Next Article in Journal
Pigeon Pea Husk for Removal of Emerging Contaminants Trimethoprim and Atenolol from Water
Previous Article in Journal
Application of Propolis in Protecting Skeletal and Periodontal Health—A Systematic Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Roles of Nrf2 in Gastric Cancer: Targeting for Therapeutic Strategies

by
Tahereh Farkhondeh
1,2,
Ali Mohammad Pourbagher-Shahri
3,
Mohsen Azimi-Nezhad
4,
Fatemeh Forouzanfar
5,
Aranka Brockmueller
6,
Milad Ashrafizadeh
7,8,
Marjan Talebi
9,
Mehdi Shakibaei
6,* and
Saeed Samarghandian
4,*
1
Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand 9717853577, Iran
2
Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand 9717853577, Iran
3
Faculty of Medicine, Birjand University of Medical Sciences, Birjand 9717853577, Iran
4
Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur 9318614139, Iran
5
Medical Toxicology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran
6
Musculoskeletal Research Group and Tumor Biology, Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University Munich, Pettenkoferstrasse 11, D-80336 Munich, Germany
7
Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla, Istanbul 34956, Turkey
8
Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, Istanbul 34956, Turkey
9
Department of Pharmacognosy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran 1996835113, Iran
*
Authors to whom correspondence should be addressed.
Molecules 2021, 26(11), 3157; https://doi.org/10.3390/molecules26113157
Submission received: 9 April 2021 / Revised: 15 May 2021 / Accepted: 19 May 2021 / Published: 25 May 2021

Abstract

:
Nuclear Factor Erythroid 2-Related Factor 2 (Nrf2) is a specific transcription factor with potent effects on the regulation of antioxidant gene expression that modulates cell hemostasis under various conditions in tissues. However, the effects of Nrf2 on gastric cancer (GC) are not fully elucidated and understood. Evidence suggests that uncontrolled Nrf2 expression and activation has been observed more frequently in malignant tumors, including GC cells, which is then associated with increased antioxidant capacity, chemoresistance, and poor clinical prognosis. Moreover, Nrf2 inhibitors and the associated modulation of tumor cell redox balance have shown that Nrf2 also has beneficial effects on the therapy of various cancers, including GC. Based on previous findings on the important role of Nrf2 in GC therapy, it is of great interest to scientists in basic and clinical tumor research that Nrf2 can be active as both an oncogene and a tumor suppressor depending on different background situations.

1. Introduction

Gastric cancer (GC) is one of the most common malignant tumor diseases with a very high mortality rate globally [1,2,3]. Symptoms of this disease do not become apparent until the advanced stage of disease. In recent decades, a continuous decrease in the incidence rates of GC has been found worldwide [4,5,6,7], as the decrease in GC rate may be related to the improvement of hygiene and food standards and the eradication of Helicobacter pylori (H. pylori) [8]. Genetic background, environmental pollutants, cigarette smoking, alcohol consumption, socioeconomic status, and H. pylori infection are known to be the main risk factors for GC [9,10,11]. Other risk factors for GC include Epstein–Barr virus (EBV), pernicious anemia, blood type A, obesity, and gastrectomy [12,13,14,15,16]. To manage the treatment of GC, classifications such as early and advanced stages have been established, and anatomically, this cancer is classified into three types: distal esophagus, cardia, and stomach distal to cardia.
The tumorigenesis and dissemination of malignant cells is modulated by the exposure of cells to detrimental or protective elements. The interaction between these factors is found at the cellular level via the strict modulation of signaling pathways for which their activation or suppression can either maintain or impair the malignant transformation of normal cells and the increased aggressiveness of cancer cells. Kelch-like ECH-associated protein 1(Keap1)–nuclear factor erythroid 2-related factor 2 (Nrf2) is the most studied signaling pathway of cellular defense against oxidative stress [12].
Nrf2 is a transcription factor that regulates the expression of genes that have antioxidant response element-like sequences in their promoter to antioxidant and reactive oxygen species (ROS) detoxification [17] such as heme oxygenase-1 (HO1), peroxiredoxins, and glutamate-cysteine ligase (GCL) [18,19,20,21,22,23]. Nrf2 protects normal cells against DNA damage induced by ROS and malignant cells against chemotherapy [24,25]. Nrf2 also stimulates several oncogenes unrelated to antioxidant activity, such as matrix metallopeptidase 9 (MMP-9), tumor necrosis factor α (TNF-α), and vascular endothelial growth factor A (VEGF-A) [23].
Several factors can modulate Nrf2 in GC cells [26,27,28,29]. Increased production of ROS disassociates the Nrf2 from Keap1, resulting in the translocation of Nrf2 to the nucleus. In addition, P62, a regulator protein, interacts with Keap1 to mediate its degradation, leading to an increase in the concentration of Nrf2 in the nucleus [30,31,32]. It has been further reported that P21 can interact with Nrf2, thereby stabilizing the Nrf2 protein [33]. Within the nucleus, the small V-Maf avian musculoaponeurotic fibrosarcoma oncogenic MAF proteins (sMAF) bind with Nrf2 to form an Nrf2/sMaf heterodimer [18]. Nrf2/sMaf heterodimers regulate the expression of target genes through binding antioxidant response elements (AREs) or MAF recognition elements (MARE) [34,35].
It has been reported that mutations of Nrf2 or its regulators may lead to the suppression or over-stimulation of Nrf2 signaling [36,37]. Nowadays, numerous scientific reports indicate the efficacy of targeting Nrf2 in various diseases [38]. In addition, there is evidence of the beneficial effect of Nrf2 activity and the suppressive role of Nrf2 inhibitors against tumor progression [39,40,41] in human cancers [42,43,44]. However, contradictory activity of Nrf2 in tumor progression has been found in recent years [39,40,41,42,45,46]. In a cancer cell, the overactivation of antioxidant defenses, in particular, Nrf2-dependent genes following ROS production, increases the ability to survive under oxidative and inflammatory stress conditions. It is mostly dependent on the expression of several genes with different antioxidant and cytoprotective activities induced by Nrf2 [9]. Various Nrf2 target genes are related to cancer cell proliferation and death, such as genes involved in the pentose phosphate pathway and responsible for NADPH and purine regeneration, leading to the stimulation of cancer cell proliferation [36,37,38]. HO-1 is another target of Nrf2 that has potent antioxidant and anti-apoptotic effects that can increase cancer cell growth and resistance to therapy [38]. High Nrf2-HO-1 levels have mostly been found in malignant tumor cells with aggressiveness and poor outcome [38]. Indeed, tumor cells use Nrf2 as a protective mechanism to increase their survival. The paradoxical activity of Nrf2 in GC suggests a “dark side profile” in the progression of GC tumors.
Nrf2 may also induce malignancy in tumor cells by binding to the ARE sequence in the promoter region of Notch1 and the p53 inhibitor Mdm2. Mdm2 expression induces high levels of p53, leading to cell death [36,37,38]. In addition, Nrf2 induces angiogenesis through stimulating hypoxia-inducible factor 1-alpha (HIF-1α)-dependent vascular endothelial growth factor (VEGF) expression in cancer cells and promoting cancer growth [36,37,38].
However, the exact mechanisms for these oncogenic effects of Nrf2 in cancer development are not clear. This study reviewed the available publications on the roles of Nrf2 in GC, focusing on the Nrf2 inhibitors and activators to summarize information for easy access for future investigations on GC therapy.

