Next Article in Journal
Preliminary Studies about Valorization of Acmella oleracea Bioactive Content in Modern Dermato-Cosmetic Applications to Combat Skin Oxidative Stress
Previous Article in Journal
Dipeptidyl Peptidase (DPP)-4 Inhibitors and Pituitary Adenylate Cyclase-Activating Polypeptide, a DPP-4 Substrate, Extend Neurite Outgrowth of Mouse Dorsal Root Ganglia Neurons: A Promising Approach in Diabetic Polyneuropathy Treatment
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Balanced Duality: H2O2-Based Therapy in Cancer and Its Protective Effects on Non-Malignant Tissues

Department of Radiation Oncology, The University of Iowa, Iowa City, IA 52242, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(16), 8885; https://doi.org/10.3390/ijms25168885
Submission received: 31 July 2024 / Revised: 10 August 2024 / Accepted: 11 August 2024 / Published: 15 August 2024
(This article belongs to the Section Molecular Oncology)

Abstract

:
Conventional cancer therapy strategies, although centered around killing tumor cells, often lead to severe side effects on surrounding normal tissues, thus compromising the chronic quality of life in cancer survivors. Hydrogen peroxide (H2O2) is a secondary signaling molecule that has an array of functions in both tumor and normal cells, including the promotion of cell survival pathways and immune cell modulation in the tumor microenvironment. H2O2 is a reactive oxygen species (ROS) crucial in cellular homeostasis and signaling (at concentrations maintained under nM levels), with increased steady-state levels in tumors relative to their normal tissue counterparts. Increased steady-state levels of H2O2 in tumor cells, make them vulnerable to oxidative stress and ultimately, cell death. Recently, H2O2-producing therapies—namely, pharmacological ascorbate and superoxide dismutase mimetics—have emerged as compelling complementary treatment strategies in cancer. Both pharmacological ascorbate and superoxide dismutase mimetics can generate excess H2O2 to overwhelm the impaired H2O2 removal capacity of cancer cells. This review presents an overview of H2O2 metabolism in the physiological and malignant states, in addition to discussing the anti-tumor and normal tissue-sparing mechanism(s) of, and clinical evidence for, two H2O2-based therapies, pharmacological ascorbate and superoxide dismutase mimetics.

1. Introduction

Hydrogen peroxide (H2O2) was discovered in 1818 by French chemist Louis Jacques Thénard for its use as a bleaching agent and disinfectant and in the late 19th to early 20th centuries for its antiseptic qualities. Over 200 years later, the knowledge of the nuanced roles of H2O2 within biological systems makes it one of the most studied molecules in biology for its complex roles in health and disease [1,2,3]. H2O2 is a reactive oxygen species and a weak acid primarily generated within the mitochondria as a byproduct of aerobic respiration and by some enzymes, including NADPH oxidases (NOXs). Mitochondria generate H2O2 through mitochondrial superoxide dismutase (SOD2), which catalyzes the removal of superoxide (O2•−) produced by a leak of electrons from redox centers (complexes I, II, and III) in the electron transport chain (ETC) and yields H2O2 [4,5]. Therefore, O2•−-generating NOX enzymes (e.g., NOX2) can generate H2O2 indirectly through the dismutation of superoxide released by the enzyme. In contrast, NOX4 can directly generate H2O2 through the activity of their peroxidase-like domain [6,7].
The ability of H2O2 to diffuse across membranes and react with various biomolecules, including DNA, proteins, and lipids, has been shown to disrupt cellular function, leading to cell death (Table 1) [8]. H2O2 can impair mitochondrial function, leading to decreased ATP synthesis and increased oxidative stress [9]. This in turn creates a bioenergetic deficiency, creating a positive feedback loop that exacerbates cell death, particularly in cancer cells. H2O2 can also cause damage through mechanisms that include Fenton chemistry, which involves the reaction between H2O2 and redox-active iron, leading to the formation of hydroxyl radicals that can, in turn, induce site-specific DNA damage, in addition to lipid and protein damage [10]. Additionally, protein modifications caused by H2O2 lead to redox changes, irreversible oxidation, protein fragmentation, mitochondrial oxidative protein accumulation, and kinase activation that subsequently affect the structural and functional integrity of proteins [11,12,13]. Therefore, H2O2 levels are tightly regulated through an antioxidant defense machinery composed of antioxidant enzymes, including catalase (CAT), glutathione peroxidase 1 (GPx1), and peroxiredoxins (Prxs) [14,15,16]. Catalase can efficiently remove H2O2 by directly converting it to oxygen (O2) and water (H2O) [14,15]. GPx1 utilizes intracellular glutathione (GSH) as a reducing equivalent for converting H2O2 into H2O, whereas Prxs utilize electrons from thioredoxin to reduce H2O2 to H2O [14,16]. Collectively this intricate antioxidant enzyme network regulates the intracellular steady-state levels of H2O2 to enable its functional utilization and maintain a non-toxic homeostasis. As an endogenous oxidant, H2O2 is a relatively stable form of reactive oxygen species (ROS) that participates in cellular activities, including proliferation, redox signaling and regulation, antioxidant function, and inflammation [17]. However, disruptions in H2O2 metabolism have been linked to a plethora of diseases, including diabetes, neurodegenerative disease, cardiovascular disease, and cancer [18,19,20,21]. Despite significant advancements in cancer therapy over the past two decades, a poor prognosis, chemo- and radioresistance, and therapy-associated toxicities persist that warrant the critical need for novel approaches to enhance conventional chemoradiation with a minimal toxicity to normal tissues. In this regard, targeting H2O2 metabolism has emerged as a promising approach in cancer therapy, due to its multifaceted role in regulating cancer cell proliferation and death. This article discusses the physiological roles of H2O2, the mechanisms of utilization and metabolism of H2O2 by cancer cells, the strategies for targeting H2O2 in cancer, and the role of H2O2 in mitigating normal tissue toxicities.

2. H2O2 as a Signaling Molecule

The conventional role of H2O2 has been synonymous with ROS and oxidative stress (the pathological accumulation of ROS); over the years, research has highlighted its function as a signaling molecule in several physiological processes. This pleiotropic nature of H2O2 underlines its dichotomy as a damaging agent and a signaling molecule.
The relative stability, tight regulation, and protein oxidation capacity of H2O2 make it an ideal second messenger for normal cell function and survival. As a second messenger, H2O2 alters protein activation via the reversible oxidation of cysteine residues on proteins into sulfonic acid or disulfide bonds with neighboring cysteines [22]. This section will highlight examples of how H2O2 as a signaling molecule can impact cell growth and survival, transcription, ion channel function, and immune modulation. A recent large-scale analysis revealed over 1000 cysteine-containing proteins potentially oxidizable by H2O2 [23]. This was consistent with previous findings indicating the involvement of H2O2 in the reversible oxidation of a variety of kinases, phosphatases, serum proteins, redox-regulating proteins, transcription factors, and oxygen carriers [22,23]. PTEN, a tumor suppressor phosphatase, is oxidized by H2O2 at Cys-124 in the active site. This leads to a disulfide bond formation with Cys-72 and the subsequent inhibition of the protein, thus promoting cell growth [24]. SH2 domain-containing phosphatases or hematopoietic cell phosphatases (SHPs), known as negative regulators of growth and proliferation, are also regulated by H2O2, where SHP-1 is oxidized at Cys-455 and SHP-2 at Cys-459, resulting in SHP inactivation and increased proliferation [25,26]. H2O2 can also promote proliferation by acting on kinases such as EGFR, where Cys-797 is oxidized by H2O2, resulting in sulfenylation of the residue and increased kinase activity and proliferation [27]. Conversely, H2O2 can suppress growth and proliferation, such as in the case of Akt2, where H2O2 acts as a negative regulator to reduce Akt2 activity by oxidizing Cys-124 [28]. These examples demonstrate the crucial role of H2O2 in cell survival, engaging the audience in the intricate mechanisms that regulate cell fates.
Some ion channels are also regulated by H2O2. As a mediator of cellular homeostasis, H2O2 contributes to vascular tone through large conductance calcium-activated potassium (BKCa) and voltage-dependent potassium (KV). The pore-forming α-subunit of the Kv channel, Kv1.5, is essential for regulating contractility, motility, proliferation, and adhesion in smooth muscles, brain cells, and disease states, including cancer and atrial fibrillation (AF) [29,30,31,32,33]. The H2O2-mediated post-translational modification of Kv1.5, via sulfonic acid formation at Cys-581, leads to internalization of the channel and a significant reduction of potassium-dependent polarization [34]. Other ion-sensitive proteins regulated by H2O2 include the neuronal calcium-binding proteins calbindin-D28k [35,36]. Calbindin D-28k contains three redox-sensitive cysteines that are thought to be targets of H2O2: Cys-187, 219, and 257 [37]. The oxidation of these residues reduces the calcium-binding affinity of calbindin D-28k and potentially leads to a reduction in calcium buffering and transport capacity [35,37].
When it comes to transcription factors, Nrf2 and NF-kB are noteworthy examples of H2O2 signaling regulating transcription. Despite being a reactive oxygen species, H2O2 plays a pivotal role in maintaining the balance between oxidative eustress (the physiologically beneficial well-tolerated accumulation of ROS) and oxidative stress. H2O2 promotes both the nuclear translocation and de novo synthesis of the transcription factor Nrf2 in a concentration-dependent manner, stimulating the transcription of several antioxidant genes, including CAT, PRDX, SOD, and GPX [38,39]. NF-kB is another transcription factor regulated by H2O2. H2O2 can regulate inflammation through its signaling action, which activates the transcription factor NF-kB by promoting the phosphorylation of IκBα, leading to NF-kB nuclear translocation and DNA binding that induces the expression of a plethora of genes associated with inflammation, such as IFNG, IL6, and CXCL11 [40,41]. In contrast, H2O2 can oxidize Cys-62 on the P50 subunit of NF-kB, thereby suppressing its DNA binding and the subsequent transcriptional events and associated inflammatory responses [42]. The effects of H2O2, therefore, may influence multiple cell populations, including immune cell populations such as neutrophils and macrophages.
Beyond NF-kB, H2O2 acts as a pro-inflammatory first messenger of cell signaling, H2O2 functionally contributes to inflammation and immune modulation. H2O2 is a chemoattractant for neutrophils via the Ca2+ permeable, transient receptor potential melastatin 2 (TRPM2) ion channel in vitro and in vivo, wherein increased concentrations of H2O2 lead to overactivation of TRPM2, flooding the cell with Ca2+ and halting neutrophil movement [43]. Conversely, an anti-inflammatory role for H2O2 has been noted in neutrophil activation in acute lung injury [44]. Zmijewski et al. showed an increase in the levels of H2O2 in acatalasemic (low levels of catalase) neutrophils and in neutrophils exposed to aminotriazole (AT) [44]. Furthermore, decreased IκB-α degradation, NF-κB nuclear accumulation, proinflammatory cytokines TNF-α, and macrophage inhibitory protein (MIP)-2 were also observed in acatalasemic neutrophils and aminotriazole-exposed neutrophils, along with a reduced severity of LPS-induced acute lung injury, indicating an anti-inflammatory role for H2O2 [44]. In addition to acting as a chemoattractant, H2O2 was also shown to aid in T cell migration upon chemokine signaling, whereby H2O2 enters T cells through aquaporin-3, activating Cdc42, thereby promoting migration [45,46]. Moreover, H2O2 was shown to activate NF-kB in monocytes/macrophages during their oxidative burst [47,48]. In addition to macrophage activation, H2O2 has been reported to regulate M1/M2 macrophage polarization in a concentration-dependent manner, where high concentrations promote M1 macrophages and lower concentrations promote M2 polarization [49]. These observations highlight the various mechanisms by which H2O2 may alter immune cell recruitment, migration, and activation.
The role of H2O2 in signaling, inflammation, the fine-tuning of cell function, and maintaining redox homeostasis makes it an essential mediator of cellular processes and, as a result, an attractive therapeutic target for various pathologies, including cancer.

