Immunotherapy for nasopharyngeal cancer—a review
Review Article

Immunotherapy for nasopharyngeal cancer—a review

Amit Jain1*, Whay Kuang Chia1*, Han Chong Toh1,2*

1Division of Medical Oncology, National Cancer Centre Singapore, 11 Hospital Drive, Singapore 169610, Singapore; 2Laboratory of Cell Therapy and Cancer Vaccine, Division of Medical Sciences, National Cancer Centre Singapore, 11 Hospital Drive, Singapore 169610, Singapore

Contributions: (I) Conception and design: All authors; (II) Administrative support: All authors; (III) Provision of study materials or patients: All authors; (IV) Collection and assembly of data: All authors; (V) Data analysis and interpretation: All authors; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

*These authors contributed equally to this manuscript.

Correspondence to: Han Chong Toh. Division of Medical Oncology, National Cancer Centre Singapore, 11 Hospital Drive, Singapore 169610, Singapore. Email: toh.han.chong@singhealth.com.sg.

Abstract: Nasopharyngeal carcinoma (NPC) is associated with the Epstein-Barr virus (EBV) and characterized by peritumoral immune infiltrate. Advanced NPC has high lethality. Immunotherapy directed against EBV antigen targets has been previously explored in clinical trials, and is likely to be validated as an important target in NPC as randomized data emerges in the future. Cancer vaccines and adoptive T cell therapy have been explored in the clinic, with the latter showing the greatest success. Recent advances in gene sequencing technology now allow personalized tumor epitope mapping, whilst the advent of immune checkpoint inhibitors targeting the PD-1/PD-L1 axis offers the opportunity to activate adaptive T cell response in vivo. Anti-PD1 antibodies have shown promising activity in early phase clinical trials, and randomized studies against chemotherapy are underway. As immunotherapy is incorporated into standard treatment paradigms, issues of optimal combinations with targeting agents, immune adjuvants, and sequence with chemotherapy and radiation therapy will need to be addressed. Effective strategies to increase tumor antigenicity, improve immunological memory and reduce immune escape, will need to be developed to improve treatment outcomes. Here we present a brief history of the evolution of immunotherapy in NPC, and highlight key concepts relevant to its further development in the clinic.

Keywords: Nasopharyngeal cancer (NPC); Epstein-Barr virus (EBV); immunotherapy; review


Submitted Dec 09, 2015. Accepted for publication Jan 28, 2016.

doi: 10.21037/cco.2016.03.08


Introduction

Nasopharyngeal cancer (NPC), an Epstein-Barr virus (EBV) associated disease, has a distinct etiology and geographic distribution. It is rare in the West with incidence of less than 1 for every 100,000 people each year, but endemic in Southern China, Hong Kong, Taiwan, and Southeast Asia, where annual incidence reaches as high as 25–50 cases per 100,000 per year. Worldwide, there are 80,000 incident cases resulting in an estimated 50,000 deaths annually (1). NPC is a chemosensitive disease and 5-year survival rate in early Stage I and II disease exceeds 80%, but outcomes are very poor in stage IV disease where the 5-year survival rate is less than 10% (2). Although the disease is highly sensitive to chemotherapy, resistance invariably develops and better treatments are urgently needed (3,4). The Epstein-Barr virus (EBV) latently infects more than 90% of the world’s adult human population and its association with NPC is thought to be mediated by an interplay of environmental (dietary, smoking, co-infectious) factors and genetic pre-disposition (high risk HLA allotypes). In NPC, the EBV virus expresses a type II latency program and is present in virtually all poorly differentiated and undifferentiated non-keratinising (WHO type II and III) NPC. The expression of viral antigens in NPC makes this disease an attractive target for immunotherapy strategies such as virus specific adoptive cell therapies. Here in this review, we summarize the range of EBV-related and unrelated antigenic targets, and discuss the crucial role of the immune-suppressive microenvironment in NPC. Significant clinical trial data for cancer vaccines and adoptive T cell therapy trials are outlined, and we explore the potential role of immune checkpoint inhibitors in NPC, and their potential combinations with conventional chemotherapy and radiation therapy. Given the disappointing clinical outcomes of all manner of targeted therapies in advanced NPC, rational immune oncology strategies become all the more crucial.


