Hostname: page-component-848d4c4894-m9kch Total loading time: 0 Render date: 2024-05-31T11:45:00.434Z Has data issue: false hasContentIssue false

Cell death mechanisms in head and neck cancer cells in response to low and high-LET radiation

Published online by Cambridge University Press:  12 January 2022

Maria Rita Fabbrizi
Affiliation:
Department of Molecular and Clinical Cancer Medicine, Cancer Research Centre, University of Liverpool, 200 London Road, Liverpool, L3 9TA, UK
Jason L. Parsons*
Affiliation:
Department of Molecular and Clinical Cancer Medicine, Cancer Research Centre, University of Liverpool, 200 London Road, Liverpool, L3 9TA, UK Clatterbridge Cancer Centre NHS Foundation Trust, Clatterbridge Road, Bebington, CH63 4JY, UK
*
Author for correspondence: Jason L. Parsons, E-mail: j.parsons@liverpool.ac.uk
Rights & Permissions [Opens in a new window]

Abstract

Head and neck squamous cell carcinoma (HNSCC) is a common malignancy that develops in or around the throat, larynx, nose, sinuses and mouth, and is mostly treated with a combination of chemo- and radiotherapy (RT). The main goal of RT is to kill enough of the cancer cell population, whilst preserving the surrounding normal and healthy tissue. The mechanisms by which conventional photon RT achieves this have been extensively studied over several decades, but little is known about the cell death pathways that are activated in response to RT of increasing linear energy transfer (LET), including proton beam therapy and heavy ions. Here, we provide an up-to-date review on the observed radiobiological effects of low- versus high-LET RT in HNSCC cell models, particularly in the context of specific cell death mechanisms, including apoptosis, necrosis, autophagy, senescence and mitotic death. We also detail some of the current therapeutic strategies targeting cell death pathways that have been investigated to enhance the radiosensitivity of HNSCC cells in response to RT, including those that may present with clinical opportunities for eventual patient benefit.

Type
Review
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution and reproduction, provided the original article is properly cited.
Copyright
Copyright © The Author(s), 2022. Published by Cambridge University Press

Introduction

Head and neck squamous cell carcinoma (HNSCC) is the eighth most common cancer in the UK, with more than 12 000 new cases every year and a one-year survival rate as low as 20% in the hypopharyngeal cancer subtype (Ref. 1). The common risk factors associated with HNSCC are tobacco and alcohol consumption and infection by high-risk (type-16/18) human papillomaviruses (HPV). Interestingly, patients with HPV-positive HNSCC are known to have a better prognosis and improved survival rates due to their improved response to radiotherapy (RT) and chemotherapy as compared with HPV-negative HNSCC (Refs Reference Ang2, Reference Lassen3). Several in vitro studies conducted using HNSCC cell lines have investigated the different molecular mechanisms and biological characteristics responsible for the increased radiosensitivity of HPV-positive cells, and have identified an association with defects in the efficiency of DNA double-strand break repair (reviewed in (Ref. Reference Zhou and Parsons4)). Treatments include surgery, chemotherapy and RT (ionising radiation; IR), where mostly conventional photon (X-ray) radiation is used. However in recent years, use of proton beam therapy (PBT) has increased and has shown significant improvement in HNSCC treatment (Ref. Reference Kim5). This is due to the fact that PBT reduces the degree of healthy tissue injury compared to photon irradiation, due to the characteristic low entrance dose and high in-depth energy deposition at a narrow and well-defined range called the Bragg peak (Fig. 1a). Moreover, X-rays are a low linear energy transfer (LET) radiation treatment which yields a reduced energy deposition along the path of the beam and a lower ionisation density resulting in spatially separated damage to vital macromolecules, particularly DNA (Fig. 1b). In comparison, PBT displays increases in LET at the Bragg peak and beyond the distal edge, which creates ionisation events and damage that is in closer proximity, such as the induction of complex DNA damage (CDD) containing multiple DNA lesions (Ref. Reference Vitti and Parsons6). This increase in CDD represents a challenge to the cellular DNA repair machinery and therefore can contribute to the therapeutic effect of PBT, and more so of heavy ions (such as carbon) that are of significantly higher LET.

Fig. 1. Depth-dose distribution of x-rays (photons) versus protons and relationship to LET leading to DNA damage. (a) Comparison of the dose delivered related to depth in tissue of photons versus protons. Proton irradiation, unlike photons, leads to targeted delivery of the radiation dose to the tumour thus minimising associated normal tissue irradiation, but which leads to associated increases in LET at and around the Bragg peak. (b) Tracks of IR of different LET and their interaction with DNA. Ionisation events (red dots) can occur indirectly (predominant with low-LET radiation) or directly (particularly with high-LET radiation) leading to DNA damage in the form of strand breaks and base damage (orange and green stars, respectively). The low-LET radiation tracks generate largely isolated DNA damage, whereas the densely ionising tracks of high-LET radiation lead to significant levels and formation of CDD.

Nevertheless, and independently from the source and type of IR used, the main goal of RT is to cause sufficient damage to macromolecules particularly DNA, but also to lipids, proteins and many metabolites and therefore to promote cancer cell death while preserving the surrounding healthy tissue. The latter is where targeted dose delivery and energy deposition by PBT have a significant advantage over conventional photon irradiation. Despite this, there are several mechanisms of cell death that may account for IR-induced cell killing, namely apoptosis (Refs Reference Ferlini, D'Amelio and Scambia7, Reference Szumiel8), necrosis (Ref. Reference Szumiel8), mitotic catastrophe (Ref. Reference Roninson, Broude and Chang9), senescence (Ref. Reference Roninson, Broude and Chang9) and autophagy (Ref. Reference Chaurasia10) (Fig. 2). The mechanisms by which photon (X-ray) irradiation kills cancer cells have been studied in depth (Refs Reference Ross11Reference Eriksson and Stigbrand13), but little is known in relation to PBT and other high-LET particles, including carbon ions. In this review, we will provide details on the different cell death mechanisms and the key proteins driving these responses, but then focus on exploring the cellular pathways reportedly involved in IR-induced cell killing particularly in HNSCC cells, highlighting any reported differences between low- and high-LET radiation. Finally, we will present the therapeutic strategies available within these cell death mechanisms that are currently considered to enhance cancer cell radiosensitivity, and which have the potential to move forward into the clinic to improve HNSCC treatment.

Fig. 2. Cell death pathways responsive to IR. Depending on the level of DNA damage and cell type, one of the pathways including apoptosis, necrosis, autophagy, senescence and mitotic death will be initiated. The key steps and proteins involved in coordinating these pathways are shown. If the cell undergoes an initiation cell death pathway (senescence and mitotic catastrophe), then an executive pathway (apoptosis, necrosis and autophagy) will follow eventually.

Initiating and executive mechanisms of cell death

Cell death is a natural consequence of the life cycle progression, although it can also occur if a cell becomes redundant, damaged beyond repair or harmful for the organism (Ref. Reference Fuchs and Steller14). The mechanism chosen by the cell to die is dependent on the type and the extent of damage, but any cellular death pathway shows specific morphological alterations. For example, apoptosis shows cytoplasmic shrinkage, chromatin condensation and nuclear fragmentation resulting finally with the formation of small vesicles or apoptotic bodies released from the cells which are phagocytosed by the surrounding cells. Autophagy includes cytoplasmic vacuolisation and similarly culminates in phagocytic uptake, while necrotic cells exhibit loss of cytoplasm and damaged nuclear membranes (Ref. Reference Galluzzi15). Intracellular vacuolisation, cellular/nuclear enlargement and altered chromatin structure are usually observed in senescent cells, while nuclear changes due to chromosomal mis-segregation and/or persistence of acentric chromosomes, such as multinucleation, are commonly seen in cells undergoing mitotic catastrophe (Ref. Reference Vitale16). Apoptosis, necrosis and autophagy can be considered as executive mechanisms of cell death, while senescence and mitotic catastrophe should be categorised as initiating as these processes do not cause cell death themselves but only act as a trigger of another cell death pathway. Several other cell death mechanisms have been observed, including necroptosis and ferroptosis (Refs Reference Galluzzi17Reference Raudenska, Balvan and Masarik20), but for the purpose of this review we will focus on those listed above.

