Free radicals and the role of plant phytochemicals as antioxidants against oxidative stress-related diseases. In Phytochemicals—Source of Antioxidants and Role in Disease Prevention; InTech: London, UK, 2018. [Google Scholar]
  75. Yang, C.S.; Landau, J.M.; Huang, M.T.; Newmark, H.L. Inhibition of carcinogenesis by dietary polyphenolic compounds. Annu. Rev. Nutr. 2001, 21, 381–406. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Wang, X.; Yang, D.Y.; Yang, L.Q.; Zhao, W.Z.; Cai, L.Y.; Shi, H.P. Anthocyanin consumption and risk of colorectal cancer: A meta-analysis of observational studies. J. Am. Coll. Nutr. 2019, 38, 470–477. [Google Scholar] [CrossRef] [PubMed]
  77. Lee, J.; Shin, A.; Oh, J.H.; Kim, J. Colors of vegetables and fruits and the risks of colorectal cancer. World J. Gastroenterol. 2017, 23, 2527–2538. [Google Scholar] [CrossRef] [PubMed]
  78. Xu, C.; Zeng, X.T.; Liu, T.Z.; Zhang, C.; Yang, Z.H.; Li, S.; Chen, X.Y. Fruits and vegetables intake and risk of bladder cancer: A PRISMA-compliant systematic review and dose-response meta-analysis of prospective cohort studies. Medicine 2015, 94, 759. [Google Scholar] [CrossRef]
  79. Larsson, S.C.; Bergkvist, L.; Wolk, A. Fruit and vegetable consumption and incidence of gastric cancer: A prospective study. Cancer Epidemiol. Biomark. Prev. 2006, 15, 1998–2001. [Google Scholar] [CrossRef] [Green Version]
  80. Clifford, M.N. Anthocyanins—Nature, occurrence and dietary burden. J. Sci. Food Agric. 2000, 80, 1063–1072. [Google Scholar] [CrossRef]
  81. He, J.; Giusti, M.M. Anthocyanins: Natural colorants with health-promoting properties. Annu. Rev. Food Sci. Technol. 2010, 1, 163–187. [Google Scholar] [CrossRef]
  82. Rupasinghe, H.P.V.; Arumuggam, N.; Amararathna, M.; De Silva, A.B.K.H. The potential health benefits of haskap (Lonicera caerulea L.): Role of cyanidin-3-O-glucoside. J. Funct. Foods. 2018, 44, 24–39. [Google Scholar] [CrossRef]
  83. Fang, J. Bioavailability of anthocyanins. Drug Metab. Rev. 2014, 46, 508–520. [Google Scholar] [CrossRef]
  84. Liu, Y.; Zhang, D.; Wu, Y.; Wang, D.; Wei, Y.; Wu, J.; Ji, B. Stability and absorption of anthocyanins from blueberries subjected to a simulated digestion process. Int. J. Food Sci. Nutr. 2014, 65, 440–448. [Google Scholar] [CrossRef] [PubMed]
  85. Sun, D.; Huang, S.; Cai, S.; Cao, J.; Han, P. Digestion property and synergistic effect on biological activity of purple rice (Oryza sativa L.) anthocyanins subjected to a simulated gastrointestinal digestion in vitro. Food Res. Int. 2015, 78, 114–123. [Google Scholar] [CrossRef] [PubMed]
  86. Czank, C.; Cassidy, A.; Zhang, Q.; Morrison, D.J.; Preston, T.; Kroon, P.A.; Botting, N.P.; Kay, C.D. Human metabolism and elimination of the anthocyanin, cyanidin-3-glucoside: A 13C-tracer study. Am. J. Clin. Nutr. 2013, 97, 995–1003. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Mueller, D.; Jung, K.; Winter, M.; Rogoll, D.; Melcher, R.; Richling, E. Human intervention study to investigate the intestinal accessibility and bioavailability of anthocyanins from bilberries. Food Chem. 2017, 231, 275–286. [Google Scholar] [CrossRef] [PubMed]
  88. Felgines, C.; Talavéra, S.; Texier, O.; Besson, C.; Fogliano, V.; Lamaison, J.L.; la Fauci, L.; Galvano, G.; Rémésy, C.; Galvano, F. Absorption and metabolism of red orange juice anthocyanins in rats. Br. J. Nutr. 2006, 95, 898–904. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Talavéra, S.; Felgines, C.; Texier, O.; Besson, C.; Lamaison, J.L.; Rémésy, C. Anthocyanins are efficiently absorbed from the stomach in anesthetized rats. J. Nutr. 2003, 133, 4178–4182. [Google Scholar] [CrossRef]
  90. Passamonti, S.; Vrhovsek, U.; Vanzo, A.; Mattivi, F. The stomach as a site for anthocyanins absorption from food. FEBS Lett. 2003, 544, 210–213. [Google Scholar] [CrossRef] [Green Version]
  91. Novotny, J.A.; Clevidence, B.A.; Kurilich, A.C. Anthocyanin kinetics are dependent on anthocyanin structure. Br. J. Nutr. 2012, 107, 504–509. [Google Scholar] [CrossRef] [Green Version]
  92. Felgines, C.; Texier, O.; Besson, C.; Vitaglione, P.; Lamaison, J.L.; Fogliano, V.; Scalbert, A.; Vanella, L.; Galvano, F. Influence of glucose on cyanidin 3-glucoside absorption in rats. Mol. Nutr. Food Res. 2008, 52, 959–964. [Google Scholar] [CrossRef]
  93. Verine Talavé, S.; Felgines, C.; Texier, O.; Besson, C.; Manach, C.; Lamaison, J.L.; Ré, C.; Sy, M. Anthocyanins are efficiently absorbed from the small intestine in rats. Nutr. Metab. 2004, 134, 2275–2279. [Google Scholar]
  94. Valdez, J.C.; Bolling, B.W. View of anthocyanins and intestinal barrier function: A review. J. Food Bioact. 2019, 5, 18–30. [Google Scholar] [CrossRef] [Green Version]
  95. Fernandes, I.; Faria, A.; de Freitas, V.; Calhau, C.; Mateus, N. Multiple-approach studies to assess anthocyanin bioavailability. Phytochem. Rev. 2015, 14, 899–919. [Google Scholar] [CrossRef]
  96. Hidalgo, M.; Oruna-Concha, M.J.; Kolida, S.; Walton, G.E.; Kallithraka, S.; Spencer, J.P.E.; Gibson, G.R.; De Pascual-Teresa, S. Metabolism of anthocyanins by human gut microflora and their influence on gut bacterial growth. J. Agric. Food Chem. 2012, 60, 3882–3890. [Google Scholar] [CrossRef] [PubMed]
  97. Chen, Y.; Li, Q.; Zhao, T.; Zhang, Z.; Mao, G.; Feng, W.; Wu, X.; Yang, L. Biotransformation and metabolism of three mulberry anthocyanin monomers by rat gut microflora. Food Chem. 2017, 237, 887–894. [Google Scholar] [CrossRef]
  98. Hanske, L.; Engst, W.; Loh, G.; Sczesny, S.; Blaut, M.; Braune, A. Contribution of gut bacteria to the metabolism of cyanidin 3-glucoside in human microbiota-associated rats. Br. J. Nutr. 2013, 109, 1433–1441. [Google Scholar] [CrossRef]
  99. Tsuda, T. Anthocyanins as functional food factors—Chemistry, nutrition and health promotion. Food Sci. Technol. Res. 2012, 18, 315–324. [Google Scholar] [CrossRef] [Green Version]
  100. Amararathna, M.; Hoskin, D.W.; Rupasinghe, H.P.V. Anthocyanin-rich haskap (Lonicera caerulea L.) berry extracts reduce nitrosamine-induced DNA damage in human normal lung epithelial cells in vitro. Food Chem. Toxicol. 2020, 141, 111404. [Google Scholar] [CrossRef]
  101. Samuels, T.L.; Pearson, A.C.S.; Wells, C.W.; Stoner, G.D.; Johnston, N. Curcumin and anthocyanin inhibit pepsin-mediated cell damage and carcinogenic changes in airway epithelial cells. Ann. Otol. Rhinol. Laryngol. 2013, 122, 632–641. [Google Scholar] [CrossRef]
  102. Guttenplan, J.B.; Chen, K.M.; Sun, Y.W.; Lajara, B.; Shalaby, N.A.E.; Kosinska, W.; Benitez, G.; Gowda, K.; Amin, S.; Stoner, G.; et al. Effects of black raspberry extract and berry compounds on repair of DNA damage and mutagenesis induced by chemical and physical agents in human oral leukoplakia and rat oral fibroblasts. Chem. Res. Toxicol. 2017, 30, 2159–2164. [Google Scholar] [CrossRef]
  103. Karlsen, A.; Retterstøl, L.; Laake, P.; Paur, I.; Kjølsrud-Bøhn, S.; Sandvik, L.; Blomhoff, R. Anthocyanins inhibit nuclear factor-kB activation in monocytes and reduce plasma concentrations of pro-inflammatory mediators in healthy adults. J. Nutr. Nutr. Dis. 2007, 137, 1951–1954. [Google Scholar]
  104. Li, S.; Wu, B.; Fu, W.; Reddivari, L. The anti-inflammatory effects of dietary anthocyanins against ulcerative colitis. Int. J. Mol. Sci. 2019, 20, 2588. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Cremonini, E.; Daveri, E.; Mastaloudis, A.; Adamo, A.M.; Mills, D.; Kalanetra, K.; Hester, S.N.; Wood, S.M.; Fraga, C.G.; Oteiza, P.I. Anthocyanins protect the gastrointestinal tract from high fat diet-induced alterations in redox signaling, barrier integrity and dysbiosis. Redox Biol. 2019, 26, 101269. [Google Scholar] [CrossRef] [PubMed]
  106. Nunes, C.; Freitas, V.; Almeida, L.; Laranjinha, J. Red wine extract preserves tight junctions in intestinal epithelial cells under inflammatory conditions: Implications for intestinal inflammation. Food Funct. 2019, 10, 1364–1374. [Google Scholar] [CrossRef]
  107. Sun, X.; Du, M.; Navarre, D.A.; Zhu, M.-J. Purple Potato Extract Promotes Intestinal Epithelial Differentiation and Barrier Function by Activating AMP-Activated Protein Kinase. Mol. Nutr. Food Res. 2018, 62, 1700536. [Google Scholar] [CrossRef]
  108. Ferrari, D.; Speciale, A.; Cristani, M.; Fratantonio, D.; Molonia, M.S.; Ranaldi, G.; Saija, A.; Cimino, F. Cyanidin-3-O-glucoside inhibits NF-kB signalling in intestinal epithelial cells exposed to TNF-α and exerts protective effects via Nrf2 pathway activation. Toxicol. Lett. 2016, 264, 51–58. [Google Scholar] [CrossRef] [PubMed]