2. Treatment Modalities of Gastric Cancer

For diagnosed gastric cancer (GC), the only curative therapeutic option is surgical resection [39]. Other therapeutic options in GC, such as perioperative and adjuvant chemotherapy along with chemoradiation, are more suitable when GC can be resected and lymph nodes need to be dissected [39]. Altogether, these strategies result in a 15% increase in survival compared with surgery alone [39].
In the classical surgical settings, for the thorough resection and staging of GC, total gastrectomy and subtotal gastrectomy along with lymphadenectomy are the common approaches [39]. For the extent of gastric resection, 2 to 6 cm gross margins are the range reported to be most appropriate for the minimal chance of microscopic positive margins. However, there are no universally acceptable extents for lymphadenectomy [47]. The emergence of minimally invasive gastrectomy using laparoscopic and robotic gastrectomy has brought advantages, such as reduced blood loss, morbidity, and hospitalization periods, and a shorter normalization of bowel function. The oncologic outcome regarding mortality and lymph node procurement are similar between open and minimally invasive surgery for GC [48]. The peritoneal spread of gastric adenocarcinoma needs different treatment modalities than surgical resection alone because of its poor prognosis and high recurrence rate.
Surgical removal of GC is feasible in only 20% of cases. Locoregional and/or metastatic recurrences reduce the median survival after surgery to 12–20 months. These problems led to the use of neoadjuvant or adjuvant treatment modalities in addition to surgery. Therapeutic strategies with adjuvant chemotherapy compared with surgery alone have shown conflicting results in terms of survival [49]. More reliable results obtained from several meta-analyses shows that adjuvant chemotherapy has a significant benefit, as it can improve the overall survival and decrease the recurrence rates compared with surgery alone [50,51,52,53].
Adjuvant chemoradiation has also shown a survival benefit for the management of GC [54]. Studies have shown an advantage for adjuvant chemoradiation compared with adjuvant radiotherapy or chemotherapy alone [55,56]. For completely resected carcinoma of the esophagogastric junction or GC, treatment with postoperative chemoradiation has been suggested as the standard of care when neoadjuvant therapy has not been used [47,57].
An open-label phase 2/3 trial highlighted the benefit of adjuvant chemotherapy for gastric adenocarcinoma [51]. In patients with stage II/III gastric or esophageal cancer, neoadjuvant therapy with epirubicin, cisplatin, and 5-fluorouracil exhibited superior outcomes compared with surgery alone, as evidenced by higher rates of progression-free and overall survival [58]. Furthermore, neoadjuvant therapy with 5-fluorouracil, leucovorin, oxaliplatin, and docetaxel showed better compliance and efficiency in terms of periods without tumor progression, overall survival, and complete the regression of pathologies compared with another perioperative regimen comprised of epirubicin, cisplatin, and 5-fluorouracil or epirubicin, cisplatin, and capecitabine [59].
The identification of tumor markers and the genetic profiling of cancers offer the possibility for personalized treatment regimens. In the case of GC, monoclonal antibodies and small-molecule inhibitors are used in targeted therapies. The elevated human epidermal growth factor receptor 2 (HER2) expression in GC makes this protein a suitable treatment target. HER2 belongs to the epidermal growth factor receptor (EGFR) family, which is involved in the tumor cell proliferation, migration, differentiation, apoptosis, and adhesion. Several studies have indicated the important role of HER2 in tumorigenesis in gastric cancer [60,61]. The overexpression of HER2 shows poor prognosis in gastric cancer patients [62]. The combination of trastuzumab (anti-HER2 monoclonal antibody) and chemotherapy showed improvements in overall survival and disease-free time compared with chemotherapy alone in advanced GC patients [63]. Another targeted therapeutic agent for GC is the anti-epidermal growth factor receptor (EGFR) monoclonal antibody nimotuzumab [64]. The combination of nimotuzumab with irinotecan improved the response rate and progression-free and overall survival in patients with advanced GC who were EGFR-overexpressed. Ramucirumab, a monoclonal antibody against vascular endothelial growth factor receptor (VEGFR)-2, prolonged median survival compared with a placebo in patients with advanced GC. Adding ramucirumab to paclitaxel prolonged the overall survival and periods without tumor progression compared with paclitaxel treatment alone in the same patients [65,66].

3. Nrf2 Inhibitors and Chemotherapy Drug Resistance

3.1. Anti-HER2 Drugs Resistance

HER2 has previously been reported to be elevated in 7 to 34% of GC patients [67,68]. The administration of anti-HER2 drugs including trastuzumab plus platinum-based chemotherapy could decrease the resistance to chemotherapy. However, resistance persists with trastuzumab plus platinum drug chemotherapy in tumors with amplified HER2 [69,70,71,72]. Gambardella and coworkers [61] investigated the mechanisms of primary and acquired resistance to the combination of trastuzumab and platinum-based chemotherapy [73]. Gambardella and coworkers [61] found that NRF2 amplified the resistance to anti-HER2 drugs through the PI3K/AKT/mTOR/RPS6 pathway, and that ribosomal protein S6 (RPS6) inhibition decreased NRF2 expression and restored sensitivity in HER2-amplified gastric cancer.
The inhibition of RPS6 reduced the expression of Nrf2 and increased the sensitivity to chemotherapy in HER2-amplified GC cells. Yang and coworkers (2020) reported that brusatol, a modern Nrf2 inhibitor, showed anti-tumor properties, which were shown as decreased cancer cell growth by suppressing the Nrf2/HO-1 and HER2-AKT/ERK1/2 signaling in HER2-positive cancer cells (SK-OV-3 and BT-474 cells) [74].
It was found that brusatol exerted a synergistic effect with trastuzumab against HER2-positive cells by suppressing Nrf2/HO-1 and HER2-AKT/ERK1/2 signaling.
The combination of trastuzumab and brusatol significantly downregulated phosphorylation-HER2, as well as phosphorylation-ERK1/2 and phospho-AKT. However, the levels of the Nrf2 and HO-1 were reduced in the two HER2-positive cell lines by the combination therapy. Therefore, brusatol may increase the antitumor effect of trastuzumab via the inhibition of the Nrf2/HO-1 and HER2-AKT/ERK1/2 signaling pathway in HER2-positive cancer cells. Brusatol alone caused a moderate elevation of ROS production and apoptosis in BT-474 and SK-OV-3 cells. Meanwhile, trastuzumab plus brusatol significantly increased ROS accumulation and apoptosis when compared to brusatol or trastuzumab treatment alone in both BT-474 and SK-OV-3 cells [62]. Therefore, combining Nrf2 inhibition with anti-HER2 drugs is a suitable therapeutic strategy for the treatment of HER2-positive cancers (Figure 1).

3.2. 5-Fluorouracil Resistance

Hu and coworkers [63] found that silencing the expression of Nrf2 decreased the resistance to fluorouracil (5-FU) and increased cell death in GC cell lines [75]. They also suggested that Nrf2 is an important predictive factor for 5-FU resistance in GC specimens. In addition, Pouremamali and coworkers [64] found that the effect of Nrf2 on malic enzyme-1 (ME-1) leads to the resistance of MKN-45 (MKN-45/DR) cells to 5-FU [76]. The ME enzyme exists in three forms in mammals, including ME-1 (cytosolic NADP+ dependent isoform), ME-2 (mitochondrial NAD+ dependent isoform), and ME-3 (mitochondrial NADP+ dependent isoform). ME-1 expression was induced in several human cancers [77]. GC cells with 5-FU resistance had decreased Nrf2 and ME-1 expressions and elevated MDR1 mRNA expression compared with 5-FU-susceptible cells. Thedministration of luteolin plus brusatol, two Nrf2 inhibitors, synergistically increased the cytotoxicity of 5-FU. MKN-45/DR cells incubated with luteolin plus brusatol elevated Nrf2 and ME-1 expression. They found that a combination of Nrf2 inhibitors is a suitable strategy for combating 5-FU resistance in GC patients (Figure 2).

3.3. Oxaliplatin Resistance

Oxaliplatin or Eloxatin is an alkylating chemotherapeutic agent that has high efficacy against gastrointestinal tract cancers in combination with 5-FU and folinic acid [78,79,80]. It has been reported that oxaliplatin produces cytotoxicity by forming platinum–DNA adducts that subsequently block DNA replication and induce cell death [81]. Resistance to oxaliplatin is caused by disturbed DNA adduct formation [82] and abnormalities in autophagy and apoptotic hypoxia following hypoxia-inducible factor-1a (HIF-1a) pathway stimulation [83,84]. Furthermore, the resistance of the human GC cell line TSGH-S3 (S3) to oxaliplatin may be due to the increased repairment of DNA and copper-transporting ATPase 1 (ATP7A) contents [85]. Furthermore, Chen and coworkers [85] investigated other resistance factors in S3 cells by using a gene array assay. The interleukin-6 (IL-6), aldo-keto reductase 1C1 (AKR1C1), and AKR1C3 genes are mostly up-regulated in S3 cells compared with TSGH parental cells. Since the AKR1Cs gene belongs to the ARE genes, Nrf2 inhibition decreased AKR1C1, AKR1C2, and AKR1C3 mRNA and reversed the oxaliplatin resistance in S3 cells (Figure 2). The findings indicated that the inhibition of the Nrf2/AKR1C pathway can reverse S3 cells’ resistance to oxaliplatin (Table 1).

3.4. Multidrug Resistance

It is well known that multi-drug resistance (MDR) is an important and serious problem for achieving success in chemotherapy. Interestingly, most ABC transporter genes, as the main regulator of MDR, are modulated via the Nrf2 signaling pathway [9,20]. Jeddi and co-workers [75] found increased expression and protein levels of Nrf2, as well as MDR1/P-gp, in GC patients [86]. The stimulation of P-gp (a drug efflux pump) was associated with Nrf2 induction, tumor size, and histological grade. Moreover, the inhibition of Nrf2 expression could increase the efficacy of chemotherapeutic agents by down-regulating the expression of P-gp in GC patients.
The overexpression of DJ-1, an oncogene product, is found in various malignant cancers, including GC [87,88]. Moreover, Zhu and co-workers [88] found elevated levels of DJ-1 protein expression in HeLa cells resistant to dihydroartemisinin (DHA), and the silencing of DJ-1 increased the DHA-sensitivity of HeLa cells. The findings indicated that DJ-1 has a major role in MDR in SGC7901 GC cells via the up-regulation of P-gp and Bcl-2 [89]. P-glycoprotein (P-gp) is a well-identified membrane transporter that is able to pump out drug molecules from the cancer cell and decrease chemotherapy efficiency. In cancer cells, the up-regulation of P-gp expression occurs as an adaptive response to combat cell death induced by chemotherapy [90]. P-gp is a well-identified membrane transporter with capability to efflux drug molecules out of the cancer cell leading to reduced efficiency of chemotherapy which is directly associated with elevated MDR in tumors. In addition, Bcl-2 overexpression can prevent the chemotherapy-induced expression of Fas/Fas ligand (FasL) and blocks chemotherapy-induced apoptosis through preventing the nuclear translocation of NFAT (nuclear factor of activated T lymphocytes, a transcription factor activated by microtubule damage). The FasL gene is not transcribed without NFAT nuclear translocation. Bcl-2 antagonizes drug-induced apoptosis by inhibiting calcineurin activation, blocking NFAT nuclear translocation and preventing FasL expression [91].
DJ-1 induces Nrf2/Keap1 uncoupling and thus can modulate Nrf2 activity [92,93]. However, DJ-1 cannot affect Nrf2 or Keap1 in a direct manner [92]. It has been found that Nrf2/Keap1 dissociation usually occurs following Nrf2 phosphorylation [94]. Nrf2 phosphorylation correlates with PI3K/Akt, PKC, etc. [95]. DJ-1 can bind to PTEN and prevents its activity, leading to the stimulation of PI3K/Akt [96,97]. A relation between the MDR of GC cells and the activation of PI3K/Akt has been found [98,99]. Qiu and co-workers (2020) showed that the DJ-1-related induction of MDR occurs through the stimulation of the PTEN/PI3K/Akt/Nrf2 signaling pathway, as well as P-gp and Bcl-2 up-regulation in SGC7901 GC cells [76] (Figure 2).

