World J Oncol
World Journal of Oncology, ISSN 1920-4531 print, 1920-454X online, Open Access
Article copyright, the authors; Journal compilation copyright, World J Oncol and Elmer Press Inc
Journal website https://www.wjon.org

Review

Volume 14, Number 5, October 2023, pages 350-357


Update on Radiotherapy Changes of Nasopharyngeal Carcinoma Tumor Microenvironment

Dao Qi Zhua, Chao Sua, Jing Jun Lib, Ai Wu Lib, Ying Luvb, Qin Fana, c

aSchool of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
bNanFang Hospital, Southern Medical University, Guangzhou 510515, China
cCorresponding Author: Qin Fan, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China

Manuscript submitted June 29, 2023, accepted September 11, 2023, published online September 20, 2023
Short title: Microenvironment of NPC After RT
doi: https://doi.org/10.14740/wjon1645

Abstract▴Top 

The utilization of radiotherapy (RT) serves as the principal approach for managing nasopharyngeal carcinoma (NPC). Consequently, it is imperative to investigate the correlation between the radiation microenvironment and radiation resistance in NPC. PubMed and China National Knowledge Infrastructure (CNKI) databases were accessed to perform a search utilizing the English keywords “nasopharyngeal cancer”, “radiotherapy”, and “microenvironment”. The search time spanned from the establishment of the database until January 20, 2023. A total of 82 articles were included. The post-radiation tumor microenvironment (TME), or the radiation microenvironment, includes several components, such as the radiation-immune microenvironment and the radiation-hypoxic microenvironment. The radiation-immune microenvironment includes various factors like immune cells, signaling molecules, and extracellular matrix. RT can reshape the TME, leading to immune responses with both cytotoxic effects (T cells, B cells, natural killer (NK) cells) and immune escape mechanisms (regulatory T cells (Tregs), macrophages). RT enhances immune responses through DNA release, type I interferons, and immune cell recruitment. Radiation-hypoxic microenvironment affects metabolism and molecular changes. RT-induced hypoxia causes vascular changes, fibrosis, and vessel compression, leading to tissue hypoxia. Hypoxia activates hypoxia-inducible factor (HIF)-1α/2α, promoting angiogenesis and glycolysis in tumor cells. TME changes due to hypoxia also involve immune suppressive cells like myeloid-derived suppressor cells (MDSCs), tumor-associated macrophages (TAMs), and Tregs. The radiation microenvironment is involved in radiation resistance and holds a significant effect on the prognosis of patients with NPC. Exploring the radiation microenvironment provides new insights into RT and NPC research.

Keywords: Nasopharyngeal carcinoma; Radiation microenvironment; Immune microenvironment; Radiation hypoxia microenvironment

Introduction▴Top 

Nasopharyngeal carcinoma (NPC) is an epithelial malignancy that occurs mainly in the upper and lateral regions of the nasopharyngeal cavity. Current consensus suggests that its occurrence and development are mainly associated with factors such as Epstein-Barr virus (EBV) infection, genetic susceptibility, as well as environmental factors [1]. Owing to the unique pathology, biological behavior, and anatomical structure of NPC, radiotherapy (RT) is considered the primary treatment option [2]. However, in the case of patients diagnosed with late-stage NPC, changes in the tumor microenvironment (TME) after RT can result in tumor resistance to treatment, leading to poor prognostic outcomes or tumor recurrence after treatment.

The TME encompasses a range of factors that are in close contact with tumor cells, such as neighboring blood vessels, immune cells, fibroblasts, bone marrow-derived inflammatory cells, various signaling molecules, and extracellular matrix (ECM) [3]. RT can remodel the TME [4], which is involved in local and systemic immune regulation and influences tumor progression. This remodeled environment is termed the radiation microenvironment and includes a radiation-immune microenvironment in which RT can induce different immune responses. For example, T cells, B cells, and associated secretory factors can have a cytotoxic effect on the tumor, and macrophages, Treg cells, and Th2 cells can mediate immune escape [5]. The hypoxic radiation microenvironment is another aspect where post-radiation vascular exhaustion can exacerbate hypoxia, leading to the signaling of hypoxia-inducible factor (HIF)-1α and promoting angiogenesis through vascular endothelial growth factors (VEGFs) [6]. Furthermore, RT can affect immune cell infiltration by altering the vascular structure of the tumor tissue. Hence, RT can trigger a series of interrelated processes within the TME, leading to tumor resistance or recurrence.

Methods▴Top 

PubMed and China National Knowledge Infrastructure (CNKI) databases were accessed to perform a search utilizing the English keywords “nasopharyngeal cancer”, “radiotherapy”, and “microenvironment”. The search time spanned from the establishment of the database until January 20, 2023. A total of 7,855 English articles and 3,798 Chinese articles were retrieved. The present study employed specific inclusion criteria, which included 1) studies on RT progress for NPC and 2) studies on the progress of the TME. The exclusion criteria were conference abstracts, Chinese theses, and other informally published documents.

Radioimmune Microenvironment▴Top 

The tumor immune microenvironment, which encompasses TME immune components while excluding other stromal and extracellular components, primarily comprises T and B lymphocytes as well as myeloid cells [7, 8]. Within this microenvironment, two functional subtypes of cells can be differentiated: one that directly exerts cytotoxic effects on tumor cells, which include T and B cells, as well as natural killer (NK) cells, and another that enables tumor immune evasion, preventing tumor destruction, including regulatory T cells (Tregs) and macrophages. There is a delicate balance between these two; therefore, RT can reshape the immune microenvironment through key mechanisms that involve the activation of immunostimulatory and inhibitory signaling pathways. This leads to tumor cell destruction and promotes immune evasion, ultimately resulting in tumor recurrence and metastasis [9].

