Skip to main content
ACS AuthorChoice logoLink to ACS AuthorChoice
. 2023 May 8;36(6):805–817. doi: 10.1021/acs.chemrestox.2c00396

An Adverse Outcome Pathway Network for Chemically Induced Oxidative Stress Leading to (Non)genotoxic Carcinogenesis

Christina H J Veltman †,§,*, Jeroen L A Pennings , Bob van de Water §, Mirjam Luijten
PMCID: PMC10283045  PMID: 37156502

Abstract

graphic file with name tx2c00396_0004.jpg

Nongenotoxic (NGTX) carcinogens induce cancer via other mechanisms than direct DNA damage. A recognized mode of action for NGTX carcinogens is induction of oxidative stress, a state in which the amount of oxidants in a cell exceeds its antioxidant capacity, leading to regenerative proliferation. Currently, carcinogenicity assessment of environmental chemicals primarily relies on genetic toxicity end points. Since NGTX carcinogens lack genotoxic potential, these chemicals may remain undetected in such evaluations. To enhance the predictivity of test strategies for carcinogenicity assessment, a shift toward mechanism-based approaches is required. Here, we present an adverse outcome pathway (AOP) network for chemically induced oxidative stress leading to (NGTX) carcinogenesis. To develop this AOP network, we first investigated the role of oxidative stress in the various cancer hallmarks. Next, possible mechanisms for chemical induction of oxidative stress and the biological effects of oxidative damage to macromolecules were considered. This resulted in an AOP network, of which associated uncertainties were explored. Ultimately, development of AOP networks relevant for carcinogenesis in humans will aid the transition to a mechanism-based, human relevant carcinogenicity assessment that involves a substantially lower number of laboratory animals.

Introduction

Carcinogenesis is a multistep process in which normal cells transform into cancer cells by acquiring various, biologically diverse characteristics referred to as cancer hallmarks.13 At present, the International Agency for Research on Cancer (IARC) has classified 534 chemical compounds as proven human carcinogen (IARC 1), probable human carcinogen (IARC 2A) or possible human carcinogen (IARC 2B).4 These chemical carcinogens can induce tumor formation either through genotoxic (GTX) or nongenotoxic (NGTX) mechanisms.5 Compounds able to directly damage or interact with DNA are generally classified as GTX carcinogens, whereas NGTX carcinogens do not directly interact with DNA nor the cellular apparatus involved in preserving genomic integrity.6 Instead, NGTX carcinogens induce carcinogenesis via mechanisms including receptor activation, chronic inflammation, immune suppression, endocrine disruption, epigenetic silencing or oxidative stress.2,7,8 Both GTX and NGTX mechanisms of carcinogenesis eventually involve the cancer hallmarks of genomic instability, loss of proliferative control and resistance to cell death.5 Despite a wide spectrum of modes of action (MoAs) for NGTX carcinogens, increased cell proliferation is shown to be a fundamental key event (KE).7,9 Receptor activation, sustained cytotoxicity, altered signal transduction, immunosuppression and induction of oxidative stress can all contribute to stimulation of cell proliferation.1,7 Contrary to GTX carcinogens, NGTX carcinogens are hypothesized to induce tumorigenesis through repeated or sustained exposure resulting in prolonged perturbation or modulation of physiological processes.10,11

Oxidative stress leading to regenerative proliferation is a MoA relevant for carcinogenesis.1216 Upon an imbalance between the generation of oxidants, such as reactive oxygen species (ROS), and their scavenging by antioxidants, oxidative stress is induced.17 This imbalance can arise from exposure to either endogenous or exogenous sources responsible for oxidant generation or from depletion of antioxidants.13 Oxidative stress is known to promote carcinogenesis through both DNA damage and impaired repair and through indirect actions influencing homeostasis and signaling.18 Additionally, ROS play a role in numerous stages of the multistep carcinogenic process.19,20

Traditionally, cancer hazard assessment requires long-term carcinogenicity studies (OECD Test Guidelines 45121/45322). There is a strong need for alternative approaches because rodent studies show limited translatability to man,23 raise ethical concerns, have questionable reproducibility7,24 and are time- and cost consuming.23 Currently, cancer hazard assessment predominantly relies on genetic toxicity end points, since genetic damage is considered to be key to carcinogenicity.25 While there is evidence of a GTX MoA for most of the substances classified by IARC, approximately 9% of these substances lack a GTX potential based on data from a battery of in vitro and in vivo genotoxicity tests,8 indicating that NGTX carcinogens may remain undetected in such evaluations since these act via other mechanism than direct genetic damage. Therefore, to enhance the prediction of carcinogenic potential of substances, transitioning toward a mechanism-based approach is deemed necessary.7,2528 To aid this transition, adverse outcome pathways (AOPs) could serve as a framework to select appropriate new approach methodologies (NAMs) and identify research gaps.

In this article, an AOP network for chemically induced oxidative stress leading to (NGTX) carcinogenesis is proposed. We start with summarizing the main findings concerning the role of oxidative stress in the cancer hallmarks and its relation to cancer in both animals and humans. Next, possible mechanisms for chemical induction of oxidative stress are considered. The relation between oxidative stress and the cancer hallmarks is further explored by discussing the biological effects of oxidative damage to macromolecules. Ultimately, we integrate this information into an AOP network that can contribute to the development of an integrated approach to the testing and assessment (IATA) for suspected (NGTX) carcinogens inducing oxidative stress. Given the breadth of literature on oxidative stress as well as carcinogenesis, we considered it necessary to demarcate the scope of this paper. This review focuses on the role of ROS, whereas the role of reactive nitrogen species in oxidative stress and carcinogenesis is considered beyond the scope of this paper. Additionally, although we are well aware of the interrelation between oxidative stress and inflammation, we deliberately describe the discussed processes from an oxidative stress perspective.

Methodology

A literature search was performed in October 2021 using Embase. As a search strategy (Figure S1), certain keywords such as oxidative stress/reactive oxygen species/oxygen radicals, chemical induction/carcinogen and carcinogenesis/tumorigenesis/cancer, etc. were used. After compiling a list of 539 papers, abstracts were read and papers focusing on genotoxins, effects in offspring, and antioxidants, or when the full text could not be accessed were discarded. The resulting papers were read and then either included or excluded based on redundancy of information. Some additional relevant papers were obtained through reference tracking. Papers published after October 2021 that sufficed our search strategy were read and incorporated if assessed relevant.

Oxidative Stress in Relation to the Cancer Hallmarks

During the process of carcinogenesis cells acquire certain characteristics referred to as cancer hallmarks.1,2 These hallmarks rationalize the complex biological processes involved in tumor formation.2,29,30 In a well-known review, Hanahan and Weinberg proposed the following six hallmarks: (1) ‘sustaining proliferative signaling’, (2) ‘evading growth suppression’, (3) ‘enabling replicative immortality’, (4) “inducing angiogenesis”, (5) “resisting cell death”, and (6) ‘activating invasion and metastasis’.1 Later, two emerging hallmarks and two enabling characteristics were added: ‘avoiding immune destruction’, ‘deregulating cellular energetics’, ‘genome instability and mutation’, and ‘tumor-promoting inflammation’.2 Since then, novel insights into carcinogenesis have led to occasional reconsideration of the cancer hallmarks3,29,30 (Figure 1).

Figure 1.

Figure 1

Hallmarks of cancer. The 10 hallmarks of cancer as described by Hanahan and Weinberg,2 supplemented with novel insights into carcinogenesis.3,29,30 Translucent hallmarks are not discussed in this paper. The inner circle schematically represents the signaling networks that connect the hallmarks and the tumor microenvironment.

A vast number of studies have described the relationship between oxidative stress and carcinogenesis.13,16,1820,3133 Generally, low concentrations of ROS are involved in regular biological processes such as transcriptional regulation, differentiation and proliferation, whereas high levels of ROS exceed the antioxidant defense system, consequently inducing oxidative stress.34 Carcinogenesis may occur when ROS exceed physiological levels sustainably while avoiding excessive cell death.20 ROS can induce tumor formation either through GTX or NGTX mechanisms.13 Genotoxicity occurs when ROS interact with DNA and the resulting oxidative DNA damage is not repaired prior to DNA replication. Alternatively, ROS can modulate expression of genes and proteins, such as growth factors and proto-oncogenes, which play a pivotal role in carcinogenesis, without inflicting direct DNA damage.13,18

Since the focus of this review is on NGTX carcinogenesis by chemical induction of oxidative stress, the cancer hallmarks ‘activating invasion and metastasis’ and ‘inducing angiogenesis’ will not be discussed. These hallmarks are considered late events in carcinogenesis, making it virtually impossible to distinguish chemically induced oxidative stress from tumor-induced oxidative stress, thereby lowering its predictive value for human health protection and relevance for chemical hazard assessment.7 For the cancer hallmarks ‘enabling replicative immortality’ and ‘deregulating cellular energetics’, we were unable to find sufficiently substantial, biologically relevant evidence for their relation with chemical induction of oxidative stress. Hence, the focus of this review is on the relationship between oxidative stress and the cancer hallmarks ‘maintaining proliferative capacity’ (fusion of sustaining proliferative signaling and evading growth suppression), ‘resisting cell death’, ‘immune modulation’ (merger of avoiding immune destruction and tumor-promoting inflammation), and ‘genome instability and epigenetic dysregulation’, excluding direct GTX effects.

Maintaining Proliferative Capacity

Cancer cells are known for their ability to maintain proliferation.2 One mechanism through which ROS are involved in sustaining proliferative signaling is through oxidation of phosphatases and subsequent activation of signaling pathways associated with cell proliferation.13,32,35 To illustrate, ROS can oxidize and subsequently inhibit phosphatase and tensin homologue (PTEN), leading to activation of phosphoinositide 3 kinase (PI3K)/protein kinase B (Akt) signaling.36,37 Alternatively, PI3K/Akt signaling activation can be mediated by oxidation of protein tyrosine phosphatase 1B (PTP1B).38 Similarly, oxidation of mitogen-activated protein kinase (MAPK) phosphatase (MKP) leads to activation of MAPK/extracellular signal-regulated kinase (ERK) signaling.39,40 MAPK/ERK signaling can be triggered through ROS-dependent intracellular calcium release and subsequent activation of protein kinase C (PKC) as well.41 Notably, induction of oxidative stress can both activate and repress nuclear factor κ-light-chain-enhancer of B cells (NF-κB) signaling.35,42 Inhibitor of NF-κB (IκB) oxidation results in dissociation, allowing nuclear translocation of NF-κB,43 whereas oxidation of IκB kinase (IKK) prevents IκB degradation and subsequent NF-κB nuclear translocation.44,45

Alternatively, cell proliferation can be maintained through activation of activator protein 1 (AP-1). Upon oxidation of thioredoxin (TRX), apoptosis signal-regulating kinase 1 (ASK1) activity is no longer inhibited, allowing c-Jun N-terminal kinase (JNK) proteins to translocate to the nucleus for induction of AP-1.46,47 Activation of AP-1 induces expression of growth-stimulatory genes and suppresses cell cycle inhibitors.13,48 Lastly, in response to oxidative modification of Kelch like ECH associated protein 1 (KEAP1), nuclear factor erythroid 2-related factor 2 (NRF2) is activated and subsequent translocation to the nucleus leads to expression of genes involved in proliferation (e.g., Ki67 and NOTCH1).49,50

Resisting Cell Death

The normal balance between proliferation and cell death is disturbed in cancer cells.29 ROS can disrupt this equilibrium either through inhibition of pro-apoptotic factors or through induction of antiapoptotic factors, ultimately mediating resistance to cell death. For example, ROS can activate the Akt pathway through oxidation of PTEN,36,37 which may lead to increased cell survival via phosphorylation and consequent inactivation of pro-apoptotic factors such as B-cell lymphoma (Bcl)-2-associated agonist of cell death (Bad), Bcl-2-associated X protein (Bax), and Bcl-2-interacting mediator of cell death (Bim).33,51,52 Additionally, oxidative stress can activate MAPK/p38,53 NF-κB43 and NRF250 signaling, resulting in reduced caspase activity. Moreover, NRF2 activation can reduce the release of cytochrome-c from mitochondria, preventing apoptosis through inhibition of apoptosome formation.54,55 Alternatively, ROS can induce expression of antiapoptotic Bcl-2 family members by means of NRF2,54,55 NF-κB, and signal transducer and activator of transcription 3/5 (STAT3/5) signaling.56