4. Natural Therapeutic Agents for Gastric Cancer and Nrf2

Cancer chemoprevention with natural agents has become a focus of attention in recent years. In this context, baicalin and its aglycone, baicalein, have been found to have anti-tumor effects. Baicalin is the main flavonoid found in several plants including Scutellaria baicalensis Georgi, S. lateriflora L. S. galericulata, S. rivularis, and Oroxylum indicum (L.) Kurz [100,101,102].
The anti-progressive effects of baicalein have been found in MGC-803, SGC-7901, SGC-7901/DDP, and HGC-27 GC cell lines [103]. Furthermore, adding baicalein to diamminedichloroplatinum (DDP) suppressed progression and elevated apoptosis and autophagy in SGC-7901 and SGC-7901/DDP GC cell lines. The modulatory effect of baicalein on Akt/mTOR and Nrf2/Keap 1 signaling affects SGC-7901/DDP GC cells’ sensitivity to chemotherapy. In addition, baicalein stimulated Akt/mTOR- and Nrf2/Keap1-mediated apoptosis and autophagy which subsequently increased the sensitivity of GC cells to DDP [95].
Baicalein significantly improved Nrf2 expression and decreased the Keap 1 level dose-dependently. This mechanism indicates the antioxidative activity of baicalein, and is not related to the stimulatory effect of baicalein on the DDP sensitivity of SGC-7901/DDP. However, baicalein down-regulated MDR1 expression in SGC-7901 and SGC-7901/DDP dose- and time-dependently [95].
Garlic (Allium sativum L.) is another natural compound with anti-cancer activity. Garlic consumption is accompanied with a decrease in the incidence of GC [104]. Diallyl trisulfide (DATS), an oil-soluble compound isolated from garlic, exerted antitumor effects [105]. Indeed, DATS acts as a chemotherapeutic agent through the induction of oxidative stress [106], cell cycle arrest, apoptosis, and also proliferation suppression [107], as well as tumor cell invasion and metastasis inhibition [108].
Jiang and co-workers [99] indicated the chemo-preventive effects of DATS in BGC-823 human GC cells, and DATS stimulated the anti-tumor effects of DDP in BGC-823 xenograft mice [109]. DATS could inhibit the viability and induced cell cycle arrest in the G2/M phase in BGC-823 cells in a dose-dependent manner. It was shown that the protective effects of DATS in BGC-823 cells were caused by activation of p38 and JNK/MAPK and the attenuation of the Nrf2/Akt signaling pathway. DATS reduced Akt phosphorylation in tumors, leading to a decrease in the Nrf2 levels both in vitro and in vivo. In anticancer therapy, the PI3K/Akt signaling pathway is involved in Nrf2 expression [101].

5. Biomarkers of Gastric Cancer and Nrf2

The lymphoid enhancer factor/T cell factor (LEF/TCF) family of transcription factors is localized in the nucleus. One member, transcription factor 7-like 1 (TCF7L1), is a DNA-specific binding protein that binds to β-catenin and regulates Wnt/β-catenin signaling [110]. TCF7L1 expression is up-regulated in malignant tumors and indicates a poor prognosis. It was found that in breast cancer and colorectal cancer, the elimination of TCF7L1 expression reduced tumor cell progression and metastasis [111]. In breast cancer and colorectal cancer, TCF7L1 regulates Wnt/β-catenin signaling [112]. Wnt signaling regulates proliferation and cell hemostasis in normal gastric mucosa; however, its activation can be found in GC development. The ability of TCF7L1 to regulate Wnt/β-catenin signaling implicates its role in GC prognosis.
In this context, Zhang and co-workers [103] showed that a higher expression of TCF7L1 is associated with a poorer prognosis in patients [113]. TCF7L1 was also found to negatively modulate the expression of Keap1, leading to Nrf2 activation.
Peng and co-workers [104] investigated the association between high Nrf2 levels and the expression of NAD(P)H: quinone oxidoreductase 1 (NQO1) in chemotherapy-resistant GC patients [114]. It was found that Nrf2 and NQO1 expressions are indicators of developed GC. They also indicated that high Nrf2 expression in GC tissues directly correlates with the progression and metastatic features of GC, as well as resistance to 5-FU [106].
BDH2, also known as DHRS6, is a short-chain dehydrogenase/reductase [115], which is mostly expressed in the epithelial cells of the kidney, oral cavity, small intestine, and breast [116]. BDH2 independently indicates the poor prognosis of acute myeloid leukemia. BDH2 affects the progression of squamous cell carcinoma of the esophagus [117]. Liu and co-workers (2020) found that suppressed BDH2 expression was an indicator of tumor growth in GC tissue. BDH2 stimulated Keap1 and Nrf2 interactions, leading to a decrease in the Nrf2 translocation to the nucleus. The ubiquitination of Nrf2 blocked ARE activity, resulting in the inhibition of AktSer473 and mTORSer2448 phosphorylation, thereby changing the PI3K/Akt /mTOR pathway and ultimately reducing GC cell growth [109].

6. Conclusions

Anti-cancer modalities can use the Nrf2 signaling pathway as a therapeutic target. The role of Nrf2 in cancer is controversial, as it has protective effects in both normal and cancerous cells. In GC cells, the inhibition of Nrf2 increased their sensitivity towards chemotherapeutic agents. Accordingly, Nrf2 inhibitors abolished the resistance of GC cells to several anti-HER2 drugs, 5-FU, and oxaliplatin. The inhibition of Nrf2 expression can be achieved by inhibiting the RPS6, PTEN/PI3K/Akt/Nrf2, PI3K/AKT/mTOR/RPS6, Nrf2/HO-1, and HER2-AKT/ERK1/2 pathways in GC. In addition, the inhibition of the Nrf2/AKR1C pathway reverses resistance in S3 cells. However, most experimental studies indicated the proactive effect of natural antioxidants against cancer through the Nrf2 signaling pathway. In this context, baicalin, DATS, and luteolin attenuated Nrf2 signaling and increased the sensitivity towards chemotherapeutic agents in different GC cell lines. There are several biomarkers related to the high expression of Nrf2, which indicates the poor prognosis of GC. In this context, TCF7L1, NQO1, and BDH2 can act as biomarkers for the poor prognosis of GC, which also indicate Nrf2 induction. It has been suggested that the evaluation of these biomarkers may be effective in providing an appropriate therapeutic approach for the management of GC. Altogether, numerous scientific reports indicate the efficacy of targeting Nrf2 in various diseases. In addition, there is evidence of the beneficial effect of Nrf2 activity and the suppressive role of Nrf2 inhibitors against tumor progression in human cancers. However, we should clearly inform that contradictory activity of Nrf2 in tumor progression has been found in recent years.

Author Contributions

Study conception and design: S.S., M.S. and T.F.; acquisition of data: T.F., A.M.P.-S., M.A.-N., M.A., F.F. and A.B.; drafting of the manuscript: T.F., A.M.P.-S., M.A., F.F., M.T. and A.B.; critical revision: S.S., M.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

5-FU 5-fluorouracil
AKR Aldo-keto reductase
AREs Antioxidant responsive elements
BCL2B-cell lymphoma 2
BDH2 3-Hydroxybutyrate dehydrogenase 2
DATS Diallyl trisulfide
DHADihydroartemisinin
DNA Deoxyribonucleic acid
EBV Epstein–Barr virus
EGFR Anti-epidermal growth factor receptor
GC Gastric cancer
GCL Glutamate–cysteine ligase
GRIM Retinoid-IFN-induced mortality
HER2 Human epidermal growth factor receptor 2
HIF-1a Hypoxia-inducible factor-1a
HO-1 Heme oxygenase-1
IL Interleukin
Keap1 Kelch-like ECH-associated protein 1
LEF Lymphoid enhancer factor
MAFMusculoaponeurotic fibrosarcoma
MARE MAF recognition elements
MDR Multidrug resistance
MEMalic enzyme
NAD Nicotinamide adenine dinucleotide
NQO1 Quinone oxidoreductase 1
Nrf2 Nuclear factor erythroid 2-related factor 2
P-gpP-glycoprotein 1
RNA Ribonucleic acid
ROS Reactive oxygen species
RPS6 Ribosomal protein S6
TCF T cell factor
VEGF Vascular endothelial growth factor
VEGFRAnti-vascular endothelial growth factor receptor