Targets for immunotherapy in nasopharyngeal cancer (NPC)

Epstein-Barr virus (EBV) targets

EBV is associated with a variety of malignancies including Hodgkin Disease, Burkitt’s Lymphoma, and NPC, with the expression of viral proteins on the tumor cell surface. The pathophysiological mechanisms involved in EBV integration with the host cell genome, latency, and transformation—is complex and incompletely understood. From a therapeutic standpoint, NPC expresses an array (albeit a limited repertoire) of EBV antigens (5,6). Hence, immunotherapeutic strategies in NPC have historically focused on EBV specific epitopes as a means of targeting this cancer.

EBV-associated NPC expresses a type II latency program, and tumor cells typically express the latent membrane proteins 1, 2A, and 2B (LMP1, LMP2A, and LMP2B), EBV nuclear antigen 1 (EBNA1), all of which have limited immunogenicity. In addition, several EBV non-coding RNAs primarily EBER1 and EBER2, and BamHI-A rightward transcripts (BARTs) and BamHI-A rightward frame 1 (BARF1) of EBV are expressed abundantly and are detected consistently in NPC (7-12).

EBNA1 is expressed frequently in NPC and is a dominant target for CD4 T cells. LMP1 and LMP2 are expressed in approximately 50% of NPC tumors. LMP1 may be poorly immunogenic, while the LMP2 proteins sufficiently more immunogenic and hence putative targets for EBV directed immunotherapy, such as cytotoxic T cells (8,13-15). NPC occurs in immunocompetent individuals, and it is likely that immunological pressure results in the expression of a limited array of EBV antigens. These proteins maintain cellular transformation in malignant cells and their poor immunogenicity likely plays a role in promoting immune escape by EBV-positive malignant cells (16,17). Immunotherapeutic approaches employed to target EBV are dependent upon the capacity to generate an immunological response against EBV latency response antigens, however in human hosts, these antigens have also co-evolved to evade immune recognition. LMP1 and LMP2 are known to play a role in activating and transforming cells following infection, allowing proliferation and survival of latently infected cells (18,19). The LMP antigens are oncogenic. LMP1 is a major transforming protein and behaves as a classical oncogene (20,21). LMP2A and LMP2B are likely non-essential in B cell transformation in vitro (22). However, LMP2A can transform epithelial cells via activation of the PI3 kinase-Akt pathway (23). These LMP antigens, particularly LMP1, are poorly immunogenic, likely due to poor antigen processing in infected cells and the subsequent limited amount of antigen available for presentation by MHC class I molecules (17). As a consequence, the LMP antigens, particularly LMP1, generate a subdominant CTL response when compared to the responses generated against lytic cycle antigens and other latent antigens, such as EBNA3 (24).

EBNA1 can be detected in all EBV-associated malignancies (15). EBNA1 is highly stable and contains a glycine-alanine repeat sequence near its N-terminus that inhibit translation and subsequent self-replication (25-27) and as result, EBNA1 is processed poorly via the MHC class I pathway. Nevertheless, the demonstration of EBNA1-specific CD8+ CTL thought to be induced via cross-presentation by professional antigen presentation cells rather than via direct recognition of infected cells (28), has established that endogenously processed EBNA1 can be detected by CD8+ T cells (29-31).

NPC cells have preserved antigen-processing function and can be recognized by major histocompatibility complex class I–restricted virus-specific CTLs in vitro (32), However, downregulation of major histocompatibility complex class I peptide expression is seen in NPC tumors as an immune evasion strategy (33). NPC patients also appear to have a lower prevalence of T cells that can recognize HLA-restricted epitopes in LMP2 and EBNA (34). Epidemiological studies have suggested that certain HLA allotypes have higher associations with nasopharyngeal carcinoma. These include HLA-A*11:01 and HLA-A*02:27 (35,36). A molecular explanation that cysteine at codon 99 of the Alpha2-helix of HLA-A protein is deleterious suggests a possible locus of susceptibility to NPC (36). Hence while NPC occurs across a variety of HLA allotypes, a meaningful strategy would be to focus initial HLA specific strategies in allotypes with demonstrated susceptibilities such as HLA-A*11:01.

Taken in entirety, NPC-related EBV antigens LMP1, LMP2A/B, EBNA1, EBER, and EBV-encoded RNA each have distinct effects on growth, differentiation, and the host immune response. Collectively, they likely contribute to the development of NPC through the promotion of transformation and angiogenesis, inhibition of apoptosis, induction of stem-cell-like phenotype, and enhancement of cell motility. EBV antigens also aid in immune escape through various mechanisms, including switching off immunodominant viral antigens, impairing the HLA I or HLA II pathway, up-regulating immune-inhibitory molecules, and recruiting T regulatory cells and inducing T-cell anergy (37). Hence, an understanding of the virus–host interaction in the NPC environment is essential for successful EBV-targeted immunotherapies. Selection pressure-driven evolution constantly stimulates the emergence of new EBV variants (38,39) which may be more oncogenic and less immunogenic than the parental strain, with for example a higher tropism for epithelial cells rather than B cells, suggesting that some EBV strains may carry an increased NPC risk (40).