  109. Peng, Y.; Yan, Y.; Wan, P.; Chen, D.; Ding, Y.; Ran, L.; Mi, J.; Lu, L.; Zhang, Z.; Li, X.; et al. Gut microbiota modulation and anti-inflammatory properties of anthocyanins from the fruits of Lycium ruthenicum Murray in dextran sodium sulfate-induced colitis in mice. Free Radic. Biol. Med. 2019, 136, 96–108. [Google Scholar] [CrossRef]
  110. Li, J.; Wu, T.; Li, N.; Wang, X.; Chen, G.; Lyu, X. Bilberry anthocyanin extract promotes intestinal barrier function and inhibits digestive enzyme activity by regulating the gut microbiota in aging rats. Food Funct. 2019, 10, 333–343. [Google Scholar] [CrossRef]
  111. Rupasinghe, H.P.V.; Boehm, M.; Sekhon-Loodu, S.; Parmar, I.; Bors, B.; Jamieson, A. Anti-inflammatory activity of haskap cultivars is polyphenols-dependent. Biomolecules 2015, 5, 1079–1098. [Google Scholar] [CrossRef]
  112. Phan, M.A.T.; Bucknall, M.P.; Arcot, J. Interferences of anthocyanins with the uptake of lycopene in Caco-2 cells, and their interactive effects on anti-oxidation and anti-inflammation in vitro and ex vivo. Food Chem. 2019, 276, 402–409. [Google Scholar] [CrossRef]
  113. Lee, S.G.; Brownmiller, C.R.; Lee, S.O.; Kang, H.W. Anti-inflammatory and antioxidant effects of anthocyanins of Trifolium pratense (red clover) in lipopolysaccharide-stimulated RAW-267.4 macrophages. Nutrients 2020, 12, 1089. [Google Scholar] [CrossRef] [Green Version]
  114. Limtrakul, P.; Yodkeeree, S.; Pitchakarn, P.; Punfa, W. Suppression of inflammatory responses by black rice extract in RAW 264.7 macrophage cells via downregulation of NF-kB and AP-1 signaling pathways. Asian Pac. J. Cancer Prev. 2015, 16, 4277–4283. [Google Scholar] [CrossRef] [Green Version]
  115. Domitrovic, R. The molecular basis for the pharmacological activity of anthocyans. Curr. Med. Chem. 2011, 18, 4454–4469. [Google Scholar] [CrossRef] [PubMed]
  116. Peng, Y.; Yan, Y.; Wan, P.; Dong, W.; Huang, K.; Ran, L.; Mi, J.; Lu, L.; Zeng, X.; Cao, Y. Effects of long-term intake of anthocyanins from Lycium ruthenicum Murray on the organism health and gut microbiota in vivo. Food Res. Int. 2020, 130, 108952. [Google Scholar] [CrossRef] [PubMed]
  117. Venancio, V.P.; Cipriano, P.A.; Kim, H.; Antunes, L.M.G.; Talcott, S.T.; Mertens-Talcott, S.U. Cocoplum (Chrysobalanus icaco L.) anthocyanins exert anti-inflammatory activity in human colon cancer and non-malignant colon cells. Food Funct. 2017, 8, 307–314. [Google Scholar] [CrossRef]
  118. Li, L.; Wang, L.; Wu, Z.; Yao, L.; Wu, Y.; Huang, L.; Liu, K.; Zhou, X.; Gou, D. Anthocyanin-rich fractions from red raspberries attenuate inflammation in both RAW264.7 macrophages and a mouse model of colitis. Sci. Rep. 2014, 4, 6234. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Zhao, L.; Zhang, Y.; Liu, G.; Hao, S.; Wang, C.; Wang, Y. Black rice anthocyanin-rich extract and rosmarinic acid, alone and in combination, protect against DSS-induced colitis in mice. Food Funct. 2018, 9, 2796–2808. [Google Scholar] [CrossRef]
  120. Ghattamaneni, N.K.; Sharma, A.; Panchal, S.K.; Brown, L. Pelargonidin 3-glucoside-enriched strawberry attenuates symptoms of DSS-induced inflammatory bowel disease and diet-induced metabolic syndrome in rats. Eur. J. Nutr. 2019, 1–14. [Google Scholar] [CrossRef]
  121. Sies, H. Oxidative Stress: Eustress and Distress in Redox Homeostasis. In Stress: Physiology, Biochemistry, and Pathology; Academic Press: Cambridge, MA, USA, 2019; pp. 153–163. [Google Scholar]
  122. Ahmed, O.M. Relationships between oxidative stress, cancer development and therapeutic interventions. J. Cancer Sci. Res. 2018, 1, 1. [Google Scholar] [CrossRef]
  123. Céspedes-Acuña, C.L.; Xiao, J.; Wei, Z.J.; Chen, L.; Bastias, J.M.; Avila, J.G.; Alarcon-Enos, J.; Werner-Navarrete, E.; Kubo, I. Antioxidant and anti-inflammatory effects of extracts from Maqui berry Aristotelia chilensis in human colon cancer cells. J. Berry Res. 2018, 8, 275–296. [Google Scholar] [CrossRef]
  124. Shih, P.H.; Yeh, C.T.; Yen, G.C. Anthocyanins induce the activation of phase II enzymes through the antioxidant response element pathway against oxidative stress-induced apoptosis. J. Agric. Food Chem. 2007, 55, 9427–9435. [Google Scholar] [CrossRef]
  125. Yi, L.; Chen, C.Y.; Jin, X.; Mi, M.T.; Yu, B.; Chang, H.; Ling, W.H.; Zhang, T. Structural requirements of anthocyanins in relation to inhibition of endothelial injury induced by oxidized low-density lipoprotein and correlation with radical scavenging activity. FEBS Lett. 2010, 584, 583–590. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Juadjur, A.; Mohn, C.; Schantz, M.; Baum, M.; Winterhalter, P.; Richling, E. Fractionation of an anthocyanin-rich bilberry extract and in vitro antioxidative activity testing. Food Chem. 2015, 167, 418–424. [Google Scholar] [CrossRef] [PubMed]
  127. Olejnik, A.; Rychlik, J.; Kidoń, M.; Czapski, J.; Kowalska, K.; Juzwa, W.; Olkowicz, M.; Dembczyński, R.; Moyer, M.P. Antioxidant effects of gastrointestinal digested purple carrot extract on the human cells of colonic mucosa. Food Chem. 2016, 190, 1069–1077. [Google Scholar] [CrossRef] [PubMed]
  128. Claudio, S.R.; Gollucke, A.P.B.; Yamamura, H.; Morais, D.R.; Bataglion, G.A.; Eberlin, M.N.; Peres, R.C.; Oshima, C.T.F.; Ribeiro, D.A. Purple carrot extract protects against cadmium intoxication in multiple organs of rats: Genotoxicity, oxidative stress and tissue morphology analyses. J. Trace Elem. Med. Biol. 2016, 33, 37–47. [Google Scholar] [CrossRef] [PubMed]
  129. Chen, W.; Su, H.; Xu, Y.; Bao, T.; Zheng, X. Protective effect of wild raspberry (Rubus hirsutus Thunb.) extract against acrylamide-induced oxidative damage is potentiated after simulated gastrointestinal digestion. Food Chem. 2016, 196, 943–952. [Google Scholar] [CrossRef] [PubMed]
  130. Jakesevic, M.; Aaby, K.; Borge, G.I.A.; Jeppsson, B.; Ahrné, S.; Molin, G. Antioxidative protection of dietary bilberry, chokeberry and Lactobacillus plantarum HEAL19 in mice subjected to intestinal oxidative stress by ischemia-reperfusion. BMC Complement. Altern. Med. 2011, 11, 8. [Google Scholar] [CrossRef] [Green Version]
  131. Jakesevic, M.; Xu, J.; Aaby, K.; Jeppsson, B.; Ahrné, S.; Molin, G. Effects of bilberry (Vaccinium myrtillus) in combination with lactic acid bacteria on intestinal oxidative stress induced by ischemia-reperfusion in mouse. J. Agric. Food Chem. 2013, 61, 3468–3478. [Google Scholar] [CrossRef]
  132. Janšáková, K.; Bábíčková, J.; Filová, B.; Lengyelová, E.; Havrlentová, M.; Kraic, J.; Celec, P.; Tóthová, L. Anthocyanin-rich diet in chemically induced colitis in mice. Folia Biol. (Praha) 2015, 61, 104–109. [Google Scholar]
  133. Thoppil, R.J.; Bhatia, D.; Barnes, K.F.; Haznagy-Radnai, E.; Hohmann, J.; Darvesh, A.S.; Bishayee, A. Black currant anthocyanins abrogate oxidative stress through Nrf2-mediated antioxidant mechanisms in a rat model of hepatocellular carcinoma. Curr. Cancer Drug Targets 2012, 12, 1244–1257. [Google Scholar]
  134. Visconti, R.; Della Monica, R.; Grieco, D. Cell cycle checkpoint in cancer: A therapeutically targetable double-edged sword. J. Exp. Clin. Cancer Res. 2016, 35, 153. [Google Scholar] [CrossRef] [Green Version]
  135. Lee, H.; Kim, W.; Kang, H.G.; Kim, W.J.; Lee, S.C.; Kim, S.J. Geranium thunbergii extract-induced G1 phase cell cycle arrest and apoptosis in gastric cancer cells. Animal Cells Syst. 2020, 24, 26–33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Zhao, Y.; Hu, X.; Zuo, X.; Wang, M. Chemopreventive effects of some popular phytochemicals on human colon cancer: A review. Food Funct. 2018, 9, 4548–4568. [Google Scholar] [CrossRef] [PubMed]
  137. Tavakolian, S.; Goudarzi, H.; Faghihloo, E. Cyclin-dependent kinases and CDK inhibitors in virus-associated cancers. Infect. Agent. Cancer 2020, 15, 1–12. [Google Scholar] [CrossRef] [PubMed]
  138. Williams, G.H.; Stoeber, K. The cell cycle and cancer. J. Pathol. 2012, 226, 352–364. [Google Scholar] [CrossRef]
  139. Seeram, N.P. Berry fruits for cancer prevention: Current status and future prospects. J. Agric. Food Chem. 2008, 56, 630–635. [Google Scholar] [CrossRef]
  140. Kristo, A.S.; Klimis-Zacas, D.; Sikalidis, A.K. Protective role of dietary berries in cancer. Antioxidants 2016, 5, 37. [Google Scholar] [CrossRef] [Green Version]
