NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.
Madame Curie Bioscience Database [Internet]. Austin (TX): Landes Bioscience; 2000-2013.
Tumor cell population in nasopharyngeal carcinomas (NPC) is highly heterogeneous. In addition of being heavily infiltrated by nonmalignant leucocytes, malignant NPC cells can display various phenotypes in terms of epithelial maturation and viral gene expression. These various cell sub-populations communicate through membrane contacts, secretion of cytokines and exosomes. Understanding their interactions is crucial for the elucidation of tumor growth and immune escape as well as for designing better therapeutic approaches. This chapter deals with 3 major questions: what are the local factors responsible for leucocyte attraction and retention in NPC tumors? What are the suspected autocrine and paracrine mechanisms of tumor growth? What are the mechanisms of tumor immune evasion which could explain the growth of malignant epithelial cells containing viral antigenic proteins in a context of local inflammation?
Introduction
One of the most striking and consistent characteristic of NPC is the presence of a very abundant leucocyte infiltrate containing mainly T-lymphocytes. This infiltrate often accounts for a large fraction of the tumor mass. Therefore, among the pathologists who originally described NPC, several authors called this tumor lympho-epithelioma (see the chapter by J. Nicholls and G. Niedobitek in this book).1,2 This designation suggested a dual phenotype of NPC cells, in other words, the combination in the same tumor of malignant lymphoid and epithelial cells. This hypothesis has been ruled out in the late years 1970.3 It is now obvious that only epithelial cells are malignant and latently infected by EBV. When NPC are successfully xenografted on nude mice, the leucocyte infiltrate is rapidly eliminated.3,4 Moreover, although the leucocyte infiltrate is very consistent in the primary tumor, it is generally absent in visceral metastatic lesions (see chapter by J. Nichols and G. Niedobitek in this book).1,5 However, there are clinical and experimental observations suggesting that the lymphoid infiltrate plays a role in tumor growth at least at the initial stage of tumor development. This chapter will address several issues related to the formation and function of this infiltrate: what are the inflammatory cytokines produced by malignant NPC cells? Which vascular factor could favor leucocyte entry in the tumor? Conversely, which factor from the leucocyte infiltrate are likely to influence NPC cell growth and survival? We will also comment the role of other potential players involved in NPC tumor growth; for example stromal fibroblasts or rare malignant epithelial cells entering the lytic cycle. Finally this chapter will address a major paradox of NPC physiopathology which is the failure of the immune system to prevent tumor growth despite the presence of antigenic viral products in malignant cells and the presence of multiple immune effectors in the tumor tissue.
Our Limited Knowledge of NPC Histogenesis. The Hypothesis of a Tubal Tonsil Epithelial Origin
Primary lympho-epithelial carcinomas with an histological appearance almost identical to NPC have been reported in various anatomic sites outside the nasopharynx, for example in the stomach and salivary glands (consistently associated to EBV), in the lung (consistently associated with EBV in Asian population) and thymus (rarely associated with EBV).6-10 However one should acknowledge that these “ectopic” primary NPCs are extremely rare. A contrario, it should be recognized that the overwhelming majority of EBV-associated lymphoepithelial carcinomas do occur in the nasopharynx. This observation suggests that NPC oncogenesis is strongly dependent on local factors related to anatomical, histological and physiological characteristics of the nasopharyngeal cavity. Such local characteristics are probably important not only for EBV-infection of epithelial cells but also for the formation of the leucocyte infiltrate. On the basis of direct or endoscopic observation of small tumors, it is well established that NPC consistently arise in the fossae of Rosenmuller which are lateral extensions of the nasopharyngeal cavity also called “pharyngeal recesses”.11 These recesses are situated just above the Eustachian tube openings. They are also in close proximity to the “tubal tonsils” which is a part of the Waldeyer ring. This ring of lymphoid structures comprises the nasopharyngeal tonsil (NT) or adenoid, attached at the roof of the pharynx; the paired tubal tonsils (TT) as mentioned previously; the paired palatine tonsils (PT) positioned in the oropharynx; and the lingual tonsil (LT) on the posterior third of the tongue.12 At least some of these human lymphoepithelial elements are homologue to a unique rodent lymphoid structure called nasopharynx-associated lymphoid tissue (NALT). In rodents, NALT is located on both sides of the nasopharyngeal duct which is homologous to the human nasopharyngeal cavity.13
There are specific characteristics applying to each elements of the Waldeyer ring. PT and LT are directly exposed to ingested pathogens and antigens whereas NT and TT are strategic for interactions with airborne pathogens and antigens. The surface of the nasopharyngeal tonsils (NT and TT) are covered mainly with a ciliated respiratory epithelium whereas that of oropharyngeal tonsils (PT and LT) are protected by stratified squamous nonkeratinized or parakeratinized epithelium respectively. Unfortunately from the point of view of NPC biology, among the elements of the Waldeyer ring, TT have not been in the focus of most biological investigations. Very few things are known about tubal tonsils (TT). However we know that all human tonsils share common structural and functional properties. The lymphoid subepithelial compartment contains germinal centers surrounded by a mantle zone which are more or less similar to their lymph node homologs. Two major epithelial components are the surface epithelium and the epithelium lining the crypts which has the structure of a sponge with interstices containing infiltrating T- and B-cells, macrophages, interdigitating dendritic cells and Langherans cells.12 This specialised reticulated crypt epithelium, often called lymphoepithelium, has the same embryonic origin as the thymic epithelium (third pharyngeal pouche) and expression of a distinct subset of cytokeratins (CK 8, 18 and 19).14 The tonsil crypt epithelium has been sometimes described as a model of “lymphoepithelial symbiosis”. In addition, tonsil lymphoepithelium has a network of intra-epithelial capillary vessels, some of them ending in high endothelial venules (HEVs) with specific expression of adhesion molecules favoring lymphocyte extravasation.12
Because tonsillar epithelium has specific features supporting lymphoepithelial interactions, there is a suggestion that NPC cells derive from tubal tonsillar epithelial cells. This hypothesis needs to be substantiated by more experimental evidence. We know that small numbers of epithelial cells from both tonsil surface and reticulated crypts can be infected by EBV in vitro but we do not know yet whether specific markers of the tonsil crypt epithelium are expressed by NPC cells.15 We also ignore whether NPC tumor vessels have some properties in common with tonsillar lymphoepithelium vessels that would favor lymphocytes infiltration. Even if such vessels are not present in the tumor, their presence in close proximity to the tumor in tonsil crypts and interfollicular spaces might facilitate leucocyte entry in the neighbouring tumor tissue. Answering these question will be essential to better understand the formation of the lymphoepithelial stroma and its possible influence on tumor growth.
