Biomolecules & Therapeutics 2022; 30(6): 479-489  https://doi.org/10.4062/biomolther.2022.017
Modulation of Reactive Oxygen Species to Overcome 5-Fluorouracil Resistance
Kyung-Soo Chun1 and Sang Hoon Joo2,*
1College of Pharmacy, Keimyung University, Daegu 42601,
2Department of Pharmacy, Daegu Catholic University, Gyeongsan 38430, Republic of Korea
*E-mail: sjoo@cu.ac.kr
Tel: +82-53-850-3614, Fax: +82-53-359-6729
Received: January 28, 2022; Revised: March 29, 2022; Accepted: March 30, 2022; Published online: April 20, 2022.
© The Korean Society of Applied Pharmacology. All rights reserved.

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.
Abstract
5-Fluorouracil (5-FU) remains to be an important chemotherapeutic drug for treating several cancers when targeted therapy is unavailable. Chemoresistance limits the clinical utility of 5-FU, and new strategies are required to overcome the resistance. Reactive oxygen species (ROS) and antioxidants are balanced differently in both normal and cancer cells. Modulating ROS can be one method of overcoming 5-FU resistance. This review summarizes selected compounds and endogenous cellular targets modulating ROS generation to overcome 5-FU resistance.
Keywords: Reactive oxygen species, Cancer, Resistance, 5-Fluorouracil
INTRODUCTION

Despite the introduction of targeted anticancer therapy, 5-fluorouracil (5-FU) remains an important chemotherapeutic drug for treating several cancers, including colorectal, breast, and gastric cancer. 5-FU’s cytotoxic mechanism involves the inhibition of thymidylate biosynthesis or the misincorporation of fluorinated nucleotides into newly synthesized DNA or RNA (Longley et al., 2003). It can be effective in the treatment of cancer when targeted therapy is unavailable. As described in previous studies, the development of prodrugs such as capecitabine has improved the limitation of 5-FU due to poor oral absorption (Pazdur et al., 1998). Furthermore, combination chemotherapy improved 5-FU’s anticancer effect, as demonstrated by FOLFOX (folinic acid, 5-FU, and oxaliplatin) and FOLFIRI (folinic acid, 5-FU, and irinotecan) (Souglakos et al., 2006). Combining chemotherapeutics with different mechanisms could overcome the heterogeneity of tumor cells and decrease the development of resistance (Frei et al., 1998). Nevertheless, the overall response rate remains less than 50% (Mehrzad et al., 2016) due to the cells being resistant to chemotherapy.

Studies have been conducted to elucidate the 5-FU resistance mechanism described elsewhere (Blondy et al., 2020). The generation of reactive oxygen species (ROS) frequently correlates with the induction of apoptosis in many cancer cells; modulation of ROS may be one mechanism by which cancer cells avoid the cytotoxicity induced by 5-FU (Mates and Sanchez-Jimenez, 2000). For example, in human lung carcinoma cells NCI-H1299, the expression of reactive oxygen modulator 1 (Romo1) is elevated, and the cellular level of ROS is high. At the same time, tumor cells maintain high levels of antioxidant enzymes and antiapoptotic Bcl-2 family proteins, most likely to reduce oxidative stress (Hwang et al., 2007). This implies that cancer cells prefer a high level of ROS while keeping the protective mechanisms running to minimize the unwanted toxicity of ROS.

ROS have a versatile role in cancer cell biology (Liou and Storz, 2010). When elevated, ROS are thought to act as mitogens, inducing cancer cell proliferation (Torres and Forman, 2003). DNA damage from oxidative stress may lead to mutations that can either activate oncogenes or inactivate tumor suppressor genes (Wei, 1992). ROS production is minimal in normal cells, and antioxidant functions effectively remove ROS (Fig. 1A). Increased ROS production is frequently observed in cancer cells with a poor prognosis (Kumar et al., 2008). Cancer cells maintain a relatively high level of ROS, likely due to the tumor-promoting effects of ROS such as angiogenesis (Ushio-Fukai and Nakamura, 2008), metastasis (Nishikawa, 2008), and proliferation (Juhasz et al., 2017). As shown in Fig. 1B, cancer cells increase the level of antioxidant systems in response to elevated levels of ROS to protect themselves from oxidative stress (Gorrini et al., 2013). Many anticancer drugs, including 5-FU, induce high levels of ROS to exert cytotoxic effects. Cancer cells adapt to the escalated ROS level by expressing even more antioxidant systems (Fig. 1C) (Liu et al., 2016b). When there is insufficient protection from high levels of ROS, cancer cells may not survive (Fig. 1D).

Figure 1. Balance between ROS production and antioxidant function. (A) ROS (water in the figure) are produced by various mechanisms (drawn as a water tap), and antioxidant function (drawn as a drain) effectively removes them, allowing physiological ROS levels to remain low. (B) Increased ROS generation is frequently observed in cancer cells, and cancer cells increase the level of antioxidant functions accordingly. The cellular level of ROS increases but not to toxic levels. (C) Even higher antioxidant function accompanies ROS overproduction when cancer cells adapt to chemotherapy. (D) Decrease of antioxidant function may result in cellular toxicity.

In this study, we summarized our understanding of natural and synthetic compounds (Table 1) and identified possible cellular targets involved with the modulation of cellular ROS levels to overcome 5-FU resistance.

Table 1 Selected compounds increasing ROS generation to overcome 5-FU resistance

CompoundCell/tissue typeEffects/Mechanisms
tetrathiomolybdateOvarian cancer cellsStress-mediated apoptosis↑, activation of JNK and p38 MAPK↑
TPENColon cancer HCT116 cellsMitochondrial membrane potential (MMP)↓
apigeninHepatocellular carcinoma cellsMitochondrial apoptosis↑
Polyphenolics from quinceColon cancer cells LS174NF-κB activation↓, cell cycle progression↓, angiogenesis↓
kaempferolColon cancer cells LS174Activation of STAT3↓, angiogenesis↓
shikoninGastric cancer SGC-7901Translocation of AIF and Endo G into nucleus
proanthocyanidinBreast cancer MDA-MB-231 cellsG2/M cell cycle arrest↑, MMP↓
B63 (curcumin analog)Gastric cancer cells SGC-7901 etc.Expression of Thioredoxin reductase 1↓
dimethoxycurcuminColon cancer cells SW480, SW620Expression of Bax and cyt C↑, expression of Bcl-2↓
Sanguisorba officinalis L. radixColorectal cancer cells RKO, HCT116Bax/Bcl-2 disruption↑, autophagy↑
manuka honeyColon cancer cells HCT116Expression of EGFR, HER2, Akt and mTOR↓
emodinBreast cancer MCF7 cellsExpression of E2F1 and NRPARP↓
gypenosideColorectal cancer cells SW-480,SW-620 and Caco2DNA damage induction↑, expression of p53↑
tubeimoside-IColorectal cancer cells SW480, SW620, HCT116, and RKOActivation of AMPK↑
oridoninColorectal cancer cells HCT115Activation of JNK/c-Jun pathway↓
Coptis herb extractsLung cancer A549 cellsROS↑
mahanineColorectal cancer cells HCT116, SW480Expression of PTEN and p53 in nucleus↑
caffeineLiver cancer cells HepG2, HLF, Huh7, etc.Cleavage of PARP↑, expression of Bcl-2 and Bcl-xL↓
selenocysteineSkin cancer cells A375Activation of ERK/Akt signaling↓
allicinLiver cancer cells SK-Hep-1, BEL-7402ROS↑, MMP↓
3-bromypyruvateLiver cancer cells SNU449, Hep3BROS↑, MMP↓

NATURAL/SYNTHETIC COMPOUNDS THAT MODULATE ROS TO OVERCOME 5-FU RESISTANCE

Metal chelators

Tetrathiomolybdate, a copper-chelating drug, was initially developed as an anticopper and antiangiogenic agent to treat Wilson’s disease (Brewer et al., 1991). Interestingly, it enhances the activity of the anticancer drug doxorubicin, a DNA intercalator in ovarian cancer cells (Kim et al., 2011). Tetrathiomolybdate increased the cytotoxicity of doxorubicin at subcytotoxic levels, likely by targeting antioxidant enzymes such as copper/zinc–superoxide dismutase (SOD). Furthermore, by generating ROS, tetrathiomolybdate increased the cytotoxicity of several anticancer drugs, including 5-FU and mitomycin C (Kim et al., 2012). The production of ROS induced by tetrathiomolybdate resulted in the activation of stress-mediated apoptosis, JNK, and p38 mitogen-activated protein kinase (MAPK), which increased cytotoxicity.