Immune Modulation

According to immune surveillance theory, cells are continually monitored by the body’s immune system and eliminated upon becoming cancerous.2 Following this principle, existing tumors have arisen from cells that somehow managed to avoid detection and elimination by the immune system. Oxidative stress can mediate avoidance of immune destruction in three ways. First, oxidative stress can induce the formation of regulatory T-cells and strengthen their immunosuppressive potency.57 Second, by inhibiting the interaction between the T-cell receptor and the major histocompatibility complex (MHC)-peptide complex, ROS can functionally impair cytotoxic T-cells, which play a pivotal role in immune destruction of cancer cells.58 Lastly, tumor-induced myeloid-derived suppressor cells (MDSCs) can inhibit T-cell proliferation in a ROS-dependent manner.59

Contrarily, oxidative stress is also involved in creating a tumor-promoting inflammatory microenvironment. The abundance of ROS can trigger pro-oncogenic signaling pathways, for example, NF-κB,43 MAPK,40 and STAT3,56 which in turn can contribute to the production of pro-inflammatory mediators such as tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and IL-6.56,60 Consecutively, these pro-inflammatory mediators can stimulate both ROS production61 and pro-oncogenic signaling pathways involved in proliferation, angiogenesis, invasion, and resistance to apoptosis.62

Genome Instability and Epigenetic Dysregulation

Besides direct induction of mutations as a result of oxidative DNA damage, oxidative stress can contribute to genome instability and epigenetic dysregulation. Repetitive sequences, which are particularly susceptible to DNA oxidation, can form secondary DNA structures upon oxidation.63 During DNA replication, recruitment of a special but error-prone DNA polymerase to these secondary DNA structures is needed for continuation of synthesis, ultimately contributing to genome instability.63 Furthermore, ROS-induced expression of Bcl-2, following NRF2,54,55 NF-κB, or STAT3/5 signaling,56 can inhibit DNA double-strand break (DSB) repair.64 Lastly, ROS-induced inflammation can cause both microsatellite and chromosomal instability via dysregulation of DNA repair enzymes, defective mitotic checkpoints, induction of DSBs, and dysregulated homologous recombination (reviewed in ref (65)).

Alternatively, ROS can cause alterations in the DNA methylation status through interaction with DNA and proteins.15 As a result of DNA oxidation, 8-hydroxy-2′-deoxyguanosine (8-OHdG) and 5-hydroxymethylcytosine (5hmC) may be formed.66,67 Consequently, DNA binding of methyl-CpG binding proteins (MBPs), epigenetic regulators responsible for DNA methyl transferase (DNMT) and histone deacetylase (HDAC) recruitment can be inhibited and these DNA regions might therefore get hypomethylated.66,67 Most demethylated regions are promoters belonging to oncogenes, which consequently can be activated.15,68,69 Additionally, methylation of repetitive and transposable elements is often lost in cancer, which can result in random integration of these elements into the genome and subsequently cause genetic instability.15,70 On the other hand, ROS can also mediate hypermethylation and subsequent loss of tumor suppressor promoter regions through upregulation and recruitment of DNMT and HDAC, a phenomenon frequently observed in human cancers.15,71 For example, hypermethylation of the human mutL homologue 1 (hMLH1) promoter region diminishes its expression,72 which is related to repressed activity of the mismatch repair system.73

Oxidative Stress in Cancer

Oxidative stress leading to regenerative proliferation is one of the best documented mechanisms for carcinogenesis.1215 About half of the IARC 1 classified substances (35 out of 86) were shown to have the ability to induce oxidative stress.74 Notably, chronic inflammation, associated with ROS formation and altered signaling, has become a well-recognized risk factor for various human cancers.14,62 Evidence for the role of oxidative stress in carcinogenesis can be found in both animals and humans.

Animal Carcinogenicity Data

Clear evidence for oxidative stress leading to regenerative proliferation as a carcinogenic MoA in animals was derived from rodent knockout studies. Loss of antioxidant genes, such as superoxide dismutase 1 (Sod1),75,76 peroxiredoxin 1 (Prdx1)77,78 and glutathione peroxidase (Gpx),79 was shown to predispose mice to oxidative DNA damage and carcinogenesis. Moreover, contrary to wildtype mice, mice with a knockout of cytochrome P450 2E1 (Cyp2e1; a known electron leaker during xenobiotic metabolism) were shown to not manifest hepatotoxicity nor regenerative proliferation upon exposure to chloroform.80 In another study, Cyp2e1-null mice showed no oxidative damage phenotype whereas wildtype and humanized Cyp2e1 mice did.81 Furthermore, impaired liver regeneration has been observed in rodents lacking either AP-1 (c-Jun monomer) or NF-κB, both are essential transcription factors for hepatic regenerative proliferation.82,83 Inhibition of IL-1α84 or hematopoietic depletion of inhibitor of NF-κB kinase β (Ikkβ)85 in mice resulted in decreased regenerative proliferation. Lastly, Jnk1 knockout mice showed impaired cellular proliferation and subsequent decreased liver carcinogenesis.86

In a project by the European Partnership for Alternative Approaches to animal testing (EPAA), 170 NGTX carcinogenic agrochemicals were evaluated, with the aim to assess the tumor types induced and organs affected and to identify the various MoAs underlying the carcinogenic potential.10 Mechanistic information collected on the NGTX carcinogens resulted in a list of nine MoAs, including sustained cytotoxicity leading to regenerative proliferation. Of the 96 substances for which a MoA could be established, 49 (51%) were presumed to induce sustained cytotoxicity. This MoA has been observed for tumors in the liver, kidney, stomach, bladder, and intestine.10 Induction of oxidative stress is a central event within the MoA sustained cytotoxicity.87 However, only for four of the 49 substances, induction of oxidative stress was explicitly mentioned.10 Together these four substances induced seven unique treatment-related tumors affecting the liver, spleen and lymphoid system.10

Human Carcinogenicity Data

Evidence for the role of oxidative stress in carcinogenesis in humans mainly comes from epidemiological studies. Increased risk of cancer of the liver, kidney, blood, immune system, bladder and gastrointestinal tract has been reported in humans after exposure to carcinogens known to operate via oxidative stress in animals.8896 For instance, GTX carcinogens (pharmaceuticals) that induce oxidative DNA damage, such as cyclophosphamide, etoposide, tamoxifen and azathioprine,97,98 have epidemiologically been linked to elevated cancer risk.89 Epidemiological evidence for NGTX carcinogens operating via oxidative stress exist as well; a few examples are described here. In a large cohort study of 23,829 sawmill workers in British Columbia, substantial evidence of an association between pentachlorophenol exposure and the incidence of liver cancer, non-Hodgkin lymphoma and multiple myeloma was found.90 Additionally, occupational exposure to pentachlorophenol might elevate the risk of hematopoietic,99 neurological and digestive tract cancer.100 Evidence for oxidative stress induction following pentachlorophenol exposure has been found both in cells with a human origin and in mice.101,102 Another example is exposure to trichloroethylene, for which an epidemiological link to increased risk of kidney cancer,88,9295 non-Hodgkin lymphoma,92,94,103 and liver cancer94,96 has been found. Trichloroethylene has been shown to induce oxidative stress in a human hepatic cell line104 and in rats.105 Moreover, in a prospective cohort of pesticide applicators, an association between thyroid cancer and lindane exposure was observed.106 Agricultural use of lindane also has been associated with an elevated risk of non-Hodgkin lymphoma.107,108 In rats, lindane exposure was shown to induce oxidative stress.109

Another line of evidence for human relevance derives from genetic variation in oxidative-stress related genes and cancer susceptibility in the human population.18 Single nucleotide polymorphisms (SNPs) in antioxidant enzymes SOD and catalase (CAT) have been linked to increased cancer incidence and susceptibility.110 Moreover, SNPs altering the function of AP-endonuclease 1 (APE1) and 8-oxo-guanine DNA glycosylase (OGG1), DNA repair genes primarily involved in base excision repair of oxidative DNA damage,18 have been linked to increased cancer risk in humans.111113

Next to this, biomarkers of oxidative stress can be found in cancer patients as well, though it should be noted that these are not indicative of a role for oxidative stress in tumor initiation. Increased total oxidant, decreased total antioxidant,114119 elevated malondialdehyde (MDA)114,116,120,121 and increased protein carbonyl serum levels117119,122 are found in cancer patients compared to healthy controls. Moreover, the catalogue of somatic mutations in cancer (COSMIC) has identified a mutational signature (SBS18) in human cancers which is associated with oxidative DNA damage.123 Finally, 8-OHdG, a biomarker for oxidative DNA damage, can be quantified in biopsies and urinary samples from cancer patients. 8-OHdG levels were found to be significantly increased in chronic hepatitis C patients with hepatocellular carcinoma (HCC) compared to chronic hepatitis C patients without HCC.124 Both 8-OHdG and NRF2 expressions were found to be significantly elevated in cancerous tissue compared to noncancerous tissue of HCC patients.125 Again, these biomarkers are indicative of oxidative stress in the cancer patient which is more likely the consequence rather than the cause of the tumor.

Chemical Induction of Oxidative Stress

Chemical substances can induce oxidative stress through direct or indirect mechanisms. Direct mechanisms include mitochondrial and extramitochondrial production of oxygen radicals, whereas impact on the antioxidant defense system is considered an indirect mechanism.13 Notably, chemical induction of oxidative stress is not restricted to carcinogens nor carcinogenesis.126128

Formation of Reactive Oxygen Species

An example of a direct mechanism of oxidative stress induction is mitochondrial production of oxygen radicals. Interaction of substances with mitochondria, especially mitochondrial complex I and III,13 during oxidative phosphorylation is considered to be central in the formation of ROS.129 By blocking electron transport, a substance can induce mitochondrial membrane depolarization, and consequently ROS production.130

Another mechanism of ROS formation is closely related to the biotransformation of a substance. During detoxification of substances, CYP enzymes are induced.14 CYP enzymes catalyze the transfer of oxygen to the substrate, a process thoroughly associated with nicotinamide adenine dinucleotide phosphate (NADPH) oxidase.48 Upon disturbance of this association, electrons derived from NADPH can reduce CYP-oxygen complexes, consequently generating ROS.14,18,48 Moreover, various CYP enzymes, specifically CYP2E1, can leak electrons during the course of their catalytic cycle, producing ROS in the process.48,131,132 CYP1A, CYP1B1, CYP1D1 and CYP3A4 have also been described to produce ROS during their catalytic cycle,133 either through direct interaction with a substance or through activation of the aryl hydrocarbon receptor (AhR) in case of CYP1A1.134

Impacting the Antioxidant Capacity

An indirect mechanism of oxidative stress induction is reduction of antioxidants, of which the primary function is to protect the body against overload of oxidants.135 The antioxidant defense system consists of three parts: enzymes, vitamins and minerals.135,136 This review focuses on enzymatic antioxidants since we consider these most relevant for exposure to environmental chemicals. The main human enzymatic antioxidants include SOD, CAT, GPX and PRDX.135,137 SOD converts superoxide radicals to hydrogen peroxide.138 Next, hydrogen peroxide can be converted by either CAT, GPX or PRDX.137 CAT directly converts hydrogen peroxide in water and oxygen,139 whereas GPX and PRDX require cofactors such as reduced glutathione (GSH) or reduced TRX, respectively, to convert hydrogen peroxide in water while oxidizing GSH and TRX.137,140 Glutathione reductase (GR) and thioredoxin reductase (TR) can convert oxidized GSH and TRX, respectively, back to their reduced states by converting NADPH to NADP+.137,140 Glutathione-S-transferase (GST) uses GSH for oxidant detoxification as well.137

Oxidative Damage to Macromolecules

ROS can interact with and damage macromolecules such as lipids, proteins and DNA.19 Through interaction with these macromolecules, ROS can affect multiple cellular processes such as proliferation, inflammation and cell death.31 Notably, different types of ROS can have distinct effects on macromolecules. For example, hydrogen peroxide mostly oxidizes cysteine residues of proteins reversibly, controlling protein activity similarly to other post-translation modifications (e.g., phosphorylation), whereas highly reactive ROS, such as superoxide and hydroxyl radicals, are most likely to induce lipid peroxidation and cell death.137

Oxidative Damage to Lipids

Lipid peroxidation is the process in which ROS break down polyunsaturated fatty acids, damaging lipid-containing cellular components, such as the cell membrane, mitochondria, and the endoplasmic reticulum (ER).141143 Membrane permeability and fluidity and activity of membrane-bound proteins can be affected by lipid peroxidation.129,144 Importantly, products of lipid peroxidation, such as MDA and 4-hydroxynonenal (4-HNE),141,142 can react with other macromolecules further impairing cellular function.143,145

Lipid peroxidation can affect cell signaling pathways involved in proliferation, inflammation and cell death (Figure 2). It is important to note that, similar to ROS itself, low levels of lipid peroxidation have proliferative effects, whereas high levels induce cell death.146 For example, low concentrations of 4-HNE can trigger activation of PI3K/Akt and MAPK/ERK signaling pathways, while high concentrations of 4-HNE are cytotoxic.146 Lipid peroxidation mediated cytotoxicity can either lead to cell death or uncontrolled cell growth.147,148 Moreover, lipid peroxidation products can induce inflammatory responses through JNK.149

Figure 2.