3. H2O2 in Cancer: A Sliding Scale

Under physiological conditions, intercellular concentrations of H2O2 are tightly regulated by antioxidants (under nM concentrations) [50,51,52,53]. Cancer cells, however, maintain elevated concentrations of H2O2 [54,55,56,57,58]. These increased concentrations of H2O2 illustrate the ability of cancer cells to manipulate normal cellular processes, maximize their proliferative capacity, and exploit the role of H2O2 as a signaling molecule to promote metastasis. Moreover, due to its role in proliferation and inflammatory signaling, H2O2 is a central metabolite in cancer biology. H2O2 has been shown to induce malignant transformation, likely due to its ability to induce DNA damage [10,59,60]. The metabolism of H2O2 in cancer can be described as a sliding scale with the slider moving across two regions: a tumor-promoting region and a tumor-killing region (Figure 1). The tumor-promoting region consists of redox characteristics such as H2O2 dependency to promote proliferation (Figure 1A), whereas the tumor-killing region can be characterized by even higher H2O2 concentrations that surpass their cytotoxic threshold, which overwhelm the acquired ROS-related adaptations in the tumor, leading to DNA, lipid, and protein damage that triggers cell death machinery in the cell [61,62,63] (Figure 1B).
In the tumor-promoting region, NOX enzymes have been shown to contribute significantly to the elevated intracellular levels of H2O2 [64,65]. Data from the cancer genome atlas (TCGA) have shown that cancers of epithelial origin, including skin, lung, prostate, liver, stomach, brain, head and neck, and breast, overexpress one or more NOX enzymes, particularly NOX4 and DUOX1/2 [65]. Using NOX4 siRNA and NOX4 inhibitors, Yamaura et al. showed that NOX4-generated ROS/H2O2 is required for tumor progression in the melanoma transformation phenotype by promoting cell cycle progression through phase G2 [66]. In non-small-cell lung cancer (NSCLC), NOX1-generated H2O2 promotes metastasis by activating TLR-4 signaling independently of mitochondrial ROS [67]. In glioblastoma multiforme (GBM), NOX4-derived ROS have been shown to promote proliferation and radioresistance [68,69]. Furthermore, NOX4 overexpression corresponded with poor clinical outcomes in human colorectal cancer patients and increased proliferation and invasion in vitro [70]. These studies highlight the significance of NOX enzymes as sources of ROS/H2O2 in tumor promotion.
In addition to NOX hyperactivation, the mitochondrial dysfunction associated with rapid proliferation further exacerbates H2O2 accumulation in cancers [71,72]. This increase in steady-state levels of H2O2 can promote the tumor landscape, including proliferation, angiogenesis, and metastasis, and facilitate alterations in the tumor microenvironment (TME). H2O2, as a signaling molecule, can promote proliferation by acting on tumor suppressors or tumor promoters through the previously discussed cysteine oxidations, and cancer cells exploit this feature to promote proliferation. For example, in lung cancer, aquaporin-3-mediated H2O2 intake resulted in the inactivation of the tumor suppressor, PTEN, resulting in tumor progression [73]. H2O2 was found to promote gastric cancer cell proliferation by activating EGFR, one of the targets for H2O2-mediated cysteine oxidation, as previously discussed [27,74]. Similarly, H2O2 induced AP-1 activation in prostate cancer cells, increasing proliferation [75]. Moreover, breast and lung cancer cells demonstrated a concentration-dependent increase in proliferation when exposed to H2O2 [76]. Jerónimo et al. showed that H2O2 regulates angiogenesis in multiple tumor cells, including breast, colon, lung, and brain cancers [77]. These data were in line with previous findings by Xia et al., reporting that H2O2 was required for angiogenesis in ovarian cancer [78]. H2O2 has been shown to act as a tumor-promoting agent in metastasis. In pancreatic tumors, H2O2 promotes epithelial-to-mesenchymal transition and invasion by acting on EGFR, NF-kB, and ERK, an effect that was suppressed by catalase overexpression [79,80,81]. Similarly, the targeted delivery of catalase inhibited H2O2-mediated metastasis in a murine model of hepatic metastasis [82]. This metastasis-promoting effect of H2O2 was also observed by Stemberger et al. in breast cancer, Nelson et al. in sarcoma, and Polytarchou et al. in prostate cancer [75,83,84].
In addition to promoting proliferation, angiogenesis, and metastasis, H2O2 is believed to alter the TME to promote pro-tumorigenic inflammation. Martinez-Outschoorn et al. showed that hydrogen peroxide from tumor cells acts as a “fertilizer” in the TME by secreting H2O2 to target neighboring cancer-associated fibroblasts (CAFs), to propagate oxidative stress that results in CAF damage and “nutrient transfer” from CAFs to tumor cells, thus supporting tumor growth [85]. The oxidative stress and inflammation due to cancer-associated H2O2 can also impact the immune cell populations in the TME. ROS, including H2O2, have been shown to suppress anti-tumor T cell responses by promoting the generation of myeloid-derived suppressor cells, which in turn suppress T cell activation and proliferation [86]. A similar effect was observed with tumor-associated macrophages, which secrete ROS to suppress anti-tumor T cell function by signaling to activate immunosuppressive T regulatory cells, leading to increased tumor growth [87]. Furthermore, high levels of H2O2 and other ROS can promote T cell exhaustion, as the function of the T cell exhaustion PD-1 protein has been shown to correlate with ROS [88]. Taken together, it can be concluded that increased H2O2 concentrations in tumors can serve a tumor-promoting function by targeting pathways that promote proliferation, angiogenesis, and metastasis and by potentially targeting other cell populations in the TME.
Cancer cells are thought to acquire adaptations that accommodate their elevated steady-state levels of H2O2. These adaptations can vary among malignancies and often involve alterations to antioxidant transcription factors (e.g., Nrf2), NADPH, and iron metabolism to maintain that high level of H2O2 [21,89,90]. However, with H2O2 concentrations increased over the cytotoxic threshold, the slider moves toward the tumor-killing region (Figure 1B), wherein cancer cells fail to remove the excess H2O2, resulting in a higher propensity for DNA damage, apoptosis, ferroptosis, and protein damage. This principle is based on studies that previously demonstrated that most cancer cells have a significantly reduced capacity to remove H2O2 compared to their non-malignant counterparts [91,92]. An endogenous increase in H2O2 and a decreased H2O2 removal capacity leads to an impairment in eliminating excess H2O2 above their threshold, leading to cancer cell death. This presents H2O2 metabolism as a therapeutic vulnerability that can be exploited in various pathologies. The rationale for targeting H2O2 is based on the premise that increasing H2O2 levels in the tumor and its microenvironment (TME) can shift the balance toward cancer cell death. This strategy promotes DNA and protein damage, induces apoptosis and ferroptosis, and fosters an anti-tumorigenic TME, thereby enhancing the overall efficacy of cancer therapy. This principle led to numerous discoveries and successful clinical trials using H2O2-generating agents, such as pharmacological ascorbate and superoxide dismutase mimetics.

4. Targeting H2O2 in Cancer: Enhancing Conventional Therapy and Minimizing Its Normal Tissue Toxicities

Numerous findings indicate the significance of H2O2 in cancer growth, angiogenesis, and metastasis. Thus, consequent efforts have been made to develop therapies targeting H2O2 in cancer. This section focuses on two H2O2-based cancer therapeutic modalities, pharmacological ascorbate (gram doses delivered intravenously reaching plasma concentrations ≥ 20 mM; P-AscH), and superoxide dismutase (SOD) mimetics, in the context of the balanced duality of these agents to potentially protect non-malignant tissues from therapy-induced toxicities.

4.1. Pharmacological Ascorbate

Ascorbate (ascorbic acid or Vitamin C) is an essential dietary vitamin for multiple cellular processes such as collagen biosynthesis, antioxidant defense, epigenetic regulation, and immune function [93]. Ascorbate can undergo autooxidation or metal-catalyzed oxidation, resulting in H2O2 formation. However, metal-catalyzed ascorbate oxidation is thermodynamically favorable at a physiological pH of 8.5. Dietary vitamin C intake accomplishes physiological plasma concentrations of ≈0.8 mM, tightly regulated through urinary excretion [94,95,96,97]. However, when administered intravenously, ascorbate demonstrates exceptional pharmacokinetics that circumvents physiological excretion and allows plasma concentrations to reach upwards of 20 mM [95,96,98]. This pharmacokinetics is precipitated in the concept of P-AscH, which indicates high doses of intravenous vitamin C, yielding supraphysiological plasma concentrations [95]. P-AscH became one of the emerging strategies for cancer therapy concurrent with conventional chemoradiation, particularly following the discoveries of Mark Levine and his collaborators in pre-clinical settings, which demonstrated that P-AscH can selectively induce cancer cell killing, while sparing non-malignant cells in an H2O2-dependent manner [61,99,100,101]. Studies by Levine et al. showed that P-AscH delivers H2O2 to the tumor cells and participates in Fenton reactions with freely chelatable, redox-active intracellular iron, leading to oxidative damage [61,99,100]. Simultaneously, in non-malignant cells, the pro-oxidant role of P-AscH is dampened by the increased metabolism of H2O2 [91,102,103]. Additionally, a tightly regulated labile iron pool in non-malignant cells alleviates the pro-oxidant effects of P-AscH, thereby allowing it to function as an antioxidant [102,103]. This conclusion was based on two studies by Chen et al. where P-AscH demonstrated selective cytotoxicity in human lymphoma and breast cancer and murine colon, lung, melanoma, and kidney cancers [61]. Conversely, minimal toxicity was observed in normal breast cells, fibroblasts, and blood cells [61]. These findings were later confirmed in another study, which showed that cancer cells are less efficient at metabolizing H2O2 than non-cancerous cells, using pancreatic, lung, liver, intestine, and skin cancer cells and normal cells from the corresponding tissues [91].
The cytotoxic effects of P-AscH on cancer cells were reproduced in vitro in numerous studies on brain, colon, pancreatic, ovarian, prostate, breast, and other cancer cells [104,105,106,107,108,109]. Consistently, P-AscH demonstrated toxicity in murine models of various cancers, along with a significant enhancement of chemotherapy and radiation. Xenograft models of ovarian and pancreatic cancers that utilized P-AscH as a single agent demonstrated that daily administration of P-AscH showed a significant reduction in tumor burden by up to 53% [99]. Similarly, in a syngeneic hepatocarcinoma model, P-AscH as a single agent significantly reduced tumor growth by 40% [110]. Single-agent P-AscH also showed a significant reduction in tumor burden and metastasis in a syngeneic model of hormone-refractory prostate cancer [101]. The findings of these studies generated interest in the clinical utilization of P-AscH, concurrently with conventional chemoradiation. P-AscH, combined with chemotherapy and/or radiation, significantly increased therapeutic responses in multiple in vivo tumor models. When combined with paclitaxel, carboplatin, and radiation in gastric cancer, P-AscH significantly increased overall survival and reduced metastases [111]. Another study on gastric cancer consistently showed that P-AscH significantly enhanced the efficacy of oxaliplatin and irinotecan [109]. Pancreatic cancers, known for their poor clinical outcomes, demonstrated striking sensitivity to concurrent P-AscH in vivo. Overall survival significantly increased when P-AscH was combined with radiation or FOLFIRINOX [112,113]. In ovarian cancer, P-AscH significantly improved the efficacy of the PARP inhibitor Olaparib [114]. Concurrently with radiation and ATM inhibition, P-AscH significantly increased overall survival in a syngeneic and a xenograft model of colorectal cancer [115]. Consistently, murine models of non-small-cell lung cancer (NSCLC) and glioblastoma (GBM) showed that P-AscH is synergistic with radiation combined with carboplatin in NSCLC and with radiation and temozolomide in glioblastoma (GBM) [103].
Encouraging advances in preclinical studies on the efficacy of P-AscH proved to be a segue into clinical studies, paving the way for multiple clinical trials that combined P-AscH with conventional chemoradiation to enhance therapeutic efficacy (summarized in Table 2). In GBM, patients in a phase 2 clinical trial who received P-AscH, in addition to radiation and temozolomide, demonstrated a remarkable increase in median overall survival, from 14.6 months in historical controls to 19.6 months in the trial [116]. In ovarian cancer patients, a phase 1/2a trial reported that patients receiving P-AscH concurrently with carboplatin and paclitaxel demonstrated an increase in progression-free survival at 25.5 months, compared to 16.75 months in patients treated with carboplatin and paclitaxel alone [117]. A phase 1 trial in pancreatic cancer reported an increased overall survival (12 months compared to 6 months in historical controls) and progression-free survival (26 weeks compared to 9 weeks in historical controls), indicating a potential for P-AscH- in the clinical management of pancreatic cancer, a malignancy known for poor outcomes [118]. P-AscH is currently being investigated in a phase 2 trial with gemcitabine in pancreatic cancer (NCT02905578). Other phase 1 and 1/2a trials reported similar findings in support of using P-AscH to treat pancreatic cancer [119,120]. Furthermore, in a phase 2 clinical trial on patients with advanced NSCLC, P-AscH combined with carboplatin and paclitaxel demonstrated a disease control rate of 84.2% and a confirmed objective response rate of 34.2%, relative to historical controls (15–19%) [103,121]. Interestingly, the phase 2 trial also reported an increase in effector CD8+ T cells demonstrated by P-AscH, thus highlighting the potential of P-AscH in eliciting an anti-tumor immune response in humans [121]. The effect of P-AscH on immune cells was also reported by recent preclinical studies that combine P-AscH with immune checkpoint inhibitors (anti-PD-1 and anti-CTLA-4) in lymphoma, renal cell carcinoma, melanoma, colorectal, pancreatic, and breast cancer [122,123]. Thus, mounting evidence indicates that P-AscH acts as a radio and chemosensitizer with the potential to modulate immunotherapy responses.
In contrast to acting as a radio and chemosensitizer, data suggest that P-AscH may also exhibit protective effects in non-malignant tissues against chemotherapy and radiation. Cancer therapy-induced normal tissue toxicities constitute a significant challenge that faces cancer survivors. Conventional chemotherapeutic agents, including platinum-based drugs (cisplatin, carboplatin, etc.), are known to induce nephrotoxicity, neurotoxicity, and hepatotoxicity in pediatric and adult cancer patients [124]. Radiation also exhibits a wide array of toxic effects on normal tissues, including organ dysfunction, chronic inflammation, and fibrosis in 25% of breast cancer patients, 20% of lung cancer patients, and up to 50% of head and neck cancer patients [125]. The severe effects of chemoradiation prove dose-limiting, such that it can limit the clinical outcome and the quality of life of cancer survivors. Preclinical studies in P-AscH have highlighted its potential protective effects in combating chemoradiation toxicities. Treatment with P-AscH in rats receiving abdominal irradiation protected goblet cells in the ileum by reducing mitochondrial damage and ER degranulation [126]. Similarly, P-AscH combined with the ATM inhibitor, KU60019, significantly reduced radiation-induced intestinal injury in mice, as evidenced by significant decreases in lipid peroxidation, 3-nitrotyrosine, and TGF-ß [115]. Additionally, P-AscH protected human erythrocytes from taxol toxicity and restored their structural integrity and antioxidant function, highlighting the potential of P-AscH in mitigating the normal tissue toxicities of taxols such as paclitaxel [127]. P-AscH also reversed DNA damage, lipid peroxidation, and jejunal injury in a study combining gold–palladium–polyethylene glycol-coated (Au@Pd-PEG) nanoparticles and radiation [128]. The protective effects of P-AscH were also observed in clinical settings, wherein patients enrolled in a phase 1/2a trial of ovarian cancer reported that P-AscH significantly reduced adverse events (grade 1) associated with carboplatin and paclitaxel, while showing a trend toward substantially reducing grade 2 adverse events [117]. In a small clinical trial sample (phase 1), P-AscH exhibited potential normal tissue protection against gemcitabine and radiation, where patients treated with P-AscH showed decreased plasma levels of F2-isoprostane, a marker of oxidative stress [118,129]. Moreover, further analysis of duodenal samples and samples from pancreatic cancer patients treated with chemoradiation ± P-AscH showed the possible reversal of villi damage and lipid peroxidation [130].
Pre-clinical data and early-phase clinical trials show that P-AscH is an effective strategy to selectively enhance tumor control with chemoradiation. While it may appear that P-AscH is an ideal approach to improving cancer therapy outcomes, multiple questions regarding its efficacy and limitations to its clinical use need to be addressed through further investigation. For example, P-AscH combined with FOLFOX ± Bevacizumab demonstrated promising patient outcomes in a randomized phase 3 trial in treatment-naïve patients with unresectable metastatic colorectal cancer [131]. However, this effect was limited to patients with RAS mutations, as P-AscH significantly increased progression-free survival to 9.2 months vs. 7.8 months in the control group [131]. This highlights the need for addressing the role of the tumor mutational profile in colorectal cancer responses to P-AscH, as it appears to be a prognostic factor for clinical outcomes. Similarly, a single-arm phase 2 trial in castration-resistant prostate cancer patients reported no significant changes in patient outcomes with the use of P-AscH, despite pre-clinical studies reporting tumor control benefits in prostate cancer [101,105,132]. The lack of clinical efficacy for P-AscH- in castration-resistant prostate cancer was recently confirmed in a placebo-controlled randomized clinical trial with P-AscH and docetaxel, where P-AscH did not demonstrate any additional improvements in patient responses or toxicities [133]. In pre-clinical models of soft tissue sarcoma, P-AscH significantly enhanced the efficacy of gemcitabine and radiation therapy by promoting DNA damage [102]. Subsequently, two clinical trials were initiated to investigate P-AscH in soft tissue sarcoma. P-AscH combined with gemcitabine was investigated in a phase 2 trial (NCT03468075) that was terminated due to the lack of any patient responses. Another trial investigated P-AscH with radiation (NCT03508726) and was completed, but reports on patient outcomes are yet to be published. These studies signify the need for further investigation to address variations from preclinical to clinical responses, heterogeneous patient outcomes, and possibly ascorbate resistance. Glioblastoma makes a case for continuing research efforts with P-AscH, wherein a heterogenous patient response was observed despite significant improvements in GBM patient outcomes with P-AscH [103,116,134]. Moreover, patients with tumors containing a high iron content, as revealed using iron-sensitive MRI, were significantly more responsive to P-AscH therapy [116,134]. As a result, a new clinical trial (NCT04900792) has been initiated to investigate the utility of iron oxide nanoparticle supplementation, to increase intratumoral iron to improve P-AscH responses in GBM patients [135]. Congruent to these results, recent investigations have revealed that steady-state differences in H2O2 clearance associated with GBM molecular subtypes and intratumoral iron content contribute to differential P-AscH- responses [116,134,135,136]. Therefore, concerted translational research efforts are needed that robustly contextualize and optimize the use of P-AscH across various disease sites.
Table 2. Summary of discussed clinical trials utilizing P-AscH and their reported outcomes.
Table 2. Summary of discussed clinical trials utilizing P-AscH and their reported outcomes.
TrialDiseaseTherapyOutcomesReference
NCT02344355
Phase 2
GBMP-AscH with radiation and temozolomide.Increased overall survival (19.6 months) compared to 14.6 months in historical controls.[116]
NCT00228319
Phase 1/2a
Ovarian cancerP-AscH with carboplatin and paclitaxel.
-
Increased progression-free survival (25.5 months compared to 16.75 months with carboplatin and paclitaxel alone).
-
Significant decrease in grade 1 adverse events.
[117]
NCT01049880
Phase 1
Pancreatic cancerP-AscH with gemcitabine.
-
12 months overall survival (versus 6 months historical control).
-
26 weeks progression-free survival (compared to 9 weeks in historical controls).
-
Potential decrease in oxidative damage effects and duodenal damage.
[118]
NCT00954525
Phase 1
Pancreatic cancerP-AscH with gemcitabine and erlotinib.P-AscH was well tolerated.[119]
NCT01364805 Phase 1/2aPancreatic cancerP-AscH with gemcitabine.Trend toward increasing overall survival.[120]
NCT02905578
Phase 2
Pancreatic cancerP-AscH with gemcitabine.Ongoing trial.-
NCT02420314
Phase 2
NSCLCP-AscH with carboplatin and paclitaxel.
-
Disease control rate 84.2%.
-
Objective response rate of 34.2% compared to 15–19% in historical controls.
-
Increased effector CD8+ T cells.
[121]
NCT03146962
Phase 3
Colorectal cancerP-AscH with FOLFOX ± Bevacizumab.Increased progression-free survival to 9.2 months vs. 7.8 in the control group. This effect was limited to patients with RAS-mutant tumors.[131]
NCT01080352
Phase 2
Prostate cancerP-AscHNo benefits to the use of P-AscH- as a treatment.[132]
NCT02516670
Phase 2
Prostate cancerP-AscH with docetaxel.No benefits from the addition of P-AscH- to the treatment regimen.[133]
NCT03468075
Phase 2
Soft tissue sarcomaP-AscH with gemcitabine.Terminated.-
NCT03508726
Phase 1b/2
Soft tissue sarcomaP-AscH with radiation.Completed.-
NCT04900792
Phase 1
GBMP-AscH with ferumoxytol, radiation, and temozolomide.Ongoing trial.-