Subpopulations of Stromal Cells in NPC Tumors
Infiltrating Leucocytes
As previously mentioned, the abundance of infiltrating leucocytes is a major characteristic of NPC tumor stroma (in many reports, the terms “tumor infiltrating lymphocytes” or TILs are synonymous of “tumor infiltrating leucocytes”). Infiltrating leucocytes are often located around malignant cell clusters but sometimes disseminated within epithelial cell nests (see the chapter by J. Nicholls and G. Niedobitek in this book).1 As shown by immunohistochemistry using an anti-CD3 antibody, most of them are CD3-positive T-cells with a morphology of small resting lymphocytes.16 Among the CD3-positive cells, CD8 and CD4 T-cells are present in varying proportions depending on the tumor specimens.17 Collagenase dispersion of cells from tumor pieces allow quantitative assessment of subpopulations of tumor infiltrating leucocytes by flow cytometry. Using this technique, Ferradini et al have found a CD4 to CD8 ratio varying from 0.4 to 2.2.18 On average, 15% of these T-cells express the integrin αEβ7 (or CD103), a surface marker frequently expressed by intra-epithelial lymphocytes. NK-cells positive for CD56 or CD94 are also detected both by flow cytometry on TILs (about 5% TIL) or immunohistochemistry.18,19 Small populations of B-cells stained with anti-CD19 or -CD20 are also consistently detected.17,18 Several studies have reported the presence of monocytes and dendritic cells in NPC biopsies.20-23 Dendritic cells are often found inside malignant cell nests whereas monocytes are more often interspersed at some distance of epithelial cell clusters.21 Some NPC dendritic cells occasionally display CD23 expression like follicular dendritic cells.24 A fraction of dendritic cells contained in NPC tumors have features of Langherans cells (also called T-zone histiocytes) (see the chapter by J. Nicholls and G. Niedobitek in this book).1,22 Eosinophils are also detected in the leucocyte infiltrate of NPC tumors.25,26
Recently, several studies have refined the description of infiltrating T-lymphocytes contained in NPCs (Fig. 1). Lau et al have shown that about 12% of TILs recovered after collagenase cell dispersion have a phenoptype of regulatory T-cells (T-reg), CD4+CD25high, most of them being Foxp3-positive and CCR7-negative.27 By immunohistology on tissue sections, it has been shown that CD25 high and Foxp3 T-cells are more abundant in tumor tissue than in nonmalignant nasopharyngeal mucosa.28 On the other hand, lymphocytes expressing CXCR3 (the CXCL10 or IP10 receptor) are consistently detected in NPC tissue sections; in contrast, they were apparently rare in squamous carcinomas of the tongue investigated in the same study.29 CXCR3 expression is generally associated with Th1 differentiation. Detection of a subpopulation of CXCR3-positive TILs suggests that Th1 differentiation is taking place inside NPC tumors despite the presence of regulatory T-cells. CXCR3-postive cells are often detected within nests of malignant cells.29 It will be interesting to know whether intra-tumoral T-reg have the same distribution or accumulate at some distance of the tumor cells.
Nonleucocyte Stromal Cells
So far, only limited attention has been paid to nonleucocytic stromal cells in NPCs, especially fibroblasts and endothelial cells although both cell types are present in the tumor tissue. Histological forms of NPC with large areas of fibroblast proliferation have been reported.30 Unfortunately, so far, there has been no biological characterization of the fibroblasts derived from NPC tumors as well as from the normal nasopharyngeal mucosa (although it is much easier to recover stromal fibroblasts than malignant NPC cells when growing tumor cells in vitro; P. Busson, personal observations). The potential growth promoting effect of stromal fibroblasts on malignant NPC cells remains almost completely unexplored. In experimental models of breast carcinomas, CXCL12 or SDF1 (stromal cell-derived factor 1) has proven to be an important mediator of the growth-promoting effect of stromal fibroblasts on carcinomas cells.31 According to a recent study, malignant NPC cells often strongly express CXCR4 which is the physiological receptor of CXCL12.32 However, CXCR4 has a nuclear localisation in malignant cells of most NPC biopsies.32 A distribution which might not be compatible with responsiveness to CXCL12. In addition, production of CXCL12 by NPC stromal fibroblasts remains to be investigated. As reported for other tumors, VEGF is often detected by immunohistochemistry in nasopharyngeal carcinomas. Its level seems to be related to tumor aggressiveness.33 However, to our knowledge, the prognostic value of the tumor blood vessel count or circulating endothelial cell detection has not been assessed in NPC patients.
Heterogeneity of Malignant Cells in NPC Tumors
In addition to being heavily infiltrated by various categories of nonmalignant cells, the tumor cell population itself is heterogeneous in NPC tumors. This heterogeneity is obvious in terms of epithelial differentiation. Although most NPCs are nonkeratinizing undifferentiated carcinomas, one can consistently find areas of squamous cell maturation in this type of tumors (see the chapter by J. Nicholls and G. Niedobitek in this book).1 One can assume that there is an inverse relationship between proliferation and differentiation, although this has not been formally proven for NPC cells. It is not yet known whether one can isolate tumor stem cells from NPC specimens. A recent publication by Wang et al has reported the presence of stem cell-like side population cells in the CNE2 cell line which was depicted as an NPC cell line.34 This “side-population” or “SP” phenotype which is demonstrated by flow cytometry rely on a capacity of active extrusion of the DNA binding dye Hoechst 33342. It is characteristic of stem cells in several normal tissue lineages as well as in some malignant tumor lines. Wang et al have found that a fraction of 2.6% CNE2 cells display an SP phenotype which is associated with a series of characteristics suggestive of stem cell behavior. By comparison with nonSP cells, CNE2 SP cells have a much higher clonogenic potential in vitro. They are more tumorigenic in SCID mice and more resistant to some cytotoxic drugs. They also have a higher production of interleukin 19. One limitation of this study is that CNE2 cells are not representative of NPC cells. For example, they are not latently EBV-infected. Nevertheless, it is obvious that characterization of NPC tumor stem cells will become a major field of investigation in the next years. It will be important to determine which factors control NPC stem cell asymmetrical division and transient proliferation of tumor cells that leaves the stem cell compartment. It will be also important to confirm that morphological epithelial maturation is associated with both loss of stemness and decrease of proliferation.
Qualitative and quantitative variations in EBV-gene expression is another major factor of heterogeneity among NPC tumor cells. Another chapter of this book by N. Raab-Traub is focused on EBV gene expression and their role in tumor development. In the scope of this chapter we will simply mention a few points in close connection with tumor heterogeneity and cellular interactions. There is a consensus that EBV-infection is mainly latent in NPC. However, the amount of EBV latent gene products is variable from one cell to another, especially the amount of the Latent Membrane Protein 1 (LMP1).35 EBV viral particles have never been observed in fresh NPC biopsies although they can be produced by NPC cells used in short term culture in vitro and incubated with BUdR or other inducers of viral replication.36-38 Nevertheless partial expression of EBV genes involved in the lytic-productive cycle can occur in NPC tumors in situ. Several EBV-proteins specifically expressed during the lytic cycle have been detected in NPC tissue sections in limited areas of the tumor; for instance, elements of the EA complex, the BZLF1 protein and the EBV-Dnase.39-41 Consistently, linear replicative forms of the EBV genome are occasionally detected in a subset of NPC biopsies.42 The factors which control partial expression of the lytic EBV-genes in NPC cells are still poorly understood. There is some evidence that maturation from a phenotype of immature basal cell toward a phenotype of intermediate squamous tumor cells favors lytic gene expression.43 Local production of TGF β is also suspected to increase lytic cycle gene expression in some malignant epithelial cells.44 Recent findings made on EBV-associated SCID lymphoma models has spurred a renewed interested for lytic cycle gene expression in NPC. Hong et al have shown that a minority of lymphoma cells entering the lytic cycle play a critical role in the emergence of the disease through production of cytokines and angiogenic factors, especially interleukin-6.45 Similar mechanisms might be important for NPC tumor growth although interleukin-6 does not seem to be very abundant in NPC tissue sections.46
Dynamic Cellular Interactions: Possible Contribution to Tumor Growth
Role of Cytokines in Leucocyte Attraction and Retention
The leucocyte infiltrate consistently account for about 50% of the tumor mass. Obviously, it cannot be accounted for by a remnant of tonsilar leucocytes pre-existing to tumor development. For this reason, we hypothesized a long time ago that malignant epithelial cells were playing an active role in the formation of the infiltrate (Fig. 1, Table 1). Initially, we could demonstrate that malignant NPC cells constitutively produce interleukine-1 alpha (IL-1α), a cytokine with various inflammatory effects, including T-cell proliferation.47 This observation was later confirmed by Huang et al (1999) who detected both IL-1 α and β in malignant NPC cells by antibody staining of tissue sections. Simultaneously, investigations made by RT-PCR demonstrated IL-1 α and β transcripts in most NPC primary tumors and a fraction of metastatic lesions and its absence in control fragments of nonmalignant nasopharyngeal mucosa.48 Production of other inflammatory cytokines was investigated in NPC specimens in the subsequent years. IL-18 which has structural similarities with IL-1 is known to stimulate the proliferation of activated T-cells, to enhance their production of γ interferon and to promote their Th1 differentiation. By immunohistochemistry, IL18 was shown to be consistently produced by malignant NPC cells but not by epithelial cells of the non malignant mucosa.19 Data regarding IL-10 in NPCs remain controversial. According to three publications based on immunohistochemistry, IL-10 is detected in malignant cells of about 60% primary tumor biopsies of NPCs.49-51 In contrast, Beck et al (2001) has failed to detect IL-10 transcripts by in situ hybridization in malignant NPC cells whereas in some cases it was detected in the leucocyte infiltrate. The same group has also found a very rare expression of IL-6 and IL-8 transcripts by malignant NPC cells contrasting with occasional expression by infiltrating leucocytes.46
More recently, several studies have been focused on CC chemokines. Using in situ hybridisation, Teichman et al demonstrated a consistent and intense expression of the CXCL10 cytokine messenger by malignant NPC cells (CXCL10 is also called IP10 for γ-interferon inducible protein 10).29 CXCL10 induces chemotaxis of activated T-cells and inhibits angiogenesis. It is the agonist of the CXCR3 receptor.
