Molecular genetic signatures of head and neck squamous cell carcinoma and their changes induced by proton irradiation
- Authors: Jumaniyazova E.D.1, Lokhonina A.V.1,2, Sentyabreva A.V.1,3, Kosyreva A.M.1,3
-
Affiliations:
- RUDN University
- National Medical Research Center of Obstetrics, Gynecology and Perinatology named after Academician V.I. Kulakov
- Avtsyn Research Institute of Human Morphology of Petrovsky National Research Centre of Surgery
- Issue: Vol 28, No 4 (2024): ONCOLOGY
- Pages: 413-426
- Section: ONCOLOGY
- URL: https://journals.rudn.ru/medicine/article/view/42007
- DOI: https://doi.org/10.22363/2313-0245-2024-28-4-413-426
- EDN: https://elibrary.ru/GKJBSB
Cite item
Full Text
Abstract
Head and neck squamous cell cancer (HNSCC) is the seventh most common malignancy in the world. The overall incidence of HNSCC is increasing and is projected to increase by about 30 % annually by 2030. Clinically, HNSCC is characterized by an aggressive course: rapid local spread, resistance to various methods of antitumor treatment, and frequent recurrences. Despite improvements in diagnostic and therapeutic approaches over the last two decades, mainly due to the respective heterogeneity of these tumors, the outcomes of patients with HNSCC have not shown significant improvements, especially for patients with late TNM stage, with an overall five-year survival rate of 50 %. Approximately 75 % of HNSCC patients are treated with radiation therapy either alone or as part of a comprehensive treatment regimen. To date, one of the main ways to improve the efficacy of radiation therapy in HNSCC is considered to be a combination of maximum allowable increase of radiation dose in the target tumor and reduction and minimization of such dose in the surrounding healthy tissues. From this point of view, proton therapy (PT) has a pronounced advantage over various types of photon irradiation. Despite the growing interest of scientists in PT, studies aimed at identifying molecular and genetic changes induced by PT are scarce, while in our opinion they are very important for understanding intracellular mechanisms leading either to tumor cell destruction or to the development of radioresistance. This review summarizes the available knowledge on the changes in the main signaling pathways of HNSCC tumor cells under the influence of PT.
Full Text
Introduction
Head and neck squamous cell cancer (HNSCC) is the seventh most common malignancy in the world. The overall incidence of HNSCC is increasing and is projected to increase by about 30 % annually by 2030 [1, 2]. The most significant risk factors for the development of HNSCC include smoking, alcohol consumption, exposure to environmental pollutants, and infection with viral agents, namely human papillomavirus (HPV) and Epstein-Barr virus [3], additional predisposing factors include betel nut chewing (a species of tree-like plants of the genus Areca of the Palm family, the use of which is common in Southeast Asian countries), malnutrition, poor oral hygiene [4, 5]. Clinically, HNSCC is characterized by an aggressive course: rapid local spread, resistance to various methods of antitumor treatment and frequent recurrences [6]. Despite improvements in diagnostic and therapeutic approaches over the last two decades, mainly due to the respective heterogeneity of these tumors, the outcomes of patients with HNSCC have not shown significant improvements, especially for patients with late TNM stage, with an overall five-year survival (OS) of 50 % [7]. Therapeutic options for HNSCC include minimally invasive, organ-preserving surgery, radiation therapy (RT), and multimodal treatment strategies. For patients with early-stage HNSCC, both surgery and intensive RT provide comparable results in terms of local disease control and overall survival [8]. After surgery, postoperative RT with or without adjuvant chemotherapy is recommended for patients with risk factors including perineural invasion and/or lymphovascular invasion and when positive resection margins (i. e., resection margins with tumor cells detected in them) are identified. As a rule, a combination of surgery, RT and chemotherapy is required at advanced stages (locally advanced stage, or in the presence of distant metastases) [9]. Approximately 75 % of HNSCC patients are treated with RT as the main (independent) or as part of complex treatment [10]. Thus, in the early stages of the disease, RT can replace the need for surgical intervention. In some complex clinical situations, for example, in tumor lesions of the larynx, RT allows to perform antitumor treatment while preserving the organ, which is a fundamentally important aspect from the psychological point of view for a number of patients [11]. However, the planning and implementation of RT in patients with HNSCC is complicated due to the close proximity of a large number of critical organs at risk (OAR). Even with the introduction of Intensity Modulated Radiation Therapy (IMRT) in the clinic, it is not possible to completely avoid irradiation of nearby organs, which subsequently leads to the development of late postradiation complications [12, 13]. To date, one of the main ways to improve the efficacy of RT in HNSCC is considered to be a combination of maximum permissible increase of radiation dose in the target tumor and reduction and minimization of such dose in the surrounding healthy tissues. From this point of view, proton therapy (PT) has a pronounced advantage over various types of photon irradiation. [14, 15]. A number of studies have demonstrated a significant reduction in the radiation dose to OAR with PT compared to IMRT [16–18].
PT is a promising variant of RT, the wide application of which is expected to solve many problems [19]. Protons are positively charged particles that penetrate tissue to a limited depth and give up most of their energy at the end of their path [20]. This physical phenomenon of protons has been called the Bragg peak [21]. The pronounced peak of ionizing radiation, or Bragg peak, occurring at the end of the protons’ run through matter causes that the integral dose is almost always lower and the irradiation of healthy tissues is less than in photon therapy [22]. Due to such a dose distribution in the substance, it is possible to carry out irradiation with high effective doses against the background of a reduced radiation load on the surrounding healthy tissues, which improves the tolerability of treatment and reduces the number of postradiation complications in HNSCC patients.
The main effect of protons on tumor cells is DNA damage, including nucleotide base modifications, a basic sites and single-strand breaks, the latter being the most common type of PT-induced direct damage. In addition to direct damage, PT has an indirect cytotoxic effect through the formation of reactive oxygen species (ROS) [23], which activate caspases to induce apoptosis [24], (Fig.1). In a past review article, we highlighted the biological effects of PT [25], (Table 1).
Figure 1. Proton therapy effects on cancer cells
Table 1. Specific features of proton therapy exposure
Evaluation parameters | Peculiarities of proton therapy | |
Physical properties | Protons emit maximum energy, reaching the “target”, i. e. the tumor; while the surrounding tissues (healthy) receive minimal radiation dose [23], [26]. | |
Action on tumor | Cancer cell DNA | Proton irradiation induces clustered DNA damage, with the formation of short DNA fragments that are difficult to repair by the cellular repair mechanism, leading to high cell death [27]. |
Cancer cells as a whole | Proton irradiation leads to the accumulation of ROS, which in turn activate caspases that trigger cancer cell apoptosis [28]. It reduces invasion and migration of cancer cells by inhibiting integrins and matrix metalloproteinases (MMPs) [29]. | |
Tumor microenvironment | Cancer-associated fibroblasts | It reduces protumorigenic properties and induces rapid senescence of сancer-associated fibroblasts [30]. |
Macrophages | Proton irradiation stimulates reprogramming of M2 macrophages possessing a pro-tumor phenotype into M1 antitumor ones through activation of NFκB, MAPK and IRF/STAT [31]. It activates the expression of high mobility group box 1 (HMGB1), which is responsible for the activation of antigen-presenting cells [32]. | |
T-lymphocytes | Proton irradiation activates the recruitment of CD8+ [33], CD4+ and T-reg lymphocytes [32]. |
Despite the growing interest of scientists in PT, studies aimed at identifying molecular and genetic changes induced by PT are scarce, whereas in our opinion they are very important for understanding the intracellular mechanisms leading to either tumor cell destruction or the development of radioresistance [34]. In this review, we summarize the available knowledge of the changes in the main signaling pathways of HNSCC tumor cells under the influence of PT.
P53 signaling pathway
HNSCC is characterized by a high level of genetic instability [35], which is primarily due to inactivating mutations in tumor growth suppressor genes [36]. Like most solid tumors, HNSCC is characterized by mutations in the TP53 gene, [37] which in “normal” mode of operation provides genomic stability and in case of detecting violations is able to stop the cell cycle and start the process of DNA repair, and in case of impossibility to correct the damage — starts apoptosis [38, 39]. The hallmark of p53 is high sensitivity to DNA damage or oncogene activation in the cell [40]. Inactivating mutations of p53 are characteristic of more than 80 % of HPV-negative HNSCC, which necessarily lead to loss of its function [41]. Inactivating mutations in the TP53 gene are events of early stages of carcinogenesis. The presence of p53 mutations in any subtypes of HNSCC is associated with poor overall survival, resistance to therapy, and increased recurrence rates [41]. Talking about RT-induced changes in the TP53 gene, it is important to note that it is assigned one of the main roles in deciding the “fate” of cancer cells after irradiation. In several studies [42, 43, 44] it has been demonstrated that under the influence of RT, tumor cells die by apoptosis rather than by necrosis or autophagy. Radiation exposure increases the amount of p53 protein in cells, mainly by stimulating its translation and inhibiting its degradation [45].
Activation of the p53‑mediated signaling pathway can cause cell cycle arrest followed by DNA repair, which promotes cell survival; if DNA repair is not possible, apoptosis is induced or the cell becomes senescent, which ultimately leads to tumor cell death [44]. Bravata et al. showed on breast cancer cell lines that proton irradiation leads to TP53 pathway activation [46]. Lee et al. demonstrated on the example of three kinds of cancer cells, lewis lung carcinoma cells, hepatoma HepG2 and Molt‑4 leukemia cells, that proton irradiation induces an increase in p53 expression with subsequent apoptosis of cancer cells [47]. Taking into the consideration that HNSCC is characterized by a high level of hypoxia (like the above tumors) in the tumor node, similar activation of p53‑dependent signaling pathway is likely to occur in this MNs. In addition, proton irradiation activates caspases and increases the generation of ROS via p53. The increase of ROS can activate p53 and vice versa according to the feedback principle [43].
EGFR signaling pathway
A well-known molecular genetic feature of HNSCC is the overexpression of a receptor with tyrosine kinase activity-epidermal growth factor receptor (EGFR) [48]. Thus, EGFR overexpression is thought to be found in approximately 80 % of HNSCC cases, and in terms of disease course, it is associated with a poor prognosis of the disease [49]. The prevalence of EGFR overexpression has led to the introduction of Cetuximab, a targeting drug that is a monoclonal antibody directed against EGFR, into the antitumor therapy of HNSCC.
HER family ligands including epidermal growth factor, heparin-binding, amphiregulin, transforming growth factor-alpha, epiregulin and beta-cellulin have affinity for EGFR. [50]. Upon binding to one of the ligands, activated EGFR activates various intracellular signaling cascades, e. g., JAK/STAT, PI3K/AKT, MAPK [51]. In cancer cells, these signaling cascades are responsible for the processes of cell proliferation, invasion, migration and metastasis [52].
Despite its transmembrane position, EGFR is able to move into the cell nucleus where it functions as a transcription factor. Such a phenomenon can be induced by ionizing radiation and it is associated with the acquisition of resistance to RT by cancer cells. In turn, EGFR inhibition sensitizes radioresistant cancer cells by modulating DNA repair. Overexpression of other receptor tyrosine kinases including HER2 and MET contribute to resistance to agents targeting EGFR [53, 54].
A study by Park et al. demonstrated the efficacy of combining the EGFR inhibitor Gefinitib with proton irradiation on non-small cell lung cancer cells [55]. Unusual results were obtained when oral squamous cell carcinoma’s cells were irradiated with protons: EGFR suppression was revealed [56]. Irradiation with another type of corpuscular irradiation, carbon ions, also led to a decrease in the activity of EGFR and PI3K/AKT/mTOR pathways [57]. In contrast, Stahler et al. did not observe activation of EGFR and downstream targets AKT and ERK1/2 after carbon ion irradiation in their experiment [58].
MET signaling pathway
The MET proto-oncogene encodes RTK and is a regulator of one of the most important signaling cascades of carcinogenesis, primarily causing epithelial-mesenchymal transition (EMT) [59]. In turn, EMT enhances cancer cell migration and invasion and thereby determines the process of metastasis in HNSCC [54]. Although c-MET has several functional domains, it binds to a single ligand-hepatocyte growth factor (HGF) [60]. The binding of HGF to c-MET iniects the catalytic activity of RTC, which activates other cell signaling pathways such as RAS/RAF/ERK, PI3K/AKT/mTOR, JAK/STAT and NOTCH, resulting in uncontrolled proliferation of cancer cells [61]. In HNSCC, c-MET mutations are infrequent, occurring in approximately 2–13 % of cases, whereas MET copy number increase and overexpression of its ligand HGF are common [62] and are associated with poor prognosis and poor overall survival [63]. Increased expression of c-MET is associated with metastasis to lymph nodes in HNSCC, and overexpression of HGF is associated with resistance to anoikis (a type of cell death) [64]. Activation of c-Met promotes increased expression of plexin containing domain 2 through activation of ERK1/2-ELK1 signaling. This leads to cancer cell plasticity through the induction of epithelial-mesenchymal transition and an increase in the number of stem cells in the tumor, resulting in RT resistance of HNSCC. Inactivation of c-Met by knockdown or an inhibitory pharmacological agent not only reverses the EMT process, but also diminishes the CD44+CD133- CSC population in radioresistant HNSCC cells, which significantly slows tumor progression [65].
In view of the fact that EGFR and c-MET activate common downstream components of signaling cascades such as MAPK and PI3K/Akt/mTOR, the MET signaling cascade may be considered as a promising target for the treatment of HNSCC in cetuximab-resistant patients [54]. In addition, this signaling pathway is believed to interact with other signaling pathways such as STAT and Wnt, contributing to tumor progression and resistance to antitumor agents and RT [66]. In writing this review, we did not find any studies on how this signaling cascade is altered after proton irradiation. However, there are reports indicating that c-Met is overexpressed in most HPV-negative HNSCC cell lines after ionizing irradiation.
PI3K/Akt/mTOR signaling pathway
Disorders in the PI3K/AKT/mTOR signaling cascade in HNSCC are common, occurring in 90 % of cases [67]. The first constituent member of the signaling cascade-PI3K, which belongs to a class of enzymes that are essential for cell growth, differentiation, and survival-is activated by RTKs (e. g., as mentioned above, such as EGFR). Other members of the pathway include mTORC1, mTORC2 and Akt. mTORC2 is required for phosphorylation of Akt and activation of other signaling molecules of the PI3K pathway [68]. Phosphorylated Akt activates mTOR or inhibits Bad, caspase 9 and other proteins, thereby regulating cell proliferation, differentiation, apoptosis and migration. PI3K is thought to phosphorylate phosphatidylinositol‑4,5‑bisphosphate and convert it to another form, phosphatidylinositol‑3, 4, 5‑triphosphate [69]. Phosphatidylinositol‑3, 4, 5‑triphosphate can be dephosphorylated by phosphatase and tensin homolog, which in turn blocks the PI3K/AKT/mTOR pathway [70, 71].
Activation of this signaling cascade in HNSCC cancer cells leads to the development of resistance to antitumor therapies, especially RT [72], which is associated with the induction of DNA repair [73]. After PT, there is a decrease in AKT phosphorylation followed by inhibition of signaling pathways, leading to decreased radioresistance [74]. Carbon ion irradiation resulted in decreased PI3K/AKT/mTOR activity in non-small cell lung cancer cells [57]. The study conducted on the cell line of glioblastoma multiforme U87 showed that proton irradiation of tumor cells under conditions of acute hypoxia leads to activation of the PI3K/AKT/mTOR signaling pathway [75].
JAK/STAT signaling pathway
Another signaling pathway associated with the malignancy of HNSCC is JAK/STAT. A transmembrane protein called Janus kinase (JAK) perceives information from outside the cell and directs the signal inside the cell by phosphorylating STAT, which is a transcription factor. Activation of this signaling cascade is associated with resistance to various anticancer treatment approaches-chemotherapy, RT, and targeted therapy [76, 77]. It is unusual that activation of the participants of this signaling cascade is associated not only with changes in cancer cells directly, but also in the tumor microenvironment. For example, STAT3 activation leads to changes in the tumor microenvironment: it acquires immunosuppressive potential due to increased synthesis of cytokines TGF-β1, IL‑6, IL‑10, and VEGF, which, in turn, ensures tumor escape from recognition and lysis by cytotoxic T-lymphocytes [78]. In vitro inhibition of STAT3 has been shown to be associated with a less pronounced development of immunosuppressive phenotype of HNSCC cells [79]. In contrast, STAT5 activation observed in PRGS correlates with increased cancer cell proliferation, invasion and EMT activity [80]. In our recent study on transcriptomic changes of HNSCC after PT, we observed suppression of STAT5 activity [81].
MAPK signaling pathway
The mitogen-activated protein kinase (MAPK) signaling cascade has an important regulatory function in cancer cells. Such processes as proliferation of cancer cells, their subsequent differentiation, metastasis, and in addition the processes of angiogenesis and resistance to antitumor treatment are caused by the activation of the MAPK signaling pathway [82]. The MAPK signaling pathway includes RAS (H/K/NRAS), RAF (A-/B-/C-RAF), mitogen-activated protein kinase (MEK, MEK1/2), extracellular signal regulated kinases (ERK, ERK1/2), adaptor molecules (GRB2, SHC1/2/3/4) and dual specificity phosphatases (DUSP3/5/6/7/9), which are negative regulators of ERK [83]. It has been demonstrated that activation of several kinases, including BRAF, KRAS, HRAS and ERK1/2, induces carcinogenesis and stimulates cancer cell invasion [84].
In HNSCC, mutations affecting members of the MAPK signaling pathway occur in about 18 % of cases [85]. These mutations are predominantly localized in BRAF, HRAS, KRAS, and ERK genes [86]; most of the mutations of the MAPK signaling pathway in HNSCC are inducers of carcinogenesis. High intratumoral expression of p-MAPK1/3 (p-ERK1/2) in HNSCC patients correlates with low survival rates [87].
Hyperexpression of one of the participants of the p38/MAPK signaling pathway, MAP2K6, in patients with HNSCC is associated with resistance to RT and poor prognosis of the disease [88]. In a study by Meerz et al., MAPK activation was detected in 3-D cell cultures of HNSCC after proton irradiation. In addition, the authors demonstrated radiosensitization of HNSCC cells when combining PT with selective inhibitors of key members of the MAPK pathway — for ERK1/2 (Ulixertinib); for JNK 1/2/3 (SP600125); for p38α/β/ɣ/δ (Ralimetinib). The most pronounced radiosensitization effect was demonstrated by a selective ERK1/2 inhibitor [89]. Another work devoted to the effects of proton irradiation on the cell line of colorectal cancer, which revealed the inhibitory effect of PT on MAPK phosphorylation in cancer cells, is also noteworthy [90].
The NOTCH signaling pathway
The NOTCH signaling cascade starts with 4 receptors (NOTCH1–4) to which 5 ligands (JAG1 and 2 and DLL1, 3 and 4) can bind. After ligand binding to the NOTCH receptor, the γ-secretase complex releases an intracellular NOTCH domain called NICD. NICD moves into the cell nucleus, which activates transcription of NOTCH target genes -HES and HEY [91]. A large-scale genomic analysis conducted in 2015 revealed that NOTCH 1–3 mutations are present in 17 % of HPV-positive and 26 % of HPV-negative HNSCC cases [41, 92].
An increase in the activity of NOTCH signaling pathway participants was observed in HNSCC cells, and their inhibition leads to a decrease in proliferation and invasion of cancer cells [91]. Activation of the signaling pathway through NOTCH1 by Wnt-signaling stabilizes the population of cancer stem cells (CSCs), which is associated with frequent recurrences and active metastasis. Hovinga et al. demonstrated that Notch inhibition significantly improves response to radiation by reducing proliferation and self-renewal of CSCs in tumor explants [93]. In addition, survival of CSCs is maintained by activation of Akt and STAT3, which are almost always activated in cancer cells. These together account for tumor radioresistance [94]. On human glioma cell lines it was shown that carbon ion irradiation suppresses Notch signaling at all levels of transcription and translation of proteins participating in this pathway [95]. Proton boron capture therapy in GBM cells induces a Notch signaling activation, able to regulate cell fate through the modulation of autophagy/apoptosis transition [96].
PDGF/PDGFR signaling pathway
The peculiarity of this signaling pathway is that simultaneous expression of platelet-derived growth factor (PDGF) and platelet derived growth factor receptor (PDGFR) is observed in cancer cells, which creates an autocrine loop that promotes aggressive tumor behavior. The PDGF ligand family includes several members: PDGFA, PDGFB, PDGFC, and PDGFD, which form homo- and heterodimers [97]. Upon ligand binding to the receptor, intracellular tyrosine kinases are activated and form multiple binding sites for downstream signaling molecules, thereby activating various signaling pathways such as PI3K/Akt/mTOR signaling cascades, MAPK signaling cascade, JAK/STAT and Notch signaling cascade [98].
The PDGF/PDGFR signaling pathway has been assigned a major role in tumor progression [99]. PDGF has been found to have a stimulatory effect on malignant cell transformation, cancer cell migration, and cancer cell survival [100, 98]. PDGF overexpression promotes tumor cell growth [101] and induces angiogenesis [102], affecting cells in the tumor microenvironment, thereby provoking tumor progression and dissemination [103, 104]. In addition, there is evidence that increased PDGF activity in tumors is associated with resistance to drug treatment, which is associated with impaired blood flow in tumors due to increased interstitial fluid pressure [105]. Aebersold et al. found that more than half of tumor samples from 95 patients with oropharyngeal cancer stained positive for PDGF-BB, and this was associated with a greater risk of metastasis [106]. After proton irradiation, suppression of this signaling pathway was observed in HNSCC cells [81].
PD‑1/PD-L1 signaling pathway
The PD‑1/PD-L1 signaling pathway controls the induction and maintenance of immune tolerance in the tumor microenvironment [107]. PD-L1 is expressed by tumor cells, it is a so-called “adaptive immune mechanism” to avoid antitumor response by inhibiting T cell activation, decreasing cytokine production and inducing T cell cytolysis [108, 109]. Primary tumors and metastases, particularly in HNSCC, and even different regions of the same tumor node can differ significantly in PD-L1 expression levels [110, 111, 112]. Data regarding the prognostic value of PD-L1 expression in HNSCC are conflicting in the literature. A meta-analysis evaluating PD-L1 expression and the association with survival in HNSCC patients, found no significant difference in overall survival between PD-L1‑positive and -negative patients [113]. Currently, five monoclonal antibodies targeting PD‑1 and PD-L1 have been approved by the FDA. Two of them target PD-L1, atezolizumab and durvalumab, and nivolumab, pembrolizumab and cemiplimab target PD‑1.
Proton irradiation of the KYSE450 (esophageal squamous cell carcinoma) cell line increased PD-L1 expression [114]. The combination of RT with immunotherapy for the treatment of HNSCC has long attracted the attention of researchers. When RT is combined with immune checkpoint inhibitors, it can potentiate the synergistic effects, where RT contributes to the normalization of the tumor vascular system, enhance the expression of leukocyte adhesion molecules on endothelial cells, and stimulate the secretion of chemokines that attract CD8+ T cells [115]. And a recent study revealed a synergistic effect of combining proton irradiation and immunotherapy on mouse oral cancer cell lines [116].
Conclusion
PT is a promising option for RT of HNSCC, minimizing the number of post-radiation complications and significantly improving the quality of life of patients undergoing antitumor treatment. However, radioresistance leads to treatment ineffectiveness in some cases. In the course of writing this review article, we encountered the fact that the described molecular-genetic signatures of this malignancy are not few, but they lack specificity, and the number of works devoted to the description of PT effects at the level of intracellular signaling pathways is limited. This determines the urgent need to search for molecular genetic biomarkers to predict response to PT. The understanding of molecular genetic changes induced by PT in tumor cells of HNSCC will allow to create effective combinations of antitumor therapies to avoid radioresistance.
About the authors
Enar D. Jumaniyazova
RUDN University
Author for correspondence.
Email: enar2017@yandex.ru
ORCID iD: 0000-0002-8226-0433
SPIN-code: 1780-5326
Moscow, Russian Federation
Anastasia V. Lokhonina
RUDN University; National Medical Research Center of Obstetrics, Gynecology and Perinatology named after Academician V.I. Kulakov
Email: enar2017@yandex.ru
ORCID iD: 0000-0001-8077-2307
SPIN-code: 4521-2250
Moscow, Russian Federation
Alexandra V. Sentyabreva
RUDN University; Avtsyn Research Institute of Human Morphology of Petrovsky National Research Centre of Surgery
Email: enar2017@yandex.ru
ORCID iD: 0000-0001-5064-219X
SPIN-code: 6966-9959
Moscow, Russian Federation
Anna M. Kosyreva
RUDN University; Avtsyn Research Institute of Human Morphology of Petrovsky National Research Centre of Surgery
Email: enar2017@yandex.ru
ORCID iD: 0000-0002-6182-1799
SPIN-code: 5421-5520
Moscow, Russian Federation
References
- Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2021;71(3):209–249. doi: 10.3322/caac.21660
- Gormley M, Creaney G, Schache A, Ingarfield K, Conway DI. Reviewing the epidemiology of head and neck cancer: definitions, trends and risk factors. Br Dent J. 2022;233(9):780–786. doi: 10.1038/s41415-022-5166-x
- IARC. List of Classifications by cancer sites with sufficient or limited evidence in humans. IARC Monograpghs; 2018. 14 p.
- Johnson DE, Burtness B, Leemans CR, Lui VWY, Bauman JE, Grandis JR. Head and neck squamous cell carcinoma. Nat Rev Dis Primers. 2020;6(1):92. doi: 10.1038/s41572-020-00224-3
- Sun Z, Sun X, Chen Z, Du J, Wu Y. Head and Neck Squamous Cell Carcinoma: Risk Factors, Molecular Alterations, Immunology and Peptide Vaccines. Int J Pept Res Ther. 2022;28(1):19. doi: 10.1007/s10989-021-10334-5.
- Georgopoulos R, Liu JC. Examination of the patient with head and neck cancer. Surg Oncol Clin N Am. 2015;24(3):409–21. doi: 10.1016/j.soc.2015.03.003
- Pulte D, Brenner H. Changes in survival in head and neck cancers in the late 20th and early 21st century: a period analysis. Oncologist. 2010;15(9):994–1001. doi: 10.1634/theoncologist.2009-0289
- Guan Z, Liu J, Zheng L. Effect of radiotherapy on head and neck cancer tissues in patients receiving radiotherapy: a bioinformatics analysis-based study. Sci Rep. 2024;14(1):6304. doi: 10.1038/s41598-024-56753-4
- Nissi L, Suilamo S, Kytö E, Vaittinen S, Irjala H, Minn H. Recurrence of head and neck squamous cell carcinoma in relation to high-risk treatment volume. Clin Transl Radiat Oncol. 2021;27:139–146. doi: 10.1016/j.ctro.2021.01.013
- Alfouzan AF. Radiation therapy in head and neck cancer. Saudi Med J. 2021;42(3):247–254. doi: 10.15537/smj.2021.42.3.20210660
- Borras JM, Barton M, Grau C, Corral J, Verhoeven R, Lemmens V, van Eycken L, Henau K, Primic-Zakelj M, Strojan P, Trojanowski M, Dyzmann-Sroka A, Kubiak A, Gasparotto C, Defourny N, Malicki J, Dunscombe P, Coffey M, Lievens Y. The impact of cancer incidence and stage on optimal utilization of radiotherapy: Methodology of a population based analysis by the ESTRO-HERO project. Radiother Oncol. 2015;116(1):45–50. doi: 10.1016/j.radonc.2015.04.021
- Nutting CM, Morden JP, Harrington KJ, Urbano TG, Bhide SA, Clark C, Miles EA, Miah AB, Newbold K, Tanay M, Adab F, Jefferies SJ, Scrase C, Yap BK, A’Hern RP, Sydenham MA, Emson M, Hall E; PARSPORT trial management group. Parotid-sparing intensity modulated versus conventional radiotherapy in head and neck cancer (PARSPORT): a phase 3 multicentre randomised controlled trial. Lancet Oncol. 2011;12(2):127–36. doi: 10.1016/S1470-2045(10)70290-4
- Rosenthal DI, Chambers MS, Fuller CD, Rebueno NC, Garcia J, Kies MS, Morrison WH, Ang KK, Garden AS. Beam path toxicities to non-target structures during intensity-modulated radiation therapy for head and neck cancer. Int J Radiat Oncol Biol Phys. 2008;72(3):747–55. doi: 10.1016/j.ijrobp.2008.01.012
- Newhauser W. Proton and Charged Particle Radiotherapy. Medical Physics. 2008;5(35). doi: 10.1118/1.2907963
- Beddok A, Vela A, Calugaru V, Tessonnier T, Kubes J, Dutheil P, Gerard A, Vidal M, Goudjil F, Florescu C, Kammerer E, Benezery K, Herault J, Poortmans P, Bourhis J, Thariat J; GORTEC, the 3 French proton centers. Proton therapy for head and neck squamous cell carcinomas: A review of the physical and clinical challenges. Radiother Oncol. 2020;147:30–39. doi: 10.1016/j.radonc.2020.03.006
- Taheri-Kadkhoda Z, Björk-Eriksson T, Nill S, Wilkens JJ, Oelfke U, Johansson KA, Huber PE, Münter MW. Intensity-modulated radiotherapy of nasopharyngeal carcinoma: a comparative treatment planning study of photons and protons. Radiat Oncol. 2008;3:4. doi: 10.1186/1748-717X-3-4
- Simone CB 2nd, Ly D, Dan TD, Ondos J, Ning H, Belard A, O’Connell J, Miller RW, Simone NL. Comparison of intensity-modulated radiotherapy, adaptive radiotherapy, proton radiotherapy, and adaptive proton radiotherapy for treatment of locally advanced head and neck cancer. Radiother Oncol. 2011;101(3):376–82. doi: 10.1016/j.radonc.2011.05.028