Figure 2

Schematic representation of ROS-induced damage and biological effects in relation to carcinogenesis. ROS can interact with and damage lipids (green/left), proteins (blue/middle) and DNA (red/right). Through signaling pathways, oxidative modification of macromolecules can contribute to various cancer hallmarks and, ultimately, carcinogenesis. Inflammation also affects neighboring cells. First layer effector molecules are depicted in orange/rectangles, second layer effector molecules in purple/rounded rectangles, and third layer effector molecules in yellow/circles. ROS: reactive oxygen species; SOD: superoxide dismutase; CAT: catalase; GPX: glutathione peroxidase; PKC: protein kinase; NF-κB: factor κ-light-chain-enhancer of B cells inhibitor; IκB: Inhibitor of NF-κB; IKK: IκB kinase; PTEN: phosphatase and tensin homologue; PI3K: phosphoinositide 3 kinase; AKT: protein kinase B; PTP1B: protein tyrosine phosphatase 1B; JAK/STAT: Janus kinase/signal transducer and activator of transcription protein; MKP: MAPK phosphatase; ERK: extracellular signal-regulated kinase; TRX: thioredoxin; AKS1: apoptosis signal-regulating kinase 1; JNK: c-Jun N-terminal kinase; AP1: activator protein 1; KEAP1: Kelch like ECH associated protein 1; NRF2: nuclear factor erythroid 2-related factor 2; HDAC: histone deacetylase; DNMT: DNA methyltransferase; MBP: methyl binding protein; Bcl2: B-cell lymphoma 2; Bcl-xL: B-cell lymphoma extra-large; Bad: Bcl-2-associated agonist of cell death; Bim: Bcl-2-interacting mediator of cell death; Bax: Bcl-2-associated X protein.

Oxidative Modification of Proteins

Oxidative damage can reduce, increase, modify or completely abrogate biological activity of proteins.143 Examples of oxidized proteins are protein carbonyl derivates, advanced oxidation protein products, and advanced glycation end products.141 During oxidative stress, proteins can be temporarily oxidized leading to altered protein function and signaling.150 Oxidized receptor proteins can modify the transfer of signals13 or enzymatic activity may be reduced due to oxidative alteration.3640,43 One important example is ROS-mediated cysteine modification of KEAP1, resulting in nuclear translocation and transcriptional activation of NRF2.50 Alternatively, protein oxidation can also be irreversible, such as protein carbonylation, resulting in loss of function due to protein aggregation and degradation.150 In addition, accumulation of oxidized proteins may result in impaired cellular function and apoptosis.143 Furthermore, oxidative stress can lead to misfolding of proteins in the ER, consequently inducing ER stress.151

Oxidative damage to proteins can affect cell proliferation, survival and inflammation (Figure 2). Upon oxidation of TRX46 or phosphatases,3640,43 such as MKP, PTEN, PTP1B, and IκB, proliferative signaling is stimulated. Alternatively, oxidative stress can result in intracellular release of calcium,152 consequently stimulating PKC signaling, which in turn can activate MAPK/ERK and NF-κB signaling pathways.41 Moreover, protein oxidation can enhance survival through oxidation of PTEN, followed by PI3K/Akt activation and subsequent inactivation of pro-apoptotic Bad, Bax, and Bim.33,51,52 Additionally, oxidation of IκB lifts its inhibitory effect on NF-κB signaling, which can both reduce caspase activity43 and induce expression of antiapoptotic Bcl-2 family members.56 This can also be mediated by oxidation of KEAP1 and subsequent activation of NRF2 signaling.50,54,55 Lastly, NF-κB signaling contributes to inflammation via the production of pro-inflammatory mediators.56,60

Oxidative Damage to DNA

ROS-induced oxidative DNA modification may result in alteration of the DNA methylation status15 or genomic instability following DNA oxidation-induced secondary structures.63 These alterations in DNA methylation, resulting from oxidative modification of MBPs, DNMTs, and HDACs,66,67 can activate proto-oncogenes, such as c-Myc, Kras and c-Jun, as a result of promoter hypomethylation,68,69 and inactivate tumor suppressors, for instance RUNX3 (RUNX family transcription factor 3) and hMLH1, following promoter hypermethylation72,153 (Figure 2). Altered regulation of proto-oncogenes and tumor suppressor genes is pivotal in carcinogenesis.154

Relation with the Cancer Hallmarks and Carcinogenesis

Oxidative damage to lipids, proteins and DNA can affect multiple cancer hallmarks, including maintaining proliferative capacity, immune modulation, resistance to cell death, and genomic instability and epigenetic dysregulation (Figure 2). Ultimately, through contribution to these cancer characteristics, carcinogenesis can be initiated and promoted.1,2 Notably, cancer hallmarks can be induced through oxidative damage to one specific class of macromolecules or to multiple (Figure 2). Importantly, oxidative damage mediated cell death can be compensated by regenerative proliferation.155,156

Proposed AOP Network for Chemical Induction of Oxidative Stress Leading to (Non)genotoxic Carcinogenesis

Through structuring the above-described information regarding the role of oxidative stress in the cancer hallmarks, chemical induction of oxidative stress and oxidative damage to macromolecules, we have developed an AOP network for chemically induced oxidative stress leading to carcinogenesis (Figure 3). Notably, this is an outline AOP network that requires further development especially a detailed description of the KEs and key event relationships (KERs). A preliminary weight of evidence (WoE) evaluation was performed (Table S1A–D) according to OECD guidance.157 Adjacent KERs were assessed for both biological plausibility (Table S1A) and empirical evidence including consideration of contradictions (Table S1C), while KEs were assessed for essentiality (Table S1B). For this, we relied on the WoE assessments available in the AOP-Wiki (aopwiki.org) as well as scientific literature. Details are given in Tables S1A–D, with Table S1D providing a summary of the WoE for each of the adjacent KERs. Overall, the biological plausibility for the direct relationships in this AOP network is fairly strong. Evidence from empirical studies partly supports this assessment, but for some KERs the number of published studies appeared to be limited. The essentiality of the KEs in the AOP network, which can be demonstrated by modulating one KE and observing concordance in the downstream KEs, was considered as moderate. Based on this, we consider the overall WoE of the AOP network moderate to strong: certain branches are very data rich, providing strong evidence, whereas other branches require more data to be generated. Additionally, to further examine this AOP network, a noncomprehensive list of possible assays for KEs and reference chemicals for these assays is provided in Table S2.

Figure 3.

Figure 3

Proposed network of AOPs for chemically induced oxidative stress leading to carcinogenesis. Chronic or prolonged activation of the molecular initiating event and subsequent key events is required for this AOP network to trigger the adverse outcome. Molecular initiating events are depicted in green, cellular effects in orange, tissue effects in bright red and the adverse outcome in dark red. Cancer hallmarks are depicted in bold. Italicized text represent associated signaling pathways. Examples of cytochrome P450 enzymes, oxidases and key proteins in protein oxidation are given in parentheses.

The AOP network consists of five molecular initiating events (MIEs), namely activation of specific CYPs, inhibition of mitochondrial complexes, impacting on cellular antioxidant capacity, sustained AhR activation, and oxidase activation, which may all lead to oxidative stress. Following the central KE oxidative stress, three parallel KEs can be induced: lipid peroxidation, protein oxidation and oxidative DNA damage. Sufficient activation of these KEs can result in ER stress, altered cell survival, and/or genomic instability. Altered cell survival can result in tumor promoting inflammation, which in turn can lead to sustained or regenerative proliferation. Moreover, both genomic instability and altered cell survival can also result in regenerative or sustained proliferation. Sufficient and persistent activation of this KE can lead to tumor formation. Notably, both sustained or regenerative proliferation and mutations are pivotal in carcinogenesis, and their relationships to carcinogenesis are interlaced. Importantly, chronic or prolonged activation of the MIEs and subsequent KEs is necessary to trigger cancer.

Discussion and Conclusion

In the preceding sections, we reported evidence for the role of oxidative stress in carcinogenesis along the lines of the cancer hallmarks. We summarized the main findings concerning the role of oxidative stress in cancer in both animals and humans. Lastly, we described how chemicals can induce oxidative stress, and how oxidative stress to macromolecules relates to cell death, ER stress, inflammation, proliferation, and genomic instability. Our efforts have led to the development of an AOP network for chemically induced oxidative stress leading to carcinogenesis (Figure 3), which can be used to guide the development of an IATA for suspected NGTX carcinogens inducing oxidative stress. Notably, many other mechanism relevant for tumorigenesis exist. These were considered beyond the scope of this review and are therefore not included in the presented AOP network.

Within the AOP-Wiki,158 53 AOPs mentioning either oxidative stress or ROS as KE exist at the time of writing this review (Table S3). Of the 53 AOPs, only 12 cover cancer as an adverse outcome (AO), highlighting the diverse role of oxidative stress. In these 12 AOPs, various organs are linked to cancer resulting from induction of oxidative stress, including the liver, lung, breast, stomach and mesothelium. The diversity in target sites for the AO has implications for hazard assessment since quantitative differences between organs presumably affect the point of departure. Additionally, several currently existing AOPs link cytotoxicity to carcinogenesis (Table S4). Although these AOPs do not specifically mention induction of oxidative stress, this is known to be closely linked to cytotoxicity.87 Notably, chronic or sustained activation of KEs is necessary for carcinogenesis to occur.

The described AOP network is exploratory and requires further development to elucidate the associated uncertainties. One uncertainty relates to the relationship between mitochondrial dysfunction and oxidative stress. While some AOPs suggest that oxidative stress is the consequence of mitochondrial dysfunction, others suggest that oxidative stress is the cause. Here it should be noted that AOPs are a simplified representation of complex biological networks. The discrepancy between different AOPs is most likely the result of this simplification. We chose to depict mitochondrial dysfunction as the causative factor for oxidative stress since there is evidence for chemical interference with mitochondrial complexes leading to oxidative stress.130 However, ROS have been described to disrupt mitochondrial function, and this should be taken into account upon quantification of the KER. Another feedback loop within the AOP network includes damage to macromolecules by lipid peroxidation products,145 causing reinforcement of the effect of oxidative stress. Moreover, inflammation can both be portrayed as the causative and consequential factor in relation to oxidative stress. This likely results from the interrelation between oxidative stress and inflammation as an abundance of ROS can trigger production of pro-inflammatory cytokines, which in turn can stimulate ROS formation, resulting in a positive feedback loop.62 Since the focus of our AOP network is chemically induced oxidative stress, we depict oxidative stress as the cause and inflammation as the consequence. Similarly, oxidative stress can induce ER stress through protein oxidation, and ER stress can, in turn, cause oxidative stress.151 Lastly, altered cell survival and inflammation appear to be intertwined KEs. Cell death is known to trigger inflammation through the production of damage-associated molecular patterns (DAMPs),159 and inflammation can induce cell death via a process referred to as pyroptosis (reviewed in160,161). Importantly, all biologically relevant feedback loops should be taken into account when quantifying the AOP network.

