key: cord-0810939-uw1v03jq authors: Wang, Jingya; Yang, Jin; Cao, Mingnan; Zhao, Zhigang; Cao, Baoshan; Yu, Siwang title: The potential roles of Nrf2/Keap1 signaling in anticancer drug interactions date: 2021-05-04 journal: Curr Res Pharmacol Drug Discov DOI: 10.1016/j.crphar.2021.100028 sha: e9d1aa8fe82a0af1797fd9ec1c1388574b93079d doc_id: 810939 cord_uid: uw1v03jq Nuclear factor (erythroid-derived 2)-related factor 2 (Nrf2), together with its suppressive binding partner Kelch-like ECH-associated protein 1 (Keap1), regulates cellular antioxidant response and drug metabolism. The roles of Nrf2/Keap1 signaling in the pathology of many diseases have been extensively investigated, and small molecules targeting Nrf2/Keap1 signaling have been developed to prevent or treat diseases such as multiple sclerosis, chronic kidney disease and cancer. Notably, Nrf2 plays dual roles in cancer development and treatment. Activation of Nrf2/Keap1 signaling in cancer cells has been reported to promote cancer progression and result in therapy resistance. Since cancer patients are often suffering comorbidities of other chronic diseases, anticancer drugs could be co-administrated with other drugs and herbs. Nrf2/Keap1 signaling modulators, especially activators, are common in drugs, herbs and dietary ingredients, even they are developed for other targets. Therefore, drug-drug or herb-drug interactions due to modulation of Nrf2/Keap1 signaling should be considered in cancer therapies. Here we briefly summarize basic biochemistry and physiology functions of Nrf2/Keap1 signaling, Nrf2/Keap1 signaling modulators that cancer patients could be exposed to, and anticancer drugs that are sensitive to Nrf2/Keap1 signaling, aiming to call attention to the potential drug-drug or herb-drug interactions between anticancer drugs and these Nrf2/Keap1 signaling modulators. When two or more drugs are concomitantly administrated or sequentially within a short period, there are risks of drug-drug interactions (DDIs). DDIs can be grouped in two classes, namely pharmacokinetic and pharmacodynamic DDIs (Beijnen and Schellens, 2004) . Pharmacokinetic DDIs refer to influences on the absorption, distribution, metabolism, or elimination of a drug. Drug metabolizing enzymes or transporters are often involved in these processes (Gay et al., 2017) . Pharmacodynamic DDIs occur when two or more concomitantly used drugs have complementary, similar or competitive mechanisms of action, which lead to synergistic, additive or antagonistic effect, respectively (Niu et al., 2019) . Since cancer is a typical age-related disease, cancer patients are frequently suffering from comorbidities such as diabetes, cardiovascular diseases, neuro-degenerative diseases, and infectious diseases (Sarfati et al., 2016) . Drugs to treat these comorbidities are often co-administrated with anticancer drugs. Indeed, polypharmacy (with five or more concurrent medications) is common among elder cancer patients (Mohamed et al., 2020) . On the other hand, anticancer drugs are one of the most toxic classes of medications with narrow therapeutic window, and they are generally prescribed in combinations to maximize therapeutic efficacy and minimize adverse effects. Furthermore, many cancer patients turn to complementary and alternative therapies such as traditional Chinese medicine or other ethnopharmacy, as well as certain functional foods or dietary supplements (Goss et al., 2014) . All the above contribute to the high risk of drug-drug or herb-drug interactions in cancer patients. These interactions may result in therapeutic failure or serious adverse events and exert important impacts on the prognosis, mortality and life quality of cancer patients (Ismail et al., 2020; Sharma et al., 2019; Jermini et al., 2019) . Nuclear factor erythroid-derived 2-related factor 2 (Nrf2, gene symbol NFE2L2), together with its suppressive binding partner, Kelch-like ECH-associated protein 1 (Keap1), regulates the expression of many drug metabolizing and cyto-protective genes, including those involved in redox homeostasis, drug metabolism and transport, mitochondrial functions and DNA repair (Yamamoto et al., 2018; Bai et al., 2016) . Nrf2/Keap1 signaling plays important and complicated roles in the initiation and progression of cancer, and has been implied in both the resistance to and the toxicities of anticancer drugs (Rojo de la Sporn and Liby, 2012) . Thus, Nrf2/Keap1 signaling may play important roles in both pharmacokinetic and pharmacodynamic anticancer DDIs. Nevertheless, its potential roles in drug interaction during cancer treatment attracted little attention. Here we briefly summarize the involvement of Nrf2/Keap1 signaling in cancer treatment, and call attention to potential drug-drug or herb-drug interactions of anticancer drugs due to modulation of Nrf2/Keap1 signaling. Nrf2 is a cap'n'collar (CNC) basic-leucine zipper (bZIP) transcription factor which regulates the transcriptional response of cells to oxidative stress and electrophilic substances. It has been more than 20 years since the first report of Nrf2-mediated induction of phase II drug metabolizing enzymes (Itoh et al., 1997) . Then the molecular regulation and physiological functions of Nrf2 have been extensively investigated and reviewed (Yamamoto et al., 2018; Baird and Yamamoto, 2020; Tonelli et al., 2018) . Therefore, it will only be briefly summarized in the present review. Under basal conditions, Nrf2 is bound to Keap1 through the DLG and ETGE motifs and located in cytoplasm at a low level with a short half-life time. Keap1 is an E3 ubiquitin ligase adaptor which targets Nrf2 for rapid ubiquitination and proteasomal degradation. When the cells are subjected to oxidative