As mentioned previously, CXCR3 which is often associated with Th1-differentiation is consistently detected in a fraction of T-cells infiltrating NPC. However, there is no precise relationships between CXCL10 and CXCR3 expression in terms of abundance or spatial distribution in the tissue sections.29 To our knowledge CXCL10 production has not yet been confirmed at the protein level in NPC tissue sections. Its status in the nonmalignant nasopharyngeal mucosa is not known. CCL20 or MIP-3α (Macrophage inflammatory protein-3 α), is another CC-chemokine that induces leukocyte migration into inflammation sites and regulates leukocyte trafficking through lymphoid tissues. It is a chemoattractant for memory regulatory T-cells.52 Chang et al have reported a high expression of CCL20 in NPC tumor cells. Interestingly, CCL20 is detected at a high concentration in serum samples from NPC patients. Its concentration is correlated with tumor mass and has prognostic value.53
In summary, on the basis of currently published data, the main inflammatory cytokines produced by malignant NPC cells are IL-1 α and β, CCL20, IL-18 and probably CXCL10 (Fig. 1, Table 2). We do not know yet which factors up-regulate their production by NPC cells. So far there is no evidence of a direct role of an EBV product in their induction. Except for CCL20 whose expression is induced by EBNA1 in the background of malignant Hodgkin cells.52 Whether or not the same applies to epithelial cells remain to be investigated. All these cytokines are produced by NPC cells not only in situ but also by several NPC tumor lines used in the laboratory, suggesting that their production is constitutive and do not require the presence of the leucocyte infiltrate.19,29,47 However some leucocytes might be involved in positive regulatory loops contributing to additional infiltration. For example CD3-positive T-lymphocytes and CD94 NK cells abundantly produce γ-interferon when they are located in primary NPC tumors whereas they do not or at a very low level when they are in nonmalignant NP mucosa.23,54 Concentration of γ-interferon is increased in the plasma of NPC patients.55 Production of γ-interferon by infiltrating lymphocytes is thought to be induced by local IL-18 whereas γ-interferon will in turn enhance CXCL10 production by malignant epithelial cells.19,29 Similarly, CD68-positive monocytes have been shown to abundantly produce two chemokines, CCL2 (also called MCP1 or monocyte chemoattractant protein 1) and CCL3 (also called MIP-1α), when they are located in the tumor infiltrate but not—or at a low level—when they are observed in nonmalignant nasopharyngeal mucosa or sub-mucosa.23 CCL2, like other monocyte chemoattractant proteins, recruits and/or activates monocytes, activated T-cells, NK cells and immature dendritic cells.23 CCL3 is an attractant for CD8+ T-cells, B-cells and dendritic cells.
Tumor infiltrating leucocytes which are so abundant in the primary tumor often disappear in the metastatic lesions.5 This might be explained by the decrease or loss of the production of some cytokines during the metastatic process. Alternatively, a positive balance between leucocyte entry and exit in the primary tumor might be dependent on specific anatomic factors in its local environment. As mentioned earlier, the proximity of the tubal tonsils with their network of specialised vessels—especially “high endothelial veinules”—probably facilitates leucocyte extravasation and tissue penetration.12
Influence of the Leucocyte Infiltrate on Malignant NPC Cells
It is obvious that malignant NPC cells are resistant to growth-inhibitory factors released by the leucocyte infiltrate, especially γ-interferon. Resistance to interferon is thought to be supported by at least 1 type of EBV-products, the small untranslated RNAs called EBERs (small EBV-encoded RNAs). EBERs 1 and 2 are about 120 base long and are very abundant in NPC cells. They have several oncogenic functions. One of these functions is to prevent the blockade of protein synthesis induced by interferons; an effect which is dependent on their direct binding to PKR.56
Not only the leucocyte infiltrate fails to block NPC tumor growth but it is suspected to enhance malignant NPC cell growth at the initial stage of primary tumor development. This hypothesis of a « ping-pong » or « folie à deux » mechanism is based on two main observations. One is the consistency of the infiltrate present in virtually all NPC primary tumors. The other is the low rate of successful NPC xenografts when using fragments from primary tumors (about 1% successful grafts) in contrast to a rate of about 50% when using metastatic fragments.4 The same contrast has been reported for other types of human tumors but probably not to the same extent. One possible interpretation is that infiltrating cells which are not retained in nude or SCID mice are required for growth of cells from primary tumors whereas cells from metastatic lesions are much better prepared for autonomous growth.
One experimental argument in favour of a cooperative effect of infiltrating leucocytes in tumor growth is provided by data from our and other groups who have investigated the role of the CD154/CD40 system in NPC cell survival and growth. Indeed CD40 is consistently and abundantly expressed by NPC cells whereas its cognate ligand CD154 or CD40-L is consistently expressed by infiltrating T-cells.17 We have shown that the CD40-receptor is functional in NPC cells in at least one respect; it has the ability to induce a cellular response called “rapid rescue from CD95-induced apoptosis”.57,58 In other models, long term permanent stimulation of CD40 has been shown to enhance the tumorigenic phenotype of epithelial cells.59 In summary, high constitutive expression of CD40 by malignant NPC cells and infiltration by T-cells bearing CD154 is likely to favour oncogenesis and tumor development. CD70 (strongly expressed by NPC cells) and CD27 (expressed by many infiltrating lymphocytes) are suspected to be involved in a similar process; a point that would require further investigation.17
In addition, Qian et al have published evidence of lymphoepithelial interactions mediated by the met receptor tyrosine kinase and its ligand the HGF receptor.60 Met is consistently expressed by NPC cells whereas its ligand HGF is only expressed by stromal cells which have not been unequivocally characterized but are probably infiltrating lymphocytes. A contribution of the Met/HGF system to tumor growth is suggested by an inverse correlation between patient survival and the level of Met expression. In many cell types, Met stimulation is known to enhance cell proliferation and motility.61 It is not yet known whether pharmacological agents which can block Met phosphorylation and signalling are beneficial to NPC patients.