According to research, the utilization of RT has been found to potentially enhance the double-stranded DNA (dsDNA) release in the nucleus, elevate the permeability of the outer membrane of mitochondria, and trigger mitochondrial DNA (mtDNA) exposure in the cytoplasm, thus enhancing the immune response [10]. For instance, dsDNA and mtDNA can initiate the transcription of type I interferons (IFNs) [11]. Signals from type I IFNs can enhance cytotoxic T-cell activation, exerting a direct cytotoxic impact on tumor cells [12]. In contrast, dsDNA accumulation in tumor-derived exosomes post-RT also enhances dendritic cells (DCs) recruitment and directly triggers DC type I IFN responses, which additionally promotes CD8+ T-cell recruitment [13] and provides signals to activate T cell. Therefore, RT changes the phenotype of tumor cells besides promoting the production of damage-related molecular patterns and also immune responses. Moreover, RT promotes not only the tumor cell sensitivity to T-cell-mediated anti-tumor effects but also the tumor cell recognition and elimination. The immune microenvironment of NPCs includes multiple immune cells, including T and B lymphocytes, NK cells, as well as myeloid-derived suppressor cells (MDSCs) [14].

In NPC, the most common infiltrates are EBV-negative CD3 T lymphocytes [15], which lack an effective immune response. Therefore, B lymphocytes are immune cells with potential anti-NPC properties. B lymphocytes represent the second most abundant and varied cell type within the NPC microenvironment. An increase in the abundance and diversity of B lymphocytes is associated with EBV positivity [16, 17]. According to previous studies, B cells in NPC can be recruited to tertiary lymphoid structures by PD-1+ exhausted CD4+ T cells of tumor origin via the CXCL13/CXCR5 axis. Additionally, a positive correlation has been observed between CD19+ B cells and EBV-positive NPC patients [18]. This finding suggests that NPC-infiltrating B cells may have an anti-NPC immune function.

Along with T and B cells, NK cells were demonstrated to possess a cytotoxic impact on tumors. NK cells are a type of immune cell that holds the ability to directly trigger the death of tumor and virus-infected cells without specific immunity. RT can lead to an overexpression of stress-induced activation ligands in tumor cells, thus increasing NK cell-mediated cytotoxic effects [19]. NK cells perform various biological functions. Effective NK cell anti-tumor activity has been shown to be a prerequisite for T-cell-mediated anti-tumor immune responses [20]. NK cells, including cytotoxic T cells, exert control over tumors by secreting perforin-containing cytotoxic granules [21] or through ligands binding to death receptors in target cells, which include tumor necrosis factor (TNF)-associated apoptosis-inducing ligand (TRAIL) and Fas ligand. Moreover, NK cells also produce multiple cytokines, including those that promote inflammatory responses and immune suppression, that include interleukin (IL)-10 and TNF-α, or growth factors, including a colony-stimulating factor of macrophage granulocytes (GM-CSF) and granulocyte colony-stimulating factor (G-CSF) [22]. Some immune mechanisms of NPCs have been shown to be downregulated, leading to immune evasion, but NK cells can secrete cytokines or directly lyse tumor cells [23]. NK cells were found to be involved in the prevention of EBV-associated malignancy progression arising from primary immunodeficiencies. These immunodeficiencies are characterized by genetic mutations that impact the function of genes contributing to NK cell differentiation and activation. A recent study has revealed that the immune features specific to NK cells in tumors are correlated to a favorable NPC prognosis. This suggests that NK cells present in the TME contribute to the regulation of the progression of EBV-associated epithelial tumors [24]. In conclusion, NK cells, including T and B cells, participate in the immune response process in different ways and undergo functional maturation to achieve cytotoxic effects on tumor cells.

In immunogenesis, tumor cells develop resistance to radiation due to the induction of anti-immune mechanisms by RT. The repetitive irradiation of tumor cells can result in the expression of IFN-stimulated genes and chronic type I IFN, which can promote radiation resistance and metastasis via inhibitory pathways [5]. IFN-γ and type I IFNs are accountable for programmed death ligand 1 (PD-L1) overexpression in tumor cells, leading to the induction of T-cell exhaustion and resistance to anti-tumor immunity [25]. Research data indicate that NPC radiosensitivity can be further enhanced by utilizing potential immunotherapy targets in conjunction with PD-1/PD-L1 blockade [26].

Recent in-depth research on exosomes has also revealed their role in suppressing tumor immunity and promoting tumor growth and metastasis. Exosomes are small vesicles between 30 and 150 nm in size and are considered key factors in intercellular communication [27]. Exosomes were found to be involved in mediating the TME through the level transfer of proteins, mRNA, and miRNAs [28]. Exosomes mediate cell communication and transfer components to recipient cells. NPC cells infuse exosomes containing LMP1 (NPC-Exo) and miRNAs into the TME, leading to the evasion of the immune system from host surveillance [29]. According to a recent study, methyltransferase 3 (METTL3), methyltransferase 14 (METTL14), and WT1-associated protein (WTAP) modify the effect of m6A on radiosensitivity and invasive metastasis of NPC. Exosome production within the NPC microenvironment may have increased, which is one possible explanation [30].

Furthermore, Tregs also affect the impact of RT on tumors. Tregs survive in RT and inhibit the proliferation of effector cells [31], and by overexpressing cytotoxic T-lymphocyte-associated protein 4 (CTLA4), they block the activation of T lymphocytes, achieving an immune suppression effect. Research has found that functional analysis at the cellular and animal levels for NPC confirms that Tregs are recruited into the TME by tumor-secreted substances, assisting tumors in escaping immune surveillance [32]. Thus, RT also induces immune resistance, leading to tumor recurrence and metastasis.