  141. Lin, B.W.; Gong, C.C.; Song, H.F.; Cui, Y.Y. Effects of anthocyanins on the prevention and treatment of cancer. Br. J. Pharmacol. 2017, 174, 1226–1243. [Google Scholar] [CrossRef] [Green Version]
  142. Malik, M.; Zhao, C.; Schoene, N.; Guisti, M.M.; Moyer, M.P.; Magnuson, B.A. Anthocyanin-rich extract from Aronia meloncarpa E. induces a cell cycle block in colon cancer but not normal colonic cells. Nutr. Cancer 2003, 46, 186–196. [Google Scholar] [CrossRef]
  143. Qi, C.; Li, S.; Jia, Y.; Wang, L. Blueberry anthocyanins induce G2/M cell cycle arrest and apoptosis of oral cancer KB cells through down-regulation methylation of p53. Yi Chuan 2014, 36, 566–573. [Google Scholar]
  144. Hsu, C.-P.P.; Shih, Y.-T.T.; Lin, B.-R.R.; Chiu, C.-F.F.; Lin, C.-C.C. Inhibitory effect and mechanisms of an anthocyanins- and anthocyanidins-rich extract from purple-shoot tea on colorectal carcinoma cell proliferation. J. Agric. Food Chem. 2012, 60, 3686–3692. [Google Scholar] [CrossRef]
  145. Yun, J.-M.M.; Afaq, F.; Khan, N.; Mukhtar, H. Delphinidin, an anthocyanidin in pigmented fruits and vegetables, induces apoptosis and cell cycle arrest in human colon cancer HCT116 cells. Mol. Carcinog. 2009, 48, 260–270. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Zhao, C.; Giusti, M.M.; Malik, M.; Moyer, M.P.; Magnuson, B.A. Effects of commercial anthocyanin-rich on colonic cancer and nontumorigenic colonic cell growth. J. Agric. Food Chem. 2004, 52, 6122–6128. [Google Scholar] [CrossRef] [PubMed]
  147. Zhang, Y.; Vareed, S.K.; Nair, M.G. Human tumor cell growth inhibition by nontoxic anthocyanidins, the pigments in fruits and vegetables. Life Sci. 2005, 76, 1465–1472. [Google Scholar] [CrossRef]
  148. Chen, P.N.; Chu, S.C.; Chiou, H.L.; Chiang, C.L.; Yang, S.F.; Hsieh, Y.S. Cyanidin 3-glucoside and peonidin 3-glucoside inhibit tumor cell growth and induce apoptosis in vitro and suppress tumor growth in vivo. Nutr. Cancer 2005, 53, 232–243. [Google Scholar] [CrossRef] [PubMed]
  149. Blanco, A.; Blanco, G. Chapter 32—Apoptosis; Academic Press: Cambridge, MA, USA, 2017; pp. 791–796. [Google Scholar]
  150. Jin, Z.; El-Deiry, W.S. Overview of cell death signaling pathways. Cancer Biol. Ther. 2005, 4, 147–171. [Google Scholar] [CrossRef] [Green Version]
  151. Fernald, K.; Kurokawa, M. Evading apoptosis in cancer. Trends Cell Biol. 2013, 23, 620–633. [Google Scholar] [CrossRef] [Green Version]
  152. Baba, A.B.; Nivetha, R.; Chattopadhyay, I.; Nagini, S. Blueberry and malvidin inhibit cell cycle progression and induce mitochondrial-mediated apoptosis by abrogating the JAK/STAT-3 signalling pathway. Food Chem. Toxicol. 2017, 109, 534–543. [Google Scholar] [CrossRef]
  153. Mazewski, C.; Liang, K.; Gonzalez de Mejia, E. Comparison of the effect of chemical composition of anthocyanin-rich plant extracts on colon cancer cell proliferation and their potential mechanism of action using in vitro, in silico, and biochemical assays. Food Chem. 2018, 242, 378–388. [Google Scholar] [CrossRef]
  154. Shin, D.Y.; Ryu, C.H.; Lee, W.S.; Kim, D.C.; Kim, S.H.; Hah, Y.-S.S.; Lee, S.J.; Shin, S.C.; Kang, H.S.; Choi, Y.H. Induction of apoptosis and inhibition of invasion in human hepatoma cells by anthocyanins from meoru. Ann. N. Y. Acad. Sci. 2009, 1171, 137–148. [Google Scholar] [CrossRef]
  155. Jo, W.S.; Jeong, M.-H.; Jin, Y.-H.; Jang, J.Y.; Nam, B.H.; Son, S.H.; Choi, S.S.; Yoo, Y.H.; Kang, C.D.; Lee, J.D.; et al. Loss of mitochondrial membrane potential and caspase activation enhance apoptosis in irradiated K562 cells treated with herbimycin A. Int. J. Radiat. Biol. 2005, 81, 531–543. [Google Scholar] [CrossRef]
  156. Lazze, M.C.; Savio, M.; Pizzala, R.; Cazzalini, O.; Perucca, P.; Scovassi, A.I.; Stivala, L.A.; Bianchi, L.; Lazz, M.C.; Savio, M.; et al. Anthocyanins induce cell cycle perturbations and apoptosis in different human cell lines. Carcinogenesis 2004, 25, 1427–1433. [Google Scholar] [CrossRef] [PubMed]
  157. Shih, P.H.; Yeh, C.T.; Yen, G.C. Effects of anthocyanidin on the inhibition of proliferation and induction of apoptosis in human gastric adenocarcinoma cells. Food Chem. Toxicol. 2005, 43, 1557–1566. [Google Scholar] [CrossRef]
  158. Wu, Q.K.; Koponen, J.M.; Mykkänen, H.M.; Törrönen, A.R. Berry phenolic extracts modulate the expression of p21WAF1 and Bax but Not Bcl-2 in HT-29 colon cancer cells. J. Agric. Food Chem. 2007, 55, 1156–1163. [Google Scholar] [CrossRef] [PubMed]
  159. Sordet, O.; Khan, Q.A.; Kohn, K.W.; Pommier, Y. Apoptosis induced by topoisomerase inhibitors. Curr. Med. Chem. Anti-Cancer Agents 2003, 3, 271–290. [Google Scholar] [CrossRef] [PubMed]
  160. Webb, M.R.; Min, K.; Ebeler, S.E. Anthocyanin interactions with DNA: Interactions, topoisomerase I inhibition and oxidative reactions. J. Food Biochem. 2008, 32, 576–596. [Google Scholar] [CrossRef] [PubMed]
  161. Habermeyer, M.; Fritz, J.; Barthelmes, H.U.; Christensen, M.O.; Larsen, M.K.; Boege, F.; Marko, D. Anthocyanidins modulate the activity of human DNA topoisomerases I and II and affect cellular DNA integrity. Chem. Res. Toxicol. 2005, 18, 1395–1404. [Google Scholar] [CrossRef]
  162. Esselen, M.; Barth, S.W.; Winkler, S.; Baechler, S.; Briviba, K.; Watzl, B.; Skrbek, S.; Marko, D. Anthocyanins suppress the cleavable complex formation by irinotecan and diminish its DNA-strand-breaking activity in the colon of Wistar rats. Carcinogenesis 2013, 34, 835–840. [Google Scholar] [CrossRef] [Green Version]
  163. Esselen, M.; Fritz, J.; Hutter, M.; Teller, N.; Baechler, S.; Boettler, U.; Marczylo, T.H.; Gescher, A.J.; Marko, D. Anthocyanin-rich extracts suppress the DNA-damaging effects of topoisomerase poisons in human colon cancer cells. Mol. Nutr. Food Res. 2011, 55, 143–153. [Google Scholar] [CrossRef]
  164. Esselen, M.; Boettler, U.; Teller, N.; Bächler, S.; Hutter, M.; Rüfer, C.E.; Skrbek, S.; Marko, D. Anthocyanin-rich blackberry extract suppresses the DNA-damaging properties of topoisomerase i and II poisons in colon carcinoma cells. J. Agric. Food Chem. 2011, 59, 6966–6973. [Google Scholar] [CrossRef]
  165. Wroblewski, L.E.; Peek, R.M.; Coburn, L.A. The role of the microbiome in gastrointestinal cancer. Gastroenterol. Clin. N. Am. 2016, 45, 543–556. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Pan, P.; Lam, V.; Salzman, N.; Huang, Y.W.; Yu, J.; Zhang, J.; Wang, L.S. Black raspberries and their anthocyanin and fiber fractions alter the composition and diversity of gut microbiota in F-344 rats. Nutr. Cancer 2017, 69, 943–951. [Google Scholar] [CrossRef] [PubMed]
  167. Liu, F.; Wang, T.T.Y.Y.; Tang, Q.; Xue, C.; Li, R.W.; Wu, V.C.H.H. Malvidin 3-glucoside modulated gut microbial dysbiosis and global metabolome disrupted in a murine colitis model induced by dextran sulfate sodium. Mol. Nutr. Food Res. 2019, 63, 1900455. [Google Scholar] [CrossRef] [PubMed]
  168. Cheng, Z.; Lin, J.; Gao, N.; Sun, X.; Meng, X.; Liu, R.; Liu, Y.; Wang, W.; Wang, Y.; Li, B. Blueberry malvidin-3-galactoside modulated gut microbial dysbiosis and microbial TCA cycle KEGG pathway disrupted in a liver cancer model induced by HepG2 cells. Food Sci. Hum. Wellness 2020, in press. [Google Scholar] [CrossRef]
  169. Khairnar, M.R.; Wadgave, U.; Jadhav, H.; Naik, R. Anticancer activity of chlorhexidine and cranberry extract: An in-vitro study. J. Exp. Ther. Oncol. 2018, 12, 201–205. [Google Scholar]
  170. Fan, M.J.; Wang, I.C.; Hsiao, Y.T.; Lin, H.Y.; Tang, N.Y.; Hung, T.C.; Quan, C.; Lien, J.C.; Chung, J.G. Anthocyanins from black rice (Oryza sativa L.) demonstrate antimetastatic properties by reducing MMPs and NF-B expressions in human oral cancer CAL 27 cells. Nutr. Cancer 2015, 67, 327–338. [Google Scholar] [CrossRef]
  171. Yue, E.; Tuguzbaeva, G.; Chen, X.; Qin, Y.; Li, A.; Sun, X.; Dong, C.; Liu, Y.; Yu, Y.; Zahra, S.M.; et al. Anthocyanin is involved in the activation of pyroptosis in oral squamous cell carcinoma. Phytomedicine 2019, 56, 286–294. [Google Scholar] [CrossRef]
  172. De Moura, C.F.G.; Soares, G.R.; Ribeiro, F.A.P.; Silva, M.J.D.; Vilegas, W.; Santamarina, A.B.; Pisani, L.P.; Estadella, D.; Ribeiro, D.A. Evaluation of the chemopreventive activity of grape skin extract using medium-term oral carcinogenesis assay induced by 4-nitroquinoline 1-oxide. Anticancer Res. 2019, 39, 177–182. [Google Scholar] [CrossRef]