- van de Water TA, Bijl HP, Schilstra C, Pijls-Johannesma M, Langendijk JA. The potential benefit of radiotherapy with protons in head and neck cancer with respect to normal tissue sparing: a systematic review of literature. Oncologist. 2011;16(3):366–77. doi: 10.1634/theoncologist.2010-0171
- Nuyts S, Bollen H, Ng SP, Corry J, Eisbruch A, Mendenhall WM, Smee R, Strojan P, Ng WT, Ferlito A. Proton Therapy for Squamous Cell Carcinoma of the Head and Neck: Early Clinical Experience and Current Challenges. Cancers (Basel). 2022;14(11):2587. doi: 10.3390/cancers14112587
- Liu H, Chang JY. Proton therapy in clinical practice. Chin J Cancer. 2011;30(5):315–26. doi: 10.5732/cjc.010.10529
- Bragg WH, Kleeman R. On the ionization curves of radium. J Philos Mag. 1904;6:726–738. doi: 10.1080/14786440409463246
- Byun HK, Han MC, Yang K, Kim JS, Yoo GS, Koom WS, Kim YB. Physical and Biological Characteristics of Particle Therapy for Oncologists. Cancer Res Treat. 2021;53(3):611–620. doi: 10.4143/crt.2021.066
- Vitti ET, Parsons JL. The Radiobiological Effects of Proton Beam Therapy: Impact on DNA Damage and Repair. Cancers (Basel). 2019;11(7):946. doi: 10.3390/cancers11070946
- Alan Mitteer R, Wang Y, Shah J, Gordon S, Fager M, Butter PP, Jun Kim H, Guardiola-Salmeron C, Carabe-Fernandez A, Fan Y. Proton beam radiation induces DNA damage and cell apoptosis in glioma stem cells through reactive oxygen species. Sci Rep. 2015;5:13961. doi: 10.1038/srep13961
- Jumaniyazova E, Smyk D, Vishnyakova P, Fatkhudinov T, Gordon K. Photon- and Proton-Mediated Biological Effects: What Has Been Learned? Life (Basel). 2022;13(1):30. doi: 10.3390/life13010030
- Bernier J, Hall EJ, Giaccia A. Radiation oncology: a century of achievements. Nat Rev Cancer. 2004;4(9):737–47. doi: 10.1038/nrc1451
- Frame CM, Chen Y, Gagnon J, Yuan Y, Ma T, Dritschilo A, Pang D. Proton induced DNA double strand breaks at the Bragg peak: Evidence of enhanced LET effect. Front Oncol. 2022;12:930393. doi: 10.3389/fonc.2022.930393
- Miszczyk J, Rawojc K, Borkowska AM, Panek A, Swakon J, Galas A, Ahmed MM, Prasanna PGS. Therapeutic proton irradiation results in apoptosis and caspase‑3 activation in human peripheral blood lymphocytes. Transl Cancer Res 2018;7(4):879–889. doi: 10.21037/tcr.2018.06.14
- Ogata T, Teshima T, Kagawa K, Hishikawa Y, Takahashi Y, Kawaguchi A, Suzumoto Y, Nojima K, Furusawa Y, Matsuura N. Particle irradiation suppresses metastatic potential of cancer cells. Cancer Res. 2005;65(1):113–120. doi: 10.1158/0008-5472.113.65.1
- Grinde MT, Vik J, Camilio KA, Martinez-Zubiaurre I, Hellevik T. Ionizing radiation abrogates the pro-tumorigenic capacity of cancer-associated fibroblasts co-implanted in xenografts. Sci Rep. 2017;7:46714. doi: 10.1038/srep46714
- Genard G, Wera AC, Huart C, Le Calve B, Penninckx S, Fattaccioli A, Tabarrant T, Demazy C, Ninane N, Heuskin AC, Lucas S, Michiels C. Proton irradiation orchestrates macrophage reprogramming through NFκB signaling. Cell Death Dis. 2018;9(7):728. doi: 10.1038/s41419-018-0757-9
- Mirjolet C, Nicol A, Limagne E, Mura C, Richard C, Morgand V, Rousseau M, Boidot R, Ghiringhelli F, Noel G, Burckel H. Impact of proton therapy on antitumor immune response. Sci Rep. 2021;11(1):13444. doi: 10.1038/s41598-021-92942-1
- Lupu-Plesu M, Claren A, Martial S, N’Diaye PD, Lebrigand K, Pons N, Ambrosetti D, Peyrottes I, Feuillade J, Hérault J, Dufies M, Doyen J, Pagès G. Effects of proton versus photon irradiation on (lymph)angiogenic, inflammatory, proliferative and anti-tumor immune responses in head and neck squamous cell carcinoma. Oncogenesis. 2017;6(7): e354. doi: 10.1038/oncsis.2017.56
- Wang L, Fossati P, Paganetti H, Ma L, Gillison M, Myers JN, Hug E, Frank SJ. The Biological Basis for Enhanced Effects of Proton Radiation Therapy Relative to Photon Radiation Therapy for Head and Neck Squamous Cell Carcinoma. Int J Part Ther. 2021;8(1):3–13. doi: 10.14338/IJPT-20-00070.1
- Ha PK, Chang SS, Glazer CA, Califano JA, Sidransky D. Molecular techniques and genetic alterations in head and neck cancer. Oral Oncol. 2009;45(4–5):335–9. doi: 10.1016/j.oraloncology.2008.05.015
- Leemans CR, Snijders PJF, Brakenhoff RH. The molecular landscape of head and neck cancer. Nat Rev Cancer. 2018;18(5):269–282. doi: 10.1038/nrc.2018.11
- Sullivan KD, Galbraith MD, Andrysik Z, Espinosa JM. Mechanisms of transcriptional regulation by p53. Cell Death Differ. 2018;25(1):133–143. doi: 10.1038/cdd.2017.174
- Bykov VJN, Eriksson SE, Bianchi J, Wiman KG. Targeting mutant p53 for efficient cancer therapy. Nat Rev Cancer. 2018;18(2):89–102. doi: 10.1038/nrc.2017.109
- Kaiser AM, Attardi LD. Deconstructing networks of p53‑mediated tumor suppression in vivo. Cell Death Differ. 2018;25(1):93–103. doi: 10.1038/cdd.2017.171
- Aubrey BJ, Kelly GL, Janic A, Herold MJ, Strasser A. How does p53 induce apoptosis and how does this relate to p53‑mediated tumour suppression? Cell Death Differ. 2018;25(1):104–113. doi: 10.1038/cdd.2017.169
- Cancer Genome Atlas Network. Comprehensive genomic characterization of head and neck squamous cell carcinomas. Nature. 2015;517(7536):576–582. doi: 10.1038/nature14129
- Di Pietro C, Piro S, Tabbì G, Ragusa M, Di Pietro V, Zimmitti V, Cuda F, Anello M, Consoli U, Salinaro ET, Caruso M, Vancheri C, Crimi N, Sabini MG, Cirrone GA, Raffaele L, Privitera G, Pulvirenti A, Giugno R, Ferro A, Cuttone G, Lo Nigro S, Purrello R, Purrello F, Purrello M. Cellular and molecular effects of protons: apoptosis induction and potential implications for cancer therapy. Apoptosis. 2006;11(1):57–66. doi: 10.1007/s10495-005-3346-1
- Lee KB, Lee JS, Park JW, Huh TL, Lee YM. Low energy proton beam induces tumor cell apoptosis through reactive oxygen species and activation of caspases. Exp Mol Med. 2008;40(1):118–29. doi: 10.3858/emm.2008.40.1.118
- Chun SY, Nam KS, Lee KS. Proton Beam Induces P53‑mediated Cell Cycle Arrest in HepG2 Hepatocellular Carcinoma Cells. Biotechnol Bioproc. E. 2020;25;141–148. doi: 10.1007/s12257-019-0390-1
- Lee CL, Blum JM, Kirsch DG. Role of p53 in regulating tissue response to radiation by mechanisms independent of apoptosis. Transl. Cancer Res. 2013;2(5):412–421. doi: 10.3978/j.issn.2218-676X.2013.09.01
- Bravatà V, Cammarata FP, Minafra L, Pisciotta P, Scazzone C, Manti L, Savoca G, Petringa G, Cirrone GAP, Cuttone G, Gilardi MC, Forte GI, Russo G. Proton-irradiated breast cells: molecular points of view. J Radiat Res. 2019;60(4):451–465. doi: 10.1093/jrr/rrz032
- Lee KB, Kim KR, Huh TL, Lee YM. Proton induces apoptosis of hypoxic tumor cells by the p53‑dependent and p38/JNK MAPK signaling pathways. Int J Oncol. 2008;33(6):1247–56. doi: 10.3892/ijo_00000115.
- Chung CH, Germain A, Subramaniam RM, Heilmann AM, Fedorchak K, Ali SM, Miller VA, Palermo RA, Fakhry C. Genomic alterations in human epidermal growth factor receptor 2 (HER2/ERBB2) in head and neck squamous cell carcinoma. Head Neck. 2017;39(1): E15-E19. doi: 10.1002/hed.24587
- Solomon B, Young RJ, Rischin D. Head and neck squamous cell carcinoma: Genomics and emerging biomarkers for immunomodulatory cancer treatments. Semin Cancer Biol. 2018;52(2):228–240. doi: 10.1016/j.semcancer.2018.01.008
- Xu MJ, Johnson DE, Grandis JR. EGFR-targeted therapies in the post-genomic era. Cancer Metastasis Rev. 2017;36(3):463–473. doi: 10.1007/s10555-017-9687-8
- Byeon HK, Ku M, Yang J. Beyond EGFR inhibition: multilateral combat strategies to stop the progression of head and neck cancer. Exp Mol Med. 2019;51(1):1–14. doi: 10.1038/s12276-018-0202-2
- Sigismund S, Avanzato D, Lanzetti L. Emerging functions of the EGFR in cancer. Mol Oncol. 2018;12(1):3–20. doi: 10.1002/1878-0261.12155
- Alsahafi E, Begg K, Amelio I, Raulf N, Lucarelli P, Sauter T, Tavassoli M. Clinical update on head and neck cancer: molecular biology and ongoing challenges. Cell Death Dis. 2019;10(8):540. doi: 10.1038/s41419-019-1769-9
- Madoz-Gúrpide J, Zazo S, Chamizo C, Casado V, Caramés C, Gavín E, Cristóbal I, García-Foncillas J, Rojo F. Activation of MET pathway predicts poor outcome to cetuximab in patients with recurrent or metastatic head and neck cancer. J Transl Med. 2015;13:282. doi: 10.1186/s12967-015-0633-7
- Park HJ, Oh JS, Chang JW, Hwang SG, Kim JS. Proton Irradiation Sensitizes Radioresistant Non-small Cell Lung Cancer Cells by Modulating Epidermal Growth Factor Receptor-mediated DNA Repair. Anticancer Res. 2016;36(1):205–12
- Juvkam IS, Zlygosteva O, Sitarz M, Thiede B, Sørensen BS, Malinen E, Edin NJ, Søland TM, Galtung HK. Proton Compared to X–Irradiation Induces Different Protein Profiles in Oral Cancer Cells and Their Derived Extracellular Vesicles. Int J Mol Sci. 2023;24(23):16983. doi: 10.3390/ijms242316983
- Ogata T, Teshima T, Inaoka M, Minami K, Tsuchiya T, Isono M, Furusawa Y, Matsuura N. Carbon ion irradiation suppresses metastatic potential of human non-small cell lung cancer A549 cells through the phosphatidylinositol‑3‑kinase/Akt signaling pathway. J Radiat Res. 2011;52(3):374–9. doi: 10.1269/jrr.10102
- Stahler C, Roth J, Cordes N, Taucher-Scholz G, Mueller-Klieser W. Impact of carbon ion irradiation on epidermal growth factor receptor signaling and glioma cell migration in comparison to conventional photon irradiation. Int J Radiat Biol. 2013;89(6):454–61. doi: 10.3109/09553002.2013.766769
- Organ SL, Tsao MS. An overview of the c-MET signaling pathway. Ther Adv Med Oncol. 2011;3(1 Suppl): S7-S19. doi: 10.1177/1758834011422556
- Ma PC, Maulik G, Christensen J, Salgia R. c-Met: structure, functions and potential for therapeutic inhibition. Cancer Metastasis Rev. 2003;22(4):309–25. doi: 10.1023/a:1023768811842
- Khedkar HN, Chen LC, Kuo YC, Wu ATH, Huang HS. Multi-Omics Identification of Genetic Alterations in Head and Neck Squamous Cell Carcinoma and Therapeutic Efficacy of HNC018 as a Novel Multi-Target Agent for c-MET/STAT3/AKT Signaling Axis. Int J Mol Sci. 2023;24(12):10247. doi: 10.3390/ijms241210247
- Cho YA, Kim EK, Heo SJ, Cho BC, Kim HR, Chung JM, Yoon SO. Alteration status and prognostic value of MET in head and neck squamous cell carcinoma. J Cancer. 2016;7(15):2197–2206. doi: 10.7150/jca.16686
- Rothenberger NJ, Stabile LP. Hepatocyte Growth Factor/c-Met Signaling in Head and Neck Cancer and Implications for Treatment. Cancers (Basel). 2017;9(4):39. doi: 10.3390/cancers9040039
- Szturz P, Raymond E, Abitbol C, Albert S, de Gramont A, Faivre S. Understanding c-MET signalling in squamous cell carcinoma of the head & neck. Crit Rev Oncol Hematol. 2017;111:39–51. doi: 10.1016/j.critrevonc.2017.01.004
- Lang L, Chen F, Li Y, Shay C, Yang F, Dan H, Chen ZG, Saba NF, Teng Y. Adaptive c-Met-PLXDC2 Signaling Axis Mediates Cancer Stem Cell Plasticity to Confer Radioresistance-associated Aggressiveness in Head and Neck Cancer. Cancer Res Commun. 2023;3(4):659–671. doi: 10.1158/2767-9764.CRC-22-0289
- Liu D, Zhong M, Zhan D, Zhang Y, Liu S. Roles of the HGF/Met signaling in head and neck squamous cell carcinoma: Focus on tumor immunity (Review). Oncol Rep. 2020;44(6):2337–2344. doi: 10.3892/or.2020.7799
- Marquard FE, Jücker M. PI3K/AKT/mTOR signaling as a molecular target in head and neck cancer. Biochem Pharmacol. 2020;172:113729. doi: 10.1016/j.bcp.2019.113729
- Vander Broek R, Mohan S, Eytan DF, Chen Z, Van Waes C. The PI3K/Akt/mTOR axis in head and neck cancer: functions, aberrations, cross-talk, and therapies. Oral Dis. 2015;21(7):815–25. doi: 10.1111/odi.12206
- Engelman JA, Luo J, Cantley LC. The evolution of phosphatidylinositol 3‑kinases as regulators of growth and metabolism. Nat Rev Genet. 2006;7(8):606–19. doi: 10.1038/nrg1879
- Zhang P, Steinberg BM. Overexpression of PTEN/MMAC1 and decreased activation of Akt in human papillomavirus-infected laryngeal papillomas. Cancer Res. 2000;60(5):1457–62
- Maehama T, Dixon JE. The tumor suppressor, PTEN/MMAC1, dephosphorylates the lipid second messenger, phosphatidylinositol 3,4,5‑trisphosphate. J Biol Chem. 1998;273(22):13375–8. doi: 10.1074/jbc.273.22.13375
- Su YC, Lee WC, Wang CC, Yeh SA, Chen WH, Chen PJ. Targeting PI3K/AKT/mTOR Signaling Pathway as a Radiosensitization in Head and Neck Squamous Cell Carcinomas. Int J Mol Sci. 2022;23(24):15749. doi: 10.3390/ijms232415749
- Glorieux M, Dok R, Nuyts S. The influence of PI3K inhibition on the radiotherapy response of head and neck cancer cells. Sci Rep. 2020;10(1):16208. doi: 10.1038/s41598-020-73249-z
- Lee KS, Lee DH, Chun SY, Nam KS. Metastatic potential in MDA-MB‑231 human breast cancer cells is inhibited by proton beam irradiation via the Akt/nuclear factor-κB signaling pathway. Mol Med Rep. 2014;10(2):1007–12. doi: 10.3892/mmr.2014.2259
- Bravatà V, Tinganelli W, Cammarata FP, Minafra L, Calvaruso M, Sokol O, Petringa G, Cirrone GAP, Scifoni E, Forte GI, Russo G. Hypoxia Transcriptomic Modifications Induced by Proton Irradiation in U87 Glioblastoma Multiforme Cell Line. J Pers Med. 2021;11(4):308. doi: 10.3390/jpm11040308
- Geiger JL, Grandis JR, Bauman JE. The STAT3 pathway as a therapeutic target in head and neck cancer: Barriers and innovations. Oral Oncol. 2016;56:84–92. doi: 10.1016/j.oraloncology.2015.11.022
- Avalle L, Camporeale A, Morciano G, Caroccia N, Ghetti E, Orecchia V, Viavattene D, Giorgi C, Pinton P, Poli V. STAT3 localizes to the ER, acting as a gatekeeper for ER-mitochondrion Ca2+ fluxes and apoptotic responses. Cell Death Differ. 2019;26(5):932–942. doi: 10.1038/s41418-018-0171-y
- Bu LL, Yu GT, Wu L, Mao L, Deng WW, Liu JF, Kulkarni AB, Zhang WF, Zhang L, Sun ZJ. STAT3 Induces Immunosuppression by Upregulating PD‑1/PD-L1 in HNSCC. J Dent Res. 2017;96(9):1027–1034. doi: 10.1177/0022034517712435
- Albesiano E, Davis M, See AP, Han JE, Lim M, Pardoll DM, Kim Y. Immunologic consequences of signal transducers and activators of transcription 3 activation in human squamous cell carcinoma. Cancer Res. 2010;70(16):6467–76. doi: 10.1158/0008-5472.CAN-09-4058
- Koppikar P, Lui VW, Man D, Xi S, Chai RL, Nelson E, Tobey AB, Grandis JR. Constitutive activation of signal transducer and activator of transcription 5 contributes to tumor growth, epithelial-mesenchymal transition, and resistance to epidermal growth factor receptor targeting. Clin Cancer Res. 2008;14(23):7682–90. doi: 10.1158/1078-0432.CCR-08-1328
- Jumaniyazova ED, Vishnyakova PA, Chirkova MV, Karpulevich EA, Eremina IZ, Gordon KB, Kaprin AD, Fatkhudinov TH. Study of head and neck squamous cell carcinoma transcriptome after proton therapy. Bull Siberian Med. 2024;1(23):37–47. doi: 10.20538/1682-0363-2024-1-37-47
- Degirmenci U, Wang M, Hu J. Targeting Aberrant RAS/RAF/MEK/ERK Signaling for Cancer Therapy. Cells. 2020;9(1):198. doi: 10.3390/cells9010198
- Barbosa R, Acevedo LA, Marmorstein R. The MEK/ERK Network as a Therapeutic Target in Human Cancer. Mol Cancer Res. 2021;19(3):361–374. doi: 10.1158/1541-7786.MCR-20-0687
- Samatar AA, Poulikakos PI. Targeting RAS-ERK signalling in cancer: promises and challenges. Nat Rev Drug Discov. 2014;13(12):928–42. doi: 10.1038/nrd4281
- Ngan HL, Liu Y, Fong AY, Poon PHY, Yeung CK, Chan SSM, Lau A, Piao W, Li H, Tse JSW, Lo KW, Chan SM, Su YX, Chan JYK, Lau CW, Mills GB, Grandis JR, Lui VWY. MAPK pathway mutations in head and neck cancer affect immune microenvironments and ErbB3 signaling. Life Sci Alliance. 2020;3(6): e201900545. doi: 10.26508/lsa.201900545
- Zhang L, MacIsaac KD, Zhou T, Huang PY, Xin C, Dobson JR, Yu K, Chiang DY, Fan Y, Pelletier M, Wang Y, Jaeger S, Krishnamurthy Radhakrishnan V, JeBailey L, Skewes-Cox P, Zhang J, Fang W, Huang Y, Zhao H, Zhao Y, Li E, Peng B, Huang A, Dranoff G, Hammerman PS, Engelman J, Bitter H, Zeng YX, Yao Y. Genomic Analysis of Nasopharyngeal Carcinoma Reveals TME-Based Subtypes. Mol Cancer Res. 2017;15(12):1722–1732. doi: 10.1158/1541-7786.MCR-17-0134
- Theocharis S, Kotta-Loizou I, Klijanienko J, Giaginis C, Alexandrou P, Dana E, Rodriguez J, Patsouris E, Sastre-Garau X. Extracellular signal-regulated kinase (ERK) expression and activation in mobile tongue squamous cell carcinoma: associations with clinicopathological parameters and patients survival. Tumour Biol. 2014;35(7):6455–65. doi: 10.1007/s13277-014-1853-9
- Li Z, Li N, Shen L. MAP2K6 is associated with radiation resistance and adverse prognosis for locally advanced nasopharyngeal carcinoma patients. Cancer Manag Res. 2018;10:6905–6912. doi: 10.2147/CMAR.S184689
- Meerz A, Deville SS, Müller J, Cordes N. Comparative Therapeutic Exploitability of Acute Adaptation Mechanisms to Photon and Proton Irradiation in 3D Head and Neck Squamous Cell Carcinoma Cell Cultures. Cancers (Basel). 2021;13(6):1190. doi: 10.3390/cancers13061190
- Ha BG, Park JE, Cho HJ, Lim YB, Shon YH. Inhibitory effects of proton beam irradiation on integrin expression and signaling pathway in human colon carcinoma HT29 cells. Int J Oncol. 2015;46(6):2621–8. doi: 10.3892/ijo.2015.2942
- Fukusumi T, Califano JA. The NOTCH Pathway in Head and Neck Squamous Cell Carcinoma. J Dent Res. 2018;97(6):645–653. doi: 10.1177/0022034518760297
- Nowell CS, Radtke F. Notch as a tumour suppressor. Nat Rev Cancer. 2017;17(3):145–159. doi: 10.1038/nrc.2016.145
- Hovinga KE, Shimizu F, Wang R, Panagiotakos G, Van Der Heijden M, Moayedpardazi H, Correia AS, Soulet D, Major T, Menon J, Tabar V. Inhibition of notch signaling in glioblastoma targets cancer stem cells via an endothelial cell intermediate. Stem Cells. 2010;28(6):1019–29. doi: 10.1002/stem.429
- Yahyanejad S, Theys J, Vooijs M. Targeting Notch to overcome radiation resistance. Oncotarget. 2016;7(7):7610–28. doi: 10.18632/oncotarget.6714
- Kumar V, Vashishta M, Kong L, Lu JJ, Wu X, Dwarakanath BS, Guha C. Carbon Ion Irradiation Downregulates Notch Signaling in Glioma Cell Lines, Impacting Cell Migration and Spheroid Formation. Cells. 2022;11(21):3354. doi: 10.3390/cells11213354
- Cammarata FP, Torrisi F, Vicario N, Bravatà V, Stefano A, Salvatorelli L, D’Aprile S, Giustetto P, Forte GI, Minafra L, Calvaruso M, Richiusa S, Cirrone GAP, Petringa G, Broggi G, Cosentino S, Scopelliti F, Magro G, Porro D, Libra M, Ippolito M, Russo G, Parenti R, Cuttone G. Proton boron capture therapy (PBCT) induces cell death and mitophagy in a heterotopic glioblastoma model. Commun Biol. 2023;6(1):388. doi: 10.1038/s42003-023-04770-w
- Demoulin JB, Essaghir A. PDGF receptor signaling networks in normal and cancer cells. Cytokine Growth Factor Rev. 2014;25(3):273–83. doi: 10.1016/j.cytogfr.2014.03.003
- Lin LH, Lin JS, Yang CC, Cheng HW, Chang KW, Liu CJ. Overexpression of Platelet-Derived Growth Factor and Its Receptor Are Correlated with Oral Tumorigenesis and Poor Prognosis in Oral Squamous Cell Carcinoma. Int J Mol Sci. 2020;21(7):2360. doi: 10.3390/ijms21072360
- Heldin CH. Targeting the PDGF signaling pathway in tumor treatment. Cell Commun Signal. 2013;11:97. doi: 10.1186/1478-811X-11-97
- Farooqi AA, Siddik ZH. Platelet-derived growth factor (PDGF) signalling in cancer: rapidly emerging signalling landscape. Cell Biochem Funct. 2015;33(5):257–65. doi: 10.1002/cbf.3120
- Pietras K, Sjöblom T, Rubin K, Heldin CH, Ostman A. PDGF receptors as cancer drug targets. Cancer Cell. 2003;3(5):439–43. doi: 10.1016/s1535-6108(03)00089-8
- Lindahl P, Johansson BR, Levéen P, Betsholtz C. Pericyte loss and microaneurysm formation in PDGF-B-deficient mice. Science. 1997;277(5323):242–5. doi: 10.1126/science.277.5323.242
- Pandey P, Khan F, Upadhyay TK, Seungjoon M, Park MN, Kim B. New insights about the PDGF/PDGFR signaling pathway as a promising target to develop cancer therapeutic strategies. Biomed Pharmacother. 2023;161:114491. doi: 10.1016/j.biopha.2023.114491
- Kondratyuk RB, Grekov IS, Seleznev EA. Microenvironment influence on the development of epithelial-mesenchymal transformation in lung cancer. RUDN Journal of Medicine. 2022;26(3):325–337. doi: 10.22363/2313-0245-2022-26-3-325-337
- Heldin CH, Rubin K, Pietras K, Ostman A. High interstitial fluid pressure — an obstacle in cancer therapy. Nat Rev Cancer. 2004;4(10):806–13. doi: 10.1038/nrc1456
- Aebersold DM, Froehlich SC, Jonczy M, Beer KT, Laissue J, Greiner RH, Djonov V. Expression of transforming growth factor-alpha, epidermal growth factor receptor and platelet-derived growth factors A and B in oropharyngeal cancers treated by curative radiation therapy. Radiother Oncol. 2002;63(3):275–83. doi: 10.1016/s0167-8140(02)00131-7
- Han Y, Liu D, Li L. PD‑1/PD-L1 pathway: current researches in cancer. Am J Cancer Res. 2020;10(3):727–742
- Ohaegbulam KC, Assal A, Lazar-Molnar E, Yao Y, Zang X. Human cancer immunotherapy with antibodies to the PD‑1 and PD-L1 pathway. Trends Mol Med. 2015;21(1):24–33. doi: 10.1016/j.molmed.2014.10.009
- Qiao XW, Jiang J, Pang X, Huang MC, Tang YJ, Liang XH, Tang YL. The Evolving Landscape of PD‑1/PD-L1 Pathway in Head and Neck Cancer. Front Immunol. 2020;11:1721. doi: 10.3389/fimmu.2020.01721
- Chen TC, Wu CT, Wang CP, Hsu WL, Yang TL, Lou PJ, Ko JY, Chang YL. Associations among pretreatment tumor necrosis and the expression of HIF‑1α and PD-L1 in advanced oral squamous cell carcinoma and the prognostic impact thereof. Oral Oncol. 2015;51(11):1004–1010. doi: 10.1016/j.oraloncology.2015.08.011
- Scognamiglio T, Chen YT. Beyond the Percentages of PD-L1-Positive Tumor Cells: Induced Versus Constitutive PD-L1 Expression in Primary and Metastatic Head and Neck Squamous Cell Carcinoma. Head Neck Pathol. 2018;12(2):221–229. doi: 10.1007/s12105-017-0857-3
- Kwon MJ, Rho YS, Nam ES, Cho SJ, Park HR, Min SK, Seo J, Choe JY, Kim ES, Park B, Hong M, Min KW. Clinical implication of programmed cell death‑1 ligand‑1 expression in tonsillar squamous cell carcinoma in association with intratumoral heterogeneity, human papillomavirus, and epithelial-to-mesenchymal transition. Hum Pathol. 2018;80:28–39. doi: 10.1016/j.humpath.2018.03.025
- Yang WF, Wong MCM, Thomson PJ, Li KY, Su YX. The prognostic role of PD-L1 expression for survival in head and neck squamous cell carcinoma: A systematic review and meta-analysis. Oral Oncol. 2018;86:81–90. doi: 10.1016/j.oraloncology.2018.09.016
- Du J, Kageyama SI, Hirata H, Motegi A, Nakamura M, Hirano Y, Okumura M, Yamashita R, Tsuchihara K, Hojo H, Hirayama R, Akimoto T. Comparative analysis of the immune responses in cancer cells irradiated with X-ray, proton and carbon-ion beams. Biochem Biophys Res Commun. 2021;585:55–60. doi: 10.1016/j.bbrc.2021.11.004
- Ko EC, Formenti SC. Radiation therapy to enhance tumor immunotherapy: a novel application for an established modality. Int J Radiat Biol. 2019;95(7):936–939. doi: 10.1080/09553002.2019.1623429
- Rykkelid AM, Sinha PM, Folefac CA, Horsman MR, Sørensen BS, Søland TM, Schreurs OJF, Malinen E, Edin NFJ. Combination of proton- or X-irradiation with anti-PDL1 immunotherapy in two murine oral cancers. Sci Rep. 2024;14(1):11569. doi: 10.1038/s41598-024-62272-z