Abstract
Reactive oxygen species (ROS) are a class of highly reactive oxidizing molecules, including superoxide anion (O2•−) and hydrogen peroxide (H2O2), among others. Moderate levels of ROS play a crucial role in regulating cellular signaling and maintaining cellular functions. However, abnormal ROS levels or persistent oxidative stress can lead to changes in the tumor microenvironment (TME) that favor cancer development. This review provides an overview of ROS generation, structure, and properties, as well as their effects on various components of the TME. Contrary to previous studies, our findings reveal a dual effect of ROS on different components of the TME, whereby ROS can either enhance or inhibit certain factors, ultimately leading to the promotion or suppression of the TME. For example, H2O2 has dual effects on immune cells and non-cellular components within the TME, while O2•− has dual effects on T cells and fibroblasts. Furthermore, each component demonstrates distinct mechanisms of action and ranges of influence. In the final section of the article, we summarize the current clinical applications of ROS in cancer treatment and identify certain limitations associated with existing therapeutic approaches. Therefore, this review aims to provide a comprehensive understanding of ROS, highlighting their dual effects on different components of the TME, and exploring the potential clinical applications that may pave the way for future treatment and prevention strategies.
Keywords: ROS, tumor, microenvironment, molecule, metabolism, therapy
1. INTRODUCTION
Tumor cells, immune cells, stromal cells (including vascular endothelial cells, fibroblasts, adipocytes, and stellate cells), and non-cellular components (such as the extracellular matrix (ECM), exosomes, and blood vessels) constitute the major constituents of the tumor microenvironment (TME) (Fig. 1). However, the specific composition of these elements can vary depending on the tumor type [1, 2]. Furthermore, the tumor microenvironment (TME) and metabolic reprogramming are recognized as significant characteristics of modern cancer research [3]. The interplay between cancer cells and ROS within the tumor microenvironment govern tumor progression at the intersection of these two pivotal biological phenomena [4-6].
Fig. (1).
Describes the components of TME and the relationship between various components. TME mainly includes immune cells, stromal cells, and non-cellular components such as ECM. The components of TME coordinate with each other and are closely related.
ROS, which are molecules with short lifespans and unpaired electrons, are formed through the partial reduction of molecular oxygen. They encompass oxygen-derived compounds comprising highly labile oxygen radicals such as superoxide (O2•−) and hydroxyl (HO∙) radicals, which can readily transform into more stable, freely diffusible non-radicals like hydrogen peroxide (H2O2) and hypochlorous acid. Furthermore, ROS encompasses 1O2, HOO•, and ClO• [7, 8]. ROS play a crucial role as signaling molecules in regulating the interactions among cellular and non-cellular components within the tumor microenvironment [9]. Nevertheless, recent research has primarily focused on exploring the dual role of ROS specifically within tumor cells [10-12]. A comprehensive examination of the impact of ROS on other components of the TME has not been thoroughly conducted. The primary focus of this paper is to investigate the dual effects of ROS on the various components of the TME.
ROS can create a “favorable” environment for tumor cells by modulating the TME, which is characterized by acidity, hypoxia, nutrient deprivation, and inflammation [13-15]. However, the role of ROS in the tumor microenvironment is multifaceted. This review comprehensively examines the generation, characteristics, and functions of ROS, as well as their impact on cancer development. Initially, it provides an overview of the sources of ROS, encompassing the mitochondrial respiratory chain, NADPH oxidases, and other enzymatic systems, while elucidating the properties and structures of ROS. Subsequently, it delves into the intricate interplay between ROS and the TME. Furthermore, it consolidates the current strategies for ROS-targeted cancer therapy and sheds light on their inherent limitations. Lastly, it synthesizes and discusses the patterns and extent of interactions between ROS and the TME, with the ultimate aim of exploring potential clinical applications.
2. THE ORIGINS, COMPOSITION, AND CHARACTERISTICS OF ROS, AS WELL AS THEIR PROPERTIES WITHIN THE TUMOR MICROENVIRONMENT
2.1. The Origin, Structure, and Properties of H2O2
The generation of endogenous H2O2 in cells is intricately associated with mitochondrial activity, the endoplasmic reticulum, and NADPH oxidase [16]. The superoxide radical is produced within the constituents of complex I and complex III of the mitochondrial electron transport chain [17]. Moreover, the superoxide radical can serve as a precursor for the production of H2O2 [18], which is then converted to H2O2 through the action of manganese superoxide dismutase (MnSOD) [19]. H2O2-producing enzymes are also present in the lumen of the endoplasmic reticulum. The most prevalent enzyme source is the flavoprotein ERO1, which collaborates with members of the protein disulfide isomerase (PDI) family of oxidoreductases. ERO1 facilitates the formation of disulfide bonds during the folding of newly synthesized polypeptides. Electrons released during disulfide bond formation are transferred from the nascent secretory protein to ERO1 via PDI. Subsequently, H2O2 is generated as ERO1 transfers the received electrons from PDI to molecular oxygen [20]. NADPH oxidase facilitates the transfer of electrons from NADPH to two distinct heme moieties through flavin adenine dinucleotide. This electron transfer process occurs towards molecular oxygen, serving as the electron acceptor. The resulting multimeric complex ultimately generates either superoxide or H2O2 [21]. Likewise, superoxide is catalyzed by superoxide dismutase (SOD), leading to the subsequent production of H2O2 [22]. Therefore, the collective production of H2O2 in the mitochondria, endoplasmic reticulum, and NADPH oxidase contributes to the overall oxidative stress response in the organism.
H2O2, with the chemical formula H-O-O-H, possesses a polar molecule structure consisting of both polar and nonpolar bonds. The oxygen atom (O) in H2O2 carries an intermediate valence of -1. Due to its specific spatial arrangement, H2O2 adopts a permanent dipole conformation, resulting in the molecule having a distinct separation of positive and negative charges [23]. Furthermore, the presence of a permanent dipole in H2O2 may have implications for cellular motor function and transport processes [24]. This structural characteristic is intricately linked to the impact of H2O2 on the invasion and metastasis of cancer cells as described below.
The properties of H2O2 in the microenvironment encompass its oxidizing, conductive, and transformative characteristics. H2O2's inherent oxidative activity is manifested through the degradation of hemoglobin, enzyme inactivation, and release of iron, as well as the oxidation of lipids, DNA, ketones, and -SH groups [25]. When H2O2 induces oxidative stress in various components of the tumor microenvironment, it can generate a diverse array of “nutrients” that promote tumor growth. H2O2 demonstrates excellent conductivity within the TME and may be the most suitable oxygen metabolite for signaling among various oxygen metabolites. This may be attributed to H2O2's ability to trigger multiple sensors and pathways during the signaling process, as well as regulate transcription factors at various levels. For instance, H2O2 synthesizes transcription factors by upregulating transcription or increasing mRNA stability and translation or activates individual genes for DNA binding and nuclear transcription by targeting specific chromosomal regions [26, 27]. Given H2O2's numerous transduction targets and mechanisms, it frequently operates as a messenger in cell proliferation induction, mechanism activation, apoptosis, and anti-apoptosis within the TME. It is noteworthy that H2O2, in its role as a signal transmitter, induces cellular apoptosis through a concentration-dependent pathway. In a study exploring H2O2-induced apoptosis in melanoma cells, it was discovered that low concentrations of H2O2 initially activate the caspase-dependent apoptotic pathway. Conversely, high concentrations of H2O2 trigger cell death and cancer cell apoptosis via a caspase-independent signaling pathway [28]. H2O2 is also capable of transformation. In a study, overexpression of rat acyl-coenzyme A oxidase (ACOX) was allowed in the non-tumorigenic rat uroepithelial cell line MYP3, leading to high levels of H2O2 production in the organism. The resulting study demonstrated that H2O2 exhibited tumorigenicity in thymus-free nude mice, suggesting a malignant transforming effect of H2O2 on normal cells [29]. These properties are closely associated with the effects of H2O2 on the TME discussed later in the text.
2.2. The Origin, Structure, and Properties of O2•−
Superoxide (O2•−), an intermediate reactive oxygen species, is primarily generated in isolated mitochondria, with complex I (ubiquinone oxidoreductase) and complex III (cytochrome bc1 complex; ubiquinone: cytochrome oxidoreductase) serving as the main sites of its production. Complex I, a multi-component complex consisting of 45 polypeptides, functions as the entry point for electrons derived from nicotinamide adenine dinucleotide (NADH) into the respiratory chain [30, 31]. Complex I generate substantial quantities of O2•− through two mechanisms. One of these mechanisms involves the reaction of isolated complex I with fully reduced flavin mononucleotide (FMN), resulting in the production of O2•−. The extent of FMN reduction depends on the ratio of NADH to nicotinamide adenine dinucleotide (NAD) [32]. The rate of superoxide anion generation through complex I is regulated by the NADH/NAD ratio, which governs the electron transfer process [33]. Another mechanism involves the generation of O2•− by complex I during reverse electron transport. In this process, a high membrane potential drives electrons to flow backward from coenzyme Q (CoQH2) to complex I, resulting in the reduction of NAD to NADH at the FMN site and the production of O2•− [34]. Ultimately, a large amount of O2•− is produced from complex-I [35]. The ubiquinone oxidation center within complex III is responsible for the production of superoxide. The cytochrome bc1 complex, which consists of two homodimers, forms two ubiquinone reaction centers from cytochrome b on each side of the membrane [36]. It forms the ubiquinone oxidation center, also known as center P or Qo site, on the positive side of the membrane, and the ubiquinone reduction center, also known as center N or Qi site, on the negative side of the membrane [37]. Moreover, the generation of the superoxide anion occurs in the ubiquinone oxidation center [38].
O2•− comprises a single anion and a single radical [39]. The O-O bond length is 1.33 pm, and there is a σ bond and a 3-electron π bond within the ion. The 17-electron O2•− is a separable paramagnetic main group ion and is one of many oxygen reduction reaction intermediate species [40].
The O2•− possesses oxidizing and signaling activating properties. Its oxidative nature is attributed to its nucleophilic characteristics. When compounds in the tumor microenvironment are present, capable of donating hydrogen (H), such as tocopherol, ascorbic acid, or other positively charged substances, O2•− demonstrates strong nucleophilic reactivity. It selectively targets positively charged centers, initiating chemical reactions [25]. The nucleophilic nature of O2•− contributes to the induction of oxidative stress in stromal cells. Additionally, O2•− possesses the ability to activate signaling pathways that have profound effects on cellular processes. For example, when exposed to O2•− and its intermediate oxidizing metabolites produced by phagocytes, the fibrosarcoma QR-32 cell line demonstrates metastatic behavior, indicating the involvement of O2•− in signaling and activation pathways. This can be attributed to the role of O2•− as a conductor of activating signals, which triggers the activation of nicotinamide adenine dinucleotide phosphate oxidase, ultimately leading to the transformation of QR-32 fibrosarcoma cells into metastatic tumors [41]. Another illustrative example is the stimulation of proliferation in lung cancer cells, wherein O2•− serves as an intracellular messenger within the ROS oncogenic signaling cascade. This highlights the involvement of O2•− in facilitating cell proliferation and underscores its significance as a key player in intracellular signaling processes associated with cancer development [42]. In cases of leukemia, O2•− functions as a messenger for activation signals, initiating crucial signaling pathways such as the mitogen-activated protein kinase (MAPK) and mammalian target of rapamycin (mTOR) upstream pathways. This activation, in turn, leads to apoptosis in leukemic cells, further emphasizing the pivotal role of O2•− in regulating cellular processes and influencing disease outcomes [43].
2.3. The Origin, Structure, and Properties of HO∙
Hydroxyl radical (HO∙) is produced within mitochondria through the Fenton and Haber-Weiss chemical reactions. In the Fenton reaction (Equation 1), H2O2 reacts with Fe2+ of low valence to generate HO∙, facilitated by the reduction of iron by O2•− in the presence of an iron catalyst (Equation 2). Additionally, ligands that promote redox cycling can enhance the chelation of iron catalysts, further influencing the generation of HO∙ [44].
Fe2+(ligand)+H2O2→Fe3+(ligand) + OH-+ HO• Eq. (1)
Fe3+(ligand)+O2•−→Fe2+(ligand) + O2 Eq. (2)
The combined reaction of Equation 1 and Equation 2 is called the iron-catalyzed Haber-Weiss reaction [45]. It is another way to generate HO∙.
Fe2+/Fe3+
O2•−+H2O2 O2+OH-+HO•
HO• and its secondary product radicals possess one or more unpaired electrons, rendering them highly chemically reactive. As a result, they can engage in redox reactions with various substances within living organisms [46].
In the microenvironment, the HO∙ exhibits oxidative, conductive, and highly reactive properties. HO∙ is known for its strong oxidizing nature and its rapid reactivity with a wide range of inorganic and organic molecules in cells. These include proteins, amino acids, DNA, lipids, metals, and sugars [25]. These characteristics of HO∙ are closely associated with its ability to modify DNA, induce DNA base lesions, and contribute to oxidative stress in the tumor microenvironment [47-49]. HO∙ exhibits signaling properties, as demonstrated by Sumkhemthong's research group. Specifically, HO∙ functions as a signaling molecule that mediates the autophagic response in lung cancer cells. It confers resistance to the apoptotic effect of the drug cisplatin on cancer cells and promotes cell survival [50]. HO∙, being the most active free radical in biological systems, exhibits high reactivity and instability. Unlike the relatively stable O2•−, HO∙ is a short-lived species that readily interacts with other molecules. In studies involving apoptosis in HeLa, MW451, and 293 cells, it was observed that HO∙ could directly target telomeres, leading to telomere shortening and induction of apoptosis in cancer cells [51].
2.4. The Origin, Structure, and Properties of 1O2
There are two main mechanisms for the production of 1O2. The first mechanism involves macrophages (including neutrophils, heart cells, and endothelial cells) accumulating in tissues exposed to oxidative stress or in various disease states. In this process, macrophages generate 1O2 through a spontaneous disproportionation reaction of O2•−, with the involvement of H2O2 and NO•. Additionally, the microenvironment plays a significant role in extracellular 1O2 production, and research has demonstrated that an acidic cellular environment enhances 1O2 production by macrophages.
The second mechanism of 1O2 generation occurs in the erythropoietin (EPO)-H2O2-bromide system and the Myeloperoxidase (MPO)-H2O2-chloride system [52] (Fig. 2).
Fig. (2).
Describes the ROS formation pathway in the body. The superoxide anion is mainly produced in complex I and complex III in mitochondria and relies on NADPH enzymosomes to transfer hydrogen. Hydrogen peroxide is mainly produced by superoxide anion catalyzed by SOD or by electron transfer from ERO1 and PDI enzymes of the endoplasmic reticulum to molecular oxygen. Hydrogen peroxide further undergoes the Fenton and Haber reactions to produce hydroxyl peroxyl. Phagocytic cells, the MPO, and the EPO systems produce singlet oxygen. The four ROS components are not produced independently but have a cascade relationship.
The excited state of molecular oxygen (1O2) is characterized by having two paired electrons in its structure and a change in electron spin within the molecular orbital. It is an extremely unstable and highly reactive species [46, 53].
1O2 is an excited form of molecular oxygen that displays high reactivity towards organic compounds rich in electrons, including polycyclic aromatic compounds, diolefins, and olefins [54]. In a study involving Hepa hepatocellular carcinoma cell lines, it was observed that 1O2 induced the peroxidation of unsaturated fatty acids within the cells. This lipid peroxidation process has been identified as a direct mediator of iron-induced apoptosis. Consequently, the pathway leading to iron-induced apoptosis in cancer cells can be activated by 1O2, acting downstream of the cysteine-glutathione axis [55]. Besides, 1O2 also inactivates SOD and peroxidase on the surface of cancer cells [56, 57], which mediates apoptosis. Due to its exceptional reactivity, 1O2 exhibits significant cytotoxicity. The oxidation of crucial biological targets, such as lipids, proteins, or nucleic acids, by 1O2, can directly trigger cell death [58].
Given the structural and property variations among different ROS components, it is logical to inquire about the potential diversity in their effects on the tumor microenvironment. In the subsequent section, we will thoroughly examine the interplay between each ROS component and the various cellular and non-cellular elements within the TME.