References

  1. Wright, N.A.; Poulsom, R.; Stamp, G.; van Noorden, S.; Sarraf, C.; Elia, G.; Ahnen, D.; Jeffery, R.; Longcroft, J.; Pike, C.; et al. Trefoil peptide gene expression in gastrointestinal epithelial cells in inflammatory bowel disease. Gastroenterology 1993, 104, 12–20. [Google Scholar] [CrossRef]
  2. Jemal, A.; Bray, F.; Center, M.M.; Ferlay, J.; Ward, E.; Forman, D. Global cancer statistics. CA Cancer J. Clin. 2011, 61, 69–90. [Google Scholar] [CrossRef] [Green Version]
  3. Abadi, A.J.; Zarrabi, A.; Hashemi, F.; Zabolian, A.; Najafi, M.; Entezari, M.; Hushmandi, K.; Aref, A.R.; Khan, H.; Makvandi, P.; et al. The role of SOX family transcription factors in gastric cancer. Int. J. Biol. Macromol. 2021, 180, 608–624. [Google Scholar] [CrossRef] [PubMed]
  4. Stock, M.; Otto, F. Gene deregulation in gastric cancer. Gene 2005, 360, 1–19. [Google Scholar] [CrossRef] [PubMed]
  5. Ashrafizadeh, M.; Zarrabi, A.; Hashemipour, M.; Vosough, M.; Najafi, M.; Shahinozzaman, M.; Hushmandi, K.; Khan, H.; Mirzaei, H. Sensing the scent of death: Modulation of microRNAs by Curcumin in gastrointestinal cancers. Pharmacol. Res. 2020, 160, 105199. [Google Scholar] [PubMed]
  6. Ashrafizadeh, M.; Najafi, M.; Ang, H.; Moghadam, E.; Mahabady, M.; Zabolian, A.; Jafaripour, L.; Bejandi, A.; Hushmandi, K.; Saleki, H.; et al. PTEN, a Barrier for Proliferation and Metastasis of Gastric Cancer Cells: From Molecular Pathways to Targeting and Regulation. Biomedicines 2020, 8, 264. [Google Scholar] [CrossRef] [PubMed]
  7. Ashrafizadeh, M.; Zarrabi, A.; Orouei, S.; Zarrin, V.; Rahmani Moghadam, E.; Zabolian, A.; Mohammadi, S.; Hushmandi, K.; Gharehaghajlou, Y.; Makvandi, P.; et al. STAT3 Pathway in Gastric Cancer: Signaling, Therapeutic Targeting and Future Prospects. Biology 2020, 9, 126. [Google Scholar] [CrossRef] [PubMed]
  8. Muñoz, N.; Franceschi, S. Epidemiology of gastric cancer and perspectives for prevention. Salud Publica Mex. 1997, 39, 318–330. [Google Scholar] [CrossRef]
  9. Ladeiras-Lopes, R.; Pereira, A.K.; Nogueira, A.; Pinheiro-Torres, T.; Pinto, I.; Santos-Pereira, R.; Lunet, N. Smoking and gastric cancer: Systematic review and meta-analysis of cohort studies. Cancer Causes Control 2008, 19, 689–701. [Google Scholar] [CrossRef]
  10. Nishino, Y.; Inoue, M.; Tsuji, I.; Wakai, K.; Nagata, C.; Mizoue, T.; Tanaka, K.; Tsugane, S. Tobacco smoking and gastric cancer risk: An evaluation based on a systematic review of epidemiologic evidence among the Japanese population. Jpn. J. Clin. Oncol. 2006, 36, 800–807. [Google Scholar] [CrossRef] [Green Version]
  11. Ashrafizadeh, M.; Rafiei, H.; Mohammadinejad, R.; Farkhondeh, T.; Samarghandian, S. Wnt-regulating microRNAs role in gastric cancer malignancy. Life Sci. 2020, 250, 117547. [Google Scholar] [PubMed]
  12. Imai, S.; Koizumi, S.; Sugiura, M.; Tokunaga, M.; Uemura, Y.; Yamamoto, N.; Tanaka, S.; Sato, E.; Osato, T. Gastric carcinoma: Monoclonal epithelial malignant cells expressing Epstein-Barr virus latent infection protein. Proc. Natl. Acad. Sci. USA 1994, 91, 9131–9135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Vaughan, T.L.; Davis, S.; Kristal, A.; Thomas, D.B. Obesity, alcohol, and tobacco as risk factors for cancers of the esophagus and gastric cardia: Adenocarcinoma versus squamous cell carcinoma. Cancer Epidemiol. Biomark. Prev. 1995, 4, 85–92. [Google Scholar]
  14. Hsing, A.W.; Hansson, L.E.; McLaughlin, J.K.; Nyren, O.; Blot, W.J.; Ekbom, A.; Fraumeni, J.F., Jr. Pernicious anemia and subsequent cancer. A population-based cohort study. Cancer 1993, 71, 745–750. [Google Scholar] [CrossRef]
  15. Aird, I.; Bentall, H.H.; Roberts, J.A. A relationship between cancer of stomach and the ABO blood groups. Br. Med. J. 1953, 1, 799–801. [Google Scholar] [CrossRef] [Green Version]
  16. Offerhaus, G.J.; Tersmette, A.C.; Huibregtse, K.; van de Stadt, J.; Tersmette, K.W.; Stijnen, T.; Hoedemaeker, P.J.; Vandenbroucke, J.P.; Tytgat, G.N. Mortality caused by stomach cancer after remote partial gastrectomy for benign conditions: 40 years of follow up of an Amsterdam cohort of 2633 postgastrectomy patients. Gut 1988, 29, 1588–1590. [Google Scholar] [CrossRef] [Green Version]
  17. Shen, J.; Rasmussen, M.; Dong, Q.R.; Tepel, M.; Scholze, A. Expression of the NRF2 Target Gene NQO1 is Enhanced in Mononuclear Cells in Human Chronic Kidney Disease. Oxid. Med. Cell. Longev. 2017, 2017, 9091879. [Google Scholar] [CrossRef] [Green Version]
  18. Loboda, A.; Damulewicz, M.; Pyza, E.; Jozkowicz, A.; Dulak, J. Role of Nrf2/HO-1 system in development, oxidative stress response and diseases: An evolutionarily conserved mechanism. Cell Mol. Life Sci. 2016, 73, 3221–3247. [Google Scholar] [CrossRef] [Green Version]
  19. Kansanen, E.; Kuosmanen, S.M.; Leinonen, H.; Levonen, A.-L. The Keap1-Nrf2 pathway: Mechanisms of activation and dysregulation in cancer. Redox Biol. 2013, 1, 45–49. [Google Scholar] [CrossRef] [Green Version]
  20. Zimta, A.A.; Cenariu, D.; Irimie, A.; Magdo, L.; Nabavi, S.M.; Atanasov, A.G.; Berindan-Neagoe, I. The Role of Nrf2 Activity in Cancer Development and Progression. Cancers 2019, 11, 1755. [Google Scholar] [CrossRef] [Green Version]
  21. Ashrafizadeh, M.; Fekri, H.S.; Ahmadi, Z.; Farkhondeh, T.; Samarghandian, S. Therapeutic and biological activities of berberine: The involvement of Nrf2 signaling pathway. J. Cell. Biochem. 2020, 121, 1575–1585. [Google Scholar] [CrossRef] [PubMed]
  22. Ashrafizadeh, M.; Ahmadi, Z.; Mohammadinejad, R.; Farkhondeh, T.; Samarghandian, S. Curcumin activates the Nrf2 pathway and induces cellular protection against oxidative injury. Curr. Mol. Med. 2020, 20, 116–133. [Google Scholar]
  23. Farkhondeh, T.; Folgado, S.L.; Pourbagher-Shahri, A.M.; Ashrafizadeh, M.; Samarghandian, S. Pharmacotherapy. The therapeutic effect of resveratrol: Focusing on the Nrf2 signaling pathway. Curr. Mol. Med. 2020, 127, 110234. [Google Scholar]
  24. Mirzaei, S.; Mohammadi, A.T.; Gholami, M.H.; Hashemi, F.; Zarrabi, A.; Zabolian, A.; Hushmandi, K.; Makvandi, P.; Samec, M.; Liskova, A.; et al. Nrf2 signaling pathway in cisplatin chemotherapy: Potential involvement in organ protection and chemoresistance. Pharmacol. Res. 2021, 167, 105575. [Google Scholar]
  25. Mirzaei, S.; Zarrabi, A.; Hashemi, F.; Zabolian, A.; Saleki, H.; Azami, N.; Hamzehlou, S.; Farahani, M.V.; Hushmandi, K.; Ashrafizadeh, M.; et al. Nrf2 Signaling Pathway in Chemoprotection and Doxorubicin Resistance: Potential Application in Drug Discovery. Antioxidants 2021, 10, 349. [Google Scholar] [CrossRef]
  26. Blagotinsek, K.; Rozman, D. Targeting Signalling Pathways in Hepatocellular Carcinoma. Curr. Pharm. Des. 2017, 23, 170–175. [Google Scholar] [CrossRef]
  27. Yagishita, Y.; Fukutomi, T.; Sugawara, A.; Kawamura, H.; Takahashi, T.; Pi, J.; Uruno, A.; Yamamoto, M. Nrf2 protects pancreatic β-cells from oxidative and nitrosative stress in diabetic model mice. Diabetes 2014, 63, 605–618. [Google Scholar] [CrossRef] [Green Version]