N,N,N′,N′-tetrakis-[2-pyridylmethyl]-ethylenediamine (TPEN) was reported to have a cancer-specific copper chelation mediated cytotoxicity (Fatfat et al., 2014). Additionally, TPEN treatment resulted in the excessive generation of ROS via the formation of the TPEN-copper complex, leading to cytotoxicity in human colon cancer HCT116 cells. Evidently, elevated copper levels may be important in maintaining the proper level of ROS generation in cancer cells, whereas intracellular copper levels are crucial to maintaining the proper level of ROS generation in cancer cells (Gupte and Mumper, 2009). Furthermore, although cellular copper levels may be a target for cancer treatment, it remains to be seen whether a copper-chelating drug can help overcome 5-FU resistance.

Phenolic compounds

Interestingly, several antioxidant compounds promote the production of ROS in cancer cells. Although more research is needed to elucidate the precise mechanisms, these antioxidant compounds are thought to modulate ROS generation and increase the cytotoxicity of 5-FU. Phenolic compounds refer to diverse natural products such as flavanols, flavonols, chalcones, tannins, curcuminoids, etc. Their antioxidant function is usually attributed to the phenolic ring structure (Cai et al., 2006). The following sections list several phenolic compounds (Fig. 2) that have been reported to have synergistic cytotoxicity when combined with 5-FU or to be cytotoxic to 5-FU resistant cancer cells.

Figure 2. Phenolic compounds.

Apigenin is a flavonoid compound found in common fruits and vegetables that exhibits anti-inflammatory, antioxidant, and anticancer activity (Shukla and Gupta, 2010). Research revealed that apigenin cotreatment with 5-FU at a subtoxic level demonstrated synergistic cytotoxicity in treating hepatocellular carcinoma (HCC) cells in vitro and in vivo (Hu et al., 2015). Moreover, the ROS level was increased, and the mitochondrial apoptotic pathway was activated, indicating that apigenin has a pro-oxidant function. Although it remains to be seen whether apigenin is cytotoxic to 5-FU resistant cancer cells, apigenin, which is well-known for its antioxidant activity, appears to also demonstrate some pro-oxidant activity.

The polyphenolic extract from quince (Cydonia oblonga Miller) has shown antiproliferative effects in kidney and colon cancer cells (Carvalho et al., 2010). A Tunisian research group reported that quince peel polyphenolic extract induced ROS production, and the cytotoxic effect of 5-FU was increased in human colon adenocarcinoma LS174 cell (Riahi-Chebbi et al., 2015). Although the potential expansion of the cellular work to a preclinical level requires further study, it is worth noting that ROS generation may be linked to the cytotoxicity of 5-FU. Riahi-Chebbi et al. (2019), conversely, reported that kaempferol, another phenolic compound derived from quince, inhibited the production of ROS while exhibiting the same cytotoxicity as other phenolic compounds and was effective even in 5-FU resistant colon cancer cells. This intriguing result cautions us not to assume that a decrease in ROS levels is cytoprotective, as other mechanisms may simultaneously be responsible for cytotoxicity.

Shikonin, a naphthoquinone derivative found in the shikonin plant (Lithospermum erythrohizon), is known for its cytotoxicity and anti-inflammatory activity (Chen et al., 2002). Similarly, Liang et al. (2016) studied the antitumor activity of shikonin on gastric cancer. They observed that shikonin induced ROS generation and enhanced the 5-FU sensitivity in vitro and in vivo. In addition to the mitochondria-mediated apoptotic pathway, they detected the caspase-independent nuclear translocation of the apoptosis-inducing factor and endonuclease G from mitochondria.

Proanthocyanidin compounds from white fig Ficus virens (Chen et al., 2017b) and Uncaria rhynchophylla (Chen et al., 2017c) have been shown to have cytotoxic activity on human breast cancer MDA-MB-231 cells. Proanthocyanidins increased cellular ROS and the mitochondrial apoptotic pathway, and synergistic cytotoxicity was observed when proanthocyanidins were combined with 5-FU. Surprisingly, the cytotoxic effect appeared to be cancer cell-specific, and proanthocyanidins alleviated intestinal mucositis in 5-FU-treated rats (Chen et al., 2017b).

Curcumin, a polyphenolic compound frequently found in curry powders, has long been considered an antioxidant (Ak and Gulcin, 2008). Several studies, however, have reported the generation of ROS by curcumin analogs. Researchers created B63, a curcumin analog, as an anticancer agent and discovered that B63 induced ROS-mediated paraptosis in gastric cancer cells (Chen et al., 2019). They showed the inhibition of thioredoxin reductase 1 (TrxR1) by B63 in vitro, and the overexpression of TrxR1 negated the proparaptotic activity of B63. Their findings indicate that TrxR1 is a target of B63 and that B63 effectively suppressed the growth of 5-FU-resistant gastric cancer cells. Similarly, dimethoxycurcumin increases ROS production in colon cancer cells, allowing it to exert cytotoxic activity against colon cancer SW480 and SW620 cells when combined with 5-FU (Zhao et al., 2017).

A Chinese research group studied the water extract of Sanguisorba officinalis L. radix, for its anticancer activity on human colorectal cancer HCT116 and RKO cells (Liu et al., 2016a). They demonstrated that treating cells with the extract and 5-FU significantly increased ROS generation and that cotreatment increased 5-FU cytotoxicity. Moreover, they reported an increase in autophagy-related markers, light chain LC3, and p62, besides ROS generation, implying that the generation of ROS is not the only explanation for the synergism between Sanguisorba officinalis L. radix and 5-FU. The study demonstrated that gallic acid, catechinic acid, and ellagic acid, three main constituents of Sanguisorba officinalis L. radix, are responsible for the synergistic activity.

Manuka honey, a type of honey collected from the manuka tree Leptospermum scoparium, has antioxidant, anti-inflammatory, and anticancer properties (Afrin et al., 2018b). Reports describe the synergistic cytotoxicity of manuka honey on human colon cancer HCT116 and LoVo cells when combined with 5-FU (Afrin et al., 2018a). Manuka honey, a polyphenol-rich natural product, suppressed cell survival signals in HCT116 and LoVo cells while inducing pro-apoptotic signals and ROS production. Furthermore, the combined treatment reduced the activity of antioxidant enzymes such as SOD, catalase, glutathione peroxidase, glutathione reductase, and the expression of Nrf2, SOD, catalase, and HO-1, resulting in increased cell death due to oxidative stress.

Emodin, a natural anthraquinone compound, has antiproliferative activity in human breast cancer MCF7 cells (Huang et al., 2007). In a later study, tests were conducted to determine whether low-dose emodin could potentiate the activity of 5-FU in MCF7 cells (Zu et al., 2018). Findings revealed that emodin increased 5-FU-induced apoptosis in breast cancer cells by generating ROS. Surprisingly, researchers observed cellular senescence after 5-FU treatment with emodin, which they believe was caused by the upregulation of cyclin-dependent kinase inhibitors and the downregulation of E2F1 and the notch-regulated ankyrin repeat protein (NRARP) protein. Their findings suggested that NRARP is a critical target for inducing cellular senescence.

Polycyclic compounds and alkaloids

Several polycyclic compounds and alkaloids (Fig. 3) have been investigated for their role in producing ROS in cancer cells. For instance, gypenosides are triterpenoid saponin compounds whose potential use in cancer treatment has been documented (Ahmad et al., 2019), and they are thought to have potentiated 5-FU’s cytotoxicity (Kong et al., 2015). Results showed that p53 and ROS generation mediates the synergism between gypenosides and 5-FU to exert anticancer activity. Additionally, the triterpenoid saponin compound, tubeimoside-I, isolated from Rhizoma Bolbostemmatis, has exhibited antitumor activity in various types of tumors (Yu et al., 1994). Yan et al. (2019) discovered that combining 5-FU and tubeimoside-I suppressed the growth of colorectal cancer SW480, SW620, HCT116, and RKO cells in a synergistic manner, whereas tubeimoside-I induced cellular ROS and the activation of AMPK, resulting in cytotoxic autophagy.

Figure 3. Polycyclic compounds and alkaloids.

Oridonin, a diterpenoid from the medicinal herb Rabdosia rubescens, exhibits antitumor activity (Li et al., 2011). Studies assessed oridonin’s anticancer effect in colorectal cancer HCT15 cells and compared the 5-FU resistant HCT15 cells and sensitive cells (Zhang et al., 2019). To exert its cytotoxicity, oridonin induced the generation of ROS in both cells and the activation of the JNK/c-Jun pathway. Notably, cotreatment with N-acetylcysteine reversed JNK/c-Jun pathway activation, indicating that ROS generation mediates JNK/c-Jun pathway activation. Although oridonin activated apoptosis in colorectal cancer cells, it appears to activate necroptosis in renal carcinoma 786-O cells (Zheng et al., 2018). Cotreatment of oridonin and 5-FU showed synergistic cytotoxicity, probably through separate mechanisms, and notably, the same compound showed a different mechanism of action.