3. ROS ALTER THE CELLULAR COMPONENTS IN THE TUMOR MICROENVIRONMENT
3.1. ROS Exert an Impact on Immune Cells within the Tumor Microenvironment
3.1.1. ROS Suppress T-cell Function and Facilitate the Establishment of the Tumor Microenvironment
H2O2 within the tumor microenvironment plays a significant role in promoting the TME by detrimentally affecting T cells, suppressing their immune functions, and impeding their proliferation, thereby facilitating tumorigenic progression. A study by Tan et al. illustrated this phenomenon in the context of hepatocarcinogenesis, where Lysyl oxidase-like 4 (LOXL4), secreted by tumor cells and predominantly localized in hepatic macrophages through exocytosis, catalyzes the conversion of amine groups, resulting in the generation of H2O2 and ammonia as byproducts in a copper-dependent manner. Consequently, H2O2 activates interferon (IFN)-associated Signal transducers and activators of transcription (STATs), leading to the upregulation of programmed cell death ligand 1 (PD-L1). This process enables macrophages to interact with tumor-killing T cells, hindering T-cell activation, inducing functional impairment, and ultimately suppressing the immune microenvironment [59-61]. In gastric and esophageal cancers, regulatory T (Treg) cells exhibit a notable resistance to apoptosis induced by H2O2 compared to conventional T cells. This characteristic may result in a higher accumulation of Treg cells in the vicinity of the tumor within the tumor microenvironment, rather than an actual increase in the number of Treg cells influenced by chemical factors [62]. Treg cells possess functional immunosuppressive properties within the tumor microenvironment, effectively suppressing immune responses of T effector cells. Moreover, elevated numbers of Treg cells are observed in various malignancies, indicating their increased presence in the tumor microenvironment [63, 64]. Kono et al. conducted a study showing that the secretion of H2O2 by melanoma-associated macrophages or activated monocytes leads to a decrease in the expression of the cell surface CD3 complex. The CD3 complex plays a crucial role in initiating T cell proliferation and cytolytic function. Based on their findings, the researchers proposed that the inhibitory impact of H2O2 on T cell function could be attributed to its influence on the expression of the CD3 complex [65]. In an experiment conducted on glioma cells, it was observed that H2O2 induced the expression of CYB561D2, an antioxidant protein. Additionally, H2O2 activated signal transducer and activator of transcription 3 (STAT3) in glioma cells. The increased expression of CYB561D2 and activation of STAT3 had immunosuppressive effects on T cells. Specifically, CYB561D2 inhibited the secretion of interleukin-2 (IL-2) from T cells in a STAT3-dependent manner. This immunosuppressive effect of H2O2 on T cells, mediated by CYB561D2 and STAT3, could ultimately lead to T cell apoptosis and contribute to the immunosuppressive environment in gliomas [66]. IL4I1, a phenylalanine oxidase, can be expressed by infiltrating macrophages and malignant cells in B-cell lymphomas and non-lymphoid tumors [67]. In vivo, IL4I1, an immune regulatory enzyme, was found to oxidize phenylalanine, resulting in the production of phenyl pyruvic acid and H2O2. This H2O2 generated by IL4I1 exhibited inhibitory effects on T lymphocyte proliferation. Interestingly, H2O2 had a more pronounced inhibitory effect on memory T cells compared to other T cell subsets. It was observed that H2O2 at micromolar concentrations had a greater impact on T lymphocyte proliferation inhibition. This suggests that H2O2, produced by IL4I1-mediated phenylalanine oxidation, plays a role in modulating T cell responses, particularly affecting memory T cells [68]. Indeed, impaired T cell function in tumor-infiltrating lymphocytes (TIL) has been associated with alterations in T-cell receptor (TCR)-mediated signal transduction. In the context of ovarian cancer, Lockhart et al. demonstrated that H2O2 produced by tumor-derived macrophages could contribute to the loss of TCR expression and affect the secretion of high molecular weight factors by certain tumor cells. The loss or decreased expression levels of TCR signaling chains can further impair T cell function, including their ability to recognize and respond to tumor antigens. These modifications in TCR-mediated signal transduction pathways highlight the role of H2O2 in the tumor microenvironment in influencing T cell functionality within the context of ovarian cancer [69]. Other mechanisms by which H2O2 interacts with T cells to inhibit TME are listed in Table 1.
Table 1. The effect of each component of ROS on TME and experimental results.
| ROS Type | Effect | Cancer Type | Components | Experimental Result | References |
|---|---|---|---|---|---|
| H2O2 | promotion | liver cancer | T cells | Impaired immune activation | [59-61] |
| gastric and esophageal cancers | Treg cells T effector cells |
Suppression of immune response | [62-64] | ||
| melanoma | T cells | Inhibition of proliferation | [65] | ||
| glioma | T cells | Immunosuppression and even apoptosis | [66] | ||
| B-cell lymphomas and non-lymphoid tumors | T cells | Inhibition of proliferation | [68] | ||
| ovarian cancer | T cells | Impaired cellular function | [69] | ||
| squamous cell carcinoma of the head and neck | T cells | Inhibition of proliferation and survival | [150] | ||
| melanoma | T cells | Inhibition of cellular responses | [151] | ||
| colon cancer | T cells | Inhibition of cellular responses | [152] | ||
| astrocytoma | T cells | Apoptosis | [153] | ||
| breast cancer | CD8 T-cells | Inhibition of cytotoxicity | [154] | ||
| inhibition | pancreatic cancer | T cells | Enhancement of immunosuppressive function | [73] | |
| mammary tumor | T cells | Enhancement of toxicity | [74] | ||
| colon cancer | T cells | Activation | [75] | ||
| promotion | lymphocytic leukemia | B lymphocytes | Oxidative damage | [79, 80] | |
| gastric and esophageal cancers | CD56dimNK cells | Apoptosis | [81] | ||
| breast cancer | NK cells | Reduced activity | [82] | ||
| melanoma | NK cells | Reduced activity | [65] | ||
| inhibition | mast cell tumor | NK cells | Accelerated Migration | [83] | |
| promotion | liver cancer | Macrophages | Suppression of immune function | [61] | |
| lung cancer | Macrophages | Dodging the immune response | [84] | ||
| inhibition | / | Macrophages | Immune Enhancement | [86] | |
| promotion | prostate cancer | Endothelial cells | Promote proliferation | [97] | |
| colon cancer | Endothelial cells | Promote proliferation | [100] | ||
| esophageal squamous cell carcinoma | Fibroblasts | Induced expression | [102] | ||
| hereditary ovarian cancer | Fibroblasts | Induction of oxidative stress | [103] | ||
| breast cancer | Fibroblasts | Induction of oxidative stress | [104] | ||
| prostate cancer | Fibroblasts | Reduced expression | [110] | ||
| inhibition | nerve tumor | Fibroblasts | Enhanced stimulation | [115] | |
| promotion | prostate cancer | Blood vessel | Increased angiogenesis | [118] | |
| inhibition | / | Blood vessel | Inhibition of angiogenesis | [121] | |
| promotion | breast cancer | ECM | Promote the formation of ECM | [123] | |
| O2•− | promotion | colon cancer | T cells | Inhibition function | [70] |
| melanoma | T cells | Inhibition function | [71] | ||
| inhibition | / | T cells | Increased cytotoxicity | [76] | |
| promotion | Retinoblastoma sarcoma breast cancer |
Macrophages | Regulation of microenvironment | [85] | |
| inhibition | prostate cancer | Macrophages | Apoptosis | [87] | |
| lung cancer | Neutrophils | Increased target cytotoxicity | [89] | ||
| promotion | melanoma | Endothelial cells | Promote proliferation | [101] | |
| breast cancer | Fibroblasts | Promote metabolism | [6, 111, 112] | ||
| breast cancer | Fibroblasts | Facilitated interaction | [113] | ||
| inhibition | prostate cancer | Fibroblasts | Inhibition of proliferation | [116] | |
| promotion | melanoma glioblastoma | Blood vessel | Angiogenesis | [119] | |
| HO∙ | inhibition | / | T cells | Activation of immunity | [77, 78] |
| breast cancer | Macrophages | Suppression of metastasis | [88] | ||
| promotion | stomach cancer | Neutrophils | Inflammatory environment | [92] | |
| inhibition | / | Neutrophils | Promotion of immunity | [90, 91] | |
| promotion | Squamous cell carcinoma of the head and neck | Blood vessel | Angiogenesis | [120] | |
| / | ECM | Invasion | [125] | ||
| 1O2 | promotion | / | T cells | Apoptosis | [72] |
| inhibition | / | ECM | Suppression of metastasis | [124] |
Indeed, O2•− plays a role in promoting the tumor microenvironment by inhibiting T-cell proliferation and immune responses. Bronte et al. demonstrated that arginase 1 (Arg1) is involved in the mechanism by which myeloid suppressor cells (MSC) inhibit T cell proliferation in colon cancer. Arg1 increases O2•− production in myeloid cells through a pathway that may involve the reductase structural domain of inducible NO synthase (iNOS). This increased O2•− production is implicated in the Arg1-dependent inhibition of T-cell function. These findings highlight the contribution of O2•− and the interplay between myeloid cells and T cells in shaping the immune response within the tumor microenvironment, particularly in colon cancer [70]. Furthermore, additional research has indicated that in cases of melanoma, CD14(+) cells exert a suppressive influence on autologous T cells through a STAT-3, prostaglandin E2 (PGE2), and O2•− dependent mechanism. This orchestration of factors not only dampens the immune response but also fosters the progression of tumor development [71].
The impact of 1O2 on T cells has received limited investigation, with only a singular study demonstrating its capacity to induce mutual oxidative damage to telomeres and mitochondria within T cells. This dual effect of damage is contingent upon both the duration and intensity of exposure, ultimately culminating in T-cell senescence and potentially even fatality [72]. Ultimately, it promotes the tumor cell microenvironment.
3.1.2. ROS Augment T-cell Activity and Impede the Progression of the Tumor Microenvironment
H2O2 within the tumor microenvironment exerts its influence by augmenting T-cell immunosuppression and cytotoxicity, while concurrently facilitating T-cell activation to repress the tumor microenvironment. In an experimental model employing in vitro and in vivo approaches, specifically focusing on K-ras-driven pancreatic cancer, Glorieux et al. implemented antioxidants to mitigate H2O2 levels. This intervention significantly attenuated the expression of programmed death-ligand 1 (PD-L1) in K-ras mutant cells, consequently leading to increased T-cell infiltration within tumor tissues. This, in turn, enhanced the immunosuppressive functionality of T cells, resulting in impeded growth of tumor cells [73]. Experimental data indicate that upon exposure of T lymphocytes from mice to mammary cell tumor cells, H2O2 production occurs. Notably, H2O2 becomes concentrated in the vicinity of target cells, rendering the sulfhydryl groups on these cells vulnerable to its effects. Through this mechanism, H2O2 serves as a mediator, amplifying T-cell cytotoxicity and impeding the survival of cancer cells [74]. In a study by Freund et al., it was observed that the treatment of CT26 colon cancer cells with an H2O2 saline solution resulted in the upregulation of cell surface markers associated with anti-tumor immune response (referred to as ICD). This treatment also promoted T-cell activation. Concurrently, the levels of two anti-inflammatory factors, IL4 and IL10, were found to be elevated. Consequently, the growth of colon cancer cells was inhibited, and apoptosis was induced in the cancer cells [75].
The presence of O2•− has been shown to enhance the cytotoxicity of T lymphocytes and suppress the tumor microenvironment. Lu et al. employed high-fluence, low-power laser irradiation (HF-LPLI) as a method to induce a significant release of O2•−. This resulted in the generation of large quantities of O2•−, which in turn extensively modified the phosphatidylserine fraction of oxidized tumor cells. This modification facilitated macrophage uptake and recognition of the tumor cells, ultimately leading to increased cytotoxicity of T lymphocytes and subsequent suppression of tumor growth [76].
The majority of studies examining the impact of HO∙ on T cells within the TME primarily concentrate on therapeutic applications. These investigations explore therapeutic strategies such as chemodynamic therapy, which involves the utilization of synergetic chemodynamic-photothermo-photocatalytic therapy combined with tetramodal imaging. This approach results in a substantial increase in the production of HO∙ through the stimulation of endogenous H2O2. Furthermore, HO∙ is implicated in the induction of mitochondrial damage. The reciprocal interaction and cyclic feedback between HO∙ and other ROS significantly augment the presence of cytotoxic cells and helper T lymphocytes in the tumor region. Consequently, these effects contribute to the effective activation of the immune response [77, 78].
3.1.3. ROS Suppress B-cell Function and Foster the Establishment of a Conducive Tumor Microenvironment
Limited research has been conducted on the influence of H2O2 on B cells within the microenvironment. One notable study by Farber et al. provided evidence that in cases of lymphocytic leukemia, B lymphocytes are susceptible to oxidative damage when exposed to H2O2 in conjunction with T cells. Interestingly, this susceptibility of B cells to oxidative damage was found to be dependent on the presence of Ca2+. In contrast, T lymphocytes exhibited relatively higher resistance to the detrimental effects of H2O2 [79, 80]. The majority of malignant lymphomas (90%) originate from B-lymphocytes within the lymphopoietic system, suggesting a potential link between B-lymphocytes and their increased susceptibility to ROS compared to T cells. However, it is important to note that the existing evidence supporting this hypothesis is limited and lacks substantial strength in the present study. Further research is needed to better understand the differential responses of B and T cells to ROS and their implications in lymphoma development.
3.1.4. ROS Hinder the Activity of NK Cells and Facilitate the Progression of the Tumor Microenvironment
H2O2 contributes to the promotion of the tumor microenvironment by compromising the activity of natural killer (NK) cells and suppressing immune function. A study conducted by Izawa et al. investigated CD56dim/CD56bright NK cells in gastric and esophageal cancers and observed that CD56dim NK cells were more susceptible to apoptosis induced by physiological levels of H2O2 compared to CD56bright NK cells. Furthermore, there was a negative correlation between CD56dim NK cell infiltration in tumors and H2O2 production. Additionally, H2O2 was found to impair the antibody-dependent cellular cytotoxicity (ADCC) activity of tumor-associated NK cells [81]. In breast cancer, Klopotowska et al. discovered that NK cells exhibit greater susceptibility to H2O2 compared to T or B cells. As a result, the functionality of NK cells is substantially inhibited, leading to a reduction in their anti-tumor activity. The study also revealed that peroxiredoxin 1 (PRDX1), an essential component of the antioxidant defense system, is deficient in NK cells within the tumor microenvironment. This deficiency in PRDX1 may explain the heightened sensitivity of NK cells to H2O2 [82]. Furthermore, in melanoma, H2O2 released by macrophages exerts an inhibitory effect on the promotion of tumor growth by NK cells. It leads to a reduction in the expression of the CD3 complex and CD16 complex in peripheral blood NK cells. This decrease in expression is associated with a decline in the cytolytic activity of NK cells and significantly impairs their effector function [65]. H2O2 plays a regulatory role in the tumor microenvironment, contributing to tumor growth by inhibiting the function of NK cells and inducing damage to these immune cells.
3.1.5. ROS Enhance the Function of NK Cells and Hinder the Development of the Tumor Microenvironment
H2O2 can enhance the migration rate of NK cells and exert an inhibitory effect on the tumor microenvironment. For instance, in the context of mast cell tumors, the P815 cell line acts as a “bystander” cell within the tumor microenvironment, releasing H2O2. This H2O2 production facilitates the migration of NK cells toward target cells, thereby enhancing the efficiency of NK cells in eliminating these target cells [83]. The impact of H2O2 on NK cells within the tumor microenvironment is concentration-dependent. Low concentrations of H2O2 have been shown to enhance the function of NK cells and their effectiveness in suppressing tumors. However, it's important to note that high concentrations of H2O2 (above 10 μM) can induce apoptosis in NK cells. Therefore, when utilizing H2O2 to promote tumor suppression through NK cells, careful consideration should be given to maintaining an appropriate and controlled concentration of H2O2 to avoid detrimental effects on NK cell viability and function.
3.1.6. ROS Regulate the Activity of Macrophages and Promote the Development of the Tumor Microenvironment
H2O2 plays a role in promoting the tumor microenvironment by impairing the immune function of macrophages and evading their immune response. In the context of hepatocellular carcinogenesis, the expression of LOXL4, an amine oxidase involved in the remodeling of the extracellular matrix, is upregulated. The increased LOXL4 expression leads to the production of H2O2, which in turn activates STATs associated with interferon (IFN) signaling and induces the expression of programmed cell death ligand 1 (PD-L1). The upregulated PD-L1 expression then acts to suppress the immune function of macrophages, contributing to an immune-suppressive microenvironment in hepatocellular carcinoma [61]. Furthermore, it has been observed that H2O2 at a concentration of 100μM in lung cancer cells induces oxidative stress, resulting in alterations in the structure of protein subunits. These structural changes may give rise to the formation of novel antigens, which could potentially aid tumor cells in evading the immune response of phagocytes. This phenomenon highlights the ability of H2O2 to contribute to immune evasion mechanisms employed by tumor cells in the tumor microenvironment [84].
O2•−, in turn, plays a crucial role in enhancing the expression of C-C motif ligand 2 (CCL2), which serves to recruit macrophages and facilitate the development of the tumor microenvironment. Specifically, in retinoblastoma-deficient tumors, CCL2 expression is upregulated through the activation of AMP-activated protein kinase (AMPK). This activation leads to an increase in fatty acid oxidation, ultimately promoting the production of mitochondrial O2•−. The activation of the CCL2-CCR2 axis within the tumor microenvironment has been shown to contribute to various tumor types, including sarcoma and breast cancer. This axis serves to facilitate tumor angiogenesis and the recruitment of tumor-associated macrophages (TAMs) as well as myeloid-derived suppressor cells (MDSCs), which are known to exert immunosuppressive effects. Ultimately, these processes regulate the tumor microenvironment, promoting tumor growth in the aforementioned tumor types [85].
3.1.7. ROS Enhance the Function of Macrophages and Inhibit the Development of the Tumor Microenvironment
H2O2 plays a pivotal role in suppressing the tumor microenvironment by inducing alterations in the phenotype of macrophages. Sang et al. employed Iron Nanotrap as a means to target iron transport specifically to tumor-associated macrophages (TAMs) within the tumor. By releasing iron from the Nanotrap, TAMs were reprogrammed and subjected to oxidative stress upon stimulation with H2O2. This reprogramming process resulted in the conversion of TAMs from a pro-tumor M2 phenotype to an anti-tumor M1 phenotype. Consequently, the reprogrammed TAMs exhibited the ability to elicit an immune response and suppress tumor survival, thereby contributing to the inhibition of tumor progression [86].
Pigment epithelium-derived factor (PEDF) has shown potential in inhibiting the tumor microenvironment by enhancing macrophage activity via O2•−. In the context of prostate cancer, PEDF plays a crucial role in macrophage recruitment and stimulates their polarization towards the classical activation pathway. By regulating the interaction between prostate cancer cells and macrophages, PEDF enhances the immune response of macrophages and promotes apoptosis of tumor cells. Experimental findings suggest that PEDF stimulates the production of O2•− by macrophages, which is implicated in the apoptotic process of cancer cells. However, further investigation is necessary to elucidate the precise regulatory mechanism underlying this phenomenon [87].
HO• utilize the disguise of macrophage membranes to confer immune evasion properties to nano-catalysts. Through interaction with the catalyst α4/VCAM-1, macrophages gain the ability to recognize tumor endothelial cells and cancer cells, effectively suppressing the metastasis of breast cancer cells [88].
3.1.8. ROS Enhance Neutrophils and Inhibit the Development of the Tumor Microenvironment
O2•− enhances the cytotoxicity of neutrophils against target cells and inhibits the tumor microenvironment. Ishihara et al. utilized PSK (a protein-bound polysaccharide) induction to elevate the levels of bronchoalveolar lavage (BAL) neutrophils, resulting in increased production of O2•− by neutrophils. This, in turn, led to heightened cytotoxicity of neutrophils towards their targets and a significant reduction in the number and size of metastatic foci in lung cancer cells. However, the precise mechanism through which O2•− enables neutrophils to exert cytotoxic effects and inhibit tumor metastasis remains unclear [89]. Furthermore, the specific mechanism by which O2•− affects B cells, NK cells, and dendritic cells within the tumor microenvironment (TME) remains uncertain and requires further investigation.
HO∙ exploits neutrophils to facilitate immune responses and suppress the tumor microenvironment. Under conditions where the neutrophil defense system is overwhelmed and the microenvironment provides oxidized iron, neutrophils generate and release HO∙ through iron-catalyzed reactions of superoxide. This process serves to eliminate potential pathogens and mediate the innate immune response [90, 91]. Ultimately, HO∙ inhibits the tumor cell microenvironment.
3.1.9. ROS Exploit Neutrophils to Promote the Development of the Tumor Microenvironment
HO∙ utilizes neutrophils, inducing oxidative stress and ultimately modulating the tumor microenvironment. In cases of gastric cancer associated with H. pylori infection, the presence of virulence factors in H. pylori can attract neutrophils that harbor γ-glutamyl transferase, leading to the generation of different ROS, including H2O2 and highly reactive HO∙. Prolonged oxidative stress in the infected area results in DNA damage and chronic inflammation, establishing a favorable inflammatory milieu for the survival of tumor cells [92].
3.2. ROS Exert a Significant Influence on Stromal Cells Within the Tumor Microenvironment
3.2.1. ROS Enhance the Activity of Endothelial Cells and Benefit the Progression of the Tumor Microenvironment
H2O2 contributes to the promotion of the tumor microenvironment and sustains tumor cell survival primarily by enhancing the proliferation and migration of endothelial cells. Vascular endothelial growth factor (VEGF) is an important factor involved in angiogenesis, and its upregulation facilitates the migration and proliferation of endothelial cells [93], and endothelial cells provide nutrients to support tumor growth and development [94]. In human prostate cancer, there exists a notable correlation between the expression of VEGF-C, a growth factor with a specific affinity for heparin and vascular endothelial cells, and the occurrence of lymph node metastasis [95, 96]. Muders et al. discovered that in prostate cancer, stimulation with H2O2 leads to an increase in the phosphorylation of serine-threonine kinase AKT-1 and promotes endothelial cell proliferation within cancer cells through the action of VEGF-C. This process renders serine-threonine kinase AKT-1 resistant to inactivation, thereby enabling its continuous activation. The activation of AKT-1 is mediated by mTOR complex 2 (mTORC-2) within the tumor microenvironment, which protects prostate cancer cells from undergoing H2O2-induced cell death [97]. Furthermore, when exposed to H2O2, CV-MSCs (mesenchymal stem cells/stromal cells derived from human-term placental chorionic villi) demonstrate a protective effect on endothelial cells, mitigating the harmful impacts of oxidative stress [98]. This phenomenon may be attributed to the modified expression of p53 in CV-MSCs within the breast cancer microenvironment, thereby endowing them with certain capabilities to counteract oxidative stress [99]. In colon cancer, low levels of H2O2 have been shown to enhance the expression of vascular endothelial growth factors in cancer cells, stimulating the migration and proliferation of endothelial cells. Consequently, this facilitates increased nutrient supply to cancer cells [100].
O2•−, also known as superoxide, has been found to enhance vascular endothelial cell migration, thereby promoting the tumor microenvironment. David et al. employed MALDI-TOF mass spectrometry to identify protein spots associated with melanoma development. Through their analysis, they observed a substantial increase in proteins involved in O2•− production, glycolysis, inflammation, and other processes. Furthermore, the induction of O2•− resulted in a significant upregulation of VEGF, which further stimulated vascular endothelial cell migration, proliferation, and angiogenesis [101].
3.2.2. ROS Modify the Behavior of Fibroblasts and Reinforce the Progression of the Tumor Microenvironment
H2O2 plays a role in promoting the tumor microenvironment in fibroblasts by influencing protein expression, oxidative stress, and phenotypic changes. Specifically, in esophageal squamous cell carcinoma (ESCC), H2O2 induces the upregulation of Hydrogen peroxide-inducible clone 5 (HIC-5) in cancer-associated fibroblasts present in the tumor stroma. HIC-5, in turn, regulates the migration and invasion of ESCC cells by modulating cytokines and altering the ECM. This interplay between H2O2, HIC-5, fibroblasts, and ESCC cells contributes to the dynamic tumor microenvironment in ESCC [102]. Frequent mutations and deletions of the BRCA1 tumor suppressor gene are observed in hereditary ovarian cancer. In the context of BRCA1-deficient cancer cells, these cells produce elevated levels of H2O2, which results in oxidative stress and catabolic effects on neighboring stromal fibroblasts. This phenomenon is mediated through the activation of stromal NF-κB, a transcription factor. Consequently, the activated NF-κB pathway leads to the reprogramming of tumor mesenchymal metabolism. This metabolic reprogramming drives mesenchymal-epithelial metabolic coupling, a critical process promoting the development of lethal tumors in the context of BRCA1 deficiency [103]. In human breast cancer, H2O2 is capable of diffusing easily through the cell membrane and exerting its effects on surrounding fibroblasts. When exposed to acute stress, fibroblasts respond by increasing the expression of Hypoxia-inducible factor-1 (HIF) and chemokine CXCL12. This molecular signaling leads to the transformation of fibroblasts into myofibroblasts, which are characterized by their high contractility and expression of smooth muscle α-actin (SM-α-actin). These activated myofibroblasts play a significant role in promoting the migration and spread of tumor cells within the breast cancer microenvironment [104]. Furthermore, H2O2 generated by cancer cells elicits oxidative stress in adjacent fibroblasts, culminating in the substantial production of copious quantities of L-lactic acid. This metabolic byproduct serves as a pivotal “fuel” for fostering the proliferation and metabolic activities of breast cancer cells [105]. This intricate process unfolds within the TME of breast cancer, whereby cancer cells emit H2O2 into the surrounding milieu, instigating oxidative stress and triggering the production of H2O2 within neighboring cancer-associated fibroblasts (CAFs). Subsequently, these CAFs undergo aerobic glycolysis, generating substantial quantities of energy-dense “fuel” such as ketone bodies, lactate, pyruvate, and fatty acids. Remarkably, this energy-rich “fuel” is then harnessed by cancer cells to sustain their growth and proliferative endeavors [106-108]. Furthermore, the secretion of H2O2 by CAFs emulates the modus operandi of immune cells such as neutrophils and macrophages, instigating the body's innate immune response mediated by NF-κB. Consequently, this cascade triggers both local and systemic inflammation, thereby fostering an environment conducive to the survival of cancer cells [109]. In the context of prostate cancer, H2O2 governs distinct translational regulations of matrix metalloproteinase-3 (MMP-3) in both cancer cells and CAFs. Specifically, H2O2 directly impedes the activity of the MMP-3 promoter in CAFs, consequently suppressing the nuclear translocation of NF-κB and leading to diminished MMP-3 expression in CAFs. Conversely, H2O2 induces the upregulation of microRNA-128 in prostate cancer cells, which in turn reduces the expression of thrombospondin 2 (a repressor of MMP-3). Consequently, MMP-3 expression in cancer cells is elevated, facilitating the maintenance of ECM homeostasis [110]. Therefore, H2O2 exerts control over the TME by modulating the behavior of fibroblasts, including cancer-associated fibroblasts (CAFs), thereby shaping the TME to create a favorable environment for tumor growth.
O2•− promotes the tumor microenvironment by stimulating autophagy in cancer cells and enhancing the role of fibroblasts. In breast cancer, O2•− induces tumor cells to trigger autophagy in neighboring cancer-associated fibroblasts by suppressing Caveolin-1 (Cav-1) expression. Autophagy occurring in the tumor stroma generates high-energy nutrients that support cancer cell metabolism and facilitate their growth [6, 111, 112]. Extracellular superoxide dismutase (EcSOD) serves as the sole extracellular enzyme responsible for scavenging O2•−. An interesting finding by Golden et al. revealed that the overexpression of EcSOD in breast cancer cells had inhibitory effects on hepatocyte growth factor (HGF)-induced phosphorylation of the c-Met receptor tyrosine kinase. This, in turn, resulted in the suppression of cancer-fibroblast interactions and the inhibition of three-dimensional (3D) stromal gel growth of MDA-MB231 cells, a breast cancer cell line. These observations suggest that O2•− production enhances HGF-mediated interactions between cancer cells and fibroblasts, thereby promoting the tumor microenvironment [113].
3.2.3. ROS Change the Resilience of Fibroblasts and Deter the Development of the Tumor Microenvironment
H2O2 plays a suppressive role in the tumor microenvironment by enhancing the stimulation of the Fibroblast-FGF2 system. Fibroblast growth factor 2 (FGF2), a key member of the fibroblast growth factor family, is primarily secreted by fibroblasts. H2O2, in this context, enhances the activation of the Fibroblast-FGF2 system, thereby exerting suppressive effects on the tumor microenvironment [114]. Ma et al. discovered that H2O2 amplifies the retention of phosphorylated extracellular signal-regulated kinase ½ (pERK1/2) in the cytoplasm and the binding of kinase pGSK3β (Tyr-216) in the peripheral primitive neuroectodermal tumor cell line SK-N-MC, under the stimulation of the Fibroblast-FGF2 system. Furthermore, the experimental findings revealed that the FGF2-induced modulation of phosphorylation and accumulation of pERK1/2 in the cytoplasm exerts a cytotoxic inhibitory effect on SK-N-MC cells, ultimately leading to apoptosis [115]. The impact of H2O2 on adipocytes and stellate cells within the stromal cells of the TME remains poorly elucidated and necessitates additional research for comprehensive understanding.
O2•−, through its action on fibroblasts, can upregulate the expression of the tumor suppressor protein p53 and inhibit the fibroblast-mediated proliferation of cancer cells. Wang et al. conducted a study on the prostate cancer cell line PC3 and found that fibroblasts secrete basic fibroblast growth factor (bFGF), which promotes the growth of PC3 cells. However, it was observed that a specific concentration of O2•− can significantly hinder the proliferation of cancer cells mediated by fibroblast-derived bFGF. This inhibitory effect is attributed to the upregulation of p53 expression induced by O2•− [116].
4. ROS ALTER THE NON-CELLULAR COMPONENTS OF THE TUMOR MICROENVIRONMENT
4.1. ROS Promote Angiogenesis and Enhance the Development of the Tumor Microenvironment
H2O2, as a signaling molecule produced by the Nox1 gene (a homolog of gp91phox), plays a role in upregulating the protein expression of VEGF and promoting angiogenesis [117]. Nox1 gene expression in tumor cells leads to a significant upregulation of VEGF mRNA expression through the action of H2O2. This molecular mechanism promotes tumor angiogenesis and enhances the tumorigenicity of DU-145 prostate epithelial cells [118].
O2•− plays a role in promoting angiogenesis by upregulating the protein expression of VEGF. Kuroki et al. demonstrated that when melanoma and glioblastoma cells were exposed to O2•−, there was a rapid increase in cellular VEGF mRNA levels. This increase was found to be dose-dependent, meaning that higher levels of O2•− resulted in greater VEGF expression. The enhanced angiogenesis induced by O2•− contributed to the promotion of tumor growth [119]. Further investigation is required to elucidate the mechanism by which O2•−, or superoxide, influences non-cellular constituents, including the ECM and exosomes.
HO∙ triggers gene expression and facilitates angiogenesis within the microenvironment. In head and neck squamous cell carcinoma (HNSCC), the oxidative stress induced by HO∙ modifies the expression of BRAK and IL-8 genes in human HNSCC cells via the EGFR/MEK/ERK pathway, thereby promoting angiogenesis and facilitating tumor progression [120].
4.2. ROS Inhibit Angiogenesis and Impede the Development of the Tumor Microenvironment
H2O2 functions as an inhibitor of tumor angiogenesis by modifying the protein structure of the VEGF receptor. Peroxiredoxin II (PrxII), an important antioxidant enzyme, plays a crucial role in this process. Dong et al. conducted a study revealing that the absence of PrxII resulted in a substantial increase in intracellular H2O2 levels, leading to oxidative inactivation caused by the altered protein structure of VEGF receptor-2. Consequently, VEGF receptor-2 loses its responsiveness to VEGF stimulation, ultimately inhibiting tumor angiogenesis in vivo [121].
4.3. ROS Promote ECM Production and Impede the Development of the Tumor Microenvironment
There is limited research exploring the effects of H2O2 on the ECM. Nevertheless, it has been demonstrated that lysyl oxidase (LOX) plays a role in catalyzing the cross-linking of elastin and collagen within the ECM. This enzymatic process contributes to the regulation of tissue tensile strength [122]. This process plays a crucial role in the migration of cancer cells. Additionally, LOX has been found to facilitate the formation of ECM adhesions and inhibit the migration of invasive breast cancer cells. These effects are mediated by H2O2 and involve the FAK/Src pathway [123].
The role of 1O2 in the ECM is not extensively investigated. However, existing studies indicate that 1O2 interacts with amino acid residues within proteins, generating active substances that facilitate cross-linking of the ECM. This cross-linking process enhances the ECM’s resistance to degradation by matrix metalloproteinases (MMPs), which are responsible for breaking down the ECM and facilitating tumor cell metastasis. As a result, 1O2 inhibits the migration of invasive tumor cells [124].
4.4. ROS Degrade ECM and Facilitate the Development of the Tumor Microenvironment
HO∙ increases the expression of MMPs, leading to the degradation of the ECM and promoting the tumor microenvironment. Furthermore, HO∙ enhances the process of macromolecular damage to cells mediated by reactive oxygen species and activates MMPs, resulting in heightened invasiveness of cancer cells [125]. However, the investigation of the effects of HO∙ on stromal cells is predominantly lacking in current research (Fig. 3).
Fig. (3).
The effect of each component of ROS on the TME component.
5. ANTICANCER THERAPIES INVOLVING ROS MANIPULATION
Based on the previous analysis, it is evident that ROS is a crucial metabolic byproduct within the tumor microenvironment. It participates in tumorigenesis and tumor progression through various mechanisms, including direct cellular damage, promotion of apoptosis, induction of cell proliferation and migration, and modulation of cell signaling pathways. Consequently, research on utilizing ROS for tumor treatment has become a prominent focus in current studies.
Currently, the use of ROS in tumor therapy primarily revolves around several approaches, namely antioxidant therapy, peroxide therapy, targeted ROS therapy, immunotherapy, and combination therapy.
5.1. Antioxidant Therapy
Antioxidant therapy is a treatment approach that involves the use of drugs or other methods to reduce excessive free radical activity within the body. Its purpose is to alleviate oxidative stress-induced damage to tissues and organs and to counteract the carcinogenic effects of ROS. This therapy primarily works by inhibiting ROS generation pathways or by enhancing the clearance of ROS through various antioxidants, thus reducing the ROS levels within tumor cells. Carvedilol, for example, exhibits the ability to eliminate ROS generation induced by Benzo(a)pyrene (BaP), thereby safeguarding normal cells from DNA damage and suppressing the malignant proliferation of breast epithelial cells [126]. In line with this, the administration of DHLA (dihydrolipoic acid) effectively prevented the formation of ROS in cells treated with Tetrachlorohydroquinone (TCHQ) and significantly suppressed the development of skin tumors induced by TCHQ [127]. The utilization of antioxidants, either as standalone agents or in conjunction with conventional anticancer medications, holds promise as an effective therapeutic approach to mitigate ROS-mediated tumorigenesis and disease progression in the organism. Extensive data derived from in vivo and in vitro experiments have substantiated the effectiveness of antioxidants (either alone or in combination) in suppressing tumor cell growth (as summarized in Table 2).
Table 2. Antioxidants reduce ROS levels in the body and inhibit tumors.
| Antioxidant Type | Tumor Type | Mechanism | References |
|---|---|---|---|
| hyperoside | breast cancer | Hyperoside induces apoptosis in breast cancer cells | [155] |
| silibinin | breast cancer | Silibinin inhibits the migration and invasion of breast cancer cells | [156] |
| N-acetylcysteine | hepatocellular carcinoma | N-acetylcysteine reduces ROS stress defense against hepatocarcinogenesis | [157] |
| catalase | / | Catalase delivery inhibits tumor cell metastasis | [158] |
| multi-antioxidant (FTP) | breast cancer | Multi-antioxidant (FTP) scavenges ROS for metastatic breast cancer | [159] |
| allopurinol complex | breast cancer | Allopurinol complex reduces ROS levels to induce breast cancer cell death | [160] |
| vitamin E | liver cancer | Vitamin E reduces ROS production to inhibit the development of liver cancer | [161] |
| carotenoids | prostate, lung, and digestive cancers | Carotenoids inactivate ROS and protect against oxidative damage to prevent cancer | [162] |
| catalase | melanoma | Targeted delivery of catalase inhibits melanoma proliferation and metastasis | [163] |
Nevertheless, certain clinical trials investigating the use of antioxidants for tumor treatment have revealed limitations and, in some cases, even an elevated risk of cancer [128-130]. Several potential explanations can account for this phenomenon. Firstly, the substantial inhibition of oxidative stress can potentially promote the proliferation of cancer cells [131-133]. Secondly, antioxidants have the ability to impede the apoptosis triggered by ROS, which can consequently hinder the normal apoptosis process in cancer cells [134]. Therefore, it is crucial to conduct research aimed at minimizing the potential risk of antioxidants promoting tumor development. This endeavor can lead to the development of more clinically appropriate treatment modalities.
5.2. Oxidative Therapy
Oxidative therapy involves further increasing ROS levels to a toxic level, activating multiple cell death pathways, enhancing chemosensitivity, and opening up new avenues for cancer treatment [10]. The primary objective is to elevate ROS levels by utilizing ROS generation inducers and suppressing antioxidants. As an illustration, upregulating p53 expression and downregulating superoxide dismutase 2 (SOD2) expression through the use of Astragalus purpureus led to heightened ROS levels and activation of the mitochondrial apoptotic pathway, effectively impeding the growth and proliferation of liver tumor cells [135]. The efficacy of Deoxyelephantopin in reducing the viability of osteosarcoma cells is dependent on the dosage. It achieves this by promoting the generation of ROS, activating apoptosis-related proteins like Bax, and inducing mitochondrial dysfunction [136]. Several studies and clinical trials have investigated the inhibition of tumor cells by increasing ROS levels (Table 3). However, it is important to note that solely increasing ROS concentration as a treatment approach for tumors may have undesirable effects. Inducing ROS through certain agents can disrupt the redox balance in tumor cells, potentially leading to DNA damage, cell genome mutations, tumor initiation, or increased survival of tumor cells [137, 138]. Overall, it appears that tumor cells have higher levels of ROS compared to normal cells, which makes anti-ROS therapy a targeted approach for treating tumors. However, achieving the right balance of anti-ROS agents and inducing ROS production in clinical research to achieve therapeutic goals without causing harmful or even contradictory side effects still requires further improvement.
Table 3. Increasing ROS levels allows tumor cells to reach a critical state and induce inhibition or death.
| Oxidizer Type | Tumor Type | Mechanism | References |
|---|---|---|---|
| astragalus | lung cancer | Astragalus enhances ROS production in cancer cells and inhibits cell migration and invasion of lung cancer cells | [164] |
| albanol B | lung cancer | Albanol B increased mitochondrial ROS production, inhibited proliferation, and induced apoptosis of lung cancer cells | [165] |
| lotus leaf flavonoids | lung cancer | Lotus leaf flavonoids increase ROS to induce apoptosis in lung cancer cells in vitro | [166] |
| chloroquine | cholangiocarcinoma | The autophagy inhibitor chloroquine increases intracellular ROS and increases the sensitivity of cisplatin in cholangiocarcinoma cells | [167] |
| fibulin 5 and beta1 integrins | pancreatic tumor | Loss of fibulin 5 binding to beta1 integrins inhibits tumor growth by increasing the level of ROS | [168] |
| oxaliplatin | oral squamous cell carcinoma | Oxaliplatin inhibits oral squamous cell carcinoma by increasing ROS production | [169] |
| chrysin | pancreatic cancer | Chrysin increases cellular ROS levels and induces ROS-dependent autophagy to induce pancreatic cancer cell death | [170] |
| flavonoids | colorectal cancer | Flavonoids increase the level of intracellular peroxide and induce apoptosis of colorectal cancer cells | [171] |
| ampelopsin | mammary tumor | Ampelopsin increases mitochondrial membrane permeability and induces mammary tumor cell apoptosis by increasing ROS levels | [172] |
| thymoquinone | / | Thymoquinone induces ROS production and prevents mediating cancer progression | [173] |
| gossypol | colorectal cancer | Gossypol induces ROS production and induces apoptosis of colorectal cancer cells | [174] |
| tempol | ovarian cancer | Tempol increase cellular ROS production and enhances cisplatin-induced apoptosis in ovarian cancer cells | [175] |
| melatonin | thyroid cancer | Melatonin induces reactive oxygen species to inhibit migration and induce apoptosis of thyroid cancer cells | [176] |
| ginsenoside Rg2 | breast cancer | Ginsenoside Rg2 induces ROS production and enhances protein and mRNA expression of breast cancer apoptotic molecules | [177] |
| ascorbate | chronic lymphocytic leukemia | ROS accumulation by ascorbate enhances the cytotoxicity of ATO in chronic lymphocytic leukemia | [178] |
5.3. Targeted ROS Therapy
Targeted ROS therapy primarily involves the use of specific targeted molecules such as nanomaterials and small molecules to selectively target specific ROS molecules for treatment. Based on their action sites and mechanisms, targeted ROS molecules can be classified into those targeting ROS generation, ROS signaling pathways, and ROS scavenging, among others. For example, researchers have developed targeted peptide precursor nanodrugs that conjugate with the ROS-sensitive sulfone group, linked with the cytotoxin epothilone B. These nanodrugs exhibit high-level targeting of ROS generation and release epothilone B upon cleavage, demonstrating remarkable tumor selectivity and excellent anti-cancer efficiency in both in vitro and in vivo studies [139]. Targeted ROS therapy also includes photodynamic therapy and combination chemotherapy [140, 141]. Targeted ROS therapy offers the advantages of strong specificity, significant efficacy, and minimal side effects. By precisely targeting ROS for treatment, this approach can minimize damage to normal cells and improve therapeutic outcomes. Currently, targeted ROS therapy has been widely applied in clinical tumor treatment, particularly for lung cancer, liver cancer, and other types of tumors [142, 143].
Although targeted ROS therapy holds certain application value, it also presents several challenges and limitations. For instance, in certain specific tumors such as melanoma, the presence of melanin enhances oxidative stress defense and may have defective apoptotic pathways, posing a challenge in developing efficient and specific targeted ROS drugs [144]. In summary, targeted ROS therapy holds promise in tumor treatment, but it also faces challenges and limitations. Therefore, future research should focus on exploring highly efficient and specific targeted ROS drugs, as well as investigating their combination with other treatment modalities to enhance therapeutic efficacy and minimize side effects.
5.4. Immunotherapy
Immunotherapy utilizes ROS as a target for treatment, aiming to enhance the body's antioxidant capacity and strengthen the immune system's ability to eliminate tumor cells, thereby achieving the goal of tumor therapy. ROS-based immunotherapy encompasses various approaches, including autophagic cell death and inflammation modulators. For instance, a chemical photothermal nano-platform has been developed to activate immune sites using ROS, mediating cancer cell apoptosis and autophagic death to enhance treatment efficacy [145].
Targeting ROS as a therapeutic target in immunotherapy holds significant importance as it can enhance the body's antioxidant capacity and augment the immune system's cytotoxicity against tumor cells, thereby facilitating tumor treatment. However, further understanding of the specific mechanisms and potential risks of ROS in immunotherapy is necessary to provide scientific guidance for its clinical application.
5.5. Combination Therapy
Combination therapy refers to the use of two or more different treatment modalities together to enhance therapeutic efficacy and reduce side effects. In the context of utilizing ROS as a target for immunotherapy, combining ROS inhibitors with chemotherapy drugs, immunotherapy agents, or other treatment modalities can enhance treatment effectiveness while minimizing adverse effects [146, 147]. Combination therapies can include ROS inhibitors in conjunction with chemotherapy drugs, ROS inhibitors combined with immunotherapy agents, ROS-releasing agents combined with chemotherapy drugs, and ROS-releasing agents combined with immunotherapy agents.
Although combination therapy can improve treatment outcomes and reduce side effects, it is crucial to consider factors such as drug interactions, dosages, and administration methods. It is important to undertake combination therapy under the guidance of a healthcare professional to ensure treatment safety and efficacy.
CONCLUSION
Based on the previous discussion, it is evident that many current therapeutic strategies are based on the direct impact of ROS on tumor cells. Moreover, whether it is antioxidation, peroxidation, targeted ROS therapy, or immunotherapy, etc., all exhibit certain drawbacks and side effects. Hence, it is apparent that solely adjusting the ROS levels may not always lead to optimal treatment outcomes. Can we, then, derive novel therapeutic approaches from the relationship between ROS and TME? Firstly, by examining their interactions, we can identify certain patterns and potentially establish a foundation for innovative treatment strategies.
First and foremost, among the various components of ROS, H2O2 exhibits the most extensive influence on the tumor microenvironment. This can be attributed to the dual impact of H2O2 on immune cells such as T cells, B cells, NK cells, and phagocytes, as well as stromal cells including endothelial cells and fibroblasts. Additionally, H2O2 affects non-cellular components such as blood vessels and ECM. In terms of magnitude, the remaining components, namely O2•−, HO∙, and 1O2, also exert dual effects, albeit to a lesser extent. This observation may be attributed to H2O2's ability to activate multiple sensors and pathways involved in signal transduction, as discussed in the previous article. For instance, H2O2 enhances signaling by promoting the upregulation of transcription or by increasing mRNA stability and translation to facilitate the synthesis of transcription factors [26, 27]. Furthermore, due to its ability to target various transduction pathways and mechanisms, H2O2 offers a unique opportunity to regulate genes and signaling with remarkable precision.
Secondly, H2O2 displayed the highest sensitivity to the influence of the tumor microenvironment. This is evident from the findings presented in Table 4, where Zhu et al. experimented to investigate the upregulation of H2O2 on vascular endothelial growth factor. Notably, even at a concentration as low as 10-3μM, H2O2 induced the migration of endothelial cells within the microenvironment of colon cancer [100]. Nevertheless, it should be noted that the other ROS exhibited minimum effective concentrations greater than 10-3μM.
Table 4. ROS action concentration.
| H2O2 (µM) | Microenvironmental Components | Direction of Action | References |
|---|---|---|---|
| 10 | T cells | It interacts with T cells to promote the tumor microenvironment | [65] |
| 50 | [68] | ||
| 10, 20, 50, 100 | [69] | ||
| 10, 50, 100 | [66] | ||
| 200, 400, 600, 800 | [153] | ||
| 1x104, 5x104 | [152] | ||
| 100 | It interacts with T cells to suppress the tumor microenvironment | [75] | |
| 500 | [74] | ||
| 2 | NK cells | It interacts with NK cells to suppress the tumor microenvironment | [83] |
| 6.25-100 | It interacts with NK cells to promote the tumor microenvironment | [82] | |
| 10 | [65] | ||
| 10, 20 | [81] | ||
| 100 | macrophages | It interacts with macrophages to promote the tumor microenvironment | [84] |
| 10, 10-1, 10-3 | endothelial cells | It interacts with endothelial cells to promote the tumor microenvironment | [100] |
| 3x103 | [97] | ||
| 50 | Fibroblast (associated with cancer) | It interacts with Fibroblast (associated with cancer) to suppress the tumor microenvironment |
[115] |
| 100 | blood vessels | It interacts with blood vessels to suppress the tumor microenvironment | [121] |
| O2•− | |||
| 9-36μM NADH induction | Fibroblast | It interacts with Fibroblast (associated with cancer) to suppress the tumor microenvironment | [116] |
| 500 | blood vessels | It interacts with blood vessels to promote the tumor microenvironment | [119] |
| HO∙ | |||
| 50, 100, 175, 250 | blood vessels | It interacts with blood vessels to promote the tumor microenvironment | [120] |
| 1O2 | |||
| 2x10-1μM of MG2I induction | T cells | It interacts with T cells to promote the tumor microenvironment | [72] |
Thirdly, it is worth noting that the action concentration of 1O2 is relatively low. Unlike the other ROS concentrations in Table 4, which were mostly above 1 μM, there was only one instance of 2x10-1 μM MG2I (a chemical fluorogen) used to induce 1O2 production. From this, we can infer that 1O2 exerts its effects on the TME under low-concentration conditions. This can be attributed to the low concentration of the inducer MG2I and the volatile nature of 1O2 itself. Despite its unstable structure and short half-life, 1O2, being a diolefin with high reactivity, remains effective even at low concentrations [54].
Fourthly, H2O2 exhibits its effects across a wide range of concentrations. As observed in Table 4, the action concentrations of H2O2 in the TME span from 10-3μM to 5x104 µM. It is crucial to emphasize that H2O2 can elicit contrasting effects on the same component of the TME at the same concentration. For instance, different experiments conducted by Bang and Freund demonstrated that 100 µM of H2O2 yielded opposing effects on T cells, namely enhancement, and inhibition, respectively [66, 75]. However, it is important to consider that the microenvironments of T cells in the two studies were different: one was in the glioma microenvironment, while the other was in the colon cancer microenvironment. Consequently, the same concentration of H2O2 may yield opposite effects on T cells in different tumor types. Therefore, when conducting research or clinical treatment involving H2O2, careful consideration should be given to the specific tumor type under investigation.