4.2. Superoxide Dismutase Mimetics

Superoxide dismutase (SOD) is an O 2 -scavenging enzyme that exists in several isoforms; manganese SOD in the mitochondria (SOD2/MnSOD), copper/zinc SOD in the cytosol (SOD1 or Cu/ZnSOD), and extracellular SOD (SOD3/ECSOD) [137,138]. The primary product of the reactions driven by these SOD enzymes is H2O2 (Equation (1)), making pharmaceutical mimetics of SOD enzymes valuable tools to generate therapeutic levels of H2O2 to enhance cancer cell killing.
2 O 2 + 2 H +   S O D ,       k     6.4 10 9   M 1 s 1   H 2 O 2 + O 2
The interest in generating H2O2 using SOD mimetics dates to the 1970s, and by the 1990s, some of the earliest SOD mimetics were characterized and studied in vitro [139]. Currently, there are several classes of SOD mimetics, such as Mn salens, Mn-metalloporphyrins, Mn (II)-cyclic polyamines, and others that exhibit dismutase activity comparable to that of the native MnSOD enzyme (kcat ≈ 2 × 109 M−1s−1) [139,140,141,142,143]. Like P-AscH, SOD mimetics yield excess amounts of H2O2 that are thought to be toxic to cancer cells but are efficiently removed by non-malignant cells. However, SOD mimetics are often more studied as protective agents in normal tissue against the toxicities of conventional cancer therapy, due to their remarkable capacity to remove toxic O 2 radicals that are generated by chemotherapy and radiation. This section focuses on the pre-clinical and clinical studies that utilize SOD mimetics to protect normal tissues from chemotherapy and radiation-induced toxicities, as well as providing recent insights into the utilization of these SOD mimetics as potential chemo and radiosensitizers.
Mn salens are a class of bioavailable low-molecular-weight SOD mimetics. The salen complex, EUK-113 (analogs EUK-207 and EUK-189), exhibits a catalytic rate constant between 8 × 105 and 1.23 × 106 M−1s−1 [144,145]. EUK-207 has been reported to mitigate radiation-induced cognitive impairment in a murine model of hippocampal radiation injury [146]. In this study, the administration of EUK-207 for up to 3 months post-irradiation resulted in a significant reduction in cognitive impairment and a potential decrease in 3-NT staining in the brain [146]. Multiple studies reported similar results in radiation-induced lung injury using EUK-207 and EUK-189. Langan et al. presented preliminary evidence on EUK-189 potentially reducing DNA damage in the lung fibroblasts of partially irradiated rat lungs [147]. However, their dosing scheme failed to prevent the long-term morbidity associated with lung irradiation [147]. Later, Mahmood et al. reported that EUK-207, in combination with genistein, successfully mitigated radiation injury in rats, as evidenced by reduced fibrosis, normalized breathing rates, reduced DNA damage, TGDF-ß, and pro-inflammatory macrophages up to 7 months post-radiation, in addition to increasing survival [148,149]. These findings were consistent with another study by Gao et al., who reported a reduction of pneumonitis and fibrosis using EUK-207 in rats exposed to whole-thorax or whole-body irradiation [150]. Lastly, EUK-207 was reported to mitigate radiation-induced dermatitis and wound healing impairment in rats [151]. In this study, EUK-207 was shown to reduce dermatitis up to 90 days post-irradiation, accelerate wound healing, increase wound vasculature, and restore skin integrity and gene expression [151]. While reports on EUK-189 and EUK-207 indicated a promising potential in radioprotection, they were not explored in a clinical setting.
Mn-metalloporphyrins are another class of SOD mimics that have been studied both as anticancer drugs and as radioprotectants for normal tissues. MnTE-2-PyP (MnE) was one of the earliest Mn-metalloporphyrins to be investigated in the context of radioprotection [139]. Although MnE effectively mitigated radiation injury in the lungs, rectum, and pelvis, the bioavailability of these drugs was limited due to their inability to cross the blood–brain barrier [152,153,154,155,156,157,158]. Two highly lipophilic analogs to MnE (MnHex and MnBuOE) were investigated in murine studies [159,160,161]. Although both drugs adequately crossed the blood–brain barrier, MnHex was associated with toxicity, thus raising concerns about long-term dosage [161]. Consequently, MnBuOE became one of the most studied bioavailable Mn-metalloporphyrins. In the field of radiation-induced brain injury, MnBuOE demonstrated the capacity to promote hippocampal neurogenesis, Dcx+ neuron production, restoration of corpus callosum axons, and restoration of cognitive function [161,162]. Similar effects were reported with chemotherapy-induced brain damage as MnBuOE was able to restore spatial memory and dendritic length following treatment with doxorubicin, cyclophosphamide, and paclitaxel [163]. Additionally, when combined with Carbenoxolone-Mediated TRAIL in GBM, MnBuOE enhanced tumor killing by selectively promoting apoptosis in GBM cells but not in normal astrocytes [164]. In head and neck cancer, MnBuOE efficiently reduced mucositis and fibrosis in murine models receiving radiation to the oral mucosa and salivary glands [165,166]. Contrarily, MnBuOE was able to sensitize head and neck tumor xenografts to radiation, as evidenced by increased tumor necrosis and immune infiltrates [165,166]. Similar anticancer effects of MnBuOE were also reported, in combination with radiation, in murine melanoma and mammary carcinoma, with carboplatin in ovarian cancer, and with cisplatin in NSCLC [167,168,169]. These data demonstrate the dual ability of MnBuOE to act as a radioprotective agent in normal tissues and enhance radiation efficacy in tumor tissues. These pre-clinical studies have resulted in the initiation of multiple ongoing clinical trials investigating MnBuOE in tumor control and normal tissue protection in locally advanced head and neck cancer (NCT02990468), in high-grade glioma (NCT02655601), anal cancer (NCT03386500), and rectal cancer (NCT05254327) (Table 3).
Mn (II)-cyclic polyamines are a relatively new class of small-molecule SOD mimetics developed in the early 2000s [170]. Avasopasem manganese (also known as AVA, GC4419, or M40419) is a Mn (II)-cyclic polyamine that has recently emerged in preclinical studies and clinical trials, with characteristics that distinguish it from other SOD mimetics. With a catalytic rate constant of 2 × 107 M−1s −1, AVA is highly specific to O 2 . This specificity of AVA to O 2 relies on the Mn(II) center of AVA, which does not react with other forms of ROS such as H2O2 and nitric oxide (NO) and, thus, cannot undergo the Fenton-like reactions associated with Mn(II) recycling [171,172]. This represents a fundamental divergence from the chemical activity of the Mn(II) centers of EUK-207 and MnBuOE, which were reported to have dual SOD/catalase activity, in addition to reacting with some nitrogen species, indicating the ability to undergo reversible oxidation reactions with H2O2 [144,170,173,174,175]. One of the earliest reports of AVA as a radioprotective agent was in a hamster model of radiation-induced oral mucositis, where it successfully reduced the severity and duration of oral mucositis [176]. Thompson and colleagues reported that AVA protected mice from lethal total-body irradiation (8.5 Gy) in a dose-dependent manner, as at the highest dose (40 mg kg−1), 100% of irradiated animals survived until day 30 post-irradiation, compared to the vehicle controls where 100% of animals died by day 17 [177]. This study also reported a marked reduction in intestinal villi loss and restored lymphoid and hematopoietic tissue integrity compared to vehicle controls [177]. In human fibroblasts, AVA was able to reverse the toxicity of radiation and chemotherapy (cisplatin and carboplatin) by reducing the aging-associated DNA damage and mitochondrial dysfunction that promote age-dependent toxicity in human dermal fibroblasts [178]. In a clinical setting, AVA has been studied as a radioprotective agent against oral mucositis in head and neck cancer patients treated with radiation and cisplatin (Table 3) [179,180,181]. AVA was reported in a phase 1/2a trial to be safe and effective at reducing the incidence of severe oral mucositis by 50% compared to historical controls [181]. A randomized double-blind phase 2b trial was initiated to follow up on these findings and reported that AVA significantly reduced the incidence of severe oral mucositis to 43%, compared to 65% in the placebo group [180]. Additionally, AVA protected against cisplatin-induced renal injury in phase 2b patients, as indicated by a reduced estimated glomerular filtration rate and epithelial growth factor [182]. These findings were later confirmed in a murine model of cisplatin-induced renal injury, where AVA was able to mitigate both chronic and acute kidney injury by restoring mitochondrial function and suppressing renal inflammation by downregulating TNFα, IL1, ICAM-1, and VCAM-1 [183]. The investigation of AVA as a therapeutic agent for oral mucositis was concluded with a phase 3 double-blind placebo-controlled trial, which showed that AVA could significantly reduce the incidence (54% compared to 64% in placebo controls) and duration (8 days compared to 18 days in placebo controls) of oral mucositis in head and neck cancer patients with radiation and cisplatin [179]. These recent studies showed that AVA is effective at enhancing radiation in lung cancer and soft tissue sarcoma and accelerating wound healing with neoadjuvant radiation [184,185,186]. Beyond these clinical outcomes with AVA, more emerging preclinical studies continue to explore AVA as an anticancer agent and a normal tissue radioprotectant. Although these data show that AVA is potentially an effective H2O2-generating agent with benefits for tumor control and normal tissue protectant, like the case of P-AscH, there are mechanistic limitations that warrant continued research with these mimetics. One study reported that two SOD mimetics (MnTePyP and EUK 134) significantly reduced endothelial cell proliferation [187]. While beneficial for suppressing tumor vascularization, these data raise concerns regarding the systemic effects of excess SOD activity and their potential long-term effects on normal endothelial function, and whether tumor-targeted delivery is required for SOD mimetics. The study also reported differential effects on NF-kB activation, where it was downregulated by EUK-134 in both cancer cells and endothelial cells and upregulated in both by MnTmPyP [187]. The effect of EUK-134 is consistent with findings reported by Mapuskar et al. that TNFα, an inducer of NF-kB activation, was downregulated by AVA [183]. These variations in effects on inflammatory responses following treatment with SOD mimetics warrant further investigation as to their effects on immune cell function and inflammatory signaling. Moreover, previous studies provided evidence on aging as a potential factor that affects responses to SOD mimetics, as the SOD mimetic AVA was shown to exert protective effects in normal tissues in fibroblasts from older patients against chemoradiation, older mice against cisplatin-induced renal injury, and in older patients with cisplatin-induced acute kidney injury [178,183]. Therefore, more studies are needed to address the responses of SOD mimetics in aging, to better target the patient populations that may benefit from them in future clinical trials.
Table 3. Summary of discussed clinical trials utilizing SOD mimetics and their reported outcomes.
Table 3. Summary of discussed clinical trials utilizing SOD mimetics and their reported outcomes.
TrialDiseaseTherapyOutcomesReference
NCT02990468
Phase 1/2
Head and neck cancerMnBuOE with radiation and cisplatinOngoing trial.-
NCT02655601
Phase 2
High-grade gliomaMnBuOE with radiation and temozolomideOngoing trial.-
NCT03386500
Phase 1
Anal cancerMnBuOE with radiation, 5FU, and mitomycinOngoing trial.-
NCT05254327
Phase 2
Rectal cancerMnBuOE with radiation and chemotherapy (oxaliplatin, leucovorin, fluorouracil, capecitabine)Ongoing trial.-
NCT01921426
Phase 1b/2a
Head and neck cancerAVA with radiation and cisplatinReduction in the incidence of severe radiation-induced oral mucositis by 50% (relative to historical controls).[183]
NCT02508389
Phase 2b
Head and neck cancerAVA with radiation and cisplatin
-
Significant reduction in severe radiation-induced oral mucositis (43% vs. 65% in the placebo control).
-
Reduction in estimated glomerular filtration rate, and epithelial growth factor indicating protection against cisplatin-induced renal toxicity.
[180,182]
NCT03689712
Phase 3
Head and neck cancerAVA with radiation and cisplatin
-
Significant reduction in the incidence of radiation-induced oral mucositis (54% compared to 64% in the placebo control).
-
Significant reduction in the duration of radiation induced oral mucositis (8 days vs. 18 days in the placebo control).
[181]