We would like to give a few considerations concerning the application of the proposed AOP network for in vitro testing. First, both the macromolecular and cellular KEs can be measured in vitro using a relatively simple model system, for example, HepG2 cells. Despite quantitative uncertainties due to the cancer origin, such cell lines can be used for high-throughput screening after which more complex and relevant model systems can be used for further testing. One important challenge here is the definition of oxidative stress. At present, no threshold to discern physiological ROS levels from pathological ROS levels exist. Second, since prolonged activation of KEs is required for carcinogenesis to occur, repeated, low-dose exposure is hypothesized to be more relevant compared to single, high-dose exposure. In vitro assays are still limited in their duration and applicability for testing repeated-dose toxicity. Possibly, recovery experiments can shed light on the sustainability of induced effects. Lastly, no regulatory accepted in vitro assays for measuring proliferation currently exist. Additionally, the AO cannot be measured in vitro. Therefore, other downstream KEs such as altered cell survival and tumor promoting inflammation are thought to be key in distinguishing carcinogens from noncarcinogens. In vitro testing of the KE tumor promoting inflammation requires a complex model system containing multiple cell types, e.g., hepatocytes, Kupffer cells and stellate cells. For the KE altered cell survival interpretation at the moment in unclear. Both an increase and a decrease in cell survival can be linked to carcinogenesis, for the latter in combination with regenerative proliferation. Future research will have to elucidate how both different types of cell death and quantitative differences in cell survival are to be interpreted in relation to carcinogenesis.

For regulatory application of this AOP network, quantification of the various KERs is essential. Not all substances that induce oxidative stress through one of the proposed MIEs are known to cause cancer. For example, although acetaminophen (MIE: CYP2E1 activation)162 and diclofenac (MIE: mitochondrial dysfunction)163 are hepatotoxicants that are able to induce oxidative stress, they are not known as (human) carcinogens. We hypothesize that quantitative differences in certain KERs, specifically those associated with the cancer hallmarks, are central in this distinction of carcinogenic potential. To illustrate, acetaminophen is highly hepatotoxic and possibly does not cause liver cancer because death by liver failure will precede carcinogenesis.131 Next to this, toxicokinetic differences between substances are presumed to affect this inequity in AO induction as well. Furthermore, target sites can differ in their antioxidant capacity and varying responses to oxidative stress by the microenvironment are thought to influence carcinogenesis.137 Lastly, increased cellular proliferation does not exclusively lead to carcinogenesis, healthy regeneration can occur as well. Therefore, sustained perturbation of signaling, ultimately impairing normal regeneration and homeostasis, is expected to be fundamental in malignant transformation. Hazard assessment will thus have to qualitatively and quantitatively cover the KERs in the proposed AOP network and consider tissue-specific differences in sensitivity.

Glossary

Abbreviations

4-HNE

4-hydroxynonenal

5hmC

5-hydroxymethylcytosine

8-OHdG

8-hydroxy-2′-deoxyguanosine

AhR

aryl hydrocarbon receptor

Akt

protein kinase B

AO

adverse outcome

AOP

adverse outcome pathway

AP-1

activator protein 1

APE1

AP-endonuclease 1

AKS1

apoptosis signal-regulating kinase 1

Bad

Bcl-2-associated agonist of cell death

Bax

Bcl-2-associated X protein

Bcl-xL

B-cell lymphoma extra-large

Bim

Bcl-2-interacting mediator of cell death

CAT

catalase

CYP

cytochrome P450

DAMP

damage-associated molecular pattern

DNMT

DNA methyl transferase

DSB

double-strand break

EPAA

European partnership in alternative approaches to animal testing

ER

endoplasmic reticulum

ERK

extracellular signal-regulated kinase

GPX

glutathione peroxidase

GR

glutathione reductase

GSH

glutathione

GST

glutathione-S-transferase

GTX

genotoxic

HCC

hepatocellular carcinoma

HDAC

histone deacetylase

hMLH1

human mutL homologue 1

IARC

International Agency for Research on Cancer

IATA

integrated approach to testing and assessment

IκB

inhibitor of NF-κB

IKK

IκB kinase

IL

interleukin

JAK

Janus kinase

JNK

c-Jun N-terminal kinase

KE

key event

KEAP1

Kelch-like ECH associated protein 1

KER

key event relationship

MAPK

mitogen-activated protein kinase

MBP

methyl binding protein

MDA

malondialdehyde

MDSC

myeloid-derived suppressor cell

MHC

major histocompatibility complex

MIE

molecular initiating event

MKP

MAPK phosphatase

MoA

mode of action

NADPH

nicotinamide adenine dinucleotide phosphate

NAM

new approach methodology

NF-κB

factor κ-light-chain-enhancer of B cells inhibitor

NGTX

nongenotoxic

NRF2

nuclear factor erythroid 2-related factor 2

OGG1

8-oxo-guanine DNA glycosylase

PI3K

phosphoinositide 3 kinase

PKC

protein kinase

PRDX

peroxiredoxin

PTEN

phosphatase and tensin homologue

PTP1B

protein tyrosine phosphatase 1B

ROS

reactive oxygen species

RUNX3

RUNX family transcription factor 3

SNP

single nucleotide polymorphism

SOD

superoxide dismutase

STAT

signal transducer and activator of transcription protein

TNF

tumor necrosis factor

TR

thioredoxin reductase

TRX

thioredoxin

WoE

weight of evidence

Biographies

Christina H. J. Veltman is currently a Ph.D. student at RIVM and LACDR under the mentorship of Dr. Jeroen L. A. Pennings, Dr. Mirjam Luijten and Prof. Dr. Bob van de Water. Her research focuses on advancing the safety assessment of suspected nongenotoxic carcinogens inducing oxidative stress without the use of animals.

Jeroen L. A. Pennings is a bioinformatician/research scientist at RIVM. He received his Ph.D. in natural sciences, mathematics and computer sciences from the University of Nijmegen in 1999. His research focuses on computational approaches and alternative toxicity testing methods in toxicology.

Bob van de Water is a professor of Drug Safety Science and chair of the division Drug Discovery and Safety at LACDR. His research focuses on mechanistic toxicology, toxicogenomics and the application of new approach methodologies in safety assessment. A focus is on liver and kidney toxicity and carcinogenesis.

Mirjam Luijten is a research scientist at RIVM. She received her Ph.D. in human genetics from the University of Amsterdam in 2001. Her research focuses on nongenotoxic carcinogens and next generation risk assessment of chemicals.

Supporting Information Available

The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acs.chemrestox.2c00396.

  • Figure S1: Flowchart search strategy. Table S1: Weight of evidence assessment of the AOP network, conducted according to OECD guidance. Table S2: Possible assays for the AOP network and reference chemicals for these assays Table S3: AOPs from the AOP-wiki with oxidative stress or reactive oxygen species as key event. Table S4: AOPs from the AOP-wiki with cytotoxicity as key event and cancer as adverse outcome not mentioning oxidative stress or reactive oxygen species as key event (PDF)

Author Contributions

All authors contributed to the outline of the manuscript. C.H.J.V. performed the literature search and wrote the manuscript. J.L.A..P, B.v.d.W., and M.L. critically revised the work. CRediT: Christina H. J. Veltman conceptualization, writing-original draft, writing-review & editing; Jeroen L. A. Pennings conceptualization, supervision, writing-review & editing; Bob Van De Water conceptualization, writing-review & editing; Mirjam Luijten conceptualization, supervision, writing-review & editing.

This research was carried out under the RISK-HUNT3R project. The RISK-HUNT3R project has received funding from the European Union’s Horizon 2020 research and innovation program under grant agreement no. 964537, which is part of the ASPIS cluster.

The authors declare no competing financial interest.

Supplementary Material

tx2c00396_si_001.pdf (413.3KB, pdf)

