ArticlePDF AvailableLiterature Review

Reactive Oxygen Species (ROS): Key Components in Cancer Therapies

Authors:
  • School of Pharmacy and Life Sciences, Centurion University of Technology & Management, Bhubaneswar, Khurda-752050, Odisha, India
  • Indira Gandhi University

Abstract and Figures

Reactive oxygen species (ROS) refer to the highly reactive substances, which contain oxygen radicals. Hypochlorous acid, peroxides, superoxide, singlet oxygen, alpha-oxygen and hydroxyl radicals are the major examples of ROS. Generally, the reduction of oxygen (O2) in molecular form produces superoxide (•O2−) anion. ROS are produced during a variety of biochemical reactions within the cell organelles, such as endoplasmic reticulum, mitochondria and peroxisome. Naturally, ROS are also formed as a byproduct of the normal metabolism of oxygen. The production of ROS can be induced by various factors such as heavy metals, tobacco, smoke, drugs, xenobiotics, pollutants and radiation. From various experimental studies, it is reported that ROS act as either tumor suppressing or tumor promoting agent. The elevated levels of ROS can arrest the growth of tumor through the persistent increase in cell cycle inhibition. The increased level of ROS can induce apoptosis by both intrinsic and extrinsic pathways. ROS are considered to be tumor suppressing agent as the production of ROS is due to the use of most of the chemotherapeutic agents in order to activate the cell death. The cytotoxic effect of ROS provides impetus towards apoptosis, but in higher levels, ROS can cause initiation of malignancy that leads to uncontrolled cell death in cancer cells. Whereas, some species of ROS can influence various activities at the cellular level that include cell proliferation. This review highlights the genesis of ROS within cells by various routes and their role in cancer therapies.
Content may be subject to copyright.
Anti-Cancer Agents in Medicinal Chemistry
The journal for in-depth reviews and high quality research
papers on Anti-Cancer Agents
Impact
Factor:
2.505
ISSN: 1871-5206
eISSN: 1875-5992
Send Orders for Reprints to reprints@benthamscience.net
Anti-Cancer Agents in Medicinal Chemistry, 2022, 22, 215-222
215
REVIEW ARTICLE
Reactive Oxygen Species (ROS): Key Components in Cancer Therapies
Biswa Mohan Sahoo1,*, Bimal Krishna Banik2, Preetismita Borah3 and Adya Jain4
1Roland Institute of Pharmaceutical Sciences (Biju Patnaik University of Technology Nodal Centre of Research), Berhampur-760010,
Odisha, India; 2Department of Mathematics and Natural Sciences, College of Sciences and Human Studies, Prince Mohammad Bin
Fahd University, Al Khobar, Kingdom of Saudi Arabia; 3CSIR-Central Scientific Instruments Organization, Chandigarh, India;
4Department of Chemistry, MRK Educational Institutions, IGU Rewari, Haryana, India
A R T I C L E H I S T O R Y
Received: November 12, 2020
Revised: March 31, 2021
Accepted: April 05, 2021
DOI:
10.2174/1871520621666210608095512
Abstract: Reactive Oxygen Species (ROS) refers to the highly reactive substances which contain oxygen radicals.
Hypochlorous acid, peroxides, superoxide, singlet oxygen, alpha-oxygen, and hydroxyl radicals are the major exam-
ples of ROS. Generally, the reduction of oxygen (O2) in molecular form produces superoxide (•O2
) anion. ROS are
produced during a variety of biochemical reactions within the cell organelles, such as endoplasmic reticulum, mito-
chondria, and peroxisome. Naturally, ROS are also formed as a byproduct of the normal metabolism of oxygen. The
production of ROS can be induced by various factors such as heavy metals, tobacco, smoke, drugs, xenobiotics, pollut-
ants, and radiation. From various experimental studies, it is reported that ROS acts as either a tumor-suppressing or a
tumor-promoting agent. The elevated level of ROS can arrest the growth of tumors through the persistent increase in
cell cycle inhibition. The increased level of ROS can induce apoptosis by both intrinsic and extrinsic pathways. ROS is
considered to be a tumor-suppressing agent as the production of ROS is due to the use of most of the chemotherapeutic
agents in order to activate cell death. The cytotoxic effect of ROS provides impetus towards apoptosis, but in higher
levels, ROS can cause initiation of malignancy that leads to uncontrolled cell death in cancer cells. In contrast, some
species of ROS can influence various activities at the cellular level, including cell proliferation. This review highlights
the genesis of ROS within cells by various routes and their role in cancer therapies.
Keywords: Reactive oxygen species, generation, cancer, pathway, therapy, free radical.
1. INTRODUCTION
Reactive Oxygen Species (ROS) are defined as reactive, unsta-
ble, and reduced forms of oxygen derivatives that are produced as a
by-product of normal metabolic processes [1]. ROS mainly com-
prise superoxide anion (O2-), singlet oxygen (1O2), hydrogen per-
oxide (H2O2), hypochlorous acid (HOCl), and hydroxyl radical
(OH). The reactive nature of ROS is due to the presence of extra-
unpaired electrons at outer shells and their half-life. The half-life
ROS is measured in milliseconds [2]. The hydroxyl radical is the
primary damaging species because it cannot be easily scavenged in
the human body. ROS acts as a secondary messenger in cell signal-
ing pathways and plays a major role in various biological processes
in the case of normal and cancer cells [3]. From various studies, it is
reported that ROS acts as a tumor-suppressing or promoting agent.
ROS is considered a tumor-suppressing agent when the production
of ROS is a mechanism shared by most of the chemotherapeutic
agents due to their involvement in producing cell death [4]. How-
ever, some species of ROS can affect various functions at the cellu-
lar level, including cell proliferation. In the case of cancerous cells,
ROS are usually observed as oncogenic because these are associ-
ated with the initiation, progression, and metastasis of tumors [5].
However, this hypothesis is not clear as ROS may also be crucial
for clearance of tumor. The effect of ROS in the development of
tumors involves damage of DNA directly during the transformation
of carcinogenesis [6].
The function of ROS is observed as conflicting in cancer che-
motherapy. In some cases, the excess levels of ROS exhibit
*Address correspondence to this author at the Roland Institute of Pharmaceutical
Sciences (Biju Patnaik University of Technology Nodal Centre of Research), Berham-
pur-760010, Odisha, India; Tel: +91 -9040442719;
E-mail: drbiswamohansahoo@gm ail.com
anti-tumorigenic activities by inducing the cell cycle arrest and cell
death. In addition to this, it is reported that tumor cells elevate their
survival through augmentation of manipulation mechanisms of
reactive oxygen species that include elevated antioxidant levels or
increased ROS production to maintain the optimum balance of ROS
level that promotes proliferation and survival [7]. In the case of
cancer cells, the oxidative stress induced by drugs results in ROS
mediated cytotoxic effect that preferentially destroys tumor cells
and reduces their proliferation.