  28. Strom, J.; Xu, B.; Tian, X.; Chen, Q.M. Nrf2 protects mitochondrial decay by oxidative stress. FASEB J. 2016, 30, 66–80. [Google Scholar] [CrossRef] [Green Version]
  29. Noel, S.; Martina, M.N.; Bandapalle, S.; Racusen, L.C.; Potteti, H.R.; Hamad, A.R.; Reddy, S.P.; Rabb, H. T Lymphocyte-Specific Activation of Nrf2 Protects from AKI. J. Am. Soc. Nephrol. 2015, 26, 2989–3000. [Google Scholar] [CrossRef] [Green Version]
  30. Jiang, T.; Harder, B.; de la Vega, M.R.; Wong, P.K.; Chapman, E.; Zhang, D.D. p62 links autophagy and Nrf2 signaling. Free Radic. Biol. Med. 2015, 88, 199–204. [Google Scholar] [CrossRef] [Green Version]
  31. Ashrafizadeh, M.; Ahmadi, Z.; Farkhondeh, T.; Samarghandian, S. Back to nucleus: Combating with cadmium toxicity using Nrf2 signaling pathway as a promising therapeutic target. Biol. Trace Elem. Res. 2020, 197, 52–62. [Google Scholar]
  32. Ashrafizadeh, M.; Ahmadi, Z.; Samarghandian, S.; Mohammadinejad, R.; Yaribeygi, H.; Sathyapalan, T.; Sahebkar, A. MicroRNA-mediated regulation of Nrf2 signaling pathway: Implications in disease therapy and protection against oxidative stress. Life Sci. 2020, 244, 117329. [Google Scholar]
  33. Chen, W.; Sun, Z.; Wang, X.J.; Jiang, T.; Huang, Z.; Fang, D.; Zhang, D.D. Direct interaction between Nrf2 and p21(Cip1/WAF1) upregulates the Nrf2-mediated antioxidant response. Mol. Cell 2009, 34, 663–673. [Google Scholar] [CrossRef] [Green Version]
  34. Moon, E.J.; Giaccia, A. Dual roles of NRF2 in tumor prevention and progression: Possible implications in cancer treatment. Free Radic. Biol. Med. 2015, 79, 292–299. [Google Scholar] [CrossRef] [Green Version]
  35. Na, H.K.; Surh, Y.J. Oncogenic potential of Nrf2 and its principal target protein heme oxygenase-1. Free Radic. Biol. Med. 2014, 67, 353–365. [Google Scholar] [CrossRef]
  36. Ma, Q. Role of nrf2 in oxidative stress and toxicity. Annu. Rev. Pharmacol. Toxicol. 2013, 53, 401–426. [Google Scholar] [CrossRef] [Green Version]
  37. Li, R.; Jia, Z.; Zhu, H. Regulation of Nrf2 Signaling. React. Oxyg. Species 2019, 8, 312–322. [Google Scholar] [CrossRef]
  38. Jaramillo, M.C.; Zhang, D.D. The emerging role of the Nrf2-Keap1 signaling pathway in cancer. Genes Dev. 2013, 27, 2179–2191. [Google Scholar] [CrossRef] [Green Version]
  39. DeNicola, G.M.; Karreth, F.A.; Humpton, T.J.; Gopinathan, A.; Wei, C.; Frese, K.; Mangal, D.; Yu, K.H.; Yeo, C.J.; Calhoun, E.S.; et al. Oncogene-induced Nrf2 transcription promotes ROS detoxification and tumorigenesis. Nature 2011, 475, 106–109. [Google Scholar] [CrossRef]
  40. Satoh, H.; Moriguchi, T.; Takai, J.; Ebina, M.; Yamamoto, M. Nrf2 Prevents Initiation but Accelerates Progression through the Kras Signaling Pathway during Lung Carcinogenesis. Cancer Res. 2013, 73, 4158–4168. [Google Scholar] [CrossRef] [Green Version]
  41. Tao, S.; de la Vega, M.R.; Chapman, E.; Ooi, A.; Zhang, D.D. The effects of NRF2 modulation on the initiation and progression of chemically and genetically induced lung cancer. Mol. Carcinog. 2018, 57, 182–192. [Google Scholar] [CrossRef] [PubMed]
  42. Lau, A.; Villeneuve, N.F.; Sun, Z.; Wong, P.K.; Zhang, D.D. Dual roles of Nrf2 in cancer. Pharmacol. Res. 2008, 58, 262–270. [Google Scholar] [CrossRef]
  43. Hayes, J.D.; McMahon, M. NRF2 and KEAP1 mutations: Permanent activation of an adaptive response in cancer. Trends Biochem. Sci. 2009, 34, 176–188. [Google Scholar]
  44. Singh, A.; Venkannagari, S.; Oh, K.H.; Zhang, Y.-Q.; Rohde, J.M.; Liu, L.; Nimmagadda, S.; Sudini, K.; Brimacombe, K.R.; Gajghate, S. Small molecule inhibitor of NRF2 selectively intervenes therapeutic resistance in KEAP1-deficient NSCLC tumors. ACS Chem. Biol. 2016, 11, 3214–3225. [Google Scholar] [CrossRef] [Green Version]
  45. Wang, H.; Liu, X.; Long, M.; Huang, Y.; Zhang, L.; Zhang, R.; Zheng, Y.; Liao, X.; Wang, Y.; Liao, Q.; et al. NRF2 activation by antioxidant antidiabetic agents accelerates tumor metastasis. Sci. Transl. Med. 2016, 8, 334ra51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Piskounova, E.; Agathocleous, M.; Murphy, M.M.; Hu, Z.; Huddlestun, S.E.; Zhao, Z.; Leitch, A.M.; Johnson, T.M.; DeBerardinis, R.J.; Morrison, S.J. Oxidative stress inhibits distant metastasis by human melanoma cells. Nature 2015, 527, 186–191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Johnston, F.M.; Beckman, M. Updates on management of gastric cancer. Curr. Oncol. Rep. 2019, 21, 67. [Google Scholar] [CrossRef]
  48. Huscher, C.G.; Mingoli, A.; Sgarzini, G.; Sansonetti, A.; di Paola, M.; Recher, A.; Ponzano, C. Laparoscopic versus open subtotal gastrectomy for distal gastric cancer: Five-year results of a randomized prospective trial. Ann. Surg. 2005, 241, 232–237. [Google Scholar] [CrossRef]
  49. Orditura, M.; Galizia, G.; Sforza, V.; Gambardella, V.; Fabozzi, A.; Laterza, M.M.; Andreozzi, F.; Ventriglia, J.; Savastano, B.; Mabilia, A. Treatment of gastric cancer. World J. Gastroenterol. 2014, 20, 1635. [Google Scholar] [CrossRef]
  50. Zhao, S.L.; Fang, J.Y. The role of postoperative adjuvant chemotherapy following curative resection for gastric cancer: A meta-analysis. Cancer Investig. 2008, 26, 317–325. [Google Scholar] [CrossRef] [PubMed]
  51. Liu, T.S.; Wang, Y.; Chen, S.Y.; Sun, Y.H. An updated meta-analysis of adjuvant chemotherapy after curative resection for gastric cancer. Eur. J. Surg. Oncol. 2008, 34, 1208–1216. [Google Scholar] [CrossRef]
  52. Paoletti, X.; Oba, K.; Burzykowski, T.; Michiels, S.; Ohashi, Y.; Pignon, J.P.; Rougier, P.; Sakamoto, J.; Sargent, D.; Sasako, M.; et al. Benefit of adjuvant chemotherapy for resectable gastric cancer: A meta-analysis. JAMA 2010, 303, 1729–1737. [Google Scholar] [CrossRef]
  53. Janunger, K.G.; Hafström, L.; Glimelius, B. Chemotherapy in gastric cancer: A review and updated meta-analysis. Eur. J. Surg. Acta Chir. 2002, 168, 597–608. [Google Scholar] [CrossRef]
  54. Schernberg, A.; del Campo, E.R.; Rousseau, B.; Matzinger, O.; Loi, M.; Maingon, P.; Huguet, F. Adjuvant chemoradiation for gastric carcinoma: State of the art and perspectives. Clin. Transl. Radiat. Oncol. 2018, 10, 13–22. [Google Scholar] [CrossRef] [Green Version]
  55. Moertel, C.; Reitemeier, R.; Childs, D.J.R.; Colby, M.J.R.; Holbrook, M. Combined 5-fluorouracil and Supervoltage Radiation Therapy of Locally Unresectable Gastrointestinal Cancer. Lancet 1969, 294, 865–867. [Google Scholar] [CrossRef]
  56. Gastrointestinal Tumor Study Group. A comparison of combination chemotherapy and combined modality therapy for locally advanced gastric carcinoma. Cancer 1982, 49, 1771–1777. [Google Scholar] [CrossRef]
  57. Smalley, S.R.; Benedetti, J.K.; Haller, D.G.; Hundahl, S.A.; Estes, N.C.; Ajani, J.A.; Gunderson, L.L.; Goldman, B.; Martenson, J.A.; Jessup, J.M.; et al. Updated Analysis of SWOG-Directed Intergroup Study 0116: A Phase III Trial of Adjuvant Radiochemotherapy Versus Observation After Curative Gastric Cancer Resection. J. Clin. Oncol. 2012, 30, 2327–2333. [Google Scholar] [CrossRef]
  58. Cunningham, D.; Allum, W.H.; Stenning, S.P.; Thompson, J.N.; van de Velde, C.J.H.; Nicolson, M.; Scarffe, J.H.; Lofts, F.J.; Falk, S.J.; Iveson, T.J.; et al. Perioperative Chemotherapy versus Surgery Alone for Resectable Gastroesophageal Cancer. N. Engl. J. Med. 2006, 355, 11–20. [Google Scholar] [CrossRef] [Green Version]