The anticancer effects of the Coptis herb extracts and the major alkaloid component berberine have been well-reported, and their cytotoxic effects have been detected in various cancer cell lines (Tang et al., 2009). Furthermore, Coptis extract showed cytotoxicity when combined with 5-FU in human lung cancer A549 cells (He et al., 2012). The cytotoxicity of either Coptis extract or berberine was associated with an increase in ROS generation in a dose-dependent manner, and when combined with 5-FU, the anticancer effect was enhanced.

Mahanine, an alkaloid from the curry leaf plant (Murraya koenigii), has exhibited various biological activities (Ramsewak et al., 1999). Das et al. (2014) showed the synergistic enhancement of cytotoxicity of 5-FU when mahanine was used together in human colorectal cancer HCT116 and SW480 cells. Interestingly, the synergistic effect was observed irrespective of p53 status, i.e., both p53wt and p53null cells were sensitive to mahanine in combination with 5-FU. Although the precise mechanism is unknown, mahanine induced ROS production and led to the accumulation of PTEN and p53 in the nucleus. The increased production of ROS appears to be linked to the activation of tumor suppressor proteins PTEN and p53, resulting in increased cytotoxicity of 5-FU.

Caffeine, a food ingredient found in coffee and tea, slows the growth of liver cancer cells, including HepG2, HLF, Huh7, and PLC/PRF/5 (Okano et al., 2008). Many studies report a synergistic effect of caffeine and cisplatin in various cancers, such as the human endometrial cancer cell line RL95-2 (Lin et al., 2021). Recently, Wang et al. (2019) reported that the antitumor activity of 5-FU was enhanced by cotreatment of caffeine in HCC HepG3 and SMMMC cells. They discovered that combining 5-FU and caffeine inhibited HCC cell growth and induced apoptosis by increasing ROS production.

Role of other small molecules in ROS production

As described below, reports suggest that other small molecules (Fig. 4) may modulate ROS generation in cancer cells. First, selenocystine is the oxidation product of selenocysteine, which has a diselenide bond connecting two amino acids. It induces apoptosis in human cancer cells such as A375, HepG2, and MCF7 by increasing ROS production (Chen and Wong, 2009). Fan et al. (2013) investigated whether selenocystine cotreatment could increase the cytotoxicity of 5-FU in human melanoma A375 cells. They observed significant selenocystine-induced DNA damage mediated by ROS production and the inactivation of the extracellular-signal-regulated kinase (ERK) and Akt signaling pathways, resulting in anticancer synergism. Furthermore, the induction of ROS-mediated apoptosis in melanoma cells by 3,3′-diselenodipropionic acid, a selenocysteine derivative, is another example of potentially overcoming anticancer drug resistance (Cao et al., 2014).

Figure 4. Other small molecules.

Allicin, a compound in garlic, has drawn considerable attention as an antimicrobial antioxidant (Chan et al., 2013). Zou et al. (2016) tested whether the anticancer activity of 5-FU in human HCC SK-Hep-1 and BEL-7402 cells and in nude mice increased with allicin and 5-FU cotreatment. They discovered that cotreatment with allicin increased ROS production and sensitization of HCC cells to 5-FU. The synergistic effect was reversed by N-acetylcysteine treatment, indicating that the anticancer activity is mediated by ROS generation. Their study also demonstrated that cotreatment with allicin and 5-FU significantly inhibited the growth of HCC xenograft tumors in nude mice; although commonly thought to be an antioxidant, allicin increased ROS generation when combined with combined 5-FU.

3-Bromopyruvate is an inhibitor of hexokinase (Ko et al., 2001), the key enzyme of glycolysis. The researchers reported that 3-bromopyruvate induced the ROS-mediated cell death of hepatoma SNU449 and Hep3B cells (Kim et al., 2008). Upon treatment with 3-bromopyruvate, both cell lines underwent necrosis and apoptosis in an ATP depletion-dependent manner due to increased intracellular ROS and the disruption of mitochondrial function. Furthermore, the combination of 3-bromopyruvate and 5-FU inhibited tumor growth in vivo and in vitro (Chong et al., 2017).

ENDOGENOUS CELLULAR TARGETS TO OVERCOME 5-FU RESISTANCE

Nuclear factor erythroid 2-related factor 2 (Nrf2)

The transcription factor Nrf2 mediates antioxidant response (Moi et al., 1994). Nrf2 exists in the cytoplasm as the Nrf2-Keap1 complex in the absence of oxidative stress. The cellular level of Nrf2 is kept low by continuous degradation via the ubiquitin–proteasome system, which is mediated by Keap1, the Nrf2 key repressor (Zhang, 2006). Several cysteine residues of Keap1 are modified when exposed to oxidative stress, resulting in the dissociation of the Nrf2-Keap1 complex. Nrf2, which is released by Keap1, enters the nucleus and binds to the DNA in the antioxidant response element (ARE) region to regulate the expression of several genes involved in antioxidant function, such as glutamate-cysteine ligase catalytic subunit (Solis et al., 2002), thioredoxin reductase (Soriano et al., 2009), and heme oxygenase-1 (HO-1) (Jarmi and Agarwal, 2009). When expressed, these antioxidants may impart some degree of protection to cells under oxidative stress. Overexpression of Nrf2 in gastric cancer serves as a prognostic marker for 5-FU resistance, lending credence to Nrf2’s prosurvival role (Hu et al., 2013). Similarly, Nrf2 has a role in developing 5-FU resistance in colon cancer HT-29 cells (Akhdar et al., 2009). Kang et al. (2014) discovered hypomethylation of Nrf2 promoter CpG islands in 5-FU resistance colorectal cancer SNU5/5-FUR cells compared with nonresistant cancer cells, indicating that Nrf2 upregulation led to 5-FU resistance.

Besides its antioxidant function, Nrf2 regulates the expression of drug-metabolizing enzymes and drug transporters, resulting in a decrease in 5-FU efficacy (Bai et al., 2016). A team of researchers reported that 2′,4′-dihydroxy-6′methoxy-3′,5′-dimethylchalcone, an inhibitor of Nrf2/ARE pathway, could reverse 5-FU resistance in HCC BEL-7402 cells by inhibiting the 5-FU efflux (Wei et al., 2018).

ROS/mitogen-activated protein kinases pathway

JNK, c-Jun N-terminal kinase, belongs to MAPKs. The function of JNK is related to both cell survival (Wu et al., 2019) and death (Dhanasekaran and Reddy, 2008). Based on the stimuli, JNK signaling can be either prosurvival or pro-apoptotic, and the signaling pathway is not directly linked to the cytotoxic effect of 5-FU. It appears that either activation or inactivation of the proper signaling pathway could place an additional burden on cells treated with 5-FU, potentially increasing 5-FU cytotoxicity.

Compared with differentiated and chemosensitive pancreatic cancer stem cells, the JNK signaling pathway is activated in pancreatic cancer stem cells (Okada et al., 2014; Suzuki et al., 2015). Researchers established that the JNK signaling pathway is activated in the pancreatic cancer stem cells (Suzuki et al., 2015). Pretreatment of cells with SP600125, a JNK inhibitor, resulted in the sensitization of the cells to 5-FU and gemcitabine. The cytotoxic effects of these chemotherapeutics were accompanied by an increase in ROS production. Furthermore, the use of N-acetylcysteine, a free radical scavenger, reduced the intracellular level of ROS and allowed the cells to remain resistant to 5-FU; this is an example of the detrimental use of an antioxidant in chemotherapy. The synergistic cytotoxicity of 5-FU and the compounds mentioned above, tetrathiomolybdate (Kim et al., 2012) and oridonin (Zhang et al., 2019), is associated with the generation of ROS and the activation of JNK. 5-FU cytotoxicity appears to be enhanced by oxidative stress and JNK activation, potentially overcoming 5-FU resistance.

Similarly, coronarin D, a diterpene compound derived from grapes, has anticancer activity. Zingiberaceae (Bailly, 2020) is involved with the activation of JNK signaling and ROS generation, as shown in human nasopharyngeal cancer cells (Chen et al., 2017a). It was recently reported that coronarin D induces the apoptosis of 5-FU resistant human oral cancer cells. The cytotoxicity is related to the JNK signaling pathway (Hsieh et al., 2020).

Besides JNK signaling, activation of p38 MAP kinase is linked to 5-FU cytotoxicity. According to Xie et al. (2016), the overexpression of nicotinamide N-methyltransferase causes a decrease in ROS levels, and the inactivation of p38 signaling is involved in 5-FU resistance in colorectal cancer SW480 cells. Moon et al. (2020) reported that the activation of p38 by yeast extract resulted in the antitumor effect on 5-FU resistant colorectal cancer SNU-C5 cells.

Nevertheless, the role of the ERK pathway in 5-FU sensitivity appears to be prosurvival. Kim et al. (2016) discovered ERK overexpression in SUNC5/FUR cells, resistant to 5-FU. Furthermore, sensitization was achieved by transfecting 5-FU resistant cells with siRNA against ERK. The role of aluminum chloride in inducing 5-FU resistance was further investigated, revealing that ERK activation facilitated the survival of HCC HepG2 cells during 5-FU treatment (Li et al., 2019a). By contrast, U0126, an ERK inhibitor, reversed aluminum chloride-induced 5-FU resistance; it also remains unclear whether ERK activation is required for cells to remain resistant to 5-FU. A Japanese group, for example, reported a 5-FU resistant human squamous carcinoma UM-SCC-23 cell line that activated both ERK and Akt signals (You et al., 2009). Nevertheless, U0126 could not reverse the resistance, whereas Akt inhibition was. Furthermore, Wang et al. (2017b) reported that 5-FU resistance caused by ADAM12 overexpression increased phosphorylated Akt but not phosphorylated ERK in breast cancer SKBR3 MDA-MB-231 cells.

PI3K/Akt pathway

Phosphatidylinositol 3-kinase (PI3K)/Akt pathway is considered one of the key signaling pathways that confers cancer cells’ resistance to chemotherapy (Liu et al., 2020). Researchers reported the constitutive activation of Akt signaling in 5-FU resistant squamous carcinoma UM-SCC-23 cells (You et al., 2009). By contrast, the inhibition of Akt signaling was noticed when the synergistic cytotoxicity of 5-FU was observed by the cotreatment of several compounds, including violacein (Kodach et al., 2006), curcumin (Zhang et al., 2017), and kaempferol (Li et al., 2019b). Recent reports state that celecoxib, a COX-2 inhibitor, induced apoptosis by inhibiting Akt in 5-FU resistant gastric carcinoma AGS cells (Choi et al., 2021). The inhibition of COX-2 is thought to have resulted in the downregulation of Akt and the induction of apoptosis. MK-2206’s direct inhibition of Akt also increases the cytotoxicity of 5-FU in gastric cancer SGC-7901 and MKN45 cells (Jin et al., 2016). Hence, it remains to be confirmed whether MK-2206 could be used to treat 5-FU resistant cells.

Autophagy pathway

Autophagy, a self-eating process involving the autophagosome (Glick et al., 2010), has two states, either prosurvival or prodeath, depending on the mode of activation and which cells are affected. Initially, the prosurvival role of autophagy was reported because inhibition of autophagy was associated with increased cytotoxicity of 5-FU in human colon cancer colon26 and HT29 cells in vitro and in vivo (Li et al., 2010). The prosurvival role of autophagy was again shown in HCT116 p53−/− cells (Sui et al., 2014) and human HCC Bel-7402 cells (Wang et al., 2017a). The involvement of prosurvival autophagy in 5-FU resistance has been demonstrated for several cellular components such as TSPAN9 (Qi et al., 2020) and claudin-1 (Tong et al., 2019). Zhang et al. (2017) reported synergistic cytotoxicity from the combination of curcumin and 5-FU, which included a reduction in prosurvival autophagy mediated by AMPK and Unc-51 Like Autophagy Activating Kinase 1 (ULK1). By contrast, several compounds have been shown to increase the cytotoxicity of 5-FU by inducing prodeath autophagy. β-Elemene, a sesquiterpene compound found in various plants, induces prodeath autophagy in 5-FU resistant colorectal cancer HCT116 p53−/− cells (Zhang et al., 2020). Similarly, when combined with 5-FU, withaferin-A, a natural product with a steroidal lactone structure, induces endoplasmic reticulum stress-mediated autophagy (Alnuqaydan et al., 2020) in CRC cells (SW480, HT29, HCT116).

Aminopeptidase N (CD13)

Aminopeptidase N, also known as CD13, is a cell-surface-anchored zinc peptidase with various functions, including peptide cleavage, endocytosis, and signaling (Mina-Osorio, 2008). It was initially identified as a cell surface marker CD13 for myeloid leukemia cells (Sakai et al., 1987), and the signaling function of aminopeptidase N appears to be independent of enzyme activity. Aminopeptidase expression has been linked to a poor prognosis and angiogenesis in cancer cells such as nonsmall cell lung cancer (Tokuhara et al., 2006), pancreatic carcinoma (Ikeda et al., 2003), and colon cancer (Hashida et al., 2002). When substrates or inhibitors bind to CD13, the conformation of the dimeric CD13 structure changes, resulting in signal transduction (Xu et al., 1997), and CD13 protects cells from apoptosis by reducing ROS-induced DNA damage.

Ubenimex, also known as bestatin, is a dipeptide compound produced by actinomycetes (Umezawa et al., 1976). It specifically blocks and antagonizes CD13 (Look et al., 1989). Haraguchi et al. (2010) reported that ubenimex or a CD13-neutralizing antibody inhibited CD13 in hepatocellular cancer HuH7 cells. They revealed that combining 5-FU and ubenimex increased ROS production and improved liver cancer therapy. Additionally, Dou et al. (2017) investigated the therapeutic potential of BC-02, a conjugation compound of ubenimex and 5-FU, in the successful inhibition of the growth and self-renewal of liver cancer stem cells. Similarly, Sun et al. (2015) reported that 4cc, a synthetic inhibitor of aminopeptidase N, could increase 5-FU cytotoxicity by generating ROS in human liver cancer HCC cells. It remains to be seen whether CD13 inhibitors can be used to treat cancer cells other than HCC.

CONCLUSION

In this study, we have compiled a list of compounds that can be used alone or combined with 5-FU to modulate ROS generation. Although the precise mechanisms underlying their cytotoxicity remain unknown, several endogenous cellular targets have been identified, as described above. These compounds and cellular targets could help develop new strategies for combating 5-FU resistance.

ACKNOWLEDGMENTS

This work was supported by research grants from Daegu Catholic University in 2020.

CONFLICT OF INTEREST

The authors claim no conflicts of interest.

References
  1. Afrin, S., Giampieri, F., Forbes-Hernandez, T. Y., Gasparrini, M., Amici, A., Cianciosi, D., Quiles, J. L. and Battino, M. (2018a) Manuka honey synergistically enhances the chemopreventive effect of 5-fluorouracil on human colon cancer cells by inducing oxidative stress and apoptosis, altering metabolic phenotypes and suppressing metastasis ability. Free Radic. Biol. Med. 126, 41-54.
    Pubmed CrossRef
  2. Afrin, S., Giampieri, F., Gasparrini, M., Forbes-Hernandez, T. Y., Cianciosi, D., Reboredo-Rodriguez, P., Amici, A., Quiles, J. L. and Battino, M. (2018b) The inhibitory effect of Manuka honey on human colon cancer HCT-116 and LoVo cell growth. Part 1: the suppression of cell proliferation, promotion of apoptosis and arrest of the cell cycle. Food Funct. 9, 2145-2157.
    Pubmed CrossRef
  3. Ahmad, B., Khan, S., Nabi, G., Gamallat, Y., Su, P., Jamalat, Y., Duan, P. and Yao, L. (2019) Natural gypenosides: targeting cancer through different molecular pathways. Cancer Manag. Res. 11, 2287-2297.
    Pubmed KoreaMed CrossRef
  4. Ak, T. and Gulcin, I. (2008) Antioxidant and radical scavenging properties of curcumin. Chem. Biol. Interact. 174, 27-37.
    Pubmed CrossRef
  5. Akhdar, H., Loyer, P., Rauch, C., Corlu, A., Guillouzo, A. and Morel, F. (2009) Involvement of Nrf2 activation in resistance to 5-fluorouracil in human colon cancer HT-29 cells. Eur. J. Cancer 45, 2219-2227.
    Pubmed CrossRef
  6. Alnuqaydan, A. M., Rah, B., Almutary, A. G. and Chauhan, S. S. (2020) Synergistic antitumor effect of 5-fluorouracil and withaferin-A induces endoplasmic reticulum stress-mediated autophagy and apoptosis in colorectal cancer cells. Am. J. Cancer Res. 10, 799-815.
    Pubmed KoreaMed
  7. Bai, X., Chen, Y., Hou, X., Huang, M. and Jin, J. (2016) Emerging role of NRF2 in chemoresistance by regulating drug-metabolizing enzymes and efflux transporters. Drug Metab. Rev. 48, 541-567.
    Pubmed CrossRef
  8. Bailly, C. (2020) Anticancer activities and mechanism of action of the labdane diterpene coronarin D. Pathol. Res. Pract. 216, 152946.
    Pubmed CrossRef
  9. Blondy, S., David, V., Verdier, M., Mathonnet, M., Perraud, A. and Christou, N. (2020) 5-Fluorouracil resistance mechanisms in colorectal cancer: from classical pathways to promising processes. Cancer Sci. 111, 3142-3154.
    Pubmed KoreaMed CrossRef
  10. Brewer, G. J., Dick, R. D., Yuzbasiyan-Gurkin, V., Tankanow, R., Young, A. B. and Kluin, K. J. (1991) Initial therapy of patients with Wilson's disease with tetrathiomolybdate. Arch. Neurol. 48, 42-47.
    Pubmed CrossRef
  11. Cai, Y. Z., Mei, S., Jie, X., Luo, Q. and Corke, H. (2006) Structure-radical scavenging activity relationships of phenolic compounds from traditional Chinese medicinal plants. Life Sci. 78, 2872-2888.
    Pubmed CrossRef
  12. Cao, W., Li, X., Zheng, S., Zheng, W., Wong, Y. S. and Chen, T. (2014) Selenocysteine derivative overcomes TRAIL resistance in melanoma cells: evidence for ROS-dependent synergism and signaling crosstalk. Oncotarget 5, 7431-7445.
    Pubmed KoreaMed CrossRef
  13. Carvalho, M., Silva, B. M., Silva, R., Valentao, P., Andrade, P. B. and Bastos, M. L. (2010) First report on Cydonia oblonga Miller anticancer potential: differential antiproliferative effect against human kidney and colon cancer cells. J. Agric. Food Chem. 58, 3366-3370.
    Pubmed CrossRef
  14. Chan, J. Y., Yuen, A. C., Chan, R. Y. and Chan, S. W. (2013) A review of the cardiovascular benefits and antioxidant properties of allicin. Phytother. Res. 27, 637-646.
    Pubmed CrossRef
  15. Chen, J. C., Hsieh, M. C., Lin, S. H., Lin, C. C., Hsi, Y. T., Lo, Y. S., Chuang, Y. C., Hsieh, M. J. and Chen, M. K. (2017a) Coronarin D induces reactive oxygen species-mediated cell death in human nasopharyngeal cancer cells through inhibition of p38 MAPK and activation of JNK. Oncotarget 8, 108006-108019.
    Pubmed KoreaMed CrossRef
  16. Chen, T. and Wong, Y. S. (2009) Selenocystine induces reactive oxygen species-mediated apoptosis in human cancer cells. Biomed. Pharmacother. 63, 105-113.
    Pubmed CrossRef
  17. Chen, X., Chen, X., Zhang, X., Wang, L., Cao, P., Rajamanickam, V., Wu, C., Zhou, H., Cai, Y., Liang, G. and Wang, Y. (2019) Curcuminoid B63 induces ROS-mediated paraptosis-like cell death by targeting TrxR1 in gastric cells. Redox. Biol. 21, 101061.
    Pubmed KoreaMed CrossRef
  18. Chen, X., Yang, L., Oppenheim, J. J. and Howard, M. Z. (2002) Cellular pharmacology studies of shikonin derivatives. Phytother. Res. 16, 199-209.
    Pubmed CrossRef
  19. Chen, X. X., Lam, K. H., Chen, Q. X., Leung, G. P., Tang, S. C. W., Sze, S. C., Xiao, J. B., Feng, F., Wang, Y., Zhang, K. Y. and Zhang, Z. J. (2017b) Ficus virens proanthocyanidins induced apoptosis in breast cancer cells concomitantly ameliorated 5-fluorouracil induced intestinal mucositis in rats. Food Chem. Toxicol. 110, 49-61.
    Pubmed CrossRef
  20. Chen, X. X., Leung, G. P., Zhang, Z. J., Xiao, J. B., Lao, L. X., Feng, F., Mak, J. C., Wang, Y., Sze, S. C. and Zhang, K. Y. (2017c) Proanthocyanidins from Uncaria rhynchophylla induced apoptosis in MDA-MB-231 breast cancer cells while enhancing cytotoxic effects of 5-fluorouracil. Food Chem. Toxicol. 107, 248-260.
    Pubmed CrossRef
  21. Choi, S. M., Cho, Y. S., Park, G., Lee, S. K. and Chun, K. S. (2021) Celecoxib induces apoptosis through Akt inhibition in 5-fluorouracil-resistant gastric cancer cells. Toxicol. Res. 37, 25-33.
    Pubmed KoreaMed CrossRef
  22. Chong, D., Ma, L., Liu, F., Zhang, Z., Zhao, S., Huo, Q., Zhang, P., Zheng, H. and Liu, H. (2017) Synergistic antitumor effect of 3-bromopyruvate and 5-fluorouracil against human colorectal cancer through cell cycle arrest and induction of apoptosis. Anticancer Drugs 28, 831-840.
    Pubmed CrossRef
  23. Das, R., Bhattacharya, K., Sarkar, S., Samanta, S. K., Pal, B. C. and Mandal, C. (2014) Mahanine synergistically enhances cytotoxicity of 5-fluorouracil through ROS-mediated activation of PTEN and p53/p73 in colon carcinoma. Apoptosis 19, 149-164.
    Pubmed CrossRef
  24. Dhanasekaran, D. N. and Reddy, E. P. (2008) JNK signaling in apoptosis. Oncogene 27, 6245-6251.
    Pubmed KoreaMed CrossRef
  25. Dou, C., Fang, C., Zhao, Y., Fu, X., Zhang, Y., Zhu, D., Wu, H., Liu, H., Zhang, J., Xu, W., Liu, Z., Wang, H., Li, D. and Wang, X. (2017) BC-02 eradicates liver cancer stem cells by upregulating the ROS-dependent DNA damage. Int. J. Oncol. 51, 1775-1784.
    Pubmed CrossRef
  26. Fan, C., Chen, J., Wang, Y., Wong, Y. S., Zhang, Y., Zheng, W., Cao, W. and Chen, T. (2013) Selenocystine potentiates cancer cell apoptosis induced by 5-fluorouracil by triggering reactive oxygen species-mediated DNA damage and inactivation of the ERK pathway. Free Radic. Biol. Med. 65, 305-316.
    Pubmed CrossRef
  27. Fatfat, M., Merhi, R. A., Rahal, O., Stoyanovsky, D. A., Zaki, A., Haidar, H., Kagan, V. E., Gali-Muhtasib, H. and Machaca, K. (2014) Copper chelation selectively kills colon cancer cells through redox cycling and generation of reactive oxygen species. BMC Cancer 14, 527.
