Next Article in Journal
A Novel Tissue-Free Method to Estimate Tumor-Derived Cell-Free DNA Quantity Using Tumor Methylation Patterns
Next Article in Special Issue
The Various Roles of PEDF in Cancer
Previous Article in Journal
Cyclooxygenase-2 Blockade Is Crucial to Restore Natural Killer Cell Activity before Anti-CTLA-4 Therapy against High-Grade Serous Ovarian Cancer
Previous Article in Special Issue
Targeting Proteasomes and the MHC Class I Antigen Presentation Machinery to Treat Cancer, Infections and Age-Related Diseases
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Inflammation-Associated Cytotoxic Agents in Tumorigenesis

by
Jürgen Arnhold
Institute of Medical Physics and Biophysics, Medical Faculty, Leipzig University, Härtelstr. 16-18, 04107 Leipzig, Germany
Cancers 2024, 16(1), 81; https://doi.org/10.3390/cancers16010081
Submission received: 3 December 2023 / Revised: 16 December 2023 / Accepted: 20 December 2023 / Published: 22 December 2023
(This article belongs to the Special Issue Inflammation, Immunity, and Cancer Progression)

Abstract

:

Simple Summary

This review summarizes the present knowledge of cytotoxic agents and protective systems against these damaging substances in tumors. In tumor cells, enhanced levels of cytotoxic agents are usually counteracted by an overexpression of protective mechanisms. In this manner, tumor cells can even survive therapeutically induced stress situations. Tumor cells also affect immune cells and other cells in their close neighborhood in such a way that cells in this tumor microenvironment change their properties and promote tumor progression. Numerous examples are given for how the disturbed balance between inflammation-associated cytotoxic agents and antagonizing principles is related to tumor growth, tumor cell invasion, and metastasis. A thorough knowledge of these mechanisms is mandatory for the implementation of novel therapeutic approaches against cancers.

Abstract

Chronic inflammatory processes are related to all stages of tumorigenesis. As inflammation is closely associated with the activation and release of different cytotoxic agents, the interplay between cytotoxic agents and antagonizing principles is highlighted in this review to address the question of how tumor cells overcome the enhanced values of cytotoxic agents in tumors. In tumor cells, the enhanced formation of mitochondrial-derived reactive species and elevated values of iron ions and free heme are antagonized by an overexpression of enzymes and proteins, contributing to the antioxidative defense and maintenance of redox homeostasis. Through these mechanisms, tumor cells can even survive additional stress caused by radio- and chemotherapy. Through the secretion of active agents from tumor cells, immune cells are suppressed in the tumor microenvironment and an enhanced formation of extracellular matrix components is induced. Different oxidant- and protease-based cytotoxic agents are involved in tumor-mediated immunosuppression, tumor growth, tumor cell invasion, and metastasis. Considering the special metabolic conditions in tumors, the main focus here was directed on the disturbed balance between the cytotoxic agents and protective mechanisms in late-stage tumors. This knowledge is mandatory for the implementation of novel anti-cancerous therapeutic approaches.

1. Introduction

Inflammation is related to all stages of tumorigenesis, such as the initiation, promotion, progression, and metastasis [1,2,3]. Inflammatory processes are primarily directed to eliminate by means of the immune systems and the activation of components in the acute phase, complement, coagulation, and contact systems any harm from the host that can disturb normal functioning and tissue homeostasis [4,5].
In tumors, the associated inflammatory process is persistent [6]. During tumor development and the interaction between immune and tumor cells, three major phases can be distinguished, according to the concept of immunoediting [3,7]. These phases are schematically presented in Figure 1. The first phase, the elimination phase, corresponds to the original concept of immune surveillance, suggesting that immune cells are able to eliminate transformed cells [8,9]. Natural killer (NK) cells and cytotoxic T lymphocytes (CTLs) are the key players in the recognition and killing of transformed cells [10,11]. It is generally assumed that the formation and elimination of unwanted transformed cells occurs often in our organism.
During the second phase, the equilibrium phase, the elimination of tumors cells and the development of tumor-mediated immune suppressing strategies co-exist. Thus, the elimination of unwanted cells is impeded, and part of these cells can survive. In this phase, tumor immunogenicity is reduced, and a pro-inflammatory tumor microenvironment (TME) is generated that promotes immune tolerance [3].
These alterations in the immune response are gradually strengthened to the immune escape that is characteristic of the third phase, the escape phase. As a result, the immune response is manipulated in multiple ways by tumor cells and the TME, ensuring that tumor growth and metastasis can occur and the processes of elimination of tumor cells by the host’s immune system are slowed down. The progressive development of tumor-mediated immune escape is closely associated with major changes in the tumor cell properties and conditions in the TME, such as increasing the deficiency of dioxygen, the accumulation of lactate, altered routes in the supply of energy substrates, the activation of stress-related genes and oncogenes, nutrient depletion, reversed pH gradient on tumor cells, and matrix remodeling. In general, these altered conditions promote the survival of tumor cells, favor the suppression of immune defense reactions against tumor components, elevate proliferative activities, and contribute to tumor progression [3,12].
During the gradual development of tumors, many immunological relevant factors are converted from an anti-tumor behavior in the early stages to tumor-promoting properties in advanced tumors. Concurrently, many therapeutic procedures are increasingly restricted in their efficiency during tumor progression and fail in the late stages. In cancer research, the focus is, therefore, directed to study and identify the immunosuppressive mechanisms in late-stage tumors with the hope of finding novel ways for therapeutical intervention [12,13,14,15].
In tumor-associated inflammations, host-derived cytotoxic agents originate from activated immune cells, damaged tissue cells in the TME, dysfunctional cellular processes in tumor cells, and can additionally result from therapeutic applications like radio- and chemotherapy. To prevent the disastrous action of these cytotoxic agents, numerous ready-to-use protective mechanisms exist that neutralize these cytotoxic agents. Disturbances in the control of host-derived cytotoxic agents by antagonizing principles are associated with the development of chronic inflammatory disease states [16].
In this review, the knowledge of chronic inflammatory processes during tumorigenesis is highlighted. Special focus is directed on the interplay between host-derived cytotoxic agents and antagonizing principles in late-stage tumors and how alterations in this balance contribute to the survival of tumor cells and tumor progression.

2. Chronic Inflammatory States and Altered Conditions in Tumors

2.1. Immunosuppression during Acute and Chronic Inflammation

Regarding the time elapsed, an acute inflammation can be subdivided into two major phases with distinct differences in the mediator profile and immune cell functions. First, in response to pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs), pro-inflammatory cascades are induced via the activation of pattern recognition receptors (PRRs) [17,18]. As a result, immune cells are recruited and activated onto inflammatory sites and the maturation of dendritic cells is induced for the presentation of antigens to T cells [19]. Typical pro-inflammatory mediators are cytokines, such as interleukin-1β (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor α (TNF-α), and the acute-phase proteins C-reactive protein (CRP) and serum amyloid A (SAA) [4,5,20,21,22]. These activities are directed to deactivate pathogens, eliminate virus-infected or transformed cells, remove any damaged cell material, and create better conditions for ongoing immune reactions. This very common concept of pattern recognition is valid for the initiation of novel inflammatory events in a broad range of diseases, including infections, rheumatoid arthritis, atherosclerosis, Alzheimer’s disease, and many others, as well as tumors.
During inflammatory response, different host-derived cytotoxic agents are released or generated, which are derived from activated immune and undergoing tissue cells, and in tumors also from tumor cells. These agents generally execute a dual role in living tissues. They are an essential part in many physiological functions, especially in immune cell-triggered processes. Cytotoxic agents are mandatory for combatting microbes, to restore tissue homeostasis after any threat, and to ensure normal functioning of the physiological processes in the organism [4]. Otherwise, they can damage unperturbed tissues and contribute to the initiation of novel inflammatory cascades. According to their mode of action, cytotoxic agents can be differentiated into oxidant-based agents (reactive species, oxidized heme proteins, free heme, transition metal ions) and protease-based agents (serine proteases, matrix metalloproteases, pro-inflammatory peptides) [16]. In healthy tissues, cytotoxic agents are inactivated by already existing antagonizing principles.
In the second phase of an acute inflammation, the termination or resolution phase, destroyed cells and tissues are replaced by novel synthesized material via the induction of proliferative processes. The pro-inflammatory activities of immune cells are downregulated, and finally the former homeostasis is restored. The typical transiently acting cytokines of this phase are transforming growth factor β (TGF-β) and IL-10 [23,24,25]. In addition, growth factors [26,27] and lipid mediators, such as lipoxins and resolvins, are involved in the resolution of inflammation [28,29]. Cell signaling is characterized by the induction of the STAT3 pathway. Macrophages are polarized from the M1 type to M2 type [30,31]. The accumulation of myeloid-derived suppressor cells (MDSCs) contributes to the development of a transient immunosuppression in inflamed areas [32,33]. MDSCs are immature immune cells that promote immunosuppressive effects on lymphocytes, natural killer cells, macrophages, and dendritic cells [34]. In general, the aforementioned mechanisms impede the hyperactivation of pro-inflammatory cells and prevent any excessive tissue damage by host-derived cytotoxic agents during the resolution of inflammation. After the resolution, the number of immune cells and mediators is reduced to a level typical of healthy tissues [35]. Cytotoxic agents and their antagonizing counterparts are also involved in the resolution of inflammation, especially in the processes of matrix remodeling.
Under chronic inflammatory conditions, inflammation is only insufficiently terminated as pro-inflammatory cascades are activated again and again due to the incomplete inactivation of host-derived cytotoxic agents and the continuing release of DAMPs and antigens. At chronic inflammatory sites, pro-inflammatory mediators, the recruitment of immune cells, and tissue damage co-exist with a plethora of immunosuppressive mechanisms, such as the prevalence of MDSCs, the presence of inflammation-resolving and proliferative factors, as well as the depletion of essential metabolites for appropriate immune defense [16]. The resulting long-lasting immunosuppression can seriously affect the health status and enhance susceptibility to chronic infections and comorbidities [36,37]. For example, in elderly persons, persistent inflammation is closely associated with immune dysfunction. This condition is known as inflammaging [38], which is regarded as a risk factor for life-threatening diseases and adverse health outcomes [39,40]. In general, pro-inflammatory mechanisms and immunosuppressive conditions are closely associated with each other in many chronic diseases, including cancer.
In chronic inflammations, serious health problems can arise from the excessive release or generation of cytotoxic agents and from the decline or exhaustion of protective mechanisms. In consequence, the following release of novel DAMPs, antigens, and cytotoxic agents promotes ongoing inflammatory events and prevents the termination of inflammatory cascades. The worst case is a very low capacity of antagonizing principles or their exhaustion with the consequence of septic complications and organ failure [16]. This concept of the incomplete inactivation of cytotoxic agents by protective principles explains under which conditions an inflammation becomes persistent. It also provides the basis for a better understanding of the underlying molecular mechanisms for a wide range of chronic inflammatory diseases, including cancer.
In the description of the molecular processes during tumorigenesis, numerous cytotoxic agents derived from tumor cells, immune cells, and other tumor-associated cells are more expressed and exhibit higher activities compared to healthy tissue areas. In general, these agents are able to damage transformed cells with the subsequent elimination of these cells by the immune system. This is mostly observed during the early phases of tumorigenesis. However, in response to stress and the increasing accumulation of cytotoxic agents, numerous antagonizing principles are markedly upregulated in advanced tumors and promote the survival of tumor cells and protection against stress induced by chemo- and radiotherapy.
In Section 3 and Section 4, details about the interplay between cytotoxic agents and the protective mechanisms will be given concerning tumor development. As the action of cytotoxic agents highly depends on the altered conditions in tumor cells and the TME, Section 2.2 summarizes the key processes of immunosuppression and metabolic reprogramming occurring during tumorigenesis.

2.2. Key Elements of Immunosuppression in Tumors

Tumor progression and chronic inflammatory processes are highly linked to each other [1,2,3,6]. In the creation of an immunosuppressive milieu, both tumor cells and the TME contribute multiple mechanisms to the manipulation of the host’s immune answer and to tissue remodeling in tumors. An overview of the major mechanisms of tumor cell-mediated processes in the TME and immunosuppression is given in Figure 2.
During tumor progression, the principal processes involved in immunosuppression and tissue remodeling during the resolution phase of an acute inflammation are also active. Whereas these mechanisms are only transiently expressed in a subsiding inflammation, they act permanently and more intensively in tumors. In cancer cells, several protective systems are highly upregulated in response to a stress-mediated increase of cytotoxic agents (see Section 3). Further, these cells actively secrete inflammation-resolving molecules (TGF-β, IL-10) and exosomes that affect the conditions and properties of immune cells in the TME [41]. Importantly, natural killer cells (NK) and cytotoxic T cells, which are able to deactivate tumor cells, are suppressed in their activity by the release of TGF-β [42,43,44]. In the TME, tumor-associated macrophages (TAMs) are triggered from the pro-inflammatory M1 type to the immune-resolving M2 type [45]. Regulatory T cells (Treg) and various types of myeloid-derived suppressor cells (MDSCs) accumulate in the TME during tumor progression. These cell types suppress the activation of other immune cells and contribute to the survival of tumors [3,13,32,35,41,46,47,48]. With their cargo, cancer cell-derived exosomes are able to modulate different processes of tumor progression such as angiogenesis, metastasis, and survival [41].
MDSCs are formed as all other myeloid cells from precursors in the bone marrow. Under inflammatory conditions, the need for novel immune cells increases and these cells undergo a forced formation process, known as emergency myelopoiesis, leading to immature properties. In most kinds of cancers, the accumulation of granulocytic MDSCs (G-MDSCs) dominates over monocytic MDSCs (M-MDSCs) [49]. In glioma, M-MDSCs are more expressed than G-MDSCs [49]. In humans, a third, less pronounced type of MDSCs, early-stage MDSCs (e-MDSCs), has been described [50,51]. The MDSC subtypes are usually identified by their morphological properties, density, and surface markers [49].
In the TME, immunosuppressive conditions can be differentiated by their underlying mechanisms as enzyme-dependent, cytokine-dependent, and immune checkpoint-dependent [14]. In addition, the expression of neoantigens and antigen-presenting molecules like MHC-1 are decreased in cancer cells [52,53].
Enzyme-dependent immunosuppressive pathways are characterized by a depletion of essential metabolites, such as ATP and tryptophan, or by an enhanced formation of metabolites like adenosine, citrulline, kynurenine, and prostaglandin E2 [14,54,55]. Several proteins (for example ectonucleotidases CD39 and CD73 [56] and indoleamine 2,3-dioxygenase [57]), which are involved in these pathways, are highly upregulated in tumors.
Cytokine-dependent immunosuppression can be induced by the activation of the IL-6/STAT-3 pathway that inhibits DC maturation and activates effector T cells [58,59]. Vascular endothelial growth factor A (VEGF-A) is immunosuppressive by the inhibition of the DC functions and maturation, by the infiltration of immunosuppressive cells (Treg, MDSC, and TAMs) into tumors, by reducing cytotoxic CD8+ T cell infiltration into tumors, and by the expression of the factors involved in the exhaustion of these cells [60,61,62]. Cytokines of the IL-10 and TGF-β pathways activate Treg and promote immunosuppression [63].
Several immune checkpoints are known to be involved in immunosuppressive activities. The inactivation of T cells is possible via the binding of a specific ligand to the inhibitory T cell receptor programmed cell death protein 1 (PD-1) [14]. This ligand known as PD-L1 or CD274 is expressed on the surface of a tumor and various other cells. The overexpression of PD-L1 on tumor cells is associated with a poor prognosis and the evasion of T cell recognition of cancers [64]. The PD-1/PD-L1 axis represents an important element in the generation of an immunosuppressive TME [14]. Other inhibitory immune checkpoint molecules on T cells are cytotoxic T lymphocyte-associated protein 4 (also known as CD 152), LAG3, TIM-3, and the tyrosine-based inhibitory domain (TIGIT) [65,66,67]. Cancer cells can express TIGIT ligands, such as CD155, and overcome cancer immunity.
In growing and metastasizing tumors, the balance between tumor-promoting and tumor-destroying factors is shifted towards the first types of factors. Although, many features of tumor-induced immunosuppression are known, this knowledge is incomplete concerning the interrelations between them and the consequences for further tumor processing. Numerous anti-tumor therapies, such as targeted therapies and chemotherapies, attempt to overcome immunosuppression in tumors. The application of immune checkpoint inhibitors provides a novel tool for the development of combined therapy approaches [14].

2.3. Poor Quality of the Tumor Vasculature

Tumorigenesis starts with the formation and uncontrolled growth of transformed cells. In developing tumors, the supply with dioxygen and nutrients is diminished due to the poor quality of the tumor vasculature, longer diffusion paths, insufficient lymphatic drainage, fluctuations in interstitial pressure, and intermittent vascular collapse in contrast to healthy tissues [68,69].
In solid tumors, the growth factor VEGF is overexpressed, whereby hypoxia promotes this expression [70,71]. This growth factor is responsible for the poor quality of the tumor vasculature with irregular, leaky, and immature vessels [72,73]. In addition, the VEGF is a key angiogenic factor in tumors and is involved in tumor progression and metastasis [73,74].

2.4. Direct Effects of Hypoxia in Tumor Cells

In tumors, hypoxia [75,76,77] provokes significant metabolic alterations in tumor cells, such as the activation and stabilization of hypoxia-inducible factor 1α (HIF-1α) [78], upregulation of glycolysis [79], downregulation of oxidative phosphorylation in the mitochondria [79], and enhanced formation of reactive species by dysfunctional mitochondria [80]. In addition, lactate accumulates [81,82] and induces numerous lactate-driven effects [81,83,84].
Cytosolic HIF-1α is a master regulator of glycolysis in many cells. This usually short-lived factor is controlled by prolyl hydroxylase that tags HIF-1α for proteasome degradation [85,86]. Hypoxia, stress-induced oxidation of Fe2+ in propyl hydroxylase, and an ascorbate deficiency prevent HIF-1α degradation [78]. As a result, HIF-1α upregulates several enzymes promoting glycolysis and downregulates pyruvate dehydrogenase that supplies acetyl-coenzyme A (Ac-CoA) for the citrate cycle [79]. Unlike HIF-1α, which is a ubiquitous protein, HIF-2α is predominantly expressed in highly vascularized tissues [87]. HIF-2α is active during prolonged hypoxia, replaces HIF-1α in a spatiotemporal manner [75,88], and is involved in controlling oxidative stress, the cell cycle, blood vessel remodeling, and RNA transport [89].
In healthy cells, lipogenesis is mostly induced by pyruvate-derived Ac-CoA. To ensure lipogenesis in hypoxic cancer cells, decreased pyruvate is replaced in the TCA cycle by glutamine, supporting the conversion of α-ketoglutarate into citrate and Ac-CoA [90]. In cancer cell growth, glutamine is an important carbon and nitrogen source for lipid, amino acid, and nucleotide synthesis [91].
In tumorigenesis, increased lactate production by tumors cells reprograms macrophages, T cells, and other immune cells in a way that they are immunosuppressive and anti-inflammatory [81,82,83,84]. Lactate also promotes angiogenesis and tumor progression [83,92], supporting the hyaluronan release from the adjacent fibroblasts. This hyaluronan cover protects additional tumor tissue from immune attacks [93].

2.5. Stress-Related Responses in Tumor Cells

In tumor cells, enhanced intracellular levels of reactive species, which result mainly from dysfunctional mitochondria, activate the transcription factor nuclear factor-erythroid 2-related factor 2 (Nrf2) that is low expressed under normal physiological conditions [94,95]. In cancer cells, Nrf2 promotes the syntheses of numerous enzymes involved in antioxidative defense and contributes to a resistance against chemo- and radiotherapy (see Section 3.3).
Prolonged hypoxia also mediates the processes of autophagy [96,97]. In the early stages of tumorigenesis, autophagy is directed to inhibit tumor growth [98,99,100]. Otherwise, in late-stage cancers, autophagy is known to stabilize cancer cells by maintaining the integrity of the mitochondria, reducing DNA damage, and increasing the resistance against stress. Under hypoxic conditions and a low supply of nutrients, autophagy can provide energy resources for the survival of cancer cells and resistance against chemotherapy [101]. With these mechanisms, autophagy contributes to tumor development [102,103,104] and facilitates metastasis [105,106,107,108].
Under stress situations, the percentage of misfolded proteins increases in the endoplasmic reticulum (ER) as a result of the action of reactive species and electrophiles. Misfolded proteins are subjected to ubiquitinylation with subsequent proteasomal degradation or autophagy. Three major stress sensors of an unfolded protein response (UPR) are activated by misfolded proteins in a wide range of tumor cells [109]. The overexpression of chaperones or mutations in the UPR pathways are used by tumor cells to antagonize ER stress [110,111,112,113]. A hyperactive UPR promotes tumorigenesis in advanced cancers [114].
To maintain a reducing environment in the cytosol of tumor cells, the enhanced uptake of cystine/cysteine by tumors and the conversion into glutathione are mandatory to protect cells from stress-mediated cell death [115]. In tumors, highly increased extracellular cysteine levels are observed, whereas cysteine is slightly enhanced within tumor cells [116]. Moreover, cysteine is mainly transported into tumor cells by the cystine/glutamate antiporter solute carrier family 7 member 11 (SLC7A11), which is widely expressed in human cancers [117,118]. In tumor cells, the conversion of cysteine into hydrogen sulfide activates the Nrf2-mediated gene expression of antioxidative proteins [119,120]. Hydrogen sulfide also accelerates the cell cycle in tumor cells [121].
In addition, a close link exists between the upregulation of reductive glutamine metabolism, also known as reductive carboxylation, and the transport of NADPH from cytosol into the mitochondria to upregulate mitochondrial GSH and protect against reactive species [122]. These pathways are important for anchorage-independent growth in cancer cells.

2.6. Intratumoral Hemorrhages

The rupture of blood vessels results in intratumoral hemorrhages, which are typical of many tumors [123,124]. The conditions in hemorrhages promote the formation of cytotoxic free heme (see Section 4 for more details) and in turn the transcriptional processes in tumor cells via the binding of free heme to guanine-rich DNA and RNA structures (known as G4 elements) [125,126]. Free heme controls the expression of key target genes, including telomeres and oncogenes (such as c-Myc), and favors malignant transformation [127,128].

2.7. Reversed pH Gradient in Cancer Cells

In cancer cells, the cytosolic pH is enhanced to 7.3–7.6 (versus about 7.2 in normal cells), whereas the extracellular pH is decreased to 6.8–7.0 (versus about 7.4 in normal cells) [129,130]. In the TME, the milieu is more acidic by about 0.3–0.7 pH units in comparison to the cytosolic pH of tumors cells [131]. Thus, the pH gradient across the plasma membrane of cancer cells is reversed in contrast to healthy cells. These deviations are mediated by hypoxia and lactate effects and accompanied by an increased expression and activity of ion transporters in the plasma membrane and intracellular pH regulators [132].
In many cell types, enhanced intracellular pH values by ~0.3–0.4 units are found in proliferating cells at the end of the S phase [133,134] and in migrating cells [135,136]. The reverse pH gradient in cancer cells promotes cell proliferation, survival, migration, and metastasis [129,130,137,138].

2.8. Matrix Remodeling in the TME

In the TME, the extracellular matrix (ECM) acts as a barrier around tumor cells against cytotoxic immune cells. Under hypoxic conditions, the stiffness of the matrix components, especially of collagen fibrils, increases due to more intense cross-linking reactions between matrix polymers [139,140]. These alterations are mainly caused by cancer-associated fibroblasts [141]. Several enzymes involved in matrix remodeling and cross-linking reactions are upregulated in tumors, namely lysyl oxidase, lysyl oxidase-like proteins, collagen prolyl 4-hydroxylase, and WINT1-inducible signaling pathway protein 1 (WISP1) [142,143,144,145].
Tumor fibrosis is closely associated with an overexpression of these enzymes [146]. As a result, ECM fibers, most of all collagens I and III, are converted into dense, linearized, and cross-linked fiber bundles with altered mechanical properties [147,148,149,150]. The enhanced stiffness of the ECM components favors the migration of single tumor cells, promotes angiogenesis, and dampens anti-tumor activities [151]. The elevated degree of tumor fibrosis is associated with a poor prognosis in many types of cancer [151,152,153].
In the TME, the deposition of the matrix components is important for angiogenesis, proliferation, tumor cell invasion, and metastasis. Several matrix metalloproteases such as MMP-2, -3, -9, and -14 are upregulated in malignant tumors [154,155]. An enhanced expression of heparinase also favors angiogenesis [156,157,158].
The enhanced proliferation of tumor cells is also supported by increased hyaluronan production [159,160,161]. Under hypoxic conditions, the production of hyaluronan is markedly increased by tumor cells [162]. M2-type macrophages accumulate preferentially in hyaluronan-rich areas of the TME [163]. In addition, hyaluronan favors the phenotype change in monocytes and macrophages to the M2-type [164].