Similarly, the cancer cells comprise elevated levels of both
intracellular and extracellular reactive oxygen species. The en-
hanced level of intracellular ROS is due to inadequate production of
ROS or detoxification methods that can transform the normal cells
into m alignant cells. Therefore, attempts are made for the accurate
measurement of ROS in order to characterize their nature, localiza-
tion, and time-course of generation and spread at the cellular level.
This review is mainly focused on the role of ROS involved in dif-
ferent pathways of killing cancer cells during the development of
anti-cancer drugs for achieving good therapeutic efficacy and
minimal side effects. Therefore, there should be a better under-
standing of the molecular mechanisms of the signaling pathway of
ROS in cancer cells and the identification of specific ROS targets
that can provide novel therapeutic avenues for the treatment of
cancer [8].
2. SOURCES AND PRODUCTION OF ROS
ROS is grouped into two categories such as radical ROS and
non-radical ROS. Superoxide anion (O2
-), hydroxyl radical (OH),
nitric oxide (NO•), alkoxyl radicals (RO•), thiyl radicals (RS•),
sulphonyl radicals (ROS•), and lipid peroxyl radical (LOO-) are
the major examples of radical ROS whereas hydrogen peroxide
(H2O2), singlet oxygen (1O2), dinitrogen dioxide (N2O2), ozone (O3)
187
-
/22 $65.00+.00 © 2022 Bentham Science Publishers
216 Anti-Cancer Agents in Medicinal Chemistry, 2021, Vol. 22, No. 2 Saho o et al.
and hypochlorous acid (HOCl) are considered as non-radical ROS
(Table 1). ROS is commonly generated by different biochemical
and physiological oxidative processes inside the cellular system.
The generation of ROS is generally involved via two routes, includ-
ing (a) in the cell membranes by utilizing NADPH oxidases (b) in
the mitochondria by using the enzyme-like NADH oxidoreductase
as depicted in Fig. (1) [9-11].
Table 1. List of radical and non-radical reactive oxygen species.
Sl. No. Free radicals Non-radicals
1 Superoxide anion (O2-) Hydrogen peroxide (H2O2)
2 Hydroxyl radical ( OH) Hypochlorous acid (HOCl)
3 Lipid peroxyl (LOO-) Singlet oxygen (1O2)
4 Alkoxyl radicals (RO•) Dinitrogen dioxide (N2O2)
5 Sulphonyl radicals (ROS•) Ozone/trioxygen (O3)
Mitochondria and the enzyme systems are considered as the
main sources of generating the reactive oxygen species. The univa-
lent oxygen undergoes a reduction in the mitochondria to produce
superoxide anion (O2
-), which can be further decreased by the
action of mitochondrial superoxide dismutase (SOD) into non-
radical species like hydrogen peroxide (H2O2) and singlet oxygen
(1O2). This reaction involves the Electron Transport Chain (ETC) of
mitochondria without any implication of enzymatic activity by the
action of oxido-reductive reaction intermediates (Fig. 2) [12, 13].
The reactive oxygen species are generated at the intracellular
part by stimulation of nicotinamide adenine dinucleotide phosphate
(NADPH), oxidases, and peroxidases. NADPH-oxidase (NOX) is
present in different isomeric forms such as Nox1, Nox3, Nox4,
Nox5, Duox1, and Duox2. These isoforms are responsible for gen-
erating the different types of ROS in various normal or pathological
cells. The elevated level of ROS by NOX4 regulates the growth
factor (EGFR) involved in the survival of cells. NOX is one of the
major factors necessary for the immunosuppressant activity of
Myeloid-Derived Suppressor Cells (MDSCs). MDSCs are hetero-
geneous groups of cells that suppress the responses of T-cell and
increase during inflammation and cancer. MDSCs also induce the
progression of cancer via ROS. MDSCs are found to enhance angi-
ogenesis, cell division, and metastasis of tumors [14].
ROS is also generated as an alternate product by other enzyme
systems such as cytochrome-P450, Xanthine Oxidase (XO), and
nitric oxide synthase. The production pathway of ROS also in-
volves the conversion of the hypoxanthine into xanthine in the
presence of XO and xanthine into uric acid that yields superoxide
radicals. Similarly, the pathological conditions involving ischemia,
reperfusion, inflammation, and infection also play a significant role
in regulating the generation of ROS. It has been noted that there is
an increase in the production of ROS in endothelial cells induced by
tumor necrosis factor (TNF) [15].
Fig. (1). Sources of reactive oxygen species.
H2O2
Hydrogen
peroxide
OH
Hydroxy
radical
Cl
HOCl
Hypochlorous acid
NO2
NO2
Nitrogen dioxide
e + 2H+
O22
Peroxide
ion
NO
ONOO
Peroxy nitrate
HO2
Perhydroxyl radical
H+
e
O2
Superoxide
radical ion
e
3O2
Triplet
Oxygen
UV
O
Oxygen
radical
O31O2
Ozone S inglet Oxygen
O2Irradiation/
Photosensitizer
Fig. (2). Production of Reactive Oxygen Species (ROS).
Role of Reactive Oxygen Species in Cancer Therap ies Anti-Cancer Agents in Medicinal Chemistry, 2022, Vol. 22, No. 2 217
The smoke of cigarette is primarily composed of carcinogenic sub-
stances such as polycyclic aromatic hydrocarbons (PAHs) and nitrosa-
mines that causes accumulation of 8-hydroxydeoxyguanosine (8-
OHdG) in lungs. Due to this effect, cigarette smoking persons exhibit 2
to 3 folds higher level of 8-OHdG that leads to mutations, some of
which might be stimulated by oxygen free radicals, which results in
inflammatory responses, fibrosis, and development of tumor. ROS is
also produced from lymphocytes via 5-lipoxygenase (5-LO) pathway.
In contrast, the cyclooxygenase (COX) generates ROS after stimulation
of TNFα, interleukin-1, bacterial lipopolysaccharide, and tumor pro-
moter-4-O-Tetradecanoyl-Phorbol-13-Acetate (TPA). The neurotrans-
mitter dopamine undergoes oxidation to produce reactive oxygen spe-
cies [16].