5. Concluding Remarks

H2O2 is a vital signaling molecule, with over 1000 target proteins that can be modified via cysteine oxidation. This cell signaling function leads to H2O2 accumulation in cancer cells, promoting tumor growth. Therefore, extensive efforts have been made to study H2O2 as a potential therapeutic target, especially given the increasing incidence of cancer, the limited treatment options for some malignancies, and the dose-limiting toxicities associated with conventional chemoradiation. H2O2-generating treatment modalities, including P-AscH and SOD mimetics, generate excess H2O2 that acts as a toxic prooxidant in cancer cells due to their reduced H2O2 clearance capacity, but as a protective antioxidant in normal cells that can efficiently remove H2O2. P-AscH was investigated in pre-clinical studies in a wide array of malignancies, including lung, ovarian, brain, skin, kidney, and colon cancer, and it was shown to exert selective anticancer effects without normal tissue toxicities. In contrast, P-AscH protected normal tissues from chemotherapy and radiation-associated toxicities. These findings were reproducible in phase 2 clinical trials in NSCLC and GBM and are currently being investigated in a phase 2 trial for pancreatic cancer. Similarly, SOD mimetics such as MnBuOE and AVA exhibited antioxidant protection in normal tissues treated with radiation and/or chemotherapy and prooxidant effects in cancerous tissues that enhanced tumor cell killing. SOD mimetics in the clinic were successful in the mitigation of severe oral mucositis in head and neck cancer patients and are actively being investigated in head and neck, glioma, anal, and rectal cancers. While both P-AscH and SOD mimetics are relatively well studied both in pre-clinical and clinical settings, further investigation is crucial due to their specified limitations. There is a critical need to improve therapeutic responses to rare, understudied malignancies such as sarcomas, clear renal cell carcinoma (ccRCC), and others. Therefore, expanding research on H2O2 metabolism, P-AscH, and SOD mimetics in rare malignancies may yield significant benefits to these patient populations. Additionally, potential combinations between P-AscH and SOD mimetics present another noteworthy research direction that may hold value in overcoming resistance to conventional therapy and/or the limitations of P-AscH and SOD mimetics, with Heer et al. recently reporting that the combination of P-AscH and AVA significantly enhanced cancer cell killing in NSCLC and squamous cell carcinoma [188]. Long-term clinical outcomes, the effects of sex and age, and the feasibility of the clinical use of P-AscH and SOD mimetics are to be considered and addressed in future pre-clinical and clinical studies. Thus, exploiting this duality of H2O2-based therapies in cancer underscores its potential to harness oxidative stress to selectively target cancer cells, while simultaneously protecting non-cancerous tissue. Future research should focus on optimizing delivery mechanisms, understanding the underlying mechanisms and molecular interactions, and assessing chronic outcomes, to decipher the clinical benefits of this highly innovative therapeutic approach.

Author Contributions

Conceptualization, A.Z.; Curation, A.Z., M.S.P., B.G.A. and K.A.M.; visualization, A.Z.; Writing—original draft preparation, A.Z.; Writing—review and editing, A.Z., M.S.P., B.G.A. and K.A.M.; Supervision, B.G.A. and K.A.M.; Funding Acquisition, M.S.P. and B.G.A. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by NIH grants P01 CA217797 and R21CA270742 and the Radiation Research Foundation (L998500-G). Core facilities were supported in part by the Carver College of Medicine and the Holden Comprehensive Cancer Center, NIH P30 CA086862.

Acknowledgments

Allen acknowledges support for his laboratory from a sponsored research agreement with Galera Therapeutics Inc.

Conflicts of Interest

The authors declare no other conflicts of interest.