or electrophilic substances including reactive oxygen/nitrogen species (ROS/RNS), the highly reactive cysteine residues on Keap1 protein are directly modified, disrupting the binding between Keap1 and Nrf2. The released Nrf2 accumulates in the nucleus and forms heterodimers with small musculoaponeurotic fibrosarcoma oncogene homologue (Maf) proteins, then the heterodimers bind to specific antioxidant response elements (AREs) in the regulatory regions of Nrf2 target genes and boost their transcription. In addition to the cysteine modification-and Keap1-dependent mechanisms, Nrf2 activity could be regulated by β-TrCP-, HRD1-or p62/SQSTM1-dependent protein degradation (Rojo de la , or by epigenetic mechanisms (Guo et al., 2015) . Post-translational modifications of Nrf2 such as acetylation and phosphorylation by various kinases also modulate Nrf2 transcriptional activity under some conditions (Baird and Yamamoto, 2020) . The regulatory network of Nrf2/Keap1 signaling is schematically depicted in Fig. 1 . In response to oxidative/electrophilic stresses, Nrf2 is known to regulate the expression of more than 500 cytoprotective genes and this number is likely to increase in the future (Tonelli et al., 2018) . These genes are profoundly involved in cellular redox homeostasis (Yamamoto et al., 2018) , drug detoxification (Bai et al., 2016) , energy metabolism (Vomhof-Dekrey and PickloSr, 2012) and other stress responses such as DNA damage response Kim et al., 2012) . Specifically, Nrf2 is a major transcription factor regulating the expression of phase II drug metabolizing enzymes that are important for conjugation and detoxification of many drugs or carcinogens. Nrf2 protects the cells against a wide range of endogenous and exogenous insults, helps to maintain cellular homeostasis and offers the cells advantages to survive Fig. 1 . Regulation of Nrf2/Keap1 signaling by drugs, herbal phytochemicals and dietary factors and its potential roles in anticancer drug-drug and drug-herb interactions. Under basal conditions, Nrf2 is bound to Keap1 through the DLG and ETGE motif, ubiquitinated by Cul3-E3 ubiquitin ligase and subjected for rapid proteasomal degradation. Upon oxidative or electrophilic stresses, the reactive cysteine residues on Keap1 protein are modified by these stressors including ROS and Nrf2 is released and translocated into nucleus, at where it forms heterodimers with small Maf proteins and boosts the transcription of ARE-driven genes, including antioxidant proteins, drug metabolizing enzymes and transporters. These genes are well known players in anticancer drug-drug and drug-herb interactions. In addition to the above core regulatory mechanism, Nrf2/Keap1 signaling could be modulated through other Keap1-independent mechanisms. Nrf2 protein can be phosphorylated and activated by MAPKs, AMPK and PI3K/Akt, while GSK3β phosphorylation promotes the exportation of Nrf2 from nucleus. The transcription of Nrf2 mRNA is regulated by DNA methylation and histone acetylation, the stability and translation of Nrf2 mRNA can be inhibited by certain compounds such as brusatol, camptothecin, luteolin and wogonin. under unfavorable environments. Some typical drug metabolizing and cytoprotective genes regulated by Nrf2/Keap1 signaling and potentialyinvolved in the DDIs of anticancer drugs are summarized in Table 1 (Hayes and Dinkova-Kostova, 2014; Shen and Kong, 2009; Hirotsu et al., 2012; Ma, 2013) . Nrf2-deficient mice were more susceptible to oxidative injuries and chemical carcinogenesis, while Nrf2 activators protect against them (Sinha et al., 2013; Slocum and Kensler, 2011) . Therefore, Nrf2 activators were initially considered as promising chemopreventive agents against various carcinogenesis (Hayes et al., 2010) . However, Nrf2 was soon found to be aberrantly activated in many tumors by oncogenes or Keap1 mutations (DeNicola et al., 2011) . Neoplastic cells can "hijack" Nrf2/Keap1 signaling to reprogram cellular metabolism, promote tumor survival and proliferation, drive tumorigenesis and result in chemo-and radio-resistance (Rojo de la Sporn and Liby, 2012) . Thus, Nrf2/Keap1 signaling inhibitors as anticancer drugs have attracted increasing academic and industrial interests (Telkoparan-Akillilar et al., 2019; Zhu et al., 2016) . To date, the roles of Nrf2/Keap1 signaling in cancer development and treatment are still controversial and highly sensitive to the context. As a multifaceted transcription factor, Nrf2 has also been implied in the pathogenesis of many other chronic diseases including diabetes, respiratory disease, cardiovascular disease, inflammatory disease, neurodegenerative disease and autoimmune disease (Michalickova et al., 2020; Cuadrado et al., 2019) . Unlike the case in cancer, the protective role of Nrf2 is generally favored for prevention or treatment of these diseases. Indeed, the first US FDA-approved Nrf2 activator, dimethyl fumarate (DMF), has been successfully marketed as multiple sclerosis (MS) treatment with impressive performance, and more Nrf2 activators are in phase II/III clinical trials and are expected to enter the market within a few years (Michalickova et al., 2020; Cuadrado et al., 2019) . Furthermore, Nrf2 has been reported to be involved in pathogen infection, and activation of Nrf2 has been proposed as a host-directed therapeutic strategy to treat infectious diseases including COVID-19 (Deramaudt et al., 2013; Olagnier et al., 2020) . 