Skip to main content

Unveiling immune resistance mechanisms in nasopharyngeal carcinoma and emerging targets for antitumor immune response: tertiary lymphoid structures

Abstract

Nasopharyngeal carcinoma (NPC) is a prevalent malignancy in China, commonly associated with undifferentiated cell types and Epstein-Barr virus (EBV) infection. The presence of intense lymphocytic infiltration and elevated expression of programmed cell death ligand 1(PD-L1) in NPC highlights its potential for immunotherapy, yet current treatment outcomes remain suboptimal. In this review, we explore the tumor microenvironment of NPC to better understand the mechanisms of resistance to immunotherapy, evaluate current therapeutic strategies, and pinpoint emerging targets, such as tertiary lymphoid structures (TLSs), that could enhance treatment outcomes and prognostic accuracy. TLSs have demonstrated positive prognostic value in NPC, making them a promising target for future therapies. This review summarizes the key characteristics of TLSs and latest research in the context of NPC. We are optimistic that targeting TLSs could improve immunotherapy outcomes for NPC patients, ultimately leading to more effective treatment strategies and better patient survival.

Introduction

Nasopharyngeal carcinoma (NPC) originates from the nasopharyngeal mucosal layer, and is categorized into keratinized, non-keratinized, and basal-like. It is notably prevalent in Asia, with the non-keratinized variant being significantly associated with Epstein-Barr virus (EBV) infection [1]. Although NPC demonstrates sensitivity to radiotherapy, late-stage diagnosis and absence of early detection biomarkers lead to suboptimal outcomes, which are further compounded by treatment complications [1, 2]. The standard treatment for NPC typically involves chemoradiotherapy [3], but relapse rates remain high. Platinum-based chemotherapy remains the conventional approach, underscoring the need to identify patients who are most likely to respond favorably [4].

With the advancements in tumor research, immunotherapy has gained prominence and has sparked significant interest in its application to NPC. Current immunotherapy strategies for NPC include active immunotherapy, adoptive immunotherapy, and immune checkpoint inhibitors. Regarding active immunotherapy, EBV infection is a known contributor to cancer development, making it a promising therapeutic target. Research is currently focused on preventive vaccines against EBV to reduce the incidence of EBV-related diseases by limiting viral infection. Vaccine targets include EBV glycoproteins gp350, gH/gL, gB, gp42 serve as vaccine targets [5]. Additionally, EBV cancer proteins like EBNA and LMP are being explored as targets to eradicate latent infection within the host [6]. Moreover, injection of EBV-associated antigens or dendritic cells (DCs) capable of presenting these antigens (antigen-presenting cells, APCs) has shown potential in amplifying EBV antigen-specific CD8+T cells [7]. Adoptive immunotherapy involves the ex vivo culture of autologous CD8+T cells, which were then reintroduced into the body to target EBV-specific antigens such as Epstein-barr virus nuclear antigen (EBNA) and Latent membrane protein (LMP). Studies have demonstrated that this immune response to EBV can produces long-lasting anti-tumor effects [8]. Additionally, adoptive immunotherapy is associated with fewer adverse effects compared to conventional therapies [9]. Immune checkpoint inhibitors (ICIs) work by restoring the immune activity of immune cells, and countering the immunosuppression induced by tumor cells [10].

Major immune checkpoints, such as cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) and PD-L1, inhibit lymphocytes activity, allowing immune cells to evade immune detection. In NPC, PD-L1 expression is prevalent and associated with a poor prognosis. Anti-PD1/PDL-1 therapies have shown promising results in clinical trials, significantly prolonging survival in patients with recurrent/metastatic platinum-refractory NPC. However, resistance to PD-1/PD-L1 inhibitors may develop through T cell dysfunction and decreased immunogenicity [10].

Face with the challenges of NPC immunotherapy, we conducted annalysis to understand the causes of resistance. In our exploration of potential new target for NPC immunotherapy, we identified TLSs, which may offer a novel approach for improving immunotherapy outcomes. Typically absent under physiological conditions, TLSs are frequently found in chronically inflamed environment. Mature TLSs characterized by the presence of T cells (CD 4 + T follicular helper cells), B cells and follicular dendritic cells (FDCs). scattered macrophages, stromal cells [11], and high endothelial venules (HEVs) at the periphery [12]. TLSs, organized aggregate of immune cells in non-lymphoid tissues, have been identified in the pathological sections of some NPC patients and have been correlated with improved prognosis [13].

The resistance mechanism of immunotherapy

The emergence of drug resistance presents a significant challenge in the treatment of recurrent NPC [14]. The TME comprises a diverse range of cellular components, including infiltrating immune cells, cancer-associated fibroblasts (CAFs), endothelial cells, pericytes, and various non-cellular elements. Within the TME, there is a delicate balance between immunostimulatory and immunosuppressive factors [15].Upon the death of tumor cells, antigens are released, captured, and presented by antigen-presenting cells (APCs), which trigger the activation of T cells. These activated T cells then migrate to the tumor site, infiltrate and identify tumor cells, leading to their elimination. This initiates an immune cycle within the TME [15]. During the anti-tumor immune response, TLSs form, and the appearance of germinal centers signals their maturation. Plasma cells, derived from B cell differentiation, produce antibodies that induce apoptosis in tumor cells. The formation of antigen-antibody complexes enhances dendritic cell-mediated antigen presentation and reinforces memory cell responses, thereby amplifying the tumor immune cycle [16](Fig. 1). Despite the considerable potential of immunotherapy in NPCs, current clinical outcomes remain suboptimal [17,18,19]. The possible mechanisms underlying immunotherapy resistance in NPC are summarized in Table 1.

