Next Article in Journal
The Nature of Metal Artifacts in X-ray Computed Tomography and Their Reduction by Optimization of Tomography Systems Parameters
Next Article in Special Issue
Potential Health Benefits of Fruits and Vegetables II
Previous Article in Journal
Energy Spatial Distribution of Behind-Armor Debris Generated by Penetration of Explosively Formed Projectiles with Different Length–Diameter Ratio
Previous Article in Special Issue
Bioactive Compounds, Antioxidant Activity, and Mineral Content of Wild Rocket (Diplotaxis tenuifolia L.) Leaves as Affected by Saline Stress and Biostimulant Application
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Vegetal-Derived Bioactive Compounds as Multidrug Resistance Modulators in Colorectal Cancer

by
Francisco Quiñonero
1,2,3,
Cristina Mesas
1,2,3,
Mercedes Peña
1,2,3,
Laura Cabeza
1,2,3,
Gloria Perazzoli
1,2,3,
Consolación Melguizo
1,2,3,*,
Raul Ortiz
1,2,3,† and
Jose Prados
1,2,3,†
1
Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain
2
Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
3
Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18014 Granada, Spain
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Appl. Sci. 2023, 13(4), 2667; https://doi.org/10.3390/app13042667
Submission received: 13 December 2022 / Revised: 15 February 2023 / Accepted: 17 February 2023 / Published: 19 February 2023
(This article belongs to the Special Issue Potential Health Benefits of Fruits and Vegetables II)

Abstract

:
Colorectal cancer is one of the leading causes of morbidity and mortality today. Knowledge of its pathogenesis has made it possible to advance the development of different therapeutic strategies. However, the appearance of drug resistance constitutes one of the main causes of treatment failure. Bioactive compounds of vegetable origin are being studied as a new strategy to improve antitumor treatment, due to their ability to regulate the pathways involved in the development of carcinogenesis or processes that are decisive in its evolution, including multidrug resistance. In vitro and in vivo studies of these substances in combination with cytotoxic drugs have shown that they reduce resistance and increase therapeutic efficacy. The objective of this review is to summarize the knowledge that is described in the scientific literature on the antitumor and chemo-sensitizing capacity of vegetable-derived biomolecules such as polyphenols, flavonoids, and terpenes. These compounds may hold a promising future in improving the treatment of colorectal cancer.

1. Introduction

Colorectal cancer (CRC) accounted for 9.39% of deaths by cancers in 2020 and is the third most commonly diagnosed cancer in the word. CRC incidence could double by 2035 due to an increase in the number of cases and their early diagnosis [1]. Current treatment is based on resection combined with chemotherapy and adjuvant radiotherapy in the early stages and larger resections that are associated with chemotherapy (oxaliplatin, irinotecan, 5-fluorouracil (5-FU) and capecitabine, among others) in metastatic stages [2]. However, despite the use of different therapeutic options, the average survival in metastatic patients is low (3 years) although the 5-year survival rates of patients that are diagnosed with localized tumors or with regional dissemination are 90% and 69%, respectively [3,4]. Acquired or intrinsic drug resistance in CRC is one the main causes of treatment failure. A clinical assay that was performed with patients from 13 European countries, Israel, and South Africa showed that more than 50% of CRC patients had resistance to the drug 5-FU [5,6]. In addition, the mechanisms that generate resistance to one drug produce resistance to others (multidrug resistance or MDR). The mechanism that is mediated by the ATP-dependent transporter family (ATP-binding cassette family, ABC) is prominent in CRC, including the p-glycoprotein (p-GP; ABCB1), a drug efflux pump which prevents cellular uptake of many structurally and functionally cytotoxic compounds [7,8]. Understanding resistance phenomena is essential for improving CRC patient prognosis.
In this context, it has been shown that some plant-derived natural products or their association with the classic cytotoxic drugs used in cancer treatment were capable of overcoming multidrug resistance and/or reducing the effective antitumor drug dose [9]. Despite the low proportion of plant species that have been explored for their antitumor activity [10], plant-derived natural products have played an important role in the treatment and prevention of cancer. Most of the compounds from vegetables and fruits with significant antitumor activity also showed clear advantages in relation to modern drugs that are used in chemotherapy [11]. Furthermore, this therapeutic strategy based on the use of natural compounds showed minimal side effects and lower cost than synthesized antitumor drugs [12]. However, biodistribution, biotransformation, and transport limitations of natural products may hinder their application in cancer patients. Furthermore, their effect on the immune system is not fully analyzed [13,14,15]. Concretely in CRC, the therapeutic potential of some natural compounds has been clearly demonstrated [16]. In addition, their combination with other conventional chemotherapeutic drugs has demonstrated a significant synergistic effect [17]. In fact, curcumin, resveratrol, and (-)-Epigallocatechin Gallate (EGCG) and terpenoids, secondary plant metabolites that are composed of isoprene units, enhanced the effect of cytotoxic drugs [18,19,20]. The antitumor effect exerted by these compounds is carried out through several pathways including cell cycle arrest and cell death by apoptosis [21]. On the other hand, polyphenols from plants also prevent multidrug resistance in several types of solid tumors, including CRC. In addition, these compounds inhibit cell proliferation, angiogenesis, and metastasis, as well as regulating the proinflammatory response [22]. Finally, quercetin, a flavonoid, not only modulates the drug resistance phenomena but is capable of increasing sensitivity to doxorubicin in CRC through the inhibition of a glutamine transporter (SLC1A5) [23,24].
The objective of this review is to study the different families of plant-derived compounds that modulate drug resistance in CRC, observing their ability to be used as a future therapy against this type of cancer.

2. Colorectal Cancer: Resistant Mechanisms

Despite the discovery of new drugs against CRC, the emergence of resistance to these agents is inevitable. Drug resistance can be innate (dysregulations of tumor cells before treatment) or acquired (resistance after treatment cycles) [25,26]. Drug efflux mediated by transmembrane transporters, specifically those of the ATP binding cassette superfamily (ABC), is one of most relevant mechanisms in CRC. These proteins are capable of expelling toxic substances from the inside of cells including different anticancer agents [27]. Specifically, p-GP, encoded by the ABCB1 gene, was overexpressed in different CRC cell lines, conferring resistance to treatment. In addition, resistant CRC cells overexpressed CD133, a protein that regulates p-GP expression through the AKT/NF-κB/MDR1 axis [28,29]. Other members of the ABC family, such as MRP1 and BCRP, are also overexpressed in some CRC cell lines, leading to multiple resistance to chemotherapeutic drugs as 5-FU, doxorubicin, irinotecan, vincristine, among others [30,31], while MRP2-mediated resistance to oxaliplatin and vincristine in CRC, being Nrf2, signaling is critical for its expression (Figure 1) [32]. Drug resistance in CRC can also arise when there are alterations in antitumor drug targets, such as mutations or changes in expression due to epigenetic variations [33]. Finally, an increase in the expression of repair protein-DNA, such as MGMT, was detected in some 5-FU-resistant CRC lines [34].
Apoptosis evasion promotes carcinogenesis and tumor progression, leading to the appearance of pharmacological resistance, especially to drugs that induce this pathway such as doxorubicin and cisplatin. Several apoptosis-resistant tumors were associated with the increased or decreased expression of antiapoptotic (BCL-2, MCL-1, and BCL-XL) and proapoptotic (p53, BAX, and BIM) genes, respectively [35]. In addition, modulation of DNA methylation, histones and chromatin remodeling can alter the expression of genes that are involved in the metabolism and activity of chemotherapeutic drugs, inducing resistance [33]. Moreover, tumor heterogeneity also plays a role in this phenomenon, as it makes treatment more difficult because of the presence of cancer stem cells which are more resistant to drugs. These cells have a self-healing and differentiation capacity and are associated with greater tumorigenicity. These cells are also capable of acquiring mesenchymal characteristics, which is related to the cell migration process and metastasis and a worse prognosis in patients [36,37]. On the other hand, it is known that the most resistant cells within the tumor sinus can transfer small miRNAs to their environment, inducing resistance in neighboring cells [38]. Finally, another important factor to highlight is the tumor microenvironment, including the extracellular matrix, blood vessels, fibroblast, and immune system cells. This microenvironment will be an additional layer of protection against drugs, making the entry of chemotherapeutics into the tumor sinus more limited [39].

3. Natural Products: A New Source of Molecules with Antitumor Activity

Many plant compounds are substances with known bioactive activity that can be used as chemotherapeutic agents in cancer. One of the best known, taxol, is a potent diterpene with antitumor activity that is derived from the bark of the yew tree (Taxus brevifolia). These compounds show advantages over traditional drugs, such as greater bioavailability, accessibility, cost-effectiveness, and lower systemic toxicity [40]. This last point is highly relevant since conventional drugs show many side effects [20]. In addition, these compounds show a synergy with conventional therapies and a benefit for the treatment of CRC by avoiding drug resistance [41,42,43]. Therefore, plant-derived natural compounds allow us to have more cancer treatment options. Polyphenols, flavonoids, and terpenes have been described as the main molecules that are able to modulate chemoresistance phenomena in CRC (Figure 2).

3.1. Polyphenols

Polyphenols (Figure 2A) comprise a heterogeneous group of phytochemicals that contain one or more phenolic rings in their chemical structure. They are divided into different groups, including stilbenes, phenolic acids, lignans, xanthones, tannins, and flavonoids [44]. These compounds are synthesized by different plants to carry out different functions, including protection against UV radiation and infection by microorganisms, as well as regulation of cell growth [45,46]. The three most studied polyphenols in CRC are resveratrol, curcumin, and kaempferol.

3.1.1. Resveratrol

Resveratrol is a polyphenol belonging to the stilbenes group. It is found in grapes, blueberries, and peanuts, intervening in the response to infection by microorganisms [47]. From its study it has been determined that it has antioxidant, anti-inflammatory, antitumor, cardioprotective, and neuroprotective properties. These effects are mainly based on its ability to suppress inflammation, regulate various enzymatic processes that are involved in the metabolism of numerous substances, eliminate reactive oxygen species, and inhibit cell proliferation through its action as a phytoestrogen. The therapeutic action of resveratrol comprises different mechanisms such as the induction of cell death through apoptosis and autophagy, the capacity to suppress tumor proliferation, or the inhibition of angiogenesis and metastasis [48]. The preventive and therapeutic effect has also been observed for CRC. In vitro studies that were carried out in different colon cancer cell lines have demonstrated the ability of this compound to inhibit cell growth and proliferation, promoting cell cycle stop and derived apoptosis activating apoptotic markers such as caspases 3 and 8. In the CRC line HT-29, this compound had an IC50 of 10 mg/mL after 72 h of treatment [49]. Particularly, in HCT116 and CaCo-2 cell lines, it has been observed that resveratrol inhibits cell cycle initiation by the downregulation of the CCND1/CDK4 complex. In addition, resveratrol treatment decreases the synthesis of proangiogenic factors such as VEGF in CRC [50]. Furthermore, it has been observed that it is able to reverse the Warburg effect in tumor cells through modulation of the pyruvate dehydrogenase complex, influencing the process of glycolysis [51]. Its in vivo antitumor capacity has also been shown. Thus, in murine models of rats and mice that had the KRAS proto-oncogene overexpressed, dietary administration of 150 or 300 ppm resveratrol for 4 weeks was shown to prevent tumor growth through the deregulation of KRAS via miR-96 [52]. On the other hand, a model of colonic tumorigenesis in situ showed that resveratrol prevented the formation of precursor lesions (dysplasia or adenomas), in addition to reducing the volume of tumors if they had been generated. The downregulation of the pro-apoptotic gene BAX was observed in the treated tumor mucosa, as well as a reduction of COX-2 and NF-κB [47,53]. Human clinical trials showed that resveratrol intake was safe and well tolerated systemically, with side effects such as diarrhea, abdominal pain, and nausea being observed in patients that were treated with more than 1 g per day. Despite this, the bioavailability of resveratrol in the body is limited with 71–98% recovery of the compound or its derivatives in urine and feces after oral administration [47,54,55].
Another possibility that was studied was the combination of resveratrol together with traditional chemotherapy drugs, showing the sensitization of tumor cells to conventional drugs and the reduction of existing resistance to these drugs. Thus, its combination with 5-FU produced a decrease in cell proliferation and an increase in apoptosis in vitro, accompanied by a reduction in tumor volume and vascular density in vivo [56,57,58]. Synergistic effects have also been shown with oxaliplatin, observing that their combination produces a greater inhibition of cell proliferation compared to the chemotherapy alone. The effect of resveratrol is found to be exerted through increased expression of miR-34c, which silences the PI3K/AKT pathway and triggers reduced expression of the resistance gene MDR1 [59]. The death that is produced by this combination is through necrotic and apoptotic pathways [60].
Due to the low bioavailability of resveratrol, formulations have been designed to allow improved transport into the cell. Khayat et al. designed zein nanoformulations that had a high encapsulation of the plant compound and were more effective in inducing apoptosis and oxidative stress in cells compared to the free drug in HCT116, Caco-2, and HT-29 cell lines [61]. Another study designed gels that were capable of encapsulating resveratrol with high efficacy for efficient oral administration. These formulations were tested in HCT116 cells, obtaining greater efficacy in cytotoxicity assays and significantly increasing the expression of pro-apoptotic genes such as Bcl-2 compared to free resveratrol [62]. Feng et al. synthesized lipid nanocapsules encapsulating resveratrol and showed 70% drug release at 48 h, increasing the cytotoxic effect of the free drug and significantly increasing the induced apoptosis in the HT-29 cell line [63]. Finally, other formulations encapsulating resveratrol in mesoporous silica nanoparticles and liposomes were developed. The former showed high drug loading capacity and improved efficacy against free drug in HT-29 and LS147T lines [64]. Meanwhile, liposomes showed a high drug release after 24 h and increased the cytotoxic effect of the free drug in the HT-29 line, increasing the delivery of this hydrophobic agent [65].