3. Nrf2/Keap1 signaling modulators that cancer patients could be exposed to As discussed above, Nrf2 is an attractive drug target for treating many chronic and even infectious diseases, and several Nrf2/Keap1 signaling modulators are under active development or even been marketed (Telkoparan-Akillilar et al., 2019; Cuadrado et al., 2019; Panieri et al., 2020) . On the other hand, some other drugs or drug candidates, though initially were developed for targets other than Nrf2/Keap1, have also been found to modulate Nrf2/Keap1 signaling. These drugs, no matter anticancer reagents or not, could be concurrently administrated to cancer patients due to comorbidities. Moreover, plants are the most abundant resource of Nrf2 activators, and cancer patients could be exposed to these phytochemicals through ingestion of herbs and dietary plants as complementary and alternative therapies. Some typical Nrf2/Keap1 signaling modulators including both activators and inhibitors that could be co-administrated to cancer patients are summarized in Table 2 , and the regulatory mechanisms of Nrf2/Keap1 signaling by these drugs, herbal phytochemicals and dietary factors, and their potential roles in anticancer drug-drug and drug-herb interactions are presented in Fig. 1. A few examples will be discussed in more details below. Nrf2 can be activated through Keap1-dependent or independent mechanisms (Baird and Yamamoto, 2020) . Most Keap1-dependent activators are electrophilic compounds that covalently modifying the sulfhydryl group of reactive cysteine residues in Keap1 protein, while some others can modulate the protein-protein interactions in Keap1-Nrf2 or Keap1-Cul3 complexes, and finally stabilize Nrf2 protein (Yamamoto et al., 2018; Baird and Yamamoto, 2020; Tonelli et al., 2018) . Some protein-protein interaction inhibitors of Keap1-Nrf2, like tetrahydroisoquinoline, thiopyrimidine and naphthalene have been discovered using virtual screening methods, but their therapeutic potential need to be further investigated (Lu et al., 2016 ; Robledinos-Ant on et al., 2019). Nrf2 could also be activated through post-translational modifications independent of Keap1. For example, Nrf2 can be phosphorylated and activated by kinases such as AMP-activated protein kinase (AMPK), protein kinase C (PKC), phosphatidylinositol-3-kinase/protein kinase B (PI3K/PKB or PI3K/Akt) and mitogen-activated protein kinases (MAPKs) (Xu et al., 2006) . The sulfhydryl-dependent Keap1 modifications are often sensitive to other sulfhydryl groups abundant in cellular context such as GSH and thioredoxin, while the selectivity of Keap1-independent activation is lower than Keap1-dependent activation. DMF (commercial name Tecfidera®) is the first Nrf2-targeting drug approved by FDA in 2013 to treat multiple sclerosis (MS). Before that DMF is used to treat psoriasis (approved in Germany as Fumaderm®) (Kourakis et al., 2020) . DMF and its metabolite monomethyl fumarate (MMF) modulate inflammatory and immune responses through both Nrf2-dependent and independent mechanisms, especially in central nervous system (Michalickova et al., 2020; Schulze-Topphoff et al., 2016; Gillard et al., 2015; Zaro et al., 2019) . And multiple molecular targets of DMF have been proposed and identified (Piroli et (Iniaghe et al., 2015; Kalinin et al., 2013) . On the other hand, higher concentrations of DMF (>25 mmol/L) may inhibit Nrf2 and induces oxidative stresses in several cancer cell lines (Saidu et al., 2017) . There are at least two publications investigated the potential drugdrug interactions between DMF and other drugs. In a clinical investigation , the potential DDI between DMF and an oral contraceptive (OC, norgestimate/ethinyl estradiol) was tested in healthy women, and no impact of DMF on the DM/PK of OC was identified. Another publication (Aubets et al., 2019) examined the direct impact of DMF on cytochrome P450 (CYP) enzymes and P-glycoprotein (P-gp) activities in vitro, and the results indicate that DMF is unlikely to interfere with CYP or P-gp activities at clinically relevant concentrations. However, it is well recognized that DMF activates Nrf2 which regulates the expression of phase II metabolizing enzymes, but this issue was not considered. Moreover, the potential interactions between DMF and anticancer drugs were not investigated. It is noteworthy that psoriasis is associated with increased risks of cancer, especially non-melanoma skin cancer, lymphoma, and lung cancer (Loft et al., 2020) . Although the prevalence of cancer in MS patients is similar to or less than that in general populations, the frequency of cancer diagnosis increased over time among MS patients but not in controls (Zecca et al., 2020; Ghajarzadeh et al., 2020) . There are unneglectable chances for cancer patients to be exposed to DMF, the potential DDIs between DMF and anticancer drugs deserve attention. Indeed, DMF has been tested as anticancer agents in vitro, and has been shown to interact with proteasome inhibitors (Booth et al., 2014) or synergize with ruxolitinib (Tavallai et al., 2016) . Moreover, several clinical trials are undergoing to test different formulations of DMF and fumarate derivatives in cancer therapies, like cutaneous T cell lymphoma, adult brain glioblastoma and chronic lymphocytic leukemia (Nicolay et al., 2016; Al-Jaderi and Maghazachi, 2016) . The outcomes of these trials may help to further clarify the potential DDIs of DMF with anticancer drugs. Bardoxolone methyl (CDDO-Me or RTA402) is a synthetic potent Nrf2-activating triterpenoid initially developed for cancer prevention and treatment (Hong et al., 2012) . Similar to the situation of DMF, multiple molecular targets including PPARγ, PML-RARα and IKKβ have been proposed for bardoxolone, but Nrf2/Keap1 is recognized as its major target. After the phase I clinical trial in lymphoma patients, bardoxolone was re-purposed for end-stage renal disease treatment, but unfortunately failed in phase III trial due to safety issues (de Zeeuw et al., 2013) . Currently