Fig. 1
figure 1

Tumor-immune circulation. The main process of tumor immunity is that dying cells release tumor cell antigens, and APCs initiate and activate T cells through antigen presentation. Activated T cells and other immune related cells are transported to the tumor site and infiltrate to recognize tumor cells, ultimately killing the tumor cells. The immune cycle process that forms the microenvironment. During the anti-tumor immune process, TLSs are formed, and the formation of germinal centers marks the maturation of TLSs, which play an effective anti-tumor response. B cells differentiate into plasma cells and secrete antibodies to promote tumor apoptosis. The formed antigen antibody complexes can amplify the antigen presentation effect of APCs, enhance memory of B cell responses, and ultimately amplify the immune cycle within the tumor

Table 1 Mechanism of drug resistance in NPC

Resistance related to EBV infection is particularly important. As a microbial component in the TME, EBV significantly influences tumor growth, invasion, and metastasis [20]. Compared to EBV-negative NPC, EBV-positive cases exhibit a stronger immunosuppressive microenvironment [21], facilitating immune evasion and tumor progression. EBV predominantly adopts type II latency, expressing proteins such as EBNA1, LMP1/2 (late membrane proteins), and EBER (small RNA encoded by EBV) [1]. EBNA1 downregulates P53, leading to gene mutations in cancer cells [22]. In advanced tumors, TGF-β1 promotes the chemotactic migration of regulatory T cells (Tregs) via the CXCL12-CXCR4 axis, reshaping the NPC microenvironment [23]. LMP1 stimulates epidermal growth factor receptor (EGFR), inhibits epithelial differentiation, and promotes cell cycle progression, thereby enhancing cell growth. Additionally, LMP1 activates NF-κB by binding to tumor necrosis factor (TNF) receptor-associated factor (TRAF), amplifying the immune response of lymphocytes and inhibiting DNA repair through the PI3K/Akt pathway [24]. TGF-βfurther suppresses the immune response while enhancing proliferation signals [25]. LMP2A promotes tumor transformation through activation of the Ras/PI3K/Akt and β-catenin/Wnt pathways [26]. LMP1 also activates the classic RAF/MEK/ERK pathway upon binding to receptors such as EGFR, FGFR, and VEGFR, inducing epithelial to mesenchymal transition (EMT) and increasing cell motility and invasiveness [27]. The JAK/STAT3 pathway inhibits apoptosis, promotes cell proliferation, and facilitates angiogenesis, thereby enhancing NPC’s invasive capabilities [28]. We have summarized and illustrated the pathway of immunosuppression induced by EBV-positive tumor cells in Fig. 2 [22,23,24,25,26,27,28].

Fig. 2
figure 2

Immunoresistance mechanisms related pathways in EBV + NPC. The interaction between the Epstein-Barr virus (EBV) glycoprotein BMRF-2 or gH/gL and integrins on epithelial cells alters the conformation of gH/gL and gB, leading to the fusion of the EBV envelope with epithelial cells. Ultimately, EBV is delivered into the cells. EBV commonly adopts type II latency, expressing like EBNA1, LMP1/2, and EBER. These oncoproteins manipulate various cellular pathways (such as P53, TGF-β1, EGFR, NF-κB, PI3K/Akt, RAF/MEK/ERK, JAK/STAT3), promoting tumor proliferation, survival, invasion

Recent advancements in oncology underscore the crucial role of TME, particularly in immune modulation [29, 30]. Immune cells infiltration correlates with favorable prognoses, with the T cells, B cells, and stromal cells increasingly recognized for their influence on therapeutic responses [31, 32]. Despite the promise of immunotherapy, resistance remains a significant challenge, particularly in NPC [17, 33]. The expression of PD-L1 in EBV-associated NPC suggests PD-1/PD-L1 inhibitors may serve as effective therapies, with varied prognostic implications [34,35,36]. Understanding the TME, including TLSs, offers valuable insights into potentialimmunotherapy targets for NPC.

TLSs as the targets for antitumor immune response

Effective induction of an anti-tumor immune response within the TME is crucial for inhibiting tumor progression. However, despite a similar composition of infiltrating immune cell in certain tumors, patients exhibit varying prognosis, suggesting that interactions between immune cells and tumors extend mere presence. TLSs have emerged as significant factors to the anti-tumor immune response in NPC and hold considerable promise for immunotherapy [37].

Composition of TLSs

Histopathological analysis of NPC reveals extensive infiltration of immune cells surrounding and within tumor lesion [31], indicating a complex TME. Various methods for detecting TLSs are outlined in Table 2. The composition of TLSs varies across in different tumors [38]. For example, TLSs in non-small cell lung cancer primarily contains DC-LAMP + DCs [39], while TFH cells are common in TLSs associated with breast cancer [11]. In NPC, studies have identified a combination of markers of TLSs components, including germinal center B cell components, follicle-assisted CD4 + T cells, CXCL13 + CD8 + T cells, CXCL13 + fibroblasts. This leads us to consider whether the differences in TLSs components across various tumors may be a factor contributing to the variability in prognostic outcomes associated with TLSs.

