Next Article in Journal
BRCA Mutation Status in Triple-Negative Breast Cancer Patients Treated with Neoadjuvant Chemotherapy: A Pivotal Role for Treatment Decision-Making
Next Article in Special Issue
Polygodial, a Sesquiterpene Dialdehyde, Activates Apoptotic Signaling in Castration-Resistant Prostate Cancer Cell Lines by Inducing Oxidative Stress
Previous Article in Journal
Addition of Chromosome 17 Polysomy and HER2 Amplification Status Improves the Accuracy of Clinicopathological Factor-Based Progression Risk Stratification and Tumor Grading of Non-Muscle-Invasive Bladder Cancer
Previous Article in Special Issue
Enriched Riceberry Bran Oil Exerts Chemopreventive Properties through Anti-Inflammation and Alteration of Gut Microbiota in Carcinogen-Induced Liver and Colon Carcinogenesis in Rats
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Advances in Dietary Phenolic Compounds to Improve Chemosensitivity of Anticancer Drugs

by
Abdelhakim Bouyahya
1,†,
Nasreddine El Omari
2,†,
Saad Bakrim
3,
Naoufal El Hachlafi
4,
Abdelaali Balahbib
5,
Polrat Wilairatana
6,* and
Mohammad S. Mubarak
7,*
1
Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, Mohammed V University, Rabat 10106, Morocco
2
Laboratory of Histology, Embryology, and Cytogenetic, Faculty of Medicine and Pharmacy, Mohammed V University, Rabat 10100, Morocco
3
Geo-Bio-Environment Engineering and Innovation Laboratory, Molecular Engineering, Biotechnologies, and Innovation Team, Polydisciplinary Faculty of Taroudant, Ibn Zohr University, Agadir 80000, Morocco
4
Microbial Biotechnology and Bioactive Molecules Laboratory, Sciences and Technologies Faculty, Sidi Mohmed Ben Abdellah University, Fez 30050, Morocco
5
Laboratory of Biodiversity, Ecology, and Genome, Faculty of Sciences, Mohammed V University, Rabat 10056, Morocco
6
Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
7
Department of Chemistry, The University of Jordan, Amman 11942, Jordan
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Cancers 2022, 14(19), 4573; https://doi.org/10.3390/cancers14194573
Submission received: 4 July 2022 / Revised: 13 September 2022 / Accepted: 16 September 2022 / Published: 21 September 2022

Abstract

:

Simple Summary

Several dietary phenolic compounds isolated from medicinal plants exert significant anticancer effects via several mechanisms. They induce apoptosis, autophagy, telomerase inhibition, and angiogenesis. Certain dietary phenolic compounds increase the effectiveness of drugs used in conventional chemotherapy. Some clinical uses of dietary phenolic compounds for treating certain cancers have shown remarkable therapeutic results, suggesting effective incorporation in anticancer treatments in combination with traditional chemotherapeutic agents.

Abstract

Despite the significant advances and mechanistic understanding of tumor processes, therapeutic agents against different types of cancer still have a high rate of recurrence associated with the development of resistance by tumor cells. This chemoresistance involves several mechanisms, including the programming of glucose metabolism, mitochondrial damage, and lysosome dysfunction. However, combining several anticancer agents can decrease resistance and increase therapeutic efficacy. Furthermore, this treatment can improve the effectiveness of chemotherapy. This work focuses on the recent advances in using natural bioactive molecules derived from phenolic compounds isolated from medicinal plants to sensitize cancer cells towards chemotherapeutic agents and their application in combination with conventional anticancer drugs. Dietary phenolic compounds such as resveratrol, gallic acid, caffeic acid, rosmarinic acid, sinapic acid, and curcumin exhibit remarkable anticancer activities through sub-cellular, cellular, and molecular mechanisms. These compounds have recently revealed their capacity to increase the sensitivity of different human cancers to the used chemotherapeutic drugs. Moreover, they can increase the effectiveness and improve the therapeutic index of some used chemotherapeutic agents. The involved mechanisms are complex and stochastic, and involve different signaling pathways in cancer checkpoints, including reactive oxygen species signaling pathways in mitochondria, autophagy-related pathways, proteasome oncogene degradation, and epigenetic perturbations.

1. Introduction

Cancer is a significant issue for physicians in multidisciplinary health care facilities. It is a complex and multifactorial pathology in which normal cells develop mutations in their genetic structure, resulting in continued cell growth, colonization, and metastasis to other organs such as the liver, prostate, breast, lungs, brain, and colon. The transformation mechanisms range from genetic and hormonal disturbances to environmental inducers and metabolic deregulations. This divergence of risk factors gives rise to various forms of cancer and, sometimes, implies therapeutic specificity even for the same type of cancer [1,2,3]. In this regard, searching for anticancer treatments requires screening several chemical molecules with functional diversity. Among the candidate molecules studied are phenolic compounds. Chemically defined as having a phenolic structure, phenolic compounds are well recognized for their extensive pharmacological properties such as anti-inflammatory, antibiotic, antiseptic, antitumor, antiallergic, cardioprotective, etc. Phenolic compounds are derived from edible plants, particularly medicinal and aromatic plants, in many food products such as vegetables, cereals, legumes, fruits, nuts, and certain beverages. Indeed, this chemical family constitutes a group of substances frequently present in the metabolism of medicinal plants and contains several subclasses, such as acids, flavonoids, and tannins, which are the most abundant molecules [4,5,6]. Various investigations have focused on phenolic compounds as anticancer bioactive compounds. These groups of molecules exert anticancer properties by acting on the multiple checkpoints of cancerous cells and can induce apoptosis, autophagy, and cell cycle arrest with high specificity [7,8].
In addition, phenolic compounds exert other actions such as inhibiting telomeres, blocking their expression and inhibiting angiogenesis and metastases. On the other hand, phenolic compounds have recently been shown to act in combination with other bioactive compounds used in chemotherapy, sometimes with a potent synergistic mechanism. Recent investigations have also highlighted the sensitization action of cancer cells to chemotherapeutic treatments [9,10]. Indeed, dietary phenolic compounds can induce chemosensitivity of human cancers towards used drugs in chemotherapy via different molecular mechanisms, which include reducing the expression of a transcription factor regulating the expression of cytoprotective genes, the down-regulation of the phosphatidylinositol 3-kinase and Akt protein kinase B (PI3K/Ak) pathway, reducing p53 activation, enhancing the cytotoxicity of used drugs, decreasing Bcl-2 expression and mitochondrial membrane potential (ΔΨm) while inhibiting tumor growth, enhancing the cytotoxicity of used drugs, reducing Bcl-2 expression and mitochondrial membrane potential (ΔΨm) while inhibiting tumor growth, suppressing the expression of hypoxia-inducible factor (HIF-1α) and P-glycoprotein (P-gp) responsible for multidrug resistance, and increasing cellular apoptosis with down-regulation of p-Akt expression and up-regulation of phosphatase and tensin homolog (PTEN) expression [9,10]. Based on the previous discussion, this study aims to investigate and demonstrate the potential benefits of dietary sources, notably phenolic compounds, in managing and preventing cancer. Additionally, the current review aims to examine combining chemotherapeutic drugs with phenolic compounds and their sensitizing effects on cancer treatments to improve the effectiveness and diminish the harmful effects of anticancer bioactive compounds.

2. Dietary Phenolic Compounds Improving the Chemosensitivity of Anticancer Drugs

Recent research findings showed that cancer cells could develop resistance to used drugs in chemotherapy. This resistance is related to different molecular mechanisms which give cancer cells a selective advantage in resisting drugs administered during cancer chemotherapy (Figure 1).

2.1. Flavonoids

Resistance to various anti-cancer treatments, whether chemotherapy or radiotherapy, remains a significant obstacle in the management of cancer patients. Therefore, the use of chemo- and radio-sensitizers of plant origin has attracted the attention of scientists to replace synthetic drugs to improve tumor sensitivity. One example of these natural compounds is flavonoids. Food products containing high levels of flavonoids include blueberries and other berries, parsley, onions, bananas, green and black tea, citrus fruits, sea buckthorn, Ginkgo biloba, and dark chocolate.
Combinatorial treatment with flavonoids has been suggested in several studies as a potential therapeutic approach to avoid drug resistance and enhance their antitumor properties. Table 1 lists flavonoids (Figure 2) that improve the chemosensitivity of chemotherapeutic drugs in cancer.

2.1.1. Flavones

Apigenin is a natural product found in numerous fruits and vegetables, but it is particularly abundant in chamomile tea, parsley, celery, propolis, and garlic oil. It was among the most investigated flavonoids in this field. In 2006, Chan, et al. [11] synthesized a series of apigenin dimers that increased the chemo-sensitivity of leukemic and breast cells, known to be multidrug-resistant (MDR), to numerous anticancer drugs, such as vinblastine (VBL), vincristine (VCR), daunomycin (DM), doxorubicin (DOX), and paclitaxel (PTX) [11]. Seven years later, the chemo-sensitive mechanism by which apigenin acts on DOX has been investigated [12]. The mechanism involves reducing the expression of a transcription factor regulating the expression of cytoprotective genes, called Nrf2, at the levels of proteins and messenger RNA by down-regulating the PI3K/Akt pathway. Compared to DOX treatment alone, the combination treatment of apigenin with DOX showed anticancer effects by inducing apoptosis, reducing cell proliferation, and inhibiting tumor growth.
Johnson and Mejia [13] evaluated the interaction effect between this flavonoid and one of the known chemotherapeutic drugs, gemcitabine (GEM), on human pancreatic cancer cells. This interaction inhibited cell proliferation and growth by 59–73%, whereas apigenin alone potentiated the anti-proliferative effect of GEM. This effect was attributed to IKK-β-mediated NF-κB activation [14]. To improve the chemo-sensitivity of another chemotherapeutic agent, called cisplatin (CP), and to overcome the chemo-resistance of laryngeal carcinoma (Hep-2 cells), apigenin was chosen in an in vitro co-targeted therapy [15]. The results showed that CP-induced Hep-2 cell growth suppression was significantly enhanced in a time- and concentration-dependent manner with suppression of p-AKT and glucose transporter-1 (GLUT-1) involved in resistance to cancer treatments. Bao, et al. also tested this a year later against the same type of cancer [16]. In human renal proximal tubular epithelial cells, apigenin ameliorated CP-induced nephrotoxicity by promoting the PI3K/Akt pathway (Figure 3) and reducing p53 activation [18].
On the other hand, a promising combined effect was recorded with apigenin and 5-fluorouracil (5-FU), a chemotherapeutic drug belonging to the class of antimetabolite drugs [17,19]. In this context, apigenin significantly improved the treatment of hepatocellular carcinoma (HCC) by enhancing the cytotoxicity of 5-FU [17]. The combination of these two elements decreased Bcl-2 expression and mitochondrial membrane potential (ΔΨm) while inhibiting tumor growth of HCC xenografts. This was in agreement with the results of Gaballah and collaborators [19], who also observed a reduction in tumor size and Mcl-1 expression, with an increase in Beclin-1 levels and caspase-3 and -9 activities. Furthermore, Gao, et al. [20,21] investigated the chemosensitivity of apigenin using BEL-7402/ADM cells, which are known for their resistance to DOX, a molecule belonging to the anthracycline family. Results showed that apigenin enhanced DOX sensitivity, induced apoptosis, and prevented HCC xenograft growth. Recently, treatment with apigenin was applied to ovarian cancer (OC) using ovarian cancer-sensitive cells (SKOV3) and drug-resistant cells (SKOV3/DDP) [22]. Results showed positive effects on the chemo-sensitivity of both cell types with apoptosis and reversal of drug resistance of these cancer cells through the down-regulation of the Mcl-1 gene.