bardoxolone is in phase II/III clinical trials for pulmonary hypertension, polycystic kidney disease, type 2 diabetes mellitus (T2DM), chronic kidney disease and most recently COVID-19 (NCT02657356, NCT03068130, NCT03366337, NCT03550443, NCT04494646, respectively). In addition, the anticancer activities of bardoxolone and its derivatives are under experimental or pre-clinical Table 2 Nrf2/Keap1 signaling modulators that cancer patients could be exposed to. investigations. Obviously, bardoxolone has a good chance to interact with other anticancer drugs in clinical trials or clinical use if approved by FDA, and the potential DDIs between bardoxolone and other anticancer drugs due to activation of Nrf2/Keap1 signaling must be taken into account. Some other FDA-approved drugs, including anticancer drugs, have been reported to activate Nrf2/Keap1 signaling. An ARE-luciferase reporter-based screening identified cisplatin, carmustine and acrolein (an active metabolite of cyclophosphamide) as Nrf2 activators (Wang et al., 2006) . Oxaliplatin, a third-generation platinum-based drug used to treat colorectal and ovarian cancer, activates Nrf2/Keap1 signaling conferring protection against the cytotoxicity of anticancer drugs Chian et al., 2014) . Statins are the most prescribed lipid-lowering drugs to prevent cardiovascular diseases. Some statins such as simvastatin, lovastatin and fluvastatin can activate Nrf2 through extracellular signal-regulated kinase (ERK) or PI3K/Akt pathway (Niedzielski et al., 2020; Jang et al., 2016; Cuadrado et al., 2018) . Second line hypoglycemic dipeptidyl peptidase-4 inhibitors (DPP-4i) saxagliptin and sitagliptin have been reported to upregulate Nrf2 signaling and promoted tumor metastasis . The mechanisms of metformin to treat T2DM are not completely understood, and it has been found to activate Nrf2/Keap1 signaling in neurons, liver and skeletal muscle in an AMPK-dependent manner (Tschop et al., 2016; Ashabi et al., 2015) . However, some other reports proposed metformin as a Nrf2 inhibitor and will be discussed later. Many herbal or dietary ingredients are strong Nrf2 activators. Sulforaphane (SFN), an isothiocyanate found in cruciferous vegetables, has been reported as a very potent Nrf2 activator through direct modification of Keap1 or by inhibiting HDAC. SFN has been well documented to prevent the initiation and development of several cancers and has been recommended as a chemopreventive agent against various cancers (Jiang et al., 2018) . Several clinical trials have been carried out to test the therapeutic efficacy of SFN on breast cancer (Atwell et al., 2015) [NCT03934905], prostate cancer (Traka et al., 2019) , melanoma (Tahata et al., 2018) , and other diseases such as schizophrenia [NCT04521868], autism [NCT02654743] and cystic fibrosis [NCT01315665]. On the other hand, SFN modulates the enzymatic drug metabolizing system and interacts with drugs (Lubelska et al., 2012) , and has been found to increase tumor growth once the tumor is already formed . Silymarin has been widely used to treat liver diseases, and activation of Nrf2/Keap1 signaling is one of its major mechanisms of action (Xie et al., 2019) . Epigallocatechin-3-gallate (EGCG) from green tea extract, curcumin from turmeric, resveratrol from grape seeds, and many other phytochemicals, are known as Nrf2 activators potentially involved in cancer care (Zhu et al., 2020; T et al., 2020) . According to epidemiological, laboratory and clinical studies, these phytochemicals have been regarded as cancer-preventive or curative agents, with activation of Nrf2 as one of their mechanisms of action. Nevertheless, they could also interact with anticancer therapies due to activation of Nrf2/Keap1 signaling. As the "dark side" of Nrf2 in cancer progression and therapy resistance is being revealed, Nrf2 inhibitors receive more and more interests for their potential applications in reversing chemoresistance or suppressing tumor growth. Though Nrf2 inhibitors are relatively rare and the underlying mechanisms are elusive, several promising Nrf2 inhibitors have been identified and some of them have progressed to clinical development. Notably, a few approved drugs have been found to inhibit Nrf2/Keap1 signaling, thus potential DDIs between these drugs and anticancer drugs should be considered in cancer treatment. Brusatol, a quassinoid from Brucea javanica seeds, might be the best characterized Nrf2 inhibitor to the date. It rapidly decreases Nrf2 protein levels in a wide array of cancer cells and sensitizes the cells to chemotherapy or radiotherapy (Ren et al., 2011) . Brusatol is located to the endoplasmic reticulum and inhibits both cap-dependent and cap-independent protein translation, especially those short-lived proteins, like Nrf2 (Harder et al., 2017) . Inhibition of Nrf2 by brusatol in hepatocytes also sensitized the cells to chemical toxicity, highlighting the involvement of Nrf2 inhibition in evaluation of potential adverse events resulted from non-target cells (Olayanju et al., 2015) . Several other natural compounds, such as apigenin, chrysin, cryptotanshinone, luteolin, trigonelline, triptolide, wogonin, etc., have been reported to inhibit Nrf2/Keap1 signaling in cancer cells at different concentrations through various mechanisms like accelerating mRNA degradation, downregulating PI3K/Akt and ERK pathways to decrease Nrf2 phosphorylation, and reducing nuclear import of Nrf2, etc. (reviewed in (Telkoparan-Akillilar et al., 2019; Zhu et al., 2016; Panieri et al., 2020) ). These compounds, though none have progressed to clinical development yet, have attracted significant research interests for their potential applications in cancer therapy. Several prescription