3. Cytotoxic Agents in Cancer Cells

3.1. Sources for Cytotoxic Agents in Cancer Cells

The shift in energy metabolism toward the dominance of glycolysis in tumor cells markedly affects the properties of the mitochondria. The tumor-associated dysfunction of the mitochondria has multiple facets, including mutations of mitochondrial DNA, defective enzymes in the TCA cycle, glutamine-induced reductive carboxylation in the TCA cycle, disturbed electron redox conversions, imbalances in the signaling pathways, resistance to apoptosis induction, and disturbed energy metabolism [165]. Dysfunctional mitochondria are a major source for reactive species, such as the superoxide anion radical (O2•−) and hydrogen peroxide (H2O2) [80]. Highly reactive species can be derived from the reactions of both these species.
In tumor cells, several mechanisms contribute to the enhanced values of catalytically active ferrous ions (Fe2+). Iron overload is involved in cell death by ferroptosis [166,167] and in the formation of highly reactive hydroxyl radicals (HO).
Another source of cytotoxic agents in tumors is free heme that results either from degradative processes in intratumoral hemorrhages or from defective cellular heme proteins. Apart from numerous cytotoxic activities, free heme promotes the oncogene activation via interactions with heme-responsive elements in promoter regions [127,128].
O2•− and H2O2 are less reactive species that can react with metal ions, radical species like NO, or iron-containing proteins to generate very reactive and tissue-damaging agents such as hydroxyl radical (HO), and peroxynitrite (ONOO) [168]. Although many (patho)physiological processes are associated with the excess formation of reactive species in living tissues, in tumor cells the main focus is directed on dysfunctional mitochondria. Other physiologically relevant highly reactive species are hypochlorous acid (HOCl), hypobromous acid (HOBr), and singlet oxygen (1O2). HOCl and HOBr are generated in H2O2-driven reactions by heme peroxidases from activated neutrophils and eosinophils [169,170,171]. The formation of 1O2 is relevant to surface-exposed locations like the skin and eyes, where ultraviolet and visible light can induce photooxidative processes [172,173].

3.2. Mitochondria-Derived Reactive Species in Cancer Cells

In intact mitochondria, only a small percentage of O2 is converted into O2•−. The main mitochondrial sources of O2•− are redox conversions in complex I and the ubiquinone cycle [174]. Spontaneous and superoxide dismutase (SOD)-catalyzed dismutation of two O2•− yields H2O2 and O2. In dysfunctional mitochondria, an enhanced production of O2•− and H2O2 is favored by reverse electron transport in complex I [175] and by disturbances in the electron flow in the ubiquinone cycle [176].
In the mitochondria, apart from dismutation reactions, O2•− is involved in two major pathways, both contributing to the formation of highly reactive species, which exhibit cytotoxic activities. The reaction of O2•− with mitochondrial aconitase and other proteins containing [4Fe-4S]2+ clusters is associated with the release of catalytically active Fe2+ [177,178], which is involved in the formation of HO.
The second pathway concerns the reaction between O2•− and nitric oxide (NO), which is synthesized by NO synthases. The mitochondria exhibit NO synthase activity [179]. However, which isoform is present in the mitochondria is still unsolved [180,181]. Otherwise, NO can also be derived from other cellular sources, as this agent diffuses well through membranes. Inducible NO synthase (iNOS) is upregulated in multiple cancers such as breast, colorectal, prostate, esophageal, and gastric cancers [182,183,184]. The very rapid reaction between NO and O2•− promotes the formation of ONOO [185,186]. NO competes with SODs for O2•− and lowers the load of O2•− within the mitochondria [187,188]. ONOO is involved in the nitration of tyrosine residues, the formation of thiyl radicals and radical species after reaction with CO2, and initiates the lipid peroxidation processes [189,190,191,192]. ONOO is scavenged and inactivated by glutathione peroxidases (GPXs), especially GPX1, heme proteins, and heme peroxidases [193,194,195,196]. Both NO and ONOO inhibit several enzymes of the mitochondrial respiratory chain [179].
H2O2, which mainly results from the dismutation of O2•−, is known to react with free transition metal ions, such as Fe2+ and Cu+, to yield the highly reactive HO in the so-called Fenton reaction [197,198,199]. HO reacts as nearly diffusion controlled with many neighbored biological substrates. Thus, no enzymatic mechanism exists to scavenge and inactivate this radical [200]. Therefore, the main physiological strategy against HO is to avoid its formation through the proper control of iron and copper metabolism during the transport, storage, and utilization of free metal ions [201,202,203,204,205,206,207].
The major antagonizing principles against small reactive species are summarized in reference [16]. O2•− is deactivated by SODs and cytochrome c [208,209,210,211,212,213]. Whereas SOD1 is found within cells at three distinct locations: cytosol, intermembrane space of mitochondria, and nucleus [210]. Within the intermembrane space of the mitochondria, O2•− is also detoxified by cytochrome c [212,213]. H2O2 is controlled by GPXs, peroxiredoxins, and catalase [214,215,216,217]. Unlike O2•−, H2O2 is able to permeate through biological membranes. Thus, the defense reactions against H2O2 concern the whole cell. These mechanisms hold down their concentrations in biological media and prevent dangerous side reactions of these species.

3.3. Antioxidant Defense Mechanisms and Stabilization of Redox Homeostasis in Cancer Cells

The enhanced content of reactive species in malignant cells, which mainly results from dysfunctional mitochondria, is associated with oxidative stress and the upregulation of antioxidative mechanisms [80]. Hypoxia stimulates the production of reactive species in the mitochondria. In turn, H2O2 stabilizes HIF-1α [218,219,220]. The participation of reactive species in carcinogenesis is supported by several studies [221,222,223,224,225]. These species affect anchorage-independent cell growth, epithelial-to-mesenchymal transition, angiogenesis, and apoptosis [226]. As high levels of reactive species are lethal, numerous antioxidative mechanisms are upregulated in cancer cells [227,228].
A key sensor for oxidative stress is Nrf2 that is low expressed under normal physiological conditions. Cytosolic Nrf2 is under the control of a Kelch-like erythroid cell-derived protein, with CNC homology [ECH]-associated protein 1 (Keap1) and Cul3 forming a complex with Nrf2 and inducing its ubiquitination and proteasomal degradation [229]. Under oxidative and electrophilic stress, Nrf2 is released from this complex and can mediate transcriptional processes in the nucleus. Nrf2 promotes the synthesis of several enzymes involved in the detoxification of reactive species (thioredoxin 1, thioredoxin reductase 1, thioredoxin interacting protein, peroxiredoxins, glutaredoxins, glutathione peroxidase, glutathione reductase, sulfiredoxin), the biosynthesis of glutathione, enzymes involved in the phase II and phase III inactivation of xenobiotics, and proteins controlling iron metabolism [94,95]. Enzymes such as glucose-6-phosphate dehydrogenase and 6-phosphogluconate dehydrogenase involved in the pentose phosphate pathway for the generation of NADPH are also upregulated by Nrf2 [230]. This pathway also produces ribulose-5-phosphate, which is necessary for enhanced nucleotide synthesis in malignant tumors [231].
Nrf2 is able to inhibit the initiation of carcinogenesis. Otherwise, it promotes the progression of already transformed cells [232,233]. In several human cancers, Nrf2 is stabilized by a reduced Nrf2-Keap1 interaction due to missense mutations in the keap1 and Nrf2 genes [228,234,235,236]. In cancer cells, Nrf2 can be activated by mutations or the epigenetic silencing of Keap1 as well as by the transcriptional activation via oncogenes like k-ras, c-myc, or b-raf [237].
Redox homeostasis is based on the production of NADPH as a universal source of electrons [238,239]. As a cofactor of glutathione reductase and thioredoxin reductase, NADPH ensures the recovery of GSH from oxidized glutathione (GS-SG) and reduced thioredoxin (Trx(-SH, -SH)) from the oxidized form (Trx(-S-S-)) [240]. GSH is an essential element in GPX-mediated reactions, the reduction in disulfides, and the reduction in glutaredoxin. Thioredoxin is involved in the reduction in peroxiredoxins, in ribonucleotide reductase-mediated reactions, and the recovery of methionine sulfoxides by methionine sulfoxide reductases (MSRs). With these reactions, NADPH ensures the maintenance of the thiol and methionine groups in cellular proteins and small molecules like GSH [238,241].
At the initiation of tumorigenesis, antioxidative systems may exert an anti-tumor role, as shown in the examples for peroxiredoxins [226], components of the thioredoxin system [242], and glutathione peroxidase 1 [243]. However, in growing tumors, the antioxidative mechanisms are essential for protecting tumor cells from excessive damage and apoptosis induction by reactive species. In late-stage cancer cells, a higher expression of NrF2, HIF-1α, and UPR promotes a marked upregulation of the aforementioned intracellular antioxidative defense mechanisms and systems promoting the maintenance of redox homeostasis, which makes cancer cells insensitive against oxidative stress and additional stress induced by chemo- and radiotherapy. Examples of the upregulation of these mechanisms in late-stage tumors are listed in Table 1. An overview of the major protective systems in cancer cells is also given in Figure 3.
Few remarks are necessary concerning the detoxification of methionine sulfoxides, which exists in two chiral forms, the R- and S-form of methionine sulfoxide. MSR-A is specific to the S-form of methionine sulfoxides. The downregulation of MSR-A is associated with a more aggressive phenotype of cancer cells in breast cancer [244]. In contrast, the upregulation of the MSR-B types, which reduce the R-forms of methionine sulfoxides, contributes to cancer growth in different cancer types [245,246,247].
As the upregulation of the aforementioned antioxidative mechanisms contributes to tumor growth and disease progression, the pharmacological inhibition of these enzymes is applied as a potential therapeutic trial against cancers.
Table 1. Overexpression of the defense systems against reactive species and systems maintaining redox homeostasis in cancer cells of late-stage cancers.
Table 1. Overexpression of the defense systems against reactive species and systems maintaining redox homeostasis in cancer cells of late-stage cancers.
Antioxidative SystemFunctionsCancer TypesReferences
SOD1Removal of O2•−, formation of H2O2Lung and breast cancer[248,249]
SOD2Removal of O2•−, formation of H2O2Esophageal cancer and others[250,251,252]
CatalaseRemoval of H2O2Glioblastoma[253]
Peroxiredoxins (six subtypes)Removal of H2O2Many types of tumors[226,254]
Thioredoxin system
(thioredoxin, thioredoxin reductase; NADPH)
Removal of H2O2, redox homeostasisLung, breast, prostate, colorectal, pancreatic, hepatocellular, gastric, and other carcinomas[255,256]
Cystine/glutamate anti-porter solute carrier family 7 member 11Cystine/cysteine uptake in cells for GSH productionMultiple types of cancer[117,118]
Glutathione (GSH)Redox homeostasis, cofactor of glutathione peroxidasesOvarian, breast, and lung cancers[257,258,259]
Glutathione reductaseRecovery of GSHGlioblastoma[260]
Glutathione peroxidase 4Removal of H2O2 and lipid hydroperoxides, inhibition of ferroptosisThyroid, colorectal, and esophageal carcinomas [261,262]
Glutathione peroxidase 1Removal of H2O2, peroxynitrite, and lipid hydroperoxidesBreast, hepatocellular, colorectal, esophageal, and lung carcinomas[243]
Glutathione peroxidase 2Removal of H2O2Liver, colorectal, breast, lung, and prostate carcinomas[263,264]
Glutaredoxins 3 and 5Redox homeostasisLiver and colorectal cancers[265]
sulfiredoxinRedox homeostasisSkin, liver, and colorectal cancers[266]
Ribonucleotide reductaseRedox homeostasisGastric, ovarian, bladder, colorectal cancers[267,268]
Methionine sulfoxide reductase B1 and B3Redox homeostasisHepatocellular carcinoma and others[245,246,247]
Glucose-6-phosphate dehydrogenaseNADPH production, redox homeostasisMultiple types of cancer[269,270]
6-phosphogluconate dehydrogenase NADPH production, redox homeostasisMultiple types of cancer[271,272]

3.4. Disturbed Iron Homeostasis in Cancer Cells

As redox-sensitive iron ions (Fe2+ and Fe3+) are involved in tissue-damaging reactions like Fenton reaction, all the aspects of iron metabolism are strictly controlled to avoid an increase in free iron ions [273,274]. In blood, iron ions are mainly sequestered by transferrin, which is the ligand to the transferrin receptor on the cell surfaces [202,203]. In cells, iron is stored inside ferritin, which represents a hollow sphere [204].
Malignant cancer cells require iron for all the processes of tumor progression. Transferrin receptor 1 is highly upregulated in cancer cells [275]. Ferritin is overexpressed in esophageal adenocarcinoma, glioblastoma, and breast and colorectal carcinomas [276]. The iron exporter protein ferroportin has a lower expression in prostate and breast cancers and is associated with a poor prognosis in pancreatic cancer [277,278,279]. These mechanisms contribute to a higher iron load in cancer cells and concomitant protection against iron-mediated damaging reactions. These data are summarized in Table 2 and included in Figure 3.

3.5. Intratumoral Hemorrhages and Free Heme

Intratumoral hemorrhages are a common clinical manifestation in patients with lung, bladder, gastric, or colorectal cancers [124]. They result from a rupture of leaky blood vessels. In these hemorrhages, hemoglobin is released from defective red blood cells and rapidly oxidized to methemoglobin, which is unable to bind dioxygen. This oxidation is mostly favored by NO. Thus, an excessive release of hemoglobin diminishes the bioavailability of NO [283,284]. In circulating blood, methemoglobin is scavenged by haptoglobin and the resulting haptoglobin–heme protein complex is removed by binding to CD163 expressed on liver and spleen macrophages [285,286].
In a similar way, myoglobin released from damaged muscles is oxidized to metmyoglobin, which is eliminated from blood via scavenging by haptoglobin [287,288]. Both methemoglobin and metmyoglobin are known to liberate ferric protoporphyrin IX, also known as free heme or labile heme [289]. In blood, free heme is antagonized by hemopexin through the formation of a high-affinity complex, which is cleared by hepatocytes via binding to CD91 [290].
In the context of inflammatory processes, free heme acts both as a DAMP being a ligand to TLR4 [291,292] and as a strong cytotoxic agent [16]. It promotes numerous cytotoxic effects, such as the induction of oxidative processes in membranes, lipoproteins, and hydrophobic areas of proteins [293,294], the induction of hemolytic processes in unperturbed red blood cells [295,296], and promotes serious damage in the kidneys and liver [297,298]. Serious health problems are associated with excessive intravascular hemolysis and rhabdomyolysis leading to a limited availability of haptoglobin and especially hemopexin. Elevated levels of free heme are found in malaria, sickle cell disease, and porphyria [299].
In tumors, hemorrhages and enhanced hemoglobin levels are associated with a poor clinical outcome for affected patients [124,300,301]. An inverse correlation between the level of free heme and hemopexin was found in the blood of patients with prostate cancer [299]. As shown in prostate cancer cells, external free heme is taken up by these cells and accumulates in the nucleus, where it intercalates into parallel guanine-rich quadruplex DNA and RNA structural elements (so-called G4 elements) [125,126]. In this way, free heme promotes the expression of heme-responsive genes that are involved in heme degradation and iron sequestration. Intercalating heme also controls the expression of the key target genes, including telomeres and oncogene promoters [127,128]. Free heme targets the promoter region of the potent oncogene c-Myc [299] that is expressed in about 80% of human malignancies [302].
In hemopexin knockout mice, the most aggressive tumor phenotype was detected supporting the view that free heme promotes tumor growth and metastasis in prostate cancer [299]. Free heme also induces the accumulation of nuclear heme oxygenase-1, which contributes to the unwinding of the G4 elements and an increase in the genomic instability in cancer cells [303].

3.6. Protection against Free Heme by Heme Oxygenase 1 (HO-1)

Within cells, free heme is detoxified by HO-1 in an O2 and NADPH-dependent reaction to yield carbon monoxide (CO), Fe2+, and biliverdin, which is further metabolized by biliverdin reductase to bilirubin [304,305].
In tumor cells, HO-1 exhibits anti-oxidative, anti-inflammatory, anti-apoptotic, and pro-angiogenic effects [306]. HO-1 also activates autophagy and contributes to the chemoresistance of breast cancer cells [307]. During tumorigenesis, HO-1 is highly upregulated in multiple cancer types, including breast, prostate, liver, gastric, pancreatic, colorectal, and esophageal cancers, and neuroblastoma (Table 2) [282]. This upregulation is associated with therapy resistance and a poor prognosis for patients. Within the mitochondria, HO-1 protects these organelles from mitochondrial free heme and prevents apoptotic cell death [308].

4. Cytotoxic Agents in the TME

4.1. Special Conditions in the TME

Tumor cells are encompassed by the TME, which contains numerous cell types, including mesenchymal stem cells, fibroblasts, endothelial cells, and a variety of immune cells as well as extracellular matrix (ECM) components. Unlike healthy tissue, the conditions in the TME are dictated by hypoxia, an enhanced basic level of H2O2, acidosis, deficiencies in essential nutrients and metabolites, and by the presence of bioactive agents such as inflammation-resolving cytokines, growth factors, and mediators released from tumor and stromal cells. Tumor cells affect their environment through the release of exosomes containing numerous active agents that promote tumor progression, immunosuppression, and remodeling of the TME. Exosomes are also involved in the angiogenesis, migration, and invasion of cancer cells, and play a role in the formation of a metastatic niche [41,309,310]. Altogether, in advanced cancers, special conditions in the TME contribute to tumor growth and progression.
The presence of TGF-β, which is derived from tumor cells and some other cells in the TME, is a key factor affecting many aspects of tumorigenesis. Although TGF-β acts as a tumor suppressor in the early stages of disease, it develops to a potent tumor promoter in the late stages of cancer [311,312,313,314]. In the TME, TGF-β is a major metabolic driver and plays a crucial role in the reprogramming of immune cells favoring immune tolerance and cancer progression [313,315,316]. Moreover, TGF-β is essentially involved in the development of resistance against anti-cancer therapies. Thus, the inhibition of TGF-β signaling in combination with other therapeutic approaches should be a promising tool for the treatment of therapy-resistant cancer forms [313].

4.2. Peculiarities of Cytotoxic Agents in the TME

In the TME, the contribution of cytotoxic agents and their protective mechanisms in tumorigenesis is more complex than in tumor cells. In addition to oxidant-based cytotoxic agents like reactive species and free heme, proteases like elastase, matrix metalloproteases (MMPs) also exhibit enhanced activities in comparison to healthy tissue areas. This also concerns angiotensin II, which is generated from angiotensin I and angiotensinogen by serine proteases. In the TME, cytotoxic agents are derived from stromal cells and immune cells, especially from neutrophils and G-MDSCs. Neutrophils and neutrophil subpopulations are able to generate O2•− and H2O2 and activate the heme protein myeloperoxidase, which exhibits a pronounced halogenation and peroxidase activity [169,317,318]. These cells also secrete the serine proteases elastase, cathepsin G, and proteinase 3 [319]. Specifically, elastase has a wide range of substrates, including the components of the ECM.
However, the status of protective mechanisms against cytotoxic agents is not well defined. In the TME, other protective principles dominate against cytotoxic agents in contrast to tumor cells. The dominating protective systems are the proteins controlling the transition free metal ions (ceruloplasmin, transferrin), proteins avoiding the formation of free heme (haptoglobin, hemopexin), antiproteases inhibiting serine proteases, TIMPs inactivating MMPs, and small antioxidative acting molecules (urate, ascorbate, polyphenols). Some of the mentioned proteins are acute-phase proteins. As the data on these antagonizing principles are usually determined in the serum and other fluids of cancer patients, these values only badly reflect real conditions in pericellular areas of tumor cells. Furthermore, the TME does not represent a unique structure. Instead, it is a poor perfused region with multiple gradients for dioxygen, nutrients, and metabolites towards the tumor center.
Often protective mechanisms are more expressed in cancer patients in comparison to healthy individuals as found, for example, in ceruloplasmin [320,321], haptoglobin [322], hemopexin [323], and some protease inhibitors [324,325,326]. This higher expression can be explained as a response to the ongoing inflammatory process. However, in some cases, opposed data are obtained for some protective mechanisms with a dependence on certain cancer types. The selected examples are diminished haptoglobin values in hepatocellular carcinoma [327], reduced hemopexin levels in prostate cancer patients [299], and downregulated ceruloplasmin in adrenocortical and hepatocellular carcinomas [328,329]. In addition, the expression of protective systems depends not only on the presence of tumors but is also related to a patient’s specific physiological status and existing non-tumorous diseases. Thus, it remains very challenging to draw meaningful conclusions for the balance between TME-related cytotoxic agents and antagonizing principles in cancer patients.
By analyzing the haptoglobin protein expression in patients with hepatocellular carcinoma using immunohistochemical staining, it was observed that this protein was highly expressed in adjacent non-tumorous cells, but rarely detectable in the tumor tissue [327]. Within the tumor, a very low haptoglobin level was found in tumors with poorly differentiated cancer, a condition with poor prognosis [327]. This study showed that great differences may exist for the expression of protective principles between tumors and healthy tissue regions.
In addition to the upregulation or downregulation of the protective principles, strategies for tumors against cytotoxic agents in the TME also comprise the depletion of essential metabolites and nutrients as well as an enhanced production of the ECM components.

4.3. Neutrophil Subpopulations with the Tumor-Promoting Phenotype

Mature neutrophils are known for their anti-tumoral activities [330,331,332]. In contrast, neutrophilia and a high ratio between neutrophils and lymphocytes are associated with a poor clinical outcome in many cancers [333].
In circulating blood of cancer patients, two subpopulations of neutrophils accumulate, which both exhibit pro-tumoral characteristics. Apart from the aforementioned G-MDSCs, which represent an immature phenotype of granulocytes, mature neutrophils are converted under the action of TGF-β released from tumor cells and different stromal cells into a tumor-promoting phenotype [334]. The latter type has been called N2-type neutrophils in analogy to macrophage phenotypes. Both G-MDSCs and N2-type neutrophils have a lower density than mature neutrophils.
In the expansion and activation of MDSCs, the activation of STAT3 plays a critical role. This transcription factor mediates the expression of the arginase 1, IL-10, PL-D1, S-100 proteins, which are all involved in immunosuppressive activities [32,35]. Moreover, the STAT3 expression is regulated by several microRNAs [335,336,337].

4.4. Involvement of Myeloperoxidase (MPO) in the Immunosuppression by G-MDSCs

In cancers, the production of reactive species by G-MDSCs mediates the immunosuppressive activity against CD8+ T cells [338,339]. Murine G-MDSCs exhibit a higher activity of arginase, MPO, and intracellular reactive species than murine neutrophils [339]. The MPO activity is lower in MDSCs from tumor-bearing mice compared to MDSCs from tumor-free mice [338].
The immunosuppressive function of G-MDSCs is related to the transfer of lipid bodies with oxidatively truncated lipids to DCs, where these lipids inhibit the cross-presentation of tumor-associated antigens and impair anti-tumor CD8+ T cell responses unlike lipid bodies with non-oxidized lipids [340]. Thereby, the interaction of modified lipids with chaperones blocks the antigen cross-presentation in DCs [341]. In G-MDSCs, a concerted action of NADPH oxidase and MPO is responsible for the formation of oxidized, truncated lipids in G-MDSCs. The involvement of MPO was evidenced in the experiments using MPO-knockout mice and a pharmacological inhibition of MPO [340]. It remains puzzling how MPO favors the oxidation of lipids in G-MDSCs. The immunosuppressive activities of G-MDSCs depend on the uptake of lipids via the scavenger receptor CD36 and with the help of the fatty acid transport protein 2, as fatty acid oxidation is the main energy source in tumor-associated G-MDSCs [342,343,344]. Apparently, MPO oxidizes lipid droplets inside G-MDSCs.
In another study, both genetic and pharmacological blockades of MPO increased the success of immune checkpoint therapy in experimental models with primary melanoma [345]. There was a significant increase in MPO in melanoma tissue arrays. However, no details were given regarding how MPO contributed to the resistance against immune checkpoint therapy [345].