2.1.2. Flavanols

Quercetin is naturally distributed in many fruits, vegetables, leaves, seeds, and grains; capers, red onions, and kale contain appreciable quantities. Regarding quercetin, several research studies have evaluated the effect of this flavonoid against multidrug resistance by several mechanisms of action in various cancer cells. Research findings indicated that a co-treatment with quercetin combined with temozolomide (TMZ), an active anticancer drug, showed positive results such as inhibition of cell viability, induction of cell apoptosis, an increase of caspase-3 activity, elimination of drug insensitivity, and improvement of TMZ inhibition [23,27]. Some of these effects, such as the decrease in colony formation, inhibition of growth, and the stimulation of apoptosis by decreasing Bcl2 gene expression and increasing p53 and caspase-9 activity in esophageal (EC9706 and Eca109) have also been observed by combining quercetin with 5-FU [24] and breast (MCF-7) [34] cancer cells.
The management of breast cancer attracted the attention of Li, et al. [25,28] who performed two experiments to evaluate the combination therapy of quercetin with DOX on MCF-7 cells. Results revealed that this treatment inhibits cell invasion and proliferation by suppressing the expression of HIF-1α and P-glycoprotein (P-gp), responsible for multidrug resistance [25]. In addition, results showed an increase in cellular apoptosis with down-regulation of p-Akt expression and up-regulation of phosphatase and tensin homolog (PTEN) expression [28]. Other effects of this combination, namely increasing cellular sensitivity to DOX and promoting DOX-induced cellular apoptosis via the mitochondrial/ROS pathway, respectively, have been noted in studies conducted by Chen, et al. [30] and by Shu, et al. [31] in the treatment of HCC (BEL-7402 cells) and prostate cancer (PC3 cells). Quercetin alone was able to down-regulate the expression of specific ABC transporters (ABCB1, ABCC1, and ABCC2) [30] with inhibition of the expression of the PI3K/AKT pathway [31].
On the other hand, the synergistic effect of quercetin was examined in vitro and in vivo with PTX, a molecule used in chemotherapy and synthesized by endophytic fungi [26]. Quercetin alone inhibited the proliferation of OC cells and increased their sensitivity to PTX [26]. Meanwhile, the combination of these two molecules showed an inhibition of the migration and proliferation of prostate cancer cells with an increase in apoptosis, and induction of G2/M cell cycle arrest, whereas the in vivo combination showed a synergistic effect in killing cancer cells [26]. Moreover, this flavonol positively affected GEM, a drug with significant cytotoxic activity, such as the improvement of cell death associated with increased caspase-3 and -9 activities in lung cancer cells. It caused considerable suppression of HSP70 chaperone protein expression compared to treatment with GEM alone [29]. This chemo-sensitivity has also been noted in pancreatic cancer cells [32]. In a recent study, Safi, et al. [35] evaluated the synergistic effect of quercetin (95 μM) with docetaxel (7 nM), an alkaloid with anticancer properties. Results revealed a decrease in STAT3, AKT, pERK1/2, and Bcl-2 proteins in MDA-MB-231 breast cancer cells.

2.1.3. Flavonols

Kaempferol is a flavonol found in numerous fruits and vegetables such as grapes, potatoes, squash, tomatoes, broccoli, onions, brussels sprouts, green beans, green tea, peaches, spinach, blackberries, lettuce, cucumber, apples, and raspberries. It has been studied for its chemo-sensitizing activity. Its association with quercetin has shown promising results, namely growth inhibition of adriamycin-resistant K562/A cells and myeloid leukemia K562 cells, increasing their sensitivity, and induction of apoptosis [36]. Additionally, this ubiquitous flavonoid chemo-sensitized 5-FU resistant colon cancer LS174-R cells and the combination of both substances provided a synergistic effect by inhibiting cell viability and inducing cell cycle arrest [37]. This was explained recently by Wu et al. [38] who attributed these results to the inhibition of PKM2-mediated glycolysis. The combinatorial effect of 5-FU with a flavonol was further evaluated (in vitro and in vivo) with myricetin against esophageal carcinoma [39]. Several favorable outcomes such as suppression of cell proliferation, increase in cell apoptosis and caspase-3 expression, and decrease in Bcl-2 and tumor xenograft growth (in vivo) were observed. In addition, kaempferol increased the PTX cytotoxicity with modulation of anti- and pro-apoptotic markers in OC cells [40].
As previously reported in breast cancer treatment with flavonoids, these secondary metabolites reverse cancer drug resistance and sensitize tumor cells to chemotherapy via several mechanisms. In this respect, Iriti et al. studied the chemo-sensitizing potential of rutin (3′,4′,5,7-Tetrahydroxy-3-[α-L-rhamnopyranosyl-(1–6)-β-D-glucopyranosyloxy]flavone) against two breast cancer cell lines (MB-MDA-231 and MCF-7 cells) [41]. At a dose of 20 μM, these researchers found that this flavonoid acts as a chemo-sensitizing agent by improving the anti-tumor effect of two chemotherapeutic agents (methotrexate and cyclophosphamide). Furthermore, rutin improved the in vivo efficacy of another anti-cancer drug (sorafenib) in a xenograft model of human HCC [42]. As seen with quercetin and berberine, another natural flavone called hispidulin enhanced cellular chemo-sensitivity by inhibiting the expression of the transcription factor HIF-1α via AMPK signaling in gallbladder cancer [43].

2.1.4. Anthocyanidins

Anthocyanins (ACNs) are the primary color of many leaves (such as purple cabbage), fruits (such as grapes and blueberries), tubers (such as purple radishes and yams), and flowers (such as roses). In a broad sense, anthocyanidins (ACNs) present a subclass of flavonoids that have not been well investigated for their chemo-sensitizing and radio-sensitizing effects. Indeed, black raspberry ACNs improved the efficacy of two chemotherapeutic agents (5-FU and celecoxib); in vitro by inhibiting the proliferation of CRC cells and in vivo by decreasing the number of CRC tumors in animals [44]. Recently, specific molecules of this family, such as delphinidin [45] and cyanidin-3-glucoside (C3G) [46], have been studied. In radiation-exposed A549 human lung adenocarcinoma cells, delphinidin enhanced the radio-therapeutic effects (induction of autophagy and apoptosis) by activating the JNK/MAPK signaling pathway [45]. Similarly, C3G improved the sensitivity to DOX and its cytotoxicity by inhibiting the phosphorylation of Akt and increasing that of p38, mainly by reducing the expression of claudin-2 [46]. Table 2 lists anthocyanidins (Figure 4) that could improve the chemosensitivity of cancer drugs.