  173. Casto, B.C.; Knobloch, T.J.; Galioto, R.L.; Yu, Z.; Accurso, B.T.; Warner, B.M. Chemoprevention of oral cancer by lyophilized strawberries. Anticancer Res. 2013, 33, 4757–4766. [Google Scholar]
  174. Medda, R.; Lyros, O.; Schmidt, J.L.; Jovanovic, N.; Nie, L.; Link, B.J.; Otterson, M.F.; Stoner, G.D.; Shaker, R.; Rafiee, P. Anti-inflammatory and anti angiogenic effect of black raspberry extract on human esophageal and intestinal microvascular endothelial cells. Microvasc. Res. 2015, 97, 167–180. [Google Scholar] [CrossRef] [Green Version]
  175. Zikri, N.N.; Riedl, K.M.; Wang, L.S.; Lechner, J.; Schwartz, S.J.; Stoner, G.D. Black raspberry components inhibit proliferation, induce apoptosis, and modulate gene expression in rat esophageal epithelial cells. Nutr. Cancer 2009, 61, 816–826. [Google Scholar] [CrossRef] [PubMed]
  176. Aiyer, H.S.; Li, Y.; Losso, J.N.; Gao, C.; Schiffman, S.C.; Slone, S.P.; Martin, R.C.G. Effect of freeze-dried berries on the development of reflux-induced esophageal adenocarcinoma. Nutr. Cancer 2011, 63, 1256–1262. [Google Scholar] [CrossRef] [PubMed]
  177. Seguin, C.M.; Wang, L.S.; Stoner, G.D. Chemopreventive effects of berries and berry components in the rodent esophagus. In Berries and Cancer Prevention; Springer: New York, NY, USA, 2011; pp. 143–161. [Google Scholar]
  178. Wang, L.S.; Hecht, S.S.; Carmella, S.G.; Yu, N.; Larue, B.; Henry, C.; McIntyre, C.; Rocha, C.; Lechner, J.F.; Stoner, G.D. Anthocyanins in black raspberries prevent esophageal tumors in rats. Cancer Prev. Res. 2009, 2, 84–93. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Stoner, G.D.; Wang, L.S.; Seguin, C.; Rocha, C.; Stoner, K.; Chiu, S.; Kinghorn, A.D. Multiple berry types prevent N-nitrosomethylbenzylamine-induced esophageal cancer in rats. Pharm. Res. 2010, 27, 1138–1145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  180. Peiffer, D.S.; Wang, L.S.; Zimmerman, N.P.; Ransom, B.W.S.; Carmella, S.G.; Kuo, C.T.; Chen, J.H.; Oshima, K.; Huang, Y.W.; Hecht, S.S.; et al. Dietary consumption of black raspberries or their anthocyanin constituents alters innate immune cell trafficking in esophageal cancer. Cancer Immunol. Res. 2016, 4, 72–82. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  181. Kim, J.M.; Kim, K.M.; Park, E.H.; Seo, J.H.; Song, J.Y.; Shin, S.C.; Kang, H.L.; Lee, W.K.; Cho, M.J.; Rhee, K.H.; et al. Anthocyanins from black soybean inhibit Helicobacter pylori-induced inflammation in human gastric epithelial AGS cells. Microbiol. Immunol. 2013, 57, 366–373. [Google Scholar] [CrossRef]
  182. Wang, Y.; Lin, J.; Tian, J.; Si, X.; Jiao, X.; Zhang, W.; Gong, E.; Li, B. Blueberry malvidin-3-galactoside suppresses hepatocellular carcinoma by regulating apoptosis, proliferation, and metastasis pathways in vivo and in vitro. J. Agric. Food Chem. 2019, 67, 625–636. [Google Scholar] [CrossRef]
  183. Shin, D.Y.; Lee, W.S.; Kim, S.H.; Kim, M.J.; Yun, J.W.; Lu, J.N.; Lee, S.J.; Tsoy, I.; Kim, H.J.; Ryu, C.H.; et al. Anti-invasive activity of anthocyanins isolated from Vitis coignetiae in human hepatocarcinoma cells. J. Med. Food 2009, 12, 967–972. [Google Scholar] [CrossRef]
  184. Savio, M.; Stivala, L.A.; Prosperi, E. Anthocyanins protect against DNA damage induced by tert-butyl-hydroperoxide in rat smooth muscle and hepatoma cells. Mutat. Res. 2003, 535, 103–115. [Google Scholar]
  185. Longo, L.; Platini, F.; Scardino, A.; Alabiso, O.; Vasapollo, G.; Tessitore, L. Autophagy inhibition enhances anthocyanin-induced apoptosis in hepatocellular carcinoma. Mol. Cancer Ther. 2008, 7, 2476–2485. [Google Scholar] [CrossRef] [Green Version]
  186. Yeh, C.T.; Yen, G.C. Induction of apoptosis by the anthocyanidins through regulation of Bcl-2 gene and activation of c-Jun N-terminal kinase cascade in hepatoma cells. J. Agric. Food Chem. 2005, 53, 1740–1749. [Google Scholar] [CrossRef] [PubMed]
  187. Bishayee, A.; Háznagy-Radnai, E.; Mbimba, T.; Sipos, P.; Morazzoni, P.; Darvesh, A.S.; Bhatia, D.; Hohmann, J. Anthocyanin-rich black currant extract suppresses the growth of human hepatocellular carcinoma cells. Nat. Prod. Commun. 2010, 5, 1613–1618. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  188. Anwar, S.; Fratantonio, D.; Ferrari, D.; Saija, A.; Cimino, F.; Speciale, A. Berry anthocyanins reduce proliferation of human colorectal carcinoma cells by inducing caspase-3 activation and p21 upregulation. Mol. Med. Rep. 2016, 14, 1397–1403. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  189. Yi, W.; Fischer, J.; Krewer, G.; Akoh, C.C. Phenolic compounds from blueberries can inhibit colon cancer cell proliferation and induce apoptosis. J. Agric. Food Chem. 2005, 53, 7320–7329. [Google Scholar] [CrossRef]
  190. Harris, G.K.; Gupta, A.; Nines, R.G.; Kresty, L.A.; Habib, S.G.; Frankel, W.L.; LaPerle, K.; Gallaher, D.D.; Schwartz, S.J.; Stoner, G.D. Effects of lyophilized black raspberries on azoxymethane-induced colon cancer and 8-hydroxy-2′-deoxyguanosine levels in the fischer 344 rat. Nutr. Cancer 2001, 40, 125–133. [Google Scholar] [CrossRef]
  191. Asadi, K.; Ferguson, L.R.; Philpott, M.; Karunasinghe, N. Cancer-preventive properties of an anthocyanin-enriched sweet potato in the APC Min mouse model. J. Cancer Prev. 2017, 22, 135–146. [Google Scholar] [CrossRef] [Green Version]
  192. Pan, P.; Skaer, C.W.; Wang, H.-T.; Stirdivant, S.M.; Young, M.R.; Oshima, K.; Stoner, G.D.; Lechner, J.F.; Huang, Y.-W.; Wang, L.-S. Black raspberries suppress colonic adenoma development in Apc Min/+ mice: Relation to metabolite profiles. Carcinogenesis 2015, 36, 1245–1253. [Google Scholar] [CrossRef] [Green Version]
  193. Lim, S.; Xu, J.; Kim, J.; Chen, T.Y.Y.; Su, X.; Standard, J.; Carey, E.; Griffin, J.; Herndon, B.; Katz, B.; et al. Role of anthocyanin-enriched purple-fleshed sweet potato p40 in colorectal cancer prevention. Mol. Nutr. Food Res. 2013, 57, 1908–1917. [Google Scholar] [CrossRef] [Green Version]
  194. Madiwale, G.P.; Reddivari, L.; Holm, D.G.; Vanamala, J. Storage elevates phenolic content and antioxidant activity but suppresses antiproliferative and pro-apoptotic properties of colored-flesh potatoes against human colon cancer cell lines. J. Agric. Food Chem. 2011, 59, 8155–8166. [Google Scholar] [CrossRef]
  195. Lee, D.Y.; Yun, S.M.; Song, M.Y.; Jung, K.; Kim, E.H. Cyanidin chloride induces apoptosis by inhibiting NF-κB signaling through activation of Nrf2 in colorectal cancer cells. Antioxidants 2020, 9, 285. [Google Scholar] [CrossRef] [Green Version]
  196. Wang, L.S.; Kuo, C.T.; Cho, S.J.; Seguin, C.; Siddiqui, J.; Stoner, K.; Yu, I.W.; Huang, T.H.M.; Tichelaar, J.; Yearsley, M.; et al. Black raspberry-derived anthocyanins demethylate tumor suppressor genes through the inhibition of DNMT1 and DNMT3B in colon cancer cells. Nutr. Cancer 2013, 65, 118–125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Dai, J.; Patel, J.D.; Mumper, R.J. Characterization of blackberry extract and its antiproliferative and anti-inflammatory properties. J. Med. Food 2007, 10, 258–265. [Google Scholar] [CrossRef] [PubMed]
  198. Lala, G.; Malik, M.; Zhao, C.; He, J.; Kwon, Y.; Giusti, M.M.; Magnuson, B.A. Anthocyanin-rich extracts inhibit multiple biomarkers of colon cancer in rats. Nutr. Cancer 2006, 54, 84–93. [Google Scholar] [CrossRef] [PubMed]
  199. Lippert, E.; Ruemmele, P.; Obermeier, F.; Goelder, S.; Kunst, C.; Rogler, G.; Dunger, N.; Messmann, H.; Hartmann, A.; Endlicher, E. Anthocyanins prevent colorectal cancer development in a mouse model. Digestion 2017, 95, 275–280. [Google Scholar] [CrossRef] [Green Version]
  200. Rivera, C. Essentials of oral cancer. Int. J. Clin. Exp. Pathol. 2015, 8, 11884–11894. [Google Scholar]
  201. Ugalde, C.M.; Liu, Z.; Ren, C.; Chan, K.K.; Rodrigo, K.A.; Ling, Y.; Larsen, P.E.; Chacon, G.E.; Stoner, G.D.; Mumper, R.J.; et al. Distribution of anthocyanins delivered from a bioadhesive black raspberry gel following topical intraoral application in normal healthy volunteers. Pharm. Res. 2009, 26, 977–986. [Google Scholar] [CrossRef] [Green Version]
  202. Mallery, S.R.; Stoner, G.D.; Larsen, P.E.; Fields, H.W.; Rodrigo, K.A.; Schwartz, S.J.; Tian, Q.; Dai, J.; Mumper, R.J. Formulation and in-vitro and in-vivo evaluation of a mucoadhesive gel containing freeze dried black raspberries: Implications for oral cancer chemoprevention. Pharm. Res. 2007, 24, 728–737. [Google Scholar] [CrossRef]