3.1.2. Curcumin

On the other hand, curcumin is one of the most studied bioactive compounds as an adjuvant in CRC. This phytochemical is derived from turmeric (Curcuma longa) and is used as a dietary supplement or food flavoring [66]. The mechanisms by which it influences CRC have been extensively studied. It is known to regulate the microbiome, protect the intestinal barrier and promote anti-inflammatory activity in the intestine through inactivation of the proinflammatory factor NF-κB and by increasing the expansion of CD4+ FOXP3+ regulatory T lymphocytes in the mucosa. In addition, it modulates tumor cell death via the autophagy mechanism by suppressing the PI3K/AKT/mTOR signaling pathway and it is able to trigger epigenetic modifications that increase its chemosensitizing power together with other drugs [67]. Pharmacokinetic and toxicity studies showed that its administration in humans is safe, although its absorption is low and it is a compound that is rapidly eliminated by the body, problems that currently prevent it from being tested as a therapeutic agent [68].
The chemopreventive action of curcumin in CRC has been demonstrated in numerous in vitro studies. In the HT116 cell line, it inhibits proliferation by blocking the WNT/β-catenin pathway that is mediated by c-MYC, producing cell cycle arrest in G2/M phase, and inducing apoptosis. In the HT-29 cell line, curcumin is also able to stop the cycle in the G1 phase and induce apoptosis by acting on the pAKT-AMPK-COX-2 signaling pathway at a dose of 20 µM [69]. In addition, curcumin has been shown to inhibit the migration and invasion of HT116 and LoVo cell lines [70]. In vivo studies that were conducted in murine models have shown that a dose of 300 mg/kg prevents progression to cancer from precancerous lesions, reducing the number of adenomas and tumor size [71,72]. Clinical studies that were conducted in humans observed that oral consumption of curcumin in CRC patients reduced serum TNF-α levels and increased apoptosis in the tumor tissue tested [73].
Curcumin has also been studied as an adjuvant and chemosensitizer in combination with drugs that were traditionally used in the treatment of CRC. Several in vitro and in vivo studies have demonstrated the ability of curcumin to decrease tumor cell resistance to drugs such as 5-FU and oxaliplatin. Particularly, in vitro studies indicate that this compound decreases the expression of several genes (IGF-1, AKT, COX-2, and cyclin D1) that are involved in resistance to these two drugs [73]. It has also been shown, in oxaliplatin-resistant HCT116 cell lines that were cultured with this chemotherapeutic and curcumin, that resistance is reversed through inhibition of the TGF-β/SMAD2/SMAD3 pathway. In mice that were transplanted with oxaliplatin-resistant HCT116 cells, the administration of curcumin together with oxaliplatin has also been observed to significantly reduce tumor volume and weight [74]. The combination of a slightly cytotoxic dose of curcumin (10 µM) with 5-FU (5 µM) produced a synergistic antitumor response, reducing cell growth, and inducing apoptosis as observed in in vitro and in vivo studies. This chemo-sensitizing effect is carried out by inhibiting STAT1 activation, which reduces PD-L1 expression [75]. Furthermore, this synergism could also lead to a decrease in CRC progression and metastasis, thanks to the ability of curcumin to decrease the expression of IGF-1 and the myc oncogene [76].
As curcumin is one of the most studied natural compounds, studies have attempted to encapsulate it to increase its bioavailability and absorption. Han et al. designed a nanoformulation based on fusion proteins that were capable of being efficiently introduced into cells with high expression of epidermal growth factor receptor (EGFR), being more effective in the HCT116 line (EGFR-positive) versus SW620 (EGFR-negative) [77]. Wang et al. encapsulated the compound in micelles and increased its bioavailability and cytotoxic activity in CRC cell lines. Moreover, after simulated in vitro digestions, 40% of the formulation was still effective against tumor cell lines, thus possessing potential for the prevention and treatment of this type of cancer [78]. Although this drug was encapsulated in other types of nanoformulations such as PLGA (poly lactic co-glycolic acid)-coated nanoparticles [79] or in β- cyclodextrins [80] showing efficacy, an interesting study that was carried out by Gupta et al. used poly(allylamine)/eudragit nanoparticles that were capable of encapsulating doxorubicin and curcumin with high efficiency. The combination of both drugs in the nanoformulation showed high cytotoxicity in CRC HCT116 cells and in Balb/C murine models, with retention of these nanoformulations in the colon region 24 h after treatment [81].
Therefore, the findings that were observed in the multiple studies employing the use of curcumin and resveratrol in CRC demonstrate their great capacity as chemopreventive and sensitizing agents in combination with other cytotoxic drugs. However, it is still essential to continue studying these compounds to be able to introduce them into clinical practice in the future.

3.1.3. Kaempferol

Kaempferol is a flavanol that is present in vegetables and plants such as broccoli, grapes, apples, brussels sprouts, or black tea. This bioactive compound possesses cardioprotective, neuroprotective, anti-inflammatory, antidiabetic, antioxidant, and antitumor properties [82]. The mechanisms of action underlying the anticancer effect of kaempferol are based on inhibition of the cell cycle in the G2/M phase to prevent tumor growth and proliferation, as well as induction of apoptosis by acting on different cell signaling pathways. Several in vitro studies have also observed its anti-angiogenic effect and anti-metastatic, inhibiting VEGF production and decreasing the expression of markers that are involved in epithelial-mesenchymal transition (EMT) at a dose of 0.1 µM in MDA cell lines [83,84]. Pharmacokinetic studies that were performed in rats determined that the bioavailable fraction of this compound after oral doses was 2% of the total administered [85]. To overcome this obstacle, the combination with quercetin has been tested, resulting in an increase in its bioavailability, in addition to improving efficacy [82]. In this way, this bioactive compound would act selectively on tumor cells, without affecting healthy ones. This compound exhibits cytotoxic effects on various CRC cell lines, such as HCT116, HT-29, HCT-15, LS174-R, and SW480, both when used alone and in combination with other chemotherapeutic agents. In HT-29 cell lines, the ability of this compound to inhibit the cell cycle has been studied, resulting in cell cycle arrest in G1 and G2/M phases through inhibition of CDK2, CDK4, and Cdc2 activity [86].
In vitro studies in the 5-FU-resistant LS174-R cell line and in other CRC lines such as HCT116 and HT-29 have shown that the antiproliferative activity that is exerted by the combination of kaempferol with this chemotherapeutic agent is through cell cycle arrest and induction of apoptosis, acting on signaling pathways such as JAK/STAT3, PI3K/AKT, MAPK, or NF-κB [87,88]. Other studies that were carried out in oxaliplatin-resistant HCT116, and HT-29 cell lines have shown that the combination of kaempferol with this drug generates a greater cytotoxic response, decreasing cell proliferation. The chemo-sensitization of these cells is mediated by the suppression of c-FOS protein expression [89].
Although not many nanoformulations have been synthesized to improve the bioavailability of kaemferol in the treatment of CRC, Meena et al. designed PEGylated gold nanoparticles that efficiently co-encapsulated DOX and the natural compound, improving the cytotoxic effect of both drugs separately and inducing apoptosis in the HT-29 line, without having high toxicity in the HTB-38 non-tumorous colorectal line. Finally, an in vivo experiment that was performed on mice showed a significant reduction in tumor volume without observing side effects [90].
Data on the use of kaempferol in vivo are still scarce, so it is imperative to continue its research to better understand its anticancer potential.

3.2. Flavonoids

Flavonoids represent a broad group of bioactive compounds from vegetables and plants. Although they have been catalogued as a subtype of polyphenols, as they are made up of a great variety of compounds, they are studied separately. The chemical structure of these substances is made up of a carbon skeleton with two aromatic rings joined by three carbons, with different modifications depending on the compound [43]. The subgroups that make up this family are chalcones, flavanols, flavones, flavanones, flavanols, isoflavones, and anthocyanins. Plants synthesize these extracts to provide color and aroma, for their protective properties against radiation or microbial infections, and their high detoxifying power [66]. There are numerous benefits of these compounds against various diseases, acting as antioxidants, anti-inflammatory, antibacterial, or antiviral agents, improving cognitive functions or preventing the onset of cancer and cardiovascular diseases [91]. Quercetin and EGCG (epigallocatechin gallate) are some of the most representative molecules of this group of compounds (Figure 2B).

3.2.1. Quercetin

Quercetin is the most representative bioactive compound of the flavanols group, and the one with the greatest presence in foods consumed daily. It can be found in onions, apples, grapes, broccoli, or tea [66]. This compound has been shown to be an excellent antioxidant and anti-inflammatory in vitro, also presenting an antitumor and antimicrobial effect [92]. Numerous in vivo and in vitro studies have analyzed the mechanisms of action by which quercetin exerts its antitumor effect. It mainly acts by regulating the cell cycle, inhibiting cell proliferation and growth through the modulation of different molecular pathways such as PI3K/AKT/mTOR and MAPK/ERK1/2. The cytotoxic effect of this compound has been shown in CRC lines HCT-15 and RKO at doses up to 300 µM [93] and with IC50 of 50, 100, and 50 µM in CRC lines CT26, MC38, and HT29 after 24 h of treatment, respectively [94]. The combination of quercetin with other bioactive compounds, such as curcumin, has also been shown to enhance the antiproliferative effect in different types of cancer by modulating the Wnt/β-catenin signaling pathway as observed in in vitro studies [95]. This compound also induces cell death through autophagy and apoptosis of tumor cells by increasing the expression of pro-apoptotic proteins and reducing the expression of anti-apoptotic proteins. In addition, it possesses anti-angiogenic and anti-metastatic activity, inhibiting VEGF protein expression and the EMT process increasing E-cadherin and decreasing mesenchymal markers such as N-cadherin, vimentin, and Snail. Lastly, in CRC quercetin inhibits tumor invasion and migration by regulating the expression of matrix metalloproteinases (MMPs) [92]. In in vivo studies, it is involved in reducing tumor size, reducing the number of precancerous lesions, suppressing metastasis, and reducing resistance to chemotherapy drugs [96].
This compound has also been shown to synergize with doxorubicin. Thus, a study that was carried out in the SW620 cell line observed that quercetin improves the cytotoxic activity of doxorubicin acting at the p-GP level [24]. On the other hand, encapsulation of the compound and its use together with doxorubicin increases cell growth inhibition in the CT26 cell line, reducing cardiac toxicity in murine models [97].
Although quercetin has not been generally encased in formulations, Wen et al. encapsulated this compound in a film containing chitosan nanoparticles and showed it to be an effective delivery system in CRC. It produced an effective cytotoxic effect on the Caco-2 line, exerting its effect through cell cycle arrest in G0/G1 phase and inducing apoptosis [98].