drugs and vitamins have been reported to inhibit Nrf2/Keap1 signaling in cancer cells. Camptothecin, a chemotherapeutic DNA topoisomerase inhibitor, can suppress Nrf2 activity in hepatocellular carcinoma cells and sensitize them to chemotherapies (Chen et al., 2017) . Camptothecin is an anticancer drug approved to treat a broad spectrum of cancers, inhibition of Nrf2 could be one of the mechanisms underlying the synergistic effects between camptothecin and other therapeutic agents. Metformin has also been found to inhibit Nrf2 and reverse chemoresistance in various cancer cell lines Bai et al., 2018) . More importantly, metformin diminished the deleterious impacts of Nrf2/Keap1 signaling on the prognosis of breast cancer patients with T2DM (Urpilainen et al., 2019) . All trans-retinoic acid (ATRA) inhibits Nrf2 activity in cancer cells via the crosstalk between RARalpha and Nrf2, but not in normal tissues (Wang et al., 2007) . Ascorbic acid was reported to sensitize cervical carcinoma and leukemia cells to anticancer drugs while protecting normal cells and tissues, and Nrf2/Keap1 signaling was involved in both effects (Wu et al., 2020; Tarumoto et al., 2004) . Identification of more Nrf2/Keap1 signaling inhibitors from approved drugs is an attractive strategy to improve therapeutic efficacy. Interestingly, many of these Nrf2/Keap1 signaling inhibitors, including natural compounds such as apigenin, chrysin and luteolin, drugs like metformin and ATRA, have also been reported to activate Nrf2 at different concentrations or in different tissues or cell lines. Such phenomena underpin the importance of clinically relevant dosages when talking about potential DDIs related to Nrf2/Keap1 signaling. The tissuespecific concentrations of specified drugs to activate or inhibit Nrf2/ Keap1 signaling must be clinically relevant for patients, especially most of these drugs are not designed to target Nrf2/Keap1 signaling. ATRA at 1 μM was found to suppress tBHQ, β-naphthoflavone, or SFN-induced activation of Nrf2 in cancer cells (Wang et al., 2007) , but itself was also reported to activate Nrf2 signaling in a dose-dependent manner in vitro and in vivo (Tan et al., 2008) . The clinical dose of metformin as an oral hypoglycemic drug ranges between 500 and 2000 mg/d, which translates into about 60-260 mg/kg in mice, and this concentration has been reported to activate Nrf2 in mice (Wu et al., 2018; Prasad et al., 2017) . Meanwhile, the drugs designed to target Nrf2/Keap1 signaling such as DMF or bardoxolone at their effective doses will activate Nrf2/Keap1 signaling, and supposedly the subsequent expression of drug metabolizing enzymes and antioxidant proteins. Cancer patients could also be exposed to some Nrf2/Keap1-targeting agents such as SFN through dietary supplements or herbs. SFN has not been formally approved for any medical usage, but broccoli sprouts or broccoli seed extract that can provide SFN are available as food additives or dietary supplements in many countries, and the allowed dosage is enough to activate Nrf2/Keap1 signaling in vivo. Thus, it is expected that these drugs could interact with anticancer drugs through modulation of Nrf2/Keap1 signaling at a clinically relevant dosage. In general, extra caution should be paid to the potential DDIs between these Nrf2 modulators and anticancer drugs, especially those are sensitive to Nrf2 modulators. Activation of Nrf2/Keap1 signaling due to mutations in NFE2L2/ KEAP1 genes or aberrant upstream signaling in cancer is well known to promote cancer progression and lead to resistance to anticancer drugs through both pharmacokinetic and pharmacodynamic mechanisms. These mechanisms include upregulation of antioxidant and detoxification enzymes, activation of drug efflux transporters, inhibition of druginduced cellular apoptotic responses and induction of Nrf2-dependent proteasome activity (Rojo de la Panieri et al., 2020) . On the other hand, adverse events resulted from anticancer drugs could also be affected by Nrf2/Keap1 signaling in normal tissues (Yarmohammadi et al., 2020; Fang et al., 2020) . Due to the broad participation of Nrf2/Keap1 signaling in the metabolism of and cellular defense against anticancer drugs in both cancerous and normal cells, it is expected that more and more anticancer drugs would be identified to be sensitive to Nrf2/Keap1 signaling. A plethora of anticancer drugs have already been confirmed to be sensitive to Nrf2/Keap1 signaling modulators, and some examples are summarized in Table 3 and discussed below. The aberrant activation of Nrf2 is an important reason accounting for the chemoresistance of platinum-based drugs. For example, Cisplatin can be detoxified by coordination to GSH and transported via MRP2, both are regulated by Nrf2/Keap1 signaling (Homma et al., 2009) . A549 human lung adenoma cells, with somatic mutation at the Keap1-Kelch domain, is resistant to cisplatin treatment; while NCI H231 human lung cancer cells, with a mutation at the Keap1-LVR domain which increases Nrf2 degradation, is sensitive to cisplatin (Silva et al., 2019) . In Keap1-deficient non-small cell lung cancer, constitutive Nrf2 activation conferred resistance to carboplatin (Singh et al., 2016) . Oxaliplatin is sensitive to Nrf2 activity, and Nrf2/Keap1 signaling inhibitor could sensitize oxaliplatin-resistant cancer cells to chemotherapeutic drugs Chian et al., 2014) . Nrf2 over-activation by Keap1 mutation reduced the sensitivity of biliary tract cancer cells to 5-fluorouracil (5-FU) (Shibata et al., 2008) . Similar effect was seen in iASPP-overexpression/knockdown cancer cells, which is an antioxidant factor competing with Nrf2 for Keap1 binding (Ge et al., 2017) . A clinical investigation on gastric