Next Article in Journal
Senescence Rejuvenation through Reduction in Mitochondrial Reactive Oxygen Species Generation by Polygonum cuspidatum Extract: In Vitro Evidence
Previous Article in Journal
Cookies Fortified with Polyphenols Extracts: Impact on Phenolic Content, Antioxidant Activity, Inhibition of α-Amylase and α-Glucosidase Enzyme, Colour and Sensory Attractiveness
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Oxidative Stress and Age-Related Tumors

by
Emma Di Carlo
1,2,* and
Carlo Sorrentino
1,2
1
Department of Medicine and Sciences of Aging, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
2
Anatomic Pathology and Immuno-Oncology Unit, Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
*
Author to whom correspondence should be addressed.
Antioxidants 2024, 13(9), 1109; https://doi.org/10.3390/antiox13091109
Submission received: 19 July 2024 / Revised: 6 September 2024 / Accepted: 10 September 2024 / Published: 13 September 2024
(This article belongs to the Special Issue Reactive Nitrogen Species (RNS) and Redox Signaling in Tumors)

Abstract

:
Oxidative stress is the result of the imbalance between reactive oxygen and nitrogen species (RONS), which are produced by several endogenous and exogenous processes, and antioxidant defenses consisting of exogenous and endogenous molecules that protect biological systems from free radical toxicity. Oxidative stress is a major factor in the aging process, contributing to the accumulation of cellular damage over time. Oxidative damage to cellular biomolecules, leads to DNA alterations, lipid peroxidation, protein oxidation, and mitochondrial dysfunction resulting in cellular senescence, immune system and tissue dysfunctions, and increased susceptibility to age-related pathologies, such as inflammatory disorders, cardiovascular and neurodegenerative diseases, diabetes, and cancer. Oxidative stress-driven DNA damage and mutations, or methylation and histone modification, which alter gene expression, are key determinants of tumor initiation, angiogenesis, metastasis, and therapy resistance. Accumulation of genetic and epigenetic damage, to which oxidative stress contributes, eventually leads to unrestrained cell proliferation, the inhibition of cell differentiation, and the evasion of cell death, providing favorable conditions for tumorigenesis. Colorectal, breast, lung, prostate, and skin cancers are the most frequent aging-associated malignancies, and oxidative stress is implicated in their pathogenesis and biological behavior. Our aim is to shed light on the molecular and cellular mechanisms that link oxidative stress, aging, and cancers, highlighting the impact of both RONS and antioxidants, provided by diet and exercise, on cellular senescence, immunity, and development of an antitumor response. The dual role of ROS as physiological regulators of cell signaling responsible for cell damage and diseases, as well as its use for anti-tumor therapeutic purposes, will also be discussed. Managing oxidative stress is crucial for promoting healthy aging and reducing the risk of age-related tumors.

1. Introduction

Oxidative stress is implicated in numerous physiological processes, including cell signaling, immune response, and the regulation of gene expression [1]. In normal conditions, the production of reactive oxygen species (ROS), such as the hydroxyl radical (OH), the superoxide anion (O2•−), hydrogen peroxide (H2O2), singlet oxygen (1O2), and reactive nitrogen species (RNS, generated when nitric oxide interacts with reactive oxygen species) [2], such as nitric oxide (NO), and peroxynitrite (ONOO), normally generated as by-products of the oxygen and nitrogen metabolism, is balanced by the presence of antioxidants, such as superoxide dismutase (SOD), glutathione, glutathione peroxidase (GPx), catalase, and thioredoxin, which neutralize reactive species and maintain cellular homeostasis [3]. Maintaining redox homeostasis is critical for cellular function and adaptation to environmental stressors. Moderate levels of RONS, namely oxidative eustress, are critical to maintaining, modulating, and regulating cellular functions through reversible interactions between RONS and the components of cellular signaling pathways that control those functions [4]. Excessive or uncontrolled oxidative stress, namely oxidative distress, causes cellular damage and dysfunction, contributing to the pathogenesis of age-related diseases [5]. Oxidative and nitrosative stresses are intimately linked to the aging process since the accumulation of damage to macromolecules such as DNA, RNA, proteins, and lipids contributes to cellular senescence and increased susceptibility to age-related pathologies, including neurodegenerative and cardiovascular diseases, diabetes, inflammatory conditions, and cancer [6,7]. The progressive increase in blood levels of factors secreted by senescent cells, known as the senescence-associated secretory phenotype (SASP), promotes chronic inflammation and low-grade chronic damage, which accelerate the senescence of immune cells, resulting in a weakened immune function and an inability to clear senescent cells and inflammatory factors, which creates a vicious cycle of inflammation and senescence, namely “inflammaging” [8]. The mechanisms underlying the two-way links of oxidative distress with inflammation and aging, which impact tumorigenesis and the response to anticancer therapies, will be explored below.

2. Antioxidant and Oxidative Stress Driving Genes

Several genes are implicated in promoting oxidative stress, either by enhancing the production of ROS or by impairing antioxidant defenses [3,9]. Antioxidant coding genes are crucial in protecting cells from damage caused by oxidative stress, which is the result of an imbalance between ROS and the body’s ability to detoxify them [10]. These genes encode proteins that function as antioxidants, repair enzymes, and regulators of redox signaling pathways.
Key genes involved in oxidative stress and their functions are outlined below.
NADPH oxidases (NOX). NADPH oxidases are a family of enzymes, including NOX1, NOX2, NOX3, NOX4, and NOX5, that generate ROS as part of the innate immune response and various signaling pathways [11,12]. NOX enzymes transfer electrons from NADPH to molecular oxygen, producing superoxide radicals (O2•−) and other ROS. The dysregulation of NOX activity is implicated in oxidative stress-related diseases.
Nitric Oxide Synthases (NOS). Enzymes such as NOS1, NOS2 (iNOS), and NOS3 (eNOS) produce NO, which has antioxidant properties and can scavenge ROS. Under pathological conditions, such as in the presence of high levels of superoxide, NO can react with superoxide to form ONOO, a highly reactive and damaging species [13]. This reaction reduces the availability of NO, leading to endothelial dysfunction and contributing to oxidative stress [14]. Factors leading to this uncoupling include a deficiency in essential cofactors like tetrahydrobiopterin (BH4) or substrates like L-arginine. Uncoupled NOS contributes to a vicious cycle of increased ROS production and further oxidative stress.
Arachidonate Lipoxygenases (ALOX). The ALOX family includes several isoforms, such as 5-LOX, 12-LOX, and 15-LOX. Each isoform catalyzes the oxidation of specific positions on polyunsaturated fatty acids to produce lipid peroxides, leading to different biological effects. During the metabolism of fatty acids, ALOX enzymes, such as ALOX5, generate lipid hydroperoxides, which contribute to oxidative stress, inflammation, and cancer [15].
Cytochrome P450 Enzymes (CYP). The major families involved in drug metabolism are CYP1, CYP2, and CYP3. CYP enzymes are primarily located in the liver and are involved in the metabolism of xenobiotics and endogenous compounds such as steroid hormones, fatty acids, and bile acids. Some CYP enzymes, such as CYP2E1, generate ROS as by-products of their catalytic activity, contributing to oxidative stress [16,17].
Xanthine Dehydrogenase (XDH)/Xanthine Oxidoreductase (XOR). XDH and XOR are enzymes that play critical roles in purine metabolism. They catalyze the oxidation of hypoxanthine into xanthine and subsequently of xanthine into uric acid. Under certain conditions, such as oxidative stress or proteolytic cleavage, XDH can be converted into xanthine oxidase (XO), which produces superoxide and hydrogen peroxide during purine degradation, contributing to oxidative stress [18].
Hypoxia-Inducible Factor 1 Alpha (HIF1A). HIF1A is a key transcription factor that responds to changes in cellular oxygen levels. Under normoxic conditions, HIF1A is hydroxylated by prolyl hydroxylase domain enzymes (PHDs), marking it for ubiquitination and proteasomal degradation [19]. Under hypoxic conditions, the hydroxylation is inhibited, stabilizing HIF1A and allowing it to translocate into the nucleus where it dimerizes with HIF1B and activates target genes involved in glycolysis and angiogenesis. It can also promote ROS production, contributing to oxidative stress [20].
These genes are deeply involved in the molecular mechanisms driving oxidative stress, which is implicated in age-related diseases, including chronic inflammatory conditions and cancer.
Key genes driving antioxidant activity, and their functions are outlined below.
SOD. SOD enzymes catalyze the dismutation of O2•− into oxygen (O2) and hydrogen peroxide (H2O2), thereby protecting cells from superoxide-mediated oxidative damage. There are several isoforms of SOD, including cytosolic SOD1 (Cu/Zn-SOD), mitochondrial SOD2 (Mn-SOD), and extracellular SOD3 (Ec-SOD). Mutations in SOD1 are associated with familial amyotrophic lateral sclerosis (ALS) [21,22]. Mutant SOD1 proteins can misfold and aggregate, leading to mitochondrial dysfunction and increased ROS production [23].
Catalase (CAT). Catalase is an enzyme that catalyzes the decomposition of hydrogen peroxide (H2O2) into water (H2O) and O2, thereby neutralizing H2O2 and preventing its harmful effects. Catalase is primarily localized within peroxisomes and plays a critical role in cellular antioxidant defense mechanisms [24].
GPX. Glutathione peroxidases are a family of enzymes that catalyze the reduction of hydroperoxides, including hydrogen peroxide (H2O2), using reduced glutathione (GSH) as a cofactor. GPX enzymes protect cells from oxidative damage by scavenging peroxides and maintaining cellular redox balance [25].
Glutathione reductase (GSR): Glutathione reductase is an enzyme that catalyzes the reduction of oxidized glutathione (GSSG) to its reduced form (GSH), which serves as a major cellular antioxidant. GSR plays a crucial role in recycling GSH and maintaining cellular redox homeostasis [26].
Nuclear factor erythroid 2-related factor 2 (Nrf2). Nrf2 is a transcription factor that regulates the expression of antioxidant and detoxification genes in response to oxidative stress [27]. Under basal conditions, Nrf2 is sequestered in the cytoplasm by Keap1 (Kelch-like ECH-associated protein 1). Upon exposure to oxidative stress, Nrf2 is released from Keap1, translocates to the nucleus, and activates the transcription of antioxidant response element (ARE)-containing genes, such as those encoding antioxidant enzymes, phase II detoxification enzymes, and NADPH-generating enzymes [28].
Heme oxygenase-1 (HO-1). Heme oxygenase-1 is an enzyme that catalyzes the degradation of heme into biliverdin, carbon monoxide (CO), and iron. HO-1 induction is a key adaptive response to oxidative stress and inflammation, serving as a cytoprotective mechanism against oxidative damage and cellular stress [29].
Peroxiredoxins (PRX). Peroxiredoxins are a family of antioxidant enzymes that catalyze the reduction of peroxides, including hydrogen peroxide (H2O2), alkyl hydroperoxides, and peroxynitrite. PRX enzymes utilize thioredoxin or glutathione as reducing equivalents to detoxify peroxides and maintain cellular redox balance [30].
These genes represent a subset of the many components involved in cellular antioxidant defense mechanisms and oxidative stress responses. Their coordinated regulation ensures that cells can effectively neutralize reactive oxygen species and mitigate oxidative damage to maintain cellular homeostasis.

3. Role of Oxidative Stress in Normal Cell Signal Transduction and Homeostasis

Reactive oxygen species play a dual role in biological systems. While they are often associated with harmful effects leading to cell damage and disease, at physiological levels, ROS play crucial roles as second messengers in many intracellular signaling cascades aimed at maintaining cells in homeostasis with their microenvironment [31].
At low to moderate levels, ROS act as secondary messengers in various signaling pathways. They modulate the activity of key proteins by oxidizing specific thiol groups on cysteine residues, leading to reversible modifications that can alter protein function.
In the context of the Epidermal Growth Factor Receptor (EGFR) pathway, ROS are generated upon the binding of growth factors like EGF to their receptors. The activation of NOX leads to the production of superoxide, which is converted into H2O2. In turn, H2O2 modulates the activity of protein tyrosine phosphatases (PTPs) by oxidizing their cysteine residues, leading to the persistence of phosphorylation events and thus amplifying the growth factor signaling. This regulation can affect cellular processes such as proliferation, migration, and differentiation [32,33].
In the context of the Hypoxia-Inducible Factor (HIF) pathway, ROS are produced in the mitochondria in response to hypoxia. These ROS stabilize HIF-1α by inhibiting prolyl hydroxylases, which usually mark HIF-1α for degradation under normal oxygen conditions. Stabilized HIF-1α translocates to the nucleus, where it activates the transcription of genes involved in angiogenesis (e.g., vascular endothelial growth factor, VEGF), erythropoiesis, and glycolysis, helping cells adapt to hypoxic conditions [34].
ROS can modulate the activity of kinases, such as protein tyrosine kinases and mitogen-activated protein kinases (MAPKs), which are involved in controlling cell proliferation, differentiation, and apoptosis [35]. ROS-mediated activation of kinases in the MAPK pathway, such as extracellular signal-regulated kinases (ERKs), c-Jun N-terminal kinases (JNKs), and p38 MAPKs, occurs through the oxidative modification of cysteine residues in upstream kinases and phosphatases, leading to cellular responses like proliferation, differentiation, stress responses, and apoptosis [35].
ROS regulate the activity of transcription factors [36], including, a. Nuclear Factor-kappa B (NF-κB). NF-κB, a transcription factor involved in inflammatory and immune responses, can be activated by ROS. The oxidative modification of inhibitory proteins like IκB leads to the release and activation of NF-κB, allowing it to translocate to the nucleus and initiate gene expression. b. Activator Protein-1 (AP-1). ROS can enhance the activity of AP-1, which regulates gene expression related to proliferation and differentiation. c. Nrf2. Under normal conditions, Nrf2 is bound to its inhibitor Keap1 and targeted for degradation. Under oxidative stress, Nrf2 dissociates from Keap1, translocates to the nucleus, and activates the expression of genes involved in antioxidant defense and detoxification, thus restoring homeostasis.
Cells maintain redox homeostasis by balancing ROS production and scavenging. This balance is critical for cellular adaptation to environmental changes. ROS can induce adaptive responses, such as the upregulation of antioxidant defense systems (e.g., glutathione peroxidase, superoxide dismutase), which help restore redox balance.
Controlled ROS levels are involved in the regulation of cell cycle progression, proliferation, and differentiation. For example, H2O2 has been shown to regulate the transition between different phases of the cell cycle, influencing cell division. ROS influence the expression and activity of key cell cycle regulators, such as cyclins, cyclin-dependent kinases (CDKs), and CDK inhibitors. For instance, moderate ROS levels can upregulate cyclin D1, driving the transition from the G1 to the S phase of the cell cycle, thereby promoting DNA synthesis and cell division. ROS can activate or inhibit Notch signaling depending on the context, influencing differentiation outcomes [37,38].
ROS can influence calcium signaling by modulating calcium channels and transporters. Calcium is a vital second messenger in numerous cellular processes, and ROS can influence intracellular calcium levels by modulating calcium channels and pumps, such as the ryanodine receptor (RyR) and the inositol 1,4,5-trisphosphate receptor (IP3R). ROS-induced calcium release from the endoplasmic reticulum can activate various calcium-dependent signaling pathways, influencing processes like muscle contraction, neurotransmitter release, and cell death [39,40].
ROS play a pivotal role in apoptosis by influencing signaling pathways that regulate cell survival and death. For example, ROS can activate pro-apoptotic proteins like Bax and Bak, leading to mitochondrial outer membrane permeabilization (MOMP) and the release of cytochrome c, which activates caspases to execute cell death. ROS also modulate signaling through the p53 pathway, which can lead to cell cycle arrest or apoptosis depending on the context [41].
ROS are critical regulators of autophagy, a cellular degradation process that helps maintain homeostasis by removing damaged organelles, misfolded proteins, and pathogens. Autophagy can be induced by various stressors, including nutrient deprivation, hypoxia, and oxidative stress. By influencing key signaling pathways and transcription factors, ROS can induce and regulate the autophagic process, promoting cell survival under stress conditions by clearing damaged components and maintaining cellular homeostasis [42].
ROS are involved in signaling processes that regulate immune cell functions, such as phagocytosis [43] and the production of cytokines [44]. Low levels of ROS contribute to the regulation of immune responses, ensuring that immune cells respond appropriately to pathogens without causing excessive tissue damage. For instance, upon activation of TLR4 by lipopolysaccharide (LPS), ROS are produced by NOX and mitochondria. These ROS participate in the activation of downstream signaling pathways, including the NF-κB and MAPK pathways, which lead to the production of pro-inflammatory cytokines and other immune responses [45,46].
In summary, oxidative stress, when tightly regulated, plays an essential role in normal cell signal transduction and homeostasis. It acts as a critical regulator of various cellular processes, including redox signaling, proliferation, differentiation, and immune responses. The fine balance between ROS production and antioxidant defense systems ensures that cells can respond to environmental cues and maintain homeostasis without tipping into pathological states associated with excessive oxidative damage.