Table 2 The advantages and features of TLSs observation methods

The function of TLSs

The function of TLSs varies among different tumor types, which may be attributed to differences in TLSs differentiation, spatial location and overall function [37, 40]. Although consistent evaluation criteria for TLSs are lacking, it is widely accepted that the maturity of TLSs significantly influences their functionality [1]. The germinal center within TLSs plays a crucial role in tumor prognosis and has emerged as an independent prognostic marker in numerous solid tumors [41]. In tumor immunotherapy, an increasing number of studies have highlighted the important role of B cells in tumor immunity [42]. In immature TLSs, B cells may acquire immunosuppressive activity, however, in mature germinal centers, B cells undergo processes such as antibody class switching and somatic hypermutation to differentiate into mature plasma cells, thereby promoting anti-tumor immune responses [43]. Research has shown that infiltrated B cells were closely associated with TLSs [44]. Both infiltrating B cells and/or TLSs are considered valuable prognostic or predictive biomarkers [45].

Studies analyzing gene expression differences between normal nasopharyngeal tissue and NPC tissue have identified a close association between tumor-infiltrating B cells and NPC. The infiltration of B cells in NPC tissues was significantly higher than that in adjacent tissues, with specific B cell marker genes, such as CD79B and CD19, positively correlating with favorable prognosis in NPC [44]. However, not all B cell subsets are linked to positive outcomes in NPC. Some subsets expressing activation-related genes, as well as high-level transcription factor key genes (e.g., JUN, JUNB, FOS, and FOSB), which play critical roles in the immune microenvironment of NPC, are associated with better prognoses and may serve as potential targets for clinical immunotherapy. In contrast, subsets expressing proliferation-related genes (e.g., MKI67, TOP2A) exhibit poorer prognoses and fewer TLSs formations [44]. Cancer-associated fibroblasts (CAFs) can promote B cell aggregation and communication through secretion of CXCL13 and MIF signaling, respectively [46]. In NPC, often associated with viral infections such as Epstein-Barr virus (EBV), a significant positive correlation exists between EBV infection-specific genes (including genes specific to EBV infected cells and LMP1-related gene signatures) and TLSs characteristics [44]. EBV-positive patients tend to display higher levels of TLSs compared to EBV-negative patients, correlating with a better therapeutic response and overall prognosis. In other virus-associated cancers like EBV-positive gastric cancer [47] and human papillomavirus(HPV)-positive head and neck squamous cell carcinoma (HNSCC) [48], viral infections influence the composition of the TME, thereby affecting the formation of TLSs. For instance, EBV alters immune infiltration characteristics in gastric cancer tissues, while SEMA4A, a membrane-bound glycoprotein, plays a central role in the formation of immune aggregates in HPV-positive HNSCC [48].

The therapeutic potential of TLSs in NPC

The presence of TLSs within the TME of NPC has emerged as a significant factor influencing anti-tumor immune responses and patient outcomes. TLSs are organized aggregates of immune cells formed in non-lymphoid tissues and play a crucial role in promoting anti-tumor immunity. Therefore, inducing the formation of TLSs is a particularly promising immunotherapy. Research has identified several factors associated with the formation and function of TLSs in NPC (Fig. 3).

Fig. 3
figure 3

The therapeutic potential of TLSs in NPC. A. Modulating the immune cells-induced TLSs formation: The subsets CD4 + Th-CXCL13PD1 + CXCR5-cells in NPC can promote TLSs formation; B. Nano-vaccine injection-induced TLSs formation: The nano-vaccine activated the LT-α and LT-β pathways, and subsequently enhanced the expression of downstream chemokines CCL19/CCL21, CXCL10 and CXCL13 in the tumor microenvironment to promote the formation of TLSs; C. Chemotherapy-induced TLSs formation: GP chemotherapy induces ILB(innate-like B cell) through toll-like receptor 9 signaling. ILB further amplifies follicular helper cells and helper type 1 T cells via the ICOS-ICOS axis and subsequently enhances cytotoxic T cells in tertiary lymphoid organ-like structures with germinal center defects after chemotherapy; D. TLSs and ICB therapy are mutually beneficial

TLSs associated immune cells

PD-1+CXCR5-CD4+TH-CXCL13 cells found in the TME of NPC samples play a critical role in recruiting tumor-associated B cells to participate in TLSs formation. These cells express the chemokine CXCL13, which attracts B cells, and secrete interleukin-21 (IL-21), promoting the differentiation of plasma cells and production of immunoglobulins. TGF-β and the transcription factor SOX-4 have been identified as key regulators of CXCL13 expression in these CD4 + TH cells. TLSs formed as a result significantly enhance the anti-tumor immune response and improve patient survival [13].

TLSs related nano-vaccines

In NPC mouse models, injecting a nano-vaccine composed of EBV’s EBNA1, combined with a bi-adjuvant of Mn2 + and cytosine-phosphate-guanine (CpG) formulated with tannic acid, has been shown to facilitate TLSs formation within tumors. This approach accelerates the anti-tumor response and inhibits tumor growth [49]. Nano-vaccines represent a promising avenue for future immunotherapy by inducing and accelerating TLSs formation in tumors.

Chemotherapy-induced TLSs formation

Gemcitabine combined with cisplatin (GP) chemotherapy is the standard treatment for nasopharyngeal carcinoma (NPC). Chemotherapy regimens such as GP, have been found to induce immune cell infiltration and form tertiary lymphoid organ (TLO)-like structures that may lack germinal centers. However, these GC-deficient TLOs may still hold clinical significance. In lung [43] and pancreatic [50] cancer patients who received neoadjuvant chemotherapy, an increase in immune cell composition suggested enhanced TLSs formation. These findings highlight the potential of chemotherapy in modulating the TME to enhance anti-tumor immunity [51], indicating that therapeutic interventions could reconstruct B cells to establish an effective anti-tumor response.