2.2. Non-Flavonoids

2.2.1. Phenolic Acids

It has been demonstrated that phenolic acids have a chemo-sensitizing activity on several types of cancer cells to different chemotherapeutics (Table 3). Data presented in Table 3 indicate that ellagic acid was the most studied molecule. It is found in large quantities in pecans, chestnuts, raspberries, peaches, cranberries, strawberries, raw grapes, walnuts, and pomegranates. Indeed, its combination with 5-FU in treating colorectal carcinoma (CRC) gave significant effects such as inhibition of apoptotic cell death and cell proliferation. In contrast, treatment alone enhanced 5-FU chemo-sensitivity in CRC cells [47]. Indeed, ellagic acid alone potentiated CP cytotoxicity and prevented the development of CP resistance in epithelial OC cells [48]. Table 3 shows the phenolic acids (Figure 5) that improve the chemosensitivity of cancer drugs.
Caffeic acid can be derived from a variety of beverages and is relatively present at high concentrations in lingonberry, thyme, sage, and spearmint as well as in spices such as Ceylon cinnamon and star anise. Caffeic acid is moderately available in sunflower seeds, applesauce, apricots, and prunes. Caffeic acid phenethyl ester (CAPE), a central component of propolis, has also been investigated for its chemo-sensitizing [51,52] and radio-sensitizing [53] effects against various types of cancer. The radio-sensitizing effect of this substance was evaluated in 2005 by Chen, et al. [49] against CT26 colorectal adenocarcinoma cells and in vivo on BALB/c mice implanted with these cells. These authors noted, in vitro, an improvement in the destruction of CT26 cells by ionizing radiation (IR) and, in vivo, an extension of animal survival and a marked inhibition of tumor growth compared to radiotherapy alone. The mechanism of action explaining this radio-sensitivity was elucidated very recently on prostate cancer cells (DU145 and PC3) by co-treatment using gamma radiation (GR) and CAPE [53]. Results showed that this combined treatment sensitizes the cells to radiotherapy by reducing the RAD50 and RAD51 proteins and the cell migration potential, mainly by inhibiting DNA damage repair. As for the chemo-sensitivity of this phenolic compound, Lin, et al. [50] did not observe any chemo-sensitizing effect of medulloblastoma Daoy cells on the chemotherapeutics studied (DOX or CP). However, in 2018, two similar studies proved otherwise by enhancing the sensitivity of gastric and lung cancer cells to DOX and CP by decreasing proteasome function [51,52].
In contrast, Muthusamy, et al. carried out two studies on the ability of ferulic acid (FA), a phenolic acid present in seeds and leaves of certain plants and found in exceptionally high amounts in popcorn and bamboo shoots, to reverse the resistance of multiresistant cells to anticancer drugs. In the first study, FA-enhanced cell cycle arrest was exerted by PTX and decreased resistance to this drug [54]. In the second study, FA increased VCR and DOX cytotoxicity and synergistically increased DOX-induced apoptotic signaling [55]. In addition, the authors showed that the synergy between FA and DOX reduced tumor xenograft size compared to the treatment with DOX alone. They associated these results with suppressing P-gp expression by inhibiting the PI3K/Akt/NF-κB signaling pathway.
Another phenolic acid constituent, called rosmarinic acid (RA), is found in culinary herbs such as Ocimum tenuiflorum (holy basil), Origanum majorana (marjoram), Melissa officinalis (lemon balm), Ocimum basilicum (basil), Salvia officinalis (sage), Salvia rosmarinus (rosemary), peppermint, and thyme. This natural compound showed remarkable potential as an anti-leukemic agent in acute promyelocytic leukemia cells by potentiating macrophage differentiation induced by all-trans retinoic acid [56]. Furthermore, Yu, et al. [57] evaluated the impact of RA on 5-FU chemo-resistance in the treatment of gastric carcinoma. In SGC7901 gastric carcinoma cells treated with 5-FU, the application of RA increased the chemo-sensitivity of these cells to 5-FU by reducing its IC50 values from 208.6 to 70.43 μg/mL and the expression levels of two miRNAs (miR-642a-3p and miR-6785-5p), with increased expression of FOXO4.

2.2.2. Tannins

Although condensed tannins (also called proanthocyanidins (PCs)), found in plants, such as cranberry, blueberry, and grape seeds, are chemically polymers of flavanols, they have not been widely investigated as anticancer agents compared to flavonoids and phenolic acids. However, they have recently been studied to overcome the problems of cancer cell resistance to chemotherapy [58]. In this context, Zhang, et al. [58] showed that PCs inhibit the growth and characteristics of platinum-resistant OC cells by inducing G1 cell cycle arrest and targeting the Wnt/β-catenin signaling pathway. On the other hand, other researchers indicated that PCs sensitize chemoresistant CC cells (HCT116 and H716) to 5-FU and oxaliplatin (OXP) [59]. In contrast, combining all these substances reduced tumor growth in chemoresistant cells and chemoresistant tumor xenografts. The mechanism suggested to overcome this chemo-resistance involves suppressing the activity of adenosine triphosphate-binding cassette transporters. Furthermore, tannic acid (TA), another plant tannin used as an anticancer agent, has been studied for its synergistic effect with chemotherapeutic drugs (5-FU, GEM, and mitomycin C) against malignant cholangiocytes [60]. Results revealed that TA exhibits a crucial synergistic effect with 5-FU and mitomycin C in modulating drug efflux pathways. The exact synergy was observed by combining TA and CP on HepG2 liver cancer cells through mitochondria-mediated apoptosis [61]. This chemotherapeutic sensitivity to CP was corroborated by co-treatment with procyanidins in TU686 laryngeal cancer cells through the apoptosis and autophagy pathway [62]. Table 4 lists condensed tannins (Figure 6) that could improve the chemosensitivity of cancer drugs.
To improve the bioavailability and bioactivity of ellagic acid in vivo, Mady, et al. [63] formulated nanoparticles loaded with this acid from a biodegradable polymer [poly(ε-caprolactone)]. This encapsulation improved the oral bioavailability and the anti-tumor effect of ellagic acid. In a glioblastoma model, Cetin, et al. carried out two studies that showed an improvement in the anticancer efficacy of bevacizumab [64] and TMZ [65] by co-treatment with ellagic acid. This treatment reduced the expression of MGMT, affected caspase-3 and p53 proteins, and its combination with the chemotherapeutics reduced cell viability and the expression of MDR1.
Chemo-resistance of bladder cancer has been a serious problem in managing this type of cancer, particularly resistance to GEM. However, the underlying resistance mechanism has not been elucidated. The effect of ellagic acid or its combinatorial effect with GEM on GEM-sensitive bladder cancer cells and GEM-resistant cells was recently evaluated [66]. Results revealed that ellagic acid exerts numerous promising anticancer effects, particularly resensitization of GEM-resistant cells by inhibiting GEM transporters and the epithelial–mesenchymal transition (EMT), responsible for GEM resistance in other types of cancer. Suppression of EMT was also observed by catechol against pancreatic cancer cells, in addition to cellular chemo-sensitivity and radio-sensitivity to GEM via inhibition of the AMPK/Hippo signaling pathway [67].

3. Conclusions and Perspectives

At present, the use of foodstuffs is attracting attention in treating and preventing diseases, including cancer. This is due to the presence of bioactive compounds such as phenolic acids, among others, in our diet. These natural compounds are gaining popularity in cancer treatment due to their lower side effects, cost, and accessibility than conventional drugs. In this review, we have shown through published research that phenolic compounds are an excellent source of natural anticancer substances providing a range of preventive and therapeutic options against several types of cancer. These compounds could be used alone or in combination with other anticancer drugs. Certain phenolic compounds such as quercetin and gallic acid have well-known mechanisms of action. These molecules act specifically on the various checkpoints of cancerous cells. Therefore, exploring these mechanisms of action could further improve the therapeutic efficacy. However, further investigations that could involve human subjects and different pharmacokinetic parameters are required to ensure the safety of these compounds before they can be used as prescription drugs. In addition, the development of a standardized extract or dosage could also be followed in clinical trials. In summary, phenolic compounds present in our food can be useful in complementary medicine for the prevention and treatment of different types of cancers due to their natural origin, safety, and low cost compared to cancer drugs.

Author Contributions

Conceptualization, A.B. (Abdelhakim Bouyahya), P.W. and M.S.M.; methodology, N.E.O., N.E.H. and S.B.; software, A.B. (Abdelaali Balahbib); validation, A.B. (Abdelhakim Bouyahya); formal analysis, A.B. (Abdelhakim Bouyahya); investigation, A.B. (Abdelhakim Bouyahya), N.E.O., N.E.H. and S.B.; writing—original draft preparation, A.B. (Abdelhakim Bouyahya); N.E.O., N.E.H. and S.B.; writing—review and editing, A.B. (Abdelhakim Bouyahya), P.W. and M.S.M.; supervision, A.B. (Abdelhakim Bouyahya) and M.S.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ACNsAnthocyanidins
AktProtein Kinase B
APGApigenin
CAPECaffeic Acid Phenethyl Ester
CLLChronic Lymphocytic Leukemia
COX-2Cyclooxygenase-2
CRCColorectal Carcinoma
DMDaunomycin
DOXDoxorubicin
ELISAEnzyme-linked immunosorbent assay
EMTEpithelial-Mesenchymal Transition
ERKExtracellular Signal-Regulated Kinase
FAFerulic acid
GLUT-1Glucose Transporter-1
GemGemcitabine
GRGamma Radiation
HCCHepatocellular Carcinoma
HIF-1αHypoxia-Inducible Factor-1α
HOHeme Oxygenase
HPDHispidulin
HPDEHuman pancreatic ductal epithelium
ILInterleukin
IRIonizing Radiation
JNKC-Jun N-Terminal Kinase
KAEKaempferol
MAPKMitogen-Activated Protein Kinase
MDR1Multidrug resistance protein 1
MMMultiple Myeloma
mTORmammalian Target of Rapamycin
MYRMyricetin
NF-ĸBNuclear Factor Kappa B
Nrf2Nuclear factor erythroid-related factor 2
NSCLCNon-Small Cell Lung Cancer
OCOvarian Cancer
OXPOxaliplatin
P-gpP-glycoprotein
PTENPhosphatase and Tensin Homolog
PTXPaclitaxel
QueQuercetin
ROSReactive Oxygen Species
RTNRutin
STAT3Signal Transducer and Activator of Transcription 3
TATannic acid
TNF-αTumor Necrosis Factor-α
TMZTemozolomide
VBLVinblastine
VCRVincristine
5-FU5-Fluorouracil