  59. Al-Batran, S.E.; Homann, N.; Pauligk, C.; Goetze, T.O.; Meiler, J.; Kasper, S.; Kopp, H.G.; Mayer, F.; Haag, G.M.; Luley, K.; et al. Perioperative chemotherapy with fluorouracil plus leucovorin, oxaliplatin, and docetaxel versus fluorouracil or capecitabine plus cisplatin and epirubicin for locally advanced, resectable gastric or gastro-oesophageal junction adenocarcinoma (FLOT4): A randomised, phase 2/3 trial. Lancet 2019, 393, 1948–1957. [Google Scholar] [CrossRef] [PubMed]
  60. Konecny, G.E.; Meng, Y.G.; Untch, M.; Wang, H.-J.; Bauerfeind, I.; Epstein, M.; Stieber, P.; Vernes, J.-M.; Gutierrez, J.; Hong, K. Association between HER-2/neu and vascular endothelial growth factor expression predicts clinical outcome in primary breast cancer patients. Clin. Cancer Res. 2004, 10, 1706–1716. [Google Scholar] [CrossRef] [Green Version]
  61. McCubrey, J.A.; Steelman, L.S.; Chappell, W.H.; Abrams, S.L.; Wong, E.W.T.; Chang, F.; Lehmann, B.; Terrian, D.M.; Milella, M.; Tafuri, A. Roles of the Raf/MEK/ERK pathway in cell growth, malignant transformation and drug resistance. Biochim. Biophys. Acta Mol. Cell Res. 2007, 1773, 1263–1284. [Google Scholar]
  62. Lei, Y.-Y.; Huang, J.-Y.; Zhao, Q.-R.; Jiang, N.; Xu, H.-M.; Wang, Z.-N.; Li, H.-Q.; Zhang, S.-B.; Sun, Z. The clinicopathological parameters and prognostic significance of HER2 expression in gastric cancer patients: A meta-analysis of literature. World J. Surg. Oncol. 2017, 15, 1–7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Bang, Y.-J.; van Cutsem, E.; Feyereislova, A.; Chung, H.C.; Shen, L.; Sawaki, A.; Lordick, F.; Ohtsu, A.; Omuro, Y.; Satoh, T.; et al. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): A phase 3, open-label, randomised controlled trial. Lancet 2010, 376, 687–697. [Google Scholar] [CrossRef]
  64. Satoh, T.; Lee, K.H.; Rha, S.Y.; Sasaki, Y.; Park, S.H.; Komatsu, Y.; Yasui, H.; Kim, T.-Y.; Yamaguchi, K.; Fuse, N.; et al. Randomized phase II trial of nimotuzumab plus irinotecan versus irinotecan alone as second-line therapy for patients with advanced gastric cancer. Gastric Cancer 2015, 18, 824–832. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Fuchs, C.S.; Tomasek, J.; Yong, C.J.; Dumitru, F.; Passalacqua, R.; Goswami, C.; Safran, H.; dos Santos, L.V.; Aprile, G.; Ferry, D.R.; et al. Ramucirumab monotherapy for previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma (REGARD): An international, randomised, multicentre, placebo-controlled, phase 3 trial. Lancet 2014, 383, 31–39. [Google Scholar] [CrossRef]
  66. Wilke, H.; Muro, K.; van Cutsem, E.; Oh, S.-C.; Bodoky, G.; Shimada, Y.; Hironaka, S.; Sugimoto, N.; Lipatov, O.; Kim, T.-Y.; et al. Ramucirumab plus paclitaxel versus placebo plus paclitaxel in patients with previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma (RAINBOW): A double-blind, randomised phase 3 trial. Lancet Oncol. 2014, 15, 1224–1235. [Google Scholar] [CrossRef]
  67. Arteaga, C.L.; Engelman, J.A. ERBB receptors: From oncogene discovery to basic science to mechanism-based cancer therapeutics. Cancer Cell 2014, 25, 282–303. [Google Scholar]
  68. Moasser, M.M. The oncogene HER2: Its signaling and transforming functions and its role in human cancer pathogenesis. Oncogene 2007, 26, 6469–6487. [Google Scholar]
  69. Hecht, J.R.; Bang, Y.-J.; Qin, S.K.; Chung, H.C.; Xu, J.M.; Park, J.O.; Jeziorski, K.; Shparyk, Y.; Hoff, P.M.; Sobrero, A.; et al. Lapatinib in combination with capecitabine plus oxaliplatin in human epidermal growth factor receptor 2–positive advanced or metastatic gastric, esophageal, or gastroesophageal adenocarcinoma: TRIO-013/LOGiC—A randomized phase III trial. J. Clin. Oncol. 2016, 34, 443–451. [Google Scholar] [CrossRef] [Green Version]
  70. Tabernero, J.; Hoff, P.M.; Shen, L.; Ohtsu, A.; Shah, M.A.; Cheng, K.; Song, C.; Wu, H.; Eng-Wong, J.; Kim, K.; et al. Pertuzumab plus trastuzumab and chemotherapy for HER2-positive metastatic gastric or gastro-oesophageal junction cancer (JACOB): Final analysis of a double-blind, randomised, placebo-controlled phase 3 study. Lancet Oncol. 2018, 19, 1372–1384. [Google Scholar] [CrossRef]
  71. Satoh, T.; Xu, R.-H.; Chung, H.C.; Sun, G.-P.; Doi, T.; Xu, J.-M.; Tsuji, A.; Omuro, Y.; Li, J.; Wang, J.-W.; et al. Lapatinib plus paclitaxel versus paclitaxel alone in the second-line treatment of HER2-amplified advanced gastric cancer in Asian populations: TyTAN—A randomized, phase III study. J. Clin. Oncol. 2014, 32, 2039–2049. [Google Scholar] [CrossRef] [PubMed]
  72. Thuss-Patience, P.C.; Shah, M.A.; Ohtsu, A.; van Cutsem, E.; Ajani, J.A.; Castro, H.; Mansoor, W.; Chung, H.C.; Bodoky, G.; Shitara, K.; et al. Trastuzumab emtansine versus taxane use for previously treated HER2-positive locally advanced or metastatic gastric or gastro-oesophageal junction adenocarcinoma (GATSBY): An international randomised, open-label, adaptive, phase 2/3 study. Lancet Oncol. 2017, 18, 640–653. [Google Scholar] [CrossRef]
  73. Gambardella, V.; Gimeno-Valiente, F.; Tarazona, N.; Ciarpaglini, C.M.; Roda, D.; Fleitas, T.; Tolosa, P.; Cejalvo, J.M.; Huerta, M.; Roselló, S.J.C.C.R. NRF2 through RPS6 activation is related to anti-HER2 drug resistance in HER2-amplified gastric cancer. Clin. Cancer Res. 2019, 25, 1639–1649. [Google Scholar] [CrossRef] [PubMed]
  74. Yang, Y.; Tian, Z.; Guo, R.; Ren, F. Nrf2 Inhibitor, Brusatol in Combination with Trastuzumab Exerts Synergistic Antitumor Activity in HER2-Positive Cancers by Inhibiting Nrf2/HO-1 and HER2-AKT/ERK1/2 Pathways. Oxid. Med. Cell. Longev. 2020, 2020, 9867595. [Google Scholar] [CrossRef] [PubMed]
  75. Hu, X.-F.; Yao, J.; Gao, S.-G.; Wang, X.-S.; Peng, X.-Q.; Yang, Y.-T.; Feng, X.-S. Nrf2 overexpression predicts prognosis and 5-FU resistance in gastric cancer. Asian Pac. J. Cancer Prev. 2013, 14, 5231–5235. [Google Scholar] [CrossRef] [Green Version]
  76. Pouremamali, F.; Jeddi, F.; Samadi, N. Nrf2-ME-1 axis is associated with 5-FU resistance in gastric cancer cell line. Process. Biochem. 2020. [Google Scholar] [CrossRef]
  77. Wen, D.; Liu, D.; Tang, J.; Dong, L.; Liu, Y.; Tao, Z.; Wan, J.; Gao, D.; Wang, L.; Sun, H.; et al. Malic enzyme 1 induces epithelial–mesenchymal transition and indicates poor prognosis in hepatocellular carcinoma. Tumor Biol. 2015, 36, 6211–6221. [Google Scholar] [CrossRef]
  78. Bécouarn, Y.; Agostini, C.; Trufflandier, N.; Boulanger, V. Oxaliplatin: Available data in non-colorectal gastrointestinal malignancies. Crit. Rev. Oncol. Hematol. 2001, 40, 265–272. [Google Scholar] [CrossRef]
  79. Gruenberger, B.; Schueller, J.; Heubrandtner, U.; Wrba, F.; Tamandl, D.; Kaczirek, K.; Roka, R.; Freimann-Pircher, S.; Gruenberger, T. Cetuximab, gemcitabine, and oxaliplatin in patients with unresectable advanced or metastatic biliary tract cancer: A phase 2 study. Lancet Oncol. 2010, 11, 1142–1148. [Google Scholar] [CrossRef]