References

  1. Hanahan D.; Weinberg R. A. The hallmarks of cancer. Cell 2000, 100 (1), 57–70. 10.1016/S0092-8674(00)81683-9. [DOI] [PubMed] [Google Scholar]
  2. Hanahan D.; Weinberg R. A. Hallmarks of cancer: the next generation. Cell 2011, 144 (5), 646–674. 10.1016/j.cell.2011.02.013. [DOI] [PubMed] [Google Scholar]
  3. Hanahan D. Hallmarks of Cancer: New Dimensions. Cancer Discov 2022, 12 (1), 31–46. 10.1158/2159-8290.CD-21-1059. [DOI] [PubMed] [Google Scholar]
  4. IARC . Agents Classified by the IARC Monographs, Vol 1–131. IARC Monographs on the Identification of Carcinogenic Hazards to Humans; International Agency for Research on Cancer - World Health Organization, 2022. https://monographs.iarc.who.int/agents-classified-by-the-iarc/ (accessed 2022-07-01).
  5. Luch A. Nature and nurture – lessons from chemical carcinogenesis. Nat. Rev. Cancer 2005, 5 (2), 113–125. 10.1038/nrc1546. [DOI] [PubMed] [Google Scholar]
  6. Adler S.; Basketter D.; Creton S.; Pelkonen O.; van Benthem J.; Zuang V.; Andersen K. E.; Angers-Loustau A.; Aptula A.; Bal-Price A.; et al. Alternative (non-animal) methods for cosmetics testing: current status and future prospects. Arch. Toxicol. 2011, 85 (5), 367–485. 10.1007/s00204-011-0693-2. [DOI] [PubMed] [Google Scholar]
  7. Jacobs M. N.; Colacci A.; Corvi R.; Vaccari M.; Aguila M. C.; Corvaro M.; Delrue N.; Desaulniers D.; Ertych N.; Jacobs A.; et al. Chemical carcinogen safety testing: OECD expert group international consensus on the development of an integrated approach for the testing and assessment of chemical non-genotoxic carcinogens. Arch. Toxicol. 2020, 94 (8), 2899–2923. 10.1007/s00204-020-02784-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Hernandez L. G.; van Steeg H.; Luijten M.; van Benthem J. Mechanisms of non-genotoxic carcinogens and importance of a weight of evidence approach. Mutat. Res. 2009, 682 (2–3), 94–109. 10.1016/j.mrrev.2009.07.002. [DOI] [PubMed] [Google Scholar]
  9. Cohen S. M.; Ellwein L. B. Genetic errors, cell proliferation, and carcinogenesis. Cancer Res. 1991, 51 (24), 6493–6505. [PubMed] [Google Scholar]
  10. Heusinkveld H.; Braakhuis H.; Gommans R.; Botham P.; Corvaro M.; van der Laan J. W.; Lewis D.; Madia F.; Manou I.; Schorsch F.; et al. Towards a mechanism-based approach for the prediction of nongenotoxic carcinogenic potential of agrochemicals. Crit Rev. Toxicol 2020, 50 (9), 725–739. 10.1080/10408444.2020.1841732. [DOI] [PubMed] [Google Scholar]
  11. Braakhuis H. M.; Slob W.; Olthof E. D.; Wolterink G.; Zwart E. P.; Gremmer E. R.; Rorije E.; van Benthem J.; Woutersen R.; van der Laan J. W.; et al. Is current risk assessment of non-genotoxic carcinogens protective?. Crit Rev. Toxicol 2018, 48 (6), 500–511. 10.1080/10408444.2018.1458818. [DOI] [PubMed] [Google Scholar]
  12. Jacobs M. N.; Colacci A.; Louekari K.; Luijten M.; Hakkert B. C.; Paparella M.; Vasseur P. International regulatory needs for development of an IATA for non-genotoxic carcinogenic chemical substances. ALTEX 2016, 33 (4), 359–392. 10.14573/altex.1601201. [DOI] [PubMed] [Google Scholar]
  13. Klaunig J. E.; Wang Z.; Pu X.; Zhou S. Oxidative stress and oxidative damage in chemical carcinogenesis. Toxicol. Appl. Pharmacol. 2011, 254 (2), 86–99. 10.1016/j.taap.2009.11.028. [DOI] [PubMed] [Google Scholar]
  14. Kakehashi A.; Wei M.; Fukushima S.; Wanibuchi H. Oxidative stress in the carcinogenicity of chemical carcinogens. Cancers 2013, 5 (4), 1332–1354. 10.3390/cancers5041332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Nishida N.; Kudo M. Oxidative Stress and Epigenetic Instability in Human Hepatocarcinogenesis. Dig Dis 2013, 31 (5–6), 447–453. 10.1159/000355243. [DOI] [PubMed] [Google Scholar]
  16. Saikolappan S.; Kumar B.; Shishodia G.; Koul S.; Koul H. K. Reactive oxygen species and cancer: A complex interaction. Cancer Lett. 2019, 452, 132–143. 10.1016/j.canlet.2019.03.020. [DOI] [PubMed] [Google Scholar]
  17. Sies H. Oxidative stress: oxidants and antioxidants. Exp Physiol 1997, 82 (2), 291–295. 10.1113/expphysiol.1997.sp004024. [DOI] [PubMed] [Google Scholar]
  18. Klaunig J. E.; Wang Z. Oxidative stress in carcinogenesis. Curr. Opin Toxicol 2018, 7, 116–121. 10.1016/j.cotox.2017.11.014. [DOI] [Google Scholar]
  19. Kumari S.; Badana A. K.; G M. M.; G S.; Malla R. Reactive Oxygen Species: A Key Constituent in Cancer Survival. Biomark Insights 2018, 13, 1. 10.1177/1177271918755391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Weinberg F.; Ramnath N.; Nagrath D. Reactive Oxygen Species in the Tumor Microenvironment: An Overview. Cancers 2019, 11 (8), 1191. 10.3390/cancers11081191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. OECD . Test No. 451: Carcinogenicity Studies. OECD Guidelines for the Testing of Chemicals, Section 4; OECD: Paris, 2018. https://www.oecd-ilibrary.org/content/publication/9789264071186-en (accessed 2022-06-21).
  22. OECD . Test No. 453: Combined Chronic Toxicity/Carcinogenicity Studies. OECD Guidelines for the Testing of Chemicals, Section 4; OECD: Paris, 2018. https://www.oecd-ilibrary.org/content/publication/9789264071223-en (accessed 2022-06-21).
  23. Marone P. A.; Hall W. C.; Hayes A. W. Reassessing the two-year rodent carcinogenicity bioassay: a review of the applicability to human risk and current perspectives. Regul. Toxicol. Pharmacol. 2014, 68 (1), 108–118. 10.1016/j.yrtph.2013.11.011. [DOI] [PubMed] [Google Scholar]
  24. Gottmann E; Kramer S; Pfahringer B; Helma C Data quality in predictive toxicology: reproducibility of rodent carcinogenicity experiments. Environ. Health Perspect 2001, 109 (5), 509–514. 10.1289/ehp.01109509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Luijten M.; Olthof E. D.; Hakkert B. C.; Rorije E.; van der Laan J. W.; Woutersen R. A.; van Benthem J. An integrative test strategy for cancer hazard identification. Crit Rev. Toxicol 2016, 46 (7), 615–639. 10.3109/10408444.2016.1171294. [DOI] [PubMed] [Google Scholar]
  26. Luijten M.; Corvi R.; Mehta J.; Corvaro M.; Delrue N.; Felter S.; Haas B.; Hewitt N. J.; Hilton G.; Holmes T. A comprehensive view on mechanistic approaches for cancer risk assessment of non-genotoxic agrochemicals. Regul. Toxicol. Pharmacol. 2020, 118, 104789. 10.1016/j.yrtph.2020.104789. [DOI] [PubMed] [Google Scholar]
  27. Julien E.; Boobis A. R.; Olin S. S. The Key Events Dose-Response Framework: A Cross-Disciplinary Mode-of-Action Based Approach to Examining Dose-Response and Thresholds. Crit Rev. Food Sci. Nutr 2009, 49 (8), 682–689. 10.1080/10408390903110692. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Doe J. E.; Boobis A. R.; Dellarco V.; Fenner-Crisp P. A.; Moretto A.; Pastoor T. P.; Schoeny R. S.; Seed J. G.; Wolf D. C. Chemical carcinogenicity revisited 2: Current knowledge of carcinogenesis shows that categorization as a carcinogen or non-carcinogen is not scientifically credible. Regul. Toxicol. Pharmacol. 2019, 103, 124–129. 10.1016/j.yrtph.2019.01.024. [DOI] [PubMed] [Google Scholar]
  29. Fouad Y. A.; Aanei C. Revisiting the hallmarks of cancer. Am. J. Cancer Res. 2017, 7 (5), 1016–1036. [PMC free article] [PubMed] [Google Scholar]
  30. Senga S. S.; Grose R. P. Hallmarks of cancer - the new statement. Open Biol. 2021, 11 (1), 200358. 10.1098/rsob.200358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Jelic M.; Mandic A.; Maricic S.; Srdjenovic B. Oxidative stress and its role in cancer. J. Cancer Res. Ther 2021, 17 (1), 22–28. 10.4103/jcrt.JCRT_862_16. [DOI] [PubMed] [Google Scholar]
  32. Hayes J. D.; Dinkova-Kostova A. T.; Tew K. D. Oxidative Stress in Cancer. Cancer Cell 2020, 38 (2), 167–197. 10.1016/j.ccell.2020.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Moloney J. N.; Cotter T. G. ROS signalling in the biology of cancer. Semin Cell Dev Biol. 2018, 80, 50–64. 10.1016/j.semcdb.2017.05.023. [DOI] [PubMed] [Google Scholar]
  34. Zheng F.; Goncalves F. M.; Abiko Y.; Li H.; Kumagai Y.; Aschner M. Redox toxicology of environmental chemicals causing oxidative stress. Redox Biol. 2020, 34, 101475. 10.1016/j.redox.2020.101475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Lingappan K. NF-kappaB in Oxidative Stress. Curr. Opin Toxicol 2018, 7, 81–86. 10.1016/j.cotox.2017.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Leslie N. R.; Bennett D.; Lindsay Y. E.; Stewart H.; Gray A.; Downes C. P. Redox regulation of PI 3-kinase signalling via inactivation of PTEN. EMBO J. 2003, 22 (20), 5501–5510. 10.1093/emboj/cdg513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Lee S. R.; Yang K. S.; Kwon J.; Lee C.; Jeong W.; Rhee S. G. Reversible Inactivation of the Tumor Suppressor PTEN by H2O2. J. Biol. Chem. 2002, 277 (23), 20336–20342. 10.1074/jbc.M111899200. [DOI] [PubMed] [Google Scholar]
  38. Salmeen A.; Andersen J. N.; Myers M. P.; Meng T. C.; Hinks J. A.; Tonks N. K.; Barford D. Redox regulation of protein tyrosine phosphatase 1B involves a sulphenyl-amide intermediate. Nature 2003, 423 (6941), 769–773. 10.1038/nature01680. [DOI] [PubMed] [Google Scholar]
  39. Seth D.; Rudolph J. Redox Regulation of MAP Kinase Phosphatase 3. Biochemistry 2006, 45 (28), 8476–8487. 10.1021/bi060157p. [DOI] [PubMed] [Google Scholar]
  40. Chan D. W.; Liu V. W. S.; Tsao G. S. W.; Yao K.-M.; Furukawa T.; Chan K. K. L.; Ngan H. Y. S. Loss of MKP3 mediated by oxidative stress enhances tumorigenicity and chemoresistance of ovarian cancer cells. Carcinogenesis 2008, 29 (9), 1742–1750. 10.1093/carcin/bgn167. [DOI] [PubMed] [Google Scholar]
  41. Hu C. T.; Wu J. R.; Cheng C. C.; Wang S.; Wang H. T.; Lee M. C.; Wang L. J.; Pan S. M.; Chang T. Y.; Wu W. S. Reactive oxygen species-mediated PKC and integrin signaling promotes tumor progression of human hepatoma HepG2. Clin Exp Metastasis 2011, 28 (8), 851–863. 10.1007/s10585-011-9416-6. [DOI] [PubMed] [Google Scholar]
  42. Morgan M. J.; Liu Z. G. Crosstalk of reactive oxygen species and NF-kappaB signaling. Cell Res. 2011, 21 (1), 103–115. 10.1038/cr.2010.178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Zhang J.; Johnston G.; Stebler B.; Keller E. T. Hydrogen peroxide activates NFkappaB and the interleukin-6 promoter through NFkappaB-inducing kinase. Antioxid Redox Signal 2001, 3 (3), 493–504. 10.1089/15230860152409121. [DOI] [PubMed] [Google Scholar]
  44. Reynaert N. L.; van der Vliet A.; Guala A. S.; McGovern T.; Hristova M.; Pantano C.; Heintz N. H.; Heim J.; Ho Y. S.; Matthews D. E.; et al. Dynamic redox control of NF-kappaB through glutaredoxin-regulated S-glutathionylation of inhibitory kappaB kinase beta. Proc. Natl. Acad. Sci. U. S. A. 2006, 103 (35), 13086–13091. 10.1073/pnas.0603290103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Korn S. H.; Wouters E. F.; Vos N.; Janssen-Heininger Y. M. Cytokine-induced activation of nuclear factor-kappa B is inhibited by hydrogen peroxide through oxidative inactivation of IkappaB kinase. J. Biol. Chem. 2001, 276 (38), 35693–35700. 10.1074/jbc.M104321200. [DOI] [PubMed] [Google Scholar]