3.2.2. Epigallocatechin Gallate

Epigallocatechin gallate (EGCG) belongs to the flavanol group and is the most studied compound of this group in CRC. It is found mainly in green tea but is also found in other foods such as pistachios, strawberries, kiwis, hazelnuts, blackberries, and cherries. This compound is characterized by its antitumoral properties, acting as a chemopreventive agent through the inhibition of carcinogenesis in numerous types of cancer. The mechanisms that are used by this catechin to carry out its antitumor effect are based on the regulation of various signaling pathways that are involved in proliferation, apoptosis, angiogenesis, and metastasis. Among them, EGCG inhibits colonic tumor cell proliferation and migration inducing apoptosis through the activation of caspase-3 and PARP, in addition to produce the downregulation of STAT3. This compound was tested on CRC lines SW480, SW620, and LS411N and IC50 values of 74.6, 99.4, and 112.1 µg/mL were obtained after 24 h of treatment, respectively [99,100].
Its chemo-sensitizing activity has been studied in CRC cells that are resistant to chemotherapeutics. The combination of EGCG with 5-FU increases the efficacy of the latter, as well as its cytotoxic action, acting on tumor target cells. This effect has been observed in HCT116 and SW480 cell lines [101,102]. One of the mechanisms that is used by EGCG to enhance sensitivity to 5-FU is the inhibition of the GRP78/NF-κB/miR-155-5p/MDR1 pathway, having been demonstrated in in vitro and in vivo studies [103]. Furthermore, in vivo experiments demonstrated that administration of 30 mg/kg EGCG for two weeks decreased the number of hepatic metastatic lesions, reduced tumor growth, and increased apoptosis in tissues. In addition, less vascularization was found in treated tumors versus controls [104].
EGCG also has synergistic effects in combination with irinotecan in HCT116 cell lines. It has been observed that this drug produces S- and G2-phase arrest, inducing genotoxicity, an effect that was not observed in cells that were treated with EGCG alone [18]. Cisplatin and oxaliplatin are two chemotherapeutics that are used in the treatment of CRC. The combination of EGCG with these two agents in DLD-1 and HT-29 cell lines has shown an increased cytotoxic effect, causing cell proliferation inhibition and inducing autophagy [105]. These studies suggest that the combination of EGCG with different chemotherapeutics in CRC produce a synergism that increases antitumor activity.
To improve the bioavailability of this compound, Wang et al. synthesized gelatin and chitosan nanoparticles encapsulating 5-FU and ECGC, the latter compound inhibited tumor growth through its anti-angiogenic action and its ability to induce apoptosis. Encapsulation in this formulation allowed both compounds to increase blood circulation time in in vivo experiments, making them effective formulations against CRC [106].
A clinical trial was carried out with green tea extract, whose main bioactive compound was EGCG, trying to show whether the administration of 150 mg twice a day was able to reduce the risk of CRC, with no significant differences between the treatment group and the placebo [107]. Moreover, the chemo-sensitizing effect of EGCG is a breakthrough in overcoming tumor resistance. However, more evidence is needed to translate these results to clinical assays.

3.2.3. Other Flavonoids of Interest

In addition to the flavonoids that were analyzed above, which are the most studied as phytochemicals in CRC, there are other compounds that are derived from citrus fruits that show interest as active compounds against cancer. Among them, narangenin is a compound that is extracted from thyme (Thymus vulgaris L.), a shrub that grows in regions all over the world, especially in Mediterranean regions. Efficacy of this compound has been shown in CRC lines such as SW1116 and SW837, showing IC50 at 24 h of 1 and 1.55 mM, respectively, while in the non-tumor line CRL1554 it did not reach an IC50 dose at the 4 mM dose [108]. Another study showed that naringenin exerted its anti-tumor effect by deregulating cyclin D1 expression, leading to cell cycle arrest in HCT116 and SW480 lines [109]. This compound also showed an in vivo chemopreventive effect against the induction of precancerous lesions, reducing processes such as lipid peroxidation, ROS formation, and the activation of proinflammatory pathways in Wistar rat models [110]. Despite its interesting effects, this compound has low water solubility, cell permeability, and bioavailability. Therefore, Shabad et al. developed nanogels that were capable of increasing its dissolution capacity, obtaining a higher toxicity in in vitro models [111]. Another studied flavonoid is aromadendrin, derived from mandarin molasses. A study in the LoVo cell line and a DOX-resistant derivative (LoVo/Dx) showed that its effect is very limited in this type of cancer, producing a low cytotoxic effect [112]. Finally, another citrus-derived compound (tangeretin, present in the peel of several citrus fruits) showed synergistic activity with the chemotherapeutic drug 5-FU through early induction of oxidative stress and apoptosis in the HCT116 cell line. Bai et al. demonstrated that this synergistic effect occurs due to the natural compound that is able to decrease the expression of miR-21, whose expression increases after treatment with 5-FU, rescuing PTEN expression and inducing cellular autophagy [113].

3.3. Terpenes

Terpenes (Figure 2C) are another large group of bioactive compounds, many of which have CRC effects. These substances are part of the leaves, fruits, flowers, or roots of many plants, giving them a characteristic odor. When these compounds are oxidized, they become terpenoids, some of them being limonene, vitamin A, or β-carotene [42]. The study of these compounds as resistance modulators in CRC is not so recent or extensive, however, some evidence of the potential antitumor effect has been found in some of them.

3.3.1. Geraniol

Geraniol is a monoterpene that is found mainly in essential oils of aromatic plants and used in perfumes. It has been used as an active ingredient in many drugs since it exhibits analgesic and anti-inflammatory activity [114]. This compound presents preventive and therapeutic activity in many types of cancer, carrying out its effects by acting on the regulation of different signaling pathways such as PI3K/AKT/mTOR, MAPK/ERK1/2, or NF-κB, and modulating the expression of different molecules such as cyclins, CDKs, interleukins, and different growth factors [115].
In the Caco-2 cell line, it has been observed that this compound triggers the inhibition of cell proliferation by inducing an S-phase cycle arrest (IC30 = 200 µM after 7 days). In addition, it was shown that it is able to induce apoptosis in in vivo models, showing a decrease in the expression of the anti-apoptotic protein BCL-2 in tumor tissue after treating mice with a dose of 250 mg/kg for 4 weeks. Its oral administration in in vivo models prevents the development of CRC, reducing the number and size of precursor lesions [116].
Furthermore, its chemosensitizing effect was observed in Caco-2 and SW620 cell lines, and in murine models originated with 5-FU-resistant TC-118 tumor cells, having shown that co-treatment sensitizes tumor cells to 5-FU [117,118]. However, these studies are scarce, which requires further analysis to understand the effects of geraniol on resistance phenomena.

3.3.2. Ginsenosides

Ginsenosides are a subgroup of bioactive chemical compounds that are found in the root of ginseng, an Asian plant. These substances are used in teas, in the preparation of creams, or capsules. Of all the compounds that make up this subgroup, panaxadiol is one of the most notable for its antitumor action in many types of cancer. In the CRC cell line HCT116, this terpene inhibits cell proliferation (IC50 lower than 10 µM after 72 h of treatment) by suppressing PD-L1 expression; modulating different cellular pathways such as mTOR, MAPK/ERK, or JAK-STAT; and molecules such as HIF-1α. It has also been observed to decrease VEGF levels to exert antiangiogenic action. This has also been shown in studies that were performed on murine models treating mice at a dose of 30 mg/kg every 2 days for 3 weeks [119,120].
Another of the best known ginsenosides is Rg3. A study demonstrated that Rg3 was able to inhibit cell proliferation (IC50 between 100–200 µM after 48 h of treatment) and promote apoptosis in the HT-29 tumor line, decreasing its pluripotency and decreasing its angiogenesis through deregulation of the AMPK molecular pathway [121]. Analysis of its in vivo activity showed that its treatment with 25 mg/kg of Rg3 for 12 consecutive days reduced tumor vascularization and increased the toxicity that was produced by the chemotherapeutics 5-FU and oxaliplatin, allowing evasion of chemoresistance to both drugs [122].
The study of panaxadiol in the HCT116 cell line resistant to 5-FU has demonstrated the ability to inhibit proliferation producing synergy in combination with this chemotherapeutic compound, stopping the cell cycle and inducing apoptosis. Its sensitizing effect has also been analyzed in mouse models combining 30 mg/kg of 5-FU and 15/30 mg/kg of panaxadiol, confirming the results that were obtained in vitro, where a significant reduction in tumor size has been observed [123]. Another in vitro study demonstrated that panaxadiol was synergistic with irinotecan in HCT116 and SW480 tumor lines [124]. On the other hand, Rg3 showed synergy with 5-FU both in vitro and in vivo in SW620 and LOVO lines, having shown suppression of proliferation and tumor development and metastasis generation through PI3K/Akt activation [125].
Among all the terpenes that were studied, Rg3 has been the most encapsulated in formulations to increase its biological availability. Qiu et al. synthesized nanoformulations of Rg3 encapsulated in pH-sensitive poly (ethylene glycol) that increased the blood circulation of the compound showing rapid release and enhanced cytotoxicity against free drug in SW480, SW620, and CL40 tumor cells. Meanwhile, these formulations did not possess toxicity in the non-tumor line CCD-18Co at the concentrations that were tested. Meanwhile, in vivo experiments showed a greater reduction of tumor volume versus the free compound, increasing apoptosis in the tumor sinus [126]. Finally, Sun et al. synthesized folato-targeted polyethylene glycol nanoparticles based on cyclodextrins that co-encapsulated Rg3 and quercetin. The microtumor environment modulating effect that was demonstrated by Rg3 together with the oxidative stress inducing ability of quercetin exhibited a high cytotoxic effect on CRC lines CT26 and HCT116. The in vitro effect was reflected in in vivo experiments, where increased survival of mice was shown in combination with an anti-PD-L1 drug [127].
Rg3 was tested in clinical trials as a possible hepatocellular carcinoma treatment (NCT02724358 and NCT01717066), although it was not tested for CRC therapy as there is no sufficient knowledge of this compound in this type of carcinoma.