    Pubmed KoreaMed CrossRef
  28. Frei, E., Elias, A., Wheeler, C., Richardson, P. and Hryniuk, W. (1998) The relationship between high-dose treatment and combination chemotherapy: the concept of summation dose intensity. Clin. Cancer Res. 4, 2027-2037.
    Pubmed
  29. Glick, D., Barth, S. and Macleod, K. F. (2010) Autophagy: cellular and molecular mechanisms. J. Pathol. 221, 3-12.
    Pubmed KoreaMed CrossRef
  30. Gorrini, C., Harris, I. S. and Mak, T. W. (2013) Modulation of oxidative stress as an anticancer strategy. Nat. Rev. Drug Discov. 12, 931-947.
    Pubmed CrossRef
  31. Gupte, A. and Mumper, R. J. (2009) Elevated copper and oxidative stress in cancer cells as a target for cancer treatment. Cancer Treat. Rev. 35, 32-46.
    Pubmed CrossRef
  32. Haraguchi, N., Ishii, H., Mimori, K., Tanaka, F., Ohkuma, M., Kim, H. M., Akita, H., Takiuchi, D., Hatano, H., Nagano, H., Barnard, G. F., Doki, Y. and Mori, M. (2010) CD13 is a therapeutic target in human liver cancer stem cells. J. Clin. Invest. 120, 3326-3339.
    Pubmed KoreaMed CrossRef
  33. Hashida, H., Takabayashi, A., Kanai, M., Adachi, M., Kondo, K., Kohno, N., Yamaoka, Y. and Miyake, M. (2002) Aminopeptidase N is involved in cell motility and angiogenesis: its clinical significance in human colon cancer. Gastroenterology 122, 376-386.
    Pubmed CrossRef
  34. He, C., Rong, R., Liu, J., Wan, J., Zhou, K. and Kang, J. X. (2012) Effects of Coptis extract combined with chemotherapeutic agents on ROS production, multidrug resistance, and cell growth in A549 human lung cancer cells. Chin. Med. 7, 11.
    Pubmed KoreaMed CrossRef
  35. Hsieh, M. Y., Hsieh, M. J., Lo, Y. S., Lin, C. C., Chuang, Y. C., Chen, M. K. and Chou, M. C. (2020) Modulating effect of Coronarin D in 5-fluorouracil resistance human oral cancer cell lines induced apoptosis and cell cycle arrest through JNK1/2 signaling pathway. Biomed. Pharmacother. 128, 110318.
    Pubmed CrossRef
  36. Hu, X. F., Yao, J., Gao, S. G., Wang, X. S., Peng, X. Q., Yang, Y. T. and Feng, X. S. (2013) Nrf2 overexpression predicts prognosis and 5-FU resistance in gastric cancer. Asian Pac. J. Cancer Prev. 14, 5231-5235.
    Pubmed CrossRef
  37. Hu, X. Y., Liang, J. Y., Guo, X. J., Liu, L. and Guo, Y. B. (2015) 5-Fluorouracil combined with apigenin enhances anticancer activity through mitochondrial membrane potential (DeltaPsim)-mediated apoptosis in hepatocellular carcinoma. Clin. Exp. Pharmacol. Physiol. 42, 146-153.
    Pubmed CrossRef
  38. Huang, Q., Lu, G., Shen, H. M., Chung, M. C. and Ong, C. N. (2007) Anti-cancer properties of anthraquinones from rhubarb. Med. Res. Rev. 27, 609-630.
    Pubmed CrossRef
  39. Hwang, I. T., Chung, Y. M., Kim, J. J., Chung, J. S., Kim, B. S., Kim, H. J., Kim, J. S. and Yoo, Y. D. (2007) Drug resistance to 5-FU linked to reactive oxygen species modulator 1. Biochem. Biophys. Res. Commun. 359, 304-310.
    Pubmed CrossRef
  40. Ikeda, N., Nakajima, Y., Tokuhara, T., Hattori, N., Sho, M., Kanehiro, H. and Miyake, M. (2003) Clinical significance of aminopeptidase N/CD13 expression in human pancreatic carcinoma. Clin. Cancer Res. 9, 1503-1508.
    Pubmed
  41. Jarmi, T. and Agarwal, A. (2009) Heme oxygenase and renal disease. Curr. Hypertens. Rep. 11, 56-62.
    Pubmed CrossRef
  42. Jin, P., Wong, C. C., Mei, S., He, X., Qian, Y. and Sun, L. (2016) MK-2206 co-treatment with 5-fluorouracil or doxorubicin enhances chemosensitivity and apoptosis in gastric cancer by attenuation of Akt phosphorylation. OncoTargets Ther. 9, 4387-4396.
    Pubmed KoreaMed CrossRef
  43. Juhasz, A., Markel, S., Gaur, S., Liu, H., Lu, J., Jiang, G., Wu, X., Antony, S., Wu, Y., Melillo, G., Meitzler, J. L., Haines, D. C., Butcher, D., Roy, K. and Doroshow, J. H. (2017) NADPH oxidase 1 supports proliferation of colon cancer cells by modulating reactive oxygen species-dependent signal transduction. J. Biol. Chem. 292, 7866-7887.
    Pubmed KoreaMed CrossRef
  44. Kang, K. A., Piao, M. J., Kim, K. C., Kang, H. K., Chang, W. Y., Park, I. C., Keum, Y. S., Surh, Y. J. and Hyun, J. W. (2014) Epigenetic modification of Nrf2 in 5-fluorouracil-resistant colon cancer cells: involvement of TET-dependent DNA demethylation. Cell Death Dis. 5, e1183.
    Pubmed KoreaMed CrossRef
  45. Kim, J. K., Kang, K. A., Piao, M. J., Ryu, Y. S., Han, X., Fernando, P. M., Oh, M. C., Park, J. E., Shilnikova, K., Boo, S. J., Na, S. Y., Jeong, Y. J., Jeong, S. U. and Hyun, J. W. (2016) Endoplasmic reticulum stress induces 5-fluorouracil resistance in human colon cancer cells. Environ. Toxicol. Pharmacol. 44, 128-133.
    Pubmed CrossRef
  46. Kim, J. S., Ahn, K. J., Kim, J. A., Kim, H. M., Lee, J. D., Lee, J. M., Kim, S. J. and Park, J. H. (2008) Role of reactive oxygen species-mediated mitochondrial dysregulation in 3-bromopyruvate induced cell death in hepatoma cells : ROS-mediated cell death by 3-BrPA. J. Bioenerg. Biomembr. 40, 607-618.
    Pubmed CrossRef
  47. Kim, K. K., Kawar, N. M., Singh, R. K., Lange, T. S., Brard, L. and Moore, R. G. (2011) Tetrathiomolybdate induces doxorubicin sensitivity in resistant tumor cell lines. Gynecol. Oncol. 122, 183-189.
    Pubmed CrossRef
  48. Kim, K. K., Lange, T. S., Singh, R. K., Brard, L. and Moore, R. G. (2012) Tetrathiomolybdate sensitizes ovarian cancer cells to anticancer drugs doxorubicin, fenretinide, 5-fluorouracil and mitomycin C. BMC Cancer 12, 147.
    Pubmed KoreaMed CrossRef
  49. Ko, Y. H., Pedersen, P. L. and Geschwind, J. F. (2001) Glucose catabolism in the rabbit VX2 tumor model for liver cancer: characterization and targeting hexokinase. Cancer Lett. 173, 83-91.
    Pubmed CrossRef
  50. Kodach, L. L., Bos, C. L., Duran, N., Peppelenbosch, M. P., Ferreira, C. V. and Hardwick, J. C. (2006) Violacein synergistically increases 5-fluorouracil cytotoxicity, induces apoptosis and inhibits Akt-mediated signal transduction in human colorectal cancer cells. Carcinogenesis 27, 508-516.
    Pubmed CrossRef
  51. Kong, L., Wang, X., Zhang, K., Yuan, W., Yang, Q., Fan, J., Wang, P. and Liu, Q. (2015) Gypenosides synergistically enhances the anti-tumor effect of 5-fluorouracil on colorectal cancer in vitro and in vivo: a role for oxidative stress-mediated DNA damage and p53 activation. PLoS ONE 10, e0137888.
    Pubmed KoreaMed CrossRef
  52. Kumar, B., Koul, S., Khandrika, L., Meacham, R. B. and Koul, H. K. (2008) Oxidative stress is inherent in prostate cancer cells and is required for aggressive phenotype. Cancer Res. 68, 1777-1785.
    Pubmed CrossRef
  53. Li, C. Y., Wang, E. Q., Cheng, Y. and Bao, J. K. (2011) Oridonin: an active diterpenoid targeting cell cycle arrest, apoptotic and autophagic pathways for cancer therapeutics. Int. J. Biochem. Cell Biol. 43, 701-704.
    Pubmed CrossRef
  54. Li, J., Hou, N., Faried, A., Tsutsumi, S. and Kuwano, H. (2010) Inhibition of autophagy augments 5-fluorouracil chemotherapy in human colon cancer in vitro and in vivo model. Eur. J. Cancer 46, 1900-1909.