TLSs correlation with immune checkpoint blockade (ICB) therapy

The presence of TLSs has been significantly associated with improved responses to ICB therapy in various tumors. Studies have shown the emergence of TLSs following treatment with PD-1 or combined PD-1 and CTLA-4 blockade inhibitors [42]. The abundance of TLSs has been correlated with PD-L1 expression on immune cells [52]. Additionally, ICB therapy has been shown to enhance TLSs formation and function in many tumors, including melanoma and urothelial carcinoma [42]. By promoting TLSs formation, ICB therapy can further enhance its therapeutic efficacy [53]. Formation of TLSs was also found to be beneficial to the prognosis of patients in NPC [13].Therefore, inducing TLSs in NPC may improve the effectiveness of ICB treatment.

Summary and prospect

The TME plays a crucial role in the prognosis and treatment of NPC, particularly regarding immunotherapy. Understanding the complex interplay between tumor cells, immune cells, and the surrounding microenvironment is essential for developing effective immunotherapy strategies for NPC. Future research should focus on elucidating the mechanisms of treatment resistance and identifying novel targets within the tumor microenvironment. Given the challenges of immunotherapy resistance in NPC, studies have identified that TLSs as potential targets to enhance the treatment effect in patients. Investigating their formation, function, and correlation with treatment outcomes could provide valuable insights into optimizing immunotherapy approaches, including predicting treatment responses and exploring novel drugs targeting TLSs. Additionally, CXCL-13 shows promise as a novel target in NPC immunotherapy. Preliminary analyses suggest its association with favorable prognosis and its potential as a predictor of response to immune checkpoint blockade therapy in NPC. Further research is needed to elucidate its mechanisms of action and explore its therapeutic potential in combination with other targets, such as PD-L1. It is essential to recognize the unique features of NPC’s TME, including strong immune infiltration and high PD-L1 expression. Strategies for immunotherapy should be tailored to these specific characteristics rather than relying solely on conclusions drawn from other tumor types. Despite advancements in immunotherapy, resistance remains a significant challenge in NPC treatment. Continued research into novel targets and therapeutic approaches is crucial for overcoming resistance and improving patient outcomes.

In summary, further exploration of the TME, TLSs, CXCL-13, and other potential targets holds promise for advancing immunotherapy in NPC. By understanding the unique characteristics of NPC and developing targeted therapeutic strategies, we can enhance treatment efficacy and provide new hope for patients facing this challenging disease.

Data availability

Not applicable.

Abbreviations

NPC:

Nasopharyngeal carcinoma

EBV:

Epstein-barr virus

TLSs:

Tertiary lymphoid structures

TME:

Tumor microenvironment

CAF:

Cancer-associated fibroblast

DC:

Dendritic cell

APC:

Antigen-presenting cell

EBNA:

Epstein-barr virus nuclear antigen

LMP:

Latent membrane protein

ACT:

Adoptive cell therapy

MDSCs:

Myeloid-derived suppressor cells

ICIs:

Immune checkpoint inhibitors

CTLA-4:

Cytotoxic T lymphocyte-associated antigen-4

PD-L1:

Programmed cell death 1 ligand 1

TME:

Tumor microenvironment

TFH cell:

T follicular helper cell

FDCs:

Follicular dendritic cells

HEVs:

High endothelial venules

HPV:

Human Papillomavirus

HNSCC:

Head and Neck Squamous Cell Carcinoma

IL-21:

interleukin-21

CXCL-13:

C-X-C motif chemokine ligand 13

CCL21:

Chemokine (C-C motif) ligand 21

ICB:

Immune checkpoint blockade

PDAC:

Pancreatic ductal adenocarcinoma

References

  1. Chen YP, Chan ATC, Le QT, Blanchard P, Sun Y, Ma J. Nasopharyngeal carcinoma. Lancet. 2019;394:64–80.

    Article  PubMed  Google Scholar 

  2. Zhang L, Chen QY, Liu H, Tang LQ, Mai HQ. Emerging treatment options for nasopharyngeal carcinoma. Drug Des Devel Ther. 2013;7:37–52.

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Chen YP, Ismaila N, Chua MLK, Colevas AD, Haddad R, Huang SH, Wee JTS, Whitley AC, Yi JL, Yom SS, et al. Chemotherapy in Combination with Radiotherapy for definitive-intent treatment of stage II-IVA nasopharyngeal carcinoma: CSCO and ASCO Guideline. J Clin Oncol. 2021;39:840–59.