  46. Liu H.; Nishitoh H.; Ichijo H.; Kyriakis J. M. Activation of apoptosis signal-regulating kinase 1 (ASK1) by tumor necrosis factor receptor-associated factor 2 requires prior dissociation of the ASK1 inhibitor thioredoxin. Mol. Cell. Biol. 2000, 20 (6), 2198–2208. 10.1128/MCB.20.6.2198-2208.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Tobiume K.; Matsuzawa A.; Takahashi T.; Nishitoh H.; Morita K. I.; Takeda K.; Minowa O.; Miyazono K.; Noda T.; Ichijo H. ASK1 is required for sustained activations of JNK/p38 MAP kinases and apoptosis. EMBO Reports 2001, 2 (3), 222–228. 10.1093/embo-reports/kve046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Hrycay E. G.; Bandiera S. M. Involvement of Cytochrome P450 in Reactive Oxygen Species Formation and Cancer. Adv. Pharmacol. 2015, 74, 35–84. 10.1016/bs.apha.2015.03.003. [DOI] [PubMed] [Google Scholar]
  49. Rojo de la Vega M.; Chapman E.; Zhang D. D. NRF2 and the Hallmarks of Cancer. Cancer Cell 2018, 34 (1), 21–43. 10.1016/j.ccell.2018.03.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Itoh K.; Wakabayashi N.; Katoh Y.; Ishii T.; Igarashi K.; Engel J. D.; Yamamoto M. Keap1 represses nuclear activation of antioxidant responsive elements by Nrf2 through binding to the amino-terminal Neh2 domain. Genes Dev. 1999, 13 (1), 76–86. 10.1101/gad.13.1.76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Xin M.; Deng X. Nicotine Inactivation of the Proapoptotic Function of Bax through Phosphorylation. J. Biol. Chem. 2005, 280 (11), 10781–10789. 10.1074/jbc.M500084200. [DOI] [PubMed] [Google Scholar]
  52. Qi X.-J.; Wildey G. M.; Howe P. H. Evidence That Ser87 of BimEL Is Phosphorylated by Akt and Regulates BimEL Apoptotic Function. J. Biol. Chem. 2006, 281 (2), 813–823. 10.1074/jbc.M505546200. [DOI] [PubMed] [Google Scholar]
  53. Nguyen Ho-Bouldoires T. H.; Clapéron A.; Mergey M.; Wendum D.; Desbois-Mouthon C.; Tahraoui S.; Fartoux L.; Chettouh H.; Merabtene F.; Scatton O.; et al. Mitogen-activated protein kinase-activated protein kinase 2 mediates resistance to hydrogen peroxide-induced oxidative stress in human hepatobiliary cancer cells. Free Radic Biol. Med. 2015, 89, 34–46. 10.1016/j.freeradbiomed.2015.07.011. [DOI] [PubMed] [Google Scholar]
  54. Niture S. K.; Jaiswal A. K. Nrf2-induced antiapoptotic Bcl-xL protein enhances cell survival and drug resistance. Free Radic Biol. Med. 2013, 57, 119–131. 10.1016/j.freeradbiomed.2012.12.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Niture S. K.; Jaiswal A. K. Nrf2 Protein Up-regulates Antiapoptotic Protein Bcl-2 and Prevents Cellular Apoptosis. J. Biol. Chem. 2012, 287 (13), 9873–9886. 10.1074/jbc.M111.312694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Liu C. W.; Lee T. L.; Chen Y. C.; Liang C. J.; Wang S. H.; Lue J. H.; Tsai J. S.; Lee S. W.; Chen S. H.; Yang Y. F. PM2.5-induced oxidative stress increases intercellular adhesion molecule-1 expression in lung epithelial cells through the IL-6/AKT/STAT3/NF-ΚB-dependent pathway. Part Fibre Toxicol 2018, 15 (1), 4. 10.1186/s12989-018-0240-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Kraaij M. D.; Savage N. D. L.; van der Kooij S. W.; Koekkoek K.; Wang J.; van den Berg J. M.; Ottenhoff T. H. M.; Kuijpers T. W.; Holmdahl R.; van Kooten C.; et al. Induction of regulatory T cells by macrophages is dependent on production of reactive oxygen species. Proc. Natl. Acad. Sci. U. S. A. 2010, 107 (41), 17686–17691. 10.1073/pnas.1012016107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Liu Y.; Wei J.; Guo G.; Zhou J. Norepinephrine-induced myeloid-derived suppressor cells block T-cell responses via generation of reactive oxygen species. Immunopharmacol Immunotoxicol 2015, 37 (4), 359–365. 10.3109/08923973.2015.1059442. [DOI] [PubMed] [Google Scholar]
  59. Wei J.; Zhang M.; Zhou J. Myeloid-derived suppressor cells in major depression patients suppress T-cell responses through the production of reactive oxygen species. Psychiatry Res. 2015, 228 (3), 695–701. 10.1016/j.psychres.2015.06.002. [DOI] [PubMed] [Google Scholar]
  60. Zhang Y. A.-O.; Cen J.; Jia Z.; Hsiao C. D.; Xia Q.; Wang X.; Chen X.; Wang R.; Jiang Z.; Zhang L. Hepatotoxicity Induced by Isoniazid-Lipopolysaccharide through Endoplasmic Reticulum Stress, Autophagy, and Apoptosis Pathways in Zebrafish. Antimicrob. Agents Chemother. 2019, 63 (5), 1. 10.1128/AAC.01639-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Yang D.; Elner S. G.; Bian Z. M.; Till G. O.; Petty H. R.; Elner V. M. Pro-inflammatory cytokines increase reactive oxygen species through mitochondria and NADPH oxidase in cultured RPE cells. Exp. Eye Res. 2007, 85 (4), 462. 10.1016/j.exer.2007.06.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Neganova M.; Liu J.; Aleksandrova Y.; Klochkov S.; Fan R. Therapeutic Influence on Important Targets Associated with Chronic Inflammation and Oxidative Stress in Cancer Treatment. Cancers 2021, 13 (23), 6062. 10.3390/cancers13236062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Stein M.; Eckert K. A. Impact of G-Quadruplexes and Chronic Inflammation on Genome Instability: Additive Effects during Carcinogenesis. Genes 2021, 12 (11), 1779. 10.3390/genes12111779. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Saintigny Y.; Dumay A.; Lambert S.; Lopez B. S. A novel role for the Bcl-2 protein family: specific suppression of the RAD51 recombination pathway. EMBO J. 2001, 20 (10), 2596–2607. 10.1093/emboj/20.10.2596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Colotta F.; Allavena P.; Sica A.; Garlanda C.; Mantovani A. Cancer-related inflammation, the seventh hallmark of cancer: links to genetic instability. Carcinogenesis 2009, 30 (7), 1073–1081. 10.1093/carcin/bgp127. [DOI] [PubMed] [Google Scholar]
  66. Bhusari S. S.; Dobosy J. R.; Fu V.; Almassi N.; Oberley T.; Jarrard D. F. Superoxide dismutase 1 knockdown induces oxidative stress and DNA methylation loss in the prostate. Epigenetics 2010, 5 (5), 402–409. 10.4161/epi.5.5.11853. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Valinluck V.; Tsai H. H.; Rogstad D. K.; Burdzy A.; Bird A.; Sowers L. C. Oxidative damage to methyl-CpG sequences inhibits the binding of the methyl-CpG binding domain (MBD) of methyl-CpG binding protein 2 (MeCP2). Nucleic Acids Res. 2004, 32 (14), 4100–4108. 10.1093/nar/gkh739. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Stefanska B.; Huang J.; Bhattacharyya B.; Suderman M.; Hallett M.; Han Z. G.; Szyf M. Definition of the landscape of promoter DNA hypomethylation in liver cancer. Cancer Res. 2011, 71 (17), 5891–5903. 10.1158/0008-5472.CAN-10-3823. [DOI] [PubMed] [Google Scholar]
  69. Kuppusamy S. P.; Kaiser J. P.; Wesselkamper S. C. Epigenetic Regulation in Environmental Chemical Carcinogenesis and its Applicability in Human Health Risk Assessment. Int. J. Toxicol 2015, 34 (5), 384–392. 10.1177/1091581815599350. [DOI] [PubMed] [Google Scholar]
  70. Howard G; Eiges R; Gaudet F; Jaenisch R; Eden A Activation and transposition of endogenous retroviral elements in hypomethylation induced tumors in mice. Oncogene 2008, 27 (3), 404–408. 10.1038/sj.onc.1210631. [DOI] [PubMed] [Google Scholar]
  71. Kang K. A.; Zhang R.; Kim G. Y.; Bae S. C.; Hyun J. W. Epigenetic changes induced by oxidative stress in colorectal cancer cells: methylation of tumor suppressor RUNX3. Tumor Biol. 2012, 33 (2), 403–412. 10.1007/s13277-012-0322-6. [DOI] [PubMed] [Google Scholar]
  72. Fleisher A. S.; Esteller M.; Harpaz N.; Leytin A.; Rashid A.; Xu Y.; Liang J.; Stine O. C.; Yin J.; Zou T.-T.; et al. Microsatellite Instability in Inflammatory Bowel Disease-associated Neoplastic Lesions Is Associated with Hypermethylation and Diminished Expression of the DNA Mismatch Repair Gene, hMLH11. Cancer Res. 2000, 60 (17), 4864–4868. [PubMed] [Google Scholar]
  73. Chang C. L.; Marra G.; Chauhan D. P.; Ha H. T.; Chang D. K.; Ricciardiello L.; Randolph A.; Carethers J. M.; Boland C. R. Oxidative stress inactivates the human DNA mismatch repair system. Am. J. Physiol Cell Physiol 2002, 283 (1), 148–154. 10.1152/ajpcell.00422.2001. [DOI] [PubMed] [Google Scholar]
  74. Krewski D.; Al-Zoughool M.; Bird M.; Birkett N.; Billard M.; Milton B.; Rice J. M.; Cogliano V. J.; Hill M. A.; Little J.; et al. Analysis of key characteristics of human carcinogens. In Tumour Site Concordance and Mechanisms of Carcinogenesis; International Agency for Research on Cancer, 2019; Vol. 165. [PubMed] [Google Scholar]
  75. Elchuri S.; Oberley T. D.; Qi W.; Eisenstein R. S.; Jackson Roberts L.; Van Remmen H.; Epstein C. J.; Huang T.-T. CuZnSOD deficiency leads to persistent and widespread oxidative damage and hepatocarcinogenesis later in life. Oncogene 2005, 24 (3), 367–380. 10.1038/sj.onc.1208207. [DOI] [PubMed] [Google Scholar]
  76. Busuttil R. A.; Garcia A. M.; Cabrera C.; Rodriguez A.; Suh Y.; Kim W. H.; Huang T.-T.; Vijg J. Organ-Specific Increase in Mutation Accumulation and Apoptosis Rate in CuZn-Superoxide Dismutase–Deficient Mice. Cancer Res. 2005, 65 (24), 11271–11275. 10.1158/0008-5472.CAN-05-2980. [DOI] [PubMed] [Google Scholar]
  77. Neumann C. A.; Krause D. S.; Carman C. V.; Das S.; Dubey D. P.; Abraham J. L.; Bronson R. T.; Fujiwara Y.; Orkin S. H.; Van Etten R. A. Essential role for the peroxiredoxin Prdx1 in erythrocyte antioxidant defence and tumour suppression. Nature 2003, 424 (6948), 561–565. 10.1038/nature01819. [DOI] [PubMed] [Google Scholar]
  78. Rani V.; Neumann C. A.; Shao C.; Tischfield J. A. Prdx1 deficiency in mice promotes tissue specific loss of heterozygosity mediated by deficiency in DNA repair and increased oxidative stress. Mutat. Res. 2012, 735 (1), 39–45. 10.1016/j.mrfmmm.2012.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Chu F. F.; Esworthy R. S.; Chu P. G.; Longmate J. A.; Huycke M. M.; Wilczynski S.; Doroshow J. H. Bacteria-Induced Intestinal Cancer in Mice with Disrupted Gpx1 and Gpx2 Genes. Cancer Res. 2004, 64 (3), 962–968. 10.1158/0008-5472.CAN-03-2272. [DOI] [PubMed] [Google Scholar]
  80. Constan A. A.; Sprankle C. S.; Peters J. M.; Kedderis G. L.; Everitt J. I.; Wong B. A.; Gonzalez F. L.; Butterworth B. E. Metabolism of Chloroform by Cytochrome P450 2E1 Is Required for Induction of Toxicity in the Liver, Kidney, and Nose of Male Mice. Toxicol. Appl. Pharmacol. 1999, 160 (2), 120–126. 10.1006/taap.1999.8756. [DOI] [PubMed] [Google Scholar]
  81. Bradford B. U.; Kono H.; Isayama F.; Kosyk O.; Wheeler M. D.; Akiyama T. E.; Bleye L.; Krausz K. W.; Gonzalez F. J.; Koop D. R.; et al. Cytochrome P450 CYP2E1, but not nicotinamide adenine dinucleotide phosphate oxidase, is required for ethanol-induced oxidative DNA damage in rodent liver. Hepatology 2005, 41 (2), 336–344. 10.1002/hep.20532. [DOI] [PubMed] [Google Scholar]
  82. Schrum L. W.; Black D.; Iimuro Y.; Rippe R. A.; Brenner D. A.; Behrns K. E. c-Jun Does Not Mediate Hepatocyte Apoptosis Following NFκB Inhibition and Partial Hepatectomy. J. Surg Res. 2000, 88 (2), 142–149. 10.1006/jsre.1999.5784. [DOI] [PubMed] [Google Scholar]