4.5. Elastase-Mediated Effects in the TME

In a serum-free incubation medium, elastase released from human neutrophils is able to kill a large variety of cancer cells [346]. However, these data are irrelevant as the role of important elastase substrates, like serum antiproteases and components of the extracellular matrix, were not considered in these experiments. The current view is that elastase promotes tumorigenesis and metastasis [347]. This was evidenced by applying elastase knockout mice and a pharmacological inhibition of elastase in cancer models [347,348,349]. A deficiency of α1-antitrypsin, an efficient antagonist against elastase, is a risk factor for the development of hepatic and lung cancer [350,351].
Moreover, elastase is significantly enhanced in several tumors [352,353,354]. This protease is mainly derived from invading neutrophils and myeloid-derived suppressor cells. The detrimental activity of elastase in tumors is associated with a proteolytic degradation of the components of the vessel wall and extracellular matrix. This facilitates tumor cell intravasation and dissemination via elastase-mediated dilated intratumoral blood vessels [347]. Elastase has a diversified substrate spectrum, including elastin, laminin, and transmembrane proteins (E-cadherin, VCAM-1, G-CSF receptor), cytokines (IL-1, G-CSF), vascular endothelial growth factors (VEGFs), and precursors of angiotensin II [347]. It activates MMP2, MMP3, and MMP9 from inactive precursors, and degrades TIMP-1 contributing to ECM degradation [355,356,357]. In the TME, some potential elastase substrates are overexpressed, like VEGFs [70,71], several MMPs [154,155], elastin [358,359], and laminins [360].
The inhibitory activity of antiproteases depends on two factors. First, the oxidation of critical residues at active site of antiproteases is associated with a loss to inhibit elastase. In α1-antitrypsin, these critical residues are Met-351 and Met-358 [361]. Second, the cationic elastase is known to bind to negatively charged components of the extracellular matrix and surfaces. Matrix- and surface-associated elastase cannot be inhibited by antiproteases [362]. In tumors, the high abundance of matrix components apparently favors the enhanced activity of elastase.
In the same cancers, the overexpression of antiproteases is associated with tumor progression and metastasis, as found for elafin in hepatocellular carcinoma [325], the secretory leukocyte protease inhibitor in colorectal cancer [324], or α1-chymotrypsin in pancreatic cancer [326]. Otherwise, the enhanced α1-chymotrypsin expression promotes survival in liver cancer [326].

4.6. Role of Angiotensin II

As part of the renin-angiotensin-aldosteron system, angiotensin II contributes to the regulation of blood pressure and water metabolism. It is formed under the action of the angiotensin converting enzyme (ACE) from the decapeptide angiotensin I. Under inflammatory conditions, neutrophil-derived serine proteases cathepsin G, elastase, and proteinase 3 release angiotensin II from angiotensin I and angiotensinogen [363,364,365]. Angiotensin II promotes actin cleavage, proteolysis, increased apoptosis, caspase-3 activation, and the lowering of IGF-1 [366,367]. Further effects of angiotensin II are the activation of NADPH oxidase, the production of reactive species, such as O2•− and H2O2 [366,367], and the promotion of the ubiquitin-proteasome proteolytic pathway [368].
Angiotensin II is under the control of ACE2, which converts it into anti-inflammatory angiotensin 1–7. The inhibition of ACE2 by coronaviruses, including SARS-CoV-2, enhanced the impact of angiotensin II on lung damage [369,370].
The effects of angiotensin II are mediated by ligation to angiotensin II receptors (ATR), most of all to ATR1. The overexpression of this receptor plays a significant role in cancer aggressiveness and angiogenesis in various kinds of cancers, such as ovary, bladder, lung, and breast cancers [371,372,373,374,375,376].
Angiotensin receptor blockers are widely used as medication for the treatment of hypertension, heart failure, diabetic nephropathy, and to prevent cardiovascular events [377,378]. The risk of cancer, especially of lung cancer, increases with treatment using these drugs [379].

4.7. NO Metabolism in the TME

Generally, the NO effects on tumors are concentration dependent. At low concentrations, NO promotes tumor progression via apoptosis inhibition and the enhancement of angiogenesis and metastasis through the activation of several survival pathways. At higher concentrations, NO becomes cytotoxic via nitrosative stress, DNA damage, and apoptosis induction [380,381].
Several factors reduce the bioavailability of NO in tumors (Figure 4). Arginase 1 and arginase 2 compete with NO synthases for L-arginine, limiting the production of NO. Both arginases are overexpressed in various cancer types, like breast, gastric, colorectal, and liver cancers [382,383,384,385]. Arginases produce L-ornithine, which is further metabolized to yield proline for collagen synthesis and polyamines involved in tumor cell proliferation, migration, and invasion [386]. In the TME, arginases from tumor cells, MDSCs, macrophages, and Tregs contribute to the depletion of L-arginine and impair T cell functions and tumor perfusion [387].
In tumor hemorrhages, the release of hemoglobin from leaky red blood cells also contributes to a reduced bioavailability of NO as the oxidation of hemoglobin to methemoglobin is usually promoted by NO [283,284]. Methemoglobin, which is under the control of haptoglobin, liberates cytotoxic free heme. When not antagonized by hemopexin, this agent is able to promote the release of hemoglobin from yet unperturbed red blood cells [289,290], diminishing the bioavailability of NO.
Further, NO reacts very rapidly with O2•− to yield peroxynitrite [185,186]. In addition to the production of O2•− by dysfunctional mitochondria inside tumor cells, O2•− can also result from the activity of NADPH oxidase (also known as NOX2) present in neutrophils and many other cells in the TME [388,389], from xanthine oxidase-mediated reactions [390,391], and from the autoxidation of hemoglobin and myoglobin [392,393]. In the extracellular medium, O2•− is under the control of SOD3, the secretory form of SODs. Although SOD3 is found in many tissues, higher SOD3 values are known for their well vascularized tissues, such as lung, kidney, uterus, placenta, and blood vessels [394]. In many tumors, SOD3 is downregulated and promotes cancer metastasis and malignant progression [395,396,397,398]. A reduced SOD3 expression diminishes the bioavailability of NO, as NO reacts easily with O2•− to yield peroxynitrite. The latter agent damages the components of the ECM and endothelial cells, contributes to leaky blood vessels in tumors, and reduces the invasion of T cells into tumors [399,400]. Peroxynitrite is also able to deactivate antiproteases through the oxidation of critical residues at active sites, enhancing the destructive reactions mediated by elastase [401].
In contrast, an overexpression of mRNA of SOD3 correlates with increased benign growth and is associated with apoptosis induction and the death of cancer cells [397]. The removal of O2•− by SOD3 improves vasorelaxation by NO and enables NO to activate HIF-2α [402]. In turn, the HIF-2α expression in endothelial cells activates the WNT signaling pathway, where the components are involved in the stabilization and normalization of the endothelium, including the enhanced transcription of vascular endothelial cadherin (VED) [399]. This improves T cell extravasation from blood vessels into the TME and enhances anti-tumor activities [402]. An overexpression of SOD3 also attenuates the activity of MMPs and heparinase, which are involved in matrix degradation, angiogenesis, and metastasis [403].

4.8. Role of the Extracellular Matrix Components in the TME

Different factors contribute to remodeling of the extracellular matrix (ECM) in the TME. This remodeling is accompanied by an increased stiffness of the ECM and an increased synthesis and decomposition of the matrix components like collagens, hyaluronan, and others.
In healthy tissues, a high molecular weight hyaluronan is essential for maintaining tissue homeostasis. It exhibits anti-inflammatory and anti-proliferative properties and contributes to tumor resistance [404,405]. The fragmentation of high molecular weight hyaluronan by hyaluronidases or reactive species into low molecular weight polymers promotes tumor cell migration and proliferation as well as immune cell influx in the TME [406,407].
During the resolution of inflammation, the initiation of repair processes is indispensable for restoring homeostasis in the affected tissue regions. Under the action of TGF-β secreted from M2-type macrophages and some other cells, the components of the extracellular matrix (ECM) proteins, like collagen, are mainly produced by myofibroblasts during tissue repair [408,409]. At the same time, different TIMPs are also released by these cells to prevent matrix degradation by MMPs. Whereas in normal wound healing the generation of ECM proteins and TIMPs is transient and counter regulated by various MMPs that destroy excess matrix components, the accumulation of ECM components and scar formation dominates in fibrotic tissues.
An overexpression of MMP-2, MMP-3, MMP-9, and MMP-14 is associated with tumor metastasis [154,155]. MMP-2 and MMP-9 are involved in collagen IV degradation in the basement membrane [410]. MMP-14 contributes to collagen digestion around tumor cells and favors cell invasion and migration [411]. MMP-9 also exhibits gene modulatory functions upon binding to the α4β1 integrin in chronic lymphocytic leukemia [412].
MMPs are controlled by TIMPs. In different cancer types, TIMP-1 is upregulated and associated with a poor prognosis [413,414]. Increased levels of TIMP-1 are also associated with cachexia in patients with chronic pancreatitis and pancreatic cancer [415]. However, in another study, TIMP-1 was indeed associated with pancreatic cancer, but not with a significant weight loss [416]. Contrariwise, TIMP-3 suppresses tumor progression and promotes apoptosis induction [417,418]. Low levels of TIMP-3 increase the activity of MMPs in cancer [419]. Different cytokines and micro RNAs are involved in regulation of the expression of TIMP-1 and TIMP-3 [413,420].
Despite chemotherapy and surgical resection, numerous patients with triple-negative breast cancer developed a recurrence of the disease. In the TME, collagen IV has been identified by proteomic approaches as a major driver of enhanced cancer cell motility and metastasis after chemotherapy in both mice and patients with this kind of cancer [421]. An overexpression of collagen IV is known for pancreatic, gastric, colorectal, and breast cancers [422]. High circulating levels of collagen IV are associated with a poor clinical outcome [423,424]. In the basement membrane, the activity of the heme peroxidase peroxidasin is essential for cross-linking reactions between collagen IV fibrils [425,426]. This peroxidase promotes angiogenesis through extracellular signal-regulated kinases 1/2, protein kinase B, the focal adhesion kinase pathways, and the induction of several pro-angiogenic genes [427]. In invasive metastatic melanoma cells, a high expression of peroxidasin was reported [428]. Peroxidasin is upregulated in several kinds of cancers [429]. Abnormal collagen IV cross-linking by peroxidasin leads to an exceedingly dense ECM and a disordered matrix structure, as reported for lung tissue. In consequence, T cell infiltration is highly disturbed [430].

5. Conclusions

In tumorigenesis, different oxidant-based and protease-based cytotoxic agents are upregulated. Nevertheless, antagonizing principles are usually highly active to diminish the disastrous effects of these cytotoxic agents and to provide survival for tumors as well as tumor progression and metastasis.
In late-stage tumor cells, the enhanced formation of reactive species, increase in iron ions, and elevated values of free heme are antagonized by an overexpression of the numerous anti-oxidative protective systems. In addition, the pathways contributing to the maintenance of redox homeostasis are also upregulated in tumor cells despite existing in oxidative stress conditions. These counter regulations limit the cytotoxic activities of reactive species, metal ions, and free heme, protect tumor cells from the induction of cell death mechanisms, and prevent them from experiencing additional stress caused by therapeutic approaches like radio- and chemotherapy.
It is necessary to emphasize that an overexpression of these protective mechanisms was developed step by step during tumorigenesis and reached their maximum characteristic in late-stage tumors, especially in those tumors, which were malignant and prone to metastasis. As long as these antagonizing systems against oxidative stress are not fully developed, the probability exists to affect tumors successfully through different therapeutic options. Of course, scientists worldwide are trying to implement novel anti-tumor therapies based on the inhibition and/or downregulation of antioxidative mechanisms in tumors. Then, the cytotoxic potential of stress-related reactive species, metal ions, and free heme can better be applied to substantially damaged tumor cells.
Tumor cells affect their immediate neighborhood through the secretion of active agents, creating a pro-tumoral environment characterized by immunosuppression and an elevated production of ECM elements. During the resolution phase of inflammation, similar processes occur at inflammatory sites with the difference that they act only temporary but not permanently, as in tumors.
In the TME, the contribution of cytotoxic agents and their protective mechanisms is more complex than in tumor cells. In addition to reactive species and free heme, elastase, angiotensin II, and MMPs also exhibit elevated activities in comparison to healthy tissue areas. In the TME, cytotoxic agents are mainly derived from immune cells and stromal cells. Strategies against these cytotoxic agents include the upregulation or downregulation of protective principles, the depletion of essential metabolites, and the enhanced production of the ECM.
Superoxide anion radicals, hemoglobin released from leaky red blood cells, and free heme favor the depletion of NO and hinder the activation of cytotoxic T cells. These effects are elevated by low values of SOD3 and hemopexin as well as by the enhanced expression of arginases and the presence of hemoglobin released from leaky red blood cells.
Collagen and other ECM components form a barrier around tumor cells against invading immune cells. The increased production of the ECM is associated with tumor fibrosis. Otherwise, elastase, MMPs, and heparinase are involved in local matrix degradation that highly promotes tumor invasion and metastasis.
To sum up, despite an enormous knowledge about the interplay between cytotoxic agents and antagonizing principles in tumorigenesis, we are far from a thorough understanding how these mechanisms are embedded into the whole machinery of tumor-induced immunosuppression and metabolic alterations. Moreover, the individual expression of cytotoxic agents and protective mechanisms can vary considerably from patient to patient. It is a very great challenge to implement novel therapeutic approaches for individual cancer patients.

Funding

This research received no external funding.

Conflicts of Interest

The author declares no conflict of interests.