cancer showed that Nrf2 could be a predictive marker for 5-FU resistance and prognosis (Hu et al., 2013) . Nrf2 inhibition enhances the therapeutic efficacy of paclitaxel and docetaxel in ovarian cancer, non-small cell lung cancer and endometrial cancer (Chen et al., 2019; Manandhar et al., 2012; Jiang et al., 2010) . Acquired resistance to anthracycline antibiotics doxorubicin is accompanied by Nrf2 over activation in various cancer cells, and inhibition of Nrf2 re-sensitized resistant cells to these drugs (Shim et al., 2009 ). In addition, the resistances to gemcitabine, temozolomide, bortezomib, arsenic trioxide, and some other chemotherapeutic drugs have all been reported to be regulated by Nrf2/Keap1 signaling (Wang et al., 2008) . Along with the increased prescription of targeted kinase/enzyme inhibitors such as tyrosine kinase inhibitors (TKIs), the potential roles of Nrf2/Keap1 signaling in targeted therapies are increasingly reported. Overexpression of Nrf2 target genes by acquired Keap1 mutation mediates resistance to gefitinib and cross-resistance to afatinib and osimertinib (Park et al., 2018) . Nrf2 regulates the sensitivity of ovarian cancer cells to lapatinib and erlotinib through HER1 signaling (Kankia et al., 2017) . Activation of Nrf2 signaling by hemin reduces cellular sensitivity to imatinib (Tarumoto et al., 2004; Nagai et al., 2008) . Oxidative stress and mitochondrial damage are the most common mechanisms for the toxicity of chemotherapy drugs. Given the crucial roles of Nrf2 in oxidative stress, mitochondrial damage, drug metabolism and transport, it is expectable that the toxicities of anticancer drugs to normal cells are also regulated by Nrf2/Keap1 signaling, though less noticed and investigated than drug resistance. The application of doxorubicin has been limited by its acute and chronic cardiotoxicity, with oxidative stress as the primary cause of this cardiotoxicity (Yarmohammadi et al., 2020) . Nrf2 deficiency aggravated doxorubicin-induced oxidative stress and abnormal autophagic activities . Cisplatin can induce Nrf2-dependent cytoprotective genes in wild type mice kidney, but this induction is blunted in Nrf2-deficient mice, accompanied by severe oxidative damage and inflammatory reaction in the kidney (Aleksunes et al., 2010) . Nrf2-deficient mice are more susceptible to oxaliplatin-induced peripheral neuropathy and hepatotoxicity (X.J. He et al., 2017) . The hematological toxicity of cyclophosphamide and its active metabolite acrolein is deteriorated in Nrf2 deficient mice (Que et al., 2016) . Nrf2 activators have been well-recognized to protect normal cells against electrophilic insults, thus it is reasonable to use Nrf2 activators to protect against the toxicity of anticancer drugs. A wide range of Nrf2 activators including natural compounds such as sulforaphane (Yarmohammadi et al., 2020; Bose et al., 2018) and silymarin (Ra skovi c et al., 2011) , and synthetic drugs such as bardoxolone (Atilano-Roque et al., 2016) and DMF (Miyagi et al., 2019) have been used to reduce the toxicities of chemotherapeutic drugs. Some of these chemo-protectants, such as SFN against doxorubicin-induced cardiotoxicity, have progressed to clinical trials [NCT03934905]. The important roles of Nrf2/Keap1 signaling in the prevention and treatment of cancers have been extensively demonstrated and recognized. It is well accepted that the resistance to and toxicities of certain Silva et al. (2019) cancer therapies are profoundly impacted by Nrf2/Keap1 signaling, either good or bad. Therefore, the potential roles of Nrf2/Keap1 signaling in drug-drug, herb-drug or diet-drug interactions of anticancer drugs cannot be neglected. The occurrence of these interactions is an integrated result of dosage, tissue distribution, metabolism and excretion, as well as activities other than modulating Nrf2/Keap1 signaling, of the drugs and herbs in consideration. These interactions could be exploited to enhance therapeutic efficacy or reduce toxicity, which is the major focus of current literatures. For instance, Nrf2 inhibitors have been developed to reverse the chemoresistance of cancer cells, while Nrf2 activators have been examined as potential chemo-protectants to reduce the toxicities or side effects of chemotherapies. On the other hand, the undesirable interactions of anticancer drugs and Nrf2/Keap1 signaling modulators, such as increased chemoresistance or deteriorated toxicities, are far less reported, possibly due to a publication bias. However, it does not mean such interactions are scarce or risk-free. As a matter of fact, Nrf2/Keap1 signaling modulators, especially activators, are widely presented in drugs in clinical application or development, herb medicines and diets. Indeed, many herbs that have been reported to interact with anticancer drugs have also been reported to activate Nrf2/Keap1 signaling. On the other hand, some of these herbs or herb ingredients have also been reported to be cancer preventive or curative under certain situations, and cancer patients could be exposed to these substances when concomitantly taking anticancer drugs. Therefore, greater attention should be paid to the potential roles of Nrf2/Keap1 signaling in interactions with anticancer drugs, in either clinical practices or drug development. The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper. Utilization of dimethyl fumarate and related molecules for treatment of multiple sclerosis, cancer, and other diseases Transcriptional regulation of renal cytoprotective genes by Nrf2 and its potential use as a therapeutic target to mitigate cisplatin-induced nephrotoxicity Pre-treatment with metformin activates Nrf2 antioxidant pathways and inhibits inflammatory