Furthermore, most of the chemotherapeutic agents produce
ROS in cancer cells. Anthracyclines such as Doxorubicin, Daun-
orubicin, and Epirubicin cause an increase in the cellular levels of
ROS. Alkylating agents (Busulfan, Cyclophosphamide, Chloram-
bucil, Cisplatin, carmustin, thiotepa) and topoisomerase inhibitors
(Topoisomerase-I inhibitors: camptothecin, topotecan, irinotecan
and Topoisomerase-II inhibitors: doxorubicin, etoposide, Daunoru-
bicin, Epirubicin, mitoxantrone and epipodophyllotoxin) induce a
high level of cellular ROS. In contrast, vinca alkaloids, taxanes,
nucleotide analogues, and anti-metabolites, including anti-folates,
create lower levels of ROS. New chemotherapeutic agents are also
bio-engineered to target the specific compartments at the cellular
level for the generation of ROS and continue for a definite period of
time [17].
3. REGULATION OF ROS
Homeostasis of ROS is needed for the survival of normal cells
and suitable cell signaling. The decrease in the level of ROS can
stimulate signaling pathways to control the metabolic process, dif-
ferentiation and cellular proliferation in a controlled way. In com-
parison with normal cells, cancer cells cause increase in production
of localized ROS that hyperactivates the cell signaling pathways
essential for cellular transformation and carcinogenesis. The accu-
mulation of ROS can arise as a result of activation of the oncogene,
loss of tumor suppressor gene, increased metabolic activity, defi-
ciency of nutrient (low glucose), or low level of oxygen (hypoxia).
The excess levels of ROS can induce oxidative damage and cell
death. So, the cancer cells regulate the accumulation of ROS by
increasing their antioxidant capacity [18].
Cancer cells activate the transcription factor NRF2 (nuclear
factor erythroid-2 related factor-2) to enhance the expression of
antioxidants such as peroxiredoxins (PRXs), superoxide dismutases
(SODs), catalase (CAT), and glutathione peroxidases (GPXs). Can-
cer cells also trigger the genes involved in NADPH production,
glutathione (GSH) synthesis and enzymes detoxification. This type
of balance between production and elimination of ROS maintains
an optimum level of ROS for pro-tumorigenic signaling (Fig. 3)
[19].
4. METHODS FOR MEASUREMENT OF ROS
There is a significant challenge in bio-analytical techniques for
the estimation of reactive oxygen species in both cells and living
organisms. So, it is difficult to identify ROS directly in complex
biological systems because most of the ROS are highly reactive,
short-lived, and do not accumulate at elevated levels. Recently, the
advancement of analytical methods, especially in electron par-
amagnetic resonance and Mass Spectroscopy (MS), afford quantita-
tive and precise measurement of ROS at the cellular level. The
assay methods involved in the measurement o f ROS are susceptible
to numerous artifacts that result from the analytical techniques and
redox sensor (Table 2). These methods are inadequate in th eir func-
tion to distinguish among different types of ROS. The real-time
imaging of oxidative changes by applying redox-sensitive green
fluorescent proteins is recently utilized to provide information
about the location of ROS at the subcellular level. The elevated
levels of ROS are identified in cancer cells due to high metabolic
activity, cellular signaling, peroxisomal activity, mitochondrial
dysfunction, activation of oncogene, and increased enzymatic activ-
ity of oxidases, cyclooxygenases, lipoxygenases, and thymidine
phosphorylases [20, 21].
5. ROLE OF ROS IN CANCER THERAPIES
Cancer is recognized as one of the major heterogeneous dis-
eases with high morbidity and mortality with poor prognosis. It was
estimated that there are 18.1 million new cases of cancer and 9.6
Fig. (3). Balance of reactive oxygen species in cancer cells. (A higher resolution / colour version of this figure is available in the electronic copy of the arti-
cle).
Table 2. Detection methods for measurement of ROS.
Sl. No. Types of ROS Detection Methods
1 Superoxide anion (O2-) Cyto chrome-c reduction Chemiluminescence reactions Electron paramagnetic resonance
2 Hydroxyl radical (OH) Horseradish peroxidase assay Dichlorofluorescein fluorescen ce Dihydrorhodamine-123 fluorescence
3 Sin glet oxygen (1O2) Scanning-laser method I nfrared phosphorescence spectroscopy Electro n paramagnetic resonance spec-
troscopy
218 Anti-Cancer Agents in Medicinal Chemistry, 2021, Vol. 22, No. 2 Saho o et al.
million cancer deaths globally during 2018. Current developments
in different research also reveal that cancer is one of the leading
causes of death and will be a major barrier to increasing life expec-
tancy worldwide. Further, more than 90% of cancer deaths are
caused due to the metastasis of tumor cells. Metastasis is the condi-
tion in which the primary tumor cells spread to distant organs of the
body, and this process is considered the primary cause of morbidity
and mortality of cancer [22].
There are no suitable therapeutic approaches available to sup-
press the metastasis of cancer cells. However, the resistance devel-
oped by conventional chemotherapy and disease relapse remains a
persistent challenge clinically. These observations suggest that
there is no clear information regarding the mechanism of action at
the cellular level and biotic heterogeneity in the cancer cells. Clini-
cally, it is also observed that cancer may be a genetic disorder that
results from both internal factors (e.g., inherited mutations, immune
conditions, hormones) and external factors (e.g., environment, diet,
tobacco, diet, infection, radiation). These factors can also affect
essential genes, including proto-oncogenes, tumor suppressor
genes, and deoxyribonucleic acid repair genes via cellular interme-
diates like reactive oxygen species [23].
In the case of normal cells, ROS are produced in a highly con-
trolled manner as these are associated with the regulation of signal-
ing processes of cell division, immune regulation, autophagy, in-
flammation, and stress-related responses. However, the uncon-
trolled production of ROS can lead to oxidative stress and cytotox-
icity, which impart loss of cellular functions and development of
heterogeneous disease states like cancer. In the early stages of can-
cer, the intracellular ROS promote the initiation of cancer via in-
duction of oxidation and base-pair substitution mutations in pro-
oncogenes such as Ras and tumor suppressor genes such as p53.
Oxidative stress mainly contributes to aging and many other dis-
eases such as cancer, diabetes, and obesity. Oxidative stress arises
due to the excess accumulation of ROS in the cell. This is again due
to an imbalance of oxido-reductive activities that results in damage
to the cellular system, as presented in Fig. (4) [24].