Other Potential Mechanisms of Growth Based on Cellular Interactions
In addition to lympho-epithelial interactions, other cellular interactions are suspected to contribute to tumor growth. We have previously mentioned an hypothetical role of stromal fibroblasts possibly mediated by the SDF1/CXCR4 ligand-receptor pair. Autrocrine growth mechanisms should also be taken in account (Fig. 2). Sheu et al have suggested a role for an autocrine loop involving the c-kit tyrosine kinase receptor and its ligand SCF (stem cell factor). Both molecules are co-expressed by malignant cells from primary tumors and metastases in about 70% NPC patients. SCF/c-kit co-expression is also observed in epithelial cells of nonmalignant nasopharyngeal mucosa but at a lower frequency.62 On the other hand NPC patients with c-kit expression in their tumors do not have a more severe prognosis.63 In our hands, there is no significant constitutive phosphorylation of c-kit in an EBV-positive, c-kit-positive NPC xenograft (P. Busson, unpublished data). As mentioned in the chapter on systemic therapy (E.P. Hui and A.T.C. Chan in this book), a therapeutic trial based on SU 11248 (sunitinib malate) is in progress for NPC patients.64 This drug is active on several tyrosine kinases including c-kit. Positive results would encourage further investigations about the role of the c-kit-SCF pair. In addition to promoting leucocyte infiltration, CCL20 or MIP-3α is suspected to have an autocrine enhancing effect on NPC cell migration and invasion.53
Several viral products are suspected to participate in autocrine growth mechanisms of NPC cells. A 33 Kd EBV protein called BARF1—according to the name of the corresponding viral ORF—is frequently produced by malignant cells in a large fraction if not all NPC tumors.65 This protein which is secreted in the extracellular medium has homology with the human CSF gene. According to Houali et al, it is detected in the plasma of NPC patients.66 A publication from the same group provides evidence that extra-cellular BARF1 has growth-promoting activity and suggest that it might be an autocrine growth factor for malignant NPC cells.67 The small viral untranslated RNAs called EBERs have been previously mentioned for their role in cell resistance to interferon. These viral RNAs which are partially double-stranded are mainly concentrated in the nucleus. However, a small fraction of them leaks in the cytoplasm and even further in the extra-cellular space. These extra-nuclear EBERs have the power to activate some cellular receptors of double-strand RNAs, such as RIG1 and TLR-3.68 Activation of these receptors can result in growth-promoting signals for example an increase in IGF-1 secretion.69 Finally, a possible contribution to tumor growth of a small number of cells entering the lytic cycle has already been mentioned in this chapter.
Mechanisms of Tumor Immune Evasion
Various Mechanisms of Tumor Evasion for EBV-Associated Malignancies
The Epstein-Barr virus is involved in a wide range of human malignancies, either of epithelial or lymphoid origin. In all these malignancies, EBV-infection is mainly latent. No viral particles are detected by electron microscopy in tumor biopsy sections. The EBV genome is under circular form in the nuclei of malignant cells and most viral genes are silent, especially genes encoding viral enzymes and structural proteins. However a few viral genes called latent genes are consistently expressed in EBV-associated malignancies. These genes encode various types of viral products, either proteins or untranslated RNAs. All these viral products are suspected to contribute to the maintenance of the malignant phenotype. Depending on the type of malignancy, three patterns of EBV latency have been identified. The type III latency is characterized by the expression of a wide range of viral products including the immunodominant EBV nuclear antigens (EBNA) 3-6 and the less immunogenic antigens, EBNA1 and latent membrane proteins (LMP) 1 and 2. This type of latency is observed mainly in lymphoid malignancies occurring in immunocompromised individuals, such as the posttransplant lymphomas. In contrast, the expression of viral products is much more restricted in type I latency characteristic of EBV-associated Burkitt's lymphomas which do not express LMP1 and LMP2. Halfway between type I and type III latency, type II latency is characterized by the frequent expression of LMP1 and LMP2 combined to the absence of EBNA 3 to 6.70
Mechanisms of immune evasion are completely different in these three categories of tumors. In malignancies with type III latency—mainly posttransplant lymphomas—immune evasion likely occurs due to the direct suppression of lymphocyte function by immunosuppressive drugs.71 In contrast, malignancies with type I and type II latency occur in patients without obvious impairment of immune functions at the systemic level. Tumor immune evasion results from various combinations of 2 types of tolerogenic mechanisms: on one hand, internal cellular alterations impairing the machinery of antigen processing and presentation and, on the other hand, release of factors which create a context of immune inhibition in the tumor microenvironment. Regarding Burkitt's lymphoma (type I), there is strong evidence that immune evasion is supported to a large extent by early defects of antigen presentation to CD8+ T-cells, especially a reduction in MHC class I surface expression.72 This does not seem to apply to NPC which is characterized by a type II latency. Defects in MHC class I molecules have been reported in NPC cells, especially in a context of high EBNA1 expression.73,74 However, malignant NPC cells consistently retain the capacity to process and present antigen to CD8+ T-cells.75,76 Recent observations suggest that several immunosuppressive factors are released by malignant cells in the tumor microenvironment. These immunosuppressive factors probably result in partial inhibition rather than in complete abrogation of the local immune response. One potential consequence of this crippled immune response might be not only a failure in tumor eradication but also the progressive emergence of more resistant malignant clones, contributing to a process of immune selection or immuno-editing.
Candidate Immunosuppressive Factors in NPC Tumor Microenvironment
Factors released by malignant cells can impair local immune reactions by a large variety of mechanisms.77 Some factors released by tumor cells have direct cytotoxic or inhibitory effects against CD8+ and CD4+ CTL. Other factors have direct promoting effects on regulatory T-cells or shift the balance of Th1 and Th2 CD4+ cells towards a predominant Th2 response. Finally other tumor factors can impair the functions of professional antigen presenting cells in a way that results in tolerogenic effects on T-cell distribution and functions.
Only a fraction of immunosuppressive factors reported in various tumor models has been investigated in NPC, including TGF-β, IL-10, Fas-ligand and nitric oxide. We will provide concise informations for each of them. TGF-β can impair anti-tumor immune reactions by its inhibitory effect on CTLs and its stimulating effect on T-reg expansion.77 An elevated concentration of TGF-β has been reported in the plasma of NPC patients.78 However, with regard to tumor environment, TGF-β transcripts are not more abundant in the tumor tissue than in the nonmalignant nasopharyngeal mucosa.48 Fas-ligand can induce apoptosis of actived T-cells which often express high amounts of plasma membrane CD95 or Fas-receptor. Production of Fas-ligand by malignant NPC cells has been reported but only in a fraction of patients generally with a high tumor mass.79 IL-10 is a cytokine which switch the Th1/Th2 balance towards Th2 polarisation. As previously mentioned, its expression in malignant NPC cells remains controversial.46,49-51 Nitric oxide (NO) is a gas which has strong inhibitory effects on T-cells.80 In humans, NO derives from arginin through the action of enzymes called NO-synthases. Endothelial NO-synthase (e-Nos) has constitutive expression in the vascular system whereas inducible NO-synthase (i-Nos) can be expressed in a wide range of cell types mainly in inflammatory conditions. A high expression of i-Nos has been reported in malignant NPC cells; it is not yet known whether its expression is under the control of latent EBV infection.81 Nevertheless a high level of NO concentration is probably achieved in NPC tumor environment.