References

  1. Balkwill, F.; Mantovani, A. Inflammation and cancer: Back to Virchow? Lancet 2001, 357, 539–545. [Google Scholar] [CrossRef] [PubMed]
  2. Mantovani, A.; Allavena, P.; Sica, A.; Balkwill, F. Cancer-related inflammation. Nature 2008, 454, 436–444. [Google Scholar] [CrossRef] [PubMed]
  3. Nakamura, K.; Smyth, M.J. Targeting cancer-related inflammation in the era of immunotherapy. Immunol. Cell Biol. 2017, 95, 325–332. [Google Scholar] [CrossRef] [PubMed]
  4. Arnhold, J. Immune response and tissue damage. In Cell and Tissue Destruction. Mechanisms, Protection, Disorders; Academic Press: London, UK; San Diego, CA, USA; Cambridge, MA, USA; Oxford, UK, 2020; pp. 155–204. [Google Scholar] [CrossRef]
  5. Arnhold, J. Acute-phase proteins and additional protective systems. In Cell and Tissue Destruction. Mechanisms, Protection, Disorders; Academic Press: London, UK; San Diego, CA, USA; Cambridge, MA, USA; Oxford, UK, 2020; pp. 205–228. [Google Scholar] [CrossRef]
  6. Wang, D.; DuBois, R.N. Immunosuppression associated with chronic inflammation in the tumor microenvironment. Carcinogenesis 2015, 36, 1085–1093. [Google Scholar] [CrossRef] [PubMed]
  7. Dunn, G.P.; Bruce, A.T.; Ikeda, H.; Old, L.J.; Schreiber, R.D. Cancer immunoediting: From immunosurveillance to tumor escape. Nat. Immunol. 2002, 3, 991–998. [Google Scholar] [CrossRef] [PubMed]
  8. Burnet, M. Cancer; a biological approach. I. The process of control. II. The significance of somatic mutation. Br. Med. J. 1957, 1, 779–786. [Google Scholar] [CrossRef] [PubMed]
  9. Thomas, L. On immunosurveillance in human cancer. Yale J. Biol. Med. 1982, 55, 329–333. [Google Scholar] [PubMed]
  10. Shimasaki, M.; Jain, A.; Campana, D. NK cells for cancer immunotherapy. Nat. Rev. Drug Discov. 2020, 19, 200–218. [Google Scholar] [CrossRef]
  11. Raskov, H.; Orhan, A.; Christensen, J.P.; Gögenur, I. Cytotoxic CD8+ T cells in cancer and cancer immunotherapy. Br. J. Cancer 2021, 124, 359–367. [Google Scholar] [CrossRef]
  12. Nakamura, K.; Smyth, M.J. Myeloid immunosuppression and immune checkpoints in the tumor microenvironment. Cell Mol. Immunol. 2020, 17, 1–12. [Google Scholar] [CrossRef]
  13. Labani-Motlagh, A.; Ashja-Mahdavi, M.; Loskog, A. The tumor microenvironment: A milieu hindering and obstructing antitumor immune response. Front. Immunol. 2020, 11, 940. [Google Scholar] [CrossRef]
  14. Barnestein, R.; Galland, L.; Kalfeist, L.; Ghiringhelli, F.; Ladoire, S.; Limagne, E. Immunosuppressive tumor microenvironment modulation by chemotherapies and targeted therapies to enhance immunotherapy effectiveness. OncoImmunology 2022, 11, 2120676. [Google Scholar] [CrossRef]
  15. Tie, Y.; Tang, F.; Wei, Y.-Q.; Wie, X.-W. Immunosuppressive cells in cancer: Mechanisms and potential therapeutic targets. J. Hematol. Oncol. 2022, 15, 61. [Google Scholar] [CrossRef]
  16. Arnhold, J. Host-derived cytotoxic agents in chronic inflammation and disease progression. Int. J. Mol. Sci. 2023, 24, 3016. [Google Scholar] [CrossRef]
  17. Matzinger, P. Tolerance, danger, and the extended family. Annu. Rev. Immunol. 1994, 12, 991–1045. [Google Scholar] [CrossRef]
  18. Janeway, C.A.; Medzhitov, R. Innate immune recognition. Annu. Rev. Immunol. 2002, 20, 197–216. [Google Scholar] [CrossRef]
  19. Suresh, R.; Moser, D.M. Pattern recognition in innate immunity, host defense, and immunopathology. Adv. Physiol. Educ. 2013, 37, 284–291. [Google Scholar] [CrossRef]
  20. Pepys, M.B.; Baltz, M.I. Acute phase proteins with special reference to C-reactive protein and related proteins (pentraxins) and serum amyloid A protein. Adv. Immunol. 1983, 34, 141–212. [Google Scholar] [CrossRef]
  21. Vandivier, R.W.; Henson, P.M.; Douglas, I.S. Burying the death: The impact of failed apoptotic cell removal (efferocytosis) on chronic inflammatory lung disease. Chest 2006, 129, 1673–1682. [Google Scholar] [CrossRef]
  22. Pober, J.S.; Sessa, W.C. Inflammation and the blood microvascular system. Cold Spring Harbor Perspect. Biol. 2015, 7, 016345. [Google Scholar] [CrossRef]
  23. Li, M.O.; Wan, Y.Y.; Sanjabi, S.; Robertson, A.K.; Flavell, R.A. Transforming growth factor-beta regulation of immune response. Annu. Rev. Immunol. 2006, 24, 99–146. [Google Scholar] [CrossRef]
  24. Li, M.O.; Flavell, R.A. Contextual regulation of inflammation: A duet of transforming growth factor-beta and interleukin-10. Immunity 2008, 28, 468–476. [Google Scholar] [CrossRef]
  25. Couper, K.N.; Blount, D.G.; Riley, E.M. IL-10: The master regulator of immunity to infection. J. Immunol. 2008, 180, 5771–5777. [Google Scholar] [CrossRef]
  26. Landén, N.X.; Li, D.; Ståhle, M. Transition from inflammation to proliferation: A critical step during wound healing. Cell Mol. Life Sci. 2016, 73, 3861–3885. [Google Scholar] [CrossRef]
  27. Marega, M.; Chen, C.; Bellusci, S. Cross-talk between inflammation and fibroblast growth factor 10 during organogenesis and pathogenesis: Lessons learnt from the lung and other organs. Front. Cell Dev. Biol. 2021, 9, 656883. [Google Scholar] [CrossRef]
  28. Serhan, C.N.; Savill, J. Resolution of inflammation: The beginning programs the end. Nat. Immunol. 2005, 6, 1191–1197. [Google Scholar] [CrossRef]
  29. Chandrasekharan, J.A.; Sharma-Walia, N. Lipoxins: Nature’s way to resolve inflammation. J. Inflamm. Res. 2015, 8, 181–192. [Google Scholar] [CrossRef]
  30. Murray, P.J.; Wynn, T.A. Protective and pathogenic functions of macrophage subsets. Nat. Rev. Immunol. 2011, 11, 723–737. [Google Scholar] [CrossRef]
  31. Martinez, F.O.; Gordon, S.; Locati, M.; Montavani, A. Transcriptional profiling of the human monocyte-to-macrophage differentiation and polarization: New molecules and patterns of gene expression. J. Immunol. 2006, 177, 7303–7311. [Google Scholar] [CrossRef]
  32. Sanchez-Pino, M.D.; Dean, M.J.; Ochoa, A.C. Myeloid-derived suppressor cells (MDSC): When good intentions go awry. Cell Immunol. 2021, 362, 104302. [Google Scholar] [CrossRef]
  33. Veglia, F.; Sanseviero, E.; Gabrilovich, D.I. Myeloid-derived suppressor cells in the era of increasing myeloid cell diversity. Nat. Rev. Immunol. 2021, 21, 485–498. [Google Scholar] [CrossRef]
  34. Groth, C.; Hu, X.; Weber, R.; Fleming, V.; Altevogt, P.; Utikal, J.; Umansky, V. Immunosuppression mediated by myeloid-derived suppressor cells (MDSCs) during tumor progression. Br. J. Cancer 2019, 120, 16–25. [Google Scholar] [CrossRef]
  35. Gabrilovich, D.I.; Nagaraj, S. Myeloid-derived suppressor cells as regulators of the immune system. Nat. Rev. Immunol. 2009, 9, 162–174. [Google Scholar] [CrossRef]
  36. Sepkowitz, K.A. Opportunistic infections in patients with and patients without acquired immunodeficiency syndrome. Clin. Infect. Dis. 2002, 34, 1098–1107. [Google Scholar] [CrossRef]
  37. Kampitak, T.; Suwanpimolkul, G.; Browne, S.; Suankratay, C. Anti-interferon-autoantibody and opportunistic infections: Case series and review of the literature. Infection 2011, 39, 65–71. [Google Scholar] [CrossRef]
  38. Franceschi, C.; Bonafè, M.; Valensin, S.; Olivieri, F.; De Luca, M.; Ottaviani, E.; De Beneticits, G. Inflammaging—An evolutionary perspective on immunosenescence. Ann. N. Y. Acad. Sci. 2000, 908, 244–254. [Google Scholar] [CrossRef]
  39. Newman, A.B.; Sanders, J.L.; Kizer, J.R.; Boudrou, R.M.; Odden, M.C.; Zeki Al Hazzouri, A.; Arnold, A.M. Trajectories of function and biomarkers with age: The CHS All stars study. Int. J. Epidemiol. 2016, 45, 1135–1145. [Google Scholar] [CrossRef]
  40. Ferrucci, L.; Fabbri, E. Inflammageing: Chronic inflammation in ageing, cardiovascular disease, and frailty. Nat. Rev. Cardiol. 2018, 15, 1135–1145. [Google Scholar] [CrossRef]
  41. Othman, N.; Jamal, R.; Abu, N. Cancer-derived exosomes as effectors of key inflammation-related players. Front. Immunol. 2019, 10, 2103. [Google Scholar] [CrossRef]
  42. Wrzesinski, S.H.; Wan, Y.Y.; Flavell, R.A. Transforming growth factor-β and immune response: Implications for anticancer therapy. Clin. Cancer Res. 2007, 13, 5262–5270. [Google Scholar] [CrossRef]
  43. Bierie, B.; Moses, H.L. Transforming growth factor β (TGF-β) and inflammation in cancer. Cytokine Growth Factor Rev. 2010, 21, 49–59. [Google Scholar] [CrossRef]
  44. Szczepanski, M.J.; Szajnik, M.; Welsh, A.; Whiteside, T.L.; Boyiadzis, M. Blast-derived microvesicles in sera from patients with acute myeloid leukemia suppress natural killer cell function via membrane-associated transforming growth factor-beta 1. Haematologia 2011, 96, 1302–1309. [Google Scholar] [CrossRef]
  45. He, Z.; Zhang, S. Tumor-associated macrophages and their functional transformation in the hypoxic tumor microenvironment. Front. Immunol. 2021, 12, 741305. [Google Scholar] [CrossRef]
  46. Ostrand-Rosenberg, S.; Sinha, P. Myeloid-derived suppressor cells: Linking inflammation and cancer. J. Immunol. 2009, 182, 4499–4506. [Google Scholar] [CrossRef]
  47. Umansky, V.; Blattner, C.; Gebhardt, C.; Utikal, J. The role of myeloid-derived suppressor cells (MDSC) in cancer progression. Vaccines 2016, 4, 36. [Google Scholar] [CrossRef]
  48. Hedge, S.; Leader, A.M.; Merad, M. MDSC: Marker, development, states, and unaddressed complexity. Immunity 2021, 54, 875–884. [Google Scholar] [CrossRef]
  49. Cassetta, L.; Bruderek, K.; Skrzeczynska-Moncznik, J.; Osiecka, O.; Hu, X.; Rundgren, I.M.; Lin, A.; Santegoets, K.; Horzum, U.; Godinhi-Santos, A.; et al. Differential expansion of circulating human MDSC subsets in patients with cancer, infection and inflammation. J. Immunother. Cancer 2020, 8, e001223. [Google Scholar] [CrossRef]
  50. Casetta, L.; Baekkevold, E.S.; Brandau, S.; Bujko, A.; Casatella, M.A.; Dorhoi, A.; Krieg, C.; Lin, A.; Loré, K.; Marini, O.; et al. Deciphering myeloid-derived suppressor cells: Isolation and markers in humans, mice and non-human primates. Cancer Immunol. Immunother. 2019, 68, 687–697. [Google Scholar] [CrossRef]
  51. Bronte, V.; Brandau, S.; Chen, S.-H.; Colombo, M.P.; Frey, A.B.; Greten, T.F.; Mandruzzato, S.; Murray, P.J.; Ochoa, A.; Ostrand-Rosenberg, S.; et al. Recommendations for myeloid-derived suppressor cell nomenclature and characterization standards. Nat. Commun. 2016, 7, 12150. [Google Scholar] [CrossRef]
  52. Sade-Feldman, M.; Jiao, Y.J.; Chen, J.H.; Rooney, M.S.; Barzily-Rokni, M.; Eliane, J.P.; Bjorgaard, S.L.; Hammond, M.R.; Vitzthum, H.; Blackmon, S.M.; et al. Resistance to checkpoint blockade therapy through inactivation of antigen presentation. Nat. Commun. 2017, 8, 1136. [Google Scholar] [CrossRef]
  53. Vinay, D.S.; Ryan, E.P.; Pawelec, G.; Talib, W.H.; Stagg, J.; Elkord, E.; Lichtor, T.; Decker, W.K.; Whelan, R.L.; Kumara, H.M.C.S.; et al. Immune evasion in cancer: Mechanistic basis and therapeutic strategies. Semin. Cancer Biol. 2015, 35, S185–S198. [Google Scholar] [CrossRef]
  54. Molinier-Frenkel, V.; Castellano, F. Immunosuppressive enzymes in the tumor microenvironment. FEBS Lett. 2017, 591, 3135–3157. [Google Scholar] [CrossRef]
  55. Bader, J.E.; Voss, K.; Rathmell, J.C. Targeting metabolism to improve the tumor microenvironment for cancer immunotherapy. Mol. Cell 2020, 78, 1019–1033. [Google Scholar] [CrossRef]
  56. Allard, B.; Longhi, M.S.; Robson, S.C.; Stagg, J. The ectonucleotidases CD39 and CD73: Novel checkpoint inhibitor targets. Immunol. Rev. 2017, 276, 121–144. [Google Scholar] [CrossRef]
  57. Moon, Y.W.; Hajjar, J.; Hwu, P.; Naing, A. Targeting the indoleamine 2,3-dioxygenase pathway in cancer. J. Immunother. Cancer 2015, 3, 51. [Google Scholar] [CrossRef]
  58. Kitamura, H.; Ohno, Y.; Toyoshima, Y.; Ohtake, J.; Homma, S.; Kawamura, H.; Takahashi, N.; Taketomi, A. Interleukin-6/STAT3 signaling as a promising target to improve the efficacy of cancer immunotherapy. Cancer Sci. 2017, 108, 1947–1952. [Google Scholar] [CrossRef]
  59. Park, S.-J.; Nakagawa, T.; Kitamura, H.; Atsumi, T.; Kamon, H.; Sawa, S.-I.; Kamimura, D.; Ueda, N.; Iwakura, Y.; Ishihara, K. IL-6 Regulates in vivo dendritic cell differentiation through STAT3 activation. J. Immunol. 2004, 1950, 3844–3854. [Google Scholar] [CrossRef]
  60. Voron, T.; Colussi, O.; Marcheteau, E.; Pernot, S.; Nizard, M.; Pointet, A.-L.; Latreche, S.; Bergaya, S.; Benhamouda, N.; Tanchot, C. VEGF-A modulates expression of inhibitory checkpoints on CD8+ T cells in tumors. J. Exp. Med. 2015, 212, 139–148. [Google Scholar] [CrossRef]
  61. Ohm, J.E.; Carbone, D.P. VEGF as a mediator of tumor-associated immunodeficiency. Immunol. Res. 2001, 23, 263–272. [Google Scholar] [CrossRef]
  62. Huang, H.; Langenkamp, E.; Georganaki, M.; Loskog, A.; Fuchs, P.F.; Dieterich, L.C.; Kreuger, J.; Dimberg, A. VEGF suppresses T-lymphocyte infiltration in the tumor microenvironment through Inhibition of NF-κB-induced endothelial activation. FASEB J. 2015, 29, 227–238. [Google Scholar] [CrossRef]
  63. Ohue, Y.; Nishikawa, H. Regulatory T (Treg) cells in cancer: Can Treg cells be a new therapeutic target? Cancer Sci. 2019, 110, 2080–2089. [Google Scholar] [CrossRef] [PubMed]
  64. Patel, S.P.; Kurzrock, R. PD-L1 expression as a predictive biomarker in cancer immunotherapy. Mol. Cancer Ther. 2015, 14, 847–856. [Google Scholar] [CrossRef] [PubMed]
  65. Walunas, T.L.; Lenschow, D.J.; Bakker, C.Y.; Linsley, P.S.; Freeman, G.J.; Green, J.M.; Thompson, C.B.; Bluestone, J.A. CTLA-4 can function as a negative regulator of T cell activation. Immunity 1994, 1, 405–413. [Google Scholar] [CrossRef]
  66. Ribas, A.; Wolchok, J.D. Cancer immunotherapy using checkpoint blockade. Science 2018, 359, 1350–1355. [Google Scholar] [CrossRef] [PubMed]
  67. Kawashima, S.; Inozume, T.; Kawazu, M.; Ueno, T.; Nagasaki, J.; Tanji, E.; Honobe, A.; Ohnuma, T.; Kawamura, T.; Umeda, Y. TIGIT/CD155 axis mediates resistance to immunotherapy in patients with melanoma with the inflamed tumor microenvironment. J. Immunother. Cancer 2021, 9, e003134. [Google Scholar] [CrossRef] [PubMed]
  68. Konerding, M.A.; Fait, E.; Gaumann, A. 3D microvascular architecture of pre-cancerous lesion and invasive carcinomas in the colon. Br. J. Cancer 2001, 84, 1354–1362. [Google Scholar] [CrossRef]
  69. Siemann, D.W. The unique characteristics of tumor vasculature and preclinical evidence for its selective disruption by tumor-vascular disrupting agents. Cancer Treat. Rev. 2011, 37, 63–74. [Google Scholar] [CrossRef] [PubMed]
  70. Jubb, A.M.; Pham, T.Q.; Hanby, A.M.; Frantz, G.D.; Peale, F.V.; Wu, T.D.; Koeppen, H.W.; Hilan, K.J. Expression of vascular endothelial growth factor, hypoxia inducible factor 1 alpha, and carbonic anhydrase IX in human tumours. J. Clin. Pathol. 2004, 57, 504–512. [Google Scholar] [CrossRef]
  71. Shweiki, D.; Itin, A.; Soffer, D.; Keshet, E. Vascular endothelial growth factor induced by hypoxia may mediate hypoxia-initiated angiogenesis. Nature 1992, 359, 843–845. [Google Scholar] [CrossRef]
  72. Roskoski, R., Jr. Vascular endothelial growth factor(VEGF) signaling in tumor progression. Crit. Rev. Oncol./Hematol. 2007, 62, 179–213. [Google Scholar] [CrossRef]
  73. Saharinen, P.; Eklund, L.; Pulkki, K.; Bono, P.; Alitalo, K. VEGF and angiopoietin signaling in tumor angiogenesis and metastasis. Trends Mol. Med. 2011, 17, 347–362. [Google Scholar] [CrossRef] [PubMed]
  74. Yang, Y.; Cao, Y. The impact of VEGF on cancer metastasis and systemic disease. Semin. Cancer Biol. 2022, 86, 251–261. [Google Scholar] [CrossRef] [PubMed]
  75. Carreau, A.; El Hafny-Rahbi, B.; Matejuk, A.; Grillon, C.; Kieda, C. Why is the partial oxygen pressure of human tissues a crucial parameter? Small molecules and hypoxia. J. Cell Mol. Med. 2011, 15, 1239–1253. [Google Scholar] [CrossRef] [PubMed]
  76. McKeown, S.R. Defined normoxia, physoxia and hypoxia in tumors—Implications for treatment response. Br. J. Radiol. 2014, 87, 20130676. [Google Scholar] [CrossRef] [PubMed]
  77. Vaupel, P.; Mayer, A.; Höckel, M. Impact of hemoglobin levels on tumor oxygenation: The higher, the better? Strahlenther. Onkol. 2006, 182, 63–71. [Google Scholar] [CrossRef] [PubMed]
  78. Kozlov, A.M.; Lone, A.; Betts, D.H.; Cumming, R.C. Lactate preconditioning promotes a HIF-1α-mediated metabolic shift from OXPHOS to glycolysis in normal human diploid fibroblasts. Sci. Rep. 2020, 10, 8388. [Google Scholar] [CrossRef] [PubMed]
  79. Nishimura, K.; Fukuda, A.; Hisatake, K. Mechanisms of the metabolic shift during somatic cell reprogramming. Int. J. Mol. Sci. 2019, 20, 2254. [Google Scholar] [CrossRef] [PubMed]
  80. Weinberg, F.; Hamanaka, R.; Wheaton, W.W.; Weinberg, S.; Joseph, J.; Lopez, M.; Kalyanaraman, B.; Mutlu, G.M.; Budinger, G.R.S.; Chandel, N.S. Mitochondrial metabolism and ROS generation are essential for Kras-mediated tumorigenicity. Proc. Natl. Acad. Sci. USA 2010, 107, 8788–8793. [Google Scholar] [CrossRef]
  81. Fischer, K.; Hoffmann, P.; Voelkl, S.; Meidenbauer, N.; Ammer, J.; Edinger, M.; Gottfried, E.; Schwarz, S.; Rothe, G.; Hoves, S.; et al. Inhibitory effect of tumor cell-derived lactic acid on human T cells. Blood 2007, 109, 3812–3819. [Google Scholar] [CrossRef]
  82. Choi, J.; Gyamfi, J.; Jang, H.; Koo, J.S. The role of tumor-associated macrophage in breast cancer biology. Histol. Histopathol. 2018, 33, 133–145. [Google Scholar] [CrossRef]
  83. Mu, X.; Shi, W.; Xu, Y.; Xu, C.; Zhao, T.; Geng, B.; Yang, J.; Pan, J.; Hu, S.; Zhang, C.; et al. Tumor-derived lactate induces M2 macrophage polarization via the activation of the ERK/STAT3 signaling pathway in breast cancer. Cell Cycle 2018, 17, 428–438. [Google Scholar] [CrossRef] [PubMed]
  84. Ratter, J.M.; Rooljackers, H.M.M.; Hoolveld, G.J.; Hijmans, A.G.M.; de Galan, B.E.; Tack, C.J.; Stienstra, R. In vitro and in vivo effects of lactate on metabolism and cytokine production of human primary PBMCs and monocytes. Front. Immunol. 2018, 9, 2564. [Google Scholar] [CrossRef] [PubMed]
  85. De Saedeleer, C.J.; Copetti, T.; Porporato, P.E.; Verrax, J.; Feron, O.; Sonveaux, P. Lactate activates HIF-1 in oxidative but not in Warburg-phenotype human tumor cells. PLoS ONE 2012, 7, e46571. [Google Scholar] [CrossRef] [PubMed]
  86. Pires, B.R.B.; Mencalha, A.L.; Ferreira, G.M.; Panis, C.; Silva, R.C.M.C.; Abdelhay, E. The hypoxia-inducible factor-1 alpha signaling pathway and its relation to cancer and immunology. Am. J. Immunol. 2014, 10, 215–224. [Google Scholar] [CrossRef]
  87. Davis, L.; Recktenwald, M.; Hutt, E.; Fuller, S.; Briggs, M.; Goel, A.; Daringer, N. Targeting HIF-2α in the tumor microenvironment: Redefining the role of HIF-2α for solid cancer therapy. Cancers 2022, 14, 1259. [Google Scholar] [CrossRef] [PubMed]
  88. Löfstedt, T.; Fredlund, E.; Holmquist-Mengelbier, L.; Pietras, A.; Overnberger, M.; Pollinger, L.; Påhlman, S. Hypoxia inducible factor-2α in cancer. Cell Cycle 2007, 6, 919–926. [Google Scholar] [CrossRef] [PubMed]
  89. Schödel, J.; Oikonomopoulos, S.; Ragoussis, J.; Pugh, C.W.; Ratcliffe, P.J.; Mole, D.R. High-resolution genome-wide mapping of HIF-binding sites by CHIP-seq. Blood 2011, 117, e207. [Google Scholar] [CrossRef] [PubMed]
  90. Stalnecker, C.A.; Cluntun, A.A.; Cerione, R.A. Balancing redox stress: Anchorage-independent growth requires reductive carboxylation. Transl. Cancer Res. 2016, 5, S433–S437. [Google Scholar] [CrossRef]
  91. Jin, J.; Byun, J.-K.; Choi, Y.-K.; Park, K.-G. Targeting glutamine metabolism as a therapeutic strategy for cancer. Exp. Mol. Med. 2023, 55, 706–715. [Google Scholar] [CrossRef]
  92. Végran, F.; Boidot, R.; Michiels, C.; Sonveaux, P.; Feron, O. Lactate influx through the endothelial cell monocarboxylate transporter MCT1 supports on NFκB/IL-8 pathway that drives tumor angiogenesis. Cancer Res. 2011, 71, 2550–2560. [Google Scholar] [CrossRef]
  93. Chamnee, T.; Ontong, P.; Itano, N. Hyaluronan: A modulator of the tumor microenvironment. Cancer Lett. 2016, 375, 20–30. [Google Scholar] [CrossRef] [PubMed]
  94. Lennicke, C.; Rahn, J.; Lichtenfels, R.; Wessjohann, L.A.; Seliger, B. Hydrogen peroxide—Production, fate and role in redox signaling of tumor cells. Cell Commun. Signal. 2015, 13, 39. [Google Scholar] [CrossRef] [PubMed]
  95. Cyran, A.M.; Zhitkovich, A. HIF1, HSF1, and NRF2: Oxidant-responsive trio raising cellular defenses and engaging immune system. Chem. Res. Toxicol. 2022, 35, 1690–1700. [Google Scholar] [CrossRef] [PubMed]
  96. Wang, S.; Song, P.; Zou, M.-H. AMP-activated protein kinase, stress responses and cardiovascular diseases. Clin. Sci. 2012, 122, 555–573. [Google Scholar] [CrossRef] [PubMed]
  97. Gwinn, D.M.; Shackelford, D.B.; Egan, D.F.; Mihaylova, M.M.; Mery, A.; Vasquez, D.S.; Turk, B.E.; Shaw, R.J. AMPK phosphorylation of raptor mediates a metabolic checkpoint. Mol. Cell 2008, 30, 214–226. [Google Scholar] [CrossRef]
  98. Barnard, R.A.; Regan, D.P.; Hansen, R.J.; Maycotte, P.; Thorborn, A.; Gustafson, D.L. Autophagy inhibition delays early but not late-stage metastatic disease. J. Pharmacol. Exp. Ther. 2016, 358, 282–293. [Google Scholar] [CrossRef]