    Pubmed CrossRef
  55. Li, M., Cui, Z. G., Zakki, S. A., Feng, Q., Sun, L., Feril, L. B. Jr and Inadera, H. (2019a) Aluminum chloride causes 5-fluorouracil resistance in hepatocellular carcinoma HepG2 cells. J. Cell. Physiol. 234, 20249-20265.
    Pubmed CrossRef
  56. Li, Q., Wei, L., Lin, S., Chen, Y., Lin, J. and Peng, J. (2019b) Synergistic effect of kaempferol and 5fluorouracil on the growth of colorectal cancer cells by regulating the PI3K/Akt signaling pathway. Mol. Med. Rep. 20, 728-734.
    CrossRef
  57. Liang, W., Cai, A., Chen, G., Xi, H., Wu, X., Cui, J., Zhang, K., Zhao, X., Yu, J., Wei, B. and Chen, L. (2016) Shikonin induces mitochondria-mediated apoptosis and enhances chemotherapeutic sensitivity of gastric cancer through reactive oxygen species. Sci. Rep. 6, 38267.
    Pubmed KoreaMed CrossRef
  58. Lin, C. K., Liu, S. T., Wu, Z. S., Wang, Y. C. and Huang, S. M. (2021) Mechanisms of cisplatin in combination with repurposed drugs against human endometrial carcinoma cells. Life (Basel) 11, 160.
    Pubmed KoreaMed CrossRef
  59. Liou, G. Y. and Storz, P. (2010) Reactive oxygen species in cancer. Free Radic. Res. 44, 479-496.
    Pubmed KoreaMed CrossRef
  60. Liu, M. P., Liao, M., Dai, C., Chen, J. F., Yang, C. J., Liu, M., Chen, Z. G. and Yao, M. C. (2016a) Sanguisorba officinalis L synergistically enhanced 5-fluorouracil cytotoxicity in colorectal cancer cells by promoting a reactive oxygen species-mediated, mitochondria-caspase-dependent apoptotic pathway. Sci. Rep. 6, 34245.
    Pubmed KoreaMed CrossRef
  61. Liu, R., Chen, Y., Liu, G., Li, C., Song, Y., Cao, Z., Li, W., Hu, J., Lu, C. and Liu, Y. (2020) PI3K/AKT pathway as a key link modulates the multidrug resistance of cancers. Cell Death Dis. 11, 797.
    Pubmed KoreaMed CrossRef
  62. Liu, Y., Li, Q., Zhou, L., Xie, N., Nice, E. C., Zhang, H., Huang, C. and Lei, Y. (2016b) Cancer drug resistance: redox resetting renders a way. Oncotarget 7, 42740-42761.
    Pubmed KoreaMed CrossRef
  63. Longley, D. B., Harkin, D. P. and Johnston, P. G. (2003) 5-Fluorouracil: mechanisms of action and clinical strategies. Nat. Rev. Cancer 3, 330-338.
    Pubmed CrossRef
  64. Look, A. T., Ashmun, R. A., Shapiro, L. H. and Peiper, S. C. (1989) Human myeloid plasma membrane glycoprotein CD13 (gp150) is identical to aminopeptidase N. J. Clin. Invest. 83, 1299-1307.
    Pubmed KoreaMed CrossRef
  65. Mates, J. M. and Sanchez-Jimenez, F. M. (2000) Role of reactive oxygen species in apoptosis: implications for cancer therapy. Int. J. Biochem. Cell Biol. 32, 157-170.
    Pubmed CrossRef
  66. Mehrzad, V., Roayaei, M., Peikar, M. S., Nouranian, E., Mokarian, F., Khani, M. and Farzannia, S. (2016) Bevacizumab plus FOLFOX or FOLFIRI regimens on patients with unresectable liver-only metastases of metastatic colorectal cancer. Adv. Biomed. Res. 5, 10.
  67. Mina-Osorio, P. (2008) The moonlighting enzyme CD13: old and new functions to target. Trends Mol. Med. 14, 361-371.
    Pubmed KoreaMed CrossRef
  68. Moi, P., Chan, K., Asunis, I., Cao, A. and Kan, Y. W. (1994) Isolation of NF-E2-related factor 2 (Nrf2), a NF-E2-like basic leucine zipper transcriptional activator that binds to the tandem NF-E2/AP1 repeat of the beta-globin locus control region. Proc. Natl. Acad. Sci. U.S.A. 91, 9926-9930.
    Pubmed KoreaMed CrossRef
  69. Moon, D., Kang, H. K., Kim, J. and Yoon, S. P. (2020) Yeast extract induces apoptosis and cell cycle arrest via activating p38 signal pathway in colorectal cancer cells. Ann. Clin. Lab. Sci. 50, 31-44.
    Pubmed
  70. Nishikawa, M. (2008) Reactive oxygen species in tumor metastasis. Cancer Lett. 266, 53-59.
    Pubmed CrossRef
  71. Okada, M., Shibuya, K., Sato, A., Seino, S., Suzuki, S., Seino, M. and Kitanaka, C. (2014) Targeting the K-Ras--JNK axis eliminates cancer stem-like cells and prevents pancreatic tumor formation. Oncotarget 5, 5100-5112.
    Pubmed KoreaMed CrossRef
  72. Okano, J., Nagahara, T., Matsumoto, K. and Murawaki, Y. (2008) Caffeine inhibits the proliferation of liver cancer cells and activates the MEK/ERK/EGFR signalling pathway. Basic Clin. Pharmacol. Toxicol. 102, 543-551.
    Pubmed CrossRef
  73. Pazdur, R., Hoff, P. M., Medgyesy, D., Royce, M. and Brito, R. (1998) The oral fluorouracil prodrugs. Oncology (Williston Park) 12, 48-51.
    Pubmed
  74. Qi, Y., Qi, W., Liu, S., Sun, L., Ding, A., Yu, G., Li, H., Wang, Y., Qiu, W. and Lv, J. (2020) TSPAN9 suppresses the chemosensitivity of gastric cancer to 5-fluorouracil by promoting autophagy. Cancer Cell Int. 20, 4.
    Pubmed KoreaMed CrossRef
  75. Ramsewak, R. S., Nair, M. G., Strasburg, G. M., DeWitt, D. L. and Nitiss, J. L. (1999) Biologically active carbazole alkaloids from Murraya koenigii. J. Agric. Food Chem. 47, 444-447.
    Pubmed CrossRef
  76. Riahi-Chebbi, I., Haoues, M., Essafi, M., Zakraoui, O., Fattouch, S., Karoui, H. and Essafi-Benkhadir, K. (2015) Quince peel polyphenolic extract blocks human colon adenocarcinoma LS174 cell growth and potentiates 5-fluorouracil efficacy. Cancer Cell Int. 16, 1.
    Pubmed KoreaMed CrossRef
  77. Riahi-Chebbi, I., Souid, S., Othman, H., Haoues, M., Karoui, H., Morel, A., Srairi-Abid, N., Essafi, M. and Essafi-Benkhadir, K. (2019) The Phenolic compound Kaempferol overcomes 5-fluorouracil resistance in human resistant LS174 colon cancer cells. Sci. Rep. 9, 195.
    Pubmed KoreaMed CrossRef
  78. Sakai, K., Hattori, T., Sagawa, K., Yokoyama, M. and Takatsuki, K. (1987) Biochemical and functional characterization of MCS-2 antigen (CD13) on myeloid leukemic cells and polymorphonuclear leukocytes. Cancer Res. 47, 5572-5576.
    Pubmed
  79. Shukla, S. and Gupta, S. (2010) Apigenin: a promising molecule for cancer prevention. Pharm. Res. 27, 962-978.
    Pubmed KoreaMed CrossRef
  80. Solis, W. A., Dalton, T. P., Dieter, M. Z., Freshwater, S., Harrer, J. M., He, L., Shertzer, H. G. and Nebert, D. W. (2002) Glutamate-cysteine ligase modifier subunit: mouse Gclm gene structure and regulation by agents that cause oxidative stress. Biochem. Pharmacol. 63, 1739-1754.
    Pubmed CrossRef
  81. Soriano, F. X., Baxter, P., Murray, L. M., Sporn, M. B., Gillingwater, T. H. and Hardingham, G. E. (2009) Transcriptional regulation of the AP-1 and Nrf2 target gene sulfiredoxin. Mol. Cells 27, 279-282.
    Pubmed KoreaMed CrossRef
  82. Souglakos, J., Androulakis, N., Syrigos, K., Polyzos, A., Ziras, N., Athanasiadis, A., Kakolyris, S., Tsousis, S., Kouroussis, C., Vamvakas, L., Kalykaki, A., Samonis, G., Mavroudis, D. and Georgoulias, V. (2006) FOLFOXIRI (folinic acid, 5-fluorouracil, oxaliplatin and irinotecan) vs FOLFIRI (folinic acid, 5-fluorouracil and irinotecan) as first-line treatment in metastatic colorectal cancer (MCC): a multicentre randomised phase III trial from the hellenic oncology research group (HORG). Br.. J. Cancer 94, 798-805.
    Pubmed KoreaMed CrossRef