    Article  CAS  PubMed  Google Scholar 

  4. Jiang Y, Chen C, Liu G, Fang T, Lu N, Bei W, Dong S, Li W, Xia W, Liang H, Xiang Y. Combination strategy exploration for prior treated recurrent or metastatic nasopharyngeal carcinoma in the era of immunotherapy. Sci Rep. 2024;14:1768.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Zhao B, Zhang X, Krummenacher C, Song S, Gao L, Zhang H, Xu M, Feng L, Feng Q, Zeng M, et al. Immunization with Fc-Based recombinant Epstein-Barr Virus gp350 elicits potent neutralizing Humoral Immune response in a BALB/c mice Model. Front Immunol. 2018;9:932.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Hui EP, Taylor GS, Jia H, Ma BB, Chan SL, Ho R, Wong WL, Wilson S, Johnson BF, Edwards C, et al. Phase I trial of recombinant modified vaccinia ankara encoding Epstein-Barr viral tumor antigens in nasopharyngeal carcinoma patients. Cancer Res. 2013;73:1676–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Palucka K, Banchereau J. Cancer immunotherapy via dendritic cells. Nat Rev Cancer. 2012;12:265–77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Li J, Chen QY, He J, Li ZL, Tang XF, Chen SP, Xie CM, Li YQ, Huang LX, Ye SB, et al. Phase I trial of adoptively transferred tumor-infiltrating lymphocyte immunotherapy following concurrent chemoradiotherapy in patients with locoregionally advanced nasopharyngeal carcinoma. Oncoimmunology. 2015;4:e976507.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Liu H, Tang L, Li Y, Xie W, Zhang L, Tang H, Xiao T, Yang H, Gu W, Wang H, Chen P. Nasopharyngeal carcinoma: current views on the tumor microenvironment’s impact on drug resistance and clinical outcomes. Mol Cancer. 2024;23:20.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Jiang Y, Zhan H. Communication between EMT and PD-L1 signaling: new insights into tumor immune evasion. Cancer Lett. 2020;468:72–81.

    Article  PubMed  Google Scholar 

  11. Gu-Trantien C, Migliori E, Buisseret L, de Wind A, Brohée S, Garaud S, Noël G, Dang Chi VL, Lodewyckx JN, Naveaux C et al. CXCL13-producing TFH cells link immune suppression and adaptive memory in human breast cancer. JCI Insight 2017, 2.

  12. Ager A. High endothelial venules and other blood vessels: critical regulators of lymphoid Organ development and function. Front Immunol. 2017;8:45.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Li JP, Wu CY, Chen MY, Liu SX, Yan SM, Kang YF, Sun C, Grandis JR, Zeng MS, Zhong Q. PD-1(+)CXCR5(-)CD4(+) Th-CXCL13 cell subset drives B cells into tertiary lymphoid structures of nasopharyngeal carcinoma. J Immunother Cancer 2021, 9.

  14. Kachalaki S, Ebrahimi M, Mohamed Khosroshahi L, Mohammadinejad S, Baradaran B. Cancer chemoresistance; biochemical and molecular aspects: a brief overview. Eur J Pharm Sci. 2016;89:20–30.

    Article  CAS  PubMed  Google Scholar 

  15. Mellman I, Chen DS, Powles T, Turley SJ. The cancer-immunity cycle: indication, genotype, and immunotype. Immunity. 2023;56:2188–205.

    Article  CAS  PubMed  Google Scholar 

  16. Fridman WH, Meylan M, Pupier G, Calvez A, Hernandez I, Sautès-Fridman C. Tertiary lymphoid structures and B cells: an intratumoral immunity cycle. Immunity. 2023;56:2254–69.

    Article  CAS  PubMed  Google Scholar 

  17. Liu Z, He J, Hu X. Ferroptosis regulators related scoring system by Gaussian finite mixture model to predict prognosis and immunotherapy efficacy in nasopharyngeal carcinoma. Front Genet. 2022;13:975190.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Hsu C, Lee SH, Ejadi S, Even C, Cohen RB, Le Tourneau C, Mehnert JM, Algazi A, van Brummelen EMJ, Saraf S, et al. Safety and Antitumor Activity of Pembrolizumab in patients with programmed death-ligand 1-Positive nasopharyngeal carcinoma: results of the KEYNOTE-028 study. J Clin Oncol. 2017;35:4050–6.

    Article  CAS  PubMed  Google Scholar 

  19. Fang W, Yang Y, Ma Y, Hong S, Lin L, He X, Xiong J, Li P, Zhao H, Huang Y, et al. Camrelizumab (SHR-1210) alone or in combination with gemcitabine plus cisplatin for nasopharyngeal carcinoma: results from two single-arm, phase 1 trials. Lancet Oncol. 2018;19:1338–50.

    Article  CAS  PubMed  Google Scholar 

  20. Helmink BA, Khan MAW, Hermann A, Gopalakrishnan V, Wargo JA. The microbiome, cancer, and cancer therapy. Nat Med. 2019;25:377–88.

    Article  CAS  PubMed  Google Scholar 

  21. Ruffin AT, Li H, Vujanovic L, Zandberg DP, Ferris RL, Bruno TC. Improving head and neck cancer therapies by immunomodulation of the tumour microenvironment. Nat Rev Cancer. 2023;23:173–88.

    Article  CAS  PubMed  Google Scholar 

  22. Capuozzo M, Santorsola M, Bocchetti M, Perri F, Cascella M, Granata V, Celotto V, Gualillo O, Cossu AM, Nasti G et al. p53: from Fundamental Biology to Clinical Applications in Cancer. Biology (Basel) 2022, 11.

  23. Huo S, Luo Y, Deng R, Liu X, Wang J, Wang L, Zhang B, Wang F, Lu J, Li X. EBV-EBNA1 constructs an immunosuppressive microenvironment for nasopharyngeal carcinoma by promoting the chemoattraction of Treg cells. J Immunother Cancer 2020, 8.

  24. Zhu QY, Zhao GX, Li Y, Talakatta G, Mai HQ, Le QT, Young LS, Zeng MS. Advances in pathogenesis and precision medicine for nasopharyngeal carcinoma. MedComm (2020) 2021, 2:175–206.

  25. Zhu L, Wang Y, Yuan X, Ma Y, Zhang T, Zhou F, Yu G. Effects of immune inflammation in head and neck squamous cell carcinoma: Tumor microenvironment, drug resistance, and clinical outcomes. Front Genet. 2022;13:1085700.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Fukuda M, Longnecker R. Epstein-Barr virus latent membrane protein 2A mediates transformation through constitutive activation of the Ras/PI3-K/Akt Pathway. J Virol. 2007;81:9299–306.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Morris MA, Laverick L, Wei W, Davis AM, O’Neill S, Wood L, Wright J, Dawson CW, Young LS. The EBV-Encoded Oncoprotein, LMP1, induces an epithelial-to-mesenchymal transition (EMT) via its CTAR1 domain through integrin-mediated ERK-MAPK signalling. Cancers (Basel) 2018, 10.

  28. Wang Z, Luo F, Li L, Yang L, Hu D, Ma X, Lu Z, Sun L, Cao Y. STAT3 activation induced by Epstein-Barr virus latent membrane protein1 causes vascular endothelial growth factor expression and cellular invasiveness via JAK3 and ERK signaling. Eur J Cancer. 2010;46:2996–3006.

    Article  CAS  PubMed  Google Scholar 

  29. Gourzones C, Barjon C, Busson P. Host-tumor interactions in nasopharyngeal carcinomas. Semin Cancer Biol. 2012;22:127–36.

    Article  CAS  PubMed  Google Scholar 

  30. Shen YC, Hsu CL, Jeng YM, Ho MC, Ho CM, Yeh CP, Yeh CY, Hsu MC, Hu RH, Cheng AL. Reliability of a single-region sample to evaluate tumor immune microenvironment in hepatocellular carcinoma. J Hepatol. 2020;72:489–97.

    Article  CAS  PubMed  Google Scholar 

  31. Wang YQ, Chen YP, Zhang Y, Jiang W, Liu N, Yun JP, Sun Y, He QM, Tang XR, Wen X, et al. Prognostic significance of tumor-infiltrating lymphocytes in nondisseminated nasopharyngeal carcinoma: a large-scale cohort study. Int J Cancer. 2018;142:2558–66.

    Article  CAS  PubMed  Google Scholar 

  32. Fridman WH, Zitvogel L, Sautès-Fridman C, Kroemer G. The immune contexture in cancer prognosis and treatment. Nat Rev Clin Oncol. 2017;14:717–34.

    Article  CAS  PubMed  Google Scholar 

  33. Chow JC, Ngan RK, Cheung KM, Cho WC. Immunotherapeutic approaches in nasopharyngeal carcinoma. Expert Opin Biol Ther. 2019;19:1165–72.

    Article  CAS  PubMed  Google Scholar 

  34. Chen BJ, Chapuy B, Ouyang J, Sun HH, Roemer MG, Xu ML, Yu H, Fletcher CD, Freeman GJ, Shipp MA, Rodig SJ. PD-L1 expression is characteristic of a subset of aggressive B-cell lymphomas and virus-associated malignancies. Clin Cancer Res. 2013;19:3462–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Lee VH, Lo AW, Leung CY, Shek WH, Kwong DL, Lam KO, Tong CC, Sze CK, Leung TW. Correlation of PD-L1 expression of Tumor cells with survival outcomes after Radical Intensity-Modulated Radiation Therapy for non-metastatic nasopharyngeal carcinoma. PLoS ONE. 2016;11:e0157969.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Zhu Q, Cai MY, Chen CL, Hu H, Lin HX, Li M, Weng DS, Zhao JJ, Guo L, Xia JC. Tumor cells PD-L1 expression as a favorable prognosis factor in nasopharyngeal carcinoma patients with pre-existing intratumor-infiltrating lymphocytes. Oncoimmunology. 2017;6:e1312240.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Sautès-Fridman C, Petitprez F, Calderaro J, Fridman WH. Tertiary lymphoid structures in the era of cancer immunotherapy. Nat Rev Cancer. 2019;19:307–25.

    Article  PubMed  Google Scholar 

  38. Colbeck EJ, Ager A, Gallimore A, Jones GW. Tertiary lymphoid structures in Cancer: drivers of Antitumor Immunity, Immunosuppression, or Bystander sentinels in Disease? Front Immunol. 2017;8:1830.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Goc J, Germain C, Vo-Bourgais TK, Lupo A, Klein C, Knockaert S, de Chaisemartin L, Ouakrim H, Becht E, Alifano M, et al. Dendritic cells in tumor-associated tertiary lymphoid structures signal a Th1 cytotoxic immune contexture and license the positive prognostic value of infiltrating CD8 + T cells. Cancer Res. 2014;74:705–15.

    Article  CAS  PubMed  Google Scholar 

  40. Schumacher TN, Thommen DS. Tertiary lymphoid structures in cancer. Science. 2022;375:eabf9419.

    Article  CAS  PubMed  Google Scholar 

  41. Siliņa K, Soltermann A, Attar FM, Casanova R, Uckeley ZM, Thut H, Wandres M, Isajevs S, Cheng P, Curioni-Fontecedro A, et al. Germinal centers determine the prognostic relevance of Tertiary lymphoid structures and are impaired by corticosteroids in Lung squamous cell carcinoma. Cancer Res. 2018;78:1308–20.

    Article  PubMed  Google Scholar 

  42. Helmink BA, Reddy SM, Gao J, Zhang S, Basar R, Thakur R, Yizhak K, Sade-Feldman M, Blando J, Han G, et al. B cells and tertiary lymphoid structures promote immunotherapy response. Nature. 2020;577:549–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Germain C, Gnjatic S, Tamzalit F, Knockaert S, Remark R, Goc J, Lepelley A, Becht E, Katsahian S, Bizouard G, et al. Presence of B cells in tertiary lymphoid structures is associated with a protective immunity in patients with lung cancer. Am J Respir Crit Care Med. 2014;189:832–44.

    Article  CAS  PubMed  Google Scholar 

  44. Chen C, Zhang Y, Wu X, Shen J. The role of tertiary lymphoid structure and B cells in nasopharyngeal carcinoma: based on bioinformatics and experimental verification. Transl Oncol. 2024;41:101885.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Fridman WH, Meylan M, Petitprez F, Sun CM, Italiano A, Sautès-Fridman C. B cells and tertiary lymphoid structures as determinants of tumour immune contexture and clinical outcome. Nat Rev Clin Oncol. 2022;19:441–57.

    Article  CAS  PubMed  Google Scholar 

  46. Klasen C, Ziehm T, Huber M, Asare Y, Kapurniotu A, Shachar I, Bernhagen J, El Bounkari O. LPS-mediated cell surface expression of CD74 promotes the proliferation of B cells in response to MIF. Cell Signal. 2018;46:32–42.

    Article  CAS  PubMed  Google Scholar 

  47. Qiu MZ, Wang C, Wu Z, Zhao Q, Zhao Z, Huang CY, Wu W, Yang LQ, Zhou ZW, Zheng Y, et al. Dynamic single-cell mapping unveils Epstein–Barr virus-imprinted T-cell exhaustion and on-treatment response. Signal Transduct Target Ther. 2023;8:370.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Ruffin AT, Cillo AR, Tabib T, Liu A, Onkar S, Kunning SR, Lampenfeld C, Atiya HI, Abecassis I, Kürten CHL, et al. B cell signatures and tertiary lymphoid structures contribute to outcome in head and neck squamous cell carcinoma. Nat Commun. 2021;12:3349.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Wen Z, Liu H, Qiao D, Chen H, Li L, Yang Z, Zhu C, Zeng Z, Chen Y, Liu L. Nanovaccines fostering Tertiary Lymphoid structure to Attack Mimicry Nasopharyngeal Carcinoma. ACS Nano. 2023;17:7194–206.

    Article  CAS  PubMed  Google Scholar 

  50. Kuwabara S, Tsuchikawa T, Nakamura T, Hatanaka Y, Hatanaka KC, Sasaki K, Ono M, Umemoto K, Suzuki T, Sato O, et al. Prognostic relevance of tertiary lymphoid organs following neoadjuvant chemoradiotherapy in pancreatic ductal adenocarcinoma. Cancer Sci. 2019;110:1853–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Lv J, Wei Y, Yin JH, Chen YP, Zhou GQ, Wei C, Liang XY, Zhang Y, Zhang CJ, He SW, et al. The tumor immune microenvironment of nasopharyngeal carcinoma after gemcitabine plus cisplatin treatment. Nat Med. 2023;29:1424–36.

    Article  CAS  PubMed  Google Scholar 

  52. Cabrita R, Lauss M, Sanna A, Donia M, Skaarup Larsen M, Mitra S, Johansson I, Phung B, Harbst K, Vallon-Christersson J, et al. Tertiary lymphoid structures improve immunotherapy and survival in melanoma. Nature. 2020;577:561–5.

    Article  CAS  PubMed  Google Scholar 

  53. Cottrell TR, Thompson ED, Forde PM, Stein JE, Duffield AS, Anagnostou V, Rekhtman N, Anders RA, Cuda JD, Illei PB, et al. Pathologic features of response to neoadjuvant anti-PD-1 in resected non-small-cell lung carcinoma: a proposal for quantitative immune-related pathologic response criteria (irPRC). Ann Oncol. 2018;29:1853–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Pelka K, Hofree M, Chen JH, Sarkizova S, Pirl JD, Jorgji V, Bejnood A, Dionne D, Ge WH, Xu KH, et al. Spatially organized multicellular immune hubs in human colorectal cancer. Cell. 2021;184:4734–e47524720.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Jiang R, Gu X, Nathan CO, Hutt-Fletcher L. Laser-capture microdissection of oropharyngeal epithelium indicates restriction of Epstein-Barr virus receptor/CD21 mRNA to tonsil epithelial cells. J Oral Pathol Med. 2008;37:626–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Lin CH, Chiang MC, Chen YJ. STAT3 mediates resistance to anoikis and promotes invasiveness of nasopharyngeal cancer cells. Int J Mol Med. 2017;40:1549–56.

    Article  CAS  PubMed  Google Scholar 

  57. Endo K, Kondo S, Shackleford J, Horikawa T, Kitagawa N, Yoshizaki T, Furukawa M, Zen Y, Pagano JS. Phosphorylated ezrin is associated with EBV latent membrane protein 1 in nasopharyngeal carcinoma and induces cell migration. Oncogene. 2009;28:1725–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Liu Y, Jiang Q, Liu X, Lin X, Tang Z, Liu C, Zhou J, Zhao M, Li X, Cheng Z, et al. Cinobufotalin powerfully reversed EBV-miR-BART22-induced cisplatin resistance via stimulating MAP2K4 to antagonize non-muscle myosin heavy chain IIA/glycogen synthase 3β/β-catenin signaling pathway. EBioMedicine. 2019;48:386–404.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Chan OS, Kowanetz M, Ng WT, Koeppen H, Chan LK, Yeung RM, Wu H, Amler L, Mancao C. Characterization of PD-L1 expression and immune cell infiltration in nasopharyngeal cancer. Oral Oncol. 2017;67:52–60.