  99. Guo, J.Y.; Xia, B.; White, E. Autophagy-mediated tumor promotion. Cell 2013, 155, 1216–1219. [Google Scholar] [CrossRef]
  100. White, E. Deconvoluting the context-dependent role for autophagy in cancer. Nat. Rev. Cancer 2012, 12, 401–410. [Google Scholar] [CrossRef]
  101. Jiang, X.; Overholtzer, M.; Thompson, C.B. Autophagy in cellular metabolism and cancer. J. Clin. Investig. 2015, 125, 47–54. [Google Scholar] [CrossRef]
  102. Jin, S.; DiPaola, R.S.; Mathew, R.; White, E. Metabolic catastrophe as a means to cancer cell death. J. Cell Sci. 2007, 120, 379–383. [Google Scholar] [CrossRef]
  103. Degenhardt, K.; Mathew, R.; Beaudoin, B.; Bray, K.; Anderson, D.; Chen, G.; Mukherjee, C.; Shi, Y.; Gélinas, C.; Fan, Y.; et al. Autophagy promotes tumor cell survival and restricts necrosis, inflammation, and tumorigenesis. Cancer Cell 2006, 10, 51–64. [Google Scholar] [CrossRef] [PubMed]
  104. Lum, J.J.; Bauer, D.E.; Kong, M.; Harris, M.H.; Li, C.; Lindsten, Z.; Thompson, C.B. Growth factor regulation of autophagy and cell survival in the absence of apoptosis. Cell 2005, 120, 237–248. [Google Scholar] [CrossRef] [PubMed]
  105. Fung, C.; Lock, R.; Gao, S.; Salas, E.; Debnath, J. Induction of autophagy during extracellular matrix detachment promotes cell survival. Mol. Biol. Cell 2008, 19, 797–806. [Google Scholar] [CrossRef] [PubMed]
  106. Macintosh, R.L.; Timpson, P.; Thorborn, J.; Anderson, K.I.; Thorborn, A.; Ryan, K.M. Inhibition of autophagy impairs tumor cell invasion in an organotypic model. Cell Cycle 2012, 11, 2022–2029. [Google Scholar] [CrossRef] [PubMed]
  107. Peng, Y.-F.; Shi, Y.-H.; Ding, Z.-B.; Ke, A.-W.; Gu, C.-Y.; Zhou, J.; Qiu, S.-J.; Dai, Z.; Fan, J. Autophagy inhibition suppresses pulmonary metastasis of HCC in mice via impairing anoikis resistance and colonization of HCC cells. Autophagy 2013, 9, 2056–2068. [Google Scholar] [CrossRef] [PubMed]
  108. Li, X.; He, S.; Ma, B. Autophagy and autophagy-related proteins in cancer. Mol. Cancer 2020, 19, 12. [Google Scholar] [CrossRef] [PubMed]
  109. Moenner, M.; Pluquet, O.; Bouchecareilh, M.; Chevet, E. Integrated endoplasmic reticulum stress responses in cancer. Cancer Res. 2007, 67, 10361–10364. [Google Scholar] [CrossRef] [PubMed]
  110. Fernandez, P.M.; Tabbara, S.O.; Jacobs, L.K.; Manning, F.C.; Tsangaris, T.N.; Schwartz, A.M.; Kennedy, K.A.; Patierno, S.R. Overexpression of the glucose-regulated stress gene GRP78 in malignant but not benign human breast lesions. Breast Cancer Res. Treat. 2000, 59, 15–26. [Google Scholar] [CrossRef]
  111. Xue, Z.; He, Y.; Ye, K.; Gu, Z.; Mao, Y.; Qi, L. A conserved structural determinant located at the interdomain region of mammalian inositol-requiring enzyme 1alpha. J. Biol. Chem. 2011, 286, 30859–30866. [Google Scholar] [CrossRef]
  112. Hong, S.Y.; Hagen, T. Multiple myeloma Leu167Ile (c.499C>A) mutation prevents XBP1 mRNA splicing. Br. J. Haematol. 2013, 161, 898–901. [Google Scholar] [CrossRef]
  113. Ghosh, R.; Wang, L.; Wang, E.S.; Perera, B.G.; Igbaria, A.; Morita, S.; Prado, K.; Thamsen, M.; Caswell, D.; Macias, H.; et al. Allosteric inhibition of the IRE1α RNase preserves cell viability and function during endoplasmic reticulum stress. Cell 2014, 158, 534–548. [Google Scholar] [CrossRef] [PubMed]
  114. Oakes, S.A. Endoplasmic reticulum stress signaling in cancer cells. Am. J. Pathol. 2020, 190, 934–946. [Google Scholar] [CrossRef] [PubMed]
  115. Min, J.-Y.; Chun, K.-S.; Kim, D.-H. The versatile utility of cysteine as a target for cancer treatment. Front. Oncol. 2023, 12, 997919. [Google Scholar] [CrossRef] [PubMed]
  116. Banjac, A.; Perisic, T.; Sato, H.; Seiler, A.; Bannai, S.; Weiss, N.; Kölle, P.; Tschoep, K.; Issels, R.D.; Daniel, P.T.; et al. The cystine/cysteine cycle: A redox cycle regulating susceptibility versus resistance to cell death. Oncogene 2008, 27, 1618–1628. [Google Scholar] [CrossRef] [PubMed]
  117. Lewerenz, J.; Hewett, S.J.; Huang, Y.; Lambros, M.; Gout, P.W.; Kalivas, P.W.; Massie, A.; Smolders, I.; Methner, A.; Pergande, M.; et al. The cystine/glutamate antiporter system xc in health and disease: From molecular mechanisms to novel therapeutic opportunities. Antioxid. Redox Signal. 2013, 18, 522–555. [Google Scholar] [CrossRef] [PubMed]
  118. Lin, W.; Wang, C.; Liu, G.; Bi, C.; Wang, X.; Zhou, Q.; Jin, H. SLC7A11/xCT in cancer: Biological functions and therapeutic implications. Am. J. Cancer Res. 2020, 10, 3106–3126. [Google Scholar]
  119. Sen, N.; Paul, B.D.; Gadalla, M.M.; Mustafa, A.K.; Sen, T.; Xu, R.; Kim, S.; Snyder, S.H. Hydrogen sulfide-linked sulfhydration of NF-κB mediates its antiapoptotic actions. Mol. Cell 2012, 45, 13–24. [Google Scholar] [CrossRef]
  120. Yang, G.; Zhao, K.; Ju, Y.; Mani, S.; Cao, Q.; Puukila, S.; Khaper, N.; Wu, L.; Wang, R. Hydrogen sulfide protects against cellular senescence via S-sulfhydration of Keap1 and activation of Nrf2. Antioxid. Redox Signal. 2013, 18, 1906–1919. [Google Scholar] [CrossRef]
  121. Ma, Z.; Bi, Q.; Wang, Y. Hydrogen sulfide accelerates cell cycle progression in oral squamous cell carcinoma cell lines. Oral Dis. 2015, 21, 156–162. [Google Scholar] [CrossRef]
  122. Jiang, L.; Shestov, A.A.; Swain, P.; Yang, C.; Parker, S.J.; Wang, Q.A.; Terada, L.S.; Adams, N.D.; McCabe, M.T.; Pietrak, B.; et al. Reductive carboxylation supports redox homeostasis during anchorage-independent growth. Nature 2016, 532, 255–258. [Google Scholar] [CrossRef]
  123. Kaya, B.; Çiçek, O.; Erdi, F.; Findig, S.; Karatas, Y.; Esen, H.; Keskin, F.; Kalkan, E. Intratumoral hemorrhage-related differences in the expression of vascular endothelial growth factor, basic fibroblast growth factor and thioredoxin reductase 1 in human glioblastoma. Mol. Clin. Oncol. 2006, 5, 343–346. [Google Scholar] [CrossRef] [PubMed]
  124. Yin, T.; He, S.; Liu, X.; Jiang, W.; Ye, T.; Lin, Z.; Sang, Y.; Su, C.; Wan, Y.; Shen, G.; et al. Extravascular red blood cells and hemoglobin promote tumor growth and therapeutic resistance as endogenous danger signals. J. Immunol. 2015, 194, 429–437. [Google Scholar] [CrossRef] [PubMed]
  125. Poon, L.C.; Methot, S.P.; Morabi-Pazocki, W.; Pio, F.; Bennet, A.J.; Sen, D. Guanine-rich RNAs and DNAs that bind heme robustly catalyze oxygen transfer reactions. J. Am. Chem. Soc. 2011, 133, 1877–1884. [Google Scholar] [CrossRef] [PubMed]
  126. Gray, L.T.; Lombardi, E.P.; Verga, D.; Nicolas, A.; Teulade-Fichou, M.-P.; Londoño-Vallejo, A.; Maizels, N. G-Quadruplexes sequester free heme in living cells. Cell Chem. Biol. 2019, 26, 1681–1689. [Google Scholar] [CrossRef] [PubMed]
  127. Brooks, T.A.; Hurley, L.H. Targeting MYC expression through G-quadruplexes. Genes Cancer 2010, 1, 641–649. [Google Scholar] [CrossRef] [PubMed]
  128. Brooks, T.A.; Kendrick, S.; Hurley, L.H. Making sense of G-quadruplex and i-motif functions in oncogene promoters. FEBS J. 2010, 277, 3459–3469. [Google Scholar] [CrossRef] [PubMed]
  129. Webb, B.A.; Chimenti, M.; Jacobson, M.P.; Barber, D.L. Dysregulated pH: A perfect storm for cancer progression. Nat. Rev. Cancer 2011, 11, 671–677. [Google Scholar] [CrossRef] [PubMed]
  130. Liu, Y.; White, K.A.; Barber, D.L. Intracellular pH regulates cancer and stem cell behaviors: A protein dynamics perspective. Front. Oncol. 2020, 10, 1401. [Google Scholar] [CrossRef]
  131. Hao, G.; Xu, Z.P.; Li, L. Manipulating extracellular tumour pH: An effective target for cancer therapy. RSC Adv. 2018, 8, 22182–22192. [Google Scholar] [CrossRef]
  132. Casey, J.R.; Grinstein, S.; Orlowski, J. Sensors and regulators of intracellular pH. Nat. Rev. Mol. Cell Biol. 2010, 11, 50–61. [Google Scholar] [CrossRef]
  133. Flinck, M.; Kramer, S.H.; Schnipper, J.; Andersen, A.P.; Pedersen, S.F. The acid-base transport proteins NHE1 and NBCn1 regulate cell cycle progression in human breast cancer cells. Cell Cycle 2018, 17, 1056–1067. [Google Scholar] [CrossRef] [PubMed]
  134. Putney, L.K.; Barber, D.L. Na-H exchange-dependent increase in intracellular pH times G2/M entry and transition. J. Biol. Chem. 2003, 278, 44645–44649. [Google Scholar] [CrossRef] [PubMed]
  135. Parks, S.K.; Pouyssegur, J. The Na(+)/HCO3(-) co-transporter SLC4A4 plays a role in growth and migration of colon and breast cancer cells. J. Cell Physiol. 2015, 230, 1954–1963. [Google Scholar] [CrossRef] [PubMed]
  136. Schwab, A.; Stock, C. Ion channels and transporters in tumour cell migration and invasion. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2014, 369, 20130102. [Google Scholar] [CrossRef]
  137. Schulze, A.; Harris, A.L. How cancer metabolism is tuned for proliferation and vulnerable to disruption. Nature 2012, 491, 364–373. [Google Scholar] [CrossRef]
  138. Lee, S.; Shanti, A. Effect of exogenous pH on cell growth of breast cancer cells. Int. J. Mol. Sci. 2021, 22, 9910. [Google Scholar] [CrossRef]
  139. Makris, E.A.; Responte, D.J.; Paschos, N.K.; Hu, J.C.; Athanasiou, K.A. Developing functional musculoskeletal tissues through hypoxia and lysyl oxidase-induced collagen cross-linking. Proc. Natl. Acad. Sci. USA 2014, 111, E4832–E4841. [Google Scholar] [CrossRef]
  140. Makris, E.A.; Hu, J.C.; Athanasiou, K.A. Hypoxia-induced collagen crosslinking as a mechanism for enhancing mechanical properties of engineered articular cartilage. Osteoarthr. Cartil. 2013, 21, 634–641. [Google Scholar] [CrossRef]
  141. Najafi, M.; Farhood, B.; Mortezaee, K. Extracellular matrix (ECM) stiffness and degradation as cancer drivers. J. Cell Biochem. 2019, 120, 2782–2790. [Google Scholar] [CrossRef]
  142. Aro, E.; Khatri, R.; Gerard-O’Riley, R.; Mangiavini, L.; Myllyharju, J.; Schipani, E. Hypoxia-inducible factor-1 (HIF-1) but not HIF-2 is essential for hypoxic induction of collagen prolyl 4-hydroxylases in primary newborn mouse epiphyseal growth plate chondrocytes. J. Biol. Chem. 2012, 287, 37134–37144. [Google Scholar] [CrossRef]
  143. Xiong, G.; Stewart, R.L.; Chen, J.; Gao, T.; Scott, T.L.; Samayoa, L.M.; O’Connor, K.; Lane, A.N.; Xu, R. Collagen prolyl 4-hydroxylase 1 is essential for HIF-1alpha stabilization and TNBC chemoresistance. Nat. Commun. 2018, 9, 4456. [Google Scholar] [CrossRef] [PubMed]
  144. Saatci, O.; Kaymak, A.; Raza, U.; Ersan, P.G.; Akbulut, O.; Banister, C.E.; Sikirzhytski, V.; Tokat, U.M.; Aykut, G.; Ansari, S.A.; et al. Targeting lysyl oxidase (LOX) overcomes chemotherapy resistance in triple negative breast cancer. Nat. Commun. 2020, 11, 2416. [Google Scholar] [CrossRef] [PubMed]
  145. Janjanam, J.; Pano, G.; Wang, R.; Minden-Birkenmaier, B.A.; Breeze-Jones, H.; Baker, E.; Garcin, C.; Clayton, G.; Shirinifard, A.; Zaske, A.M.; et al. Matricellular protein WISP2 is an endogenous inhibitor of collagen linearization and cancer metastasis. Cancer Res. 2021, 81, 5666–5677. [Google Scholar] [CrossRef] [PubMed]
  146. Murdocca, M.; De Masi, C.; Pucci, S.; Mango, R.; Novelli, G.; Di Natale, C.; Sangiuolo, F. LOX-1 and cancer: An indissoluble liaison. Cancer Gene Ther. 2021, 28, 1088–1098. [Google Scholar] [CrossRef] [PubMed]
  147. Bodelon, C.; Mullooly, M.; Pfeiffer, R.M.; Fan, S.; Abubakar, M.; Lenz, P.; Vacek, P.M.; Weaver, D.L.; Herschorn, S.D.; Johnson, J.M.; et al. Mammary collagen architecture and its association with mammographic density and lesion severity among women undergoing image-guided breast biopsy. Breast Cancer Res. 2021, 23, 105. [Google Scholar] [CrossRef] [PubMed]
  148. Chen, D.; Liu, Z.; Liu, W.; Fu, M.; Jiang, W.; Xu, S.; Wang, G.; Chen, F.; Lu, J.; Chen, H.; et al. Predicting postoperative peritoneal metastasis in gastric cancer with serosal invasion using a collagen nomogram. Nat. Commun. 2021, 12, 179. [Google Scholar] [CrossRef]
  149. Angenendt, L.; Mikesch, J.H.; Gorlich, D.; Busch, A.; Arnhold, I.; Rudack, C.; Hartmann, W.; Wardelmann, E.; Berdel, W.E.; Stenner, M.; et al. Stromal collagen type VI associates with features of malignancy and predicts poor prognosis in salivary gland cancer. Cell. Oncol. Dordr. 2018, 41, 517–525. [Google Scholar] [CrossRef]
  150. Salmon, H.; Franciszkiewicz, K.; Damotte, D.; Dieu-Nosjean, M.C.; Validire, P.; Trautmann, A.; Mami-Chouaib, F.; Donnadieu, E. Matrix architecture defines the preferential localization and migration of T cells into the stroma of human lung tumors. J. Clin. Investig. 2012, 122, 899–910. [Google Scholar] [CrossRef]
  151. Piersma, B.; Hayward, M.K.; Weaver, V.M. Fibrosis and cancer: A strained relationship. Biochim. Biophys. Acta Rev. Cancer 2020, 1873, 188356. [Google Scholar] [CrossRef]
  152. Landoldt, L.; Spagnoli, G.C.; Hertig, A.; Brocheriou, I.; Marti, H.-P. Fibrosis and cancer: Shared features and mechanisms suggest common targeted therapeutic approaches. Nephrol. Dial. Transplant. 2022, 37, 1024–1032. [Google Scholar] [CrossRef]
  153. Wu, B.; Sodji, Q.H.; Oyelere, A.K. inflammation, fibrosis and cancer: Mechanisms, therapeutic options and challenges. Cancers 2022, 14, 552. [Google Scholar] [CrossRef]
  154. Luo, L.; Yang, J.-X.; Luo, T.; Liu, D.; Wu, G.-H.; He, J.-M. A study on the mechanism of PP2A in the recovery of SCI in rats through downregulation of MMP-9 via MAPK signaling pathway. Eur. Rev. Med. Pharmacol. Sci. 2021, 25, 7195–7203. [Google Scholar] [CrossRef] [PubMed]
  155. Turk, V.; Stoka, V.; Vasiljeva, O.; Renko, M.; Sun, T.; Turk, B.; Turk, D. Cysteine cathepsins: From structure, function and regulation to new frontiers. Biochim. Biophys. Acta 2012, 1824, 68–88. [Google Scholar] [CrossRef] [PubMed]
  156. Piperigkou, Z.; Kyriakopoulou, K.; Koutsakis, C.; Mastronikolis, S.; Karamanos, N.K. Key Matrix Remodeling Enzymes: Functions and Targeting in Cancer. Cancers 2021, 13, 1441. [Google Scholar] [CrossRef] [PubMed]
  157. Barash, U.; Cohen-Kaplan, V.; Dowek, I.; Sanderson, R.D.; Ilan, N.; Vlodavsky, I. Proteoglycans in health and disease: New concepts for heparanase function in tumor progression and metastasis. FEBS J. 2010, 277, 3890–3903. [Google Scholar] [CrossRef] [PubMed]
  158. Wei, R.-R.; Sun, D.-N.; Yang, H.; Yan, J.; Zhang, X.; Zheng, X.-L.; Fu, X.-H.; Geng, M.-Y.; Huang, X.; Ding, J. CTC clusters induced by heparanase enhance breast cancer metastasis. Acta Pharmacol. Sin. 2018, 39, 1326–1337. [Google Scholar] [CrossRef] [PubMed]
  159. Song, J.M.; Im, J.; Nho, R.S.; Han, Y.H.; Upadhyaya, P.; Kassie, F. Hyaluronan-CD44/RHAMM interaction-dependent cell proliferation and survival in lung cancer cells. Mol. Carcinog. 2019, 58, 321–333. [Google Scholar] [CrossRef]
  160. Makkar, S.; Riehl, T.E.; Chen, B.; Yan, Y.; Alvarado, D.M.; Ciorba, M.A.; Stenson, W.F. Hyaluronic acid binding to TLR4 promotes proliferation and blocks apoptosis in colon cancer. Mol. Cancer Ther. 2019, 18, 2446–2456. [Google Scholar] [CrossRef]
  161. Yang, Y.M.; Noureddin, M.; Liu, C.; Ohashi, K.; Kim, S.Y.; Ramnath, D.; Powell, E.E.; Sweet, M.J.; Roh, Y.S.; Hsin, I.-F.; et al. Hyaluronan synthase 2-mediated hyaluronan production mediates Notch1 activation and liver fibrosis. Sci. Transl. Med. 2019, 11, eaat9284. [Google Scholar] [CrossRef]
  162. Chen, J.E.; Lumibao, J.; Blazek, A.; Gaskins, H.R.; Harley, B. Hypoxia activates enhanced invasive potential and endogenous hyaluronic acid production by glioblastoma cells. Biomater. Sci. 2018, 6, 854–862. [Google Scholar] [CrossRef]
  163. Kobayashi, N.; Miyoshi, S.; Mikami, T.; Koyama, H.; Kitazawa, M.; Takeoka, M.; Sano, K.; Amano, J.; Isogai, Z.; Niida, S.; et al. Hyaluronan deficiency in tumor stroma impairs macrophage trafficking and tumor neovascularization. Cancer Res. 2010, 70, 7073–7083. [Google Scholar] [CrossRef] [PubMed]
  164. Kuang, D.M.; Wu, Y.; Chen, N.; Cheng, J.; Zhuang, S.M.; Zheng, L. Tumor-derived hyaluronan induces formation of immunosuppressive macrophages through transient early activation of monocytes. Blood 2007, 110, 587–595. [Google Scholar] [CrossRef] [PubMed]
  165. Luo, Y.; Ma, J.; Lu, W. The significance of mitochondrial dysfunction in cancer. Int. J. Mol. Sci. 2020, 21, 5598. [Google Scholar] [CrossRef] [PubMed]
  166. Yang, W.S.; Stockwell, B.R. Ferroptosis: Death by lipid peroxidation. Trends Cell Biol. 2016, 26, 165–176. [Google Scholar] [CrossRef] [PubMed]
  167. Xie, Y.; Hou, W.; Song, X.; Yu, Y.; Huang, J.; Sun, X.; Kang, R.; Tang, D. Ferroptosis: Process and function. Cell Death Differ. 2016, 23, 360–379. [Google Scholar] [CrossRef] [PubMed]
  168. Arnhold, J. Role of reactive species in destructions. In Cell and Tissue Destruction. Mechanisms, Protection, Disorders; Academic Press: London, UK; San Diego, CA, USA; Cambridge, MA, USA; Oxford, UK, 2020; pp. 23–54. [Google Scholar] [CrossRef]
  169. Arnhold, J.; Flemmig, J. Human myeloperoxidase in innate and acquired immunity. Arch. Biochem. Biophys. 2010, 500, 92–106. [Google Scholar] [CrossRef] [PubMed]
  170. Klebanoff, S.J. Myeloperoxidase: Friend and foe. J. Leukoc. Biol. 2005, 77, 598–625. [Google Scholar] [CrossRef]
  171. Rothenberg, M.E.; Hogan, S.P. The eosinophil. Annu. Rev. Immunol. 2006, 24, 147–174. [Google Scholar] [CrossRef]
  172. Cadet, J.; Teoule, R. Comparative study of oxidation of nucleic acid components by hydroxyl radicals, singlet oxygen and superoxide anion radicals. Photochem. Photobiol. 1978, 28, 661–667. [Google Scholar] [CrossRef]
  173. Agnez-Lima, L.F.; Melo, J.T.A.; Silva, A.E.; Oliviera, A.H.S.; Timoteo, A.R.S.; Lima-Bessa, K.M.; Martinez, G.R.; Medeiros, M.H.G.; Di Mascio, P.; Galhardo, R.S.; et al. DNA damage by singlet oxygen and cellular protective mechanisms. Mutat. Res. 2012, 751, 15–28. [Google Scholar] [CrossRef]
  174. Brand, M.D. mitochondrial generation of superoxide and hydrogen peroxide as the source of mitochondrial redox signaling. Free Radic. Biol. Med. 2016, 100, 14–31. [Google Scholar] [CrossRef] [PubMed]
  175. Robb, E.L.; Hall, A.R.; Prime, T.A.; Eaton, S.; Szibor, M.; Viscomi, C.; James, A.M.; Murphy, M.P. Control of mitochondrial superoxide production by reverse electron transport at complex I. J. Biol. Chem. 2011, 286, 9869–9879. [Google Scholar] [CrossRef]
  176. Demin, O.V.; Khodolenko, B.N.; Skulachev, V.P. A model of O2•− generation in the complex III of the electron transport chain. Mol. Cell Biochem. 1998, 184, 21–33. [Google Scholar] [CrossRef] [PubMed]
  177. Gardner, P.R. Superoxide-driven aconitase Fe-S cycling. Biosci. Rep. 1997, 17, 33–42. [Google Scholar] [CrossRef]
  178. Gardner, P.R. Aconitase: Sensitive target and measure of superoxide. Meth. Enzymol. 2002, 349, 9–23. [Google Scholar] [CrossRef]
  179. Tengan, C.H.; Moraes, C.T. NO control of mitochondrial function in normal and transformed cells. Biochim. Biophys. Acta 2017, 1858, 573–581. [Google Scholar] [CrossRef]
  180. Tengan, C.H.; Rodrigues, G.S.; Godinho, R.O. Nitric oxide in skeletal muscle: Role on mitochondrial biogenesis and function. Int. J. Mol. Sci. 2012, 13, 17160–17184. [Google Scholar] [CrossRef]
  181. Figueira, T.R.; Barros, M.H.; Camargo, A.A.; Castilho, R.F.; Ferreira, J.C.; Kowaltowski, A.J.; Sluse, F.E.; Souza-Pinto, N.C.; Vercesi, A.E. Mitochondria as a source of reactive oxygen and nitrogen species: From molecular mechanisms to human health. Antioxid. Redox Signal. 2013, 18, 2029–2074. [Google Scholar] [CrossRef]
  182. Aaltoma, S.H.; Lipponen, P.K.; Kosma, V.M. Inducible nitric oxide synthase (iNOS) expression and its prognostic value in prostate cancer. Anticancer Res. 2001, 21, 3101–3106. [Google Scholar]
  183. Jabłońska, E.; Puzewska, W.; Marcińczyk, M.; Grabowska, Z.; Jabłoński, J. iNOS expression and NO production by neutrophils in cancer patients. Arch. Immunol. Ther. Exp. 2005, 53, 175–179. [Google Scholar]
  184. Barani, R.; Motalleb, G.; Maghsoudi, H. Evaluation of iNOS expression in esophageal eancer eatients. Gastrointest. Tumors 2016, 3, 44–58. [Google Scholar] [CrossRef] [PubMed]
  185. Huie, R.E.; Padmaja, S. Reactions of NO and O2•−. Free Radic. Res. Commun. 1993, 18, 195–199. [Google Scholar] [CrossRef] [PubMed]
  186. Kissner, R.; Nauser, T.; Bugnon, P.; Lye, P.G.; Koppenol, W.H. Formation and properties of peroxynitrite as studied by laser flash photolysis, high-pressure stopped-flow technique, and pulse radiolysis. Chem. Res. Toxicol. 1997, 10, 1285–1292. [Google Scholar] [CrossRef] [PubMed]
  187. Wenzel, U.; Kuntz, S.; De Sousa, U.J.; Daniel, H. Nitric oxide suppresses apoptosis in human colon cancer cells by scavenging mitochondrial superoxide anions. Int. J. Cancer 2003, 106, 666–675. [Google Scholar] [CrossRef] [PubMed]
  188. Wang, Q.; Ye, S.; Chen, X.; Xu, P.; Li, K.; Zeng, S.; Huang, M.; Gao, W.; Chen, J.; Zhang, Q.; et al. Mitochondrial NOS1 suppresses apoptosis in colon cancer cells through increasing SIRT3 activity. Biochem. Biophys. Res. Commun. 2019, 515, 517–523. [Google Scholar] [CrossRef] [PubMed]
  189. Radi, R.; Beckman, J.S.; Bush, K.M.; Freeman, B.A. Peroxynitrite-induced membrane lipid peroxidation: The cytotoxic potential of superoxide and nitric oxide. Arch. Biochem. Biophys. 1991, 288, 481–487. [Google Scholar] [CrossRef] [PubMed]
  190. Goldstein, S.; Czapski, G. Mechanism of the nitrosation of thiols and amines by oxygenated NO solutions: The nature of the nitrosating intermediates. J. Am. Chem. Soc. 1996, 118, 3419–3425. [Google Scholar] [CrossRef]
  191. Schopfer, F.J.; Baker, P.R.S.; Freeman, B.A. NO-dependent protein nitration: A cell signaling event or an oxidative inflammatory response? Trends Biochem. Sci. 2003, 28, 646–654. [Google Scholar] [CrossRef]
  192. Ferrer-Sueta, G.; Radi, R. Chemical biology of peroxynitrite: Kinetics, diffusion, and radicals. ACS Chem. Biol. 2009, 4, 161–177. [Google Scholar] [CrossRef]
  193. Furtmüller, P.G.; Jantschko, W.; Zederbauer, M.; Schwanninger, M.; Jakoptisch, C.; Herold, S.; Koppenol, W.H.; Obinger, C. Peroxynitrite efficiently mediates the interconversion of redox intermediates of myeloperoxidase. Biochem. Biophys. Res. Commun. 2005, 337, 944–954. [Google Scholar] [CrossRef]
  194. Koyani, C.N.; Flemmig, J.; Malle, E.; Arnhold, J. Myeloperoxidase scavenges peroxynitrite: A novel anti-inflammatory action of the heme enzyme. Arch. Biochem. Biophys. 2015, 571, 1–9. [Google Scholar] [CrossRef] [PubMed]
  195. Su, J.; Groves, J.T. Mechanisms of peroxynitrite interactions with heme proteins. Inorg. Chem. 2010, 49, 6317–6329. [Google Scholar] [CrossRef] [PubMed]
  196. Gebicka, L.; Gebicki, J.L. Reaction of heme peroxidases with peroxynitrite. IUBMB Life 2000, 49, 11–15. [Google Scholar] [CrossRef] [PubMed]
  197. Halliwell, B.; Gutteridge, J.M.C. Iron as a biological pro-oxidant. ISI Atlas Sci. Biochem. 1988, 1, 48–52. [Google Scholar]
  198. Koppenol, W.H. The centennial of the Fenton reaction. Free Radic. Biol. Chem. 1993, 15, 645–651. [Google Scholar] [CrossRef]
  199. Gunther, M.R.; Hanna, P.M.; Mason, R.P.; Cohen, M.S. Hydroxyl radical formation from cuprous ion and hydrogen peroxide: A spin-trapping study. Arch. Biochem. Biophys. 1995, 316, 515–522. [Google Scholar] [CrossRef]
  200. Bors, W.; Erben-Russ, M.; Saran, M. Fatty acid peroxyl radicals: Their generation and reactivities. Bioelectrochem. Bioenergy 1987, 18, 37–49. [Google Scholar] [CrossRef]
  201. Zhao, N.; Zhang, A.-S.; Enns, C.A. Iron regulation by hepcidin. J. Clin. Investig. 2013, 123, 2337–2343. [Google Scholar] [CrossRef]
  202. Gkouvatsos, K.; Papanikolaou, G.; Pantopoulos, K. Regulation of iron transport and the role of transferrin. Biochim. Biophys. Acta 2011, 1820, 188–202. [Google Scholar] [CrossRef]
  203. Gamella, E.; Buratti, P.; Cairo, G.; Recalcati, S. The transferrin receptor: The cellular iron gate. Metallomics 2017, 9, 1367–1375. [Google Scholar] [CrossRef]
  204. Massover, W.H. Ultrastructure of ferritin and apoferritin: A review. Micron 1993, 24, 389–437. [Google Scholar] [CrossRef]
  205. Prohaska, J.R. Role of copper transporters in copper homeostasis. Am. J. Clin. Nutr. 2008, 88, 826S–829S. [Google Scholar] [CrossRef] [PubMed]
  206. Stoj, C.; Kosman, D.J. Cuprous oxidase activity of yeast Fet3p and human ceruloplasmin: Implication for function. FEBS Lett. 2003, 554, 422–426. [Google Scholar] [CrossRef] [PubMed]
  207. Sokolov, A.V.; Pulina, M.O.; Ageeva, K.V.; Runova, O.I.; Zakharova, E.T.; Vasilyev, V.B. Identification of leukocyte cationic proteins that interact with ceruloplasmin. Biochemistry 2007, 72, 872–877. [Google Scholar] [CrossRef] [PubMed]
  208. McCord, J.; Fridovich, I. Superoxide dismutase: An enzymic function for erythrocuprein (hemocuprein). J. Biol. Chem. 1960, 224, 6049–6055. [Google Scholar]
  209. Chang, L.Y.; Slot, J.W.; Geuze, H.J.; Crapo, J.D. Molecular immunocytochemistry of the CuZn superoxide dismutase in rat hepatocytes. J. Cell Biol. 1988, 107, 2169–2179. [Google Scholar] [CrossRef]
  210. Weisiger, R.A.; Fridovich, I. Mitochondrial superoxide dismutase. Site of synthesis and intramolecular localization. J. Biol. Chem. 1973, 248, 4793–4796. [Google Scholar] [CrossRef]
  211. Antonyuk, S.V.; Strange, R.W.; Marklund, S.L.; Hasnain, S.S. The structure of human extracellular copper-zinc superoxide dismutase at 1.7 Å resolution: Insights into heparin and collagen binding. J. Mol. Biol. 2009, 388, 310–326. [Google Scholar] [CrossRef]
  212. Starkov, A.A. The role of mitochondria in reactive oxygen species metabolism and signaling. Ann. N. Y. Acad. Sci. 2008, 1147, 37–52. [Google Scholar] [CrossRef]
  213. Pasdois, P.; Parker, J.E.; Griffiths, E.J.; Halestrap, A.P. The role of oxidized cytochrome c in regulating mitochondrial reactive species production and its perturbation in ischemia. Biochem. J. 2011, 436, 493–505. [Google Scholar] [CrossRef]
  214. Ursini, F.; Maiorino, M.; Roveri, A. Phospholipid hydroperoxide glutathione peroxidase (PHGPx): More than an antioxidant enzyme? Biomed. Environm. Sci. 1997, 10, 327–332. [Google Scholar]
  215. Lubos, E.; Loscalzo, J.; Handy, D.E. Glutathione peroxidase-1 in health and disease: From molecular mechanisms to therapeutic opportunities. Antioxid. Redox Signal. 2011, 15, 1957–1997. [Google Scholar] [CrossRef] [PubMed]
  216. Low, F.M.; Hampton, M.P.; Winterbourn, C.C. Prx2 and peroxide metabolism in the erythrocyte. Antioxid. Redox Signal. 2008, 10, 1621–1630. [Google Scholar] [CrossRef] [PubMed]
  217. Goyal, M.M.; Basak, A. Human catalase: Looking for complete identity. Protect. Cell 2010, 1, 888–897. [Google Scholar] [CrossRef] [PubMed]
  218. Chandel, N.S.; McClintock, D.S.; Feliciano, C.E.; Wood, T.M.; Melendez, J.A.; Rodriguez, A.M.; Schumacker, P.T. Reactive oxygen species generated at mitochondrial complex III stabilize hypoxia-inducible factor-1alpha during hypoxia: A mechanism of O2 sensing. J. Biol. Chem. 2000, 275, 25130–25138. [Google Scholar] [CrossRef] [PubMed]
  219. Sabharwal, S.S.; Waypa, G.B.; Marks, J.D.; Schumacker, P.T. Peroxiredoxin-5 targeted to the mitochondrial intermembrane space attenuates hypoxia-induced reactive oxygen species signalling. Biochem. J. 2013, 456, 337–346. [Google Scholar] [CrossRef] [PubMed]
  220. Guzy, R.D.; Hoyos, B.; Robin, E.; Chen, H.; Liu, L.; Mansfield, K.D.; Simon, M.C.; Hammerling, U.; Schumacker, P.T. Mitochondrial complex III is required for hypoxia-induced ROS production and cellular oxygen sensing. Cell Metab. 2005, 1, 401–408. [Google Scholar] [CrossRef] [PubMed]
  221. Szatrowski, T.P.; Nathan, C.F. Production of large amounts of hydrogen peroxide by human tumor cells. Cancer Res. 1991, 51, 794–798. [Google Scholar]
  222. Kamiguti, A.S.; Serrander, L.; Lin, K.; Harris, R.J.; Cawley, J.C.; Allsup, D.J.; Slupsky, J.R.; Krause, K.H.; Zuzel, M. Expression and activity of NOX5 in the circulating malignant B cells of hairy cell leukemia. J. Immunol. 2005, 175, 8424–8430. [Google Scholar] [CrossRef]
  223. Tsao, S.M.; Yin, M.C.; Liu, W.H. Oxidant stress and B vitamins status in patients with non-small cell lung cancer. Nutr. Cancer 2007, 59, 8–13. [Google Scholar] [CrossRef]
  224. Khandrika, L.; Kumar, B.; Koul, S.; Maroni, P.; Koul, H.K. Oxidative stress in prostate cancer. Cancer Lett. 2009, 282, 125–136. [Google Scholar] [CrossRef] [PubMed]
  225. Patel, B.P.; Rawal, U.M.; Dave, T.K.; Rawal, R.M.; Shukla, S.N.; Shah, P.M.; Patel, P.S. Lipid peroxidation, total antioxidant status, and total thiol levels predict overall survival in patients with oral squamous cell carcinoma. Integr. Cancer Ther. 2007, 6, 365–372. [Google Scholar] [CrossRef] [PubMed]
  226. Nicolussi, A.; D’Inezo, S.; Capalbo, C.; Giannini, G.; Coppa, A. the role of peroxiredoxins in cancer. Mol. Clin. Oncol. 2017, 6, 139–153. [Google Scholar] [CrossRef] [PubMed]
  227. Park, M.H.; Jo, M.; Kim, Y.R.; Lee, C.K.; Hong, J.T. Roles of peroxiredoxins in cancer, neurodegenerative diseases and inflammatory diseases. Pharmacol. Ther. 2016, 163, 1–23. [Google Scholar] [CrossRef] [PubMed]
  228. Taguchi, K.; Motohashi, H.; Yamamoto, M. Molecular mechanisms of the Keap1-Nrf2 pathway in stress response and cancer evolution. Genes Cells 2011, 16, 123–140. [Google Scholar] [CrossRef] [PubMed]
  229. Jaramillo, M.C.; Zhang, D.D. The emerging role of the Nrf2-Keap1 signaling pathway in cancer. Genes Dev. 2013, 27, 2179–2191. [Google Scholar] [CrossRef]
  230. Mitsuishi, Y.; Taguchi, K.; Kawatani, Y.; Shibata, T.; Nukiwa, T.; Aburatani, H.; Yamamoto, M.; Motohashi, H. Nrf2 redirects glucose and glutamine into anabolic pathways in metabolic reprogramming. Cancer Cell 2012, 22, 66–79. [Google Scholar] [CrossRef]
  231. Jin, L.; Zhou, Y. Crucial role of the pentose phosphate pathway in malignant tumors. Oncol. Lett. 2019, 17, 4213–4221. [Google Scholar] [CrossRef]
  232. Menegon, S.; Columbano, A.; Giordano, S. The dual role of NRF2 in cancer. Trends Mol. Med. 2016, 22, 578–593. [Google Scholar] [CrossRef]
  233. Schmidlin, C.J.; Shakya, A.; Dodson, M.; Chapman, E.; Zhang, D.D. The intricacies of NFR2 regulation in cancer. Semin. Cancer Biol. 2021, 76, 110–119. [Google Scholar] [CrossRef]
  234. Singh, A.; Misra, V.; Thimmulappa, R.K.; Lee, H.; Ames, S.; Hoque, M.O.; Herman, J.G.; Baylin, S.B.; Sidransky, D.; Gabrielson, E.; et al. Dysfunctional KEAP1-NRF2 interaction in non-small-cell lung cancer. PLoS Med. 2006, 3, e420. [Google Scholar] [CrossRef] [PubMed]
  235. Shibata, T.; Kokubu, A.; Gotoh, M.; Ojima, H.; Ohta, T.; Yamamoto, M.; Hirohashi, S. Genetic alteration of Keap1 confers constitutive Nrf2 activation and resistance to chemotherapy in gallbladder cancer. Gastroenterology 2008, 135, 1358–1368.e4. [Google Scholar] [CrossRef] [PubMed]
  236. Solis, L.M.; Behrens, C.; Dong, W.; Suraokar, M.; Ozburn, N.C.; Moran, C.A.; Corvalan, A.H.; Biswal, S.; Swisher, S.G.; Bekele, B.N.; et al. Nrf2 and Keap1 abnormalities in non-small cell lung carcinoma and association with clinicopathologic features. Clin. Cancer Res. 2010, 16, 3743–3753. [Google Scholar] [CrossRef] [PubMed]
  237. Kansanen, E.; Kuosmanen, S.M.; Leinonen, H.; Levonen, A.L. The Keap1-Nrf2 pathway: Mechanisms of activation and dysregulation in cancer. Redox Biol. 2013, 1, 45–49. [Google Scholar] [CrossRef] [PubMed]
  238. Schafer, F.Q.; Buettner, G.R. Redox environment of the cell as viewed through the redox state of the glutathione disulfide/glutathione couple. Free Radic. Biol. Med. 2001, 30, 1191–1212. [Google Scholar] [CrossRef] [PubMed]
  239. Mullarky, E.; Cantley, L.C. Diverting glycolysis to combat oxidative stress. In Innovative Medicine. Basic Research and Development; Nakao, K., Minato, N., Uemoto, S., Eds.; Springer Open: Tokyo, Japan, 2015; pp. 3–24. [Google Scholar] [CrossRef]
  240. Arnhold, J. Oxidation and reduction of biological material. In Cell and Tissue Destruction. Mechanisms, Protection, Disorders; Academic Press: London, UK; San Diego, CA, USA; Cambridge, MA, USA; Oxford, UK, 2020; pp. 55–97. [Google Scholar] [CrossRef]
  241. Meister, A. Glutathione metabolism and its selective modification. J. Biol. Chem. 1988, 263, 17205–17208. [Google Scholar] [CrossRef] [PubMed]
  242. Arnér, E.S.; Holmgren, A. The thioredoxin system in cancer. Semin. Cancer Biol. 2006, 16, 420–426. [Google Scholar] [CrossRef]
  243. Zhao, Y.; Wang, H.; Zhou, J.; Shao, Q. Glutathione peroxidase GPX1 and its dichotomous roles in cancer. Cancers 2022, 14, 2560. [Google Scholar] [CrossRef]
  244. De Luca, A.; Sanna, F.; Sallese, M.; Ruggiero, C.; Grossi, M.; Sacchetta, P.; Rossi, C.; de Laurenzi, V.; de Ilio, C.; Favaloro, B. Methionine sulfoxide reductase A down-regulation in human breast cancer cells results in a more aggressive phenotype. Proc. Natl. Acad. Sci. USA 2010, 107, 18628–18633. [Google Scholar] [CrossRef]
  245. Kwak, G.-H.; Kim, T.-H.; Kim, H.-Y. Down-regulation of MsrB3 induces cancer cell apoptosis through reactive oxygen species production and intrinsic mitochondrial pathway activation. Biochem. Biophys. Res. Commun. 2017, 483, 468–474. [Google Scholar] [CrossRef]
  246. Jiang, B.; Moskovitz, J. The functions of the mammalian sulfoxide reductase system and related diseases. Antioxidants 2018, 7, 122. [Google Scholar] [CrossRef] [PubMed]
  247. He, D.; Feng, H.; Sundberg, B.; Yang, J.; Powers, J.; Christian, A.H.; Wilkinson, J.E.; Monnin, C.; Avizonis, D.; Thomas, C.J.; et al. Methionine oxidation activates pyruvate kinase M2 to promote pancreatic cancer metastasis. Mol. Cell 2022, 82, 3045–3060. [Google Scholar] [CrossRef]
  248. Glasauer, A.; Sena, L.A.; Diebold, L.P.; Mazar, A.P.; Chandel, N.S. Targeting SOD1 reduces experimental non-small-cell lung cancer. J. Clin. Investig. 2014, 124, 117–128. [Google Scholar] [CrossRef]
  249. Papa, L.; Hahn, M.; Marsh, E.L.; Evans, B.S.; Germain, D. SOD2 to SOD1 switch in breast cancer. J. Biol. Chem. 2014, 289, 5412–5416. [Google Scholar] [CrossRef] [PubMed]
  250. Dhar, S.K.; St. Clair, D.K. Manganese superoxide dismutase regulation and cancer. Free Radic. Biol. Med. 2012, 52, 2209–2222. [Google Scholar] [CrossRef] [PubMed]
  251. Che, M.; Wang, R.; Wang, H.-Y.; Zheng, X.F.S. Expanding roles of superoxide dismutases in cell regulation and cancer. Drug Discov. Today 2016, 21, 143–149. [Google Scholar] [CrossRef]
  252. Kinnula, V.L.; Crapo, J.D. Superoxide dismutases in malignant cells and human tumors. Free Radic. Biol. Med. 2004, 36, 718–744. [Google Scholar] [CrossRef]
  253. Flor, S.; Oliva, C.R.; Ali, M.Y.; Coleman, K.L.; Greenlee, J.D.; Jones, K.A.; Monga, V.; Griguer, C.E. Catalase overexpression drives an aggressive phenotype in glioblastoma. Antioxidants 2021, 10, 1988. [Google Scholar] [CrossRef]
  254. Forshaw, T.E.; Holmila, R.; Nelson, K.J.; Lewis, J.E.; Kemp, M.L.; Tsang, A.W.; Poole, L.B.; Lowther, W.T.; Furdui, C.M. Peroxiredoxins in cancer and response to radiation therapies. Antioxidants 2019, 8, 11. [Google Scholar] [CrossRef]
  255. Jia, J.-J.; Geng, W.-S.; Wang, Z.-Q.; Chen, L.; Zeng, X.-S. The role of thioredoxin system in cancer: Strategy for cancer therapy. Cancer Chemother. Pharmacol. 2019, 84, 453–470. [Google Scholar] [CrossRef]
  256. Ghareeb, H.; Metanis, N. The thioredoxin system: A promising target for cancer drug development. Chem. Eur. J. 2020, 26, 10175–10184. [Google Scholar] [CrossRef] [PubMed]
  257. Gamcsik, M.P.; Kasibhatla, M.S.; Teeter, S.D.; Colvin, O.M. Glutathione levels in human tumors. Biomarkers 2012, 17, 671–691. [Google Scholar] [CrossRef] [PubMed]
  258. Kennedy, L.; Sandhu, J.K.; Harper, M.-E.; Cuperlovic-Culf, M. Role of glutathione in cancer: From mechanisms to therapies. Biomolecules 2020, 10, 1429. [Google Scholar] [CrossRef] [PubMed]
  259. Valenti, G.E.; Tasso, B.; Traverso, N.; Domenicotti, C.; Marengo, B. Glutathione in cancer progression and chemoresistance: An update. Redox Exp. Med. 2023, 2023, e220023. [Google Scholar] [CrossRef]
  260. Zhu, Z.; Du, S.; Du, Y.; Ren, J.; Ying, G.; Yan, Z. Glutathione reductase mediates drug resistance in glioblastoma cells by regulating redox homeostasis. J. Neurochem. 2018, 144, 93–104. [Google Scholar] [CrossRef] [PubMed]
  261. Sekhar, K.R.; Hanna, D.N.; Cyr, S.; Baechle, J.J.; Kuravi, S.; Balusu, R.; Rathmell, K.; Baregamian, N. Glutathione peroxidase 4 inhibition induces ferroptosis and mTOR pathway suppression in thyroid cancer. Sci. Rep. 2022, 12, 19396. [Google Scholar] [CrossRef] [PubMed]
  262. Shi, Z.-Z.; Tao, H.; Fan, Z.-W.; Song, S.-J.; Bal, J. Prognostic and immunological role of key genes of ferroptosis in pan-cancer. Front. Cell Dev. Biol. 2021, 9, 748925. [Google Scholar] [CrossRef] [PubMed]
  263. Ahmed, K.M.; Veeramachaneni, R.; Deng, D.; Putluri, N.; Putluri, V.; Cardenas, M.F.; Wheeler, D.A.; Decker, W.K.; Frederick, A.I.; Kazi, S.; et al. Glutathione peroxidase 2 is a metabolic driver of the tumor immune microenvironment and immune checkpoint inhibitor response. J. Immunother. Cancer 2022, 10, e004752. [Google Scholar] [CrossRef]
  264. Wang, M.; Chen, X.; Fu, G.; Ge, M. Glutathione peroxidase 2 overexpression promotes malignant progression and cisplatin resistance of KRAS-mutated lung cancer cells. Oncol. Rep. 2022, 48, 207. [Google Scholar] [CrossRef]
  265. Mollbrink, A.; Jawad, R.; Vlamis-Gardikas, A.; Edenvik, P.; Isaksson, B.; Danielsson, O.; Stål, F.A.P. Expression of thioredoxins and glutaredoxins in human hepatocellular carcinoma: Correlation to cell proliferation, tumor size and metabolic syndrome. Int. J. Immunopathol. Pharmacol. 2014, 27, 169–183. [Google Scholar] [CrossRef]
  266. Kim, H.; Lee, G.-R.; Kim, J.; Baek, J.Y.; Jo, Y.-J.; Hong, S.-E.; Kim, S.H.; Lee, J.; Lee, H.I.; Park, S.-K.; et al. Sulfiredoxin inhibitor induces preferential death of cancer cells through reactive species-mediated mitochondrial damage. Free Radic. Biol. Med. 2016, 91, 264–274. [Google Scholar] [CrossRef] [PubMed]
  267. Aye, Y.; Li, M.; Long, M.J.C.; Weiss, R.S. Ribonucleotide reductase and cancer: Biological mechanisms and targeted therapies. Oncogene 2015, 34, 2011–2021. [Google Scholar] [CrossRef] [PubMed]
  268. Zhan, Y.; Jiang, L.; Jin, X.; Ying, S.; Wu, Z.; Wang, L.; Yu, W.; Tong, J.; Zhang, L.; Lou, Y.; et al. Inhibiting RRM2 to enhance the anticancer activity of chemotherapy. Biomed. Pharmacother. 2021, 133, 110996. [Google Scholar] [CrossRef] [PubMed]
  269. Song, J.; Sun, H.; Zhang, S.; Shan, C. The multiple roles of glucose-6-phosphate dehydrogenase in tumorigenesis and cancer chemoresistance. Life 2022, 12, 271. [Google Scholar] [CrossRef] [PubMed]
  270. Li, R.; Wang, W.; Yang, Y.; Gu, C. Exploring the role of glucose-6-phospahte dehydrogenase in cancer. Oncol. Rep. 2020, 44, 2325–2336. [Google Scholar] [CrossRef] [PubMed]
  271. Sarfraz, I.; Rasul, A.; Hussain, G.; Shah, M.A.; Zahoor, A.F.; Asrar, M.; Selamoglu, Z.; Ji, X.-Y.; Adem, S.; Sarker, S.D. 6-Phosphogluconate dehydrogenase fuels multiple aspects of cancer cells: From cancer initiation to metastasis and chemoresistance. BioFactors 2020, 46, 550–562. [Google Scholar] [CrossRef] [PubMed]
  272. Chen, C.; Du, P.; Zhang, Z.; Bao, D. 6-Phosphoglucoronate dehydrogenase inhibition arrests growth and induces apoptosis in gastric cancer via AMPK activation and oxidative stress. Open Life Sci. 2023, 18, 20220514. [Google Scholar] [CrossRef] [PubMed]
  273. Chrichton, R.R.; Ward, R.J. Iron homeostasis. Met. Ions Biol. Syst. 1998, 35, 633–665. [Google Scholar]
  274. Ponka, P. Cellular iron metabolism. Kidney Int. 1999, 55, S2–S11. [Google Scholar] [CrossRef]
  275. Adachi, M.; Kai, K.; Yamaji, K.; Ide, T.; Noshiro, H.; Kawaguchi, A.; Aishima, S. Transferrin receptor 1 overexpression is associated with tumour de-differentiation and acts as a potential prognostic indicator of hepatocellular carcinoma. Histopathology 2019, 75, 63–73. [Google Scholar] [CrossRef]
  276. Brown, R.A.; Richardson, K.L.; Kabir, T.D.; Trinder, D.; Ganss, R.; Leedman, P.J. Altered iron metabolism and impact in cancer biology, metastasis, and immunology. Front. Oncol. 2020, 10, 476. [Google Scholar] [CrossRef] [PubMed]
  277. Xue, D.; Zhou, C.X.; Shi, Y.B.; Lu, H.; He, X.Z. Decreased expression of ferroportin in prostate cancer. Oncol. Lett. 2015, 10, 913–916. [Google Scholar] [CrossRef] [PubMed]
  278. Pinnix, Z.K.; Miller, L.D.; Wang, W.; D’Agostino, R.; Kute, T.; Willingham, M.C.; Hatcher, H.; Tesfay, L.; Sui, G.; Di, X.; et al. Ferroportin and iron regulation in breast cancer progression and prognosis. Sci. Transl. Med. 2010, 2, 43ra56. [Google Scholar] [CrossRef] [PubMed]
  279. Toshiyama, R.; Konno, M.; Eguchi, H.; Asai, A.; Noda, T.; Koseki, J.; Asukai, K.; Ohashi, T.; Matsushita, K.; Iwagami, Y.; et al. Association of iron metabolic enzyme hepcidin expression levels with the prognosis of patients with pancreatic cancer. Oncol. Lett. 2018, 15, 8125–8133. [Google Scholar] [CrossRef] [PubMed]
  280. Shen, Y.; Li, X.; Dong, D.; Zhang, B.; Xue, Y.; Shang, P. Transferrin receptor 1 in cancer: A new sight for cancer therapy. Am. J. Cancer Res. 2018, 8, 916–931. [Google Scholar] [PubMed]
  281. Forciniti, S.; Greco, L.; Grizzi, F.; Malesci, A.; Laghi, L. Iron metabolism in cancer progression. Int. J. Mol. Sci. 2020, 21, 2257. [Google Scholar] [CrossRef] [PubMed]
  282. Wang, H.; Cheng, Q.; Bao, L.; Li, M.; Chang, K.; Yi, X. Cytoprotective role of heme oxygenase-1 in cancer chemoresistance: Focus on antioxidant, antiapoptotic, and pro-autophagy properties. Antioxidants 2023, 12, 1217. [Google Scholar] [CrossRef]
  283. Reiter, C.D.; Wang, X.; Tanus-Santos, J.E.; Hogg, N.; Cannon, R.O., III; Schechter, A.N.; Gladwin, M.T. Cell-free hemoglobin limits nitric oxide bioavailability in sickle-cell disease. Nat. Med. 2002, 8, 1383–1389. [Google Scholar] [CrossRef]
  284. Rother, R.P.; Bell, L.; Hillman, P.; Gladwin, M.T. The clinical sequelae of intravascular hemolysis and extracellular plasma hemoglobin. J. Am. Med. Assoc. 2005, 293, 1653–1662. [Google Scholar] [CrossRef]
  285. Kristiansen, M.; Graversen, J.H.; Jacobsen, C.; Sonne, O.; Hoffmann, H.J.; Law, S.K.; Moestrup, S.K. Identification of the haemoglobin scavenger receptor. Nature 2001, 409, 198–201. [Google Scholar] [CrossRef]
  286. Chiabrando, D.; Vinchi, F.; Fiorito, V.; Tolosano, E. Haptoglobin and hemopexin in heme detoxification and iron recycling. In Acute Phase Proteins-Regulation and Functions of Acute Phase Proteins; Veas, F., Ed.; Intech: Rijeka, Croatia, 2011; pp. 261–288. [Google Scholar] [CrossRef]