responses through induction of AMPK after transient global cerebral ischemia. Metab. Brain Dis Bardoxolone methyl modulates efflux transporter and detoxifying enzyme expression in cisplatin-induced kidney cell injury Sulforaphane bioavailability and chemopreventive activity in women scheduled for breast biopsy No evidence for interactions of dimethylfumarate (DMF) and its main metabolite monomethylfumarate (MMF) with human cytochrome P450 (CYP) enzymes and the P-glycoprotein (P-gp) drug transporter Emerging role of NRF2 in chemoresistance by regulating drug-metabolizing enzymes and efflux transporters Metformin sensitizes endometrial cancer cells to chemotherapy through IDH1-induced Nrf2 expression via an epigenetic mechanism The molecular mechanisms regulating the KEAP1-NRF2 pathway Drug interactions in oncology Regulation of dimethyl-fumarate toxicity by proteasome inhibitors Sulforaphane potentiates anticancer effects of doxorubicin and attenuates its cardiotoxicity in a breast cancer model Metformin suppresses Nrf2-mediated chemoresistance in hepatocellular carcinoma cells by increasing glycolysis Camptothecin suppresses NRF2-ARE activity and sensitises hepatocellular carcinoma cells to anticancer drugs Diosmetin induces apoptosis and enhances the chemotherapeutic efficacy of paclitaxel in non-small cell lung cancer cells via Nrf2 inhibition Luteolin sensitizes two oxaliplatinresistant colorectal cancer cell lines to chemotherapeutic drugs via inhibition of the Nrf2 pathway. Asian Pac Transcription factor NRF2 as a therapeutic target for chronic diseases: a systems medicine approach Therapeutic targeting of the NRF2 and KEAP1 partnership in chronic diseases Bardoxolone methyl in type 2 diabetes and stage 4 chronic kidney disease Oncogene-induced Nrf2 transcription promotes ROS detoxification and tumorigenesis Regulation of oxidative stress by Nrf2 in the pathophysiology of infectious diseases Formononetin ameliorates oxaliplatin-induced peripheral neuropathy via the KEAP1-NRF2-GSTP1 axis BG-12 (dimethyl fumarate): a review of mechanism of action, efficacy, and safety Pharmacokinetic drug-drug interactions of tyrosine kinase inhibitors: a focus on cytochrome P450, transporters, and acid suppression therapy iASPP is an antioxidative factor and drives cancer growth and drug resistance by competing with Nrf2 for Keap1 binding Risk of cancer in multiple sclerosis (MS): a systematic review and meta-analysis DMF, but not other fumarates, inhibits NF-κB activity in vitro in an Nrf2-independent manner Epigenetic regulation of Keap1-Nrf2 signaling. Free Radic Brusatol overcomes chemoresistance through inhibition of protein translation The Nrf2 regulatory network provides an interface between redox and intermediary metabolism Cancer chemoprevention mechanisms mediated through the Keap1-Nrf2 pathway Nrf2-MafG heterodimers contribute globally to antioxidant and metabolic networks Nrf2 enhances cell proliferation and resistance to anticancer drugs in human lung cancer A phase I firstin-human trial of bardoxolone methyl in patients with advanced solid tumors and lymphomas Nrf2 overexpression predicts prognosis and 5-FU resistance in gastric cancer. Asian Pac Dimethyl fumarate confers neuroprotection by casein kinase 2 phosphorylation of Nrf2 in murine intracerebral hemorrhage Prevalence and significance of potential drug-drug interactions among cancer patients receiving chemotherapy An Nrf2/small Maf heterodimer mediates the induction of phase II detoxifying enzyme genes through antioxidant response elements Simvastatin induces heme oxygenase-1 via NF-E2-related factor 2 (Nrf2) activation through ERK and PI3K/Akt pathway in colon cancer Complementary medicine use during cancer treatment and potential herbdrug interactions from a cross-sectional study in an academic centre High levels of Nrf2 determine chemoresistance in type II endometrial cancer Chemopreventive activity of sulforaphane Dimethyl fumarate regulates histone deacetylase expression in astrocytes NRF2 regulates HER1 signaling pathway to modulate the sensitivity of ovarian cancer cells to lapatinib and erlotinib Targeting of Nrf2 induces DNA damage signaling and protects colonic epithelial cells from ionizing radiation Dimethyl fumarate and its esters: a drug with broad clinical utility Nrf2 deficiency exaggerates doxorubicin-induced cardiotoxicity and cardiac dysfunction Cancer risk in patients with psoriasis: should we be paying more attention? Expet Rev The keap1-nrf2-ARE pathway as a potential preventive and therapeutic target: an update Interactions between drugs and sulforaphane modulate the drug metabolism enzymatic system Role of nrf2 in oxidative stress and toxicity NRF2 inhibition represses ErbB2 signaling in ovarian carcinoma cells: implications for tumor growth retardation and docetaxel sensitivity. Free Radic Targeting Keap1/Nrf2/ARE signaling pathway in multiple sclerosis Dimethyl fumarate attenuates oxaliplatin-induced peripheral neuropathy without affecting the anti-tumor activity of oxaliplatin in rodents Associations of polypharmacy and inappropriate medications with adverse outcomes in older adults with cancer: a systematic review and meta-analysis Hemin reduces cellular sensitivity to imatinib and anthracyclins via Nrf2 Dimethyl fumarate restores apoptosis sensitivity and inhibits tumor growth and metastasis in CTCL by targeting NF-κB New possible pharmacological targets for statins and ezetimibe Pharmacodynamic drug-drug interactions SARS-CoV2-mediated suppression of NRF2-signaling reveals potent antiviral and anti-inflammatory activity of 4-octyl-itaconate and dimethyl fumarate Brusatol provokes a rapid and transient inhibition of Nrf2 signaling and sensitizes mammalian cells to