There are a lot of other potential tumor immunosuppressive factors which, to our knowledge, have not yet been investigated in NPC, for example the enzyme indoleamine 2, 3-dioxygenase (IDO) which is constitutively expressed by some human tumors.77
Role of Tumor Exosomes
Exosomes are bi-lamellar nanovesicles secreted by many cell types which are paradoxically derived from structures of the endosomal pathway called multivesicular bodies.82 Exosomes contains various types of cellular proteins which are either luminal or membrane-inserted. They also carry RNAs.83 There is growing evidence that exosomes are major players in cell communications including developmental processes, neural communications and immune responses.82,84 Exosomes also appear to play an important role in tumor growth and host-tumor relationships.85 Initial evidence that exosomes could play a role in immune evasion of EBV-infected cells came from a study by J. Middeldorp's group in Amsterdam dealing with the EBV-encoded LMP1 oncoprotein. They could show that this type III membrane protein contains an immunosuppressive motif in its first transmembrane segment and is secreted by EBV-transformed B-cells in association with exosomes.86 These LMP1-positive exosomes have an inhibitory effect on T-cell proliferation. More recently, inspired by this study, our group could demonstrate that NPC cells also produce exosomes. These NPC exosomes contain LMP1 only when they are produced by NPC cells with strong LMP1 expression (for example cells from the C15 xenograft).87 In contrast, regardless of the presence of LMP1, they have a high content of HLA class II molecules and galectin 9. Galectin-9 is a β-galactosyl binding lectin which is very abundant in NPC and has been identified as one specific ligand of the Tim-3 receptor.88 Tim-3 is expressed by mature CD4+ Th1 lymphocytes. In the context of CD4+ Th1 lymphocytes, galectin 9 binding to Tim-3 triggers a very rapid process of apoptosis. We have shown that the galectin 9 CRDs (Carbohydrate Recognition Domains) are presented at the surface of NPC exosomes. Accordingly, these exosomes can induce massive apoptosis of CD4+ T-cells expressing Tim-3, including CD4+ T-cells directed to EBV-antigens.89 In addition, we have demonstrated that HLA-class II-positive exosomes carrying galectin 9 are detected specifically in plasma samples from NPC patients but not from control patients, for example patients with nonNPC head and neck tumors.89 We believe that this presence of galectin 9 positive exosomes in the plasma reflects passive diffusion from tumor interstitial fluids to the blood stream. In addition to cytotoxic effects against mature CD4+ Th1 lymphocytes, NPC exosomes are suspected to enhance T-reg expansion possibly in cooperation with CCL20 (C. Durieu and P. Busson, personal data).52 They probably also have some influence on dendritic cell maturation.90 According to our current hypothesis depicted in Figure 3, NPC exosomes might subvert CD4+ Th1 functions by combining effects on dendritic cells favouring initial Th1 maturation with direct cytotoxic effects on fully mature Tim-3 positive Th1 cells. Development of novel in vitro and in vivo models will be required to validate this hypothesis.
Evidence of Immuno-Editing
The concept of immuno-editing or immuno-selection in NPC tumors is supported by two series of observations. One series about EBV strain restriction in tumor tissue and the other about acquired resistance to apoptosis in malignant cells.
EBV strain diversity is consistently more restricted in tumor tissue than in saliva or circulating blood. In most cases, only one strain is present in the tumor. Remarkably, this unique tumor-associated strain often has genetic polymorphisms that selectively invalidate viral CTL-epitopes restricted in the HLA alleles of the patient.91 At the single patient level, these data strongly suggest that genomes of certain viral isolates are selectively retained in the tumor cells for their ability to escape immune recognition. With regard to large groups of population, the same data suggest that combinations of HLA alleles and EBV-strain repertoires prevalent in certain geographic areas might favor a high incidence of NPCs.
Another form of immune-editing is suggested by a more aggressive phenotype of NPC tumors infiltrated by T-lymphocytes positive for granzyme B and perforin.73 This observation suggests that intra-tumoral CTLs, although unable to eradicate all malignant cells, nevertheless select a subpopulation of tumor cells resistant to apoptosis. One possible consequence of these observations is that the immune response in NPC not only fails to block tumor growth but might result in an increase of NPC cell resistance to therapeutic agents.
Conclusion
In conclusion, it is interesting to address these two questions: what can we do to make further progress in this field? What will be the practical consequences of our knowledge of cellular interactions in NPC tumors?
Detection of cell population markers and cytokines on tumor tissue sections will have more and more importance in this field for the years to come. One important challenge is to achieve simultaneous detection of multiple targets on the same tissue section. One recent exciting trend in clinical investigations of NPC has been the discovery of a series of novel tumor markers in the peripheral blood of NPC patients.53,92,93 Interestingly many of these markers are related to cellular interactions in NPC tumor microenvironment. 53,92,93 Their detection in the blood is related to their production in the tumor and provide indirect informations on cellular interactions. Several of these markers which have relevance to cellular interactions inside NPC tumors are listed in Table 2. In the coming years one may expect that some of them will contribute to assessment of intra-tumoral cellular interactions in combination with data obtained by immunohistology.
A major challenge is to connect observations made on clinical samples with in vitro experimental systems allowing assessment of functional roles of cytokines, exosomes and other cell communication agents. For this aim, novel methods allowing 3D in vitro cultures of cells derived from NPC biopsies need to be explored more intensively. In the past, some attempts to culture NPC cells in vitro under the form of spheroids have resulted in some partial success.94,95
In terms of therapeutic, there are several consequences of our knowledge of cellular interactions in NPC. Some therapeutic agents will be useful to block stimuli given by infiltrating cells to malignant cells. For example inhibitors of the met-receptor which are currently in phase I or II trials might be useful to block tumor growth promoting effects by HGF released by stromal lymphocytes or fibroblasts.60,61 Other therapeutic approaches will aim to antagonize the immunosuppressive effects of cytokines or exosomes released by malignant epithelial cells. In this regard, it is noteworthy that a monoclonal antibody directed to the CRD (carbohydrate recognition domain) of galectin 9 can block the stimulation of Tim-3 by galectin 9 exosomes and protect CD4+ Th1 lymphocytes.89 Finally based on our appreciation of cellular interactions in NPC tumor growth, it is useful to end with a word of caution about therapeutic strategies based on induction of the lytic cycle which might favour the release of a wide range of cytokines with potential enhancement of tumor growth and angiogenesis.
References
- 1.
- Nicholls J, Niedobitek G. Histopathological diagnosis of nasopharyngeal carcinoma—Looking beyond the blue book. In: Busson P, editor. Nasopharyngeal carcinoma: from basic sciences to clinical management. 2008.
- 2.
- Godtfredsen E. On the histopathology of malignant nasopharyngeal tumours. Acta Path et Microbiol Scand Supp. 1944;55:38–319.
- 3.
- Klein G, Giovanella BC, Lindahl T, et al. Direct evidence for the presence of Epstein-Barr virus DNA and nuclear antigen in malignant epithelial cells from patients with poorly differentiated carcinoma of the nasopharynx. Proc Natl Acad Sci USA. 1974;71(12):4737–4741. [PMC free article: PMC433971] [PubMed: 4373728]
- 4.
- Busson P, Ganem G, Flores P, et al. Establishment and characterization of three transplantable EBV-containing nasopharyngeal carcinomas. Int J Cancer. 1988;42(4):599–606. [PubMed: 2971626]
- 5.
- Teoh T. Epidermoid carcinoma of the nasopharynx among Chinese: a study of 31 necropsies. J Pathol Bacteriol. 1957;73:451–465.
- 6.
- Oda K, Tamaru J, Takenouchi T, et al. Association of Epstein-Barr virus with gastric carcinoma with lymphoid stroma. Am J Pathol. 1993;143(4):1063–1071. [PMC free article: PMC1887057] [PubMed: 8214002]
- 7.
- Hsu YC, Lu HF, Huang CC, et al. Malignant lymphoepithelial lesions of the salivary gland. Otolaryngol Head Neck Surg. 2006;134(4):661–666. [PubMed: 16564393]
- 8.
- Abdulla AK, Mian MY. Lymphoepithelial carcinoma of salivary glands. Head Neck. 1996;18(6):577–581. [PubMed: 8902573]
- 9.
- Ho JC, Wong MP, Lam WK. Lymphoepithelioma-like carcinoma of the lung. Respirology. 2006;11(5):539–545. [PubMed: 16916325]
- 10.
- Wu TC, Kuo TT. Study of Epstein-Barr virus early RNA 1 (EBER1) expression by in situ hybridization in thymic epithelial tumors of Chinese patients in Taiwan. Hum Pathol. 1993;24(3):235–238. [PubMed: 8454269]
- 11.
- Loh LE, Chee TS, John AB. The anatomy of the Fossa of Rosenmuller—its possible influence on the detection of occult nasopharyngeal carcinoma. Singapore Med J. 1991;32(3):154–155. [PubMed: 1876886]
- 12.