References

  1. Hurson, A.N.; Ahearn, T.U.; Keeman, R.; Abubakar, M.; Jung, A.Y.; Kapoor, P.M.; Koka, H.; Yang, X.R.; Chang-Claude, J.; Martínez, E. Systematic Literature Review of Risk Factor Associations with Breast Cancer Subtypes in Women of African, Asian, Hispanic, and European Descents. Cancer Res. 2022, 82, 3670. [Google Scholar] [CrossRef]
  2. Stopsack, K.H.; Nandakumar, S.; Arora, K.; Nguyen, B.; Vasselman, S.E.; Nweji, B.; McBride, S.M.; Morris, M.J.; Rathkopf, D.E.; Slovin, S.F. Differences in Prostate Cancer Genomes by Self-Reported Race: Contributions of Genetic Ancestry, Modifiable Cancer Risk Factors, and Clinical FactorsRacial Differences in Prostate Cancer Genomes. Clin. Cancer Res. 2022, 28, 318–326. [Google Scholar] [CrossRef]
  3. Tan, D.A.; Dayu, A.R.B. Menopausal Hormone Therapy: Why We Should No Longer Be Afraid of the Breast Cancer Risk. Climacteric 2022, 25, 362–368. [Google Scholar] [CrossRef]
  4. Dobroslavić, E.; Repajić, M.; Dragović-Uzelac, V.; Elez Garofulić, I. Isolation of Laurus Nobilis Leaf Polyphenols: A Review on Current Techniques and Future Perspectives. Foods 2022, 11, 235. [Google Scholar] [CrossRef]
  5. Mitra, S.; Tareq, A.M.; Das, R.; Emran, T.B.; Nainu, F.; Chakraborty, A.J.; Ahmad, I.; Tallei, T.E.; Idris, A.M.; Simal-Gandara, J. Polyphenols: A First Evidence in the Synergism and Bioactivities. Food Rev. Int. 2022, 1–23. [Google Scholar] [CrossRef]
  6. Rosero, S.; Del Pozo, F.; Simbaña, W.; Álvarez, M.; Quinteros, M.F.; Carrillo, W.; Morales, D. Polyphenols and Flavonoids Composition, Anti-Inflammatory and Antioxidant Properties of Andean Baccharis Macrantha Extracts. Plants 2022, 11, 1555. [Google Scholar] [CrossRef]
  7. 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]
  8. Rauf, A.; Shariati, M.A.; Imran, M.; Bashir, K.; Khan, S.A.; Mitra, S.; Emran, T.B.; Badalova, K.; Uddin, M.; Mubarak, M.S. Comprehensive Review on Naringenin and Naringin Polyphenols as a Potent Anticancer Agent. Environ. Sci. Pollut. Res. 2022, 29, 31025–31041. [Google Scholar] [CrossRef]
  9. Mottaghi, S.; Abbaszadeh, H. Natural Lignans Honokiol and Magnolol as Potential Anticarcinogenic and Anticancer Agents. A Comprehensive Mechanistic Review. Nutr. Cancer 2022, 74, 761–778. [Google Scholar] [CrossRef]
  10. Yoganathan, S.; Alagaratnam, A.; Acharekar, N.; Kong, J. Ellagic Acid and Schisandrins: Natural Biaryl Polyphenols with Therapeutic Potential to Overcome Multidrug Resistance in Cancer. Cells 2021, 10, 458. [Google Scholar] [CrossRef]
  11. Chan, K.-F.; Zhao, Y.; Burkett, B.A.; Wong, I.L.; Chow, L.M.; Chan, T.H. Flavonoid Dimers as Bivalent Modulators for P-Glycoprotein-Based Multidrug Resistance: Synthetic Apigenin Homodimers Linked with Defined-Length Poly (Ethylene Glycol) Spacers Increase Drug Retention and Enhance Chemosensitivity in Resistant Cancer Cells. J. Med. Chem. 2006, 49, 6742–6759. [Google Scholar] [CrossRef]
  12. Gao, A.-M.; Ke, Z.-P.; Wang, J.-N.; Yang, J.-Y.; Chen, S.-Y.; Chen, H. Apigenin Sensitizes Doxorubicin-Resistant Hepatocellular Carcinoma BEL-7402/ADM Cells to Doxorubicin via Inhibiting PI3K/Akt/Nrf2 Pathway. Carcinogenesis 2013, 34, 1806–1814. [Google Scholar] [CrossRef]
  13. Johnson, J.L.; de Mejia, E.G. Interactions between Dietary Flavonoids Apigenin or Luteolin and Chemotherapeutic Drugs to Potentiate Anti-Proliferative Effect on Human Pancreatic Cancer Cells, in Vitro. Food Chem. Toxicol. 2013, 60, 83–91. [Google Scholar] [CrossRef]
  14. Wu, D.-G.; Yu, P.; Li, J.-W.; Jiang, P.; Sun, J.; Wang, H.-Z.; Zhang, L.-D.; Wen, M.-B.; Bie, P. Apigenin Potentiates the Growth Inhibitory Effects by IKK-β-Mediated NF-ΚB Activation in Pancreatic Cancer Cells. Toxicol. Lett. 2014, 224, 157–164. [Google Scholar] [CrossRef]
  15. Xu, Y.-Y.; Wu, T.-T.; Zhou, S.-H.; Bao, Y.-Y.; Wang, Q.-Y.; Fan, J.; Huang, Y.-P. Apigenin Suppresses GLUT-1 and p-AKT Expression to Enhance the Chemosensitivity to Cisplatin of Laryngeal Carcinoma Hep-2 Cells: An in Vitro Study. Int. J. Clin. Exp. Pathol. 2014, 7, 3938. [Google Scholar]
  16. Bao, Y.-Y.; Zhou, S.-H.; Lu, Z.-J.; Fan, J.; Huang, Y.-P. Inhibiting GLUT-1 Expression and PI3K/Akt Signaling Using Apigenin Improves the Radiosensitivity of Laryngeal Carcinoma in Vivo. Oncol. Rep. 2015, 34, 1805–1814. [Google Scholar] [CrossRef]
  17. Hu, X.-Y.; Liang, J.-Y.; Guo, X.-J.; Liu, L.; Guo, Y.-B. 5-Fluorouracil Combined with Apigenin Enhances Anticancer Activity through Mitochondrial Membrane Potential (ΔΨm)-Mediated Apoptosis in Hepatocellular Carcinoma. Clin. Exp. Pharmacol. Physiol. 2015, 42, 146–153. [Google Scholar] [CrossRef]
  18. Ju, S.M.; Kang, J.G.; Bae, J.S.; Pae, H.O.; Lyu, Y.S.; Jeon, B.H. The Flavonoid Apigenin Ameliorates Cisplatin-Induced Nephrotoxicity through Reduction of P53 Activation and Promotion of PI3K/Akt Pathway in Human Renal Proximal Tubular Epithelial Cells. Evid. -Based Complementary Altern. Med. 2015, 2015, 186436 . [Google Scholar] [CrossRef]
  19. Gaballah, H.H.; Gaber, R.A.; Mohamed, D.A. Apigenin Potentiates the Antitumor Activity of 5-FU on Solid Ehrlich Carcinoma: Crosstalk between Apoptotic and JNK-Mediated Autophagic Cell Death Platforms. Toxicol. Appl. Pharmacol. 2017, 316, 27–35. [Google Scholar] [CrossRef]
  20. Gao, A.-M.; Zhang, X.-Y.; Ke, Z.-P. Apigenin Sensitizes BEL-7402/ADM Cells to Doxorubicin through Inhibiting MiR-101/Nrf2 Pathway. Oncotarget 2017, 8, 82085. [Google Scholar] [CrossRef]
  21. Gao, A.-M.; Zhang, X.-Y.; Hu, J.-N.; Ke, Z.-P. Apigenin Sensitizes Hepatocellular Carcinoma Cells to Doxorubic through Regulating MiR-520b/ATG7 Axis. Chem.-Biol. Interact. 2018, 280, 45–50. [Google Scholar] [CrossRef]
  22. Li, Q.; Li, L.; Zhao, X.; Cheng, Z.; Ma, J. Apigenin Induces Apoptosis and Reverses the Drug Resistance of Ovarian Cancer Cells. Int. J. Clin. Exp. Med. 2020, 13, 1987–1994. [Google Scholar]
  23. Thangasamy, T.; Sittadjody, S.; Mitchell, G.C.; Mendoza, E.E.; Radhakrishnan, V.M.; Limesand, K.H.; Burd, R. Quercetin Abrogates Chemoresistance in Melanoma Cells by Modulating ΔNp73. BMC Cancer 2010, 10, 282. [Google Scholar] [CrossRef]
  24. Chuang-Xin, L.; Wen-Yu, W.; Yao, C.U.I.; Xiao-Yan, L.; Yun, Z. Quercetin Enhances the Effects of 5-Fluorouracil-Mediated Growth Inhibition and Apoptosis of Esophageal Cancer Cells by Inhibiting NF-ΚB. Oncol. Lett. 2012, 4, 775–778. [Google Scholar] [CrossRef]
  25. Li, S.; Li, K.; Zhang, J.; Dong, Z. The Effect of Quercetin on Doxorubicin Cytotoxicity in Human Breast Cancer Cells. Anti-Cancer Agents Med. Chem. (Former. Curr. Med. Chem. -Anti-Cancer Agents) 2013, 13, 352–355. [Google Scholar] [CrossRef]
  26. Maciejczyk, A.; Surowiak, P. Quercetin Inhibits Proliferation and Increases Sensitivity of Ovarian Cancer Cells to Cisplatin and Paclitaxel. Ginekol. Pol. 2013, 84, 590–595. [Google Scholar] [CrossRef]
  27. Sang, D.; Li, R.; Lan, Q. Quercetin Sensitizes Human Glioblastoma Cells to Temozolomide in Vitro via Inhibition of Hsp27. Acta Pharmacol. Sin. 2014, 35, 832–838. [Google Scholar] [CrossRef]
  28. Li, S.; Qiao, S.; Zhang, J.; Li, K. Quercetin Increase the Chemosensitivity of Breast Cancer Cells to Doxorubicin via PTEN/Akt Pathway. Anti-Cancer Agents Med. Chem. (Former. Curr. Med. Chem. -Anti-Cancer Agents) 2015, 15, 1185–1189. [Google Scholar] [CrossRef]
  29. Lee, S.H.; Lee, E.J.; Min, K.H.; Hur, G.Y.; Lee, S.H.; Lee, S.Y.; Kim, J.H.; Shin, C.; Shim, J.J.; In, K.H. Quercetin Enhances Chemosensitivity to Gemcitabine in Lung Cancer Cells by Inhibiting Heat Shock Protein 70 Expression. Clin. Lung Cancer 2015, 16, e235–e243. [Google Scholar] [CrossRef]
  30. Chen, Z.; Huang, C.; Ma, T.; Jiang, L.; Tang, L.; Shi, T.; Zhang, S.; Zhang, L.; Zhu, P.; Li, J. Reversal Effect of Quercetin on Multidrug Resistance via FZD7/β-Catenin Pathway in Hepatocellular Carcinoma Cells. Phytomedicine 2018, 43, 37–45. [Google Scholar] [CrossRef]
  31. Shu, Y.; Xie, B.; Liang, Z.; Chen, J. Quercetin Reverses the Doxorubicin Resistance of Prostate Cancer Cells by Downregulating the Expression of C-Met. Oncol. Lett. 2018, 15, 2252–2258. [Google Scholar] [CrossRef]
  32. Lan, C.-Y.; Chen, S.-Y.; Kuo, C.-W.; Lu, C.-C.; Yen, G.-C. Quercetin Facilitates Cell Death and Chemosensitivity through RAGE/PI3K/AKT/MTOR Axis in Human Pancreatic Cancer Cells. J. Food Drug Anal. 2019, 27, 887–896. [Google Scholar] [CrossRef]