References

  1. Thenard, L. Observations sur l’Influence de l’Eau dans la Formation des Acides Oxigénés. Ann. Chim. 1818, 9, 314–317. [Google Scholar]
  2. Thenard, L.J. Nouveaux résultats sur la combination de l’oxigène avec l’eau. Annales de Chimie et de Physique 1819, 10, 335–336. [Google Scholar]
  3. Thénard, L.J. Observations sur des nouvelles combinaisons entre l’oxigène et divers acides. Ann. Chim. Phys. 1818, 8, 306–312. [Google Scholar]
  4. Chen, Q.; Vazquez, E.J.; Moghaddas, S.; Hoppel, C.L.; Lesnefsky, E.J. Production of reactive oxygen species by mitochondria: Central role of complex III. J. Biol. Chem. 2003, 278, 36027–36031. [Google Scholar] [CrossRef]
  5. Wong, H.S.; Dighe, P.A.; Mezera, V.; Monternier, P.A.; Brand, M.D. Production of superoxide and hydrogen peroxide from specific mitochondrial sites under different bioenergetic conditions. J. Biol. Chem. 2017, 292, 16804–16809. [Google Scholar] [CrossRef]
  6. Bernard, K.; Hecker, L.; Luckhardt, T.R.; Cheng, G.; Thannickal, V.J. NADPH oxidases in lung health and disease. Antioxid. Redox Signal. 2014, 20, 2838–2853. [Google Scholar] [CrossRef]
  7. Takac, I.; Schröder, K.; Zhang, L.; Lardy, B.; Anilkumar, N.; Lambeth, J.D.; Shah, A.M.; Morel, F.; Brandes, R.P. The E-loop is involved in hydrogen peroxide formation by the NADPH oxidase Nox4. J. Biol. Chem. 2011, 286, 13304–13313. [Google Scholar] [CrossRef] [PubMed]
  8. Juan, C.A.; Pérez de la Lastra, J.M.; Plou, F.J.; Pérez-Lebeña, E. The Chemistry of Reactive Oxygen Species (ROS) Revisited: Outlining Their Role in Biological Macromolecules (DNA, Lipids and Proteins) and Induced Pathologies. Int. J. Mol. Sci. 2021, 22, 4642. [Google Scholar] [CrossRef]
  9. Palma, F.R.; Gantner, B.N.; Sakiyama, M.J.; Kayzuka, C.; Shukla, S.; Lacchini, R.; Cunniff, B.; Bonini, M.G. ROS production by mitochondria: Function or dysfunction? Oncogene 2024, 43, 295–303. [Google Scholar] [CrossRef]
  10. Henle, E.S.; Linn, S. Formation, prevention, and repair of DNA damage by iron/hydrogen peroxide. J. Biol. Chem. 1997, 272, 19095–19098. [Google Scholar] [CrossRef]
  11. Gottfredsen, R.H.; Larsen, U.G.; Enghild, J.J.; Petersen, S.V. Hydrogen peroxide induce modifications of human extracellular superoxide dismutase that results in enzyme inhibition. Redox Biol. 2013, 1, 24–31. [Google Scholar] [CrossRef] [PubMed]
  12. Mailloux, R.J.; Grayson, C.; Koufos, O. Regulation of Mitochondrial Hydrogen Peroxide Availability by Protein S-glutathionylation. Cells 2022, 12, 107. [Google Scholar] [CrossRef] [PubMed]
  13. Zmijewski, J.W.; Banerjee, S.; Bae, H.; Friggeri, A.; Lazarowski, E.R.; Abraham, E. Exposure to hydrogen peroxide induces oxidation and activation of AMP-activated protein kinase. J. Biol. Chem. 2010, 285, 33154–33164. [Google Scholar] [CrossRef] [PubMed]
  14. Molavian, H.; Madani Tonekaboni, A.; Kohandel, M.; Sivaloganathan, S. The Synergetic Coupling among the Cellular Antioxidants Glutathione Peroxidase/Peroxiredoxin and Other Antioxidants and its Effect on the Concentration of H2O2. Sci. Rep. 2015, 5, 13620. [Google Scholar] [CrossRef]
  15. Deisseroth, A.; Dounce, A.L. Catalase: Physical and chemical properties, mechanism of catalysis, and physiological role. Physiol. Rev. 1970, 50, 319–375. [Google Scholar] [CrossRef] [PubMed]
  16. Lubos, E.; Loscalzo, J.; Handy, D.E. Glutathione peroxidase-1 in health and disease: From molecular mechanisms to therapeutic opportunities. Antioxid. Redox Signal 2011, 15, 1957–1997. [Google Scholar] [CrossRef]
  17. Di Marzo, N.; Chisci, E.; Giovannoni, R. The Role of Hydrogen Peroxide in Redox-Dependent Signaling: Homeostatic and Pathological Responses in Mammalian Cells. Cells 2018, 7, 156. [Google Scholar] [CrossRef]
  18. Shimokawa, H. Reactive oxygen species in cardiovascular health and disease: Special references to nitric oxide, hydrogen peroxide, and Rho-kinase. J. Clin. Biochem. Nutr. 2020, 66, 83–91. [Google Scholar] [CrossRef]
  19. Korac, B.; Kalezic, A.; Pekovic-Vaughan, V.; Korac, A.; Jankovic, A. Redox changes in obesity, metabolic syndrome, and diabetes. Redox Biol. 2021, 42, 101887. [Google Scholar] [CrossRef]
  20. Lee, Y.M.; He, W.; Liou, Y.C. The redox language in neurodegenerative diseases: Oxidative post-translational modifications by hydrogen peroxide. Cell Death Dis. 2021, 12, 58. [Google Scholar] [CrossRef]
  21. Jelic, M.D.; Mandic, A.D.; Maricic, S.M.; Srdjenovic, B.U. Oxidative stress and its role in cancer. J. Cancer Res. Ther. 2021, 17, 22–28. [Google Scholar] [CrossRef]
  22. Devarie-Baez, N.O.; Silva Lopez, E.I.; Furdui, C.M. Biological chemistry and functionality of protein sulfenic acids and related thiol modifications. Free Radic. Res. 2016, 50, 172–194. [Google Scholar] [CrossRef]
  23. Garrido Ruiz, D.; Sandoval-Perez, A.; Rangarajan, A.V.; Gunderson, E.L.; Jacobson, M.P. Cysteine Oxidation in Proteins: Structure, Biophysics, and Simulation. Biochemistry 2022, 61, 2165–2176. [Google Scholar] [CrossRef]
  24. Lee, S.R.; Yang, K.S.; Kwon, J.; Lee, C.; Jeong, W.; Rhee, S.G. Reversible inactivation of the tumor suppressor PTEN by H2O2. J. Biol. Chem. 2002, 277, 20336–20342. [Google Scholar] [CrossRef]
  25. Chen, C.Y.; Willard, D.; Rudolph, J. Redox regulation of SH2-domain-containing protein tyrosine phosphatases by two backdoor cysteines. Biochemistry 2009, 48, 1399–1409. [Google Scholar] [CrossRef] [PubMed]
  26. Crump, K.E.; Juneau, D.G.; Poole, L.B.; Haas, K.M.; Grayson, J.M. The reversible formation of cysteine sulfenic acid promotes B-cell activation and proliferation. Eur. J. Immunol. 2012, 42, 2152–2164. [Google Scholar] [CrossRef]
  27. Paulsen, C.E.; Truong, T.H.; Garcia, F.J.; Homann, A.; Gupta, V.; Leonard, S.E.; Carroll, K.S. Peroxide-dependent sulfenylation of the EGFR catalytic site enhances kinase activity. Nat. Chem. Biol. 2012, 8, 57–64. [Google Scholar] [CrossRef]
  28. Wani, R.; Qian, J.; Yin, L.; Bechtold, E.; King, S.B.; Poole, L.B.; Paek, E.; Tsang, A.W.; Furdui, C.M. Isoform-specific regulation of Akt by PDGF-induced reactive oxygen species. Proc. Natl. Acad. Sci. USA 2011, 108, 10550–10555. [Google Scholar] [CrossRef] [PubMed]
  29. Stuhmer, W.; Ruppersberg, J.P.; Schroter, K.H.; Sakmann, B.; Stocker, M.; Giese, K.P.; Perschke, A.; Baumann, A.; Pongs, O. Molecular basis of functional diversity of voltage-gated potassium channels in mammalian brain. EMBO J. 1989, 8, 3235–3244. [Google Scholar] [CrossRef]
  30. Yuan, X.J.; Wang, J.; Juhaszova, M.; Golovina, V.A.; Rubin, L.J. Molecular basis and function of voltage-gated K+ channels in pulmonary arterial smooth muscle cells. Am. J. Physiol. 1998, 274, L621–L635. [Google Scholar] [CrossRef] [PubMed]
  31. Tamargo, J.; Caballero, R.; Gomez, R.; Delpon, E. I(Kur)/Kv1.5 channel blockers for the treatment of atrial fibrillation. Expert Opin. Investig. Drugs 2009, 18, 399–416. [Google Scholar] [CrossRef] [PubMed]
  32. Comes, N.; Bielanska, J.; Vallejo-Gracia, A.; Serrano-Albarras, A.; Marruecos, L.; Gomez, D.; Soler, C.; Condom, E.; Ramon, Y.C.S.; Hernandez-Losa, J.; et al. The voltage-dependent K(+) channels Kv1.3 and Kv1.5 in human cancer. Front. Physiol. 2013, 4, 283. [Google Scholar] [CrossRef]
  33. Wettwer, E.; Terlau, H. Pharmacology of voltage-gated potassium channel Kv1.5--impact on cardiac excitability. Curr. Opin. Pharmacol. 2014, 15, 115–121. [Google Scholar] [CrossRef] [PubMed]
  34. Svoboda, L.K.; Reddie, K.G.; Zhang, L.; Vesely, E.D.; Williams, E.S.; Schumacher, S.M.; O’Connell, R.P.; Shaw, R.; Day, S.M.; Anumonwo, J.M.; et al. Redox-sensitive sulfenic acid modification regulates surface expression of the cardiovascular voltage-gated potassium channel Kv1.5. Circ. Res. 2012, 111, 842–853. [Google Scholar] [CrossRef] [PubMed]
  35. Schmidt, H. Three functional facets of calbindin D-28k. Front. Mol. Neurosci. 2012, 5, 25. [Google Scholar] [CrossRef] [PubMed]
  36. Zang, J.; Neuhauss, S.C.F. The Binding Properties and Physiological Functions of Recoverin. Front. Mol. Neurosci. 2018, 11, 473. [Google Scholar] [CrossRef] [PubMed]
  37. Cedervall, T.; Berggard, T.; Borek, V.; Thulin, E.; Linse, S.; Akerfeldt, K.S. Redox sensitive cysteine residues in calbindin D28k are structurally and functionally important. Biochemistry 2005, 44, 684–693. [Google Scholar] [CrossRef] [PubMed]
  38. Jung, K.-A.; Kwak, M.-K. The Nrf2 System as a Potential Target for the Development of Indirect Antioxidants. Molecules 2010, 15, 7266–7291. [Google Scholar] [CrossRef] [PubMed]
  39. Covas, G.; Marinho, H.S.; Cyrne, L.; Antunes, F. Chapter Nine—Activation of Nrf2 by H2O2: De Novo Synthesis Versus Nuclear Translocation. In Methods in Enzymology; Cadenas, E., Packer, L., Eds.; Academic Press: Cambridge, MA, USA, 2013; Volume 528, pp. 157–171. [Google Scholar]
  40. Takada, Y.; Mukhopadhyay, A.; Kundu, G.C.; Mahabeleshwar, G.H.; Singh, S.; Aggarwal, B.B. Hydrogen peroxide activates NF-kappa B through tyrosine phosphorylation of I kappa B alpha and serine phosphorylation of p65: Evidence for the involvement of I kappa B alpha kinase and Syk protein-tyrosine kinase. J. Biol. Chem. 2003, 278, 24233–24241. [Google Scholar] [CrossRef]
  41. Pahl, H.L. Activators and target genes of Rel/NF-kappaB transcription factors. Oncogene 1999, 18, 6853–6866. [Google Scholar] [CrossRef]
  42. Pineda-Molina, E.; Klatt, P.; Vazquez, J.; Marina, A.; Garcia de Lacoba, M.; Perez-Sala, D.; Lamas, S. Glutathionylation of the p50 subunit of NF-kappaB: A mechanism for redox-induced inhibition of DNA binding. Biochemistry 2001, 40, 14134–14142. [Google Scholar] [CrossRef]
  43. Morad, H.; Luqman, S.; Tan, C.H.; Swann, V.; McNaughton, P.A. TRPM2 ion channels steer neutrophils towards a source of hydrogen peroxide. Sci. Rep. 2021, 11, 9339. [Google Scholar] [CrossRef] [PubMed]
  44. Zmijewski, J.W.; Lorne, E.; Zhao, X.; Tsuruta, Y.; Sha, Y.; Liu, G.; Abraham, E. Antiinflammatory effects of hydrogen peroxide in neutrophil activation and acute lung injury. Am. J. Respir. Crit. Care Med. 2009, 179, 694–704. [Google Scholar] [CrossRef]
  45. Hara-Chikuma, M.; Chikuma, S.; Sugiyama, Y.; Kabashima, K.; Verkman, A.S.; Inoue, S.; Miyachi, Y. Chemokine-dependent T cell migration requires aquaporin-3-mediated hydrogen peroxide uptake. J. Exp. Med. 2012, 209, 1743–1752. [Google Scholar] [CrossRef] [PubMed]
  46. Guo, F. RhoA and Cdc42 in T cells: Are they targetable for T cell-mediated inflammatory diseases? Precis. Clin. Med. 2021, 4, 56–61. [Google Scholar] [CrossRef]
  47. Kaul, N.; Forman, H.J. Activation of NFκB by the respiratory burst of macrophages. Free Radic. Biol. Med. 1996, 21, 401–405. [Google Scholar] [CrossRef]
  48. Lu, Y.; Wahl, L.M. Oxidative Stress Augments the Production of Matrix Metalloproteinase-1, Cyclooxygenase-2, and Prostaglandin E2 through Enhancement of NF-κB Activity in Lipopolysaccharide-Activated Human Primary Monocytes1. J. Immunol. 2005, 175, 5423–5429. [Google Scholar] [CrossRef]
  49. Tan, H.Y.; Wang, N.; Li, S.; Hong, M.; Wang, X.; Feng, Y. The Reactive Oxygen Species in Macrophage Polarization: Reflecting Its Dual Role in Progression and Treatment of Human Diseases. Oxidative Med. Cell. Longev. 2016, 2016, 2795090. [Google Scholar] [CrossRef] [PubMed]
  50. Forman, H.J.; Bernardo, A.; Davies, K.J. What is the concentration of hydrogen peroxide in blood and plasma? Arch. Biochem. Biophys. 2016, 603, 48–53. [Google Scholar] [CrossRef]
  51. Halliwell, B.; Clement, M.V.; Long, L.H. Hydrogen peroxide in the human body. FEBS Lett. 2000, 486, 10–13. [Google Scholar] [CrossRef]
  52. Chance, B.; Sies, H.; Boveris, A. Hydroperoxide metabolism in mammalian organs. Physiol. Rev. 1979, 59, 527–605. [Google Scholar] [CrossRef] [PubMed]
  53. Sies, H. Role of metabolic H2O2 generation: Redox signaling and oxidative stress. J. Biol. Chem. 2014, 289, 8735–8741. [Google Scholar] [CrossRef]
  54. Lennicke, C.; Rahn, J.; Lichtenfels, R.; Wessjohann, L.A.; Seliger, B. Hydrogen peroxide—Production, fate and role in redox signaling of tumor cells. Cell Commun. Signal. 2015, 13, 39. [Google Scholar] [CrossRef]
  55. Wu, Y.; Guo, T.; Qiu, Y.; Lin, Y.; Yao, Y.; Lian, W.; Lin, L.; Song, J.; Yang, H. An inorganic prodrug, tellurium nanowires with enhanced ROS generation and GSH depletion for selective cancer therapy. Chem. Sci. 2019, 10, 7068–7075. [Google Scholar] [CrossRef] [PubMed]
  56. Szatrowski, T.P.; Nathan, C.F. Production of large amounts of hydrogen peroxide by human tumor cells. Cancer Res. 1991, 51, 794–798. [Google Scholar]
  57. Lopez-Lazaro, M. Dual role of hydrogen peroxide in cancer: Possible relevance to cancer chemoprevention and therapy. Cancer Lett. 2007, 252, 1–8. [Google Scholar] [CrossRef]
  58. Ali, T.; Li, D.; Ponnamperumage, T.N.F.; Peterson, A.K.; Pandey, J.; Fatima, K.; Brzezinski, J.; Jakusz, J.A.R.; Gao, H.; Koelsch, G.E.; et al. Generation of Hydrogen Peroxide in Cancer Cells: Advancing Therapeutic Approaches for Cancer Treatment. Cancers 2024, 16, 2171. [Google Scholar] [CrossRef]