- Perry M, Whyte A. Immunology of the tonsils. Immunol Today. 1998;19(9):414–421. [PubMed: 9745205]
- 13.
- Brandtzaeg P. Immunology of tonsils and adenoids: everything the ENT surgeon needs to know. Int J Pediatr Otorhinolaryngol. 2003;67(Suppl 1):S69–76. [PubMed: 14662171]
- 14.
- Clark MA, Wilson C, Sama A, et al. Differential cytokeratin and glycoconjugate expression by the surface and crypt epithelia of human palatine tonsils. Histochem Cell Biol. 2000;114(4):311–321. [PubMed: 11131096]
- 15.
- Pegtel DM, Middeldorp J, Thorley-Lawson DA. Epstein-Barr virus infection in ex vivo tonsil epithelial cell cultures of asymptomatic carriers. J Virol. 2004;78(22):12613–12624. [PMC free article: PMC525079] [PubMed: 15507648]
- 16.
- Herait P, Ganem G, Lipinski M, et al. Lymphocyte subsets in tumour of patients with undifferentiated nasopharyngeal carcinoma: presence of lymphocytes with the phenotype of activated T-cells. Br J Cancer. 1987;55(2):135–139. [PMC free article: PMC2002096] [PubMed: 3545275]
- 17.
- Agathanggelou A, Niedobitek G, Chen R, et al. Expression of immune regulatory molecules in Epstein-Barr virus-associated nasopharyngeal carcinomas with prominent lymphoid stroma. Evidence for a functional interaction between epithelial tumor cells and infiltrating lymphoid cells. Am J Pathol. 1995;147(4):1152–1160. [PMC free article: PMC1871000] [PubMed: 7573360]
- 18.
- Ferradini L, Miescher S, Stoeck M, et al. Cytotoxic potential despite impaired activation pathways in T-lymphocytes infiltrating nasopharyngeal carcinoma. Int J Cancer. 1991;47(3):362–370. [PubMed: 1899651]
- 19.
- Hu H, Tang KF, Chua YN, et al. Expression of interleukin-18 by nasopharyngeal carcinoma cells: a factor that possibly initiates the massive leukocyte infiltration. Hum Pathol. 2004;35(6):722–728. [PubMed: 15188138]
- 20.
- Zong YS, Zhang CQ, Zhang F, et al. Infiltrating lymphocytes and accessory cells in nasopharyngeal carcinoma. Jpn J Cancer Res. 1993;84(8):900–905. [PMC free article: PMC5919276] [PubMed: 8407554]
- 21.
- Giannini A, Bianchi S, Messerini L, et al. Prognostic significance of accessory cells and lymphocytes in nasopharyngeal carcinoma. Pathol Res Pract. 1991;187(4):496–502. [PubMed: 1876530]
- 22.
- Ma CX, Jia TC, Li XR, et al. Langerhans cells in nasopharyngeal carcinoma in relation to prognosis. In Vivo. 1995;9(3):225–229. [PubMed: 8562888]
- 23.
- Tang KF, Tan SY, Chan SH, et al. A distinct expression of CC chemokines by macrophages in nasopharyngeal carcinoma: implication for the intense tumor infiltration by T-lymphocytes and macrophages. Hum Pathol. 2001;32(1):42–49. [PubMed: 11172294]
- 24.
- Niedobitek G, Young LS, Sam CK, et al. Expression of Epstein-Barr virus genes and of lymphocyte activation molecules in undifferentiated nasopharyngeal carcinomas. Am J Pathol. 1992;140(4):879–887. [PMC free article: PMC1886364] [PubMed: 1314028]
- 25.
- Leighton SE, Teo JG, Leung SF, et al. Prevalence and prognostic significance of tumor-associated tissue eosinophilia in nasopharyngeal carcinoma. Cancer. 1996;77(3):436–440. [PubMed: 8630949]
- 26.
- Liu CM, Ko JJ, Shun CT, et al. Soluble adhesion molecules and cytokines in tumor-associated tissue eosinophilia of nasopharyngeal carcinoma. Acta Otolaryngol. 2001;121(4):534–538. [PubMed: 11508518]
- 27.
- Lau KM, Cheng SH, Lo KW, et al. Increase in circulating Foxp3+CD4+CD25(high) regulatory T-cells in nasopharyngeal carcinoma patients. Br J Cancer. 2007;96(4):617–622. [PMC free article: PMC2360054] [PubMed: 17262084]
- 28.
- Yip WK, Abdullah MA, Yusoff SM, et al. Increase in tumour-infiltrating lymphocytes with regulatory T-cell immunophenotypes and reduced zeta-chain expression in nasopharyngeal carcinoma patients. Clin Exp Immunol. 2009;155(3):412–422. [PMC free article: PMC2669517] [PubMed: 19220831]
- 29.
- Teichmann M, Meyer B, Beck A, et al. Expression of the interferon-inducible chemokine IP-10 (CXCL10), a chemokine with proposed anti-neoplastic functions, in Hodgkin lymphoma and nasopharyngeal carcinoma. J Pathol. 2005;206(1):68–75. [PubMed: 15751051]
- 30.
- Braham H, Trimeche M, Ziadi S, et al. CD10 expression by fusiform stromal cells in nasopharyngeal carcinoma correlates with tumor progression. Virchows Arch. 2006;449(2):220–224. [PubMed: 16673118]
- 31.
- Orimo A, Gupta PB, Sgroi DC, et al. Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion. Cell. 2005;121(3):335–348. [PubMed: 15882617]
- 32.
- Wang N, Wu QL, Fang Y, et al. Expression of chemokine receptor CXCR4 in nasopharyngeal carcinoma: pattern of expression and correlation with clinical outcome. J Transl Med. 2005;3:26. [PMC free article: PMC1188078] [PubMed: 15978137]
- 33.
- Hui EP, Chan AT, Pezzella F, et al. Coexpression of hypoxia-inducible factors 1alpha and 2alpha, carbonic anhydrase IX and vascular endothelial growth factor in nasopharyngeal carcinoma and relationship to survival. Clin Cancer Res. 2002;8(8):2595–2604. [PubMed: 12171890]
- 34.
- Wang J, Guo LP, Chen LZ, et al. Identification of cancer stem cell-like side population cells in human nasopharyngeal carcinoma cell line. Cancer Res. 2007;67(8):3716–3724. [PubMed: 17440084]
- 35.
- Khabir A, Karray H, Rodriguez S, et al. EBV latent membrane protein 1 abundance correlates with patient age but not with metastatic behavior in north African nasopharyngeal carcinomas. Virol J. 2005;2(1):39. [PMC free article: PMC1112617] [PubMed: 15842731]
- 36.
- Trumper PA, Epstein MA, Giovanella BC. Activation in vitro by BUdR of a productive EB virus infection in the epithelial cells of nasopharyngeal carcinoma. Int J Cancer. 1976;17(5):578–587. [PubMed: 178612]
- 37.
- Trumper PA, Epstein MA, Giovanella BC, et al. Isolation of infectious EB virus from the epithelial tumour cells of nasopharyngeal carcinoma. Int J Cancer. 1977;20(5):655–662. [PubMed: 200568]
- 38.
- Hitt MM, Allday MJ, Hara T, et al. EBV gene expression in an NPC-related tumour. Embo J. 1989;8(9):2639–2651. [PMC free article: PMC401270] [PubMed: 2479554]
- 39.
- Lung ML, Chan KH, Lam WP, et al. In situ detection of Epstein-Barr virus markers in nasopharyngeal carcinoma patients. Oncology. 1989;46(5):310–317. [PubMed: 2550863]
- 40.
- Cochet C, Martel-Renoir D, Grunewald V, et al. Expression of the Epstein-Barr virus immediate early gene, BZLF1, in nasopharyngeal carcinoma tumor cells. Virology. 1993;197(1):358–365. [PubMed: 8212572]
- 41.