4. Oxidative Stress and Ageing

Oxidative stress promotes aging through a series of interconnected biological processes, and age is one of the main risk factors for cancer [47]. The following subsections present detailed explanations of the reasons behind this connection.
Cellular and DNA Damage and Mutations. Over the years, oxidative stress and DNA damage caused by life-long exposure to endogenous metabolic insults (e.g., free radicals) and exogenous factors (e.g., UV irradiation, foods, etc.) accumulate. ROS can react with DNA, leading to various types of damage, including base modifications, single-strand breaks, double-strand breaks, and mutagenic lesions such as the formation of 8-oxo-2′-deoxyguanosine (8-oxo-dG), which can lead to erroneous base pairing during DNA replication. 8-oxo-dG-induced DNA damage can activate cellular senescence [48], a state of permanent growth arrest, resulting in the secretion of inflammatory factors, which further contribute to tissue degeneration and aging. The accumulation of 8-oxo-dG in DNA leads to increased genomic instability, which is a hallmark of aging and age-related diseases. Overall, the accumulation of DNA damage can result in faulty protein production and disrupted cellular processes, which impair cellular functions and contribute to genomic instability, promoting aging processes and increasing the risk of developing cancer [49].
Mitochondrial Dysfunction. Mitochondria are the powerhouses of the cell and are a major source of ROS. Excessive ROS production can damage mitochondrial components, including lipids, proteins, and mitochondrial DNA (mtDNA), leading to mitochondrial dysfunction. Mitochondrial membranes are particularly vulnerable to lipid peroxidation, which can impair the function of membrane-bound enzymes and ion channels. Damage to mtDNA can lead to mutations. Unlike nuclear DNA, mtDNA is more susceptible to oxidative damage due to its proximity to the electron transport chain and the lack of protective histones [50]. Accumulated mutations in mtDNA can disrupt the synthesis of key proteins required for the electron transport chain (ETC), further impairing mitochondrial function and increasing ROS production in a vicious cycle that accelerates cellular aging and reduces energy production [51].
Telomere Shortening. Telomeres are protective caps at the ends of chromosomes that shorten with each cell division. Oxidative stress accelerates telomere shortening through different mechanisms [52,53]: 1. Direct Oxidative Damage to Telomeres. Telomeres are particularly susceptible to oxidative damage due to their high guanine content (G-rich sequences). Guanine is the most easily oxidized of the four DNA bases, and oxidation leads to the formation of 8-oxoguanine (8-oxoG), a mutagenic lesion. ROS-induced damage at telomeres results in single-strand breaks and base modifications. DNA repair at telomeres is less efficient compared to other genomic regions, due to the limited access of DNA repair machinery at these specialized chromosomal ends, leading to unresolved damage and faster telomere attrition [54]. 2. Impaired Telomerase Function. Oxidative stress can impair the function of telomerase (the enzyme responsible for adding telomeric repeats to the ends of chromosomes) through several pathways: a. telomerase inhibition. ROS can directly damage the telomerase RNA component (TERC) or the reverse transcriptase (TERT) catalytic subunit, leading to reduced telomerase activity and accelerated telomere shortening; b. decreased telomerase recruitment. Oxidative stress can interfere with the recruitment of telomerase to telomeres, further preventing telomere elongation. 3. Telomere Loop (T-loop) Disruption. Telomeres form a protective structure called the T-loop, which hides the chromosome end from being recognized as a DNA break [55]. Oxidative stress can disrupt this structure, exposing the telomere ends and making them more prone to degradation and fusion. 4. Replication Stress at Telomeres. ROS can induce replication stress, leading to stalled replication forks, particularly at telomeres. Telomeres are difficult to replicate because of their repetitive sequence and secondary structures like G-quadruplexes, which make them more vulnerable to replication fork stalling and collapse [56]. Fork stalling can result in incomplete replication of telomeres, leading to shortened telomeres with each cell division.
When telomeres become critically short, cells enter a state of senescence or apoptosis, contributing to tissue aging and dysfunction, or leading to genomic instability by causing chromosome end-to-end fusions, breakage-fusion-bridge cycles, and aneuploidy. This genomic instability can drive the progression of cancer [57,58].
Protein Oxidation and Aggregation. ROS can affect protein structure and function. ROS can directly modify the side chains of amino acids, leading to the formation of disulfide bonds (in the case of cysteine) or other oxidized derivatives, which can disrupt the protein’s tertiary and quaternary structure. ROS can attack the carbon backbone of amino acids, leading to the introduction of carbonyl groups (C=O) into the protein structure, resulting in a loss of protein function or increased susceptibility to proteolytic degradation. ROS can either disrupt or form incorrect disulfide linkages, which can lead to the misfolding or aggregation of proteins. ROS can break peptide bonds within the protein, leading to fragmentation and loss of structural integrity [59]. Oxidized proteins can lose their enzymatic activity or alter cellular signaling when involved in signaling pathways. The impairment of structural integrity makes proteins susceptible to aggregation. Protein aggregation is sufficient to activate the stress response and inflammation, impairing protein synthesis and quality control mechanisms, which are associated with age-related diseases [60] and are implicated in anti-tumor therapy resistance [61].
Inflammation. Oxidative stress can trigger inflammatory responses, through the activation of redox-sensitive transcription factors such as NF-κB and AP-1, leading to chronic inflammation. “Inflammaging” refers to the chronic, low-grade inflammation that typically accompanies aging, which is characterized by increased levels of pro-inflammatory markers in the blood and tissues [8] which feeds oxidative stress, creating a feedback loop that exacerbates cellular damage. This phenomenon is thought to be driven by cellular senescence, mitochondrial dysfunction, altered immune response, and epigenetic changes, which promote cancer development and the response to anti-cancer immunotherapy [62,63].
Cellular Senescence. Cells that experience significant oxidative damage may enter a state of senescence, where they no longer divide but remain metabolically active. Senescent cells secrete pro-inflammatory cytokines, growth factors (including IL-6, IL-8, TNF-α, MCP-2/CCL8, MCP-1/CCL2, and GRO-α/CXCL1), proteases (such as matrix metalloproteinases, MMPs), and other signaling molecules known as the senescence-associated secretory phenotype (SASP). SASP factors can damage surrounding tissues and disrupt normal cellular function, contributing to aging and inflammation [64] and creating a tumorigenic microenvironment.
Impaired Autophagy. Autophagy is a cellular process that degrades and recycles damaged organelles, misfolded proteins, and other cellular debris. This process is crucial for maintaining cellular homeostasis, especially under stress conditions. Oxidative stress can impair autophagy, leading to the accumulation of damaged cellular components. This impairment further exacerbates cellular dysfunction and contributes to the aging process [65]. The decline in autophagy with age contributes to the accumulation of cellular damage, increasing the risk of cancer development.
Antioxidant Defense Decline. Aging is associated with a decline in antioxidant defense mechanisms, including enzymatic antioxidants such as SOD, catalase, and glutathione peroxidase, as well as non-enzymatic antioxidants like vitamins C and E, glutathione, and flavonoids. Reduced antioxidant capacity contributes to increased susceptibility to oxidative damage and impaired cellular homeostasis with age [66,67].
In summary, oxidative stress promotes aging by causing cumulative damage to DNA, proteins, lipids, and cellular organelles. This damage leads to mitochondrial dysfunction, telomere shortening, protein aggregation, chronic inflammation, cellular senescence, and impaired autophagy. Together, these processes drive the decline in cellular and tissue function that characterizes aging.

5. Oxidative Stress and Tumorigenesis

Oxidative stress is implicated in the development and progression of age-related cancers by promoting DNA damage, inflammation, cellular senescence, mitochondrial dysfunction, and impaired antioxidant defenses [68,69].
The link between oxidative stress and DNA damage, which plays a significant role in aging and cancer, arises from the imbalance between the production of ROS and the body’s ability to neutralize them with antioxidants. When the levels of highly reactive molecules, such as free radicals like O2•−, OH, and non-radical molecules like H2O2, exceed the capacity of antioxidant defenses, they can attack cellular components, including DNA, and cause multiple types of damage: a. Single-strand breaks (SSBs). ROS can cause breaks in one strand of the DNA helix, leading to replication errors or mutations. Mutations leading to oncogene activation and tumor suppressor gene [70] inactivation can drive tumorigenesis [71]. b. Double-strand breaks (DSBs). More severe than SSBs, DSBs occur when both strands of the DNA helix are broken, which can lead to chromosomal rearrangements or cell death if not repaired. DNA damage sensors, such as the Ataxia-Telangiectasia Mutated (ATM) kinase, which is a critical sensor for double-strand breaks, and Poly ADP-Ribose Polymerase (PARP), which recognizes single-strand breaks, are the first responders in the DNA repair process, and their functions are critical to the maintenance of genome integrity. c. Base modifications. ROS can oxidize DNA bases, such as the conversion of guanine to 8-oxo-7,8-dihydroguanine (8-oxoG), one of the most common markers of oxidative DNA damage. This can lead to mispairing during replication and mutagenesis [72]. d. DNA cross-linking. ROS can also induce covalent bonding between DNA strands or between DNA and proteins, impairing normal cellular processes [73].
To address oxidative DNA damage, cells have evolved several DNA repair mechanisms [74] namely, a. Base excision repair (BER). This pathway is the primary repair mechanism for oxidative base modifications like 8-oxoG. Enzymes such as glycosylases recognize and remove damaged bases, and DNA polymerase fills the gap. b. Nucleotide excision repair (NER). This pathway repairs bulky DNA lesions, such as those caused by cross-linking. c. Non-homologous end joining (NHEJ) and homologous recombination (HR). These pathways repair DSBs, with HR being the more accurate of the two as it uses a sister chromatid as a template, ensuring error-free repair [75]. However, if not properly repaired, DNA damage can lead to several adverse outcomes, including apoptosis or necrosis, genomic instability, and mutations.
Accumulation of mutations in genes involved in cell cycle regulation, DNA repair, and tumor suppression increases the risk of age-related cancers, such as breast, colorectal, prostate, and lung cancers and hematological malignancies such as non-Hodgkin lymphoma, multiple myeloma, and leukemia [76]. Oxidative stress plays a multifaceted role in all stages of tumorigenesis [76] by promoting DNA damage and mutations, which foster cell proliferation, angiogenesis, invasion, metastasis, and resistance to therapy [77,78], as outlined below and in Figure 1.
Initiation. ROS can cause DNA strand breaks, base modifications, and chromosomal rearrangements, leading to mutations in oncogenes, tumor suppressor genes, and genes involved in DNA repair, which promote uncontrolled cell proliferation and the development of pre-cancerous lesions [77]. Oncogenes such as RAS, MYC, and RAF can be activated by oxidative stress through mutations or epigenetic modifications, leading to cell proliferation and survival. By contrast, tumor suppressor genes such as TP53 (p53), RB, and PTEN can be inactivated, either through direct mutations or through signaling pathways that degrade these proteins [79]. Oxidative stress also supports the cancer stem cell (CSC) compartment, which contributes to tumor initiation, progression, metastasis, and therapy resistance by activating signaling pathways involved in self-renewal and pluripotency. For example, the ROS-mediated activation of signaling pathways, such as Wnt/β-catenin, Notch, and Hedgehog, can enhance CSC self-renewal capacity and maintain their stemness properties [80]. The accumulation of mutations in these genes increases the risk of age-related cancers [67].
Promotion. Chronic exposure to oxidative stress promotes tumor growth and progression by inducing genomic instability, enhancing cell proliferation, and fostering a pro-tumorigenic microenvironment [81]. ROS-mediated activation of signaling pathways, such as NF-κB and AP-1, stimulates the expression of genes involved in inflammation, angiogenesis, and cell survival. ROS produced during oxidative stress can activate inflammatory signaling pathways, leading to the production of cytokines and chemokines that promote inflammatory cell recruitment. Intratumorally recruited myeloid-derived suppressor cells (MDSCs), macrophages, and neutrophils can produce ROS, further exacerbating oxidative stress and creating a pro-tumorigenic milieu that supports cancer growth and progression [82]. Oxidative stress-dependent inactivation of tumor suppressor genes and the activation of oncogenes contribute to tumor progression [79].
Angiogenesis. Oxidative stress promotes angiogenesis, the formation of new blood vessels, which is essential for supplying nutrients and oxygen to growing tumors [83,84]. ROS stimulate the expression of angiogenic factors, such as VEGF and HIF1A, promoting endothelial cell proliferation and vessel sprouting [85]. Increased angiogenesis facilitates tumor growth, invasion, and metastasis, contributing to the progression of cancer [86].
Metastasis. Oxidative stress plays a role in the metastatic spread of cancer cells to distant sites. ROS-mediated activation of epithelial-to-mesenchymal (EMT)-associated transcription factors, such as Snail, Slug, and Twist, contributes to the invasive phenotype of CSCs and facilitates metastatic dissemination [87]. Additionally, oxidative stress promotes the activation of proteases involved in extracellular matrix degradation, facilitating cancer cell invasion and metastasis [88].
Effects on the tumor microenvironment (TME) and immune cell functions. Immuno-senescence contributes to a higher incidence of cancer due to reduced immune surveillance and impaired antitumor immunity [89], to which oxidative stress contributes [90,91]. ROS can influence the activation and differentiation of immune cells. Moderate levels of ROS are necessary for T cell activation, but excessive ROS can cause T cell exhaustion and apoptosis. High levels of ROS in the TME can lead to immunosuppression by promoting regulatory T cells and MDSCs.
Resistance to therapy. High levels of ROS are mostly involved in programmed cell death and/or apoptosis [92]. Radiation therapy kills cancer cells by generating high levels of ROS, which contributes to inducing severe DNA damages and cancer cell death. However, oxidative stress can contribute to the development of resistance to chemotherapy and radiation therapy in cancer cells since the self-renewal capability of CSCs, the main responsible for tumor growth and progression, is firmly regulated by ROS levels [93,94]. In quiescent stem cells, very low levels of ROS may be indispensable for their stemness maintenance; however, physiological ROS levels may encourage stem cell proliferation and/or differentiation [95]. CSCs also exhibit increased resistance to oxidative stress-induced cell death compared to non-CSCs. This resistance is attributed to elevated expression of antioxidant enzymes such as SOD, catalase, and glutathione peroxidase, which help CSCs neutralize ROS and protect them from oxidative damage. As a result, CSCs can survive under conditions of oxidative stress induced by chemotherapy, radiation therapy, or the TME [92,96]. Moreover, oxidative stress-induced alterations in DNA repair pathways can reduce the efficacy of DNA-damaging agents, further contributing to therapy resistance.
Interplay with immunotherapy. Chemotherapeutics, such as oxaliplatin, bleomycin, and doxorubicin, can induce ROS-mediated immunotherapy. Excessive ROS promote the immunogenic cell death (ICD) of tumors, providing a potential antigenic stimulation for the immune system [97]. Studies suggest that ROS can increase the infiltration of dendritic cells (DCs) and T cells in the TME and transform “cold” (low immunogenicity) tumors into “hot” (high immunogenicity) tumors, thereby increasing a more effective antitumor immune response [98,99]. Nevertheless, it has been reported that, in breast cancer, the elimination of ROS alleviates the immunosuppressive ICD in the TME and increases antitumor immunity and T-lymphocyte infiltration, resulting in a potent antitumor effect [100]. Collectively, these contradictory results suggest that ICD can be induced by modulating ROS to obtain enhanced immunotherapy.
Understanding the mechanisms underlying the interplay of oxidative stress with both the immune environment and CSCs is essential for developing effective therapeutic approaches to target immune-resistant and aggressive tumor cell populations. Strategies aimed at reducing oxidative stress and its effects may help mitigate cancer risk and improve outcomes in aging populations.

6. Antioxidants in the Prevention and Treatment of Age-Related Cancer

Antioxidant compounds have garnered considerable interest in cancer prevention and therapy due to their ability to neutralize free radicals, reduce oxidative stress, and mitigate some of the cellular damage associated with aging processes and the risk of developing cancer [101]. Here, we provide an overview of the current understanding of the role of antioxidant compounds in cancer prevention and therapy resulting from ongoing research and completed studies.

6.1. Antioxidant Mechanisms of Action

Antioxidants scavenge free radicals, preventing oxidative damage to proteins, lipids, and DNA that can lead to mutations and cancer development. Antioxidants modulate signaling pathways involved in cell proliferation, apoptosis, and inflammation, all of which are relevant to cancer development and progression.
Recent clinical trials have found that antioxidant supplementation can significantly improve certain immune responses [102]. Vitamins C and E, selenium, and zinc play vital roles in supporting the proper functioning of the immune system [103]. Specifically, supplementation with vitamins C, E, and A or beta-carotene increased the activation of cells involved in tumor immunity in the elderly [104,105].

6.2. Types of Antioxidants

While endogenous antioxidants are produced by the body and include enzymes like SOD, catalase, and glutathione peroxidase, exogenous antioxidants are obtained from the diet and supplements, as follows.
Vitamins. Vitamin C is found in citrus fruits, strawberries, bell peppers, and broccoli. It helps regenerate other antioxidants and is crucial for skin health and immune function. Vitamin E is found in nuts, seeds, spinach, and vegetable oils. It protects cell membranes from oxidative damage.
Carotenoids. Beta-carotene is found in carrots, sweet potatoes, and spinach. It is a precursor to vitamin A and plays a role in vision and immune function. Lycopene is found in tomatoes, watermelon, and pink grapefruit. It has been linked to a reduced risk of certain cancers.
Polyphenols. Flavonoids are found in berries, tea, onions, and apples. They have anti-inflammatory and anti-carcinogenic properties. Resveratrol is found in red wine, grapes, and peanuts. It is known for its potential cardiovascular benefits.
Minerals. Selenium is found in Brazil nuts, seafood, and meats. It is a component of antioxidant enzymes like glutathione peroxidase. Zinc is found in meat, shellfish, legumes, and seeds. It helps maintain the function of antioxidant enzymes.

6.3. Evidence from Research and Clinical Studies

Epidemiological Studies. Many studies suggest a correlation between high dietary intake of antioxidants and a reduced risk of certain cancers. For example, a diet rich in fruits and vegetables is associated with a lower risk of cancers, such as colorectal, prostate, and lung cancer [106,107].
Clinical Trials. The results of clinical trials are mixed. Some studies have shown benefits, while others have not demonstrated significant effects or have even suggested potential harm in certain populations. For instance, the ATBC (Alpha-Tocopherol, Beta-Carotene Cancer Prevention) Study found that beta-carotene supplementation increased the incidence of lung cancer in smokers. Conversely, the SELECT (Selenium and Vitamin E Cancer Prevention Trial) did not show a protective effect of selenium and vitamin E supplements against prostate cancer and indicated an increased risk of prostate cancer with vitamin E supplementation [108].
Preclinical Studies. In vivo and in vitro studies often show that antioxidants can inhibit cancer cell growth and induce apoptosis [109,110]. However, applying these findings to humans is complex because antioxidants’ effects may depend on complex in vivo conditions, such as a. the low bioavailability and bio-accessibility in some specific organs, b. their concentration and the levels of oxidative stress and hypoxia in the TME, and c. their potential pro-oxidant activity. In aerobic conditions, they generate superoxide radicals and dismutate to H2O2, which reacts with reduced metal ions and superoxide to ROS [111].

6.4. Antioxidants in Cancer Treatment

Chemoprevention. Antioxidants have been explored as agents to prevent the development of cancer in high-risk populations. For example, retinoids (vitamin A derivatives) are used in the prevention of certain types of skin cancer.
Adjunct Therapy. Antioxidants are sometimes used alongside conventional cancer treatments to mitigate side effects and improve overall health. For instance, studies on curcumin and resveratrol have shown promising results in sensitizing cancer cells to chemotherapy [112]. Antioxidants may enhance the efficacy of conventional cancer treatments, such as chemotherapy and radiation, and reduce their side effects by protecting normal cells from oxidative damage [113]. However, there is concern that antioxidants might interfere with the efficacy of chemotherapy and radiation therapy, which rely on generating oxidative stress to kill cancer cells. Ongoing research is evaluating the effects of antioxidants for cancer prevention and as adjuvants in cancer therapy. Table 1 reports active clinical trials testing antioxidants for cancer prevention and treatment.
Controversies and Considerations. While antioxidants have potential in the prevention and treatment of age-related cancers, their role is complex and not fully understood. Different antioxidant compounds failed to demonstrate efficacy in clinical practice. Table 2 reports the effects of clinically tested antioxidant supplementation in cancer treatment.
Pro-oxidant Activity. At high doses, some antioxidants may have pro-oxidant effects, potentially contributing to oxidative stress and cancer risk.
Population-Specific Effects. The impact of antioxidants may vary based on genetic, environmental, and lifestyle factors. What is beneficial for one group may not be for another.
Dosage and Formulation. The form and dose of antioxidants are crucial as excessive antioxidant intake might interfere with the oxidative stress required for the effectiveness of certain cancer therapies. Natural sources (e.g., whole fruits and vegetables) are generally preferred over high-dose supplements due to the potential for adverse effects with the latter.
In conclusion, individual variability in response to antioxidants exists, influenced by genetic factors, the type and stage of cancer, and overall health status. Antioxidant compounds hold significant potential in the prevention and treatment of cancer due to their ability to mitigate oxidative stress and modulate various biological processes related to cancer development. While the incorporation of antioxidants from a balanced diet is generally beneficial, further research is needed to fully understand the optimal use of antioxidant supplements in cancer therapy.