  203. Cui, L.; Liu, X.; Tian, Y.; Xie, C.; Li, Q.; Cui, H.; Sun, C. Flavonoids, flavonoid subclasses, and esophageal cancer risk: A meta-analysis of epidemiologic studies. Nutrients 2016, 8, 350. [Google Scholar] [CrossRef]
  204. Zhang, Y. Epidemiology of esophageal cancer. World J. Gastroenterol. 2013, 19, 5598–5606. [Google Scholar] [CrossRef]
  205. Reen, R.K.; Nines, R.; Stoner, G.D. Modulation of N-nitrosomethylbenzylamine metabolism by black raspberries in the esophagus and liver of Fischer 344 rats. Nutr Cancer 2006, 54, 47–57. [Google Scholar] [CrossRef] [Green Version]
  206. Stoner, G.D.; Chen, T.; Kresty, L.A.; Aziz, R.M.; Reinemann, T.; Nines, R. Protection against esophageal cancer in rodents with lyophilized berries: Potential mechanisms. Nutr Cancer 2006, 54, 33–46. [Google Scholar] [CrossRef] [Green Version]
  207. Peiffer, D.S.; Zimmerman, N.P.; Wang, L.S.; Ransom, B.W.S.; Carmella, S.G.; Kuo, C.T.; Siddiqui, J.; Chen, J.H.; Oshima, K.; Huang, Y.W.; et al. Chemoprevention of esophageal cancer with black raspberries, their component anthocyanins, and a major anthocyanin metabolite, protocatechuic acid. Cancer Prev. Res. 2014, 7, 574–584. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  208. Lu, Z.; Lu, C.; Li, C.; Jiao, Y.; Li, Y.; Zhang, G. Dracorhodin perchlorate induces apoptosis and G2/M cell cycle arrest in human esophageal squamous cell carcinoma through inhibition of the JAK2/STAT3 and AKT/FOXO3a pathways. Mol. Med. Rep. 2019, 20, 2091–2100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  209. Shi, N.; Riedl, K.M.; Schwartz, S.J.; Zhang, X.; Clinton, S.K.; Chen, T. Efficacy comparison of lyophilised black raspberries and combination of celecoxib and PBIT in prevention of carcinogen-induced oesophageal cancer in rats. J. Funct. Foods 2016, 27, 84–94. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  210. Cheng, X.J.; Lin, J.C.; Tu, S.P. Etiology and prevention of gastric cancer. Gastrointest. Tumors 2016, 3, 25–36. [Google Scholar] [CrossRef] [PubMed]
  211. Yang, D.Y.; Wang, X.; Yuan, W.J.; Chen, Z.H. Intake of anthocyanins and gastric cancer risk: A comprehensive meta-analysis on cohort and case-control studies. J. Nutr. Sci. Vitaminol. 2019, 65, 72–81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  212. Woo, H.D.; Lee, J.; Choi, I.J.; Kim, C.G.; Lee, J.Y.; Kwon, O.; Kim, J. Dietary flavonoids and gastric cancer risk in a Korean population. Nutrients 2014, 6, 4961–4973. [Google Scholar] [CrossRef] [Green Version]
  213. Kim, S.H.; Lee, M.H.; Park, M.; Woo, H.J.; Kim, Y.S.; Tharmalingam, N.; Seo, W.D.; Kim, J.B. Regulatory effects of black rice extract on Helicobacter pylori infection-induced apoptosis. Mol. Nutr. Food Res. 2018, 62, 1700586. [Google Scholar] [CrossRef]
  214. Zhang, L.; Zhou, J.; Luo, J.; Wang, Q.; Liu, J.; Zeng, Q. Study on mulberry anthocyanins induced autophagy and apoptosis of human gastric cancer SGC-7901 cell autophagy. Zhong Yao Cai 2016, 39, 1134–1138. [Google Scholar]
  215. Lu, J.N.; Lee, W.S.; Nagappan, A.; Chang, S.-H.; Choi, Y.H.; Kim, H.J.; Kim, G.S.; Ryu, C.H.; Shin, S.C.; Jung, J.-M.; et al. Anthocyanins from the fruit of Vitis coignetiae Pulliat potentiate the cisplatin activity by inhibiting PI3K/Akt signaling pathways in human gastric cancer cells. J. Cancer Prev. 2015, 20, 50–56. [Google Scholar] [CrossRef] [Green Version]
  216. Savitha, G.; Vishnupriya, V.; Krishnamohan, S. Hepatocellular carcinoma—A review. J. Pharm. Sci. Res. 2017, 9, 1276–1280. [Google Scholar]
  217. Liu, C.Y.; Chen, K.F.; Chen, P.J. Treatment of liver cancer. Cold Spring Harb. Perspect. Med. 2015, 5, a021535. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  218. Zhou, L.; Wang, H.; Yi, J.; Yang, B.; Li, M.; He, D.; Yang, W.; Zhang, Y.; Ni, H. Anti-tumor properties of anthocyanins from Lonicera caerulea ‘Beilei’ fruit on human hepatocellular carcinoma: In vitro and in vivo study. Biomed. Pharmacother. 2018, 104, 520–529. [Google Scholar] [CrossRef] [PubMed]
  219. Bishayee, A.; Thoppil, R.J.; Mandal, A.; Darvesh, A.S.; Ohanyan, V.; Meszaros, J.G.; Háznagy-Radnai, E.; Hohmann, J.; Bhatia, D. Black currant phytoconstituents exert chemoprevention of diethylnitrosamine-initiated hepatocarcinogenesis by suppression of the inflammatory response. Mol. Carcinog. 2013, 52, 304–317. [Google Scholar] [CrossRef]
  220. Suwannakul, N.; Punvittayagul, C.; Jarukamjorn, K.; Wongpoomchai, R. Purple rice bran extract attenuates the aflatoxin B1-induced initiation stage of hepatocarcinogenesis by alteration of xenobiotic metabolizing enzymes. Asian Pac. J. Cancer Prev. 2015, 16, 3371–3376. [Google Scholar] [CrossRef] [Green Version]
  221. Marley, A.R.; Nan, H. Epidemiology of colorectal cancer. Int. J. Mol. Epidemiol. Genet. 2016, 7, 105–114. [Google Scholar]
  222. Moghimi-Dehkordi, B. An overview of colorectal cancer survival rates and prognosis in Asia. World J. Gastrointest. Oncol. 2012, 4, 71. [Google Scholar] [CrossRef]
  223. Kuipers, E.J.; Grady, W.M.; Lieberman, D.; Seufferlein, T.; Sung, J.J.; Boelens, P.G.; Van De Velde, C.J.H.; Watanabe, T. Colorectal cancer. Nat. Rev. Dis. Prim. 2015, 1, 15065. [Google Scholar] [CrossRef] [Green Version]
  224. Medic, N.; Tramer, F.; Passamonti, S. Anthocyanins in colorectal cancer prevention. A systematic review of the literature in search of molecular oncotargets. Front. Pharmacol. 2019, 10, 675. [Google Scholar] [CrossRef]
  225. Ding, L.; Lan, Z.; Xiong, X.; Ao, H.; Feng, Y.; Gu, H.; Yu, M.; Cui, Q. The dual role of microRNAs in colorectal cancer progression. Int. J. Mol. Sci. 2018, 19, 2791. [Google Scholar] [CrossRef] [Green Version]
  226. Zhang, H.; Guo, J.; Mao, L.; Li, Q.; Guo, M.; Mu, T.; Zhang, Q.; Bi, X. Up-regulation of MIR-24-1-5p is involved in the chemoprevention of colorectal cancer by black raspberry anthocyanins. Br. J. Nutr. 2019, 122, 518–526. [Google Scholar] [CrossRef] [PubMed]
  227. Guo, J.; Yang, Z.; Zhou, H.; Yue, J.; Mu, T.; Zhang, Q.; Bi, X. Upregulation of DKK3 by miR-483-3p plays an important role in the chemoprevention of colorectal cancer mediated by black raspberry anthocyanins. Mol. Carcinog. 2020, 59, 168–178. [Google Scholar] [CrossRef] [PubMed]
  228. Coates, E.M.; Popa, G.; Gill, C.I.R.; McCann, M.J.; McDougall, G.J.; Stewart, D.; Rowland, I. Colon-available raspberry polyphenols exhibit anti-cancer effects on in vitro models of colon cancer. J. Carcinog. 2007, 6, 1477–3143. [Google Scholar] [CrossRef] [PubMed]
  229. Zhou, Y.; Xia, L.; Wang, H.; Oyang, L.; Su, M.; Liu, Q.; Lin, J.; Tan, S.; Tian, Y.; Liao, Q.; et al. Cancer stem cells in progression of colorectal cancer. Oncotarget 2018, 9, 33403–33415. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  230. Charepalli, V.; Reddivari, L.; Radhakrishnan, S.; Vadde, R.; Agarwal, R.; Vanamala, J.K.P.P. Anthocyanin-containing purple-fleshed potatoes suppress colon tumorigenesis via elimination of colon cancer stem cells. J. Nutr. Biochem. 2015, 26, 1641–1649. [Google Scholar] [CrossRef]
  231. Shin, D.Y.; Lu, J.N.; Kim, G.Y.; Jung, J.M.; Kang, H.S.; Lee, W.S.; Choi, Y.H. Anti-invasive activities of anthocyanins through modulation of tight junctions and suppression of matrix metalloproteinase activities in HCT-116 human colon carcinoma cells. Oncol. Rep. 2011, 25, 567–572. [Google Scholar]
  232. Jing, P.; Bomser, J.A.; Schwartz, S.J.; He, J.; Magnuson, B.A.; Giusti, M.M. Structure-function relationships of anthocyanins from various anthocyanin-rich extracts on the inhibition of colon cancer cell growth. J. Agric. Food Chem. 2008, 56, 9391–9398. [Google Scholar] [CrossRef]
  233. Vishnu, V.R.; Renjith, R.S.; Mukherjee, A.; Anil, S.R.; Sreekumar, J.; Jyothi, A.N. Comparative study on the chemical structure and in vitro antiproliferative activity of anthocyanins in purple root tubers and leaves of sweet potato (Ipomoea batatas). J. Agric. Food Chem. 2019, 67, 2467–2475. [Google Scholar] [CrossRef]
  234. Wang, L.; Liu, F.; Liu, Y.; Gao, H.; Dong, M. Cyanidin-3-O-glucoside inhibits proliferation of colorectal cancer cells by targeting TOPK. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi 2019, 35, 1101–1108. [Google Scholar]
  235. Mazewski, C.; Kim, M.S.; Gonzalez de Mejia, E. Anthocyanins, delphinidin-3-O-glucoside and cyanidin-3-O-glucoside, inhibit immune checkpoints in human colorectal cancer cells in vitro and in silico. Sci. Rep. 2019, 9, 11560. [Google Scholar] [CrossRef] [Green Version]