- Sbih-Lammali F, Berger F, Busson P, et al. Expression of the DNase encoded by the BGLF5 gene of Epstein-Barr virus in nasopharyngeal carcinoma epithelial cells. Virology. 1996;222(1):64–74. [PubMed: 8806488]
- 42.
- Raab-Traub N, Flynn K. The structure of the termini of the Epstein-Barr virus as a marker of clonal cellular proliferation. Cell. 1986;47(6):883–889. [PubMed: 3022942]
- 43.
- Zhang JX, Chen HL, Zong YS, et al. Epstein-Barr virus expression within keratinizing nasopharyngeal carcinoma. J Med Virol. 1998;55(3):227–233. [PubMed: 9624611]
- 44.
- Fahmi H, Cochet C, Hmama Z, et al. Transforming growth factor beta 1 stimulates expression of the Epstein-Barr virus BZLF1 immediate-early gene product ZEBRA by an indirect mechanism which requires the MAPK kinase pathway. J Virol. 2000;74(13):5810–5818. [PMC free article: PMC112075] [PubMed: 10846060]
- 45.
- Hong GK, Gulley ML, Feng WH, et al. Epstein-Barr virus lytic infection contributes to lymphoproliferative disease in a SCID mouse model. J Virol. 2005;79(22):13993–14003. [PMC free article: PMC1280209] [PubMed: 16254335]
- 46.
- Beck A, Pazolt D, Grabenbauer GG, et al. Expression of cytokine and chemokine genes in Epstein-Barr virus-associated nasopharyngeal carcinoma: comparison with Hodgkin's disease. J Pathol. 2001;194(2):145–151. [PubMed: 11400141]
- 47.
- Busson P, Braham K, Ganem G, et al. Epstein-Barr virus-containing epithelial cells from nasopharyngeal carcinoma produce interleukin 1 alpha. Proc Natl Acad Sci USA. 1987;84(17):6262–6266. [PMC free article: PMC299051] [PubMed: 2819868]
- 48.
- Huang YT, Sheen TS, Chen CL, et al. Profile of cytokine expression in nasopharyngeal carcinomas: a distinct expression of interleukin 1 in tumor and CD4+ T-cells. Cancer Res. 1999;59(7):1599–1605. [PubMed: 10197635]
- 49.
- Yao M, Ohshima K, Suzumiya J, et al. Interleukin-10 expression and cytotoxic-T-cell response in Epstein-Barr-virus-associated nasopharyngeal carcinoma. Int J Cancer. 1997;72(3):398–402. [PubMed: 9247280]
- 50.
- Fujieda S, Lee K, Sunaga H, et al. Staining of interleukin-10 predicts clinical outcome in patients with nasopharyngeal carcinoma. Cancer. 1999;85(7):1439–1445. [PubMed: 10193932]
- 51.
- Ozyar E, Ayhan A, Korcum AF, et al. Prognostic role of Epstein-Barr virus latent membrane protein-1 and interleukin-10 expression in patients with nasopharyngeal carcinoma. Cancer Invest. 2004;22(4):483–491. [PubMed: 15565804]
- 52.
- Baumforth KR, Birgersdotter A, Reynolds GM, et al. Expression of the Epstein-Barr virus-encoded Epstein-Barr virus nuclear antigen 1 in Hodgkin's lymphoma cells mediates Up-regulation of CCL20 and the migration of regulatory T-cells. Am J Pathol. 2008;173(1):195–204. [PMC free article: PMC2438297] [PubMed: 18502823]
- 53.
- Chang KP, Hao SP, Chang JH, et al. Macrophage inflammatory protein-3alpha is a novel serum marker for nasopharyngeal carcinoma detection and prediction of treatment outcomes. Clin Cancer Res. 2008;14(21):6979–6987. [PubMed: 18980993]
- 54.
- Tang KF, Chan SH, Loh KS, et al. Increased production of interferon-gamma by tumour infiltrating T-lymphocytes in nasopharyngeal carcinoma: indicative of an activated status. Cancer Lett. 1999;140(1-2):93–98. [PubMed: 10403546]
- 55.
- Li J, Zeng XH, Mo HY, et al. Functional Inactivation of EBV-Specific T-Lymphocytes in Nasopharyngeal Carcinoma: Implications for Tumor Immunotherapy. PLoS ONE. 2007;2(11):e1122. [PMC free article: PMC2048575] [PubMed: 17987110]
- 56.
- Nanbo A, Takada K. The role of Epstein-Barr virus-encoded small RNAs (EBERs) in oncogenesis. Rev Med Virol. 2002;12(5):321–326. [PubMed: 12211044]
- 57.
- Sbih-Lammali F, Clausse B, Ardila-Osorio H, et al. Control of apoptosis in Epstein Barr virus-positive nasopharyngeal carcinoma cells: opposite effects of CD95 and CD40 stimulation. Cancer Res. 1999;59(4):924–930. [PubMed: 10029086]
- 58.
- Benson RJ, Hostager BS, Bishop GA. Rapid CD40-mediated rescue from CD95-induced apoptosis requires TNFR-associated factor-6 and PI3K. Eur J Immunol. 2006;36(9):2535–2543. [PubMed: 16897814]
- 59.
- Baxendale AJ, Dawson CW, Stewart SE, et al. Constitutive activation of the CD40 pathway promotes cell transformation and neoplastic growth. Oncogene. 2005;24(53):7913–7923. [PubMed: 16091748]
- 60.
- Qian CN, Guo X, Cao B, et al. Met protein expression level correlates with survival in patients with late-stage nasopharyngeal carcinoma. Cancer Res. 2002;62(2):589–596. [PubMed: 11809714]
- 61.
- Naran S, Zhang X, Hughes SJ. Inhibition of HGF/MET as therapy for malignancy. Expert Opin Ther Targets. 2009;13(5):569–581. [PubMed: 19397476]
- 62.
- Sheu LF, Lee WC, Lee HS, et al. Co-expression of c-kit and stem cell factor in primary and metastatic nasopharyngeal carcinomas and nasopharyngeal epithelium. J Pathol. 2005;207(2):216–223. [PubMed: 16021677]
- 63.
- Bar-Sela G, Ben Arush MW, Sabo E, et al. Pediatric nasopharyngeal carcinoma: better prognosis and increased c-Kit expression as compared to adults. Pediatr Blood Cancer. 2005;45(3):291–297. [PubMed: 15558705]
- 64.
- Hui EP, Chan ATC. The evolving role of systemic therapy in nasopharyngeal carcinoma: Current strategies and perspectives. In: Busson P, editor. Nasopharyngeal carcinoma: from basic sciences to clinical management. Landes; 2008.
- 65.
- Seto E, Yang L, Middeldorp J, et al. Epstein-Barr virus (EBV)-encoded BARF1 gene is expressed in nasopharyngeal carcinoma and EBV-associated gastric carcinoma tissues in the absence of lytic gene expression. J Med Virol. 2005;76(1):82–88. [PubMed: 15778977]
- 66.
- Houali K, Wang X, Shimizu Y, et al. A new diagnostic marker for secreted Epstein-Barr virus encoded LMP1 and BARF1 oncoproteins in the serum and saliva of patients with nasopharyngeal carcinoma. Clin Cancer Res. 2007;13(17):4993–5000. [PubMed: 17785549]
- 67.
- Sall A, Caserta S, Jolicoeur P, et al. Mitogenic activity of Epstein-Barr virus-encoded BARF1 protein. Oncogene. 2004;23(28):4938–4944. [PubMed: 15064715]
- 68.
- Samanta M, Iwakiri D, Kanda T, et al. EB virus-encoded RNAs are recognized by RIG-I and activate signaling to induce type I IFN. Embo J. 2006;25(18):4207–4214. [PMC free article: PMC1570431] [PubMed: 16946700]
- 69.