7. Imaging Techniques to Study Oxidative Damage In Vivo

Studying oxidative damage in vivo requires advanced imaging techniques that can detect and quantify ROS and oxidative damage to biomolecules, such as DNA, proteins, and lipids, providing insights into the related pathological implications in real-time, within living organisms. The main imaging techniques used to study oxidative damage in vivo are the following.
  • Fluorescence and Bioluminescence Imaging, which provide sensitive and non-invasive ways to visualize and quantify oxidative stress and related cellular damage, by means of fluorescent probes [128] or bioluminescent reporters [129], in living tissues and organisms [130]:
    • ROS-sensitive probes can be used to visualize and quantify ROS in living cells. These probes react with specific ROS to emit fluorescence, which can be detected using fluorescence or confocal microscopy. Dihydroethidium (DHE) reacts with O2•− to produce fluorescent ethidium, which intercalates into DNA. 2′,7′-Dichlorodihydrofluorescein diacetate (DCFH-DA) reacts with hydrogen peroxide (H2O2) to produce a fluorescent product, dichlorofluorescein (DCF). MitoSOX™ (Thermo Fisher Scientific, Waltham, MA, USA), a mitochondria-targeted version of DHE, specifically detects superoxide production within mitochondria, a key site of oxidative stress.
    • Lipid Peroxidation Probes, such as BODIPY C11, are fluorescent dyes that detect lipid peroxidation, a process driven by oxidative stress that damages cell membranes.
    • Protein and DNA Damage Probes, such as a. Oxidation-Sensitive GFP (roGFP), which is a genetically encoded fluorescent protein that changes its fluorescence in response to changes in the redox state of cells, allowing real-time visualization of oxidative stress; and b. DNA Damage Sensors consisting of fluorescent probes or labeled antibodies that detect 8-oxoguanine, a marker of oxidative DNA damage.
    • Bioluminescent Reporters are genetically encoded or chemically synthesized fluorescent proteins that emit light when excited by a specific wavelength of light. These fluorophores can be targeted to specific cells, organelles, or molecules involved in oxidative stress. For example, luciferase-based reporters, where oxidative stress-induced gene expression triggers bioluminescence, allow real-time imaging of oxidative damage [129].
  • Magnetic Resonance Imaging (MRI) is a powerful non-invasive imaging technique with excellent spatial resolution and the ability to visualize soft tissues without ionizing radiation. MRI has been adapted to visualize oxidative damage by using redox-sensitive contrast agents or combined with Electron Paramagnetic Resonance (EPR) [131]:
    • Redox-Sensitive MRI Contrast Agents that are sensitive to the redox state of tissues can be used to assess oxidative damage. For example, manganese-based contrast agents are oxidized or reduced depending on the oxidative environment, altering their MRI signal.
    • EPR is a technique that detects unpaired electrons, such as those in free radicals, making it well-suited for studying oxidative stress [132]. EPR can be combined with MRI to create EPR-MRI, allowing for spatial mapping of free radicals in tissues. Nitroxide-based probes are commonly used in EPR-MRI to detect ROS [133]. These probes are stable free radicals that change their magnetic properties upon reduction by antioxidants or reaction with ROS, providing imaging of oxidative damage in vivo.
  • Positron Emission Tomography (PET) is traditionally used to visualize metabolic processes, such as glucose metabolism or blood flow, and can also be applied to study oxidative damage, using specialized radiotracers that target oxidative stress or its related consequences:
    • PET Imaging with Redox-Sensitive Radiotracers that are sensitive to redox changes, such as [18F]-labeled dihydroethidine (18F-DHE), and especially [18F]ROStrace, have proven effective for visualizing ROS production and oxidative stress in live models of neuroinflammation [134].
    • Hypoxia Imaging. PET imaging with tracers like [18F]FMISO (fluoromisonidazole) is used to study oxidative stress associated with hypoxic conditions (leading to oxidative stress) in ischemic tissues or to identify hypoxic regions of tumors [135], which may be more aggressive and resistant to treatment. PET imaging with 62Cu-ATSM, which accumulates in the presence of an over-reductive state, detects oxidative stress in the disease-related brain regions of patients with mitochondrial disease, Parkinson’s disease, and ALS and can be useful for monitoring antioxidant therapies [136].
  • Optical Imaging (Near-Infrared Fluorescence, NIRF) takes advantage of the tissue-penetrating properties of near-infrared light and the use of fluorescent probes that can penetrate deep tissues and be activated by ROS or that target specific molecular markers of oxidative stress. ROS-activated probes are non-fluorescent in their native state but become fluorescent upon reacting with ROS, such as H2O2, O2•−, or OH. Examples include IR-775c, a near-infrared dye (with absorption and emission typically between 650–900 nm, and good photostability), which becomes fluorescent upon oxidation by ROS, and Cy7-Based Probes, a family of cyanine dyes (with absorption typically around 740 nm and emission around 770–790 nm), which are commonly used in NIRF imaging and can be modified to become activated by ROS.
  • Multiphoton Microscopy employs two or more photons of low energy (near-infrared light) to excite a fluorophore, which then emits light at a higher energy level. The use of longer wavelength light allows for deeper penetration into tissues and minimizes photodamage, making it ideal for studying processes such as oxidative stress in living cells and tissues [137,138]:
    • Multiphoton Excitation Microscopy allows the viewing of oxidative stress-induced changes in cellular structures and can also image ROS by using fluorescent probes, such as dihydroethidium (DHE), which becomes fluorescent when oxidized by superoxide; and MitoSOX™, which is a mitochondria-targeted fluorescent probe that selectively detects superoxide production within mitochondria, a major site of oxidative stress [139].
    • Multiphoton microscopy can also detect the intrinsic autofluorescence of naturally occurring fluorophores like NADH/NADPH and flavoproteins (involved in oxidative phosphorylation and used to assess mitochondrial function), whose fluorescence can change in response to oxidative stress, allowing for indirect detection of oxidative damage.
  • Raman Spectroscopy and Imaging provide non-invasive, label-free detection of biochemical alterations, making them highly suitable for investigating oxidative modifications in lipids, proteins, and nucleic acids [140] and enabling real-time monitoring of oxidative stress and damage under physiological conditions:
    • Surface-Enhanced Raman Scattering (SERS) imaging can detect molecular vibrations that provide detailed information about chemical structures. SERS enhances the Raman signal, enabling the detection of biomolecules involved in oxidative stress, such as lipid peroxidation or protein oxidation, and nucleic acid alterations, providing information on base modifications, backbone damage, and strand breaks.
    • Label-Free Raman Imaging can be used to detect oxidative damage to biomolecules without using external probes, thus providing a label-free approach to studying oxidative stress in vivo [141].
  • Super-Resolution Microscopy, which includes STORM (Stochastic Optical Reconstruction Microscopy) and PALM (Photoactivated Localization Microscopy), relies on the precise localization of individual fluorescent molecules. By turning subsets of fluorophores on and off, their exact positions can be calculated, allowing the construction of super-resolution images (at the nanoscale, 20–30 nm) from the aggregated data. STORM and PALM allow for visualizing oxidative damage at the molecular level within cells (cellular organelles, such as mitochondria and endoplasmic reticulum) and are especially useful in studying oxidative damage to DNA or proteins in situ [142].
In conclusion, the study of oxidative damage in vivo relies on a range of imaging techniques, each with its strengths, depending on the type of oxidative damage being studied, the depth of tissue penetration required, and the resolution needed to observe cellular and molecular events in living organisms. These techniques are critical for understanding the role of oxidative stress in diseases such as cancer, neurodegeneration, and cardiovascular disorders, and they also help in evaluating the efficacy of antioxidants and other therapeutic interventions.

8. Overgeneration of ROS as Anti-Cancer Therapy

ROS play a dual role in cancer biology. They can promote tumorigenesis at low levels, while at high levels, they cause cell damage and apoptosis [143]. Therefore, ROS overproduction, which can be generated by a variety of tools, including photodynamic therapy (PDT) [144] and sonodynamic therapy (SDT) [145], is a promising anti-cancer strategy. ROS overproduction for anti-cancer purposes is a subject of active clinical investigation and is especially used in combination therapies with inhibitors of antioxidant pathways (e.g., targeting glutathione or thioredoxin systems) or traditional chemotherapeutics and with radiotherapy (which induce ROS overproduction) to enhance cancer cell killing and achieve synergistic effects.
Notably, many chemotherapeutic drugs, such as doxorubicin, cisplatin, and paclitaxel, induce ROS production as part of their cytotoxic effects. These drugs either directly generate ROS or disrupt cellular processes that lead to ROS accumulation [146,147].
Doxorubicin is an anthracycline chemotherapy drug, widely used to treat a variety of cancers, including breast and lung cancers, lymphomas, leukemias, and sarcomas, due to its ability to intercalate DNA, inhibit topoisomerase II, and generate free radicals [148]. Doxorubicin generates ROS primarily through redox cycling of its quinone moiety. This process involves the enzymatic reduction of the quinone group to a semiquinone radical by NADPH-dependent enzymes, such as NADPH cytochrome P450 reductase or NADH dehydrogenase (Complex I of the electron transport chain). The semiquinone radical is unstable and can rapidly react with O2, leading to the formation of O2, which can further generate other ROS, such as H2O2 and OH·, through dismutation and Fenton reactions, respectively. This cycling between the quinone and semiquinone forms can occur repeatedly, amplifying the production of ROS. Doxorubicin accumulates in mitochondria, where it can interact with components of the electron transport chain (ETC), especially Complexes I and III, and disrupt the normal flow of electrons through the ETC, leading to electron leakage and production of O2, which in turn generate more ROS.
Alkylating agents, like cisplatin, a platinum-based chemotherapeutic used for the treatment of solid tumors [149] such as testicular [150], ovarian [151], bladder [152], lung [153], and head and neck cancers [154] that can bind to and damage mitochondrial DNA (mtDNA), lead to the formation of crosslinks that distort the DNA structure and inhibit transcription of essential components of the ETC. Cisplatin can also directly impair the function of the ETC, leading to electron leakage from the mitochondrial inner membrane and consequent overproduction of ROS, particularly O2. Highly reactive ROS can cause oxidative damage to lipids, proteins, and mitochondrial membranes, further exacerbating mitochondrial dysfunction.
Paclitaxel (Taxol) is a widely used chemotherapeutic agent primarily for the treatment of solid tumors, including breast [155], ovarian [156], and lung [157] cancers, which exerts its antitumor effects by stabilizing microtubules, preventing their depolymerization, and thereby disrupting cell division. Beyond its effects on the cytoskeleton, paclitaxel induces ROS overproduction [158] through several mechanisms that contribute to both its therapeutic efficacy and its side effects. Paclitaxel induces mitochondrial dysfunction by disrupting the ETC, leading to the leakage of electrons from the ETC, which can prematurely react with molecular oxygen to form O2•−. These superoxides can be further converted into other ROS like H2O2 and OH radicals. Furthermore, paclitaxel causes changes in the mitochondrial membrane potential (ΔΨm), leading to mitochondrial depolarization and dysfunction, further increasing ROS production. Additional mechanisms of paclitaxel-induced ROS overproduction include endoplasmic reticulum stress, due to the accumulation of misfolded proteins within the endoplasmic reticulum, which triggers the unfolded protein response (UPR) and activates pathways that increase the production of ROS. Endoplasmic reticulum stress also leads to the release of calcium ions (Ca2+) from the endoplasmic reticulum into the cytosol, resulting in abnormal calcium signaling, which can exacerbate mitochondrial dysfunction, amplifying ROS production.
ROS overproduction is central to the mechanism of action of PDT as an anti-cancer treatment [144,159]. PDT is a non-invasive therapeutic approach that uses 1. a light-sensitive compound (non-toxic until activated by light) known as photosensitizers, which is administered to the patient and accumulates preferentially in cancer cells; 2. light exposure (specific wavelength of light, usually in the visible or near-infrared spectrum) required to activate the photosensitizer; and 3. molecular oxygen present in the tissue, to generate ROS, leading to the selective destruction of cancer cells. The most significant ROS produced in PDT is 1O2, O2, OH, and hydrogen peroxide (H2O2), which can also be generated depending on the environment and the type of photosensitizer used.
The advantages of PDT consist of a. selective targeting since the production of ROS is localized to the site of light exposure, allowing for precise targeting of the tumor while minimizing damage to surrounding healthy tissues, b. low risks of systemic toxicity since the photosensitizer remains relatively inert until activated by light, and c. efficiency in overcoming tumor resistance to conventional treatments since the types of cellular damage caused by ROS in PDT are different from those caused by traditional chemotherapy or radiotherapy, which makes PDT a powerful tool in cancer therapy, especially for surface or accessible tumors. PDT is mainly used for localized tumors and is particularly effective for certain types of cancers and non-cancerous conditions, such as skin cancers (basal cell carcinoma and squamous cell carcinoma in situ), pre-cancerous skin conditions (actinic keratosis), early-stage esophageal cancer, early-stage bladder cancer, and cervical intraepithelial neoplasia (CIN) [160]. However, challenges such as oxygen dependence and light penetration still need to be addressed to fully realize the potential of PDT in broader clinical applications.
SDT is an emerging cancer treatment that combines ultrasound with a sonosensitizer to produce ROS and induce selective tumor cell death [145]. As in the case of PDT, SDT relies on ROS as the primary agents of cytotoxicity, but it uses ultrasound, typically in the low-frequency range (20 kHz to 3 MHz), instead of light to activate the sensitizer, a compound that is sensitive to ultrasound, such as Hematoporphyrin Derivatives (HPDs) or Protoporphyrin IX (PpIX) or doxorubicin, which is administered to the patient. The ultrasound waves activate the sonosensitizer, leading to the generation of ROS such as singlet oxygen and free radicals, and cause damage to cellular components like DNA, proteins, and lipids, ultimately leading to cancer cell death. The selective accumulation of sonosensitizers in tumor tissues, combined with the localized application of ultrasound, allows for targeted cancer treatment with minimal damage to surrounding healthy tissues.
The advantage of SDT lies in its ability to treat deeply located tumors. On the other hand, photodynamic therapy uses visible light, which attenuates rapidly in tissues, has limited penetration, can be employed only superficially or intra-operatively, and is not as effective as therapy in the deeper regions of the tumor [145]. SDT recently emerged as a promising noninvasive alternative to chemotherapy and radiation therapy in the treatment of aggressive gliomas. Studies have suggested that coupling sonosensitizers such as 5-aminolevulinic acid, hematoporphyrin monomethyl ether, and sinoporphyrin sodium with focused ultrasound induces robust cytotoxic activity in tumor cells, in vitro and in vivo. These effects are likely mediated by the oxidative stress induced by reactive oxygen species production, apoptotic signaling cascades, and intracellular calcium overload [161].
SDT is currently under investigation in preclinical and early clinical studies. Researchers are focusing on optimizing sonosensitizer formulations, improving ultrasound delivery methods, and understanding the mechanisms of ROS generation and tumor destruction. Some sonosensitizers, such as porphyrins and nanoparticles, are being tested for their efficacy in specific cancer types, including hepatocellular carcinoma and pancreatic adenocarcinoma, which are often resistant to conventional therapies [162].

9. Understanding the Mechanisms Underlying the Oxidative Stress–Aging–Carcinogenesis Relationships to Fight Age-Related Tumors

Geroscience is only beginning to inform oncology of the relationship between oxidative stress, aging, and cancer. It has been established that oxidative stress, in the form of excess ROS or RNS, promotes both aging and cancer development by causing structural damage in intracellular proteins and lipids and by mutating nuclear or mitochondrial DNA. Prostate, breast, lung, and colorectal cancer incidence increases with age. However, there is epidemiological evidence that cancer incidence and aggressiveness decrease after the age of 80 [163,164], contradicting the aging–cancer paradigm and raising questions that need to be urgently addressed given the worldwide aging population and the demand for healthy aging.

9.1. Cancers Associated with Oxidative Stress and Aging

Lung Cancer. The lung is constantly exposed to higher oxygen pressures compared to other tissues [165], therefore making it a major target for air pollutants, many of which are electrophilic molecules that can induce ROS production [166,167]. Cigarette smoke is a major source of oxidative stress [168] and can significantly increase the production of ROS in the lung parenchyma, which can, in turn, induce IL-8 release and SNAIL1 expression in cancer cells, leading to the epithelial–mesenchymal transition, which promotes tumor progression [169].
The accumulation of harmful peroxides and lipid peroxidation products in the lung parenchyma is also promoted by the impairment of antioxidant defense mechanisms [170]. In squamous cell lung carcinoma and adenocarcinoma, the activity of glutathione peroxidase and catalase is significantly decreased, compared to normal lung tissue, and this is accompanied by a significant increase in reactive aldehydes [170], which can induce high levels of oxidative stress.
The presence of massive oxidative damage in lung cancer patients is also demonstrated by the elevated levels of oxidative stress markers, such as protein carbonylation and malondialdehyde (MDA)-protein adducts, in the bronchial epithelium and peripheral blood [171].
Breast Cancer. Oxidative stress has been implicated in breast carcinogenesis in relation to age-related hormonal changes [172] and environmental factors [173]. In particular, the relationship between estrogen metabolism and oxidative stress in breast cancer involves various mechanisms and pathways.
Estrogens can increase mitochondrial ROS production by down-regulating uncoupling proteins (UCPs), leading to oxidative stress in estrogen receptor (ER)-positive breast cancer cells [174,175]. This oxidative stress is implicated in the onset and progression of breast cancer through the formation of DNA adducts and subsequent mutations [176,177,178]. In addition, chronic oxidative stress can lead to epigenetic changes, such as the inactivation of the ERα gene, contributing to the development of more aggressive, estrogen-independent breast cancer subtypes [179].
Colorectal Cancer. Diet, chronic inflammation, and gut microbiota dysbiosis [180] can contribute to oxidative stress in the colon. ROS-induced DNA damage in colonic epithelial cells is a critical step in colorectal cancer development [181].
Epidemiological and experimental studies have proven that a high-fat diet is an important risk factor for colon cancer [182], particularly due to the interactions between dietary fats, gut microbiota, and inflammatory processes. High-fat diets can promote obesity, resulting in insulin resistance and inflammation and the development of oxidative stress, increased cell proliferation, and suppression of apoptosis [182].
Increased levels of oxidative DNA damage markers, such as 8-hydroxy-2′-deoxyguanosine (8-OHdG), are observed in colon cancer tissues, compared to normal colonic mucosa [183]. In addition, the altered function of enzymes involved in the repair of oxidative DNA damage, e.g., 8-oxoguanine DNA N-glycosylase 1 (hOGG1) and mutY DNA glycosylase (MUTYH), results in the accumulation of DNA mutations and contributes to the adenoma-adenocarcinoma transition [184].
The gut microbiota can also be involved in the development of oxidative stress since the microbial expression of ROS-scavenging enzymes is compromised in cancer. This evidence suggests that the gut microbiota, under normal conditions, may protect the colonic mucosa from oxidative stress and that the impairment of this activity may contribute to the onset and progression of colon cancer [185].
Prostate Cancer. Oxidative stress plays a role in the initiation and progression of prostate cancer. Age-related increases in ROS production and decreased antioxidant defenses are contributing factors [186].
The serum levels of protein oxidation and lipid peroxidation products increase in prostate cancer patients with a Gleason score >7 compared to patients with a Gleason score <7, and the presence of perineural invasion significantly is associated with an increase in the concentration of protein oxidation products in the urine [187]. These results suggest a significant role for oxidative damage in prostate cancer onset and progression [187]. Furthermore, oxidative stress can induce, in prostate cancer cells, the expression of genes such as Lanthionine synthase C-like protein 1 (LanCL1) [188], which promotes tumor growth and progression, and protects cells from damage caused by oxidative stress resulting in resistance to therapies [188].
Liver Cancer. Conditions such as hepatitis, alcohol abuse, and non-alcoholic fatty liver disease can lead to chronic oxidative stress in the liver. This persistent oxidative damage promotes hepatocellular carcinoma [189,190].
Due to their high metabolic activity, hepatocytes are characterized by a high number of mitochondria that serve as sources of ROS. ROS overproduction is a common occurrence in chronic liver diseases and leads to DNA damage and inflammatory infiltration, which collectively exacerbate liver damage and promote malignant transformation [191,192]. Analyses of serum markers and histological findings indicate that oxidative stress increases the likelihood of developing hepatocarcinoma [193]. Oxidative damage can also promote the progression of hepatocellular carcinoma since it can induce the degradation of E-cadherin, mediated by ring finger protein 25 (RNF25), which is associated with tumor metastases [194].
Skin Cancer. As the greatest defense organ of the body, the skin is continuously exposed to endogenous and external factors that can induce the production of ROS [195]. Ultraviolet (UV) radiation from the sun is the primary source of ROS in skin cells. UV-induced oxidative stress is a major cause of skin cancers, including melanoma [196] and non-melanoma skin cancers [197].
Two main mechanisms underlie oxidative stress-induced skin cancers: 1. the direct degradation of proteins, DNA, and lipids by ROS, and 2. the alteration of cell signaling pathways, such as MAPK and JAK/STAT [195]. ROS can be involved in all stages of carcinogenesis, including initiation, promotion, and progression [198]. Oxidative stress parameters (superoxide anion radical levels and manganese superoxide dismutase and catalase activity) are significantly higher in melanoma patients than in healthy controls and increase with the disease progression, achieving the maximum in stage IV [199]. Patients with nonmelanoma skin cancer have high blood levels of all oxidative stress biomarkers compared to healthy subjects [200].
It has been recently demonstrated that Staphylococcus aureus strains isolated from biopsies of cutaneous squamous cell carcinoma can secrete molecules, which can have cancer-promoting effects as they are able to induce the ROS production and DNA damage in keratinocytes [201].