In addition to epithelial cells in the immune microenvironment, stromal cells constitute a considerable portion, mainly composed of tumor-associated macrophages (TAMs) and MDSC. MDSC directly enhances immune tolerance [33]. MDSCs, a heterogeneous group of pathologically activated myeloid cells, can enhance tumor cell metastasis by infiltrating primary tumors and suppressing immune function to accelerate tumor progression [34]. MDSCs can negate the associated immune response during RT [35]. Clinical data show that the percentage of monocytic MDSCs among the total live cells is significantly increased in high-risk groups after NPC chemotherapy, limiting the growth of MDSCs, which is vital for patient treatment [36]. TAMs are the primary components of immune infiltration in solid tumors [37]. Current research suggests that TAMs in the TME originate mainly from bone marrow-derived monocyte precursor cells recruited to tumor sites, affected by TGF-β, CCL2, IL-1/4, and other cytokines, chemokines, and even microorganisms in the tumor tissue, polarizing toward the M2 phenotype, thus inducing immune infiltration [38, 39]. Relevant reports show that TAM infiltration exhibited a close correlation to the poor prognosis of EBV-infected NPC [40], and it was experimentally proven that EBV-infected NPC cells could recruit monocytes through VEGF and activate monocytes in TAMs in a manner dependent on GM-CSF- and nuclear factor kappa B (NF-κB)-dependent manner. Gleichzeitig TAM promotes tumor metastasis and NF-κB activation [41].

In recent years, immune checkpoints have become a hotspot. These immune checkpoint molecules, such as PD-1, PD-L1, and CTLA-4, are fundamentally a component of the immune system’s regulatory apparatus. They assist in maintaining equilibrium between a robust immune response when necessary and the ability to prevent typical tissue damage and destruction. Immune checkpoint inhibitors are medications used in the treatment of cancer that are developed to prevent these interactions. By obstructing the inhibitory signals given by these checkpoint molecules, the immune system is better able to target and eliminate cancer cells. However, the immune checkpoints are a significant contributor to immune tolerance [42] and radiation resistance during the onset and progression of tumors. Recent research based on different immune subtypes in NPC has demonstrated the predictive power of the immune checkpoint inhibitor (ICI) response and has increased sensitivity to RT [43]. Therefore, the application of ICIs can inhibit tumor immune evasion and enhance tumor killing by the immune system through various pathways and mechanisms, thereby increasing radiosensitivity.

In summary, RT places the body’s immune microenvironment in a very delicate balance between killing tumors and immune suppression. RT has the potential to enhance the presentation of tumor antigens, trigger immunogenic cell death of tumor cells, activate pathways that mimic viral responses, and overexpress chemokines, ultimately promoting tumor cell death by T cells. Conversely, RT was found to be involved in promoting immune evasion mechanisms such as PD-L1 overexpression, immunosuppressive chemokine release, and exosome activation, thereby impeding the activation and function of T cells and similar cells.

Radiation-Hypoxic Microenvironment▴Top 

During tumor growth, tumor vessels provide tumor cells with abundant nutrients and oxygen and remove metabolic waste, which enables the tumor to grow rapidly and potentially invade and metastasize [44]. However, tumor cells have high metabolic activity and require considerable energy. When demand exceeds supply, hypoxic areas form, altering metabolic capacity [45]. Furthermore, when tumor cells are stimulated by hypoxia, a series of changes occur at the molecular and cellular levels. Acid waste produced by anaerobic glycolysis can improve tolerance to hypoxia and resistance to external damage, thus disrupting the TME [46]. RT, a key measure against tumors, has been shown to elicit stress responses, repair damage, and restores cellular homeostasis [47]. Radioactive hypoxia is a part of the microenvironment after RT in tumor cells. The tumor cell resistance to RT can be attributed to their capacity for DNA damage repair and tolerance. Therefore, the impact of a hypoxic microenvironment on radiation resistance is a significant environmental factor. The etiology of hypoxia can be succinctly categorized into two components: a decrease in tumor angiogenesis and an increase in destruction.

Substantial research has been conducted on the mechanisms of angiogenesis. VEGF specifically enhances endothelial cell growth and increases the permeability of newly formed vessels [48]. VEGF overexpression is common in NPC [49], and VEGF secretion is highly correlated with NPC angiogenesis, metastasis, and poor prognosis. One study emphasized the relationship between JAK2, STAT3, and VEGF expression and NPC development and progression [50]. Recently, a method of angiogenesis that is different from endothelial-derived angiogenesis, namely vascular mimicry (VM), has been discovered. VM is a vascular network pattern that is formed by highly invasive tumor cells and replaces endothelial cells and has been confirmed in various tumors [51], including lung, stomach, and ovarian cancers [52-54]. However, there is little research on NPC, with only a few studies reporting that Foxq1 can regulate VM to promote NPC metastasis [55] and the synergistic effect of anti-VM and anti-VEGF treatment in NPC [56]. To summarize, the hypoxic environment of solid tumors is strongly related to the synergistic action of VEGF and VM [56]. RT can induce endothelial cell apoptosis, leading to vessel destruction and endothelial cell aging. Aging endothelial cells cause endothelial dysfunction, thus inhibiting angiogenesis and inducing oxidative stress and inflammation [57], ultimately leading to the formation of a hypoxic microenvironment.

Furthermore, non-tumor cell components and functions in the tumor region were validated to also undergo significant changes, especially the activation and proliferation of matrix cells (such as cancer-associated fibroblasts (CAFs)) and an increase in matrix components (such as fibrin), leading to morphological tumor remodeling [58]. As a result, vessels are compressed or aggregated, and fibrin clots block vessels [59], causing damage to blood circulation, insufficient oxygen supply, and increased tissue hypoxia. In a recent study, a degradable magnesium alloy (Mg) with anticancer activity was designed by reducing cancer cell proliferation. This experiment verified that the physiological functions of elements such as Mg can change with changes in the hypoxic environment. Magnesium has been observed to promote angiogenesis in normoxic conditions, while conversely, it has been found to impede angiogenesis in hypoxic conditions [60]. Many factors can lead to the formation of a hypoxic microenvironment. Hypoxia has a close correlation cancer cell proliferation, migration, invasion, and angiogenesis, interacts with other cells and pathways, and has a significant impact on the TME.