Apoptosis

Apoptosis is a form of programmed cell death characterised by specific morphological changes (Ref. Reference Kerr, Wyllie and Currie21), which consists of two major subtypes, namely the extrinsic and intrinsic apoptotic pathways (Fig. 3). Extrinsic apoptosis is mediated by membrane receptors, especially by death receptors (e.g. Fas cell surface death receptor and tumour necrosis factor (TNF) receptor superfamily member), and is driven mostly by the initiator caspases 8 and 10 (Ref. Reference Schulze-Osthoff22). Initiator caspase 9 can also trigger extrinsic apoptosis together with unc-5 netrin receptor B and DCC netrin 1 receptor (Ref. Reference Bredesen, Mehlen and Rabizadeh23), although this is mostly involved in the activation of the intrinsic apoptotic pathway. Intrinsic apoptosis starts with mitochondrial outer membrane permeabilisation which is controlled via a fine balance of BCL2 family pro- and anti-apoptotic members, including BCL2-associated X, apoptosis regulator (BAX), BCL2 antagonist/killer 1 (BAK1) and BCL2 and BCL2-like 1 (BCL2L1) (Refs Reference Singh, Letai and Sarosiek24, Reference Czabotar25). When the pro-apoptotic signal overcomes the anti-apoptotic one, mitochondrial proteins are released into the cytoplasm (e.g. cytochrome C and diablo IAP-binding mitochondrial protein) and this triggers initiator caspase 9 activation (Ref. Reference Chipuk, Bouchier-Hayes and Green26). Both the intrinsic and extrinsic pathways of apoptosis proceed with the activation of effector caspases (caspases 3, 6 and 7), which in turn catalyse the specific cleavage of many key cellular proteins. Other members of the cysteine-dependent aspartate-specific protease family are caspase 2 (initiator caspase), caspases 1, 4, 5, 11 and 12 (inflammatory caspases) and caspase 14 (keratinisation-relevant caspase). In terms of morphological features, apoptotic cells show chromatin condensation which progresses into nuclear fragmentation as the apoptotic process proceeds, and this ends with the formation of apoptotic bodies and phagocytosis by the surrounding cells (Ref. Reference Kerr, Wyllie and Currie21).

Fig. 3. The extrinsic and intrinsic pathways of apoptosis. The extrinsic death receptor pathway is activated by death receptor ligands, including FasL, TNF-α or TRAIL, which in turn activates caspase 8 and downstream executing caspases. The intrinsic death receptor pathway is initiated by several intracellular stresses, leading to activation of Bax and Bak on the mitochondrial membrane and which result in the release of cytochrome c from the mitochondria. Cytoplasmic cytochrome c activates caspase 9 and downstream executing caspases.

Necrosis

Necrosis is usually induced by several physical or chemical stress factors, including ischemia and hypoxia. The main event in necrosis is mitochondrial inner membrane depolarisation and outer mitochondrial membrane rupture, due mostly as a consequence of an increase in Ca2+ ions, ATP depletion and reactive oxygen species production (Ref. Reference Vanlangenakker27). However, other metabolic changes have been observed in cells undergoing necrotic death (Ref. Reference Brown28). Necrotic cell death can also be induced when ligands bind to specific receptors, such as TNF receptor 1, Fas and TRAIL receptor, although these activation pathways are equally shared within apoptosis (Refs Reference Peter and Krammer29, Reference Wajant, Pfizenmaier and Scheurich30). Necrosis is also induced by biological stress triggers such as external pathogens, which are recognised by pathogen recognition receptor family members, including the membrane-associated Toll-like receptors, the cytosolic NOD-like receptors and RIG-I-like receptors (Refs Reference Gay and Gangloff31Reference Loo and Gale33). At a macroscopic level, necrosis stimulates the cell membrane to become permeable early in the process, followed by leakage of the cellular contents. In the case of autophagic-like necrosis, numerous vacuoles are observed in the cytoplasm, while dilation of organelles and empty spaces is detectable if the cell undergoes non-lysosomal-type necrosis (Ref. Reference Ziegler and Groscurth34).

Autophagy

Autophagy is a highly regulated mechanism by which the cell removes unnecessary or dysfunctional components by self-degradation and recycling within the cell. Autophagy can be categorised into three forms: macroautophagy (or simply autophagy), microautophagy and chaperone-mediated autophagy (Fig. 4). Macroautophagy requires an intermediate single-membrane vesicle called a phagophore that engulfs the cytoplasm containing the components to be degraded, and rounds up becoming a double-membrane autophagosome. This will then fuse with an endosome or lysosome and releases its contents into the lytic organelle for degradation. Microautophagy, on the other hand, does not have any intermediates and the cellular content to be removed is directly engulfed by an endosome or lysosome (Ref. Reference Schuck35). Chaperone-mediated autophagy requires firstly the recognition of a specific motif in the protein to be degraded by heat shock cognate 71 kDa protein (hsc70), which will then bind the cytosolic tail of lysosome-associated membrane protein type 2A on the lysosomal membrane and transport the substrate into the lysosomal lumen where it will be rapidly degraded (Ref. Reference Tekirdag and Cuervo36). The proteins involved in vesicle formation can be grouped together as the autophagy-related (ATG) proteins, which collectively provide a very dedicated and fine machinery (Ref. Reference Yu, Chen and Tooze37). Autophagic responses are usually triggered by nutrient deprivation, but also by organelle damage or by intracellular pathogens (Ref. Reference Mizushima and Komatsu38). Since autophagy may act as a tumour suppressor, an impaired mechanism could lead to the accumulation of toxic proteins and organelles, such as dysfunctional mitochondria, thus promoting oxidative stress, accumulation of DNA lesions and genomic instability, ultimately leading to promotion of cancer transformation (Ref. Reference Gozuacik and Kimchi39). Recently, it has been suggested that basic autophagy can actually maintain the survival of cancer cells in their unique environment (Ref. Reference Kang40).

Fig. 4. The mechanisms of autophagy. Schematic representation of the three main autophagy pathways: macro-, micro- and chaperone-mediated autophagy. Macroautophagy sequesters cytosolic cargo inside a phagophore formed by specific ATG proteins and lipids. The membrane then seals into an autophagosome and fuses with lysosomes causing the degradation of the trapped cargo. Microautophagy entraps cytosolic cargo in small vesicles formed by invagination of the lysosomal membrane. Chaperone-mediated autophagy involves the selective degradation of KFRQ-like motif-bearing proteins delivered to the lysosomes via chaperone HSC70 and their internalisation in lysosomes via the receptor lysosome-associated membrane protein type 2A (LAMP2A).

Senescence

The term ‘replicative cellular senescence’ indicates a process in which cells are alive but are unable to undergo further cell division and therefore are in a permanent state of growth arrest (Ref. Reference Ben-Porath and Weinberg41). Senescence can be triggered by a variety of factors and is usually associated with increased levels of p53, p21WAF–1 and/or p16INK4A, but telomere-associated DNA damage foci are also often present (Ref. Reference Kuilman42). The molecular mechanisms involved in senescence pathway activation and execution have been extensively studied and reviewed (Refs Reference Ben-Porath and Weinberg41, Reference Chakradeo, Elmore and Gewirtz43). Assessment of senescent cells can be easily performed with histochemical staining of β-galactosidase (Refs Reference Kurz44, Reference Yang and Hu45), although other markers of senescence that have been identified include Cathepsin D and Dec-1 (Refs Reference Byun46, Reference Kim47). Morphological changes in senescent cells include increased granularity and cytoplasmic vacuolar content, followed by cell flattening. Senescence has always been considered an irreversible process, due to the major metabolic modifications the cell undergoes together with genetic and structural alterations, although several studies have highlighted the possibility of cells escaping senescence. In fact, in the case of senescence induced by chemotherapy or RT, several studies have reported that cells were able to re-enter the cell cycle due to overexpression of the cyclin-dependent kinase, Cdc2/Cdk1 or its downstream target survivin (Refs Reference Roberson48Reference La Porta, Zapperi and Sethna50). Considering that most tumours have inactive p53 and/or p16INK4A/Rb signalling pathways, escaping senescence should be considered as a possibility.

Mitotic catastrophe

The term ‘mitotic catastrophe’, or the more appropriate phrase ‘mitotic death’, indicates cell death induced by aberrant mitosis (Ref. Reference Galluzzi17). Cells undergoing mitotic catastrophe show unique morphologically defined features, such as multinucleation and micronucleation, mainly due to the chromosomal mis-segregation characteristic of this process (Ref. Reference Vitale16). Mitotic death can be triggered by either exogenous or endogenous sources, which ultimately cause several cell dysfunctions, such as altered DNA replication and chromosome segregation, and interference with microtubular dynamics (Refs Reference Dominguez-Brauer51Reference Denisenko53). The molecular mechanisms involved in mitotic catastrophe are still under investigation, although p53 seems to play a role in the trigger signal (Ref. Reference Vitale16) and to activate the transduction cascade via caspase 2, which in turn initiates a variant of intrinsic apoptosis regulated by members of the BCL2 protein family (Refs Reference Dawar54, Reference Lopez-Garcia55). However, many cancer cell types lack functional p53 and in those cases it appears that cells showing massive chromosomal aberration are driven preferentially into necrosis (Ref. Reference Surova and Zhivotovsky56). It is interesting to note that cell death is not the only possible fate aberrant mitotic cells can face, as it has been observed that cells escaping the mitotic block can instead enter p53-mediated or hippo-mediated cellular senescence (Refs Reference Ganem57Reference Meitinger60), or survive as polyploid and aneuploid cells and initiate neoplastic transformation and progression (Ref. Reference Vitale16). However, a recent study suggests that cells escaping mitotic catastrophe stimulate the immunological response which in turn will trigger cell degradation (Ref. Reference Mackenzie61), providing an efficient mechanism for the control of tissue homeostasis.

Radiation-induced cell death mechanisms

Generally, the therapeutic effect of RT is achieved through sufficient cell injury, particularly in terms of macromolecule and DNA damage, to overcome the cancer cells ability to repair the damage and therefore forcing the cell into initiating a cell death-activated pathway. The form of cell death induced by a particular anti-cancer agent such as IR depends on several factors, including cell type, the type of DNA damage to which the cell is exposed and the dose of the agent used (Refs Reference Surova and Zhivotovsky56, Reference Abend62). For example, γ-radiation exposure can cause a massive apoptotic response in T and B cells but not in monocyte-derived macrophages and immature dendritic cells ex vivo (Ref. Reference Heylmann63). Furthermore, it is known that X-rays and PBT cause a different spectrum of DNA damage in cancer cells due to changes in energy/LET (Refs Reference Vitti and Parsons6, Reference Carter64), which is likely to trigger a different cell death response. Therefore, whilst low-LET X-rays generally induce a high proportion of DNA base damage and single DNA strand breaks (SSB) relative to DNA double-strand breaks (DSB), higher LET radiation exposure including PBT but more so heavy ions can induce increased amounts and complexity of CDD. CDD is defined as multiple DNA damage types within close proximity (1–2 helical turns of the DNA), and can be classified as either DSB-associated or non-DSB-associated (Ref. Reference Goodhead65). This though suggests that depending on the spectrum of DNA damage induced, different radiation sources can cause different cell death mechanisms to be activated that should be considered. Furthermore in addition to the type of damage, the dose of IR leading to a specific level of DNA damage may also have a major influence on the selection of cell death mechanism triggered, as well as the switching between these events given that many cell death mechanisms share several initiating factors. Finally, the long held concept that cells either repair their damage or undergo apoptosis after IR treatment is outdated, and the role of apoptosis in the tumour response to radiation has been minimised considering that most tumours actually lose the ability to initiate the apoptotic pathway (Ref. Reference Gudkov and Komarova66). A more important role in the anti-tumour effect of radiation is played by mitotic catastrophe or senescence, although as already stated previously, both these mechanisms cannot be considered strictly cell death and therefore rely on other pathways (e.g. apoptosis or autophagy) that trigger this phenotype.

Cell death mechanisms after low LET exposure

As mentioned above, tumour cells post-IR can undergo apoptosis, although recent studies have implicated a reduced contribution of this particular pathway to the total amount of cell death to a relatively low level (Refs Reference Rodel67, Reference Gewirtz68). Apoptosis detection in several tumour cell lines, including breast cancer, non-small-cell lung cancer and colorectal cancer, has been reported to never exceed 30% of the total even at significantly high doses of radiation (Ref. Reference Chakradeo69). HNSCC cell lines appear to show no difference in the levels of IR-induced apoptosis, with the vast majority of the studies agreeing that low-LET radiation (e.g. X-rays and γ-rays) does not cause significant apoptosis activation (summarised in Table 1). In fact, thyroid cancer cells show no apoptotic response after a relatively low dose of 3 Gy (Ref. Reference Suzuki70), and only a modest increase after 20 Gy treatment (Ref. Reference Kim71), whilst laryngeal squamous carcinoma cells appear to reach an increase in 20% of apoptotic cells after a fractionated 10 Gy dose (Ref. Reference Wang72). The HNSCC cell lines UM-SCC1, UM-SCC6 (both HPV-negative) and UPCI-SCC-154 (HPV-positive) showed no difference in caspase 3 activation and Annexin V detection at 2 and 4 Gy X-ray exposure (Refs Reference Kahler73, Reference Kashin74). Similarly in nasopharyngeal carcinoma cell lines, no difference in apoptosis was observed after 8 Gy (Ref. Reference Zhan and Ni75), although in contrast another study showed a 10-fold increase in the apoptotic response after 10 Gy (Ref. Reference Nie76).

Table 1. Biological effects of low-LET radiation on head and neck cancer cells

a X-ray radiation treatment unless differently stated.

Necrosis pathway activation has mostly been considered more as a side-effect of RT in surrounding healthy tissue, rather than a cell death mechanism for cancer cells. Patients undergoing RT for HNSCC can experience significant side-effects, including osteoradionecrosis and oral cavity necrosis, which may require surgical intervention (Ref. Reference Zehr and Cooper87). The mechanism underlying this specific necrotic transformation is not fully understood, although fibrosis may play a major role. This is due to the fact that RT increases the levels of reactive oxygen species-mediated cytokines, such as TNF-α, transforming growth factor-β1 and connective tissue growth factor, resulting in unregulated fibroblastic activation (Ref. Reference Dambrain88). Human and mouse leukaemia cells have been shown to undergo necrosis after 300 and 9 Gy X-ray exposure, respectively (Refs Reference Nakano and Shinohara89, Reference Akagi, Ito and Sawada90), suggesting the radiosensitive nature of the murine cells. HNSCC cells appear to undergo necrosis after X-ray radiation exposure at different degrees depending on cell type and HPV status (summarised in Table 1). In fact 48 h after irradiation, HPV-negative SqCC/Y1 cells showed a significantly higher percentage of necrotic cells compared to the unirradiated controls, but this was not observed in another HPV-negative cell line (HN5) or in two HPV-positive cell lines (UPCI-SCC-154 and UMSCC-47) tested (Ref. Reference Wang77). One explanation, at least in the HPV-positive HNSCC cell lines, could be provided by the fact that the HPV oncoprotein E7 inhibits necrosis activation, thus making these cell lines less prone to this mechanism of cell death (Ref. Reference Shankar91).

Since autophagy is mostly associated with tumour suppression, increases in ATG proteins are to be expected following RT, and have been observed in HNSCC (summarised in Table 1). The ratio of LC3II/LC3I (an autophagic marker) has been shown to increase significantly in nasopharyngeal carcinoma cells after exposure to 4 Gy X-rays (Ref. Reference Liu78), and protein level analysis showed that autophagic signalling proteins including Beclin1, Atg5, Atg7 and LC3B were upregulated in a time- and dose-dependent manner post-irradiation (Ref. Reference Jing92). However, the same treatment did not appear to trigger any LC3B activation in SQ20B carcinoma cells (Ref. Reference Cerniglia79), suggesting a role for a differential gene expression profile in contributing to autophagy specific for each cell line. Exposure to 6 Gy of low-LET X-rays radiation caused a 2–5-fold increase in several ATG proteins in Cal-33 carcinoma cells, including LC3B, p62, Atg4A and Atg4B (Ref. Reference Deville80). However, although autophagy directly contributes to death in stressed cells (Ref. Reference Ryter, Mizumura and Choi93), several studies have actually suggested a protective role for autophagy, which actually helps cancer cells to survive by reducing cell damage (Ref. Reference Mathew, Karantza-Wadsworth and White94). Autophagy is frequently activated in radioresistant cancer cells (Ref. Reference Han95) and the pathway shares upstream mediators with apoptosis, indicating a clear cross-talk between the two mechanisms and a fine balance between cell death and cell survival in response to IR. It has been proposed, in fact, that low-energy X-rays trigger the NF-κB pathway and induce Beclin1 gene expression, which consequently activates autophagy and confers radioresistance in HNSCC cancer cells (Ref. Reference Shu96). Moreover, apoptosis can be inhibited by autophagy proteins, such as Beclin1, which can degrade caspase 8 and interfere with the activation of Bid (Ref. Reference Nikoletopoulou97), reducing cell death levels after irradiation (Ref. Reference Shu96).

Senescent cells induced by RT usually trigger inflammation, which in turn leads to the targeted removal of cancer cells (Ref. Reference Chang98), making senescence an outcome at the initial treatment stages. In contrast, some studies suggest the possibility of senescence being a protective mechanism for cancer cells, causing increased aggressiveness and metastasis (Refs Reference Milanovic99, Reference Kim100). Relatively radiosensitive SCC61 HNSCC cells were demonstrated to accumulate 76% of senescent cells, while radioresistant SQ20B cells only displayed 18% of cells positive for senescence markers after 10 Gy γ-ray exposure (Ref. Reference Ricco81) (summarised in Table 1). X-ray (4 Gy) treatment significantly increased the percentages of senescent cells at 4 and 6 days post-IR in two HPV-negative HNSCC cell lines (SqCC/Y1 and HN5) and two HPV-positive cell lines (UPCI-SCC-154 and UMSCC-47), although the absolute percentage only ranged from 5 to 18% (Ref. Reference Wang77). In contrast, Detroit562 HNSCC cells (HPV-negative) showed no increase in the levels of senescence markers, such as p21 and β-galactosidase, after 2 Gy X-rays (Ref. Reference Frederick82). The potentially higher level of senescent cells present following irradiation of HPV-positive cells can be partially explained by the ability of the E2 oncoprotein to induce overexpression of p21 and downregulation of E7, resulting in increased levels of hypophosphorylated pRb protein and ultimately to G1 cell cycle arrest (Ref. Reference Wells101). The nasopharyngeal carcinoma cell line CNE2 showed around 56 and 79% IR-induced cellular senescence 72 h after 6 and 10 Gy X-ray exposure, respectively (Ref. Reference Yu83). Since CNE2 cells are p16-deficient, only p53 and p21 protein levels were elevated together with the anti-apoptotic protein Bcl-xL, suggesting senescence was preferred over apoptosis as the cell death mechanism. Conversely, it has been shown that only 5% of p53-deficient HNSCC cancer cells undergo senescence after 4 Gy X-rays radiation treatment, whilst p53-wild type containing cells reach up to 60% senescence-associated β-galactosidase-positive cells (Ref. Reference Fitzgerald102). This confirms the prominent role of p53 in driving senescence compared to p16. This confirmed previous observations in 38 different HNSCC cell lines where p53 mutation significantly decreased IR-induced senescence and conferred radioresistance (Ref. Reference Skinner103). Furthermore, the importance of the p53–p21 interaction for the initiation of the senescence pathway has been highlighted (Refs Reference Fitzgerald102, Reference Skinner103).

HNSCC cells exposed to 1 Gy γ-radiation have been shown not to display any significant induction of mitotic catastrophe (Ref. Reference Gottgens104), whilst a higher dose (6 Gy) of X-rays caused 20% of FaDu cells to undergo this process (Ref. Reference Yun84) (summarised in Table 1). The increase in mitotic catastrophe induction appears to be p53-independent, in line with previously reported evidence for HCT116 colorectal cancer cell lines after 4 Gy X-ray treatment (Ref. Reference Hafsi105). Oesophageal squamous cell carcinoma cell lines have demonstrated a modest increase in mitotic catastrophe markers after 6 Gy X-ray irradiation (Ref. Reference Cheng85), while nasopharyngeal carcinoma cells displayed around 50% of cells undergoing mitotic catastrophe after 20 Gy of X-ray treatment (Ref. Reference Lv86). Whilst the dose used is different, this could suggest that triggering of mitotic catastrophe as a cell death mechanism could be tumour cell-dependent. Mitotic catastrophe was observed to be the dominant mechanism of cell death in HPV-positive and HPV-negative HNSCC cell lines after a single 4 Gy X-ray radiation exposure, with no major difference between 4 and 72 h timepoint (Ref. Reference Wang77). Moreover, both types of cell lines tested responded in a similar manner, suggesting that HPV infection has no dramatic impact on the degree of mitotic catastrophe induction.

In summary, evidence suggests that low-LET radiation exposure may trigger different types of HNSCC cell death mechanism, and that the cells undergo a specific pathway depending on the radiation dose but also the cellular genetic profile.

Cell death mechanisms after high LET exposure

Mechanistic modelling has suggested that high-LET radiation, such as α-particles and heavy ions, can cause up to 90% of CDD within the total amount of DNA lesions induced, compared to ~30% in the case of low-LET radiation exposure (Refs Reference Nikjoo106, Reference Watanabe, Rahmanian and Nikjoo107). CDD has been clearly demonstrated to be a challenge to the DNA repair machinery and can persist in cells and tissues several (6–24) hours after IR, while simple SSBs and DSBs resolve in less than 30 min and 2 h, respectively (Refs Reference Carter64, Reference Magnander108Reference Nickson111). High-LET radiation can therefore kill cancer cells more efficiently compared to low LET treatments. For example, and as reported for SQ20B laryngeal squamous cells, exposure to 4 Gy low-LET X-rays produced 56% cell survival whereas high-LET carbon ion (LET = 184 keV/μm) at the same dose yielded only 9% of cells surviving (Ref. Reference Bertrand112) (Table 2). A reduction in cell survival from 55 to 15% was also observed in UMSCC74A after 4 Gy high-LET proton treatment (LET = 12 keV/μm) compared to 4 Gy low-LET proton exposure (LET = 1 keV/μm), while UMSCC6 cells displayed a less marked difference (35% cell survival for low-LET versus 20% for high-LET) (Refs Reference Carter64, Reference Nickson113). This demonstrates the differences in inherent radioresistance of the different HNSCC cells to both low and high-LET radiation. Nevertheless, CDD formation is likely the major contributor to this observed reduced survival in response to high-LET radiation thereby triggering apoptosis or mitotic catastrophe, although other mechanisms in cell death pathway activation are likely to be involved once the cell fails to resolve the damage. For example, effects on centrosome biology in HNSCC cell lines have recently been shown in response to high-LET protons (Ref. Reference Nickson113).

Table 2. Biological effects of high-LET radiation on head and neck cancer cells

Interestingly, the mechanisms of cell death in response to high-LET radiation in HNSCC cells have not been studied extensively. Human tongue carcinoma cells have been shown to display less than a 5% increase in apoptotic marker activation after 5 Gy high-LET carbon ion exposure (LET = 70 keV/μm) compared to X-ray radiation (Ref. Reference Asakawa114), although this was not apparent when the dose was reduced to 1 Gy (Ref. Reference Takahashi115) (Table 2). Apoptosis activation has also been reported in a study on radiosensitive human oesophageal carcinoma cells, where caspase 3 activation was 1.8-fold higher after heavy ion irradiation versus X-rays, while only marginal differences were observed in their radioresistant cell counterparts (Ref. Reference Hartfiel116). This suggests that the cell-killing effect is cell line and radiation quality-dependent, and that perhaps other types of cell death may play a more prominent role. This confirms previous observations in radiosensitive laryngeal squamous cancer cells and their appropriate radioresistant cell lines (Ref. Reference Maalouf117). Considering the significant increased therapeutic use of PBT, but also heavy ion radiotherapy, further and more extensive studies are needed to fully elucidate the cell death mechanisms activated in specific HNSCC cell types in response to high-LET radiation exposure.

Therapeutic strategies

Targeting key proteins involved in cell death mechanisms is a feasible approach in increasing HNSCC radiosensitivity, which can lead to more effective treatment of the tumour. Some targets involved in cell death pathway activation and/or execution have been already identified and explored in vitro. In most cases, tumour cells highly express anti-apoptotic proteins, therefore their inhibition could potentially increase cellular radiosensitivity. Survivin is a protein inhibitor of the terminal effector caspases and is highly expressed in several types of cancer, including head and neck malignancies (Refs Reference Ambrosini, Adida and Altieri118, Reference Frassanito119). Efficient downregulation of survivin has been extensively studied with promising results in terms of enhancing tumour sensitivity to various therapeutic interventions (reviewed in (Ref. Reference Santarelli120)). Livin, another member of the apoptosis inhibitor protein family, has been found to be associated with tumour progression and poor prognosis in various human cancers. Its inhibition has been shown to suppress tumour cell invasion and enhance cell apoptosis, with elevated expression levels of cleaved caspases 3 and 7 and cleaved PARP in anaplastic thyroid cancer (Ref. Reference Kim71) and in laryngohypopharyngeal cancer models (Ref. Reference Yoon121). Other anti-apoptotic proteins, including Bcl-2 and vitronectin, have been investigated in several HNSCC subtypes, with their inhibition significantly increasing apoptosis in response to X-rays (Refs Reference Guy122Reference Jiang124) or γ-radiation (Ref. Reference Zerp125). A phase I/II clinical trial initiated to investigate mitochondria-derived activator of caspases as a mimetic therapy in patients with previously untreated stage III/IV HNSCC in combination with cisplatin and radiation is currently ongoing (ClinicalTrials.gov identifier NCT02022098).

Post-radiotherapy, necrosis usually occurs in the oral cavity, maxilla, mandible and salivary glands more than the tumour itself, and the aim therefore is to prevent this to improve patient quality of life. Although several therapies have been reported, there is not a universally accepted approach to tackle necrosis and the treatment options are variable. Surgical treatments are still of preference, but recently some pharmacological options have been investigated, including treatments with antioxidants or biological molecules, such as basic fibroblast growth factor or bone morphogenetic protein-1 (reviewed in (Refs Reference Pitak-Arnnop126, Reference Madrid, Abarca and Bouferrache127)). Nevertheless, inducing necrosis in HNSCC cells is a possibility that has been partially explored. The monoclonal anti-IGF-1R antibody A12 in combination with γ-rays has been shown to dramatically induce necrotic death in FaDu-derived xenografts compared to unirradiated controls (Ref. Reference Riesterer128), while the second mitochondria-derived activator of caspase mimetic birinapant has proven effective both in caspase-8-deficient and FADD-overexpressing HNSCC cells, inducing programmed necrosis and increasing cell radiosensitivity after X-ray treatment (Refs Reference Uzunparmak129, Reference Eytan130).

Although ATG proteins are often upregulated in tumour cells, the activation of this specific pathway could lead to massive cell death after radiotherapy. The phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) pathway is a critical regulator of autophagy, and targeting this could be an important therapeutic strategy enhancing the sensitivity of tumour cells to IR. Several pharmacological inhibitors have been tested on various cancer cell types, including HNSCC cells, both in vitro and in xenograft models with promising results (reviewed in (Ref. Reference Wanigasooriya131)). A phase I clinical trial is currently ongoing in high-risk patients with locally advanced squamous cell carcinoma to test a potent and highly specific oral pan-class I PI3K inhibitor, BKM120 (ClinicalTrials.gov Identifier NCT02113878). Other ATG targets have been investigated, including Kelch-like ECH-associated protein 1 (Ref. Reference Deville80) and p62, whose overexpression has been reported to induce autophagy in HNSCC cells (Ref. Reference Lee132).

The induction of senescence can be clinically beneficial, but currently there are no specific senescence-inducing agents available. The ability of cells to enter radiation-induced senescence is almost strictly linked to p53. In fact, it has been reported that HNSCC cells with wild-type or non-disruptive mutations of p53 undergo senescence after radiotherapy, while p53 mutant cells are more radioresistant and show higher levels of senescence markers after treatment with metformin (anti-diabetic agent that has been shown to induce reactive oxygen species) (Ref. Reference Skinner103). Reactivation of p53 restored senescence and increased radiosensitivity in p53 mutant HNSCC cells, although interestingly the study reported that the mechanism is only partially p53-dependent (Ref. Reference Chuang133). Treatment with rapamycin or the mTOR inhibitor PP242 in parotid carcinoma cells in vitro increased heterochromatin formation, and induced irreversible growth arrest and premature senescence. Whilst in tumour xenografts, PP242 delayed tumour regrowth after X-ray irradiation and increased senescence-associated β-galactosidase staining (Ref. Reference Nam134).

Inhibition of proteins involved in DNA replication and mitosis could be a valid therapeutic solution to force the cells into mis-segregation of chromosomes and promote aberrant mitotic division. As mitotic catastrophe is a relatively new cellular endpoint, the current literature is scarce and only a few candidates have been investigated as chemotherapeutic targets. Polo-like kinase 1 (PLK1) is a serine/threonine kinase which functions as a pleiotropic master regulator of mitosis and regulates DNA replication after stress (Ref. Reference Joukov and De Nicolo135). PLK1 depletion has been reported to induce mitotic cell cycle arrest and inhibit the separation of sister chromatids in oesophageal cancer cells, causing failure of cytokinesis followed by massive apoptotic cell death (Ref. Reference Bu136). These results have been confirmed in human nasopharyngeal cancer cells, where PDK1 inhibition was found to greatly reduce cell survival, alone or in combination with radiation, due to G2/M cell cycle arrest and aberrant spindle formation, which in turn caused mitotic catastrophe (Ref. Reference Shi137). Furthermore, co-treatment with PLK1 inhibitor and inhibitors targeting Aurora A and Aurora B enhanced metaphase arrest and mitotic slippage in nasopharyngeal cancer cells, ultimately inducing mitotic catastrophe (Ref. Reference Li138). The Aurora A and B protein kinases are key players in mitotic control and therefore another set of potential targets to increase cancer radiosensitivity. Aurora B inhibition in fact has been demonstrated to lead to G2/M accumulation, polyploidy and subsequent cell death by mitotic catastrophe in anaplastic thyroid carcinoma in vitro and reduced growth of ATC tumour xenogratfs (Ref. Reference Libertini139). Aurora A depletion appears to have a limited effect on HPV-negative HNSCC cells when administered alone, although still causing spindle defects and cytostasis, while co-treatment with a WEE1 cell cycle checkpoint kinase inhibitor triggered mitotic catastrophe in vitro and reduced tumour growth in FaDu and Detroit 562-derived xenografts (Ref. Reference Lee140). Other potential cellular targets for inhibitors that have yielded promising results as chemotherapeutic agents against HNSCC cells include WEE1 (Refs Reference Mak141Reference Deneka143), CHK1/2 (Refs Reference Mak141, Reference Deneka143) and PP2A (Ref. Reference Lv86), although further studies are needed in order to determine their specific potential as radiosensitisers.

Conclusions

Conventional photon (X-ray) radiotherapy has been used for several decades in the treatment of HNSCC, while particle beam therapy including protons and carbon ions have only recently gained increasing utility but which benefit from their high energy localised deposition coupled with the preservation of the surrounding healthy tissue. Nevertheless, current knowledge regarding the molecular mechanisms involved in cancer cell death in response to high-LET radiation compared to conventional low-LET photon therapy is still quite limited, and requires further investigation. Elucidating the different cell death mechanisms activated by tumour cells after high-LET radiotherapy would allow a more targeted therapeutic strategy, with the use of inhibitors specifically designed for proteins involved in enhancing the determined cell death pathway. This will ultimately lead to a more personalised and effective approach during radiotherapy, whilst also enabling a reduction in the dose of radiation needed to obtain a full tumour eradication and therefore limiting the possibility of acute and long-term adverse side-effects associated with irradiation of the associated normal tissues.

Financial support

Related research is currently funded by North West Cancer Research (J.P., grant number CR1197), the Medical Research Council (J.P., grant number MR/V028944/1) and the National Institutes of Health (1R01CA256854-01).

Conflict of interest

None.

References

Ang, KK et al. (2010) Human papillomavirus and survival of patients with oropharyngeal cancer. New England Journal of Medicine 363, 2435.CrossRefGoogle ScholarPubMed
Lassen, P et al. (2009) Effect of HPV-associated p16INK4A expression on response to radiotherapy and survival in squamous cell carcinoma of the head and neck. Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology 27, 19921998.CrossRefGoogle ScholarPubMed
Zhou, C and Parsons, JL (2020) The radiobiology of HPV-positive and HPV-negative head and neck squamous cell carcinoma. Expert Reviews in Molecular Medicine 22, e3.CrossRefGoogle ScholarPubMed
Kim, JK et al. (2018) Proton therapy for head and neck cancer. Current Treatment Options in Oncology 19, 28.CrossRefGoogle ScholarPubMed
Vitti, ET and Parsons, JL (2019) The radiobiological effects of proton beam therapy: impact on DNA damage and repair. Cancers (Basel) 11, 946.CrossRefGoogle ScholarPubMed
Ferlini, C, D'Amelio, R and Scambia, G (2002) Apoptosis induced by ionizing radiation. The biological basis of radiosensitivity. Subcellular Biochemistry 36, 171186.CrossRefGoogle ScholarPubMed
Szumiel, I (1994) Ionizing radiation-induced cell-death. International Journal of Radiation Biology 66, 329341.CrossRefGoogle ScholarPubMed
Roninson, IB, Broude, EV and Chang, BD (2001) If not apoptosis, then what? Treatment-induced senescence and mitotic catastrophe in tumor cells. Drug Resistance Updates 4, 303313.CrossRefGoogle ScholarPubMed
Chaurasia, M et al. (2016) Radiation-induced autophagy: mechanisms and consequences. Free Radical Research 50, 273290.CrossRefGoogle ScholarPubMed
Ross, GM (1999) Induction of cell death by radiotherapy. Endocrine-Related Cancer 6, 4144.CrossRefGoogle ScholarPubMed
Prise, KM et al. (2005) New insights on cell death from radiation exposure. The Lancet. Oncology 6, 520528.CrossRefGoogle ScholarPubMed
Eriksson, D and Stigbrand, T (2010) Radiation-induced cell death mechanisms. Tumour Biology 31, 363372.CrossRefGoogle ScholarPubMed
Fuchs, Y and Steller, H (2015) Live to die another way: modes of programmed cell death and the signals emanating from dying cells. Nature Reviews Molecular Cell Biology 16, 329344.CrossRefGoogle ScholarPubMed
Galluzzi, L et al. (2007) Cell death modalities: classification and pathophysiological implications. Cell Death and Differentiation 14, 12371243.CrossRefGoogle ScholarPubMed
Vitale, I et al. (2011) Mitotic catastrophe: a mechanism for avoiding genomic instability. Nature Reviews Molecular Cell Biology 12, 384391.CrossRefGoogle ScholarPubMed
Galluzzi, L et al. (2018) Molecular mechanisms of cell death: recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death and Differentiation 25, 486541.CrossRefGoogle ScholarPubMed
Tang, D et al. (2019) The molecular machinery of regulated cell death. Cell Research 29, 347364.CrossRefGoogle ScholarPubMed
Chen, X et al. (2021) Broadening horizons: the role of ferroptosis in cancer. Nature Reviews. Clinical Oncology 18, 280296.CrossRefGoogle ScholarPubMed
Raudenska, M, Balvan, J and Masarik, M (2021) Cell death in head and neck cancer pathogenesis and treatment. Cell Death & Disease 12, 192.CrossRefGoogle ScholarPubMed
Kerr, JF, Wyllie, AH and Currie, AR (1972) Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics. British Journal of Cancer 26, 239257.CrossRefGoogle ScholarPubMed
Schulze-Osthoff, K et al. (1998) Apoptosis signaling by death receptors. European Journal of Biochemistry 254, 439459.CrossRefGoogle ScholarPubMed
Bredesen, DE, Mehlen, P and Rabizadeh, S (2004) Apoptosis and dependence receptors: a molecular basis for cellular addiction. Physiological Reviews 84, 411430.CrossRefGoogle ScholarPubMed
Singh, R, Letai, A and Sarosiek, K (2019) Regulation of apoptosis in health and disease: the balancing act of BCL-2 family proteins. Nature Reviews Molecular Cell Biology 20, 175193.CrossRefGoogle ScholarPubMed
Czabotar, PE et al. (2014) Control of apoptosis by the BCL-2 protein family: implications for physiology and therapy. Nature Reviews Molecular Cell Biology 15, 4963.CrossRefGoogle ScholarPubMed
Chipuk, JE, Bouchier-Hayes, L and Green, DR (2006) Mitochondrial outer membrane permeabilization during apoptosis: the innocent bystander scenario. Cell Death & Differentiation 13, 13961402.CrossRefGoogle ScholarPubMed
Vanlangenakker, N et al. (2008) Molecular mechanisms and pathophysiology of necrotic cell death. Current Molecular Medicine 8, 207220.CrossRefGoogle ScholarPubMed
Brown, GC (2007) Nitric oxide and mitochondria. Frontiers in Bioscience-Landmark 12, 10241033.CrossRefGoogle ScholarPubMed
Peter, ME and Krammer, PH (2003) The CD95(APO-1/Fas) DISC and beyond. Cell Death and Differentiation 10, 2635.CrossRefGoogle ScholarPubMed
Wajant, H, Pfizenmaier, K and Scheurich, P (2003) Tumor necrosis factor signaling. Cell Death and Differentiation 10, 4565.CrossRefGoogle ScholarPubMed
Gay, NJ and Gangloff, M (2007) Structure and function of Toll receptors and their ligands. Annual Review of Biochemistry 76, 141165.CrossRefGoogle ScholarPubMed
Franchi, L et al. (2009) Function of Nod-like receptors in microbial recognition and host defense. Immunological Reviews 227, 106128.CrossRefGoogle ScholarPubMed
Loo, YM and Gale, M (2011) Immune signaling by RIG-I-like receptors. Immunity 34, 680692.CrossRefGoogle ScholarPubMed
Ziegler, U and Groscurth, P (2004) Morphological features of cell death. News in Physiological Sciences 19, 124128.Google ScholarPubMed
Schuck, S (2020) Microautophagy – distinct molecular mechanisms handle cargoes of many sizes. Journal of Cell Science 133, jcs246322.CrossRefGoogle ScholarPubMed
Tekirdag, K and Cuervo, AM (2018) Chaperone-mediated autophagy and endosomal microautophagy: jointed by a chaperone. Journal of Biological Chemistry 293, 54145424.CrossRefGoogle Scholar
Yu, L, Chen, Y and Tooze, SA (2018) Autophagy pathway: cellular and molecular mechanisms. Autophagy 14, 207215.CrossRefGoogle ScholarPubMed
Mizushima, N and Komatsu, M (2011) Autophagy: renovation of cells and tissues. Cell 147, 728741.CrossRefGoogle ScholarPubMed
Gozuacik, D and Kimchi, A (2004) Autophagy as a cell death and tumor suppressor mechanism. Oncogene 23, 28912906.CrossRefGoogle ScholarPubMed
Kang, R et al. (2020) The multifaceted effects of autophagy on the tumor microenvironment. Advances in Experimental Medicine and Biology 1225, 99114.CrossRefGoogle ScholarPubMed
Ben-Porath, I and Weinberg, RA (2005) The signals and pathways activating cellular senescence. International Journal of Biochemistry & Cell Biology 37, 961976.CrossRefGoogle ScholarPubMed
Kuilman, T et al. (2010) The essence of senescence. Genes & Development 24, 24632479.CrossRefGoogle Scholar
Chakradeo, S, Elmore, LW and Gewirtz, DA (2016) Is senescence reversible? Current Drug Targets 17, 460466.CrossRefGoogle ScholarPubMed
Kurz, DJ et al. (2000) Senescence-associated (beta)-galactosidase reflects an increase in lysosomal mass during replicative ageing of human endothelial cells. Journal of Cell Science 113, 36133622.CrossRefGoogle ScholarPubMed
Yang, NC and Hu, ML (2005) The limitations and validities of senescence associated-beta-galactosidase activity as an aging marker for human foreskin fibroblast Hs68 cells. Experimental Gerontology 40, 813819.CrossRefGoogle ScholarPubMed
Byun, HO et al. (2009) Cathepsin D and eukaryotic translation elongation factor 1 as promising markers of cellular senescence. Cancer Research 69, 46384647.CrossRefGoogle ScholarPubMed
Kim, BC et al. (2014) Evaluation of premature senescence and senescence biomarkers in carcinoma cells and xenograft mice exposed to single or fractionated irradiation. Oncology Reports 31, 22292235.CrossRefGoogle ScholarPubMed
Roberson, RS et al. (2005) Escape from therapy-induced accelerated cellular senescence in p53-null lung cancer cells and in human lung cancers. Cancer Research 65, 27952803.CrossRefGoogle ScholarPubMed
Wang, Q et al. (2011) Survivin and escaping in therapy-induced cellular senescence. International Journal of Cancer 128, 15461558.CrossRefGoogle ScholarPubMed
La Porta, CA, Zapperi, S and Sethna, JP (2012) Senescent cells in growing tumors: population dynamics and cancer stem cells. PLoS Computational Biology 8, e1002316.CrossRefGoogle ScholarPubMed
Dominguez-Brauer, C et al. (2015) Targeting mitosis in cancer: emerging strategies. Molecular Cell 60, 524536.CrossRefGoogle ScholarPubMed
Neelsen, KJ et al. (2013) Oncogenes induce genotoxic stress by mitotic processing of unusual replication intermediates. Journal of Cell Biology 200, 699708.CrossRefGoogle ScholarPubMed
Denisenko, TV et al. (2016) Mitotic catastrophe and cancer drug resistance: a link that must to be broken. Drug Resistance Updates 24, 112.CrossRefGoogle ScholarPubMed
Dawar, S et al. (2017) Caspase-2-mediated cell death is required for deleting aneuploid cells. Oncogene 36, 27042714.CrossRefGoogle ScholarPubMed
Lopez-Garcia, C et al. (2017) BCL9L dysfunction impairs caspase-2 expression permitting aneuploidy tolerance in colorectal cancer. Cancer Cell 31, 7993.CrossRefGoogle ScholarPubMed
Surova, O and Zhivotovsky, B (2013) Various modes of cell death induced by DNA damage. Oncogene 32, 37893797.CrossRefGoogle ScholarPubMed
Ganem, NJ et al. (2014) Cytokinesis failure triggers hippo tumor suppressor pathway activation. Cell 158, 833848.CrossRefGoogle ScholarPubMed
Hinchcliffe, EH et al. (2016) Chromosome missegregation during anaphase triggers p53 cell cycle arrest through histone H3.3 Ser31 phosphorylation. Nature Cell Biology 18, 668.CrossRefGoogle ScholarPubMed
Lambrus, BG et al. (2016) A USP28-53BP1-p53-p21 signaling axis arrests growth after centrosome loss or prolonged mitosis. Journal of Cell Biology 214, 143153.CrossRefGoogle ScholarPubMed
Meitinger, F et al. (2016) 53BP1 and USP28 mediate p53 activation and G1 arrest after centrosome loss or extended mitotic duration. Journal of Cell Biology 214, 155166.CrossRefGoogle ScholarPubMed
Mackenzie, KJ et al. (2017) cGAS surveillance of micronuclei links genome instability to innate immunity. Nature 548, 461465.CrossRefGoogle ScholarPubMed
Abend, M (2003) Reasons to reconsider the significance of apoptosis for cancer therapy. International Journal of Radiation Biology 79, 927941.CrossRefGoogle ScholarPubMed
Heylmann, D et al. (2021) Comparison of DNA repair and radiosensitivity of different blood cell populations. Scientific Reports 11, 2478.CrossRefGoogle ScholarPubMed
Carter, RJ et al. (2018) Complex DNA damage induced by high linear energy transfer alpha-particles and protons triggers a specific cellular DNA damage response. International Journal of Radiation Oncology Biology Physics 100, 776784.CrossRefGoogle ScholarPubMed
Goodhead, DT (2006) Energy deposition stochastics and track structure: what about the target? Radiation Protection Dosimetry 122, 315.CrossRefGoogle ScholarPubMed
Gudkov, AV and Komarova, EA (2003) The role of p53 in determining sensitivity to radiotherapy. Nature Reviews Cancer 3, 117129.CrossRefGoogle ScholarPubMed
Rodel, C et al. (2003) Spontaneous and radiation-induced apoptosis in colorectal carcinoma cells with different intrinsic radiosensitivities: survivin as a radioresistance factor. International Journal of Radiation Oncology Biology Physics 55, 13411347.CrossRefGoogle ScholarPubMed
Gewirtz, DA et al. (2016) Tumor cell recovery from senescence induced by radiation with PARP inhibition. Radiation Research 186, 327332.CrossRefGoogle ScholarPubMed
Chakradeo, S et al. (2015) Yet another function of p53 – the switch that determines whether radiation-induced autophagy will Be cytoprotective or nonprotective: implications for autophagy inhibition as a therapeutic strategy. Molecular Pharmacology 87, 803814.CrossRefGoogle ScholarPubMed
Suzuki, K et al. (2021) Combination therapy with lenvatinib and radiation significantly inhibits thyroid cancer growth by uptake of tyrosine kinase inhibitor. Experimental Cell Research 398, 112390.CrossRefGoogle ScholarPubMed
Kim, HK et al. (2021) Inhibitor of apoptosis protein Livin promotes tumor progression and chemoradioresistance in human anaplastic thyroid cancer. Oncology Reports 45, 1.CrossRefGoogle ScholarPubMed
Wang, S et al. (2020) Up-regulation of Tiam1 promotes the radioresistance of laryngeal squamous cell carcinoma through activation of the JNK/ATF-2 signaling pathway. Oncotargets and Therapy 13, 70657074.CrossRefGoogle ScholarPubMed
Kahler, J et al. (2020) Heterogeneous nuclear ribonucleoprotein K is overexpressed and contributes to radioresistance irrespective of HPV status in head and neck squamous cell carcinoma. International Journal of Molecular Medicine 46, 17331742.Google ScholarPubMed
Kashin, M et al. (2020) Gold nanoparticles enhance EGFR inhibition and irradiation effects in head and neck squamous carcinoma cells. Biomed Research International 2020, 1281645.CrossRefGoogle ScholarPubMed
Zhan, S and Ni, B (2021) hsa-miR-9-5p down-regulates HK2 and confers radiosensitivity to nasopharyngeal carcinoma. Technology in Cancer Research & Treatment 20, 1533033821997822.CrossRefGoogle ScholarPubMed
Nie, X et al. (2019) SALL4 induces radioresistance in nasopharyngeal carcinoma via the ATM/Chk2/p53 pathway. Cancer Medicine 8, 17791792.CrossRefGoogle ScholarPubMed
Wang, L et al. (2019) Proton versus photon radiation-induced cell death in head and neck cancer cells. Head & Neck 41, 4655.CrossRefGoogle ScholarPubMed
Liu, H et al. (2021) lncRNA CASC19 contributes to radioresistance of nasopharyngeal carcinoma by promoting autophagy via AMPK-mTOR pathway. International Journal of Molecular Sciences 22, 1407.Google ScholarPubMed
Cerniglia, GJ et al. (2012) Inhibition of autophagy as a strategy to augment radiosensitization by the dual phosphatidylinositol 3-kinase/mammalian target of rapamycin inhibitor NVP-BEZ235. Molecular Pharmacology 82, 12301240.CrossRefGoogle ScholarPubMed
Deville, SS et al. (2020) Keap1 inhibition sensitizes head and neck squamous cell carcinoma cells to ionizing radiation via impaired non-homologous end joining and induced autophagy. Cell Death & Disease 11, 887.CrossRefGoogle ScholarPubMed
Ricco, N et al. (2019) Mevalonate pathway activity as a determinant of radiation sensitivity in head and neck cancer. Molecular Oncology 13, 19271943.CrossRefGoogle ScholarPubMed
Frederick, BA et al. (2020) Combined EGFR1 and PARP1 inhibition enhances the effect of radiation in head and neck squamous cell carcinoma models. Radiation Research 194, 519531.CrossRefGoogle ScholarPubMed
Yu, XH et al. (2019) Radiation-promoted CDC6 protein stability contributes to radioresistance by regulating senescence and epithelial to mesenchymal transition. Oncogene 38, 549563.CrossRefGoogle ScholarPubMed
Yun, MY et al. (2018) Carbonyl reductase 1 is a new target to improve the effect of radiotherapy on head and neck squamous cell carcinoma. Journal of Experimental & Clinical Cancer Research 37, 264.CrossRefGoogle ScholarPubMed
Cheng, JJ et al. (2015) XRCC3 is a promising target to improve the radiotherapy effect of esophageal squamous cell carcinoma. Cancer Science 106, 16781686.CrossRefGoogle ScholarPubMed
Lv, P et al. (2014) Inhibition of protein phosphatase 2A with a small molecule LB100 radiosensitizes nasopharyngeal carcinoma xenografts by inducing mitotic catastrophe and blocking DNA damage repair. Oncotarget 5, 75127524.CrossRefGoogle ScholarPubMed
Zehr, LJ and Cooper, JS (2021) Mandible osteoradionecrosis. In StatPearls, Treasure Island (FL).Google Scholar
Dambrain, R (1993) [The pathogenesis of osteoradionecrosis]. Revue de Stomatologie et de Chirurgie Maxillo-Faciale 94, 140147.Google Scholar
Nakano, H and Shinohara, K (1994) X-ray-induced cell death: apoptosis and necrosis. Radiation Research 140, 19.CrossRefGoogle ScholarPubMed
Akagi, Y, Ito, K and Sawada, S (1993) Radiation-induced apoptosis and necrosis in Molt-4 cells: a study of dose-effect relationships and their modification. International Journal of Radiation Biology 64, 4756.CrossRefGoogle ScholarPubMed
Shankar, S et al. (2017) TALEN based HPV-E7 editing triggers necrotic cell death in cervical cancer cells. Scientific Reports 7, 5500.CrossRefGoogle ScholarPubMed
Jing, QC et al. (2019) Wnt3a promotes radioresistance via autophagy in squamous cell carcinoma of the head and neck. Journal of Cellular and Molecular Medicine 23, 47114722.CrossRefGoogle ScholarPubMed
Ryter, SW, Mizumura, K and Choi, AM (2014) The impact of autophagy on cell death modalities. International Journal of Cell Biology 2014, 502676.CrossRefGoogle ScholarPubMed
Mathew, R, Karantza-Wadsworth, V and White, E (2007) Role of autophagy in cancer. Nature Reviews Cancer 7, 961967.CrossRefGoogle ScholarPubMed
Han, MW et al. (2014) Autophagy inhibition can overcome radioresistance in breast cancer cells through suppression of TAK1 activation. Anticancer Research 34, 14491455.Google ScholarPubMed
Shu, CW et al. (2016) RelA-mediated BECN1 expression is required for reactive oxygen species-induced autophagy in oral cancer cells exposed to low-power laser irradiation. PLoS ONE 11, e0160586.CrossRefGoogle ScholarPubMed
Nikoletopoulou, V et al. (2013) Crosstalk between apoptosis, necrosis and autophagy. Biochimica Et Biophysica Acta-Molecular Cell Research 1833, 34483459.CrossRefGoogle ScholarPubMed
Chang, J et al. (2016) Clearance of senescent cells by ABT263 rejuvenates aged hematopoietic stem cells in mice. Nature Medicine 22, 7883.CrossRefGoogle ScholarPubMed
Milanovic, M et al. (2018) Senescence-associated reprogramming promotes cancer stemness. Nature 553, 96100.CrossRefGoogle ScholarPubMed
Kim, YH et al. (2017) Senescent tumor cells lead the collective invasion in thyroid cancer. Nature Communications 8, 15208.CrossRefGoogle ScholarPubMed
Wells, SI et al. (2000) Papillomavirus E2 induces senescence in HPV-positive cells via pRB- and p21(CIP)-dependent pathways. Embo Journal 19, 57625771.CrossRefGoogle ScholarPubMed
Fitzgerald, AL et al. (2015) Reactive oxygen species and p21Waf1/Cip1 are both essential for p53-mediated senescence of head and neck cancer cells. Cell Death & Disease 6, e1678.CrossRefGoogle ScholarPubMed
Skinner, HD et al. (2012) TP53 disruptive mutations lead to head and neck cancer treatment failure through inhibition of radiation-induced senescence. Clinical Cancer Research 18, 290300.CrossRefGoogle ScholarPubMed
Gottgens, EL et al. (2019) Inhibition of CDK4/CDK6 enhances radiosensitivity of HPV negative head and neck squamous cell carcinomas. International Journal of Radiation Oncology Biology Physics 105, 548558.CrossRefGoogle ScholarPubMed
Hafsi, H et al. (2018) Combined ATR and DNA-PK inhibition radiosensitizes tumor cells independently of their p53 status. Frontiers in Oncology 8, 245.CrossRefGoogle ScholarPubMed
Nikjoo, H et al. (2001) Computational approach for determining the spectrum of DNA damage induced by ionizing radiation. Radiation Research 156, 577583.CrossRefGoogle ScholarPubMed
Watanabe, R, Rahmanian, S and Nikjoo, H (2015) Spectrum of radiation-induced clustered non-DSB damage – a Monte Carlo track structure modeling and calculations. Radiation Research 183, 525540.CrossRefGoogle Scholar
Magnander, K et al. (2010) Clustered DNA damage in irradiated human diploid fibroblasts: influence of chromatin organization. Radiation Research 173, 272282.CrossRefGoogle ScholarPubMed
Gollapalle, E et al. (2007) Detection of oxidative clustered DNA lesions in X-irradiated mouse skin tissues and human MCF-7 breast cancer cells. Radiation Research 167, 207216.CrossRefGoogle ScholarPubMed
Carter, RJ et al. (2019) Characterisation of deubiquitylating enzymes in the cellular response to high-LET ionizing radiation and complex DNA damage. International Journal of Radiation Oncology Biology Physics 104, 656665.CrossRefGoogle ScholarPubMed
Nickson, CM et al. (2017) Misregulation of DNA damage repair pathways in HPV-positive head and neck squamous cell carcinoma contributes to cellular radiosensitivity. Oncotarget 8, 2996329975.CrossRefGoogle ScholarPubMed
Bertrand, G et al. (2014) Targeting head and neck cancer stem cells to overcome resistance to photon and carbon ion radiation. Stem Cell Reviews and Reports 10, 114126.CrossRefGoogle ScholarPubMed
Nickson, CM et al. (2021) USP9X is required to maintain cell survival in response to high-LET radiation. Frontiers in Oncology 11, 671431.CrossRefGoogle ScholarPubMed
Asakawa, I et al. (2002) Radiation-induced growth inhibition in transplanted human tongue carcinomas with different p53 gene status. Anticancer Research 22, 20372043.Google ScholarPubMed
Takahashi, A et al. (2003) p53-dependent hyperthermic enhancement of tumour growth inhibition by X-ray or carbon-ion beam irradiation. International Journal of Hyperthermia 19, 145153.CrossRefGoogle ScholarPubMed
Hartfiel, S et al. (2019) Differential response of esophageal cancer cells to particle irradiation. Radiation Oncology 14, 119.CrossRefGoogle ScholarPubMed
Maalouf, M et al. (2009) Different mechanisms of cell death in radiosensitive and radioresistant p53 mutated head and neck squamous cell carcinoma cell lines exposed to carbon ions and x-rays. International Journal of Radiation Oncology Biology Physics 74, 200209.CrossRefGoogle ScholarPubMed
Ambrosini, G, Adida, C and Altieri, DC (1997) A novel anti-apoptosis gene, survivin, expressed in cancer and lymphoma. Nature Medicine 3, 917921.CrossRefGoogle ScholarPubMed
Frassanito, MA et al. (2019) Survivin overexpression in head and neck squamous cell carcinomas as a new therapeutic target. Oncology Reports 41, 26152624.Google ScholarPubMed
Santarelli, A et al. (2018) Survivin-based treatment strategies for squamous cell carcinoma. International Journal of Molecular Sciences 19, 971.CrossRefGoogle ScholarPubMed
Yoon, TM et al. (2014) Expression of Livin and the inhibition of tumor progression by Livin silencing in laryngohypopharyngeal cancer. In Vivo 28, 751759.Google ScholarPubMed
Guy, JB et al. (2021) Combining radiation to EGFR and Bcl-2 blockade: a new approach to target cancer stem cells in head and neck squamous cell carcinoma. Journal of Cancer Research and Clinical Oncology 147, 19051916.CrossRefGoogle ScholarPubMed
Wei, F et al. (2018) BPIFB1 (LPLUNC1) inhibits radioresistance in nasopharyngeal carcinoma by inhibiting VTN expression. Cell Death & Disease 9, 432.CrossRefGoogle ScholarPubMed
Jiang, F et al. (2016) Slug inhibition increases radiosensitivity of oral squamous cell carcinoma cells by upregulating PUMA. International Journal of Oncology 49, 709719.CrossRefGoogle ScholarPubMed
Zerp, SF et al. (2015) Targeting anti-apoptotic Bcl-2 by AT-101 to increase radiation efficacy: data from in vitro and clinical pharmacokinetic studies in head and neck cancer. Radiation Oncology 10, 158.CrossRefGoogle ScholarPubMed
Pitak-Arnnop, P et al. (2008) Management of osteoradionecrosis of the jaws: an analysis of evidence. European Journal of Surgical Oncology 34, 11231134.CrossRefGoogle ScholarPubMed
Madrid, C, Abarca, M and Bouferrache, K (2010) Osteoradionecrosis: an update. Oral Oncology 46, 471474.CrossRefGoogle ScholarPubMed
Riesterer, O et al. (2011) Combination of anti-IGF-1R antibody A12 and ionizing radiation in upper respiratory tract cancers. International Journal of Radiation Oncology Biology Physics 79, 11791187.CrossRefGoogle ScholarPubMed
Uzunparmak, B et al. (2020) Caspase-8 loss radiosensitizes head and neck squamous cell carcinoma to SMAC mimetic-induced necroptosis. JCI Insight 5, e139837.CrossRefGoogle ScholarPubMed
Eytan, DF et al. (2016) SMAC mimetic birinapant plus radiation eradicates human head and neck cancers with genomic amplifications of cell death genes FADD and BIRC2. Cancer Research 76, 54425454.CrossRefGoogle ScholarPubMed
Wanigasooriya, K et al. (2020) Radiosensitising cancer using phosphatidylinositol-3-kinase (PI3K). protein kinase B (AKT) or mammalian target of rapamycin (mTOR) inhibitors. Cancers (Basel) 12, 1278.CrossRefGoogle ScholarPubMed
Lee, M et al. (2021) Epigenetic regulation of p62/SQSTM1 overcomes the radioresistance of head and neck cancer cells via autophagy-dependent senescence induction. Cell Death & Disease 12, 250.CrossRefGoogle ScholarPubMed
Chuang, HC et al. (2014) The p53-reactivating small molecule RITA induces senescence in head and neck cancer cells. PLoS ONE 9, e104821.CrossRefGoogle ScholarPubMed
Nam, HY et al. (2013) Radioresistant cancer cells can be conditioned to enter senescence by mTOR inhibition. Cancer Research 73, 42674277.CrossRefGoogle ScholarPubMed
Joukov, V and De Nicolo, A (2018) Aurora-PLK1 cascades as key signaling modules in the regulation of mitosis. Science Signaling 11, eaar4195.CrossRefGoogle ScholarPubMed
Bu, YQ et al. (2008) Silencing of polo-like kinase (Plk) 1 via siRNA causes inhibition of growth and induction of apoptosis in human esophageal cancer cells. Oncology 74, 198206.CrossRefGoogle ScholarPubMed
Shi, W et al. (2010) Significance of Plk1 regulation by miR-100 in human nasopharyngeal cancer. International Journal of Cancer 126, 20362048.Google ScholarPubMed
Li, JJ et al. (2015) Co-inhibition of polo-like kinase 1 and aurora kinases promotes mitotic catastrophe. Oncotarget 6, 93279340.CrossRefGoogle ScholarPubMed
Libertini, S et al. (2011) AZD1152 negatively affects the growth of anaplastic thyroid carcinoma cells and enhances the effects of oncolytic virus dl922-947. Endocrine-Related Cancer 18, 129141.CrossRefGoogle ScholarPubMed
Lee, JW et al. (2019) Combined Aurora kinase A (AURKA) and WEE1 inhibition demonstrates synergistic antitumor effect in squamous cell carcinoma of the head and neck. Clinical Cancer Research 25, 34303442.CrossRefGoogle ScholarPubMed
Mak, JPY et al. (2015) Pharmacological inactivation of CHK1 and WEE1 induces mitotic catastrophe in nasopharyngeal carcinoma cells. Oncotarget 6, 2107421084.CrossRefGoogle ScholarPubMed
Patel, P et al. (2019) Enhancing direct cytotoxicity and response to immune checkpoint blockade following ionizing radiation with Wee1 kinase inhibition. Oncoimmunology 8, e1638207.CrossRefGoogle ScholarPubMed
Deneka, AY et al. (2020) Synthetic lethal targeting of mitotic checkpoints in HPV-negative head and neck cancer. Cancers 12, 306.CrossRefGoogle ScholarPubMed
Figure 0