9.2. Strategies to Mitigate Oxidative Stress or Improve Antioxidant Defenses

Current strategies to combat oxidative stress focus on reducing the production of ROS, enhancing antioxidant defenses, and repairing or removing damaged molecules. These strategies span from lifestyle modifications to pharmaceutical interventions and emerging biotechnologies.
Lifestyle Modifications. Consuming a diet rich in natural antioxidants helps neutralize ROS. Foods rich in vitamins C and E, carotenoids (e.g., beta-carotene, lycopene), polyphenols (e.g., flavonoids in fruits and vegetables), and minerals like selenium and zinc play a key role in reducing oxidative stress. A diet rich in fruits, vegetables, and whole grains can enhance the body’s antioxidant defenses [202]. Supplements or compounds that enhance levels of endogenous antioxidants, such as a. N-acetylcysteine (NAC), which boosts glutathione production and can be administered orally (typically given over 72 h) or intravenously (given over 20–24 h), is particularly useful in treating acetaminophen (paracetamol) overdose and hepatotoxicity. Its mucolytic and antioxidant properties make it useful in managing respiratory illness, liver diseases [203], and psychiatric disorders [204]; b. Coenzyme Q10, also known as ubiquinone, supports mitochondrial antioxidant defenses and is often used as an adjunctive therapy in patients with heart failure, hypertension, ischemic heart disease, and neurological disorders for its potential neuroprotective effects [205].
Caloric restriction has been shown to reduce oxidative stress and extend lifespan in a variety of organisms [206,207], decreasing the overall metabolic rate and improving mitochondrial efficiency, meaning that less ROS is produced per unit of energy generated. Regular physical activity and avoiding smoking [166] and excessive alcohol consumption [208] can reduce oxidative stress. Limiting exposure to environmental pollutants (e.g., air pollution, heavy metals) and UV radiation can reduce oxidative stress by minimizing external sources of ROS.
Pharmacological Interventions. Drugs that mimic natural antioxidants or neutralize ROS directly are being developed. For example, edaravone, also known by its chemical name MCI-186, is a free-radical scavenger used as a neuroprotective agent to treat oxidative stress-related conditions like acute ischemic stroke and ALS [209,210]. Edaravone is administered intravenously, which ensures high bioavailability, and it is rapidly metabolized in the liver and excreted through the kidneys, primarily as glucuronide and sulfate conjugates. For the treatment of ALS, edaravone is administered as an i.v. infusion at a dose of 60 mg per day for 10–14 days, followed by a break, and then a continuation with a similar on–off cycle. In the treatment of acute ischemic stroke, edaravone is given at 30 mg twice daily for 14 days, proving more effective when given within 24 h of stroke onset. Although edaravone is primarily an antioxidant, excessive dosing or specific biochemical conditions (e.g., metal ion interactions since edaravone can interact with metal ions such as iron and copper, potentially catalyzing redox reactions and more free radical production) could result in pro-oxidant effects. Careful monitoring and personalized dosing strategies can help mitigate the risks associated with promoting oxidation.
Due to its antioxidant and anti-inflammatory properties, edaravone has also been investigated in preclinical models for its potential activities in treating cancer [211].
Additional drugs targeting oxidative stress pathways are being investigated for cancer prevention and treatment, including inhibitors of ROS-producing enzymes, such as NOX Inhibitors [212], Cyclooxygenase (COX) Inhibitors [213], Lipoxygenase (LOX) Inhibitors [214], and agents that boost endogenous antioxidant defenses [215]. Since chronic inflammation is often associated with increased ROS production, anti-inflammatory drugs such as nonsteroidal anti-inflammatory drugs, NSAIDs, and corticosteroids can indirectly reduce oxidative stress by curbing inflammation.
Gene and Molecular Therapy. These biotechnological approaches aim to enhance the expression of endogenous antioxidant enzymes like SOD, catalase, and GPx to reduce oxidative stress. Gene editing tools like CRISPR/Cas9 and RNA-based therapies, e.g., Small Interfering RNA (siRNA) and antisense oligonucleotides (ASOs) are being explored to specifically target genes involved in oxidative stress pathways, potentially offering new therapeutic approaches for diseases associated with oxidative damage.
  • The CRISPR/Cas9 system allows for precise editing of genes that contribute to oxidative stress, such as Kelch-like ECH-associated protein 1 (Keap1), which regulates the activity of Nrf2 [216] and other redox regulators. CRISPR has been used to knock out mutant forms of proteins that lead to oxidative damage, offering a potential cure for neurodegenerative diseases (Alzheimer’s, Parkinson’s, and ALS), cardiovascular disorders, and cancer.
     In neurodegenerative diseases, CRISPR has been used to selectively knock out the mutant form of the huntingtin (HTT) gene, reducing the toxic protein burden and mitigating oxidative stress in neuronal cells [217]. Mutations in the SOD1 gene are implicated in familial ALS. CRISPR/Cas9 has been used to knock out the mutant SOD1 gene in cellular and animal models of ALS, reducing oxidative stress and extending survival in preclinical studies [218]. Mutations in the PARK7 (DJ-1) gene are associated with oxidative damage in Parkinson’s Disease. CRISPR/Cas9 has been employed to knock out these mutant genes or restore normal gene function, reducing oxidative stress and improving cellular survival in experimental models [219].
     In cardiovascular diseases, including atherosclerosis and ischemia-reperfusion injury, which are characterized by oxidative stress-induced damage to the vascular endothelium and heart tissue, CRISPR/Cas9 has been used to target genes that contribute to ROS production or impair antioxidant defenses. CRISPR/Cas9-mediated knockout of the PCSK9 gene, which plays a role in cholesterol metabolism and oxidative stress in vascular cells, has been explored as a therapeutic strategy to reduce oxidative damage in atherosclerosis, with promising results in animal models [220].
     In the oncology field, CRISPR/Cas9 is being used to target genes that exacerbate oxidative damage in cancer cells, either by knocking out pro-oxidant genes or modulating antioxidant defenses. CRISPR/Cas9-mediated knockout mutant forms of the TP53 gene restore normal redox balance and reduce oxidative damage in cancer cells [221]. Glutathione (GSH) is a major antioxidant in cells. CRISPR/Cas9 has been used to knock out genes like the Glutamate–cysteine ligase catalytic subunit, GCLC (a key enzyme in glutathione synthesis), in Acute myeloid leukemia (AML) and solid tumors [222] to increase their sensitivity to oxidative damage, making them more susceptible to chemotherapy and radiotherapy.
  • siRNA or ASOs can silence genes involved in ROS production or modulate antioxidant pathways. NOX enzymes are key contributors to ROS production. siRNA or ASOs targeting NOX subunits can reduce ROS levels in various cell types. siRNA-targeting NOX2 has shown promise in reducing oxidative stress in models of cardiovascular disease and neuroinflammation [223,224,225].
     The mechanistic target of rapamycin (mTOR) pathway regulates various cellular processes, including oxidative stress. siRNA-targeting components of the mTOR pathway, such as mTOR itself or downstream effectors, can modulate ROS production in cancer and metabolic disorders [226,227]. siRNA or ASOs targeting negative regulators of SOD (e.g., SOD1 mutations in ALS) aim to enhance SOD activity indirectly by silencing genes that inhibit its function [228,229].
     GPx enzymes reduce hydrogen peroxide to water. siRNA targeting genes involved in the regulation or production of GPx (e.g., GPx4) [230] can enhance the cellular antioxidant capacity in models of cancer and neurodegenerative diseases [231,232].
     Nrf2 is a key transcription factor that regulates antioxidant gene expression. siRNA can target negative regulators of Nrf2 (e.g., Keap1) to increase the activity of Nrf2 and enhance antioxidant responses. In diseases where Nrf2 is dysfunctional, such as autoimmune (Multiple Sclerosis), chronic inflammatory diseases (Rheumatoid Arthritis), and neurodegenerative disorders [233], siRNA can help restore its normal function.
     While gene and molecular therapies hold significant promise in developing therapies for oxidative stress, further studies are needed to overcome challenges related to delivery, immune responses, off-target effects, and safety concerns such as the risk of insertional mutagenesis or unintended gene activation.
Microbiome Modulation. The gut microbiome plays a pivotal role in modulating oxidative stress through various mechanisms, including the production of antioxidant compounds, regulation of inflammation, and maintenance of mitochondrial health. Dysbiosis can lead to the overproduction of ROS by pathogenic bacteria and the immune system, increasing oxidative stress [234]. A healthy and balanced gut microbiome can help reduce systemic oxidative stress and protect against a range of chronic diseases. Certain probiotic strains, such as Lactobacillus rhamnosus and Bifidobacterium breve, have been shown to reduce oxidative stress by enhancing antioxidant defenses and reducing pro-inflammatory cytokines [235,236]. Dietary fibers that act as prebiotics (e.g., inulin, fructooligosaccharides) promote the growth of beneficial bacteria that produce SCFAs, thereby enhancing antioxidant defenses and reducing oxidative stress [237,238,239].
Overall, oxidative stress is a significant factor in the development and progression of age-related cancers, and its modulation could improve anti-tumor treatments [240]. While antioxidant therapies and lifestyle modifications hold promise, ongoing research is crucial to fully elucidate the complex relationship between oxidative stress, aging, and cancer to reduce the incidence and impact of age-related malignancies.