  83. Sui, X., Kong, N., Wang, X., Fang, Y., Hu, X., Xu, Y., Chen, W., Wang, K., Li, D., Jin, W., Lou, F., Zheng, Y., Hu, H., Gong, L., Zhou, X., Pan, H. and Han, W. (2014) JNK confers 5-fluorouracil resistance in p53-deficient and mutant p53-expressing colon cancer cells by inducing survival autophagy. Sci. Rep. 4, 4694.
    Pubmed KoreaMed CrossRef
  84. Sun, Z. P., Zhang, J., Shi, L. H., Zhang, X. R., Duan, Y., Xu, W. F., Dai, G. and Wang, X. J. (2015) Aminopeptidase N inhibitor 4cc synergizes antitumor effects of 5-fluorouracil on human liver cancer cells through ROS-dependent CD13 inhibition. Biomed. Pharmacother. 76, 65-72.
    Pubmed CrossRef
  85. Suzuki, S., Okada, M., Shibuya, K., Seino, M., Sato, A., Takeda, H., Seino, S., Yoshioka, T. and Kitanaka, C. (2015) JNK suppression of chemotherapeutic agents-induced ROS confers chemoresistance on pancreatic cancer stem cells. Oncotarget 6, 458-470.
    Pubmed KoreaMed CrossRef
  86. Tang, J., Feng, Y., Tsao, S., Wang, N., Curtain, R. and Wang, Y. (2009) Berberine and Coptidis rhizoma as novel antineoplastic agents: a review of traditional use and biomedical investigations. J. Ethnopharmacol. 126, 5-17.
    Pubmed CrossRef
  87. Tokuhara, T., Hattori, N., Ishida, H., Hirai, T., Higashiyama, M., Kodama, K. and Miyake, M. (2006) Clinical significance of aminopeptidase N in non-small cell lung cancer. Clin. Cancer Res. 12, 3971-3978.
    Pubmed CrossRef
  88. Tong, H., Li, T., Qiu, W. and Zhu, Z. (2019) Claudin-1 silencing increases sensitivity of liver cancer HepG2 cells to 5-fluorouracil by inhibiting autophagy. Oncol. Lett. 18, 5709-5716.
    Pubmed KoreaMed CrossRef
  89. Torres, M. and Forman, H. J. (2003) Redox signaling and the MAP kinase pathways. BioFactors 17, 287-296.
    Pubmed CrossRef
  90. Umezawa, H., Aoyagi, T., Suda, H., Hamada, M. and Takeuchi, T. (1976) Bestatin, an inhibitor of aminopeptidase B, produced by actinomycetes. J. Antibiot. 29, 97-99.
    Pubmed CrossRef
  91. Ushio-Fukai, M. and Nakamura, Y. (2008) Reactive oxygen species and angiogenesis: NADPH oxidase as target for cancer therapy. Cancer Lett. 266, 37-52.
    Pubmed KoreaMed CrossRef
  92. Wang, M., Huang, C., Su, Y., Yang, C., Xia, Q. and Xu, D. J. (2017a) Astragaloside II sensitizes human hepatocellular carcinoma cells to 5-fluorouracil via suppression of autophagy. J. Pharm. Pharmacol. 69, 743-752.
    Pubmed CrossRef
  93. Wang, X., Wang, Y., Gu, J., Zhou, D., He, Z., Wang, X. and Ferrone, S. (2017b) ADAM12-L confers acquired 5-fluorouracil resistance in breast cancer cells. Sci. Rep. 7, 9687.
    Pubmed KoreaMed CrossRef
  94. Wang, Z., Gu, C., Wang, X., Lang, Y., Wu, Y., Wu, X., Zhu, X., Wang, K. and Yang, H. (2019) Caffeine enhances the anti-tumor effect of 5-fluorouracil via increasing the production of reactive oxygen species in hepatocellular carcinoma. Med. Oncol. 36, 97.
    Pubmed CrossRef
  95. Wei, H. (1992) Activation of oncogenes and/or inactivation of anti-oncogenes by reactive oxygen species. Med. Hypotheses 39, 267-270.
    Pubmed CrossRef
  96. Wei, X., Mo, X., An, F., Ji, X. and Lu, Y. (2018) 2',4'-Dihydroxy-6'-methoxy-3',5'-dimethylchalcone, a potent Nrf2/ARE pathway inhibitor, reverses drug resistance by decreasing glutathione synthesis and drug efflux in BEL-7402/5-FU cells. Food Chem. Toxicol. 119, 252-259.
    Pubmed CrossRef
  97. Wu, Q., Wu, W., Fu, B., Shi, L., Wang, X. and Kuca, K. (2019) JNK signaling in cancer cell survival. Med. Res. Rev. 39, 2082-2104.
    Pubmed CrossRef
  98. Xie, X., Liu, H., Wang, Y., Zhou, Y., Yu, H., Li, G., Ruan, Z., Li, F., Wang, X. and Zhang, J. (2016) Nicotinamide N-methyltransferase enhances resistance to 5-fluorouracil in colorectal cancer cells through inhibition of the ASK1-p38 MAPK pathway. Oncotarget 7, 45837-45848.
    Pubmed KoreaMed CrossRef
  99. Xu, Y., Wellner, D. and Scheinberg, D. A. (1997) Cryptic and regulatory epitopes in CD13/aminopeptidase N. Exp. Hematol. 25, 521-529.
    Pubmed
  100. Yan, J., Dou, X., Zhou, J., Xiong, Y., Mo, L., Li, L. and Lei, Y. (2019) Tubeimoside-I sensitizes colorectal cancer cells to chemotherapy by inducing ROS-mediated impaired autophagolysosomes accumulation. J. Exp. Clin. Cancer Res. 38, 353.
    Pubmed KoreaMed CrossRef
  101. You, F., Aoki, K., Ito, Y. and Nakashima, S. (2009) AKT plays a pivotal role in the acquisition of resistance to 5-fluorouracil in human squamous carcinoma cells. Mol. Med. Rep. 2, 609-613.
    Pubmed CrossRef
  102. Yu, L., Ma, R., Wang, Y. and Nishino, H. (1994) Potent anti-tumor activity and low toxicity of tubeimoside 1 isolated from Bolbostemma paniculatum. Planta Med. 60, 204-208.
    Pubmed CrossRef
  103. Zhang, D., Zhou, Q., Huang, D., He, L., Zhang, H., Hu, B., Peng, H. and Ren, D. (2019) ROS/JNK/c-Jun axis is involved in oridonin-induced caspase-dependent apoptosis in human colorectal cancer cells. Biochem. Biophys. Res. Commun. 513, 594-601.
    Pubmed CrossRef
  104. Zhang, D. D. (2006) Mechanistic studies of the Nrf2-Keap1 signaling pathway. Drug Metab. Rev. 38, 769-789.
    Pubmed CrossRef
  105. Zhang, P., Lai, Z. L., Chen, H. F., Zhang, M., Wang, A., Jia, T., Sun, W. Q., Zhu, X. M., Chen, X. F., Zhao, Z. and Zhang, J. (2017) Curcumin synergizes with 5-fluorouracil by impairing AMPK/ULK1-dependent autophagy, AKT activity and enhancing apoptosis in colon cancer cells with tumor growth inhibition in xenograft mice. J. Exp. Clin. Cancer Res. 36, 190.
    Pubmed KoreaMed CrossRef
  106. Zhang, R., Pan, T., Xiang, Y., Zhang, M., Feng, J., Liu, S., Duan, T., Chen, P., Zhai, B., Chen, X., Wang, W., Chen, B., Han, X., Chen, L., Yan, L., Jin, T., Liu, Y., Li, G., Huang, X., Zhang, W., Sun, Y., Li, Q., Zhang, Q., Zhuo, L., Xie, T., Wu, Q. and Sui, X. (2020) beta-Elemene reverses the resistance of p53-deficient colorectal cancer cells to 5-fluorouracil by inducing pro-death autophagy and cyclin D3-dependent cycle arrest. Front. Bioeng. Biotechnol. 8, 378.
    Pubmed KoreaMed CrossRef
  107. Zhao, H., Liu, Q., Wang, S., Dai, F., Cheng, X., Cheng, X., Chen, W., Zhang, M. and Chen, D. (2017) In vitro additive antitumor effects of dimethoxycurcumin and 5-fluorouracil in colon cancer cells. Cancer Med. 6, 1698-1706.
    Pubmed KoreaMed CrossRef
  108. Zheng, W., Zhou, C. Y., Zhu, X. Q., Wang, X. J., Li, Z. Y., Chen, X. C., Chen, F., Che, X. Y. and Xie, X. (2018) Oridonin enhances the cytotoxicity of 5-FU in renal carcinoma cells by inducting necroptotic death. Biomed. Pharmacother. 106, 175-182.
    Pubmed CrossRef
  109. Zou, X., Liang, J., Sun, J., Hu, X., Lei, L., Wu, D. and Liu, L. (2016) Allicin sensitizes hepatocellular cancer cells to anti-tumor activity of 5-fluorouracil through ROS-mediated mitochondrial pathway. J. Pharmacol. Sci. 131, 233-240.
    Pubmed CrossRef
  110. Zu, C., Qin, G., Yang, C., Liu, N., He, A., Zhang, M. and Zheng, X. (2018) Low dose Emodin induces tumor senescence for boosting breast cancer chemotherapy via silencing NRARP. Biochem. Biophys. Res. Commun. 505, 973-978.
    Pubmed CrossRef


This Article


Cited By Articles

Funding Information

Services
Social Network Service

e-submission

Archives