Next Article in Journal
Evaluation of the Impact of Population Management on the Genetic Parameters of Selected Spiral-Horned Antelopes
Next Article in Special Issue
Overnutrition and Lipotoxicity: Impaired Efferocytosis and Chronic Inflammation as Precursors to Multifaceted Disease Pathogenesis
Previous Article in Journal
Cellulolytic Aerobic Bacteria Isolated from Agricultural and Forest Soils: An Overview
Previous Article in Special Issue
Types of Cell Death from a Molecular Perspective
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Exploring beyond Common Cell Death Pathways in Oral Cancer: A Systematic Review

by
Leonardo de Oliveira Siquara da Rocha
1,2,
Everton Freitas de Morais
3,
Lilianny Querino Rocha de Oliveira
3,
Andressa Vollono Barbosa
2,
Daniel W. Lambert
4,
Clarissa A. Gurgel Rocha
1,2,5,6,* and
Ricardo D. Coletta
3,*
1
Department of Pathology and Forensic Medicine, School of Medicine, Federal University of Bahia, Salvador 40110-100, BA, Brazil
2
Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, BA, Brazil
3
Graduate Program in Oral Biology and Department of Oral Diagnosis, School of Dentistry, University of Campinas, Piracicaba 13414-018, SP, Brazil
4
School of Clinical Dentistry, The University of Sheffield, Sheffield S10 2TA, UK
5
Department of Propaedeutics, School of Dentistry, Federal University of Bahia, Salvador 40110-909, BA, Brazil
6
D’Or Institute for Research and Education (IDOR), Salvador 41253-190, BA, Brazil
*
Authors to whom correspondence should be addressed.
Biology 2024, 13(2), 103; https://doi.org/10.3390/biology13020103
Submission received: 5 December 2023 / Revised: 17 January 2024 / Accepted: 1 February 2024 / Published: 6 February 2024

Abstract

:

Simple Summary

Oral squamous cell carcinoma (OSCC), the major malignant tumor of the oral cavity, is one of the most common cancers in the world. Its treatment response rate mainly depends on the clinical stage, and there is an urgent need to develop more effective therapeutic alternatives. Our understanding of the molecular mechanisms regulating different types of tumor cell death is evolving and providing new perspectives to increase the efficacy of conventional therapies combined with the manipulation of signaling cascades to promote tumor cell death. In this systematic review, we provide an overview of emerging types of cell death in OSCC, highlighting opportunities to capitalize on this increased understanding to inform diagnosis, prognosis and treatment.

Abstract

Oral squamous cell carcinoma (OSCC) is the most common and lethal type of head and neck cancer in the world. Variable response and acquisition of resistance to traditional therapies show that it is essential to develop novel strategies that can provide better outcomes for the patient. Understanding of cellular and molecular mechanisms of cell death control has increased rapidly in recent years. Activation of cell death pathways, such as the emerging forms of non-apoptotic programmed cell death, including ferroptosis, pyroptosis, necroptosis, NETosis, parthanatos, mitoptosis and paraptosis, may represent clinically relevant novel therapeutic opportunities. This systematic review summarizes the recently described forms of cell death in OSCC, highlighting their potential for informing diagnosis, prognosis and treatment. Original studies that explored any of the selected cell deaths in OSCC were included. Electronic search, study selection, data collection and risk of bias assessment tools were realized. The literature search was carried out in four databases, and the extracted data from 79 articles were categorized and grouped by type of cell death. Ferroptosis, pyroptosis, and necroptosis represented the main forms of cell death in the selected studies, with links to cancer immunity and inflammatory responses, progression and prognosis of OSCC. Harnessing the potential of these pathways may be useful in patient-specific prognosis and individualized therapy. We provide perspectives on how these different cell death types can be integrated to develop decision tools for diagnosis, prognosis, and treatment of OSCC.

1. Introduction

Among the hallmarks of cancer, the acquisition of resistance to cell death plays an important role in cancer initiation and progression to high-grade malignant states, which are frequently unresponsive to conventional anti-cancer therapies [1]. In recent years, there has been a significant improvement in the understanding of different mechanisms of cell death beyond the traditionally observed apoptosis and necrosis [2,3]. Alternative cell death pathways, collectively called programmed cell death (PCD), have been identified in a variety of pathological processes, including oral cancer [4]. These pathways not only contribute to the understanding of cancer pathophysiology [5] but also reveal an intricate balance between molecules involved in cell survival and death, with significant implications, for example, as prognostic biomarkers and in the development of new therapeutic strategies that are beginning to be explored [6].
While malignant cells develop strategies to escape or limit conventional cell death pathways, understanding of their ability to escape death by other mechanisms is much more limited [5]. Since 2018, the Nomenclature Committee on Cellular Death has classified PCD into 12 subtypes of death that differ in molecular mechanisms but may share small similarities in their morphological characteristics, ranging from a necrotic profile, that is, unprogrammed and with a disordered appearance, to an apoptotic profile with an organized profile [2,7]. However, an update proposed by Yan, Elbadawi and Efferth [3] divided cell deaths into three large groups based on activation of specific signaling pathways: (1) Non-programmed cell death (NPCD) or necrosis, (2) apoptotic programmed cell death (APCD) and (3) non-apoptotic programmed cell death (NAPCD), which differs from the apoptotic form because it does not maintain the integrity of the cell membrane and is independent of caspases. Similar to apoptosis, NAPCD has a highly regulated molecular machinery that can be targeted or modulated by molecular strategies [2].
The most studied emerging NAPCD types in oncology are ferroptosis, pyroptosis and necroptosis [8]. Ferroptosis involves the accumulation of lipid peroxides due to disrupted cellular antioxidant defenses, leading to oxidative stress-induced cell death [9]. Cancer cells can escape ferroptosis by enhancing antioxidant defenses and modifying lipid metabolism, enabling them to survive and proliferate despite conditions that typically trigger ferroptotic cell death [10]. Meanwhile, during pyroptosis and necroptosis, the intracellular content is expelled from the cell through membrane pores formed by proteins such as those from the Gasdermin family (GSDM) in pyroptosis and mixed lineage kinase domain-like (MLKL) in necroptosis, resulting in recruitment of inflammatory cells [11,12]. The pro-inflammatory environment induced by the extravasation of intracellular contents may promote the transformation and progression of tumor cells [13,14]. However, the role of NAPCD in cancer progression is only partially understood, and the literature is conflicting. This systematic review aims to summarize the available literature on NAPCD in oral squamous cell carcinomas (OSCCs), which represent more than 90% of cancer cases in the head and neck region, emphasizing the application of these different types of cell death in diagnosis, prognosis, and treatment of OSCC.

2. Materials and Methods

2.1. Review Approach

This systematic review followed the methodological principles outlined in the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines [15]. The PICO format was used to construct the research questions with the following inclusion criteria: Population—in vitro studies, animal models or studies of patients with OSCC; Intervention—studies exploring NAPCD in OSCC; Comparison—control group; Outcome—analysis of the behavior of the tumor cells using functional assays or prognostic significance of NAPCD-related genes. In this context, the following research questions were established: 1. What is the role of NAPCD in OSCC? 2. What are the underlying mechanisms of NAPCD in the context of OSCC?

2.2. Search Strategy

Bibliographic searches were conducted in four databases: Embase, Medline/PubMed, Scopus and Web of Science. In each database, different combinations of the following descriptors and their synonyms were used: “oral cancer” OR “mouth cancer” OR “oral cavity cancer” OR “head neck cancer” AND “entosis” OR “ferroptosis” OR “pyroptosis” OR “NETosis” OR “Necroptosis” OR “Parthanatos” OR “Mitoptosis” OR “Methuosis”. The strategy adopted sought to rescue as many studies as possible related to the subject. Boolean operators AND, OR and NOT were used. All manuscripts published in English from 1960 to October 2023 were considered and analyzed according to the other steps of the review. The complete search strategy is depicted in Appendix A.

2.3. Eligibility Criteria

The following inclusion criteria were adopted: (1) articles using in silico, in vitro or in vivo methods and (2) studies investigating NAPCD in OSCC. The following criteria were used to exclude articles: (1) articles unavailable as full texts, (2) articles without the selected descriptors, (3) duplicated studies and (4) review articles.

2.4. Study Selection

After elimination of duplicates, the initial screening was based on titles and abstracts. The articles that passed the initial screening and those for which our preliminary analysis raised uncertainties underwent a thorough examination of their full texts.

2.5. Data Extraction and Data Synthesis

Data extraction was carried out by the researchers independently using a predetermined extraction table, and disagreements were resolved by consensus. The reported activities were subdivided into in silico, in vitro and in vivo. The following information was extracted from each paper: study design, sample, type of cell death, markers/pathways, detection methods and main results/conclusion. The findings were presented using a narrative synthesis because it was not feasible to perform a meta-analysis due to the significant heterogeneity among the studies included in this review.

2.6. Quality Assessment

The risk of bias and methodological quality of the selected studies exploring animal models were independently assessed by applying the Systematic Review Center for Laboratory Animal Experimentation SYRCLE tool [16]. For in vitro studies, the quality of evidence was assessed with the tool developed by the United States national toxicology program, with modifications incorporated by Bezemer et al. [17].

3. Results

A total of 4506 articles were identified in the initial search. After the exclusion of duplicates and the screening of the titles and abstracts, 4396 studies were excluded (first exclusion criterion), and 110 studies remained for full-text evaluation. After the full-text evaluation, 79 articles were included in this review (second exclusion criterion). The PRISMA flowchart for the study is reported in Figure 1. The list of excluded studies (n = 31) and reasons for exclusion are shown in Appendix B.
The selected studies were published between 2015 and 2023 and were all written in English. They were conducted in 12 different countries, with 55 (68.7%) of the articles performed by Chinese researchers, followed by studies from the Republic of Korea (n = 5, 6.3%) and Taiwan (n = 4, 5%). Three studies were performed in collaboration: one involving India and the United States, one involving Japan and Australia, and one from collaboration between Japan and China.
Among the selected studies, 46 studies evaluated ferroptosis [18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63], 22 studies focused on pyroptosis [28,33,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83], 8 studies on necroptosis [84,85,86,87,88,89,90,91] and 5 studies on other types of cell death [92,93,94,95,96]. One study [28] explored both ferroptosis and pyroptosis, revealing that Quisinostat, a broad-spectrum epigenetic drug acting as a histone deacetylase inhibitor, shows antitumor effects by promoting the caspase-1-related pathway of pyroptosis and the glutathione peroxidase 4 (GPX4)-related pathway associated with ferroptosis in OSCC cells, besides caspase-3-related pathway-induced apoptosis. Additionally, another study [33], while investigating tumor hypoxia and oxidative stress in cancer stem cell (CSC) reprogramming, noticed an upregulation of pyroptosis and an inhibition of ferroptosis and necroptosis in bacteria-infected CSCs.
Most of the articles included in this review performed experiments based on in vitro assays (n = 68, 85%), with 31 studies exploring only in vitro assays, 34 combining in vitro and in vivo assays, and 3 combining in vitro and in silico predictions. In vivo studies, either with human cancer samples or xenograft murine models, were presented in 38 (47.5%) of the studies. Amongst these studies, the most common type involved in vivo and in vitro experiments (n = 34). In silico prediction analyses were found in 14 (17.5%) studies, with 6 of them combining in vitro or in vivo experiments for validation of the evidence. The main features and findings of the studies included in this review are presented in Table 1.
The risk of bias assessment of the in vitro studies revealed 56 (84.8%) studies with low risk and 10 (15.2%) with moderate risk (Appendix C). Concerning the risk of experimental conditions, performance, outcome assessment and reporting, most studies have been categorized as low risk of bias because the descriptions were well-detailed and classical methods with clear outcomes were adopted. The risk of blinding could not be confirmed because none of the studies provided complete information. Based on the SYRCLE tool, none of the in vivo studies using animal models showed a high risk of bias (Appendix D). Most studies did not provide sufficient information to assess the strategies of selection, including baseline characteristics of the animals and allocation concealment, and the methods of performance and evaluation of the outcomes (there was no description of whether the researchers who manipulated the animals had any knowledge of the groups). Moreover, 29 out of 31 studies did not properly describe how they deal with potential bias due to incomplete outcome data.

4. Discussion

4.1. Ferroptosis

Ferroptosis is a distinguished type of NAPCD characterized by iron-dependent lipid peroxide accumulation, particularly of polyunsaturated fatty acids [97,98]. The research landscape of this type of cell death and its implications in diseases is relatively recent [99,100] but promising for increased knowledge on mechanisms and therapeutic strategies in cancer [99,101]. Ferroptosis can occur through two major pathways: the extrinsic pathway or transporter-dependent, and the intrinsic or enzyme-regulated pathway [99,102] (Figure 2). Despite being distinct pathways, it is important to highlight that one can influence the other, as both rely on iron metabolism and glutathione (GSH)-dependent antioxidant mechanisms [103]. The extrinsic mechanism depends on the balance of iron and amino acid transport across the cell membrane [102]. A higher intracellular iron level increases the production of reactive oxygen species (ROS) through the Fenton reaction [104]. Moreover, inhibition of the Xc-system reduces cystine uptake, which is essential for synthesizing GSH [102]. The depletion of GSH reduces the cell’s antioxidant defenses and favors lipid peroxidation. In turn, the intrinsic pathway is mainly induced by inhibiting glutathione peroxidase 4 (GPX4), an enzyme that plays a pivotal role in reducing lipid hydroperoxides to non-toxic lipid alcohols using GSH as a cofactor [105]. The inhibition of GPX4 activity occurs in an unchecked accumulation of lipid hydroperoxides, leading to cellular damage and eventual ferroptosis cell death [102,105].
Tumor cells are more susceptible to ferroptosis due to altered metabolism and increased iron uptake [98]. Although ferroptosis is expected to be associated with tumor suppression [99], evidence demonstrates that ferroptosis regulatory pathways may promote tumor growth or progression [106]. The p53 gene can promote ferroptosis by repression of solute carrier family 7 member 11 (SLC7A11), a component of the cystine/glutamate antiporter, reducing GSH and increasing cellular oxidative stress and lipid peroxidation [107]. In addition, tumor cells display increased iron uptake, which can increase the Fenton reaction, producing reactive oxygen species and lipid peroxidation [108]. In other circumstances, the oxidative stress, a prominent feature of ferroptosis, can activate the nuclear erythroid factor 2-related factor 2 (NRF2), a well-known transcription factor activated in response to oxidative stress [109]. Although NRF2 may be cytoprotective, chronic NRF2 activation in cancer cells supports proliferation, metabolic reprogramming and resistance to therapy [110,111]. The hypoxia inducible factor 1 subunit alpha (HIF-1α) has also been implicated in increasing iron storage and transport proteins, influencing cellular propensity for ferroptosis [112]. While HIF-1α is primarily known for its role in the cellular response to hypoxia, oxidative stress can also stabilize HIF-1α, resulting in the transcription of genes involved in angiogenesis, glucose metabolism, and cell survival, and supporting tumor growth, angiogenesis, and metastasis [113]. The tumor microenvironment is also influenced by ferroptosis once tumor cells release damage-associated molecular patterns (DAMPs) that recruit and activate immune cells such as dendritic cells and macrophages, initiating an antitumor immune response [114]. Increased lipid peroxidation resulting from ferroptosis may also increase T cell-mediated cytotoxicity against tumor cells [115].
Targeting ferroptosis in OSCC may offer a new path in tumor treatment [116], especially those resistant to traditional therapy [99], and holds the potential to be integrated into combination therapies for enhanced efficacy [117]. Cancer cells, due to their high metabolic activity and dependency on iron and lipid metabolism [118,119], are vulnerable targets for ferroptosis induction, which could inhibit their growth and proliferation. This is further enhanced by targeting CSCs, which are often responsible for tumor initiation and recurrence [120,121]. Iron metabolism plays a crucial role in CSC maintenance and survival [122,123]. These cells exhibit an “iron addiction” by showing a higher concentration of iron compared to the non-stem cell population in the tumor [108]. By hijacking iron metabolism in these cells, ferroptosis becomes a potential tumor suppressor agent to control a significantly powerful population of cells within cancer. Several strategies have been described to induce ferroptosis in this manner, such as manipulation of tumor-suppressor p53 [124,125], cysteine deprivation [126], and inhibition of ferritinophagy, a selective autophagic process capable of repressing accumulation of iron and lipid ROS [127]. In a similar manner, many studies use the collectively termed “ferroptosis inducers” in OSCC as a potential therapeutic approach, essentially by interfering with intracellular iron levels or ROS accumulation [50]. This has been evaluated both in vitro and in vivo by tampering with the signaling cascades that trigger ferroptosis or silencing of ferroptosis inhibitors, which can be achieved through direct genetic modifications or chemical compounds. Some of the compounds used to achieve such effects are erastin [21,42,57], carnosic acid [34], piperlongumine [58] and Disulfiram [63].
Some studies used strategies to induce ferroptosis in OSCC by silencing target genes. One study silenced the circular RNA FNDC3B (circFNDC3B) with interference RNA, which inhibited GPX4 and SLC7A11 expression (negative regulators of ferroptosis), inducing intracellular ROS and iron accumulation [29]. Another study silenced eukaryotic translation initiation factor 3 subunit B (EIF3B), commonly associated with unfavorable head and neck squamous cell carcinoma (HNSCC) prognosis [44]. EIF3B knockdown resulted in decreased invasion and migration in OSCC, as well as induction of cell death [44]. The knockdowns of enhancer of zeste 2 polycomb repressive complex 2 subunit (EZH2) and SLC7A11, both highly expressed in OSCC, resulted in ferroptosis induction [61]. Other target genes whose knockdown promoted ferroptosis were heat shock protein family A (Hsp70) member 5 (HSPA5) [54], adipocyte enhancer-binding protein 1 (AEBP1) [48], glutaredoxin 5 (GLRX5) [23], and miR-7-5p [27]. AEBP1 silencing was especially effective after sulfasalazine treatment, which significantly increased levels of ROS and free intracellular iron [48]. Another study targeting GPX4 with circular RNA showed increased levels of ROS and intracellular iron, meanwhile repressing tumor growth in OSCC cells [29]. Inhibition of GPX4 in Erlotinib-tolerant persisted cancer cells (erPCC) was also effective in increasing sensitivity to ferroptosis [30].
Many studies evaluated the potential of compounds or nanoparticles in controlling tumor progression by inducing ferroptosis. The recent study by Wu et al. [59] reported the effect of A-GSP (aqueous-soluble sporoderm-removed G. lucidum spore power) in tumor suppression by activating ferroptosis, which was confirmed by the assessment of GSH, malondialdehyde (MDA) and ROS levels, as well as ferroptosis-marker expression and mitochondrial morphological alterations—key in confirming the occurrence of this pathway [128]. Additionally, the authors evaluated this compound in vivo in a tumorigenesis assay, and their results showed that there was a decrease in tumor growth among the treated groups while maintaining low toxicity to the treated animals. These results strongly suggest the effectiveness and specificity of this compound in suppressing the tumor through ferroptosis induction. Another study achieved similar ferroptosis induction using nanoparticles termed ginseng-based carbon dots [56]. Another interesting study was performed by Huang et al. [19] using on-oxidized zero-valent iron (ZVI) nanoparticles. The authors treated several oral cancer cell lines with ZVI nanoparticles, and cell death was observed, together with ROS accumulation and mitochondrial damage. However, some cell lines managed to acquire resistance to treatment, and further examination revealed that ferroptosis-related genes were associated with this resistance. Glutathione reductase replenishes cellular GSH stock and circumvents ferroptotic cell death [129]. Therefore, by targeting these cells with ferroptosis-inducers, the authors were able to overcome treatment resistance without affecting the viability of other non-tumoral cells in vitro [19]. These results provide further evidence for the role of ferroptosis as a target while minimizing toxic side effects.
Ferroptosis induction is especially powerful in treatment-resistant cell lines, as was shown in a study that overcame Cetuximab resistance in oral cancer by inducing ferroptosis [52]. It was noticed that the utilization of Cetuximab in resistant cells was not enough to suppress tumor growth or reduce viability. However, when Cetuximab was associated with RSL3, a ferroptosis-inducer agent, there was mitochondrial damage and increased cellular sensitivity to ferroptosis. RSL3 acts by depletion of GTX4, which is responsible for regulating a GTX4 protein depletor, which is, in turn, responsible for reducing the amount of intracellular lipid peroxide [130]. However, the study does not suggest through which mechanism this chemical synergy occurs, despite this effect being described in other studies [131,132]. Ferroptosis can also be induced by hyperbaric oxygen and X-ray radiation in a synergic effect [38]. It has been evaluated as a mechanism to overcome radiotherapy resistance by oral cancer cells, as has been done in other cancers [133]. Another interesting approach is the development of carrier particles to increase intracellular iron and trigger cell death [62]. OSCC-specific nanoprobes carrying iron can be internalized by endocytosis, much like “Trojan horses”. The acidic lysosomal conditions lead to the release of iron from the nanoprobes, inducing ferroptosis [62]. Non-thermal plasma treatment also showed an effect in oral squamous cell carcinoma cells by promoting lipid peroxidation and, ultimately, cell death by ferroptosis [20]. Moreover, studies show that regulation of ferroptosis inhibitor proteins is a promising therapeutic approach, as is the case of caveolin 1 (CAV1) downregulation [40]. CAV1 is known for its ferroptosis-inhibiting capabilities [40]. By knocking down CAV1, authors increased ROS and intracellular iron levels while reducing tumor cell growth in vitro. One of the main obstacles to inducing ferroptosis is assuring target specificity in order to avoid non-tumoral cell toxicity and side effects. In most studies, this has been achieved with a certain degree of success, such as the use of A-GSP, which was able to promote ferroptosis in oral cancer cells by inducing Fe2+ influx and GSH depletion, as well as lipid peroxide and ROS accumulation, while not producing significant toxic side effects [59].
Aside from exploring ferroptosis as a therapeutic target in oral cancer, developing a prognosis model based on ferroptosis-related genes is also useful for directing patient-specific treatments, as has been done in melanoma [134], colon cancer [135], and lung adenocarcinoma [136]. The establishment of prognosis indicators is relevant in the context of oral cancer, as they hold the potential for directing personalized treatment of specific cases [137]. In this context, the pursuit of determining a prognostic model was frequent in several papers, by associating genes/proteins with a better or worse prognosis. Through bioinformatic methods in gene expression detection, several authors were able to identify differentially expressed ferroptosis-related genes in OSCC tissues, used to classify cases according to expression levels. Cases were usually split into two groups: high-risk and low-risk. The cases within the high-risk groups usually correlate with lower overall survival rates [26,47,51]. High-risk groups can show higher expression levels of ferroptosis-related genes (FRGs), considered “risk genes”, which are related to ferroptosis inhibition, or they might show low expression of FRGs, considered “protector” genes, related to ferroptosis promotion. The opposite is also true for the low-risk groups [26,51]. This evidence shows a tumor-suppressing role of ferroptosis in oral cancer: when inhibited or impaired, overall survival rates drop.
Gu, Kim, Wang et al. [25] developed the FPscore, a ferroptosis-specific gene-expression signature linked to outcomes and clinical relevance. High FPscore groups in OSCC were associated with better prognosis, activation of a ferroptosis-related immune phenotype, and better response to chemo and immunotherapy. Overall, common findings of prognosis-related ferroptosis-associated genes were ferritin heavy chain 1 (FTH1) [47], autophagy related 5 (ATG5) [26,51], arachidonate 15-lipoxygenase (ALOX15) [26,51], microtubule associated protein 1 light chain 3 alpha (MAP1LC3A) [26,51], mitogen-activated protein kinase kinase kinase 5 (MAP3K5) [26], and carbonic anhydrase 9 (CA9) [49,51]. According to Yin et al. [47], the main genes whose increased expression would be correlated with poor prognosis are, besides FTH1 and ATG5, BCL2 interacting protein 3 (BNIP3) and peroxiredoxin 6 (PRDX6). On the other hand, increased expression of MAP1LC3A, MAP3K5 and suppressor of cytokine signaling 1 (SOCS1) are related to a better prognosis [51]. Gene enrichment strategies were also relevant in mapping the association between ferroptosis and other functional and metabolic pathways in oral cancer. Overall, findings indicated that the ferroptosis-related gene signature may relate to the dysregulation of cancer-related and immune-related pathways [25,26,51]. Low-risk groups presented a higher immune cell content in the tumor microenvironment [47]. Better prognosis and treatment response are associated with a higher “immunoscore” in oral cancer due to the presence of T cells and macrophages [25,138] and the expression of HLA molecules [47]. Research in OSCC has shown that overexpression of immune checkpoint receptors such as indoleamine 2,3-dioxygenase 1 (IDO1) and programmed cell death ligand 1 (PD-L1) has been correlated with better response to treatment with pembrolizumab and nimotuzumab [25,139,140]. The opposite was also seen in high-risk groups, which showed downregulated expression of immune-related components [25,26]. In the same manner, it is possible to establish a link between response to immunotherapy and several ferroptosis-related prognosis genes, such as FTH1, fms-related receptor tyrosine kinase 3 (FLT3), cyclin-dependent kinase inhibitor 2A (CDKN2A), and DNA damage inducible transcript 3 (DDIT3) [47], which also relate to the tumor’s immunoscore [25,47]. Gene-signature models were also constructed with long-non-coding RNA, and high-risk groups were successfully correlated with lower overall survival and lower immunogenic score [36,41]. Regarding chemotherapy, several studies identified the association between the established ferroptosis-related gene signature and higher or lower treatment success rates, which can help personalize the therapeutic protocol in different profile cases. In the case of the FPscore [25], higher expression of FRGs (higher FPscore) was associated with higher chemosensitivity, while low expression was associated with drug resistance [25].
In summary, several studies have shown promising results in using the existent ferroptosis-related cellular machinery as potential therapeutic strategies. Both knockdown strategies targeting key proteins involved in promoting/regulating this type of cell death and application of compounds with activity to induce ferroptosis of the OSCC cells were used. Additionally, the characterization of several gene signatures of ferroptosis-associated genes for both prognostication and response to treatment is relevant, but further verification in large-scale clinical studies is required. Thereafter, the ongoing exploration of ferroptosis in oral cancer not only deepens our understanding of cancer mechanisms but also holds the potential to translate this knowledge to improve patient outcomes.

4.2. Pyroptosis

Pyroptosis, an inflammatory type of caspase-mediated cell death, can modulate the immunogenic potential of specific cancers [82]. The role of pyroptosis in OSCC is an area of ongoing research, and the mechanisms and implications are not fully understood, but there is evidence to suggest that pyroptosis may play a role in both development and progression of OSCC [76,77,78,79,80,81,82,83]. Pyroptosis derives its name from the combination of “pyro” and “ptosis”. “Pyro” signifies fire, highlighting its inflammatory properties, while “ptosis” refers to falling, which aligns with other forms of programmed cell death [141]. There are notable similarities between pyroptosis and apoptosis, including features like DNA damage and chromatin condensation [142]. Interestingly, pyroptotic cells exhibit swelling and numerous bubble-like protrusions on the cellular membrane before rupture, a phenomenon reminiscent of membrane blebbing observed in apoptosis. Pyroptosis was officially defined as Gasdermin-mediated programmed cell death in 2015 [141]. The Gasdermin superfamily in humans includes Gasdermin A/B/C/D (GSDMA/B/C/D), Gasdermin E (GSDME, also known as DFNA5), and DFNB59 (Pejvakin, PJVK). Inflammasomes are responsible for initiating pyroptosis via two distinct pathways (Figure 3). The canonical inflammasome pathway is reliant on the activation of caspase-1, and the noncanonical inflammasome pathway involves the activation of caspase-4, caspase-5, or caspase-11 [143]. Furthermore, certain studies have demonstrated that proapoptotic caspase-3 activation can also initiate pyroptosis by cleaving GSDME [144,145].
Inflammation is a critical component of tumor progression [146], and inflammation intensified by chemotherapy can lead to therapy failure and metastasis [147]. In the Nod-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome is one of the critical components of the innate immune system and plays an important role in cancer [148,149]. Many factors can activate NLRP3 inflammasomes, including potassium efflux, intracellular calcium, endoplasmic reticulum (ER) stress and ROS [149]. Chronic inflammation has the potential to impact every phase of the carcinogenic process, increasing the risk of tumorigenesis with prolonged exposure to an inflammatory milieu [150]. Pyroptosis, as a form of lytic cell death, amplifies the release of mature interleukin-1 (IL-1) and interleukin-18 (IL-18), potentially influencing the development of cancer [151]. Furthermore, pyroptosis serves as the mechanism for inflammatory cell death in cancer cells, thereby restraining the proliferation and migration of these cancer cells [28,33,68,69,70,78,79]. Consequently, pyroptosis assumes a dual role, both promoting and inhibiting tumorigenesis [152]. Previous findings revealed that 5-fluorouracil (5-FU) treatment increased NLRP3 expression in OSCC, which mediated drug resistance. It was also proven that NLRP3 could promote tumor growth and metastasis in OSCC [64,69,153]. Activation of pyroptosis has also been directly associated with increased chemoresistance to cisplatin and 5-FU treatment [64], and inhibition of pyroptosis has been associated with increased sensitivity of neoplastic cells to cisplatin treatment [66].
Methods of pyroptosis inhibition are gaining the attention of the scientific community [65,69,74,154]. Yang et al. [69] highlighted that extracellular vesicles derived from bitter melon led to a significant decrease in the expression of NLRP3, reducing OSCC resistance to 5-FU treatment. The study developed by Yue et al. [65] evaluated the effect of anthocyanin on OSCC. Anthocyanin reduced the viability of OSCC cells and inhibited migration and invasion capacity, concomitantly increasing pyroptosis. Simultaneously, activation of pyroptosis was associated with increased expression of NLRP3, caspase-1 and interleukin-1β (IL-1β). After administration of caspase-1 inhibitors, anthocyanin-activated pyroptosis was suppressed, and cell viability, migration and invasion rates increased concomitantly. In vitro studies in monoculture do not show the real dimension of the role of pyroptosis in OSCC. Therefore, conflicting results can be seen depending on the methodology used in different studies. The poor prognosis of pyroptosis is associated with its ability to activate inflammation; however, the development of in vitro and in vivo studies capable of more broadly evaluating the tumor microenvironment and all mechanisms triggered by the activation of pyroptosis should be encouraged. The inflammation associated with pyroptosis can lead to the recruitment of immune cells and other factors that support tumor growth and metastasis [154]. The pro-inflammatory environment can also contribute to resistance to therapy and promote angiogenesis, which is the formation of new blood vessels that supply nutrients to the tumor. The study developed by Xin et al. [74] used The Cancer Genome Atlas (TCGA) dataset to investigate the predictive value of pyroptosis-related lncRNAs in the prognosis of OSCC. The authors identified eight pyroptosis-related lncRNAs associated with overall survival in patients with OSCC by multivariate regression analysis.
Taken together, our analysis indicates that the number of studies exploring pyroptosis in OSCC is still restricted, limiting our knowledge of the mechanisms of how molecules related to this type of cell death affect OSCC cells. However, the studies highlighted the potential of pyroptosis in the control of OSCC development and progression and described drugs and molecules related to both blockage and induction of it. Moreover, two studies demonstrated that pyroptosis gene clusters are correlated with clinical characteristics, infiltration of immune cells, susceptibility to chemotherapy and immunotherapy, and prognosis of patients with OSCC [74,75]. It will be important to validate these risk models and to explore potential drugs that could induce cell death through pyroptosis and concomitantly inhibit the pro-tumor inflammatory response in OSCCs.

4.3. Necroptosis

Necroptosis is considered a programmed form of necrosis mediated by receptor-interacting protein kinase 1 (RIPK1) and receptor-interacting protein kinase 3 (RIPK3) [155,156]. The stimulus is initiated by tumor necrosis factor (TNF) binding to its receptors [157,158]. The interaction of ligand and receptor leads to the formation of a signaling complex, which may include adaptor proteins such as Fas-associated protein with death domain (FADD) and TNF receptor-associated death domain (TRADD). RIPK1 is recruited to the signaling complex and is activated by phosphorylation. Depending on cellular conditions, RIPK1 can associate with caspases, promoting apoptosis, or with RIPK3, initiating the necroptosis pathway. If the necroptosis pathway is activated, RIPK1 recruits RIPK3 and the protein MLKL, forming the necrosome complex. RIPK3 phosphorylates MLKL, activating it. Activated MLKL oligomerizes and translocates to the plasma membrane, where it causes damage, leading to membrane rupture and necroptosis (Figure 4) [159].
The formation of an MLKL oligomer opens a pore in the membrane, allowing the entrance of ROS and DAMPs [160]. In this manner, death by necroptosis, despite being activated by specific signals, leads to a similar end as necrosis and ferroptosis, triggering the entrance of ROS in the cell and leading to membrane damage [160,161]. To allow for necroptosis to occur, the apoptotic pathway must be impaired or damaged [155,162,163], which can be quite common in cancer since the inhibition of healthy apoptosis as a control allows for the unchecked reproduction of damaged cells [164]. This phenomenon holds significant implications for oral cancer, where dysregulation of cell death mechanisms can tip the balance in favor of tumor progression.
In this context, necroptosis, which is characterized by a regulated inflammatory response, becomes a last-resort mechanism to eliminate aberrant cells [165,166]. However, cancer cells can hijack this mechanism to promote their survival and evade the body’s natural defenses, using pro-tumoral inflammation to their advantage [165]. Additionally, necroptosis can generate an immunosuppressive tumor microenvironment, which may further contribute to cancer cell survival and progression [167]. It comes as no surprise that the expression level of key mediators of necroptosis is elevated in cancer [165,168], indicating that necroptosis may play a role in promoting oncogenesis and cancer metastasis [169]. However, it is possible to interfere in this process by targeting necroptosis as an ally in halting cancer progression and survival, as has been evaluated in oral cancer studies, by targeting focal adhesion molecules [170,171] or even caspase-8 itself, which is responsible for deciding the pathway outcome of apoptosis versus necrosis [170,172]. In the context of OSCC, studies have explored the induction of necroptosis using different agents, such as Obatoclax [84]. This agent targets members of the BCL-2 family, specifically the myeloid cell leukemia sequence 1 (MCL-1). The study suggests that Obatoclax induces cell death in OSCC cells through autophagy-dependent necroptosis [173], with mitochondrial stress and dysfunction as detectable upstream events. Additionally, capsaicin was found to inhibit cell proliferation and induce endoplasmic reticulum stress and autophagy in oral cancer [87]. This mechanism negatively regulates ribophorin II, impairing P-glycoprotein functions and sensitizing cells to anticancer therapy [174]. The association of capsaicin with anticancer agents promotes necroptosis rather than apoptosis, showcasing a unique pathway for inhibiting OSCC cell viability. Similarly, chelerythrine chloride (CS) demonstrated necroptosis induction in OSCC, impairing cell proliferation and inducing morphological alterations in a dose-dependent manner, such as membrane rupture, and dose-dependent cell death [87]. Moreover, the development of targeted delivery systems such as PLGA-Dtx (poly-lactic-co-glycolic acid nanoparticles containing docetaxel) has shown enhanced efficacy in inhibiting cancer cell proliferation. This strategy induced both apoptosis and necroptosis in oral cancer cells [89]. These results altogether suggest a potential role for these compounds in triggering necroptosis in OSCC cells.
Studies on necroptosis-related genes may reveal potential targets, either enhancers or inhibitors of this NAPCD. HNSCC studies have associated CASP8 mutations with radioresistance and poor survival outcomes [86], as is the case for OSCC [175]. In this manner, knockdown of CASP8 enhances the radiosensitizing effects of certain compounds through the induction of necroptosis. Additionally, as seen in other cell death mechanisms, the investigation of necroptosis-related genes that may be relevant in prognosis prediction has also been explored. Bioinformatic analyses and in vitro experiments identified six genes (hypoxanthine phosphoribosyltransferase 1-HPRT1, PGAM family member 5, mitochondrial serine/threonine protein phosphatase-PGAM5, BH3 interacting domain death agonist-BID, survival of motor neuron 1, telomeric-SMN1, FADD, and KIAA1191) contributing to OSCC development, metastasis, and immune modulation. These genes may play a role in the regulation of cell death pathways, including necroptosis [90]. Moreover, a study in HNSCC emphasized the prevalence of necroptosis and its association with poor overall survival and progression-free survival. Approximately half of the necrosis in HNSCC was attributed to necroptosis, indicating its significance as an independent risk factor for adverse clinical outcomes [85].
In summary, necroptosis induction is a promising alternative in the treatment of oral cancers, and the expression of necroptosis-related genes depicts prognostic potential for predicting OSCC outcomes. However, one of the main challenges in inducing necroptosis as a cancer treatment strategy remains in establishing specificity in a manner by which little toxicity is archived [172].

4.4. Other Emerging Types of Cell Death

Other forms of NAPCD, such as NETosis [93,94], parthanatos [96], mitoptosis [92] and paraptosis [95], were also investigated in OSCC.
The term “NETotic cell death” refers to a somewhat controversial form of NAPCD initially identified in neutrophils due to its association with the extrusion of a meshwork composed of chromatin and histone fibers bound to granular and cytoplasmic proteins [176]. This structure is known as neutrophil extracellular traps (NETs), a process commonly referred to as NETosis [177,178]. NETs, generated in response to various microbial and sterile activators or upon the stimulation of specific receptors such as Toll-like receptors (TLRs), essentially serve as a stable extracellular platform for capturing and breaking down microbes [179]. Several reports indicate that a significant portion of the nucleic acids present in NETs originates from mitochondria rather than the cell nucleus [179]. In addition to their antimicrobial effects, NETs are reported to contribute to the development of certain human pathologies, including diabetes and cancer [178]. It is worth noting that structures resembling NETs can be released by cell types other than neutrophils, including mast cells, eosinophils, and basophils [3]. Garley et al. [93], aiming to evaluate the role of NETosis in oral cancer, evaluated the following groups of patients: (1) 10 patients with odontogenic infection/inflammation, (2) 17 patients with OSCC and (3) 15 healthy people (blood donor volunteers). In the study, neutrophils from patients with inflammation and individuals with oral cancer produced increased amounts of NETs. Garley et al. [94] observed an increase in NET formation when co-culturing an oral cancer cell line (CAL-27). Recent study findings highlight the crucial role of circulating cancer cells (CTCs) in interacting with and modulating the blood microenvironment for metastatic development. It has been demonstrated that neutrophils mobilize and accumulate at future metastatic sites, releasing NETs that bind to CTCs. This process contributes to the creation of a “pre-metastatic niche” and supports the development of tumors with aggressive phenotypes. Consequently, the increased formation of NETs in oral cancer may carry significant implications for its biological behavior, serving as an indicator of a worse prognosis. While studies in tumor models have yielded satisfactory results, research involving actual tumor patients has not been as successful. There is a need to intensify research efforts toward achieving a better understanding of the regulation and formation of NETosis, focusing on considering NETosis as a therapeutic target without compromising immune function [3,179].
Parthanatos is a type of NAPCD triggered by the hyperactivation of a specific component of the DNA damage response (DDR) machinery, such as poly(ADP-ribose) polymerase 1 (PARP1) [180]. Notably, parthanatos appears to occur not only as a result of severe or prolonged alkylating DNA damage but also in response to oxidative stress, hypoxia, hypoglycemia, or inflammatory signals [3]. Existing studies have demonstrated a close association between parthanatos and tumorigenesis development. In one study, microarray analysis was conducted on PARP-1 gene expression in over 8000 tumor samples [181], revealing higher expression levels of PARP-1 in breast, ovarian, endometrial, lung and skin cancers compared to equivalent amounts of normal tissues. This suggests a relationship between parthanatos and these tumors. Moreover, the construction of PARP-1 knockout mice showed a significant reduction in the risk of epithelial cancer in these mice [181]. The study developed by Li et al. [96] evaluated the effect of oxaliplatin (a new third-generation platinum-based chemotherapy drug); the authors pointed out that oxaliplatin can increase the production of ROS and then can induce the overactivation of PARP1, the depolarization of mitochondria and the nuclear translocation of apoptosis-inducing factor (AIF) and macrophage migration inhibitory factor (MIF), leading to parthanatos in oral cancer. However, the authors did not assess the impact of parthanatos on inducing inflammation. The impact of parthanatos on oral cancer remains an underexplored area, requiring further study.
The process of mitoptosis, or the death program affecting mitochondria, is a relatively poorly understood phenomenon primarily characterized by its morphological features. The induction of mitoptosis, coupled with the disruption of ATP supply by mitochondria, is often accompanied by the activation of autophagy to ensure the maintenance of the energy supply [182,183]. Mitoptosis can manifest in various forms; for instance, an inner membrane mitoptosis may occur, during which only the internal matrix and cristae undergo degradation while the external mitochondrial envelope remains unchanged. Alternatively, an outer membrane mitoptosis may occur, where only swollen internal cristae are detected as remnants. In the study developed by Ruggieri et al. [92], after treatment with dichloroacetate, strong mitochondrial fragmentation was observed in HSC-2 oral cancer lines and, to a lesser extent, in HSC-3 cells. The study data presented indicates that dichloroacetate can make oral cancer cell lines more sensitive to cancer treatment via mitochondrial damage.
Paraptosis is an alternative cell death pathway distinguished by vacuolation and damage to the endoplasmic reticulum and mitochondria [184]. The study conducted by Chen et al. [95] investigated the impact of isorhamnetin, a flavonoid, in OSCC cell lines. The findings demonstrated a dose- and time-dependent inhibition of cell proliferation, as evidenced by reduced cell viability and decreased cell colonies. The study also revealed cell cycle arrest in the G2/M phase, accompanied by the suppression of cyclin B1 and CDC2 protein levels. Moreover, the research showed inhibition of cell migration with modulation of related protein levels. Notably, the presence of abundant cytoplasmic vacuoles, originating from mitochondria and the endoplasmic reticulum, was observed. Importantly, the study confirmed that cell death did not occur through apoptosis but suggested a propensity towards paraptosis. Isorhamnetin was found to upregulate phosphorylated extracellular regulated MAP kinase (ERK) cascades and elevate intracellular reactive oxygen species levels. The collective results of the study suggest that the induction of paraptosis holds promising potential for promoting cell death in oral cancer.
Some limitations of this review should be considered. First, from the eight types of NAPCD explored in this study, only ferroptosis and pyroptosis were reported in several studies involving in vitro, in vivo and in silico approaches. However, even for those, several studies had to be excluded due to lack of complete data reporting, and the heterogeneity of the data did not allow any type of quantitative analysis. Another limitation arises from the variety of terminology used in the past to define the types of cell death under NAPCD, in the period in which the biological features and markers were not well-defined. This inconsistency in nomenclature could lead to missing articles. Finally, regarding the risk of bias from the included studies, no information about blinding was reported by the majority of the in vitro studies, and most of the in vivo studies did not provide sufficient information to assess the strategies of selection, allocation concealment, and the methods of performance and evaluation of the outcomes, reducing the certainty of evidence. The large majority of the in silico studies exploring the prognostic potential of emerging subtypes of NAPCD used the OSCC cohort from TCGA, applying different pipelines to describe prognostic cancer gene expression signatures. As validation in independent, large and multicenter cohorts was limited, the translational potential requires further examination. In this scenario, false-positive discoveries cannot be ignored.

5. Conclusions

Exploring novel NAPCD modalities holds considerable potential for identifying novel prognostic markers and/or therapeutic targets for OSCC. NAPCD can also make tumors more responsive to immunotherapy by regulating tumor immunogenicity and enhancing lymphocyte infiltration in the tumor microenvironment. In spite of the discovery of many compounds and agents that induce or modulate NAPCD programming, exerting strong antitumor effects, more well-designed studies are needed to improve the certainty of evidence. Our review postulates that understanding the role of novel types of tumor cell death may have potential in cancer treatment, and we encourage future studies using animal models or more complex in vitro models to identify therapeutic opportunities. Furthermore, we expect that more clinical trials will be carried out investigating the use of new agents modulating cell death in patients with OSCC.

Author Contributions

Conceptualization, C.A.G.R. and R.D.C.; methodology, L.d.O.S.d.R., E.F.d.M., L.Q.R.d.O., C.A.G.R. and R.D.C.; validation, D.W.L., C.A.G.R. and R.D.C.; formal analysis, L.d.O.S.d.R., E.F.d.M., L.Q.R.d.O., A.V.B., C.A.G.R. and R.D.C.; investigation, L.d.O.S.d.R., E.F.d.M., L.Q.R.d.O., A.V.B., C.A.G.R. and R.D.C.; resources, D.W.L., C.A.G.R. and R.D.C.; data curation, L.d.O.S.d.R., E.F.d.M., L.Q.R.d.O., A.V.B., C.A.G.R. and R.D.C.; writing—original draft preparation, L.d.O.S.d.R., E.F.d.M., L.Q.R.d.O., A.V.B., C.A.G.R. and R.D.C.; writing—review and editing, L.d.O.S.d.R., E.F.d.M., L.Q.R.d.O., A.V.B., D.W.L., C.A.G.R. and R.D.C.; visualization, L.d.O.S.d.R., E.F.d.M., L.Q.R.d.O., A.V.B., D.W.L., C.A.G.R. and R.D.C.; supervision, D.W.L., C.A.G.R. and R.D.C.; project administration, D.W.L., C.A.G.R. and R.D.C.; funding acquisition, C.A.G.R. and R.D.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Academy of Medical Sciences/Newton Advanced Fellowship Grant (NAFR12n1035) and the Program for Excellence in Research of the Oswaldo Cruz Foundation (PROEP-FIOCRUZ-BA) (02.385.669/0001-74, ID 2034). L.d.O. Siquara da Rocha is a research student supported by the Conselho Nacional de Desenvolvimento Científico e Tecnológico–CNPq Brasil (140066/2022-5). E.F. de Moraes is a research fellow supported by Fundação de Amparo à Pesquisa do Estado de São Paulo-FAPESP (2022/00994-5).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

Appendix A. Search Strategies with Key Words and MeSH Terms

  • EMBASE
  • Search 1: ENTOSIS (Results: 7)
((‘mouth cancer’/exp OR ‘cancer, mouth’ OR ‘intraoral cancer’ OR ‘mouth mucosa cancer’ OR ‘oral cancer’ OR ‘oral cavity cancer’ OR ‘mouth cancer’) OR (‘head and neck cancer’/exp OR ‘cancer, head and neck’ OR ‘cervicofacial cancer’ OR ‘ear nose throat cancer’ OR ‘ENT cancer’ OR ‘head neck cancer’ OR ‘ORL cancer’ OR ‘otorhinolaryngeal cancer’ OR ‘otorhinolaryngologic cancer’ OR ‘otorhinolaryngological cancer’ OR ‘head and neck cancer’)) AND (‘entosis’/exp).
  • Search 2: FERROPTOSIS (Results: 265)
((‘mouth cancer’/exp OR ‘cancer, mouth’ OR ‘intraoral cancer’ OR ‘mouth mucosa cancer’ OR ‘oral cancer’ OR ‘oral cavity cancer’ OR ‘mouth cancer’) OR (‘head and neck cancer’/exp OR ‘cancer, head and neck’ OR ‘cervicofacial cancer’ OR ‘ear nose throat cancer’ OR ‘ENT cancer’ OR ‘head neck cancer’ OR ‘ORL cancer’ OR ‘otorhinolaryngeal cancer’ OR ‘otorhinolaryngologic cancer’ OR ‘otorhinolaryngological cancer’ OR ‘head and neck cancer’)) AND (‘ferroptosis’/exp OR ‘ferroptotic cell death’ OR ‘ferroptotic death’ OR ‘ferroptosis’).
  • Search 3: PYROPTOSIS (Results: 124)
((‘mouth cancer’/exp OR ‘cancer, mouth’ OR ‘intraoral cancer’ OR ‘mouth mucosa cancer’ OR ‘oral cancer’ OR ‘oral cavity cancer’ OR ‘mouth cancer’) OR (‘head and neck cancer’/exp OR ‘cancer, head and neck’ OR ‘cervicofacial cancer’ OR ‘ear nose throat cancer’ OR ‘ENT cancer’ OR ‘head neck cancer’ OR ‘ORL cancer’ OR ‘otorhinolaryngeal cancer’ OR ‘otorhinolaryngologic cancer’ OR ‘otorhinolaryngological cancer’ OR ‘head and neck cancer’)) AND (‘pyroptosis’/exp OR ‘caspase 1-dependent cell death’ OR ‘caspase-1 dependent cell death’ OR ‘caspase-1-dependent cell death’ OR ‘inflammatory apoptosis’ OR ‘pyroptotic cell death’ OR ‘pyroptotic cellular death’ OR ‘pyroptotic death’ OR ‘pyroptotic inflammatory cell death’ OR ‘pyroptosis’).
  • Search 4: NETOSIS (Results: 7)
((‘mouth cancer’/exp OR ‘cancer, mouth’ OR ‘intraoral cancer’ OR ‘mouth mucosa cancer’ OR ‘oral cancer’ OR ‘oral cavity cancer’ OR ‘mouth cancer’) OR (‘head and neck cancer’/exp OR ‘cancer, head and neck’ OR ‘cervicofacial cancer’ OR ‘ear nose throat cancer’ OR ‘ENT cancer’ OR ‘head neck cancer’ OR ‘ORL cancer’ OR ‘otorhinolaryngeal cancer’ OR ‘otorhinolaryngologic cancer’ OR ‘otorhinolaryngological cancer’ OR ‘head and neck cancer’)) AND (‘NETosis’/exp OR ‘ENTosis cell death’ OR NETosis cellular death’ OR ‘NETosis death’).
  • Search 5: NECROPTOSIS (Results: 211)
((‘mouth cancer’/exp OR ‘cancer, mouth’ OR ‘intraoral cancer’ OR ‘mouth mucosa cancer’ OR ‘oral cancer’ OR ‘oral cavity cancer’ OR ‘mouth cancer’) OR (‘head and neck cancer’/exp OR ‘cancer, head and neck’ OR ‘cervicofacial cancer’ OR ‘ear nose throat cancer’ OR ‘ENT cancer’ OR ‘head neck cancer’ OR ‘ORL cancer’ OR ‘otorhinolaryngeal cancer’ OR ‘otorhinolaryngologic cancer’ OR ‘otorhinolaryngological cancer’ OR ‘head and neck cancer’)) AND (‘Necroptosis’/exp OR ‘Necroptosis cell death’ OR ‘Necroptosis cellular death’ OR ‘Necroptosis death’).
  • Search 6: PARTHANATOS (Results: 0)
((‘mouth cancer’/exp OR ‘cancer, mouth’ OR ‘intraoral cancer’ OR ‘mouth mucosa cancer’ OR ‘oral cancer’ OR ‘oral cavity cancer’ OR ‘mouth cancer’) OR (‘head and neck cancer’/exp OR ‘cancer, head and neck’ OR ‘cervicofacial cancer’ OR ‘ear nose throat cancer’ OR ‘ENT cancer’ OR ‘head neck cancer’ OR ‘ORL cancer’ OR ‘otorhinolaryngeal cancer’ OR ‘otorhinolaryngologic cancer’ OR ‘otorhinolaryngological cancer’ OR ‘head and neck cancer’)) AND (‘Parthanatos’/exp OR ‘Parthanatos cell death’ OR ‘Parthanatos cellular death’ OR ‘Parthanatos death’).
  • Search 7: MITOPTOSIS (Results: 7)
((‘mouth cancer’/exp OR ‘cancer, mouth’ OR ‘intraoral cancer’ OR ‘mouth mucosa cancer’ OR ‘oral cancer’ OR ‘oral cavity cancer’ OR ‘mouth cancer’) OR (‘head and neck cancer’/exp OR ‘cancer, head and neck’ OR ‘cervicofacial cancer’ OR ‘ear nose throat cancer’ OR ‘ENT cancer’ OR ‘head neck cancer’ OR ‘ORL cancer’ OR ‘otorhinolaryngeal cancer’ OR ‘otorhinolaryngologic cancer’ OR ‘otorhinolaryngological cancer’ OR ‘head and neck cancer’)) AND (‘Mitoptosis’/exp OR ‘Mitoptosis cell death’ OR ‘Mitoptosis cellular death’ OR ‘Mitoptosis death’).
  • Search 8: PARAPTOSIS (Results: 21)
((‘mouth cancer’/exp OR ‘cancer, mouth’ OR ‘intraoral cancer’ OR ‘mouth mucosa cancer’ OR ‘oral cancer’ OR ‘oral cavity cancer’ OR ‘mouth cancer’) OR (‘head and neck cancer’/exp OR ‘cancer, head and neck’ OR ‘cervicofacial cancer’ OR ‘ear nose throat cancer’ OR ‘ENT cancer’ OR ‘head neck cancer’ OR ‘ORL cancer’ OR ‘otorhinolaryngeal cancer’ OR ‘otorhinolaryngologic cancer’ OR ‘otorhinolaryngological cancer’ OR ‘head and neck cancer’)) AND (‘Paraptosis’/exp OR ‘ Paraptosis cell death’ OR ‘Paraptosis cellular death’ OR ‘Paraptosis death’).
  • Search 9: METHUOSIS (Results: 12)
((‘mouth cancer’/exp OR ‘cancer, mouth’ OR ‘intraoral cancer’ OR ‘mouth mucosa cancer’ OR ‘oral cancer’ OR ‘oral cavity cancer’ OR ‘mouth cancer’) OR (‘head and neck cancer’/exp OR ‘cancer, head and neck’ OR ‘cervicofacial cancer’ OR ‘ear nose throat cancer’ OR ‘ENT cancer’ OR ‘head neck cancer’ OR ‘ORL cancer’ OR ‘otorhinolaryngeal cancer’ OR ‘otorhinolaryngologic cancer’ OR ‘otorhinolaryngological cancer’ OR ‘head and neck cancer’)) AND (‘Methuosis’/exp OR ‘Methuosis cell death’ OR ‘Methuosis cellular death’ OR ‘Methuosis death’).
  • PUBMED
  • Search 1: ENTOSIS (Results: 3)
((“Mouth Neoplasms”[Mesh]) OR (Mouth Neoplasm) OR (Neoplasm, Mouth) OR (Neoplasms, Oral) OR (Neoplasm, Oral) OR (Oral Neoplasm) OR (Oral Neoplasms) OR (Neoplasms, Mouth) OR (Cancer of Mouth) OR (Mouth Cancers) OR (Oral Cancer) OR (Cancer, Oral) OR (Cancers, Oral) OR (Oral Cancers) OR (Cancer of the Mouth) OR (Mouth Cancer) OR (Cancer, Mouth) OR (Cancers, Mouth) OR (“Head and Neck Neoplasms”[Mesh]) OR (Neoplasms, Head and Neck) OR (Head, Neck Neoplasms) OR (Head and Neck Neoplasm) OR (Cancer of Head and Neck) OR (Head and Neck Cancer) OR (Cancer of the Head and Neck) OR (Upper Aerodigestive Tract Neoplasms) OR (UADT Neoplasm) OR (Neoplasm, UADT) OR (Neoplasms, UADT) OR (UADT Neoplasms) OR (Neoplasms, Upper Aerodigestive Tract) OR (Upper Aerodigestive Tract Neoplasm) OR (Head Neoplasms) OR (Neoplasms, Head) OR (Head Neoplasm) OR (Neoplasm, Head) OR (Neck Neoplasms) OR (Neoplasms, Neck) OR (Neck Neoplasm) OR (Neoplasm, Neck) OR (Cancer of Head) OR (Head Cancers) OR (Head Cancer) OR (Cancer, Head) OR (Cancers, Head) OR (Cancer of the Head) OR (Cancer of Neck) OR (Neck Cancers) OR (Neck Cancer) OR (Cancer, Neck) OR (Cancers, Neck) OR (Cancer of the Neck)) AND ((“Entosis”[Mesh]) OR (Entoses)).
  • Search 2: FERROPTOSIS (Results: 256)
((“Mouth Neoplasms”[Mesh]) OR (Mouth Neoplasm) OR (Neoplasm, Mouth) OR (Neoplasms, Oral) OR (Neoplasm, Oral) OR (Oral Neoplasm) OR (Oral Neoplasms) OR (Neoplasms, Mouth) OR (Cancer of Mouth) OR (Mouth Cancers) OR (Oral Cancer) OR (Cancer, Oral) OR (Cancers, Oral) OR (Oral Cancers) OR (Cancer of the Mouth) OR (Mouth Cancer) OR (Cancer, Mouth) OR (Cancers, Mouth) OR (“Head and Neck Neoplasms”[Mesh]) OR (Neoplasms, Head and Neck) OR (Head, Neck Neoplasms) OR (Head and Neck Neoplasm) OR (Cancer of Head and Neck) OR (Head and Neck Cancer) OR (Cancer of the Head and Neck) OR (Upper Aerodigestive Tract Neoplasms) OR (UADT Neoplasm) OR (Neoplasm, UADT) OR (Neoplasms, UADT) OR (UADT Neoplasms) OR (Neoplasms, Upper Aerodigestive Tract) OR (Upper Aerodigestive Tract Neoplasm) OR (Head Neoplasms) OR (Neoplasms, Head) OR (Head Neoplasm) OR (Neoplasm, Head) OR (Neck Neoplasms) OR (Neoplasms, Neck) OR (Neck Neoplasm) OR (Neoplasm, Neck) OR (Cancer of Head) OR (Head Cancers) OR (Head Cancer) OR (Cancer, Head) OR (Cancers, Head) OR (Cancer of the Head) OR (Cancer of Neck) OR (Neck Cancers) OR (Neck Cancer) OR (Cancer, Neck) OR (Cancers, Neck) OR (Cancer of the Neck)) AND ((“Ferroptosis”[Mesh]) OR (Oxytosis)).
  • Search 3: PYROPTOSIS (Results: 77)
((“Mouth Neoplasms”[Mesh]) OR (Mouth Neoplasm) OR (Neoplasm, Mouth) OR (Neoplasms, Oral) OR (Neoplasm, Oral) OR (Oral Neoplasm) OR (Oral Neoplasms) OR (Neoplasms, Mouth) OR (Cancer of Mouth) OR (Mouth Cancers) OR (Oral Cancer) OR (Cancer, Oral) OR (Cancers, Oral) OR (Oral Cancers) OR (Cancer of the Mouth) OR (Mouth Cancer) OR (Cancer, Mouth) OR (Cancers, Mouth) OR (“Head and Neck Neoplasms”[Mesh]) OR (Neoplasms, Head and Neck) OR (Head, Neck Neoplasms) OR (Head and Neck Neoplasm) OR (Cancer of Head and Neck) OR (Head and Neck Cancer) OR (Cancer of the Head and Neck) OR (Upper Aerodigestive Tract Neoplasms) OR (UADT Neoplasm) OR (Neoplasm, UADT) OR (Neoplasms, UADT) OR (UADT Neoplasms) OR (Neoplasms, Upper Aerodigestive Tract) OR (Upper Aerodigestive Tract Neoplasm) OR (Head Neoplasms) OR (Neoplasms, Head) OR (Head Neoplasm) OR (Neoplasm, Head) OR (Neck Neoplasms) OR (Neoplasms, Neck) OR (Neck Neoplasm) OR (Neoplasm, Neck) OR (Cancer of Head) OR (Head Cancers) OR (Head Cancer) OR (Cancer, Head) OR (Cancers, Head) OR (Cancer of the Head) OR (Cancer of Neck) OR (Neck Cancers) OR (Neck Cancer) OR (Cancer, Neck) OR (Cancers, Neck) OR (Cancer of the Neck)) AND ((“Pyroptosis”[Mesh]) OR (Pyroptoses) OR (Pyroptotic Cell Death) OR (Cell Death, Pyroptotic) OR (Cell Deaths, Pyroptotic) OR (Death, Pyroptotic Cell) OR (Deaths, Pyroptotic Cell) OR (Pyroptotic Cell Deaths) OR (Caspase-1 Dependent Cell Death) OR (Caspase 1 Dependent Cell Death) OR (Inflammatory Apoptosis) OR (Apoptoses, Inflammatory) OR (Apoptosis, Inflammatory) OR (Inflammatory Apoptoses)).
  • Search 4: NETOSIS (Results: 2)
((“Mouth Neoplasms”[Mesh]) OR (Mouth Neoplasm) OR (Neoplasm, Mouth) OR (Neoplasms, Oral) OR (Neoplasm, Oral) OR (Oral Neoplasm) OR (Oral Neoplasms) OR (Neoplasms, Mouth) OR (Cancer of Mouth) OR (Mouth Cancers) OR (Oral Cancer) OR (Cancer, Oral) OR (Cancers, Oral) OR (Oral Cancers) OR (Cancer of the Mouth) OR (Mouth Cancer) OR (Cancer, Mouth) OR (Cancers, Mouth) OR (“Head and Neck Neoplasms”[Mesh]) OR (Neoplasms, Head and Neck) OR (Head, Neck Neoplasms) OR (Head and Neck Neoplasm) OR (Cancer of Head and Neck) OR (Head and Neck Cancer) OR (Cancer of the Head and Neck) OR (Upper Aerodigestive Tract Neoplasms) OR (UADT Neoplasm) OR (Neoplasm, UADT) OR (Neoplasms, UADT) OR (UADT Neoplasms) OR (Neoplasms, Upper Aerodigestive Tract) OR (Upper Aerodigestive Tract Neoplasm) OR (Head Neoplasms) OR (Neoplasms, Head) OR (Head Neoplasm) OR (Neoplasm, Head) OR (Neck Neoplasms) OR (Neoplasms, Neck) OR (Neck Neoplasm) OR (Neoplasm, Neck) OR (Cancer of Head) OR (Head Cancers) OR (Head Cancer) OR (Cancer, Head) OR (Cancers, Head) OR (Cancer of the Head) OR (Cancer of Neck) OR (Neck Cancers) OR (Neck Cancer) OR (Cancer, Neck) OR (Cancers, Neck) OR (Cancer of the Neck)) AND ((“NETosis “[Mesh]) OR (NETosis Cell Death) OR (Cell Death, NETosis) OR (Cell Deaths, NETosis) OR (Death, NETosis Cell) OR (Deaths, NETosis Cell) OR (NETosis Cell Deaths)).
  • Search 5: NECROPTOSIS (Results: 124)
((“Mouth Neoplasms”[Mesh]) OR (Mouth Neoplasm) OR (Neoplasm, Mouth) OR (Neoplasms, Oral) OR (Neoplasm, Oral) OR (Oral Neoplasm) OR (Oral Neoplasms) OR (Neoplasms, Mouth) OR (Cancer of Mouth) OR (Mouth Cancers) OR (Oral Cancer) OR (Cancer, Oral) OR (Cancers, Oral) OR (Oral Cancers) OR (Cancer of the Mouth) OR (Mouth Cancer) OR (Cancer, Mouth) OR (Cancers, Mouth) OR (“Head and Neck Neoplasms”[Mesh]) OR (Neoplasms, Head and Neck) OR (Head, Neck Neoplasms) OR (Head and Neck Neoplasm) OR (Cancer of Head and Neck) OR (Head and Neck Cancer) OR (Cancer of the Head and Neck) OR (Upper Aerodigestive Tract Neoplasms) OR (UADT Neoplasm) OR (Neoplasm, UADT) OR (Neoplasms, UADT) OR (UADT Neoplasms) OR (Neoplasms, Upper Aerodigestive Tract) OR (Upper Aerodigestive Tract Neoplasm) OR (Head Neoplasms) OR (Neoplasms, Head) OR (Head Neoplasm) OR (Neoplasm, Head) OR (Neck Neoplasms) OR (Neoplasms, Neck) OR (Neck Neoplasm) OR (Neoplasm, Neck) OR (Cancer of Head) OR (Head Cancers) OR (Head Cancer) OR (Cancer, Head) OR (Cancers, Head) OR (Cancer of the Head) OR (Cancer of Neck) OR (Neck Cancers) OR (Neck Cancer) OR (Cancer, Neck) OR (Cancers, Neck) OR (Cancer of the Neck)) AND ((“Necroptosis”[Mesh]) OR (Necroptosis Cell Death) OR (Cell Death, Necroptosis) OR (Cell Deaths, Necroptosis) OR (Death, Necroptosis Cell) OR (Deaths, Necroptosis Cell) OR (Necroptosis Cell Deaths).
  • Search 6: PARTHANATOS (Results: 4)
((“Mouth Neoplasms”[Mesh]) OR (Mouth Neoplasm) OR (Neoplasm, Mouth) OR (Neoplasms, Oral) OR (Neoplasm, Oral) OR (Oral Neoplasm) OR (Oral Neoplasms) OR (Neoplasms, Mouth) OR (Cancer of Mouth) OR (Mouth Cancers) OR (Oral Cancer) OR (Cancer, Oral) OR (Cancers, Oral) OR (Oral Cancers) OR (Cancer of the Mouth) OR (Mouth Cancer) OR (Cancer, Mouth) OR (Cancers, Mouth) OR (“Head and Neck Neoplasms”[Mesh]) OR (Neoplasms, Head and Neck) OR (Head, Neck Neoplasms) OR (Head and Neck Neoplasm) OR (Cancer of Head and Neck) OR (Head and Neck Cancer) OR (Cancer of the Head and Neck) OR (Upper Aerodigestive Tract Neoplasms) OR (UADT Neoplasm) OR (Neoplasm, UADT) OR (Neoplasms, UADT) OR (UADT Neoplasms) OR (Neoplasms, Upper Aerodigestive Tract) OR (Upper Aerodigestive Tract Neoplasm) OR (Head Neoplasms) OR (Neoplasms, Head) OR (Head Neoplasm) OR (Neoplasm, Head) OR (Neck Neoplasms) OR (Neoplasms, Neck) OR (Neck Neoplasm) OR (Neoplasm, Neck) OR (Cancer of Head) OR (Head Cancers) OR (Head Cancer) OR (Cancer, Head) OR (Cancers, Head) OR (Cancer of the Head) OR (Cancer of Neck) OR (Neck Cancers) OR (Neck Cancer) OR (Cancer, Neck) OR (Cancers, Neck) OR (Cancer of the Neck)) AND ((“Parthanatos”[Mesh]) OR OR (Parthanatos Cell Death) OR (Cell Death, Parthanatos) OR (Cell Deaths, Parthanatos) OR (Death, PARTHANATOS Cell) OR (Deaths, Parthanatos Cell) OR (Parthanatos Cell Deaths)).
  • Search 7: MITOPTOSIS (Results: 2)
((“Mouth Neoplasms”[Mesh]) OR (Mouth Neoplasm) OR (Neoplasm, Mouth) OR (Neoplasms, Oral) OR (Neoplasm, Oral) OR (Oral Neoplasm) OR (Oral Neoplasms) OR (Neoplasms, Mouth) OR (Cancer of Mouth) OR (Mouth Cancers) OR (Oral Cancer) OR (Cancer, Oral) OR (Cancers, Oral) OR (Oral Cancers) OR (Cancer of the Mouth) OR (Mouth Cancer) OR (Cancer, Mouth) OR (Cancers, Mouth) OR (“Head and Neck Neoplasms”[Mesh]) OR (Neoplasms, Head and Neck) OR (Head, Neck Neoplasms) OR (Head and Neck Neoplasm) OR (Cancer of Head and Neck) OR (Head and Neck Cancer) OR (Cancer of the Head and Neck) OR (Upper Aerodigestive Tract Neoplasms) OR (UADT Neoplasm) OR (Neoplasm, UADT) OR (Neoplasms, UADT) OR (UADT Neoplasms) OR (Neoplasms, Upper Aerodigestive Tract) OR (Upper Aerodigestive Tract Neoplasm) OR (Head Neoplasms) OR (Neoplasms, Head) OR (Head Neoplasm) OR (Neoplasm, Head) OR (Neck Neoplasms) OR (Neoplasms, Neck) OR (Neck Neoplasm) OR (Neoplasm, Neck) OR (Cancer of Head) OR (Head Cancers) OR (Head Cancer) OR (Cancer, Head) OR (Cancers, Head) OR (Cancer of the Head) OR (Cancer of Neck) OR (Neck Cancers) OR (Neck Cancer) OR (Cancer, Neck) OR (Cancers, Neck) OR (Cancer of the Neck)) AND ((“Mitoptosis”[Mesh]) OR OR (Mitoptosis Cell Death) OR (Cell Death, Mitoptosis) OR (Cell Deaths, Mitoptosis) OR (Death, Mitoptosis Cell) OR (Deaths, Mitoptosis Cell) OR (Mitoptosis Cell Deaths)).
  • Search 8: PARAPTOSIS (Results: 11)
((“Mouth Neoplasms”[Mesh]) OR (Mouth Neoplasm) OR (Neoplasm, Mouth) OR (Neoplasms, Oral) OR (Neoplasm, Oral) OR (Oral Neoplasm) OR (Oral Neoplasms) OR (Neoplasms, Mouth) OR (Cancer of Mouth) OR (Mouth Cancers) OR (Oral Cancer) OR (Cancer, Oral) OR (Cancers, Oral) OR (Oral Cancers) OR (Cancer of the Mouth) OR (Mouth Cancer) OR (Cancer, Mouth) OR (Cancers, Mouth) OR (“Head and Neck Neoplasms”[Mesh]) OR (Neoplasms, Head and Neck) OR (Head, Neck Neoplasms) OR (Head and Neck Neoplasm) OR (Cancer of Head and Neck) OR (Head and Neck Cancer) OR (Cancer of the Head and Neck) OR (Upper Aerodigestive Tract Neoplasms) OR (UADT Neoplasm) OR (Neoplasm, UADT) OR (Neoplasms, UADT) OR (UADT Neoplasms) OR (Neoplasms, Upper Aerodigestive Tract) OR (Upper Aerodigestive Tract Neoplasm) OR (Head Neoplasms) OR (Neoplasms, Head) OR (Head Neoplasm) OR (Neoplasm, Head) OR (Neck Neoplasms) OR (Neoplasms, Neck) OR (Neck Neoplasm) OR (Neoplasm, Neck) OR (Cancer of Head) OR (Head Cancers) OR (Head Cancer) OR (Cancer, Head) OR (Cancers, Head) OR (Cancer of the Head) OR (Cancer of Neck) OR (Neck Cancers) OR (Neck Cancer) OR (Cancer, Neck) OR (Cancers, Neck) OR (Cancer of the Neck)) AND ((“Paraptosis”[Mesh]) OR (Paraptosis Cell Death) OR (Cell Death, Paraptosis) OR (Cell Deaths, Paraptosis) OR (Death, Paraptosis Cell) OR (Deaths, Paraptosis Cell) OR (Paraptosis Cell Deaths)).
  • Search 9: METHUOSIS (Results: 3)
((“Mouth Neoplasms”[Mesh]) OR (Mouth Neoplasm) OR (Neoplasm, Mouth) OR (Neoplasms, Oral) OR (Neoplasm, Oral) OR (Oral Neoplasm) OR (Oral Neoplasms) OR (Neoplasms, Mouth) OR (Cancer of Mouth) OR (Mouth Cancers) OR (Oral Cancer) OR (Cancer, Oral) OR (Cancers, Oral) OR (Oral Cancers) OR (Cancer of the Mouth) OR (Mouth Cancer) OR (Cancer, Mouth) OR (Cancers, Mouth) OR (“Head and Neck Neoplasms”[Mesh]) OR (Neoplasms, Head and Neck) OR (Head, Neck Neoplasms) OR (Head and Neck Neoplasm) OR (Cancer of Head and Neck) OR (Head and Neck Cancer) OR (Cancer of the Head and Neck) OR (Upper Aerodigestive Tract Neoplasms) OR (UADT Neoplasm) OR (Neoplasm, UADT) OR (Neoplasms, UADT) OR (UADT Neoplasms) OR (Neoplasms, Upper Aerodigestive Tract) OR (Upper Aerodigestive Tract Neoplasm) OR (Head Neoplasms) OR (Neoplasms, Head) OR (Head Neoplasm) OR (Neoplasm, Head) OR (Neck Neoplasms) OR (Neoplasms, Neck) OR (Neck Neoplasm) OR (Neoplasm, Neck) OR (Cancer of Head) OR (Head Cancers) OR (Head Cancer) OR (Cancer, Head) OR (Cancers, Head) OR (Cancer of the Head) OR (Cancer of Neck) OR (Neck Cancers) OR (Neck Cancer) OR (Cancer, Neck) OR (Cancers, Neck) OR (Cancer of the Neck)) AND ((“Methuosis”[Mesh]) OR (Methuosis Cell Death) OR (Cell Death, Methuosis) OR (Cell Deaths, Methuosis) OR (Death, Methuosis Cell) OR (Deaths, Methuosis Cell) OR (Methuosis Cell Deaths)).
  • SCOPUS
  • Search 1: ENTOSIS (Results: 6)
((Mouth Neoplasms) OR (Mouth Neoplasm) OR (Neoplasm, Mouth) OR (Neoplasms, Oral) OR (Neoplasm, Oral) OR (Oral Neoplasm) OR (Oral Neoplasms) OR (Neoplasms, Mouth) OR (Cancer of Mouth) OR (Mouth Cancers) OR (Oral Cancer) OR (Cancer, Oral) OR (Cancers, Oral) OR (Oral Cancers) OR (Cancer of the Mouth) OR (Mouth Cancer) OR (Cancer, Mouth) OR (Cancers, Mouth) OR (Head and Neck Neoplasms) OR (Neoplasms, Head and Neck) OR (Head, Neck Neoplasms) OR (Head and Neck Neoplasm) OR (Cancer of Head and Neck) OR (Head and Neck Cancer) OR (Cancer of the Head and Neck) OR (Upper Aerodigestive Tract Neoplasms) OR (UADT Neoplasm) OR (Neoplasm, UADT) OR (Neoplasms, UADT) OR (UADT Neoplasms) OR (Neoplasms, Upper Aerodigestive Tract) OR (Upper Aerodigestive Tract Neoplasm) OR (Head Neoplasms) OR (Neoplasms, Head) OR (Head Neoplasm) OR (Neoplasm, Head) OR (Neck Neoplasms) OR (Neoplasms, Neck) OR (Neck Neoplasm) OR (Neoplasm, Neck) OR (Cancer of Head) OR (Head Cancers) OR (Head Cancer) OR (Cancer, Head) OR (Cancers, Head) OR (Cancer of the Head) OR (Cancer of Neck) OR (Neck Cancers) OR (Neck Cancer) OR (Cancer, Neck) OR (Cancers, Neck) OR (Cancer of the Neck)) AND ((Entosis) OR (Entoses)).
  • Search 2: FERROPTOSIS (Results: 218)
((Mouth Neoplasms) OR (Mouth Neoplasm) OR (Neoplasm, Mouth) OR (Neoplasms, Oral) OR (Neoplasm, Oral) OR (Oral Neoplasm) OR (Oral Neoplasms) OR (Neoplasms, Mouth) OR (Cancer of Mouth) OR (Mouth Cancers) OR (Oral Cancer) OR (Cancer, Oral) OR (Cancers, Oral) OR (Oral Cancers) OR (Cancer of the Mouth) OR (Mouth Cancer) OR (Cancer, Mouth) OR (Cancers, Mouth) OR (Head and Neck Neoplasms) OR (Neoplasms, Head and Neck) OR (Head, Neck Neoplasms) OR (Head and Neck Neoplasm) OR (Cancer of Head and Neck) OR (Head and Neck Cancer) OR (Cancer of the Head and Neck) OR (Upper Aerodigestive Tract Neoplasms) OR (UADT Neoplasm) OR (Neoplasm, UADT) OR (Neoplasms, UADT) OR (UADT Neoplasms) OR (Neoplasms, Upper Aerodigestive Tract) OR (Upper Aerodigestive Tract Neoplasm) OR (Head Neoplasms) OR (Neoplasms, Head) OR (Head Neoplasm) OR (Neoplasm, Head) OR (Neck Neoplasms) OR (Neoplasms, Neck) OR (Neck Neoplasm) OR (Neoplasm, Neck) OR (Cancer of Head) OR (Head Cancers) OR (Head Cancer) OR (Cancer, Head) OR (Cancers, Head) OR (Cancer of the Head) OR (Cancer of Neck) OR (Neck Cancers) OR (Neck Cancer) OR (Cancer, Neck) OR (Cancers, Neck) OR (Cancer of the Neck)) AND ((Ferroptosis) OR (Oxytosis)).
  • Search 3: PYROPTOSIS (Results: 1.431)
((Mouth Neoplasms) OR (Mouth Neoplasm) OR (Neoplasm, Mouth) OR (Neoplasms, Oral) OR (Neoplasm, Oral) OR (Oral Neoplasm) OR (Oral Neoplasms) OR (Neoplasms, Mouth) OR (Cancer of Mouth) OR (Mouth Cancers) OR (Oral Cancer) OR (Cancer, Oral) OR (Cancers, Oral) OR (Oral Cancers) OR (Cancer of the Mouth) OR (Mouth Cancer) OR (Cancer, Mouth) OR (Cancers, Mouth) OR (Head and Neck Neoplasms) OR (Neoplasms, Head and Neck) OR (Head, Neck Neoplasms) OR (Head and Neck Neoplasm) OR (Cancer of Head and Neck) OR (Head and Neck Cancer) OR (Cancer of the Head and Neck) OR (Upper Aerodigestive Tract Neoplasms) OR (UADT Neoplasm) OR (Neoplasm, UADT) OR (Neoplasms, UADT) OR (UADT Neoplasms) OR (Neoplasms, Upper Aerodigestive Tract) OR (Upper Aerodigestive Tract Neoplasm) OR (Head Neoplasms) OR (Neoplasms, Head) OR (Head Neoplasm) OR (Neoplasm, Head) OR (Neck Neoplasms) OR (Neoplasms, Neck) OR (Neck Neoplasm) OR (Neoplasm, Neck) OR (Cancer of Head) OR (Head Cancers) OR (Head Cancer) OR (Cancer, Head) OR (Cancers, Head) OR (Cancer of the Head) OR (Cancer of Neck) OR (Neck Cancers) OR (Neck Cancer) OR (Cancer, Neck) OR (Cancers, Neck) OR (Cancer of the Neck)) AND ((Pyroptosis) OR (Pyroptoses) OR (Pyroptotic Cell Death) OR (Cell Death, Pyroptotic) OR (Cell Deaths, Pyroptotic) OR (Death, Pyroptotic Cell) OR (Deaths, Pyroptotic Cell) OR (Pyroptotic Cell Deaths) OR (Caspase-1 Dependent Cell Death) OR (Caspase 1 Dependent Cell Death) OR (Inflammatory Apoptosis) OR (Apoptoses, Inflammatory) OR (Apoptosis, Inflammatory) OR (Inflammatory Apoptoses)).
  • Search 4: NETOSIS (Results: 8)
((Mouth Neoplasms) OR (Mouth Neoplasm) OR (Neoplasm, Mouth) OR (Neoplasms, Oral) OR (Neoplasm, Oral) OR (Oral Neoplasm) OR (Oral Neoplasms) OR (Neoplasms, Mouth) OR (Cancer of Mouth) OR (Mouth Cancers) OR (Oral Cancer) OR (Cancer, Oral) OR (Cancers, Oral) OR (Oral Cancers) OR (Cancer of the Mouth) OR (Mouth Cancer) OR (Cancer, Mouth) OR (Cancers, Mouth) OR (Head and Neck Neoplasms) OR (Neoplasms, Head and Neck) OR (Head, Neck Neoplasms) OR (Head and Neck Neoplasm) OR (Cancer of Head and Neck) OR (Head and Neck Cancer) OR (Cancer of the Head and Neck) OR (Upper Aerodigestive Tract Neoplasms) OR (UADT Neoplasm) OR (Neoplasm, UADT) OR (Neoplasms, UADT) OR (UADT Neoplasms) OR (Neoplasms, Upper Aerodigestive Tract) OR (Upper Aerodigestive Tract Neoplasm) OR (Head Neoplasms) OR (Neoplasms, Head) OR (Head Neoplasm) OR (Neoplasm, Head) OR (Neck Neoplasms) OR (Neoplasms, Neck) OR (Neck Neoplasm) OR (Neoplasm, Neck) OR (Cancer of Head) OR (Head Cancers) OR (Head Cancer) OR (Cancer, Head) OR (Cancers, Head) OR (Cancer of the Head) OR (Cancer of Neck) OR (Neck Cancers) OR (Neck Cancer) OR (Cancer, Neck) OR (Cancers, Neck) OR (Cancer of the Neck)) AND (Netosis) OR (Netosis Cell Death) OR (Cell Death, Netosis) OR (Cell Deaths, Netosis) OR (Death, Netosis Cell) OR (Deaths, Netosis Cell)) OR (Netosis Cell Deaths)).
  • Search 5: NECROPTOSIS (Results: 108)
((Mouth Neoplasms) OR (Mouth Neoplasm) OR (Neoplasm, Mouth) OR (Neoplasms, Oral) OR (Neoplasm, Oral) OR (Oral Neoplasm) OR (Oral Neoplasms) OR (Neoplasms, Mouth) OR (Cancer of Mouth) OR (Mouth Cancers) OR (Oral Cancer) OR (Cancer, Oral) OR (Cancers, Oral) OR (Oral Cancers) OR (Cancer of the Mouth) OR (Mouth Cancer) OR (Cancer, Mouth) OR (Cancers, Mouth) OR (Head and Neck Neoplasms) OR (Neoplasms, Head and Neck) OR (Head, Neck Neoplasms) OR (Head and Neck Neoplasm) OR (Cancer of Head and Neck) OR (Head and Neck Cancer) OR (Cancer of the Head and Neck) OR (Upper Aerodigestive Tract Neoplasms) OR (UADT Neoplasm) OR (Neoplasm, UADT) OR (Neoplasms, UADT) OR (UADT Neoplasms) OR (Neoplasms, Upper Aerodigestive Tract) OR (Upper Aerodigestive Tract Neoplasm) OR (Head Neoplasms) OR (Neoplasms, Head) OR (Head Neoplasm) OR (Neoplasm, Head) OR (Neck Neoplasms) OR (Neoplasms, Neck) OR (Neck Neoplasm) OR (Neoplasm, Neck) OR (Cancer of Head) OR (Head Cancers) OR (Head Cancer) OR (Cancer, Head) OR (Cancers, Head) OR (Cancer of the Head) OR (Cancer of Neck) OR (Neck Cancers) OR (Neck Cancer) OR (Cancer, Neck) OR (Cancers, Neck) OR (Cancer of the Neck)) AND (Necroptosis) OR (Necroptosis Cell Death) OR (Cell Death, Necroptosis) OR (Cell Deaths, Necroptosis) OR (Death, Necroptosis Cell) OR (Deaths, Necroptosis Cell) OR (Necroptosis Cell Deaths).
  • Search 6: PARTHANATOS (Results: 7)
((Mouth Neoplasms) OR (Mouth Neoplasm) OR (Neoplasm, Mouth) OR (Neoplasms, Oral) OR (Neoplasm, Oral) OR (Oral Neoplasm) OR (Oral Neoplasms) OR (Neoplasms, Mouth) OR (Cancer of Mouth) OR (Mouth Cancers) OR (Oral Cancer) OR (Cancer, Oral) OR (Cancers, Oral) OR (Oral Cancers) OR (Cancer of the Mouth) OR (Mouth Cancer) OR (Cancer, Mouth) OR (Cancers, Mouth) OR (Head and Neck Neoplasms) OR (Neoplasms, Head and Neck) OR (Head, Neck Neoplasms) OR (Head and Neck Neoplasm) OR (Cancer of Head and Neck) OR (Head and Neck Cancer) OR (Cancer of the Head and Neck) OR (Upper Aerodigestive Tract Neoplasms) OR (UADT Neoplasm) OR (Neoplasm, UADT) OR (Neoplasms, UADT) OR (UADT Neoplasms) OR (Neoplasms, Upper Aerodigestive Tract) OR (Upper Aerodigestive Tract Neoplasm) OR (Head Neoplasms) OR (Neoplasms, Head) OR (Head Neoplasm) OR (Neoplasm, Head) OR (Neck Neoplasms) OR (Neoplasms, Neck) OR (Neck Neoplasm) OR (Neoplasm, Neck) OR (Cancer of Head) OR (Head Cancers) OR (Head Cancer) OR (Cancer, Head) OR (Cancers, Head) OR (Cancer of the Head) OR (Cancer of Neck) OR (Neck Cancers) OR (Neck Cancer) OR (Cancer, Neck) OR (Cancers, Neck) OR (Cancer of the Neck)) AND (Parthanatos) OR (Parthanatos Cell Death) OR (Cell Death, Parthanatos) OR (Cell Deaths, Parthanatos) OR (Death, Parthanatos Cell) OR (Deaths, Parthanatos Cell) OR (Parthanatos Cell Deaths)).
  • Search 7: MITOPTOSIS (Results: 4)
((Mouth Neoplasms) OR (Mouth Neoplasm) OR (Neoplasm, Mouth) OR (Neoplasms, Oral) OR (Neoplasm, Oral) OR (Oral Neoplasm) OR (Oral Neoplasms) OR (Neoplasms, Mouth) OR (Cancer of Mouth) OR (Mouth Cancers) OR (Oral Cancer) OR (Cancer, Oral) OR (Cancers, Oral) OR (Oral Cancers) OR (Cancer of the Mouth) OR (Mouth Cancer) OR (Cancer, Mouth) OR (Cancers, Mouth) OR (Head and Neck Neoplasms) OR (Neoplasms, Head and Neck) OR (Head, Neck Neoplasms) OR (Head and Neck Neoplasm) OR (Cancer of Head and Neck) OR (Head and Neck Cancer) OR (Cancer of the Head and Neck) OR (Upper Aerodigestive Tract Neoplasms) OR (UADT Neoplasm) OR (Neoplasm, UADT) OR (Neoplasms, UADT) OR (UADT Neoplasms) OR (Neoplasms, Upper Aerodigestive Tract) OR (Upper Aerodigestive Tract Neoplasm) OR (Head Neoplasms) OR (Neoplasms, Head) OR (Head Neoplasm) OR (Neoplasm, Head) OR (Neck Neoplasms) OR (Neoplasms, Neck) OR (Neck Neoplasm) OR (Neoplasm, Neck) OR (Cancer of Head) OR (Head Cancers) OR (Head Cancer) OR (Cancer, Head) OR (Cancers, Head) OR (Cancer of the Head) OR (Cancer of Neck) OR (Neck Cancers) OR (Neck Cancer) OR (Cancer, Neck) OR (Cancers, Neck) OR (Cancer of the Neck)) AND (Mitoptosis) OR (Mitoptosis Cell Death) OR (Cell Death, Mitoptosis) OR (Cell Deaths, Mitoptosis) OR (Death, Mitoptosis Cell) OR (Deaths, Mitoptosis Cell) OR (Mitoptosis Cell Deaths)).
  • Search 8: PARAPTOSIS (Results: 2)
((Mouth Neoplasms) OR (Mouth Neoplasm) OR (Neoplasm, Mouth) OR (Neoplasms, Oral) OR (Neoplasm, Oral) OR (Oral Neoplasm) OR (Oral Neoplasms) OR (Neoplasms, Mouth) OR (Cancer of Mouth) OR (Mouth Cancers) OR (Oral Cancer) OR (Cancer, Oral) OR (Cancers, Oral) OR (Oral Cancers) OR (Cancer of the Mouth) OR (Mouth Cancer) OR (Cancer, Mouth) OR (Cancers, Mouth) OR (Head and Neck Neoplasms) OR (Neoplasms, Head and Neck) OR (Head, Neck Neoplasms) OR (Head and Neck Neoplasm) OR (Cancer of Head and Neck) OR (Head and Neck Cancer) OR (Cancer of the Head and Neck) OR (Upper Aerodigestive Tract Neoplasms) OR (UADT Neoplasm) OR (Neoplasm, UADT) OR (Neoplasms, UADT) OR (UADT Neoplasms) OR (Neoplasms, Upper Aerodigestive Tract) OR (Upper Aerodigestive Tract Neoplasm) OR (Head Neoplasms) OR (Neoplasms, Head) OR (Head Neoplasm) OR (Neoplasm, Head) OR (Neck Neoplasms) OR (Neoplasms, Neck) OR (Neck Neoplasm) OR (Neoplasm, Neck) OR (Cancer of Head) OR (Head Cancers) OR (Head Cancer) OR (Cancer, Head) OR (Cancers, Head) OR (Cancer of the Head) OR (Cancer of Neck) OR (Neck Cancers) OR (Neck Cancer) OR (Cancer, Neck) OR (Cancers, Neck) OR (Cancer of the Neck)) AND (Paraptosis) OR (Paraptosis Cell Death) OR (Cell Death, Paraptosis) OR (Cell Deaths, Paraptosis) OR (Death, Paraptosis Cell) OR (Deaths, Paraptosis Cell) OR (Paraptosis Cell Deaths).
  • Search 9: METHUOSIS (Results: 3)
((Mouth Neoplasms) OR (Mouth Neoplasm) OR (Neoplasm, Mouth) OR (Neoplasms, Oral) OR (Neoplasm, Oral) OR (Oral Neoplasm) OR (Oral Neoplasms) OR (Neoplasms, Mouth) OR (Cancer of Mouth) OR (Mouth Cancers) OR (Oral Cancer) OR (Cancer, Oral) OR (Cancers, Oral) OR (Oral Cancers) OR (Cancer of the Mouth) OR (Mouth Cancer) OR (Cancer, Mouth) OR (Cancers, Mouth) OR (Head and Neck Neoplasms) OR (Neoplasms, Head and Neck) OR (Head, Neck Neoplasms) OR (Head and Neck Neoplasm) OR (Cancer of Head and Neck) OR (Head and Neck Cancer) OR (Cancer of the Head and Neck) OR (Upper Aerodigestive Tract Neoplasms) OR (UADT Neoplasm) OR (Neoplasm, UADT) OR (Neoplasms, UADT) OR (UADT Neoplasms) OR (Neoplasms, Upper Aerodigestive Tract) OR (Upper Aerodigestive Tract Neoplasm) OR (Head Neoplasms) OR (Neoplasms, Head) OR (Head Neoplasm) OR (Neoplasm, Head) OR (Neck Neoplasms) OR (Neoplasms, Neck) OR (Neck Neoplasm) OR (Neoplasm, Neck) OR (Cancer of Head) OR (Head Cancers) OR (Head Cancer) OR (Cancer, Head) OR (Cancers, Head) OR (Cancer of the Head) OR (Cancer of Neck) OR (Neck Cancers) OR (Neck Cancer) OR (Cancer, Neck) OR (Cancers, Neck) OR (Cancer of the Neck)) AND (Methuosis) OR (Methuosis Cell Death) OR (Cell Death, Methuosis) OR (Cell Deaths, Methuosis) OR (Death, Methuosis Cell) OR (Deaths, Methuosis Cell) OR (Methuosis Cell Deaths).
  • WEB OF SCIENCE
  • Search 1: ENTOSIS (Results: 5)
(TS = ((Mouth Neoplasms) OR (Mouth Neoplasm) OR (Neoplasm, Mouth) OR (Neoplasms, Oral) OR (Neoplasm, Oral) OR (Oral Neoplasm) OR (Oral Neoplasms) OR (Neoplasms, Mouth) OR (Cancer of Mouth) OR (Mouth Cancers) OR (Oral Cancer) OR (Cancer, Oral) OR (Cancers, Oral) OR (Oral Cancers) OR (Cancer of the Mouth) OR (Mouth Cancer) OR (Cancer, Mouth) OR (Cancers, Mouth) OR (Head and Neck Neoplasms) OR (Neoplasms, Head and Neck) OR (Head, Neck Neoplasms) OR (Head and Neck Neoplasm) OR (Cancer of Head and Neck) OR (Head and Neck Cancer) OR (Cancer of the Head and Neck) OR (Upper Aerodigestive Tract Neoplasms) OR (UADT Neoplasm) OR (Neoplasm, UADT) OR (Neoplasms, UADT) OR (UADT Neoplasms) OR (Neoplasms, Upper Aerodigestive Tract) OR (Upper Aerodigestive Tract Neoplasm) OR (Head Neoplasms) OR (Neoplasms, Head) OR (Head Neoplasm) OR (Neoplasm, Head) OR (Neck Neoplasms) OR (Neoplasms, Neck) OR (Neck Neoplasm) OR (Neoplasm, Neck) OR (Cancer of Head) OR (Head Cancers) OR (Head Cancer) OR (Cancer, Head) OR (Cancers, Head) OR (Cancer of the Head) OR (Cancer of Neck) OR (Neck Cancers) OR (Neck Cancer) OR (Cancer, Neck) OR (Cancers, Neck) OR (Cancer of the Neck))) AND TS = ((Entosis) OR (Entoses)).
  • Search 2: FERROPTOSIS (Results: 151)
(TS = ((Mouth Neoplasms) OR (Mouth Neoplasm) OR (Neoplasm, Mouth) OR (Neoplasms, Oral) OR (Neoplasm, Oral) OR (Oral Neoplasm) OR (Oral Neoplasms) OR (Neoplasms, Mouth) OR (Cancer of Mouth) OR (Mouth Cancers) OR (Oral Cancer) OR (Cancer, Oral) OR (Cancers, Oral) OR (Oral Cancers) OR (Cancer of the Mouth) OR (Mouth Cancer) OR (Cancer, Mouth) OR (Cancers, Mouth) OR (Head and Neck Neoplasms) OR (Neoplasms, Head and Neck) OR (Head, Neck Neoplasms) OR (Head and Neck Neoplasm) OR (Cancer of Head and Neck) OR (Head and Neck Cancer) OR (Cancer of the Head and Neck) OR (Upper Aerodigestive Tract Neoplasms) OR (UADT Neoplasm) OR (Neoplasm, UADT) OR (Neoplasms, UADT) OR (UADT Neoplasms) OR (Neoplasms, Upper Aerodigestive Tract) OR (Upper Aerodigestive Tract Neoplasm) OR (Head Neoplasms) OR (Neoplasms, Head) OR (Head Neoplasm) OR (Neoplasm, Head) OR (Neck Neoplasms) OR (Neoplasms, Neck) OR (Neck Neoplasm) OR (Neoplasm, Neck) OR (Cancer of Head) OR (Head Cancers) OR (Head Cancer) OR (Cancer, Head) OR (Cancers, Head) OR (Cancer of the Head) OR (Cancer of Neck) OR (Neck Cancers) OR (Neck Cancer) OR (Cancer, Neck) OR (Cancers, Neck) OR (Cancer of the Neck))) AND TS = ((Ferroptosis) OR (Oxytosis)).
  • Search 3: PYROPTOSIS (Results: 1090)
(TS = ((Mouth Neoplasms) OR (Mouth Neoplasm) OR (Neoplasm, Mouth) OR (Neoplasms, Oral) OR (Neoplasm, Oral) OR (Oral Neoplasm) OR (Oral Neoplasms) OR (Neoplasms, Mouth) OR (Cancer of Mouth) OR (Mouth Cancers) OR (Oral Cancer) OR (Cancer, Oral) OR (Cancers, Oral) OR (Oral Cancers) OR (Cancer of the Mouth) OR (Mouth Cancer) OR (Cancer, Mouth) OR (Cancers, Mouth) OR (Head and Neck Neoplasms) OR (Neoplasms, Head and Neck) OR (Head, Neck Neoplasms) OR (Head and Neck Neoplasm) OR (Cancer of Head and Neck) OR (Head and Neck Cancer) OR (Cancer of the Head and Neck) OR (Upper Aerodigestive Tract Neoplasms) OR (UADT Neoplasm) OR (Neoplasm, UADT) OR (Neoplasms, UADT) OR (UADT Neoplasms) OR (Neoplasms, Upper Aerodigestive Tract) OR (Upper Aerodigestive Tract Neoplasm) OR (Head Neoplasms) OR (Neoplasms, Head) OR (Head Neoplasm) OR (Neoplasm, Head) OR (Neck Neoplasms) OR (Neoplasms, Neck) OR (Neck Neoplasm) OR (Neoplasm, Neck) OR (Cancer of Head) OR (Head Cancers) OR (Head Cancer) OR (Cancer, Head) OR (Cancers, Head) OR (Cancer of the Head) OR (Cancer of Neck) OR (Neck Cancers) OR (Neck Cancer) OR (Cancer, Neck) OR (Cancers, Neck) OR (Cancer of the Neck))) AND TS = ((Pyroptosis) OR (Pyroptoses) OR (Pyroptotic Cell Death) OR (Cell Death, Pyroptotic) OR (Cell Deaths, Pyroptotic) OR (Death, Pyroptotic Cell) OR (Deaths, Pyroptotic Cell) OR (Pyroptotic Cell Deaths) OR (Caspase-1 Dependent Cell Death) OR (Caspase 1 Dependent Cell Death) OR (Inflammatory Apoptosis) OR (Apoptoses, Inflammatory) OR (Apoptosis, Inflammatory) OR (Inflammatory Apoptoses)).
  • Search 4: NETOSIS (Results: 11)
(TS = ((Mouth Neoplasms) OR (Mouth Neoplasm) OR (Neoplasm, Mouth) OR (Neoplasms, Oral) OR (Neoplasm, Oral) OR (Oral Neoplasm) OR (Oral Neoplasms) OR (Neoplasms, Mouth) OR (Cancer of Mouth) OR (Mouth Cancers) OR (Oral Cancer) OR (Cancer, Oral) OR (Cancers, Oral) OR (Oral Cancers) OR (Cancer of the Mouth) OR (Mouth Cancer) OR (Cancer, Mouth) OR (Cancers, Mouth) OR (Head and Neck Neoplasms) OR (Neoplasms, Head and Neck) OR (Head, Neck Neoplasms) OR (Head and Neck Neoplasm) OR (Cancer of Head and Neck) OR (Head and Neck Cancer) OR (Cancer of the Head and Neck) OR (Upper Aerodigestive Tract Neoplasms) OR (UADT Neoplasm) OR (Neoplasm, UADT) OR (Neoplasms, UADT) OR (UADT Neoplasms) OR (Neoplasms, Upper Aerodigestive Tract) OR (Upper Aerodigestive Tract Neoplasm) OR (Head Neoplasms) OR (Neoplasms, Head) OR (Head Neoplasm) OR (Neoplasm, Head) OR (Neck Neoplasms) OR (Neoplasms, Neck) OR (Neck Neoplasm) OR (Neoplasm, Neck) OR (Cancer of Head) OR (Head Cancers) OR (Head Cancer) OR (Cancer, Head) OR (Cancers, Head) OR (Cancer of the Head) OR (Cancer of Neck) OR (Neck Cancers) OR (Neck Cancer) OR (Cancer, Neck) OR (Cancers, Neck) OR (Cancer of the Neck))) AND TS = ((Netosis) OR (Netosis Cell Death) OR (Cell Death, Netosis) OR (Cell Deaths, Netosis) OR (Death, Netosis Cell) OR (Deaths, Netosis Cell) OR (Netosis Cell Deaths)).
  • Search 5: NECROPTOSIS (Results: 298)
(TS = ((Mouth Neoplasms) OR (Mouth Neoplasm) OR (Neoplasm, Mouth) OR (Neoplasms, Oral) OR (Neoplasm, Oral) OR (Oral Neoplasm) OR (Oral Neoplasms) OR (Neoplasms, Mouth) OR (Cancer of Mouth) OR (Mouth Cancers) OR (Oral Cancer) OR (Cancer, Oral) OR (Cancers, Oral) OR (Oral Cancers) OR (Cancer of the Mouth) OR (Mouth Cancer) OR (Cancer, Mouth) OR (Cancers, Mouth) OR (Head and Neck Neoplasms) OR (Neoplasms, Head and Neck) OR (Head, Neck Neoplasms) OR (Head and Neck Neoplasm) OR (Cancer of Head and Neck) OR (Head and Neck Cancer) OR (Cancer of the Head and Neck) OR (Upper Aerodigestive Tract Neoplasms) OR (UADT Neoplasm) OR (Neoplasm, UADT) OR (Neoplasms, UADT) OR (UADT Neoplasms) OR (Neoplasms, Upper Aerodigestive Tract) OR (Upper Aerodigestive Tract Neoplasm) OR (Head Neoplasms) OR (Neoplasms, Head) OR (Head Neoplasm) OR (Neoplasm, Head) OR (Neck Neoplasms) OR (Neoplasms, Neck) OR (Neck Neoplasm) OR (Neoplasm, Neck) OR (Cancer of Head) OR (Head Cancers) OR (Head Cancer) OR (Cancer, Head) OR (Cancers, Head) OR (Cancer of the Head) OR (Cancer of Neck) OR (Neck Cancers) OR (Neck Cancer) OR (Cancer, Neck) OR (Cancers, Neck) OR (Cancer of the Neck))) AND TS = ((Necroptosis) OR (Necroptosis Cell Death) OR (Cell Death, Necroptosis) OR (Cell Deaths, Necroptosis) OR (Death, Necroptosis Cell) OR (Deaths, Necroptosis Cell) OR (Necroptosis Cell Deaths)).
  • Search 6: PARTHANATOS (Results: 12)
(TS = ((Mouth Neoplasms) OR (Mouth Neoplasm) OR (Neoplasm, Mouth) OR (Neoplasms, Oral) OR (Neoplasm, Oral) OR (Oral Neoplasm) OR (Oral Neoplasms) OR (Neoplasms, Mouth) OR (Cancer of Mouth) OR (Mouth Cancers) OR (Oral Cancer) OR (Cancer, Oral) OR (Cancers, Oral) OR (Oral Cancers) OR (Cancer of the Mouth) OR (Mouth Cancer) OR (Cancer, Mouth) OR (Cancers, Mouth) OR (Head and Neck Neoplasms) OR (Neoplasms, Head and Neck) OR (Head, Neck Neoplasms) OR (Head and Neck Neoplasm) OR (Cancer of Head and Neck) OR (Head and Neck Cancer) OR (Cancer of the Head and Neck) OR (Upper Aerodigestive Tract Neoplasms) OR (UADT Neoplasm) OR (Neoplasm, UADT) OR (Neoplasms, UADT) OR (UADT Neoplasms) OR (Neoplasms, Upper Aerodigestive Tract) OR (Upper Aerodigestive Tract Neoplasm) OR (Head Neoplasms) OR (Neoplasms, Head) OR (Head Neoplasm) OR (Neoplasm, Head) OR (Neck Neoplasms) OR (Neoplasms, Neck) OR (Neck Neoplasm) OR (Neoplasm, Neck) OR (Cancer of Head) OR (Head Cancers) OR (Head Cancer) OR (Cancer, Head) OR (Cancers, Head) OR (Cancer of the Head) OR (Cancer of Neck) OR (Neck Cancers) OR (Neck Cancer) OR (Cancer, Neck) OR (Cancers, Neck) OR (Cancer of the Neck))) AND TS = (Parthanatos) OR (Parthanatos Cell Death) OR (Cell Death, Parthanatos) OR (Cell Deaths, Parthanatos) OR (Death, Parthanatos Cell) OR (Deaths, Parthanatos Cell) OR (Parthanatos Cell Deaths)).
  • Search 7: MITOPTOSIS (Results: 9)
(TS = ((Mouth Neoplasms) OR (Mouth Neoplasm) OR (Neoplasm, Mouth) OR (Neoplasms, Oral) OR (Neoplasm, Oral) OR (Oral Neoplasm) OR (Oral Neoplasms) OR (Neoplasms, Mouth) OR (Cancer of Mouth) OR (Mouth Cancers) OR (Oral Cancer) OR (Cancer, Oral) OR (Cancers, Oral) OR (Oral Cancers) OR (Cancer of the Mouth) OR (Mouth Cancer) OR (Cancer, Mouth) OR (Cancers, Mouth) OR (Head and Neck Neoplasms) OR (Neoplasms, Head and Neck) OR (Head, Neck Neoplasms) OR (Head and Neck Neoplasm) OR (Cancer of Head and Neck) OR (Head and Neck Cancer) OR (Cancer of the Head and Neck) OR (Upper Aerodigestive Tract Neoplasms) OR (UADT Neoplasm) OR (Neoplasm, UADT) OR (Neoplasms, UADT) OR (UADT Neoplasms) OR (Neoplasms, Upper Aerodigestive Tract) OR (Upper Aerodigestive Tract Neoplasm) OR (Head Neoplasms) OR (Neoplasms, Head) OR (Head Neoplasm) OR (Neoplasm, Head) OR (Neck Neoplasms) OR (Neoplasms, Neck) OR (Neck Neoplasm) OR (Neoplasm, Neck) OR (Cancer of Head) OR (Head Cancers) OR (Head Cancer) OR (Cancer, Head) OR (Cancers, Head) OR (Cancer of the Head) OR (Cancer of Neck) OR (Neck Cancers) OR (Neck Cancer) OR (Cancer, Neck) OR (Cancers, Neck) OR (Cancer of the Neck))) AND TS = ((Mitoptosis) OR (Mitoptosis) OR (Mitoptosis Cell Death) OR (Cell Death, Mitoptosis) OR (Cell Deaths, Mitoptosis) OR (Death, Mitoptosis Cell) OR (Deaths, Mitoptosis Cell) OR (Mitoptosis Cell Deaths).
  • Search 8: PARAPTOSIS (Results: 5)
(TS = ((Mouth Neoplasms) OR (Mouth Neoplasm) OR (Neoplasm, Mouth) OR (Neoplasms, Oral) OR (Neoplasm, Oral) OR (Oral Neoplasm) OR (Oral Neoplasms) OR (Neoplasms, Mouth) OR (Cancer of Mouth) OR (Mouth Cancers) OR (Oral Cancer) OR (Cancer, Oral) OR (Cancers, Oral) OR (Oral Cancers) OR (Cancer of the Mouth) OR (Mouth Cancer) OR (Cancer, Mouth) OR (Cancers, Mouth) OR (Head and Neck Neoplasms) OR (Neoplasms, Head and Neck) OR (Head, Neck Neoplasms) OR (Head and Neck Neoplasm) OR (Cancer of Head and Neck) OR (Head and Neck Cancer) OR (Cancer of the Head and Neck) OR (Upper Aerodigestive Tract Neoplasms) OR (UADT Neoplasm) OR (Neoplasm, UADT) OR (Neoplasms, UADT) OR (UADT Neoplasms) OR (Neoplasms, Upper Aerodigestive Tract) OR (Upper Aerodigestive Tract Neoplasm) OR (Head Neoplasms) OR (Neoplasms, Head) OR (Head Neoplasm) OR (Neoplasm, Head) OR (Neck Neoplasms) OR (Neoplasms, Neck) OR (Neck Neoplasm) OR (Neoplasm, Neck) OR (Cancer of Head) OR (Head Cancers) OR (Head Cancer) OR (Cancer, Head) OR (Cancers, Head) OR (Cancer of the Head) OR (Cancer of Neck) OR (Neck Cancers) OR (Neck Cancer) OR (Cancer, Neck) OR (Cancers, Neck) OR (Cancer of the Neck))) AND TS = (Paraptosis) OR (Paraptosis Cell Death) OR (Cell Death, Paraptosis) OR (Cell Deaths, Paraptosis) OR (Death, Paraptosis Cell) OR (Deaths, Paraptosis Cell) OR (Paraptosis Cell Deaths)
  • Search 9: METHUOSIS (Results: 2)
(TS = ((Mouth Neoplasms) OR (Mouth Neoplasm) OR (Neoplasm, Mouth) OR (Neoplasms, Oral) OR (Neoplasm, Oral) OR (Oral Neoplasm) OR (Oral Neoplasms) OR (Neoplasms, Mouth) OR (Cancer of Mouth) OR (Mouth Cancers) OR (Oral Cancer) OR (Cancer, Oral) OR (Cancers, Oral) OR (Oral Cancers) OR (Cancer of the Mouth) OR (Mouth Cancer) OR (Cancer, Mouth) OR (Cancers, Mouth) OR (Head and Neck Neoplasms) OR (Neoplasms, Head and Neck) OR (Head, Neck Neoplasms) OR (Head and Neck Neoplasm) OR (Cancer of Head and Neck) OR (Head and Neck Cancer) OR (Cancer of the Head and Neck) OR (Upper Aerodigestive Tract Neoplasms) OR (UADT Neoplasm) OR (Neoplasm, UADT) OR (Neoplasms, UADT) OR (UADT Neoplasms) OR (Neoplasms, Upper Aerodigestive Tract) OR (Upper Aerodigestive Tract Neoplasm) OR (Head Neoplasms) OR (Neoplasms, Head) OR (Head Neoplasm) OR (Neoplasm, Head) OR (Neck Neoplasms) OR (Neoplasms, Neck) OR (Neck Neoplasm) OR (Neoplasm, Neck) OR (Cancer of Head) OR (Head Cancers) OR (Head Cancer) OR (Cancer, Head) OR (Cancers, Head) OR (Cancer of the Head) OR (Cancer of Neck) OR (Neck Cancers) OR (Neck Cancer) OR (Cancer, Neck) OR (Cancers, Neck) OR (Cancer of the Neck))) AND TS = (Methuosis) OR (Methuosis Cell Death) OR (Cell Death, Methuosis) OR (Cell Deaths, Methuosis) OR (Death, Methuosis Cell) OR (Deaths, Methuosis Cell) OR (Methuosis Cell Deaths)).

Appendix B. List of Excluded Studies along with Reasons for Exclusion

Study (Year)DOIReason for Exclusion *
Almangush et al. (2020)https://doi.org/10.1186/s12885-020-07342-xDid not evaluate non-apoptotic programmed cell death
Deng et al. (2022)https://doi.org/10.1002/jcla.24292Study not specific to OSCC
Fan et al. (2021)https://doi.org/10.3389/fgene.2021.732211Study not specific to OSCC
Fukuda et al. (2021)https://doi.org/10.21873/anticanres.14944Unable to access the full article
Gohara et al. (2022)https://doi.org/10.1016/j.omto.2022.10.001Study not specific to OSCC
Goreger et al. (2017)https://doi.org/10.1186/s12865-016-0185-5Study not specific to OSCC
He et al. (2021)https://doi.org/10.1016/j.intimp.2021.107789Study not specific to OSCC
Huang et al. (2022)https://doi.org/10.1016/j.intimp.2021.108431Study not specific to OSCC
Kosim et al. (2023)https://doi.org/10.3389/fphar.2022.988335Did not evaluate non-apoptotic programmed cell death
Li et al. (2022)https://doi.org/10.1002/cam4.4825Study not specific to OSCC
Li et al. (2023)https://doi.org/10.1186/s13027-023-00507-wStudy not specific to OSCC
Liu et al. (2023)https://doi.org/10.3389/fgene.2022.988606Study not specific to OSCC
Lu et al. (2021)https://doi.org/10.3389/fgene.2021.755486Study not specific to OSCC
Lu et al. (2022)https://doi.org/10.1155/2022/1539659Study not specific to OSCC
Qian et al. (2021)https://doi.org/10.2147/IJGM.S337089Study not specific to OSCC
Roh et al. (2017)https://doi.org/10.1016/j.redox.2016.12.010Study not specific to OSCC
Savic et al. (2023)https://doi.org/10.3390/cells12020336Study not specific to OSCC
Shin et al. (2018)https://doi.org/10.1016/j.freeradbiomed.2018.10.426Study not specific to OSCC
Takasu et al. (2016)https://doi.org/10.1038/cgt.2016.8Study not specific to OSCC
Tian et al. (2022)https://doi.org/10.4103/2221-1691.357743Study not specific to OSCC
Wang et al. (2022)https://doi.org/10.1016/j.csbj.2022.06.046Mixed together tumors from several locations
Wei et al. (2020)https://doi.org/10.21037/atm.2020.02.36Study not specific to OSCC
Wei et al. (2022)https://doi.org/10.3892/etm.2022.11449Study not specific to OSCC
Wu et al. (2022)https://doi.org/10.1155/2022/7602482Study not specific to OSCC
Wu et al. (2022)https://doi.org/10.3389/fcell.2022.702224Study not specific to OSCC
Xu et al. (2021)https://doi.org/10.1155/2021/5759927Study not specific to OSCC
Yaghmaei et al. (2017)https://doi.org/10.2174/1871520616666160725110844Unable to access the full article
Yu et al. (2020)https://doi.org/10.3390/cancers12061670Study not specific to OSCC
Yu et al. (2022)https://doi.org/10.1155/2022/3713929Unable to access the full article
Zhou et al. (2022)https://doi.org/10.1002/cam4.4819Study not specific to OSCC
Zhu et al. (2021)https://doi.org/10.1016/j.intimp.2021.108268Study not specific to OSCC
* Abbreviation: OSCC: oral squamous cell carcinoma.

Appendix C. Risk of Bias Assessment of In Vitro Studies

Experimental ConditionsBlinding *Complete OutcomeExposure CharacterizationOutcome AssessmentReportingOtherOverall Risk
Ruggieri et al. [92]LowNILowLowLowLowLowLow Risk
Sulkshane and Teni [84]HighNILowLowLowLowHighModerate Risk
Feng et al. [64]LowNILowLowLowLowLowLow Risk
Okazaki et al. [18]LowNILowLowLowLowLowLow Risk
Garley et al. [93]LowNILowHighLowLowLowModerate Risk
Yue et al. [65]LowNILowLowLowLowLowLow Risk
Huang et al. [19]LowNILowLowLowLowLowLow Risk
Zhu et al. [21]LowNILowLowLowLowLowLow Risk
Sato et al. [20]LowNILowLowLowLowLowLow Risk
Huang et al. [66]LowNILowLowLowLowLowLow Risk
Lee et al. [23]LowNILowLowLowLowLowLow Risk
Lin et al. [24]LowNILowLowLowLowLowLow Risk
Hémon et al. [22]LowNILowLowLowLowLowLow Risk
Uzunparmak et al. [86]LowNILowLowLowLowLowLow Risk
Li et al. [96]LowNILowLowLowLowLowLow Risk
Garley et al. [94]LowNILowHighLowLowLowModerate Risk
Chen et al. [95]LowNILowLowLowLowLowLow Risk
Yang et al. [69]LowNILowLowLowLowLowLow Risk
Wang et al. [28]LowNILowLowLowLowLowLow Risk
Yao et al. [70]LowNILowLowLowLowLowLow Risk
Luo et al. [68]LowNILowLowLowLowLowLow Risk
Jiang et al. [67]LowNILowLowLowLowLowLow Risk
Yang et al. [29]LowNILowLowLowLowLowLow Risk
Tomita et al. [27]LowNILowNILowLowLowLow Risk
You et al. [30]LowNILowLowLowLowLowLow Risk
You et al. [31]LowNILowLowLowLowLowLow Risk
Zhang et al. [32]LowNILowLowLowLowLowLow Risk
Huang et al. [87]LowNILowLowLowLowLowLow Risk
Li et al. [35]LowNILowLowLowLowLowLow Risk
Rioja-Blanco et al. [71]LowNILowLowLowLowLowLow Risk
Zhu et al. [77]LowNILowLowLowLowLowLow Risk
Wang et al. [73]LowNILowLowLowLowLowLow Risk
Shen et al. [72]LowNILowLowLowLowLowLow Risk
Zhu et al. [78]LowNILowLowLowLowLowLow Risk
Bhuyan et al. [33]LowNILowLowLowLowLowLow Risk
Lu et al. [40]LowNILowHighLowLowLowModerate Risk
Liu et al. [39]LowNILowLowLowLowLowLow Risk
Xu C. et al. [44]LowNILowLowLowLowLowLow Risk
Liu et al. [38]LowNILowNILowLowLowLow Risk
Han and Wu [34]LowNILowHighLowLowLowModerate Risk
Sun et al. [42]LowNILowLowLowLowLowLow Risk
Wang et al. [58]LowNILowHighLowLowLowModerate Risk
Wang et al. [43]LowNILowLowLowLowLowLow Risk
Li J, Tang and Ma [35]LowNILowLowLowLowLowLow Risk
Yang et al. [46]LowNILowNILowLowHighModerate Risk
Kaokaen et al. [88]LowNILowLowLowLowLowLow Risk
Liu et al. [80]LowNILowLowLowLowLowLow Risk
Wang et al. [82]LowNILowLowLowLowLowLow Risk
Nan et al. [81]LowNILowLowLowLowLowLow Risk
Zi et al. [79]LowNILowLowLowLowLowLow Risk
Yan et al. [83]LowNILowLowLowLowLowLow Risk
Wu et al. [59]LowNILowLowLowLowLowLow Risk
Pan et al. [55]LowNILowLowLowLowLowLow Risk
Jehl et al. [52]LowNILowLowLowLowLowLow Risk
Xie et al. [60]LowNILowLowLowLowLowLow Risk
Zhang et al. [62]LowNILowLowLowLowLowLow Risk
Yu et al. [61]LowNILowLowLowLowLowLow Risk
Wang et al. [56]LowNILowLowLowLowLowLow Risk
Li et al. [54]LowNILowLowLowLowLowLow Risk
Wang et al. [57]LowNILowHighLowLowLowModerate Risk
Zhao and Zhu [63]LowNILowHighLowLowLowModerate Risk
Lee and Roh [53]LowNILowHighLowLowLowModerate Risk
Gupta et al. [89]LowNILowLowLowLowLowLow Risk
Yun et al. [91]LowNILowLowLowLowLowLow Risk
Huang et al. [90]LowNILowLowLowLowLowLow Risk
Zhou et al. [76]LowNILowLowLowLowLowLow Risk
* Abbreviation: NI: not informed.

Appendix D. Risk of Bias of In Vivo Studies Based on Animal Models Using the SYRCLE Risk of Bias Tool

Sequence Generation (Selection Bias)Baseline Characteristics (Selection Bias)Allocation Concealment (Selection Bias)Random Housing (Performance Bias)Blinding (Performance Bias)Random Outcome Assessment (Detection Bias)Blinding (Detection Bias)Incomplete Outcome Data (Attrition Bias)Selective Outcome Reporting (Reporting Bias)Other Sources of Bias
Feng et al. [64]LowUnclearUnclearUnclearUnclearUnclearHighHighLowHigh
Sulkshane and Teni [84]LowUnclearUnclearUnclearUnclearUnclearHighHighLowLow
Okazaki et al. [18]UnclearUnclearUnclearUnclearUnclearUnclearUnclearHighUnclearHigh
Huang et al. [19]LowUnclearUnclearUnclearUnclearUnclearHighHighLowLow
Huang et al. [66]UnclearUnclearUnclearUnclearUnclearUnclearHighHighLowLow
Lee et al. [23]UnclearUnclearUnclearUnclearUnclearUnclearHighHighLowHigh
Uzunparmak et al. [86]LowUnclearUnclearUnclearUnclearUnclearHighHighLowLow
Yang et al. [29]UnclearUnclearUnclearUnclearUnclearUnclearHighHighLowLow
Wang et al. [28]LowLowUnclearUnclearUnclearUnclearHighHighLowLow
Yao et al. [70]LowUnclearUnclearUnclearUnclearUnclearHighHighLowLow
Luo et al. [68]LowUnclearUnclearUnclearUnclearUnclearHighHighLowLow
Yang et al. [69]UnclearUnclearUnclearUnclearUnclearUnclearHighHighLowLow
You et al. [30]UnclearUnclearUnclearUnclearUnclearUnclearHighHighLowLow
Zhang et al. [32]LowUnclearUnclearUnclearUnclearUnclearHighHighLowLow
Li et al. [96]LowLowUnclearUnclearUnclearUnclearHighHighLowLow
Rioja-Blanco et al. [71]LowLowUnclearUnclearUnclearUnclearHighHighLowLow
Shen et al. [72]LowLowUnclearUnclearUnclearUnclearHighHighLowHigh
Zhu et al. [77]LowUnclearUnclearUnclearUnclearUnclearUnclearHighLowHigh
Xu et al. [44]UnclearUnclearUnclearUnclearUnclearUnclearHighHighLowLow
Wang et al. [57]LowUnclearUnclearUnclearUnclearUnclearHighHighLowLow
Yang et al. [46]LowLowUnclearUnclearUnclearUnclearHighHighLowLow
Liu et al. [44]LowLowUnclearUnclearUnclearUnclearHighHighLowLow
Wang et al. [82]LowLowUnclearUnclearUnclearUnclearHighHighLowLow
Zi et al. [79]LowUnclearUnclearUnclearUnclearUnclearHighHighLowHigh
Wu et al. [59]LowLowUnclearUnclearUnclearUnclearHighHighLowLow
Chung et al. [50]UnclearUnclearUnclearUnclearUnclearUnclearHighHighLowLow
Wang et al. [56]LowUnclearUnclearUnclearUnclearUnclearHighHighLowLow
Wang et al. [43]LowUnclearUnclearUnclearUnclearUnclearHighHighLowLow
Zhao and Zhu [63]UnclearUnclearUnclearUnclearUnclearUnclearHighHighUnclearLow
Zhou et al. [76]LowUnclearUnclearUnclearUnclearUnclearHighHighLowLow
Zhou et al. [48]UnclearUnclearUnclearUnclearUnclearUnclearHighHighLowLow

References

  1. Safa, A.R. Resistance to Cell Death and Its Modulation in Cancer Stem Cells. Crit. Rev. Oncog. 2016, 21, 203. [Google Scholar] [CrossRef] [PubMed]
  2. 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]
  3. Yan, G.; Elbadawi, M.; Efferth, T. Multiple cell death modalities and their key features (Review). World Acad. Sci. J. 2020, 2, 39–48. [Google Scholar] [CrossRef]
  4. Peng, F.; Liao, M.; Qin, R.; Zhu, S.; Peng, C.; Fu, L.; Chen, Y.; Han, B. Regulated cell death (RCD) in cancer: Key pathways and targeted therapies. Signal Transduct. Target. Ther. 2022, 7, 286. [Google Scholar] [CrossRef]
  5. Tong, X.; Tang, R.; Xiao, M.; Xu, J.; Wang, W.; Zhang, B.; Liu, J.; Yu, X.; Shi, S. Targeting cell death pathways for cancer therapy: Recent developments in necroptosis, pyroptosis, ferroptosis, and cuproptosis research. J. Hematol. Oncol. 2022, 15, 174. [Google Scholar] [CrossRef] [PubMed]
  6. Wang, X.; Hua, P.; He, C.; Chen, M. Non-apoptotic cell death-based cancer therapy: Molecular mechanism, pharmacological modulators, and nanomedicine. Acta Pharm. Sin. B 2022, 12, 3567–3593. [Google Scholar] [CrossRef]
  7. Xi, Y.; Gao, L.; Li, S.; Sun, K.; Chen, P.; Cai, Z.; Ren, W.; Zhi, K. The role of novel programmed cell death in head and neck squamous cell carcinoma: From mechanisms to potential therapies. Front. Pharmacol. 2023, 14, 1228985. [Google Scholar] [CrossRef] [PubMed]
  8. Bedoui, S.; Herold, M.J.; Strasser, A. Emerging connectivity of programmed cell death pathways and its physiological implications. Nat. Rev. Mol. Cell Biol. 2020, 21, 678–695. [Google Scholar] [CrossRef]
  9. Li, J.; He, D.; Li, S.; Xiao, J.; Zhu, Z. Ferroptosis: The emerging player in remodeling triple-negative breast cancer. Front. Immunol. 2023, 14, 1284057. [Google Scholar] [CrossRef]
  10. Jia, Y.-J.; Zhang, Y.; Ma, X.-B.; Wang, Y.; Tian, Y.-Q.; He, P.-X.; Xu, Y.-C. Ferroptosis: Opportunities and Challenges in Cancer. J. Explor. Res. Pharmacol. 2023, 8, 243–253. [Google Scholar] [CrossRef]
  11. Frank, D.; Vince, J.E. Pyroptosis versus necroptosis: Similarities, differences, and crosstalk. Cell Death Differ. 2019, 26, 99–114. [Google Scholar] [CrossRef]
  12. Bertheloot, D.; Latz, E.; Franklin, B.S. Necroptosis, pyroptosis and apoptosis: An intricate game of cell death. Cell. Mol. Immunol. 2021, 18, 1106–1121. [Google Scholar] [CrossRef] [PubMed]
  13. Kayagaki, N.; Webster, J.D.; Newton, K. Control of Cell Death in Health and Disease. Annu. Rev. Pathol. Mech. Dis. 2023, 19, 157–180. [Google Scholar] [CrossRef]
  14. Gulia, S.; Chandra, P.; Das, A. The Prognosis of Cancer Depends on the Interplay of Autophagy, Apoptosis, and Anoikis within the Tumor Microenvironment. Cell Biochem. Biophys. 2023, 81, 621–658. [Google Scholar] [CrossRef] [PubMed]
  15. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. BMJ 2021, 372, 71. [Google Scholar] [CrossRef] [PubMed]
  16. Hooijmans, C.R.; Rovers, M.M.; de Vries, R.B.M.; Leenaars, M.; Ritskes-Hoitinga, M.; Langendam, M.W. SYRCLE’s risk of bias tool for animal studies. BMC Med. Res. Methodol. 2014, 14, 43. [Google Scholar] [CrossRef] [PubMed]
  17. Bezemer, J.M.; van der Ende, J.; Limpens, J.; de Vries, H.J.C.; Schallig, H.D.F.H. Safety and efficacy of allylamines in the treatment of cutaneous and mucocutaneous leishmaniasis: A systematic review. PLoS ONE 2021, 16, e0249628. [Google Scholar] [CrossRef]
  18. Okazaki, S.; Shintani, S.; Hirata, Y.; Suina, K.; Semba, T.; Yamasaki, J.; Umene, K.; Ishikawa, M.; Saya, H.; Nagano, O. Synthetic lethality of the ALDH3A1 inhibitor dyclonine and xCT inhibitors in glutathione deficiency-resistant cancer cells. Oncotarget 2018, 9, 33832–33843. [Google Scholar] [CrossRef]
  19. Huang, K.-J.; Wei, Y.-H.; Chiu, Y.-C.; Wu, S.-R.; Shieh, D.-B. Assessment of zero-valent iron-based nanotherapeutics for ferroptosis induction and resensitization strategy in cancer cells. Biomater. Sci. 2019, 7, 1311–1322. [Google Scholar] [CrossRef]
  20. Sato, K.; Shi, L.; Ito, F.; Ohara, Y.; Motooka, Y.; Tanaka, H.; Mizuno, M.; Hori, M.; Hirayama, T.; Hibi, H.; et al. Non-thermal plasma specifically kills oral squamous cell carcinoma cells in a catalytic Fe(II)-dependent manner. J. Clin. Biochem. Nutr. 2019, 65, 8–15. [Google Scholar] [CrossRef] [PubMed]
  21. Zhu, T.; Shi, L.; Yu, C.; Dong, Y.; Qiu, F.; Shen, L.; Qian, Q.; Zhou, G.; Zhu, X. Ferroptosis Promotes Photodynamic Therapy: Supramolecular Photosensitizer-Inducer Nanodrug for Enhanced Cancer Treatment. Theranostics 2019, 9, 3293–3307. [Google Scholar] [CrossRef] [PubMed]
  22. Hémon, A.; Louandre, C.; Lailler, C.; Godin, C.; Bottelin, M.; Morel, V.; François, C.; Galmiche, A.; Saidak, Z. SLC7A11 as a biomarker and therapeutic target in HPV-positive head and neck Squamous Cell Carcinoma. Biochem. Biophys. Res. Commun. 2020, 533, 1083–1087. [Google Scholar] [CrossRef] [PubMed]
  23. Lee, J.; You, J.H.; Shin, D.; Roh, J.-L. Inhibition of Glutaredoxin 5 predisposes Cisplatin-resistant Head and Neck Cancer Cells to Ferroptosis. Theranostics 2020, 10, 7775–7786. [Google Scholar] [CrossRef] [PubMed]
  24. Lin, Y.H.; Chiu, V.; Huang, C.Y.; Tzeng, I.S.; Hsieh, P.C.; Kuo, C.Y. Promotion of Ferroptosis in Oral Cancer Cell Lines by Chrysophanol. Curr. Top. Nutraceutical Res. 2020, 18, 273–276. [Google Scholar] [CrossRef]
  25. Gu, W.; Kim, M.; Wang, L.; Yang, Z.; Nakajima, T.; Tsushima, Y. Multi-omics Analysis of Ferroptosis Regulation Patterns and Characterization of Tumor Microenvironment in Patients with Oral Squamous Cell Carcinoma. Int. J. Biol. Sci. 2021, 17, 3476–3492. [Google Scholar] [CrossRef] [PubMed]
  26. Li, H.; Zhang, X.; Yi, C.; He, Y.; Chen, X.; Zhao, W.; Yu, D. Ferroptosis-related gene signature predicts the prognosis in Oral squamous cell carcinoma patients. BMC Cancer 2021, 21, 835. [Google Scholar] [CrossRef] [PubMed]
  27. Tomita, K.; Nagasawa, T.; Kuwahara, Y.; Torii, S.; Igarashi, K.; Roudkenar, M.H.; Roushandeh, A.M.; Kurimasa, A.; Sato, T. MiR-7-5p Is Involved in Ferroptosis Signaling and Radioresistance Thru the Generation of ROS in Radioresistant HeLa and SAS Cell Lines. Int. J. Mol. Sci. 2021, 22, 8300. [Google Scholar] [CrossRef]
  28. Wang, X.; Liu, K.; Gong, H.; Li, D.; Chu, W.; Zhao, D.; Wang, X.; Xu, D. Death by histone deacetylase inhibitor quisinostat in tongue squamous cell carcinoma via apoptosis, pyroptosis, and ferroptosis. Toxicol. Appl. Pharmacol. 2020, 410, 115363. [Google Scholar] [CrossRef]
  29. Yang, J.; Cao, X.-H.; Luan, K.-F.; Huang, Y.-D. Circular RNA FNDC3B Protects Oral Squamous Cell Carcinoma Cells From Ferroptosis and Contributes to the Malignant Progression by Regulating miR-520d-5p/SLC7A11 Axis. Front. Oncol. 2021, 11, 672724. [Google Scholar] [CrossRef]
  30. You, J.H.; Lee, J.; Roh, J.-L. Mitochondrial pyruvate carrier 1 regulates ferroptosis in drug-tolerant persister head and neck cancer cells via epithelial-mesenchymal transition. Cancer Lett. 2021, 507, 40–54. [Google Scholar] [CrossRef]
  31. You, J.H.; Lee, J.; Roh, J.-L. PGRMC1-dependent lipophagy promotes ferroptosis in paclitaxel-tolerant persister cancer cells. J. Exp. Clin. Cancer Res. 2021, 40, 1–18. [Google Scholar] [CrossRef]
  32. Zhang, X.; Yang, S.; Wang, Q.; Ye, W.; Liu, S.; Wang, X.; Zhang, Z.; Cao, L.; Jiang, X. Tailored theranostic nanoparticles cause efficient ferroptosis in head and neck squamous cell carcinoma through a reactive oxygen species “butterfly effect”. Chem. Eng. J. 2021, 423, 130083. [Google Scholar] [CrossRef]
  33. Bhuyan, S.; Pal, B.; Pathak, L.; Saikia, P.J.; Mitra, S.; Gayan, S.; Mokhtari, R.B.; Li, H.; Ramana, C.V.; Baishya, D.; et al. Targeting hypoxia-induced tumor stemness by activating pathogen-induced stem cell niche defense. Front. Immunol. 2022, 13, 933329. [Google Scholar] [CrossRef]
  34. Han, L.; Li, L.; Wu, G. Induction of ferroptosis by carnosic acid-mediated inactivation of Nrf2/HO-1 potentiates cisplatin responsiveness in OSCC cells. Mol. Cell. Probes 2022, 64, 101821. [Google Scholar] [CrossRef]
  35. Li, J.; Tang, L.-L.; Ma, J. Survival-related indicators ALOX12B and SPRR1A are associated with DNA damage repair and tumor microenvironment status in HPV 16-negative head and neck squamous cell carcinoma patients. BMC Cancer 2022, 22, 714. [Google Scholar] [CrossRef]
  36. Li, T.; Wang, Y.; Xiang, X.; Chen, C. Development and Validation of a Ferroptosis-Related lncRNAs Prognosis Model in Oral Squamous Cell Carcinoma. Front. Genet. 2022, 13, 847940. [Google Scholar] [CrossRef]
  37. Liu, F.; Tang, L.; Li, Q.; Chen, L.; Pan, Y.; Yin, Z.; He, J.; Tian, J. Single-cell transcriptomics uncover the key ferroptosis regulators contribute to cancer progression in head and neck squamous cell carcinoma. Front. Mol. Biosci. 2022, 9, 962742. [Google Scholar] [CrossRef]
  38. Liu, J.; An, W.; Zhao, Q.; Liu, Z.; Jiang, Y.; Li, H.; Wang, D. Hyperbaric oxygen enhances X-ray induced ferroptosis in oral squamous cell carcinoma cells. Oral Dis. 2022. online ahead of print. [Google Scholar] [CrossRef]
  39. Liu, S.; Yan, S.; Zhu, J.; Lu, R.; Kang, C.; Tang, K.; Zeng, J.; Ding, M.; Guo, Z.; Lai, X.; et al. Combination RSL3 Treatment Sensitizes Ferroptosis- and EGFR-Inhibition-Resistant HNSCCs to Cetuximab. Int. J. Mol. Sci. 2022, 23, 9014. [Google Scholar] [CrossRef]
  40. Lu, T.; Zhang, Z.; Pan, X.; Zhang, J.; Wang, X.; Wang, M.; Li, H.; Yan, M.; Chen, W. Caveolin-1 promotes cancer progression via inhibiting ferroptosis in head and neck squamous cell carcinoma. J. Oral Pathol. Med. 2022, 51, 52–62. [Google Scholar] [CrossRef]
  41. Qiu, L.; Tao, A.; Liu, F.; Ge, X.; Li, C. Potential prognostic value of a eight ferroptosis-related lncRNAs model and the correlative immune activity in oral squamous cell carcinoma. BMC Genet. 2022, 23, 80. [Google Scholar] [CrossRef] [PubMed]
  42. Sun, K.; Ren, W.; Li, S.; Zheng, J.; Huang, Y.; Zhi, K.; Gao, L. MiR-34c-3p upregulates erastin-induced ferroptosis to inhibit proliferation in oral squamous cell carcinomas by targeting SLC7A11. Pathol. Res. Pract. 2022, 231, 153778. [Google Scholar] [CrossRef] [PubMed]
  43. Wang, M.; Li, F.; Lu, T.; Wu, R.; Yang, S.; Chen, W. Photodynamic and ferroptotic Ce6@ZIF-8@ssPDA for head and neck cancer treatment. Mater. Des. 2022, 224, 111403. [Google Scholar] [CrossRef]
  44. Xu, C.; Shen, Y.; Shi, Y.; Zhang, M.; Zhou, L. Eukaryotic translation initiation factor 3 subunit B promotes head and neck cancer via CEBPB translation. Cancer Cell Int. 2022, 22, 161. [Google Scholar] [CrossRef] [PubMed]
  45. Xu, Y.; Hong, M.; Kong, D.; Deng, J.; Zhong, Z.; Liang, J. Ferroptosis-associated DNA methylation signature predicts overall survival in patients with head and neck squamous cell carcinoma. BMC Genom. 2022, 23, 63. [Google Scholar] [CrossRef] [PubMed]
  46. Yang, Y.; Tang, H.; Zheng, J.; Yang, K. The PER1/HIF-1alpha negative feedback loop promotes ferroptosis and inhibits tumor progression in oral squamous cell carcinoma. Transl. Oncol. 2022, 18, 101360. [Google Scholar] [CrossRef]
  47. Yin, J.; Fu, J.; Zhao, Y.; Xu, J.; Chen, C.; Zheng, L.; Wang, B. Comprehensive Analysis of the Significance of Ferroptosis-Related Genes in the Prognosis and Immunotherapy of Oral Squamous Cell Carcinoma. Bioinform. Biol. Insights 2022, 16, 1–13. [Google Scholar] [CrossRef]
  48. Zhou, Q.; Wang, X.; Zhang, Y.; Wang, L.; Chen, Z. Inhibition of AEBP1 predisposes cisplatin-resistant oral cancer cells to ferroptosis. BMC Oral Health 2022, 22, 478. [Google Scholar] [CrossRef]
  49. Zhu, H.; Tao, Y.; Huang, Q.; Chen, Z.; Jiang, L.; Yan, H.; Zhong, J.; Liang, L. Identification of ferroptosis-related genes as potential biomarkers of tongue squamous cell carcinoma using an integrated bioinformatics approach. FEBS Open Bio 2022, 12, 412–429. [Google Scholar] [CrossRef]
  50. Chung, C.; Lin, C.; Chen, C.; Hsueh, C.; Chang, Y.; Wang, C.; Chu, P.; Tai, S.; Yang, M. Ferroptosis Signature Shapes the Immune Profiles to Enhance the Response to Immune Checkpoint Inhibitors in Head and Neck Cancer. Adv. Sci. 2023, 10, 2204514. [Google Scholar] [CrossRef] [PubMed]
  51. Fan, X.; Zhong, Y.; Yuan, F.; Zhang, L.; Cai, Y.; Liao, L. A ferroptosis-related prognostic model with excellent clinical performance based on the exploration of the mechanism of oral squamous cell carcinoma progression. Sci. Rep. 2023, 13, 1461. [Google Scholar] [CrossRef]
  52. Jehl, A.; Conrad, O.; Burgy, M.; Foppolo, S.; Vauchelles, R.; Ronzani, C.; Etienne-Selloum, N.; Chenard, M.-P.; Danic, A.; Dourlhes, T.; et al. Blocking EREG/GPX4 Sensitizes Head and Neck Cancer to Cetuximab through Ferroptosis Induction. Cells 2023, 12, 733. [Google Scholar] [CrossRef] [PubMed]
  53. Lee, J.; Roh, J.-L. Promotion of ferroptosis in head and neck cancer with divalent metal transporter 1 inhibition or salinomycin. Hum. Cell 2023, 36, 1090–1098. [Google Scholar] [CrossRef] [PubMed]
  54. Li, J.; Xiao, W.; Wei, W.; Wu, M.; Xiong, K.; Lyu, J.; Li, Y. HSPA5, as a ferroptosis regulator, may serve as a potential therapeutic for head and neck squamous cell carcinoma. Mol. Immunol. 2023, 158, 79–90. [Google Scholar] [CrossRef] [PubMed]
  55. Pan, X.; Xu, X.; Wang, L.; Zhang, S.; Chen, Y.; Yang, R.; Chen, X.; Cheng, B.; Xia, J.; Ren, X. BASP1 is a prognostic biomarker associated with immunotherapeutic response in head and neck squamous cell carcinoma. Front. Oncol. 2023, 13, 1021262. [Google Scholar] [CrossRef]
  56. Wang, Z.; Han, J.; Guo, Z.; Wu, H.; Liu, Y.; Wang, W.; Zhang, C.; Liu, J. Ginseng-based carbon dots inhibit the growth of squamous cancer cells by increasing ferroptosis. Front. Oncol. 2023, 13, 1097692. [Google Scholar] [CrossRef] [PubMed]
  57. Wang, L.; Wang, C.; Li, X.; Tao, Z.; Zhu, W.; Su, Y.; Choi, W.S. Melatonin and erastin emerge synergistic anti-tumor effects on oral squamous cell carcinoma by inducing apoptosis, ferroptosis, and inhibiting autophagy through promoting ROS. Cell. Mol. Biol. Lett. 2023, 28, 36. [Google Scholar] [CrossRef] [PubMed]
  58. Wang, Z.-Q.; Li, Y.-Q.; Wang, D.-Y.; Shen, Y.-Q. Natural product piperlongumine inhibits proliferation of oral squamous carcinoma cells by inducing ferroptosis and inhibiting intracellular antioxidant capacity. Transl. Cancer Res. 2022, 12, 2911–2922. [Google Scholar] [CrossRef] [PubMed]
  59. Wu, X.; Wu, Q.; Wang, Y.; Liu, Y.; Li, Z.; Liu, Q.; Huang, Z.; Li, M.; Zhang, B.; Zhan, Q. Aqueous-soluble components of sporoderm-removed Ganoderma lucidum spore powder promote ferroptosis in oral squamous cell carcinoma. Chin. J. Cancer Res. 2023, 35, 176–190. [Google Scholar] [CrossRef]
  60. Xie, J.; Lan, T.; Zheng, D.-L.; Ding, L.-C.; Lu, Y.-G. CDH4 inhibits ferroptosis in oral squamous cell carcinoma cells. BMC Oral Health 2023, 23, 329. [Google Scholar] [CrossRef]
  61. Yu, Y.; MohamedAl-Sharani, H.; Zhang, B. EZH2-mediated SLC7A11 upregulation via miR-125b-5p represses ferroptosis of TSCC. Oral Dis. 2021, 29, 880–891. [Google Scholar] [CrossRef]
  62. Zhang, P.; Cui, Y.; Wang, J.; Cheng, J.; Zhu, L.; Liu, C.; Yue, S.; Pang, R.; Guan, J.; Xie, B.; et al. Dual-stimuli responsive smart nanoprobe for precise diagnosis and synergistic multi-modalities therapy of superficial squamous cell carcinoma. J. Nanobiotechnology 2023, 21, 4. [Google Scholar] [CrossRef]
  63. Zhao, Y.; Zhu, S. Nrf2/HO-1 Alleviates Disulfiram/Copper-Induced Ferroptosis in Oral Squamous Cell Carcinoma. Biochem. Genet. 2023. online ahead of print. [Google Scholar] [CrossRef] [PubMed]
  64. Feng, X.; Luo, Q.; Zhang, H.; Wang, H.; Chen, W.; Meng, G.; Chen, F. The role of NLRP3 inflammasome in 5-fluorouracil resistance of oral squamous cell carcinoma. J. Exp. Clin. Cancer Res. 2017, 36, 81. [Google Scholar] [CrossRef] [PubMed]
  65. Yue, E.; Tuguzbaeva, G.; Chen, X.; Qin, Y.; Li, A.; Sun, X.; Dong, C.; Liu, Y.; Yu, Y.; Zahra, S.M.; et al. Anthocyanin is involved in the activation of pyroptosis in oral squamous cell carcinoma. Phytomedicine 2019, 56, 286–294. [Google Scholar] [CrossRef] [PubMed]
  66. Huang, Z.; Zhang, Q.; Wang, Y.; Chen, R.; Wang, Y.; Huang, Z.; Zhou, G.; Li, H.; Rui, X.; Jin, T.; et al. Inhibition of caspase-3-mediated GSDME-derived pyroptosis aids in noncancerous tissue protection of squamous cell carcinoma patients during cisplatin-based chemotherapy. Am. J. Cancer Res. 2020, 10, 4287. [Google Scholar] [PubMed]
  67. Jiang, L.; Ge, W.; Cui, Y.; Wang, X. The regulation of long non-coding RNA 00958 (LINC00958) for oral squamous cell carcinoma (OSCC) cells death through absent in melanoma 2 (AIM2) depending on microRNA-4306 and Sirtuin1 (SIRT1) in vitro. Bioengineered 2021, 12, 5085–5098. [Google Scholar] [CrossRef] [PubMed]
  68. Luo, Q.; Li, X.; Gan, G.; Yang, M.; Chen, X.; Chen, F. PPT1 Reduction Contributes to Erianin-Induced Growth Inhibition in Oral Squamous Carcinoma Cells. Front. Cell Dev. Biol. 2021, 9, 764263. [Google Scholar] [CrossRef] [PubMed]
  69. Yang, M.; Luo, Q.; Chen, X.; Chen, F. Bitter melon derived extracellular vesicles enhance the therapeutic effects and reduce the drug resistance of 5-fluorouracil on oral squamous cell carcinoma. J. Nanobiotechnol. 2021, 19, 259. [Google Scholar] [CrossRef] [PubMed]
  70. Yao, Y.; Shen, X.; Zhou, M.; Tang, B. Periodontal Pathogens Promote Oral Squamous Cell Carcinoma by Regulating ATR and NLRP3 Inflammasome. Front. Oncol. 2021, 11, 722797. [Google Scholar] [CrossRef]
  71. Rioja-Blanco, E.; Arroyo-Solera, I.; Álamo, P.; Casanova, I.; Gallardo, A.; Unzueta, U.; Serna, N.; Sánchez-García, L.; Quer, M.; Villaverde, A.; et al. CXCR4-targeted nanotoxins induce GSDME-dependent pyroptosis in head and neck squamous cell carcinoma. J. Exp. Clin. Cancer Res. 2022, 41, 49. [Google Scholar] [CrossRef]
  72. Shen, X.; Zhang, B.; Hu, X.; Li, J.; Wu, M.; Yan, C.; Yang, Y.; Li, Y. Neisseria sicca and Corynebacterium matruchotii inhibited oral squamous cell carcinomas by regulating genome stability. Bioengineered 2022, 13, 14094–14106. [Google Scholar] [CrossRef]
  73. Wang, S.; Zhang, M.; Wu, Z.; Zhu, S.; Wan, S.; Zhang, B.; Yang, Q.; Xiao, Y.; Chen, L.; Sun, Z. GSDME Is Related to Prognosis and Response to Chemotherapy in Oral Cancer. J. Dent. Res. 2022, 101, 848–858. [Google Scholar] [CrossRef]
  74. Xin, Y.; Zhang, J.; Jiang, Q.; Qiu, J. Construction of prognostic signature of patients with oral squamous cell carcinoma based on pyroptosis-related long non-coding RNAs. Front. Surg. 2022, 9, 935765. [Google Scholar] [CrossRef]
  75. Zeng, D.; Wang, X.; Zhang, S.; Zheng, A.; Huang, Q.; Cao, L. Pyroptosis-related gene-based prognostic signature for predicting the overall survival of oral squamous cell carcinoma patients. Front. Surg. 2022, 9, 903271. [Google Scholar] [CrossRef]
  76. Zhou, J.-Y.; Wang, W.-J.; Zhang, C.-Y.; Ling, Y.-Y.; Hong, X.-J.; Su, Q.; Li, W.-G.; Mao, Z.-W.; Cheng, B.; Tan, C.-P.; et al. Ru(II)-modified TiO2 nanoparticles for hypoxia-adaptive photo-immunotherapy of oral squamous cell carcinoma. Biomaterials 2022, 289, 121757. [Google Scholar] [CrossRef]
  77. Zhu, S.-W.; Ye, M.; Ma, X.; Wu, Z.-Z.; Wan, S.-C.; Yang, S.-C.; Li, H.; Xu, Z.; Sun, Z.-J. pH-responsive nanoprodrugs combining a Src inhibitor and chemotherapy to potentiate antitumor immunity via pyroptosis in head and neck cancer. Acta Biomater. 2022, 154, 497–509. [Google Scholar] [CrossRef] [PubMed]
  78. Zhu, W.; Zhang, J.; Wang, M.; Zhai, R.; Xu, Y.; Wang, J.; Wang, M.; Zhang, H.; Liu, L. Development of a prognostic pyroptosis-related gene signature for head and neck squamous cell carcinoma patient. Cancer Cell Int. 2022, 22, 62. [Google Scholar] [CrossRef] [PubMed]
  79. Zi, M.; Xingyu, C.; Yang, C.; Xiaodong, S.; Shixian, L.; Shicheng, W. Improved antitumor immunity of chemotherapy in OSCC treatment by Gasdermin-E mediated pyroptosis. Apoptosis 2022, 28, 348–361. [Google Scholar] [CrossRef] [PubMed]
  80. Liu, X.; Zhan, W.; Gao, G.; Jiang, Q.; Zhang, X.; Zhang, H.; Sun, X.; Han, W.; Wu, F.-G.; Liang, G. Apoptosis-Amplified Assembly of Porphyrin Nanofiber Enhances Photodynamic Therapy of Oral Tumor. J. Am. Chem. Soc. 2023, 145, 7918–7930. [Google Scholar] [CrossRef] [PubMed]
  81. Nan, Z.; Dou, Y.; Chen, A.; Wang, K.; Sun, J.; Meng, Z.; Neckenig, M.; Ai, D.; Liu, S.; Dong, Z.; et al. Identification and validation of a prognostic signature of autophagy, apoptosis and pyroptosis-related genes for head and neck squamous cell carcinoma: To imply therapeutic choices of HPV negative patients. Front. Immunol. 2023, 13, 1100417. [Google Scholar] [CrossRef]
  82. Wang, S.; Wu, Z.-Z.; Zhu, S.-W.; Wan, S.-C.; Zhang, M.-J.; Zhang, B.-X.; Yang, Q.-C.; Xiao, Y.; Li, H.; Mao, L.; et al. CTLA-4 blockade induces tumor pyroptosis via CD8+ T cells in head and neck squamous cell carcinoma. Mol. Ther. 2023, 31, 2154–2168. [Google Scholar] [CrossRef]
  83. Yan, L.; Sun, Y.; Guo, J.; Jia, R. PD-L1 Exon 3 Is a Hidden Switch of Its Expression and Function in Oral Cancer Cells. Int. J. Mol. Sci. 2023, 24, 8193. [Google Scholar] [CrossRef] [PubMed]
  84. Sulkshane, P.; Teni, T. BH3 mimetic Obatoclax (GX15-070) mediates mitochondrial stress predominantly via MCL-1 inhibition and induces autophagy-dependent necroptosis in human oral cancer cells. Oncotarget 2016, 8, 60060–60079. [Google Scholar] [CrossRef] [PubMed]
  85. Li, J.; Huang, S.; Zeng, L.; Li, K.; Yang, L.; Gao, S.; Guan, C.; Zhang, S.; Lao, X.; Liao, G.; et al. Necroptosis in head and neck squamous cell carcinoma: Characterization of clinicopathological relevance and in vitro cell model. Cell Death Dis. 2020, 11, 391. [Google Scholar] [CrossRef] [PubMed]
  86. Uzunparmak, B.; Gao, M.; Lindemann, A.; Erikson, K.; Wang, L.; Lin, E.; Frank, S.J.; Gleber-Netto, F.O.; Zhao, M.; Skinner, H.D.; et al. Caspase-8 loss radiosensitizes head and neck squamous cell carcinoma to SMAC mimetic–induced necroptosis. J. Clin. Investig. 2020, 5, 139837. [Google Scholar] [CrossRef]
  87. Huang, Y.-C.; Yuan, T.-M.; Liu, B.-H.; Liu, K.-L.; Wung, C.-H.; Chuang, S.-M. Capsaicin Potentiates Anticancer Drug Efficacy Through Autophagy-Mediated Ribophorin II Downregulation and Necroptosis in Oral Squamous Cell Carcinoma Cells. Front. Pharmacol. 2021, 12, 676813. [Google Scholar] [CrossRef] [PubMed]
  88. Kaokaen, P.; Chaicharoenaudomrung, N.; Kunhorm, P.; Mesil, K.; Binlateh, T.; Noisa, P.; Jitprasertwong, P. Nanoencapsulation of Cordycepin Induces Switching from Necroptosis to Apoptosis in Human Oral Cancer Cells (HSC-4) Through Inhibition of Receptor-Interacting Serine/Threonine-Protein Kinase 3 (RIPK3) and Autophagy Modulation. Nat. Prod. Commun. 2022, 17, 1934578X221074838. [Google Scholar] [CrossRef]
  89. Gupta, P.; Singh, A.; Verma, A.K.; Kant, S.; Pandey, A.K.; Mishra, A.; Khare, P.; Prakash, V. Nanoencapsulation of Docetaxel Induces Concurrent Apoptosis and Necroptosis in Human Oral Cancer Cells (SCC-9) via TNF-α/RIP1/RIP3 Pathway. Indian J. Clin. Biochem. 2022, 38, 351–360. [Google Scholar] [CrossRef] [PubMed]
  90. Huang, K.; Gu, X.; Xu, H.; Li, H.; Shi, M.; Wei, D.; Wang, S.; Li, Y.; Liu, B.; Li, Y. Prognostic Value of Necroptosis-Related Genes Signature in Oral Squamous Cell Carcinoma. Cancers 2023, 15, 4539. [Google Scholar] [CrossRef]
  91. Yun, H.-M.; Kwon, Y.-J.; Kim, E.; Chung, H.-J.; Park, K.-R. Machilin D Promotes Apoptosis and Autophagy, and Inhibits Necroptosis in Human Oral Squamous Cell Carcinoma Cells. Int. J. Mol. Sci. 2023, 24, 4576. [Google Scholar] [CrossRef] [PubMed]
  92. Ruggieri, V.; Agriesti, F.; Scrima, R.; Laurenzana, I.; Perrone, D.; Tataranni, T.; Mazzoccoli, C.; Muzio, L.L.; Capitanio, N.; Piccoli, C. Dichloroacetate, a selective mitochondria-targeting drug for oral squamous cell carcinoma: A metabolic perspective of treatment. Oncotarget 2014, 6, 1217–1230. [Google Scholar] [CrossRef] [PubMed]
  93. Garley, M.; Dziemiańczyk-Pakieła, D.; Grubczak, K.; Surażyński, A.; Dąbrowska, D.; Ratajczak-Wrona, W.; Sawicka-Powierza, J.; Borys, J.; Moniuszko, M.; Pałka, J.A.; et al. Differences and similarities in the phenomenon of NETs formation in oral inflammation and in oral squamous cell carcinoma. J. Cancer 2018, 9, 1958–1965. [Google Scholar] [CrossRef]
  94. Garley, M.; Jabłońska, E.; Miltyk, W.; Grubczak, K.; Surażyński, A.; Ratajczak-Wrona, W.; Grudzińska, M.; Nowacka, K.H.; Moniuszko, M.; Pałka, J.A.; et al. Cancers Cells in Traps? The Pathways of NETs Formation in Response to OSCC in Humans—A Pilot Study. Cancer Control. 2020, 27, 1073274820960473. [Google Scholar] [CrossRef]
  95. Chen, Q.; Song, S.; Wang, Z.; Shen, Y.; Xie, L.; Li, J.; Jiang, L.; Zhao, H.; Feng, X.; Zhou, Y.; et al. Isorhamnetin induces the paraptotic cell death through ROS and the ERK/MAPK pathway in OSCC cells. Oral Dis. 2020, 27, 240–250. [Google Scholar] [CrossRef] [PubMed]
  96. Li, D.; Kou, Y.; Gao, Y.; Liu, S.; Yang, P.; Hasegawa, T.; Su, R.; Guo, J.; Li, M. Oxaliplatin induces the PARP1-mediated parthanatos in oral squamous cell carcinoma by increasing production of ROS. Aging 2021, 13, 4242–4257. [Google Scholar] [CrossRef] [PubMed]
  97. Li, J.; Cao, F.; Yin, H.; Huang, Z.; Lin, Z.; Mao, N.; Sun, B.; Wang, G. Ferroptosis: Past, present and future. Cell Death Dis. 2020, 11, 88. [Google Scholar] [CrossRef]
  98. Stockwell, B.R.; Angeli, J.P.F.; Bayir, H.; Bush, A.I.; Conrad, M.; Dixon, S.J.; Fulda, S.; Gascón, S.; Hatzios, S.K.; Kagan, V.E.; et al. Ferroptosis: A Regulated Cell Death Nexus Linking Metabolism, Redox Biology, and Disease. Cell 2017, 171, 273–285. [Google Scholar] [CrossRef]
  99. Zhang, C.; Liu, X.; Jin, S.; Chen, Y.; Guo, R. Ferroptosis in cancer therapy: A novel approach to reversing drug resistance. Mol. Cancer 2022, 21, 47. [Google Scholar] [CrossRef]
  100. Hou, W.; Xie, Y.; Song, X.; Sun, X.; Lotze, M.T.; Zeh, H.J., 3rd; Kang, R.; Tang, D. Autophagy promotes ferroptosis by degradation of ferritin. Autophagy 2016, 12, 1425–1428. [Google Scholar] [CrossRef] [PubMed]
  101. Chen, Y.; Fan, Z.; Hu, S.; Lu, C.; Xiang, Y.; Liao, S. Ferroptosis: A New Strategy for Cancer Therapy. Front. Oncol. 2022, 12, 830561. [Google Scholar] [CrossRef]
  102. Tang, D.; Chen, X.; Kang, R.; Kroemer, G. Ferroptosis: Molecular mechanisms and health implications. Cell Res. 2021, 31, 107–125. [Google Scholar] [CrossRef] [PubMed]
  103. Cao, J.Y.; Dixon, S.J. Mechanisms of ferroptosis. Cell. Mol. Life Sci. 2016, 73, 2195–2209. [Google Scholar] [CrossRef] [PubMed]
  104. Ying, J.F.; Lu, Z.B.; Fu, L.Q.; Tong, Y.; Wang, Z.; Li, W.F.; Mou, X.Z. The role of iron homeostasis and iron-mediated ROS in cancer. Am. J. Cancer Res. 2021, 11, 1895. [Google Scholar] [PubMed]
  105. Seibt, T.M.; Proneth, B.; Conrad, M. Role of GPX4 in ferroptosis and its pharmacological implication. Free. Radic. Biol. Med. 2018, 133, 144–152. [Google Scholar] [CrossRef]
  106. Dodson, M.; Castro-Portuguez, R.; Zhang, D.D. NRF2 plays a critical role in mitigating lipid peroxidation and ferroptosis. Redox Biol. 2019, 23, 101107. [Google Scholar] [CrossRef]
  107. Jiang, L.; Kon, N.; Li, T.; Wang, S.-J.; Su, T.; Hibshoosh, H.; Baer, R.; Gu, W. Ferroptosis as a p53-mediated activity during tumour suppression. Nature 2015, 520, 57–62. [Google Scholar] [CrossRef]
  108. Hassannia, B.; Vandenabeele, P.; Berghe, T.V. Targeting Ferroptosis to Iron Out Cancer. Cancer Cell 2019, 35, 830–849. [Google Scholar] [CrossRef]
  109. Hsieh, C.-H.; Hsieh, H.-C.; Shih, F.-H.; Wang, P.-W.; Yang, L.-X.; Shieh, D.-B.; Wang, Y.-C. An innovative NRF2 nano-modulator induces lung cancer ferroptosis and elicits an immunostimulatory tumor microenvironment. Theranostics 2021, 11, 7072–7091. [Google Scholar] [CrossRef]
  110. Ma, Q. Role of Nrf2 in Oxidative Stress and Toxicity. Annu. Rev. Pharmacol. Toxicol. 2013, 53, 401–426. [Google Scholar] [CrossRef] [PubMed]
  111. Sun, X.; Ou, Z.; Chen, R.; Niu, X.; Chen, D.; Kang, R.; Tang, D. Activation of the p62-Keap1-NRF2 pathway protects against ferroptosis in hepatocellular carcinoma cells. Hepatology 2016, 63, 173–184. [Google Scholar] [CrossRef] [PubMed]
  112. Zheng, S.; Mo, J.; Zhang, J.; Chen, Y. HIF-1α inhibits ferroptosis and promotes malignant progression in non-small cell lung cancer by activating the Hippo-YAP signalling pathway. Oncol. Lett. 2023, 25, 1–9. [Google Scholar] [CrossRef] [PubMed]
  113. Jun, J.C.; Rathore, A.; Younas, H.; Gilkes, D.; Polotsky, V.Y. Hypoxia-Inducible Factors and Cancer. Curr. Sleep Med. Rep. 2017, 3, 1–10. [Google Scholar] [CrossRef]
  114. Wu, J.; Minikes, A.M.; Gao, M.; Bian, H.; Li, Y.; Stockwell, B.R.; Chen, Z.-N.; Jiang, X. Intercellular interaction dictates cancer cell ferroptosis via NF2–YAP signalling. Nature 2019, 572, 402–406. [Google Scholar] [CrossRef] [PubMed]
  115. Wang, W.; Green, M.; Choi, J.E.; Gijón, M.; Kennedy, P.D.; Johnson, J.K.; Liao, P.; Lang, X.; Kryczek, I.; Sell, A.; et al. CD8+ T cells regulate tumour ferroptosis during cancer immunotherapy. Nature 2019, 569, 270–274. [Google Scholar] [CrossRef] [PubMed]
  116. Sun, S.; Shen, J.; Jiang, J.; Wang, F.; Min, J. Targeting ferroptosis opens new avenues for the development of novel therapeutics. Signal Transduct. Target. Ther. 2023, 8, 372. [Google Scholar] [CrossRef]
  117. Lei, G.; Mao, C.; Yan, Y.; Zhuang, L.; Gan, B. Ferroptosis, radiotherapy, and combination therapeutic strategies. Protein Cell 2021, 12, 836–857. [Google Scholar] [CrossRef]
  118. Lei, G.; Zhuang, L.; Gan, B. Targeting ferroptosis as a vulnerability in cancer. Nat. Rev. Cancer 2022, 22, 381–396. [Google Scholar] [CrossRef]
  119. Luo, L.; Wang, H.; Tian, W.; Zeng, J.; Huang, Y.; Luo, H. Targeting ferroptosis for cancer therapy: Iron metabolism and anticancer immunity. Am. J. Cancer Res. 2021, 11, 5508–5525. [Google Scholar]
  120. Cosialls, E.; El Hage, R.; Dos Santos, L.; Gong, C.; Mehrpour, M.; Hamaï, A. Ferroptosis: Cancer Stem Cells Rely on Iron until “to Die for” It. Cells 2021, 10, 2981. [Google Scholar] [CrossRef] [PubMed]
  121. Taylor, W.R.; Fedorka, S.R.; Gad, I.; Shah, R.; Alqahtani, H.D.; Koranne, R.; Kuganesan, N.; Dlamini, S.; Rogers, T.; Al-Hamashi, A.; et al. Small-Molecule Ferroptotic Agents with Potential to Selectively Target Cancer Stem Cells. Sci. Rep. 2019, 9, 5926. [Google Scholar] [CrossRef] [PubMed]
  122. Wang, Y.; Yu, L.; Ding, J.; Chen, Y. Iron Metabolism in Cancer. Int. J. Mol. Sci. 2019, 20, 95. [Google Scholar] [CrossRef] [PubMed]
  123. Wang, H.; Zhang, Z.; Ruan, S.; Yan, Q.; Chen, Y.; Cui, J.; Wang, X.; Huang, S.; Hou, B. Regulation of iron metabolism and ferroptosis in cancer stem cells. Front. Oncol. 2023, 13, 1251561. [Google Scholar] [CrossRef] [PubMed]
  124. Gnanapradeepan, K.; Basu, S.; Barnoud, T.; Budina-Kolomets, A.; Kung, C.-P.; Murphy, M.E. The p53 Tumor Suppressor in the Control of Metabolism and Ferroptosis. Front. Endocrinol. 2018, 9, 124. [Google Scholar] [CrossRef]
  125. Liu, Y.; Gu, W. p53 in ferroptosis regulation: The new weapon for the old guardian. Cell Death Differ. 2022, 29, 895–910. [Google Scholar] [CrossRef] [PubMed]
  126. Poltorack, C.D.; Dixon, S.J. Understanding the role of cysteine in ferroptosis: Progress & paradoxes. FEBS J. 2021, 289, 374–385. [Google Scholar] [CrossRef] [PubMed]
  127. Liu, M.-Z.; Kong, N.; Zhang, G.-Y.; Xu, Q.; Xu, Y.; Ke, P.; Liu, C. The critical role of ferritinophagy in human disease. Front. Pharmacol. 2022, 13, 933732. [Google Scholar] [CrossRef]
  128. Li, J.; Jia, Y.-C.; Ding, Y.-X.; Bai, J.; Cao, F.; Li, F. The crosstalk between ferroptosis and mitochondrial dynamic regulatory networks. Int. J. Biol. Sci. 2023, 19, 2756–2771. [Google Scholar] [CrossRef]
  129. Bartolacci, C.; Andreani, C.; El-Gammal, Y.; Scaglioni, P.P. Lipid Metabolism Regulates Oxidative Stress and Ferroptosis in RAS-Driven Cancers: A Perspective on Cancer Progression and Therapy. Front. Mol. Biosci. 2021, 8, 706650. [Google Scholar] [CrossRef]
  130. Hirata, Y.; Cai, R.; Volchuk, A.; Steinberg, B.E.; Saito, Y.; Matsuzawa, A.; Grinstein, S.; Freeman, S.A. Lipid peroxidation increases membrane tension, Piezo1 gating, and cation permeability to execute ferroptosis. Curr. Biol. 2023, 33, 1282–1294. [Google Scholar] [CrossRef]
  131. Yang, J.; Mo, J.; Dai, J.; Ye, C.; Cen, W.; Zheng, X.; Jiang, L.; Ye, L. Cetuximab promotes RSL3-induced ferroptosis by suppressing the Nrf2/HO-1 signalling pathway in KRAS mutant colorectal cancer. Cell Death Dis. 2021, 12, 1079. [Google Scholar] [CrossRef] [PubMed]
  132. Zhang, C.; Wang, C.; Yang, Z.; Bai, Y.; Shukuya, T.; Poh, M.-E.; Ekman, S.; Li, J.; Xu, Y.; Deng, S. Identification of GPX4 as a therapeutic target for lung adenocarcinoma after EGFR-TKI resistance. Transl. Lung Cancer Res. 2022, 11, 786–801. [Google Scholar] [CrossRef] [PubMed]
  133. Stępień, K.; Ostrowski, R.P.; Matyja, E. Hyperbaric oxygen as an adjunctive therapy in treatment of malignancies, including brain tumours. Med. Oncol. 2016, 33, 101. [Google Scholar] [CrossRef] [PubMed]
  134. Xu, Z.; Xie, Y.; Mao, Y.; Huang, J.; Mei, X.; Song, J.; Sun, Y.; Yao, Z.; Shi, W. Ferroptosis-Related Gene Signature Predicts the Prognosis of Skin Cutaneous Melanoma and Response to Immunotherapy. Front. Genet. 2021, 12, 758981. [Google Scholar] [CrossRef] [PubMed]
  135. Qi, X.; Wang, R.; Lin, Y.; Yan, D.; Zuo, J.; Chen, J.; Shen, B. A Ferroptosis-Related Gene Signature Identified as a Novel Prognostic Biomarker for Colon Cancer. Front. Genet. 2021, 12, 692426. [Google Scholar] [CrossRef] [PubMed]
  136. Jin, J.; Liu, C.; Yu, S.; Cai, L.; Sitrakiniaina, A.; Gu, R.; Li, W.; Wu, F.; Xue, X. A novel ferroptosis-related gene signature for prognostic prediction of patients with lung adenocarcinoma. Aging 2021, 13, 16144–16164. [Google Scholar] [CrossRef] [PubMed]
  137. Qian, Y.; Daza, J.; Itzel, T.; Betge, J.; Zhan, T.; Marmé, F.; Teufel, A. Prognostic Cancer Gene Expression Signatures: Current Status and Challenges. Cells 2021, 10, 648. [Google Scholar] [CrossRef] [PubMed]
  138. Shimizu, S.; Hiratsuka, H.; Koike, K.; Tsuchihashi, K.; Sonoda, T.; Ogi, K.; Miyakawa, A.; Kobayashi, J.; Kaneko, T.; Igarashi, T.; et al. Tumor-infiltrating CD8+ T-cell density is an independent prognostic marker for oral squamous cell carcinoma. Cancer Med. 2019, 8, 80–93. [Google Scholar] [CrossRef]
  139. Foy, J.-P.; Bertolus, C.; Michallet, M.-C.; Deneuve, S.; Incitti, R.; Bendriss-Vermare, N.; Albaret, M.-A.; Ortiz-Cuaran, S.; Thomas, E.; Colombe, A.; et al. The immune microenvironment of HPV-negative oral squamous cell carcinoma from never-smokers and never-drinkers patients suggests higher clinical benefit of IDO1 and PD1/PD-L1 blockade. Ann. Oncol. 2017, 28, 1934–1941. [Google Scholar] [CrossRef]
  140. Wang, H.; Mao, L.; Zhang, T.; Zhang, L.; Wu, Y.; Guo, W.; Hu, J.; Ju, H.; Ren, G. Altered expression of TIM-3, LAG-3, IDO, PD-L1, and CTLA-4 during nimotuzumab therapy correlates with responses and prognosis of oral squamous cell carcinoma patients. J. Oral Pathol. Med. 2019, 48, 669–676. [Google Scholar] [CrossRef]
  141. Yu, P.; Zhang, X.; Liu, N.; Tang, L.; Peng, C.; Chen, X. Pyroptosis: Mechanisms and diseases. Signal Transduct. Target. Ther. 2021, 6, 128. [Google Scholar] [CrossRef] [PubMed]
  142. Fang, Y.; Tian, S.; Pan, Y.; Li, W.; Wang, Q.; Tang, Y.; Yu, T.; Wu, X.; Shi, Y.; Ma, P.; et al. Pyroptosis: A new frontier in cancer. Biomed. Pharmacother. 2020, 121, 109595. [Google Scholar] [CrossRef] [PubMed]
  143. Kelley, N.; Jeltema, D.; Duan, Y.; He, Y. The NLRP3 Inflammasome: An Overview of Mechanisms of Activation and Regulation. Int. J. Mol. Sci. 2019, 20, 3328. [Google Scholar] [CrossRef]
  144. Jiang, M.; Qi, L.; Li, L.; Li, Y. The caspase-3/GSDME signal pathway as a switch between apoptosis and pyroptosis in cancer. Cell Death Discov. 2020, 6, 112. [Google Scholar] [CrossRef]
  145. Bhat, A.A.; Thapa, R.; Afzal, O.; Agrawal, N.; Almalki, W.H.; Kazmi, I.; Alzarea, S.I.; Altamimi, A.S.A.; Prasher, P.; Singh, S.K.; et al. The pyroptotic role of Caspase-3/GSDME signalling pathway among various cancer: A Review. Int. J. Biol. Macromol. 2023, 242, 124832. [Google Scholar] [CrossRef]
  146. Zhang, Z.; Li, X.; Wang, Y.; Wei, Y.; Wei, X. Involvement of inflammasomes in tumor microenvironment and tumor therapies. J. Hematol. Oncol. 2023, 16, 24. [Google Scholar] [CrossRef]
  147. Vyas, D.; Laput, G.; Vyas, A. Chemotherapy-enhanced inflammation may lead to the failure of therapy and metastasis. OncoTargets Ther. 2014, 7, 1015–1023. [Google Scholar] [CrossRef] [PubMed]
  148. Moossavi, M.; Parsamanesh, N.; Bahrami, A.; Atkin, S.L.; Sahebkar, A. Role of the NLRP3 inflammasome in cancer. Mol. Cancer 2018, 17, 158. [Google Scholar] [CrossRef]
  149. Sharma, B.R.; Kanneganti, T.-D. NLRP3 inflammasome in cancer and metabolic diseases. Nat. Immunol. 2021, 22, 550–559. [Google Scholar] [CrossRef]
  150. Liu, X.; Yin, L.; Shen, S.; Hou, Y. Inflammation and cancer: Paradoxical roles in tumorigenesis and implications in immunotherapies. Genes Dis. 2023, 10, 151–164. [Google Scholar] [CrossRef]
  151. Lu, L.; Zhang, Y.; Tan, X.; Merkher, Y.; Leonov, S.; Zhu, L.; Deng, Y.; Zhang, H.; Zhu, D.; Tan, Y.; et al. Emerging mechanisms of pyroptosis and its therapeutic strategy in cancer. Cell Death Discov. 2022, 8, 338. [Google Scholar] [CrossRef]
  152. Lu, X.; Guo, T.; Zhang, X. Pyroptosis in Cancer: Friend or Foe? Cancers 2021, 13, 3620. [Google Scholar] [CrossRef]
  153. Feng, X.; Luo, Q.; Wang, H.; Zhang, H.; Chen, F. MicroRNA-22 suppresses cell proliferation, migration and invasion in oral squamous cell carcinoma by targeting NLRP3. J. Cell. Physiol. 2018, 233, 6705–6713. [Google Scholar] [CrossRef]
  154. Wei, X.; Xie, F.; Zhou, X.; Wu, Y.; Yan, H.; Liu, T.; Huang, J.; Wang, F.; Zhou, F.; Zhang, L. Role of pyroptosis in inflammation and cancer. Cell. Mol. Immunol. 2022, 19, 971–992. [Google Scholar] [CrossRef]
  155. Dhuriya, Y.K.; Sharma, D. Necroptosis: A regulated inflammatory mode of cell death. J. Neuroinflammation 2018, 15, 199. [Google Scholar] [CrossRef] [PubMed]
  156. Nicolè, L.; Sanavia, T.; Cappellesso, R.; Maffeis, V.; Akiba, J.; Kawahara, A.; Naito, Y.; Radu, C.M.; Simioni, P.; Serafin, D.; et al. Necroptosis-driving genes RIPK1, RIPK3 and MLKL-p are associated with intratumoral CD3+ and CD8+ T cell density and predict prognosis in hepatocellular carcinoma. J. Immunother. Cancer 2022, 10, e004031. [Google Scholar] [CrossRef] [PubMed]
  157. Grootjans, S.; Vanden Berghe, T.; Vandenabeele, P. Initiation and execution mechanisms of necroptosis: An overview. Cell Death Differ. 2017, 24, 1184–1195. [Google Scholar] [CrossRef] [PubMed]
  158. Pinci, F.; Gaidt, M.M.; Jung, C.; Nagl, D.; Kuut, G.; Hornung, V. Tumor necrosis factor is a necroptosis-associated alarmin. Front. Immunol. 2022, 13, 1074440. [Google Scholar] [CrossRef] [PubMed]
  159. Seo, J.; Nam, Y.W.; Kim, S.; Oh, D.-B.; Song, J. Necroptosis molecular mechanisms: Recent findings regarding novel necroptosis regulators. Exp. Mol. Med. 2021, 53, 1007–1017. [Google Scholar] [CrossRef]
  160. Deragon, M.A.; McCaig, W.D.; Truong, P.V.; Metz, K.R.; Carron, K.A.; Hughes, K.J.; Knapp, A.R.; Dougherty, M.J.; LaRocca, T.J. Mitochondrial Trafficking of MLKL, Bak/Bax, and Drp1 Is Mediated by RIP1 and ROS which Leads to Decreased Mitochondrial Membrane Integrity during the Hyperglycemic Shift to Necroptosis. Int. J. Mol. Sci. 2023, 24, 8609. [Google Scholar] [CrossRef]
  161. Juan, C.A.; de la Lastra, J.M.P.; Plou, F.J.; Pérez-Lebeña, E. The Chemistry of Reactive Oxygen Species (ROS) Revisited: Outlining Their Role in Biological Macromolecules (DNA, Lipids and Proteins) and Induced Pathologies. Int. J. Mol. Sci. 2021, 22, 4642. [Google Scholar] [CrossRef]
  162. Choi, M.E.; Price, D.R.; Ryter, S.W.; Choi, A.M.K. Necroptosis: A crucial pathogenic mediator of human disease. J. Clin. Investig. 2019, 4, 128834. [Google Scholar] [CrossRef]
  163. Galluzzi, L.; Kepp, O.; Chan, F.K.-M.; Kroemer, G. Necroptosis: Mechanisms and Relevance to Disease. Annu. Rev. Pathol. Mech. Dis. 2017, 12, 103–130. [Google Scholar] [CrossRef]
  164. Pistritto, G.; Trisciuoglio, D.; Ceci, C.; Garufi, A.; D’Orazi, G. Apoptosis as anticancer mechanism: Function and dysfunction of its modulators and targeted therapeutic strategies. Aging 2016, 8, 603–619. [Google Scholar] [CrossRef]
  165. Gong, Y.; Fan, Z.; Luo, G.; Yang, C.; Huang, Q.; Fan, K.; Cheng, H.; Jin, K.; Ni, Q.; Yu, X.; et al. The role of necroptosis in cancer biology and therapy. Mol. Cancer 2019, 18, 100. [Google Scholar] [CrossRef] [PubMed]
  166. Sprooten, J.; De Wijngaert, P.; Vanmeerbeek, I.; Martin, S.; Vangheluwe, P.; Schlenner, S.; Krysko, D.V.; Parys, J.B.; Bultynck, G.; Vandenabeele, P.; et al. Necroptosis in Immuno-Oncology and Cancer Immunotherapy. Cells 2020, 9, 1823. [Google Scholar] [CrossRef] [PubMed]
  167. Yan, J.; Wan, P.; Choksi, S.; Liu, Z.-G. Necroptosis and tumor progression. Trends Cancer 2022, 8, 21–27. [Google Scholar] [CrossRef]
  168. Khamseh, M.E.; Sheikhi, A.; Shahsavari, Z.; Ghorbani, M.; Akbari, H.; Imani, M.; Panahi, M.; Alimohammadi, A.; Ameri, M.; Nazem, S.; et al. Evaluation of the expression of necroptosis pathway mediators and its association with tumor characteristics in functional and non-functional pituitary adenomas. BMC Endocr. Disord. 2022, 22, 1. [Google Scholar] [CrossRef] [PubMed]
  169. Zhang, T.; Wang, Y.; Inuzuka, H.; Wei, W. Necroptosis pathways in tumorigenesis. Semin. Cancer Biol. 2022, 86, 32–40. [Google Scholar] [CrossRef] [PubMed]
  170. Wu, Y.; Dong, G.; Sheng, C. Targeting necroptosis in anticancer therapy: Mechanisms and modulators. Acta Pharm. Sin. B 2020, 10, 1601–1618. [Google Scholar] [CrossRef] [PubMed]
  171. Murphy, J.M.; Rodriguez, Y.A.R.; Jeong, K.; Ahn, E.-Y.E.; Lim, S.-T.S. Targeting focal adhesion kinase in cancer cells and the tumor microenvironment. Exp. Mol. Med. 2020, 52, 877–886. [Google Scholar] [CrossRef]
  172. Su, Z.; Yang, Z.; Xie, L.; DeWitt, J.P.; Chen, Y. Cancer therapy in the necroptosis era. Cell Death Differ. 2016, 23, 748–756. [Google Scholar] [CrossRef]
  173. Basit, F.; Cristofanon, S.; Fulda, S. Obatoclax (GX15-070) triggers necroptosis by promoting the assembly of the necrosome on autophagosomal membranes. Cell Death Differ. 2013, 20, 1161–1173. [Google Scholar] [CrossRef]
  174. Yuan, T.-M.; Liang, R.-Y.; Chueh, P.J.; Chuang, S.-M. Role of ribophorin II in the response to anticancer drugs in gastric cancer cell lines. Oncol. Lett. 2015, 9, 1861–1868. [Google Scholar] [CrossRef] [PubMed]
  175. Bhosale, P.G.; Kennedy, R.A.; Watt, F.M. Caspase activation in tumour-infiltrating lymphocytes is associated with lymph node metastasis in oral squamous cell carcinoma. J. Pathol. 2023, 261, 43–54. [Google Scholar] [CrossRef] [PubMed]
  176. Vorobjeva, N.V.; Chernyak, B.V. NETosis: Molecular Mechanisms, Role in Physiology and Pathology. Biochemistry 2020, 85, 1178–1190. [Google Scholar] [CrossRef] [PubMed]
  177. Ronchetti, L.; Boubaker, N.S.; Barba, M.; Vici, P.; Gurtner, A.; Piaggio, G. Neutrophil extracellular traps in cancer: Not only catching microbes. J. Exp. Clin. Cancer Res. 2021, 40, 231. [Google Scholar] [CrossRef] [PubMed]
  178. Jaboury, S.; Wang, K.; O’sullivan, K.M.; Ooi, J.D.; Ho, G.Y. NETosis as an oncologic therapeutic target: A mini review. Front. Immunol. 2023, 14, 1170603. [Google Scholar] [CrossRef] [PubMed]
  179. Garley, M. Unobvious Neutrophil Extracellular Traps Signification in the Course of Oral Squamous Cell Carcinoma: Current Understanding and Future Perspectives. Cancer Control. 2023, 30, 1–8. [Google Scholar] [CrossRef] [PubMed]
  180. Huang, P.; Chen, G.; Jin, W.; Mao, K.; Wan, H.; He, Y. Molecular Mechanisms of Parthanatos and Its Role in Diverse Diseases. Int. J. Mol. Sci. 2022, 23, 7292. [Google Scholar] [CrossRef] [PubMed]
  181. Galia, A.; Calogero, A.; Condorelli, R.; Fraggetta, F.; La Corte, A.; Ridolfo, F.; Bosco, P.; Castiglione, R.; Salemi, M. PARP-1 protein expression in glioblastoma multiforme. Eur. J. Histochem. 2012, 56, 9. [Google Scholar] [CrossRef] [PubMed]
  182. Jangamreddy, J.R.; Los, M.J. Mitoptosis, a Novel Mitochondrial Death Mechanism Leading Predominantly to Activation of Autophagy. Hepat. Mon. 2012, 12, e6159. [Google Scholar] [CrossRef] [PubMed]
  183. Pimenova, E.A.; Reunova, Y.A.; Menchinskaya, E.S.; Reunov, A.A.; Aminin, D.L. An Unusual Pathway of Mitoptosis Found in Ehrlich Carcinoma Cells. Dokl. Biol. Sci. 2020, 494, 240–243. [Google Scholar] [CrossRef] [PubMed]
  184. Hanson, S.; Dharan, A.; PV, J.; Pal, S.; Nair, B.G.; Kar, R.; Mishra, N. Paraptosis: A unique cell death mode for targeting cancer. Front. Pharmacol. 2023, 14, 1159409. [Google Scholar] [CrossRef]
Figure 1. Flow diagram of literature search and selection criteria adapted from Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA).
Figure 1. Flow diagram of literature search and selection criteria adapted from Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA).
Biology 13 00103 g001
Figure 2. Ferroptosis manifests through two primary pathways. In the first pathway, transferrin (TRF1), a crucial player in iron homeostasis, binds to Fe3+, forming a complex, which then binds to the transferrin receptor. Endocytosis of this complex occurs, leading to the formation of the endosome. Within the endosome, there will be acidification of the environment, causing the dissociation of the complex and the reduction of Fe3+ to Fe2+. This Fe2+ is released through the endosomal membrane. However, surplus free iron ions form a labile iron pool (LIP), which partakes in Fenton’s reactions, causing the generation of the reactive oxygen species (ROS) free radical hydroxyl. This cascade of events culminates in the initiation of lipoperoxidation, a critical step in the ferroptotic process. In the second pathway, System Xc-, a cystine/glutamate antiporter, imports extracellular cystine. Intracellularly, cystine undergoes conversion into glutathione (GSH), a vital antioxidant. The availability of GSH is integral to the function of glutathione peroxidase 4 (GPX4), which tackles intracellular lipid peroxides, preventing ferroptosis. However, the inhibition of GPX4 leads to a disruption in this protective mechanism, resulting in an augmented presence of ROS and ultimately contributing to the progression of ferroptosis. [Image was created using Biorender.com (accessed on 16 November 2023)].
Figure 2. Ferroptosis manifests through two primary pathways. In the first pathway, transferrin (TRF1), a crucial player in iron homeostasis, binds to Fe3+, forming a complex, which then binds to the transferrin receptor. Endocytosis of this complex occurs, leading to the formation of the endosome. Within the endosome, there will be acidification of the environment, causing the dissociation of the complex and the reduction of Fe3+ to Fe2+. This Fe2+ is released through the endosomal membrane. However, surplus free iron ions form a labile iron pool (LIP), which partakes in Fenton’s reactions, causing the generation of the reactive oxygen species (ROS) free radical hydroxyl. This cascade of events culminates in the initiation of lipoperoxidation, a critical step in the ferroptotic process. In the second pathway, System Xc-, a cystine/glutamate antiporter, imports extracellular cystine. Intracellularly, cystine undergoes conversion into glutathione (GSH), a vital antioxidant. The availability of GSH is integral to the function of glutathione peroxidase 4 (GPX4), which tackles intracellular lipid peroxides, preventing ferroptosis. However, the inhibition of GPX4 leads to a disruption in this protective mechanism, resulting in an augmented presence of ROS and ultimately contributing to the progression of ferroptosis. [Image was created using Biorender.com (accessed on 16 November 2023)].
Biology 13 00103 g002
Figure 3. Pathways related to activation of pyroptosis. In the canonical signaling pathway, intracellular sensors Nod-like receptor family, pyrin domain containing 1 (NLRP1), 3 (NLRP3), 4 (NLRC4), absent in melanoma 2 (AIM2) and other inflammasome sensors are responsible for detecting microbial signals. Upon detection, they initiate a response by recruiting the adaptor protein ASC (apoptosis-associated speck-like protein containing a C-terminal caspase recruitment domain), which subsequently recruits pro-caspase-1. Once activated, caspase-1 cleaves Gasdermin D (GSDMD), generating GSDMD-NT fragments. These GSDMD-NT fragments create pores in the plasma membrane that are associated with phosphoinositides. Simultaneously, caspase-1 itself undergoes cleavage, giving rise to caspase-1 P10/P20 and P33/P10 tetramers. These tetramers play a crucial role in the maturation of pro-interleukin-18 (IL-18) and pro-interleukin-1β (IL-1β) into their active forms, IL-18 and IL-1β. These mature cytokines are subsequently released into the extracellular matrix, leading to the initiation of inflammatory responses. In the noncanonical pathway, the presence of lipopolysaccharides (LPS) from Gram-negative bacteria triggers the activation of caspase-4 and caspase-5 (in humans) or caspase-11 (in mice). These caspases, in turn, cleave GSDMD, forming pores in the plasma membrane. These GSDMD pores permit the release of potassium ions, which further activate the NLRP3 inflammasome and contribute to the maturation of IL-1β and IL-18. Additionally, GSDMD pores release mature cytokines, ultimately leading to pyroptosis. [Image was created using Biorender.com (accessed on 13 December 2023)].
Figure 3. Pathways related to activation of pyroptosis. In the canonical signaling pathway, intracellular sensors Nod-like receptor family, pyrin domain containing 1 (NLRP1), 3 (NLRP3), 4 (NLRC4), absent in melanoma 2 (AIM2) and other inflammasome sensors are responsible for detecting microbial signals. Upon detection, they initiate a response by recruiting the adaptor protein ASC (apoptosis-associated speck-like protein containing a C-terminal caspase recruitment domain), which subsequently recruits pro-caspase-1. Once activated, caspase-1 cleaves Gasdermin D (GSDMD), generating GSDMD-NT fragments. These GSDMD-NT fragments create pores in the plasma membrane that are associated with phosphoinositides. Simultaneously, caspase-1 itself undergoes cleavage, giving rise to caspase-1 P10/P20 and P33/P10 tetramers. These tetramers play a crucial role in the maturation of pro-interleukin-18 (IL-18) and pro-interleukin-1β (IL-1β) into their active forms, IL-18 and IL-1β. These mature cytokines are subsequently released into the extracellular matrix, leading to the initiation of inflammatory responses. In the noncanonical pathway, the presence of lipopolysaccharides (LPS) from Gram-negative bacteria triggers the activation of caspase-4 and caspase-5 (in humans) or caspase-11 (in mice). These caspases, in turn, cleave GSDMD, forming pores in the plasma membrane. These GSDMD pores permit the release of potassium ions, which further activate the NLRP3 inflammasome and contribute to the maturation of IL-1β and IL-18. Additionally, GSDMD pores release mature cytokines, ultimately leading to pyroptosis. [Image was created using Biorender.com (accessed on 13 December 2023)].
Biology 13 00103 g003
Figure 4. Necroptosis signaling pathway. The binding of tumor necrosis factor (TNF) induces the formation of the membrane-associated complex I, composed of TNF receptor-associated death domain (TRADD), TNF receptor-associated factor 2 (TRAF2), cellular inhibitor of apoptosis protein 1 and 2 (cIAP1/2), receptor-interacting serine/threonine kinase 1 (RIPK1), and LUBAC, an E3 ubiquitin ligase. cIAP1/2 and LUBAC induce the polyubiquitination of RIPK1, recruiting the IκB kinase (IkappaB kinase) complex (IKKa, IKKB, and NEMO) and TGF-β-activated kinase 1 (TAK1) complex (TAK1, TAB1, and TAB2). These two complexes can eventually lead to the activation of the NF-κB pathway and cell survival. A20CYLD promotes the deubiquitination of RIPK1, inducing the dissociation of TRADD and RIPK1 from TNFR1 and leading to the formation of complex IIa or complex IIb. Complex IIa, consisting of TRADD and Fas-associated protein with death domain (FADD), activates caspase-8 and induces apoptosis through cleavage. Inhibition of RIPK1 ubiquitination results in the induction of complex IIb, composed of RIPK1, FADD and caspase-8. When caspase-8 is inhibited (in either complex IIa or IIb), RIPK1 and RIPK3 form necrosome complexes, activating MLKL through a phosphorylation cascade. Phosphorylated MLKL undergoes oligomerization and migrates to the membrane, which induces necroptosis by membrane rupture or regulating ion flow. [Image was created using Biorender.com (accessed on 16 November 2023)].
Figure 4. Necroptosis signaling pathway. The binding of tumor necrosis factor (TNF) induces the formation of the membrane-associated complex I, composed of TNF receptor-associated death domain (TRADD), TNF receptor-associated factor 2 (TRAF2), cellular inhibitor of apoptosis protein 1 and 2 (cIAP1/2), receptor-interacting serine/threonine kinase 1 (RIPK1), and LUBAC, an E3 ubiquitin ligase. cIAP1/2 and LUBAC induce the polyubiquitination of RIPK1, recruiting the IκB kinase (IkappaB kinase) complex (IKKa, IKKB, and NEMO) and TGF-β-activated kinase 1 (TAK1) complex (TAK1, TAB1, and TAB2). These two complexes can eventually lead to the activation of the NF-κB pathway and cell survival. A20CYLD promotes the deubiquitination of RIPK1, inducing the dissociation of TRADD and RIPK1 from TNFR1 and leading to the formation of complex IIa or complex IIb. Complex IIa, consisting of TRADD and Fas-associated protein with death domain (FADD), activates caspase-8 and induces apoptosis through cleavage. Inhibition of RIPK1 ubiquitination results in the induction of complex IIb, composed of RIPK1, FADD and caspase-8. When caspase-8 is inhibited (in either complex IIa or IIb), RIPK1 and RIPK3 form necrosome complexes, activating MLKL through a phosphorylation cascade. Phosphorylated MLKL undergoes oligomerization and migrates to the membrane, which induces necroptosis by membrane rupture or regulating ion flow. [Image was created using Biorender.com (accessed on 16 November 2023)].
Biology 13 00103 g004
Table 1. Main characteristics of the articles included in the systematic review.
Table 1. Main characteristics of the articles included in the systematic review.
AuthorCountryYearCell DeathType of StudyCell LinesIn Silico Model *In Vivo ModelMain Finding
Ruggieri et al. [92]Italy2015MitoptosisIn vitroHSC-2, HSC-3, PE15NANADichloroacetate (DCA) promoted mitochondria fragmentation and enhanced production of reactive oxygen species
Sulkshane and Teni [84]India2016NecroptosisIn vitro and in vivoAW8507, AW13516, SCC029BNAXenograft murine modelObatoclax induced members of the BCL-2 (B-cell lymphoma 2) family, specifically antagonizing the myeloid cell leukemia sequence 1 (MCL-1) protein, which induces necroptosis through extensive stress and mitochondrial dysfunction
Feng et al. [64]China2017PyroptosisIn vitro and in vivoCAL-27NAXenograft murine modelActivation of Nod-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome was slightly increased in OSCC tissues from patients who received 5-fluorouracil (5-FU), and 5-FU increased the expression and activation of NLRP3 xenograft model
Garley et al. [93]Poland2018NETosisIn vitroPrimary culture of neutrophils from OSCC patientsNANACells stimulated with lipopolysaccharides (LPS) and interleukin 17 (IL-17) produced more neutrophil extracellular traps (NETs) compared to unstimulated cells
Okazaki et al. [18]Japan2018FerroptosisIn vitro and in vivoOSC19, HSC-2, HSC-3, HSC-4NAXenograft murine modelxCT (also called SLC7A11, solute carrier family 7 member 11) regulated lipid peroxidation, reactive oxygen species (ROS) and ferroptosis, while aldehyde dehydrogenase 3 family member A1 (ALDH3A1) mediated the detoxification of 4-hydroxynonenal (4-HNE) derived from lipid peroxides
Huang et al. [19]Taiwan2019FerroptosisIn vitro and in vivoOEC-M1, SCC-9, HSC-3, SAS, OC-3NAXenograft murine modelZero-valent iron nanoparticles promoted ferroptosis
Sato et al. [20]Japan/Australia2019FerroptosisIn vitroSAS, HSC-3, HSC-4, Ca9-22, Sa3, HSC-2, Ho-1-u-1NANANon-thermal plasma induced ferroptosis of cancer cells
Yue et al. [65]China2019PyroptosisIn vitroSCC-15NANAAnthocyanin induced NLRP3 inflammasome, Gasdermin-D (GSDMD), caspase-1 and interleukin-1β (IL-1β)
Zhu et al. [21]China2019FerroptosisIn vitroCAL-27NANAThe photosensitizer chlorin e6 inhibited SLC7A11 and promoted ROS accumulation, increasing ferroptosis
Garley et al. [94]Poland2020NETosisin vitroCAL-27NANAThe enhanced process of NET formation was accompanied by changes in the expression of proteins from the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) pathway
Hémon et al. [22]France2020FerroptosisIn vitroPECAPJ41NANADefect in cystine transport sensibilized cells to ferroptosis
Huang et al. [66]China2020PyroptosisIn vitro and in vivoCAL-27, SCC-9NAXenograft murine model and human tumorsVitamin D inhibited caspase-3-mediated Gasdermin-E (GSDME) cleavage, reducing pyroptosis
Lee et al. [23]Republic of Korea2020FerroptosisIn vitro and in vivoHN-4NAXenograft murine modelSulfasalazine, cysteine deprivation and glutaredoxin 5 (GLRX5) silencing resulted in ferroptosis
Li et al. [85]China2020NecroptosisIn vitro and in vivoSCC-9, SCC-25, CAL-27, SCC-15, HSC-3, HSC-4, CALL-33, HSC6, SCC1NAHuman tumorsOSCC cells displayed a high tendency to necroptosis, and necroptosis was an independent risk factor for poor overall survival and progression-free survival
Lin et al. [24]Taiwan2020FerroptosisIn vitroSASNANAThe natural compound Chrysophanol promoted ferroptosis by decreasing glutathione peroxidase 4 (GPX4) and increasing lipocalin-2 (LCN2)
Uzunparmak et al. [86]United States2020NecroptosisIn vitro and in vivoMOC-1, TR146, UMSCC-1NAXenograft murine modelCaspase-8 knockdown enhanced the radiosensitizing effects of birinapant and Z-VAD-FMK through the induction of necroptosis
Chen et al. [95]China2021Paraptosisin vitroHSC-3, HSC-4NANAIsorhamnetin induced paraptosis cell death, which was mediated by ROS and extracellular regulated MAP kinase (ERK) signal pathway
Gu et al. [25]Japan/China2021FerroptosisIn silicoNAmRNA expression data from TCGA and GEONADeveloped a ferroptosis score, which was associated with prognosis and treatment of OSCC patients
Huang et al. [66]Taiwan2021NecroptosisIn vitroHSC-3, SASNANACoadministration of capsaicin with conventional anticancer agents increased levels of the necroptosis markers phospho-MLKL (mixed lineage kinase domain like pseudokinase) and phospho-RIPK3 (receptor interacting protein kinase 3)
Jiang et al. [67]China2021PyroptosisIn vitroHSC-4, CAL-27NANAKnockdown of the LINC00958 reduced GSDMD, inflammasome-mediated proteins and pyroptosis
Li et al. [26]China2021aFerroptosisIn silicoNAmRNA expression data from TCGANAEstablished a 10-ferroptosis-related gene signature and nomogram that were associated with OSCC prognosis
Li et al. [96]China2021bParthanatosin vitro and in vivoCAL-27, SCC-25NAXenograft murine modelOxaliplatin inhibited poly(ADP-ribose) polymerase 1 (PARP1)-mediated parthanatos through increasing ROS production
Luo et al. [68]China2021PyroptosisIn vitro and in vivoWSU-HN4, CAL-27, SCC-9NAXenograft murine modelGSDME was cleaved by activated caspase-3 to generate the GSDME-N fragment, which promoted pyroptosis. Although Erianine increased GSDME-N fragment, it was unable to induce pyroptosis
Tomita et al. [27]Japan2021FerroptosisIn vitroSASNANAmiR-7-5p control radioresistance via ROS generation that leads to ferroptosis.
Wang et al. [28]China2021Ferroptosis and PyroptosisIn vitro and in vivoCAL-27, TCA-8113NAXenograft murine modelQuisinostat, a histone inhibitor, induced both pyroptosis and ferroptosis
Yao et al. [70]China2021PyroptosisIn vitro and in vivoHSC-3, SCC-7NAXenograft murine modelThe periodontitis-related bacteria (P. gingivalis and F. nucleatum) overexpressed NLRP3 inflammasome, activating upstream signal molecules of ataxia telangiectasia and Rad3-related (ATR) checkpoint kinase 1 (CHK1) pathway and inhibiting CHK1
Yang et al. [29]China2021aFerroptosisIn vitro and in vivoCAL-27, SCC-15NAXenograft murine modelSilencing of circFNDC3B/miR-520d-5p/SLC7A11 axis inhibited GPX4 and SLC7A11 and increased ROS and Fe2+, attenuating ferroptosis
Yang et al. [69]China2021bPyroptosisIn vitro and in vivoCal-27, WSU-HN-6NAXenograft murine modelBitter melon-derived extracellular vesicles decreased NLRP3 inflammasome
You et al. [30]Republic of Korea2021aFerroptosisIn vitro and in vivoAMC-HN4NAXenograft murine modelLysine demethylase 5A (KDM5A)/mitochondrial pyruvate carrier 1 (MPC-1) axis promoted cancer ferroptosis
You et al. [31]Republic of Korea2021bFerroptosisIn vitroHN-4NANASilencing of progesterone receptor membrane component 1 (PGRMC1) caused ferroptosis by xCT inhibitors
Zhang et al. [32]China2021FerroptosisIn vitro and in vivoHN-6, CAL-27NAXenograft murine modelThe pH-sensitive Zif-8 particles led to ferroptosis
Bhuyan et al. [33]India/United States2022Ferroptosis and PyroptosisIn vitroSCC-25 and Cancer stem cells derived from SCC-25NANAFerrostatin-1, a ferroptosis inhibitor, prevented the effect of conditioned media from Bacillus Calmette Guerin (BCG) and induced loss of cell viability; infected cancer stem cells exhibited significant upregulation of caspase-3, caspase-1 and Gasdermin D (GSDMD), well-known markers of pyroptosis
Han et al. [34]China2022FerroptosisIn vitroCAL-27, SCC-9NANAThe carnosic acid sensitized cisplatin-resistant cells to cisplatin by evoking ferroptosis, which involves the inactivation of nuclear erythroid factor 2-related factor 2 (Nrf2)/heme oxygenase 1 (HO-1)/xCT pathway
Kaokaen et al. [88]Thailand2022NecroptosisIn vitroHSC-4NANANanoencapsulation of cordycepin induced a switch from necroptosis to apoptosis
Li et al. [35]China2022aFerroptosisIn silico and in vitroHN-6, CAL-27mRNA expression data from TCGA and GEONAArachidonate 12-lipoxygenase, 12R type (ALOX12B) and small proline rich protein 1A (SPRR1A) were associated with overall survival and correlated with the number of cancer-associated fibroblasts
Li et al. [36]China2022bFerroptosisIn silicoNAmRNA expression data from TCGANAThe combination of ferroptosis-related non-coding RNA was associated with prognosis
Liu et al. [37]China2022aFerroptosisIn silico and in vivoNAmRNA expression data from GEO single cellsHuman tumorsThe acyl-CoA synthetase long chain family member 1 (ACSL1), solute carrier family 39 member 14 (SLC39A14), transferrin receptor (TFRC) and prion protein (PRNP) expressions were closely associated with ferroptosis-related development and tumor progression
Liu et al. [38]China2022bFerroptosisIn vitro and in vivoSCC-15NAHuman tumorsCo-exposure of hyperbaric oxygen and X-ray radiation promoted ferroptosis by regulating GPX4
Liu et al. [39]China2022cFerroptosisIn vitroCAL-33NANAErastin and ferroptosis-inducer agent RSL3 promoted ferroptosis independently of GPX4 or HO-1, and RSL3, together with epidermal growth factor receptor inhibitor Gefitinib or monoclonal antibody Cetuximab, significantly reduced the viability of the tumor cells
Lu et al. [40]China2022FerroptosisIn vitroHN-6, CAL-27NANAOverexpression of caveolin 1 (CAV1) inhibited ferroptosis
Qiu et al. [41]China2022FerroptosisIn silicoNAmRNA expression data from TCGANAEight ferroptosis-related lncRNAs (FIRRE, LINC01305, AC099850.3, AL512274.1, AC090246.1, MIAT, AC079921.2 and LINC00524) were associated with prognosis
Rioja-Blanco et al. [71]Spain2022PyroptosisIn vitro and in vivoUM-SCC-74BNAXenograft murine modelNanotoxins T22-PE24-H6 and T22-DITOX-H6 triggered caspase-3/GSDME-mediated pyroptosis
Shen et al. [72]China2022PyroptosisIn vitro and in vivoHSC-3, SCC-7NAXenograft murine modelThe natural compounds N. sicca and C. matruchotii promoted pyroptosis
Sun et al. [42]China2022FerroptosisIn vitroSCC-25NANAmiR-34c-3p negatively regulated SLC7A11 expression, promoting ferroptosis
Wang et al. [43]China2022aFerroptosisIn vitro and in vivoHN-6NAXenograft murine modelZIF-8@ssPDA as a drug carrier for the chlorin e6 photosensitizer induced ferroptosis
Wang et al. [73]China2022bPyroptosisIn vitro and in vivoSCC-7NAHuman tumorsGSDME expression in OSCC was related to better prognosis, and knockdown of GSDME attenuated the antitumor effect induced by cisplatin
Xin et al. [74]China2022PyroptosisIn silico and in vivoNAmRNA expression data from TCGAHuman tumorsThe expression levels of pyroptosis-related lncRNA genes ZJPX, ZFAS1, TNFRSF10A-AS1, LINC00847, AC099850.3 and IER3-AS1 were associated with the prognosis of OSCC patients
Xu et al. [44]China2022aFerroptosisIn vitro and in vivoCAL-27NAXenograft murine modelFerroptosis was pointed out as a top activated pathway after eukaryotic translation initiation factor 3 subunit B (EIF3B) knockdown
Xu et al. [45]China2022bFerroptosisIn silicoNAmRNA expression data from TCGA and GEONAEstablished a ferroptosis-related 16-DNA methylation signature with potential to predict prognosis outcome
Yang et al. [46]China2022FerroptosisIn vitro and in vivoTSCCA, SCC-15, CAL-27NAXenograft murine modelPeriod 1 promoted ferroptosis in a hypoxia inducible factor 1 subunit alpha (HIF-1α)-dependent manner
Yin et al. [47]China2022FerroptosisIn silicoNAmRNA expression data from TCGANAA gene expression signature containing 4 ferroptosis-related genes was associated with prognosis and response to immunotherapy of OSCC patients
Zeng et al. [75]China2022PyroptosisIn silicoNAmRNA expression data from TCGANAEstablished a pyroptosis-related prognostic signature associated with OSCC prognosis
Zhou et al. [48]China2022aFerroptosisIn vitro and in vivoCAL-27, SCC15NAXenograft murine modelSilencing of the adipocyte enhancer-binding protein 1 (AEBP1) predisposed cisplatin-resistant oral cancer cells to ferroptosis via the JNK/p38/ERK pathway
Zhou et al. [76]China2022bPyroptosisIn vivoNANAPatient-derived xenograft modelTiO2@Ru@siRNA induced pyroptosis
Zhu et al. [49]China2022aFerroptosisIn silico and in vivoNAmRNA expression data from TCGA and GEOHuman tumorsA model based on ferroptosis-related genes showed a good ability to predict overall survival
Zhu et al. [77]China2022bPyroptosisIn vitro and in vivoSCC-7, 4MOSC2NAXenograft murine modelGSDME-mediated pyroptosis could awaken potent antitumor immunity
Zhu et al. [78]China2022cPyroptosisIn vitroHN-6, Cal-27NANANLRP3 inflammasome inhibited the invasion and migration of OSCC cells
Zi et al. [79]China2022PyroptosisIn vitro and in vivoHN-6, Cal-27NAXenograft murine modelGSDME expression improved the sensitivity of chemotherapeutics, activated pyroptosis and altered the tumor immune-suppressive microenvironment
Chung et al. [50]Taiwan2023FerroptosisIn vitro and in vivoHSC-3, CAL-27NAXenograft murine model and Human tumorsFerroptosis induced programmed cell death ligand 1 (PD-L1) expression through the membrane damage-independent (NF-κB pathway) and -dependent (calcium influx) mechanisms
Fan et al. [51]China2023FerroptosisIn silicoNAmRNA expression data from TCGA, GEO and ICGCNAThe model composed of 9 prognostic-related differently expressed ferroptosis-related genes (CISD2, DDIT4, CA9, ALOX15, ATG5, BECN1, BNIP3, PRDX5 and MAP1LC3A) was able to predict overall survival
Gupta et al. [89]India2023NecroptosisIn vitroSCC-9NANADocetaxel nanoformulation (PLGA-Dtx) induced both apoptosis and necroptosis via TNF-α/RIP1/RIP3 and caspase-dependent pathway
Huang et al. [90]China2023NecroptosisIn silico and in vitroSAS, SCC-9mRNA expression data from TCGA and GTExNAThe signature containing necroptosis-related genes (HPRT1, PGAM5, BID, SMN1, FADD, and KIAA1191) was associated with survival of OSCC patients, and hypoxanthine phosphoribosyltransferase 1 (HPRT1) was an independent prognostic factor
Jehl et al. [52]France2023FerroptosisIn vitroCAL-27, CALL-33, SCC-9NANASilencing of epiregulin sensitized cells to Cetuximab with induction of ferroptosis
Lee and Roh [53]Republic of Korea2023FerroptosisIn vitroHN3, HN-6, HN12NANADivalent metal transporter 1 silencing or salinomycin promoted ferroptosis
Li et al. [54]China2023FerroptosisIn silico and in vitroCAL-27, HSC-3mRNA expression data from TCGA and GEONAHeat shock protein family A (Hsp70) member 5 (HSPA5) is a ferroptosis regulator by reducing GPX4 and ferritin heavy chain 1 (FTH1) mRNA amounts and increasing acyl-CoA synthetase long chain family member 4 (ACSL4) expression
Liu et al. [80]China2023PyroptosisIn vitro and in vivoSCC-7NAXenograft murine modelThe porphyrin derivative Ac-Asp-Glu-Val-Asp-Asp-TPP (Ac-DEVDD-TPP) was able to induce pyroptosis and apoptosis
Nan et al. [81]China2023PyroptosisIn vitroCAL-27, SCC-15NANAAn increase in the expression of pyroptosis markers was observed in cells treated with radiotherapy
Pan et al. [55]China2023FerroptosisIn vitroHSC-3, HSC-4NANABrain abundant membrane attached signal protein 1 (BASP1) suppressed immunogenic ferroptosis
Wang et al. [57]China2023aFerroptosisIn vitro and in vivoSCC-15NAXenograft murine modelMelatonin combined with erastin exhibited synergistic anticancer effects by inducing ferroptosis
Wang et al. [56]China2023bFerroptosisIn vitro and in vivoCAL-27, SCC-25, SCC-7NAXenograft murine modelGinseng-based carbon dots inhibited cancer invasion and migration in vitro and tumor growth in vivo by inducing ferroptosis
Wang et al. [58]China2023cFerroptosisIn vitroHSC-3, H400NANAPiperlongumine (PL) induced ferroptosis, with synergic effects with CB-839
Wang et al. [82]China2023dPyroptosisIn vitro and in vivoSCC4, SCC-9, SCC-25, CAL-27NAXenograft murine modelThe blockade of cytotoxic T-lymphocyte associated protein 4 (CTLA-4) triggered pyroptosis via the release of interferon-gamma (IFN-γ) and tumor necrosis factor alpha (TNF-α) from activated CD8+ T cells
Wu et al. [59]China2023FerroptosisIn vitro and in vivoSCC-15, SCC-25NAXenograft murine modelAqueous-soluble sporoderm-removed Ganoderma lucidum spore powder promotes ferroptosis of OSCC cells
Xie et al. [60]China2023FerroptosisIn vitroCAL-27, SCC-9NANACadherin 4 (CDH4) positively correlated with ferroptosis suppressor genes
Yan et al. [83]China2023PyroptosisIn vitroCAL-27NANAThe blockage of two exonic splicing enhancers in PD-L1 inhibited cell growth and induced pyroptosis
Yu et al. [61]China2023FerroptosisIn vitro and in vivoCAL-27, SCC-9NAHuman tumorsEnhancer of zeste 2 polycomb repressive complex 2 subunit (EZH2) inhibited erastin-induced ferroptosis in tongue cancer cells via miR-125b-5p/SLC7A11 axis
Yun et al. [91]Republic of Korea2023NecroptosisIn vitroYD-8, YD-10BNANAMachilin D, a lignin from the roots of Saururus chinensis, promoted apoptosis and autophagy and inhibited necroptosis
Zhao and Zhu [63]China2023FerroptosisIn vitro and in vivoSCC-9, CAL-27NAXenograft murine modelDisulfiram/copper complex induced ferroptosis, and inhibition of NRF2 or HO-1 enhanced the sensitivity of OSCC cells
Zhang et al. [62]China2023FerroptosisIn vitroSCC-9NANApH/HAase dual-stimuli triggered smart nanoprobe FeIIITA@HA-induced ferroptosis
* Abbreviations: NA: not applicable, TCGA: The Cancer Genome Atlas, GEO: Gene Expression Omnibus, ICGC: International Cancer Genome Consortium, GTEx: Genotype-Tissue Expression Portal.
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

Siquara da Rocha, L.d.O.; de Morais, E.F.; de Oliveira, L.Q.R.; Barbosa, A.V.; Lambert, D.W.; Gurgel Rocha, C.A.; Coletta, R.D. Exploring beyond Common Cell Death Pathways in Oral Cancer: A Systematic Review. Biology 2024, 13, 103. https://doi.org/10.3390/biology13020103

AMA Style

Siquara da Rocha LdO, de Morais EF, de Oliveira LQR, Barbosa AV, Lambert DW, Gurgel Rocha CA, Coletta RD. Exploring beyond Common Cell Death Pathways in Oral Cancer: A Systematic Review. Biology. 2024; 13(2):103. https://doi.org/10.3390/biology13020103

Chicago/Turabian Style

Siquara da Rocha, Leonardo de Oliveira, Everton Freitas de Morais, Lilianny Querino Rocha de Oliveira, Andressa Vollono Barbosa, Daniel W. Lambert, Clarissa A. Gurgel Rocha, and Ricardo D. Coletta. 2024. "Exploring beyond Common Cell Death Pathways in Oral Cancer: A Systematic Review" Biology 13, no. 2: 103. https://doi.org/10.3390/biology13020103

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