Author Contributions

Conceptualization, E.D.C.; writing—original draft preparation, E.D.C.; writing—review and editing, E.D.C. and C.S.; formal analysis, E.D.C. and C.S.; data curation, C.S. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by: AIRC: IG 2019—ID. 23264 project, P.I. Emma Di Carlo; European Union—NextGenerationEU, program “MUR-Fondo Promozione e Sviluppo—DM 737/2021”, acronym of the project “TAILOR-30”, P.I. Emma Di Carlo; Ministero della Salute, PNRR: M6/C2_CALL 2022 Full Proposal, ID: PNRR-MAD-2022-12375909, to Emma Di Carlo.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Flohé, L. Looking Back at the Early Stages of Redox Biology. Antioxidants 2020, 9, 1254. [Google Scholar] [CrossRef] [PubMed]
  2. Auger, C.; Lemire, J.; Cecchini, D.; Bignucolo, A.; Appanna, V.D. The metabolic reprogramming evoked by nitrosative stress triggers the anaerobic utilization of citrate in Pseudomonas fluorescens. PLoS ONE 2011, 6, e28469. [Google Scholar] [CrossRef] [PubMed]
  3. Sies, H.; Berndt, C.; Jones, D.P. Oxidative Stress. Annu. Rev. Biochem. 2017, 86, 715–748. [Google Scholar] [CrossRef]
  4. Sies, H. Oxidative Stress: Concept and Some Practical Aspects. Antioxidants 2020, 9, 852. [Google Scholar] [CrossRef]
  5. Jomova, K.; Raptova, R.; Alomar, S.Y.; Alwasel, S.H.; Nepovimova, E.; Kuca, K.; Valko, M. Reactive oxygen species, toxicity, oxidative stress, and antioxidants: Chronic diseases and aging. Arch. Toxicol. 2023, 97, 2499–2574. [Google Scholar] [CrossRef]
  6. Liguori, I.; Russo, G.; Curcio, F.; Bulli, G.; Aran, L.; Della-Morte, D.; Gargiulo, G.; Testa, G.; Cacciatore, F.; Bonaduce, D.; et al. Oxidative stress, aging, and diseases. Clin. Interv. Aging 2018, 13, 757–772. [Google Scholar] [CrossRef]
  7. Maldonado, E.; Morales-Pison, S.; Urbina, F.; Solari, A. Aging Hallmarks and the Role of Oxidative Stress. Antioxidants 2023, 12, 651. [Google Scholar] [CrossRef]
  8. Li, X.; Li, C.; Zhang, W.; Wang, Y.; Qian, P.; Huang, H. Inflammation and aging: Signaling pathways and intervention therapies. Signal Transduct. Target. Ther. 2023, 8, 239. [Google Scholar] [CrossRef] [PubMed]
  9. Holmström, K.M.; Finkel, T. Cellular mechanisms and physiological consequences of redox-dependent signalling. Nat. Rev. Mol. Cell Biol. 2014, 15, 411–421. [Google Scholar] [CrossRef]
  10. Halliwell, B. Understanding mechanisms of antioxidant action in health and disease. Nat. Rev. Mol. Cell Biol. 2024, 25, 13–33. [Google Scholar] [CrossRef]
  11. Bedard, K.; Krause, K.H. The NOX family of ROS-generating NADPH oxidases: Physiology and pathophysiology. Physiol. Rev. 2007, 87, 245–313. [Google Scholar] [CrossRef] [PubMed]
  12. Brown, D.I.; Griendling, K.K. Nox proteins in signal transduction. Free Radic. Biol. Med. 2009, 47, 1239–1253. [Google Scholar] [CrossRef]
  13. Calcerrada, P.; Peluffo, G.; Radi, R. Nitric oxide-derived oxidants with a focus on peroxynitrite: Molecular targets, cellular responses and therapeutic implications. Curr. Pharm. Des. 2011, 17, 3905–3932. [Google Scholar] [CrossRef] [PubMed]
  14. Kar, S.; Kavdia, M. Endothelial NO and O2 production rates differentially regulate oxidative, nitroxidative, and nitrosative stress in the microcirculation. Free Radic. Biol. Med. 2013, 63, 161–174. [Google Scholar] [CrossRef]
  15. Wisastra, R.; Dekker, F.J. Inflammation, Cancer and Oxidative Lipoxygenase Activity are Intimately Linked. Cancers 2014, 6, 1500–1521. [Google Scholar] [CrossRef] [PubMed]
  16. Veith, A.; Moorthy, B. Role of cytochrome p450s in the generation and metabolism of reactive oxygen species. Curr. Opin. Toxicol. 2018, 7, 44–51. [Google Scholar] [CrossRef]
  17. Hrycay, E.G.; Bandiera, S.M. Involvement of Cytochrome P450 in Reactive Oxygen Species Formation and Cancer. Adv. Pharmacol. 2015, 74, 35–84. [Google Scholar] [CrossRef]
  18. Seychell, B.C.; Vella, M.; Hunter, G.J.; Hunter, T. The Good and the Bad: The Bifunctional Enzyme Xanthine Oxidoreductase in the Production of Reactive Oxygen Species. In Reactive Oxygen Species—Advances and Developments; IntechOpen: London, UK, 2024. [Google Scholar] [CrossRef]
  19. Li, H.S.; Zhou, Y.N.; Li, L.; Li, S.F.; Long, D.; Chen, X.L.; Zhang, J.B.; Feng, L.; Li, Y.P. HIF-1α protects against oxidative stress by directly targeting mitochondria. Redox Biol. 2019, 25, 101109. [Google Scholar] [CrossRef]
  20. Luo, Z.; Tian, M.; Yang, G.; Tan, Q.; Chen, Y.; Li, G.; Zhang, Q.; Li, Y.; Wan, P.; Wu, J. Hypoxia signaling in human health and diseases: Implications and prospects for therapeutics. Signal Transduct. Target. Ther. 2022, 7, 218. [Google Scholar] [CrossRef]
  21. Wang, X.X.; Chen, W.Z.; Li, C.; Xu, R.S. Current potential pathogenic mechanisms of copper-zinc superoxide dismutase 1 (SOD1) in amyotrophic lateral sclerosis. Rev. Neurosci. 2024, 35, 549–563. [Google Scholar] [CrossRef]
  22. Abati, E.; Bresolin, N.; Comi, G.; Corti, S. Silence superoxide dismutase 1 (SOD1): A promising therapeutic target for amyotrophic lateral sclerosis (ALS). Expert Opin. Ther. Targets 2020, 24, 295–310. [Google Scholar] [CrossRef] [PubMed]
  23. Malik, S.; Waquar, S.; Idrees, N.; Malik, A. Impending role of inflammatory markers and their specificity and sensitivity in breast cancer patients. Sci. Rep. 2024, 14, 15117. [Google Scholar] [CrossRef] [PubMed]
  24. Nandi, A.; Yan, L.J.; Jana, C.K.; Das, N. Role of Catalase in Oxidative Stress- and Age-Associated Degenerative Diseases. Oxidative Med. Cell. Longev. 2019, 2019, 9613090. [Google Scholar] [CrossRef] [PubMed]
  25. Pei, J.; Pan, X.; Wei, G.; Hua, Y. Research progress of glutathione peroxidase family (GPX) in redoxidation. Front. Pharmacol. 2023, 14, 1147414. [Google Scholar] [CrossRef] [PubMed]
  26. Couto, N.; Wood, J.; Barber, J. The role of glutathione reductase and related enzymes on cellular redox homoeostasis network. Free Radic. Biol. Med. 2016, 95, 27–42. [Google Scholar] [CrossRef]
  27. Yang, Y.; Liu, Z.; Wu, J.; Bao, S.; Wang, Y.; Li, J.; Song, T.; Sun, Y.; Pi, J. Nrf2 Mitigates RANKL and M-CSF Induced Osteoclast Differentiation via ROS-Dependent Mechanisms. Antioxidants 2023, 12, 2094. [Google Scholar] [CrossRef]
  28. Kim, J.; Cha, Y.N.; Surh, Y.J. A protective role of nuclear factor-erythroid 2-related factor-2 (Nrf2) in inflammatory disorders. Mutat. Res. 2010, 690, 12–23. [Google Scholar] [CrossRef]
  29. Chiang, S.K.; Chen, S.E.; Chang, L.C. The Role of HO-1 and Its Crosstalk with Oxidative Stress in Cancer Cell Survival. Cells 2021, 10, 2401. [Google Scholar] [CrossRef]
  30. Gozzelino, R.; Jeney, V.; Soares, M.P. Mechanisms of cell protection by heme oxygenase-1. Annu. Rev. Pharmacol. Toxicol. 2010, 50, 323–354. [Google Scholar] [CrossRef]
  31. Schieber, M.; Chandel, N.S. ROS function in redox signaling and oxidative stress. Curr. Biol. 2014, 24, R453–R462. [Google Scholar] [CrossRef]
  32. Weng, M.S.; Chang, J.H.; Hung, W.Y.; Yang, Y.C.; Chien, M.H. The interplay of reactive oxygen species and the epidermal growth factor receptor in tumor progression and drug resistance. J. Exp. Clin. Cancer Res. 2018, 37, 61. [Google Scholar] [CrossRef] [PubMed]
  33. Lee, H.-C.; An, S.; Lee, H.; Woo, S.-H.; Jin, H.-O.; Seo, S.-K.; Choe, T.-B.; Yoo, D.-H.; Lee, S.-J.; Hong, Y.-J.; et al. Activation of epidermal growth factor receptor and its downstream signaling pathway by nitric oxide in response to ionizing radiation. Mol. Cancer Res. 2008, 6, 996–1002. [Google Scholar] [CrossRef]
  34. Movafagh, S.; Crook, S.; Vo, K. Regulation of hypoxia-inducible factor-1a by reactive oxygen species: New developments in an old debate. J. Cell. Biochem. 2015, 116, 696–703. [Google Scholar] [CrossRef]
  35. Zhang, J.; Wang, X.; Vikash, V.; Ye, Q.; Wu, D.; Liu, Y.; Dong, W. ROS and ROS-mediated cellular signaling. Oxidative Med. Cell. Longev. 2016, 2016, 4350965. [Google Scholar] [CrossRef] [PubMed]
  36. Kohlgrüber, S.; Upadhye, A.; Dyballa-Rukes, N.; McNamara, C.A.; Altschmied, J. Regulation of transcription factors by reactive oxygen species and nitric oxide in vascular physiology and pathology. Antioxid. Redox Signal. 2017, 26, 679–699. [Google Scholar] [CrossRef] [PubMed]
  37. Milkovic, L.; Cipak Gasparovic, A.; Cindric, M.; Mouthuy, P.A.; Zarkovic, N. Short overview of ROS as cell function regulators and their implications in therapy concepts. Cells 2019, 8, 793. [Google Scholar] [CrossRef] [PubMed]
  38. Guo, C.L. Self-sustained regulation or self-perpetuating dysregulation: ROS-dependent HIF-YAP-Notch signaling as a double-edged sword on stem cell physiology and tumorigenesis. Front. Cell Dev. Biol. 2022, 10, 862791. [Google Scholar] [CrossRef]
  39. Bertero, E.; Maack, C. Calcium signaling and reactive oxygen species in mitochondria. Circ. Res. 2018, 122, 1460–1478. [Google Scholar] [CrossRef]
  40. De Nicolo, B.; Cataldi-Stagetti, E.; Diquigiovanni, C.; Bonora, E. Calcium and reactive oxygen species signaling interplays in cardiac physiology and pathologies. Antioxidants 2023, 12, 353. [Google Scholar] [CrossRef]
  41. Redza-Dutordoir, M.; Averill-Bates, D.A. Activation of apoptosis signalling pathways by reactive oxygen species. Biochim. Biophys. Acta 2016, 1863, 2977–2992. [Google Scholar] [CrossRef]
  42. Redza-Dutordoir, M.; Averill-Bates, D.A. Interactions between reactive oxygen species and autophagy: Special issue: Death mechanisms in cellular homeostasis. Biochim. Biophys. Acta Mol. Cell Res. 2021, 1868, 119041. [Google Scholar] [CrossRef] [PubMed]
  43. Liu, S.; Huang, B.; Cao, J.; Wang, Y.; Xiao, H.; Zhu, Y.; Zhang, H. ROS fine-tunes the function and fate of immune cells. Int. Immunopharmacol. 2023, 119, 110069. [Google Scholar] [CrossRef]
  44. Morris, G.; Gevezova, M.; Sarafian, V.; Maes, M. Redox regulation of the immune response. Cell. Mol. Immunol. 2022, 19, 1079–1101. [Google Scholar] [CrossRef] [PubMed]
  45. Asehnoune, K.; Strassheim, D.; Mitra, S.; Kim, J.Y.; Abraham, E. Involvement of reactive oxygen species in Toll-like receptor 4-dependent activation of NF-kappa B. J. Immunol. 2004, 172, 2522–2529. [Google Scholar] [CrossRef]
  46. Pushpakumar, S.; Ren, L.; Kundu, S.; Gamon, A.; Tyagi, S.C.; Sen, U. Toll-like receptor 4 deficiency reduces oxidative stress and macrophage mediated inflammation in hypertensive kidney. Sci. Rep. 2017, 7, 6349. [Google Scholar] [CrossRef]
  47. Montégut, L.; López-Otín, C.; Kroemer, G. Aging and cancer. Mol. Cancer 2024, 23, 106. [Google Scholar] [CrossRef] [PubMed]
  48. Hahm, J.Y.; Park, J.; Jang, E.S.; Chi, S.W. 8-Oxoguanine: From oxidative damage to epigenetic and epitranscriptional modification. Exp. Mol. Med. 2022, 54, 1626–1642. [Google Scholar] [CrossRef]
  49. Liu, B.; Yip RKh Zhou, Z. Chromatin remodeling, DNA damage repair and aging. Curr. Genom. 2012, 13, 533–547. [Google Scholar] [CrossRef]
  50. Yu, C.; Asadian, S.; Tigano, M. Molecular and cellular consequences of mitochondrial DNA double-stranded breaks. Hum. Mol. Genet. 2024, 33, R12–R18. [Google Scholar] [CrossRef]
  51. Srivastava, S. The Mitochondrial Basis of Aging and Age-Related Disorders. Genes 2017, 8, 398. [Google Scholar] [CrossRef]
  52. Lin, J.; Epel, E. Stress and telomere shortening: Insights from cellular mechanisms. Ageing Res. Rev. 2022, 73, 101507. [Google Scholar] [CrossRef]
  53. von Zglinicki, T. Oxidative stress shortens telomeres. Trends Biochem. Sci. 2002, 27, 339–344. [Google Scholar] [CrossRef] [PubMed]
  54. Barnes, R.P.; Fouquerel, E.; Opresko, P.L. The impact of oxidative DNA damage and stress on telomere homeostasis. Mech. Ageing Dev. 2019, 177, 37–45. [Google Scholar] [CrossRef] [PubMed]
  55. Tomaska, L.; Nosek, J.; Kar, A.; Willcox, S.; Griffith, J.D. A new view of the T-loop junction: Implications for self-primed telomere extension, expansion of disease-related nucleotide repeat blocks, and telomere evolution. Front. Genet. 2019, 10, 792. [Google Scholar] [CrossRef]
  56. Alanazi, A.F.R.; Parkinson, G.N.; Haider, S. Structural motifs at the telomeres and their role in regulatory pathways. Biochemistry 2024, 63, 827–842. [Google Scholar] [CrossRef]
  57. Cuanalo-Contreras, K.; Schulz, J.; Mukherjee, A.; Park, K.W.; Armijo, E.; Soto, C. Extensive accumulation of misfolded protein aggregates during natural aging and senescence. Front. Aging Neurosci. 2023, 14, 1090109. [Google Scholar] [CrossRef] [PubMed]
  58. Gu, L.; Liu, M.; Zhang, Y.; Zhou, H.; Wang, Y.; Xu, Z.X. Telomere-related DNA damage response pathways in cancer therapy: Prospective targets. Front. Pharmacol. 2024, 15, 1379166. [Google Scholar] [CrossRef] [PubMed]
  59. Lévy, E.; El Banna, N.; Baïlle, D.; Heneman-Masurel, A.; Truchet, S.; Rezaei, H.; Huang, M.-E.; Béringue, V.; Martin, D.; Vernis, L. Causative links between protein aggregation and oxidative stress: A review. Int. J. Mol. Sci. 2019, 20, 3896. [Google Scholar] [CrossRef] [PubMed]
  60. Guang, M.H.Z.; Kavanagh, E.L.; Dunne, L.P.; Dowling, P.; Zhang, L.; Lindsay, S.; Bazou, D.; Goh, C.Y.; Hanley, C.; Bianchi, G.; et al. Targeting Proteotoxic Stress in Cancer: A Review of the Role that Protein Quality Control Pathways Play in Oncogenesis. Cancers 2019, 11, 66. [Google Scholar] [CrossRef]
  61. Leonardi, G.C.; Accardi, G.; Monastero, R.; Nicoletti, F.; Libra, M. Ageing: From inflammation to cancer. Immun. Ageing 2018, 15, 1. [Google Scholar] [CrossRef]
  62. Hou, C.; Wang, Z.; Lu, X. Impact of immunosenescence and inflammaging on the effects of immune checkpoint inhibitors. Cancer Pathog. Ther. 2023, 2, 24–30. [Google Scholar] [CrossRef] [PubMed]
  63. Finkel, T.; Serrano, M.; Blasco, M.A. The common biology of cancer and ageing. Nature 2007, 448, 767–774. [Google Scholar] [CrossRef] [PubMed]
  64. Aman, Y.; Schmauck-Medina, T.; Hansen, M.; Morimoto, R.I.; Simon, A.K.; Bjedov, I.; Palikaras, K.; Simonsen, A.; Johansen, T.; Tavernarakis, N.; et al. Autophagy in healthy aging and disease. Nat. Aging 2021, 1, 634–650. [Google Scholar] [CrossRef]
  65. Kozakiewicz, M.; Kornatowski, M.; Krzywińska, O.; Kędziora-Kornatowska, K. Changes in the blood antioxidant defense of advanced age people. Clin. Interv. Aging 2019, 14, 763–771. [Google Scholar] [CrossRef]
  66. Tan, B.L.; Norhaizan, M.E.; Liew, W.P.; Sulaiman Rahman, H. Antioxidant and Oxidative Stress: A Mutual Interplay in Age-Related Diseases. Front. Pharmacol. 2018, 9, 1162. [Google Scholar] [CrossRef]
  67. Campisi, J. Aging, cellular senescence, and cancer. Annu. Rev. Physiol. 2013, 75, 685–705. [Google Scholar] [CrossRef] [PubMed]
  68. Calcinotto, A.; Kohli, J.; Zagato, E.; Pellegrini, L.; Demaria, M.; Alimonti, A. Cellular Senescence: Aging, Cancer, and Injury. Physiol. Rev. 2019, 99, 1047–1078. [Google Scholar] [CrossRef]
  69. Benz, C.C.; Yau, C. Ageing, oxidative stress and cancer: Paradigms in parallax. Nat. Rev. Cancer 2008, 8, 875–879. [Google Scholar] [CrossRef]
  70. Kontomanolis, E.N.; Koutras, A.; Syllaios, A.; Schizas, D.; Mastoraki, A.; Garmpis, N.; Diakosavvas, M.; Angelou, K.; Tsatsaris, G.; Pagkalos, A.; et al. Role of oncogenes and tumor-suppressor genes in carcinogenesis: A review. Anticancer Res. 2020, 40, 6009–6015. [Google Scholar] [CrossRef] [PubMed]
  71. Dakal, T.C.; Dhabhai, B.; Pant, A.; Moar, K.; Chaudhary, K.; Yadav, V.; Ranga, V.; Sharma, N.K.; Kumar, A.; Maurya, P.K.; et al. Oncogenes and tumor suppressor genes: Functions and roles in cancers. MedComm 2024, 5, e582. [Google Scholar] [CrossRef]
  72. Maynard, S.; Schurman, S.H.; Harboe, C.; de Souza-Pinto, N.C.; Bohr, V.A. Base excision repair of oxidative DNA damage and association with cancer and aging. Carcinogenesis 2009, 30, 2–10. [Google Scholar] [CrossRef] [PubMed]
  73. Chen, W.; Balakrishnan, K.; Kuang, Y.; Han, Y.; Fu, M.; Gandhi, V.; Peng, X. Reactive oxygen species (ROS) inducible DNA cross-linking agents and their effect on cancer cells and normal lymphocytes. J. Med. Chem. 2014, 57, 4498–4510. [Google Scholar] [CrossRef] [PubMed]
  74. Huang, R.; Zhou, P.K. DNA damage repair: Historical perspectives, mechanistic pathways and clinical translation for targeted cancer therapy. Signal Transduct. Target. Ther. 2021, 6, 254. [Google Scholar] [CrossRef] [PubMed]
  75. Chen, J.; Potlapalli, R.; Quan, H.; Chen, L.; Xie, Y.; Pouriyeh, S.; Sakib, N.; Liu, L.; Xie, Y. Exploring DNA damage and repair mechanisms: A review with computational insights. BioTech 2024, 13, 3. [Google Scholar] [CrossRef] [PubMed]
  76. Kirtonia, A.; Sethi, G.; Garg, M. The multifaceted role of reactive oxygen species in tumorigenesis. Cell. Mol. Life Sci. 2020, 77, 4459–4483. [Google Scholar] [CrossRef]
  77. Hayes, J.D.; Dinkova-Kostova, A.T.; Tew, K.D. Oxidative Stress in Cancer. Cancer Cell 2020, 38, 167–197. [Google Scholar] [CrossRef]
  78. Arfin, S.; Jha, N.K.; Jha, S.K.; Kesari, K.K.; Ruokolainen, J.; Roychoudhury, S.; Rathi, B.; Kumar, D. Oxidative Stress in Cancer Cell Metabolism. Antioxidants 2021, 10, 642. [Google Scholar] [CrossRef]
  79. Iqbal, M.J.; Kabeer, A.; Abbas, Z.; Siddiqui, H.A.; Calina, D.; Sharifi-Rad, J.; Cho, W.C. Interplay of oxidative stress, cellular communication and signaling pathways in cancer. Cell Commun. Signal. 2024, 22, 7. [Google Scholar] [CrossRef]
  80. Najafi, M.; Farhood, B.; Mortezaee, K. Cancer stem cells (CSCs) in cancer progression and therapy. J. Cell. Physiol. 2019, 234, 8381–8395. [Google Scholar] [CrossRef]
  81. Li, K.; Deng, Z.; Lei, C.; Ding, X.; Li, J.; Wang, C. The Role of Oxidative Stress in Tumorigenesis and Progression. Cells 2024, 13, 441. [Google Scholar] [CrossRef]
  82. Zhao, H.; Wu, L.; Yan, G.; Chen, Y.; Zhou, M.; Wu, Y.; Li, Y. Inflammation and tumor progression: Signaling pathways and targeted intervention. Signal Transduct. Target. Ther. 2021, 6, 263. [Google Scholar] [CrossRef] [PubMed]
  83. Biagioni, A.; Peri, S.; Versienti, G.; Fiorillo, C.; Becatti, M.; Magnelli, L.; Papucci, L. Gastric Cancer Vascularization and the Contribution of Reactive Oxygen Species. Biomolecules 2023, 13, 886. [Google Scholar] [CrossRef] [PubMed]
  84. Ushio-Fukai, M.; Nakamura, Y. Reactive oxygen species and angiogenesis: NADPH oxidase as target for cancer therapy. Cancer Lett. 2008, 266, 37–52. [Google Scholar] [CrossRef]
  85. 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]
  86. Kuczynski, E.A.; Vermeulen, P.B.; Pezzella, F.; Kerbel, R.S.; Reynolds, A.R. Vessel co-option in cancer. Nat. Rev. Clin. Oncol. 2019, 16, 469–493. [Google Scholar] [CrossRef] [PubMed]
  87. Farahzadi, R.; Valipour, B.; Fathi, E.; Pirmoradi, S.; Molavi, O.; Montazersaheb, S.; Sanaat, Z. Oxidative stress regulation and related metabolic pathways in epithelial-mesenchymal transition of breast cancer stem cells. Stem Cell Res. Ther. 2023, 14, 342. [Google Scholar] [CrossRef]
  88. Mori, K.; Uchida, T.; Yoshie, T.; Mizote, Y.; Ishikawa, F.; Katsuyama, M.; Shibanuma, M. A mitochondrial ROS pathway controls matrix metalloproteinase 9 levels and invasive properties in RAS-activated cancer cells. FEBS J. 2019, 286, 459–478. [Google Scholar] [CrossRef]
  89. Cisneros, B.; García-Aguirre, I.; Unzueta, J.; Arrieta-Cruz, I.; González-Morales, O.; Domínguez-Larrieta, J.M.; Tamez-González, A.; Leyva-Gómez, G.; Magaña, J.J. Immune system modulation in aging: Molecular mechanisms and therapeutic targets. Front. Immunol. 2022, 13, 1059173. [Google Scholar] [CrossRef]
  90. Cannizzo, E.S.; Clement, C.C.; Sahu, R.; Follo, C.; Santambrogio, L. Oxidative stress, inflamm-aging and immunosenescence. J. Proteom. 2011, 74, 2313–2323. [Google Scholar] [CrossRef]
  91. Pangrazzi, L.; Meryk, A.; Naismith, E.; Koziel, R.; Lair, J.; Krismer, M.; Trieb, K.; Grubeck-Loebenstein, B. “Inflamm-aging” influences immune cell survival factors in human bone marrow. Eur. J. Immunol. 2017, 47, 481–492. [Google Scholar] [CrossRef]
  92. Olivier, C.; Oliver, L.; Lalier, L.; Vallette, F.M. Drug Resistance in Glioblastoma: The Two Faces of Oxidative Stress. Front. Mol. Biosci. 2021, 7, 620677. [Google Scholar] [CrossRef] [PubMed]
  93. Wang, K.; Zhang, T.; Dong, Q.; Nice, E.C.; Huang, C.; Wei, Y. Redox homeostasis: The linchpin in stem cell self-renewal and differentiation. Cell Death Dis. 2013, 4, e537. [Google Scholar] [CrossRef] [PubMed]
  94. Bigarella, C.L.; Liang, R.; Ghaffari, S. Stem cells and the impact of ROS signaling. Development 2014, 141, 4206–4218. [Google Scholar] [CrossRef]
  95. Movahed, Z.G.; Yarani, R.; Mohammadi, P.; Mansouri, K. Sustained oxidative stress instigates differentiation of cancer stem cells into tumor endothelial cells: Pentose phosphate pathway, reactive oxygen species and autophagy crosstalk. Biomed. Pharmacother. 2021, 139, 111643. [Google Scholar] [CrossRef]
  96. Najafi, M.; Mortezaee, K.; Majidpoor, J. Cancer stem cell (CSC) resistance drivers. Life Sci. 2019, 234, 116781. [Google Scholar] [CrossRef] [PubMed]
  97. Yang, J.; Ma, S.; Xu, R.; Wei, Y.; Zhang, J.; Zuo, T.; Wang, Z.; Deng, H.; Yang, N.; Shen, Q. Smart biomimetic metal organic frameworks based on ROS-ferroptosis-glycolysis regulation for enhanced tumor chemo-immunotherapy. J. Control. Release 2021, 334, 21–33. [Google Scholar] [CrossRef]
  98. Teng, M.W.; Ngiow, S.F.; Ribas, A.; Smyth, M.J. Classifying Cancers Based on T-cell Infiltration and PD-L1. Cancer Res. 2015, 75, 2139–2145. [Google Scholar] [CrossRef]
  99. Li, Z.; Zhu, L.; Sun, H.; Shen, Y.; Hu, D.; Wu, W.; Wang, Y.; Qian, C.; Sun, M. Fluorine assembly nanocluster breaks the shackles of immunosuppression to turn the cold tumor hot. Proc. Natl. Acad. Sci. USA 2020, 117, 32962–32969. [Google Scholar] [CrossRef]
  100. Deng, H.; Yang, W.; Zhou, Z.; Tian, R.; Lin, L.; Ma, Y.; Song, J.; Chen, X. Targeted scavenging of extracellular ROS relieves suppressive immunogenic cell death. Nat. Commun. 2020, 11, 4951. [Google Scholar] [CrossRef]
  101. Marino, P.; Pepe, G.; Basilicata, M.G.; Vestuto, V.; Marzocco, S.; Autore, G.; Procino, A.; Gomez-Monterrey, I.M.; Manfra, M.; Campiglia, P. Potential Role of Natural Antioxidant Products in Oncological Diseases. Antioxidants 2023, 12, 704. [Google Scholar] [CrossRef]
  102. Pisoschi, A.M.; Pop, A.; Iordache, F.; Stanca, L.; Geicu, O.I.; Bilteanu, L.; Serban, A.I. Antioxidant, anti-inflammatory and immunomodulatory roles of vitamins in COVID-19 therapy. Eur. J. Med. Chem. 2022, 232, 114175. [Google Scholar] [CrossRef] [PubMed]
  103. Ferencík, M.; Ebringer, L. Modulatory effects of selenium and zinc on the immune system. Folia Microbiol. 2003, 48, 417–426. [Google Scholar] [CrossRef] [PubMed]
  104. Hughes, D.A. Effects of dietary antioxidants on the immune function of middle-aged adults. Proc. Nutr. Soc. 1999, 58, 79–84. [Google Scholar] [CrossRef]
  105. Bendich, A. Physiological role of antioxidants in the immune system. J. Dairy Sci. 1993, 76, 2789–2794. [Google Scholar] [CrossRef]
  106. Key, T.J.; Schatzkin, A.; Willett, W.C.; Allen, N.E.; Spencer, E.A.; Travis, R.C. Diet, nutrition and the prevention of cancer. Public Health Nutr. 2004, 7, 187–200. [Google Scholar] [CrossRef]
  107. Turati, F.; Rossi, M.; Pelucchi, C.; Levi, F.; La Vecchia, C. Fruit and vegetables and cancer risk: A review of southern European studies. Br. J. Nutr. 2015, 113 (Suppl. 2), S102–S110. [Google Scholar] [CrossRef]
  108. Klein, E.A.; Thompson, I.M., Jr.; Tangen, C.M.; Crowley, J.J.; Lucia, M.S.; Goodman, P.J.; Minasian, L.M.; Ford, L.G.; Parnes, H.L.; Gaziano, J.M.; et al. Vitamin E and the risk of prostate cancer: The Selenium and Vitamin E Cancer Prevention Trial (SELECT). JAMA 2011, 306, 1549–1556. [Google Scholar] [CrossRef] [PubMed]
  109. Olsson, M.E.; Andersson, C.S.; Oredsson, S.; Berglund, R.H.; Gustavsson, K.E. Antioxidant levels and inhibition of cancer cell proliferation in vitro by extracts from organically and conventionally cultivated strawberries. J. Agric. Food Chem. 2006, 54, 1248–1255. [Google Scholar] [CrossRef]
  110. Aiello, P.; Sharghi, M.; Mansourkhani, S.M.; Ardekan, A.P.; Jouybari, L.; Daraei, N.; Peiro, K.; Mohamadian, S.; Rezaei, M.; Heidari, M.; et al. Medicinal Plants in the Prevention and Treatment of Colon Cancer. Oxidative Med. Cell. Longev. 2019, 2019, 2075614. [Google Scholar] [CrossRef]
  111. Kozlov, A.V.; Javadov, S.; Sommer, N. Cellular ROS and Antioxidants: Physiological and Pathological Role. Antioxidants 2024, 13, 602. [Google Scholar] [CrossRef]
  112. Patra, S.; Pradhan, B.; Nayak, R.; Behera, C.; Rout, L.; Jena, M.; Efferth, T.; Bhutia, S.K. Chemotherapeutic efficacy of curcumin and resveratrol against cancer: Chemoprevention, chemoprotection, drug synergism and clinical pharmacokinetics. Semin. Cancer Biol. 2021, 73, 310–320. [Google Scholar] [CrossRef] [PubMed]
  113. Imran, M.; Insaf, A.; Hasan, N.; Sugandhi, V.V.; Shrestha, D.; Paudel, K.R.; Jha, S.K.; Hansbro, P.M.; Dua, K.; Devkota, H.P.; et al. Exploring the Remarkable Chemotherapeutic Potential of Polyphenolic Antioxidants in Battling Various Forms of Cancer. Molecules 2023, 28, 3475. [Google Scholar] [CrossRef] [PubMed]
  114. Toledo, E.; Salas-Salvadó, J.; Donat-Vargas, C.; Buil-Cosiales, P.; Estruch, R.; Ros, E.; Corella, D.; Fitó, M.; Hu, F.B.; Arós, F.; et al. Mediterranean Diet and Invasive Breast Cancer Risk Among Women at High Cardiovascular Risk in the PREDIMED Trial: A Randomized Clinical Trial. JAMA Intern. Med. 2015, 175, 1752–1760, Erratum in: JAMA Intern. Med. 2018, 178, 1731–1732. [Google Scholar] [CrossRef] [PubMed]
  115. Alpha-Tocopherol, Beta Carotene Cancer Prevention Study Group. The effect of vitamin E and beta carotene on the incidence of lung cancer and other cancers in male smokers. N. Engl. J. Med. 1994, 330, 1029–1035. [Google Scholar] [CrossRef]
  116. Rock, C.L.; Natarajan, L.; Pu, M.; Thomson, C.A.; Flatt, S.W.; Caan, B.J.; Gold, E.B.; Al-Delaimy, W.K.; Newman, V.A.; Hajek, R.A.; et al. Longitudinal biological exposure to carotenoids is associated with breast cancer-free survival in the Women’s Healthy Eating and Living Study. Cancer Epidemiol. Biomark. Prev. 2009, 18, 486–494. [Google Scholar] [CrossRef]
  117. Romney, S.L.; Ho, G.Y.; Palan, P.R.; Basu, J.; Kadish, A.S.; Klein, S.; Mikhail, M.; Hagan, R.J.; Chang, C.J.; Burk, R.D. Effects of beta-carotene and other factors on outcome of cervical dysplasia and human papillomavirus infection. Gynecol. Oncol. 1997, 65, 483–492. [Google Scholar] [CrossRef]
  118. Manetta, A.; Schubbert, T.; Chapman, J.; Schell, M.J.; Peng, Y.M.; Liao, S.Y.; Meyskens, F.J., Jr. beta-Carotene treatment of cervical intraepithelial neoplasia: A phase II study. Cancer Epidemiol. Biomark. Prev. 1996, 5, 929–932. [Google Scholar]
  119. Green, A.; Williams, G.; Neale, R.; Hart, V.; Leslie, D.; Parsons, P.; Marks, G.C.; Gaffney, P.; Battistutta, D.; Frost, C.; et al. Daily sunscreen application and betacarotene supplementation in prevention of basal-cell and squamous-cell carcinomas of the skin: A randomised controlled trial. Lancet 1999, 354, 723–729. [Google Scholar] [CrossRef]
  120. Omenn, G.S.; Goodman, G.E.; Thornquist, M.D.; Balmes, J.; Cullen, M.R.; Glass, A.; Keogh, J.P.; Meyskens, F.L., Jr.; Valanis, B.; Williams, J.H., Jr.; et al. Risk factors for lung cancer and for intervention effects in CARET, the Beta-Carotene and Retinol Efficacy Trial. J. Natl. Cancer Inst. 1996, 88, 1550–1559. [Google Scholar] [CrossRef]
  121. Goodman, G.E.; Thornquist, M.D.; Balmes, J.; Cullen, M.R.; Meyskens, F.L., Jr.; Omenn, G.S.; Valanis, B.; Williams, J.H., Jr. The Beta-Carotene and Retinol Efficacy Trial: Incidence of lung cancer and cardiovascular disease mortality during 6-year follow-up after stopping beta-carotene and retinol supplements. J. Natl. Cancer Inst. 2004, 96, 1743–1750. [Google Scholar] [CrossRef]
  122. Carotene and Retinol Efficacy Trial (CARET). Available online: https://clinicaltrials.gov/ct2/show/NCT00712647 (accessed on 19 July 2024).
  123. Greenberg, E.R.; Baron, J.A.; Stevens, M.M.; Stukel, T.A.; Mandel, J.S.; Spencer, S.K.; Elias, P.M.; Nicholas, L.; Nierenberg, D.N.; Bayrd, G.; et al. The Skin Cancer Prevention Study: Design of a clinical trial of beta-carotene among persons at high risk for nonmelanoma skin cancer. Control. Clin. Trials 1989, 10, 153–166. [Google Scholar] [CrossRef] [PubMed]
  124. Frieling, U.M.; Schaumberg, D.A.; Kupper, T.S.; Muntwyler, J.; Hennekens, C.H. A randomized, 12-year primary-prevention trial of beta carotene supplementation for nonmelanoma skin cancer in the physician’s health study. Arch. Dermatol. 2000, 136, 179–184. [Google Scholar] [CrossRef] [PubMed]
  125. Popat, R.; Plesner, T.; Davies, F.; Cook, G.; Cook, M.; Elliott, P.; Jacobson, E.; Gumbleton, T.; Oakervee, H.; Cavenagh, J. A phase 2 study of SRT501 (resveratrol) with bortezomib for patients with relapsed and or refractory multiple myeloma. Br. J. Haematol. 2013, 160, 714–717. [Google Scholar] [CrossRef] [PubMed]
  126. Paller, C.J.; Zhou, X.C.; Heath, E.I.; Taplin, M.E.; Mayer, T.; Stein, M.N.; Bubley, G.J.; Pili, R.; Hudson, T.; Kakarla, R.; et al. Muscadine Grape Skin Extract (MPX) in Men with Biochemically Recurrent Prostate Cancer: A Randomized, Multicenter, Placebo-Controlled Clinical Trial. Clin. Cancer Res. 2018, 24, 306–315. [Google Scholar] [CrossRef]
  127. Paller, C.J.; Rudek, M.A.; Zhou, X.C.; Wagner, W.D.; Hudson, T.S.; Anders, N.; Hammers, H.J.; Dowling, D.; King, S.; Antonarakis, E.S.; et al. A phase I study of muscadine grape skin extract in men with biochemically recurrent prostate cancer: Safety, tolerability, and dose determination. Prostate 2015, 75, 1518–1525. [Google Scholar] [CrossRef]
  128. Winterbourn, C.C. The challenges of using fluorescent probes to detect and quantify specific reactive oxygen species in living cells. Biochim. Biophys. Acta 2014, 1840, 730–738. [Google Scholar] [CrossRef]
  129. Van de Bittner, G.C.; Dubikovskaya, E.A.; Bertozzi, C.R.; Chang, C.J. In vivo imaging of hydrogen peroxide production in a murine tumor model with a chemoselective bioluminescent reporter. Proc. Natl. Acad. Sci. USA 2010, 107, 21316–21321. [Google Scholar] [CrossRef]
  130. Duanghathaipornsuk, S.; Farrell, E.J.; Alba-Rubio, A.C.; Zelenay, P.; Kim, D.S. Detection technologies for reactive oxygen species: Fluorescence and electrochemical methods and their applications. Biosensors 2021, 11, 30. [Google Scholar] [CrossRef] [PubMed]
  131. Towner, R.A.; Smith, N.; Saunders, D.; Lupu, F.; Silasi-Mansat, R.; West, M.; Ramirez, D.C.; Gomez-Mejiba, S.E.; Bonini, M.G.; Mason, R.P.; et al. In vivo detection of free radicals using molecular MRI and immuno-spin trapping in a mouse model for amyotrophic lateral sclerosis. Free Radic. Biol. Med. 2013, 63, 351–360. [Google Scholar] [CrossRef]
  132. Berliner, L.J. (Ed.) In Vivo EPR (ESR); Springer: Berlin/Heidelberg, Germany, 2012. [Google Scholar] [CrossRef]
  133. Caia, G.L.; Efimova, O.V.; Velayutham, M.; El-Mahdy, M.A.; Abdelghany, T.M.; Kesselring, E.; Petryakov, S.; Sun, Z.; Samouilov, A.; Zweier, J.L. Organ specific mapping of in vivo redox state in control and cigarette smoke-exposed mice using EPR/NMR co-imaging. J. Magn. Reson. 2012, 216, 21–27. [Google Scholar] [CrossRef]
  134. Hou, C.; Hsieh, C.-J.; Li, S.; Lee, H.; Graham, T.J.; Xu, K.; Weng, C.-C.; Doot, R.K.; Chu, W.; Chakraborty, S.K.; et al. Development of a positron emission tomography radiotracer for imaging elevated levels of superoxide in neuroinflammation. ACS Chem. Neurosci. 2018, 9, 578–586. [Google Scholar] [CrossRef] [PubMed]
  135. Greenwood, H.E.; Witney, T.H. Latest advances in imaging oxidative stress in cancer. J. Nucl. Med. 2021, 62, 1506–1510. [Google Scholar] [CrossRef] [PubMed]
  136. Ikawa, M.; Okazawa, H.; Nakamoto, Y.; Yoneda, M. PET imaging for oxidative stress in neurodegenerative disorders associated with mitochondrial dysfunction. Antioxidants 2020, 9, 861. [Google Scholar] [CrossRef] [PubMed]
  137. Tsakanova, G.; Arakelova, E.; Ayvazyan, V.; Ayvazyan, A.; Tatikyan, S.; Aroutiounian, R.; Dalyan, Y.; Haroutiunian, S.; Tsakanov, V.; Arakelyan, A. Two-photon microscopy imaging of oxidative stress in human living erythrocytes. Biomed. Opt. Express 2017, 8, 5834–5846. [Google Scholar] [CrossRef]
  138. Wang, H.; Zhang, R.; Bridle, K.R.; Jayachandran, A.; Thomas, J.A.; Zhang, W.; Yuan, J.; Xu, Z.P.; Crawford, D.H.G.; Liang, X.; et al. Two-photon dual imaging platform for in vivo monitoring cellular oxidative stress in liver injury. Sci. Rep. 2017, 7, 45374. [Google Scholar] [CrossRef]
  139. Calvo-Rodriguez, M.; Kharitonova, E.K.; Snyder, A.C.; Hou, S.S.; Sanchez-Mico, M.V.; Das, S.; Fan, Z.; Shirani, H.; Nilsson, K.P.R.; Serrano-Pozo, A.; et al. Real-time imaging of mitochondrial redox reveals increased mitochondrial oxidative stress associated with amyloid β aggregates in vivo in a mouse model of Alzheimer’s disease. Mol. Neurodegener. 2024, 19, 6. [Google Scholar] [CrossRef]
  140. Ripanti, F.; Fasolato, C.; Mazzarda, F.; Palleschi, S.; Ceccarini, M.; Li, C.; Bignami, M.; Bodo, E.; Bell, S.E.; Mazzei, F.; et al. Advanced Raman spectroscopy detection of oxidative damage in nucleic acid bases: Probing chemical changes and intermolecular interactions in guanosine at ultralow concentration. Anal. Chem. 2021, 93, 10825–10833. [Google Scholar] [CrossRef]
  141. Surmacki, J.M.; Quiros-Gonzalez, I.; Bohndiek, S.E. Evaluation of label-free confocal Raman microspectroscopy for monitoring oxidative stress in vitro in live human cancer cells. Antioxidants 2022, 11, 573. [Google Scholar] [CrossRef] [PubMed]
  142. Godin, A.G.; Lounis, B.; Cognet, L. Super-resolution microscopy approaches for live cell imaging. Biophys. J. 2014, 107, 1777–1784. [Google Scholar] [CrossRef]
  143. Wu, K.; El Zowalaty, A.E.; Sayin, V.I.; Papagiannakopoulos, T. The pleiotropic functions of reactive oxygen species in cancer. Nat. Cancer 2024, 5, 384–399. [Google Scholar] [CrossRef]
  144. Jiang, W.; Liang, M.; Lei, Q.; Li, G.; Wu, S. The current status of photodynamic therapy in cancer treatment. Cancers 2023, 15, 585. [Google Scholar] [CrossRef] [PubMed]
  145. Wood, A.K.W.; Sehgal, C.M. A review of low-intensity ultrasound for cancer therapy. Ultrasound Med. Biol. 2015, 41, 905–928. [Google Scholar] [CrossRef]
  146. Zhang, Y.; Ding, C.; Zhu, W.; Li, X.; Chen, T.; Liu, Q.; Zhou, S.; Zhang, T.-C.; Ma, W. Chemotherapeutic drugs induce oxidative stress associated with DNA repair and metabolism modulation. Life Sci. 2022, 289, 120242. [Google Scholar] [CrossRef] [PubMed]
  147. Padi, S.K.R.; Chauhan, S.S.; Singh, N. ROS Induced by Chemo- and Targeted Therapy Promote Apoptosis in Cancer Cells. In Handbook of Oxidative Stress in Cancer: Mechanistic Aspects; Springer Nature: Singapore, 2022; pp. 583–598. [Google Scholar] [CrossRef]
  148. Kciuk, M.; Gielecińska, A.; Mujwar, S.; Kołat, D.; Kałuzińska-Kołat, Ż.; Celik, I.; Kontek, R. Doxorubicin-an agent with multiple mechanisms of anticancer activity. Cells 2023, 12, 659. [Google Scholar] [CrossRef] [PubMed]
  149. Dasari, S.; Tchounwou, P.B. Cisplatin in cancer therapy: Molecular mechanisms of action. Eur. J. Pharmacol. 2014, 740, 364–378. [Google Scholar] [CrossRef]
  150. de Vries, G.; Rosas-Plaza, X.; van Vugt, M.A.T.M.; Gietema, J.A.; de Jong, S. Testicular cancer: Determinants of cisplatin sensitivity and novel therapeutic opportunities. Cancer Treat. Rev. 2020, 88, 102054. [Google Scholar] [CrossRef]
  151. Kleih, M.; Böpple, K.; Dong, M.; Gaißler, A.; Heine, S.; Olayioye, M.A.; Aulitzky, W.E.; Essmann, F. Direct impact of cisplatin on mitochondria induces ROS production that dictates cell fate of ovarian cancer cells. Cell Death Dis. 2019, 10, 851. [Google Scholar] [CrossRef]
  152. Taber, A.; Christensen, E.; Lamy, P.; Nordentoft, I.; Prip, F.; Lindskrog, S.V.; Birkenkamp-Demtröder, K.; Okholm, T.L.H.; Knudsen, M.; Pedersen, J.S.; et al. Molecular correlates of cisplatin-based chemotherapy response in muscle invasive bladder cancer by integrated multi-omics analysis. Nat. Commun. 2020, 11, 4858. [Google Scholar] [CrossRef]
  153. Wang, G.; Reed, E.; Li, Q.Q. Molecular basis of cellular response to cisplatin chemotherapy in non-small cell lung cancer (Review). Oncol. Rep. 2004, 12, 955–965. [Google Scholar] [CrossRef]
  154. Helfenstein, S.; Riesterer, O.; Meier, U.R.; Papachristofilou, A.; Kasenda, B.; Pless, M.; Rothschild, S.I. 3-weekly or weekly cisplatin concurrently with radiotherapy for patients with squamous cell carcinoma of the head and neck—A multicentre, retrospective analysis. Radiat. Oncol. 2019, 14, 32. [Google Scholar] [CrossRef]
  155. Abu Samaan, T.M.; Samec, M.; Liskova, A.; Kubatka, P.; Büsselberg, D. Paclitaxel’s mechanistic and clinical effects on breast cancer. Biomolecules 2019, 9, 789. [Google Scholar] [CrossRef] [PubMed]
  156. Baird, R.D.; Tan, D.S.P.; Kaye, S.B. Weekly paclitaxel in the treatment of recurrent ovarian cancer. Nat. Rev. Clin. Oncol. 2010, 7, 575–582. [Google Scholar] [CrossRef] [PubMed]
  157. von Eiff, D.; Bozorgmehr, F.; Chung, I.; Bernhardt, D.; Rieken, S.; Liersch, S.; Muley, T.; Kobinger, S.; Thomas, M.; Christopoulos, P.; et al. Paclitaxel for treatment of advanced small cell lung cancer (SCLC): A retrospective study of 185 patients. J. Thorac. Dis. 2020, 12, 782–793. [Google Scholar] [CrossRef] [PubMed]
  158. Alexandre, J.; Hu, Y.; Lu, W.; Pelicano, H.; Huang, P. Novel action of paclitaxel against cancer cells: Bystander effect mediated by reactive oxygen species. Cancer Res. 2007, 67, 3512–3517. [Google Scholar] [CrossRef]
  159. Dolmans, D.E.J.G.J.; Fukumura, D.; Jain, R.K. Photodynamic therapy for cancer. Nat. Rev. Cancer 2003, 3, 380–387. [Google Scholar] [CrossRef]
  160. Gunaydin, G.; Gedik, M.E.; Ayan, S. Photodynamic therapy for the treatment and diagnosis of cancer-A Review of the current clinical status. Front. Chem. 2021, 9, 686303. [Google Scholar] [CrossRef]
  161. Mehta, N.H.; Shah, H.A.; D’Amico, R.S. Sonodynamic therapy and sonosensitizers for glioma treatment: A systematic qualitative review. World Neurosurg. 2023, 178, 60–68. [Google Scholar] [CrossRef]
  162. Yamaguchi, T.; Kitahara, S.; Kusuda, K.; Okamoto, J.; Horise, Y.; Masamune, K.; Muragaki, Y. Current landscape of sonodynamic therapy for treating cancer. Cancers 2021, 13, 6184. [Google Scholar] [CrossRef] [PubMed]
  163. Harding, C.; Pompei, F.; Lee, E.E.; Wilson, R. Cancer suppression at old age. Cancer Res. 2008, 68, 4465–4478. [Google Scholar] [CrossRef]
  164. Ledford, H. Why cancer risk declines sharply in old age. Nature 2024, 631, 261–262. [Google Scholar] [CrossRef]
  165. Esme, H.; Cemek, M.; Sezer, M.; Saglam, H.; Demir, A.; Melek, H.; Unlu, M. High levels of oxidative stress in patients with advanced lung cancer. Respirology 2008, 13, 112–116. [Google Scholar] [CrossRef] [PubMed]
  166. Caliri, A.W.; Tommasi, S.; Besaratinia, A. Relationships among smoking, oxidative stress, inflammation, macromolecular damage, and cancer. Mutat. Res. Rev. Mutat. Res. 2021, 787, 108365. [Google Scholar] [CrossRef] [PubMed]
  167. Valavanidis, A.; Vlachogianni, T.; Fiotakis, K.; Loridas, S. Pulmonary oxidative stress, inflammation and cancer: Respirable particulate matter, fibrous dusts and ozone as major causes of lung carcinogenesis through reactive oxygen species mechanisms. Int. J. Environ. Res. Public Health 2013, 10, 3886–3907. [Google Scholar] [CrossRef]
  168. Lawless, M.W.; O’Byrne, K.J.; Gray, S.G. Oxidative stress induced lung cancer and COPD: Opportunities for epigenetic therapy. J. Cell. Mol. Med. 2009, 13, 2800–2821. [Google Scholar] [CrossRef]
  169. Ferraro, M.; Di Vincenzo, S.; Lazzara, V.; Pinto, P.; Patella, B.; Inguanta, R.; Bruno, A.; Pace, E. Formoterol exerts anti-cancer effects modulating oxidative stress and epithelial-mesenchymal transition processes in cigarette smoke extract exposed lung adenocarcinoma cells. Int. J. Mol. Sci. 2023, 24, 16088. [Google Scholar] [CrossRef]
  170. Gęgotek, A.; Nikliński, J.; Žarković, N.; Žarković, K.; Waeg, G.; Łuczaj, W.; Charkiewicz, R.; Skrzydlewska, E. Lipid mediators involved in the oxidative stress and antioxidant defence of human lung cancer cells. Redox Biol. 2016, 9, 210–219. [Google Scholar] [CrossRef] [PubMed]
  171. Barreiro, E.; Fermoselle, C.; Mateu-Jimenez, M.; Sánchez-Font, A.; Pijuan, L.; Gea, J.; Curull, V. Oxidative stress and inflammation in the normal airways and blood of patients with lung cancer and COPD. Free Radic. Biol. Med. 2013, 65, 859–871. [Google Scholar] [CrossRef]
  172. Kotsopoulos, J. Menopausal hormones: Definitive evidence for breast cancer. Lancet 2019, 394, 1116–1118. [Google Scholar] [CrossRef]
  173. Jezierska-Drutel, A.; Rosenzweig, S.A.; Neumann, C.A. Role of oxidative stress and the microenvironment in breast cancer development and progression. Adv. Cancer Res. 2013, 119, 107–125. [Google Scholar] [CrossRef]
  174. Sastre-Serra, J.; Valle, A.; Company, M.M.; Garau, I.; Oliver, J.; Roca, P. Estrogen down-regulates uncoupling proteins and increases oxidative stress in breast cancer. Free Radic. Biol. Med. 2010, 48, 506–512. [Google Scholar] [CrossRef]
  175. Nadal-Serrano, M.; Sastre-Serra, J.; Pons, D.G.; Miró, A.M.; Oliver, J.; Roca, P. The ERalpha/ERbeta ratio determines oxidative stress in breast cancer cell lines in response to 17beta-estradiol. J. Cell. Biochem. 2012, 113, 3178–3185. [Google Scholar] [CrossRef] [PubMed]
  176. Singh, B.; Chatterjee, A.; Ronghe, A.M.; Bhat, N.K.; Bhat, H.K. Antioxidant-mediated up-regulation of OGG1 via NRF2 induction is associated with inhibition of oxidative DNA damage in estrogen-induced breast cancer. BMC Cancer 2013, 13, 253. [Google Scholar] [CrossRef]
  177. Singh, B.; Bhat, N.K.; Bhat, H.K. Partial inhibition of estrogen-induced mammary carcinogenesis in rats by tamoxifen: Balance between oxidant stress and estrogen responsiveness. PLoS ONE 2011, 6, e25125. [Google Scholar] [CrossRef]
  178. Smolarek, A.K.; Suh, N. Chemopreventive activity of vitamin E in breast cancer: A focus on γ- and δ-tocopherol. Nutrients 2011, 3, 962–986. [Google Scholar] [CrossRef] [PubMed]
  179. Mahalingaiah, P.K.S.; Ponnusamy, L.; Singh, K.P. Chronic oxidative stress causes estrogen-independent aggressive phenotype, and epigenetic inactivation of estrogen receptor alpha in MCF-7 breast cancer cells. Breast Cancer Res. Treat. 2015, 153, 41–56. [Google Scholar] [CrossRef] [PubMed]
  180. Wu, Y.; Zhuang, J.; Zhang, Q.; Zhao, X.; Chen, G.; Han, S.; Hu, B.; Wu, W.; Han, S. Aging characteristics of colorectal cancer based on gut microbiota. Cancer Med. 2023, 12, 17822–17834. [Google Scholar] [CrossRef]
  181. Carini, F.; Mazzola, M.; Rappa, F.; Jurjus, A.; Geagea, A.G.; Al Kattar, S.; Bou-Assi, T.; Jurjus, R.; Damiani, P.; Leone, A.; et al. Colorectal Carcinogenesis: Role of Oxidative Stress and Antioxidants. Anticancer Res. 2017, 37, 4759–4766. [Google Scholar] [CrossRef]
  182. Pettan-Brewer, C.; Morton, J.; Mangalindan, R.; Ladiges, W. Curcumin suppresses intestinal polyps in APC Min mice fed a high fat diet. Pathobiol. Aging Age-Relat. Dis. 2011, 1, 7013. [Google Scholar] [CrossRef]
  183. Tsuchiya, H.; Kanbe, T.; Harada, K.; Yashima, K.; Nozaka, K.; Tanida, O.; Kohno, M.; Mukoyama, T.; Nishimuki, E.; Kojo, H.; et al. Aberrant expression of selenoproteins in the progression of colorectal cancer. Cancer Lett. 2008, 259, 218–230. [Google Scholar] [CrossRef]
  184. Kavec, M.J.; Urbanova, M.; Makovicky, P.; Opattová, A.; Tomasova, K.; Kroupa, M.; Kostovcikova, K.; Siskova, A.; Navvabi, N.; Schneiderova, M.; et al. Oxidative damage in sporadic colorectal cancer: Molecular mapping of base excision repair glycosylases MUTYH and hOGG1 in colorectal cancer patients. Int. J. Mol. Sci. 2022, 23, 5704. [Google Scholar] [CrossRef]
  185. Lamaudière, M.T.F.; Arasaradnam, R.; Weedall, G.D.; Morozov, I.Y. The colorectal cancer Microbiota alter their transcriptome to adapt to the acidity, reactive oxygen species, and metabolite availability of gut microenvironments. mSphere 2023, 8, e0062722. [Google Scholar] [CrossRef] [PubMed]
  186. Udensi, U.K.; Tchounwou, P.B. Oxidative stress in prostate hyperplasia and carcinogenesis. J. Exp. Clin. Cancer Res. 2016, 35, 139. [Google Scholar] [CrossRef]
  187. Biesiadecki, M.; Mołoń, M.; Balawender, K.; Kobylińska, Z.; Galiniak, S. Shedding light on the shadows: Oxidative stress and its pivotal role in prostate cancer progression. Front. Oncol. 2024, 14, 1393078. [Google Scholar] [CrossRef]
  188. Wang, J.; Xiao, Q.; Chen, X.; Tong, S.; Sun, J.; Lv, R.; Wang, S.; Gou, Y.; Tan, L.; Xu, J.; et al. LanCL1 protects prostate cancer cells from oxidative stress via suppression of JNK pathway. Cell Death Dis. 2018, 9, 197. [Google Scholar] [CrossRef]
  189. Fu, Y.; Chung, F.L. Oxidative stress and hepatocarcinogenesis. Hepatoma Res. 2018, 4, 39. [Google Scholar] [CrossRef] [PubMed]
  190. McLoughlin, M.R.; Orlicky, D.J.; Prigge, J.R.; Krishna, P.; Talago, E.A.; Cavigli, I.R.; Eriksson, S.; Miller, C.G.; Kundert, J.A.; Sayin, V.I.; et al. TrxR1, Gsr, and oxidative stress determine hepatocellular carcinoma malignancy. Proc. Natl. Acad. Sci. USA 2019, 116, 11408–11417. [Google Scholar] [CrossRef]
  191. Liu, Y.; Hao, C.; Li, L.; Zhang, H.; Zha, W.; Ma, L.; Chen, L.; Gan, J. The role of oxidative stress in the development and therapeutic intervention of hepatocellular carcinoma. Curr. Cancer Drug Targets 2023, 23, 792–804. [Google Scholar] [CrossRef]
  192. Huang, P.S.; Wang, C.S.; Yeh, C.T.; Lin, K.H. Roles of thyroid hormone-associated microRNAs affecting oxidative stress in human hepatocellular carcinoma. Int. J. Mol. Sci. 2019, 20, 5220. [Google Scholar] [CrossRef]
  193. Takaki, A.; Yamamoto, K. Control of oxidative stress in hepatocellular carcinoma: Helpful or harmful? World J. Hepatol. 2015, 7, 968–979. [Google Scholar] [CrossRef]
  194. Huang, Z.; Zhou, L.; Duan, J.; Qin, S.; Jiang, J.; Chen, H.; Wang, K.; Liu, R.; Yuan, M.; Tang, X.; et al. Oxidative stress promotes liver cancer metastasis via RNF25-mediated E-cadherin protein degradation. Adv. Sci. 2024, 11, e2306929. [Google Scholar] [CrossRef]
  195. Liu, H.-M.; Cheng, M.-Y.; Xun, M.-H.; Zhao, Z.-W.; Zhang, Y.; Tang, W.; Cheng, J.; Ni, J.; Wang, W. Possible mechanisms of oxidative stress-induced skin cellular senescence, inflammation, and cancer and the therapeutic potential of plant polyphenols. Int. J. Mol. Sci. 2023, 24, 3755. [Google Scholar] [CrossRef]
  196. Pizzimenti, S.; Ribero, S.; Cucci, M.A.; Grattarola, M.; Monge, C.; Dianzani, C.; Barrera, G.; Muzio, G. Oxidative Stress-Related Mechanisms in Melanoma and in the Acquired Resistance to Targeted Therapies. Antioxidants 2021, 10, 1942. [Google Scholar] [CrossRef] [PubMed]
  197. Brand, R.M.; Wipf, P.; Durham, A.; Epperly, M.W.; Greenberger, J.S.; Falo, L.D., Jr. Targeting Mitochondrial Oxidative Stress to Mitigate UV-Induced Skin Damage. Front. Pharmacol. 2018, 9, 920. [Google Scholar] [CrossRef] [PubMed]
  198. Denat, L.; Kadekaro, A.L.; Marrot, L.; Leachman, S.A.; Abdel-Malek, Z.A. Melanocytes as instigators and victims of oxidative stress. J. Investig. Dermatol. 2014, 134, 1512–1518. [Google Scholar] [CrossRef] [PubMed]
  199. Bisevac, J.P.; Djukic, M.; Stanojevic, I.; Stevanovic, I.; Mijuskovic, Z.; Djuric, A.; Gobeljic, B.; Banovic, T.; Vojvodic, D. Association between oxidative stress and melanoma progression. J. Med. Biochem. 2018, 37, 12–20. [Google Scholar] [CrossRef] [PubMed]
  200. Karampinis, E.; Nechalioti, P.-M.; Georgopoulou, K.E.; Goniotakis, G.; Schulze, A.V.R.; Zafiriou, E.; Kouretas, D. Systemic oxidative stress parameters in skin cancer patients and patients with benign lesions. Stresses 2023, 3, 785–812. [Google Scholar] [CrossRef]
  201. Krueger, A.; Mohamed, A.; Kolka, C.M.; Stoll, T.; Zaugg, J.; Linedale, R.; Morrison, M.; Soyer, H.P.; Hugenholtz, P.; Frazer, I.H.; et al. Skin cancer-associated S. aureus strains can induce DNA damage in human keratinocytes by downregulating DNA repair and promoting oxidative stress. Cancers 2022, 14, 2143. [Google Scholar] [CrossRef]
  202. Poljsak, B. Strategies for reducing or preventing the generation of oxidative stress. Oxidative Med. Cell. Longev. 2011, 2011, 194586. [Google Scholar] [CrossRef]
  203. Schwalfenberg, G.K. N-acetylcysteine: A review of clinical usefulness (an old drug with new tricks). J. Nutr. Metab. 2021, 2021, 9949453. [Google Scholar] [CrossRef]
  204. Smaga, I.; Frankowska, M.; Filip, M. N-acetylcysteine as a new prominent approach for treating psychiatric disorders. Br. J. Pharmacol. 2021, 178, 2569–2594. [Google Scholar] [CrossRef]
  205. Garrido-Maraver, J. Clinical applications of coenzyme Q10. Front. Biosci. 2014, 19, 619. [Google Scholar] [CrossRef] [PubMed]
  206. Gredilla, R.; Barja, G. Minireview: The role of oxidative stress in relation to caloric restriction and longevity. Endocrinology 2005, 146, 3713–3717. [Google Scholar] [CrossRef] [PubMed]
  207. Strilbytska, O.; Klishch, S.; Storey, K.B.; Koliada, A.; Lushchak, O. Intermittent fasting and longevity: From animal models to implication for humans. Ageing Res. Rev. 2024, 96, 102274. [Google Scholar] [CrossRef] [PubMed]
  208. Wu, D.; Cederbaum, A.I. Alcohol, oxidative stress, and free radical damage. Alcohol Res. Health 2003, 27, 277–284. [Google Scholar] [PubMed]
  209. Lee, B.J.; Egi, Y.; van Leyen, K.; Lo, E.H.; Arai, K. Edaravone, a free radical scavenger, protects components of the neurovascular unit against oxidative stress in vitro. Brain Res. 2010, 1307, 22–27. [Google Scholar] [CrossRef]
  210. Maragakis, N.J.; de Carvalho, M.; Weiss, M.D. Therapeutic targeting of ALS pathways: Refocusing an incomplete picture. Ann. Clin. Transl. Neurol. 2023, 10, 1948–1971. [Google Scholar] [CrossRef]
  211. Duranti, E.; Cordani, N.; Villa, C. Edaravone: A novel possible drug for cancer treatment? Int. J. Mol. Sci. 2024, 25, 1633. [Google Scholar] [CrossRef]
  212. Konaté, M.M.; Antony, S.; Doroshow, J.H. Inhibiting the activity of NADPH oxidase in cancer. Antioxid. Redox. Signal 2020, 33, 435–454. [Google Scholar] [CrossRef]