  236. Duthie, S.J.; Gardner, P.T.; Morrice, P.C.; Wood, S.G.; Pirie, L.; Bestwick, C.C.; Milne, L.; Duthie, G.G. DNA stability and lipid peroxidation in vitamin E-deficient rats in vivo and colon cells in vitro: Modulation by the dietary anthocyanin, cyanidin-3-glycoside. Eur. J. Nutr. 2005, 44, 195–203. [Google Scholar] [CrossRef] [PubMed]
  237. Pervin, M.; Hasnat, M.A.; Lim, J.H.; Lee, Y.M.; Kim, E.O.; Um, B.H.; Lim, B.O. Preventive and therapeutic effects of blueberry (Vaccinium corymbosum) extract against DSS-induced ulcerative colitis by regulation of antioxidant and inflammatory mediators. J. Nutr. Biochem. 2016, 28, 103–113. [Google Scholar] [CrossRef] [PubMed]
  238. Bibi, S.; Du, M.; Zhu, M.J. Dietary red raspberry reduces colorectal inflammation and carcinogenic risk in mice with dextran sulfate sodium-induced colitis. J. Nutr. 2018, 148, 667–674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  239. De Robertis, M.; Massi, E.; Poeta, M.; Carotti, S.; Morini, S.; Cecchetelli, L.; Signori, E.; Fazio, V. The AOM/DSS murine model for the study of colon carcinogenesis: From pathways to diagnosis and therapy studies. J. Carcinog. 2011, 10, 1477–3163. [Google Scholar]
  240. Shi, N.; Clinton, S.K.; Liu, Z.; Wang, Y.; Riedl, K.M.; Schwartz, S.J.; Zhang, X.; Pan, Z.; Chen, T. Strawberry phytochemicals inhibit azoxymethane/dextran sodium sulfate-induced colorectal carcinogenesis in Crj: CD-1 mice. Nutrients 2015, 7, 1696–1715. [Google Scholar] [CrossRef] [Green Version]
  241. Zhou, G.; Chen, L.; Sun, Q.; Mo, Q.G.; Sun, W.C.; Wang, Y.W. Maqui berry exhibited therapeutic effects against DSS-induced ulcerative colitis in C57BL/6 mice. Food Funct. 2019, 10, 6655–6665. [Google Scholar] [CrossRef]
  242. Zhang, W.; Xu, L.; Cho, S.Y.; Min, K.J.; Oda, T.; Zhang, L.J.; Yu, Q.; Jin, J.O. Ginseng berry extract attenuates dextran sodium sulfate-induced acute and chronic colitis. Nutrients 2016, 8, 199. [Google Scholar] [CrossRef] [Green Version]
  243. Cai, X.; Han, Y.; Gu, M.; Song, M.; Wu, X.; Li, Z.; Li, F.; Goulette, T.; Xiao, H. Dietary cranberry suppressed colonic inflammation and alleviated gut microbiota dysbiosis in dextran sodium sulfate-treated mice. Food Funct. 2019, 10, 6331–6341. [Google Scholar] [CrossRef]
  244. Piberger, H.; Oehme, A.; Hofmann, C.; Dreiseitel, A.; Sand, P.G.; Obermeier, F.; Schoelmerich, J.; Schreier, P.; Krammer, G.; Rogler, G. Bilberries and their anthocyanins ameliorate experimental colitis. Mol. Nutr. Food Res. 2011, 55, 1724–1729. [Google Scholar] [CrossRef]
  245. Xia, Y.; Tian, L.M.; Liu, Y.; Guo, K.S.; Lv, M.; Li, Q.T.; Hao, S.Y.; Ma, C.H.; Chen, Y.X.; Tanaka, M.; et al. Low Dose of cyanidin-3-O-glucoside alleviated dextran sulfate sodium-induced colitis, mediated by CD169+ macrophage pathway. Inflamm. Bowel Dis. 2019, 25, 1510–1521. [Google Scholar] [CrossRef]
  246. Akiyama, S.; Nesumi, A.; Maeda-Yamamoto, M.; Uehara, M.; Murakami, A. Effects of anthocyanin-rich tea “Sunrouge” on dextran sodium sulfate-induced colitis in mice. BioFactors 2012, 38, 226–233. [Google Scholar] [CrossRef] [PubMed]
  247. Kim, Y.J.; Ju, J.; Song, J.L.; Yang, S.G.; Park, K.Y. Anti-colitic effect of purple carrot on dextran sulfate sodium (DSS)-induced colitis in C57BL/6J Mice. Prev. Nutr. Food Sci. 2018, 23, 77–83. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  248. Mallery, S.R.; Tong, M.; Shumway, B.S.; Curran, A.E.; Larsen, P.E.; Ness, G.M.; Kennedy, K.S.; Blakey, G.H.; Kushner, G.M.; Vickers, A.M.; et al. Topical application of a mucoadhesive freeze-dried black raspberry gel induces clinical and histologic regression and reduces loss of heterozygosity events in premalignant oral intraepithelial lesions: Results from a multicentered, placebo-controlled clin. Clin. Cancer Res. 2014, 20, 1910–1924. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  249. Shumway, B.S.; Kresty, L.A.; Larsen, P.E.; Zwick, J.C.; Lu, B.; Fields, H.W.; Mumper, R.J.; Stoner, G.D.; Mallery, S.R. Effects of a topically applied bioadhesive berry gel on loss of heterozygosity indices in premalignant oral lesions. Clin. Cancer Res. 2008, 14, 2421–2430. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  250. Mallery, S.R.; Zwick, J.C.; Pei, P.; Tong, M.; Larsen, P.E.; Shumway, B.S.; Lu, B.; Fields, H.W.; Mumper, R.J.; Stoner, G.D. Topical application of a bioadhesive black raspberry gel modulates gene expression and reduces cyclooxygenase 2 protein in human premalignant oral lesions. Cancer Res. 2008, 68, 4945–4957. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  251. Kresty, L.A.; Fromkes, J.J.; Frankel, W.L.; Hammond, C.D.; Seeram, N.P.; Baird, M.; Stoner, G.D. A phase I pilot study evaluating the beneficial effects of black raspberries in patients with Barrett’s esophagus. Oncotarget 2018, 9, 35356–35372. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  252. Sun, L.; Subar, A.F.; Bosire, C.; Dawsey, S.M.; Kahle, L.L.; Zimmerman, T.P.; Abnet, C.C.; Heller, R.; Graubard, B.I.; Cook, M.B.; et al. Dietary flavonoid intake reduces the risk of head and neck but not esophageal or gastric cancer in US men and women. J. Nutr. 2017, 147, 1729–1738. [Google Scholar]
  253. Pan, P.; Skaer, C.W.; Stirdivant, S.M.; Young, M.R.; Stoner, G.D.; Lechner, J.F.; Huang, Y.W.; Wang, L.S. Beneficial regulation of metabolic profiles by black raspberries in human colorectal cancer patients. Cancer Prev. Res. 2015, 8, 743–750. [Google Scholar] [CrossRef] [Green Version]
  254. Wajed, S.A.; Laird, P.W.; DeMeester, T.R. DNA methylation: An alternative pathway to cancer. Ann. Surg. 2001, 234, 10–20. [Google Scholar] [CrossRef]
  255. Yamaguchi, M.; Okamura, S.; Yamaji, T.; Iwasaki, M.; Tsugane, S.; Shetty, V.; Koizumi, T. Plasma cytokine levels and the presence of colorectal cancer. PLoS ONE 2019, 14, e0213602. [Google Scholar] [CrossRef] [Green Version]
  256. Mentor-Marcel, R.A.; Bobe, G.; Sardo, C.; Wang, L.S.; Kuo, C.T.; Stoner, G.; Colburn, N.H. Plasma cytokines as potential response indicators to dietary freeze-dried black raspberries in colorectal cancer patients. Nutr. Cancer 2012, 64, 820–825. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  257. Wang, L.S.; Burke, C.A.; Hasson, H.; Kuo, C.T.; Molmenti, C.L.S.; Seguin, C.; Liu, P.; Huang, T.H.M.; Frankel, W.L.; Stoner, G.D. A phase Ib study of the effects of black raspberries on rectal polyps in patients with familial adenomatous polyposis. Cancer Prev. Res. 2014, 7, 666–674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  258. Molan, A.-L.; Liu, Z.; Plimmer, G. Evaluation of the effect of blackcurrant products on gut microbiota and on markers of risk for colon cancer in humans. Phyther. Res. 2014, 28, 416–422. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Two-hit theory of the initiation of hereditary and non-hereditary cancer. People with a hereditary susceptibility to GI cancers harbor an inherited genetic mutation on one of the chromosomes at the time of conception and receive the 1st somatic mutation due to the endogenous (e.g., chronic inflammation) or exogenous (e.g., exposure to carcinogens) rare events which in turn inactivate the full function of the respective gene and initiate neoplastic transformation. Non-inherited forms of GI cancer occur by acquiring two somatic mutations in later life, resulting in the inactivation of a gene leading to the initiation of malignancy.
Figure 1. Two-hit theory of the initiation of hereditary and non-hereditary cancer. People with a hereditary susceptibility to GI cancers harbor an inherited genetic mutation on one of the chromosomes at the time of conception and receive the 1st somatic mutation due to the endogenous (e.g., chronic inflammation) or exogenous (e.g., exposure to carcinogens) rare events which in turn inactivate the full function of the respective gene and initiate neoplastic transformation. Non-inherited forms of GI cancer occur by acquiring two somatic mutations in later life, resulting in the inactivation of a gene leading to the initiation of malignancy.
Ijms 21 06555 g001
Figure 2. Major anthocyanins derived from the basic anthocyanin structure. Based on the changes in R1 and R2 chemical groups, six major anthocyanins have been identified.
Figure 2. Major anthocyanins derived from the basic anthocyanin structure. Based on the changes in R1 and R2 chemical groups, six major anthocyanins have been identified.