  59. Okamoto, M.; Kawai, K.; Reznikoff, C.A.; Oyasu, R. Transformation in vitro of a nontumorigenic rat urothelial cell line by hydrogen peroxide. Cancer Res. 1996, 56, 4649–4653. [Google Scholar]
  60. 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, 5550–5555. [Google Scholar] [CrossRef]
  61. Chen, Q.; Espey, M.G.; Krishna, M.C.; Mitchell, J.B.; Corpe, C.P.; Buettner, G.R.; Shacter, E.; Levine, M. Pharmacologic ascorbic acid concentrations selectively kill cancer cells: Action as a pro-drug to deliver hydrogen peroxide to tissues. Proc. Natl. Acad. Sci. USA 2005, 102, 13604–13609. [Google Scholar] [CrossRef] [PubMed]
  62. Erudaitius, D.; Huang, A.; Kazmi, S.; Buettner, G.R.; Rodgers, V.G. Peroxiporin Expression Is an Important Factor for Cancer Cell Susceptibility to Therapeutic H2O2: Implications for Pharmacological Ascorbate Therapy. PLoS ONE 2017, 12, e0170442. [Google Scholar] [CrossRef]
  63. Erudaitius, D.; Mantooth, J.; Huang, A.; Soliman, J.; Doskey, C.M.; Buettner, G.R.; Rodgers, V.G.J. Calculated cell-specific intracellular hydrogen peroxide concentration: Relevance in cancer cell susceptibility during ascorbate therapy. Free Radic. Biol. Med. 2018, 120, 356–367. [Google Scholar] [CrossRef]
  64. Blanchetot, C.; Boonstra, J. The ROS-NOX connection in cancer and angiogenesis. Crit. Rev. Eukaryot. Gene Expr. 2008, 18, 35–45. [Google Scholar] [CrossRef] [PubMed]
  65. Meitzler, J.L.; Konate, M.M.; Doroshow, J.H. Hydrogen peroxide-producing NADPH oxidases and the promotion of migratory phenotypes in cancer. Arch. Biochem. Biophys. 2019, 675, 108076. [Google Scholar] [CrossRef] [PubMed]
  66. Yamaura, M.; Mitsushita, J.; Furuta, S.; Kiniwa, Y.; Ashida, A.; Goto, Y.; Shang, W.H.; Kubodera, M.; Kato, M.; Takata, M.; et al. NADPH oxidase 4 contributes to transformation phenotype of melanoma cells by regulating G2-M cell cycle progression. Cancer Res. 2009, 69, 2647–2654. [Google Scholar] [CrossRef]
  67. Liu, X.; Pei, C.; Yan, S.; Liu, G.; Liu, G.; Chen, W.; Cui, Y.; Liu, Y. NADPH oxidase 1-dependent ROS is crucial for TLR4 signaling to promote tumor metastasis of non-small cell lung cancer. Tumour Biol. 2015, 36, 1493–1502. [Google Scholar] [CrossRef] [PubMed]
  68. Hsieh, C.H.; Shyu, W.C.; Chiang, C.Y.; Kuo, J.W.; Shen, W.C.; Liu, R.S. NADPH oxidase subunit 4-mediated reactive oxygen species contribute to cycling hypoxia-promoted tumor progression in glioblastoma multiforme. PLoS ONE 2011, 6, e23945. [Google Scholar] [CrossRef]
  69. Garcia-Gomez, P.; Golan, I.; Dadras, M.S.; Mezheyeuski, A.; Bellomo, C.; Tzavlaki, K.; Moren, A.; Carreras-Puigvert, J.; Caja, L. NOX4 regulates TGFbeta-induced proliferation and self-renewal in glioblastoma stem cells. Mol. Oncol. 2022, 16, 1891–1912. [Google Scholar] [CrossRef]
  70. Lin, X.L.; Yang, L.; Fu, S.W.; Lin, W.F.; Gao, Y.J.; Chen, H.Y.; Ge, Z.Z. Overexpression of NOX4 predicts poor prognosis and promotes tumor progression in human colorectal cancer. Oncotarget 2017, 8, 33586–33600. [Google Scholar] [CrossRef]
  71. Laurent, A.; Nicco, C.; Chereau, C.; Goulvestre, C.; Alexandre, J.; Alves, A.; Levy, E.; Goldwasser, F.; Panis, Y.; Soubrane, O.; et al. Controlling tumor growth by modulating endogenous production of reactive oxygen species. Cancer Res. 2005, 65, 948–956. [Google Scholar] [CrossRef]
  72. Nicco, C.; Laurent, A.; Chereau, C.; Weill, B.; Batteux, F. Differential modulation of normal and tumor cell proliferation by reactive oxygen species. Biomed. Pharmacother. 2005, 59, 169–174. [Google Scholar] [CrossRef]
  73. Wang, Y.; Chen, D.; Liu, Y.; Zhang, Y.; Duan, C.; Otkur, W.; Chen, H.; Liu, X.; Xia, T.; Qi, H.; et al. AQP3-mediated H(2) O(2) uptake inhibits LUAD autophagy by inactivating PTEN. Cancer Sci. 2021, 112, 3278–3292. [Google Scholar] [CrossRef]
  74. Wang, Q.; Shen, W.; Tao, G.Q.; Sun, J.; Shi, L.P. Study on the proliferation of human gastric cancer cell AGS by activation of EGFR in H2O2. Eur. Rev. Med. Pharmacol. Sci. 2017, 21, 1006–1012. [Google Scholar] [PubMed]
  75. Polytarchou, C.; Hatziapostolou, M.; Papadimitriou, E. Hydrogen peroxide stimulates proliferation and migration of human prostate cancer cells through activation of activator protein-1 and up-regulation of the heparin affin regulatory peptide gene. J. Biol. Chem. 2005, 280, 40428–40435. [Google Scholar] [CrossRef]
  76. Galli, S.; Labato, M.I.; Bal de Kier Joffé, E.; Carreras, M.a.C.; Poderoso, J.J. Decreased Mitochondrial Nitric Oxide Synthase Activity and Hydrogen Peroxide Relate Persistent Tumoral Proliferation to Embryonic Behavior1. Cancer Res. 2003, 63, 6370–6377. [Google Scholar]
  77. Jeronimo, A.; Rodrigues, G.; Vilas-Boas, F.; Martins, G.G.; Bagulho, A.; Real, C. Hydrogen peroxide regulates angiogenesis-related factors in tumor cells. Biochem. Cell Biol. 2017, 95, 679–685. [Google Scholar] [CrossRef]
  78. Xia, C.; Meng, Q.; Liu, L.Z.; Rojanasakul, Y.; Wang, X.R.; Jiang, B.H. Reactive oxygen species regulate angiogenesis and tumor growth through vascular endothelial growth factor. Cancer Res. 2007, 67, 10823–10830. [Google Scholar] [CrossRef]
  79. Li, W.; Ma, Q.; Li, J.; Guo, K.; Liu, H.; Han, L.; Ma, G. Hyperglycemia enhances the invasive and migratory activity of pancreatic cancer cells via hydrogen peroxide. Oncol. Rep. 2011, 25, 1279–1287. [Google Scholar] [CrossRef]
  80. Li, W.; Ma, Z.; Ma, J.; Li, X.; Xu, Q.; Duan, W.; Chen, X.; Lv, Y.; Zhou, S.; Wu, E.; et al. Hydrogen peroxide mediates hyperglycemia-induced invasive activity via ERK and p38 MAPK in human pancreatic cancer. Oncotarget 2015, 6, 31119–31133. [Google Scholar] [CrossRef] [PubMed]
  81. Li, W.; Zhang, L.; Chen, X.; Jiang, Z.; Zong, L.; Ma, Q. Hyperglycemia Promotes the Epithelial-Mesenchymal Transition of Pancreatic Cancer via Hydrogen Peroxide. Oxidative Med. Cell. Longev. 2016, 2016, 5190314. [Google Scholar] [CrossRef] [PubMed]
  82. Nishikawa, M.; Tamada, A.; Hyoudou, K.; Umeyama, Y.; Takahashi, Y.; Kobayashi, Y.; Kumai, H.; Ishida, E.; Staud, F.; Yabe, Y.; et al. Inhibition of experimental hepatic metastasis by targeted delivery of catalase in mice. Clin. Exp. Metastasis 2004, 21, 213–221. [Google Scholar] [CrossRef] [PubMed]
  83. Nelson, K.K.; Ranganathan, A.C.; Mansouri, J.; Rodriguez, A.M.; Providence, K.M.; Rutter, J.L.; Pumiglia, K.; Bennett, J.A.; Melendez, J.A. Elevated Sod2 Activity Augments Matrix Metalloproteinase Expression: Evidence for the Involvement of Endogenous Hydrogen Peroxide in Regulating Metastasis1. Clin. Cancer Res. 2003, 9, 424–432. [Google Scholar] [PubMed]
  84. Stemberger, M.B.; Ju, J.A.; Thompson, K.N.; Mathias, T.J.; Jerrett, A.E.; Chang, K.T.; Ory, E.C.; Annis, D.A.; Mull, M.L.; Gilchrist, D.E.; et al. Hydrogen Peroxide Induces alpha-Tubulin Detyrosination and Acetylation and Impacts Breast Cancer Metastatic Phenotypes. Cells 2023, 12, 1266. [Google Scholar] [CrossRef] [PubMed]
  85. 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: Implications for PET imaging of human tumors. Cell Cycle 2011, 10, 2504–2520. [Google Scholar] [CrossRef] [PubMed]
  86. Liu, Y.; Wei, J.; Guo, G.; Zhou, J. Norepinephrine-induced myeloid-derived suppressor cells block T-cell responses via generation of reactive oxygen species. Immunopharmacol. Immunotoxicol. 2015, 37, 359–365. [Google Scholar] [CrossRef] [PubMed]
  87. Kraaij, M.D.; Savage, N.D.; van der Kooij, S.W.; Koekkoek, K.; Wang, J.; van den Berg, J.M.; Ottenhoff, T.H.; Kuijpers, T.W.; Holmdahl, R.; van Kooten, C. Induction of regulatory T cells by macrophages is dependent on production of reactive oxygen species. Proc. Natl. Acad. Sci. USA 2010, 107, 17686–17691. [Google Scholar] [CrossRef] [PubMed]
  88. Tkachev, V.; Goodell, S.; Opipari, A.W.; Hao, L.Y.; Franchi, L.; Glick, G.D.; Ferrara, J.L.; Byersdorfer, C.A. Programmed death-1 controls T cell survival by regulating oxidative metabolism. J. Immunol. 2015, 194, 5789–5800. [Google Scholar] [CrossRef] [PubMed]
  89. Hayes, J.D.; Dinkova-Kostova, A.T.; Tew, K.D. Oxidative Stress in Cancer. Cancer Cell 2020, 38, 167–197. [Google Scholar] [CrossRef] [PubMed]
  90. Petronek, M.S.; Spitz, D.R.; Buettner, G.R.; Allen, B.G. Linking Cancer Metabolic Dysfunction and Genetic Instability through the Lens of Iron Metabolism. Cancers 2019, 11, 1077. [Google Scholar] [CrossRef]
  91. Doskey, C.M.; Buranasudja, V.; Wagner, B.A.; Wilkes, J.G.; Du, J.; Cullen, J.J.; Buettner, G.R. Tumor cells have decreased ability to metabolize H(2)O(2): Implications for pharmacological ascorbate in cancer therapy. Redox Biol. 2016, 10, 274–284. [Google Scholar] [CrossRef]
  92. Wagner, B.A.; Witmer, J.R.; van ‘t Erve, T.J.; Buettner, G.R. An Assay for the Rate of Removal of Extracellular Hydrogen Peroxide by Cells. Redox Biol. 2013, 1, 210–217. [Google Scholar] [CrossRef] [PubMed]
  93. Carr, A.C.; Maggini, S. Vitamin C and Immune Function. Nutrients 2017, 9, 1211. [Google Scholar] [CrossRef] [PubMed]
  94. Spellberg, M.A. Excretion of Ascorbic Acid in Relation to Saturation and Utilization. Arch. Intern. Med. 1939, 63, 1095–1116. [Google Scholar] [CrossRef]
  95. Padayatty, S.J.; Sun, H.; Wang, Y.; Riordan, H.D.; Hewitt, S.M.; Katz, A.; Wesley, R.A.; Levine, M. Vitamin C pharmacokinetics: Implications for oral and intravenous use. Ann. Intern. Med. 2004, 140, 533–537. [Google Scholar] [CrossRef] [PubMed]
  96. Lykkesfeldt, J.; Tveden-Nyborg, P. The Pharmacokinetics of Vitamin C. Nutrients 2019, 11, 2412. [Google Scholar] [CrossRef] [PubMed]
  97. Rumsey, S.C.; Levine, M. Absorption, transport, and disposition of ascorbic acid in humans. J. Nutr. Biochem. 1998, 9, 116–130. [Google Scholar] [CrossRef]
  98. Levine, M.; Conry-Cantilena, C.; Wang, Y.; Welch, R.W.; Washko, P.W.; Dhariwal, K.R.; Park, J.B.; Lazarev, A.; Graumlich, J.F.; King, J.; et al. Vitamin C pharmacokinetics in healthy volunteers: Evidence for a recommended dietary allowance. Proc. Natl. Acad. Sci. USA 1996, 93, 3704–3709. [Google Scholar] [CrossRef] [PubMed]
  99. Chen, Q.; Espey, M.G.; Sun, A.Y.; Pooput, C.; Kirk, K.L.; Krishna, M.C.; Khosh, D.B.; Drisko, J.; Levine, M. Pharmacologic doses of ascorbate act as a prooxidant and decrease growth of aggressive tumor xenografts in mice. Proc. Natl. Acad. Sci. USA 2008, 105, 11105–11109. [Google Scholar] [CrossRef] [PubMed]
  100. Espey, M.G.; Sun, A.Y.; Drisko, J.; Krishna, M.C.; Pooput, C.; Kirk, K.; Levine, M. Ascorbic acid as a pharmacologic agent decreases tumor growth by generating hydrogen peroxide in vivo. In Proceedings of the 14th Biennial Meeting of the Society for Free Radical Research International, Beijing, China, 18–22 October 2008. [Google Scholar]
  101. Pollard, H.B.; Levine, M.A.; Eidelman, O.; Pollard, M. Pharmacological ascorbic acid suppresses syngeneic tumor growth and metastases in hormone-refractory prostate cancer. In Vivo 2010, 24, 249–255. [Google Scholar]
  102. Schoenfeld, J.D.; Sibenaller, Z.A.; Mapuskar, K.A.; Bradley, M.D.; Wagner, B.A.; Buettner, G.R.; Monga, V.; Milhem, M.; Spitz, D.R.; Allen, B.G. Redox active metals and H(2)O(2) mediate the increased efficacy of pharmacological ascorbate in combination with gemcitabine or radiation in pre-clinical sarcoma models. Redox Biol. 2018, 14, 417–422. [Google Scholar] [CrossRef]
  103. Schoenfeld, J.D.; Sibenaller, Z.A.; Mapuskar, K.A.; Wagner, B.A.; Cramer-Morales, K.L.; Furqan, M.; Sandhu, S.; Carlisle, T.L.; Smith, M.C.; Hejleh, T.A. O2•− and H2O2-mediated disruption of Fe metabolism causes the differential susceptibility of NSCLC and GBM cancer cells to pharmacological ascorbate. Cancer Cell 2017, 31, 487–500.e8. [Google Scholar] [CrossRef] [PubMed]
  104. Fromberg, A.; Gutsch, D.; Schulze, D.; Vollbracht, C.; Weiss, G.; Czubayko, F.; Aigner, A. Ascorbate exerts anti-proliferative effects through cell cycle inhibition and sensitizes tumor cells towards cytostatic drugs. Cancer Chemother. Pharmacol. 2011, 67, 1157–1166. [Google Scholar] [CrossRef] [PubMed]
  105. Chen, P.; Yu, J.; Chalmers, B.; Drisko, J.; Yang, J.; Li, B.; Chen, Q. Pharmacological ascorbate induces cytotoxicity in prostate cancer cells through ATP depletion and induction of autophagy. Anticancer Drugs 2012, 23, 437–444. [Google Scholar] [CrossRef] [PubMed]
  106. Mamede, A.C.; Pires, A.S.; Abrantes, A.M.; Tavares, S.D.; Goncalves, A.C.; Casalta-Lopes, J.E.; Sarmento-Ribeiro, A.B.; Maia, J.M.; Botelho, M.F. Cytotoxicity of ascorbic acid in a human colorectal adenocarcinoma cell line (WiDr): In vitro and in vivo studies. Nutr. Cancer 2012, 64, 1049–1057. [Google Scholar] [CrossRef]
  107. Herst, P.M.; Broadley, K.W.; Harper, J.L.; McConnell, M.J. Pharmacological concentrations of ascorbate radiosensitize glioblastoma multiforme primary cells by increasing oxidative DNA damage and inhibiting G2/M arrest. Free Radic. Biol. Med. 2012, 52, 1486–1493. [Google Scholar] [CrossRef]
  108. Ma, E.; Chen, P.; Wilkins, H.M.; Wang, T.; Swerdlow, R.H.; Chen, Q. Pharmacologic ascorbate induces neuroblastoma cell death by hydrogen peroxide mediated DNA damage and reduction in cancer cell glycolysis. Free Radic. Biol. Med. 2017, 113, 36–47. [Google Scholar] [CrossRef] [PubMed]