  33. Zhang, X.; Huang, J.; Yu, C.; Xiang, L.; Li, L.; Shi, D.; Lin, F. Quercetin Enhanced Paclitaxel Therapeutic Effects towards PC-3 Prostate Cancer through ER Stress Induction and ROS Production. OncoTargets Ther. 2020, 13, 513. [Google Scholar] [CrossRef]
  34. Mawalizadeh, F.; Mohammadzadeh, G.; Khedri, A.; Rashidi, M. Quercetin Potentiates the Chemosensitivity of MCF-7 Breast Cancer Cells to 5-Fluorouracil. Mol. Biol. Rep. 2021, 48, 7733–7742. [Google Scholar] [CrossRef]
  35. Safi, A.; Heidarian, E.; Ahmadi, R. Quercetin Synergistically Enhances the Anticancer Efficacy of Docetaxel through Induction of Apoptosis and Modulation of PI3K/AKT, MAPK/ERK, and JAK/STAT3 Signaling Pathways in MDA-MB-231 Breast Cancer Cell Line. Int. J. Mol. Cell. Med. 2021, 10, 11. [Google Scholar]
  36. Yanqiu, H.; Linjuan, C.; Jin, W.; Hongjun, H.; Yongjin, S.; Guobin, X.; Hanyun, R. The Effects of Quercetin and Kaempferol on Multidrug Resistance and the Expression of Related Genes in Human Erythroleukemic K562/A Cells. Afr. J. Biotechnol. 2011, 10, 13399–13406. [Google Scholar] [CrossRef]
  37. 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]
  38. Wu, H.; Du, J.; Li, C.; Li, H.; Guo, H.; Li, Z. Kaempferol Can Reverse the 5-Fu Resistance of Colorectal Cancer Cells by Inhibiting PKM2-Mediated Glycolysis. Int. J. Mol. Sci. 2022, 23, 3544. [Google Scholar] [CrossRef]
  39. Wang, L.; Feng, J.; Chen, X.; Guo, W.; Du, Y.; Wang, Y.; Zang, W.; Zhang, S.; Zhao, G. Myricetin Enhance Chemosensitivity of 5-Fluorouracil on Esophageal Carcinoma in Vitro and in Vivo. Cancer Cell Int. 2014, 14, 71. [Google Scholar] [CrossRef]
  40. Zheng, A.-W.; Chen, Y.-Q.; Zhao, L.-Q.; Feng, J.-G. Myricetin Induces Apoptosis and Enhances Chemosensitivity in Ovarian Cancer Cells. Oncol. Lett. 2017, 13, 4974–4978. [Google Scholar] [CrossRef]
  41. Iriti, M.; Vitalini, S.; Arnold Apostolides, N.; El Beyrouthy, M. Chemical Composition and Antiradical Capacity of Essential Oils from Lebanese Medicinal Plants. J. Essent. Oil Res. 2014, 26, 466–472. [Google Scholar] [CrossRef]
  42. Zhou, M.; Zhang, G.; Hu, J.; Zhu, Y.; Lan, H.; Shen, X.; Lv, Y.; Huang, L. Rutin Attenuates Sorafenib-Induced Chemoresistance and Autophagy in Hepatocellular Carcinoma by Regulating BANCR/MiRNA-590-5P/OLR1 Axis. Int. J. Biol. Sci. 2021, 17, 3595. [Google Scholar] [CrossRef]
  43. Gao, H.; Xie, J.; Peng, J.; Han, Y.; Jiang, Q.; Han, M.; Wang, C. Hispidulin Inhibits Proliferation and Enhances Chemosensitivity of Gallbladder Cancer Cells by Targeting HIF-1α. Exp. Cell Res. 2015, 332, 236–246. [Google Scholar] [CrossRef]
  44. Li, X.; Chen, L.; Gao, Y.; Zhang, Q.; Chang, A.K.; Yang, Z.; Bi, X. Black Raspberry Anthocyanins Increased the Antiproliferative Effects of 5-Fluorouracil and Celecoxib in Colorectal Cancer Cells and Mouse Model. J. Funct. Foods 2021, 87, 104801. [Google Scholar] [CrossRef]
  45. Kang, S.H.; Bak, D.-H.; Chung, B.Y.; Bai, H.-W.; Kang, B.S. Delphinidin Enhances Radio-Therapeutic Effects via Autophagy Induction and JNK/MAPK Pathway Activation in Non-Small Cell Lung Cancer. Korean J. Physiol. Pharmacol. Off. J. Korean Physiol. Soc. Korean Soc. Pharmacol. 2020, 24, 413–422. [Google Scholar] [CrossRef]
  46. Eguchi, H.; Matsunaga, H.; Onuma, S.; Yoshino, Y.; Matsunaga, T.; Ikari, A. Down-Regulation of Claudin-2 Expression by Cyanidin-3-Glucoside Enhances Sensitivity to Anticancer Drugs in the Spheroid of Human Lung Adenocarcinoma A549 Cells. Int. J. Mol. Sci. 2021, 22, 499. [Google Scholar] [CrossRef]
  47. Kao, T.-Y.; Chung, Y.-C.; Hou, Y.-C.; Tsai, Y.-W.; Chen, C.-H.; Chang, H.-P.; Chou, J.-L.; Hsu, C.-P. Effects of Ellagic Acid on Chemosensitivity to 5-Fluorouracil in Colorectal Carcinoma Cells. Anticancer Res. 2012, 32, 4413–4418. [Google Scholar]
  48. Engelke, L.H.; Hamacher, A.; Proksch, P.; Kassack, M.U. Ellagic Acid and Resveratrol Prevent the Development of Cisplatin Resistance in the Epithelial Ovarian Cancer Cell Line A2780. J. Cancer 2016, 7, 353. [Google Scholar] [CrossRef]
  49. Chen, Y.-J.; Liao, H.-F.; Tsai, T.-H.; Wang, S.-Y.; Shiao, M.-S. Caffeic Acid Phenethyl Ester Preferentially Sensitizes CT26 Colorectal Adenocarcinoma to Ionizing Radiation without Affecting Bone Marrow Radioresponse. Int. J. Radiat. Oncol. Biol. Phys. 2005, 63, 1252–1261. [Google Scholar] [CrossRef]
  50. Lin, Y.-H.; Chiu, J.-H.; Tseng, W.-S.; Wong, T.-T.; Chiou, S.-H.; Yen, S.-H. Antiproliferation and Radiosensitization of Caffeic Acid Phenethyl Ester on Human Medulloblastoma Cells. Cancer Chemother. Pharmacol. 2006, 57, 525–532. [Google Scholar] [CrossRef]
  51. Matsunaga, T.; Tsuchimura, S.; Azuma, N.; Endo, S.; Ichihara, K.; Ikari, A. Caffeic Acid Phenethyl Ester Potentiates Gastric Cancer Cell Sensitivity to Doxorubicin and Cisplatin by Decreasing Proteasome Function. Anti-Cancer Drugs 2019, 30, 251–259. [Google Scholar] [CrossRef]
  52. Sonoki, H.; Tanimae, A.; Furuta, T.; Endo, S.; Matsunaga, T.; Ichihara, K.; Ikari, A. Caffeic Acid Phenethyl Ester Down-Regulates Claudin-2 Expression at the Transcriptional and Post-Translational Levels and Enhances Chemosensitivity to Doxorubicin in Lung Adenocarcinoma A549 Cells. J. Nutr. Biochem. 2018, 56, 205–214. [Google Scholar] [CrossRef]
  53. Anjaly, K.; Tiku, A.B. Caffeic Acid Phenethyl Ester Induces Radiosensitization via Inhibition of DNA Damage Repair in Androgen-Independent Prostate Cancer Cells. Environ. Toxicol. 2022, 37, 995–1006. [Google Scholar] [CrossRef]
  54. Muthusamy, G.; Balupillai, A.; Ramasamy, K.; Shanmugam, M.; Gunaseelan, S.; Mary, B.; Prasad, N.R. Ferulic Acid Reverses ABCB1-Mediated Paclitaxel Resistance in MDR Cell Lines. Eur. J. Pharmacol. 2016, 786, 194–203. [Google Scholar] [CrossRef]
  55. Muthusamy, G.; Gunaseelan, S.; Prasad, N.R. Ferulic Acid Reverses P-Glycoprotein-Mediated Multidrug Resistance via Inhibition of PI3K/Akt/NF-ΚB Signaling Pathway. J. Nutr. Biochem. 2019, 63, 62–71. [Google Scholar] [CrossRef]
  56. Heo, S.-K.; Noh, E.-K.; Yoon, D.-J.; Jo, J.-C.; Koh, S.; Baek, J.H.; Park, J.-H.; Min, Y.J.; Kim, H. Rosmarinic Acid Potentiates ATRA-Induced Macrophage Differentiation in Acute Promyelocytic Leukemia NB4 Cells. Eur. J. Pharmacol. 2015, 747, 36–44. [Google Scholar] [CrossRef]
  57. Yu, C.; Chen, D.; Liu, H.; Li, W.; Lu, J.; Feng, J. Rosmarinic Acid Reduces the Resistance of Gastric Carcinoma Cells to 5-Fluorouracil by Downregulating FOXO4-Targeting MiR-6785-5p. Biomed. Pharmacother. 2019, 109, 2327–2334. [Google Scholar] [CrossRef]
  58. Zhang, Y.; Chen, S.; Wei, C.; Rankin, G.O.; Rojanasakul, Y.; Ren, N.; Ye, X.; Chen, Y.C. Dietary Compound Proanthocyanidins from Chinese Bayberry (Myrica Rubra Sieb. et Zucc.) Leaves Inhibit Angiogenesis and Regulate Cell Cycle of Cisplatin-Resistant Ovarian Cancer Cells via Targeting Akt Pathway. J. Funct. Foods 2018, 40, 573–581. [Google Scholar] [CrossRef]
  59. Ravindranathan, P.; Pasham, D.; Goel, A. Oligomeric Proanthocyanidins (OPCs) from Grape Seed Extract Suppress the Activity of ABC Transporters in Overcoming Chemoresistance in Colorectal Cancer Cells. Carcinogenesis 2019, 40, 412–421. [Google Scholar] [CrossRef]
  60. Naus, P.J.; Henson, R.; Bleeker, G.; Wehbe, H.; Meng, F.; Patel, T. Tannic Acid Synergizes the Cytotoxicity of Chemotherapeutic Drugs in Human Cholangiocarcinoma by Modulating Drug Efflux Pathways. J. Hepatol. 2007, 46, 222–229. [Google Scholar] [CrossRef]
  61. Geng, N.; Zheng, X.; Wu, M.; Yang, L.; Li, X.; Chen, J. Tannic Acid Synergistically Enhances the Anticancer Efficacy of Cisplatin on Liver Cancer Cells through Mitochondria-Mediated Apoptosis. Oncol. Rep. 2019, 42, 2108–2116. [Google Scholar] [CrossRef]
  62. Yu, F.; Liu, W.; Gong, X.R.; Zhou, Y.B.; Lin, Y. Procyanidins Enhance the Chemotherapeutic Sensitivity of Laryngeal Carcinoma Cells to Cisplatin through Autophagy Pathway. Lin Chuang Er Bi Yan Hou Tou Jing Wai Ke Za Zhi J. Clin. Otorhinolaryngol. Head Neck Surg. 2018, 32, 447–456. [Google Scholar]
  63. Mady, F.M.; Shaker, M.A. Enhanced Anticancer Activity and Oral Bioavailability of Ellagic Acid through Encapsulation in Biodegradable Polymeric Nanoparticles. Int. J. Nanomed. 2017, 12, 7405. [Google Scholar] [CrossRef]
  64. Cetin, A.; Biltekin, B.; Degirmencioglu, S. Ellagic Acid Enhances the Antitumor Efficacy of Bevacizumab in an in Vitro Glioblastoma Model. World Neurosurg. 2019, 132, e59–e65. [Google Scholar] [CrossRef]
  65. Cetin, A.; Biltekin, B. Ellagic Acid Enhances Antitumor Efficacy of Temozolomide in an in Vitro Glioblastoma Model. Turk Neurosurg 2020, 30, 813–821. [Google Scholar] [CrossRef]
  66. Wu, Y.-S.; Ho, J.-Y.; Yu, C.-P.; Cho, C.-J.; Wu, C.-L.; Huang, C.-S.; Gao, H.-W.; Yu, D.-S. Ellagic Acid Resensitizes Gemcitabine-Resistant Bladder Cancer Cells by Inhibiting Epithelial-Mesenchymal Transition and Gemcitabine Transporters. Cancers 2021, 13, 2032. [Google Scholar] [CrossRef]
  67. Moon, J.Y.; Ediriweera, M.K.; Ryu, J.Y.; Kim, H.Y.; Cho, S.K. Catechol Enhances Chemo-and Radio-Sensitivity by Targeting AMPK/Hippo Signaling in Pancreatic Cancer Cells. Oncol. Rep. 2021, 45, 1133–1141. [Google Scholar] [CrossRef]
Figure 1. Mechanisms of chemoresistance of cancer cells against anticancer drugs.
Figure 1. Mechanisms of chemoresistance of cancer cells against anticancer drugs.
Cancers 14 04573 g001
Figure 2. Chemical structures of flavonoids that improve the chemosensitivity of anticancer drugs.
Figure 2. Chemical structures of flavonoids that improve the chemosensitivity of anticancer drugs.
Cancers 14 04573 g002
Figure 3. Mechanisms of chemosensitivity of apigenin towards sisplatin.
Figure 3. Mechanisms of chemosensitivity of apigenin towards sisplatin.
Cancers 14 04573 g003
Figure 4. Chemical structures of anthocyanidins that improve chemosensitivity of anticancer drugs.
Figure 4. Chemical structures of anthocyanidins that improve chemosensitivity of anticancer drugs.
Cancers 14 04573 g004
Figure 5. Chemical structures of phenolic acids that improve the chemosensitivity of anticancer drugs.
Figure 5. Chemical structures of phenolic acids that improve the chemosensitivity of anticancer drugs.
Cancers 14 04573 g005
Figure 6. Chemical structures of tannins that improve chemosensitivity of anticancer drugs.
Figure 6. Chemical structures of tannins that improve chemosensitivity of anticancer drugs.
Cancers 14 04573 g006
Table 1. Flavonoids improve the chemosensitivity of chemotherapeutic drugs in cancer.
Table 1. Flavonoids improve the chemosensitivity of chemotherapeutic drugs in cancer.
MoleculesOriginsExperimental ApproachesKey ResultsReferences
Apigenin SynthetizedMDA435/LCC6 and P388 cells
Cell proliferation assay
ATPase assay
Enhanced the cytotoxicity of paclitaxel (PTX), doxorubicin (Dox), daunomycin (DM), vincristine (VCR), and vinblastine, resulting in a reduction of IC50 by 5–50 times[11]
PurchasedParental human HCC cells (BEL-7402) and BEL-7402/ADM cells
MTT assay
Cell cycle analysis
Real-time quantitative PCR
Western blot analysis
In vivo xenograft studies
Sensitized Dox-resistant BEL-7402 (BEL- 7402/ADM) cells to Dox
Increased intracellular concentration of Dox
Reduced Nrf2 expression
APG + Dox (in vivo) inhibited tumor growth, reduced cell proliferation, and induced apoptosis more substantially when compared with Dox treatment alone
[12]
PurchasedHuman pancreatic cancer cell line BxPC-3 Human pancreatic ductal epithelium (HPDE) cells
Western blot analysis
MTS cell proliferation assay
APG (13 μM) + gemcitabine (Gem) (13 μM) inhibited cell proliferation
APG (11–19 μM) + Gem (10 μM) inhibited growth by 59–73%
Enhanced the anti-proliferative activity of chemotherapeutic drugs
[13]
PurchasedHuman pancreatic cancer cell lines AsPc-1, Panc-1, and MiaPaCa-2
MTT assay
Cell apoptosis assay
Western blot analysis
In vitro IKK-β kinase activity assay
Xenograft model
Reduced cell growth
Induced cell apoptosis
Down-regulated the TNF-α-induced NF-κB DNA binding activity
Suppressed pancreatic cancer growth and IKK-β activation in nude mice xenograft
[14]
PurchasedLaryngeal carcinoma Hep-2 cell line
RT-PCR
Cell counting Kit-8 (CCK-8) system
Western blot analysis
Enhanced the cisplatin (CP)-induced suppression of Hep-2 cell growth in a concentration- and time-dependent manner
Reduced the levels of GLUT-1 mRNA and GLUT-1 and p-Akt proteins in CP-treated Hep-2 cells in a concentration and time-dependent manner
[15]
Purchased Laryngeal hep-2 carcinoma cell line
Nude mouse model of laryngeal carcinoma
Western blot analysis
Improved xenograft radio-sensitivity
Reduced the expression of PI3K mRNA, Akt, and GLUT-1 after X-ray radiation
[16]
PurchasedTumor xenografts in nude mice
SK-Hep-1 and BEL-7402 cells
MTT assay
Annexin V/PI assay
Western blotting analysis
Cellular ROS detection
Enhanced the cytotoxicity of 5-FU in HCC cells
APG + 5-fluorouracil (5-FU) (in vivo) inhibited HCC xenograft tumor growth
APG + 5-FU increased the levels of reactive oxygen species (ROS)
APG + 5-FU decreased the mitochondrial membrane potential (ΔΨm)
APG + 5-FU decreased Bcl-2 expression
[17]
PurchasedHuman renal proximal tubular epithelial (HK-2) cells
MTT assay
Analysis of cell morphology and cell cycleCaspase-3 activity assay
Western blot analysis
ROS production assay
Inhibited the CP-induced apoptosis of HK-2 cells
Induced cell cycle arrest
Inhibited caspase-3 activity and PARP cleavage
Reduced CP-induced phosphorylation and expression of p53
Promoted the CP-induced Akt phosphorylation
[18]
Purchased80 Swiss albino male mice
ELISA
Quantitative real-time RT-PCR
Histopathological and immunohistochemical analysis
APG alone or combined with 5-FU
Increased Beclin-1 levels, caspase-3 and -9, and JNK activities,
decreased tumor volume, Mcl-1expression, and total antioxidant capacity,
alleviated histopathological changes, and
decreased Ki-67 proliferation index
[19]
Not reported BEL-7402 and BEL-7402/ADM cells
TUNEL assay
qRT-PCR
Annexin V-FITC/PI apoptosis assay
Western blot analysis
Reversed Dox sensitivity
Induced the caspase-dependent apoptosis in BEL-7402/ADM cells
Induced the miR-101 expression
[20]
Not reportedHuman hepatocellular carcinoma (HCC) and adjacent normal tissue specimens
qRT-PCR
MTT assay
Western blot analysis
In vivo xenograft studies
Enhanced Dox sensitivity
Induced miR-520b expression
Inhibited ATG7-dependent autophagy in BEL-7402/ADM cells
Inhibited hepatocellular carcinoma xenograft growth
[21]
PurchasedOvarian cancer-sensitive cell line SKOV3
Ovarian cancer drug-resistant cell line SKOV3/DDP
MTT assay
PCR test
Western blot test
Apoptosis test
Enhanced the chemosensitivity of ovarian cancer-sensitive cells and drug-resistant cells
Induced the apoptosis of ovarian cancer cells by down-regulating the Mcl-1 gene
[22]
Quercetin PurchasedDB-1 melanoma and SK Mel 28 cell lines Western blot analysis
Annexin V-FITC staining
RNA isolation and RT-PCR
Immunocytochemistry
siRNA transfection
Induced a redistribution of ΔNp73 in the cytoplasm and nucleus
Que + temozolomide (TMZ) abolished drug insensitivity and caused a more than additive induction of apoptosis
[23]
Not reported Human esophageal cancer cells (EC9706 and Eca109)
MTT assay
Annexin V-FITC/propidium iodide (PI)-stained fluorescence-activated cell sorting (FACS)
Western blot analysis
Que + 5-FU inhibited growth and stimulated apoptosis in EC9706 and Eca109 esophageal cancer cells compared to Que[24]
PurchasedMCF-7 and MCF-7/Dox cells
MTT assay
Flow cytometry
Matrigel invasion assay
Western blot analysis
Increased intracellular concentration of Dox
Improved Dox cytotoxicity
Que + Dox inhibited cell proliferation and invasion and suppressed HIF-1α and P-gp expression
[25]
Not reportedHuman ovarian cancer cell lines, SKOV-3, EFO27, OVCAR-3, and A2780P
Evaluation of quercetin toxicity
SRB staining
Inhibited proliferation and increased sensitivity of ovarian cancer cells to CP and PTX[26]
PurchasedU251 and U87 human glioblastoma cells
MTT assay
Flow cytometry
Western blot analysis
Que (30 μmol/L) + TMZ (100 μmol/L) inhibited cell viability and enhanced TMZ inhibition
Que did not affect the caspase-3 activity and cell apoptosis, whereas combined with TMZ, it increased the caspase-3 activity and induced cell apoptosis.
[27]
PurchasedMCF-7 cells and MCF-7/Dox cells
MTT assay
Flow cytometry
Que + Dox inhibited cell proliferation and invasion
Que + Dox increased cell apoptosis
Que + Dox up-regulated PTEN expression
Que + Dox down-regulated p-Akt expression
[28]
PurchasedLung cancer cells (A549 and H460 cells)
Western blot analysis
Reduced cell viability
Suppressed HSP70 expression
Improved Gem-induced cell death linked to increased caspase-3 and caspase-9 activities