4. Discussion

Chemotherapeutic treatment of colon cancer has been compromised mainly by the appearance of resistance, which reduces therapeutic efficacy and leads to a lower cure rate and worse prognosis. These can be produced by the existence of previous mutations in genes that are involved in resistance, by the activation of cellular pathways that are involved in cellular detoxification, and the existence of transmembrane transporters that expel the drugs to the exterior (such as p-glycoprotein) [128]. In addition to this protein, other members of the transmembrane protein family such as MRP1 and BCRP are also overexpressed in CRC, and a relationship has been observed between their expression and resistance phenomena against drugs that are frequently used in this type of tumor, such as 5-FU and doxorubicin [30,31]. It has long been shown that plant compounds can be used as a therapy for different types of cancer. Thus, a plant-derived compound such as taxol has been used for years as a chemotherapeutic agent and is currently used as a therapy in non-small cell lung cancer (NSCLC), breast, pancreatic, and cervical cancer [129]. These types of compounds can exert their actions at the molecular level through processes such as the regulation of oxidative stress or epigenetic modification in cells [130] (Figure 3).
It has been shown that the most studied compounds of plant origin in CRC are polyphenols, specifically resveratrol and curcumin (Table 1). Resveratrol belongs to the stilbenes group and has been shown to possess high antioxidant and antitumor activity, causing cell death through the induction of apoptosis and autophagy. Similar to resveratrol, the other polyphenols that were studied induce death through these pathways [48,67,73,83].
Polyphenols have been classified as chemopreventives and chemotherapeutics, although their sensitizing effect has also been observed in in vitro and in vivo models. These compounds can modulate drug resistance by increasing drug internalization into the cell, decreasing enzymes that are responsible for drug degradation (such as glutathione-S-transferases and cytochromes) and reducing the expression of transmembrane detoxifying proteins in the cell. In addition, they can induce apoptosis, oxidative stress damage, and inhibit metastasis-triggering processes such as EMT [131]. The alteration of all these resistance mechanisms implies that these polyphenols have been shown to be effective in combination with traditional drugs such as 5-FU or oxaliplatin [56,73,75,87,88,89]. This effect is also observed in foods with high polyphenol contents such as the strawberry tree honey, that chemosensitizes the drug 5-FU in colon cancer lines such as HCT116 and LoVo [132]. In addition to this, it has been observed that curcumin and resveratrol had the capacity to inhibit tumor proliferation in in vivo models and prevented the formation of tumor precursor lesions, with an increase in apoptosis of induced tumor tissues [47,53,76]. Pharmacokinetic studies have been carried out in humans, where low bioavailability has been observed [47,52,54,71,75]. Given the low bioavailability of these compounds, the use of nanotechnology for their encapsulation could help stabilize the compound and prevent its degradation in blood. In addition, the use of nanotechnology allows specific targeting of tumor cells through their functionalization with antibodies or peptides [14,15].
On the other hand, it has been observed that flavonoid compounds also have anticarcinogenic potential. Among them, the most investigated in CRC have been quercetin and EGCG. These compounds exerted their cytotoxic effect by producing cycle arrest, inhibiting key pathways in tumor development such as PI3K/AKT/mTOR and the MAPK pathway, and inhibiting processes that are linked to tumor progression such as cell migration [93,99,100]. In addition, they showed a great chemosensitizing capacity in combination with traditional drugs such as doxorubicin, irinotecan, 5-FU, cisplatin, and oxaliplatin under in vitro and in vivo conditions. A study that was conducted by Hassanein et al. [133] showed the chemopreventive effect of EGCG administration together with sulindac, a non-steroidal anti-inflammatory drug, showing that it was able to decrease the production of neoplastic lesions in in vivo models of CRC. As polyphenols, these compounds have a low bioavailability, which is a limitation for their use in humans [18,24,87,88,89,97,101,102,103,105]. In this context, the synthesis of gold NPs encapsulating EGCG has been shown to be an effective therapy against tumor cells while it has been shown that the co-encapsulation of EGCG and 5-FU in NPs allows an increase in the effect of both compounds separately, producing an anti-angiogenic and pro-apoptotic effect [106,134]. The encapsulation of this compound in nanoformulations would increase the half-life of the compound in serum, increasing its bioavailability and increasing its antitumor effect [135].
The bibliographic analysis showed that terpenes are the least studied bioactive compounds as chemosensitizers in this type of cancer. Among this family, geraniol and ginsenosides have been the most studied compounds with sensitizing properties in CRC [114,117,118]. Due to the small number of studies that have been conducted on these compounds, it is complicated for these results to be transferred to clinical studies at present. These compounds exert their antiproliferative activity by producing cell cycle arrest and inducing apoptotic pathways. In addition, it has been observed that they suppress pathways essential for tumor development such as PI3K/AKT/mTOR [115,119,120]. Results in in vivo models showed that geraniol sensitized tumors that were induced in mice from a 5-FU-resistant CRC line to the drug, while the major ginsenoside (Rg3) showed synergy with 5-FU in tumors that were generated from the SW620 and LoVo cell lines. However, it has been shown that these compounds have clear preventive and therapeutic properties in CRC [115,116,123].
Therefore, after reviewing the existing literature, it is necessary to continue investigating the antitumor properties and possible chemosensitizing actions of these compounds, trying to transfer these results to future clinical trials in humans. In addition, it is important to study their bioavailability, trying to limit their elimination in blood using these natural compounds encapsulated in nanoformulations.

5. Conclusions

The bioactive compounds of plant origin that were described in this review have been shown to have therapeutic and chemosensitizing action in vitro and in vivo. However, their low bioavailability in the human body presents a serious limitation for their application in therapy. Future larger studies including clinical trials and the development of future effective nanoformulations to increase their bioavailability will be necessary to determine its real utility in improving the treatment of CRC.

Author Contributions

Conceptualization, J.P. and C.M (Consolación Melguizo).; methodology, F.Q., C.M. (Cristina Mesas) and M.P; software, L.C.; validation, L.C. and G.P.; formal analysis, F.Q.; investigation, F.Q., C.M. (Cristina Mesas) and M.P.; data curation, G.P.; writing—original draft preparation, F.Q., C.M. (Cristina Mesas) and M.P.; writing—review and editing, L.C., G.P. and R.O.; visualization, C.M. (Consolación Melguizo) and R.O; supervision, C.M (Consolación Melguizo), R.O. and J.P.; funding acquisition, J.P. and C.M (Consolación Melguizo). All authors have read and agreed to the published version of the manuscript.