  109. Lu, Y.X.; Wu, Q.N.; Chen, D.L.; Chen, L.Z.; Wang, Z.X.; Ren, C.; Mo, H.Y.; Chen, Y.; Sheng, H.; Wang, Y.N.; et al. Pharmacological Ascorbate Suppresses Growth of Gastric Cancer Cells with GLUT1 Overexpression and Enhances the Efficacy of Oxaliplatin Through Redox Modulation. Theranostics 2018, 8, 1312–1326. [Google Scholar] [CrossRef] [PubMed]
  110. Verrax, J.; Calderon, P.B. Pharmacologic concentrations of ascorbate are achieved by parenteral administration and exhibit antitumoral effects. Free Radic. Biol. Med. 2009, 47, 32–40. [Google Scholar] [CrossRef]
  111. O’Leary, B.R.; Houwen, F.K.; Johnson, C.L.; Allen, B.G.; Mezhir, J.J.; Berg, D.J.; Cullen, J.J.; Spitz, D.R. Pharmacological Ascorbate as an Adjuvant for Enhancing Radiation-Chemotherapy Responses in Gastric Adenocarcinoma. Radiat. Res. 2018, 189, 456–465. [Google Scholar] [CrossRef]
  112. Du, J.; Cieslak, J.A., 3rd; Welsh, J.L.; Sibenaller, Z.A.; Allen, B.G.; Wagner, B.A.; Kalen, A.L.; Doskey, C.M.; Strother, R.K.; Button, A.M.; et al. Pharmacological Ascorbate Radiosensitizes Pancreatic Cancer. Cancer Res. 2015, 75, 3314–3326. [Google Scholar] [CrossRef]
  113. O’Leary, B.R.; Ruppenkamp, E.K.; Steers, G.J.; Du, J.; Carroll, R.S.; Wagner, B.A.; Buettner, G.R.; Cullen, J.J. Pharmacological Ascorbate Enhances Chemotherapies in Pancreatic Ductal Adenocarcinoma. Pancreas 2022, 51, 684–693. [Google Scholar] [CrossRef] [PubMed]
  114. Ma, Y.; Chen, P.; Drisko, J.A.; Khabele, D.; Godwin, A.K.; Chen, Q. Pharmacological ascorbate induces ‘BRCAness’ and enhances the effects of Poly(ADP-Ribose) polymerase inhibitors against BRCA1/2 wild-type ovarian cancer. Oncol. Lett. 2020, 19, 2629–2638. [Google Scholar] [CrossRef] [PubMed]
  115. Callaghan, C.M.; Abukhiran, I.M.; Masaadeh, A.; Van Rheeden, R.V.; Kalen, A.L.; Rodman, S.N., 3rd; Petronek, M.S.; Mapuskar, K.A.; George, B.N.; Coleman, M.C.; et al. Manipulation of Redox Metabolism Using Pharmacologic Ascorbate Opens a Therapeutic Window for Radio-Sensitization by ATM Inhibitors in Colorectal Cancer. Int. J. Radiat. Oncol. Biol. Phys. 2023, 115, 933–944. [Google Scholar] [CrossRef] [PubMed]
  116. Petronek, M.S.; Monga, V.; Bodeker, K.L.; Kwofie, M.; Lee, C.Y.; Mapuskar, K.A.; Stolwijk, J.M.; Zaher, A.; Wagner, B.A.; Smith, M.C.; et al. Magnetic Resonance Imaging of Iron Metabolism with T2* Mapping Predicts an Enhanced Clinical Response to Pharmacologic Ascorbate in Patients with GBM. Clin. Cancer Res. 2024, 30, 283–293. [Google Scholar] [CrossRef] [PubMed]
  117. Ma, Y.; Chapman, J.; Levine, M.; Polireddy, K.; Drisko, J.; Chen, Q. High-dose parenteral ascorbate enhanced chemosensitivity of ovarian cancer and reduced toxicity of chemotherapy. Sci. Transl. Med. 2014, 6, 222ra218. [Google Scholar] [CrossRef] [PubMed]
  118. Welsh, J.L.; Wagner, B.A.; van’t Erve, T.J.; Zehr, P.S.; Berg, D.J.; Halfdanarson, T.R.; Yee, N.S.; Bodeker, K.L.; Du, J.; Roberts, L.J., 2nd; et al. Pharmacological ascorbate with gemcitabine for the control of metastatic and node-positive pancreatic cancer (PACMAN): Results from a phase I clinical trial. Cancer Chemother. Pharmacol. 2013, 71, 765–775. [Google Scholar] [CrossRef]
  119. Monti, D.A.; Mitchell, E.; Bazzan, A.J.; Littman, S.; Zabrecky, G.; Yeo, C.J.; Pillai, M.V.; Newberg, A.B.; Deshmukh, S.; Levine, M. Phase I evaluation of intravenous ascorbic acid in combination with gemcitabine and erlotinib in patients with metastatic pancreatic cancer. PLoS ONE 2012, 7, e29794. [Google Scholar] [CrossRef] [PubMed]
  120. Polireddy, K.; Dong, R.; Reed, G.; Yu, J.; Chen, P.; Williamson, S.; Violet, P.C.; Pessetto, Z.; Godwin, A.K.; Fan, F.; et al. High Dose Parenteral Ascorbate Inhibited Pancreatic Cancer Growth and Metastasis: Mechanisms and a Phase I/IIa study. Sci. Rep. 2017, 7, 17188. [Google Scholar] [CrossRef]
  121. Furqan, M.; Abu-Hejleh, T.; Stephens, L.M.; Hartwig, S.M.; Mott, S.L.; Pulliam, C.F.; Petronek, M.; Henrich, J.B.; Fath, M.A.; Houtman, J.C.; et al. Pharmacological ascorbate improves the response to platinum-based chemotherapy in advanced stage non-small cell lung cancer. Redox Biol. 2022, 53, 102318. [Google Scholar] [CrossRef]
  122. Magri, A.; Germano, G.; Lorenzato, A.; Lamba, S.; Chila, R.; Montone, M.; Amodio, V.; Ceruti, T.; Sassi, F.; Arena, S.; et al. High-dose vitamin C enhances cancer immunotherapy. Sci. Transl. Med. 2020, 12, eaay8707. [Google Scholar] [CrossRef]
  123. Luchtel, R.A.; Bhagat, T.; Pradhan, K.; Jacobs, W.R., Jr.; Levine, M.; Verma, A.; Shenoy, N. High-dose ascorbic acid synergizes with anti-PD1 in a lymphoma mouse model. Proc. Natl. Acad. Sci. USA 2020, 117, 1666–1677. [Google Scholar] [CrossRef] [PubMed]
  124. van den Boogaard, W.M.C.; Komninos, D.S.J.; Vermeij, W.P. Chemotherapy Side-Effects: Not All DNA Damage Is Equal. Cancers 2022, 14, 627. [Google Scholar] [CrossRef] [PubMed]
  125. De Ruysscher, D.; Niedermann, G.; Burnet, N.G.; Siva, S.; Lee, A.W.M.; Hegi-Johnson, F. Radiotherapy toxicity. Nat. Rev. Dis. Primers 2019, 5, 13. [Google Scholar] [CrossRef] [PubMed]
  126. Kanter, M.; Akpolat, M. Vitamin C protects against ionizing radiation damage to goblet cells of the ileum in rats. Acta Histochem. 2008, 110, 481–490. [Google Scholar] [CrossRef] [PubMed]
  127. Faridi, U.; Alatawi, F.; Mostafa, M. Protective role of tocopherol and ascorbic acid in taxol-treated human erythrocytes in vitro. Toxicol. Res. Appl. 2017, 1, 2397847317705813. [Google Scholar] [CrossRef]
  128. Liu, Q.; Shi, Y.; Chong, Y.; Ge, C. Pharmacological Ascorbate Promotes the Tumor Radiosensitization of Au@Pd Nanoparticles with Simultaneous Protection of Normal Tissues. ACS Appl. Bio Mater. 2021, 4, 1843–1851. [Google Scholar] [CrossRef]
  129. Alexander, M.S.; Wilkes, J.G.; Schroeder, S.R.; Buettner, G.R.; Wagner, B.A.; Du, J.; Gibson-Corley, K.; O’Leary, B.R.; Spitz, D.R.; Buatti, J.M.; et al. Pharmacologic Ascorbate Reduces Radiation-Induced Normal Tissue Toxicity and Enhances Tumor Radiosensitization in Pancreatic Cancer. Cancer Res. 2018, 78, 6838–6851. [Google Scholar] [CrossRef] [PubMed]
  130. Chen, G.Y.; O’Leary, B.R.; Du, J.; Carroll, R.S.; Steers, G.J.; Buettner, G.R.; Cullen, J.J. Pharmacologic Ascorbate Radiosensitizes Pancreatic Cancer but Radioprotects Normal Tissue: The Role of Oxidative Stress-Induced Lipid Peroxidation. Antioxidants 2024, 13, 361. [Google Scholar] [CrossRef] [PubMed]
  131. Wang, F.; He, M.M.; Xiao, J.; Zhang, Y.Q.; Yuan, X.L.; Fang, W.J.; Zhang, Y.; Wang, W.; Hu, X.H.; Ma, Z.G.; et al. A Randomized, Open-Label, Multicenter, Phase 3 Study of High-Dose Vitamin C Plus FOLFOX ± Bevacizumab versus FOLFOX ± Bevacizumab in Unresectable Untreated Metastatic Colorectal Cancer (VITALITY Study). Clin. Cancer Res. 2022, 28, 4232–4239. [Google Scholar] [CrossRef]
  132. Nielsen, T.K.; Hojgaard, M.; Andersen, J.T.; Jorgensen, N.R.; Zerahn, B.; Kristensen, B.; Henriksen, T.; Lykkesfeldt, J.; Mikines, K.J.; Poulsen, H.E. Weekly ascorbic acid infusion in castration-resistant prostate cancer patients: A single-arm phase II trial. Transl. Androl. Urol. 2017, 6, 517–528. [Google Scholar] [CrossRef]
  133. Paller, C.J.; Zahurak, M.L.; Mandl, A.; Metri, N.A.; Lalji, A.; Heath, E.; Kelly, W.K.; Hoimes, C.; Barata, P.; Taksey, J.; et al. High-Dose Intravenous Vitamin C Combined with Docetaxel in Men with Metastatic Castration-Resistant Prostate Cancer: A Randomized Placebo-Controlled Phase 2 Trial. Cancer Res. Commun. 2024. [Google Scholar] [CrossRef]
  134. Cushing, C.M.; Petronek, M.S.; Bodeker, K.L.; Vollstedt, S.; Brown, H.A.; Opat, E.; Hollenbeck, N.J.; Shanks, T.; Berg, D.J.; Smith, B.J.; et al. Magnetic resonance imaging (MRI) of pharmacological ascorbate-induced iron redox state as a biomarker in subjects undergoing radio-chemotherapy. Redox Biol. 2021, 38, 101804. [Google Scholar] [CrossRef] [PubMed]
  135. Petronek, M.S.; Teferi, N.; Caster, J.M.; Stolwijk, J.M.; Zaher, A.; Buatti, J.M.; Hasan, D.; Wafa, E.I.; Salem, A.K.; Gillan, E.G.; et al. Magnetite nanoparticles as a kinetically favorable source of iron to enhance GBM response to chemoradiosensitization with pharmacological ascorbate. Redox Biol. 2023, 62, 102651. [Google Scholar] [CrossRef]
  136. Zaher, A.; Mapuskar, K.A.; Sarkaria, J.N.; Spitz, D.R.; Petronek, M.S.; Allen, B.G. Differential H2O2 Metabolism among Glioblastoma Subtypes Confers Variable Responses to Pharmacological Ascorbate Therapy Combined with Chemoradiation. Int. J. Mol. Sci. 2023, 24, 17158. [Google Scholar] [CrossRef]
  137. Fridovich, I. Superoxide dismutases. Annu. Rev. Biochem. 1975, 44, 147–159. [Google Scholar] [CrossRef] [PubMed]
  138. Marklund, S.L.; Holme, E.; Hellner, L. Superoxide dismutase in extracellular fluids. Clin. Chim. Acta 1982, 126, 41–51. [Google Scholar] [CrossRef]
  139. Batinic-Haberle, I.; Benov, L.; Spasojevic, I.; Fridovich, I. The ortho effect makes manganese(III) meso-tetrakis(N-methylpyridinium-2-yl)porphyrin a powerful and potentially useful superoxide dismutase mimic. J. Biol. Chem. 1998, 273, 24521–24528. [Google Scholar] [CrossRef]
  140. Policar, C.; Bouvet, J.; Bertrand, H.C.; Delsuc, N. SOD mimics: From the tool box of the chemists to cellular studies. Curr. Opin. Chem. Biol. 2022, 67, 102109. [Google Scholar] [CrossRef]
  141. Borgstahl, G.E.O.; Oberley-Deegan, R.E. Superoxide Dismutases (SODs) and SOD Mimetics. Antioxidants 2018, 7, 156. [Google Scholar] [CrossRef]
  142. Chidambaram, S.B.; Anand, N.; Varma, S.R.; Ramamurthy, S.; Vichitra, C.; Sharma, A.; Mahalakshmi, A.M.; Essa, M.M. Superoxide dismutase and neurological disorders. IBRO Neurosci. Rep. 2024, 16, 373–394. [Google Scholar] [CrossRef] [PubMed]
  143. Miriyala, S.; Spasojevic, I.; Tovmasyan, A.; Salvemini, D.; Vujaskovic, Z.; St Clair, D.; Batinic-Haberle, I. Manganese superoxide dismutase, MnSOD and its mimics. Biochim. Biophys. Acta 2012, 1822, 794–814. [Google Scholar] [CrossRef] [PubMed]
  144. Friedel, F.C.; Lieb, D.; Ivanovic-Burmazovic, I. Comparative studies on manganese-based SOD mimetics, including the phosphate effect, by using global spectral analysis. J. Inorg. Biochem. 2012, 109, 26–32. [Google Scholar] [CrossRef] [PubMed]
  145. Riley, D.P. Functional mimics of superoxide dismutase enzymes as therapeutic agents. Chem. Rev. 1999, 99, 2573–2588. [Google Scholar] [CrossRef]
  146. Raber, J.; Davis, M.J.; Pfankuch, T.; Rosenthal, R.; Doctrow, S.R.; Moulder, J.E. Mitigating effect of EUK-207 on radiation-induced cognitive impairments. Behav. Brain Res. 2017, 320, 457–463. [Google Scholar] [CrossRef] [PubMed]
  147. Langan, A.R.; Khan, M.A.; Yeung, I.W.; Van Dyk, J.; Hill, R.P. Partial volume rat lung irradiation: The protective/mitigating effects of Eukarion-189, a superoxide dismutase-catalase mimetic. Radiother. Oncol. 2006, 79, 231–238. [Google Scholar] [CrossRef] [PubMed]
  148. Mahmood, J.; Jelveh, S.; Calveley, V.; Zaidi, A.; Doctrow, S.R.; Hill, R.P. Mitigation of radiation-induced lung injury by genistein and EUK-207. Int. J. Radiat. Biol. 2011, 87, 889–901. [Google Scholar] [CrossRef]
  149. Mahmood, J.; Jelveh, S.; Zaidi, A.; Doctrow, S.R.; Hill, R.P. Mitigation of radiation-induced lung injury with EUK-207 and genistein: Effects in adolescent rats. Radiat. Res. 2013, 179, 125–134. [Google Scholar] [CrossRef] [PubMed]
  150. Gao, F.; Fish, B.L.; Szabo, A.; Doctrow, S.R.; Kma, L.; Molthen, R.C.; Moulder, J.E.; Jacobs, E.R.; Medhora, M. Short-term treatment with a SOD/catalase mimetic, EUK-207, mitigates pneumonitis and fibrosis after single-dose total-body or whole-thoracic irradiation. Radiat. Res. 2012, 178, 468–480. [Google Scholar] [CrossRef] [PubMed]
  151. Doctrow, S.R.; Lopez, A.; Schock, A.M.; Duncan, N.E.; Jourdan, M.M.; Olasz, E.B.; Moulder, J.E.; Fish, B.L.; Mader, M.; Lazar, J.; et al. A synthetic superoxide dismutase/catalase mimetic EUK-207 mitigates radiation dermatitis and promotes wound healing in irradiated rat skin. J. Investig. Dermatol. 2013, 133, 1088–1096. [Google Scholar] [CrossRef]
  152. Vujaskovic, Z.; Batinic-Haberle, I.; Rabbani, Z.N.; Feng, Q.F.; Kang, S.K.; Spasojevic, I.; Samulski, T.V.; Fridovich, I.; Dewhirst, M.W.; Anscher, M.S. A small molecular weight catalytic metalloporphyrin antioxidant with superoxide dismutase (SOD) mimetic properties protects lungs from radiation-induced injury. Free Radic. Biol. Med. 2002, 33, 857–863. [Google Scholar] [CrossRef]
  153. Gauter-Fleckenstein, B.; Fleckenstein, K.; Owzar, K.; Jiang, C.; Batinic-Haberle, I.; Vujaskovic, Z. Comparison of two Mn porphyrin-based mimics of superoxide dismutase in pulmonary radioprotection. Free Radic. Biol. Med. 2008, 44, 982–989. [Google Scholar] [CrossRef]
  154. Spasojevic, I.; Chen, Y.; Noel, T.J.; Fan, P.; Zhang, L.; Reboucas, J.S.; St Clair, D.K.; Batinic-Haberle, I. Pharmacokinetics of the potent redox-modulating manganese porphyrin, MnTE-2-PyP(5+), in plasma and major organs of B6C3F1 mice. Free Radic. Biol. Med. 2008, 45, 943–949. [Google Scholar] [CrossRef]