Fifth, it is noteworthy that the four ROS components do not exhibit dual actions simultaneously on the same component within the TME. For instance, H2O2 promotes ECM, O2•− promotes endothelial cells, HO∙ inhibits ECM, and 1O2 inhibits T cells. As a result, whether the ROS components have a dual effect on the overall TME environment requires individual investigation and cannot be generalized.
Based on the legislation and the effects of ROS on various components of the TME outlined in Table 1, it may be possible to select specific ROS treatments based on different tumor types, tumor microenvironments, and concentrations employed. For instance, taking into consideration the abundance of cancer-associated fibroblasts (CAFs) in breast cancer, Agnieszka's research team discovered that H2O2 excessively activated the JNK1 stress signaling pathway within the cancer stroma. This resulted in the acquisition of a CAF-like phenotype and promoted tumor invasion in breast fibroblasts. Conversely, inhibiting the activation of JNK1 signaling by reducing H2O2 levels using the peroxidase PRDX1 effectively mitigated breast fibroblast invasion and tumor development [148]. Moreover, the synergistic administration of lactoferrin and black tea polyphenols has been shown to effectively suppress the generation of HO∙ and O2•− in hamster buccal pouch carcinoma. This combined treatment strategy exhibits inhibitory effects on tumor angiogenesis, providing a promising approach for impeding the formation of new blood vessels in the tumor microenvironment [149]. Consequently, the treatment of tumors with ROS is not solely centered around directly suppressing malignant cells but also revolves around the pivotal influence of ROS on the protective barrier of tumors, known as the tumor microenvironment.
In pursuing ROS-based interventions for the TME, adherence to three overarching guidelines is warranted. Firstly, drug administration ought to align with the specific tumor type and its distinct microenvironment. Secondly, diverse ROS components manifest distinct impacts, thus cautioning against a generalized approach to ROS dosing. Thirdly, even when dealing with the same ROS, disparate concentrations yield inconsistent outcomes, necessitating vigilance regarding alterations in blood levels.
To adhere to these recommendations, several strategies can be employed in the development and utilization of clinical drugs. Primarily, it is advisable to focus on designing drugs that specifically target individual ROS components. Additionally, the utilization of in vitro cell/organ drug sensitivity assays can aid in determining the optimal efficacy and dosage concentrations before drug administration. Furthermore, each ROS component should be thoroughly investigated to identify specific dosing markers that provide precise dosing instructions and therapeutic targets (Fig 4). However, it is worth noting that there is a dearth of therapeutic studies exploring the modulation of ROS in the tumor microenvironment for tumor eradication, despite its promising potential in targeting TME's crucial role in tumor development. Therefore, further research focusing on ROS components with high specificity for the TME is eagerly anticipated to advance effective tumor treatments.
Fig. (4).
Depicts the change of ROS effect on TME with the change of ROS level. At low concentrations, ROS mainly plays a role in signal transduction. With the occurrence of the tumor, ROS promotes tumor invasion and metastasis, proliferation, and mitochondrial damage. When the concentration of ROS reached a certain level, the apoptosis and DNA damage of cells in the TME environment were induced more. The graph shows existing treatment strategies for this condition. In addition, future research directions are proposed based on the relationship between ROS and TME.
ACKNOWLEDGEMENTS
Declared none.
LIST OF ABBREVIATIONS
- ROS
Reactive Oxygen Species
- TME
Total Mesorectal Excision
AUTHORS’ CONTRIBUTIONS
All authors contributed to the conception and design, writing, critical revision, and final approval of the article. S.Y.H, D.C, and S.G conceived the article, Y.L, L.L, X.L.T constructed the article structure, Y.C, Y.S.Z, J.S, X.W, M.X.L, M.J.C, X.B.L, Y.H.S, L.G, W.P.L, F.W corrected the article. C.H.C, Z.G.X, and F.K.D supervised the study and coordinated the writing of the manuscript. All authors have read and agreed to the published version of the manuscript.
CONSENT FOR PUBLICATION
Not applicable.
FUNDING
This work was supported by Grants from the Sichuan Science and Technology Program, China (No. 2022NSFSC0783), Joint Founds of Southwest Medical University and Luzhou Government (No. 2020LZXNYDJ08), Grants of Southwest Medical University (2021ZKMS038), Funds of talent introduction and scientific research of Southwest Medical University (No. 05-00040140), the Strategic Cooperation Project for Transfer and Transformation of Scientific and Technological achievements of Southwest Medical University and Lu County Government (grant no. 2019LXXNYKD-07).
CONFLICT OF INTEREST
The authors declare no conflict of interest, financial or otherwise.
REFRENCES
- 1.Anderson N.M., Simon M.C. The tumor microenvironment. Curr. Biol. 2020;30(16):R921–R925. doi: 10.1016/j.cub.2020.06.081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Deepak K.G.K., Vempati R., Nagaraju G.P., Dasari V.R., S N., Rao D.N., Malla R.R. Tumor microenvironment: Challenges and opportunities in targeting metastasis of triple negative breast cancer. Pharmacol. Res. 2020;153:104683. doi: 10.1016/j.phrs.2020.104683. [DOI] [PubMed] [Google Scholar]
- 3.Hanahan D., Weinberg R.A. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–674. doi: 10.1016/j.cell.2011.02.013. [DOI] [PubMed] [Google Scholar]
- 4.Weinberg F., Hamanaka R., Wheaton W.W., Weinberg S., Joseph J., Lopez M., Kalyanaraman B., Mutlu G.M., Budinger G.R.S., Chandel N.S. Mitochondrial metabolism and ROS generation are essential for Kras-mediated tumorigenicity. Proc. Natl. Acad. Sci. USA. 2010;107(19):8788–8793. doi: 10.1073/pnas.1003428107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Kong H., Chandel N.S. Regulation of redox balance in cancer and T cells. J. Biol. Chem. 2018;293(20):7499–7507. doi: 10.1074/jbc.TM117.000257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Martinez-Outschoorn U.E., Balliet R.M., Rivadeneira D., Chiavarina B., Pavlides S., Wang C., Whitaker-Menezes D., Daumer K., Lin Z., Witkiewicz A., Flomenberg N., Howell A., Pestell R., Knudsen E., Sotgia F., Lisanti M.P. Oxidative stress in cancer associated fibroblasts drives tumor-stroma co-evolution. Cell Cycle. 2010;9(16):3276–3296. doi: 10.4161/cc.9.16.12553. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Moloney J.N., Cotter T.G. ROS signalling in the biology of cancer. Semin. Cell Dev. Biol. 2018;80:50–64. doi: 10.1016/j.semcdb.2017.05.023. [DOI] [PubMed] [Google Scholar]
- 8.Mittler R. ROS are good. Trends Plant Sci. 2017;22(1):11–19. doi: 10.1016/j.tplants.2016.08.002. [DOI] [PubMed] [Google Scholar]
- 9.Weinberg F., Ramnath N., Nagrath D. Reactive oxygen species in the tumor microenvironment: An overview. Cancers (Basel) 2019;11(8):1191. doi: 10.3390/cancers11081191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Galadari S., Rahman A., Pallichankandy S., Thayyullathil F. Reactive oxygen species and cancer paradox: To promote or to suppress? Free Radic. Biol. Med. 2017;104:144–164. doi: 10.1016/j.freeradbiomed.2017.01.004. [DOI] [PubMed] [Google Scholar]
- 11.Kirtonia A., Sethi G., Garg M. The multifaceted role of reactive oxygen species in tumorigenesis. Cell. Mol. Life Sci. 2020;77(22):4459–4483. doi: 10.1007/s00018-020-03536-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Halliwell B. Oxidative stress and cancer: have we moved forward? Biochem. J. 2007;401(1):1–11. doi: 10.1042/BJ20061131. [DOI] [PubMed] [Google Scholar]
- 13.Nakazawa M.S., Keith B., Simon M.C. Oxygen availability and metabolic adaptations. Nat. Rev. Cancer. 2016;16(10):663–673. doi: 10.1038/nrc.2016.84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Xiao Z., Dai Z., Locasale J.W. Metabolic landscape of the tumor microenvironment at single cell resolution. Nat. Commun. 2019;10(1):3763. doi: 10.1038/s41467-019-11738-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Kumar S., Sharife H., Kreisel T., Mogilevsky M., Bar-Lev L., Grunewald M., Aizenshtein E., Karni R., Paldor I., Shlomi T., Keshet E. Intra-tumoral metabolic zonation and resultant phenotypic diversification are dictated by blood vessel proximity. Cell Metab. 2019;30(1):201–211.e6. doi: 10.1016/j.cmet.2019.04.003. [DOI] [PubMed] [Google Scholar]
- 16.Velayutham M., Hemann C., Zweier J.L. Removal of H2O2 and generation of superoxide radical: Role of cytochrome c and NADH. Free Radic. Biol. Med. 2011;51(1):160–170. doi: 10.1016/j.freeradbiomed.2011.04.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Edmondson D. Hydrogen peroxide produced by mitochondrial monoamine oxidase catalysis: biological implications. Curr. Pharm. Des. 2014;20(2):155–160. doi: 10.2174/13816128113190990406. [DOI] [PubMed] [Google Scholar]
- 18.Loschen G., Azzi A., Richter C., Flohé L. Superoxide radicals as precursors of mitochondrial hydrogen peroxide. FEBS Lett. 1974;42(1):68–72. doi: 10.1016/0014-5793(74)80281-4. [DOI] [PubMed] [Google Scholar]
- 19.Buettner G.R., Ng C.F., Wang M., Rodgers V.G.J., Schafer F.Q. A new paradigm: manganese superoxide dismutase influences the production of H2O2 in cells and thereby their biological state. Free Radic. Biol. Med. 2006;41(8):1338–1350. doi: 10.1016/j.freeradbiomed.2006.07.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Roscoe J.M., Sevier C.S. Pathways for sensing and responding to hydrogen peroxide at the endoplasmic reticulum. Cells. 2020;9(10):2314. doi: 10.3390/cells9102314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Aviello G., Knaus U.G. NADPH oxidases and ROS signaling in the gastrointestinal tract. Mucosal Immunol. 2018;11(4):1011–1023. doi: 10.1038/s41385-018-0021-8. [DOI] [PubMed] [Google Scholar]
- 22.Sirokmány G., Geiszt M. The relationship of NADPH oxidases and heme peroxidases: Fallin’ in and out. Front. Immunol. 2019;10:394. doi: 10.3389/fimmu.2019.00394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Bienert G.P., Schjoerring J.K., Jahn T.P. Membrane transport of hydrogen peroxide. Biochim. Biophys. Acta Biomembr. 2006;1758(8):994–1003. doi: 10.1016/j.bbamem.2006.02.015. [DOI] [PubMed] [Google Scholar]
- 24.Lefèvre C.T., Bennet M., Landau L., Vach P., Pignol D., Bazylinski D.A., Frankel R.B., Klumpp S., Faivre D. Diversity of magneto-aerotactic behaviors and oxygen sensing mechanisms in cultured magnetotactic bacteria. Biophys. J. 2014;107(2):527–538. doi: 10.1016/j.bpj.2014.05.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Katakwar P., Metgud R., Naik S., Mittal R. Oxidative stress marker in oral cancer: A review. J. Cancer Res. Ther. 2016;12(2):438–446. doi: 10.4103/0973-1482.151935. [DOI] [PubMed] [Google Scholar]
- 26.Marinho H.S., Real C., Cyrne L., Soares H., Antunes F. Hydrogen peroxide sensing, signaling and regulation of transcription factors. Redox Biol. 2014;2:535–562. doi: 10.1016/j.redox.2014.02.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Forman H.J., Maiorino M., Ursini F. Signaling functions of reactive oxygen species. Biochemistry. 2010;49(5):835–842. doi: 10.1021/bi9020378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Tochigi M., Inoue T., Suzuki-Karasaki M., Ochiai T., Ra C., Suzuki-Karasaki Y. Hydrogen peroxide induces cell death in human TRAIL-resistant melanoma through intracellular superoxide generation. Int. J. Oncol. 2013;42(3):863–872. doi: 10.3892/ijo.2013.1769. [DOI] [PubMed] [Google Scholar]
- 29.Okamoto M., Reddy J.K., Oyasu R. Tumorigenic conversion of a non-tumorigenic rat urothelial cell line by overexpression of H2O2-generating peroxisomal fatty acyl-CoA oxidase. Int. J. Cancer. 1997;70(6):716–721. doi: 10.1002/(SICI)1097-0215(19970317)70:6<716::AID-IJC14>3.0.CO;2-7. [DOI] [PubMed] [Google Scholar]
- 30.Hirst J., Carroll J., Fearnley I.M., Shannon R.J., Walker J.E. The nuclear encoded subunits of complex I from bovine heart mitochondria. Biochim. Biophys. Acta Bioenerg. 2003;1604(3):135–150. doi: 10.1016/S0005-2728(03)00059-8. [DOI] [PubMed] [Google Scholar]
- 31.Sazanov L.A. Respiratory complex I: mechanistic and structural insights provided by the crystal structure of the hydrophilic domain. Biochemistry. 2007;46(9):2275–2288. doi: 10.1021/bi602508x. [DOI] [PubMed] [Google Scholar]
- 32.Murphy M.P. How mitochondria produce reactive oxygen species. Biochem. J. 2009;417(1):1–13. doi: 10.1042/BJ20081386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Kussmaul L., Hirst J. The mechanism of superoxide production by NADH:ubiquinone oxidoreductase (complex I) from bovine heart mitochondria. Proc. Natl. Acad. Sci. USA. 2006;103(20):7607–7612. doi: 10.1073/pnas.0510977103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Chance B., Hollunger G. The interaction of energy and electron transfer reactions in mitochondria. I. General properties and nature of the products of succinate-linked reduction of pyridine nucleotide. J. Biol. Chem. 1961;236(5):1534–1543. doi: 10.1016/S0021-9258(18)64210-3. [DOI] [PubMed] [Google Scholar]
- 35.Cino M., Del Maestro R.F. Generation of hydrogen peroxide by brain mitochondria: The effect of reoxygenation following postdecapitative ischemia. Arch. Biochem. Biophys. 1989;269(2):623–638. doi: 10.1016/0003-9861(89)90148-3. [DOI] [PubMed] [Google Scholar]
- 36.Hunte C., Palsdottir H., Trumpower B.L. Protonmotive pathways and mechanisms in the cytochrome bc 1 complex. FEBS Lett. 2003;545(1):39–46. doi: 10.1016/S0014-5793(03)00391-0. [DOI] [PubMed] [Google Scholar]
- 37.Dröse S., Brandt U. Molecular mechanisms of superoxide production by the mitochondrial respiratory chain. Adv. Exp. Med. Biol. 2012;748:145–169. doi: 10.1007/978-1-4614-3573-0_6. [DOI] [PubMed] [Google Scholar]