Research has found that hypoxia and HIF-1α/2α play important roles in tumor angiogenesis [61]. Post-RT vascular injury can lead to significant activation of HIFs [62]. Under hypoxic conditions, enzymes involved in HIF degradation are deactivated, allowing HIF to enter the cell nucleus, where it participates in angiogenesis, anaerobic glycolysis, inhibition of apoptosis, and transcription of other genes. This increases oxygen delivery and normalizes tumor vessels [63]. Additionally, the endogenous PI3K-AKT-mTOR pathway activation induces an increase in the HIF pathway, resulting in a comparable hypoxic microenvironment [64]. Both exogenous and endogenous hypoxia disrupt autophagy mechanisms by upregulating metabolic processes, DNA repair, and anti-apoptotic pathways, thereby enhancing the radiation resistance of tumor cells. Clinical studies revealed that 100% of primary NPC cases and 58% of neck lymph node metastases contain hypoxic areas and HIF-1α overexpression and risks of metastasis and eventual death [65]. Thus, HIF-1α has emerged as a significant prognostic factor for NPC and a promising therapeutic target. Moreover, research has shown that hypoxia can regulate CAFs, which are spindle-shaped cells that can synthesize collagen in connective tissues and also contribute to wound healing, tissue fibrosis, and inflammatory processes [66]. HIF was suggested to be involved in the metabolic reprogramming of CAFs and mediates their tumor-promoting effects [67]. It has been found that CAFs induce radiation resistance and promote the survival of NPC cells post-RT through the IL-8/NF-κB pathway to decrease radiation-induced DNA damage. The administration of trabectedin, a CAF inhibitor, has the potential to restrict the survival of NPC cells induced by CAF and mitigate the impact of radiation [68].

It is well known that under cellular hypoxia, cells reduce their dependence on mitochondrial oxidative phosphorylation and preferentially use the anaerobic glycolysis pathway, which does not rely on oxygen consumption, to maintain sufficient ATP production to meet body energy needs [69]. Consequently, the metabolic feature of most tumors is increased glycolysis, which is caused by hypoxia, forcing tumor cells to switch from oxidative phosphorylation to glycolysis. These changes are mediated by HIF-1α-driven transcription, resulting in upregulating glucose transporter proteins and glycolysis-involved genes [70]. This phenomenon promotes tumor cell survival and metastasis under hypoxic conditions. This glycolysis is observed in various cells, and recent research has discovered a new circular RNA that can directly bind and stabilize ubiquitinated mRNA, thereby inhibiting the glycolysis process and NPC proliferation and migration through ubiquitination and upregulation of protein levels [71], thus demonstrating the impact of glycolysis on NPC proliferation and migration.

Hypoxia also has a significant impact on alterations in the immune microenvironment. This is because hypoxia can improve the immune attack resistance of tumor cells, resulting in immune evasion [72]. Research has shown that hypoxic regions in solid tumors are infiltrated by high levels of immune suppressive cells, including MDSCs, TAMs, and Treg cells [73]. As mentioned previously, MDSCs can directly promote immune tolerance, making them one of the main components of the immunosuppressive network that leads to tumor T-cell defects, with direct evidence that HIF-1α can regulate MDSC function and differentiation in a hypoxic microenvironment. TAMs, a significant constituent of immune infiltration in solid tumors [37], were found to tend to be situated primarily in hypoxic regions, and HIF-1α-induced under hypoxic conditions has an inhibitory effect on cytotoxic T cells [74]. Treg cells can also be driven by HIF-1α-dependent transcription factors (FoxP3) or attracted by the cytokine spectrum in the tumor bed microenvironment [75]. Recent research suggests that ICI not only causes T-cell attacks on tumor cells but might also regulate the TME through the normalization of tumor vessels [76, 77]. The novel interaction has the potential to offer opportunities for immunotherapies aimed at decreasing tumor hypoxia and enhancing radiosensitivity. Unfortunately, there is no strong evidence that biomarkers for ICIs in NPC have been identified.

Conclusion and Future Perspectives▴Top 

Numerous studies have shown that, despite continued improvements in the efficacy and safety of RT, tumor recurrence after RT remains an issue, and the TME plays a critical role in this process. The activation of the immune system and the immune suppression induced by RT are delicately balanced. The process of metabolic reprogramming and the induction of angiogenesis within the TME is crucial for the effective elimination of tumor cells by cytotoxic T cells and other immune cells. The composition of multiple cells and the ECM within this particular environment has demonstrated a capacity to mitigate the immune responses of the host toward malignant neoplastic cells. The proliferation of tumors is facilitated by various mechanisms, including the secretion of immunosuppressive cytokines, metabolic alterations, and other factors. Conversely, TMEs that are hypoxic and acidic have been observed to provide radioresistance by impeding the proliferation of anti-tumor cells, including CD8 T lymphocytes and NK cells, while promoting the proliferation of tumor-tolerant cells such as MDSCs and Treg cells [78].

These characteristics of the TME may be helpful in NPC treatment. For example, potential immune therapy targets could be used in combination with the PD-1/PD-L1 blockade to further increase the radiosensitivity of NPC [79]. The heterogeneity of the NPC TME can identify biologically different immune subtypes, predict prognosis, and predict immune therapy responses [43]. EBV-infected NPC cells can secrete cytokines and exosomes containing viral products to modulate the function of matrix cells in the TME, thus promoting NPC progression and avoiding host immune attack [80].

In summary, RT induces remodeling of the TME, with hypoxia and immunity being the two most critical aspects. The two are interrelated and inseparable. Most malignant tumors create a hypoxic microenvironment that is conducive to their development, thus suppressing immune responses. Currently, the immune stimulation process is inseparable from the stimulation of HIFs, thereby immunogenically activating key progressions.

Acknowledgments

We express gratitude to our research team members for their dedication and hard work.

Financial Disclosure

Project was supported by the National Natural Science Foundation of China (82074132), Project of Administration of Traditional Chinese Medicine of Guangdong Province of China (20221258, 20213009), Natural Science Foundation of Guangdong Province of China (2314050000165).

Conflict of Interest

None to declare.

Author Contributions

Dao Qi Zhu: conceptualization, writing - original draft, literature review, funding acquisition; Chao Su: writing - review and editing; Jing Jun Li: writing - review and editing, compilation of references; Ai Wu Li: writing - review and editing, overall structure and organization; Ying Luv: writing - review and editing, final proofreading, funding acquisition; Qin Fan: supervision, project administration, funding acquisition.

Data Availability

The data supporting the findings of this study are available from the corresponding author upon reasonable request.


References▴Top 
  1. Chen YP, Chan ATC, Le QT, Blanchard P, Sun Y, Ma J. Nasopharyngeal carcinoma. Lancet. 2019;394(10192):64-80.
    doi pubmed
  2. Liu S, Wang Z, Zhu D, Yang J, Lou D, Gao R, Wang Z, et al. Effect of Shengmai Yin on the DNA methylation status of nasopharyngeal carcinoma cell and its radioresistant strains. J Pharm Anal. 2021;11(6):783-790.
    doi pubmed pmc
  3. de Visser KE, Joyce JA. The evolving tumor microenvironment: From cancer initiation to metastatic outgrowth. Cancer Cell. 2023;41(3):374-403.
    doi pubmed
  4. McLaughlin M, Patin EC, Pedersen M, Wilkins A, Dillon MT, Melcher AA, Harrington KJ. Inflammatory microenvironment remodelling by tumour cells after radiotherapy. Nat Rev Cancer. 2020;20(4):203-217.
    doi pubmed
  5. Jarosz-Biej M, Smolarczyk R, Cichon T, Kulach N. Tumor microenvironment as a "Game Changer" in cancer radiotherapy. Int J Mol Sci. 2019;20(13):3212.
    doi pubmed pmc
  6. Suwa T, Kobayashi M, Nam JM, Harada H. Tumor microenvironment and radioresistance. Exp Mol Med. 2021;53(6):1029-1035.
    doi pubmed pmc
  7. Tugues S, Ducimetiere L, Friebel E, Becher B. Innate lymphoid cells as regulators of the tumor microenvironment. Semin Immunol. 2019;41:101270.
    doi pubmed
  8. Tofilon PJ, Camphausen K. Remodeling the irradiated tumor microenvironment: the fifth R of radiobiology? Switzerland: Springer International Publishing AG. 2016:135-149
  9. Charpentier M, Spada S, Van Nest SJ, Demaria S. Radiation therapy-induced remodeling of the tumor immune microenvironment. Semin Cancer Biol. 2022;86(Pt 2):737-747.
    doi pubmed
  10. Patrushev M, Kasymov V, Patrusheva V, Ushakova T, Gogvadze V, Gaziev A. Mitochondrial permeability transition triggers the release of mtDNA fragments. Cell Mol Life Sci. 2004;61(24):3100-3103.
    doi pubmed
  11. Yamazaki T, Galluzzi L. Mitochondrial control of innate immune signaling by irradiated cancer cells. Oncoimmunology. 2020;9(1):1797292.
    doi pubmed pmc
  12. Lim JY, Gerber SA, Murphy SP, Lord EM. Type I interferons induced by radiation therapy mediate recruitment and effector function of CD8(+) T cells. Cancer Immunol Immunother. 2014;63(3):259-271.
    doi pubmed pmc
  13. Diamond JM, Vanpouille-Box C, Spada S, Rudqvist NP, Chapman JR, Ueberheide BM, Pilones KA, et al. Exosomes Shuttle TREX1-Sensitive IFN-Stimulatory dsDNA from Irradiated Cancer Cells to DCs. Cancer Immunol Res. 2018;6(8):910-920.
    doi pubmed pmc
  14. Gong L, Kwong DL, Dai W, Wu P, Li S, Yan Q, Zhang Y, et al. Comprehensive single-cell sequencing reveals the stromal dynamics and tumor-specific characteristics in the microenvironment of nasopharyngeal carcinoma. Nat Commun. 2021;12(1):1540.
    doi pubmed pmc
  15. Gourzones C, Barjon C, Busson P. Host-tumor interactions in nasopharyngeal carcinomas. Semin Cancer Biol. 2012;22(2):127-136.
    doi pubmed
  16. Su ZY, Siak PY, Leong CO, Cheah SC. Nasopharyngeal carcinoma and its microenvironment: past, current, and future perspectives. Front Oncol. 2022;12:840467.
    doi pubmed pmc
  17. Li JP, Wu CY, Chen MY, Liu SX, Yan SM, Kang YF, Sun C, et al. PD-1(+)CXCR5(-)CD4(+) Th-CXCL13 cell subset drives B cells into tertiary lymphoid structures of nasopharyngeal carcinoma. J Immunother Cancer. 2021;9(7):e002101.
    doi pubmed pmc
  18. Xu T, Huang Z, Su B, Wang S, Wang D, Wang C, Wei W, et al. Prognostic significance of circulating CD19+ B lymphocytes in EBV-associated nasopharyngeal carcinoma. Med Oncol. 2014;31(10):198.
    doi pubmed
  19. Li MO, Wolf N, Raulet DH, Akkari L, Pittet MJ, Rodriguez PC, Kaplan RN, et al. Innate immune cells in the tumor microenvironment. Cancer Cell. 2021;39(6):725-729.
    doi pubmed
  20. Hsu J, Hodgins JJ, Marathe M, Nicolai CJ, Bourgeois-Daigneault MC, Trevino TN, Azimi CS, et al. Contribution of NK cells to immunotherapy mediated by PD-1/PD-L1 blockade. J Clin Invest. 2018;128(10):4654-4668.
    doi pubmed pmc
  21. Vivier E, Raulet DH, Moretta A, Caligiuri MA, Zitvogel L, Lanier LL, Yokoyama WM, et al. Innate or adaptive immunity? The example of natural killer cells. Science. 2011;331(6013):44-49.
    doi pubmed pmc
  22. Walzer T, Dalod M, Robbins SH, Zitvogel L, Vivier E. Natural-killer cells and dendritic cells: "l'union fait la force". Blood. 2005;106(7):2252-2258.
    doi pubmed
  23. Singh S, Banerjee S. Downregulation of HLA-ABC expression through promoter hypermethylation and downmodulation of MIC-A/B surface expression in LMP2A-positive epithelial carcinoma cell lines. Sci Rep. 2020;10(1):5415.
    doi pubmed pmc
  24. Duan X, Chen H, Zhou X, Liu P, Zhang X, Zhu Q, Zhong L, et al. EBV infection in epithelial malignancies induces resistance to antitumor natural killer cells via F3-mediated platelet aggregation. Cancer Res. 2022;82(6):1070-1083.
    doi pubmed
  25. Shevtsov M, Sato H, Multhoff G, Shibata A. novel approaches to improve the efficacy of immuno-radiotherapy. Front Oncol. 2019;9:156.
    doi pubmed pmc
  26. Chen YP, Yin JH, Li WF, Li HJ, Chen DP, Zhang CJ, Lv JW, et al. Single-cell transcriptomics reveals regulators underlying immune cell diversity and immune subtypes associated with prognosis in nasopharyngeal carcinoma. Cell Res. 2020;30(11):1024-1042.
    doi pubmed pmc
  27. S ELA, Mager I, Breakefield XO, Wood MJ. Extracellular vesicles: biology and emerging therapeutic opportunities. Nat Rev Drug Discov. 2013;12(5):347-357.
    doi pubmed
  28. Vader P, Breakefield XO, Wood MJ. Extracellular vesicles: emerging targets for cancer therapy. Trends Mol Med. 2014;20(7):385-393.
    doi pubmed pmc
  29. Mrizak D, Martin N, Barjon C, Jimenez-Pailhes AS, Mustapha R, Niki T, Guigay J, et al. Effect of nasopharyngeal carcinoma-derived exosomes on human regulatory T cells. J Natl Cancer Inst. 2015;107(1):363.
    doi pubmed
  30. Wang YM, Peng ZY, Zhang LY, Wang YX, Fan RH, Zhang H, Jiang WH. N6-methyladenosine RNA modification landscape in the occurrence and recurrence of nasopharyngeal carcinoma. World J Oncol. 2022;13(4):205-215.
    doi pubmed pmc
  31. Persa E, Balogh A, Safrany G, Lumniczky K. The effect of ionizing radiation on regulatory T cells in health and disease. Cancer Lett. 2015;368(2):252-261.
    doi pubmed
  32. Lau KM, Cheng SH, Lo KW, Lee SA, Woo JK, van Hasselt CA, Lee SP, et al. Increase in circulating Foxp3+CD4+CD25(high) regulatory T cells in nasopharyngeal carcinoma patients. Br J Cancer. 2007;96(4):617-622.
    doi pubmed pmc
  33. Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol. 2009;9(3):162-174.
    doi pubmed pmc
  34. Yin Z, Li C, Wang J, Xue L. Myeloid-derived suppressor cells: Roles in the tumor microenvironment and tumor radiotherapy. Int J Cancer. 2019;144(5):933-946.
    doi pubmed
  35. Vatner RE, Formenti SC. Myeloid-derived cells in tumors: effects of radiation. Semin Radiat Oncol. 2015;25(1):18-27.
    doi pubmed
  36. Hopkins R, Xiang W, Marlier D, Au VB, Ching Q, Wu LX, Guan R, et al. Monocytic myeloid-derived suppressor cells underpin resistance to adoptive T cell therapy in nasopharyngeal carcinoma. Mol Ther. 2021;29(2):734-743.
    doi pubmed pmc
  37. Corzo CA, Condamine T, Lu L, Cotter MJ, Youn JI, Cheng P, Cho HI, et al. HIF-1alpha regulates function and differentiation of myeloid-derived suppressor cells in the tumor microenvironment. J Exp Med. 2010;207(11):2439-2453.
    doi pubmed pmc
  38. Donadon M, Torzilli G, Cortese N, Soldani C, Di Tommaso L, Franceschini B, Carriero R, et al. Macrophage morphology correlates with single-cell diversity and prognosis in colorectal liver metastasis. J Exp Med. 2020;217(11):e20191847.
    doi pubmed pmc
  39. Mantovani A, Allavena P, Marchesi F, Garlanda C. Macrophages as tools and targets in cancer therapy. Nat Rev Drug Discov. 2022;21(11):799-820.
    doi pubmed pmc
  40. Liao Q, Zeng Z, Guo X, Li X, Wei F, Zhang W, Li X, et al. LPLUNC1 suppresses IL-6-induced nasopharyngeal carcinoma cell proliferation via inhibiting the Stat3 activation. Oncogene. 2014;33(16):2098-2109.
    doi pubmed
  41. Huang D, Song SJ, Wu ZZ, Wu W, Cui XY, Chen JN, Zeng MS, et al. Epstein-barr virus-induced VEGF and GM-CSF drive nasopharyngeal carcinoma metastasis via recruitment and activation of macrophages. Cancer Res. 2017;77(13):3591-3604.
    doi pubmed
  42. Zheng X, Fang Z, Liu X, Deng S, Zhou P, Wang X, Zhang C, et al. Increased vessel perfusion predicts the efficacy of immune checkpoint blockade. J Clin Invest. 2018;128(5):2104-2115.
    doi pubmed pmc
  43. Chen YP, Lv JW, Mao YP, Li XM, Li JY, Wang YQ, Xu C, et al. Unraveling tumour microenvironment heterogeneity in nasopharyngeal carcinoma identifies biologically distinct immune subtypes predicting prognosis and immunotherapy responses. Mol Cancer. 2021;20(1):14.
    doi pubmed pmc
  44. Bagchi S, Yuan R, Engleman EG. Immune Checkpoint Inhibitors for the Treatment of Cancer: Clinical Impact and Mechanisms of Response and Resistance. Annu Rev Pathol. 2021;16:223-249.
    doi pubmed
  45. Huang RX, Zhou PK. DNA damage response signaling pathways and targets for radiotherapy sensitization in cancer. Signal Transduct Target Ther. 2020;5(1):60.
    doi pubmed pmc
  46. Vaupel P. Tumor microenvironmental physiology and its implications for radiation oncology. Semin Radiat Oncol. 2004;14(3):198-206.
    doi pubmed
  47. Wennerberg E, Vanpouille-Box C, Bornstein S, Yamazaki T, Demaria S, Galluzzi L. Immune recognition of irradiated cancer cells. Immunol Rev. 2017;280(1):220-230.
    doi pubmed pmc
  48. Semenza GL, Wang GL. A nuclear factor induced by hypoxia via de novo protein synthesis binds to the human erythropoietin gene enhancer at a site required for transcriptional activation. Mol Cell Biol. 1992;12(12):5447-5454.
    doi pubmed pmc
  49. Tang CE, Guan YJ, Yi B, Li XH, Liang K, Zou HY, Yi H, et al. Identification of the amyloid beta-protein precursor and cystatin C as novel epidermal growth factor receptor regulated secretory proteins in nasopharyngeal carcinoma by proteomics. J Proteome Res. 2010;9(12):6101-6111.
    doi pubmed
  50. Cheng JZ, Chen JJ, Xue K, Wang ZG, Yu D. Clinicopathologic and prognostic significance of VEGF, JAK2 and STAT3 in patients with nasopharyngeal carcinoma. Cancer Cell Int. 2018;18:110.
    doi pubmed pmc
  51. Maniotis AJ, Folberg R, Hess A, Seftor EA, Gardner LM, Pe'er J, Trent JM, et al. Vascular channel formation by human melanoma cells in vivo and in vitro: vasculogenic mimicry. Am J Pathol. 1999;155(3):739-752.
    doi pubmed pmc
  52. Williamson SC, Metcalf RL, Trapani F, Mohan S, Antonello J, Abbott B, Leong HS, et al. Vasculogenic mimicry in small cell lung cancer. Nat Commun. 2016;7:13322.
    doi pubmed pmc
  53. Liu R, Yang K, Meng C, Zhang Z, Xu Y. Vasculogenic mimicry is a marker of poor prognosis in prostate cancer. Cancer Biol Ther. 2012;13(7):527-533.
    doi pubmed
  54. Li M, Gu Y, Zhang Z, Zhang S, Zhang D, Saleem AF, Zhao X, et al. Vasculogenic mimicry: a new prognostic sign of gastric adenocarcinoma. Pathol Oncol Res. 2010;16(2):259-266.
    doi pubmed
  55. Luo Y, Wang J, Wang F, Liu X, Lu J, Yu X, Ma X, et al. Foxq1 promotes metastasis of nasopharyngeal carcinoma by inducing vasculogenic mimicry via the EGFR signaling pathway. Cell Death Dis. 2021;12(5):411.
    doi pubmed pmc
  56. Xiang T, Lin YX, Ma W, Zhang HJ, Chen KM, He GP, Zhang X, et al. Vasculogenic mimicry formation in EBV-associated epithelial malignancies. Nat Commun. 2018;9(1):5009.
    doi pubmed pmc
  57. Wang Y, Boerma M, Zhou D. Ionizing radiation-induced endothelial cell senescence and cardiovascular diseases. Radiat Res. 2016;186(2):153-161.
    doi pubmed pmc
  58. Gilkes DM, Semenza GL, Wirtz D. Hypoxia and the extracellular matrix: drivers of tumour metastasis. Nat Rev Cancer. 2014;14(6):430-439.
    doi pubmed pmc
  59. Bai R, Li Y, Jian L, Yang Y, Zhao L, Wei M. The hypoxia-driven crosstalk between tumor and tumor-associated macrophages: mechanisms and clinical treatment strategies. Mol Cancer. 2022;21(1):177.
    doi pubmed pmc
  60. Globig P, Madurawala R, Willumeit-Romer R, Martini F, Mazzoni E, Luthringer-Feyerabend BJC. Mg-based materials diminish tumor spreading and cancer metastases. Bioact Mater. 2023;19:594-610.
    doi pubmed pmc
  61. Conway EM, Collen D, Carmeliet P. Molecular mechanisms of blood vessel growth. Cardiovasc Res. 2001;49(3):507-521.
    doi pubmed
  62. Moeller BJ, Cao Y, Li CY, Dewhirst MW. Radiation activates HIF-1 to regulate vascular radiosensitivity in tumors: role of reoxygenation, free radicals, and stress granules. Cancer Cell. 2004;5(5):429-441.
    doi pubmed
  63. Rockwell S, Dobrucki IT, Kim EY, Marrison ST, Vu VT. Hypoxia and radiation therapy: past history, ongoing research, and future promise. Curr Mol Med. 2009;9(4):442-458.
    doi pubmed pmc
  64. Fokas E, Im JH, Hill S, Yameen S, Stratford M, Beech J, Hackl W, et al. Dual inhibition of the PI3K/mTOR pathway increases tumor radiosensitivity by normalizing tumor vasculature. Cancer Res. 2012;72(1):239-248.
    doi pubmed
  65. Pan WL, Wong JH, Fang EF, Chan YS, Ng TB, Cheung RC. Preferential cytotoxicity of the type I ribosome inactivating protein alpha-momorcharin on human nasopharyngeal carcinoma cells under normoxia and hypoxia. Biochem Pharmacol. 2014;89(3):329-339.
    doi pubmed pmc
  66. Kalluri R. The biology and function of fibroblasts in cancer. Nat Rev Cancer. 2016;16(9):582-598.
    doi pubmed
  67. Fang Z, Meng Q, Xu J, Wang W, Zhang B, Liu J, Liang C, et al. Signaling pathways in cancer-associated fibroblasts: recent advances and future perspectives. Cancer Commun (Lond). 2023;43(1):3-41.
    doi pubmed pmc
  68. Huang W, Zhang L, Yang M, Wu X, Wang X, Huang W, Yuan L, et al. Cancer-associated fibroblasts promote the survival of irradiated nasopharyngeal carcinoma cells via the NF-kappaB pathway. J Exp Clin Cancer Res. 2021;40(1):87.
    doi pubmed pmc
  69. Kierans SJ, Taylor CT. Regulation of glycolysis by the hypoxia-inducible factor (HIF): implications for cellular physiology. J Physiol. 2021;599(1):23-37.
    doi pubmed
  70. Pugachev A, Ruan S, Carlin S, Larson SM, Campa J, Ling CC, Humm JL. Dependence of FDG uptake on tumor microenvironment. Int J Radiat Oncol Biol Phys. 2005;62(2):545-553.
    doi pubmed
  71. Mo Y, Wang Y, Zhang S, Xiong F, Yan Q, Jiang X, Deng X, et al. Circular RNA circRNF13 inhibits proliferation and metastasis of nasopharyngeal carcinoma via SUMO2. Mol Cancer. 2021;20(1):112.
    doi pubmed pmc
  72. Li Y, Patel SP, Roszik J, Qin Y. Hypoxia-driven immunosuppressive metabolites in the tumor microenvironment: new approaches for combinational immunotherapy. Front Immunol. 2018;9:1591.
    doi pubmed pmc
  73. Noman MZ, Hasmim M, Messai Y, Terry S, Kieda C, Janji B, Chouaib S. Hypoxia: a key player in antitumor immune response. A Review in the Theme: Cellular Responses to Hypoxia. Am J Physiol Cell Physiol. 2015;309(9):C569-579.
    doi pubmed pmc
  74. White JR, Harris RA, Lee SR, Craigon MH, Binley K, Price T, Beard GL, et al. Genetic amplification of the transcriptional response to hypoxia as a novel means of identifying regulators of angiogenesis. Genomics. 2004;83(1):1-8.
    doi pubmed
  75. Facciabene A, Peng X, Hagemann IS, Balint K, Barchetti A, Wang LP, Gimotty PA, et al. Tumour hypoxia promotes tolerance and angiogenesis via CCL28 and T(reg) cells. Nature. 2011;475(7355):226-230.
    doi pubmed
  76. Huang Y, Kim BYS, Chan CK, Hahn SM, Weissman IL, Jiang W. Improving immune-vascular crosstalk for cancer immunotherapy. Nat Rev Immunol. 2018;18(3):195-203.
    doi pubmed pmc
  77. Tian L, Goldstein A, Wang H, Ching Lo H, Sun Kim I, Welte T, Sheng K, et al. Mutual regulation of tumour vessel normalization and immunostimulatory reprogramming. Nature. 2017;544(7649):250-254.
    doi pubmed pmc
  78. de la Cruz-Lopez KG, Castro-Munoz LJ, Reyes-Hernandez DO, Garcia-Carranca A, Manzo-Merino J. Lactate in the regulation of tumor microenvironment and therapeutic approaches. Front Oncol. 2019;9:1143.
    doi pubmed pmc
  79. Blake SJ, Dougall WC, Miles JJ, Teng MW, Smyth MJ. Molecular pathways: targeting CD96 and TIGIT for cancer immunotherapy. Clin Cancer Res. 2016;22(21):5183-5188.
    doi pubmed
  80. Huang SCM, Tsao SW, Tsang CM. Interplay of viral infection, host cell factors and tumor microenvironment in the pathogenesis of nasopharyngeal carcinoma. Cancers (Basel). 2018;10(4):106.
    doi pubmed pmc


This article is distributed under the terms of the Creative Commons Attribution Non-Commercial 4.0 International License, which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.


World Journal of Oncology is published by Elmer Press Inc.

 

Browse  Journals  

 

Journal of Clinical Medicine Research

Journal of Endocrinology and Metabolism

Journal of Clinical Gynecology and Obstetrics

 

World Journal of Oncology

Gastroenterology Research

Journal of Hematology

 

Journal of Medical Cases

Journal of Current Surgery

Clinical Infection and Immunity

 

Cardiology Research

World Journal of Nephrology and Urology

Cellular and Molecular Medicine Research

 

Journal of Neurology Research

International Journal of Clinical Pediatrics

 

 
       
 

World Journal of Oncology, bimonthly, ISSN 1920-4531 (print), 1920-454X (online), published by Elmer Press Inc.                     
The content of this site is intended for health care professionals.
This is an open-access journal distributed under the terms of the Creative Commons Attribution-NonCommercial 4.0 International License, which permits unrestricted
non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.
Creative Commons Attribution license (Attribution-NonCommercial 4.0 International CC-BY-NC 4.0)


This journal follows the International Committee of Medical Journal Editors (ICMJE) recommendations for manuscripts submitted to biomedical journals,
the Committee on Publication Ethics (COPE) guidelines, and the Principles of Transparency and Best Practice in Scholarly Publishing.

website: www.wjon.org   editorial contact: editor@wjon.org
Address: 9225 Leslie Street, Suite 201, Richmond Hill, Ontario, L4B 3H6, Canada

© Elmer Press Inc. All Rights Reserved.


Disclaimer: The views and opinions expressed in the published articles are those of the authors and do not necessarily reflect the views or opinions of the editors and Elmer Press Inc. This website is provided for medical research and informational purposes only and does not constitute any medical advice or professional services. The information provided in this journal should not be used for diagnosis and treatment, those seeking medical advice should always consult with a licensed physician.