  83. Behrens A.; Sibilia M.; David J. P.; Möhle-Steinlein U.; Tronche F.; Schütz G.; Wagner E. F. Impaired postnatal hepatocyte proliferation and liver regeneration in mice lacking. EMBO J. 2002, 21, 1782–1790. 10.1093/emboj/21.7.1782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Sakurai T.; He G.; Matsuzawa A.; Yu G.; Maeda S.; Hardiman G.; Karin M. Hepatocyte Necrosis Induced by Oxidative Stress and IL-1α Release Mediate Carcinogen-Induced Compensatory Proliferation and Liver Tumorigenesis. Cancer Cell 2008, 14 (2), 156–165. 10.1016/j.ccr.2008.06.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Maeda S.; Kamata H.; Luo J. L.; Leffert H.; Karin M. IKKbeta couples hepatocyte death to cytokine-driven compensatory proliferation that promotes chemical hepatocarcinogenesis. Cell 2005, 121 (7), 977–990. 10.1016/j.cell.2005.04.014. [DOI] [PubMed] [Google Scholar]
  86. Hui L.; Zatloukal K.; Scheuch H.; Stepniak E.; Wagner E. F. Proliferation of human HCC cells and chemically induced mouse liver cancers requires JNK1-dependent p21 downregulation. J. Clin Invest 2008, 118 (12), 3943–3953. 10.1172/JCI37156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Klaunig J. E. Oxidative Stress and Cancer. Curr. Pharm. Des. 2019, 24 (40), 4771. 10.2174/1381612825666190215121712. [DOI] [PubMed] [Google Scholar]
  88. Charbotel B.; Fevotte J.; Hours M.; Martin J. L.; Bergeret A. Case-control study on renal cell cancer and occupational exposure to trichloroethylene. Part II: Epidemiological aspects. Ann. Occup. Hyg. 2006, 50 (8), 777–787. 10.1093/annhyg/mel039. [DOI] [PubMed] [Google Scholar]
  89. IARC . Pharmaceuticals, Vol. 100A, A review of human carcinogens; IARC, World Health Organization: Lyon, France, 2012. https://publications.iarc.fr/118 (accessed 2022-06-21).
  90. Demers P. A.; Davies H. W.; Friesen M. C.; Hertzman C.; Ostry A.; Hershler R.; Teschke K. Cancer and occupational exposure to pentachlorophenol and tetrachlorophenol (Canada). Cancer Causes Control 2006, 17 (6), 749–758. 10.1007/s10552-006-0007-9. [DOI] [PubMed] [Google Scholar]
  91. IARC . Polychlorinated Biphenyls and Polybrominated Biphenyls Vol. 107; IARC, World Health Organization: Lyon, France, 2016. https://publications.iarc.fr/131 (accessed 2022-06-21).
  92. US EPA . Toxicological review of trichloroethylene (CAS No. 79-01-6) in support of summary Information on the Integrated Risk Information System (IRIS). EPA635R09011F; US EPA: Washington, DC, 2011. https://cfpub.epa.gov/ncea/iris/iris_documents/documents/toxreviews/0199tr/0199tr.pdf (accessed 2022-09-29).
  93. Bruning T.; Pesch B.; Wiesenhutter B.; Rabstein S.; Lammert M.; Baumuller A.; Bolt H. M. Renal cell cancer risk and occupational exposure to trichloroethylene: results of a consecutive case-control study in Arnsberg, Germany. Am. J. Ind. Med. 2003, 43 (3), 274–285. 10.1002/ajim.10185. [DOI] [PubMed] [Google Scholar]
  94. Scott C. S.; Jinot J. Trichloroethylene and cancer: systematic and quantitative review of epidemiologic evidence for identifying hazards. Int. J. Environ. Res. Public Health 2011, 8 (11), 4238–4272. 10.3390/ijerph8114238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Karami S.; Lan Q.; Rothman N.; Stewart P. A.; Lee K. M.; Vermeulen R.; Moore L. E. Occupational trichloroethylene exposure and kidney cancer risk: a meta-analysis. Occup Environ. Med. 2012, 69 (12), 858–867. 10.1136/oemed-2012-100932. [DOI] [PubMed] [Google Scholar]
  96. Hansen J.; Sallmen M.; Selden A. I.; Anttila A.; Pukkala E.; Andersson K.; Bryngelsson I. L.; Raaschou-Nielsen O.; Olsen J. H.; McLaughlin J. K. Risk of cancer among workers exposed to trichloroethylene: analysis of three Nordic cohort studies. J. Natl. Cancer Inst 2013, 105 (12), 869–877. 10.1093/jnci/djt107. [DOI] [PubMed] [Google Scholar]
  97. Ahmed N. S.; Samec M.; Liskova A.; Kubatka P.; Saso L. Tamoxifen and oxidative stress: an overlooked connection. Discov Oncol 2021, 12 (1), 17. 10.1007/s12672-021-00411-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Heusinkveld H. J.; Wackers P. F. K.; Schoonen W. G.; van der Ven L.; Pennings J. L. A.; Luijten M. Application of the comparison approach to open TG-GATEs: A useful toxicogenomics tool for detecting modes of action in chemical risk assessment. Food Chem. Toxicol. 2018, 121, 115–123. 10.1016/j.fct.2018.08.007. [DOI] [PubMed] [Google Scholar]
  99. Zheng R.; Zhang Q.; Zhang Q.; Yang L.; Zhang Z.; Huang F. Occupational exposure to pentachlorophenol causing lymphoma and hematopoietic malignancy for two generations. Toxicol Ind. Health 2015, 31 (4), 328–342. 10.1177/0748233712472520. [DOI] [PubMed] [Google Scholar]
  100. Cheng P.; Zhang Q.; Shan X.; Shen D.; Wang B.; Tang Z.; Jin Y.; Zhang C.; Huang F. Cancer risks and long-term community-level exposure to pentachlorophenol in contaminated areas, China. Environ. Sci. Pollut Res. Int. 2015, 22 (2), 1309–1317. 10.1007/s11356-014-3469-4. [DOI] [PubMed] [Google Scholar]
  101. Fraser D. L.; Stander B. A.; Steenkamp V. Cytotoxic activity of pentachlorophenol and its active metabolites in SH-SY5Y neuroblastoma cells. Toxicol In Vitro 2019, 58, 118–125. 10.1016/j.tiv.2019.03.024. [DOI] [PubMed] [Google Scholar]
  102. Tasaki M.; Kuroiwa Y.; Inoue T.; Hibi D.; Matsushita K.; Ishii Y.; Maruyama S.; Nohmi T.; Nishikawa A.; Umemura T. Oxidative DNA damage and in vivo mutagenicity caused by reactive oxygen species generated in the livers of p53-proficient or -deficient gpt delta mice treated with non-genotoxic hepatocarcinogens. Journal of Applied Toxicology 2013, 33 (12), 1433–1441. 10.1002/jat.2807. [DOI] [PubMed] [Google Scholar]
  103. Hansen J.; Raaschou-Nielsen O.; Christensen J. M.; Johansen I.; McLaughlin J. K.; Lipworth L.; Blot W. J.; Olsen J. H. Cancer incidence among Danish workers exposed to trichloroethylene. J. Occup Environ. Med. 2001, 43 (2), 133–139. 10.1097/00043764-200102000-00012. [DOI] [PubMed] [Google Scholar]
  104. Toyooka T.; Yanagiba Y.; Ibuki Y.; Wang R. S. Trichloroethylene exposure results in the phosphorylation of histone H2AX in a human hepatic cell line through cytochrome P450 2E1-mediated oxidative stress. J. Appl. Toxicol 2018, 38 (9), 1224–1232. 10.1002/jat.3632. [DOI] [PubMed] [Google Scholar]
  105. Rehman M. U.; Tahir M.; Quaiyoom Khan A.; Khan R.; Lateef A.; Hamiza O. O.; Ali F.; Sultana S. Diosmin protects against trichloroethylene-induced renal injury in Wistar rats: plausible role of p53. Bax and caspases. Br J. Nutr 2013, 110 (4), 699–710. 10.1017/S0007114512005752. [DOI] [PubMed] [Google Scholar]
  106. Lerro C. C.; Beane Freeman L. E.; DellaValle C. T.; Andreotti G.; Hofmann J. N.; Koutros S.; Parks C. G.; Shrestha S.; Alavanja M. C. R.; Blair A.; et al. Pesticide exposure and incident thyroid cancer among male pesticide applicators in agricultural health study. Environ. Int. 2021, 146, 106187. 10.1016/j.envint.2020.106187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Kachuri L.; Beane Freeman L. E.; Spinelli J. J.; Blair A.; Pahwa M.; Koutros S.; Hoar Zahm S.; Cantor K. P.; Weisenburger D. D.; Pahwa P.; et al. Insecticide use and risk of non-Hodgkin lymphoma subtypes: A subset meta-analysis of the North American Pooled Project. Int. J. Cancer 2020, 147 (12), 3370–3383. 10.1002/ijc.33164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Alavanja M. C. R.; Hofmann J. N.; Lynch C. F.; Hines C. J.; Barry K. H.; Barker J.; Buckman D. W.; Thomas K.; Sandler D. P.; Hoppin J. A. Non-Hodgkin Lymphoma Risk and Insecticide, Fungicide and Fumigant Use in the Agricultural Health Study. PLoS One 2014, 9 (10), e109332. 10.1371/journal.pone.0109332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Padma V. V.; Baskaran R.; Roopesh R. S.; Poornima P. Quercetin attenuates lindane induced oxidative stress in Wistar rats. Mol. Biol. Rep 2012, 39 (6), 6895–6905. 10.1007/s11033-012-1516-0. [DOI] [PubMed] [Google Scholar]
  110. Jamhiri I.; Saadat I.; Omidvari S. Genetic polymorphisms of superoxide dismutase-1 A251G and catalase C-262T with the risk of colorectal cancer. Mol. Biol. Res. Commun. 2017, 6 (2), 85–90. [PMC free article] [PubMed] [Google Scholar]
  111. Jiao L.; Bondy M. L.; Hassan M. M.; Wolff R. A.; Evans D. B.; Abbruzzese J. L.; Li D. Selected polymorphisms of DNA repair genes and risk of pancreatic cancer. Cancer Detect Prev 2006, 30 (3), 284–291. 10.1016/j.cdp.2006.05.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Hung R. J.; Hall J.; Brennan P.; Boffetta P. Genetic polymorphisms in the base excision repair pathway and cancer risk: a HuGE review. Am. J. Epidemiol 2005, 162 (10), 925–942. 10.1093/aje/kwi318. [DOI] [PubMed] [Google Scholar]
  113. Xu J.; Zheng S. L.; Turner A.; Isaacs S. D.; Wiley K. E.; Hawkins G. A.; Chang B. L.; Bleecker E. R.; Walsh P. C.; Meyers D. A.; et al. Associations between hOGG1 sequence variants and prostate cancer susceptibility. Cancer Res. 2002, 62 (8), 2253–2257. [PubMed] [Google Scholar]
  114. Naidu M. S. K.; Suryakar A. S.; Swami S. C.; Katkam R. V.; Kumbar K. M. Oxidative stress and antioxidant status in cervical cancer patients. Indian J. Clin Biochem 2007, 22 (2), 140–144. 10.1007/BF02913333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Yang Y. W.; Dai C. M.; Chen X. H.; Feng J. F. The Relationship between Serum Trace Elements and Oxidative Stress of Patients with Different Types of Cancer. Oxid Med. Cell Longev 2021, 2021, 1. 10.1155/2021/4846951. [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Battisti V.; Maders L. D. K.; Bagatini M. D.; Reetz L. G. B.; Chiesa J.; Battisti I. E.; Gonçalves J. F.; Duarte M. M. F.; Schetinger M. R. C.; Morsch V. M. Oxidative stress and antioxidant status in prostate cancer patients: Relation to Gleason score, treatment and bone metastasis. Biomed Pharmacother 2011, 65 (7), 516–524. 10.1016/j.biopha.2011.06.003. [DOI] [PubMed] [Google Scholar]
  117. Poungpairoj P.; Whongsiri P.; Suwannasin S.; Khlaiphuengsin A.; Tangkijvanich P.; Boonla C. Increased Oxidative Stress and RUNX3 Hypermethylation in Patients with Hepatitis B Virus-Associated Hepatocellular Carcinoma (HCC) and Induction of RUNX3 Hypermethylation by Reactive Oxygen Species in HCC Cells. Asian Pac J. Cancer Prev 2015, 16 (13), 5343–5348. 10.7314/APJCP.2015.16.13.5343. [DOI] [PubMed] [Google Scholar]
  118. Tsamesidis I.; Pantaleo A.; Pekou A.; Gusani A.; Iliadis S.; Manca A.; Carruale A.; Lymperaki E.; Fozza C. Correlation of oxidative stress biomarkers and hematological parameters in blood cancer patients from Sardinia Italy. Int. J. Hematol Oncol Stem Cell Res. 2019, 13 (2), 49–57. [PMC free article] [PubMed] [Google Scholar]
  119. Ahmad R.; Tripathi A. K. T.; Singh R.; Singh S.; Singh R. K. Studies on lipid peroxidation and non-enzymatic antioxidant status as indices of oxidative stress in patients with chronic leukaemia. Singapore Med. Journal 2010, 51 (2), 110–115. [PubMed] [Google Scholar]
  120. Nathan F. M.; Singh V. A.; Dhanoa A.; Palanisamy U. D. Oxidative stress and antioxidant status in primary bone and soft tissue sarcoma. BMC Cancer 2011, 11, 382. 10.1186/1471-2407-11-382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. Mahdavi R.; Faramarzi E.; Seyedrezazadeh E.; Mohammad-Zadeh M.; Pourmoghaddam M. Evaluation of oxidative stress, antioxidant status and serum vitamin C levels in cancer patients. Biol. Trace Elem Res. 2009, 130 (1), 1–6. 10.1007/s12011-008-8309-2. [DOI] [PubMed] [Google Scholar]