- Iwakiri D, Sheen TS, Chen JY, et al. Epstein-Barr virus-encoded small RNA induces insulin-like growth factor 1 and supports growth of nasopharyngeal carcinoma-derived cell lines. Oncogene. 2005;24(10):1767–1773. [PubMed: 15608666]
- 70.
- Klein E, Kis LL, Klein G. Epstein-Barr virus infection in humans: from harmless to life endangering virus-lymphocyte interactions. Oncogene. 2007;26(9):1297–1305. [PubMed: 17322915]
- 71.
- Sebelin-Wulf K, Nguyen TD, Oertel S, et al. Quantitative analysis of EBV-specific CD4/CD8 T-cell numbers, absolute CD4/CD8 T-cell numbers and EBV load in solid organ transplant recipients with PLTD. Transpl Immunol. 2007;17(3):203–210. [PubMed: 17331848]
- 72.
- Rowe M, Khanna R, Jacob CA, et al. Restoration of endogenous antigen processing in Burkitt's lymphoma cells by Epstein-Barr virus latent membrane protein-1: coordinate up-regulation of peptide transporters and HLA-class I antigen expression. Eur J Immunol. 1995;25(5):1374–1384. [PubMed: 7774641]
- 73.
- Oudejans JJ, Harijadi H, Kummer JA, et al. High numbers of granzyme B/CD8-positive tumour-infiltrating lymphocytes in nasopharyngeal carcinoma biopsies predict rapid fatal outcome in patients treated with curative intent. J Pathol. 2002;198(4):468–475. [PubMed: 12434416]
- 74.
- Sengupta S, den Boon JA, Chen IH, et al. Genome-wide expression profiling reveals EBV-associated inhibition of MHC class I expression in nasopharyngeal carcinoma. Cancer Res. 2006;66(16):7999–8006. [PubMed: 16912175]
- 75.
- Khanna R, Busson P, Burrows SR, et al. Molecular characterization of antigen-processing function in nasopharyngeal carcinoma (NPC): evidence for efficient presentation of Epstein-Barr virus cytotoxic T-cell epitopes by NPC cells. Cancer Res. 1998;58(2):310–314. [PubMed: 9443410]
- 76.
- Lee SP, Chan AT, Cheung ST, et al. CTL control of EBV in nasopharyngeal carcinoma (NPC): EBV-specific CTL responses in the blood and tumors of NPC patients and the antigen-processing function of the tumor cells. J Immunol. 2000;165(1):573–582. [PubMed: 10861098]
- 77.
- Zitvogel L, Tesniere A, Kroemer G. Cancer despite immunosurveillance: immunoselection and immunosubversion. Nat Rev Immunol. 2006;6(10):715–727. [PubMed: 16977338]
- 78.
- Xu J, Ahmad A, Jones JF, et al. Elevated serum transforming growth factor beta1 levels in Epstein-Barr virus-associated diseases and their correlation with virus-specific immunoglobulin A (IgA) and IgM. J Virol. 2000;74(5):2443–2446. [PMC free article: PMC111728] [PubMed: 10666277]
- 79.
- Ho SY, Guo HR, Chen HH, et al. Prognostic implications of Fas-ligand expression in nasopharyngeal carcinoma. Head Neck. 2004;26(11):977–983. [PubMed: 15390195]
- 80.
- Tripathi P. Nitric oxide and immune response. Indian J Biochem Biophys. 2007;44(5):310–319. [PubMed: 18341206]
- 81.
- Ma N, Kawanishi M, Hiraku Y, et al. Reactive nitrogen species-dependent DNA damage in EBV-associated nasopharyngeal carcinoma: the relation to STAT3 activation and EGFR expression. Int J Cancer. 2008;122(11):2517–2525. [PubMed: 18307254]
- 82.
- Schorey JS, Bhatnagar S. Exosome function: from tumor immunology to pathogen biology. Traffic. 2008;9(6):871–881. [PMC free article: PMC3636814] [PubMed: 18331451]
- 83.
- Valadi H, Ekstrom K, Bossios A, et al. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 2007;9(6):654–659. [PubMed: 17486113]
- 84.
- Lakkaraju A, Rodriguez-Boulan E. Itinerant exosomes: emerging roles in cell and tissue polarity. Trends Cell Biol. 2008;18(5):199–209. [PMC free article: PMC3754907] [PubMed: 18396047]
- 85.
- Iero M, Valenti R, Huber V, et al. Tumour-released exosomes and their implications in cancer immunity. Cell Death Differ. 2008;15(1):80–88. [PubMed: 17932500]
- 86.
- Dukers DF, Meij P, Vervoort MB, et al. Direct immunosuppressive effects of EBV-encoded latent membrane protein 1. J Immunol. 2000;165(2):663–670. [PubMed: 10878338]
- 87.
- Keryer-Bibens C, Pioche-Durieu C, Villemant C, et al. Exosomes released by EBV-infected nasopharyngeal carcinoma cells convey the viral latent membrane protein 1 and the immunomodulatory protein galectin 9. BMC Cancer. 2006;6:283. [PMC free article: PMC1779799] [PubMed: 17156439]
- 88.
- Zhu C, Anderson AC, Schubart A, et al. The Tim-3 ligand galectin-9 negatively regulates T helper type 1 immunity. Nat Immunol. 2005;6(12):1245–1252. [PubMed: 16286920]
- 89.
- Klibi J, Niki T, Riedel A, et al. Blood diffusion and Th1-suppressive effects of galectin-9-containing exosomes released by Epstein-Barr virus-infected nasopharyngeal carcinoma cells. Blood. 2009;113(9):1957–1966. [PubMed: 19005181]
- 90.
- Dai SY, Nakagawa R, Itoh A, et al. Galectin-9 induces maturation of human monocyte-derived dendritic cells. J Immunol. 2005;175(5):2974–2981. [PubMed: 16116184]
- 91.
- Edwards RH, Sitki-Green D, Moore DT, et al. Potential selection of LMP1 variants in nasopharyngeal carcinoma. J Virol. 2004;78(2):868–881. [PMC free article: PMC368819] [PubMed: 14694118]
- 92.
- Rousselet G, Bachouchi M, Busson P, et al. Clinical implications of the serum level of CD23 in patients with undifferentiated nasopharyngeal carcinoma. J Clin Oncol. 1993;11(11):2143–2149. [PubMed: 8229128]
- 93.
- Caggiari L, Guidoboni M, Vaccher E, et al. High serum levels of soluble CD40-L in patients with undifferentiated nasopharyngeal carcinoma: pathogenic and clinical relevance. Infect Agent Cancer. 2007;2:5. [PMC free article: PMC1819365] [PubMed: 17331231]
- 94.
- Crawford DH, Achong BG, Teich NM, et al. Identification of murine endogenous xenotropic retrovirus in cultured multicellular tumour spheroids from nude-mouse-passaged nasopharyngeal carcinoma. Int J Cancer. 1979;23(1):1–7. [PubMed: 215558]
- 95.
- Vicat JM, Ardila-Osorio H, Khabir A, et al. Apoptosis and TRAF-1 cleavage in Epstein-Barr virus-positive nasopharyngeal carcinoma cells treated with doxorubicin combined with a farnesyl-transferase inhibitor. Biochem Pharmacol. 2003;65(3):423–433. [PubMed: 12527335]
- Introduction
- Our Limited Knowledge of NPC Histogenesis. The Hypothesis of a Tubal Tonsil Epithelial Origin
- Subpopulations of Stromal Cells in NPC Tumors
- Heterogeneity of Malignant Cells in NPC Tumors
- Dynamic Cellular Interactions: Possible Contribution to Tumor Growth
- Mechanisms of Tumor Immune Evasion
- Conclusion
- References
- Cellular Interactions in Nasopharyngeal Carcinomas - Madame Curie Bioscience Dat...Cellular Interactions in Nasopharyngeal Carcinomas - Madame Curie Bioscience Database
Your browsing activity is empty.
Activity recording is turned off.
See more...