Fig. 1. Depth-dose distribution of x-rays (photons) versus protons and relationship to LET leading to DNA damage. (a) Comparison of the dose delivered related to depth in tissue of photons versus protons. Proton irradiation, unlike photons, leads to targeted delivery of the radiation dose to the tumour thus minimising associated normal tissue irradiation, but which leads to associated increases in LET at and around the Bragg peak. (b) Tracks of IR of different LET and their interaction with DNA. Ionisation events (red dots) can occur indirectly (predominant with low-LET radiation) or directly (particularly with high-LET radiation) leading to DNA damage in the form of strand breaks and base damage (orange and green stars, respectively). The low-LET radiation tracks generate largely isolated DNA damage, whereas the densely ionising tracks of high-LET radiation lead to significant levels and formation of CDD.

Figure 1

Fig. 2. Cell death pathways responsive to IR. Depending on the level of DNA damage and cell type, one of the pathways including apoptosis, necrosis, autophagy, senescence and mitotic death will be initiated. The key steps and proteins involved in coordinating these pathways are shown. If the cell undergoes an initiation cell death pathway (senescence and mitotic catastrophe), then an executive pathway (apoptosis, necrosis and autophagy) will follow eventually.

Figure 2

Fig. 3. The extrinsic and intrinsic pathways of apoptosis. The extrinsic death receptor pathway is activated by death receptor ligands, including FasL, TNF-α or TRAIL, which in turn activates caspase 8 and downstream executing caspases. The intrinsic death receptor pathway is initiated by several intracellular stresses, leading to activation of Bax and Bak on the mitochondrial membrane and which result in the release of cytochrome c from the mitochondria. Cytoplasmic cytochrome c activates caspase 9 and downstream executing caspases.

Figure 3

Fig. 4. The mechanisms of autophagy. Schematic representation of the three main autophagy pathways: macro-, micro- and chaperone-mediated autophagy. Macroautophagy sequesters cytosolic cargo inside a phagophore formed by specific ATG proteins and lipids. The membrane then seals into an autophagosome and fuses with lysosomes causing the degradation of the trapped cargo. Microautophagy entraps cytosolic cargo in small vesicles formed by invagination of the lysosomal membrane. Chaperone-mediated autophagy involves the selective degradation of KFRQ-like motif-bearing proteins delivered to the lysosomes via chaperone HSC70 and their internalisation in lysosomes via the receptor lysosome-associated membrane protein type 2A (LAMP2A).

Figure 4

Table 1. Biological effects of low-LET radiation on head and neck cancer cells

Figure 5

Table 2. Biological effects of high-LET radiation on head and neck cancer cells