Funding

This work was partially supported by the Project P20_00540 (Proyectos I+D+i Junta de Andalucía 2020), PYC20 RE 035 and P18-TP-1420 (Junta de Andalucía), PI19/01478 (Instituto de Salud Carlos III, Innbio INB-009 (Granada University and ibs. GRANADA) and RTC2019-006870-1 (Spanish Minsistry of Science, Innovation and Universities). FQ and MP acknowledges the FPU (2019 and 2020) grant from Ministerio de Educación, Ciencia y Deporte y Competitividad (Spain).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We thank Instrumentation Scientific Center (CIC) from University of Granada for technical assistance.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Hossain, M.S.; Karuniawati, H.; Jairoun, A.A.; Urbi, Z.; Ooi, D.J.; John, A.; Lim, Y.C.; Kaderi Kibria, K.M.; Mohiuddin, A.K.M.; Ming, L.C.; et al. Colorectal Cancer: A Review of Carcinogenesis, Global Epidemiology, Current Challenges, Risk Factors, Preventive and Treatment Strategies. Cancers 2022, 14, 1732. [Google Scholar] [CrossRef]
  2. Cervantes, A.; Adam, R.; Roselló, S.; Arnold, D.; Normanno, N.; Taïeb, J.; Seligmann, J.; de Baere, T.; Osterlund, P.; Yoshino, T.; et al. Metastatic Colorectal Cancer: ESMO Clinical Practice Guideline for Diagnosis, Treatment and Follow-Up. Ann. Oncol. 2023, 34, 10–32. [Google Scholar] [CrossRef] [PubMed]
  3. Wu, C. Systemic Therapy for Colon Cancer. Surg. Oncol. Clin. N. Am. 2018, 27, 235–242. [Google Scholar] [CrossRef] [PubMed]
  4. Dekker, E.; Tanis, P.J.; Vleugels, J.L.A.; Kasi, P.M.; Wallace, M.B. Colorectal Cancer. Lancet 2019, 394, 1467–1480. [Google Scholar] [CrossRef] [PubMed]
  5. Van der Jeught, K.; Xu, H.C.; Li, Y.J.; Lu, X.B.; Ji, G. Drug Resistance and New Therapies in Colorectal Cancer. World J. Gastroenterol. 2018, 24, 3834–3848. [Google Scholar] [CrossRef]
  6. Douillard, J.Y.; Cunningham, D.; Roth, A.D.; Navarro, M.; James, R.D.; Karasek, P.; Jandik, P.; Iveson, T.; Carmichael, J.; Alakl, M.; et al. Irinotecan Combined with Fluorouracil Compared with Fluorouracil Alone. as First-Line Treatment for Metastatic Colorectal Cancer: A Multicentre Randomised Trial. Lancet 2000, 355, 1041–1047. [Google Scholar] [CrossRef]
  7. Karthika, C.; Sureshkumar, R.; Zehravi, M.; Akter, R.; Ali, F.; Ramproshad, S.; Mondal, B.; Kundu, M.K.; Dey, A.; Rahman, M.H.; et al. Multidrug Resistance in Cancer Cells: Focus on a Possible Strategy Plan to Address Colon Carcinoma Cells. Life 2022, 12, 811. [Google Scholar] [CrossRef]
  8. Elfadadny, A.; El-Husseiny, H.M.; Abugomaa, A.; Ragab, R.F.; Mady, E.A.; Aboubakr, M.; Samir, H.; Mandour, A.S.; El-Mleeh, A.; El-Far, A.H.; et al. Role of Multidrug Resistance-Associated Proteins in Cancer Therapeutics: Past, Present, and Future Perspectives. Environ. Sci. Pollut. Res. 2021, 28, 49447–49466. [Google Scholar] [CrossRef]
  9. Ng, C.X.; Affendi, M.M.; Chong, P.P.; Lee, S.H. The Potential of Plant-Derived Extracts and Compounds to Augment Anticancer Effects of Chemotherapeutic Drugs. Nutr. Cancer 2022, 74, 3058–3076. [Google Scholar] [CrossRef]
  10. Veeresham, C. Natural Products Derived from Plants as a Source of Drugs. J. Adv. Pharm. Technol. Res. 2012, 3, 200–201. [Google Scholar] [CrossRef]
  11. Najmi, A.; Javed, S.A.; al Bratty, M.; Alhazmi, H.A. Modern Approaches in the Discovery and Development of Plant-Based Natural Products and Their Analogues as Potential Therapeutic Agents. Molecules 2022, 27, 349. [Google Scholar] [CrossRef] [PubMed]
  12. Islam, B.U.; Suhail, M.; Khan, M.K.; Zughaibi, T.A.; Alserihi, R.F.; Zaidi, S.K.; Tabrez, S. Polyphenols as Anticancer Agents: Toxicological Concern to Healthy Cells. Phytother. Res. 2021, 35, 6063–6079. [Google Scholar] [CrossRef] [PubMed]
  13. Kubczak, M.; Szustka, A.; Rogalińska, M. Molecular Targets of Natural Compounds with Anti-Cancer Properties. Int. J. Mol. Sci. 2021, 22, 13659. [Google Scholar] [CrossRef] [PubMed]
  14. Guo, Y.; Sun, Q.; Wu, F.-G.; Dai, Y.; Chen, X.; Guo, Y.; Sun, Q.; Wu, F.; Dai, Y.; Chen Yong Loo, X. Polyphenol-Containing Nanoparticles: Synthesis, Properties, and Therapeutic Delivery. Adv. Mater. 2021, 33, 2007356. [Google Scholar] [CrossRef]
  15. Mitchell, M.J.; Billingsley, M.M.; Haley, R.M.; Wechsler, M.E.; Peppas, N.A.; Langer, R. Engineering Precision Nanoparticles for Drug Delivery. Nat. Rev. Drug Discov. 2020, 20, 101–124. [Google Scholar] [CrossRef]
  16. Esmeeta, A.; Adhikary, S.; Dharshnaa, V.; Swarnamughi, P.; Ummul Maqsummiya, Z.; Banerjee, A.; Pathak, S.; Duttaroy, A.K. Plant-Derived Bioactive Compounds in Colon Cancer Treatment: An Updated Review. Biomed. Pharmacother. 2022, 153, 113384. [Google Scholar] [CrossRef]
  17. Wei, Y.; Yang, P.; Cao, S.; Zhao, L. The Combination of Curcumin and 5-Fluorouracil in Cancer Therapy. Arch. Pharmacal Res. 2017, 41, 1–13. [Google Scholar] [CrossRef]
  18. Wu, W.; Dong, J.; Gou, H.; Geng, R.; Yang, X.; Chen, D.; Xiang, B.; Zhang, Z.; Ren, S.; Chen, L.; et al. EGCG Synergizes the Therapeutic Effect of Irinotecan through Enhanced DNA Damage in Human Colorectal Cancer Cells. J. Cell Mol. Med. 2021, 25, 7913–7921. [Google Scholar] [CrossRef]
  19. Lu, W.D.; Qin, Y.; Yang, C.; Li, L.; Fu, Z.X. Effect of Curcumin on Human Colon Cancer Multidrug Resistance in Vitro and in Vivo. Clinics 2013, 68, 694–701. [Google Scholar] [CrossRef]
  20. Huang, L.; Zhang, S.; Zhou, J.; Li, X. Effect of Resveratrol on Drug Resistance in Colon Cancer Chemotherapy. RSC Adv. 2019, 9, 2572–2580. [Google Scholar] [CrossRef] [Green Version]
  21. Kamran, S.; Sinniah, A.; Abdulghani, M.A.M.; Alshawsh, M.A. Therapeutic Potential of Certain Terpenoids as Anticancer Agents: A Scoping Review. Cancers 2022, 14, 1100. [Google Scholar] [CrossRef] [PubMed]
  22. Costea, T.; Vlad, O.C.; Miclea, L.C.; Ganea, C.; Szöllősi, J.; Mocanu, M.M. Alleviation of Multidrug Resistance by Flavonoid and Non-Flavonoid Compounds in Breast, Lung, Colorectal and Prostate Cancer. Int. J. Mol. Sci. 2020, 21, 401. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Liskova, A.; Samec, M.; Koklesova, L.; Brockmueller, A.; Zhai, K.; Abdellatif, B.; Siddiqui, M.; Biringer, K.; Kudela, E.; Pec, M.; et al. Flavonoids as an Effective Sensitizer for Anti-Cancer Therapy: Insights into Multi-Faceted Mechanisms and Applicability towards Individualized Patient Profiles. EPMA J. 2021, 12, 155–176. [Google Scholar] [CrossRef] [PubMed]
  24. Zhou, Y.; Zhang, J.; Wang, K.; Han, W.; Wang, X.; Gao, M.; Wang, Z.; Sun, Y.; Yan, H.; Zhang, H.; et al. Quercetin Overcomes Colon Cancer Cells Resistance to Chemotherapy by Inhibiting Solute Carrier Family 1, Member 5 Transporter. Eur. J. Pharmacol. 2020, 881, 173185. [Google Scholar] [CrossRef] [PubMed]
  25. Bukowski, K.; Kciuk, M.; Kontek, R. Mechanisms of Multidrug Resistance in Cancer Chemotherapy. Int. J. Mol. Sci. 2020, 21, 3233. [Google Scholar] [CrossRef] [PubMed]
  26. Wang, X.; Zhang, H.; Chen, X. Drug Resistance and Combating Drug Resistance in Cancer. Cancer Drug Resist. 2019, 2, 141–160. [Google Scholar] [CrossRef] [Green Version]
  27. Housman, G.; Byler, S.; Heerboth, S.; Lapinska, K.; Longacre, M.; Snyder, N.; Sarkar, S. Drug Resistance in Cancer: An Overview. Cancers 2014, 6, 1769–1792. [Google Scholar] [CrossRef] [Green Version]
  28. Linn, S.C.; Giaccone, G. MDR1/P-Glycoprotein Expression in Colorectal Cancer. Eur. J. Cancer 1995, 31A, 1291–1294. [Google Scholar] [CrossRef]
  29. Yuan, Z.; Liang, X.; Zhan, Y.; Wang, Z.; Xu, J.; Qiu, Y.; Wang, J.; Cao, Y.; Le, V.-M.; Ly, H.-T.; et al. Targeting CD133 Reverses Drug-Resistance via the AKT/NF-ΚB/MDR1 Pathway in Colorectal Cancer. Br. J. Cancer 2020, 122, 1342–1353. [Google Scholar] [CrossRef]
  30. Cao, D.; Qin, S.; Mu, Y.; Zhong, M. The Role of MRP1 in the Multidrug Resistance of Colorectal Cancer. Oncol. Lett. 2017, 13, 2471–2476. [Google Scholar] [CrossRef] [Green Version]
  31. Hu, T.; Li, Z.; Gao, C.-Y.; Cho, C.H. Mechanisms of Drug Resistance in Colon Cancer and Its Therapeutic Strategies. World J. Gastroenterol. 2016, 22, 6876–6889. [Google Scholar] [CrossRef] [PubMed]
  32. Wang, Z.; Sun, X.; Feng, Y.; Wang, Y.; Zhang, L.; Wang, Y.; Fang, Z.; Azami, N.L.B.; Sun, M.; Li, Q. Dihydromyricetin Reverses MRP2-Induced Multidrug Resistance by Preventing NF-ΚB-Nrf2 Signaling in Colorectal Cancer Cell. Phytomedicine 2021, 82, 153414. [Google Scholar] [CrossRef] [PubMed]
  33. Asano, T. Drug Resistance in Cancer Therapy and the Role of Epigenetics. J. Nippon. Med. Sch. 2020, 87, 244–251. [Google Scholar] [CrossRef] [PubMed]
  34. Das, P.K.; Islam, F.; Lam, A.K. The Roles of Cancer Stem Cells and Therapy Resistance in Colorectal Carcinoma. Cells 2020, 9, 1392. [Google Scholar] [CrossRef]
  35. Neophytou, C.M.; Trougakos, I.P.; Erin, N.; Papageorgis, P. Apoptosis Deregulation and the Development of Cancer Multi-Drug Resistance. Cancers 2021, 13, 4363. [Google Scholar] [CrossRef]
  36. Prasetyanti, P.R.; Medema, J.P. Intra-Tumor Heterogeneity from a Cancer Stem Cell Perspective. Mol. Cancer 2017, 16, 41. [Google Scholar] [CrossRef] [Green Version]
  37. Phi, L.T.H.; Sari, I.N.; Yang, Y.G.; Lee, S.H.; Jun, N.; Kim, K.S.; Lee, Y.K.; Kwon, H.Y. Cancer Stem Cells (CSCs) in Drug Resistance and Their Therapeutic Implications in Cancer Treatment. Stem. Cells Int. 2018, 2018, 5416923. [Google Scholar] [CrossRef] [Green Version]
  38. Burrell, R.A.; Swanton, C. Tumour Heterogeneity and the Evolution of Polyclonal Drug Resistance. Mol. Oncol. 2014, 8, 1095–1111. [Google Scholar] [CrossRef]
  39. Junttila, M.R.; de Sauvage, F.J. Influence of Tumour Micro-Environment Heterogeneity on Therapeutic Response. Nature 2013, 501, 346–354. [Google Scholar] [CrossRef]
  40. Dutta, S.; Mahalanobish, S.; Saha, S.; Ghosh, S.; Sil, P.C. Natural Products: An Upcoming Therapeutic Approach to Cancer. Food Chem. Toxicol. 2019, 128, 240–255. [Google Scholar] [CrossRef]
  41. Castañeda, A.M.; Meléndez, C.M.; Uribe, D.; Pedroza-Díaz, J. Synergistic Effects of Natural Compounds and Conventional Chemotherapeutic Agents: Recent Insights for the Development of Cancer Treatment Strategies. Heliyon 2022, 8, e09519. [Google Scholar] [CrossRef]
  42. Rejhová, A.; Opattová, A.; Čumová, A.; Slíva, D.; Vodička, P. Natural Compounds and Combination Therapy in Colorectal Cancer Treatment. Eur. J. Med. Chem. 2018, 144, 582–594. [Google Scholar] [CrossRef]