Generally, it is reported that ROS may promote either cell pro-
liferation or cell death depending on the intensity or location of the
oxidative burst and th e activity of the antioxidant system. The abil-
ity of ROS to stimulate cell growth or cell death mainly d epends on
the intensity or duration of redox signals and the defense mecha-
nisms of antioxidants. The existing anti-cancer drugs exert harmful
effects on normal cells, which are partially activated by ROS. These
species exert reverse cellular effects by promoting either cell prolif-
eration and tumor progression or cell death. ROS act as “double-
edged sword” by acting not only as disease inducers or sustainers
but also therapeutic weapons in cancer cells. The increased level of
ROS in mitochondria is found to induce cell proliferation, cell sur-
vival, cell migration, and epithelial-mesenchymal transition through
mitogen-activated protein kinase (MAPK) and R as-ERK activation,
as presented in Fig. (5) and Table 3 [25, 26].
Edderkaoui et al. demonstrated that ROS produced by an ex-
tracellular matrix increases the survival of pancreatic cancer cells
through 5-lipoxygenase (LOX) and NOX [27]. For pancreatic can-
cer, some of the treatment strategies have proved to be effective
that include combination therapy of gemcitabine with trichostatin-
A, epigallocate-3-gallate (EGCG), capsaicin, and benzyl-
isothiocyanate (BITC) [28]. All of these drugs produce their action
based on the same mechanism via elevation of intracellular ROS
levels to trigger apoptosis (Fig. 6). Currently, FDA-approved non-
steroidal anti-inflammatory drugs (NSAIDs) like Sulindac are
tested for their potential in cancer therapy. Sulindac enhances the
intracellular level of ROS that renders colon and lung cancer cells
more sensitive to H2O2 promoted apoptosis [29, 30].
Similarly, the anticancer drug like Aminoflavone promotes cell
death in MCF-7 and MDA-MB-468 breast cancer cells. Chemi-
cally, it is 5-amino-2-(4-amino-3-fluorophenyl)-6,8-difluoro-7-
methylchromen-4-one. The treatment of cancer cells with
Fig. (4). Role of Reactive Oxygen Species (ROS) in carcinogenesis.
... revealed a significant increase in ROS levels in vivo and in vitro following treatment with 125 I seed, indicating the activation of ROS-mediated apoptotic pathways. ROS, known for their dual roles as signaling molecules and cytotoxic agents, play a crucial role in modulating cellular responses to radiation therapy (Sahoo et al. 2022). This aligns with previous research demonstrating that combined treatment of 125 I seed and salinomycin achieved enhanced growth inhibition and apoptosis in human glioma in vitro and in vivo through triggering ROS-mediated apoptosis (Chao et al. 2018;Chen and Wang 2017). ...
Article
Full-text available
Objectives Cholangiocarcinoma (CCA) is a rare tumor with a poor prognosis and poses significant therapeutic challenges. Herein, we investigated the mechanism of efficacy of ¹²⁵I seed implantation therapy in CCA, focusing on the induction of reactive oxygen species (ROS)-mediated apoptosis and the involvement of glutathione peroxidase 2 (GPX2). Materials and methods Human cholangiocarcinoma cell lines QBC939 and RBE were purchased for in vitro studies. In vivo studies were performed using a rabbit VX2 CCA model. Apoptosis and proliferation were detected by TUNEL staining and clone formation, respectively. ROS generation was detected by dihydroethidium staining. Histological evaluation was performed by hematoxylin and eosin staining. Protein expression was determined by Western blotting and immunohistochemistry. Results Our results demonstrate that ¹²⁵I seeds effectively inhibited tumor growth in the rabbit VX2 tumor model and promoted the apoptosis of CCA cells in vitro in a dose-dependent manner. Molecular analyses indicate a marked increase in reactive oxygen species (ROS) levels following treatment with ¹²⁵I seeds, suggesting the involvement of ROS-mediated apoptosis in the therapeutic mechanism. Furthermore, the downregulation of glutathione peroxidase 2 (GPX2) was observed, indicating its potential role in modulating ROS-mediated apoptosis in CCA. Conclusion ¹²⁵I seed implantation therapy exerts therapeutic effects on CCA by inducing ROS-mediated apoptosis. The downregulation of GPX2 may contribute to enhanced ROS accumulation and apoptotic cell death. These findings provide mechanistic insights into the therapeutic potential of ¹²⁵I seed implantation for CCA and highlight ROS-mediated apoptosis and GPX2 regulation as promising targets for further investigation and therapeutic intervention in this malignancy.
... The binding energies of ligands and targets and their interacting amino acids are presented in Table 1. (Sahoo et al., 2022). In this study, we found that β-Glucan and the quercetin loaded β-Glucan particles have the potential to generate an e cient amount of ROS in DU145 cells as shown in Fig. 9. Determination of nuclear morphology and DNA fragmentation with DAPI staining The in uence of particles on the DU145 cells was assessed with DAPI staining that is speci c for the binding with the nucleus. ...
Preprint
Full-text available
Background Prostate cancer remains a challenge in healthcare, being the second most common male cancer demanding innovative therapeutic approaches and treatment techniques. This study integrates in silico and in vitro methods for the investigation of the potential anticancer effects of quercetin-loaded and alginate-sealed β-Glucan particles derived from mushroom Agaricus bisporus and yeast against the DU145 cell line. Methods Prostate cancer-related genes were identified from DisGeNET and GeneCards databases, followed by target prioritization using Swiss Target Prediction software. Venny 2.1 was used for the determination of common targets between β-Glucan, Quercetin, and prostate cancer and PPI network was constructed using STRING database. CB dock online server was used for molecular docking and DU145, RAW264.7 cell lines were used for the determination of cytotoxicity against prostate cancer and healthy cells. Results Molecular docking revealed that quercetin has superior binding affinity compared to β-Glucan with selected prostate cancer-related targets. In vitro evaluation using MTT assays demonstrated the cytotoxic effects of quercetin-loaded and alginate-sealed particles against DU145 prostate cancer cells. Apoptosis induction, ROS generation, and lysosomal pH alterations underscore the potential of quercetin-loaded and alginate-sealed β-Glucan particles as promising therapeutic agents for prostate cancer. Conclusions Our study showed systematic analyses of the effect of hollow β-Glucan particles, Quercetin, and Quercetin alginate sealed particles against DU145 cells and found that formulation exhibits superior anticancer activity against prostate cancer cell line. Quercetin-loaded alginate-sealed particles showed very little cytotoxicity against healthy cell line RAW264.7. Future studies focusing on preclinical validation, pharmacokinetic profiling, and clinical trials to assess translational potential and optimize therapeutic strategies can help get impactful findings.