  122. Singh R. K.; Tripathi A. K.; Tripathi P.; Singh S.; Singh R.; Ahmad R. Studies on biomarkers for oxidative stress in patients with chronic myeloid leukemia. Hematol Oncol Stem Cell Ther 2009, 2 (1), 285–288. 10.1016/S1658-3876(09)50039-8. [DOI] [PubMed] [Google Scholar]
  123. Rose Li Y.; Halliwill K. D.; Adams C. J.; Iyer V.; Riva L.; Mamunur R.; Jen K. Y.; Del Rosario R.; Fredlund E.; Hirst G.; et al. Mutational signatures in tumours induced by high and low energy radiation in Trp53 deficient mice. Nat. Commun. 2020, 11 (1), 394. 10.1038/s41467-019-14261-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Tanaka H.; Fujita N.; Sugimoto R.; Urawa N.; Horiike S.; Kobayashi Y.; Iwasa M.; Ma N.; Kawanishi S.; Watanabe S.; et al. Hepatic oxidative DNA damage is associated with increased risk for hepatocellular carcinoma in chronic hepatitis C. Br. J. Cancer 2008, 98 (3), 580–586. 10.1038/sj.bjc.6604204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Ma-On C.; Sanpavat A.; Whongsiri P.; Suwannasin S.; Hirankarn N.; Tangkijvanich P.; Boonla C. Oxidative stress indicated by elevated expression of Nrf2 and 8-OHdG promotes hepatocellular carcinoma progression. Med. Oncol 2017, 34 (4), 57. 10.1007/s12032-017-0914-5. [DOI] [PubMed] [Google Scholar]
  126. Tanner C. M.; Kamel F.; Ross G. W.; Hoppin J. A.; Goldman S. M.; Korell M.; Marras C.; Bhudhikanok G. S.; Kasten M.; Chade A. R.; et al. Rotenone, paraquat, and Parkinson’s disease. Environ. Health Perspect 2011, 119 (6), 866–872. 10.1289/ehp.1002839. [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. Chen Q.; Zhang X.; Zhao J. Y.; Lu X. N.; Zheng P. S.; Xue X. Oxidative damage of the male reproductive system induced by paraquat. J. Biochem Mol. Toxicol 2017, 31 (3), e21870. 10.1002/jbt.21870. [DOI] [PubMed] [Google Scholar]
  128. Lam P. L.; Wong M. M.; Hung L. K.; Yung L. H.; Tang J. C. O.; Lam K. H.; Chung P. Y.; Wong W. Y.; Ho Y. W.; Wong R. S. M.; et al. Miconazole and terbinafine induced reactive oxygen species accumulation and topical toxicity in human keratinocytes. Drug Chem. Toxicol 2022, 45, 834–5. 10.1080/01480545.2020.1778019. [DOI] [PubMed] [Google Scholar]
  129. Yang C.; Lim W.; Song G. Mediation of oxidative stress toxicity induced by pyrethroid pesticides in fish. Comp Biochem Physiol C Toxicol Pharmacol 2020, 234, 108758. 10.1016/j.cbpc.2020.108758. [DOI] [PubMed] [Google Scholar]
  130. Katila N.; Bhurtel S.; Park P. H.; Choi D. Y. Metformin attenuates rotenone-induced oxidative stress and mitochondrial damage via the AKT/Nrf2 pathway. Neurochem. Int. 2021, 148, 105120. 10.1016/j.neuint.2021.105120. [DOI] [PubMed] [Google Scholar]
  131. Webster F.; Lambert I. B.; Yauk C. L.. Cyp2E1 Activation Leading to Liver Cancer. Society for the Advancement of Adverse Outcome Pathways, 2017. https://aopwiki.org/aops/220 (accessed 2022-03-28).
  132. Leung T.; Rajendran R.; Singh S.; Garva R.; Krstic-Demonacos M.; Demonacos C. Cytochrome P450 2E1 (CYP2E1) regulates the response to oxidative stress and migration of breast cancer cells. Breast Cancer Res. 2013, 15 (6), R107. 10.1186/bcr3574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Veith A.; Moorthy B. Role of cytochrome P450s in the generation and metabolism of reactive oxygen species. Curr. Opin Toxicol 2018, 7, 44–51. 10.1016/j.cotox.2017.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Kopf P. G.; Walker M. K. 2,3,7,8-Tetrachlorodibenzo-p-dioxin increases reactive oxygen species production in human endothelial cells via induction of cytochrome P4501A1. Toxicol. Appl. Pharmacol. 2010, 245 (1), 91–99. 10.1016/j.taap.2010.02.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. Patlevic P.; Vaskova J.; Svorc P. Jr; Vasko L.; Svorc P. Reactive oxygen species and antioxidant defense in human gastrointestinal diseases. Integr Med. Res. 2016, 5 (4), 250–258. 10.1016/j.imr.2016.07.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Baby B.; Antony P.; Vijayan R. Antioxidant and anticancer properties of berries. Crit Rev. Food Sci. Nutr 2018, 58 (15), 2491–2507. 10.1080/10408398.2017.1329198. [DOI] [PubMed] [Google Scholar]
  137. Cheung E. C.; Vousden K. H. The role of ROS in tumour development and progression. Nat. Rev. Cancer 2022, 22 (5), 280–297. 10.1038/s41568-021-00435-0. [DOI] [PubMed] [Google Scholar]
  138. Griess B.; Tom E.; Domann F.; Teoh-Fitzgerald M. Extracellular superoxide dismutase and its role in cancer. Free Radic Biol. Med. 2017, 112, 464–479. 10.1016/j.freeradbiomed.2017.08.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  139. Sepasi Tehrani H.; Moosavi-Movahedi A. A. Catalase and its mysteries. Prog. Biophys. Mol. Biol. 2018, 140, 5–12. 10.1016/j.pbiomolbio.2018.03.001. [DOI] [PubMed] [Google Scholar]
  140. Chang C.; Worley B. L.; Phaeton R.; Hempel N. Extracellular Glutathione Peroxidase GPx3 and Its Role in Cancer. Cancers 2020, 12 (8), 2197. 10.3390/cancers12082197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Kaweme N. M.; Zhou S.; Changwe G. J.; Zhou F. The significant role of redox system in myeloid leukemia: from pathogenesis to therapeutic applications. Biomark Res. 2020, 8 (1), 63. 10.1186/s40364-020-00242-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  142. Hauck A. K.; Bernlohr D. A. Oxidative stress and lipotoxicity. J. Lipid Res. 2016, 57 (11), 1976–1986. 10.1194/jlr.R066597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Jabłońska-Trypuć A. Pesticides as Inducers of Oxidative Stress. React. Oxyg Species 2017, 3 (8), 96–110. 10.20455/ros.2017.823. [DOI] [Google Scholar]
  144. Cermak A. M. M.; Pavicic I.; Zeljezic D. Redox imbalance caused by pesticides: a review of OPENTOX-related research. Arh Hig Rada Toksikol 2018, 69 (2), 126–134. 10.2478/aiht-2018-69-3105. [DOI] [PubMed] [Google Scholar]
  145. Patinen T.; Adinolfi S.; Cortes C. C.; Harkonen J.; Jawahar Deen A.; Levonen A. L. Regulation of stress signaling pathways by protein lipoxidation. Redox Biol. 2019, 23, 101114. 10.1016/j.redox.2019.101114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  146. Vatsyayan R.; Chaudhary P.; Sharma A.; Sharma R.; Rao Lelsani P. C.; Awasthi S.; Awasthi Y. C. Role of 4-hydroxynonenal in epidermal growth factor receptor-mediated signaling in retinal pigment epithelial cells. Exp. Eye Res. 2011, 92 (2), 147. 10.1016/j.exer.2010.11.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. Bartsch H.; Nair J. Oxidative stress and lipid peroxidation-derived DNA-lesions in inflammation driven carcinogenesis. Cancer Detect Prev 2004, 28 (6), 385. 10.1016/j.cdp.2004.07.004. [DOI] [PubMed] [Google Scholar]
  148. Yadav U. C.; Ramana K. V. Regulation of NF-kappaB-induced inflammatory signaling by lipid peroxidation-derived aldehydes. Oxid Med. Cell Longev 2013, 2013, 1. 10.1155/2013/690545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  149. Lei L.; Yang J.; Zhang J.; Zhang G. The lipid peroxidation product EKODE exacerbates colonic inflammation and colon tumorigenesis. Redox Biol. 2021, 42, 101880 10.1016/j.redox.2021.101880. [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Deavall D. G.; Martin E. A.; Horner J. M.; Roberts R. Drug-induced oxidative stress and toxicity. J. Toxicol 2012, 2012, 645460 10.1155/2012/645460. [DOI] [PMC free article] [PubMed] [Google Scholar]
  151. Chen A. C. H.; Burr L.; McGuckin M. A. Oxidative and endoplasmic reticulum stress in respiratory disease. Clin Transl Immunology 2018, 7 (6), e1019. 10.1002/cti2.1019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Bruce J. I.; Elliott A. C. Oxidant-impaired intracellular Ca2+ signaling in pancreatic acinar cells: role of the plasma membrane Ca2+-ATPase. Am. J. Physiol Cell Physiol 2007, 293 (3), C938. 10.1152/ajpcell.00582.2006. [DOI] [PubMed] [Google Scholar]
  153. Nishida N.; Arizumi T.; Takita M.; Kitai S.; Yada N.; Hagiwara S.; Inoue T.; Minami Y.; Ueshima K.; Sakurai T.; et al. Reactive oxygen species induce epigenetic instability through the formation of 8-hydroxydeoxyguanosine in human hepatocarcinogenesis. Dig Dis 2013, 31 (5–6), 459–466. 10.1159/000355245. [DOI] [PubMed] [Google Scholar]
  154. Kontomanolis E. N.; Koutras A.; Syllaios A.; Schizas A.; Mastoraki A.; Garmpis N.; Diakosavvas M.; Angelou K.; Tsatsaris G.; Pagkalos A.; et al. Role of Oncogenes and Tumor-suppressor Genes in Carcinogenesis: A Review. Anticancer Res. 2020, 40 (11), 6009. 10.21873/anticanres.14622. [DOI] [PubMed] [Google Scholar]
  155. Boobis A. R.; Daston G. P.; Preston R. J.; Olin S. S. Application of key events analysis to chemical carcinogens and noncarcinogens. Crit Rev. Food Sci. Nutr 2009, 49 (8), 690–707. 10.1080/10408390903098673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  156. Meek M. E.; Boobis A.; Cote I.; Dellarco V.; Fotakis G.; Munn S.; Seed J.; Vickers C. New developments in the evolution and application of the WHO/IPCS framework on mode of action/species concordance analysis. J. Appl. Toxicol 2014, 34 (1), 1–18. 10.1002/jat.2949. [DOI] [PMC free article] [PubMed] [Google Scholar]
  157. OECD . Handbook supplement to the Guidance Document for developing and assessing Adverse Outcome Pathways; OECD: Paris, 2018. https://www.oecd-ilibrary.org/environment/users-handbook-supplement-to-the-guidance-document-for-developing-and-assessing-adverse-outcome-pathways_5jlv1m9d1g32-en (accessed 2022-03-21).
  158. Collaborative Adverse Outcome Pathway Wiki (AOP-Wiki), 2021. https://aopwiki.org/ (accessed 2022-07-01).
  159. Brenner C.; Galluzzi L.; Kepp O.; Kroemer G. Decoding cell death signals in liver inflammation. J. Hepatol 2013, 59 (3), 583–594. 10.1016/j.jhep.2013.03.033. [DOI] [PubMed] [Google Scholar]
  160. Fang Y.; Tian S.; Pan Y.; Li W.; Wang Q.; Tang Y.; Yu T.; Wu X.; Shi Y.; Ma P.; et al. Pyroptosis: A new frontier in cancer. Biomed Pharmacother 2020, 121, 109595 10.1016/j.biopha.2019.109595. [DOI] [PubMed] [Google Scholar]
  161. Xia X.; Wang X.; Cheng Z.; Qin W.; Lei L.; Jiang J.; Hu J. The role of pyroptosis in cancer: pro-cancer or pro-″host″?. Cell Death Dis 2019, 10 (9), 650. 10.1038/s41419-019-1883-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  162. Ramachandran A.; Jaeschke H. Acetaminophen Toxicity: Novel Insights Into Mechanisms and Future Perspectives. Gene Expr 2018, 18 (1), 19–30. 10.3727/105221617X15084371374138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Jung S. H.; Lee W.; Park S. H.; Lee K. Y.; Choi Y. J.; Choi S.; Kang D.; Kim S.; Chang T. S.; Hong S. S.; et al. Diclofenac impairs autophagic flux via oxidative stress and lysosomal dysfunction: Implications for hepatotoxicity. Redox Biol. 2020, 37, 101751 10.1016/j.redox.2020.101751. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

tx2c00396_si_001.pdf (413.3KB, pdf)

Articles from Chemical Research in Toxicology are provided here courtesy of American Chemical Society

RESOURCES