Ijms 21 06555 g002
Figure 3. The possible anticarcinogenic mechanisms of anthocyanins in GI cancer prevention. Anthocyanins inhibit the pro-inflammatory COX-2 and NF-κB pathways and inhibit cell proliferation via reducing the nuclear translocation of β-catenin, upregulating cyclin-dependent kinase inhibitors, and downregulating cyclin proteins. Anthocyanins reduce the degradation of components of the extracellular matrix by suppressing the activity of MMPs and tight junction (TJ) proteins. Anthocyanins act as topoisomerase inhibitors and stimulate the DNA strand break response, leading to apoptosis. Anthocyanins induce apoptosis via the mitochondrial pathway and activation of caspase-9. Anthocyanins modulate gut microbial dysbiosis, hence reducing the production of ROS in macrophages and suppressing chronic inflammation. P21, P27, P53, cyclin-dependent kinase inhibitors; COX-2, cyclooxygenase-2; NF-κB, nuclear factor κ-light-chain-enhancer of activated B cells; MMP-2 and 9, matrix metalloproteinases 2 and 9; ROS, reactive oxygen species. Blue upwards arrow, promote; orange downwards arrow, inhibit.
Figure 3. The possible anticarcinogenic mechanisms of anthocyanins in GI cancer prevention. Anthocyanins inhibit the pro-inflammatory COX-2 and NF-κB pathways and inhibit cell proliferation via reducing the nuclear translocation of β-catenin, upregulating cyclin-dependent kinase inhibitors, and downregulating cyclin proteins. Anthocyanins reduce the degradation of components of the extracellular matrix by suppressing the activity of MMPs and tight junction (TJ) proteins. Anthocyanins act as topoisomerase inhibitors and stimulate the DNA strand break response, leading to apoptosis. Anthocyanins induce apoptosis via the mitochondrial pathway and activation of caspase-9. Anthocyanins modulate gut microbial dysbiosis, hence reducing the production of ROS in macrophages and suppressing chronic inflammation. P21, P27, P53, cyclin-dependent kinase inhibitors; COX-2, cyclooxygenase-2; NF-κB, nuclear factor κ-light-chain-enhancer of activated B cells; MMP-2 and 9, matrix metalloproteinases 2 and 9; ROS, reactive oxygen species. Blue upwards arrow, promote; orange downwards arrow, inhibit.
Ijms 21 06555 g003
Table 1. Hereditary basis of GI cancers.
Table 1. Hereditary basis of GI cancers.
Type of the CancerSyndromeAssociated Germline MutationsReference
EsophagealFamilial Barrett’s esophagus, Familial esophageal adenocarcinomaMSR1, ASCC1 and CTHRC1 [29]
Tylosis with esophageal cancer-squamous cell carcinomaRHBDF2 [30]
GastricDiffuse hereditary gastric cancer-adenocarcinomaCDH1 (E-cadherin) [31]
PancreaticHereditary pancreatitisPRSS1, CFTR, SPINK1, CTRC[32]
Hereditary breast and ovarian cancerBRCA1/2
Peutz-Jeghers syndromeSTK11/LKB1
Familial atypical multiple
mole melanoma syndrome
CDKN2A/p16
Familial adenomatous polyposisAPC
ColorectalFamilial adenomatous polyposisAPC [33,34]
Lynch syndromeEPCAM, MLH1, MSH2, MSH6, PMS2
MYH associated polyposisMUTYH
Hamartomatous polyposis syndromePeutz-Jeghers syndromeSTK11
Juvenile polyposis syndromeSMAD4, BMPR1A
Attenuated Familial adenomatous polyposisAPC
Small intestineFamilial adenomatous polyposisAPC[35]
Lynch syndromeMutations in mismatch repair genes
Juvenile polyposis syndromeSMAD4
Peutz-Jeghers syndromeSTK11
Liverα-1 antitrypsin deficiencySERPINA1[36,37,38,39,40]
Hereditary hemochromatosisHFE
Hereditary tyrosinemia type 1FAH
Glycogen storage disease type 1G6PC, SLC37A4
Wilson’s diseaseATP7B
Niemann-park diseaseSMPD1 AND NPC1 OR NPC2
BiliaryBile salt export pump deficiencyABCB11[41]
Table 2. Evidence that anthocyanins have chemopreventive properties against GI cancer and their potential cellular mechanisms.
Table 2. Evidence that anthocyanins have chemopreventive properties against GI cancer and their potential cellular mechanisms.
Source of AnthocyaninDosageCell Line/Animal ModelObservationsReference
Oral Cancer
Blueberry and malvidin50 µg/mLHuman oral SCC131 cellsReduced STAT-3 phosphorylation and nuclear translocation
Induced cell cycle arrest at G1/S phase and apoptosis
[152]
Cranberry extracts25–200 µg/mLHuman oral epidermal KB, CAL-27 cancer cellsInhibited cell proliferation[169]
Black rice (Oryza Sativa L.)100–500 µg/mLHuman tongue epithelial CAL 27 cellsInhibited cell migration and invasion
Inhibited activity of MMP-2
Inhibited NF-κB p65 protein expression
Suppressed Pl3K/Akt pathway
[170]
Commercial anthocyanin250 µg/mLHuman oral SCCReduced cell viability, Inhibited migration, and invasion abilities
Increased NLRP3, caspase-1, IL-1β protein expression
[171]
Grape skin extract2.5 mg/kg of body weightMale Wistar rats; 4-nitroquinoline 1-oxide induced tongue carcinogenesisReduced epithelial dysplasia
Reduced p-NF-κB p50 and MyD88 protein expression
No change in copper-zinc superoxide dismutase, manganese superoxide dismutase, and catalase gene expression
[172]
Lyophilized strawberry5% or 10% w/w for 12 weeksHamster cheek pouch (HCP) model of oral cancerReduced number of tumors
Mild and severe dysplasia
[173]
Esophageal Cancer
Lyophilized black raspberry100 μg/mLHuman esophageal microvascular endothelial cells (HEMEC)Inhibited TNF-α/IL-1β-induced NFκB p65 nuclear translocation, PGE2 production
Reduced COX-2, ICAM-1 and VCAM-1 mRNA and protein expression and leukocyte binding
Inhibited Akt, MAPK and JNK phosphorylation
[174]
Lyophilized black raspberry, C3G, C3R10–50 µg/mLRE-149DHD and RE-149 rat esophageal cancer cell linesInhibited cell growth
Induced apoptosis
Reduced COX-2, iNOS mRNA expression
[175]
Lyophilized black raspberry2.5% w/w of the dietMale Sprague-Dawley rats, EDA surgery-induced carcinogenesisNo change in COX-2 level
Reduced MnSOD levels
Not effective in the prevention of reflux-induced esophageal adenocarcinoma
[176]
Lyophilized black raspberry5% w/w for 10 weeksNMBA-induced carcinogenesis in F344 ratsInfluenced the metabolic activation and detoxification of NMBA
Reduced cell proliferation, inflammation, and angiogenesis
Inhibited CYP2a2 mRNA expression
[177]
Lyophilized black raspberry5% w/w for 30 weeksNMBA induced carcinogenesis in F344 ratsReduced NF-κB protein expression
Reduced number and volume of NMBA-induced papillomas
Inhibited cell proliferation and, inflammation
Induced apoptosis
[178]
Either black or red raspberries, strawberries, blueberries, noni, açaí or wolfberry5% w/w for 35 weeksNMBA induced carcinogenesis in F344 ratsReduced serum cytokines, IL-5, and GRO/KC protein expression
No change in serum IL-1ß, IL-4, IL-13, and TNF-α protein expression
Increased IFN-γ protein expression
[179]
Lyophilized black raspberry, anthocyanin extract, PCA6.1% w/w, 0.35 ppm and 500 ppm respectivelyNMBA induced carcinogenesis in F344 ratsReduced IL-1β protein expression
Increased IL-10, IL-12 protein expression
Increased infiltration of both macrophages and neutrophils into the esophagus
[180]
Gastric Cancer
Malvidin50–200 µg/mLHuman AGS cellsInduced apoptosis-arrest G0/G1 phase
Loss of mitochondrial membrane potential
Increased BAX/Bcl-2 ration and P38 kinase expression
Inhibited ERK activity
[157]
Black soybean anthocyanin12.5–50 µg/mLH. pylori-induced inflammation in AGS cells Reduced H. pylori-induced ROS production
Inhibited phosphorylation of mitogen-activated protein kinases, translocation of NF-κB, iNOS, Cox-2 mRNA expressions, IL-8 production
[181]
Liver Cancer
Black currant100, 500 mg/kg body weight for 22 weeksDENA-induced carcinogenesis in ratsReduced abnormal lipid peroxidation, protein oxidation and expression of iNOS, 3-nitrotyrosine, Nrf-2[133]
Malvidin-3-galactoside50–200 µg/mLHuman HepG2 cellsReduced P-AKT level, MMP-2 and, MMP-9 protein expression
Induced apoptosis
Increased cyclin-D1, B, E, Caspase-3 protein expression
[182]
Meoru anthocyanin400 µg/mLHuman Hep3B cellsReduced MMP-2, MMP-9 protein expression
Activated NF-κB
Promoted anti-invasive effects
[183]
Isolated anthocyanins100 or 500 µg/mLRat hepatoma cells (MH1C1)-DNA damaged induced by TBHPReduced DNA single-strand formation and lipid peroxidation
No change in redox state
[184]
Meoru anthocyanin400 µg/mLHuman Hep3B cellsReduced cell proliferation, invasion
Induced mitochondrial dysfunction
Reduced Bcl-2, XlAP, ClAP-1, ClAP-2 protein expression
[154]
Berry anthocyanin0.001–0.1 mg/mLHuman HCC cell lines PLC/PRF/5Increased Bax, cytochrome c, caspase 3 and, elF2-α protein expression
Reduced mTOR, Bcl-2 protein expression
[185]
Delphinidin, cyanidin, and malvidin100 µg/mLHuman HepG2 cellsReduced cell growth
Induced apoptosis-internucleosomal DNA fragmentation
Increased Bax: Bcl-2 protein expression
Activated c-Jun-N-terminal cascade
[186]
Black currant0.125%, 0.625% w/w for 22 weeksDENA-induced carcinogenesis in Sprague-Dawley ratsIncreased incidence, total number, multiplicity, size, and volume of preneoplastic hepatic nodules
Abnormal cell proliferation
Induced apoptosis
Increased Bax: Bcl-2 protein expression
[187]
Colorectal Cancer
Anthocyanin metabolites (gallic acid, 3-O-methylgallic acid, and 2,4,6-trihydroxybenzaldehyde10–100 µmol/LHuman Caco-2 cellsReduced cell viability
Induced cell cycle arrest at G0/G1
Increased caspase-3 activation
Inhibited transcription factors NF-κB, AP-1, STAT-1, and OCT-1
[22]
Standardized anthocyanin-rich extract50–500 μg/mLHuman Caco-2 cellsInhibited cell proliferation
Caspase-3 activation
Induced apoptosis
Increased cellular ROS
[188]