  43. Redondo-Blanco, S.; Fernández, J.; Gutiérrez-del-Río, I.; Villar, C.J.; Lombó, F. New Insights toward Colorectal Cancer Chemotherapy Using Natural Bioactive Compounds. Front. Pharmacol. 2017, 8, 109. [Google Scholar] [CrossRef] [Green Version]
  44. Durazzo, A.; Lucarini, M.; Souto, E.B.; Cicala, C.; Caiazzo, E.; Izzo, A.A.; Novellino, E.; Santini, A. Polyphenols: A Concise Overview on the Chemistry, Occurrence, and Human Health. Phytother. Res. 2019, 33, 2221–2243. [Google Scholar] [CrossRef] [Green Version]
  45. Mileo, A.M.; Nisticò, P.; Miccadei, S. Polyphenols: Immunomodulatory and Therapeutic Implication in Colorectal Cancer. Front. Immunol. 2019, 10, 729. [Google Scholar] [CrossRef]
  46. Bracci, L.; Fabbri, A.; del Cornò, M.; Conti, L. Dietary Polyphenols: Promising Adjuvants for Colorectal Cancer Therapies. Cancers 2021, 13, 4499. [Google Scholar] [CrossRef]
  47. Ko, J.H.; Sethi, G.; Um, J.Y.; Shanmugam, M.K.; Arfuso, F.; Kumar, A.P.; Bishayee, A.; Ahn, K.S. The Role of Resveratrol in Cancer Therapy. Int. J. Mol. Sci. 2017, 18, 2589. [Google Scholar] [CrossRef] [Green Version]
  48. Elshaer, M.; Chen, Y.; Wang, X.J.; Tang, X. Resveratrol: An Overview of Its Anti-Cancer Mechanisms. Life Sci. 2018, 207, 340–349. [Google Scholar] [CrossRef] [PubMed]
  49. Fuggetta, M.P.; Lanzilli, G.; Tricarico, M.; Cottarelli, A.; Falchetti, R.; Ravagnan, G.; Bonmassar, E. Effect of Resveratrol on Proliferation and Telomerase Activity of Human Colon Cancer Cells in Vitro. J. Exp. Clin. Cancer Res. 2006, 25, 189–193. [Google Scholar] [PubMed]
  50. Rotelli, M.T.; Bocale, D.; de Fazio, M.; Ancona, P.; Scalera, I.; Memeo, R.; Travaglio, E.; Zbar, A.P.; Altomare, D.F. IN-VITRO Evidence for the Protective Properties of the Main Components of the Mediterranean Diet against Colorectal Cancer: A Systematic Review. Surg Oncol. 2015, 24, 145–152. [Google Scholar] [CrossRef] [PubMed]
  51. Saunier, E.; Antonio, S.; Regazzetti, A.; Auzeil, N.; Laprévote, O.; Shay, J.W.; Coumoul, X.; Barouki, R.; Benelli, C.; Huc, L.; et al. Resveratrol Reverses the Warburg Effect by Targeting the Pyruvate Dehydrogenase Complex in Colon Cancer Cells. Sci. Rep. 2017, 7, 6945. [Google Scholar] [CrossRef] [Green Version]
  52. Saud, S.M.; Li, W.; Morris, N.L.; Matter, M.S.; Colburn, N.H.; Kim, Y.S.; Young, M.R. Resveratrol Prevents Tumorigenesis in Mouse Model of Kras Activated Sporadic Colorectal Cancer by Suppressing Oncogenic Kras Expression. Carcinogenesis 2014, 35, 2778–2786. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Carter, L.G.; D’Orazio, J.A.; Pearson, K.J. Resveratrol and Cancer: Focus on in Vivo Evidence. Endocr. Relat. Cancer 2014, 21, R209–R225. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Aires, V.; Limagne, E.; Cotte, A.K.; Latruffe, N.; Ghiringhelli, F.; Delmas, D. Resveratrol Metabolites Inhibit Human Metastatic Colon Cancer Cells Progression and Synergize with Chemotherapeutic Drugs to Induce Cell Death. Mol. Nutr. Food Res. 2013, 57, 1170–1181. [Google Scholar] [CrossRef] [PubMed]
  55. Cottart, C.H.; Nivet-Antoine, V.; Laguillier-Morizot, C.; Beaudeux, J.L. Resveratrol Bioavailability and Toxicity in Humans. Mol. Nutr. Food Res. 2010, 54, 7–16. [Google Scholar] [CrossRef]
  56. Huang, X.M.; Yang, Z.J.; Xie, Q.; Zhang, Z.K.; Zhang, H.; Ma, J.Y. Natural Products for Treating Colorectal Cancer: A Mechanistic Review. Biomed Pharm. 2019, 117, 109142. [Google Scholar] [CrossRef] [PubMed]
  57. Abdel Latif, Y.; El-Bana, M.; Hussein, J.; El-Khayat, Z.; Farrag, A.R. Effects of Resveratrol in Combination with 5-Fluorouracil on N-Methylnitrosourea-Induced Colon Cancer in Rats. Comp. Clin. Path 2019, 28, 1351–1362. [Google Scholar] [CrossRef]
  58. Buhrmann, C.; Yazdi, M.; Popper, B.; Shayan, P.; Goel, A.; Aggarwal, B.B.; Shakibaei, M. Resveratrol Chemosensitizes TNF-β-Induced Survival of 5-FU-Treated Colorectal Cancer Cells. Nutrients 2018, 10, 888. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Yang, S.; Li, W.; Sun, H.; Wu, B.; Ji, F.; Sun, T.; Chang, H.; Shen, P.; Wang, Y.; Zhou, D. Resveratrol Elicits Anti-Colorectal Cancer Effect by Activating MiR-34c-KITLG in Vitro and in Vivo. BMC Cancer 2015, 15, 969. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Kaminski, B.M.; Weigert, A.; Scherzberg, M.C.; Ley, S.; Gilbert, B.; Brecht, K.; Brüne, B.; Steinhilber, D.; Stein, J.; Ulrich-Rückert, S. Resveratrol-Induced Potentiation of the Antitumor Effects of Oxaliplatin Is Accompanied by an Altered Cytokine Profile of Human Monocyte-Derived Macrophages. Apoptosis 2014, 19, 1136–1147. [Google Scholar] [CrossRef]
  61. Khayat, M.T.; Zarka, M.A.; El-Telbany, D.; Farag, A.; El-Halawany, A.M.; Kutbi, H.I.; Elkhatib, W.F.; Noreddin, A.M.; Khayyat, A.N.; El-Telbany, R.F.A.; et al. Intensification of Resveratrol Cytotoxicity, pro-Apoptosis, Oxidant Potentials in Human Colorectal Carcinoma HCT-116 Cells Using Zein Nanoparticles. Sci. Rep. 2022, 12, 15235. [Google Scholar] [CrossRef] [PubMed]
  62. Md, S.; Abdullah, S.; Alhakamy, N.A.; Alharbi, W.S.; Ahmad, J.; Shaik, R.A.; Ansari, M.J.; Ibrahim, I.M.; Ali, J. Development, Optimization, and In Vitro Evaluation of Novel Oral Long-Acting Resveratrol Nanocomposite In-Situ Gelling Film in the Treatment of Colorectal Cancer. Gels 2021, 7, 276. [Google Scholar] [CrossRef] [PubMed]
  63. Feng, M.; Zhong, L.-X.; Zhan, Z.-Y.; Huang, Z.-H.; Xiong, J.-P. Enhanced Antitumor Efficacy of Resveratrol-Loaded Nanocapsules in Colon Cancer Cells: Physicochemical and Biological Characterization. Eur. Rev. Med. Pharmacol. Sci. 2017, 21, 375–382. [Google Scholar] [PubMed]
  64. Summerlin, N.; Qu, Z.; Pujara, N.; Sheng, Y.; Jambhrunkar, S.; McGuckin, M.; Popat, A. Colloidal Mesoporous Silica Nanoparticles Enhance the Biological Activity of Resveratrol. Colloids Surf. B Biointerfaces 2016, 144, 1–7. [Google Scholar] [CrossRef] [Green Version]
  65. Soo, E.; Thakur, S.; Qu, Z.; Jambhrunkar, S.; Parekh, H.S.; Popat, A. Enhancing Delivery and Cytotoxicity of Resveratrol through a Dual Nanoencapsulation Approach. J. Colloid Interface Sci. 2016, 462, 368–374. [Google Scholar] [CrossRef] [Green Version]
  66. Montané, X.; Kowalczyk, O.; Reig-Vano, B.; Bajek, A.; Roszkowski, K.; Tomczyk, R.; Pawliszak, W.; Giamberini, M.; Mocek-Płóciniak, A.; Tylkowski, B. Current Perspectives of the Applications of Polyphenols and Flavonoids in Cancer Therapy. Molecules 2020, 25, 3342. [Google Scholar] [CrossRef]
  67. Weng, W.; Goel, A. Curcumin and Colorectal Cancer: An Update and Current Perspective on This Natural Medicine. Semin. Cancer Biol. 2022, 80, 73–86. [Google Scholar] [CrossRef]
  68. Shehzad, A.; Wahid, F.; Lee, Y.S. Curcumin in Cancer Chemoprevention: Molecular Targets, Pharmacokinetics, Bioavailability, and Clinical Trials. Arch. Pharm. 2010, 343, 489–499. [Google Scholar] [CrossRef]
  69. Jaiswal, A.S.; Marlow, B.P.; Gupta, N.; Narayan, S. Beta-Catenin-Mediated Transactivation and Cell-Cell Adhesion Pathways Are Important in Curcumin (Diferuylmethane)-Induced Growth Arrest and Apoptosis in Colon Cancer Cells. Oncogene 2002, 21, 8414–8427. [Google Scholar] [CrossRef] [Green Version]
  70. Basbinar, Y.; Calibasi-Kocal, G.; Pakdemirli, A.; Bayrak, S.; Ozupek, N.M.; Sever, T.; Ellidokuz, H.; Yigitbasi, T. Curcumin Effects on Cell Proliferation, Angiogenesis and Metastasis in Colorectal Cancer. JBUON 2019, 24, 1482–1487. [Google Scholar]
  71. Pricci, M.; Girardi, B.; Giorgio, F.; Losurdo, G.; Ierardi, E.; di Leo, A. Curcumin and Colorectal Cancer: From Basic to Clinical Evidences. Int. J. Mol. Sci. 2020, 21, 2364. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Tunstall, R.G.; Sharma, R.A.; Perkins, S.; Sale, S.; Singh, R.; Farmer, P.B.; Steward, W.P.; Gescher, A.J. Cyclooxygenase-2 Expression and Oxidative DNA Adducts in Murine Intestinal Adenomas: Modification by Dietary Curcumin and Implications for Clinical Trials. Eur. J. Cancer 2006, 42, 415–421. [Google Scholar] [CrossRef] [PubMed]
  73. Yin, T.F.; Wang, M.; Qing, Y.; Lin, Y.M.; Wu, D. Research Progress on Chemopreventive Effects of Phytochemicals on Colorectal Cancer and Their Mechanisms. World J. Gastroenterol. 2016, 22, 7068. [Google Scholar] [CrossRef] [PubMed]
  74. Yin, J.; Wang, L.; Wang, Y.; Shen, H.; Wang, X.; Wu, L. Curcumin Reverses Oxaliplatin Resistance in Human Colorectal Cancer via Regulation of TGF-β/Smad2/3 Signaling Pathway. Onco. Targets Ther. 2019, 12, 3893–3903. [Google Scholar] [CrossRef] [Green Version]
  75. Zheng, X.; Yang, X.; Lin, J.; Song, F.; Shao, Y. Low Curcumin Concentration Enhances the Anticancer Effect of 5-Fluorouracil against Colorectal Cancer. Phytomedicine 2021, 85, 153547. [Google Scholar] [CrossRef]
  76. Hosseini, S.A.; Zand, H.; Cheraghpour, M. The Influence of Curcumin on the Downregulation of MYC, Insulin and IGF-1 Receptors: A Possible Mechanism Underlying the Anti-Growth and Anti-Migration in Chemoresistant Colorectal Cancer Cells. Medicina 2019, 55, 90. [Google Scholar] [CrossRef] [Green Version]
  77. Han, Z.; Song, B.; Yang, J.; Wang, B.; Ma, Z.; Yu, L.; Li, Y.; Xu, H.; Qiao, M. Curcumin-Encapsulated Fusion Protein-Based Nanocarrier Demonstrated Highly Efficient Epidermal Growth Factor Receptor-Targeted Treatment of Colorectal Cancer. J. Agric. Food Chem. 2022, 70, 15464–15473. [Google Scholar] [CrossRef]
  78. Wang, A.; Jain, S.; Dia, V.; Lenaghan, S.C.; Zhong, Q. Shellac Micelles Loaded with Curcumin Using a PH Cycle to Improve Dispersibility, Bioaccessibility, and Potential for Colon Delivery. J. Agric. Food Chem. 2022, 70, 15166–15177. [Google Scholar] [CrossRef]
  79. Elbassiouni, F.E.; El-Kholy, W.M.; Elhabibi, E.-S.M.; Albogami, S.; Fayad, E. Comparative Study between Curcumin and Nanocurcumin Loaded PLGA on Colon Carcinogenesis Induced Mice. Nanomaterials 2022, 12, 324. [Google Scholar] [CrossRef]
  80. Low, Z.X.; Teo, M.Y.M.; Nordin, F.J.; Dewi, F.R.P.; Palanirajan, V.K.; In, L.L.A. Biophysical Evaluation of Water-Soluble Curcumin Encapsulated in β-Cyclodextrins on Colorectal Cancer Cells. Int. J. Mol. Sci. 2022, 23, 12866. [Google Scholar] [CrossRef]
  81. Gupta, A.; Sood, A.; Dhiman, A.; Shrimali, N.; Singhmar, R.; Guchhait, P.; Agrawal, G. Redox Responsive Poly(Allylamine)/Eudragit S-100 Nanoparticles for Dual Drug Delivery in Colorectal Cancer. Biomater. Adv. 2022, 143, 213184. [Google Scholar] [CrossRef]