... Reactive oxygen species (ROS) plays an important role in regulating signals in the cell and participates in the whole aerobic process of the organism. When ROS accumulates in excess in the cell, it will promote the formation of oxidative stress [1]. ROS substances include hydroxyl radical, superoxide ion, hydrogen peroxide (H 2 O 2 ) singlet oxygen and hypochlorite [2][3][4]. ...
Article
Full-text available
In this study, a ratiometric fluorescence nanoprobe is developed for the analysis of hydrogen peroxide (H2O2). Silver nanoclusters (AgNCs) were synthesized by chemical reduction method using sodium borohydride (NaBH4) as reducing agent, and were coupled with CdSe/ZnS quantum dots (QDs) to form the ratiometric fluorescence nanoprobe silver nanoclusters-quantum dots (AgNCs-QDs). The effect of the volume ratio of CdSe/ZnS QDs to AgNCs on the fluorescence ratio of AgNCs-QDs was investigated. The fluorescence characterization results show that two emission peaks of AgNCs-QDs are located at 473 nm and 661 nm, respectively. Transmission electron microscope (TEM) and X-ray photoelectron spectroscopy (XPS) results show that H2O2 can cause the fluorescence probe to aggregate, while etching AgNCs to produce silver ions, which together cause the fluorescence of the QDs in the ratiometric fluorescent probe to be quenched. Based on this strategy, the fluorescence intensity ratio of the two emission peaks F473/F661 exhibits a strong linear correlation with the concentration of H2O2. The detection range is 3.32 µM ~ 2.65 mM with a detection limit of 3.32 µM. In addition, the ratiometric fluorescence probe can specifically recognize H2O2 and has excellent anti-interference performance and good fluorescence stability. Importantly, the probe was utilized for the detection of H2O2 in serum, showing the possibility of the probe in clinical detection applications.
... This comprehensive profiling enhances our ability to harness the beneficial properties of A. arabicum justifying any potential risks associated with its toxicity. Various research indicates that reactive oxygen species (ROS) are associated with several human diseases, encompassing cancer, autoimmune and cardiovascular conditions, ulcers, inflammation, and diabetes (Dumanović et al. 2021;Mansoor et al. 2022;Sahoo et al. 2022). Research is currently exploring the chromatographic, phytochemical and antioxidant properties from leaves extracts of A. arabicum. ...
Article
Full-text available
The inherent natural antioxidants found in numerous medicinal plants (MPs) play a crucial role in counteracting the detrimental impacts of oxidative stress. MPs contains polyphenols and flavonoids, which serve as scavengers for free radicals, mitigating oxidative stress and offering potential as alternative treatments for a range of human ailments. The significance of this study lies in its chromatographic spectrum identification, providing a in-depth exploration of the antioxidant capacities and toxicological profiles within the genetic resources of Argyrolobium arabicum. This study described the chromatographic, spectroscopic, antioxidant and hepatoprotective actions of A. arabicum leaves collected from arid zones of Saudi Arabia. Argyrolobium arabicum chromatographic separations of bioactive compounds (HPLC detection isolate prominent compounds D-Pinitol and vitexin) and GC–MS spectra identified five phenolics and six flavonoids organic compounds. FT-IR spectroscopy revealed prominent functional group of alcohols and carboxylic acids bonds stretching. The hepatic toxicity and protective effects were notably administrated of A. arabicum extracts at a dosage of 300 mg/kg, leading to the restoration of ALT levels to 51 (U/L), AST levels to 71.6 (U/L), ALP levels to 169 U/L, and total bilirubin levels to 0.58 (g/dL), which had been altered by the negative control. This research basically contributing to the understanding of medicinally important A. arabicum potential applications in pharmaceutical and environmental fields and used as to elucidate the complex genomic mechanisms explaining the bioactivity and safety of medicinal species.
... Mitochondrial oxidative phosphorylation is a key cellular process to generate ATP (for detailed descriptions and additional references, see [81,82,[90][91][92][93][94][95]). Furthermore, reactive oxygen species are reactive, unstable and reduced forms of oxygen derivatives that are metabolic by-products [96] formed especially by mitochondrial metabolism [90]. Reactive oxygen species promote genomic instability and can thereby contribute to leukemogenesis and chemoresistance [97][98][99], but they also modulate intracellular signaling through the oxidation of lysine residues of, for example, redox-sensitive transcription factors and enzymes [100]. ...
Article
Full-text available
We review the importance of monocytic differentiation and differentiation induction in non-APL (acute promyelocytic leukemia) variants of acute myeloid leukemia (AML), a malignancy characterized by proliferation of immature myeloid cells. Even though the cellular differentiation block is a fundamental characteristic, the AML cells can show limited signs of differentiation. According to the French–American–British (FAB-M4/M5 subset) and the World Health Organization (WHO) 2016 classifications, monocytic differentiation is characterized by morphological signs and the expression of specific molecular markers involved in cellular communication and adhesion. Furthermore, monocytic FAB-M4/M5 patients are heterogeneous with regards to cytogenetic and molecular genetic abnormalities, and monocytic differentiation does not have any major prognostic impact for these patients when receiving conventional intensive cytotoxic therapy. In contrast, FAB-M4/M5 patients have decreased susceptibility to the Bcl-2 inhibitor venetoclax, and this seems to be due to common molecular characteristics involving mitochondrial regulation of the cellular metabolism and survival, including decreased dependency on Bcl-2 compared to other AML patients. Thus, the susceptibility to Bcl-2 inhibition does not only depend on general resistance/susceptibility mechanisms known from conventional AML therapy but also specific mechanisms involving the molecular target itself or the molecular context of the target. AML cell differentiation status is also associated with susceptibility to other targeted therapies (e.g., CDK2/4/6 and bromodomain inhibition), and differentiation induction seems to be a part of the antileukemic effect for several targeted anti-AML therapies. Differentiation-associated molecular mechanisms may thus become important in the future implementation of targeted therapies in human AML.
... The glutathione system serves as a vital antioxidant defense mechanism against ROS, playing a critical role in the maintenance of cellular redox equilibrium and normal physiological functions by regulating the balance of redox-regulated molecules such as NAD/NADH, NADP/NADPH, and GSH/GSSG ratios [3,84,85]. ...
Article
Full-text available
Glutathione (GSH), a prominent antioxidant in organisms, exhibits diverse biological functions and is crucial in safeguarding cells against oxidative harm and upholding a stable redox milieu. The metabolism of GSH is implicated in numerous diseases, particularly in the progression of malignant tumors. Consequently, therapeutic strategies targeting the regulation of GSH synthesis and metabolism to modulate GSH levels represent a promising avenue for future research. This study aimed to elucidate the intricate relationship between GSH metabolism and ferroptosis, highlighting how modulation of GSH metabolism can impact cellular susceptibility to ferroptosis and consequently influence the development of tumors and other diseases. The paper provides a comprehensive overview of the physiological functions of GSH, including its structural characteristics, physicochemical properties, sources, and metabolic pathways, as well as investigate the molecular mechanisms underlying GSH regulation of ferroptosis and potential therapeutic interventions. Unraveling the biological role of GSH holds promise for individuals afflicted with tumors.
... These results provide evidence supporting the adverse effects of cigarette smoking on liver fibrosis and cirrhosis. Cigarette smoke contains reactive (53)(54)(55). Several studies have shown that oxidative stress plays an important role in the development of liver fibrosis and cirrhosis (56)(57)(58). ...
Article
Full-text available
Background Liver fibrosis significantly impacts public health globally. Untreated liver fibrosis eventually results in cirrhosis. Cigarette smoking is the main etiologic factor for various diseases. However, the causal effects of cigarette smoking on liver fibrosis and cirrhosis have yet to be fully elucidated. Methods In this study, Mendelian randomization (MR) analysis was performed to assess the association between cigarette smoking, liver fibrosis, and cirrhosis. Single-nucleotide polymorphisms (SNPs) were selected as instrumental variables from a genome-wide association study (GWAS) of European ancestry. Patients were divided into six exposure categories as follows: “ever smoked,” “pack years of smoking,” “age of smoking initiation,” “smoking status: never,” “smoking status: current,” and “smoking status: previous.” The outcomes of this study included liver fibrosis and cirrhosis. MR-Egger, weighted median, inverse variance weighted, simple mode, and weighted mode were selected as the analysis methods. Cochran’s Q and the MR-PRESSO tests were conducted to measure heterogeneity. The MR-Egger method was performed to evaluate horizontal pleiotropy, while the “leave-one-out” analysis was performed for sensitivity testing. Results The results of this study showed that having a smoking history increases the risk of liver fibrosis and cirrhosis [“ever smoked”: odds ratio (OR) = 5.704, 95% CI: 1.166–27.910, p = 0.032; “smoking status: previous”: OR = 99.783, 95% CI: 2.969–3.353e+03, p = 0.010]. A negative correlation was observed between patients who never smoked and liver fibrosis and cirrhosis (“smoking status: never”: OR = 0.171, 95% CI: 0.041–0.719, p = 0.016). However, there were no significant associations between “smoking status: current,” “pack years of smoking,” and “age of smoking initiation” and liver fibrosis and cirrhosis. Cigarette smoking did not have a significant horizontal pleiotropic effect on liver fibrosis and cirrhosis. The “Leave-one-out” sensitivity analysis indicated that the results were stable. Conclusion The study confirmed the causal effects of cigarette smoking on liver fibrosis and cirrhosis.
... ROS are highly reactive and can cause oxidative stress, damaging vital cellular components like proteins, lipids, and DNA, especially in these radiosensitive cells. This leads to disruption in DNA repair processes and cell cycle progression, culminating in reduced cell viability and enhanced apoptosis [37]. This resulted in the observed synergistic effects. ...
Article
Full-text available
This research underscores the potential of combining nanotechnology with conventional therapies in cancer treatment, particularly for challenging cases like pancreatic cancer. We aimed to enhance pancreatic cancer treatment by investigating the synergistic effects of gold nanoparticles (GNPs) and docetaxel (DTX) as potential radiosensitizers in radiotherapy (RT) both in vitro and in vivo, utilizing a MIA PaCa-2 monoculture spheroid model and NRG mice subcutaneously implanted with MIA PaCa-2 cells, respectively. Spheroids were treated with GNPs (7.5 μg/mL), DTX (100 nM), and 2 Gy of RT using a 6 MV linear accelerator. In parallel, mice received treatments of GNPs (2 mg/kg), DTX (6 mg/kg), and 5 Gy of RT (6 MV linear accelerator). In vitro results showed that though RT and DTX reduced spheroid size and increased DNA DSBs, the triple combination of DTX/RT/GNPs led to a significant 48% (p = 0.05) decrease in spheroid size and a 45% (p = 0.05) increase in DNA DSBs. In vivo results showed a 20% (p = 0.05) reduction in tumor growth 20 days post-treatment with (GNPs/RT/DTX) and an increase in mice median survival. The triple combination exhibited a synergistic effect, enhancing anticancer efficacy beyond individual treatments, and thus could be employed to improve radiotherapy and potentially reduce adverse effects.
... Oxidative mechanisms are crucial in both the induction and progression of cancer [46]. Elevated concentrations of reactive oxygen species (ROS) in cancer cells contribute to heightened oxidative stress, which inflicts damage on cellular components, ending in cell death [47]. Within biological systems, catalase (CAT) serves an essential role in safeguarding against the detrimental impacts of free radicals and ROS. ...
Article
In this study, novel substituted 1,3,5-triazine candidates (4a-d, 5a-j, and 6a-d) were designed as second-generation small molecules to act as dual IDH1 and IDH2 inhibitors according to the pharmacophoric features of both vorasidenib and enasidenib. Compounds 6a and 6b for leukemia cell lines showed from low to sub-micromolar GI50. Moreover, compounds 4c, 5f, and 6b described the frontier antitumor activity against THP1 and Kasumi Leukemia cancer cells with IC50 values of (10 and 12), (10.5 and 7), and (6.2 and 5.9) µg/mL, which were superior to those of cisplatin (25 and 28) µg/mL, respectively. Interestingly, compounds 4c, 6b, and 6d represented the best dual IDH1(R132H)/IDH2(R140Q) inhibitory potentials with IC50 values of (0.72 and 1.22), (0.12 and 0.93), and (0.50 and 1.28) µg/mL, respectively, compared to vorasidenib (0.02 and 0.08) µg/mL and enasidenib (0.33 and 1.80) µg/mL. Furthermore, the most active candidate (6b) has very promising inhibitory potentials towards HIF-1α, VEGF, and SDH, besides, a marked increase of ROS was observed as well. Besides, compound 6b induced the upregulation of P53, BAX, Caspases 3, 6, 8, and 9 proteins by 3.70, 1.99, 2.06, 1.73, 1.75, and 1.85-fold changes, respectively, and the downregulation for the BCL-2 protein by 0.55-fold change compared to the control. Besides, the in vivo behavior of compound 6b as an antitumor agent was evaluated in female mice bearing solid Ehrlich carcinoma tumors. Notably, compound 6b administration resulted in a prominent decrease in the weight and volume of the tumors, accompanied by improvements in biochemical, hematological, and histological parameters.
... Through the experimental studies, we summarized the representative drugs and the corresponding targeting and applications in Table 1, it has been found that ferroptosis can reduce drug resistance in several types of cancer cells during ongoing cancer treatment, which provides a new direction for cancer treatment. 70 As displayed in Figure 1, new avenues for the development of novel therapeutics targeting ferroptosis for the treatment of cancer have been developed and summarized. 71,72 ...
Article
Full-text available
Ferroptosis is a distinct mode of cell death, distinguishing itself from typical apoptosis by its reliance on the accumulation of iron ions and lipid peroxides. Cells manifest an imbalance between oxidative stress and antioxidant equilibrium during certain pathological contexts, such as tumours, resulting in oxidative stress. Notably, recent investigations propose that heightened intracellular reactive oxygen species (ROS) due to oxidative stress can heighten cellular susceptibility to ferroptosis inducers or expedite the onset of ferroptosis. Consequently, comprehending role of ROS in the initiation of ferroptosis has significance in elucidating disorders related to oxidative stress. Moreover, an exhaustive exploration into the mechanism and control of ferroptosis might offer novel targets for addressing specific tumour types. Within this context, our review delves into recent fundamental pathways and the molecular foundation of ferroptosis. Four classical ferroptotic molecular pathways are well characterized, namely, glutathione peroxidase 4‐centred molecular pathway, nuclear factor erythroid 2‐related factor 2 molecular pathway, mitochondrial molecular pathway, and mTOR‐dependent autophagy pathway. Furthermore, we seek to elucidate the regulatory contributions enacted by ROS. Additionally, we provide an overview of targeted medications targeting four molecular pathways implicated in ferroptosis and their potential clinical applications. Here, we review the role of ROS and oxidative stress in ferroptosis, and we discuss opportunities to use ferroptosis as a new strategy for cancer therapy and point out the current challenges persisting within the domain of ROS‐regulated anticancer drug research and development.
Article
Full-text available
Reactive oxygen species (ROS) are by-products of aerobic metabolism and can also act as signaling molecules to participate in multiple regulation of biological and physiological processes. The occurrence, growth and metastasis of tumors, and even the apoptosis, necrosis and autophagy of tumor cells are all closely related to ROS. However, ROS levels in the body are usually maintained at a stable status. ROS produced by oxidative stress can cause damage to cell lipids, protein and DNA. In recent years, ROS have achieved satisfactory results on the treatment of tumors. Therefore, this review summarizes some research results of tumor treatments from the perspective of ROS in recent years, and analyzes how to achieve the mechanism of inhibition and treatment of tumors by ROS or how to affect the tumor microenvironment by influencing ROS. At the same time, the detection methods of ROS, problems encountered in the research process and solutions are also summarized. The purpose of this review is to provide a clearer understanding of the ROS role in tumor treatment, so that researchers might have more inspiration and thoughts for cancer prevention and treatment in the next stage.
Article
Full-text available
Background Cancer is one of the major heterogeneous disease with high morbidity and mortality with poor prognosis. Elevated levels of reactive oxygen species (ROS), alteration in redox balance, and deregulated redox signaling are common hallmarks of cancer progression and resistance to treatment. Mitochondria contribute mainly in the generation of ROS during oxidative phosphorylation. Elevated levels of ROS have been detected in cancers cells due to high metabolic activity, cellular signaling, peroxisomal activity, mitochondrial dysfunction, activation of oncogene, and increased enzymatic activity of oxidases, cyclooxygenases, lipoxygenases, and thymidine phosphorylases. Cells maintain intracellular homeostasis by developing an immense antioxidant system including catalase, superoxide dismutase, and glutathione peroxidase. Besides these enzymes exist an important antioxidant glutathione and transcription factor Nrf2 which contribute in balancing oxidative stress. Reactive oxygen species–mediated signaling pathways activate pro-oncogenic signaling which eases in cancer progression, angiogenesis, and survival. Concomitantly, to maintain ROS homeostasis and evade cancer cell death, an increased level of antioxidant capacity is associated with cancer cells. Conclusions This review focuses the role of ROS in cancer survival pathways and importance of targeting the ROS signal involved in cancer development, which is a new strategy in cancer treatment.
Article
Full-text available
The large doses of vitamin C and E and β-carotene used to reduce reactive oxygen species (ROS) production and oxidative damages in cancerous tissue, have produced disappointing and contradictory results. This therapeutic conundrum was attributed to the double-faced role of ROS, notably, their ability to induce either proliferation or apoptosis of cancer cells. However, for a ROS-inhibitory approach to be effective, it must targets ROS when they induce proliferation rather than apoptosis. Based on recent advances in redox biology, this review underlined a differential regulation of pro- and antioxidant system, respective to the stage of cancer. At early precancerous and neoplastic stages, antioxidant activity decreases and ROS appear to promote cancer initiation via inducing oxidative damage and base pair substitution mutations in pro-oncogenes and tumor suppressor genes, such as RAS and TP53, respectively. Whereas in late stages of cancer progression, tumor cells escape apoptosis by producing high levels of intracellular antioxidants, like NADPH and GSH, via the pentose phosphate pathway to buffer the excessive production of ROS and related intra-tumor oxidative injuries. Therefore, antioxidants should be prohibited in patients with advanced stages of cancer and/or undergoing anticancer therapies. Interestingly, the biochemical and biophysical properties of some polyphenols allow them to selectively recognize tumor cells. This characteristic was exploited to design and deliver nanoparticles coated with low doses of polyphenols and containing chemotherapeutic drugs into tumor-bearing animals. First results are encouraging, which may revolutionize the conventional use of antioxidants in cancer.
Article
Full-text available
Purpose Reactive oxygen species (ROS) are produced in cancer cells as a result of increased metabolic rate, dysfunction of mitochondria, elevated cell signaling, expression of oncogenes and increased peroxisome activities. Certain level of ROS is required by cancer cells, above or below which lead to cytotoxicity in cancer cells. This biochemical aspect can be exploited to develop novel therapeutic agents to preferentially and selectively target cancer cells. Methods We searched various electronic databases including PubMed, Web of Science, and Google Scholar for peer-reviewed english-language articles. Selected articles ranging from research papers, clinical studies, and review articles on the ROS production in living systems, its role in cancer development and cancer treatment, and the role of microbiota in ROS-dependent cancer therapy were analyzed. Results This review highlights oxidative stress in tumors, underlying mechanisms of different relationships of ROS and cancer cells, different ROS-mediated therapeutic strategies and the emerging role of microbiota in cancer therapy. Conclusion Cancer cells exhibit increased ROS stress and disturbed redox homeostasis which lead to ROS adaptations. ROS-dependent anticancer therapies including ROS scavenging anticancer therapy and ROS boosting anticancer therapy have shown promising results in vitro as well as in vivo. In addition, response to cancer therapy is modulated by the human microbiota which plays a critical role in systemic body functions.
Article
Full-text available
Pharmacological concentrations of small molecule natural products, such as ascorbic acid, have exhibited distinct cell killing outcomes between cancer and normal cells whereby cancer cells undergo apoptosis or necrosis while normal cells are not adversely affected. Here, we develop a mathematical model for ascorbic acid that can be utilized as a tool to understand the dynamics of reactive oxygen species (ROS) induced cell death. We determine that not only do endogenous antioxidants such as catalase contribute to ROS-induced cell death, but also cell membrane properties play a critical role in the efficacy of ROS as a cytotoxic mechanism against cancer cells vs. normal cells. Using in vitro assays with breast cancer cells, we have confirmed that cell membrane properties are essential for ROS, in the form of hydrogen peroxide (H 2 O 2), to induce cell death. Interestingly, we did not observe any correlation between intracellular H 2 O 2 and cell survival, suggesting that cell death by H 2 O 2 is triggered by interaction with the cell membrane and not necessarily due to intracellular levels of H 2 O 2. These findings provide a putative mechanistic explanation for the efficacy and selectivity of therapies such as ascorbic acid that rely on ROS-induced cell death for their anti-tumor properties.
Article
Background: Patients with low response rates to cancer vaccines, short duration of anti-tumor response after vaccination, and relatively weak curative effects are problems that have not been resolved effectively during the development and application of cancer vaccines. With the continuous improvement of knowledge and awareness regarding the immune system and cancer cells, many researches have helped to explain the reasons for poor vaccine efficacy. Input from researchers accompanied by some newly emerged strategies could bring hope to improve the therapeutic effects of vaccines. Methods: Data were collected from Web of Science, Medline, Pubmed, through searching of these keywords: "cancer vaccine", "cancer stem cell", "targeted agent", "immune checkpoint blockade" and "neoantigen". Results: It may be more effective in immunotherapy of human cancers, including cancer stem cell vaccines, combination vaccines with targeted agents or immune checkpoint blockade, and neoantigen-based vaccines. Conclusion: Personalized vaccines will become the mainstream solution of cancer treatment program with the continuous improvement of human understanding of the immune system and the progress of related experiments.
Article
Increased reactive oxygen species (ROS) production has been detected in various cancers and has been shown to have several roles, for example, they can activate pro-tumourigenic signalling, enhance cell survival and proliferation, and drive DNA damage and genetic instability. Counterintuitively ROS can also promote anti-tumourigenic signalling, initiating oxidative stress-induced tumour cell death. Tumour cells express elevated levels of antioxidant proteins to detoxify elevated ROS levels, establish a redox balance, while maintaining pro-tumourigenic signalling and resistance to apoptosis. Tumour cells have an altered redox balance to that of their normal counterparts and this identifies ROS manipulation as a potential target for cancer therapies. This review discusses the generation and sources of ROS within tumour cells, the regulation of ROS by antioxidant defence systems, as well as the effect of elevated ROS production on their signalling targets in cancer. It also provides an insight into how pro- and anti-tumourigenic ROS signalling pathways could be manipulated in the treatment of cancer.
Article
An unanswered question in human health is whether antioxidation prevents or promotes cancer. Antioxidation has historically been viewed as chemopreventive, but emerging evidence suggests that antioxidants may be supportive of neoplasia. We posit this contention to be rooted in the fact that ROS do not operate as one single biochemical entity, but as diverse secondary messengers in cancer cells. This cautions against therapeutic strategies to increase ROS at a global level. To leverage redox alterations towards the development of effective therapies necessitates the application of biophysical and biochemical approaches to define redox dynamics and to functionally elucidate specific oxidative modifications in cancer versus normal cells. An improved understanding of the sophisticated workings of redox biology is imperative to defeating cancer.
Article
Reactive oxygen species (ROS) are short-lived and highly reactive molecules. The generation of ROS in cells exists in equilibrium with a variety of antioxidant defences. At low to modest doses, ROS are considered to be essential for regulation of normal physiological functions involved in development such as cell cycle progression and proliferation, differentiation, migration and cell death. ROS also play an important role in the immune system, maintenance of the redox balance and have been implicated in activation of various cellular signalling pathways. Excess cellular levels of ROS cause damage to proteins, nucleic acids, lipids, membranes and organelles, which can lead to activation of cell death processes such as apoptosis. Apoptosis is a highly regulated process that is essential for the development and survival of multicellular organisms. These organisms often need to discard cells that are superfluous or potentially harmful, having accumulated mutations or become infected by pathogens. Apoptosis features a characteristic set of morphological and biochemical features whereby cells undergo a cascade of self-destruction. Thus, proper regulation of apoptosis is essential for maintaining normal cellular homeostasis. ROS play a central role in cell signalling as well as in regulation of the main pathways of apoptosis mediated by mitochondria, death receptors and the endoplasmic reticulum (ER). This review focuses on current understanding of the role of ROS in each of these three main pathways of apoptosis. The role of ROS in the complex interplay and crosstalk between these different signalling pathways remains to be further unravelled during the coming years.
Article
Reactive oxygen species (ROS), now appreciated for their cellular signaling capabilities, have a dual role in cancer. On the one hand, ROS can promote protumorigenic signaling, facilitating cancer cell proliferation, survival, and adaptation to hypoxia.Onthe other hand, ROS can promote antitumorigenic signaling and trigger oxidative stress–induced cancer cell death. To hyperactivate the cell signaling pathways necessary for cellular transformation and tumorigenesis, cancer cells increase their rate of ROS production compared with normal cells. Concomitantly, in order to maintain ROS homeostasis and evade cell death, cancer cells increase their antioxidant capacity. Compared with normal cells, this altered redox environment of cancer cells may increase their susceptibility to ROS-manipulation therapies. In this review, we discuss the two faces of ROS in cancer, the potential mechanisms underlying ROS signaling and the opposing cancer therapeutic approaches to targeting ROS. Expected final online publication date for the Annual Review of Cancer Biology Volume 1 is March 04, 2017. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.