Lyophilized blueberry70–100 μg/mL
50–100 μg/mL
Human HT-29
Human Caco-2 cells
Inhibited cell proliferation
2–7 times increased DNA fragmentation
Induced apoptosis
[189]
Lyophilized black raspberries0%, 2.5%, 5%, or 10% wt/wt for 33 weeksAOM-induced carcinogenesis in F344 ratsReduced ACF, tumor multiplicity, adenocarcinoma multiplicity by the dose-depended manner[190]
Purple fleshed sweet potato10% w/w of potato skin, potato flesh & 0.12% w/w anthocyanin-rich extracted for 18 weeksC57BL/6J-APCMIN/+ miceReduced adenoma number (0.12% w/w anthocyanin-rich extracted more effective)[191]
Lyophilized black raspberries5% w/w for 8 weeksAPCMIN/+ miceReduced intestinal and colonic polyp number and size
Reversed 23 APC-regulated metabolites, including 13 colonic mucosa, 8 liver and 2 fecal metabolites
Reduced putrescine and linolenate levels
[192]
Cocoplum anthocyanin1 to 20 μg/mLTNF-α stimulated Human HT-29 cells, CCD-18Co non-malignant colonic fibroblastsInhibited cell proliferation
Increased cellular ROS
Reduced TNF-α, IL-1β, IL-6, and NF-κB1 mRNA expression
[117]
Purple-sweet potato anthocyanin0–40 μMHuman colonic SW480 cancer cellsInhibited cell proliferation
Cell cycle arrest at G1 phase
[193]
Purple fleshed potato10–30 μg/mLHuman HCT-116 and HT-29 cellsInhibited cell proliferation
Induced apoptosis
[194]
Cyanidin chloride0–50 µMTNF-α stimulated Human HCT116, HT29, and SW620Suppressed NF-κB signaling
Activated the Nrf2 pathway
Increased Bax: Bcl-2 protein and mRNA expression
Reduced protein and mRNA expression of TNF-α, IL-6, and IL-8
[195]
Black raspberry powder0.5,5,25 μg/mLHuman HCT116, Caco2 and SW480 cellsIncreased protein expression of DNMT1 and DNMT3B
Reduced mRNA expression of β-catenin
Inhibited cell proliferation
Induced apoptosis
[196]
Anthocyanin-rich extract from Hull blackberries0–40 μg/mLHuman HT-29 cellsInhibited cell proliferation
Increased release of IL-12
[197]
Anthocyanin-rich extracts from bilberry, chokeberry, grape3.85 g/kg for 4 weeksAOM-induced carcinogenesis in F344 ratsReduced ACF, fecal bile acids and, colonic cellular proliferation
Reduced COX-2 mRNA expression (bilberry, grape diets)
[198]
Anthocyanin-rich extracts from bilberry10% w/w supplementation for 9 weeksAOM/DSS-induced colitis-associated carcinogenesis in Balb/c miceLess reduced colon length
Less inflammation
Less mean tumor number
[199]
Abbreviations used: AKT, protein kinase B; AP-1, activator protein 1; bcl-2, B-cell lymphoma 2; BAX, Bcl-2 associated X; COX-2, cyclooxygenase 2; CIAP-1, cellular inhibitor of apoptosis protein-1; CIAP-2, cellular inhibitor of apoptosis protein-2; DNMT1, DNA (cytosine-5)-methyltransferase 1; DNMT2, DNA (cytosine-5)-methyltransferase 2; elF2-α, eukaryotic initiation factor 2; ERK, extracellular-signal-regulated kinase; GRO/KC, growth related oncogene; CXCL1; IL-4,5,10,12,13,1β, Interleukin-4,5,10,12,13,1β; IFN-γ, interferon γ; iNOS, inducible nitrogen oxide synthase; JAK, Janus kinase; LC3-I, LC3-I, microtubule-associated protein light chain 3; MMP-2,9, matrix metalloproteinase-2,9; MAPK, mitogen-activated protein kinase; MnSOD, manganese superoxide dismutase; MyD88, myeloid differentiation primary response 88; MTOR, mammalian target of rapamycin; NLRP3, NLR family pyrin domain containing 3; NMBA, N-nitroso methylbenzylamine; NF-κB, Nuclear factor κ-light-chain-enhancer of activated B cells; OCT-1, Octamer proteins in humans; PI3K, phosphoinositide 3-kinases; P-NF-κB, phosphorylated nuclear factor κ-light-chain-enhancer of activated B cells; ROS, reactive oxygen species; STAT-1,3, signal transducer and activator of transcription 1,3; XIAP, X-linked inhibitor of apoptosis protein.
Table 3. Experimental findings on the effect of anthocyanin-supplementation on the DSS-induced colitis in experimental animals.
Table 3. Experimental findings on the effect of anthocyanin-supplementation on the DSS-induced colitis in experimental animals.
Source of AnthocyaninDosageTreatmentObservationsReference
Black rice anthocyanin-rich extract25, 50, and 100 mg/kg of body weight8 weeks old female C57BL/6 mice: administration of 3% DSS for 5 consecutive days in drinking waterReduced DAI and the histological score of colons, myeloperoxidase (MPO) and nitric oxide (NO) levels and, mRNA expression of IL-6, IL-1β, TNF-α, iNOS, and COX-2[119]
Malvidin 3-glucoside24 mg/kg of feed weight4–5 weeks old C57BL/6J male mice: 2 cycles (7 days of 2.5% DSS and 14 days of fresh tap water)Improved histopathological scores
mRNA expression of IL-10
Promoted microbial interactions and restored the Firmicutes/Bacteroidetes ratio repressed by DSS
Reduced abundance of Ruminococcus gnavus
[167]
Blueberry extract50 mg/kg body weightFemale Balb/C mice: administration of 3% DSS for 1 week in drinking waterReduced DAI and improved the macroscopic and histological score of colons
Reduced myeloperoxidase accumulation and malondialdehyde in the colon
Increased prostaglandin E2 level in serum
Reduced levels of superoxide dismutase and catalase
Reduced mRNA expression of COX-2 and IL-1β in colonic tissue
Reduced nuclear translocation of NF-kB
[237]
Dietary red raspberry5% w/w of feed weight Six-week-old male C57BL/6J mice: administration of 2 repeated cycles of 1% DSS (7-d DSS treatment plus 14-d recovery)Reduced DAI score and histologic damage
Reduced expression of inflammatory mediators
Facilitated epithelial repair
Reduced β-catenin, STAT3 signaling
[238]
Maqui berry water extract50–200 mg/kg of body weight6 weeks old
wild-type C57BL/6 male mice: administration of 3% (w/v) DSS
for 1 week in drinking water
Reduced protein expression of COX2 and IL-6 in LPS-stimulated RAW 264.7 cells
Reduced inflammatory bowel disease index, MDA, NO, i-NOS, COX-2 protein expression in colon tissue
Reduced MPO, TNF-α, and IL-1β protein expression in blood serums
Increased protein expression of occludin (Dose-dependent manner)
[241]
Ginseng berry extract50 mg/kg of body weightC57BL/6 mice: administration of 3% DSS for 8 days in drinking waterReduced DAI score and histologic damage
Reduced numbers and inhibited the activation of colon-infiltrating T cells, neutrophils, intestinal CD103−CD11c+ dendritic cells and macrophages
[242]
Cranberry extract1.5% w/w of feed weight6 weeks old male CD-1 mice: 1.5% DSS for 4 cycles (4 days/cycle, with a 7-day
recovery after each of the first 3 DSS cycles)
Inhibited reduction in colon length
Reduced DAI and histologic score
Increased colonic levels of IL-1β, IL-6, and TNF-α proteins
Altered the microbial structure of fecal microbiota in mice
Reduced DSS-induced decline in α-diversity
Increased abundance of Lactobacillus and Bifidobacterium
Reduced abundance of Sutterella and Bilophila
[243]
Dried bilberries10% w/w of feed weightBalb/c mice: 2.5% DSS for 1 week in drinking waterReduced DAI and histologic score
Reduced secretion of IFN-γ and TNF-α from mesenteric lymph node cells
Intestinal inflammation
Prevented inflammation-induced apoptosis in colonic epithelial cells
[244]
C3GIntraperitoneal injected with 1ug C3G every 2 days, a total of 3 times8–12 weeks old C57BL/6 mice: 3.5% DSS for 1 week in drinking waterNo change in body weight and colon length
Reduced mRNA expression of IL- 6, IL-1β, IL-18, TNF-α, IFN-γ in colons and mesenteric lymph nodes
Reduced CCL22 levels and Tregs induction
[245]
Anthocyanin-rich tea0.13 or 0.16 mg/day by gavage5 weeks old female ICR mice: 3% DSS for 2 weeks in drinking waterLowered body weight loss, spleen hypertrophy, and shortening of the colon
Reduced deteriorations in survival rate, liver function, colon mucosal IL-1β level (mRNA)
[246]
Purple carrot extract5% w/w of feed weight6–7 weeks old C57BL/6 mice: 2% DSS for 1 week in drinking waterReduced DSS-induced colon shortening and inflammatory cell infiltration
Reduced serum levels of TNF-α and IL-6 (protein)
Inhibited colonic mRNA expression of iNOS, COX-2
[247]
Abbreviations used: CCL22, C-C motif chemokine ligand 22; COX-2, cyclooxygenase 2; DAI, disease activity index; IL-6, 1β, interleukin-6, 1β; IFN-γ, interferon-γ; iNOS, inducible nitrogen oxide synthase; MDA, malondialdehyde; MPO, myeloperoxidase; NO, nitrogen oxide; NF-κB, nuclear factor κ-light-chain-enhancer of activated B cells; STAT-3, signal transducer and activator of transcription 3; TNL-α, tumor necrosis factor-α.

Share and Cite

MDPI and ACS Style

Dharmawansa, K.V.S.; Hoskin, D.W.; Rupasinghe, H.P.V. Chemopreventive Effect of Dietary Anthocyanins against Gastrointestinal Cancers: A Review of Recent Advances and Perspectives. Int. J. Mol. Sci. 2020, 21, 6555. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21186555

AMA Style

Dharmawansa KVS, Hoskin DW, Rupasinghe HPV. Chemopreventive Effect of Dietary Anthocyanins against Gastrointestinal Cancers: A Review of Recent Advances and Perspectives. International Journal of Molecular Sciences. 2020; 21(18):6555. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21186555

Chicago/Turabian Style

Dharmawansa, K.V. Surangi, David W. Hoskin, and H. P. Vasantha Rupasinghe. 2020. "Chemopreventive Effect of Dietary Anthocyanins against Gastrointestinal Cancers: A Review of Recent Advances and Perspectives" International Journal of Molecular Sciences 21, no. 18: 6555. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21186555

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

Article Metrics

Back to TopTop