  82. Imran, M.; Salehi, B.; Sharifi-Rad, J.; Gondal, T.A.; Saeed, F.; Imran, A.; Shahbaz, M.; Fokou, P.V.T.; Arshad, M.U.; Khan, H.; et al. Kaempferol: A Key Emphasis to Its Anticancer Potential. Molecules 2019, 24, 2277. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Chen, A.Y.; Chen, Y.C. A Review of the Dietary Flavonoid, Kaempferol on Human Health and Cancer Chemoprevention. Food Chem. 2013, 138, 2099–2107. [Google Scholar] [CrossRef] [Green Version]
  84. Schindler, R.; Mentlein, R. Flavonoids and Vitamin E Reduce the Release of the Angiogenic Peptide Vascular Endothelial Growth Factor from Human Tumor Cells. J. Nutr. 2006, 136, 1477–1482. [Google Scholar] [CrossRef] [Green Version]
  85. Barve, A.; Chen, C.; Hebbar, V.; Desiderio, J.; Saw, C.L.-L.; Kong, A.-N. Metabolism, Oral Bioavailability and Pharmacokinetics of Chemopreventive Kaempferol in Rats. Biopharm. Drug Dispos. 2009, 30, 356–365. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Cho, H.J.; Park, J.H.Y. Kaempferol Induces Cell Cycle Arrest in HT-29 Human Colon Cancer Cells. J. Cancer Prev. 2013, 18, 257–263. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Riahi-Chebbi, I.; Souid, S.; Othman, H.; Haoues, M.; Karoui, H.; Morel, A.; Srairi-Abid, N.; Essafi, M.; Essafi-Benkhadir, K. The Phenolic Compound Kaempferol Overcomes 5-Fluorouracil Resistance in Human Resistant LS174 Colon Cancer Cells. Sci. Rep. 2019, 9, 195. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Li, Q.; Wei, L.; Lin, S.; Chen, Y.; Lin, J.; Peng, J. Synergistic Effect of Kaempferol and 5-fluorouracil on the Growth of Colorectal Cancer Cells by Regulating the PI3K/Akt Signaling Pathway. Mol. Med. Rep. 2019, 20, 728–734. [Google Scholar] [CrossRef] [PubMed]
  89. Park, J.; Lee, G.E.; An, H.J.; Lee, C.J.; Cho, E.S.; Kang, H.C.; Lee, J.Y.; Lee, H.S.; Choi, J.S.; Kim, D.J.; et al. Kaempferol Sensitizes Cell Proliferation Inhibition in Oxaliplatin-Resistant Colon Cancer Cells. Arch. Pharm. Res. 2021, 44, 1091–1108. [Google Scholar] [CrossRef] [PubMed]
  90. Meena, D.; Vimala, K.; Kannan, S. Combined Delivery of DOX and Kaempferol Using PEGylated Gold Nanoparticles to Target Colon Cancer. J. Clust. Sci. 2022, 33, 173–187. [Google Scholar] [CrossRef]
  91. Kumar, S.; Pandey, A.K. Chemistry and Biological Activities of Flavonoids: An Overview. Sci. World J. 2013, 2013, 162750. [Google Scholar] [CrossRef] [Green Version]
  92. Tang, S.M.; Deng, X.T.; Zhou, J.; Li, Q.P.; Ge, X.X.; Miao, L. Pharmacological Basis and New Insights of Quercetin Action in Respect to Its Anti-Cancer Effects. Biomed Pharm. 2020, 121, 109604. [Google Scholar] [CrossRef] [PubMed]
  93. Reyes-Farias, M.; Carrasco-Pozo, C. The Anti-Cancer Effect of Quercetin: Molecular Implications in Cancer Metabolism. Int. J. Mol. Sci. 2019, 20, 3177. [Google Scholar] [CrossRef] [Green Version]
  94. Kee, J.Y.; Han, Y.H.; Kim, D.S.; Mun, J.G.; Park, J.; Jeong, M.Y.; Um, J.Y.; Hong, S.H. Inhibitory Effect of Quercetin on Colorectal Lung Metastasis through Inducing Apoptosis, and Suppression of Metastatic Ability. Phytomedicine 2016, 23, 1680–1690. [Google Scholar] [CrossRef] [PubMed]
  95. Srivastava, N.S.; Srivastava, R.A.K. Curcumin and Quercetin Synergistically Inhibit Cancer Cell Proliferation in Multiple Cancer Cells and Modulate Wnt/β-Catenin Signaling and Apoptotic Pathways in A375 Cells. Phytomedicine 2019, 52, 117–128. [Google Scholar] [CrossRef]
  96. Neamtu, A.A.; Maghiar, T.A.; Alaya, A.; Olah, N.K.; Turcus, V.; Pelea, D.; Totolici, B.D.; Neamtu, C.; Maghiar, A.M.; Mathe, E. A Comprehensive View on the Quercetin Impact on Colorectal Cancer. Molecules 2022, 27, 1973. [Google Scholar] [CrossRef] [PubMed]
  97. Chang, C.E.; Hsieh, C.M.; Huang, S.C.; Su, C.Y.; Sheu, M.T.; Ho, H.O. Lecithin-Stabilized Polymeric Micelles (LsbPMs) for Delivering Quercetin: Pharmacokinetic Studies and Therapeutic Effects of Quercetin Alone and in Combination with Doxorubicin. Sci. Rep. 2018, 8, 17640. [Google Scholar] [CrossRef] [Green Version]
  98. Wen, P.; Hu, T.-G.; Li, L.; Zong, M.-H.; Wu, H. A Colon-Specific Delivery System for Quercetin with Enhanced Cancer Prevention Based on Co-Axial Electrospinning. Food Funct. 2018, 9, 5999–6009. [Google Scholar] [CrossRef]
  99. Almatrood, S.A.; Almatroudi, A.; Khan, A.A.; Alhumaydh, F.A.; Alsahl, M.A.; Rahmani, A.H. Potential Therapeutic Targets of Epigallocatechin Gallate (EGCG), the Most Abundant Catechin in Green Tea, and Its Role in the Therapy of Various Types of Cancer. Molecules 2020, 25, 3146. [Google Scholar] [CrossRef] [PubMed]
  100. Luo, K.W.; Xia, J.; Cheng, B.H.; Gao, H.C.; Fu, L.W.; Luo, X. le Tea Polyphenol EGCG Inhibited Colorectal-Cancer-Cell Proliferation and Migration via Downregulation of STAT3. Gastroenterol. Rep. 2020, 9, 59–70. [Google Scholar] [CrossRef] [PubMed]
  101. Wubetu, G.Y.; Shimada, M.; Morine, Y.; Ikemoto, T.; Ishikawa, D.; Iwahashi, S.; Yamada, S.; Saito, Y.; Arakawa, Y.; Imura, S. Epigallocatechin Gallate Hinders Human Hepatoma and Colon Cancer Sphere Formation. J. Gastroenterol. Hepatol. 2016, 31, 256–264. [Google Scholar] [CrossRef] [PubMed]
  102. Toden, S.; Tran, H.M.; Tovar-Camargo, O.A.; Okugawa, Y.; Goel, A. Epigallocatechin-3-Gallate Targets Cancer Stem-like Cells and Enhances 5-Fluorouracil Chemosensitivity in Colorectal Cancer. Oncotarget 2016, 7, 16158–16171. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. La, X.; Zhang, L.; Li, Z.; Li, H.; Yang, Y. (-)-Epigallocatechin Gallate (EGCG) Enhances the Sensitivity of Colorectal Cancer Cells to 5-FU by Inhibiting GRP78/NF-ΚB/MiR-155-5p/MDR1 Pathway. J. Agric. Food Chem. 2019, 67, 2510–2518. [Google Scholar] [CrossRef]
  104. Maruyama, T.; Murata, S.; Nakayama, K.; Sano, N.; Ogawa, K.; Nowatari, T.; Tamura, T.; Nozaki, R.; Fukunaga, K.; Ohkohchi, N. (-)-Epigallocatechin-3-Gallate Suppresses Liver Metastasis of Human Colorectal Cancer. Oncol. Rep. 2014, 31, 625–633. [Google Scholar] [CrossRef] [Green Version]
  105. Hu, F.; Wei, F.; Wang, Y.; Wu, B.; Fang, Y.; Xiong, B. EGCG Synergizes the Therapeutic Effect of Cisplatin and Oxaliplatin through Autophagic Pathway in Human Colorectal Cancer Cells. J. Pharmacol. Sci. 2015, 128, 27–34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Wang, R.; Huang, J.; Chen, J.; Yang, M.; Wang, H.; Qiao, H.; Chen, Z.; Hu, L.; Di, L.; Li, J. Enhanced Anti-Colon Cancer Efficacy of 5-Fluorouracil by Epigallocatechin-3- Gallate Co-Loaded in Wheat Germ Agglutinin-Conjugated Nanoparticles. Nanomedicine 2019, 21, 102068. [Google Scholar] [CrossRef]
  107. Seufferlein, T.; Ettrich, T.J.; Menzler, S.; Messmann, H.; Kleber, G.; Zipprich, A.; Frank-Gleich, S.; Algül, H.; Metter, K.; Odemar, F.; et al. Green Tea Extract to Prevent Colorectal Adenomas, Results of a Randomized, Placebo-Controlled Clinical Trial. Am. J. Gastroenterol. 2022, 117, 884–894. [Google Scholar] [CrossRef]
  108. Abaza, M.S.I.; Orabi, K.Y.; Al-Quattan, E.; Al-Attiyah, R.J. Growth Inhibitory and Chemo-Sensitization Effects of Naringenin, a Natural Flavanone Purified from Thymus Vulgaris, on Human Breast and Colorectal Cancer. Cancer Cell Int. 2015, 15, 46. [Google Scholar] [CrossRef] [Green Version]
  109. Song, H.M.; Park, G.H.; Eo, H.J.; Lee, J.W.; Kim, M.K.; Lee, J.R.; Lee, M.H.; Koo, J.S.; Jeong, J.B. Anti-Proliferative Effect of Naringenin through P38-Dependent Downregulation of Cyclin D1 in Human Colorectal Cancer Cells. Biomol. Ther. 2015, 23, 339–344. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Rehman, M.U.; Rahman Mir, M.U.; Farooq, A.; Rashid, S.M.; Ahmad, B.; Bilal Ahmad, S.; Ali, R.; Hussain, I.; Masoodi, M.; Muzamil, S.; et al. Naringenin (4,5,7-Trihydroxyflavanone) Suppresses the Development of Precancerous Lesions via Controlling Hyperproliferation and Inflammation in the Colon of Wistar Rats. Environ. Toxicol. 2018, 33, 422–435. [Google Scholar] [CrossRef]
  111. Md, S.; Abdullah, S.; Awan, Z.A.; Alhakamy, N.A. Smart Oral PH-Responsive Dual Layer Nano-Hydrogel for Dissolution Enhancement and Targeted Delivery of Naringenin Using Protein-Polysaccharides Complexation Against Colorectal Cancer. J. Pharm. Sci. 2022, 111, 3155–3164. [Google Scholar] [CrossRef] [PubMed]
  112. Wesołowska, O.; Wiśniewski, J.; Środa-Pomianek, K.; Bielawska-Pohl, A.; Paprocka, M.; Duś, D.; Duarte, N.; Ferreira, M.J.U.; Michalak, K. Multidrug Resistance Reversal and Apoptosis Induction in Human Colon Cancer Cells by Some Flavonoids Present in Citrus Plants. J. Nat. Prod. 2012, 75, 1896–1902. [Google Scholar] [CrossRef] [PubMed]
  113. Bai, Y.; Xiong, Y.; Zhang, Y.Y.; Cheng, L.; Liu, H.; Xu, K.; Wu, Y.Y.; Field, J.; Wang, X.D.; Zhou, L.M. Tangeretin Synergizes with 5-Fluorouracil to Induce Autophagy through MicroRNA-21 in Colorectal Cancer Cells. Am. J. Chin. Med. 2022, 50, 1681–1701. [Google Scholar] [CrossRef]
  114. De Cássia Da Silveira, E.; Sá, R.; Andrade, L.N.; de Sousa, D.P. A Review on Anti-Inflammatory Activity of Monoterpenes. Molecules 2013, 18, 1227–1254. [Google Scholar] [CrossRef] [PubMed]
  115. Cho, M.; So, I.; Chun, J.N.; Jeon, J.H. The Antitumor Effects of Geraniol: Modulation of Cancer Hallmark Pathways (Review). Int. J. Oncol. 2016, 48, 1772–1782. [Google Scholar] [CrossRef] [Green Version]
  116. Vieira, A.; Heidor, R.; Cardozo, M.T.; Scolastici, C.; Purgatto, E.; Shiga, T.M.; Barbisan, L.F.; Ong, T.P.; Moreno, F.S. Efficacy of Geraniol but Not of β-Ionone or Their Combination for the Chemoprevention of Rat Colon Carcinogenesis. Braz. J. Med. Biol. Res. 2011, 44, 538–545. [Google Scholar] [CrossRef]
  117. Carnesecchi, S.; Bras-Gonçalves, R.; Bradaia, A.; Zeisel, M.; Gossé, F.; Poupon, M.F.; Raul, F. Geraniol, a Component of Plant Essential Oils, Modulates DNA Synthesis and Potentiates 5-Fluorouracil Efficacy on Human Colon Tumor Xenografts. Cancer Lett. 2004, 215, 53–59. [Google Scholar] [CrossRef]
  118. Carnesecchi, S.; Langley, K.; Exinger, F.; Gosse, F.; Raul, F. Geraniol, a Component of Plant Essential Oils, Sensitizes Human Colonic Cancer Cells to 5-Fluorouracil Treatment. J. Pharmacol. Exp. Ther. 2002, 301, 625–630. [Google Scholar] [CrossRef] [Green Version]
  119. Gao, J.L.; Lv, G.Y.; He, B.C.; Zhang, B.Q.; Zhang, H.; Wang, N.; Wang, C.Z.; Du, W.; Yuan, C.S.; He, T.C. Ginseng Saponin Metabolite 20(S)-Protopanaxadiol Inhibits Tumor Growth by Targeting Multiple Cancer Signaling Pathways. Oncol. Rep. 2013, 30, 292–298. [Google Scholar] [CrossRef] [Green Version]
  120. Wang, Z.; Li, M.Y.; Zhang, Z.H.; Zuo, H.X.; Wang, J.Y.; Xing, Y.; Ri, M.H.; Jin, H.L.; Jin, C.H.; Xu, G.H.; et al. Panaxadiol Inhibits Programmed Cell Death-Ligand 1 Expression and Tumour Proliferation via Hypoxia-Inducible Factor (HIF)-1α and STAT3 in Human Colon Cancer Cells. Pharmacol. Res. 2020, 155, 104727. [Google Scholar] [CrossRef]
  121. Yuan, H.D.; Quan, H.Y.; Zhang, Y.; Kim, S.H.; Chung, S.H. 20(S)-Ginsenoside Rg3-Induced Apoptosis in HT-29 Colon Cancer Cells Is Associated with AMPK Signaling Pathway. Mol. Med. Rep. 2010, 3, 825–831. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Tang, Y.C.; Zhang, Y.; Zhou, J.; Zhi, Q.; Wu, M.Y.; Gong, F.R.; Shen, M.; Liu, L.; Tao, M.; Shen, B.; et al. Ginsenoside Rg3 Targets Cancer Stem Cells and Tumor Angiogenesis to Inhibit Colorectal Cancer Progression in Vivo. Int. J. Oncol. 2018, 52, 127–138. [Google Scholar] [CrossRef] [Green Version]
  123. Wang, C.Z.; Zhang, Z.; Wan, J.Y.; Zhang, C.F.; Anderson, S.; He, X.; Yu, C.; He, T.C.; Qi, L.W.; Yuan, C.S. Protopanaxadiol, an Active Ginseng Metabolite, Significantly Enhances the Effects of Fluorouracil on Colon Cancer. Nutrients 2015, 7, 799–814. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Du, G.-J.; Wang, C.-Z.; Zhang, Z.-Y.; Wen, X.-D.; Somogyi, J.; Calway, T.; He, T.-C.; Du, W.; Yuan, C.-S. Caspase-Mediated pro-Apoptotic Interaction of Panaxadiol and Irinotecan in Human Colorectal Cancer Cells. J. Pharm. Pharmacol. 2012, 64, 727–734. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Hong, S.; Cai, W.; Huang, Z.; Wang, Y.; Mi, X.; Huang, Y.; Lin, Z.; Chen, X. Ginsenoside Rg3 Enhances the Anticancer Effect of 5-FU in Colon Cancer Cells via the PI3K/AKTpathway. Oncol. Rep. 2020, 44, 1333–1342. [Google Scholar] [CrossRef]
  126. Qiu, R.; Qian, F.; Wang, X.; Li, H.; Wang, L. Targeted Delivery of 20(S)-Ginsenoside Rg3-Based Polypeptide Nanoparticles to Treat Colon Cancer. Biomed Microdevices 2019, 21, 18. [Google Scholar] [CrossRef]
  127. Sun, D.; Zou, Y.; Song, L.; Han, S.; Yang, H.; Chu, D.; Dai, Y.; Ma, J.; O’Driscoll, C.M.; Yu, Z.; et al. A Cyclodextrin-Based Nanoformulation Achieves Co-Delivery of Ginsenoside Rg3 and Quercetin for Chemo-Immunotherapy in Colorectal Cancer. Acta Pharm. Sin. B 2022, 12, 378–393. [Google Scholar] [CrossRef]
  128. Ortíz, R.; Quiñonero, F.; García-Pinel, B.; Fuel, M.; Mesas, C.; Cabeza, L.; Melguizo, C.; Prados, J. Nanomedicine to Overcome Multidrug Resistance Mechanisms in Colon and Pancreatic Cancer: Recent Progress. Cancers 2021, 13, 2058. [Google Scholar] [CrossRef]
  129. Gallego-Jara, J.; Lozano-Terol, G.; Sola-Martínez, R.A.; Cánovas-Díaz, M.; Puente, T.d.D. A Compressive Review about Taxol®: History and Future Challenges. Molecules 2020, 25, 5986. [Google Scholar] [CrossRef]
  130. Yuan, M.; Zhang, G.; Bai, W.; Han, X.; Li, C.; Bian, S. The Role of Bioactive Compounds in Natural Products Extracted from Plants in Cancer Treatment and Their Mechanisms Related to Anticancer Effects. Oxid. Med. Cell Longev. 2022, 2022, 1429869. [Google Scholar] [CrossRef]
  131. Maleki Dana, P.; Sadoughi, F.; Asemi, Z.; Yousefi, B. The Role of Polyphenols in Overcoming Cancer Drug Resistance: A Comprehensive Review. Cell Mol. Biol. Lett. 2022, 27, 1. [Google Scholar] [CrossRef]
  132. Afrin, S.; Giampieri, F.; Cianciosi, D.; Alvarez-Suarez, J.M.; Bullon, B.; Amici, A.; Quiles, J.L.; Forbes-Hernández, T.Y.; Battino, M. Strawberry Tree Honey in Combination with 5-Fluorouracil Enhances Chemosensitivity in Human Colon Adenocarcinoma Cells. Food Chem. Toxicol. 2021, 156, 112484. [Google Scholar] [CrossRef]
  133. Hassanein, N.M.A.; Hassan, E.S.G.; Hegab, A.M.; Elahl, H.M.S. Chemopreventive Effect of Sulindac in Combination with Epigallocatechin Gallate or Kaempferol against 1,2-Dimethyl Hydrazine-Induced Preneoplastic Lesions in Rats: A Comparative Study. J. Biochem. Mol. Toxicol. 2018, 32, e22198. [Google Scholar] [CrossRef] [PubMed]
  134. Chavva, S.R.; Deshmukh, S.K.; Kanchanapally, R.; Tyagi, N.; Coym, J.W.; Singh, A.P.; Singh, S. Epigallocatechin Gallate-Gold Nanoparticles Exhibit Superior Antitumor Activity Compared to Conventional Gold Nanoparticles: Potential Synergistic Interactions. Nanomaterials 2019, 9, 396. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Li, Z.; Jiang, H.; Xu, C.; Gu, L. A Review: Using Nanoparticles to Enhance Absorption and Bioavailability of Phenolic Phytochemicals. Food Hydrocoll 2015, 43, 153–164. [Google Scholar] [CrossRef]
Figure 1. Representative image of modulation of the MDR mechanism of resistance by natural bioactive compounds. (A) Main membrane transporters that are involved in chemoresistance by expelling them into the extracellular medium. (B) Action of natural bioactive compounds in the regulation of MDR by (1) MRP-2 downregulation through the inhibition of the Nrf2/MRP2 pathway, (2) P-glycoprotein and MRP-1 blocking by natural bioactive compounds, and (3) inhibition of glutamine cell intake producing an MRP-1 disfunction.
Figure 1. Representative image of modulation of the MDR mechanism of resistance by natural bioactive compounds. (A) Main membrane transporters that are involved in chemoresistance by expelling them into the extracellular medium. (B) Action of natural bioactive compounds in the regulation of MDR by (1) MRP-2 downregulation through the inhibition of the Nrf2/MRP2 pathway, (2) P-glycoprotein and MRP-1 blocking by natural bioactive compounds, and (3) inhibition of glutamine cell intake producing an MRP-1 disfunction.
Applsci 13 02667 g001
Figure 2. Sensitizing effect on chemotherapeutic drugs that are frequently used in CRC exerted by natural compounds of the following families (A) polyphenols, (B) flavonoids, and (C) terpenes, analyzed in this review. DOXO (doxorubicin); 5-FU (5-fluorouracil); OXA (oxaliplatin).
Figure 2. Sensitizing effect on chemotherapeutic drugs that are frequently used in CRC exerted by natural compounds of the following families (A) polyphenols, (B) flavonoids, and (C) terpenes, analyzed in this review. DOXO (doxorubicin); 5-FU (5-fluorouracil); OXA (oxaliplatin).
Applsci 13 02667 g002
Figure 3. Main signaling pathways that were altered after treatment with several of the natural compounds that were analyzed in this review. Most pathways are linked to multidrug resistance processes and their inhibition leads to cell death processes that are induced by apoptosis, necrosis, autophagy, or mediated by genotoxicity.
Figure 3. Main signaling pathways that were altered after treatment with several of the natural compounds that were analyzed in this review. Most pathways are linked to multidrug resistance processes and their inhibition leads to cell death processes that are induced by apoptosis, necrosis, autophagy, or mediated by genotoxicity.
Applsci 13 02667 g003
Table 1. Summary of the in vitro and in vivo effects that were exerted by the bioactive natural compounds that were analyzed.
Table 1. Summary of the in vitro and in vivo effects that were exerted by the bioactive natural compounds that were analyzed.
Fam.Comp.SinergyIn VitroIn VivoClinical TrialsRefs.
PolyphenolsResveratrol5-FU, OXAApoptosis and decreased AC (VEGF inhibition). IC50 of 10 mg/mL in HT-29 (72 h).150 or 300 ppm doses prevented cancerous lesions and induced apoptosis via BAXSafe intake up to a 1g/day dose. Limited BAV (2–29%).[20,47,49,50,52,54,55,57,58]
CurcuminDOX, 5-FU, OXAG1-CCA and apoptosis at 20 µM. Reduced cell migration and invasion.300 mg/kg dose prevented precancerous lesions and decreased tumor size.Low absorption and BAV. Increased tumor apoptosis.[69,70,71,72,73,74,75,76,81]
KaempferolDOX, 5-FU, OXAG1 and G2/M-CCA and apoptosis induction. Decreased AC at 0.1 µM in MDA cell line.Decreased AC and MC.Low toxicity and BAV (2%).[82,83,84,85,86,87,88,89,90]
FlavonoidsQuercetinDOXCCA, apoptosis and decreased AC and MC. CYT in up to 300 µM doses in HCT-15, RKO, CT26, MC38, and HT29 cells10 and 50 mg/kg reduced precancerous lesions and tumor size. Decreased MC and CHR.Not performed.[24,92,93,94,95,96,97]
EGCG5-FU, IRI, CPT, OXAS and G2-CCA, apoptosis, CYT IC50 between 74.6 and 112.1 in CRC cell lines SW480, SW620, and LS411N. Decreased MC.30 mg/kg for 2 weeks decreased MC, tumor growth and induced apoptosis 150 mg twice a day of green tea extract did not show any effect in CRC development risk [18,99,100,101,102,103,104,105,106,107]
TerpenesGeraniol5-FUS-CCA. CYT IC30 of 200 µM for 7 days treatment in Caco-2 cell line.250 mg/kg for 4 weeks prevented CRC precancerous lesions. Reduced tumor growth and apoptosis induction.Not performed.[116,117,118]
Panaxadiol5-FU, IRI CYT IC50 lower than 10 µM in HCT116 cell line (72 h). Decreased AC (VEGF inhibition)30 mg/kg for 3 weeks reduced tumor growth and AC.Not performed.[119,123,124,125]
Rg35-FUCYT IC50 100–200 µM in HT-29 cell line (48 h). Induction of apoptosis and AC via AMPK dysregulation.25 mg/kg for 12 days reduced tumor vascularization and decreased CHR to 5-FU and OXA.Not performed.[121,122,125]
AC (antiangiogenic capacity); BAV (bioavailability); CCA (cell cycle arrest), Comp. (Compound); CPT (cisplatin); CHR (chemoresistance); CRC (colorectal cancer); CYT (cytotoxicity); DOX (doxorubicin); EGCG (epigallocatechin gallate); EMT (epithelial-mesenchymal transition); Fam. (Family); 5-FU (5-fluorouracil); IRI (irinotecan); MC (metastatic capacity); OXA (oxaliplatin); Refs. (references); VEGF (vascular endothelial growth factor).
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Quiñonero, F.; Mesas, C.; Peña, M.; Cabeza, L.; Perazzoli, G.; Melguizo, C.; Ortiz, R.; Prados, J. Vegetal-Derived Bioactive Compounds as Multidrug Resistance Modulators in Colorectal Cancer. Appl. Sci. 2023, 13, 2667. https://doi.org/10.3390/app13042667

AMA Style

Quiñonero F, Mesas C, Peña M, Cabeza L, Perazzoli G, Melguizo C, Ortiz R, Prados J. Vegetal-Derived Bioactive Compounds as Multidrug Resistance Modulators in Colorectal Cancer. Applied Sciences. 2023; 13(4):2667. https://doi.org/10.3390/app13042667

Chicago/Turabian Style

Quiñonero, Francisco, Cristina Mesas, Mercedes Peña, Laura Cabeza, Gloria Perazzoli, Consolación Melguizo, Raul Ortiz, and Jose Prados. 2023. "Vegetal-Derived Bioactive Compounds as Multidrug Resistance Modulators in Colorectal Cancer" Applied Sciences 13, no. 4: 2667. https://doi.org/10.3390/app13042667

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