  155. Gauter-Fleckenstein, B.; Fleckenstein, K.; Owzar, K.; Jiang, C.; Reboucas, J.S.; Batinic-Haberle, I.; Vujaskovic, Z. Early and late administration of MnTE-2-PyP5+ in mitigation and treatment of radiation-induced lung damage. Free Radic. Biol. Med. 2010, 48, 1034–1043. [Google Scholar] [CrossRef]
  156. Archambeau, J.O.; Tovmasyan, A.; Pearlstein, R.D.; Crapo, J.D.; Batinic-Haberle, I. Superoxide dismutase mimic, MnTE-2-PyP(5+) ameliorates acute and chronic proctitis following focal proton irradiation of the rat rectum. Redox Biol. 2013, 1, 599–607. [Google Scholar] [CrossRef]
  157. Gauter-Fleckenstein, B.; Reboucas, J.S.; Fleckenstein, K.; Tovmasyan, A.; Owzar, K.; Jiang, C.; Batinic-Haberle, I.; Vujaskovic, Z. Robust rat pulmonary radioprotection by a lipophilic Mn N-alkylpyridylporphyrin, MnTnHex-2-PyP(5+). Redox Biol. 2014, 2, 400–410. [Google Scholar] [CrossRef] [PubMed]
  158. Shrishrimal, S.; Kosmacek, E.A.; Chatterjee, A.; Tyson, M.J.; Oberley-Deegan, R.E. The SOD Mimic, MnTE-2-PyP, Protects from Chronic Fibrosis and Inflammation in Irradiated Normal Pelvic Tissues. Antioxidants 2017, 6, 87. [Google Scholar] [CrossRef] [PubMed]
  159. Tovmasyan, A.; Sheng, H.; Weitner, T.; Arulpragasam, A.; Lu, M.; Warner, D.S.; Vujaskovic, Z.; Spasojevic, I.; Batinic-Haberle, I. Design, mechanism of action, bioavailability and therapeutic effects of mn porphyrin-based redox modulators. Med. Princ. Pract. 2013, 22, 103–130. [Google Scholar] [CrossRef]
  160. Weitner, T.; Kos, I.; Sheng, H.; Tovmasyan, A.; Reboucas, J.S.; Fan, P.; Warner, D.S.; Vujaskovic, Z.; Batinic-Haberle, I.; Spasojevic, I. Comprehensive pharmacokinetic studies and oral bioavailability of two Mn porphyrin-based SOD mimics, MnTE-2-PyP5+ and MnTnHex-2-PyP5+. Free Radic. Biol. Med. 2013, 58, 73–80. [Google Scholar] [CrossRef]
  161. Leu, D.; Spasojevic, I.; Nguyen, H.; Deng, B.; Tovmasyan, A.; Weitner, T.; Sampaio, R.S.; Batinic-Haberle, I.; Huang, T.-T. CNS bioavailability and radiation protection of normal hippocampal neurogenesis by a lipophilic Mn porphyrin-based superoxide dismutase mimic, MnTnBuOE-2-PyP5+. Redox Biol. 2017, 12, 864–871. [Google Scholar] [CrossRef] [PubMed]
  162. Weitzel, D.H.; Tovmasyan, A.; Ashcraft, K.A.; Rajic, Z.; Weitner, T.; Liu, C.; Li, W.; Buckley, A.F.; Prasad, M.R.; Young, K.H.; et al. Radioprotection of the brain white matter by Mn(III) n-Butoxyethylpyridylporphyrin-based superoxide dismutase mimic MnTnBuOE-2-PyP5+. Mol. Cancer Ther. 2015, 14, 70–79. [Google Scholar] [CrossRef]
  163. McElroy, T.; Brown, T.; Kiffer, F.; Wang, J.; Byrum, S.D.; Oberley-Deegan, R.E.; Allen, A.R. Assessing the Effects of Redox Modifier MnTnBuOE-2-PyP 5+ on Cognition and Hippocampal Physiology Following Doxorubicin, Cyclophosphamide, and Paclitaxel Treatment. Int. J. Mol. Sci. 2020, 21, 1867. [Google Scholar] [CrossRef] [PubMed]
  164. Yulyana, Y.; Tovmasyan, A.; Ho, I.A.; Sia, K.C.; Newman, J.P.; Ng, W.H.; Guo, C.M.; Hui, K.M.; Batinic-Haberle, I.; Lam, P.Y. Redox-Active Mn Porphyrin-based Potent SOD Mimic, MnTnBuOE-2-PyP(5+), Enhances Carbenoxolone-Mediated TRAIL-Induced Apoptosis in Glioblastoma Multiforme. Stem Cell Rev. Rep. 2016, 12, 140–155. [Google Scholar] [CrossRef]
  165. Ashcraft, K.A.; Boss, M.K.; Tovmasyan, A.; Roy Choudhury, K.; Fontanella, A.N.; Young, K.H.; Palmer, G.M.; Birer, S.R.; Landon, C.D.; Park, W.; et al. Novel Manganese-Porphyrin Superoxide Dismutase-Mimetic Widens the Therapeutic Margin in a Preclinical Head and Neck Cancer Model. Int. J. Radiat. Oncol. Biol. Phys. 2015, 93, 892–900. [Google Scholar] [CrossRef]
  166. Birer, S.R.; Lee, C.T.; Choudhury, K.R.; Young, K.H.; Spasojevic, I.; Batinic-Haberle, I.; Crapo, J.D.; Dewhirst, M.W.; Ashcraft, K.A. Inhibition of the Continuum of Radiation-Induced Normal Tissue Injury by a Redox-Active Mn Porphyrin. Radiat. Res. 2017, 188, 94–104. [Google Scholar] [CrossRef] [PubMed]
  167. Shin, S.W.; Choi, C.; Lee, G.H.; Son, A.; Kim, S.H.; Park, H.C.; Batinic-Haberle, I.; Park, W. Mechanism of the Antitumor and Radiosensitizing Effects of a Manganese Porphyrin, MnHex-2-PyP. Antioxid. Redox Signal. 2017, 27, 1067–1082. [Google Scholar] [CrossRef]
  168. Chaiswing, L.; Yarana, C.; St Clair, W.; Tovmasyan, A.; Batinic-Haberle, I.; Spasojevic, I.; St Clair, D. A Redox-active Mn Porphyrin, MnTnBuOE-2-PyP(5+), Synergizes with Carboplatin in Treatment of Chemoresistant Ovarian Cell Line. Oxidative Med. Cell. Longev. 2022, 2022, 9664636. [Google Scholar] [CrossRef]
  169. Soares, R.B.; Manguinhas, R.; Costa, J.G.; Saraiva, N.; Gil, N.; Rosell, R.; Camões, S.P.; Batinic-Haberle, I.; Spasojevic, I.; Castro, M.; et al. The Redox-Active Manganese(III) Porphyrin, MnTnBuOE-2-PyP5+, Impairs the Migration and Invasion of Non-Small Cell Lung Cancer Cells, Either Alone or Combined with Cisplatin. Cancers 2023, 15, 3814. [Google Scholar] [CrossRef] [PubMed]
  170. Aston, K.; Rath, N.; Naik, A.; Slomczynska, U.; Schall, O.F.; Riley, D.P. Computer-aided design (CAD) of Mn(II) complexes: Superoxide dismutase mimetics with catalytic activity exceeding the native enzyme. Inorg. Chem. 2001, 40, 1779–1789. [Google Scholar] [CrossRef]
  171. Glenn, J.K.; Akileswaran, L.; Gold, M.H. Mn(II) oxidation is the principal function of the extracellular Mn-peroxidase from Phanerochaete chrysosporium. Arch. Biochem. Biophys. 1986, 251, 688–696. [Google Scholar] [CrossRef]
  172. Stadtman, E.R.; Berlett, B.S.; Chock, P.B. Manganese-dependent disproportionation of hydrogen peroxide in bicarbonate buffer. Proc. Natl. Acad. Sci. USA 1990, 87, 384–388. [Google Scholar] [CrossRef]
  173. Ferrer-Sueta, G.; Vitturi, D.; Batinic-Haberle, I.; Fridovich, I.; Goldstein, S.; Czapski, G.; Radi, R. Reactions of manganese porphyrins with peroxynitrite and carbonate radical anion. J. Biol. Chem. 2003, 278, 27432–27438. [Google Scholar] [CrossRef]
  174. Sharpe, M.A.; Ollosson, R.; Stewart, V.C.; Clark, J.B. Oxidation of nitric oxide by oxomanganese-salen complexes: A new mechanism for cellular protection by superoxide dismutase/catalase mimetics. Biochem. J. 2002, 366 Pt 1, 97–107. [Google Scholar] [CrossRef]
  175. Doctrow, S.R.; Huffman, K.; Marcus, C.B.; Tocco, G.; Malfroy, E.; Adinolfi, C.A.; Kruk, H.; Baker, K.; Lazarowych, N.; Mascarenhas, J.; et al. Salen-manganese complexes as catalytic scavengers of hydrogen peroxide and cytoprotective agents: Structure-activity relationship studies. J. Med. Chem. 2002, 45, 4549–4558. [Google Scholar] [CrossRef]
  176. Murphy, C.K.; Fey, E.G.; Watkins, B.A.; Wong, V.; Rothstein, D.; Sonis, S.T. Efficacy of superoxide dismutase mimetic M40403 in attenuating radiation-induced oral mucositis in hamsters. Clin. Cancer Res. 2008, 14, 4292–4297. [Google Scholar] [CrossRef] [PubMed]
  177. Thompson, J.S.; Chu, Y.; Glass, J.; Tapp, A.A.; Brown, S.A. The manganese superoxide dismutase mimetic, M40403, protects adult mice from lethal total body irradiation. Free Radic. Res. 2010, 44, 529–540. [Google Scholar] [CrossRef]
  178. Mapuskar, K.A.; Flippo, K.H.; Schoenfeld, J.D.; Riley, D.P.; Strack, S.; Hejleh, T.A.; Furqan, M.; Monga, V.; Domann, F.E.; Buatti, J.M.; et al. Mitochondrial Superoxide Increases Age-Associated Susceptibility of Human Dermal Fibroblasts to Radiation and Chemotherapy. Cancer Res. 2017, 77, 5054–5067. [Google Scholar] [CrossRef]
  179. Anderson, C.M.; Lee, C.M.; Kelley, J.R.; Walker, G.V.; Dunlap, N.E.; Bar-Ad, V.C.; Miller, D.A.; King, V.J.; Peddada, A.V.; Ciuba, D.F.; et al. ROMAN: Phase 3 trial of avasopasem manganese (GC4419) for severe oral mucositis (SOM) in patients receiving chemoradiotherapy (CRT) for locally advanced, nonmetastatic head and neck cancer (LAHNC). J. Clin. Oncol. 2022, 40 (Suppl. S16), 6005. [Google Scholar] [CrossRef]
  180. Anderson, C.M.; Lee, C.M.; Saunders, D.P.; Curtis, A.; Dunlap, N.; Nangia, C.; Lee, A.S.; Gordon, S.M.; Kovoor, P.; Arevalo-Araujo, R.; et al. Phase IIb, Randomized, Double-Blind Trial of GC4419 Versus Placebo to Reduce Severe Oral Mucositis Due to Concurrent Radiotherapy and Cisplatin For Head and Neck Cancer. J. Clin. Oncol. 2019, 37, 3256–3265. [Google Scholar] [CrossRef] [PubMed]
  181. Anderson, C.M.; Sonis, S.T.; Lee, C.M.; Adkins, D.; Allen, B.G.; Sun, W.; Agarwala, S.S.; Venigalla, M.L.; Chen, Y.; Zhen, W.; et al. Phase 1b/2a Trial of the Superoxide Dismutase Mimetic GC4419 to Reduce Chemoradiotherapy-Induced Oral Mucositis in Patients With Oral Cavity or Oropharyngeal Carcinoma. Int. J. Radiat. Oncol. Biol. Phys. 2018, 100, 427–435. [Google Scholar] [CrossRef] [PubMed]
  182. Mapuskar, K.A.; Vasquez Martinez, G.; Pulliam, C.F.; Petronek, M.S.; Steinbach, E.J.; Monga, V.; Furqan, M.; Jetton, J.G.; Saunders, D.P.; Pearce, A.; et al. Avasopasem manganese (GC4419) protects against cisplatin-induced chronic kidney disease: An exploratory analysis of renal metrics from a randomized phase 2b clinical trial in head and neck cancer patients. Redox Biol. 2023, 60, 102599. [Google Scholar] [CrossRef]
  183. Mapuskar, K.A.; Pulliam, C.F.; Tomanek-Chalkley, A.; Rastogi, P.; Wen, H.; Dayal, S.; Griffin, B.R.; Zepeda-Orozco, D.; Sindler, A.L.; Anderson, C.M.; et al. The antioxidant and anti-inflammatory activities of avasopasem manganese in age-associated, cisplatin-induced renal injury. Redox Biol. 2024, 70, 103022. [Google Scholar] [CrossRef] [PubMed]
  184. El-Mahdy, M.A.; Alzarie, Y.A.; Hemann, C.; Badary, O.A.; Nofal, S.; Zweier, J.L. The novel SOD mimetic GC4419 increases cancer cell killing with sensitization to ionizing radiation while protecting normal cells. Free Radic. Biol. Med. 2020, 160, 630–642. [Google Scholar] [CrossRef]
  185. Sishc, B.J.; Ding, L.; Nam, T.K.; Heer, C.D.; Rodman, S.N.; Schoenfeld, J.D.; Fath, M.A.; Saha, D.; Pulliam, C.F.; Langen, B.; et al. Avasopasem manganese synergizes with hypofractionated radiation to ablate tumors through the generation of hydrogen peroxide. Sci. Transl. Med. 2021, 13, eabb3768. [Google Scholar] [CrossRef]
  186. Zaher, A.; Mapuskar, K.A.; Petronek, M.S.; Tanas, M.R.; Isaacson, A.L.; Dodd, R.D.; Milhem, M.; Furqan, M.; Spitz, D.R.; Miller, B.J.; et al. Superoxide Dismutase Mimetic Avasopasem Manganese Enhances Radiation Therapy Effectiveness in Soft Tissue Sarcomas and Accelerates Wound Healing. Antioxidants 2024, 13, 587. [Google Scholar] [CrossRef] [PubMed]
  187. Shah, M.H.; Liu, G.S.; Thompson, E.W.; Dusting, G.J.; Peshavariya, H.M. Differential effects of superoxide dismutase and superoxide dismutase/catalase mimetics on human breast cancer cells. Breast Cancer Res. Treat. 2015, 150, 523–534. [Google Scholar] [CrossRef]
  188. Heer, C.D.; Davis, A.B.; Riffe, D.B.; Wagner, B.A.; Falls, K.C.; Allen, B.G.; Buettner, G.R.; Beardsley, R.A.; Riley, D.P.; Spitz, D.R. Superoxide Dismutase Mimetic GC4419 Enhances the Oxidation of Pharmacological Ascorbate and Its Anticancer Effects in an H(2)O(2)-Dependent Manner. Antioxidants 2018, 7, 18. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The sliding scale of the concentration-dependent effects of intracellular H2O2 on cancer cells. (A) The tumor-promoting region of H2O2 in cancer and its associated effects. (B) The tumor-killing region of H2O2 in cancer and its related effects.
Figure 1. The sliding scale of the concentration-dependent effects of intracellular H2O2 on cancer cells. (A) The tumor-promoting region of H2O2 in cancer and its associated effects. (B) The tumor-killing region of H2O2 in cancer and its related effects.
Ijms 25 08885 g001
Table 1. Summary of some discussed mechanisms by which H2O2 may induce cellular damage.
Table 1. Summary of some discussed mechanisms by which H2O2 may induce cellular damage.
Damage MechanismOutcomeReference
Mitochondrial DysfunctionDecreased ATP synthesis, impaired mitochondrial function. [9]
Fenton ChemistryDNA, lipid, and protein damage. [10]
Protein ModificationIrreversible protein oxidation and fragmentation, mitochondrial dysfunction, hyperactivation or inactivation of essential proteins (e.g., kinases). [11,12,13]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Zaher, A.; Petronek, M.S.; Allen, B.G.; Mapuskar, K.A. Balanced Duality: H2O2-Based Therapy in Cancer and Its Protective Effects on Non-Malignant Tissues. Int. J. Mol. Sci. 2024, 25, 8885. https://doi.org/10.3390/ijms25168885

AMA Style

Zaher A, Petronek MS, Allen BG, Mapuskar KA. Balanced Duality: H2O2-Based Therapy in Cancer and Its Protective Effects on Non-Malignant Tissues. International Journal of Molecular Sciences. 2024; 25(16):8885. https://doi.org/10.3390/ijms25168885

Chicago/Turabian Style

Zaher, Amira, Michael S. Petronek, Bryan G. Allen, and Kranti A. Mapuskar. 2024. "Balanced Duality: H2O2-Based Therapy in Cancer and Its Protective Effects on Non-Malignant Tissues" International Journal of Molecular Sciences 25, no. 16: 8885. https://doi.org/10.3390/ijms25168885

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop