Next Article in Journal
Ozone in Chemotherapy-Induced Peripheral Neuropathy—Current State of Art, Possibilities, and Perspectives
Next Article in Special Issue
Regulation of Cell Plasticity by Bromodomain and Extraterminal Domain (BET) Proteins: A New Perspective in Glioblastoma Therapy
Previous Article in Journal
The Role of Light-Regulated Auxin Signaling in Root Development
Previous Article in Special Issue
Impacts of Environmental Pollution on Brain Tumorigenesis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Dialog beyond the Grave: Necrosis in the Tumor Microenvironment and Its Contribution to Tumor Growth

1
Laboratory for Personalized Medicine, Division of Molecular Medicine, Rudjer Boskovic Institute, Bijenicka 54, 10000 Zagreb, Croatia
2
CNRS UMR 9018-METSY, Université Paris-Saclay, Gustave Roussy, 39 Rue Camille Desmoulins, F-94805 Villejuif, France
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(6), 5278; https://doi.org/10.3390/ijms24065278
Submission received: 31 January 2023 / Revised: 27 February 2023 / Accepted: 7 March 2023 / Published: 9 March 2023
(This article belongs to the Special Issue Latest Review Papers in Molecular Oncology 2023)

Abstract

:
Damage-associated molecular patterns (DAMPs) are endogenous molecules released from the necrotic cells dying after exposure to various stressors. After binding to their receptors, they can stimulate various signaling pathways in target cells. DAMPs are especially abundant in the microenvironment of malignant tumors and are suspected to influence the behavior of malignant and stromal cells in multiple ways often resulting in promotion of cell proliferation, migration, invasion, and metastasis, as well as increased immune evasion. This review will start with a reminder of the main features of cell necrosis, which will be compared to other forms of cell death. Then we will summarize the various methods used to assess tumor necrosis in clinical practice including medical imaging, histopathological examination, and/or biological assays. We will also consider the importance of necrosis as a prognostic factor. Then the focus will be on the DAMPs and their role in the tumor microenvironment (TME). We will address not only their interactions with the malignant cells, frequently leading to cancer progression, but also with the immune cells and their contribution to immunosuppression. Finally, we will emphasize the role of DAMPs released by necrotic cells in the activation of Toll-like receptors (TLRs) and the possible contributions of TLRs to tumor development. This last point is very important for the future of cancer therapeutics since there are attempts to use TLR artificial ligands for cancer therapeutics.

1. Introduction

For a long time, biological investigations of malignant diseases have been mainly focused on the malignant cells and their genetic and epigenetic alterations. Since the first years of the 2000s, more emphasis has been placed on the “dialog” between malignant cells and stromal cells. In this review, we will try to illustrate this crosstalk between necrotic cells and live cells (malignant and stromal), and its consequences for tumor growth.
Cell death by necrosis can be caused by acute physical (extreme temperature, radiation, electric shock) or chemical injuries, mechanical trauma, infections, toxins, and ischemia [1]. Necrosis is characterized by cell and organelle swelling, nuclear condensation (pyknosis), and loss of plasma membrane integrity. Swollen cells and membrane rupture caused by membrane permeabilization represent an early event in necrotic cells, while the same event in apoptotic cells occurs later [2,3]. Necrosis is a quasi-constant phenomenon in solid tumors, although of variable magnitude. Sometimes it is amplified by anti-tumor therapy. One consistent characteristic of necrosis is the fact that it simultaneously affects a large number of cells, whereas apoptosis generally affects scattered, individual cells [4]. As soon as a solid tumor reaches 4 mm in diameter, the core region of the tumor, due to inadequate vascularization, experiences hypoxia and nutrient deprivation that leads to necrosis [5]. The presence of hypoxia and necrosis is one key difference between tumors and normal tissues that could potentially become an angle of attack for more selective cancer therapies. Currently, tumor hypoxia is mainly a factor of therapeutic resistance. Hypoxic cells are 2- to 3-fold more resistant than normoxic cells to radiotherapy due to the lack of oxygen in their environment [6]. Tumor core hypoxic cells also exhibit increased chemoresistance because less of the drug reaches the core due to poor vascularization and high interstitial fluid pressures [7]. New strategies to selectively target those resistant hypoxic tumor cells are cytotoxic drugs that only work under hypoxic conditions, gene therapy constructs with hypoxia-inducible promotors, and obligate anaerobic bacteria delivery vectors that selectively colonize and replicate within the tumor necrotic core [6,7,8].
Efficient anti-tumor therapy sometimes leads to necrotic cell death, which unlike apoptosis, induces inflammatory responses that may contribute to tumor regression [4]. However, spontaneous necrosis is often correlated with rapid tumor growth [4,5,9]. One explanation is the fact that angiogenesis is often all the more insufficient and unsuitable as the malignant cells proliferate rapidly. On the other hand, necrosis by itself, in a kind of vicious circle, may promote malignant cell proliferation, genomic instability, metastasis and even (imperfect) angiogenesis. There is a close link between the undesirable effects of necrosis and those of chronic inflammation [4,5,9].

2. Tumor Necrosis in Human Solid Tumors: Clinical and Pathological Aspects

Necrosis is an important prognostic factor. Richards et al. reported in 2011 that there are over 50 published studies confirming the prognostic value of tumor necrosis in patients with solid organ malignant diseases [10]. Nowadays, its number has increased as other meta-analyses and other studies have emerged [11,12,13,14].

2.1. Necrosis Assessment by Medical Imaging

2.1.1. Conventional Magnetic Resonance (MR) Imaging

Conventional contrast enhanced MR imaging has been the main technique to diagnose tumor and its recurrence [15]. Necrosis at initial MRI is often associated with metastatic disease at presentation or disease progression and is mostly studied in renal cell carcinoma and glioblastoma. Increase of necrosis, or de novo necrosis development during treatment, is a negative prognostic factor for some tumors and it precedes progression. Detection of changes (of necrosis) on MRI scans that precede tumor progression are clinically important because they save time and give the opportunity to change ineffective treatment [16,17]. However, MRI offers only limited power to differentiate between tumor recurrence and necrosis because they manifest similar features on MRI scans [18].

2.1.2. Functional Imaging Techniques

Advanced imaging techniques that are available for discrimination between treatment outcomes include functional magnetic resonance (MR) perfusion techniques, diffusion-weighted imaging (DWI), magnetic resonance spectroscopy (MRS), positron emission tomography (PET), and single photon emission CT (SPECT) [18,19]. PET and SPECT use radiopharmaceuticals to image the different functional properties of organs and tissues. Compared with necrotic regions, tumor recurrence is characterized by an increased metabolism of the growing tumor, which is expected to result in higher tracer uptake. A commonly used tracer for PET is fludeoxyglucose (18F-FDG), a glucose analogue, while the potential of amino acid analogs has also been explored [18]. Multimodal functional imaging can give increased accuracy when structural MRI or PET are combined with perfusion techniques or MRS [18,19]. However, the use of multiple techniques is costly, time-consuming, limited by low accessibility, and remains impractical in the clinical setting [18].

2.1.3. Molecular Imaging Techniques

There are several classes of necrosis-avid contrast agents that can bind cell components exposed by the loss of membrane integrity, and can thus be used to visualize necrosis [20,21]. Rhein and its derivatives are anthraquinone compounds, a class of DNA intercalators that exhibit intrinsic fluorescence [22]. They are used for non-invasive visualization of myocardial necrosis [23] and necrosis in tumors [24,25], therefore, representing a base for the development new PET and SPECT tracers [20,26].
A promising PET tracer for in vivo detection of tumor necrosis is gallium-68-labeled IRDye800CW, a cyanine-based fluorescent dye that exhibits excellent necrosis avidity by binding cytoplasmic proteins [21]. Another cyanine dye that shows avidity for necrotic tissues is an FDA-approved fluorescent probe indocyanine green (ICG). It can selectively bind necrotic tissue due to interactions with lipoproteins and phospholipids. In the preclinical model of fluorescence molecular imaging, ICG was used in the hybrid modality system PET/CT/FMI for imaging tumor progression and therapy outcomes in vivo [27].
The known in situ biomarkers of necrosis are also exploited to visualize necrosis or to target therapy to the necrotic area of solid tumors. In situ biomarkers of necrosis, targeted with antibodies and different small molecular compounds, include DNA/histone H1 complex, exposed DNA, heat shock protein 90 (HSP90), fumarase, and high mobility group box1 (HMGB1). Several molecular imaging probes designed for imaging and/or delivery of therapeutics are assessed in preclinical and clinical trials (for an extensive review see [20]).

2.2. Necrosis Assessment by Histopathological Examination

A typical feature of malignant tumors is the formation of large necrotic areas, mainly due to inadequate clearance by the macrophage phagocytic response in addition to hypoxia. In histological sections, tumor necrosis has two morphologically distinct patterns. Coagulative necrosis is characterized by clusters of eosinophilic and anucleated necrotic cells while the architecture of tissue is still preserved. This necrosis is typical for ischemia-induced injuries in all organs except the brain [1,28]. In the brain, hypoxic cell death often causes liquefactive necrosis in which necrotic tissue is completely degraded into a liquid viscous mass, similar to that seen in bacterial infections [1,29].

2.3. Circulating Biomarkers—Biological Assays for Necrosis Assessment

Assays used for necrosis measurement are based on the loss of membrane integrity. Accidental necrosis is characterized by a sudden release of cell content that finds its way into the blood flow. Circulating biomarkers are a measure of excessive cell death and inadequate phagocytic clearance [2]. The most commonly used in vitro assay for assessing the necrosis amounts is the release of cytosolic enzyme lactate dehydrogenase (LDH) [30]. LDH is present in almost all cells, and released molecules are detectable in peripheral blood. Elevated plasma LDH is a sign of necrosis and tissue damage. Free HMGB1 is passively released from necrotic or damaged cells. It is not released from apoptotic cells even after the secondary necrosis due to its binding to nucleosomes [31]. Free circulating HMGB1 can be measured by ELISA [2]. Cytokeratin 18 (CK18) is a structural protein of epithelial cells, highly expressed in many epithelial tissues, including the liver, intestine, lung, kidney, and endocrine glands, as well as many solid tumors [2,32]. Necrotic cells release an unmodified form of CK18, while apoptotic cells release a caspase-cleaved form, whose epitope can be detected with M30 ELISA. M65 ELISA quantitates total circulating CK18 i.e., caspase-cleaved and non-cleaved form [33]. Using both ELISAs, it is possible to estimate the release of non-cleaved CK18, which provides an estimation of necrosis occurring in distant foci [2]. Tumor-derived cell-free DNAs (cfDNAs) are of increasing interest for the early detection of various tumors and their metastasis. cfDNAs found in serum consist mainly of ~166 bp long fragments that correspond to the length of DNA wrapped around nucleosomes. Those fragments are assumed to be released by apoptosis and may contain tumor-specific sequences [2,34]. Longer and shorter fragments were detected in several studies and ascribed to necrosis [35]. A study using massively parallel sequencing proposed that shifts in cfDNA fragment sizes can be used for disease follow-up in hepatocellular carcinoma patients [36]. In some tumors, necrosis may be the main mechanism contributing to cfDNA release in response to ionizing radiation [35]. Another form of nucleic acids available to assay from peripheral blood is extracellular microRNAs (ex-miRNA), which are ribonucleoprotein particles containing short (~20 nucleotides long) non-coding RNA molecules that regulate gene expression at a post-transcriptional level. Serum ex-miRNAs are increased in diseases that induce tissue damage and represent tissue-specific cytotoxicity markers [2]. The release of miR-21, ubiquitous and abundant miRNA [37], was found to be related to necrosis [3].

3. Biology of Tumor Necrosis and Extra-Cellular Necrotic Products

Cell death was initially categorized into three types: type I cell death (apoptosis), type II cell death (autophagy), and type III cell death (necrosis). However, recent studies have identified additional types of cell death. They are classified based on their biochemical properties, functional potential, and morphology according to the Nomenclature Committee on Cell Death (NCCD). The types of cell deaths described include necroptosis, immunogenic cell death, apoptosis (intrinsic and extrinsic), cellular senescence, pyroptosis, lysosome-dependent cell death, mitochondrial permeability transition (MPT)-driven necrosis, entotic cell death, ferroptosis, parthanatos, NETotic cell death, lysosome-dependent cell death, mitotic catastrophe, and autophagy-dependent cell death [38] (Figure 1).
Besides their differences, apoptosis, autophagy, and necrosis share some common characteristics depending on the stimulatory onset and the signaling pathway that follows afterward. When comparing necrosis and apoptosis, the main difference is the cause of cell death. Apoptosis is a programmed process of cellular death that is a result of genetically-induced cell self-destruction with members of the Bcl-2 family and the caspases 3, 7, 8, 9, and 10 as key regulators. Apoptosis is a part of the natural and preplanned cellular mechanisms that allows the system to maintain the balance of multiplication, and to thus maintain the smooth functioning of the body. Meaning, if cells do not undergo their programmed death, it can lead to cancer formation and the accumulation of redundant cells. Apoptotic cell remnants are recognized and destroyed by the immune system.
Necrosis is also a process of cellular death; however, it happens when the cell is exposed to environmental conditions that are not physiological. The result is the destruction of the inner cellular components causing swift cellular and tissue destruction, leading to inflammation that ends with cell death. Necrosis is a pathological process that can be caused by anything, from a change in oxygen level, pathogens, toxins, and temperature leading to damage of the cell membrane. Necrosis is a random unregulated event that does not require energy on a biochemical level, while apoptosis does require energy as it is an active process [39].
Necroptosis is a type of necrosis that is triggered by innate immune activity and results in the rupture of dead cells with leakage of intracellular elements. Necroptosis is a programmed form of necrosis that comes from environmental factors. Necroptosis is a hybrid of necrosis and apoptosis: it is a regulated necrosis mediated by cellular death receptors. Unlike necrosis, necroptosis is a highly regulated programmed cell death, which unlike apoptosis, does not involve caspase activation. It shares some similarities with necrosis, such as the loss of ATP, swelling of the cell, generation of ROS, and release of lysosomal enzymes. Necroptosis has been implicated in the pathology of many diseases, such as acute tissue damage, ischemia-reperfusion injury, stroke, and myocardial infarction [40]. In apoptosis, cytokines release is either not present or significantly decreased. However, in necroptosis, the release of inflammatory cytokines is one of the hallmarks. It is strongly associated with robust inflammation that induces immune activation. Necroptosis is specific to vertebrates and may have developed as a supportive mechanism against pathogens. Pathogens have developed a system of survival within the attacked cell by utilizing virus-encoded inhibitors of caspase activity that can block caspase (and apoptosis), therefore, necroptosis assures cell death. It is a reliable mechanism during viral infection which leads to anti-viral inflammation [41].

3.1. Stages in Necrosis

Necrosis is considered an irreversible injury due to membrane damage. It starts with a persistent cell injury as a result of a pathological process where, after a certain point, the injury is irreversible. It is caused by the environmental conditions of the exposed cell, which are not physiological to the cell. Stages of necrosis start with swelling of the cell. It continues to the chromatin digestion and membrane disruption. The inner content of the cell, vacuoles, and organelles break down and the cell is decayed. This leads to leakage of the intracellular content, which provokes the inflammation and the immune response. Necrosis leads to a completely irreversible state of the cell since the final stage is enzymatic degradation of the cell.

3.2. Relationships with Hypoxia—Metabolic Reprogramming (Warburg Effect)

Cancer cells are ravenous due to their steady inappropriate growth. They tend to shape their environment to ensure their proliferation, despite poor blood and oxygen supply, and, therefore, require metabolic reprogramming. In addition to their survival and uninterrupted proliferation, the metabolic reprogramming of malignant cells often facilitates tissue invasion, immune escape, and resistance to therapy. At the heart of this metabolic reprogramming is the Warburg effect, which is a tendency to switch off oxidative phosphorylation and to activate non-mitochondrial glycolysis. Incoming glucose is converted to pyruvate, but instead of entering the citric acid cycle, most of the pyruvate is converted to lactate. One advantage of non-mitochondrial glycolysis for the malignant cells is to allow the production of ATP in the absence of oxygen, albeit with a very low yield compared to oxidative phosphorylation (overall production of 2 ATPs for one glucose molecule instead of 32 ATPs). This explains why the malignant cells are often very hungry for glucose. Another consequence of the Warburg effect is to favor a flux of substrates towards biosynthetic pathways enabling the synthesis of the nucleic acids, proteins, and lipids that are necessary for tumor cell survival and proliferation. Another consequence is the increase of lactate concentration in the tumor microenvironment (TME) because lactate can freely diffuse across biological membranes. Lowering the pH in the TME seems to favor tumor invasion [42].

3.3. General Features of Extracellular Products Released by Necrotic Cells: Damage-Associated Molecular Patterns (DAMPs)

Damage-associated molecular patterns (DAMPs), also called alarmins or danger signals, are endogenous molecules released from the cells exposed to different stressors, especially following injury or cell death. After ligation to their specific receptors, they act as sensors, inducers, or mediators of stress or immune response. DAMP receptors include advanced glycosylation end product-specific receptor (RAGE/AGER), Toll-like receptors (TLRs), NOD1-like receptors (NLRs), RIG-I-like receptors (RLRs), AIM2-like receptors (ALRs), transmembrane C-type lectin receptors, P2 × 7, P2Y2, CD91, CD14, CD36, and FPR1 [43] (Table 1).
DAMPs usually have normal functions inside the cell of origin, however, after release, their function is usually altered. Production of intracellular DAMPs may increase genomic instability [44,45], epigenetic, and telomere modifications [46,47], while extracellular DAMPs induce inflammation [31,48,49] that can contribute on the long term to cancer development. DAMPs may also be involved in the metabolic re-programming towards non-mitochondrial glycolysis, which is often associated to inflammatory processes. For example, this induces metabolic switch, which contributes to the release of HMGB1 in sepsis [50]. Reciprocally, a recent study demonstrated that extra-cellular HMGB1/RAGE promote anaerobic glycolysis of fibroblasts that is required for their activation by breast cancer cells, leading to breast cancer cell metastasis [51].

3.3.1. HMGB1 and RAGE Receptor

One of the most studied DAMPs is HMGB1, which belongs to a group of non-histone nuclear proteins. HMGB1 is an evolutionary highly-conserved protein and appears to be essential for mammalian organisms: HMGB1 knock-out mice live very shortly [52]. It is a nuclear protein that acts as a chromatin-binding factor and DNA chaperone, and is responsible for numerous DNA-associated processes (replication, transcription, recombination, and repair) [53]. HMGB1 has a dual function, which depends on whether it is inside or outside the cell. Loss of intracellular HMGB1 increases DNA damage, genomic instability, cell death, and nuclear DAMP release. Contrarily, extracellular HMGB1 functions as a regulator of inflammation, immunity, metabolism, migration, and autophagy [54]. The basic mechanism for HMGB1 release is oxidative stress and it has been shown that several antioxidants may prevent or reduce its secretion [55,56,57]. However, HMGB1 release may be mediated by several other processes: post-transcriptional modifications (acetylation, ADP-ribosylation, methylation, phosphorylation, glycosylation and oxidation) [54,58,59], nuclear export receptor (chromosome-region maintenance (CRM1)) [60], pyroptosis [61], apoptosis [62], necrosis [31], and autophagy [63]. Receptors that bind HMGB1 are RAGE, TLR2, and TLR4, which in turn activate the MAPKs, NF-κB, and PI3K/AKT signaling pathways [54]. Generally, intracellular HMGB1 acts as a tumor suppressor and may enhance other tumor suppressors’ activity [64]. Extracellular HMGB1 acts as a tumor promoter by accelerating cancer development. By binding to its receptors (RAGE and TLRs), it can enhance multiple aspects of the malignant phenotype. Direct effects can be observed in vitro, for example, the enhancement of tumor sphere formation. The effects observed in vivo are remarkably diverse—metabolic changes, epithelial to mesenchymal transition (EMT), stimulation of autophagy, enhancement of immune suppression, local invasion, angiogenesis, metastasis, radio-resistance, and chemoresistance. These protumoral effects are probably due a direct impact of HMGB1 on malignant cells and to indirect mechanisms involved in inflammatory processes [65,66,67,68,69,70,71,72,73,74,75,76,77].
The receptor for advanced glycation products (RAGE) is a pattern recognition receptor (PRR) involved in the recognition of endogenous molecules released from tissue damage. It is a single transmembrane receptor that is a member of the immunoglobulin superfamily [78]. Ligands for RAGE include advanced glycation end products (AGE), members of the S100 family, extracellular HMGB1, amyloid β peptide and amyloid fibrils, β2 integrin Mac-1, glycosaminoglycans and lysophosphaditic acid [79,80,81]. Following ligand binding to RAGE, adaptor proteins (TIRAP, MyD88, diaphanous-1) associate with the RAGE cytoplasmic domain, resulting in signal transduction. The main signaling pathways activated by RAGE include Rho GTPases (cell migration), NF-κB (inflammation), and mitogen-activated protein kinases (MAPK, proliferation) [82]. A soluble form of RAGE (sRAGE) is a naturally occurring competitive inhibitor of RAGE. It originates from the receptor’s ectodomain shedding (cleaved RAGE) or splice variant (endogenous secretory RAGE) [83].

3.3.2. Other Proteins Released by Necrotic Cells

Histones

Following infection, sterile inflammation, or cell death (apoptosis, necrosis, NETosis), histones as well as nucleosomes are released from the cells. Extra-cellular histones can bind TLRs of neighboring cells (2, 4, and 9) [84]. Histone binding may activate several signaling pathways including MAPKs, NF-κB, and MyD88 [85].

S100

The S100 protein family consists of 24 low molecular weight proteins (9–13 kDa) which form homo-, hetero- and, oligomers. Intracellular S100 proteins are involved in many important cellular processes: Ca2+ homeostasis, energy metabolism, apoptosis, cell differentiation and proliferation, inflammation, migration and cytoskeletal interactions, protein phosphorylation, and degradation [86]. Extracellular S100 have been detected in the extracellular space and body fluids where they are associated with different diseases. In the TME, S100 proteins contribute to the formation of the pre-metastatic niche, neutrophil extracellular traps (NETs), and activation of the immune response [53].

Heat Shock Proteins (HSPs)

Heat shock proteins (HSPs) are conserved ubiquitously-expressed proteins that are overexpressed in the conditions of cellular stress (hyperthermia, hypoxia, changes in pH, toxins, etc.). HSPs were named according to their molecular mass and include HSP27, HSP40, HSP60, HSP70, HSP90, and large HSPs (HSP110 and glucose-regulated protein 170, GRP170) [87]. They are molecular chaperones, which means their main function is to ensure proper protein folding and activation of signaling proteins. If HSPs are dysfunctional, misfolded proteins form aggregates, leading to cell death.

Annexin A1/FPR1

Formyl peptide receptor 1 (FPR1) is a PRR that recognizes N-formylated peptides from bacteria. However, it can also serve as a receptor for DAMPs, such as annexin A1.

3.3.3. Lipids and Carbohydrates Released by Necrotic Cells

Serum amyloid A protein (SAA) is a lipoprotein involved in cholesterol transport and the production of pro-inflammatory cytokines [88]. SAA is a known ligand for TLR2 and TLR4. Hyaluronic acid (HA) is a polysaccharide that is a major component of the extracellular matrix and also an endogenous ligand for TLR2, TLR4, and, NLRP3.

3.3.4. Metabolite-Related DAMPs

ATP

Adenosine 5′-triphosphate (ATP) is a nucleotide present in all living cells, and its main role is in energy metabolism. However, in addition to its intracellular role, extracellular ATP is involved in other important biological processes, such as neurotransmission, inflammation, bone and liver glycogen metabolism, cardiac function, and vasodilatation [89].

Uric Acid

Extracellular uric acid originates from intracellular stores of uric acid and enzymatic degradation of purine nucleotides.

3.3.5. Nucleic Acids Released by Necrotic Cells

Genomic DNA in its B-form is capable of immune system activation when present in the cytosol [90]. Other forms of DNA that may induce the immune system are mitochondrial DNA [91] and single-stranded DNA (AT-rich stem-loop regions) [92]. The DNA-binding receptor is TLR9. Kariko et al. showed that mRNA is an endogenous ligand for TLR3 and that RNA released from necrotic cells may induce an inflammatory response [93]. Additionally, UV irradiation-induced the release of RNA from keratinocytes, which activates TLR3 resulting in the production of inflammatory cytokines [94].

3.4. Necrotic Products and Tumor Microenvironment

In recent years, the critical role of the tumor microenvironment (TME) in cancer initiation and progression has been recognized. Released DAMPs can crucially impact the TME by enhancing vascular stroma formation and angiogenesis, and/or by modifying the immune response (Figure 2). Secretion of HMGB1 from cancer-associated fibroblasts (CAFs) promotes metastatic potential of non-small cell lung cancer cells [95]. HMGB1 signaling between esophageal adenocarcinoma cells and macrophages in the vicinity forms an inflammatory TME, which aids cancer progression [96]. Furthermore, exosomal HMGB1 promotes cancer cell survival, protects cells from doxorubicin cytotoxicity [97] and increases angiogenesis [98]. Secreted HSP90 plays an important role in cancer cell invasion through both binding to the surface receptors, such as CD91, and interacting with matrix metalloprotease 2 on the cellular surface. HSP90 mediates invasiveness, EMT, and modulation of the immune system response [99,100,101]. Ignacio et al. revealed that SAA can predispose inflammatory TME in triple-negative breast cancer [102]. Cancer and CAFs produce SAA, which in the TME can contribute to tumor initiation, progression, metastasis, and immune suppression [103,104,105]. DNA released from neutrophils activates pancreatic stellate cells that form compact, fibrous stroma that can promote and facilitate tumor proliferation [106]. In epithelial ovarian cancer, mtDNA in the TME induces NET formation and suppressive neutrophils, therefore, facilitating metastasis and obstructing the anti-tumor immunity [107]. Extracellular DNA in the TME promotes colorectal tumor cell survival after chemotherapy through induction of autophagy via TLR-9 signaling [108]. Furthermore, many recent studies show DAMPs are released from cancer cells in extracellular vesicles, hence, enabling their dissemination to distant organs (reviewed in [109]). Nabet et al. revealed that breast cancer stromal fibroblasts shed exosomes containing RNA that, in its protein-free/unshielded form, induces RIG-I signaling in breast cancer cells, leading to tumor growth, metastasis, and therapy resistance [110].

4. Direct Impact of Necrotic Products on Malignant Cells—Role of TLR Ligands

4.1. Role of TLR Ligands, Especially TLR3 Ligands

TLR ligands are important for cancer development and progression through their ability to stimulate chronic inflammation and activate signaling pathways, leading to the upregulation of molecules involved in cell proliferation, invasion, and metastasis, or decreased apoptosis [111]. TLR3 ligands are dsRNAs often released by necrotic cancer cells. Liu et al. revealed the mechanism by which TLR3 is involved in pre-metastatic niche formation. Primary tumor releases tumor-derived exosomes, which contain small nuclear RNAs that can activate TLR3 in alveolar cells to produce chemokines and induce neutrophil infiltration [112]. Another study showed a direct link between TLR3 activation by extracellular HSP27 and angiogenesis [113]. We have previously shown that TLR3 activation in head and neck cancer cells can induce metabolic reprogramming and the Warburg effect. TLR3 stimulation induced cancer growth under low serum conditions and metabolic switch from oxidative phosphorylation to extra-mitochondrial glycolysis in a HIF-1α dependent mechanism [114]. In another study, we have demonstrated that, besides metabolic changes, TLR3 activation increases cancer cell migration, ROS production, and decreases anti-oxidative response [115]. All this evidence indicates that TLR3 activation in a tumor and its microenvironment by endogenous ligands can induce tumor survival and progression. A recent study showed that TLR3 overexpression in prostate cancer cells induces cancer invasion while its activation triggers apoptosis, confirming once again the double-edged sword nature of TLR3 [116]. Tavora et al. recently revealed that endothelial cells have a direct instructive role in driving metastatic dissemination: tumor-derived dsRNAs induce TLR3 and SLIT2, leading to intravasation [117]. Bugge et al. established that non-metastatic, healthy intestinal epithelial cells do not express TLR3, while metastatic cells do and that TLR3 promotes their invasiveness [118].

4.2. HMGB1 and RAGE

Overexpression of HMGB1 in cancer tissue and increased HMGB1 serum levels have been documented for almost all solid tumors: colon, gastric, lung, breast, ovarian, pancreatic, and prostate [54]. Hoste et al. demonstrated that TLR5 and HMGB1 are crucial in chronic inflammation, tissue damage, and skin cancer induction [119]. During hypoxia, HMGB1 may translocate to cytoplasm where it binds mtDNA and activates TLR9, resulting in hepatocellular carcinoma growth [120]. HMGB1 and its receptor TLR2 have a crucial role in mammary cancer stem cell self-renewal, tumorigenesis, and metastatic ability [121,122]. Irradiated colorectal tumor cells stimulate non-irradiated tumor cell proliferation when co-cultured through HMGB1 [123], while in bladder cancer, HMGB1 was connected with radioresistance, in vitro and in vivo, through the upregulation of autophagy [124]. Melanoma tumor cells release HMGB1 in response to hypoxia, which promotes M2-like tumor-associated macrophage accumulation and IL-10 production, leading to an increased tumor growth and metastasis [71]. Zhu et al. revealed that the redox state of HMGB1 is crucial in colorectal carcinoma angiogenesis. The all-thiol variant of HMGB1 interacting with RAGE was important for endothelial cell migration while disulfide HMGB1 binding to TLR4 was essential for VEGF up-regulation [125]. Chen et al. demonstrated how TLR2 and TLR4 have completely different roles during interactions with HMGB1: HMGB1 released from dying cells after radiotherapy contributed to stemness maintenance via interaction with TLR2, however, TLR4 antagonized this process [126]. Recent studies revealed how HMGB1 is involved in immunosuppression through galectin-9 induction [127] and chemotherapy resistance [128].
RAGE has been connected with tumorigenesis, cancer growth, and metastasis, probably through cancer-induced carbonyl stress (increased production of AGE) and hypoxia. The overexpression of RAGE and its ligands was detected in different cancer tissues [78]. AGEs derived from glucose may promote the invasion and metastasis of colorectal cancer through the RAGE/ERK/SP1/MMP2 axis [129]. RAGE is important for breast cancer cell invasion and metastasis in vitro and in vivo [130]. In melanoma cells, the interaction of extracellular S100A4 with RAGE induced pre-metastatic events, such as increased migration, invasion, and reduced adhesion [131]. RAGE was also associated with endometrial [132], prostate [133], and liver cancer progression [134]. All these findings point out the extreme importance of HMGB1 and RAGE in cancer development and dissemination, especially as many studies show HMGB1 may arise from cancer chemo- and radiotherapy-induced necrotic cell death.

4.3. Other Product and Receptors

TLR4 signaling is activated by lipopolysaccharide (LPS) originating from gram-negative bacteria. However, endogenous fatty acids can also activate TLR4, which makes this receptor a molecular link between nutrition, lipids, and inflammation [135,136]. Additionally, as pro-inflammatory cytokines production is dysregulated in obese adipose tissue, obesity may be observed and studied as an inflammatory disease caused by fatty acids serving as DAMPs molecules [137]. Iannucci et al. also showed recently that TLR4 can mediate inflammation by extracellular IFI16, a novel DAMP and TLR4 ligand, which is released from chronically inflamed tissue [138]. Several recent studies demonstrated the role of TLR4 in the creation of an immunosuppressive environment: by stimulating immunosuppressive myeloid cells [139], by small extracellular vesicles released from malignant cells [140], and by HMGB1 stimulation of TLR4 resulting in the production of immunosuppressive protein galectin-9 [127].
There are not many studies about SAA and HA serving as DAMPs in cancer regulation. It was shown that TLR4 induction by SAA3 leads to facilitated metastasis through the NF-κB signaling pathway [141]. Small fragments of the extracellular matrix component HA (sHA) also enhance the motility of cancer cells through the TLR4 signaling pathway in melanoma [142] and papillary thyroid carcinoma [143]. HA binds to TLR4, resulting in proliferation and apoptosis inhibition in colon cancer cells [144].
The S100 family of proteins have been implicated in the promotion of growth and dissemination of many different types of cancer: hepatocellular [145], colon [146], hypopharyngeal [147], prostate [148], endometrial [149], melanoma [131], breast [150], glioma [151], thyroid [152], renal [153], lung [154], and pancreas [155]. Zhuang et al. recently demonstrated that overexpression of S100A2, S100A6, S100A10, S100A11, S100A14, and S100A16 was associated with higher T-stage, advanced histologic grade, worse prognosis, and impaired immune response in pancreatic cancer [156]. S100 proteins seem to be important in the interplay between cancer cells and immune cells in the TME. Fang et al. demonstrated that S100A9 expression in monocytes stimulated the aggressiveness of co-cultured oral cancer cells [157]. Tumor-infiltrating monocytes/macrophages in the TME play an important role in promoting tumor invasion and migration by upregulating S100A8 and S100A9 expression in cancer cells [158], while a high number of S100A9-positive inflammatory cells in cancer stroma is associated with poor outcomes in prostate cancer patients [159]. RAGE and S100A7 also modulate TME by recruiting tumor-associated macrophages (TAMs) in breast cancer [160]. Jo et al. also recently observed that co-culture of normal cells with breast cancer cells induced EMT and increased proliferation, migration, and sphere formation, which was linked to S100A8/9 overexpression [161]. Moreover, Shen et al. demonstrated that transcription factor SOX9 regulates S100P expression, resulting in metastasis and invasion of colon carcinoma [162]. S100A4 stimulation was also connected with metabolic reprogramming in melanoma [163].
HSPs are often overexpressed in tumors due to the stressful conditions in the TME, leading to hypoxia, acidity, and deprivation of nutrients. Increased HSPs levels may also cause impaired apoptotic response and promote tumor growth by stabilizing proteins involved in cancer survival [164,165] as well as promote radioresistance [166] or EMT [167,168]. HSPs are also the key target proteins in novel cancer therapies (recently reviewed in [169]) since they may promote chemoresistance [170,171]. Moreover, HSP90α converts monocytes to immunosuppressive myeloid cells in melanoma through TLR4 signaling [139]. A recent study demonstrated that plasma HSP90α level can be used as a prognostic biomarker for hepatocellular carcinoma [172]. HSP90 inhibition might be a novel strategy for advanced papillary renal cell carcinoma [173], metastatic triple-negative breast cancer [174], pancreatic carcinoma [175], prostate cancer [176], and glioma [177] treatment. HSP90 inhibition also improves the survival of patients with gastrointestinal stromal tumors [178] and overcomes resistance to molecular targeted therapy in glioma [179]. HSP27 associates with EMT and stemness in several different types of cancer [180,181,182,183]. The potential of HSP27 inhibition as a target for cancer therapy has also recently been reviewed [184].
An interesting novel study by Katakam et al. revealed that DAMPs released by necrotic tumor cells promote the growth of spheroids but not 2D cultures of Ewing sarcoma. Stimulation by DAMPs of cells grown in 3D resulted in an increased expression of genes associated with cholesterol synthesis and in enhanced cellular cholesterol load. On the other hand, activation of the stimulator of interferon genes (STING) by its natural ligand cGAMP inhibited cell growth and reduced the cellular cholesterol load. This reveals a link between the innate immune response driven by STING and cholesterol homeostasis, which may have important implications for tumor growth. [185].

5. Impacts of Necrotic Products on the Other Components of the Tumor Microenvironment

5.1. Recruitment and Action of Immune Cells

DAMPs can promote cancer growth and progression by acting upon the immune system via enhanced inflammation and through immunosuppression. DAMPs released from necrotic cells act as chemo-attractants and initiate the immune response. Phagocytosis represents one of the preventive responses against released necrotic debris. Macrophages recruited to pre-necrotic zones are capable of only a limited number of cycles of clearing debris after which they downregulate their phagocytic machinery [186]. The hypoxic microenvironment also initiates sequential changes from anti-tumor M1 to pro-tumor M2 macrophage phenotype. TAMs increase in number as the tumor grows. In some tumors, TAMs contribute to a significant proportion of the tumor mass, and are associated with disease progression and poor prognosis [186,187].
Eosinophil infiltration of tumors is observed from the earliest palpable stages, with significant accumulations only in the necrotic and capsule zones of solid tumors [187]. DAMPs present in the necrotic zone induce eosinophil recruitment, their degranulation (the release of toxic cationic granule proteins), and oxidative burst (the release of ROS). Eosinophils are thus capable of inducing an oxidative environment and have a role in the inactivation and clearance of necrotic debris [188,189].

5.2. Deleterious Effects of Neutrophils

Immune responses induced by HMGB1 strongly depend on its oxidation status. Necrotic cells release a fully active, reduced form of HMGB1. That form, by binding on its receptors, induces the release of proinflammatory cytokines, creating an inflammatory microenvironment. HMGB1 also promotes the recruitment of inflammatory cells, preferentially neutrophils [190]. Neutrophil infiltration is observed in pre-clinical and clinical cancer models and contributes to tumor cell proliferation and metastasis. The high levels of neutrophil infiltrate, due to the release of high levels of ROS and cytotoxic compounds, promote tumor necrosis, sustain chronic inflammation, and negatively correlate with prognosis and survival [190,191]. Simultaneously with the infiltration of neutrophils, cytokines released from tumor cells cause pathologically-enhanced hematopoiesis, skewed from lymphocytic to granulocytic, that produces more neutrophils [189]. A high neutrophil-to-lymphocyte ratio in the peripheral blood is shown to be associated with poor outcomes in many solid tumors [191]. Overproduction in bone marrow gives rise to immature neutrophils that lack cytotoxic granules and are immunosuppressive. New cohorts of immature cells represent a heterogeneous population of immature myeloid-derived suppressor cells (MDSCs) that promote tumor growth and suppress other effector cells (cytotoxic T lymphocytes and NK cells) [190].

5.3. Immunosuppressive Effects of Necrotic Products

Extracellular adenosine is a potent immunosuppressive metabolite. Its level is normally low, however, in response to hypoxic stimulation and inflammation, adenosine level can be induced over a hundred fold [192]. Adenosine, through its receptor A2aR, affects T cells, Treg cells [193], NK cells, and myeloid-derived suppressor cells, leading to an immunosuppressive effect. Blockade of A2aR enhanced NK cell maturation and cytotoxic function, reduced metastasis [194], and increased the number of tumor-infiltrating cytotoxic lymphocytes and decreased Treg cells [195]. ATP was also shown to be involved in immunosuppression: MDSCs from tumor-bearing mice express P2X7 receptor, which promotes the release of immunosuppressive cytokines after triggering with ATP [196]. Baghdadi et al. showed that DAMPs released from chemotherapy-damaged tumor cells upregulate T cell immunoglobulin and mucin domain-containing molecule-4 (TIM-4) on tumor-associated myeloid cells, leading to the repression of tumor-specific immunity [197]. HMGB1 may suppress the antitumor immune response by interacting with TIM-3 [198] and through the promotion of Treg cells survival while limiting the functional activity of conventional T cells [199]. HMGB1 may also act through RAGE and affect pDCs to induce a tolerogenic response during cervical/vulvar carcinogenesis [200]. A recent study showed that HMGB1 derived from hepatocellular carcinoma triggers M2 macrophage polarization through TLR2 and autophagy [201]. Exosome-derived HMGB1 may activate B cells and promote TIM-1+Breg cell expansion via the TLR2/4 and MAPK signaling pathways, therefore, generating an immunosuppressive milieu [202]. HMGB1 also mediates immune suppression through the upregulation of PD-L1 [203].
S100A8/A9 induces the accumulation of MDSCs and is secreted by MDSCs and tumor cells, which forms a positive autocrine feedback loop within the inflammatory tumor environment [204]. Similarly, Cheng et al. demonstrated that S100A9, whose expression is regulated by STAT3, is crucial for the inhibition of DC differentiation and the stimulation of the accumulation of MDSCs in cancer [205]. Moreover, overexpression of S100A6, S100A10, S100A11, S100A14, and S100A16 may impair the infiltration and cytolytic activity of cytotoxic lymphocytes in pancreatic cancer [156]. HP70 and HSP90α both induce the immunosuppressive effect of MDSCs [139,206].
Recent findings demonstrated that DAMPs are able to induce NET formation [207]. A study by Munir et al. provided insight into a novel mechanism by which CAFs stimulate t-NETosis at local and systemic levels via the production of amyloid β that serves as DAMP [208]. Several studies have shown that different kinds of traumas induce DAMPs in patients’ plasma resulting in immune suppression [209,210].
Contrary to all this, DAMPs also act as a double-edged sword by inhibiting cancer progression via immunogenic cell death (ICD). Important effectors of ICD are calreticulin, ATP, and HMGB1 [211,212,213]. This has recently been reviewed by [214].

6. Conclusions

DAMPs are produced and released in the TME by cells dying from spontaneous necrosis mainly linked to nutrients/oxygen deprivation. They can also be released by cells dying from the action of therapeutic agents during chemotherapy or radiotherapy. DAMPs can bind different receptors, including TLRs, either on cancer cells, immune cells or other cells in the TME, such as CAFs or other stromal cells. Since TLR agonists, and especially TLR3 agonists because of their ability to induce apoptosis, are already being used in various clinical studies, either in form of anticancer drugs or as immunoadjuvants, their potential detrimental role must again be emphasized. Further studies are needed in order to reveal which signaling pathways induce cell death and what are those pathways or conditions that promote tumorigenesis before the introduction of TLR ligands into clinical practice.

Author Contributions

Conceptualization, P.B.; writing—original draft preparation, E.Z., T.V. and T.M.G.; writing—review and editing, T.M.G. and P.B.; supervision, T.M.G. and P.B.; funding acquisition, T.M.G. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by Croatian Science Foundation (project number: IP-2020-02-4225 Toll-like receptor 3 in the development and treatment of human head and neck cancer: the role of endogenous ligands) and Young Researchers’ career development project- training of doctoral students (Croatian Science foundation).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Kumar, V.; Abbas, A.K.; Aster, J.C. Robbins and Cotran Pathologic Basis of Disease, 9th ed.; Elsevier/Saunders: Philadelphia, PA, USA, 2015; pp. 31–68. [Google Scholar]
  2. Wimmer, K.; Sachet, M.; Oehler, R. Circulating biomarkers of cell death. Clin. Chim. Acta 2020, 500, 87–97. [Google Scholar] [CrossRef] [PubMed]
  3. Hu, X.M.; Li, Z.X.; Lin, R.H.; Shan, J.Q.; Yu, Q.W.; Wang, R.X.; Liao, L.S.; Yan, W.T.; Wang, Z.; Shang, L.; et al. Guidelines for Regulated Cell Death Assays: A Systematic Summary, A Categorical Comparison, A Prospective. Front. Cell Dev. Biol. 2021, 9, 634690. [Google Scholar] [CrossRef]
  4. Proskuryakov, S.Y.; Gabai, V.L. Mechanisms of tumor cell necrosis. Curr. Pharm. Des. 2010, 16, 56–68. [Google Scholar] [CrossRef] [PubMed]
  5. Lee, S.Y.; Ju, M.K.; Jeon, H.M.; Jeong, E.K.; Lee, Y.J.; Kim, C.H.; Park, H.G.; Han, S.I.; Kang, H.S. Regulation of Tumor Progression by Programmed Necrosis. Oxid. Med. Cell Longev. 2018, 2018, 3537471. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Brown, J.M. Tumor microenvironment and the response to anticancer therapy. Cancer Biol. Ther. 2002, 1, 453–458. [Google Scholar] [CrossRef] [Green Version]
  7. Khatun, S.; Appidi, T.; Rengan, A.K. The role played by bacterial infections in the onset and metastasis of cancer. Curr. Res. Microb. Sci. 2021, 2, 100078. [Google Scholar] [CrossRef]
  8. Cummins, J.; Tangney, M. Bacteria and tumours: Causative agents or opportunistic inhabitants? Infect. Agent Cancer 2013, 8, 11. [Google Scholar] [CrossRef] [Green Version]
  9. Karsch-Bluman, A.; Feiglin, A.; Arbib, E.; Stern, T.; Shoval, H.; Schwob, O.; Berger, M.; Benny, O. Tissue necrosis and its role in cancer progression. Oncogene 2019, 38, 1920–1935. [Google Scholar] [CrossRef]
  10. Richards, C.H.; Mohammed, Z.; Qayyum, T.; Horgan, P.G.; McMillan, D.C. The prognostic value of histological tumor necrosis in solid organ malignant disease: A systematic review. Future Oncol. 2011, 7, 1223–1235. [Google Scholar] [CrossRef]
  11. Gkogkou, C.; Frangia, K.; Saif, M.W.; Trigidou, R.; Syrigos, K. Necrosis and apoptotic index as prognostic factors in non-small cell lung carcinoma: A review. Springerplus 2014, 3, 120. [Google Scholar] [CrossRef] [Green Version]
  12. Ling, Y.H.; Chen, J.W.; Wen, S.H.; Huang, C.Y.; Li, P.; Lu, L.H.; Mei, J.; Li, S.H.; Wei, W.; Cai, M.Y.; et al. Tumor necrosis as a poor prognostic predictor on postoperative survival of patients with solitary small hepatocellular carcinoma. BMC Cancer 2020, 20, 607. [Google Scholar] [CrossRef] [PubMed]
  13. Friebele, J.C.; Peck, J.; Pan, X.; Abdel-Rasoul, M.; Mayerson, J.L. Osteosarcoma: A Meta-Analysis and Review of the Literature. Am. J. Orthop. 2015, 44, 547–553. [Google Scholar]
  14. Choudhury, A.; West, C.M.; Porta, N.; Hall, E.; Denley, H.; Hendron, C.; Lewis, R.; Hussain, S.A.; Huddart, R.; James, N. The predictive and prognostic value of tumour necrosis in muscle invasive bladder cancer patients receiving radiotherapy with or without chemotherapy in the BC2001 trial (CRUK/01/004). Br. J. Cancer 2017, 116, 649–657. [Google Scholar] [CrossRef] [Green Version]
  15. Nael, K.; Bauer, A.H.; Hormigo, A.; Lemole, M.; Germano, I.M.; Puig, J.; Stea, B. Multiparametric MRI for Differentiation of Radiation Necrosis From Recurrent Tumor in Patients With Treated Glioblastoma. AJR Am. J. Roentgenol. 2018, 210, 18–23. [Google Scholar] [CrossRef] [PubMed]
  16. Beddy, P.; Genega, E.M.; Ngo, L.; Hindman, N.; Wei, J.; Bullock, A.; Bhatt, R.S.; Atkins, M.B.; Pedrosa, I. Tumor necrosis on magnetic resonance imaging correlates with aggressive histology and disease progression in clear cell renal cell carcinoma. Clin. Genitourin. Cancer 2014, 12, 55–62. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Nowosielski, M.; Gorlia, T.; Bromberg, J.E.C.; Sahm, F.; Harting, I.; Kickingereder, P.; Brandes, A.A.; Taphoorn, M.J.B.; Taal, W.; Domont, J.; et al. Imaging necrosis during treatment is associated with worse survival in EORTC 26101 study. Neurology 2019, 92, e2754–e2763. [Google Scholar] [CrossRef] [PubMed]
  18. Verma, N.; Cowperthwaite, M.C.; Burnett, M.G.; Markey, M.K. Differentiating tumor recurrence from treatment necrosis: A review of neuro-oncologic imaging strategies. Neuro Oncol. 2013, 15, 515–534. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Strauss, S.B.; Meng, A.; Ebani, E.J.; Chiang, G.C. Imaging Glioblastoma Posttreatment: Progression, Pseudoprogression, Pseudoresponse, Radiation Necrosis. Neuroimaging Clin. N. Am. 2021, 31, 103–120. [Google Scholar] [CrossRef]
  20. Zhang, D.; Gao, M.; Jin, Q.; Ni, Y.; Zhang, J. Updated developments on molecular imaging and therapeutic strategies directed against necrosis. Acta Pharm. Sin. B 2019, 9, 455–468. [Google Scholar] [CrossRef]
  21. Stroet, M.C.M.; de Blois, E.; Haeck, J.; Seimbille, Y.; Mezzanotte, L.; de Jong, M.; Lowik, C.; Panth, K.M. In Vivo Evaluation of Gallium-68-Labeled IRDye800CW as a Necrosis Avid Contrast Agent in Solid Tumors. Contrast Media Mol. Imaging 2021, 2021, 2853522. [Google Scholar] [CrossRef]
  22. Zhang, D.; Jin, Q.; Ni, Y.; Zhang, J. Discovery of necrosis avidity of rhein and its applications in necrosis imaging. J. Drug Target 2020, 28, 904–912. [Google Scholar] [CrossRef] [PubMed]
  23. Luo, Q.; Jin, Q.; Su, C.; Zhang, D.; Jiang, C.; Fish, A.F.; Feng, Y.; Ni, Y.; Zhang, J.; Yin, Z. Radiolabeled Rhein as Small-Molecule Necrosis Avid Agents for Imaging of Necrotic Myocardium. Anal. Chem. 2017, 89, 1260–1266. [Google Scholar] [CrossRef] [PubMed]
  24. Wu, T.; Zhang, J.; Jin, Q.; Gao, M.; Zhang, D.; Zhang, L.; Feng, Y.; Ni, Y.; Yin, Z. Rhein-based necrosis-avid MRI contrast agents for early evaluation of tumor response to microwave ablation therapy. Magn. Reason. Med. 2019, 82, 2212–2224. [Google Scholar] [CrossRef] [PubMed]
  25. Bian, L.; Gao, M.; Zhang, D.; Ji, A.; Su, C.; Duan, X.; Luo, Q.; Huang, D.; Feng, Y.; Ni, Y.; et al. Synthesis and Biological Evaluation of Rhein-Based MRI Contrast Agents for in Vivo Visualization of Necrosis. Anal. Chem. 2018, 90, 13249–13256. [Google Scholar] [CrossRef]
  26. Zhang, A.; Wu, T.; Bian, L.; Li, P.; Liu, Q.; Zhang, D.; Jin, Q.; Zhang, J.; Huang, G.; Song, S. Synthesis and Evaluation of Ga-68-Labeled Rhein for Early Assessment of Treatment-Induced Tumor Necrosis. Mol. Imaging Biol. 2020, 22, 515–525. [Google Scholar] [CrossRef] [PubMed]
  27. Kang, Y.; Zhai, X.; Lu, S.; Vuletic, I.; Wang, L.; Zhou, K.; Peng, Z.; Ren, Q.; Xie, Z. A Hybrid Imaging Platform(CT/PET/FMI) for Evaluating Tumor Necrosis and Apoptosis in Real-Time. Front. Oncol. 2022, 12, 772392. [Google Scholar] [CrossRef]
  28. Tonnus, W.; Meyer, C.; Paliege, A.; Belavgeni, A.; von Massenhausen, A.; Bornstein, S.R.; Hugo, C.; Becker, J.U.; Linkermann, A. The pathological features of regulated necrosis. J. Pathol. 2019, 247, 697–707. [Google Scholar] [CrossRef]
  29. Chung, A.G.; Frye, J.B.; Zbesko, J.C.; Constantopoulos, E.; Hayes, M.; Figueroa, A.G.; Becktel, D.A.; Antony Day, W.; Konhilas, J.P.; McKay, B.S.; et al. Liquefaction of the Brain following Stroke Shares a Similar Molecular and Morphological Profile with Atherosclerosis and Mediates Secondary Neurodegeneration in an Osteopontin-Dependent Mechanism. eNeuro 2018, 5, ENEURO.0076-18.2018. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Chan, F.K.; Moriwaki, K.; De Rosa, M.J. Detection of necrosis by release of lactate dehydrogenase activity. Methods Mol. Biol. 2013, 979, 65–70. [Google Scholar]
  31. Scaffidi, P.; Misteli, T.; Bianchi, M.E. Release of chromatin protein HMGB1 by necrotic cells triggers inflammation. Nature 2002, 418, 191–195. [Google Scholar] [CrossRef]
  32. Omary, M.B.; Ku, N.O.; Strnad, P.; Hanada, S. Toward unraveling the complexity of simple epithelial keratins in human disease. J. Clin. Investig. 2009, 119, 1794–1805. [Google Scholar] [CrossRef] [Green Version]
  33. Kramer, G.; Erdal, H.; Mertens, H.J.; Nap, M.; Mauermann, J.; Steiner, G.; Marberger, M.; Biven, K.; Shoshan, M.C.; Linder, S. Differentiation between cell death modes using measurements of different soluble forms of extracellular cytokeratin 18. Cancer Res. 2004, 64, 1751–1756. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Ulz, P.; Thallinger, G.G.; Auer, M.; Graf, R.; Kashofer, K.; Jahn, S.W.; Abete, L.; Pristauz, G.; Petru, E.; Geigl, J.B.; et al. Inferring expressed genes by whole-genome sequencing of plasma DNA. Nat. Genet. 2016, 48, 1273–1278. [Google Scholar] [CrossRef]
  35. Rostami, A.; Lambie, M.; Yu, C.W.; Stambolic, V.; Waldron, J.N.; Bratman, S.V. Senescence, Necrosis, and Apoptosis Govern Circulating Cell-free DNA Release Kinetics. Cell Rep. 2020, 31, 107830. [Google Scholar] [CrossRef]
  36. Jiang, P.; Chan, C.W.; Chan, K.C.; Cheng, S.H.; Wong, J.; Wong, V.W.; Wong, G.L.; Chan, S.L.; Mok, T.S.; Chan, H.L.; et al. Lengthening and shortening of plasma DNA in hepatocellular carcinoma patients. Proc. Natl. Acad. Sci. USA 2015, 112, E1317–E1325. [Google Scholar] [CrossRef] [Green Version]
  37. McCall, M.N.; Kim, M.S.; Adil, M.; Patil, A.H.; Lu, Y.; Mitchell, C.J.; Leal-Rojas, P.; Xu, J.; Kumar, M.; Dawson, V.L.; et al. Toward the human cellular microRNAome. Genome Res. 2017, 27, 1769–1781. [Google Scholar] [CrossRef] [Green Version]
  38. Galluzzi, L.; Vitale, I.; Aaronson, S.A.; Abrams, J.M.; Adam, D.; Agostinis, P.; Alnemri, E.S.; Altucci, L.; Amelio, I.; Andrews, D.W.; et al. Molecular mechanisms of cell death: Recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ. 2018, 25, 486–541. [Google Scholar] [CrossRef] [PubMed]
  39. Vanden Berghe, T.; Linkermann, A.; Jouan-Lanhouet, S.; Walczak, H.; Vandenabeele, P. Regulated necrosis: The expanding network of non-apoptotic cell death pathways. Nat. Rev. Mol. Cell Biol. 2014, 15, 135–147. [Google Scholar] [CrossRef] [PubMed]
  40. Khoury, M.K.; Gupta, K.; Franco, S.R.; Liu, B. Necroptosis in the Pathophysiology of Disease. Am. J. Pathol. 2020, 190, 272–285. [Google Scholar] [CrossRef] [Green Version]
  41. Dhuriya, Y.K.; Sharma, D. Necroptosis: A regulated inflammatory mode of cell death. J. Neuroinflamm. 2018, 15, 199. [Google Scholar] [CrossRef] [Green Version]
  42. Vander Heiden, M.G.; Cantley, L.C.; Thompson, C.B. Understanding the Warburg effect: The metabolic requirements of cell proliferation. Science 2009, 324, 1029–1033. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Huang, J.; Xie, Y.; Sun, X.; Zeh, H.J., 3rd; Kang, R.; Lotze, M.T.; Tang, D. DAMPs, ageing, and cancer: The ‘DAMP Hypothesis’. Ageing Res. Rev. 2015, 24 Pt A, 3–16. [Google Scholar] [CrossRef] [Green Version]
  44. Kang, R.; Zhang, Q.; Hou, W.; Yan, Z.; Chen, R.; Bonaroti, J.; Bansal, P.; Billiar, T.R.; Tsung, A.; Wang, Q.; et al. Intracellular Hmgb1 inhibits inflammatory nucleosome release and limits acute pancreatitis in mice. Gastroenterology 2014, 146, 1097–1107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Liu, Y.; Prasad, R.; Wilson, S.H. HMGB1: Roles in base excision repair and related function. Biochim. Biophys. Acta 2010, 1799, 119–130. [Google Scholar] [CrossRef] [Green Version]
  46. Polanska, E.; Dobsakova, Z.; Dvorackova, M.; Fajkus, J.; Stros, M. HMGB1 gene knockout in mouse embryonic fibroblasts results in reduced telomerase activity and telomere dysfunction. Chromosoma 2012, 121, 419–431. [Google Scholar] [CrossRef] [PubMed]
  47. Hu, Z.; Chen, K.; Xia, Z.; Chavez, M.; Pal, S.; Seol, J.H.; Chen, C.C.; Li, W.; Tyler, J.K. Nucleosome loss leads to global transcriptional up-regulation and genomic instability during yeast aging. Genes Dev. 2014, 28, 396–408. [Google Scholar] [CrossRef] [Green Version]
  48. Gebhardt, C.; Riehl, A.; Durchdewald, M.; Nemeth, J.; Furstenberger, G.; Muller-Decker, K.; Enk, A.; Arnold, B.; Bierhaus, A.; Nawroth, P.P.; et al. RAGE signaling sustains inflammation and promotes tumor development. J. Exp. Med. 2008, 205, 275–285. [Google Scholar] [CrossRef] [Green Version]
  49. Bettum, I.J.; Vasiliauskaite, K.; Nygaard, V.; Clancy, T.; Pettersen, S.J.; Tenstad, E.; Maelandsmo, G.M.; Prasmickaite, L. Metastasis-associated protein S100A4 induces a network of inflammatory cytokines that activate stromal cells to acquire pro-tumorigenic properties. Cancer Lett. 2014, 344, 28–39. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Yang, L.; Xie, M.; Yang, M.; Yu, Y.; Zhu, S.; Hou, W.; Kang, R.; Lotze, M.T.; Billiar, T.R.; Wang, H.; et al. PKM2 regulates the Warburg effect and promotes HMGB1 release in sepsis. Nat. Commun. 2014, 5, 4436. [Google Scholar] [CrossRef] [Green Version]
  51. Chen, Y.; Cai, L.; Guo, X.; Li, Z.; Liao, X.; Zhang, X.; Huang, L.; He, J. HMGB1-activated fibroblasts promote breast cancer cells metastasis via RAGE/aerobic glycolysis. Neoplasma 2021, 68, 71–78. [Google Scholar] [CrossRef]
  52. Calogero, S.; Grassi, F.; Aguzzi, A.; Voigtlander, T.; Ferrier, P.; Ferrari, S.; Bianchi, M.E. The lack of chromosomal protein Hmg1 does not disrupt cell growth but causes lethal hypoglycaemia in newborn mice. Nat. Genet. 1999, 22, 276–280. [Google Scholar] [CrossRef] [PubMed]
  53. Bertheloot, D.; Latz, E. HMGB1, IL-1alpha, IL-33 and S100 proteins: Dual-function alarmins. Cell Mol. Immunol. 2017, 14, 43–64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Kang, R.; Zhang, Q.; Zeh, H.J., 3rd; Lotze, M.T.; Tang, D. HMGB1 in cancer: Good, bad, or both? Clin. Cancer Res. 2013, 19, 4046–4057. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Tang, D.; Shi, Y.; Kang, R.; Li, T.; Xiao, W.; Wang, H.; Xiao, X. Hydrogen peroxide stimulates macrophages and monocytes to actively release HMGB1. J. Leukoc. Biol. 2007, 81, 741–747. [Google Scholar] [CrossRef] [Green Version]
  56. Tang, D.; Kang, R.; Xiao, W.; Zhang, H.; Lotze, M.T.; Wang, H.; Xiao, X. Quercetin prevents LPS-induced high-mobility group box 1 release and proinflammatory function. Am. J. Respir. Cell Mol. Biol. 2009, 41, 651–660. [Google Scholar] [CrossRef] [Green Version]
  57. Xu, W.; Lu, Y.; Yao, J.; Li, Z.; Chen, Z.; Wang, G.; Jing, H.; Zhang, X.; Li, M.; Peng, J.; et al. Novel role of resveratrol: Suppression of high-mobility group protein box 1 nucleocytoplasmic translocation by the upregulation of sirtuin 1 in sepsis-induced liver injury. Shock 2014, 42, 440–447. [Google Scholar] [CrossRef]
  58. Kim, Y.H.; Kwak, M.S.; Park, J.B.; Lee, S.A.; Choi, J.E.; Cho, H.S.; Shin, J.S. N-linked glycosylation plays a crucial role in the secretion of HMGB1. J. Cell Sci. 2016, 129, 29–38. [Google Scholar]
  59. Ito, I.; Fukazawa, J.; Yoshida, M. Post-translational methylation of high mobility group box 1 (HMGB1) causes its cytoplasmic localization in neutrophils. J. Biol. Chem. 2007, 282, 16336–16344. [Google Scholar] [CrossRef] [Green Version]
  60. Bonaldi, T.; Talamo, F.; Scaffidi, P.; Ferrera, D.; Porto, A.; Bachi, A.; Rubartelli, A.; Agresti, A.; Bianchi, M.E. Monocytic cells hyperacetylate chromatin protein HMGB1 to redirect it towards secretion. EMBO J. 2003, 22, 5551–5560. [Google Scholar] [CrossRef] [Green Version]
  61. Nystrom, S.; Antoine, D.J.; Lundback, P.; Lock, J.G.; Nita, A.F.; Hogstrand, K.; Grandien, A.; Erlandsson-Harris, H.; Andersson, U.; Applequist, S.E. TLR activation regulates damage-associated molecular pattern isoforms released during pyroptosis. EMBO J. 2013, 32, 86–99. [Google Scholar] [CrossRef] [Green Version]
  62. Bell, C.W.; Jiang, W.; Reich, C.F., 3rd; Pisetsky, D.S. The extracellular release of HMGB1 during apoptotic cell death. Am. J. Physiol. Cell Physiol. 2006, 291, C1318–C1325. [Google Scholar] [CrossRef] [PubMed]
  63. Tang, D.; Kang, R.; Livesey, K.M.; Cheh, C.W.; Farkas, A.; Loughran, P.; Hoppe, G.; Bianchi, M.E.; Tracey, K.J.; Zeh, H.J., 3rd; et al. Endogenous HMGB1 regulates autophagy. J. Cell Biol. 2010, 190, 881–892. [Google Scholar] [CrossRef]
  64. Jiao, Y.; Wang, H.C.; Fan, S.J. Growth suppression and radiosensitivity increase by HMGB1 in breast cancer. Acta Pharmacol. Sin. 2007, 28, 1957–1967. [Google Scholar] [CrossRef] [Green Version]
  65. Yang, H.; Pellegrini, L.; Napolitano, A.; Giorgi, C.; Jube, S.; Preti, A.; Jennings, C.J.; De Marchis, F.; Flores, E.G.; Larson, D.; et al. Aspirin delays mesothelioma growth by inhibiting HMGB1-mediated tumor progression. Cell Death Dis. 2015, 6, e1786. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Kang, R.; Tang, D.; Schapiro, N.E.; Loux, T.; Livesey, K.M.; Billiar, T.R.; Wang, H.; Van Houten, B.; Lotze, M.T.; Zeh, H.J. The HMGB1/RAGE inflammatory pathway promotes pancreatic tumor growth by regulating mitochondrial bioenergetics. Oncogene 2014, 33, 567–577. [Google Scholar] [CrossRef] [Green Version]
  67. Parker, K.H.; Sinha, P.; Horn, L.A.; Clements, V.K.; Yang, H.; Li, J.; Tracey, K.J.; Ostrand-Rosenberg, S. HMGB1 enhances immune suppression by facilitating the differentiation and suppressive activity of myeloid-derived suppressor cells. Cancer Res. 2014, 74, 5723–5733. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Abe, A.; Kuwata, T.; Yamauchi, C.; Higuchi, Y.; Ochiai, A. High Mobility Group Box1 (HMGB1) released from cancer cells induces the expression of pro-inflammatory cytokines in peritoneal fibroblasts. Pathol. Int. 2014, 64, 267–275. [Google Scholar] [CrossRef] [PubMed]
  69. Yan, H.X.; Wu, H.P.; Zhang, H.L.; Ashton, C.; Tong, C.; Wu, H.; Qian, Q.J.; Wang, H.Y.; Ying, Q.L. p53 promotes inflammation-associated hepatocarcinogenesis by inducing HMGB1 release. J. Hepatol. 2013, 59, 762–768. [Google Scholar] [CrossRef] [Green Version]
  70. Yang, S.; Xu, L.; Yang, T.; Wang, F. High-mobility group box-1 and its role in angiogenesis. J. Leukoc. Biol. 2014, 95, 563–574. [Google Scholar] [CrossRef]
  71. Huber, R.; Meier, B.; Otsuka, A.; Fenini, G.; Satoh, T.; Gehrke, S.; Widmer, D.; Levesque, M.P.; Mangana, J.; Kerl, K.; et al. Tumour hypoxia promotes melanoma growth and metastasis via High Mobility Group Box-1 and M2-like macrophages. Sci. Rep. 2016, 6, 29914. [Google Scholar] [CrossRef] [Green Version]
  72. Dong, J.; Zhang, X.; Du, X.; Zou, N.; Shen, W.; Ma, M.; Wang, Y.; Zhu, S. HMGB1 overexpression promotes a malignant phenotype and radioresistance in ESCC. J. Cancer 2022, 13, 2717–2726. [Google Scholar] [CrossRef] [PubMed]
  73. Amornsupak, K.; Thongchot, S.; Thinyakul, C.; Box, C.; Hedayat, S.; Thuwajit, P.; Eccles, S.A.; Thuwajit, C. HMGB1 mediates invasion and PD-L1 expression through RAGE-PI3K/AKT signaling pathway in MDA-MB-231 breast cancer cells. BMC Cancer 2022, 22, 578. [Google Scholar] [CrossRef] [PubMed]
  74. Jung, A.R.; Kim, G.E.; Kim, M.Y.; Ha, U.S.; Hong, S.H.; Lee, J.Y.; Kim, S.W.; Park, Y.H. HMGB1 promotes tumor progression and invasion through HMGB1/TNFR1/NF-kappaB axis in castration-resistant prostate cancer. Am. J. Cancer Res. 2021, 11, 2215–2227. [Google Scholar] [PubMed]
  75. Song, Y.; Zou, X.; Zhang, D.; Liu, S.; Duan, Z.; Liu, L. Self-enforcing HMGB1/NF-kappaB/HIF-1alpha Feedback Loop Promotes Cisplatin Resistance in Hepatocellular Carcinoma Cells. J. Cancer 2020, 11, 3893–3902. [Google Scholar] [CrossRef]
  76. Ma, H.; Zheng, S.; Zhang, X.; Gong, T.; Lv, X.; Fu, S.; Zhang, S.; Yin, X.; Hao, J.; Shan, C.; et al. High mobility group box 1 promotes radioresistance in esophageal squamous cell carcinoma cell lines by modulating autophagy. Cell Death Dis. 2019, 10, 136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Li, J.; Ren, H.; Wang, J.; Zhang, P.; Shi, X. Extracellular HMGB1 promotes CD44 expression in hepatocellular carcinoma via regulating miR-21. Aging 2021, 13, 8380–8395. [Google Scholar] [CrossRef] [PubMed]
  78. Tesarova, P.; Cabinakova, M.; Mikulova, V.; Zima, T.; Kalousova, M. RAGE and its ligands in cancer-culprits, biomarkers, or therapeutic targets? Neoplasma 2015, 62, 353–364. [Google Scholar] [CrossRef] [Green Version]
  79. Fritz, G. RAGE: A single receptor fits multiple ligands. Trends Biochem. Sci. 2011, 36, 625–632. [Google Scholar] [CrossRef]
  80. Mizumoto, S.; Takahashi, J.; Sugahara, K. Receptor for advanced glycation end products (RAGE) functions as receptor for specific sulfated glycosaminoglycans, and anti-RAGE antibody or sulfated glycosaminoglycans delivered in vivo inhibit pulmonary metastasis of tumor cells. J. Biol. Chem. 2012, 287, 18985–18994. [Google Scholar] [CrossRef] [Green Version]
  81. Rai, V.; Toure, F.; Chitayat, S.; Pei, R.; Song, F.; Li, Q.; Zhang, J.; Rosario, R.; Ramasamy, R.; Chazin, W.J.; et al. Lysophosphatidic acid targets vascular and oncogenic pathways via RAGE signaling. J. Exp. Med. 2012, 209, 2339–2350. [Google Scholar] [CrossRef] [Green Version]
  82. Xie, J.; Mendez, J.D.; Mendez-Valenzuela, V.; Aguilar-Hernandez, M.M. Cellular signalling of the receptor for advanced glycation end products (RAGE). Cell. Signal 2013, 25, 2185–2197. [Google Scholar] [CrossRef] [PubMed]
  83. Santilli, F.; Vazzana, N.; Bucciarelli, L.G.; Davi, G. Soluble forms of RAGE in human diseases: Clinical and therapeutical implications. Curr. Med. Chem. 2009, 16, 940–952. [Google Scholar] [CrossRef] [PubMed]
  84. Marsman, G.; Zeerleder, S.; Luken, B.M. Extracellular histones, cell-free DNA, or nucleosomes: Differences in immunostimulation. Cell Death Dis. 2016, 7, e2518. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Chen, R.; Kang, R.; Fan, X.G.; Tang, D. Release and activity of histone in diseases. Cell Death Dis. 2014, 5, e1370. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Donato, R.; Cannon, B.R.; Sorci, G.; Riuzzi, F.; Hsu, K.; Weber, D.J.; Geczy, C.L. Functions of S100 proteins. Curr. Mol. Med. 2013, 13, 24–57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Wu, J.; Liu, T.; Rios, Z.; Mei, Q.; Lin, X.; Cao, S. Heat Shock Proteins and Cancer. Trends Pharmacol. Sci. 2017, 38, 226–256. [Google Scholar] [CrossRef]
  88. He, R.; Sang, H.; Ye, R.D. Serum amyloid A induces IL-8 secretion through a G protein-coupled receptor, FPRL1/LXA4R. Blood 2003, 101, 1572–1581. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Bours, M.J.; Swennen, E.L.; Di Virgilio, F.; Cronstein, B.N.; Dagnelie, P.C. Adenosine 5′-triphosphate and adenosine as endogenous signaling molecules in immunity and inflammation. Pharmacol. Ther. 2006, 112, 358–404. [Google Scholar] [CrossRef] [PubMed]
  90. Ishii, K.J.; Coban, C.; Kato, H.; Takahashi, K.; Torii, Y.; Takeshita, F.; Ludwig, H.; Sutter, G.; Suzuki, K.; Hemmi, H.; et al. A Toll-like receptor-independent antiviral response induced by double-stranded B-form DNA. Nat. Immunol. 2006, 7, 40–48. [Google Scholar] [CrossRef]
  91. Mathew, A.; Lindsley, T.A.; Sheridan, A.; Bhoiwala, D.L.; Hushmendy, S.F.; Yager, E.J.; Ruggiero, E.A.; Crawford, D.R. Degraded mitochondrial DNA is a newly identified subtype of the damage associated molecular pattern (DAMP) family and possible trigger of neurodegeneration. J. Alzheimers Dis. 2012, 30, 617–627. [Google Scholar] [CrossRef]
  92. Sharma, S.; DeOliveira, R.B.; Kalantari, P.; Parroche, P.; Goutagny, N.; Jiang, Z.; Chan, J.; Bartholomeu, D.C.; Lauw, F.; Hall, J.P.; et al. Innate immune recognition of an AT-rich stem-loop DNA motif in the Plasmodium falciparum genome. Immunity 2011, 35, 194–207. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Kariko, K.; Ni, H.; Capodici, J.; Lamphier, M.; Weissman, D. mRNA is an endogenous ligand for Toll-like receptor 3. J. Biol. Chem. 2004, 279, 12542–12550. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Bernard, J.J.; Cowing-Zitron, C.; Nakatsuji, T.; Muehleisen, B.; Muto, J.; Borkowski, A.W.; Martinez, L.; Greidinger, E.L.; Yu, B.D.; Gallo, R.L. Ultraviolet radiation damages self noncoding RNA and is detected by TLR3. Nat. Med. 2012, 18, 1286–1290. [Google Scholar] [CrossRef]
  95. Ren, Y.; Cao, L.; Wang, L.; Zheng, S.; Zhang, Q.; Guo, X.; Li, X.; Chen, M.; Wu, X.; Furlong, F.; et al. Autophagic secretion of HMGB1 from cancer-associated fibroblasts promotes metastatic potential of non-small cell lung cancer cells via NFkappaB signaling. Cell Death Dis. 2021, 12, 858. [Google Scholar] [CrossRef] [PubMed]
  96. Flis, E.; Barber, G.; Nulty, C.; Keogh, B.; McGuirk, P.; Anand, A.; O’Sullivan, J.; Quante, M.; Creagh, E.M. Identification of TLR2 Signalling Mechanisms Which Contribute to Barrett’s and Oesophageal Adenocarcinoma Disease Progression. Cancers 2021, 13, 2065. [Google Scholar] [CrossRef] [PubMed]
  97. Wang, J.D.; Wang, Y.Y.; Lin, S.Y.; Chang, C.Y.; Li, J.R.; Huang, S.W.; Chen, W.Y.; Liao, S.L.; Chen, C.J. Exosomal HMGB1 Promoted Cancer Malignancy. Cancers 2021, 13, 877. [Google Scholar] [CrossRef]
  98. Gao, W.; He, R.; Ren, J.; Zhang, W.; Wang, K.; Zhu, L.; Liang, T. Exosomal HMGB1 derived from hypoxia-conditioned bone marrow mesenchymal stem cells increases angiogenesis via the JNK/HIF-1alpha pathway. FEBS Open Bio 2021, 11, 1364–1373. [Google Scholar] [CrossRef]
  99. Hance, M.W.; Dole, K.; Gopal, U.; Bohonowych, J.E.; Jezierska-Drutel, A.; Neumann, C.A.; Liu, H.; Garraway, I.P.; Isaacs, J.S. Secreted Hsp90 is a novel regulator of the epithelial to mesenchymal transition (EMT) in prostate cancer. J. Biol. Chem. 2012, 287, 37732–37744. [Google Scholar] [CrossRef] [Green Version]
  100. Sims, J.D.; McCready, J.; Jay, D.G. Extracellular heat shock protein (Hsp)70 and Hsp90alpha assist in matrix metalloproteinase-2 activation and breast cancer cell migration and invasion. PLoS ONE 2011, 6, e18848. [Google Scholar] [CrossRef] [Green Version]
  101. Diao, J.; Yang, X.; Song, X.; Chen, S.; He, Y.; Wang, Q.; Chen, G.; Luo, C.; Wu, X.; Zhang, Y. Exosomal Hsp70 mediates immunosuppressive activity of the myeloid-derived suppressor cells via phosphorylation of Stat3. Med. Oncol. 2015, 32, 453. [Google Scholar] [CrossRef]
  102. Ignacio, R.M.C.; Gibbs, C.R.; Kim, S.; Lee, E.S.; Adunyah, S.E.; Son, D.S. Serum amyloid A predisposes inflammatory tumor microenvironment in triple negative breast cancer. Oncotarget 2019, 10, 511–526. [Google Scholar] [CrossRef] [Green Version]
  103. Fourie, C.; Shridas, P.; Davis, T.; de Villiers, W.J.S.; Engelbrecht, A.M. Serum amyloid A and inflammasome activation: A link to breast cancer progression? Cytokine Growth Factor Rev. 2021, 59, 62–70. [Google Scholar] [CrossRef] [PubMed]
  104. Lee, J.W.; Stone, M.L.; Porrett, P.M.; Thomas, S.K.; Komar, C.A.; Li, J.H.; Delman, D.; Graham, K.; Gladney, W.L.; Hua, X.; et al. Hepatocytes direct the formation of a pro-metastatic niche in the liver. Nature 2019, 567, 249–252. [Google Scholar] [CrossRef] [PubMed]
  105. Lee, J.M.; Kim, E.K.; Seo, H.; Jeon, I.; Chae, M.J.; Park, Y.J.; Song, B.; Kim, Y.S.; Kim, Y.J.; Ko, H.J.; et al. Serum amyloid A3 exacerbates cancer by enhancing the suppressive capacity of myeloid-derived suppressor cells via TLR2-dependent STAT3 activation. Eur. J. Immunol. 2014, 44, 1672–1684. [Google Scholar] [CrossRef]
  106. Miller-Ocuin, J.L.; Liang, X.; Boone, B.A.; Doerfler, W.R.; Singhi, A.D.; Tang, D.; Kang, R.; Lotze, M.T.; Zeh, H.J., 3rd. DNA released from neutrophil extracellular traps (NETs) activates pancreatic stellate cells and enhances pancreatic tumor growth. Oncoimmunology 2019, 8, e1605822. [Google Scholar] [CrossRef] [PubMed]
  107. Singel, K.L.; Grzankowski, K.S.; Khan, A.; Grimm, M.J.; D’Auria, A.C.; Morrell, K.; Eng, K.H.; Hylander, B.; Mayor, P.C.; Emmons, T.R.; et al. Mitochondrial DNA in the tumour microenvironment activates neutrophils and is associated with worse outcomes in patients with advanced epithelial ovarian cancer. Br. J. Cancer 2019, 120, 207–217. [Google Scholar] [CrossRef] [Green Version]
  108. Anunobi, R.; Boone, B.A.; Cheh, N.; Tang, D.; Kang, R.; Loux, T.; Lotze, M.T.; Zeh, H.J. Extracellular DNA promotes colorectal tumor cell survival after cytotoxic chemotherapy. J. Surg. Res. 2018, 226, 181–191. [Google Scholar] [CrossRef] [Green Version]
  109. Abu, N.; Rus Bakarurraini, N.A.A.; Nasir, S.N. Extracellular Vesicles and DAMPs in Cancer: A Mini-Review. Front. Immunol. 2021, 12, 740548. [Google Scholar] [CrossRef]
  110. Nabet, B.Y.; Qiu, Y.; Shabason, J.E.; Wu, T.J.; Yoon, T.; Kim, B.C.; Benci, J.L.; DeMichele, A.M.; Tchou, J.; Marcotrigiano, J.; et al. Exosome RNA Unshielding Couples Stromal Activation to Pattern Recognition Receptor Signaling in Cancer. Cell 2017, 170, 352–366.e13. [Google Scholar] [CrossRef] [Green Version]
  111. Rakoff-Nahoum, S.; Medzhitov, R. Toll-like receptors and cancer. Nat. Rev. Cancer 2009, 9, 57–63. [Google Scholar] [CrossRef]
  112. Liu, Y.; Gu, Y.; Han, Y.; Zhang, Q.; Jiang, Z.; Zhang, X.; Huang, B.; Xu, X.; Zheng, J.; Cao, X. Tumor Exosomal RNAs Promote Lung Pre-metastatic Niche Formation by Activating Alveolar Epithelial TLR3 to Recruit Neutrophils. Cancer Cell 2016, 30, 243–256. [Google Scholar] [CrossRef] [Green Version]
  113. Thuringer, D.; Jego, G.; Wettstein, G.; Terrier, O.; Cronier, L.; Yousfi, N.; Hebrard, S.; Bouchot, A.; Hazoume, A.; Joly, A.L.; et al. Extracellular HSP27 mediates angiogenesis through Toll-like receptor 3. FASEB J. 2013, 27, 4169–4183. [Google Scholar] [CrossRef]
  114. Veyrat, M.; Durand, S.; Classe, M.; Glavan, T.M.; Oker, N.; Kapetanakis, N.I.; Jiang, X.; Gelin, A.; Herman, P.; Casiraghi, O.; et al. Stimulation of the toll-like receptor 3 promotes metabolic reprogramming in head and neck carcinoma cells. Oncotarget 2016, 7, 82580–82593. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Matijevic Glavan, T.; Cipak Gasparovic, A.; Verillaud, B.; Busson, P.; Pavelic, J. Toll-like receptor 3 stimulation triggers metabolic reprogramming in pharyngeal cancer cell line through Myc, MAPK, and HIF. Mol. Carcinog. 2016, 56, 1214–1226. [Google Scholar] [CrossRef] [PubMed]
  116. Muresan, X.M.; Slabakova, E.; Prochazkova, J.; Drapela, S.; Fedr, R.; Pickova, M.; Vacek, O.; Vichova, R.; Suchankova, T.; Bouchal, J.; et al. Toll-Like Receptor 3 Overexpression Induces Invasion of Prostate Cancer Cells, whereas Its Activation Triggers Apoptosis. Am. J. Pathol. 2022, 192, 1321–1335. [Google Scholar] [CrossRef] [PubMed]
  117. Tavora, B.; Mederer, T.; Wessel, K.J.; Ruffing, S.; Sadjadi, M.; Missmahl, M.; Ostendorf, B.N.; Liu, X.; Kim, J.Y.; Olsen, O.; et al. Tumoural activation of TLR3-SLIT2 axis in endothelium drives metastasis. Nature 2020, 586, 299–304. [Google Scholar] [CrossRef] [PubMed]
  118. Bugge, M.; Bergstrom, B.; Eide, O.K.; Solli, H.; Kjonstad, I.F.; Stenvik, J.; Espevik, T.; Nilsen, N.J. Surface Toll-like receptor 3 expression in metastatic intestinal epithelial cells induces inflammatory cytokine production and promotes invasiveness. J. Biol. Chem. 2017, 292, 15408–15425. [Google Scholar] [CrossRef] [Green Version]
  119. Hoste, E.; Arwert, E.N.; Lal, R.; South, A.P.; Salas-Alanis, J.C.; Murrell, D.F.; Donati, G.; Watt, F.M. Innate sensing of microbial products promotes wound-induced skin cancer. Nat. Commun. 2015, 6, 5932. [Google Scholar] [CrossRef] [Green Version]
  120. Liu, Y.; Yan, W.; Tohme, S.; Chen, M.; Fu, Y.; Tian, D.; Lotze, M.; Tang, D.; Tsung, A. Hypoxia induced HMGB1 and mitochondrial DNA interactions mediate tumor growth in hepatocellular carcinoma through Toll-like receptor 9. J. Hepatol. 2015, 63, 114–121. [Google Scholar] [CrossRef] [Green Version]
  121. Gao, X.Y.; Zang, J.; Zheng, M.H.; Zhang, Y.F.; Yue, K.Y.; Cao, X.L.; Cao, Y.; Li, X.X.; Han, H.; Jiang, X.F.; et al. Temozolomide Treatment Induces HMGB1 to Promote the Formation of Glioma Stem Cells via the TLR2/NEAT1/Wnt Pathway in Glioblastoma. Front. Cell Dev. Biol. 2021, 9, 620883. [Google Scholar] [CrossRef]
  122. Conti, L.; Lanzardo, S.; Arigoni, M.; Antonazzo, R.; Radaelli, E.; Cantarella, D.; Calogero, R.A.; Cavallo, F. The noninflammatory role of high mobility group box 1/Toll-like receptor 2 axis in the self-renewal of mammary cancer stem cells. FASEB J. 2013, 27, 4731–4744. [Google Scholar] [CrossRef]
  123. Zhang, Z.; Wang, M.; Zhou, L.; Feng, X.; Cheng, J.; Yu, Y.; Gong, Y.; Zhu, Y.; Li, C.; Tian, L.; et al. Increased HMGB1 and cleaved caspase-3 stimulate the proliferation of tumor cells and are correlated with the poor prognosis in colorectal cancer. J. Exp. Clin. Cancer Res. 2015, 34, 51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Shrivastava, S.; Mansure, J.J.; Almajed, W.; Cury, F.; Ferbeyre, G.; Popovic, M.; Seuntjens, J.; Kassouf, W. The Role of HMGB1 in Radioresistance of Bladder Cancer. Mol. Cancer Ther. 2016, 15, 471–479. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Zhu, L.; Ren, L.; Chen, Y.; Fang, J.; Ge, Z.; Li, X. Redox status of high-mobility group box 1 performs a dual role in angiogenesis of colorectal carcinoma. J. Cell Mol. Med. 2015, 19, 2128–2135. [Google Scholar] [CrossRef] [PubMed]
  126. Chen, X.; Cheng, F.; Liu, Y.; Zhang, L.; Song, L.; Cai, X.; You, T.; Fan, X.; Wang, D.; Gong, A.; et al. Toll-like receptor 2 and Toll-like receptor 4 exhibit distinct regulation of cancer cell stemness mediated by cell death-induced high-mobility group box 1. EBioMedicine 2019, 40, 135–150. [Google Scholar] [CrossRef]
  127. Teo Hansen Selno, A.; Schlichtner, S.; Yasinska, I.M.; Sakhnevych, S.S.; Fiedler, W.; Wellbrock, J.; Berger, S.M.; Klenova, E.; Gibbs, B.F.; Fasler-Kan, E.; et al. High Mobility Group Box 1 (HMGB1) Induces Toll-like Receptor 4-Mediated Production of the Immunosuppressive Protein Galectin-9 in Human Cancer Cells. Front. Immunol. 2021, 12, 675731. [Google Scholar] [CrossRef] [PubMed]
  128. Di Lorenzo, A.; Bolli, E.; Ruiu, R.; Ferrauto, G.; Di Gregorio, E.; Avalle, L.; Savino, A.; Poggio, P.; Merighi, I.F.; Riccardo, F.; et al. Toll-like receptor 2 promotes breast cancer progression and resistance to chemotherapy. Oncoimmunology 2022, 11, 2086752. [Google Scholar] [CrossRef]
  129. Deng, R.; Wu, H.; Ran, H.; Kong, X.; Hu, L.; Wang, X.; Su, Q. Glucose-derived AGEs promote migration and invasion of colorectal cancer by up-regulating Sp1 expression. Biochim. Biophys. Acta 2017, 1861 Pt A, 1065–1074. [Google Scholar] [CrossRef] [Green Version]
  130. Kwak, T.; Drews-Elger, K.; Ergonul, A.; Miller, P.C.; Braley, A.; Hwang, G.H.; Zhao, D.; Besser, A.; Yamamoto, Y.; Yamamoto, H.; et al. Targeting of RAGE-ligand signaling impairs breast cancer cell invasion and metastasis. Oncogene 2017, 36, 1559–1572. [Google Scholar] [CrossRef]
  131. Herwig, N.; Belter, B.; Wolf, S.; Haase-Kohn, C.; Pietzsch, J. Interaction of extracellular S100A4 with RAGE prompts prometastatic activation of A375 melanoma cells. J. Cell Mol. Med. 2016, 20, 825–835. [Google Scholar] [CrossRef] [Green Version]
  132. Zheng, L.; Li, D.; Zhou, Y.M.; Yang, H.; Cheng, D.; Ma, X.X. Effects of receptor for advanced glycation endproducts on microvessel formation in endometrial cancer. BMC Cancer 2016, 16, 93. [Google Scholar] [CrossRef] [Green Version]
  133. Bao, J.M.; He, M.Y.; Liu, Y.W.; Lu, Y.J.; Hong, Y.Q.; Luo, H.H.; Ren, Z.L.; Zhao, S.C.; Jiang, Y. AGE/RAGE/Akt pathway contributes to prostate cancer cell proliferation by promoting Rb phosphorylation and degradation. Am. J. Cancer Res. 2015, 5, 1741–1750. [Google Scholar] [CrossRef] [Green Version]
  134. Pusterla, T.; Nemeth, J.; Stein, I.; Wiechert, L.; Knigin, D.; Marhenke, S.; Longerich, T.; Kumar, V.; Arnold, B.; Vogel, A.; et al. Receptor for advanced glycation endproducts (RAGE) is a key regulator of oval cell activation and inflammation-associated liver carcinogenesis in mice. Hepatology 2013, 58, 363–373. [Google Scholar] [CrossRef]
  135. Shi, H.; Kokoeva, M.V.; Inouye, K.; Tzameli, I.; Yin, H.; Flier, J.S. TLR4 links innate immunity and fatty acid-induced insulin resistance. J. Clin. Investig. 2006, 116, 3015–3025. [Google Scholar] [CrossRef] [Green Version]
  136. Konner, A.C.; Bruning, J.C. Toll-like receptors: Linking inflammation to metabolism. Trends Endocrinol. Metab. 2011, 22, 16–23. [Google Scholar] [CrossRef] [PubMed]
  137. Jounai, N.; Kobiyama, K.; Takeshita, F.; Ishii, K.J. Recognition of damage-associated molecular patterns related to nucleic acids during inflammation and vaccination. Front. Cell Infect. Microbiol. 2012, 2, 168. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Iannucci, A.; Caneparo, V.; Raviola, S.; Debernardi, I.; Colangelo, D.; Miggiano, R.; Griffante, G.; Landolfo, S.; Gariglio, M.; De Andrea, M. Toll-like receptor 4-mediated inflammation triggered by extracellular IFI16 is enhanced by lipopolysaccharide binding. PLoS Pathog. 2020, 16, e1008811. [Google Scholar] [CrossRef] [PubMed]
  139. Arkhypov, I.; Ozbay Kurt, F.G.; Bitsch, R.; Novak, D.; Petrova, V.; Lasser, S.; Hielscher, T.; Groth, C.; Lepper, A.; Hu, X.; et al. HSP90alpha induces immunosuppressive myeloid cells in melanoma via TLR4 signaling. J. Immunother Cancer 2022, 10, e005551. [Google Scholar] [CrossRef]
  140. Pucci, M.; Raimondo, S.; Urzi, O.; Moschetti, M.; Di Bella, M.A.; Conigliaro, A.; Caccamo, N.; La Manna, M.P.; Fontana, S.; Alessandro, R. Tumor-Derived Small Extracellular Vesicles Induce Pro-Inflammatory Cytokine Expression and PD-L1 Regulation in M0 Macrophages via IL-6/STAT3 and TLR4 Signaling Pathways. Int. J. Mol. Sci. 2021, 22, 12118. [Google Scholar] [CrossRef]
  141. Hiratsuka, S.; Watanabe, A.; Sakurai, Y.; Akashi-Takamura, S.; Ishibashi, S.; Miyake, K.; Shibuya, M.; Akira, S.; Aburatani, H.; Maru, Y. The S100A8-serum amyloid A3-TLR4 paracrine cascade establishes a pre-metastatic phase. Nat. Cell Biol. 2008, 10, 1349–1355. [Google Scholar] [CrossRef]
  142. Voelcker, V.; Gebhardt, C.; Averbeck, M.; Saalbach, A.; Wolf, V.; Weih, F.; Sleeman, J.; Anderegg, U.; Simon, J. Hyaluronan fragments induce cytokine and metalloprotease upregulation in human melanoma cells in part by signalling via TLR4. Exp. Dermatol. 2008, 17, 100–107. [Google Scholar] [CrossRef] [PubMed]
  143. Dang, S.; Peng, Y.; Ye, L.; Wang, Y.; Qian, Z.; Chen, Y.; Wang, X.; Lin, Y.; Zhang, X.; Sun, X.; et al. Stimulation of TLR4 by LMW-HA induces metastasis in human papillary thyroid carcinoma through CXCR7. Clin. Dev. Immunol. 2013, 2013, 712561. [Google Scholar] [CrossRef] [Green Version]
  144. 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] [Green Version]
  145. Wu, R.; Duan, L.; Cui, F.; Cao, J.; Xiang, Y.; Tang, Y.; Zhou, L. S100A9 promotes human hepatocellular carcinoma cell growth and invasion through RAGE-mediated ERK1/2 and p38 MAPK pathways. Exp. Cell Res. 2015, 334, 228–238. [Google Scholar] [CrossRef]
  146. Duan, L.; Wu, R.; Ye, L.; Wang, H.; Yang, X.; Zhang, Y.; Chen, X.; Zuo, G.; Zhang, Y.; Weng, Y.; et al. S100A8 and S100A9 are associated with colorectal carcinoma progression and contribute to colorectal carcinoma cell survival and migration via Wnt/beta-catenin pathway. PLoS ONE 2013, 8, e62092. [Google Scholar] [CrossRef]
  147. Wu, P.; Quan, H.; Kang, J.; He, J.; Luo, S.; Xie, C.; Xu, J.; Tang, Y.; Zhao, S. Downregulation of Calcium Binding Protein S100A9 Inhibits Hypopharyngeal Cancer Cell Proliferation and Invasion Ability Through Inactivation of NFkappaB Signaling. Oncol. Res. 2017, 25, 1479–1488. [Google Scholar] [CrossRef]
  148. Zhu, W.; Xue, Y.; Liang, C.; Zhang, R.; Zhang, Z.; Li, H.; Su, D.; Liang, X.; Zhang, Y.; Huang, Q.; et al. S100A16 promotes cell proliferation and metastasis via AKT and ERK cell signaling pathways in human prostate cancer. Tumour Biol. 2016, 37, 12241–12250. [Google Scholar] [CrossRef]
  149. Hua, T.; Liu, S.; Xin, X.; Cai, L.; Shi, R.; Chi, S.; Feng, D.; Wang, H. S100A4 promotes endometrial cancer progress through epithelial-mesenchymal transition regulation. Oncol. Rep. 2016, 35, 3419–3426. [Google Scholar] [CrossRef] [Green Version]
  150. Grottke, A.; Ewald, F.; Lange, T.; Norz, D.; Herzberger, C.; Bach, J.; Grabinski, N.; Graser, L.; Hoppner, F.; Nashan, B.; et al. Downregulation of AKT3 Increases Migration and Metastasis in Triple Negative Breast Cancer Cells by Upregulating S100A4. PLoS ONE 2016, 11, e0146370. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  151. Jin, T.; Zhang, Z.; Yang, X.F.; Luo, J.S. S100A4 expression is closely linked to genesis and progression of glioma by regulating proliferation, apoptosis, migration and invasion. Asian Pac. J. Cancer Prev. 2015, 16, 2883–2887. [Google Scholar] [CrossRef] [Green Version]
  152. Medapati, M.R.; Dahlmann, M.; Ghavami, S.; Pathak, K.A.; Lucman, L.; Klonisch, T.; Hoang-Vu, C.; Stein, U.; Hombach-Klonisch, S. RAGE Mediates the Pro-Migratory Response of Extracellular S100A4 in Human Thyroid Cancer Cells. Thyroid 2015, 25, 514–527. [Google Scholar] [CrossRef] [PubMed]
  153. Kuper, C.; Beck, F.X.; Neuhofer, W. NFAT5-mediated expression of S100A4 contributes to proliferation and migration of renal carcinoma cells. Front. Physiol. 2014, 5, 293. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Chen, N.; Sato, D.; Saiki, Y.; Sunamura, M.; Fukushige, S.; Horii, A. S100A4 is frequently overexpressed in lung cancer cells and promotes cell growth and cell motility. Biochem. Biophys. Res. Commun. 2014, 447, 459–464. [Google Scholar] [CrossRef]
  155. Tsukamoto, N.; Egawa, S.; Akada, M.; Abe, K.; Saiki, Y.; Kaneko, N.; Yokoyama, S.; Shima, K.; Yamamura, A.; Motoi, F.; et al. The expression of S100A4 in human pancreatic cancer is associated with invasion. Pancreas 2013, 42, 1027–1033. [Google Scholar] [CrossRef] [PubMed]
  156. Zhuang, H.; Chen, X.; Dong, F.; Zhang, Z.; Zhou, Z.; Ma, Z.; Huang, S.; Chen, B.; Zhang, C.; Hou, B. Prognostic values and immune suppression of the S100A family in pancreatic cancer. J. Cell Mol. Med. 2021, 25, 3006–3018. [Google Scholar] [CrossRef] [PubMed]
  157. Fang, W.Y.; Chen, Y.W.; Hsiao, J.R.; Liu, C.S.; Kuo, Y.Z.; Wang, Y.C.; Chang, K.C.; Tsai, S.T.; Chang, M.Z.; Lin, S.H.; et al. Elevated S100A9 expression in tumor stroma functions as an early recurrence marker for early-stage oral cancer patients through increased tumor cell invasion, angiogenesis, macrophage recruitment and interleukin-6 production. Oncotarget 2015, 6, 28401–28424. [Google Scholar] [CrossRef] [Green Version]
  158. Lim, S.Y.; Yuzhalin, A.E.; Gordon-Weeks, A.N.; Muschel, R.J. Tumor-infiltrating monocytes/macrophages promote tumor invasion and migration by upregulating S100A8 and S100A9 expression in cancer cells. Oncogene 2016, 35, 5735–5745. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Tidehag, V.; Hammarsten, P.; Egevad, L.; Granfors, T.; Stattin, P.; Leanderson, T.; Wikstrom, P.; Josefsson, A.; Hagglof, C.; Bergh, A. High density of S100A9 positive inflammatory cells in prostate cancer stroma is associated with poor outcome. Eur. J. Cancer 2014, 50, 1829–1835. [Google Scholar] [CrossRef]
  160. Nasser, M.W.; Wani, N.A.; Ahirwar, D.K.; Powell, C.A.; Ravi, J.; Elbaz, M.; Zhao, H.; Padilla, L.; Zhang, X.; Shilo, K.; et al. RAGE mediates S100A7-induced breast cancer growth and metastasis by modulating the tumor microenvironment. Cancer Res. 2015, 75, 974–985. [Google Scholar] [CrossRef] [Green Version]
  161. Jo, S.H.; Heo, W.H.; Son, H.Y.; Quan, M.; Hong, B.S.; Kim, J.H.; Lee, H.B.; Han, W.; Park, Y.; Lee, D.S.; et al. S100A8/A9 mediate the reprograming of normal mammary epithelial cells induced by dynamic cell-cell interactions with adjacent breast cancer cells. Sci. Rep. 2021, 11, 1337. [Google Scholar] [CrossRef]
  162. Shen, Z.; Deng, H.; Fang, Y.; Zhu, X.; Ye, G.T.; Yan, L.; Liu, H.; Li, G. Identification of the interplay between SOX9 and S100P in the metastasis and invasion of colon carcinoma. Oncotarget 2015, 6, 20672–20684. [Google Scholar] [CrossRef] [Green Version]
  163. Bettum, I.J.; Gorad, S.S.; Barkovskaya, A.; Pettersen, S.; Moestue, S.A.; Vasiliauskaite, K.; Tenstad, E.; Oyjord, T.; Risa, O.; Nygaard, V.; et al. Metabolic reprogramming supports the invasive phenotype in malignant melanoma. Cancer Lett. 2015, 366, 71–83. [Google Scholar] [CrossRef] [PubMed]
  164. Hendriks, L.E.; Dingemans, A.C. Heat shock protein antagonists in early stage clinical trials for NSCLC. Expert Opin. Investig. Drugs 2017, 26, 541–550. [Google Scholar] [CrossRef]
  165. Bagatell, R.; Whitesell, L. Altered Hsp90 function in cancer: A unique therapeutic opportunity. Mol. Cancer Ther. 2004, 3, 1021–1030. [Google Scholar] [CrossRef] [PubMed]
  166. Song, Q.; Wen, J.; Li, W.; Xue, J.; Zhang, Y.; Liu, H.; Han, J.; Ning, T.; Lu, Z. HSP90 promotes radioresistance of cervical cancer cells via reducing FBXO6-mediated CD147 polyubiquitination. Cancer Sci. 2022, 113, 1463–1474. [Google Scholar] [CrossRef] [PubMed]
  167. Shiota, M.; Bishop, J.L.; Nip, K.M.; Zardan, A.; Takeuchi, A.; Cordonnier, T.; Beraldi, E.; Bazov, J.; Fazli, L.; Chi, K.; et al. Hsp27 regulates epithelial mesenchymal transition, metastasis, and circulating tumor cells in prostate cancer. Cancer Res. 2013, 73, 3109–3119. [Google Scholar] [CrossRef] [Green Version]
  168. Cordonnier, T.; Bishop, J.L.; Shiota, M.; Nip, K.M.; Thaper, D.; Vahid, S.; Heroux, D.; Gleave, M.; Zoubeidi, A. Hsp27 regulates EGF/beta-catenin mediated epithelial to mesenchymal transition in prostate cancer. Int. J. Cancer 2015, 136, E496–E507. [Google Scholar] [CrossRef]
  169. Ren, X.; Li, T.; Zhang, W.; Yang, X. Targeting Heat-Shock Protein 90 in Cancer: An Update on Combination Therapy. Cells 2022, 11, 2556. [Google Scholar] [CrossRef]
  170. Xue, N.; Du, T.; Lai, F.; Jin, J.; Ji, M.; Chen, X. Secreted HSP90alpha-LRP1 Signaling Promotes Tumor Metastasis and Chemoresistance in Pancreatic Cancer. Int. J. Mol. Sci. 2022, 23, 5532. [Google Scholar] [CrossRef]
  171. Feng, H.; Guo, Z.; Chen, X.; Liu, K.; Li, H.; Jia, W.; Wang, C.; Luo, F.; Ji, X.; Zhang, T.; et al. Excessive HSP70/TLR2 activation leads to remodeling of the tumor immune microenvironment to resist chemotherapy sensitivity of mFOLFOX in colorectal cancer. Clin. Immunol. 2022, 245, 109157. [Google Scholar] [CrossRef]
  172. Su, K.; Liu, Y.; Wang, P.; He, K.; Wang, F.; Chi, H.; Rao, M.; Li, X.; Wen, L.; Song, Y.; et al. Heat-shock protein 90alpha is a potential prognostic and predictive biomarker in hepatocellular carcinoma: A large-scale and multicenter study. Hepatol. Int. 2022, 16, 1208–1219. [Google Scholar] [CrossRef]
  173. Pahwa, R.; Dubhashi, J.; Singh, A.; Jailwala, P.; Lobanov, A.; Thomas, C.J.; Ceribelli, M.; Wilson, K.; Ricketts, C.J.; Vocke, C.D.; et al. Inhibition of HSP 90 is associated with potent anti-tumor activity in Papillary Renal Cell Carcinoma. J. Exp. Clin. Cancer Res. 2022, 41, 208. [Google Scholar] [CrossRef] [PubMed]
  174. Kim, J.Y.; Cho, T.M.; Park, J.M.; Park, S.; Park, M.; Nam, K.D.; Ko, D.; Seo, J.; Kim, S.; Jung, E.; et al. A novel HSP90 inhibitor SL-145 suppresses metastatic triple-negative breast cancer without triggering the heat shock response. Oncogene 2022, 41, 3289–3297. [Google Scholar] [CrossRef] [PubMed]
  175. Liu, J.; Kang, R.; Kroemer, G.; Tang, D. Targeting HSP90 sensitizes pancreas carcinoma to PD-1 blockade. Oncoimmunology 2022, 11, 2068488. [Google Scholar] [CrossRef] [PubMed]
  176. Rice, M.A.; Kumar, V.; Tailor, D.; Garcia-Marques, F.J.; Hsu, E.C.; Liu, S.; Bermudez, A.; Kanchustambham, V.; Shankar, V. SU086, an inhibitor of HSP90, impairs glycolysis and represents a treatment strategy for advanced prostate cancer. Cell Rep. Med. 2022, 3, 100502. [Google Scholar] [CrossRef]
  177. Wei, S.; Yin, D.; Yu, S.; Lin, X.; Savani, M.R.; Du, K.; Ku, Y.; Wu, D.; Li, S.; Liu, H.; et al. Antitumor Activity of a Mitochondrial-Targeted HSP90 Inhibitor in Gliomas. Clin. Cancer Res. 2022, 28, 2180–2195. [Google Scholar] [CrossRef]
  178. Killock, D. HSP90 inhibition improves GIST survival. Nat. Rev. Clin. Oncol. 2022, 19, 568. [Google Scholar] [CrossRef]
  179. Sasame, J.; Ikegaya, N.; Kawazu, M.; Natsumeda, M.; Hayashi, T.; Isoda, M.; Satomi, K.; Tomiyama, A.; Oshima, A.; Honma, H.; et al. HSP90 Inhibition Overcomes Resistance to Molecular Targeted Therapy in BRAFV600E-mutant High-grade Glioma. Clin. Cancer Res. 2022, 28, 2425–2439. [Google Scholar] [CrossRef] [PubMed]
  180. Chen, W.; Ren, X.; Wu, J.; Gao, X.; Cen, X.; Wang, S.; Sheng, S.; Chen, Q.; Tang, Y.J.; Liang, X.H.; et al. HSP27 associates with epithelial-mesenchymal transition, stemness and radioresistance of salivary adenoid cystic carcinoma. J. Cell Mol. Med. 2018, 22, 2283–2298. [Google Scholar] [CrossRef] [Green Version]
  181. Yao, K.; He, L.; Gan, Y.; Liu, J.; Tang, J.; Long, Z.; Tan, J. HMGN5 promotes IL-6-induced epithelial-mesenchymal transition of bladder cancer by interacting with Hsp27. Aging 2020, 12, 7282–7298. [Google Scholar] [CrossRef]
  182. Fang, Z.; Liang, W.; Luo, L. HSP27 promotes epithelial-mesenchymal transition through activation of the beta-catenin/MMP3 pathway in pancreatic ductal adenocarcinoma cells. Transl. Cancer Res. 2019, 8, 1268–1278. [Google Scholar] [CrossRef] [PubMed]
  183. Han, L.; Jiang, Y.; Han, D.; Tan, W. Hsp27 regulates epithelial mesenchymal transition, metastasis and proliferation in colorectal carcinoma. Oncol. Lett. 2018, 16, 5309–5316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Choi, S.K.; Kam, H.; Kim, K.Y.; Park, S.I.; Lee, Y.S. Targeting Heat Shock Protein 27 in Cancer: A Druggable Target for Cancer Treatment? Cancers 2019, 11, 1195. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Katakam, S.; Anand, S.; Martin, P.; Riggi, N.; Stamenkovic, I. Necrotic debris and STING exert therapeutically relevant effects on tumor cholesterol homeostasis. Life Sci. Alliance 2022, 5, e202101256. [Google Scholar] [CrossRef]
  186. Mehrabi, M.; Amini, F.; Mehrabi, S. Active Role of the Necrotic Zone in Desensitization of Hypoxic Macrophages and Regulation of CSC-Fate: A hypothesis. Front. Oncol. 2018, 8, 235. [Google Scholar] [CrossRef] [Green Version]
  187. ElShamy, W.M.; Sinha, A.; Said, N. Aggressiveness Niche: Can It Be the Foster Ground for Cancer Metastasis Precursors? Stem Cells Int. 2016, 2016, 4829106. [Google Scholar] [CrossRef] [Green Version]
  188. Cormier, S.A.; Taranova, A.G.; Bedient, C.; Nguyen, T.; Protheroe, C.; Pero, R.; Dimina, D.; Ochkur, S.I.; O’Neill, K.; Colbert, D.; et al. Pivotal Advance: Eosinophil infiltration of solid tumors is an early and persistent inflammatory host response. J. Leukoc. Biol. 2006, 79, 1131–1139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  189. Lotfi, R.; Kaltenmeier, C.; Lotze, M.T.; Bergmann, C. Until Death Do Us Part: Necrosis and Oxidation Promote the Tumor Microenvironment. Transfus Med. Hemother 2016, 43, 120–132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  190. Yee, P.P.; Li, W. Tumor necrosis: A synergistic consequence of metabolic stress and inflammation. Bioessays 2021, 43, e2100029. [Google Scholar] [CrossRef]
  191. Ocana, A.; Nieto-Jimenez, C.; Pandiella, A.; Templeton, A.J. Neutrophils in cancer: Prognostic role and therapeutic strategies. Mol. Cancer 2017, 16, 137. [Google Scholar] [CrossRef] [Green Version]
  192. Di Virgilio, F.; Sarti, A.C.; Coutinho-Silva, R. Purinergic signaling, DAMPs, and inflammation. Am. J. Physiol. Cell Physiol. 2020, 318, C832–C835. [Google Scholar] [CrossRef] [Green Version]
  193. Deaglio, S.; Dwyer, K.M.; Gao, W.; Friedman, D.; Usheva, A.; Erat, A.; Chen, J.F.; Enjyoji, K.; Linden, J.; Oukka, M.; et al. Adenosine generation catalyzed by CD39 and CD73 expressed on regulatory T cells mediates immune suppression. J. Exp. Med. 2007, 204, 1257–1265. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Antonioli, L.; Blandizzi, C.; Pacher, P.; Hasko, G. Immunity, inflammation and cancer: A leading role for adenosine. Nat. Rev. Cancer 2013, 13, 842–857. [Google Scholar] [CrossRef] [PubMed]
  195. Ma, S.R.; Deng, W.W.; Liu, J.F.; Mao, L.; Yu, G.T.; Bu, L.L.; Kulkarni, A.B.; Zhang, W.F.; Sun, Z.J. Blockade of adenosine A2A receptor enhances CD8(+) T cells response and decreases regulatory T cells in head and neck squamous cell carcinoma. Mol. Cancer 2017, 16, 99. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Bianchi, G.; Vuerich, M.; Pellegatti, P.; Marimpietri, D.; Emionite, L.; Marigo, I.; Bronte, V.; Di Virgilio, F.; Pistoia, V.; Raffaghello, L. ATP/P2X7 axis modulates myeloid-derived suppressor cell functions in neuroblastoma microenvironment. Cell Death Dis. 2014, 5, e1135. [Google Scholar] [CrossRef] [Green Version]
  197. Baghdadi, M.; Yoneda, A.; Yamashina, T.; Nagao, H.; Komohara, Y.; Nagai, S.; Akiba, H.; Foretz, M.; Yoshiyama, H.; Kinoshita, I.; et al. TIM-4 glycoprotein-mediated degradation of dying tumor cells by autophagy leads to reduced antigen presentation and increased immune tolerance. Immunity 2013, 39, 1070–1081. [Google Scholar] [CrossRef] [Green Version]
  198. Chiba, S.; Baghdadi, M.; Akiba, H.; Yoshiyama, H.; Kinoshita, I.; Dosaka-Akita, H.; Fujioka, Y.; Ohba, Y.; Gorman, J.V.; Colgan, J.D.; et al. Tumor-infiltrating DCs suppress nucleic acid-mediated innate immune responses through interactions between the receptor TIM-3 and the alarmin HMGB1. Nat. Immunol. 2012, 13, 832–842. [Google Scholar] [CrossRef] [Green Version]
  199. Wild, C.A.; Bergmann, C.; Fritz, G.; Schuler, P.; Hoffmann, T.K.; Lotfi, R.; Westendorf, A.; Brandau, S.; Lang, S. HMGB1 conveys immunosuppressive characteristics on regulatory and conventional T cells. Int. Immunol. 2012, 24, 485–494. [Google Scholar] [CrossRef] [Green Version]
  200. Demoulin, S.; Herfs, M.; Somja, J.; Roncarati, P.; Delvenne, P.; Hubert, P. HMGB1 secretion during cervical carcinogenesis promotes the acquisition of a tolerogenic functionality by plasmacytoid dendritic cells. Int. J. Cancer 2015, 137, 345–358. [Google Scholar] [CrossRef] [Green Version]
  201. Shiau, D.J.; Kuo, W.T.; Davuluri, G.V.N.; Shieh, C.C.; Tsai, P.J.; Chen, C.C.; Lin, Y.S.; Wu, Y.Z.; Hsiao, Y.P.; Chang, C.P. Hepatocellular carcinoma-derived high mobility group box 1 triggers M2 macrophage polarization via a TLR2/NOX2/autophagy axis. Sci. Rep. 2020, 10, 13582. [Google Scholar] [CrossRef]
  202. Ye, L.; Zhang, Q.; Cheng, Y.; Chen, X.; Wang, G.; Shi, M.; Zhang, T.; Cao, Y.; Pan, H.; Zhang, L.; et al. Tumor-derived exosomal HMGB1 fosters hepatocellular carcinoma immune evasion by promoting TIM-1(+) regulatory B cell expansion. J. Immunother Cancer 2018, 6, 145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. Wang, W.; Chapman, N.M.; Zhang, B.; Li, M.; Fan, M.; Laribee, R.N.; Zaidi, M.R.; Pfeffer, L.M.; Chi, H.; Wu, Z.H. Upregulation of PD-L1 via HMGB1-Activated IRF3 and NF-kappaB Contributes to UV Radiation-Induced Immune Suppression. Cancer Res. 2019, 79, 2909–2922. [Google Scholar] [CrossRef] [PubMed]
  204. Sinha, P.; Okoro, C.; Foell, D.; Freeze, H.H.; Ostrand-Rosenberg, S.; Srikrishna, G. Proinflammatory S100 proteins regulate the accumulation of myeloid-derived suppressor cells. J. Immunol. 2008, 181, 4666–4675. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Cheng, P.; Corzo, C.A.; Luetteke, N.; Yu, B.; Nagaraj, S.; Bui, M.M.; Ortiz, M.; Nacken, W.; Sorg, C.; Vogl, T.; et al. Inhibition of dendritic cell differentiation and accumulation of myeloid-derived suppressor cells in cancer is regulated by S100A9 protein. J. Exp. Med. 2008, 205, 2235–2249. [Google Scholar] [CrossRef] [PubMed]
  206. Gao, Y.; Xu, H.; Li, N.; Wang, H.; Ma, L.; Chen, S.; Liu, J.; Zheng, Y.; Zhang, Y. Renal cancer-derived exosomes induce tumor immune tolerance by MDSCs-mediated antigen-specific immunosuppression. Cell Commun. Signal 2020, 18, 106. [Google Scholar] [CrossRef]
  207. Block, H.; Rossaint, J.; Zarbock, A. The Fatal Circle of NETs and NET-Associated DAMPs Contributing to Organ Dysfunction. Cells 2022, 11, 1919. [Google Scholar] [CrossRef] [PubMed]
  208. Munir, H.; Jones, J.O.; Janowitz, T.; Hoffmann, M.; Euler, M.; Martins, C.P.; Welsh, S.J.; Shields, J.D. Stromal-driven and Amyloid beta-dependent induction of neutrophil extracellular traps modulates tumor growth. Nat. Commun. 2021, 12, 683. [Google Scholar] [CrossRef] [PubMed]
  209. Leijte, G.P.; Custers, H.; Gerretsen, J.; Heijne, A.; Roth, J.; Vogl, T.; Scheffer, G.J.; Pickkers, P.; Kox, M. Increased Plasma Levels of Danger-Associated Molecular Patterns Are Associated With Immune Suppression and Postoperative Infections in Patients Undergoing Cytoreductive Surgery and Hyperthermic Intraperitoneal Chemotherapy. Front. Immunol. 2018, 9, 663. [Google Scholar] [CrossRef]
  210. Timmermans, K.; Kox, M.; Vaneker, M.; van den Berg, M.; John, A.; van Laarhoven, A.; van der Hoeven, H.; Scheffer, G.J.; Pickkers, P. Plasma levels of danger-associated molecular patterns are associated with immune suppression in trauma patients. Intensive Care Med. 2016, 42, 551–561. [Google Scholar] [CrossRef]
  211. Obeid, M.; Tesniere, A.; Ghiringhelli, F.; Fimia, G.M.; Apetoh, L.; Perfettini, J.L.; Castedo, M.; Mignot, G.; Panaretakis, T.; Casares, N.; et al. Calreticulin exposure dictates the immunogenicity of cancer cell death. Nat. Med. 2007, 13, 54–61. [Google Scholar] [CrossRef]
  212. Ghiringhelli, F.; Apetoh, L.; Tesniere, A.; Aymeric, L.; Ma, Y.; Ortiz, C.; Vermaelen, K.; Panaretakis, T.; Mignot, G.; Ullrich, E.; et al. Activation of the NLRP3 inflammasome in dendritic cells induces IL-1beta-dependent adaptive immunity against tumors. Nat. Med. 2009, 15, 1170–1178. [Google Scholar] [CrossRef] [PubMed]
  213. Apetoh, L.; Ghiringhelli, F.; Tesniere, A.; Obeid, M.; Ortiz, C.; Criollo, A.; Mignot, G.; Maiuri, M.C.; Ullrich, E.; Saulnier, P.; et al. Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat. Med. 2007, 13, 1050–1059. [Google Scholar] [CrossRef] [PubMed]
  214. Ahmed, A.; Tait, S.W.G. Targeting immunogenic cell death in cancer. Mol. Oncol. 2020, 14, 2994–3006. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic illustrations of various types of cell death. (A) Two broad categories are regulated (RCD) and accidental (ACD) cell death. RCD is the most common type. It includes necrotic and non-necrotic forms of cell death. Non-necrotic cell death includes apoptosis, also called programmed cell death (PCD), and cell death by autophagy. Necrotic regulated cell death includes necroptosis, ferroptosis, and pyroptosis. Other types of RCD are entotic cell death, NETotic cell death, parthanatos, lysosome-dependent cell death, and mitochondrial permeability transition (MPT)-driven necrosis. Accidental cell death usually has the form of a non-regulated necrosis. (B) represents the summary of different sorts of cell death.
Figure 1. Schematic illustrations of various types of cell death. (A) Two broad categories are regulated (RCD) and accidental (ACD) cell death. RCD is the most common type. It includes necrotic and non-necrotic forms of cell death. Non-necrotic cell death includes apoptosis, also called programmed cell death (PCD), and cell death by autophagy. Necrotic regulated cell death includes necroptosis, ferroptosis, and pyroptosis. Other types of RCD are entotic cell death, NETotic cell death, parthanatos, lysosome-dependent cell death, and mitochondrial permeability transition (MPT)-driven necrosis. Accidental cell death usually has the form of a non-regulated necrosis. (B) represents the summary of different sorts of cell death.
Ijms 24 05278 g001
Figure 2. Impact of biomolecules released by necrotic cells on malignant and stromal live cells in the tumor microenvironment (TME). Necrotic cells release DAMPs (HMGB1, HSPs, S100A, RNAs, and DNAs) into the TME. Malignant cells, cancer-associated fibroblasts (CAFs), and various types of infiltrating immune cells have pattern recognition receptors (PRRs). When binding these PRRs, damage-associated molecular patterns (DAMPs) can induce substantial changes in gene expression, resulting in inflammation, epithelial to mesenchymal transition (EMT), immunosuppression, local invasion, and metastases.
Figure 2. Impact of biomolecules released by necrotic cells on malignant and stromal live cells in the tumor microenvironment (TME). Necrotic cells release DAMPs (HMGB1, HSPs, S100A, RNAs, and DNAs) into the TME. Malignant cells, cancer-associated fibroblasts (CAFs), and various types of infiltrating immune cells have pattern recognition receptors (PRRs). When binding these PRRs, damage-associated molecular patterns (DAMPs) can induce substantial changes in gene expression, resulting in inflammation, epithelial to mesenchymal transition (EMT), immunosuppression, local invasion, and metastases.
Ijms 24 05278 g002
Table 1. List of damage-associated molecular patterns (DAMPs) and their receptors.
Table 1. List of damage-associated molecular patterns (DAMPs) and their receptors.
Type of DAMPDAMPsReceptors
ProteinsHMGB1TLR2, TLR4, RAGE
HistoneTLR2, TLR4
S100TLR2, TLR4, RAGE
HSPsTLR2, TLR4, CD91, RAGE
Annexin A1FPR1
VersicanTLR2, TLR6, CD14
Fibronectin (EDA domain)TLR4
FibrinogenTLR4
Tenascin CTLR4
F-actinDNGR-1
Cyclophilin ACD147
TLR2, NLRP1, NLRP3, CD36, RAGE
IL1αIL-1R
IL33ST2
Formyl peptideFPR1
CalreticulinCD91
DefensinsTLR4
Cathelicidin (LL37)P2X7, FPR2
GranulysinTLR4
Lipids and carbohydratesLMW hyaluronanTLR2, TLR4, NLRP3
SAATLR2, TLR4
Heparan sulfateTLR4
Metabolite-related DAMPsATPP2X7, P2Y2
Uric acidNLRP3, P2X7
Nucleic acidsDNATLR9, AIM2
RNATLR3, TLR7/8, RIG-I, MDA5
mtDNATLR9
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

Zapletal, E.; Vasiljevic, T.; Busson, P.; Matijevic Glavan, T. Dialog beyond the Grave: Necrosis in the Tumor Microenvironment and Its Contribution to Tumor Growth. Int. J. Mol. Sci. 2023, 24, 5278. https://doi.org/10.3390/ijms24065278

AMA Style

Zapletal E, Vasiljevic T, Busson P, Matijevic Glavan T. Dialog beyond the Grave: Necrosis in the Tumor Microenvironment and Its Contribution to Tumor Growth. International Journal of Molecular Sciences. 2023; 24(6):5278. https://doi.org/10.3390/ijms24065278

Chicago/Turabian Style

Zapletal, Emilija, Tea Vasiljevic, Pierre Busson, and Tanja Matijevic Glavan. 2023. "Dialog beyond the Grave: Necrosis in the Tumor Microenvironment and Its Contribution to Tumor Growth" International Journal of Molecular Sciences 24, no. 6: 5278. https://doi.org/10.3390/ijms24065278

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