  80. Sun, W.; Sohal, D.; Haller, D.G.; Mykulowycz, K.; Rosen, M.; Soulen, M.C.; Caparro, M.; Teitelbaum, U.R.; Giantonio, B.; O’Dwyer, P.J.; et al. Phase 2 trial of bevacizumab, capecitabine, and oxaliplatin in treatment of advanced hepatocellular carcinoma. Cancer 2011, 117, 3187–3192. [Google Scholar] [CrossRef]
  81. Kelland, L. The resurgence of platinum-based cancer chemotherapy. Nat. Rev. Cancer 2007, 7, 573–584. [Google Scholar]
  82. Mishima, M.; Samimi, G.; Kondo, A.; Lin, X.; Howell, S.B. The cellular pharmacology of oxaliplatin resistance. Eur. J. Cancer 2002, 38, 1405–1412. [Google Scholar] [CrossRef]
  83. Gourdier, I.; del Rio, M.; Crabbé, L.; Candeil, L.; Copois, V.; Ychou, M.; Auffray, C.; Martineau, P.; Mechti, N.; Pommier, Y.; et al. Drug specific resistance to oxaliplatin is associated with apoptosis defect in a cellular model of colon carcinoma. FEBS Lett. 2002, 529, 232–236. [Google Scholar] [CrossRef]
  84. Ding, Z.-B.; Hui, B.; Shi, Y.-H.; Zhou, J.; Peng, Y.-F.; Gu, C.-Y.; Yang, H.; Shi, G.-M.; Ke, A.-W.; Wang, X.-Y. Autophagy activation in hepatocellular carcinoma contributes to the tolerance of oxaliplatin via reactive oxygen species modulation. Clin. Cancer Res. 2011, 17, 6229–6238. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Chen, C.C.; Chen, L.T.; Tsou, T.C.; Pan, W.Y.; Kuo, C.C.; Liu, J.F.; Yeh, S.C.; Tsai, F.Y.; Hsieh, H.P.; Chang, J.Y. Combined modalities of resistance in an oxaliplatin-resistant human gastric cancer cell line with enhanced sensitivity to 5-fluorouracil. Br. J. Cancer 2007, 97, 334–344. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Jeddi, F.; Soozangar, N.; Sadeghi, M.R.; Somi, M.H.; Shirmohamadi, M.; Eftekhar-Sadat, A.-T.; Samadi, N. Nrf2 overexpression is associated with P-glycoprotein upregulation in gastric cancer. Biomed. Pharmacother. 2018, 97, 286–292. [Google Scholar] [CrossRef] [PubMed]
  87. Qiu, L.; Ma, Z.; Li, X.; Deng, Y.; Duan, G.; Zhao, L.; Xu, X.; Xiao, L.; Liu, H.; Zhu, Z. DJ-1 is involved in the multidrug resistance of SGC7901 gastric cancer cells through PTEN/PI3K/Akt/Nrf2 pathway. Acta Biochim. Biophys. Sin. 2020, 52, 1202–1214. [Google Scholar] [CrossRef]
  88. Zhu, Z.-M.; Li, Z.-R.; Huang, Y.; Yu, H.-H.; Huang, X.-S.; Yan, Y.-F.; Shao, J.-H.; Chen, H.-P. DJ-1 is involved in the peritoneal metastasis of gastric cancer through activation of the Akt signaling pathway. Oncol. Rep. 2014, 31, 1489–1497. [Google Scholar] [CrossRef] [Green Version]
  89. Liu, H.-Y.; Duan, G.-L.; Xu, R.-Y.; Li, X.-R.; Xiao, L.; Zhao, L.; Ma, Z.-X.; Xu, X.-W.; Qiu, L.-J.; Zhu, Z.-M.; et al. DJ-1 overexpression confers the multidrug resistance phenotype to SGC7901 cells by upregulating P-gp and Bcl-2. Biochem. Biophys. Res. Commun. 2019, 519, 73–80. [Google Scholar] [CrossRef]
  90. Robinson, K.; Tiriveedhi, V. Perplexing role of P-glycoprotein in tumor microenvironment. Front. Oncol. 2020, 10, 265. [Google Scholar]
  91. Srivastava, R.K.; Sasaki, C.Y.; Hardwick, J.M.; Longo, D.L. Bcl-2–mediated drug resistance: Inhibition of apoptosis by blocking nuclear factor of activated T lymphocytes (NFAT)-induced Fas ligand transcription. J. Exp. Med. 1999, 190, 253–266. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Clements, C.M.; McNally, R.S.; Conti, B.J.; Mak, T.W.; Ting, J.P.Y. DJ-1, a cancer-and Parkinson’s disease-associated protein, stabilizes the antioxidant transcriptional master regulator Nrf2. Proc. Natl. Acad. Sci. USA 2006, 103, 15091–15096. [Google Scholar] [CrossRef] [Green Version]
  93. Malhotra, D.; Thimmulappa, R.; Navas-Acien, A.; Sandford, A.; Elliott, M.; Singh, A.; Chen, L.; Zhuang, X.; Hogg, J.; Pare, P. Decline in NRF2-regulated antioxidants in chronic obstructive pulmonary disease lungs due to loss of its positive regulator, DJ-1. Am. J. Respir. Crit. Care Med. 2008, 178, 592–604. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Bryan, H.K.; Olayanju, A.; Goldring, C.E.; Park, B.K. The Nrf2 cell defence pathway: Keap1-dependent and-independent mechanisms of regulation. Biochem. Pharmacol. 2013, 85, 705–717. [Google Scholar] [CrossRef] [Green Version]
  95. Lee, J.-S.; Surh, Y.-J. Nrf2 as a novel molecular target for chemoprevention. Cancer Lett. 2005, 224, 171–184. [Google Scholar] [CrossRef] [PubMed]
  96. Kim, R.H.; Peters, M.; Jang, Y.; Shi, W.; Pintilie, M.; Fletcher, G.C.; DeLuca, C.; Liepa, J.; Zhou, L.; Snow, B. DJ-1, a novel regulator of the tumor suppressor PTEN. Cancer Cell 2005, 7, 263–273. [Google Scholar] [CrossRef] [Green Version]
  97. Kim, Y.-C.; Kitaura, H.; Taira, T.; Iguchi-Ariga, S.M.M.; Ariga, H. Oxidation of DJ-1-dependent cell transformation through direct binding of DJ-1 to PTEN. Int. J. Oncol. 2009, 35, 1331–1341. [Google Scholar] [PubMed]
  98. Han, Z.; Hong, L.; Han, Y.; Wu, K.; Han, S.; Shen, H.; Li, C.; Yao, L.; Qiao, T.; Fan, D. Phospho Akt mediates multidrug resistance of gastric cancer cells through regulation of P-gp, Bcl-2 and Bax. J. Exp. Clin. Cancer Res. 2007, 26, 261–268. [Google Scholar]
  99. Michl, P.; Downward, J. Mechanisms of disease: PI3K/AKT signaling in gastrointestinal cancers. Z. Gastroenterol. 2005, 43, 1133–1139. [Google Scholar] [CrossRef] [PubMed]
  100. Shao, Z.-H.; Hoek, T.L.V.; Qin, Y.; Becker, L.B.; Schumacker, P.T.; Li, C.-Q.; Dey, L.; Barth, E.; Halpern, H.; Rosen, G.M.; et al. Baicalein attenuates oxidant stress in cardiomyocytes. Am. J. Physiol. Circ. Physiol. 2002, 282, H999–H1006. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Bie, B.; Sun, J.; Guo, Y.; Li, J.; Jiang, W.; Yang, J.; Huang, C.; Li, Z. Baicalein: A review of its anti-cancer effects and mechanisms in Hepatocellular Carcinoma. Biomed. Pharmacother. 2017, 93, 1285–1291. [Google Scholar] [CrossRef] [PubMed]
  102. Gao, Y.; Snyder, S.A.; Smith, J.N.; Chen, Y.C. Anticancer properties of baicalein: A review. Med. Chem. Res. 2016, 25, 1515–1523. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Li, P.; Hu, J.; Shi, B.; Tie, J. Baicalein enhanced cisplatin sensitivity of gastric cancer cells by inducing cell apoptosis and autophagy via Akt/mTOR and Nrf2/Keap 1 pathway. Biochem. Biophys. Res. Commun. 2020, 531, 320–327. [Google Scholar] [CrossRef] [PubMed]
  104. Fleischauer, A.T.; Poole, C.; Arab, L. Garlic consumption and cancer prevention: Meta-analyses of colorectal and stomach cancers. Am. J. Clin. Nutr. 2000, 72, 1047–1052. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Powolny, A.A.; Singh, S.V. Multitargeted prevention and therapy of cancer by diallyl trisulfide and related Allium vegetable-derived organosulfur compounds. Cancer Lett. 2008, 269, 305–314. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Chandra-Kuntal, K.; Lee, J.; Singh, S.V. Critical role for reactive oxygen species in apoptosis induction and cell migration inhibition by diallyl trisulfide, a cancer chemopreventive component of garlic. Breast Cancer Res. Treat. 2013, 138, 69–79. [Google Scholar] [CrossRef] [PubMed]
  107. Antony, M.L.; Singh, S.V. Molecular mechanisms and targets of cancer chemoprevention by garlic-derived bioactive compound diallyl trisulfide. Indian J. Exp. Biol. 2011, 49, 805. [Google Scholar]
  108. Lai, K.C.; Hsu, S.C.; Yang, J.S.; Yu, C.C.; Lein, J.C.; Chung, J.G. Diallyl trisulfide inhibits migration, invasion and angiogenesis of human colon cancer HsT-29 cells and umbilical vein endothelial cells, and suppresses murine xenograft tumour growth. J. Cell. Mol. Med. 2015, 19, 474–484. [Google Scholar] [CrossRef]
  109. Jiang, X.-Y.; Zhu, X.-S.; Xu, H.-Y.; Zhao, Z.-X.; Li, S.-Y.; Li, S.-Z.; Cai, J.-H.; Cao, J.-M. Diallyl trisulfide suppresses tumor growth through the attenuation of Nrf2/Akt and activation of p38/JNK and potentiates cisplatin efficacy in gastric cancer treatment. Acta Pharmacol. Sin. 2017, 38, 1048–1058. [Google Scholar] [CrossRef]
  110. Waterman, M.L. Lymphoid enhancer factor/T cell factor expression in colorectal cancer. Cancer Metastasis Rev. 2004, 23, 41–52. [Google Scholar] [CrossRef]
  111. Murphy, M.; Chatterjee, S.S.; Jain, S.; Katari, M.; DasGupta, R. TCF7L1 modulates colorectal cancer growth by inhibiting expression of the tumor-suppressor gene EPHB3. Sci. Rep. 2016, 6, 1–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Slyper, M.; Shahar, A.; Bar-Ziv, A.; Granit, R.Z.; Hamburger, T.; Maly, B.; Peretz, T.; Ben-Porath, I. Control of Breast Cancer Growth and Initiation by the Stem Cell–Associated Transcription Factor TCF3. Cancer Res. 2012, 72, 5613–5624. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Zhang, B.; Wu, J.; Cai, Y.; Luo, M.; Wang, B.; Gu, Y. TCF7L1 indicates prognosis and promotes proliferation through activation of Keap1/NRF2 in gastric cancer. Acta Biochim. Biophys. Sin. 2019, 51, 375–385. [Google Scholar] [CrossRef] [PubMed]
  114. Peng, L.; Duan, Y.; Zhang, Y.; Zhao, D.; Wen, Y.; Yao, J.; Da, M. Expression of Nrf2 and NQO1 in human gastric cancer and their clinical significance. Int. J. Clin. Exp. Pathol. 2016, 9, 1635–1643. [Google Scholar]
  115. Yang, W.-C.; Tsai, W.-C.; Lin, P.-M.; Yang, M.-Y.; Liu, Y.-C.; Chang, C.-S.; Yu, W.-H.; Lin, S.-F. Human BDH2, an anti-apoptosis factor, is a novel poor prognostic factor for de novo cytogenetically normal acute myeloid leukemia. J. Biomed. Sci. 2013, 20, 58. [Google Scholar] [CrossRef] [Green Version]
  116. Guo, K.; Lukacik, P.; Papagrigoriou, E.; Meier, M.; Lee, W.H.; Adamski, J.; Oppermann, U. Characterization of Human DHRS6, an Orphan Short Chain Dehydrogenase/Reductase Enzyme a novel, cytosolic type 2 R-β-hydroxybutyrate dehydrogenase. J. Biol. Chem. 2006, 281, 10291–10297. [Google Scholar] [CrossRef] [Green Version]
  117. Zang, W.; Wang, T.; Wang, Y.; Chen, X.; Du, Y.; Sun, Q.; Li, M.; Dong, Z.; Zhao, G. Knockdown of long non-coding RNA TP73-AS1 inhibits cell proliferation and induces apoptosis in esophageal squamous cell carcinoma. Oncotarget 2016, 7, 19960. [Google Scholar] [CrossRef] [Green Version]
Figure 1. The inhibitory effect of anti-HER2 drugs on Nrf2 inhibition and gastric cancer prevention.
Figure 1. The inhibitory effect of anti-HER2 drugs on Nrf2 inhibition and gastric cancer prevention.
Molecules 26 03157 g001
Figure 2. Role of Nrf2 pathways in gastric cancer.
Figure 2. Role of Nrf2 pathways in gastric cancer.
Molecules 26 03157 g002
Table 1. Modulation of gastric cancer through the Nrf2 pathway.
Table 1. Modulation of gastric cancer through the Nrf2 pathway.
AuthorsInvestigation AimsSubjectsPossible Roles of Nrf2
Gambardella et al. [73]Mechanisms of primary and acquired resistance to the combination of trastuzumab and platinum-based chemotherapyLapatinib- and trastuzumab-resistant clones derived from two different HER2-amplified gastric cancer cell linesNRF2 amplified the resistance to anti-HER2 drugs through the PI3K/AKT/mTOR/RPS6 pathway; RPS6 inhibition decreased NRF2 expression and restored sensitivity in HER2-amplified gastric cancer.
Yang et al. [74]Efficiency of combination chemotherapy with trastuzumab and brusatolHER2-positive SK-OV-3 and BT-474 cancer cellsBrusatol increased the antitumor activity of trastuzumab through the inhibition of the Nrf2/HO-1 and HER2-AKT/ERK1/2 signaling pathways.
Hu et al. [75]Nrf2 protein expression in gastric cancer specimens + association between Nrf2 expression and 5-FU resistanceSamples from GC patients with gastrectomy and from GC patients who received first-line combination chemotherapy of either A) docetaxel, cisplatin, and 5-FU or B) S-1 plus cisplatinNrf2 was an independent prognostic factor in GC and caused resistance to the chemotherapeutic drug 5-FU in GC cells.
Pouremamali et al. [76]The role of Nrf2 and its downstream target malic enzyme-1 (ME-1) in the resistance to 5-FU chemotherapyResistant MKN-45 (MKN-45/DR)
cell line
In resistant cells, decreased Nrf2 and ME-1 expression levels increased MDR1 mRNA levels; the inhibition of Nrf2 with luteolin and brusatol increased the effects on 5-FU-induced cytotoxicity.
Jeddi et al. [75]Association of Nrf2 and MDR1/P-gp in GC patientsEndoscopic biopsy samples from GC patientsHigher Nrf2 expression in GC patients compared with non-GC individuals; the induction of P-gp was associated with Nrf2 overexpression; there was correlation between Nrf2 overexpression and tumor size, histological grade, lymph node, and distant metastasis; P-gp up-regulation was associated with the histological grade and tumor size; the inhibition of Nrf2 expression improved the efficacy of chemotherapeutic agents for GC patients by the downregulation of P-gp expression.
Chen et al. [85]Mechanisms of oxaliplatin resistanceHuman gastric carcinoma cell line TSGH-S3 (S3)The activation of Nrf2/AKR1C axis contributed to oxaliplatin resistance as the suppression of Nrf2 expression decreased the levels of AKR1C1, AKR1C2, and AKR1C3 and reversed oxaliplatin resistance in S3 cells.
Qiu et al. [86]Mechanisms of DJ-1-induced multidrug resistance (MDR)SGC7901 + MDR variant SGC7901/VCR gastric cancer cellsDJ-1 mediated the development of MDR in SGC7901 gastric cancer cells through inhibiting PTEN, activating the PI3k/Akt pathway and consequently resulting in Nrf2 activation, thereby inducing Nrf2-dependent P-glycoprotein (P-gp) and Bcl-2 expressions.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Farkhondeh, T.; Pourbagher-Shahri, A.M.; Azimi-Nezhad, M.; Forouzanfar, F.; Brockmueller, A.; Ashrafizadeh, M.; Talebi, M.; Shakibaei, M.; Samarghandian, S. Roles of Nrf2 in Gastric Cancer: Targeting for Therapeutic Strategies. Molecules 2021, 26, 3157. https://doi.org/10.3390/molecules26113157

AMA Style

Farkhondeh T, Pourbagher-Shahri AM, Azimi-Nezhad M, Forouzanfar F, Brockmueller A, Ashrafizadeh M, Talebi M, Shakibaei M, Samarghandian S. Roles of Nrf2 in Gastric Cancer: Targeting for Therapeutic Strategies. Molecules. 2021; 26(11):3157. https://doi.org/10.3390/molecules26113157

Chicago/Turabian Style

Farkhondeh, Tahereh, Ali Mohammad Pourbagher-Shahri, Mohsen Azimi-Nezhad, Fatemeh Forouzanfar, Aranka Brockmueller, Milad Ashrafizadeh, Marjan Talebi, Mehdi Shakibaei, and Saeed Samarghandian. 2021. "Roles of Nrf2 in Gastric Cancer: Targeting for Therapeutic Strategies" Molecules 26, no. 11: 3157. https://doi.org/10.3390/molecules26113157

Article Metrics

Back to TopTop