chemical toxicity-implications for therapeutic targeting of Nrf2. Free Radic Potential applications of NRF2 modulators in cancer therapy Resistance to gefitinib and cross-resistance to irreversible EGFR-TKIs mediated by disruption of the Keap1-Nrf2 pathway in human lung cancer cells Identification of novel protein targets of dimethyl fumarate modification in neurons and astrocytes reveals actions independent of Nrf2 stabilization Role of Nrf2 and protective effects of Metformin against tobacco smoke-induced cerebrovascular toxicity Activation of Nrf2-ARE signaling mitigates cyclophosphamide-induced myelosuppression The protective effects of silymarin against doxorubicin-induced cardiotoxicity and hepatotoxicity in rats Brusatol enhances the efficacy of chemotherapy by inhibiting the Nrf2-mediated defense mechanism Activators and inhibitors of NRF2: a review of their potential for clinical development NRF2 and the hallmarks of cancer Dimethyl fumarate controls the NRF2/DJ-1 Axis in cancer cells: therapeutic applications The impact of comorbidity on cancer and its treatment Dimethyl fumarate treatment induces adaptive and innate immune modulation independent of Nrf2 Clinical outcomes associated with drug-drug interactions of oral chemotherapeutic agents: a comprehensive evidence-based literature review Nrf2 plays an important role in coordinated regulation of Phase II drug metabolism enzymes and Phase III drug transporters Genetic alteration of Keap1 confers constitutive Nrf2 activation and resistance to chemotherapy in gallbladder cancer Acquisition of doxorubicin resistance in ovarian carcinoma cells accompanies activation of the NRF2 pathway. Free Radic The balance between NRF2/GSH antioxidant mediated pathway and DNA repair modulates cisplatin resistance in lung cancer cells Small molecule inhibitor of NRF2 selectively intervenes therapeutic resistance in KEAP1-deficient NSCLC tumors Nrf2-mediated redox signaling in arsenic carcinogenesis: a review Nrf2: control of sensitivity to carcinogens NRF2 and cancer: the good, the bad and the importance of context NRF2 preserves genomic integrity by facilitating ATR activation and G2 cell cycle arrest Regulation of nrf2/ARE pathway by dietary flavonoids: a friend or foe for cancer management Evaluation of biodistribution of sulforaphane after administration of oral broccoli sprout extract in melanoma patients with multiple atypical nevi NRF2 as a determinant of cellular resistance in retinoic acid cytotoxicity. Free Radic The effects of NRF2 modulation on the initiation and progression of chemically and genetically induced lung cancer Ascorbic acid restores sensitivity to imatinib via suppression of Nrf2-dependent gene expression in the imatinib-resistant cell line Ruxolitinib synergizes with DMF to kill via BIMþBAD-induced mitochondrial dysfunction and via reduced SOD2/TRX expression and ROS Pharmacological applications of Nrf2 inhibitors as potential antineoplastic drugs Transcriptional regulation by Nrf2 Transcriptional changes in prostate of men on active surveillance after a 12-mo glucoraphanin-rich broccoli intervention-results from the Effect of Sulforaphane on prostate CAncer PrEvention (ESCAPE) randomized controlled trial Opposing effects of antidiabetic interventions on malignant growth and metastasis Metformin diminishes the unfavourable impact of Nrf2 in breast cancer patients with type 2 diabetes The Nrf2-antioxidant response element pathway: a target for regulating energy metabolism Generation of a stable antioxidant response element-driven reporter gene cell line and its use to show redox-dependent activation of nrf2 by cancer chemotherapeutic agents Identification of retinoic acid as an inhibitor of transcription factor Nrf2 through activation of retinoic acid receptor alpha Nrf2 enhances resistance of cancer cells to chemotherapeutic drugs, the dark side of Nrf2 Oxaliplatin activates the Keap1/Nrf2 antioxidant system conferring protection against the cytotoxicity of anticancer drugs. Free Radic NRF2 activation by antioxidant antidiabetic agents accelerates tumor metastasis Metformin protects against LPSinduced intestinal barrier dysfunction by activating AMPK pathway Mechanisms and applications of the anti-cancer effect of pharmacological ascorbic acid in cervical cancer cells Metabolism, transport and drug-drug interactions of silymarin Nrf2/are Signaling Protects against Oxaliplatin-Induced Hepatotoxicity in Mice Mechanism of action of isothiocyanates: the induction of ARE-regulated genes is associated with activation of ERK and JNK and the phosphorylation and nuclear translocation of Nrf2 The KEAP1-NRF2 system: a thiolbased sensor-effector apparatus for maintaining redox homeostasis Nrf2 inhibits oxaliplatin-induced peripheral neuropathy via protection of mitochondrial function. Free Radic Natural compounds against doxorubicin-induced cardiotoxicity: a review on the involvement of Nrf2/ARE signaling pathway Dimethyl fumarate disrupts human innate immune signaling by targeting the IRAK4-MyD88 complex Increasing cancer risk over calendar year in people with multiple sclerosis: a case-control study An overview of chemical inhibitors of the Nrf2-ARE signaling pathway and their potential applications in cancer therapy. Free Radic Evaluation of potential drug-drug interaction between delayed-release dimethyl fumarate and a commonly used oral contraceptive (Norgestimate/Ethinyl estradiol) in healthy women Phytochemical compounds targeting on Nrf2 for chemoprevention in colorectal cancer This work was supported by National Natural Science Foundation of China (NSFC) (81472657 and 81272468 to Yu S.). The authors thank all the members of Yu's group for technical assistance and critical discussions.