    Article  CAS  PubMed  Google Scholar 

  60. Wu X, Zhou Z, Xu S, Liao C, Chen X, Li B, Peng J, Li D, Yang L. Extracellular vesicle packaged LMP1-activated fibroblasts promote tumor progression via autophagy and stroma-tumor metabolism coupling. Cancer Lett. 2020;478:93–106.

    Article  CAS  PubMed  Google Scholar 

  61. Liu Y, He S, Wang XL, Peng W, Chen QY, Chi DM, Chen JR, Han BW, Lin GW, Li YQ, et al. Tumour heterogeneity and intercellular networks of nasopharyngeal carcinoma at single cell resolution. Nat Commun. 2021;12:741.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Biswas SK, Mantovani A. Macrophage plasticity and interaction with lymphocyte subsets: cancer as a paradigm. Nat Immunol. 2010;11:889–96.

    Article  CAS  PubMed  Google Scholar 

  63. Noy R, Pollard JW. Tumor-associated macrophages: from mechanisms to therapy. Immunity. 2014;41:49–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Jin S, Li R, Chen MY, Yu C, Tang LQ, Liu YM, Li JP, Liu YN, Luo YL, Zhao Y, et al. Single-cell transcriptomic analysis defines the interplay between tumor cells, viral infection, and the microenvironment in nasopharyngeal carcinoma. Cell Res. 2020;30:950–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol. 2009;9:162–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Safarzadeh E, Orangi M, Mohammadi H, Babaie F, Baradaran B. Myeloid-derived suppressor cells: important contributors to tumor progression and metastasis. J Cell Physiol. 2018;233:3024–36.

    Article  CAS  PubMed  Google Scholar 

  67. Lasser SA, Ozbay Kurt FG, Arkhypov I, Utikal J, Umansky V. Myeloid-derived suppressor cells in cancer and cancer therapy. Nat Rev Clin Oncol. 2024;21:147–64.

    Article  CAS  PubMed  Google Scholar 

  68. Kok VC. Current understanding of the mechanisms underlying Immune Evasion from PD-1/PD-L1 Immune Checkpoint Blockade in Head and Neck Cancer. Front Oncol. 2020;10:268.

    Article  PubMed  PubMed Central  Google Scholar 

  69. Theodoraki MN, Yerneni SS, Hoffmann TK, Gooding WE, Whiteside TL. Clinical significance of PD-L1(+) exosomes in plasma of Head and Neck Cancer patients. Clin Cancer Res. 2018;24:896–905.

    Article  CAS  PubMed  Google Scholar 

  70. Ogino T, Moriai S, Ishida Y, Ishii H, Katayama A, Miyokawa N, Harabuchi Y, Ferrone S. Association of immunoescape mechanisms with Epstein-Barr virus infection in nasopharyngeal carcinoma. Int J Cancer. 2007;120:2401–10.

    Article  CAS  PubMed  Google Scholar 

  71. Cascone T, McKenzie JA, Mbofung RM, Punt S, Wang Z, Xu C, Williams LJ, Wang Z, Bristow CA, Carugo A, et al. Increased tumor glycolysis characterizes Immune Resistance to adoptive T cell therapy. Cell Metab. 2018;27:977–e987974.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Wang Y, Lin H, Yao N, Chen X, Qiu B, Cui Y, Liu Y, Li B, Han C, Li Z, et al. Computerized tertiary lymphoid structures density on H&E-images is a prognostic biomarker in resectable lung adenocarcinoma. iScience. 2023;26:107635.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Barmpoutis P, Di Capite M, Kayhanian H, Waddingham W, Alexander DC, Jansen M, Kwong FNK. Tertiary lymphoid structures (TLS) identification and density assessment on H&E-stained digital slides of lung cancer. PLoS ONE. 2021;16:e0256907.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Buisseret L, Garaud S, de Wind A, Van den Eynden G, Boisson A, Solinas C, Gu-Trantien C, Naveaux C, Lodewyckx JN, Duvillier H, et al. Tumor-infiltrating lymphocyte composition, organization and PD-1/ PD-L1 expression are linked in breast cancer. Oncoimmunology. 2017;6:e1257452.

    Article  PubMed  Google Scholar 

  75. Shang T, Jiang T, Lu T, Wang H, Cui X, Pan Y, Xu M, Pei M, Ding Z, Feng X, et al. Tertiary lymphoid structures predict the prognosis and immunotherapy response of cholangiocarcinoma. Front Immunol. 2023;14:1166497.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Yang M, Che Y, Li K, Fang Z, Li S, Wang M, Zhang Y, Xu Z, Luo L, Wu C, et al. Detection and quantitative analysis of tumor-associated tertiary lymphoid structures. J Zhejiang Univ Sci B. 2023;24:779–95.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by the National Nature Science Foundation of China (grant number: 81972838; 82272722; 82200019).

Author information

Authors and Affiliations

Authors

Contributions

SQF designed the research. HLW drafted the manuscript. YTZ and JDL improved the structure. SQF and WYW revised the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Songqing Fan.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no known competing financial interests or personal relationship that could have appeared to influence the work reported in this paper.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, H., Zhan, Y., Luo, J. et al. Unveiling immune resistance mechanisms in nasopharyngeal carcinoma and emerging targets for antitumor immune response: tertiary lymphoid structures. J Transl Med 23, 38 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12967-024-05880-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12967-024-05880-7

Keywords