  287. Sauret, J.M.; Marinides, G.; Wang, G.K. Rhabdomyolysis. Am. Fam. Phys. 2002, 65, 907–912. [Google Scholar]
  288. Hunter, J.D.; Gregg, K.; Damani, Z. Rhabdomyolysis. Contin. Educ. Anaesth. Crit. Care Pain 2006, 6, 141–143. [Google Scholar] [CrossRef]
  289. Bunn, H.F.; Jandl, J.H. Exchange of heme among hemoglobins and hemoglobin and albumin. J. Biol. Chem. 1968, 243, 465–475. [Google Scholar] [CrossRef] [PubMed]
  290. Hvidberg, V.; Maniecki, M.B.; Jacobson, C.; Højrup, P.; Møller, H.J.; Moestrup, S.K. Identification of the receptor scavenging hemopexin-heme complexes. Blood 2005, 106, 2572–2579. [Google Scholar] [CrossRef] [PubMed]
  291. Figueiredo, R.T.; Fernandez, P.L.; Mourao-Sa, D.S.; Porto, B.N.; Dutra, F.F.; Alves, L.S.; Oliviera, M.F.; Oliviera, P.L.; Graca-Souza, A.V.; Bozza, M.T. Characterization of heme as activator of toll-like receptor 4. J. Biol. Chem. 2007, 282, 20221–20229. [Google Scholar] [CrossRef] [PubMed]
  292. Belcher, J.D.; Chen, C.; Nguyen, J.; Milbauer, L.; Abdulla, F.; Alayash, A.I.; Smith, A.; Nath, K.A.; Hebbel, R.P.; Vercelotti, G.M. Heme triggers TLR4 signaling leading to endothelial cell activation and vaso-occlusion in murine sickle cell disease. Blood 2014, 123, 377–390. [Google Scholar] [CrossRef] [PubMed]
  293. Balla, G.; Jacob, H.S.; Eaton, J.W.; Belcher, J.D.; Vercelotti, G.M. Hemin: A possible physiological mediator of low density lipoprotein oxidation and endothelial injury. Arterioscler. Thromb. 1991, 11, 1700–1711. [Google Scholar] [CrossRef] [PubMed]
  294. Jeney, V.; Balla, J.; Yachie, A.; Varga, Z.; Vercelotti, G.M.; Eaton, J.W.; Balla, G. Pro-oxidant and cytotoxic effects of circulating heme. Blood 2002, 100, 879–887. [Google Scholar] [CrossRef]
  295. Kumar, S.; Bandyopadhyay, U. Free heme toxicity and its detoxification systems in human. Toxicol. Lett. 2005, 157, 175–188. [Google Scholar] [CrossRef]
  296. Flemmig, J.; Schlorke, D.; Kühne, F.-W.; Arnhold, J. Inhibition of the heme-induced hemolysis of red blood cells by the chlorite-based drug WF10. Free Radic. Res. 2016, 50, 1386–1395. [Google Scholar] [CrossRef]
  297. Lin, T.; Sammy, F.; Yang, H.; Thundivalappil, S.; Hellman, J.; Tracey, K.C.; Warren, H.S. Identification of hemopexin as an anti-inflammatory factor that inhibits synergy of hemoglobin with HMGB1 in sterile and infectious inflammation. J. Immunol. 2012, 189, 2017–2022. [Google Scholar] [CrossRef] [PubMed]
  298. Schaer, D.J.; Buehler, P.W.; Alayash, A.I.; Belcher, J.D.; Vercelotti, G.M. Hemolysis and free hemoglobin revisited: Exploring hemoglobin and hemin scavengers as a novel class of therapeutic proteins. Blood 2013, 121, 1276–1284. [Google Scholar] [CrossRef] [PubMed]
  299. Canesin, G.; Di Ruscio, A.; Li, M.; Ummarino, S.; Hedblom, A.; Choudhury, R.; Krzyzanowska, A.; Csizmadia, E.; Palominos, M.; Stiehm, A.; et al. Scavenging of labile heme by hemopexin is a key checkpoint in cancer growth and metastases. Cell Rep. 2020, 32, 108181. [Google Scholar] [CrossRef] [PubMed]
  300. Tarnawski, R.; Skladowski, K.; Maciejewski, B. Prognostic value of hemoglobin concentration in radiotherapy for cancer of supraglottic larynx. Int. J. Radiat. Oncol. Biol. Phys. 1997, 38, 1007–1011. [Google Scholar] [CrossRef] [PubMed]
  301. Grogan, M.; Thomas, G.M.; Malemed, J.; Wong, F.L.; Pearcey, R.G.; Joseph, P.K.; Portelance, L.; Crook, J.; Jones, K.D. The importance of hemoglobin levels during radiotherapy for carcinoma of the cervix. Cancer 1999, 86, 1528–1536. [Google Scholar] [CrossRef]
  302. Dejure, F.R.; Eilers, M. MYC and tumor metabolism: Chicken and egg. EMBO J. 2017, 36, 3409–3420. [Google Scholar] [CrossRef] [PubMed]
  303. Canesin, G.; Muralidharam, A.M.; Swanson, K.D.; Wegiel, B. HO-1 and heme: G-quadruplex interaction choreograph DNA damage response and cancer growth. Cells 2021, 10, 1801. [Google Scholar] [CrossRef] [PubMed]
  304. Santoro, A.M.; Lo Giudice, M.C.; D’Urso, A.; Lauceri, R.; Purello, R.; Milardi, D. Cationic porphyrins are reversible proteasome inhibitors. J. Am. Chem. Soc. 2012, 134, 10451–10457. [Google Scholar] [CrossRef]
  305. Vallelian, F.; Deuel, J.W.; Opitz, L.; Schaer, C.A.; Puglia, M.; Lönn, M.; Engelsberger, W.; Schauer, S.; Karnaukhova, E.; Spahn, D.R.; et al. Proteasome inhibition and oxidative reactions disrupt cellular homeostasis during heme stress. Cell Death Differ. 2015, 22, 597–611. [Google Scholar] [CrossRef]
  306. Nowis, D.; Legat, M.; Grzela, T.; Niderla, J.; Wilczek, E.; Wilczynski, G.M.; Głodkowska, E.; Mrówka, P.; Issat, T.; Dulak, J.; et al. Heme oxygenase-1 protects tumor cells against photodynamic therapy-mediated cytotoxicity. Oncogene 2006, 25, 3365–3374. [Google Scholar] [CrossRef]
  307. Pei, L.; Kong, Y.; Shao, C.; Yue, X.; Wang, Z.; Zhang, N. Heme oxygenase-1 induction mediates chemoresistance of breast cancer cells to pharmorubicin by promoting autophagy via PI3K/Akt pathway. J. Cell Mol. Med. 2018, 22, 5311–5321. [Google Scholar] [CrossRef] [PubMed]
  308. Slebos, D.J.; Ryter, S.W.; van der Toorn, M.; Liu, F.; Guo, F.; Baty, C.J.; Karlsson, J.M.; Watkins, S.C.; Kim, H.P.; Wang, X.; et al. Mitochondrial localization and function of heme oxygenase-1 in cigarette smoke-induced cell death. Am. J. Respir. Cell Mol. Biol. 2007, 36, 409–417. [Google Scholar] [CrossRef] [PubMed]
  309. Liu, S.L.; Sun, P.; Li, Y.; Liu, S.-S.; Lu, Y. Exosomes as critical mediators to cell-to-cell communication in cancer pathogenesis and their potential clinical application. Transl. Cancer Res. 2019, 8, 298–311. [Google Scholar] [CrossRef] [PubMed]
  310. Osaki, M.; Okada, F. Exosomes and their role in cancer progression. Yonago Acta Med. 2019, 62, 182–190. [Google Scholar] [CrossRef] [PubMed]
  311. Wu, F.; Weigel, K.J.; Zhou, H.; Wang, X.J. paradoxical roles of TGF-β signaling in suppressing and promoting squamous cell carcinoma. Acta Biochim. Biophys. Sin. 2018, 50, 98–105. [Google Scholar] [CrossRef] [PubMed]
  312. Yeh, H.W.; Lee, S.S.; Chang, C.Y.; Lang, Y.D.; Jou, Y.S. A new switch for TGFβ in cancer. Cancer Res. 2019, 79, 3797–3805. [Google Scholar] [CrossRef] [PubMed]
  313. Zhang, M.; Zhang, Y.Y.; Chen, Y.; Wang, J.; Wang, Q.; Lu, H. TGF-β signaling and resistance to cancer therapy. Front. Cell Dev. Biol. 2021, 9, 786728. [Google Scholar] [CrossRef] [PubMed]
  314. Shi, X.; Yang, J.; Deng, S.; Xu, H.; Wu, D.; Zeng, Q.; Wang, S.; Hu, T.; Wu, F.; Zhou, H. TGF-β signaling in the tumor metabolic microenvironment and targeted therapies. J. Hematol. Oncol. 2022, 15, 135. [Google Scholar] [CrossRef]
  315. Hua, W.; Ten Dijke, P.; Kostidis, S.; Giera, M.; Hornsveld, M. TGFβ-induced metabolic reprogramming during epithelial-to-mesenchymal transition in cancer. Cell Mol. Life Sci. 2020, 77, 2103–2123. [Google Scholar] [CrossRef]
  316. Angioni, R.; Sanchez-Rodriguez, R.; Viola, A.; Molon, B. TGF-β in cancer: Metabolic driver of the tolerogenic crosstalk in the tumor microenvironment. Cancers 2021, 13, 401. [Google Scholar] [CrossRef]
  317. Arnhold, J. The dual role of myeloperoxidase in immune response. Int. J. Mol. Sci. 2020, 21, 8057. [Google Scholar] [CrossRef] [PubMed]
  318. Arnhold, J.; Malle, E. Halogenation activity of mammalian heme peroxidases. Antioxidants 2022, 11, 890. [Google Scholar] [CrossRef] [PubMed]
  319. Korkmaz, B.; Horwitz, M.S.; Jenne, D.E.; Gauthier, F. Neutrophil elastase, proteinase 3, and cathepsin G as therapeutic targets in human diseases. Pharmacol. Rev. 2010, 62, 726–759. [Google Scholar] [CrossRef] [PubMed]
  320. Matsuoka, R.; Shiba-Ishii, A.; Nakano, N.; Togayachi, A.; Sakashita, S.; Sato, Y.; Minami, Y.; Noguchi, M. Heterotopic production of ceruloplasmin by lung adenocarcinoma is significantly correlated with prognosis. Lung Cancer 2018, 118, 97–104. [Google Scholar] [CrossRef] [PubMed]
  321. Chen, F.; Han, B.; Meng, Y.; Han, Y.; Liu, B.; Zhang, B.; Chang, Y.; Cao, P.; Fan, Y.; Tan, K. Ceruloplasmin correlates with immune infiltration and serves as a prognostic factor in breast cancer. Aging 2021, 13, 20438–20467. [Google Scholar] [CrossRef] [PubMed]
  322. Chen, J.; Cheuk, I.W.-J.; Siu, M.-T.; Yang, W.; Chen, A.S.L.; Sin, V.Y.; Kwong, A. Human haptoglobin contributes to breast cancer oncogenesis through glycolytic activity modulation. Am. J. Cancer Res. 2020, 10, 2865–2877. [Google Scholar] [PubMed]
  323. Fiorito, V.; Tolosano, E. Hemopexin and cancer. Int. J. Mol. Sci. 2022, 23, 997. [Google Scholar] [CrossRef] [PubMed]
  324. Wei, Z.; Liu, G.; Jia, R.; Zhang, W.; Li, L.; Zhang, Y.; Wang, Z.; Bai, X. Targeting secretory leukocyte protease inhibitor (SLPI) inhibits colorectal cancer cell growth, migration and invasion via downregulation of AKT. Peer J. 2020, 8, e9400. [Google Scholar] [CrossRef]
  325. Wang, C.; Liao, Y.; He, W.; Zhang, H.; Zuo, D.; Liu, W.; Yang, Z.; Qiu, J.; Yuan, Y.; Li, K.; et al. Elafin promotes tumor metastasis and attenuates the anti-metastatic effects of erlotinib via binding to EGFR in hepatocellular carcinoma. J. Exp. Clin. Cancer Res. 2021, 40, 113. [Google Scholar] [CrossRef]
  326. Jin, Y.; Wang, W.; Wang, Q.; Zhang, Y.; Zahid, K.R.; Raza, U.; Gong, Y. Alpha-1-antichymotrypsin as a novel biomarker for diagnosis, prognosis, and therapy prediction in human diseases. Cancer Cell Int. 2022, 22, 156. [Google Scholar] [CrossRef]
  327. Tai, C.-S.; Lin, Y.-R.; Teng, T.-H.; Tu, S.-J.; Chou, C.-H.; Huang, Y.-R.; Huang, W.-C.; Weng, S.-L.; Huang, H.-D.; Chen, Y.-L.; et al. Haptoglobin expression correlates with tumor differentiation and five-year overall survival rate in hepatocellular carcinoma. PLoS ONE 2017, 12, e0171269. [Google Scholar] [CrossRef] [PubMed]
  328. Zhu, B.; Zhi, Q.; Xie, Q.; Wu, X.; Gao, Y.; Chen, X.; Shi, L. Reduced expression of ferroportin1 and ceruloplasmin predicts poor prognosis in adrenocortical carcinoma. J. Trace Elem. Med. Biol. 2019, 56, 52–59. [Google Scholar] [CrossRef] [PubMed]
  329. Tan, M.G.; Kumarasinghe, M.P.; Wang, S.M.; Ooi, L.L.; Aw, S.E.; Hui, K.M. Modulation of iron-regulatory genes in human hepatocellular carcinoma and its physiological consequences. Exp. Biol. Med. 2009, 234, 693–702. [Google Scholar] [CrossRef] [PubMed]
  330. Colombo, M.P.; Lombardi, L.; Stoppacciaro, A.; Melani, C.; Parenza, M.; Bottazzi, B.; Parmiani, G. Granulocyte colony-stimulating factor (G-CSF) gene transduction in murine adenocarcinoma drives neutrophil-mediated tumor inhibition in vivo. Neutrophils discriminate between G-CSF-producing and G-CSF-nonproducing tumor cells. J. Immunol. 1992, 149, 113–119. [Google Scholar] [CrossRef] [PubMed]
  331. Di Carlo, E.; Forni, G.; Lollini, P.; Colombo, M.P.; Modesti, A.; Musiani, P. The intriguing role of polymorphonuclear neutrophils in antitumor reactions. Blood 2001, 97, 339–345. [Google Scholar] [CrossRef] [PubMed]
  332. Hicks, A.M.; Riedlinger, G.; Willingham, M.C.; Alexander-Miller, M.A.; von Kap-Herr, C.; Pettenati, M.J.; Sanders, A.M.; Weir, H.M.; Du, W.; Kim, J.; et al. Transferable anticancer innate immunity in spontaneous regression/complete resistance mice. Proc. Natl. Acad. Sci. USA 2006, 103, 7753–7758. [Google Scholar] [CrossRef] [PubMed]
  333. Fridlender, Z.G.; Sun, J.; Kim, S.; Kapoor, V.; Cheng, G.; Ling, L.; Worthen, G.S.; Albelda, S.M. Polarization of tumor-associated neutrophil phenotype by TGF-beta: “N1” versus “N2” TAN. Cancer Cell 2009, 16, 183–194. [Google Scholar] [CrossRef] [PubMed]
  334. Sagiv, J.Y.; Michaeli, J.; Assi, S.; Mishalian, I.; Kisos, H.; Levy, L.; Damti, P.; Lumbroso, D.; Polyansky, L.; Sionov, R.V.; et al. Phenotypic diversity and plasticity in circulating neutrophil subpopulations in cancer. Cell Rep. 2015, 10, 562–573. [Google Scholar] [CrossRef]
  335. Su, Y.; Qiu, Y.; Qiu, Z.; Qu, P. MicroRNA networks regulate the differentiation, expansion and suppression function of myeloid-derived suppressor cells in tumor microenvironment. J. Cancer 2019, 10, 4350–4356. [Google Scholar] [CrossRef]
  336. Basso, D.; Gnatta, E.; Padoan, A.; Fogar, P.; Furlanello, S.; Aita, A.; Bozzato, D.; Zambon, C.F.; Arrigoni, G.; Frasson, C.; et al. PDAC-derived exosomes enrich the microenvironment in MDSCs in a SMAD4-dependent manner through a new calcium related axis. Oncotarget 2017, 8, 84928–84944. [Google Scholar] [CrossRef]
  337. Shang, W.; Tang, Z.; Gao, Y.; Qi, H.; Su, X.; Zhang, Y.; Yang, R. LncRNA RNCR3 promotes Chop expression by sponging miR-185–5p during MDSC differentiation. Oncotarget 2017, 8, 111754–111769. [Google Scholar] [CrossRef] [PubMed]
  338. Kusmartsev, S.; Nefedova, Y.; Yoder, D.; Gabrilovich, D.I. Antigen-specific inhibition of CD8+ T cell response by immature myeloid cells in cancer is mediated by reactive oxygen species. J. Immunol. 2004, 172, 989–999. [Google Scholar] [CrossRef] [PubMed]
  339. Youn, J.-I.; Collazo, M.; Shalova, I.N.; Biswas, S.K.; Gabrilovich, D.I. Characterization of the nature of granulocytic myeloid-derived suppressor cells in tumor-bearing mice. J. Leukoc. Biol. 2012, 91, 167–181. [Google Scholar] [CrossRef] [PubMed]
  340. Ugolini, A.; Tyurin, V.A.; Tyurina, Y.Y.; Tcyganov, E.N.; Donthireddy, L.; Kagan, V.E.; Gabrilovich, D.I.; Veglia, F. Polymorphonuclear myeloid-derived suppressor cells limit antigen cross-presentation by dendritic cells in cancer. JCI Insight 2020, 5, e138581. [Google Scholar] [CrossRef] [PubMed]
  341. Veglia, F.; Tyurin, V.A.; Mohammadyani, D.; Blasi, M.; Duperret, E.K.; Donthireddy, L.; Hashimoto, A.; Kapralov, A.; Amoscato, A.; Angelini, R.; et al. Lipid bodies containing oxidatively truncated lipids block antigen cross-presentation by dendritic cells in cancer. Nat. Commun. 2017, 8, 2122. [Google Scholar] [CrossRef] [PubMed]
  342. Al-Khami, A.A.; Zheng, L.; Del Valle, L.; Hossain, F.; Wyczechowska, D.; Zabaleta, J.; Sanchez, M.D.; Dean, M.J.; Rodriguez, P.C.; Ochoa, A.C. Exogenous lipid uptake induces metabolic and functional reprogramming of tumor-associated myeloid-derived suppressor cells. Oncoimmunology 2017, 6, e1344804. [Google Scholar] [CrossRef] [PubMed]
  343. Hossain, F.; Al-Khami, A.A.; Wyczechowska, D.; Hernandez, C.; Zheng, L.; Reiss, K.; Del Valle, L.; Trillo-Tinoco, J.; Maj, T.; Zou, W.; et al. Inhibition of fatty acid oxidation modulates immunosuppressive functions of myeloid-derived suppressor cells and enhances cancer therapies. Cancer Immunol. Res. 2015, 3, 1236–1247. [Google Scholar] [CrossRef] [PubMed]
  344. Veglia, F.; Tyurin, V.A.; Blasi, M.; De Leo, A.; Kossenkov, A.V.; Donthireddy, L.; To, T.S.J.; Schug, Z.; Basu, S.; Wang, F.; et al. Fatty acid transport protein 2 reprograms neutrophils in cancer. Nature 2019, 569, 73–78. [Google Scholar] [CrossRef]
  345. Liu, T.W.; Gammon, S.T.; Yang, P.; Ma, W.; Wang, J.; Piwnica-Worms, D. Inhibition of myeloperoxidase enhances immune checkpoint therapy for melanoma. J. Immunother. Cancer 2023, 11, e005837. [Google Scholar] [CrossRef]
  346. Cui, C.; Chakraborty, K.; Tang, X.A.; Zhou, G.; Schoenfelt, K.Q.; Becker, K.M.; Hoffmann, A.; Chang, Y.-F.; Blank, A.; Reardon, C.A.; et al. Neutrophil elastase selectively kills cancer cells and attenuates tumorigenesis. Cell 2021, 184, 3163–3177. [Google Scholar] [CrossRef]
  347. Deryugina, E.; Carré, A.; Ardi, V.; Muramatsu, T.; Schmidt, J.; Pham, C.; Quigley, J.P. neutrophil elastase facilitates tumor cell intravasation and early metastatic events. iScience 2020, 23, 101799. [Google Scholar] [CrossRef] [PubMed]
  348. Hougton, A.M.; Rzymkiewicz, D.M.; Ji, H.; Gregory, A.D.; Egea, E.E.; Metz, H.E.; Stolz, D.B.; Land, S.R.; Marconcini, L.A.; Kliment, C.R.; et al. Neutrophil elastase-mediated degradation of IRS-1 accelerates lung tumor growth. Nat. Med. 2010, 16, 219–223. [Google Scholar] [CrossRef] [PubMed]
  349. Lerman, I.; Hammes, S.R. Neutrophil elastase in the tumor microenvironment. Steroids 2018, 133, 96–101. [Google Scholar] [CrossRef] [PubMed]
  350. Tubio-Perez, R.A.; Torres-Durán, M.; Fernández-Villar, A.; Ruano-Raviña, A. Alpha-1 antitrypsin deficiency and risk of lung cancer: A systematic review. Transl. Oncol. 2021, 14, 100914. [Google Scholar] [CrossRef] [PubMed]
  351. Hiller, A.-M.; Eksröm, M.; Piitulainen, E.; Lindberg, A.; Rönmark, E.; Tanash, H. Cancer risk in severe alpha-1-antitrypsin deficiency. Eur. Respir. J. 2022, 60, 103200. [Google Scholar] [CrossRef] [PubMed]
  352. Kistowski, M.; Debski, J.; Karczmarski, J.; Paziewska, A.; Oledzki, J.; Mikula, M.; Ostrowski, J.; Dadlez, M. A Strong neutrophil elastase proteolytic fingerprint marks the carcinoma tumor proteome. Mol. Cell Proteom. 2017, 16, 213–227. [Google Scholar] [CrossRef] [PubMed]
  353. Akizuki, M.; Fukutomi, T.; Takasugi, M.; Takahashi, S.; Sato, T.; Harao, M.; Mizumoto, T.; Yamashita, J.-I. Prognostic significance of immunoreactive neutrophil elastase in human breast cancer: Long-term follow-up results in 313 patients. Neoplasia 2007, 9, 260–264. [Google Scholar] [CrossRef] [PubMed]
  354. Foekens, J.A.; Ries, C.; Look, M.P.; Gippner-Steppert, C.; Klijn, J.G.; Jochum, M. Elevated expression of polymorphonuclear leukocyte elastase in breast cancer tissue is associated with tamoxifen failure in patients with advanced disease. Br. J. Cancer 2003, 88, 1084–1090. [Google Scholar] [CrossRef]
  355. Okada, Y.; Nakanishi, I. Activation of matrix metalloproteinase 3 (stromelysin) and matrix metalloproteinase 2 (‘gelatinase’) by human neutrophil elastase and cathepsin G. FEBS Lett. 1989, 249, 353–356. [Google Scholar] [CrossRef]
  356. Shamanian, P.; Schwartz, J.D.; Pocock, B.J.Z.; Monea, S.; Whiting, D.; Marcus, S.G.; Mignatti, P. Activation of progelatinase (MMP-2) by neutrophil elastase, cathepsin G, and proteinase-3: A role for inflammatory cells in tumor invasion and angiogenesis. J. Cell Physiol. 2001, 189, 197–206. [Google Scholar] [CrossRef]
  357. Gaggar, A.; Li, Y.; Weathington, N.; Winkler, M.; Kong, M.; Jackson, P.; Blalock, J.E.; Clancy, J.P. Matrix metalloprotease-9 dysregulation in lower airway secretions of cystic fibrosis patients. Am. J. Physiol. Lung Cell Mol. Physiol. 2007, 293, L96–L104. [Google Scholar] [CrossRef] [PubMed]
  358. Li, J.; Xu, X.; Jiang, Y.; Hansbro, N.G.; Hansbro, P.M.; Xu, J.; Liu, G. Elastin is a key factor of tumor development in colorectal cancer. BMC Cancer 2020, 20, 217. [Google Scholar] [CrossRef] [PubMed]
  359. Fang, T.; Zhang, L.; Yin, X.; Wang, Y.; Zhang, X.; Bian, X.; Jiang, X.; Yang, S.; Xue, Y. The prognostic marker elastin correlates with epithelial-mesenchymal transition and vimentin-positive fibroblasts in gastric cancer. J. Pathol. Clin. Res. 2023, 9, 56–72. [Google Scholar] [CrossRef]
  360. Henke, E.; Nandigama, R.; Ergün, S. Extracellular matrix in the tumor microenvironment and its impact on cancer therapy. Front. Mol. Biosci. 2020, 6, 160. [Google Scholar] [CrossRef] [PubMed]
  361. Taggart, C.; Cervantes-Laurean, D.; Kim, G.; McElvaney, N.G.; Wehr, N.; Moss, J.; Levine, R.L. Oxidation of either methionine 351 or methionine 358 in alpha 1-antitrypsin causes loss of anti-neutrophil elastase activity. J. Biol. Chem. 2000, 275, 27258–27265. [Google Scholar] [CrossRef] [PubMed]
  362. Dittrich, A.S.; Kühbandner, I.; Gehrig, S.; Rickert-Zacharias, V.; Twigg, M.; Wege, S.; Taggart, C.C.; Herth, F.; Schultz, C.; Mall, M.A. Elastase activity on sputum neutrophils correlates with severity of lung disease in cystic fibrosis. Eur. Respir. J. 2018, 51, 1701910. [Google Scholar] [CrossRef] [PubMed]
  363. Ramaha, A.; Patston, P.A. Release and degradation of angiotensin I and II from angiotensinogen by neutrophil serine proteases. Arch. Biochem. Biophys. 2002, 397, 77–83. [Google Scholar] [CrossRef] [PubMed]
  364. Vidotti, D.B.; Casarini, D.E.; Christovam, P.C.; Leite, C.A.; Schor, N.; Boim, M.A. High glucose concentration stimulates intracellular renin activity and angiotensin II generation in rat mesangial cells. Am. J. Physiol. Renal Physiol. 2004, 286, F1039–F1045. [Google Scholar] [CrossRef]
  365. Penafuerte, C.A.; Gagnon, B.; Sirois, J.; Murphy, J.; MacDonald, N.; Tremblay, M.L. Identification of neutrophil-derived proteases and angiotensin II as biomarkers of cancer cachexia. Br. J. Cancer 2016, 114, 680–687. [Google Scholar] [CrossRef]
  366. Song, Y.H.; Li, Y.; Du, J.; Mitch, W.E.; Rosenthal, N.; Delafontaine, P. Muscle-specific expression of IGF-1 blocks angiotensin II-induced skeletal muscle wasting. J. Clin. Investig. 2005, 115, 451–458. [Google Scholar] [CrossRef]
  367. Trobec, K.; von Haehling, S.; Anker, S.D.; Lainsack, M. Growth hormone, insulin-like growth factor 1, and insulin signaling—A pharmacological target in body wasting and cachexia. J. Cachexia Sarcopenia Muscle 2011, 2, 191–200. [Google Scholar] [CrossRef] [PubMed]
  368. Sanders, P.M.; Russell, S.T.; Tisdale, M.J. Angiotensin II directly induces muscle protein catabolism through the ubiquitin proteasome proteolytic pathway and may play a role in cancer cachexia. Br. J. Cancer 2005, 93, 425–434. [Google Scholar] [CrossRef] [PubMed]