  213. Zhang, Z.; Ghosh, A.; Connolly, P.J.; King, P.; Wilde, T.; Wang, J.; Dong, Y.; Li, X.; Liao, D.; Chen, H.; et al. Gut-restricted selective cyclooxygenase-2 (COX-2) inhibitors for chemoprevention of colorectal cancer. J. Med. Chem. 2021, 64, 11570–11596. [Google Scholar] [CrossRef]
  214. Orafaie, A.; Matin, M.M.; Sadeghian, H. The importance of 15-lipoxygenase inhibitors in cancer treatment. Cancer Metastasis Rev. 2018, 37, 397–408. [Google Scholar] [CrossRef]
  215. Reis, J.; Gorgulla, C.; Massari, M.; Marchese, S.; Valente, S.; Noce, B.; Basile, L.; Törner, R.; Cox, H., 3rd; Viennet, T.; et al. Targeting ROS production through inhibition of NADPH oxidases. Nat. Chem. Biol. 2023, 19, 1540–1550. [Google Scholar] [CrossRef] [PubMed]
  216. Kurzhagen, J.T.; Noel, S.; Lee, K.; Sadasivam, M.; Gharaie, S.; Ankireddy, A.; Lee, S.A.; Newman-Rivera, A.; Gong, J.; Arend, L.J.; et al. T cell Nrf2/Keap1 gene editing using CRISPR/Cas9 and experimental kidney ischemia-reperfusion injury. Antioxid. Redox Signal. 2023, 38, 959–973. [Google Scholar] [CrossRef] [PubMed]
  217. Ekman, F.K.; Ojala, D.S.; Adil, M.M.; Lopez, P.A.; Schaffer, D.V.; Gaj, T. CRISPR-Cas9-mediated genome editing increases lifespan and improves motor deficits in a Huntington’s disease mouse model. Mol. Ther. Nucleic Acids 2019, 17, 829–839. [Google Scholar] [CrossRef] [PubMed]
  218. Duan, W.; Guo, M.; Yi, L.; Liu, Y.; Li, Z.; Ma, Y.; Zhang, G.; Liu, Y.; Bu, H.; Song, X.; et al. The deletion of mutant SOD1 via CRISPR/Cas9/sgRNA prolongs survival in an amyotrophic lateral sclerosis mouse model. Gene Ther. 2020, 27, 157–169. [Google Scholar] [CrossRef]
  219. Rahman, M.U.; Bilal, M.; Shah, J.A.; Kaushik, A.; Teissedre, P.L.; Kujawska, M. CRISPR-Cas9-based technology and its relevance to gene editing in Parkinson’s disease. Pharmaceutics 2022, 14, 1252. [Google Scholar] [CrossRef]
  220. Shan, H.; Fei, T. CRISPR screening in cardiovascular research. Front. Cell Dev. Biol. 2023, 11, 1175849. [Google Scholar] [CrossRef] [PubMed]
  221. Mirgayazova, R.; Khadiullina, R.; Chasov, V.; Mingaleeva, R.; Miftakhova, R.; Rizvanov, A.; Bulatov, E. Therapeutic editing of the TP53 gene: Is CRISPR/Cas9 an option? Genes 2020, 11, 704. [Google Scholar] [CrossRef]
  222. Lin, C.H.; Vu, J.P.; Yang, C.Y.; Sirisawad, M.; Chen, C.T.; Dao, H.; Liu, J.; Ma, X.; Pan, C.; Cefalu, J.; et al. Glutamate-cysteine ligase catalytic subunit as a therapeutic target in acute myeloid leukemia and solid tumors. Am. J. Cancer Res. 2021, 11, 2911–2927. [Google Scholar]
  223. Li, J.M.; Newburger, P.E.; Gounis, M.J.; Dargon, P.; Zhang, X.; Messina, L.M. Local arterial nanoparticle delivery of siRNA for NOX2 knockdown to prevent restenosis in an atherosclerotic rat model. Gene Ther. 2010, 17, 1279–1287. [Google Scholar] [CrossRef]
  224. Koenig, O.; Walker, T.; Perle, N.; Zech, A.; Neumann, B.; Schlensak, C.; Wendel, H.-P.; Nolte, A. New aspects of gene-silencing for the treatment of cardiovascular diseases. Pharmaceuticals 2013, 6, 881–914. [Google Scholar] [CrossRef]
  225. Ma, M.W.; Wang, J.; Zhang, Q.; Wang, R.; Dhandapani, K.M.; Vadlamudi, R.K.; Brann, D.W. NADPH oxidase in brain injury and neurodegenerative disorders. Mol. Neurodegener. 2017, 12, 7. [Google Scholar] [CrossRef] [PubMed]
  226. Zhou, P.; Zhao, M.W.; Li, X.X.; Yu, W.Z.; Bian, Z.M. siRNA targeting mammalian target of rapamycin (mTOR) attenuates experimental proliferative vitreoretinopathy. Curr. Eye Res. 2007, 32, 973–984. [Google Scholar] [CrossRef] [PubMed]
  227. Zhang, C.; Liu, J.; Jin, N.; Zhang, G.; Xi, Y.; Liu, H. SiRNA targeting mTOR effectively prevents the proliferation and migration of human lens epithelial cells. PLoS ONE 2016, 11, e0167349. [Google Scholar] [CrossRef] [PubMed]
  228. Weiss, A.; Gilbert, J.W.; Rivera Flores, I.V.; Belgrad, J.; Ferguson, C.; Dogan, E.O.; Wightman, N.; Mocarski, K.; Echeverria, D.; Summers, A.; et al. RNAi-mediated silencing of SOD1 profoundly extends survival and functional outcomes in ALS mice. bioRxiv 2024. [Google Scholar] [CrossRef]
  229. Iannitti, T.; Scarrott, J.M.; Likhite, S.; Coldicott, I.R.; Lewis, K.E.; Heath, P.R.; Higginbottom, A.; Myszczynska, M.A.; Milo, M.; Hautbergue, G.M.; et al. Translating SOD1 gene silencing toward the clinic: A highly efficacious, off-target-free, and biomarker-supported strategy for fALS. Mol. Ther. Nucleic Acids 2018, 12, 75–88. [Google Scholar] [CrossRef]
  230. Zhang, W.; Liu, Y.; Liao, Y.; Zhu, C.; Zou, Z. GPX4, ferroptosis, and diseases. Biomed. Pharmacother. 2024, 174, 116512. [Google Scholar] [CrossRef]
  231. Sun, S.; Shen, J.; Jiang, J.; Wang, F.; Min, J. Targeting ferroptosis opens new avenues for the development of novel therapeutics. Signal Transduct. Target. Ther. 2023, 8, 372. [Google Scholar] [CrossRef]
  232. Wang, Y.; Wu, S.; Li, Q.; Sun, H.; Wang, H. Pharmacological inhibition of ferroptosis as a therapeutic target for neurodegenerative diseases and strokes. Adv. Sci. 2023, 10, e2300325. [Google Scholar] [CrossRef]
  233. Dinkova-Kostova, A.T.; Kostov, R.V.; Kazantsev, A.G. The role of Nrf2 signaling in counteracting neurodegenerative diseases. FEBS J. 2018, 285, 3576–3590. [Google Scholar] [CrossRef]
  234. Kunst, C.; Schmid, S.; Michalski, M.; Tümen, D.; Buttenschön, J.; Müller, M.; Gülow, K. The influence of gut Microbiota on oxidative stress and the immune system. Biomedicines 2023, 11, 1388. [Google Scholar] [CrossRef]
  235. Kim, H.; Kim, J.-S.; Kim, Y.; Jeong, Y.; Kim, J.-E.; Paek, N.-S.; Kang, C.-H. Antioxidant and probiotic properties of lactobacilli and bifidobacteria of human origins. Biotechnol. Bioprocess Eng. 2020, 25, 421–430. [Google Scholar] [CrossRef]
  236. Virk, M.S.; Virk, M.A.; He, Y.; Tufail, T.; Gul, M.; Qayum, A.; Rehman, A.; Rashid, A.; Ekumah, J.-N.; Han, X.; et al. The anti-inflammatory and curative exponent of probiotics: A comprehensive and authentic ingredient for the sustained functioning of major human organs. Nutrients 2024, 16, 546. [Google Scholar] [CrossRef] [PubMed]
  237. Carlson, J.L.; Erickson, J.M.; Lloyd, B.B.; Slavin, J.L. Health effects and sources of prebiotic dietary fiber. Curr. Dev. Nutr. 2018, 2, nzy005. [Google Scholar] [CrossRef] [PubMed]
  238. Nance, S.A.; Duncan, A.V.; Gwathmey, T.M.; Hairston, K.G. Soluble dietary fiber in obesity-associated inflammation and oxidative stress in African American women. FASEB J. 2017, 31, 434.2. [Google Scholar] [CrossRef]
  239. Sánchez, D.; Quiñones, M.; Moulay, L.; Muguerza, B.; Miguel, M.; Aleixandre, A. Soluble fiber-enriched diets improve inflammation and oxidative stress biomarkers in Zucker fatty rats. Pharmacol. Res. 2011, 64, 31–35. [Google Scholar] [CrossRef]
  240. Gorrini, C.; Harris, I.S.; Mak, T.W. Modulation of oxidative stress as an anticancer strategy. Nat. Rev. Drug Discov. 2013, 12, 931–947. [Google Scholar] [CrossRef]
Figure 1. Role of oxidative stress in cancer development and progression. Reactive oxygen species (ROS) may contribute to tumor initiation, progression, metastasis, and therapy resistance by activating signaling pathways involved in cancer cell proliferation and metastatization, angiogenesis, and expansion of the cancer stem cell (CSC) compartment. Furthermore, oxidative stress plays a key role in the regulation of immune responses in the tumor microenvironment (TME). Original image created using Microsoft PhotoDraw (version 2.0.0.0822). MDSCs: myeloid-derived suppressor cells. Tregs: T regulatory cells.
Figure 1. Role of oxidative stress in cancer development and progression. Reactive oxygen species (ROS) may contribute to tumor initiation, progression, metastasis, and therapy resistance by activating signaling pathways involved in cancer cell proliferation and metastatization, angiogenesis, and expansion of the cancer stem cell (CSC) compartment. Furthermore, oxidative stress plays a key role in the regulation of immune responses in the tumor microenvironment (TME). Original image created using Microsoft PhotoDraw (version 2.0.0.0822). MDSCs: myeloid-derived suppressor cells. Tregs: T regulatory cells.
Antioxidants 13 01109 g001
Table 1. Active clinical trials testing antioxidants for cancer prevention and treatment.
Table 1. Active clinical trials testing antioxidants for cancer prevention and treatment.
NCT NumberStudy TitleConditionsPhases
NCT02891538Chemopreventive Effects of Epigallocatechin Gallate (EGCG) in Colorectal Cancer (CRC) PatientsColon CancerEARLY PHASE1
NCT05932511Topical Ascorbic Acid for Treatment of Squamous Cell Skin CancerSquamous Cell Cancer; Squamous Cell Carcinoma; Skin Cancer; Non-melanoma Skin CancerEARLY PHASE1
NCT05985278Clinical Application of Lutetium [177Lu]-Catalase in Tumor Radionuclide TherapyAdvanced Malignant NeoplasmEARLY PHASE1
NCT01752491A Phase I Trial of High-Dose Ascorbate in Glioblastoma MultiformeGlioblastoma; GBM; Glioblastoma MultiformePHASE1
NCT01852890Gemcitabine, Ascorbate, Radiation Therapy for Pancreatic CancerPancreatic NeoplasmsPHASE1
NCT03278925Defined Green Tea Catechin Extract in Preventing Liver Cancer in Participants With CirrhosisCirrhosisPHASE1
NCT03602235High Dose Ascorbic Acid for Plasma Cell DisordersMultiple MyelomaPHASE1
NCT04900792A Safety Study of Pharmacologic Ascorbate and Ferumoxytol in Addition to Standard of Care Chemoradiation in GlioblastomaGlioblastoma; Glioblastoma MultiformePHASE1
NCT04952129Optimal Selenium for Bowel Polyps (OSCAR)Colorectal AdenomaPHASE1
NCT05081479A Study of N-Acetylcysteine (N-AC)in People Receiving CAR T cell Therapy for LymphomaLymphomaPHASE1
NCT04046094Intravenous (IV) Vitamin C With Chemotherapy for Cisplatin Ineligible Bladder Cancer PatientsBladder CancerPHASE1 PHASE2
NCT05123365An Optimal Dose Finding Study of N-Acetylcysteine in Patients With Myeloproliferative NeoplasmsMyeloproliferative Neoplasm; MPN; Essential Thrombocythemia; Polycythemia Vera; MyelofibrosisPHASE1 PHASE2
NCT05363631Seleno-L Methionine (SLM)-Axitinib-PembrolizumabClear Cell Renal Cell Carcinoma; Clear Cell Renal Cell Carcinoma MetastaticPHASE1 PHASE2
NCT05721872Efficacy, Tolerability and Safety of Intravenous D-VC With ATO in Patients With Advanced/Metastatic Colorectal CancerColorectal Cancer; Metastatic Colorectal CancerPHASE1 PHASE2
NCT01871454Safety of Pentoxifylline and Vitamin E With Stereotactic Ablative Radiotherapy (SABR) in Non-small Cell Lung CancersNon-small Cell Lung CancersPHASE2
NCT02344355A Phase 2 Trial of High-Dose Ascorbate in Glioblastoma MultiformeGlioblastoma MultiformePHASE2
NCT02705300Side Effects to FOLFOXIRI + Tocotrienol/Placebo as First Line Treatment of Metastatic Colorectal CancerColorectal CancerPHASE2
NCT02905578A Phase 2 Trial of High-dose Ascorbate for Pancreatic Cancer (PACMAN 2.1)Pancreatic Neoplasms; Cancer of Pancreas; Cancer of the Pancreas; Neoplasms, Pancreatic; Pancreas Cancer; Pancreas Neoplasms; AdenocarcinomaPHASE2
NCT02905591A Phase 2 Study Adding Ascorbate to Chemotherapy and Radiation Therapy for NSCLCCarcinoma, Non-Small-Cell Lung; Non-Small Cell Lung Cancer; Nonsmall Cell Lung Cancer; Non-Small-Cell Lung Carcinoma; NSCLCPHASE2
NCT03418038Ascorbic Acid and Chemotherapy for the Treatment of Relapsed or Refractory Lymphoma, CCUS, and Chronic Myelomonocytic LeukemiaClonal Cytopenia of Undetermined Significance; High Grade B-Cell Lymphoma With MYC and BCL2 or BCL6 Rearrangements; Recurrent Diffuse Large B-Cell Lymphoma; Recurrent Hodgkin Lymphoma; Recurrent Lymphoma; Refractory Diffuse Large B-Cell Lymphoma; Refractory Lymphoma; Chronic Myelomonocytic LeukemiaPHASE2
NCT03476330Quercetin Chemoprevention for Squamous Cell Carcinoma in Patients With Fanconi AnemiaFanconi Anemia; Squamous Cell CarcinomaPHASE2
NCT04033107High Dose Vitamin C Combined With Metformin in the Treatment of Malignant TumorsHepatocellular Cancer; Pancreatic Cancer; Gastric Cancer; Colorectal CancerPHASE2
NCT04175470Bevacizumab and Tocotrienol in Recurrent Ovarian CancerOvarian Cancer RecurrentPHASE2
NCT04245865Tocotrienol and Bevacizumab in Metastatic Colorectal CancerColorectal Cancer MetastaticPHASE2
NCT04801511Preoperative IMRT With Concurrent High-dose Vitamin C and mFOLFOX6 in Locally Advanced Rectal CancerRectal CancerPHASE2
NCT05724329Neoadjuvant Tislelizumab in Combination With Dasatinib and Quercetin in Resectable HNSCC (COIS-01)Head and Neck Squamous Cell CarcinomasPHASE2
NCT06087237The Efficacy of Using Pentoxifylline in Patients Undergoing Breast Cancer SurgeryPost-Surgical Complication; Breast Cancer SurgeryPHASE2
NCT06176339Assessing the Clinical Utility of Adding Pentoxifylline to Neoadjuvant Chemotherapy Protocols in Breast Cancer Patients”Breast CancerPHASE2
NCT06355037Dasatinib Combined With Quercetin to Reverse Chemo Resistance in Triple Negative Breast CancerTriple-negative Breast CancerPHASE2
NCT06398405A Phase II Clinical Study of Epigallocatechin-3-gallate in Patients With Esophageal Squamous CancerEsophageal Cancer; Dysphagia, Esophageal; Epigallocatechin GallatePHASE2
NCT06115408Evaluation of Using Dienogest and N-Acetyl Cysteine on the Volume of Uterine LeiomyomaUterine LeiomyomaPHASE2 PHASE3
NCT05364008FRIEND: Fibroids and Unexplained Infertility Treatment With Epigallocatechin Gallate; A Natural CompounD in Green TeaUterine LeiomyomaPHASE3
NCT05448365Vitamin D, Epigallocatechin Gallate, D-chiro-inositol and Vitamin B6 in Uterine FibroidUterine Fibroids; D-chiro-inositol; Inositol; Epigallocatechin Gallate; Vitamin D; Vitamin B6PHASE3
NCT05502900Adjuvant Melatonin for Uveal MelanomaUveal Melanoma; Uveal Melanoma, Posterior, Medium/Large Size; Eye Cancer, Intraocular MelanomaPHASE3
NCT05631041Effect of Silymarin in Metastatic Colorectal Cancer PatientsMetastatic Colorectal CancerPHASE3
Table 2. Effects of antioxidant supplementation in cancer treatment.
Table 2. Effects of antioxidant supplementation in cancer treatment.
AntioxidantsConditionsDosage and Additional SubstancesResults
PhytosterolsBreast cancer920 g/week of extra
virgin olive oil
62% decrease in the risk of malignant breast cancer [114]
30 g/day of mixed nutsNon-significant decrease in the risk of breast cancer [114]
Beta-caroteneLung cancer20 mg/dayInverse association between dietary intake and the risk of lung cancer; higher mortality among recipients of beta carotene [115]
20 mg + 50 mg/day of
alpha-tocopherol
Breast cancerDaily intake of 5 vegetable servings, 454 g of vegetable juice or vegetable equivalents, 3 fruit servings, 30 g fiber, and 15–20% energy intake from fat21% decrease in the risk of malignant breast cancer [116]
Cervical intraepithelial neoplasia (CIN)30 mg/dayNon-significant effect [117]
30 mg/dayResponse rate of 70%
at 6 months and 43%
at 12 months [118]
Basal-cell carcinoma (BCC)
Squamous-cell
carcinoma (SCC)
30 mg/day + and a sun protection factor 15+ sunscreenSlightly lower incidence for BCC (not significant);
Slightly higher incidence of squamous-cell carcinoma (SCC) (not significant) [119]
15 mg + 0.3 g vitamin C
30 mg/day
Lung cancer30 mg + 25,000 IU retinil palmitateLack of chemopreventive benefit and increase of lung cancer incidence and mortality [120,121,122]
Non-melanoma (NMSC) skin cancer50 mg/dayNo effect on the incidence of NMSC [123]
50 mg on alternate daysNo effect on the incidence of the first NMSC, including BCC and SCC, prevention [124]
ResveratrolMultiple myeloma (MM)5 g/dayMinimal efficacy in patients with relapsed/refractory MM; Poorly tolerated side effects [125]
Prostate cancerCurcumin 0.1 g/day; resveratrol 30 mg/day; catechins 0.1 g/day; fresh broccoli sprouts equivalent to 2 g/dayNon-significant increase in the slope of PSA [126]
Muscadine Grape Skin Extract (MPX)Prostate cancer4 g/day of MPX, 0.5 g/day of MPX (1.2 mg of ellagic acid, 9.2 μg of quercetin, and 4.4 μg of trans-resveratrol)No significant difference in PSA Doubling Time [127]
1.2 mg of ellagic acid, 9.2 μg of quercetin, and 4.4 μg of trans-resveratrol
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

Di Carlo, E.; Sorrentino, C. Oxidative Stress and Age-Related Tumors. Antioxidants 2024, 13, 1109. https://doi.org/10.3390/antiox13091109

AMA Style

Di Carlo E, Sorrentino C. Oxidative Stress and Age-Related Tumors. Antioxidants. 2024; 13(9):1109. https://doi.org/10.3390/antiox13091109

Chicago/Turabian Style

Di Carlo, Emma, and Carlo Sorrentino. 2024. "Oxidative Stress and Age-Related Tumors" Antioxidants 13, no. 9: 1109. https://doi.org/10.3390/antiox13091109

APA Style

Di Carlo, E., & Sorrentino, C. (2024). Oxidative Stress and Age-Related Tumors. Antioxidants, 13(9), 1109. https://doi.org/10.3390/antiox13091109

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