Que + Gem down-regulated HSP70 expression more significantly than treatment with Que or Gem alone
[29]
Not reportedBEL-7402 and multidrug-resistant cell line BEL/5-FU
MTT assay
Flow cytometry
Real-time PCR
Western blot analysis
Increased intracellular accumulation of Dox
Increased sensitivity of BEL/5-FU cells to chemotherapeutic drugs
Down-regulated the expressions of ABCB1, ABCC1, and ABCC2
Inhibited the functions and expressions of ABCB1, ABCC1, and ABCC2 efflux pump
[30]
PurchasedHuman prostate cancer cell line PC3
MTT assay
Western blot analysis
Flow cytometry
Inhibited c-met expression and the downstream PI3K/AKT pathway
Que + Dox promoted the Dox-induced cell apoptosis through the mitochondrial/ROS pathway
[31]
Purchased Human pancreatic cancer cell lines
Transfection of small interfering RNA
MTT assay
Western blot analysis
Cell cycle measurement
Attenuated RAGE expression to facilitate cell cycle arrest, autophagy, apoptosis, and GEM chemosensitivity in MIA Paca-2 GEMR cells[32]
Purchased Human prostate cancer (PC-3) cell lines
Nude male BALB/c mice
MTT assay
Intracellular ROS content assays
RNA extraction and qRT-PCR
Western blot analysis
Immunohistochemistry
Que + PTX inhibited cell proliferation, increased apoptosis, arrested cell cycle at the G2/M phase, inhibited cell migration, induced ER stress, and increased ROS generation
Que + PTX exerted the most beneficial therapeutic effects (in vivo)
Increased the cancer cell-killing effects of PTX (in vivo)
[33]
Not reportedMCF 7 cells
MTT assay
Flow cytometry
qRT-PCR
ELISA
Quer + 5-FU improved apoptosis by increasing the gene expression of Bax and p53 and caspase-9 activity and decreasing Bcl2 gene expression
Quer + 5-FU decreased colony formation
[34]
Purchased MDA-MB-231 human breast cancer cell line
MTT assay
Flow cytometry
qRT-PCR
Western blot analysis
Decreased cell viability
Que (95 μM) + docetaxel (7 nM) up-regulated p53, increased BAX levels, and decreased levels of BCL2, pERK1/2, AKT, and STAT3 proteins
[35]
Kaempferol PurchasedHuman myelogenous leukemia K562 cells and the adriamycin-resistant variant K562/A cells
MTT assay
Annexin V/PI analysis
PCR array
Kae + Que inhibited the growth of both cells
Kae + Que increased the sensitivity of both cells
Kae + Que induced apoptosis
Kae + Que influenced the expression of drug transporter genes
[36]
PurchasedLS174 colon cancer cells
MTT assay
Colony formation assay
Spheroid generation
Sensitization assay
Measurement of ROS
Western blot analysis
qRT-PCR
Chemo-sensitized 5-FU-resistant LS174-R cells
Blocked the production of ROS and modulated the expression of JAK/STAT3, MAPK, PI3K/AKT, and NF-κBKae + 5-FU exerted a synergistic inhibitory effect on cell viability
Kae + 5-FU enhanced apoptosis and induced cell cycle arrest in chemo-resistant and sensitive cells
[37]
PurchasedHuman colorectal cancer cell line HCT8
5-FU-resistant cell line HCT8-R
CCK-8 assay
qPCR assay
Western blot analysis
Clonogenic assay
Reversed the drug resistance of HCT8-R cells to 5-FU
Reduced glucose uptake and lactic acid production in drug-resistant colorectal cancer cells
Promoted the expression of microRNA-326 in colon cancer cells
Reversed the resistance of colorectal cancer cells to 5-FU
[38]
Myricetin Purchased Esophageal carcinoma EC9706 cells
Colony formation assays
Flow cytometry
Western blot analysis
Nude mouse tumor xenograft model
MYR + 5-FU suppressed cell survival fraction and proliferation, and increased cell apoptosis
MYR + 5-FU decreased survivin, cyclin D, and Bcl-2, and increased the expression level of caspase-3 and p53
MYR + 5-FU reduced the growth rate of tumor xenografts in mice
[39]
PurchasedA2780 and OVCAR3 ovarian cancer cells
MTT assay
Apoptosis assay
Boyden chamber assay
Western blot analysis
Induced cytotoxicity, with an IC50 value of 25 μM
Induced cell apoptosis, accompanied by the modulation of certain pro- and anti-apoptotic markers
Increased paclitaxel cytotoxicity
[40]
RutinPurchasedHuman breast cancer MDA- MB-231 cellsCalcein acetoxymethyl accumulation assayRhodamine-123 uptake assay
Annexin V and 7-aminoactinomycin D
Propidium iodide staining
Increased the anticancer activity of both chemotherapeutic agents
Decreased the activity of adenosine triphosphate binding cassette transporters
RTN (20 μM) enhanced cytotoxicity related to cyclophosphamide and methotrexate
RTN (20 and 50 μM) arrested the cell cycle at the G2/M and G0/G1 phases, respectively, thus promoting cell apoptosis
[41]
PurchasedHuman HCC cell lines
qRT-PCR
Luciferase reporter assay
Cell viability assay
Flow cytometry
In vivo tumor xenograft
Attenuated autophagy and BANCR expression in SO-resistant cells
Decreased the number of autophagosomes in HepG2/SO and HCCLM3/SO cells
Enhanced the efficacy of SO in a xenograft model of HCC in nude mice
[42]
HispidulinNot reported Human gallbladder carcinoma cell line GBC-SD
MTT assay
Western blot analysis
Flow cytometry
Caspase-3 activity assay
qRT-PCR
In vivo xenograft experiments
Inhibited the growth of GBC cells
Promoted apoptosis in GBC cells
Induced cell arrest at the G0/G1 phase
Exerted antitumor effect mediated through HIF-1α inhibition
Repressed the transactivation activity and expression of HIF-1α
Suppressed the HIF-1α expression via AMPK signaling
[43]
Table 2. Anthocyanidins that could enhance the chemosensitivity of cancer drugs.
Table 2. Anthocyanidins that could enhance the chemosensitivity of cancer drugs.
MoleculesOriginsExperimental ApproachesKey ResultsReferences
DelphinidinPurchasedA549 cell line (human, lung, and carcinoma)
MTT assay
Immunofluorescence staining
Western blot analysis
qRT-PCR
Induced apoptosis in A549 cells
Promoted apoptosis in the radiation-exposed A549 cells
Induced autophagy in radiation-exposed A549 cellsActivated autophagic cell death and the JNK/MAPK signaling pathway in radiation-exposed A549 cells
[45]
Cyanidin-3-glucoside (C3G)Purchased Human lung adenocarcinoma A549 cells
Immunoblotting
RNA isolation and qRT-PCR
Immunofluorescence measurement
Luciferase reporter assay
Reduced protein level of CLDN2 in A549 cells
Inhibited Akt phosphorylation
Increased p38 phosphorylation
Reduced CLDN2 expression at transcriptional and post-translational steps mediated by Akt inhibition and p38 activation, respectively
Improved Dox accumulation and cytotoxicity in spheroid models
Increased the percentages of apoptotic and necrotic cells induced by Dox
[46]
Anthocyanins (ACNs)Black raspberryColon cancer cell lines, SW480 and Caco2MTT assay
Colony formation assays
Western blot analysis
Establishment of colitis-induced colon cancer mice model
Improved the chemotherapy efficacy of 5-FU and celecoxib
ACNs + (5-FU or celecoxib) inhibited CRC cell proliferation (in vitro) and decreased the number of tumors in AOM-induced CRC mice (in vivo)
[44]
Table 3. Phenolic acids that improve the chemosensitivity of cancer drugs.
Table 3. Phenolic acids that improve the chemosensitivity of cancer drugs.
MoleculesOriginsExperimental ApproachesKey ResultsReferences
Caffeic acid phenethyl ester (CAPE)Not reportedMouse CT26 colorectal adenocarcinoma cells
BALB/c mouse with CT26 cells implantation
Colony formation assay
RT-PCR
Flow cytometry
Depleted intracellular GSH in CT26 cells, but not in bone marrow cells
Enhanced cell killing by IR
Increased glutathione peroxidase, decreased glutathione reductase in CT26 cells
Reversed radiation-activated NF-κB
Induced a significant inhibition of tumor growth and prolongation of survival compared to IR alone (in vivo)
[49]
Purchased Human medulloblastoma Daoy cell line and Human astroglia SVGp12
MTT and trypan blue exclusion assays ELISATUNEL assay
Flow cytometry
Western blot analysis
Inhibited Daoy cell growth in a time- and concentration-dependent manner
Decreased G2/M fraction and increased S phase fractionDown-regulated expression of cyclin B1 protein
Reduced the viability of irradiated Daoy cells
No chemosensitizing effect on Dox or CP
[50]
PurchasedParental and the drug-resistant cells of stomach (MKN45) and colon (LoVo) cancersPotentiated the apoptotic effects of Dox and CP against parental cells
Reduced the production of Dox-induced ROS
Reduced 26S proteasome-based proteolytic activities in parental MKN45 cells
Up-regulated and significantly decreased chymotrypsin-like activity in Dox- or CP-resistant cells
[51]
Not reported Human lung adenocarcinoma A549 and RERF-LC-MS cell lines
Immunoblotting
RNA isolation and PCR
Luciferase reporter assay
Immunocytochemistry
Decreased claudin-2 protein level in a concentration-dependent manner
Decreased (at 50 µM) mRNA level and promoter activity
Decreased (at 50 µM) the level of p-NF−κB, and increased that of IκB
Increased the expression and activity of protein phosphatase (PP) 1 and 2A
Suppressed cell proliferation
Enhanced Dox toxicity and accumulation in 3D spheroid cells
[52]
Not reported Prostate cancer (PCa) cells, DU145 and PC3 Evaluated the radiomodulatory potential of CAPECAPE + gamma radiation (GR) sensitized PCa cells to radiation in a concentration-dependent manner
Improved the level of ionizing radiation (IR)-induced gamma H2AX foci and cell death by apoptosis
CAPE + GR decreased the migration potential of PCa cells
Sensitized PCa cells to radiation in vitro and induced apoptosis, increased Akt/mTOR phosphorylation and hampered cell migration
CAPE + IR inhibited cell growth by decreasing RAD50 and RAD51 proteins
[53]
Ferulic acid (FA)PurchasedMultidrug resistance (MDR) cell lines
MTT assay
Colony formation assay
Fluorescence microscopic analysis
Cell cycle analysis
Tryptophan fluorescence quenching
PCR array
Western blot analysis
Inhibited P-glycoprotein transport function in drug-resistant
KB ChR8-5 cell lines
Down-regulated ABCB1 expression in a concentration-dependent manner
Decreased paclitaxel resistance in KBChR8-5 and HEK293/ABCB1 cells
Enhanced paclitaxel-mediated cell cycle arrest and up-regulated paclitaxel-induced apoptotic signaling in KB-resistant cells
[54]
Purchased Parental KB cells and P−gp overexpressing KB ChR8-5 cell lines
MTT assay
γH2AX assay
Western blot analysis
Immunocytochemistry
Animals and tumor xenograft experiments
Increased the cytotoxicity of Dox and VCR in the P-gp overexpressing KB ChR8-5 cells
Enhanced the formation of Dox-induced γH2AX foci and synergistically increased Dox-induced apoptotic signaling in drug-resistant cells
FA + Dox reduced KB ChR8−5 tumor xenograft size three-fold compared to the group treated with Dox alone
Reversed MDR by suppressing P-gp expression via inhibition of PI3K/Akt/NF−κB signaling pathway
[55]
Rosmarinic acid (RA)PurchasedHuman acute promyelocytic leukemia NB4 cells
Flow cytometry analysis
Phagocytosis assay
qRT-PCR
Potentiated ATRA-induced macrophage differentiation in APL cells[56]
Not reportedHuman gastric carcinoma cell line SGC7901
Apoptosis assay
CCK8 assay
Apoptosis assay
RNA isolation and microarray
qRT-PCR
Luciferase reporter assay
Western blot analysis
Increased the chemosensitivity of SGC7901 cells to 5-FUReduced IC50 of 5-FU (70.43 ± 1.06 μg/mL) compared to untreated SGC7901/5-FU cells (208.6 ± 1.09 μg/mL)
RA + 5-FU increased apoptosis rate
Reduced the expression levels of two miRNAs (miR-642a−3p and miR−6785-5p)
Reduced P-gp expression and increased Bax expression in SGC7901/5-FU and SGC7901/5-FU -Si cells
[57]
Table 4. Condensed tannins that could improve the chemosensitivity of cancer drugs.
Table 4. Condensed tannins that could improve the chemosensitivity of cancer drugs.
MoleculesOriginsExperimental ApproachesKey ResultsReferences
Proanthocyanidins Chinese bayberry leavesPlatinum-resistant human ovarian cancer cell line OVCAR-3
Flow cytometry
MTT assay
Colony formation assay
Western blot assay
Induced inhibitory effects on the growth and CSC characteristics of OVCAR−3 SP cells
Reduced the expression of β-catenin, cyclin D1, and c-Myc and inhibited the self-renewal capacity of cells
Induced G1 cell cycle arrest in OVCAR−3 SP cells
[58]
Grape seed extractColorectal cancer cell lines, HCT116 and H716
Cell cycle and apoptosis analysis
Cell viability and proliferation
mRNA expression analysis
Genome-wide RNA-sequencing analysisXenograft animal experiments
Sensitized acquired (HCT116-FOr cells) and innately chemoresistant (H716 cells) cancer cells to 5-FU and oxaliplatin (OXP)
PCs + (5-FU and OXP) inhibited the growth of chemoresistant cells and decreased the expression of several key adenosine triphosphate-binding cassette (ABC) transporters
Sensitized chemoresistant cells to 5-FU and OXPPCs + (5-FU and OXP) reduced chemoresistant xenograft tumor growth in mice
[59]
Tannic acid Purchased Malignant human cholangiocytes
Calcein retention assays
Western blot analysis
RT-PCR
Decreased malignant cholangiocyte growth
Exhibited a synergistic effect with mitomycin C and 5-FU but not with Gem
Decreased calcein efflux and expression of PGP, MRP1, and MRP2 membrane efflux pumps
[60]
Purchased Liver cancer cell line HepG2
MTT assay
Mitochondrial transmembrane potential qRT-PCR
Western blot analysis
Inhibited HepG2 cell growth
TA + CP induced mitochondria-mediated apoptosis in HepG2 cells and enhanced growth inhibitory effect compared to treatment alone
[61]
ProcyanidinsNot reported Laryngeal cancer cell line TU686
Flow cytometry
Cell immunofluorescence staining
Western blot analysis
Inhibited TU686 cells in a concentration-dependent manner for 24 h
Induced apoptosis of TU686 cells
Increased expression of LC3−Ⅱ and Caspase-3
[62]
Ellagic acid PurchasedColorectal carcinoma HT−29, Colo 320DM, SW480, and LoVo cells
Trypan blue exclusion
Annexin−V labeling
Mitochondrial membrane potential (Δψm)
Immunoblotting
EA + 5-FU inhibited cell proliferation of HT-29, Colo 320DM and SW480 cells
EA + 5-FU increased apoptotic cell death of HT−29 and Colo 320DM cells
EA potentiated 5-FU chemosensitivity in at least three colorectal cancer cell lines
[47]
PurchasedEpithelial ovarian cancer cell line A2780MTT assay
Immunoblot analysis
Signal pathway analysis
Cell cycle analysis
Enhanced CP cytotoxicity in A2780CisR cells
Prevented the development of CP resistance
[48]
Purchased Caco-2 and HTC-116 cells
MTT assay
In vitro drug release
Male New Zealand white rabbits
Induced higher cell viability than EA-NP treated HCT−116 cells
Oral administration of EA-NPs caused a 3.6-fold increase in the area under the curve compared to that of EA (in vivo)
[63]
Purchased Rat C6 glioma cells
Immunohistochemistry
RT-PCR
Reduced MGMT expression
Affected the apoptotic proteins of p53 and caspase-3 at the protein level, but not at the gene level
EA + bevacizumab (BEV) reduced cell viability
EA + BEV reduced MDR1 expression only at 72 h
[64]
PurchasedRat C6 glioma cells
Immunocytochemistry
RT-PCR
EA + TMZ reduced cell viability
Down-regulated MGMT expression independent of the presence of TMZ
EA + TMZ reduced MDR1 expression only over 48 h compared to TMZ alone
Up-regulated caspase-3 at 48 h, but up-regulated p53 at 48 and 72 h
EA + TMZ enhanced immunoreactivities of p53 and caspase-3 proteins, but not of the genes
[65]
PurchasedFour human bladder cancer cell lines, TSGH−8301, TSGH-9202, T24, and J82
MTT assay
Flow cytometry
Cell migration and invasion assays
Western blot analysis
qRT−PCR
Xenograft model
Induced high cytotoxicity of Gem in GEM−resistant cells
EA + Gem increased apoptosis and reduced cell motility in GCB-resistant cells
Resensitized bladder cancer cells to Gem by reducing the epithelial–mesenchymal transition
Reduced EMT by inhibiting the TGFβ−SMAD2/3 upward signaling pathway
Inhibited the growth of bladder cancer tumors and increased the in vivo inhibitory effects of Gen on tumors
[66]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Bouyahya, A.; Omari, N.E.; Bakrim, S.; Hachlafi, N.E.; Balahbib, A.; Wilairatana, P.; Mubarak, M.S. Advances in Dietary Phenolic Compounds to Improve Chemosensitivity of Anticancer Drugs. Cancers 2022, 14, 4573. https://doi.org/10.3390/cancers14194573

AMA Style

Bouyahya A, Omari NE, Bakrim S, Hachlafi NE, Balahbib A, Wilairatana P, Mubarak MS. Advances in Dietary Phenolic Compounds to Improve Chemosensitivity of Anticancer Drugs. Cancers. 2022; 14(19):4573. https://doi.org/10.3390/cancers14194573

Chicago/Turabian Style

Bouyahya, Abdelhakim, Nasreddine El Omari, Saad Bakrim, Naoufal El Hachlafi, Abdelaali Balahbib, Polrat Wilairatana, and Mohammad S. Mubarak. 2022. "Advances in Dietary Phenolic Compounds to Improve Chemosensitivity of Anticancer Drugs" Cancers 14, no. 19: 4573. https://doi.org/10.3390/cancers14194573

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