  369. Lumbers, E.R.; Delforce, S.J.; Pringle, K.G.; Smith, G.R. The lung, the heart, the novel coronavirus, and the renin-angiotensin system; the need for clinical trials. Front. Med. 2020, 7, 248. [Google Scholar] [CrossRef] [PubMed]
  370. Lamas-Barreiro, J.M.; Alonso-Suárez, M.; Fernández-Martín, J.J.; Saavedra-Alonso, J.A. Angiotensin II suppression in SARS-CoV-2 infection; a therapeutic approach. Nefrologia 2020, 40, 213–216. [Google Scholar] [CrossRef]
  371. Ino, K.; Shibata, K.; Kajiyama, H.; Yamamoto, E.; Nagasaka, T.; Nawa, A.; Nomura, S.; Kikkawa, F. Angiotensin II type 1 receptor expression in ovarian cancer and its correlation with tumour angiogenesis and patient survival. Br. J. Cancer 2006, 94, 552–560. [Google Scholar] [CrossRef] [PubMed]
  372. Shirotake, S.; Miyajima, A.; Kosaka, T.; Tanaka, N.; Maeda, T.; Kikuchi, E.; Oya, M. Angiotensin II type 1 receptor expression and microvessel density in human bladder cancer. Urology 2011, 77, 1009.e19–1009.e25. [Google Scholar] [CrossRef]
  373. Imai, N.; Hashimoto, T.; Kihara, M.; Yoshida, S.; Kawana, I.; Yazawa, T.; Kitamura, H.; Umemura, S. Roles for host and tumor angiotensin II type 1 receptor in tumor growth and tumor-associated angiogenesis. Lab. Investig. 2007, 87, 189–198. [Google Scholar] [CrossRef]
  374. Oh, E.; Kim, J.Y.; Cho, Y.; An, H.; Lee, N.; Jo, H.; Ban, C.; Seo, J.H. Overexpression of angiotensin II type 1 receptor in breast cancer cells induces epithelial-mesenchymal transition and promotes tumor growth and angiogenesis. Biochim. Biophys. Acta 2016, 1863, 1071–1081. [Google Scholar] [CrossRef]
  375. Smith, G.R.; Missailidis, S. Cancer, inflammation and the AT1 and AT2 receptors. J. Inflamm. 2004, 1, 3. [Google Scholar] [CrossRef]
  376. Catarata, M.J.; Ribeiro, R.; Oliviera, M.J.; Cordeiro, C.R.; Medeiros, R. Renin-angiotensin system in lung tumor and microenvironment interactions. Cancers 2020, 12, 1457. [Google Scholar] [CrossRef]
  377. Carey, R.M.; Whelton, P.K. Prevention, detection, evaluation, and management of high blood pressure in adults: Synopsis of the 2017 American College of Cardiology/American Heart Association Hypertension Guideline. Ann. Int. Med. 2018, 168, 351–358. [Google Scholar] [CrossRef] [PubMed]
  378. Brenner, B.M.; Cooper, M.E.; de Zeeuw, D.; Keane, W.F.; Mitch, W.E.; Parving, H.H.; Remuzzi, G.; Snapinn, S.M.; Zhang, Z.; Shanifar, S.; et al. Effects of losartan on renal and cardiovascular outcomes in patients with type 2 diabetes and nephropathy. N. Engl. J. Med. 2001, 345, 861–869. [Google Scholar] [CrossRef] [PubMed]
  379. Sipahi, I. Risk of cancer with angiotensin-receptor blockers increases with increasing cumulative exposure: Meta-regression analysis of randomized trials. PLoS ONE 2022, 17, e0263461. [Google Scholar] [CrossRef] [PubMed]
  380. Choudhari, S.K.; Choudhari, M.; Bagde, S.; Gadbail, A.R.; Joshi, V. Nitric oxide and cancer: A review. World J. Surg. Oncol. 2013, 11, 18. [Google Scholar] [CrossRef] [PubMed]
  381. Niu, F.; Yu, Y.; Li, Z.; Ren, Y.; Li, Z.; Ye, Q.; Liu, P.; Ji, C.; Qian, L.; Xiong, Y. Arginase: A emerging and promising therapeutic target for cancer treatment. Biomed. Pharmacother. 2022, 149, 112840. [Google Scholar] [CrossRef] [PubMed]
  382. Wu, C.W.; Chi, C.W.; Lin, E.C.; Lui, W.Y.; P’Eng, F.K.; Wang, S.R. Serum arginase level in patients with gastric cancer. J. Clin. Gastroenterol. 1994, 18, 84–85. [Google Scholar] [CrossRef]
  383. Leu, S.Y.; Wang, S.R. Clinical significance of arginase in colorectal cancer. Cancer 1992, 70, 733–736. [Google Scholar] [CrossRef]
  384. Chrzanowska, A.; Graboń, W.; Mielczarek-Puta, M.; Barańczyk-Kuźma, A. Significance of arginase determination in body fluids of patients with hepatocellular carcinoma and liver cirrhosis before and after surgical treatment. Clin. Biochem. 2014, 47, 1056–1059. [Google Scholar] [CrossRef]
  385. Roci, I.; Watrous, J.D.; Lagerborg, K.A.; Lafranchi, L.; Lindqvist, A.; Jain, M.; Nilsson, R. Mapping metabolic events in the cancer cell cycle reveals arginine catabolism in the committed SG2M phase. Cell Rep. 2019, 26, 1691–1700.e5. [Google Scholar] [CrossRef]
  386. Srivastava, S.; Ghosh, S.K. Modulation of L-arginine-arginase metabolic pathway enzymes: Immunocytochemistry and mRNA expression in peripheral blood and tissue levels in head and neck squamous cell carcinomas in North East India. Asian Pac. J. Cancer Prev. 2015, 16, 7031–7038. [Google Scholar] [CrossRef]
  387. Munder, M.; Engelhardt, M.; Knies, D.; Medenhoff, S.; Wabnitz, G.; Luckner-Minden, C.; Feldmeyer, N.; Voss, R.H.; Kropf, P.; Müller, I.; et al. Cytotoxicity of tumor antigen specific human T cells is impaired by arginine depletion. PLoS ONE 2013, 5, e63521. [Google Scholar] [CrossRef]
  388. Segal, A.W.; Jones, O.T.G. Novel cytochrome b system in phagocytic vacuoles from human granulocytes. Nature 1978, 276, 515–517. [Google Scholar] [CrossRef] [PubMed]
  389. Wientjes, F.B.; Segal, A.W. NADPH oxidase and the respiratory burst. Semin. Cell Biol. 1995, 6, 357–365. [Google Scholar] [CrossRef] [PubMed]
  390. Olson, J.S.; Ballou, D.P.; Palmer, G.; Massey, V. The reaction of xanthine oxidase with molecular oxygen. J. Biol. Chem. 1974, 249, 4350–4362. [Google Scholar] [CrossRef] [PubMed]
  391. Anderson, R.F.; Hille, R.; Massey, V. The radical chemistry of milk xanthine oxidase as studied by radiation chemistry technique. J. Biol. Chem. 1986, 261, 15870–15876. [Google Scholar] [CrossRef] [PubMed]
  392. Carrell, R.W.; Winterbourn, C.C.; Rachmilewitz, E.A. Activated oxygen and hemolysis. Br. J. Hematol. 1975, 30, 259–264. [Google Scholar] [CrossRef]
  393. Harel, S.; Kanner, J. Hemoglobin and myoglobin as inhibitors of hydroxyl radical generation in a model system of “iron redox” cycle. Free Radic. Res. Commun. 1989, 6, 1–10. [Google Scholar] [CrossRef] [PubMed]
  394. Nozik-Grayck, E.; Suliman, H.B.; Piantadosi, C.A. Extracellular superoxide dismutase. Int. J. Biochem. Cell Biol. 2005, 37, 2466–2471. [Google Scholar] [CrossRef]
  395. Teoh-Fitzgerald, M.L.; Fitzgerald, M.P.; Jensen, T.J.; Futscher, B.W.; Domann, F.E. Genetic and epigenetic inactivation of extracellular superoxide dismutase promotes an invasive phenotype in human lung cancer by disrupting ECM homeostasis. Mol. Cancer Res. 2012, 10, 40–51. [Google Scholar] [CrossRef]
  396. Teoh-Fitzgerald, M.L.; Fitzgerald, M.P.; Zhong, W.; Askeland, R.W.; Domann, F.E. Epigenetic reprogramming governs EcSOD expression during human mammary epithelial cell differentiation, tumorigenesis and metastasis. Oncogene 2014, 33, 358–368. [Google Scholar] [CrossRef]
  397. Laukkanen, M.O. Extracellular superoxide dismutase: Growth promotor or tumor suppressor? Oxid. Med. Cell Longev. 2016, 2016, 3612589. [Google Scholar] [CrossRef] [PubMed]
  398. Zhang, Y.; Lu, X.; Zhang, Y.; Zhao, D.; Gong, H.; Du, Y.; Sun, H. The effect of extracellular superoxide dismutase (SOD3) gene in lung cancer. Front. Oncol. 2022, 12, 722646. [Google Scholar] [CrossRef] [PubMed]
  399. Mira, E.; Carmona-Rodriguez, L.; Pérez-Villamil, B.; Casas, J.; Fernández-Aceñero, M.J.; Martinez-Rey, D.; Martin-González, P.; Heras-Murillo, I.; Paz-Cabezas, M.; Tardáguila, M.; et al. SOD3 improves the tumor response to chemotherapy by stabilizing endothelial HIF-2alpha. Nat. Commun. 2018, 9, 575. [Google Scholar] [CrossRef] [PubMed]
  400. Carmona-Rodriguez, L.; Martinez-Rey, D.; Fernández-Aceñero, M.J.; González-Martin, A.; Paz-Cabezas, M.; Rodriguez-Rodriguez, N.; Pérez-Villamil, B.; Sáez, M.E.; Diaz-Rubio, E.; Mira, E.; et al. SOD3 induces a HIF-2a-dependent program in endothelial cells that provides a selective signal for tumor infiltration by T cells. J. Immunother. Cancer 2020, 8, e000432. [Google Scholar] [CrossRef] [PubMed]
  401. Evans, M.D.; Pryor, W.A. Cigarette smoking. emphysema and damage to alpha 1-proteinase inhibitor. Am. J. Physiol. Lung Cell Mol. Physiol. 1994, 266, L593–L611. [Google Scholar] [CrossRef]
  402. Martinez-Ray, D.; Carmona-Rodriguez, L.; Fernández-Aceñero, M.J.; Mira, E.; Mañes, S. Extracellular superoxide dismutase, the endothelial basement membrane, and the WNT pathway: New players in vascular normalization and tumor infiltration by T cells. Front. Immunol. 2020, 11, 579552. [Google Scholar] [CrossRef]
  403. Teoh, M.L.T.; Fitzgerald, M.P.; Oberley, L.W.; Domann, F.E. Overexpression of extracellular superoxide dismutase attenuates heparanase expression and inhibits breast carcinoma cell growth and invasion. Cancer Res. 2009, 69, 6355–6363. [Google Scholar] [CrossRef]
  404. Mueller, B.M.; Schraufstatter, I.U.; Goncharova, V.; Povaliy, T.; DiScipio, R.; Khaldoyanidi, S.K. Hyaluronan inhibits postchemotherapy tumor regrowth in a colon carcinoma xenograft model. Mol. Cancer Ther. 2010, 9, 3024–3032. [Google Scholar] [CrossRef]
  405. Tian, X.; Azpurua, J.; Hine, C.; Vaidya, A.; Myakishev-Rempel, M.; Ablaeva, J.; Mao, Z.; Nevo, E.; Gorbunova, V.; Seluanov, A. High molecular weight hyaluronan mediates the cancer resistance of the naked mole-rat. Nature 2013, 499, 346–349. [Google Scholar] [CrossRef]
  406. Toole, B.P. Hyaluronan: From extracellular glue to pericellular cue. Nat. Rev. Cancer 2004, 4, 528–539. [Google Scholar] [CrossRef]
  407. Liu, M.; Tolg, C.; Turley, E. Dissecting the dual nature of hyaluronan in the tumor microenvironment. Front. Immunol. 2019, 10, 947. [Google Scholar] [CrossRef] [PubMed]
  408. Frangociannis, N.G. Transforming growth factor-β in tissue fibrosis. J. Exp. Med. 2020, 217, e20190103. [Google Scholar] [CrossRef] [PubMed]
  409. Budi, E.H.; Schaub, J.R.; Decaris, M.; Turner, S.; Derynck, R. TGF-β as a driver of fibrosis: Physiological roles and therapeutic opportunities. J. Pathol. 2021, 254, 358–373. [Google Scholar] [CrossRef] [PubMed]
  410. Taleb, S.; Cancello, R.; Clément, K.; Lacasa, D. Cathepsin S promotes human preadipocyte differentiation: Possible involvement of fibronectin degradation. Endocrinology 2006, 147, 4950–4959. [Google Scholar] [CrossRef] [PubMed]
  411. Chen, N.; Zhang, G.; Fu, J.; Wu, Q. Matrix metalloproteinase-14 (MMP-14) down-regulation inhibits esophageal squamous cell carcinoma cell migration, invasion, and proliferation. Thorac. Cancer 2020, 11, 3168–3174. [Google Scholar] [CrossRef] [PubMed]
  412. Aguilera-Montilla, N.; Bailón, E.; Uceda-Castro, R.; Ugarte-Bercal, E.; Santos, A.; Guitérrez-González, A.; Pérez-Sánchez, C.; Van den Steen, P.E.; Opdenakker, G.; Garcia-Marco, J.E.; et al. MMP-9 affects gene expression in chronic lymphocytic leukemia revealing CD99 as an MMP-9 target and a novel partner in malignant cell migration/arrest. Oncogene 2019, 38, 4605–4619. [Google Scholar] [CrossRef] [PubMed]
  413. Cabral-Pacheco, G.A.; Garca-Veloz, I.; Castruita-De la Rosa, C.; Ramirez, J.M.; Perez-Romero, B.; Guerrero-Rodriguez, J.F.; Martinez-Avila, N.; Martinez-Fierro, M.L. The role of matrix metalloproteinases and their inhibitors in human diseases. Int. J. Mol. Sci. 2020, 21, 9739. [Google Scholar] [CrossRef] [PubMed]
  414. Cao, Z.; Zhao, K.; Jose, I.; Hoogenraad, N.J.; Osellame, L.D. Biomarkers for cancer cachexia: A mini review. Int. J. Mol. Sci. 2021, 22, 4501. [Google Scholar] [CrossRef]
  415. Prokopchuk, O.; Grünwald, B.; Nitsche, U.; Jäger, C.; Prokopchuk, O.L.; Schubert, E.C.; Friess, H.; Martignoni, M.E.; Krüger, A. Elevated systemic levels of the matrix metalloproteinase inhibitor TIMP-1 correlate with clinical markers of cachexia in patients with chronic pancreatitis and pancreatic cancer. BMC Cancer 2018, 18, 128. [Google Scholar] [CrossRef]
  416. Narasimhan, A.; Shahda, S.; Kays, J.K.; Perkins, S.M.; Cheng, L.; Schloss, K.N.H.; Schloss, D.E.I.; Koniaris, L.G.; Zimmers, T.A. Identification of potential serum protein biomarkers and pathways for pancreatic cancer cachexia using an aptamer-based discovery platform. Cancers 2020, 12, 3787. [Google Scholar] [CrossRef]
  417. Bachman, K.E.; Herman, J.G.; Corn, P.G.; Merlo, A.; Costello, J.F.; Cavenee, W.K.; Baylin, S.B.; Graff, J.R. Methylation-associated silencing of the tissue inhibitor of metalloproteinase-3 gene suggest a suppressor role in kidney, brain, and other human cancers. Cancer Res. 1999, 59, 798–802. [Google Scholar] [PubMed]
  418. Ahonen, M.; Baker, A.H.; Kahari, V.M. Adenovirus-mediated gene delivery of tissue inhibitor of metalloproteinases-3 inhibits invasion and induces apoptosis in melanoma cells. Cancer Res. 1998, 58, 2310–2315. [Google Scholar] [PubMed]
  419. Baker, A.H.; George, S.J.; Zaltsman, A.B.; Murphy, G.; Newby, A.C. Inhibition of invasion and induction of apoptotic cell death of cancer cell lines by overexpression of TIMP-3. Br. J. Cancer 1999, 79, 1347–1355. [Google Scholar] [CrossRef] [PubMed]
  420. Nagao, Y.; Hisaoka, M.; Matsuyama, A.; Kanemitsu, S.; Hamada, T.; Fukuyama, T.; Nakano, R.; Uchiyama, A.; Kawamoto, M.; Yamaguchi, K.; et al. Association of microRNA-21 expression with its targets, PDCD4 and TIMP3, in pancreatic ductal adenocarcinoma. Mod. Pathol. 2012, 25, 112–121. [Google Scholar] [CrossRef] [PubMed]
  421. Fatheree, J.P.; Guarin, J.R.; McGinn, R.A.; Naber, S.P.; Oudin, M.J. Chemotherapy-induced collagen IV drives cancer cell motility through activation of Src and focal adhesion kinase. Cancer Res. 2022, 82, 2031–2044. [Google Scholar] [CrossRef] [PubMed]
  422. Jansson, M.; Lindberg, J.; Rask, G.; Svensson, J.; Billing, O.; Nazemroaya, A.; Berglund, A.; Wärnberg, F.; Sund, M. Prognostic value of stromal type IV collagen expression in small invasive breast cancers. Front. Mol. Biosci. 2022, 9, 904526. [Google Scholar] [CrossRef]
  423. Lipton, A.; Leitzel, K.; Ali, S.M.; Polimera, H.V.; Nagabhairu, V.; Marks, E.; Richardson, A.E.; Krecko, L.; Ali, A.; Koestler, W.; et al. High turnover of extracellular matrix reflected by specific protein fragments measured in serum Is associated with poor outcomes in two metastatic breast cancer cohorts. Int. J. Cancer 2018, 143, 3027–3034. [Google Scholar] [CrossRef] [PubMed]
  424. Lindgren, M.; Jansson, M.; Tavelin, B.; Dirix, L.; Vermeulen, P.; Nyström, H. Type IV collagen as a potential biomarker of metastatic breast cancer. Clin. Exp. Metastasis 2021, 38, 175–185. [Google Scholar] [CrossRef]
  425. Lee, S.W.; Kim, H.K.; Naidansuren, P.; Ham, K.A.; Choi, H.S.; Ahn, H.Y.; Kim, M.; Kang, D.H.; Kang, S.W.; Joe, A.E. Peroxidasin is essential for endothelial cell survival and growth signaling by sulflimine crosslink-dependent matrix assembly. FASEB J. 2020, 34, 10228–10241. [Google Scholar] [CrossRef]
  426. He, C.; Song, W.; Weston, T.A.; Tran, C.; Kurtz, I.; Zuckerman, J.E.; Guagliardo, P.; Miner, J.H.; Ivanov, S.V.; Bougoure, J.; et al. Peroxidasin-mediated bromine enrichment of basement membranes. Proc. Natl. Acad. Sci. USA 2020, 117, 15827–15836. [Google Scholar] [CrossRef]
  427. Medfai, H.; Khalil, A.; Rousseau, A.; Nuyens, V.; Paumann-Page, M.; Sevcnikar, B.; Furtmüller, P.G.; Obinger, C.; Moguilevsky, N.; Peulen, O.; et al. Human peroxidasin 1 promotes angiogenesis through ERK1/2, Akt, and FAK pathways. Cardiovasc. Res. 2019, 115, 463–475. [Google Scholar] [CrossRef] [PubMed]
  428. Paumann-Page, M.; Kienzl, N.F.; Motwani, J.; Bathish, B.; Paton, L.N.; Magon, N.J.; Sevcnikar, B.; Furtmüller, P.G.; Traxlmayr, M.W.; Obinger, C.; et al. Peroxidasin protein expression and enzymatic activity in metastatic melanoma cell lines are associated with invasive potential. Redox Biol. 2021, 46, 102090. [Google Scholar] [CrossRef] [PubMed]
  429. Wyllie, K.; Panagopoulos, V.; Cox, T.R. The role of peroxidasin in solid cancer progression. Biochem. Soc. Trans. 2023, 51, 1881–1895. [Google Scholar] [CrossRef] [PubMed]
  430. Wang, Z.; Zhai, Z.; Chen, C.; Tian, X.; Xing, Z.; Xing, P.; Yang, Y.; Zhang, J.; Wang, C.; Dong, L. Air pollution particles hijack peroxidasin to disrupt immunosurveillance and promote lung cancer. eLife 2022, 11, e75345. [Google Scholar] [CrossRef]
Figure 1. Concept of immunoediting in tumorigenesis. The three major phases of the interplay between tumor cells and immune cells are schematically presented. Further explanations are given in the text. Abbreviations: MDSCs—myeloid-derived suppressor cells, NK cells—natural killer cells, TME—tumor microenvironment, Tregs—regulatory T cells.
Figure 1. Concept of immunoediting in tumorigenesis. The three major phases of the interplay between tumor cells and immune cells are schematically presented. Further explanations are given in the text. Abbreviations: MDSCs—myeloid-derived suppressor cells, NK cells—natural killer cells, TME—tumor microenvironment, Tregs—regulatory T cells.
Cancers 16 00081 g001
Figure 2. Major effects of tumor cells on the surrounding cells in the TME. The effects are mediated by the release of mediators from tumors cells and special conditions typical of tumors. Further explanations are given in the text. Abbreviations: IL-10—interleukin 10, MDSCs—myeloid-derived suppressor cells, NK cells—natural killer cells, TGF-β—transforming growth factor-β, Tregs—regulatory T cells, VEGF—vascular endothelial growth factor.
Figure 2. Major effects of tumor cells on the surrounding cells in the TME. The effects are mediated by the release of mediators from tumors cells and special conditions typical of tumors. Further explanations are given in the text. Abbreviations: IL-10—interleukin 10, MDSCs—myeloid-derived suppressor cells, NK cells—natural killer cells, TGF-β—transforming growth factor-β, Tregs—regulatory T cells, VEGF—vascular endothelial growth factor.
Cancers 16 00081 g002
Figure 3. Major antagonizing mechanisms against the enhanced levels of reactive species, iron ions, and free heme in cancer cells. The upregulated enzymes, proteins, and pathways are displayed on red backgrounds. The downregulated mechanisms are given on blue background. For more details, see Section 3.3, Section 3.4, Section 3.5 and Section 3.6. Abbreviations: GPX—glutathione peroxidase, GSH—glutathione, GS-SG—oxidized glutathione, HO-1—heme oxygenase 1, MSR—methionine sulfoxide reductase, SLC7A11—cystine/glutamate anti-porter solute carrier family 7 member 11, SOD—superoxide dismutase, Trx—thioredoxin.
Figure 3. Major antagonizing mechanisms against the enhanced levels of reactive species, iron ions, and free heme in cancer cells. The upregulated enzymes, proteins, and pathways are displayed on red backgrounds. The downregulated mechanisms are given on blue background. For more details, see Section 3.3, Section 3.4, Section 3.5 and Section 3.6. Abbreviations: GPX—glutathione peroxidase, GSH—glutathione, GS-SG—oxidized glutathione, HO-1—heme oxygenase 1, MSR—methionine sulfoxide reductase, SLC7A11—cystine/glutamate anti-porter solute carrier family 7 member 11, SOD—superoxide dismutase, Trx—thioredoxin.
Cancers 16 00081 g003
Figure 4. Factors contributing to the depletion of nitric oxide (NO) in the TME of advanced tumors. NO is generated by NO synthases from L-arginine. It is essential for the activation of CD8+ T cells and vasorelaxation. NO depletion is promoted by upregulated or enhanced factors (displayed on the red background) as well as by low-expressed proteins (presented on the blue background). Further explanations are given in the text. Abbreviations: SOD—superoxide dismutase.
Figure 4. Factors contributing to the depletion of nitric oxide (NO) in the TME of advanced tumors. NO is generated by NO synthases from L-arginine. It is essential for the activation of CD8+ T cells and vasorelaxation. NO depletion is promoted by upregulated or enhanced factors (displayed on the red background) as well as by low-expressed proteins (presented on the blue background). Further explanations are given in the text. Abbreviations: SOD—superoxide dismutase.
Cancers 16 00081 g004
Table 2. Effects of the proteins involved in iron and heme metabolism in cancer cells of late-stage cancers.
Table 2. Effects of the proteins involved in iron and heme metabolism in cancer cells of late-stage cancers.
Antioxidative SystemFunctionsEffects in Cancer CellsCancer TypesReferences
Transferrin receptor Iron uptake to cellsUpregulationLiver, breast, colon, and lung cancer[275,280]
FerritinCellular iron storage protein UpregulationEsophageal adenocarcinoma, glioblastoma, and breast and colorectal cancer[276,281]
FerroportinIron export from cellsDownregulationProstate and breast cancers[277,278,279]
Heme oxygenase 1Removal of free hemeUpregulationMultiple types of cancer[282]
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

Arnhold, J. Inflammation-Associated Cytotoxic Agents in Tumorigenesis. Cancers 2024, 16, 81. https://doi.org/10.3390/cancers16010081

AMA Style

Arnhold J. Inflammation-Associated Cytotoxic Agents in Tumorigenesis. Cancers. 2024; 16(1):81. https://doi.org/10.3390/cancers16010081

Chicago/Turabian Style

Arnhold, Jürgen. 2024. "Inflammation-Associated Cytotoxic Agents in Tumorigenesis" Cancers 16, no. 1: 81. https://doi.org/10.3390/cancers16010081

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