- 38.Cadenas E., Boveris A., Ragan C.I., Stoppani A.O.M. Production of superoxide radicals and hydrogen peroxide by NADH-ubiquinone reductase and ubiquinol-cytochrome c reductase from beef-heart mitochondria. Arch. Biochem. Biophys. 1977;180(2):248–257. doi: 10.1016/0003-9861(77)90035-2. [DOI] [PubMed] [Google Scholar]
- 39.Hayyan M., Hashim M.A., AlNashef I.M. Superoxide ion: Generation and chemical implications. Chem. Rev. 2016;116(5):3029–3085. doi: 10.1021/acs.chemrev.5b00407. [DOI] [PubMed] [Google Scholar]
- 40.Dietzel P.D.C., Kremer R.K., Jansen M. Tetraorganylammonium superoxide compounds: close to unperturbed superoxide ions in the solid state. J. Am. Chem. Soc. 2004;126(14):4689–4696. doi: 10.1021/ja039880i. [DOI] [PubMed] [Google Scholar]
- 41.Okada F., Kobayashi M., Tanaka H., Kobayashi T., Tazawa H., Iuchi Y., Onuma K., Hosokawa M., Dinauer M.C., Hunt N.H. The role of nicotinamide adenine dinucleotide phosphate oxidase-derived reactive oxygen species in the acquisition of metastatic ability of tumor cells. Am. J. Pathol. 2006;169(1):294–302. doi: 10.2353/ajpath.2006.060073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Liu R., Li B., Qiu M. Elevated superoxide production by active H-ras enhances human lung WI-38VA-13 cell proliferation, migration and resistance to TNF-α. Oncogene. 2001;20(12):1486–1496. doi: 10.1038/sj.onc.1204214. [DOI] [PubMed] [Google Scholar]
- 43.Chaudhuri J., Chowdhury A.A., Biswas N., Manna A., Chatterjee S., Mukherjee T., Chaudhuri U., Jaisankar P., Bandyopadhyay S. Superoxide activates mTOR–eIF4E–Bax route to induce enhanced apoptosis in leukemic cells. Apoptosis. 2014;19(1):135–148. doi: 10.1007/s10495-013-0904-9. [DOI] [PubMed] [Google Scholar]
- 44.Thomas C., Mackey M.M., Diaz A.A., Cox D.P. Hydroxyl radical is produced via the Fenton reaction in submitochondrial particles under oxidative stress: implications for diseases associated with iron accumulation. Redox Rep. 2009;14(3):102–108. doi: 10.1179/135100009X392566. [DOI] [PubMed] [Google Scholar]
- 45.Chen S., Schopfer P. Hydroxyl-radical production in physiological reactions. A novel function of peroxidase. Eur. J. Biochem. 1999;260(3):726–735. doi: 10.1046/j.1432-1327.1999.00199.x. [DOI] [PubMed] [Google Scholar]
- 46.Babior B.M. Phagocytes and oxidative stress. Am. J. Med. 2000;109(1):33–44. doi: 10.1016/S0002-9343(00)00481-2. [DOI] [PubMed] [Google Scholar]
- 47.Wright R.M., McManaman J.L., Repine J.E. Alcohol-induced breast cancer: a proposed mechanism. Free Radic. Biol. Med. 1999;26(3-4):348–354. doi: 10.1016/S0891-5849(98)00204-4. [DOI] [PubMed] [Google Scholar]
- 48.Malins D.C., Gunselman S.J., Holmes E.H., Polissar N.L. The etiology of breast cancer characteristic alterations in hydroxyl radical-induced DNA base lesions during oncogenesis with potential for evaluating incidence risk. Cancer. 1993;71(10):3036–3043. doi: 10.1002/1097-0142(19930515)71:10<3036::AID-CNCR2820711025>3.0.CO;2-P. [DOI] [PubMed] [Google Scholar]
- 49.Wang Z., Li S., Cao Y., Tian X., Zeng R., Liao D.F., Cao D. Oxidative stress and carbonyl lesions in ulcerative colitis and associated colorectal cancer. Oxid. Med. Cell. Longev. 2016:1–15. doi: 10.1155/2016/9875298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Sumkhemthong S., Prompetchara E., Chanvorachote P., Chaotham C. Cisplatin-induced hydroxyl radicals mediate pro-survival autophagy in human lung cancer H460 cells. Biol. Res. 2021;54(1):22. doi: 10.1186/s40659-021-00346-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Ren J.G., Xia H.L., Just T., Dai Y.R. Hydroxyl radical-induced apoptosis in human tumor cells is associated with telomere shortening but not telomerase inhibition and caspase activation. FEBS Lett. 2001;488(3):123–132. doi: 10.1016/S0014-5793(00)02377-2. [DOI] [PubMed] [Google Scholar]
- 52.Tarr M., Valenzeno D.P. Singlet oxygen: the relevance of extracellular production mechanisms to oxidative stress in vivo. Photochem. Photobiol. Sci. 2003;2(4):355–361. doi: 10.1039/b211778a. [DOI] [PubMed] [Google Scholar]
- 53.Dogra V., Kim C. Singlet oxygen metabolism: From genesis to signaling. Front. Plant Sci. 2020;10:1640. doi: 10.3389/fpls.2019.01640. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Di Mascio P., Martinez G.R., Miyamoto S., Ronsein G.E., Medeiros M.H.G., Cadet J. Singlet molecular oxygen reactions with nucleic acids, lipids, and proteins. Chem. Rev. 2019;119(3):2043–2086. doi: 10.1021/acs.chemrev.8b00554. [DOI] [PubMed] [Google Scholar]
- 55.Homma T., Kobayashi S., Fujii J. Induction of ferroptosis by singlet oxygen generated from naphthalene endoperoxide. Biochem. Biophys. Res. Commun. 2019;518(3):519–525. doi: 10.1016/j.bbrc.2019.08.073. [DOI] [PubMed] [Google Scholar]
- 56.Bauer G. HOCl-dependent singlet oxygen and hydroxyl radical generation modulate and induce apoptosis of malignant cells. Anticancer Res. 2013;33(9):3589–3602. [PubMed] [Google Scholar]
- 57.Bauer G. Autoamplificatory singlet oxygen generation sensitizes tumor cells for intercellular apoptosis-inducing signaling. Mech. Ageing Dev. 2018;172:59–77. doi: 10.1016/j.mad.2017.11.005. [DOI] [PubMed] [Google Scholar]
- 58.Klotz L.O., Kröncke K.D., Sies H. Singlet oxygen-induced signaling effects in mammalian cells. Photochem. Photobiol. Sci. 2003;2(2):88–94. doi: 10.1039/b210750c. [DOI] [PubMed] [Google Scholar]
- 59.Kuang D.M., Zhao Q., Peng C., Xu J., Zhang J.P., Wu C., Zheng L. Activated monocytes in peritumoral stroma of hepatocellular carcinoma foster immune privilege and disease progression through PD-L1. J. Exp. Med. 2009;206(6):1327–1337. doi: 10.1084/jem.20082173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Kryczek I., Zou L., Rodriguez P., Zhu G., Wei S., Mottram P., Brumlik M., Cheng P., Curiel T., Myers L., Lackner A., Alvarez X., Ochoa A., Chen L., Zou W. B7-H4 expression identifies a novel suppressive macrophage population in human ovarian carcinoma. J. Exp. Med. 2006;203(4):871–881. doi: 10.1084/jem.20050930. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Tan H.Y., Wang N., Zhang C., Chan Y.T., Yuen M.F., Feng Y. Lysyl oxidase-like 4 fosters an immunosuppressive microenvironment during hepatocarcinogenesis. Hepatology. 2021;73(6):2326–2341. doi: 10.1002/hep.31600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Izawa S., Mimura K., Watanabe M., Maruyama T., Kawaguchi Y., Fujii H., Kono K. Increased prevalence of tumor-infiltrating regulatory T cells is closely related to their lower sensitivity to H2O2-induced apoptosis in gastric and esophageal cancer. Cancer Immunol. Immunother. 2013;62(1):161–170. doi: 10.1007/s00262-012-1327-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Sakaguchi S., Ono M., Setoguchi R., Yagi H., Hori S., Fehervari Z., Shimizu J., Takahashi T., Nomura T. Foxp3+CD25+CD4+ natural regulatory T cells in dominant self-tolerance and autoimmune disease. Immunol. Rev. 2006;212(1):8–27. doi: 10.1111/j.0105-2896.2006.00427.x. [DOI] [PubMed] [Google Scholar]
- 64.Beyer M., Schultze J.L. Regulatory T cells in cancer. Blood. 2006;108(3):804–811. doi: 10.1182/blood-2006-02-002774. [DOI] [PubMed] [Google Scholar]
- 65.Kono K., Salazar-Onfray F., Petersson M., Hansson J., Masucci G., Wasserman K., Nakazawa T., Anderson P., Kiessling R. Hydrogen peroxide secreted by tumor-derived macrophages down-modulates signal-transducing zeta molecules and inhibits tumor-specific T cell-and natural killer cell-mediated cytotoxicity. Eur. J. Immunol. 1996;26(6):1308–1313. doi: 10.1002/eji.1830260620. [DOI] [PubMed] [Google Scholar]
- 66.Tao B., Shi J., Shuai S., Zhou H., Zhang H., Li B., Wang X., Li G., He H., Zhong J. CYB561D2 up-regulation activates STAT3 to induce immunosuppression and aggression in gliomas. J. Transl. Med. 2021;19(1):338. doi: 10.1186/s12967-021-02987-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Cousin C., Aubatin A., Le Gouvello S., Apetoh L., Castellano F., Molinier-Frenkel V. The immunosuppressive enzyme IL4I1 promotes FoxP3 + regulatory T lymphocyte differentiation. Eur. J. Immunol. 2015;45(6):1772–1782. doi: 10.1002/eji.201445000. [DOI] [PubMed] [Google Scholar]
- 68.Boulland M.L., Marquet J., Molinier-Frenkel V., Möller P., Guiter C., Lasoudris F., Copie-Bergman C., Baia M., Gaulard P., Leroy K., Castellano F. Human IL4I1 is a secreted l-phenylalanine oxidase expressed by mature dendritic cells that inhibits T-lymphocyte proliferation. Blood. 2007;110(1):220–227. doi: 10.1182/blood-2006-07-036210. [DOI] [PubMed] [Google Scholar]
- 69.Lockhart D.C., Chan A.K., Mak S., Joo H.G., Daust H.A., Carritte A., Douville C.C., Goedegebuure P.S., Eberlein T.J. Loss of T-cell receptor-CD3ζ and T-cell function in tumor-infiltrating lymphocytes but not in tumor-associated lymphocytes in ovarian carcinoma. Surgery. 2001;129(6):749–756. doi: 10.1067/msy.2001.114554. [DOI] [PubMed] [Google Scholar]
- 70.Bronte V., Serafini P., De Santo C., Marigo I., Tosello V., Mazzoni A., Segal D.M., Staib C., Lowel M., Sutter G., Colombo M.P., Zanovello P. IL-4-induced arginase 1 suppresses alloreactive T cells in tumor-bearing mice. J. Immunol. 2003;170(1):270–278. doi: 10.4049/jimmunol.170.1.270. [DOI] [PubMed] [Google Scholar]
- 71.Mao Y., Poschke I., Wennerberg E., Pico de Coaña Y., Egyhazi Brage S., Schultz I., Hansson J., Masucci G., Lundqvist A., Kiessling R. Melanoma-educated CD14+ cells acquire a myeloid-derived suppressor cell phenotype through COX-2-dependent mechanisms. Cancer Res. 2013;73(13):3877–3887. doi: 10.1158/0008-5472.CAN-12-4115. [DOI] [PubMed] [Google Scholar]
- 72.Wang L., Lu Z., Zhao J., Schank M., Cao D., Dang X., Nguyen L.N., Nguyen L.N.T., Khanal S., Zhang J., Wu X.Y., El Gazzar M., Ning S., Moorman J.P., Yao Z.Q. Selective oxidative stress induces dual damage to telomeres and mitochondria in human T cells. Aging Cell. 2021;20(12):e13513. doi: 10.1111/acel.13513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Glorieux C., Xia X., He Y.Q., Hu Y., Cremer K., Robert A., Liu J., Wang F., Ling J., Chiao P.J., Huang P. Regulation of PD-L1 expression in K-ras-driven cancers through ROS-mediated FGFR1 signaling. Redox Biol. 2021;38:101780. doi: 10.1016/j.redox.2020.101780. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Thorne K.J., Svvennsen R.J., Franks D. Role of hydrogen peroxide in the cytotoxic reaction of T lymphocytes. Clin. Exp. Immunol. 1980;39(2):486–495. [PMC free article] [PubMed] [Google Scholar]
- 75.Freund E., Liedtke K.R., van der Linde J., Metelmann H.R., Heidecke C.D., Partecke L.I., Bekeschus S. Physical plasma-treated saline promotes an immunogenic phenotype in CT26 colon cancer cells in vitro and in vivo. Sci. Rep. 2019;9(1):634. doi: 10.1038/s41598-018-37169-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Lu C., Zhou F., Wu S., Liu L., Xing D. Phototherapy-induced antitumor immunity: Long-term tumor suppression effects via photoinactivation of respiratory chain oxidase-triggered superoxide anion burst. Antioxid. Redox Signal. 2016;24(5):249–262. doi: 10.1089/ars.2015.6334. [DOI] [PubMed] [Google Scholar]
- 77.Wang Y., Sun X., Han Y., Wang K., Cheng L., Sun Y., Besenbacher F., Yu M. Au@MnSe 2 core–shell nanoagent enabling immediate generation of hydroxyl radicals and simultaneous glutathione deletion free of pre-reaction for chemodynamic-photothermo-photocatalytic therapy with significant immune response. Adv. Healthc. Mater. 2022;11(14):2200041. doi: 10.1002/adhm.202200041. [DOI] [PubMed] [Google Scholar]
- 78.Zhao X., Wan X., Huang T., Yao S., Wang S., Ding Y., Zhao Y., Li Z., Li L. Acidity-responsive nanocages as robust reactive oxygen species generators with butterfly effects for maximizing oxidative damage and enhancing cancer therapy. J. Colloid Interface Sci. 2022;618:270–282. doi: 10.1016/j.jcis.2022.03.084. [DOI] [PubMed] [Google Scholar]
- 79.Farber C.M., Liebes L.F., Kanganis D.N., Silber R. Human B lymphocytes show greater susceptibility to H2O2 toxicity than T lymphocytes. J. Immunol. 1984;132(5):2543–2546. doi: 10.4049/jimmunol.132.5.2543. [DOI] [PubMed] [Google Scholar]
- 80.Silber R., Stahl R.L., Farber C.M., Kanganis D., Astrow A., Liebes L.F. Chronic lymphocytic leukemia lymphocytes: membrane anomalies and H2O2 vulnerability. Blood Cells. 1984;10(2-3):233–239. [PubMed] [Google Scholar]
- 81.Izawa S., Kono K., Mimura K., Kawaguchi Y., Watanabe M., Maruyama T., Fujii H. H2O2 production within tumor microenvironment inversely correlated with infiltration of CD56dim NK cells in gastric and esophageal cancer: possible mechanisms of NK cell dysfunction. Cancer Immunol. Immunother. 2011;60(12):1801–1810. doi: 10.1007/s00262-011-1082-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Klopotowska M., Bajor M., Graczyk-Jarzynka A., Kraft A., Pilch Z., Zhylko A., Firczuk M., Baranowska I., Lazniewski M., Plewczynski D., Goral A., Soroczynska K., Domagala J., Marhelava K., Slusarczyk A., Retecki K., Ramji K., Krawczyk M., Temples M.N., Sharma B., Lachota M., Netskar H., Malmberg K.J., Zagozdzon R., Winiarska M. PRDX-1 supports the survival and antitumor activity of primary and CAR-modified nk cells under oxidative stress. Cancer Immunol. Res. 2022;10(2):228–244. doi: 10.1158/2326-6066.CIR-20-1023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Zhou X., Zhao R., Schwarz K., Mangeat M., Schwarz E.C., Hamed M., Bogeski I., Helms V., Rieger H., Qu B. Bystander cells enhance NK cytotoxic efficiency by reducing search time. Sci. Rep. 2017;7(1):44357. doi: 10.1038/srep44357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Upadhyay S., Vaish S., Dhiman M. Hydrogen peroxide-induced oxidative stress and its impact on innate immune responses in lung carcinoma A549 cells. Mol. Cell. Biochem. 2019;450(1-2):135–147. doi: 10.1007/s11010-018-3380-2. [DOI] [PubMed] [Google Scholar]
- 85.Li F., Kitajima S., Kohno S., Yoshida A., Tange S., Sasaki S., Okada N., Nishimoto Y., Muranaka H., Nagatani N., Suzuki M., Masuda S., Thai T.C., Nishiuchi T., Tanaka T., Barbie D.A., Mukaida N., Takahashi C. Retinoblastoma inactivation induces a protumoral microenvironment via enhanced CCL2 secretion. Cancer Res. 2019;79(15):3903–3915. doi: 10.1158/0008-5472.CAN-18-3604. [DOI] [PubMed] [Google Scholar]
- 86.Sang Y., Deng Q., Cao F., Liu Z., You Y., Liu H., Ren J., Qu X. Remodeling macrophages by an iron nanotrap for tumor growth suppression. ACS Nano. 2021;15(12):19298–19309. doi: 10.1021/acsnano.1c05392. [DOI] [PubMed] [Google Scholar]
- 87.Martinez-Marin D., Jarvis C., Nelius T., de Riese W., Volpert O.V., Filleur S. PEDF increases the tumoricidal activity of macrophages towards prostate cancer cells in vitro. PLoS One. 2017;12(4):e0174968. doi: 10.1371/journal.pone.0174968. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Zuo W., Chen W., Liu J., Huang S., Chen L., Liu Q., Liu N., Jin Q., Li Y., Wang P., Zhu X. Macrophage-mimic hollow mesoporous fe-based nanocatalysts for self-amplified chemodynamic therapy and metastasis inhibition via tumor microenvironment remodeling. ACS Appl. Mater. Interfaces. 2022;14(4):5053–5065. doi: 10.1021/acsami.1c22432. [DOI] [PubMed] [Google Scholar]
- 89.Ishihara Y., Fujii T., Iijima H., Saito K., Matsunaga K. The role of neutrophils as cytotoxic cells in lung metastasis: suppression of tumor cell metastasis by a biological response modifier (PSK). In vivo . 1998;12(2):175–182. [PubMed] [Google Scholar]
- 90.Rosen G.M., Pou S., Ramos C.L., Cohen M.S., Britigan B.E. Free radicals and phagocytic cells. FASEB J. 1995;9(2):200–209. doi: 10.1096/fasebj.9.2.7540156. [DOI] [PubMed] [Google Scholar]
- 91.Murphy M.S.C., Britigan B.E., Hassett D.J., Rosen G.M. Phagocytes, O2 reduction, and hydroxyl radical. Clin. Infect. Dis. 1988;10(6):1088–1096. doi: 10.1093/clinids/10.6.1088. [DOI] [PubMed] [Google Scholar]
- 92.Butcher L.D., den Hartog G., Ernst P.B., Crowe S.E. Oxidative stress resulting from Helicobacter pylori infection contributes to gastric carcinogenesis. Cell. Mol. Gastroenterol. Hepatol. 2017;3(3):316–322. doi: 10.1016/j.jcmgh.2017.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Hoeben A., Landuyt B., Highley M.S., Wildiers H., Van Oosterom A.T., De Bruijn E.A. Vascular endothelial growth factor and angiogenesis. Pharmacol. Rev. 2004;56(4):549–580. doi: 10.1124/pr.56.4.3. [DOI] [PubMed] [Google Scholar]
- 94.Arneth B. Tumor microenvironment. Medicina (Kaunas) 2019;56(1):15. doi: 10.3390/medicina56010015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Zeng Y., Opeskin K., Horvath L.G., Sutherland R.L., Williams E.D. Lymphatic vessel density and lymph node metastasis in prostate cancer. Prostate. 2005;65(3):222–230. doi: 10.1002/pros.20288. [DOI] [PubMed] [Google Scholar]
- 96.Jennbacken K., Vallbo C., Wang W., Damber J.E. Expression of vascular endothelial growth factor C (VEGF-C) and VEGF receptor-3 in human prostate cancer is associated with regional lymph node metastasis. Prostate. 2005;65(2):110–116. doi: 10.1002/pros.20276. [DOI] [PubMed] [Google Scholar]
- 97.Muders M.H., Zhang H., Wang E., Tindall D.J., Datta K. Vascular endothelial growth factor-C protects prostate cancer cells from oxidative stress by the activation of mammalian target of rapamycin complex-2 and AKT-1. Cancer Res. 2009;69(15):6042–6048. doi: 10.1158/0008-5472.CAN-09-0552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Abumaree M.H., Hakami M., Abomaray F.M., Alshabibi M.A., Kalionis B., Al Jumah M.A., AlAskar A.S. Human chorionic villous mesenchymal stem/stromal cells modify the effects of oxidative stress on endothelial cell functions. Placenta. 2017;59:74–86. doi: 10.1016/j.placenta.2017.05.001. [DOI] [PubMed] [Google Scholar]
- 99.Basmaeil Y., Al Subayyil A., Abumaree M., Khatlani T. Conditions mimicking the cancer microenvironment modulate the functional outcome of human chorionic villus mesenchymal stem/stromal cells in vitro. Front. Cell Dev. Biol. 2021;9:650125. doi: 10.3389/fcell.2021.650125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Zhu J.W., Yu B.M., Ji Y.B., Zheng M.H., Li D.H. Upregulation of vascular endothelial growth factor by hydrogen peroxide in human colon cancer. World J. Gastroenterol. 2002;8(1):153–157. doi: 10.3748/wjg.v8.i1.153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Culp W.D., Neal R., Massey R., Egevad L., Pisa P., Garland D. Proteomic analysis of tumor establishment and growth in the B16-F10 mouse melanoma model. J. Proteome Res. 2006;5(6):1332–1343. doi: 10.1021/pr060059q. [DOI] [PubMed] [Google Scholar]
- 102.Du X., Xu Q., Pan D., Xu D., Niu B., Hong W., Zhang R., Li X., Chen S. HIC-5 in cancer-associated fibroblasts contributes to esophageal squamous cell carcinoma progression. Cell Death Dis. 2019;10(12):873. doi: 10.1038/s41419-019-2114-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Martinez-Outschoorn U.E., Balliet R.M., Lin Z., Whitaker-Menezes D., Howell A., Sotgia F., Lisanti M.P. Hereditary ovarian cancer and two-compartment tumor metabolism. Cell Cycle. 2012;11(22):4152–4166. doi: 10.4161/cc.22226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Toullec A., Gerald D., Despouy G., Bourachot B., Cardon M., Lefort S., Richardson M., Rigaill G., Parrini M.C., Lucchesi C., Bellanger D., Stern M.H., Dubois T., Sastre-Garau X., Delattre O., Vincent-Salomon A., Mechta-Grigoriou F. Oxidative stress promotes myofibroblast differentiation and tumour spreading. EMBO Mol. Med. 2010;2(6):211–230. doi: 10.1002/emmm.201000073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Balliet R.M., Capparelli C., Guido C., Pestell T.G., Martinez-Outschoorn U.E., Lin Z., Whitaker-Menezes D., Chiavarina B., Pestell R.G., Howell A., Sotgia F., Lisanti M.P. Mitochondrial oxidative stress in cancer-associated fibroblasts drives lactate production, promoting breast cancer tumor growth. Cell Cycle. 2011;10(23):4065–4073. doi: 10.4161/cc.10.23.18254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Liang L., Li W., Li X., Jin X., Liao Q., Li Y., Zhou Y. ‘Reverse Warburg effect’ of cancer-associated fibroblasts (Review). Int. J. Oncol. 2022;60(6):67. doi: 10.3892/ijo.2022.5357. [DOI] [PubMed] [Google Scholar]
- 107.Jezierska-Drutel A., Rosenzweig S.A., Neumann C.A. Role of oxidative stress and the microenvironment in breast cancer development and progression. Adv. Cancer Res. 2013;119:107–125. doi: 10.1016/B978-0-12-407190-2.00003-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Martinez-Outschoorn U.E., Lin Z., Trimmer C., Flomenberg N., Wang C., Pavlides S., Pestell R.G., Howell A., Sotgia F., Lisanti M.P. Cancer cells metabolically “fertilize” the tumor microenvironment with hydrogen peroxide, driving the Warburg effect. Cell Cycle. 2011;10(15):2504–2520. doi: 10.4161/cc.10.15.16585. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Lisanti M.P., Martinez-Outschoorn U.E., Lin Z., Pavlides S., Whitaker-Menezes D., Pestell R.G., Howell A., Sotgia F. Hydrogen peroxide fuels aging, inflammation, cancer metabolism and metastasis. Cell Cycle. 2011;10(15):2440–2449. doi: 10.4161/cc.10.15.16870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Hsieh C.L., Liu C.M., Chen H.A., Yang S.T., Shigemura K., Kitagawa K., Yamamichi F., Fujisawa M., Liu Y.R., Lee W.H., Chen K.C., Shen C.N., Lin C.C., Chung L.W.K., Sung S.Y. Reactive oxygen species–mediated switching expression of MMP-3 in stromal fibroblasts and cancer cells during prostate cancer progression. Sci. Rep. 2017;7(1):9065. doi: 10.1038/s41598-017-08835-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Martinez-Outschoorn U.E., Trimmer C., Lin Z., Whitaker-Menezes D., Chiavarina B., Zhou J., Wang C., Pavlides S., Martinez-Cantarin M.P., Capozza F., Witkiewicz A.K., Flomenberg N., Howell A., Pestell R.G., Caro J., Lisanti M.P., Sotgia F. Autophagy in cancer associated fibroblasts promotes tumor cell survival. Cell Cycle. 2010;9(17):3515–3533. doi: 10.4161/cc.9.17.12928. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Trimmer C., Sotgia F., Whitaker-Menezes D., Balliet R.M., Eaton G., Martinez-Outschoorn U.E., Pavlides S., Howell A., Iozzo R.V., Pestell R.G., Scherer P.E., Capozza F., Lisanti M.P. Caveolin-1 and mitochondrial SOD2 (MnSOD) function as tumor suppressors in the stromal microenvironment. Cancer Biol. Ther. 2011;11(4):383–394. doi: 10.4161/cbt.11.4.14101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Golden B.O., Griess B., Mir S., Fitzgerald M., Kuperwasser C., Domann F., Teoh-Fitzgerald M. Extracellular superoxide dismutase inhibits hepatocyte growth factor-mediated breast cancer-fibroblast interactions. Oncotarget. 2017;8(64):107390–107408. doi: 10.18632/oncotarget.22379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Abdian N., Ghasemi-Dehkordi P., Hashemzadeh-Chaleshtori M., Ganji-Arjenaki M., Doosti A., Amiri B. Comparison of human dermal fibroblasts (HDFs) growth rate in culture media supplemented with or without basic fibroblast growth factor (bFGF). Cell Tissue Bank. 2015;16(4):487–495. doi: 10.1007/s10561-015-9494-9. [DOI] [PubMed] [Google Scholar]
- 115.Ma C., Bower K.A., Chen G., Shi X., Ke Z.J., Luo J. Interaction between ERK and GSK3beta mediates basic fibroblast growth factor-induced apoptosis in SK-N-MC neuroblastoma cells. J. Biol. Chem. 2008;283(14):9248–9256. doi: 10.1074/jbc.M707316200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Wang Z., Zheng R., Fu S., Chen Y., Duan G., Qin D., Liu G. Role of superoxide anion on the proliferation and c-Ha- ras or p53 expression in prostate cancer cell line PC3. Urol. Res. 1998;26(5):349–353. doi: 10.1007/s002400050068. [DOI] [PubMed] [Google Scholar]
- 117.Arnold R.S., Shi J., Murad E., Whalen A.M., Sun C.Q., Polavarapu R., Parthasarathy S., Petros J.A., Lambeth J.D. Hydrogen peroxide mediates the cell growth and transformation caused by the mitogenic oxidase Nox1. Proc. Natl. Acad. Sci. USA. 2001;98(10):5550–5555. doi: 10.1073/pnas.101505898. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Arbiser J.L., Petros J., Klafter R., Govindajaran B., McLaughlin E.R., Brown L.F., Cohen C., Moses M., Kilroy S., Arnold R.S., Lambeth J.D. Reactive oxygen generated by Nox1 triggers the angiogenic switch. Proc. Natl. Acad. Sci. USA. 2002;99(2):715–720. doi: 10.1073/pnas.022630199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Kuroki M., Voest E.E., Amano S., Beerepoot L.V., Takashima S., Tolentino M., Kim R.Y., Rohan R.M., Colby K.A., Yeo K.T., Adamis A.P. Reactive oxygen intermediates increase vascular endothelial growth factor expression in vitro and in vivo. J. Clin. Invest. 1996;98(7):1667–1675. doi: 10.1172/JCI118962. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Maehata Y., Ozawa S., Kobayashi K., Kato Y., Yoshino F., Miyamoto C., Izukuri K., Kubota E., Hata R.I., Lee M.C.I. Reactive oxygen species (ROS) reduce the expression of BRAK/CXCL14 in human head and neck squamous cell carcinoma cells. Free Radic. Res. 2010;44(8):913–924. doi: 10.3109/10715762.2010.490836. [DOI] [PubMed] [Google Scholar]
- 121.Kang D.H., Lee D.J., Lee K.W., Park Y.S., Lee J.Y., Lee S.H., Koh Y.J., Koh G.Y., Choi C., Yu D.Y., Kim J., Kang S.W. Peroxiredoxin II is an essential antioxidant enzyme that prevents the oxidative inactivation of VEGF receptor-2 in vascular endothelial cells. Mol. Cell. 2011;44(4):545–558. doi: 10.1016/j.molcel.2011.08.040. [DOI] [PubMed] [Google Scholar]
- 122.Pez F., Dayan F., Durivault J., Kaniewski B., Aimond G., Le Provost G.S., Deux B., Clézardin P., Sommer P., Pouysségur J., Reynaud C. The HIF-1-inducible lysyl oxidase activates HIF-1 via the Akt pathway in a positive regulation loop and synergizes with HIF-1 in promoting tumor cell growth. Cancer Res. 2011;71(5):1647–1657. doi: 10.1158/0008-5472.CAN-10-1516. [DOI] [PubMed] [Google Scholar]
- 123.Payne S.L., Fogelgren B., Hess A.R., Seftor E.A., Wiley E.L., Fong S.F.T., Csiszar K., Hendrix M.J.C., Kirschmann D.A. Lysyl oxidase regulates breast cancer cell migration and adhesion through a hydrogen peroxide-mediated mechanism. Cancer Res. 2005;65(24):11429–11436. doi: 10.1158/0008-5472.CAN-05-1274. [DOI] [PubMed] [Google Scholar]
- 124.Milla Sanabria L., Rodríguez M.E., Cogno I.S., Rumie Vittar N.B., Pansa M.F., Lamberti M.J., Rivarola V.A. Direct and indirect photodynamic therapy effects on the cellular and molecular components of the tumor microenvironment. Biochim. Biophys. Acta. 2013;1835(1):36–45. doi: 10.1016/j.bbcan.2012.10.001. [DOI] [PubMed] [Google Scholar]
- 125.Burlaka A.P., Ganusevich I.I., Lozovska Y.V., Lukianova N.Y., Chekhun V.F. Redox-regulation of gelatinases during growth of cisplatin-sensitive and resistant Guerin carcinoma. Exp. Oncol. 2015;37(1):36–39. doi: 10.31768/2312-8852.2015.37(1):36-39. [DOI] [PubMed] [Google Scholar]
- 126.Ma Z., Liu X., Zhang Q., Yu Z., Gao D. Carvedilol suppresses malignant proliferation of mammary epithelial cells through inhibition of the ROS-mediated PI3K/AKT signaling pathway. Oncol. Rep. 2019;41(2):811–818. doi: 10.3892/or.2018.6873. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Wang Y.J., Yang M.C., Pan M.H. Dihydrolipoic acid inhibits tetrachlorohydroquinone-induced tumor promotion through prevention of oxidative damage. Food Chem. Toxicol. 2008;46(12):3739–3748. doi: 10.1016/j.fct.2008.09.064. [DOI] [PubMed] [Google Scholar]
- 128.Omenn G.S., Goodman G.E., Thornquist M.D., Balmes J., Cullen M.R., Glass A., Keogh J.P., Meyskens F.L., Jr, Valanis B., Williams J.H., Jr, Barnhart S., Hammar S. Effects of a combination of beta carotene and vitamin A on lung cancer and cardiovascular disease. N. Engl. J. Med. 1996;334(18):1150–1155. doi: 10.1056/NEJM199605023341802. [DOI] [PubMed] [Google Scholar]
- 129.Klein E.A., Thompson I.M., Jr, Tangen C.M., Crowley J.J., Lucia M.S., Goodman P.J., Minasian L.M., Ford L.G., Parnes H.L., Gaziano J.M., Karp D.D., Lieber M.M., Walther P.J., Klotz L., Parsons J.K., Chin J.L., Darke A.K., Lippman S.M., Goodman G.E., Meyskens F.L., Jr, Baker L.H. Vitamin E and the risk of prostate cancer. JAMA. 2011;306(14):1549–1556. doi: 10.1001/jama.2011.1437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Alpha-Tocopherol, Beta Carotene Cancer Prevention Study Group. The effect of vitamin E and beta carotene on the incidence of lung cancer and other cancers in male smokers. N. Engl. J. Med. 1994;330(15):1029–1035. doi: 10.1056/NEJM199404143301501. [DOI] [PubMed] [Google Scholar]
- 131.Singh A., Boldin-Adamsky S., Thimmulappa R.K., Rath S.K., Ashush H., Coulter J., Blackford A., Goodman S.N., Bunz F., Watson W.H., Gabrielson E., Feinstein E., Biswal S. RNAi-mediated silencing of nuclear factor erythroid-2-related factor 2 gene expression in non-small cell lung cancer inhibits tumor growth and increases efficacy of chemotherapy. Cancer Res. 2008;68(19):7975–7984. doi: 10.1158/0008-5472.CAN-08-1401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.DeNicola G.M., Karreth F.A., Humpton T.J., Gopinathan A., Wei C., Frese K., Mangal D., Yu K.H., Yeo C.J., Calhoun E.S., Scrimieri F., Winter J.M., Hruban R.H., Iacobuzio-Donahue C., Kern S.E., Blair I.A., Tuveson D.A. Oncogene-induced Nrf2 transcription promotes ROS detoxification and tumorigenesis. Nature. 2011;475(7354):106–109. doi: 10.1038/nature10189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Bauer A.K., Cho H.Y., Miller-DeGraff L., Walker C., Helms K., Fostel J., Yamamoto M., Kleeberger S.R. Targeted deletion of Nrf2 reduces urethane-induced lung tumor development in mice. PLoS One. 2011;6(10):e26590. doi: 10.1371/journal.pone.0026590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Watson J. Oxidants, antioxidants and the current incurability of metastatic cancers. Open Biol. 2013;3(1):120144. doi: 10.1098/rsob.120144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Guo L., Tan K., Wang H., Zhang X. Pterostilbene inhibits hepatocellular carcinoma through p53/SOD2/ROS-mediated mitochondrial apoptosis. Oncol. Rep. 2016;36(6):3233–3240. doi: 10.3892/or.2016.5151. [DOI] [PubMed] [Google Scholar]
- 136.Zou J., Zhang Y., Sun J., Wang X., Tu H., Geng S., Liu R., Chen Y., Bi Z. Deoxyelephantopin induces reactive oxygen species-mediated apoptosis and autophagy in human osteosarcoma cells. Cell. Physiol. Biochem. 2017;42(5):1812–1821. doi: 10.1159/000479537. [DOI] [PubMed] [Google Scholar]
- 137.Renaudin X. Reactive oxygen species and DNA damage response in cancer. Int. Rev. Cell Mol. Biol. 2021;364:139–161. doi: 10.1016/bs.ircmb.2021.04.001. [DOI] [PubMed] [Google Scholar]
- 138.Jackson A.L., Loeb L.A. The contribution of endogenous sources of DNA damage to the multiple mutations in cancer. Mutat. Res. 2001;477(1-2):7–21. doi: 10.1016/S0027-5107(01)00091-4. [DOI] [PubMed] [Google Scholar]
- 139.Xia X., Yang X., Huang P., Yan D. ROS-responsive nanoparticles formed from RGD–epothilone b conjugate for targeted cancer therapy. ACS Appl. Mater. Interfaces. 2020;12(16):18301–18308. doi: 10.1021/acsami.0c00650. [DOI] [PubMed] [Google Scholar]
- 140.Zhang Z., Lu Z., Yuan Q., Zhang C., Tang Y. ROS-Responsive and active targeted drug delivery based on conjugated polymer nanoparticles for synergistic chemo-/photodynamic therapy. J. Mater. Chem. B Mater. Biol. Med. 2021;9(9):2240–2248. doi: 10.1039/D0TB02996C. [DOI] [PubMed] [Google Scholar]
- 141.Yun K., Guo J., Zhu R., Wang T., Zhang X., Pan H., Pan W. Design of ROS-responsive hyaluronic acid–methotrexate conjugates for synergistic chemo-photothermal therapy for cancer. Mol. Pharm. 2022;19(9):3323–3335. doi: 10.1021/acs.molpharmaceut.2c00472. [DOI] [PubMed] [Google Scholar]
- 142.Mokwena M.G., Kruger C.A., Ivan M.T., Heidi A. A review of nanoparticle photosensitizer drug delivery uptake systems for photodynamic treatment of lung cancer. Photodiagn. Photodyn. Ther. 2018;22:147–154. doi: 10.1016/j.pdpdt.2018.03.006. [DOI] [PubMed] [Google Scholar]
- 143.Li S.Y., Cheng H., Qiu W.X., Zhang L., Wan S.S., Zeng J.Y., Zhang X.Z. Cancer cell membrane-coated biomimetic platform for tumor targeted photodynamic therapy and hypoxia-amplified bioreductive therapy. Biomaterials. 2017;142:149–161. doi: 10.1016/j.biomaterials.2017.07.026. [DOI] [PubMed] [Google Scholar]
- 144.Baldea I., Giurgiu L., Teacoe I.D., Olteanu D.E., Olteanu F.C., Clichici S., Filip G.A. Photodynamic therapy in melanoma - where do we stand? Curr. Med. Chem. 2019;25(40):5540–5563. doi: 10.2174/0929867325666171226115626. [DOI] [PubMed] [Google Scholar]
- 145.Ming H., Li B., Tian H., Zhou L., Jiang J., Zhang T., Qiao L., Wu P., Nice E.C., Zhang W., He W., Huang C., Zhang H. A minimalist and robust chemo-photothermal nanoplatform capable of augmenting autophagy-modulated immune response against breast cancer. Mater. Today Bio. 2022;15:100289. doi: 10.1016/j.mtbio.2022.100289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Wang C., Wang J., Zhang X., Yu S., Wen D., Hu Q., Ye Y., Bomba H., Hu X., Liu Z., Dotti G., Gu Z. In situ formed reactive oxygen species–responsive scaffold with gemcitabine and checkpoint inhibitor for combination therapy. Sci. Transl. Med. 2018;10(429):eaan3682. doi: 10.1126/scitranslmed.aan3682. [DOI] [PubMed] [Google Scholar]
- 147.Paduch R., Kandefer-Szerszeń M., Piersiak T. The importance of release of proinflammatory cytokines, ROS, and NO in different stages of colon carcinoma growth and metastasis after treatment with cytotoxic drugs. Oncol. Res. 2009;18(9):419–436. doi: 10.3727/096504010X12671222663593. [DOI] [PubMed] [Google Scholar]
- 148.Jezierska-Drutel A., Attaran S., Hopkins B.L., Skoko J.J., Rosenzweig S.A., Neumann C.A. The peroxidase PRDX1 inhibits the activated phenotype in mammary fibroblasts through regulating c-Jun N-terminal kinases. BMC Cancer. 2019;19(1):812. doi: 10.1186/s12885-019-6031-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Letchoumy P.V., Chandra Mohan K.V.P., Stegeman J.J., Gelboin H.V., Hara Y., Nagini S. in vitro antioxidative potential of lactoferrin and black tea polyphenols and protective effects in vivo on carcinogen activation, DNA damage, proliferation, invasion, and angiogenesis during experimental oral carcinogenesis. Oncol. Res. 2008;17(5):193–203. doi: 10.3727/096504008786111365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Horinaka A., Sakurai D., Ihara F., Makita Y., Kunii N., Motohashi S., Nakayama T., Okamoto Y. Invariant NKT cells are resistant to circulating CD15+ myeloid-derived suppressor cells in patients with head and neck cancer. Cancer Sci. 2016;107(3):207–216. doi: 10.1111/cas.12866. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Lasoudris F., Cousin C., Prevost-Blondel A., Martin-Garcia N., Abd-Alsamad I., Ortonne N., Farcet J.P., Castellano F., Molinier-Frenkel V. IL4I1: an inhibitor of the CD8+ antitumor T-cell response in vivo. Eur. J. Immunol. 2011;41(6):1629–1638. doi: 10.1002/eji.201041119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Otsuji M., Kimura Y., Aoe T., Okamoto Y., Saito T. Oxidative stress by tumor-derived macrophages suppresses the expression of CD3 ζ chain of T-cell receptor complex and antigen-specific T-cell responses. Proc. Natl. Acad. Sci. USA. 1996;93(23):13119–13124. doi: 10.1073/pnas.93.23.13119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Kwon D., Choi I.H. Hydrogen peroxide upregulates TNF-related apoptosis-inducing ligand (TRAIL) expression in human astroglial cells, and augments apoptosis of T cells. Yonsei Med. J. 2006;47(4):551–557. doi: 10.3349/ymj.2006.47.4.551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Kitamura T., Doughty-Shenton D., Cassetta L., Fragkogianni S., Brownlie D., Kato Y., Carragher N., Pollard J.W. Monocytes differentiate to immune suppressive precursors of metastasis-associated macrophages in mouse models of metastatic breast cancer. Front. Immunol. 2018;8:2004. doi: 10.3389/fimmu.2017.02004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Qiu J., Zhang T., Zhu X., Yang C., Wang Y., Zhou N., Ju B., Zhou T., Deng G., Qiu C. Hyperoside induces breast cancer cells apoptosis via ROS-mediated NF-κB signaling pathway. Int. J. Mol. Sci. 2019;21(1):131. doi: 10.3390/ijms21010131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Si L., Fu J., Liu W., Hayashi T., Nie Y., Mizuno K., Hattori S., Fujisaki H., Onodera S., Ikejima T. Silibinin inhibits migration and invasion of breast cancer MDA-MB-231 cells through induction of mitochondrial fusion. Mol. Cell. Biochem. 2020;463(1-2):189–201. doi: 10.1007/s11010-019-03640-6. [DOI] [PubMed] [Google Scholar]
- 157.Lin H., Liu X., Yu J., Hua F., Hu Z. Antioxidant N-acetylcysteine attenuates hepatocarcinogenesis by inhibiting ROS/ER stress in TLR2 deficient mouse. PLoS One. 2013;8(10):e74130. doi: 10.1371/journal.pone.0074130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Nishikawa M., Hashida M., Takakura Y. Catalase delivery for inhibiting ROS-mediated tissue injury and tumor metastasis. Adv. Drug Deliv. Rev. 2009;61(4):319–326. doi: 10.1016/j.addr.2009.01.001. [DOI] [PubMed] [Google Scholar]
- 159.Zhang Q., Huang Y., Yang R., Mu J., Zhou Z., Sun M. Poly-antioxidants for enhanced anti-miR-155 delivery and synergistic therapy of metastatic breast cancer. Biomater. Sci. 2022;10(13):3637–3646. doi: 10.1039/D1BM02022F. [DOI] [PubMed] [Google Scholar]
- 160.Travassos I.O., Mello-Andrade F., Caldeira R.P., Pires W.C., da Silva P.F.F., Correa R.S., Teixeira T., Martins-Oliveira A., Batista A.A., de Silveira-Lacerda E.P. Ruthenium (II)/allopurinol complex inhibits breast cancer progression via multiple targets. J. Biol. Inorg. Chem. 2021;26(4):385–401. doi: 10.1007/s00775-021-01862-y. [DOI] [PubMed] [Google Scholar]
- 161.Factor V.M., Laskowska D., Jensen M.R., Woitach J.T., Popescu N.C., Thorgeirsson S.S. Vitamin E reduces chromosomal damage and inhibits hepatic tumor formation in a transgenic mouse model. Proc. Natl. Acad. Sci. USA. 2000;97(5):2196–2201. doi: 10.1073/pnas.040428797. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Tapiero H., Townsend D.M., Tew K.D. The role of carotenoids in the prevention of human pathologies. Biomed. Pharmacother. 2004;58(2):100–110. doi: 10.1016/j.biopha.2003.12.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Nishikawa M., Hyoudou K., Kobayashi Y., Umeyama Y., Takakura Y., Hashida M. Inhibition of metastatic tumor growth by targeted delivery of antioxidant enzymes. J. Control. Release. 2005;109(1-3):101–107. doi: 10.1016/j.jconrel.2005.09.017. [DOI] [PubMed] [Google Scholar]
- 164.Xu G., Yu B., Wang R., Jiang J., Wen F., Shi X. Astragalin flavonoid inhibits proliferation in human lung carcinoma cells mediated via induction of caspase-dependent intrinsic pathway, ROS production, cell migration and invasion inhibition and targeting JAK/STAT signalling pathway. Cell. Mol. Biol. 2021;67(2):44–49. doi: 10.14715/cmb/2021.67.2.7. [DOI] [PubMed] [Google Scholar]
- 165.Phan T.N., Kim O., Ha M.T., Hwangbo C., Min B.S., Lee J.H. Albanol B from mulberries exerts anti-cancer effect through mitochondria ROS production in lung cancer cells and suppresses in vivo tumor growth. Int. J. Mol. Sci. 2020;21(24):9502. doi: 10.3390/ijms21249502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Jia X.B., Zhang Q., Xu L., Yao W.J., Wei L. Lotus leaf flavonoids induce apoptosis of human lung cancer A549 cells through the ROS/p38 MAPK pathway. Biol. Res. 2021;54(1):7. doi: 10.1186/s40659-021-00330-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Qu X., Sheng J., Shen L., Su J., Xu Y., Xie Q., Wu Y., Zhang X., Sun L. Autophagy inhibitor chloroquine increases sensitivity to cisplatin in QBC939 cholangiocarcinoma cells by mitochondrial ROS. PLoS One. 2017;12(3):e0173712. doi: 10.1371/journal.pone.0173712. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Schluterman M.K., Chapman S.L., Korpanty G., Ozumi K., Fukai T., Yanagisawa H., Brekken R.A. Loss of fibulin-5 binding to β1 integrins inhibits tumor growth by increasing the level of ROS. Dis. Model. Mech. 2010;3(5-6):333–342. doi: 10.1242/dmm.003707. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Li D., Kou Y., Gao Y., Liu S., Yang P., Hasegawa T., Su R., Guo J., Li M. Oxaliplatin induces the PARP1-mediated parthanatos in oral squamous cell carcinoma by increasing production of ROS. Aging (Albany NY) 2021;13(3):4242–4257. doi: 10.18632/aging.202386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Zhou L., Yang C., Zhong W., Wang Q., Zhang D., Zhang J., Xie S., Xu M. Chrysin induces autophagy-dependent ferroptosis to increase chemosensitivity to gemcitabine by targeting CBR1 in pancreatic cancer cells. Biochem. Pharmacol. 2021;193:114813. doi: 10.1016/j.bcp.2021.114813. [DOI] [PubMed] [Google Scholar]
- 171.Chen Y.C., Shen S.C., Chow J.M., Ko C., Tseng S.W. Flavone inhibition of tumor growth via apoptosis in vitro and in vivo. Int. J. Oncol. 2004;25(3):661–670. doi: 10.3892/ijo.25.3.661. [DOI] [PubMed] [Google Scholar]
- 172.Li Y., Zhou Y., Wang M., Lin X., Zhang Y., Laurent I., Zhong Y., Li J. Ampelopsin inhibits breast cancer cell growth through mitochondrial apoptosis pathway. Biol. Pharm. Bull. 2021;44(11):1738–1745. doi: 10.1248/bpb.b21-00470. [DOI] [PubMed] [Google Scholar]
- 173.Alam M., Hasan G.M., Ansari M.M., Sharma R., Yadav D.K., Hassan M.I. Therapeutic implications and clinical manifestations of thymoquinone. Phytochemistry. 2022;200:113213. doi: 10.1016/j.phytochem.2022.113213. [DOI] [PubMed] [Google Scholar]
- 174.Ko C.H., Shen S.C., Yang L.Y., Lin C.W., Chen Y.C. Gossypol reduction of tumor growth through ROS-dependent mitochondria pathway in human colorectal carcinoma cells. Int. J. Cancer. 2007;121(8):1670–1679. doi: 10.1002/ijc.22910. [DOI] [PubMed] [Google Scholar]
- 175.Wang M., Li K., Zou Z., Li L., Zhu L., Wang Q., Gao W., Wang Y., Huang W., Liu R., Yao K., Liu Q. Piperidine nitroxide Tempol enhances cisplatin-induced apoptosis in ovarian cancer cells. Oncol. Lett. 2018;16(4):4847–4854. doi: 10.3892/ol.2018.9289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Zou Z.W., Liu T., Li Y., Chen P., Peng X., Ma C., Zhang W.J., Li P.D. Melatonin suppresses thyroid cancer growth and overcomes radioresistance via inhibition of p65 phosphorylation and induction of ROS. Redox Biol. 2018;16:226–236. doi: 10.1016/j.redox.2018.02.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Jeon H., Jin Y., Myung C.S., Heo K.S. Ginsenoside-Rg2 exerts anti-cancer effects through ROS-mediated AMPK activation associated mitochondrial damage and oxidation in MCF-7 cells. Arch. Pharm. Res. 2021;44(7):702–712. doi: 10.1007/s12272-021-01345-3. [DOI] [PubMed] [Google Scholar]
- 178.Biswas S., Zhao X., Mone A.P., Mo X., Vargo M., Jarjoura D., Byrd J.C., Muthusamy N. Arsenic trioxide and ascorbic acid demonstrate promising activity against primary human CLL cells in vitro. Leuk. Res. 2010;34(7):925–931. doi: 10.1016/j.leukres.2010.01.020. [DOI] [PMC free article] [PubMed] [Google Scholar]