It is important to note that NPC, while associated with EBV and the expression EBV proteins, is an entity that encompasses a broader range of other distinct molecular aberrations that may also represent immune targets.

Non-EBV targets

Genomic alterations in NPC represent neoantigens that may be immunogenic. Studies in this area are few given the limitation of accessible tissue for interrogation in this disease and the paucity of pre-clinical models. Nevertheless, a landmark study of comprehensive sequencing analysis of 56 NPC patients (41) has shed light on cancer mutations relevant in NPC. Nine significantly mutated genes included BAP1, MLL2, TSHZ3, TP53, PIK3CA, ERBB3, ERBB2, KRAS and NRAS. Copy number alterations in MAPKAPK2 have been shown to be associated with NPC risk (42). Epigenetic alteration in NPC include the CpG island methylator phenotype and a high load of hypermethylated tumor suppressor genes (43). These genomic and proteomic alterations and more, can contribute to the production of oncogenic and immunogenic alterations.

Immunogenic alterations can broadly be categorized into (I) tumor specific mutations that result in neoantigens; (II) tumor specific antigens and proteins overexpressed in tumors but not expressed or are expressed at very low levels in normal cells including proteins such as surviving; (III) lineage specific antigens expressed on tumor cells as well as on normal cells such as gp100; and (IV) cancer/testis antigens including MAGE and NY-ESO-1 (44). Emerging sequencing technologies with predictive computational algorithms now offer the possibility of developing HLA-restricted epitope maps for each tumor and the corresponding mutational landscape. These technologies will accelerate neo-antigen discovery and improve efficiency of immune targeting strategies in trials.

Immune checkpoints

PD-1 is an inhibitory receptor expressed on the surface of activated T cells. PD-1 is a known marker of T-cell exhaustion in animal models of viral infection. This manifests itself as loss of effector functions such as the secretion of cytokines (IFN-γ, IL-2, and TNF-α), production of the cytolytic effector molecules perforin and granzyme B, and eventually apoptosis (45-47). The immune infiltrates of chronic inflammation frequently employ the B7-H1/PD-1 axis. Both PD-1 ligands, B7-H1 (PD-L1) and B7-DC (PD-L2) are up-regulated in peripheral tissues during an inflammatory response to infectious agents, in response to type 1 (α, β) and type 2 (IFN-γ) interferons (48). The biologic role of this upregulation is the prevention of collateral tissue damage mediated by antigen-experienced T cells during inflammation (49-52). Other immune-checkpoint molecules such as 2B4, CD160, T cell Immunoglobulin and Mucin domain-3 (TIM3), Lymphocyte Activation Gene-3 (LAG3) are upregulated in conjunction with PD-1 on “exhausted” CD8 T cells in tumor and chronic viral models (53). Programmed cell death ligand-1 (PD-L1) is highly expressed by cancer cells and tumor-infiltrating macrophages in virus-associated malignancies including NPC (54). PD-L1 expression on tumor correlates with advanced tumor stage and lymphatic metastasis (55) while PD-1 overexpression is associated with shorter overall survival and recurrence free survival and is an independent risk factor for death, treatment failure and local recurrence of NPC (56). These early studies in NPC have added to rationale to apply immune checkpoint inhibitor antibodies to this disease.

Tumor microenvironment

NPC is characterized by substantial immune infiltrate in the primary tumor that consists of T cells, B cells, dendritic cells, monocytes, and eosinophils. This massive lymphoid infiltrate in the primary tumor is likely favored by inflammatory cytokines produced by tumor cells (57-60). There is evidence that despite the immunogenic nature of EBV antigen expressing cancer cells, there is a marked local tolerogenic immune suppression. T regulatory cells (Treg) within the tumor site may contribute to the functional inactivation of innate cytotoxic T cell responses. Significant expansion of circulating naïve and memory CD4+CD25high Foxp3+ was identified in 56 patients (61) and a smaller number was also noted to have infiltrating Treg in the tumor microenvironment. Another study of 40 untreated patients implicated the suppressive role of Treg cells with its findings of rich populations of Treg amongst tumor-infiltrating lymphocytes (TILs). A further finding in this study was that EBV-specific T cells are enriched but inactivated in the tumor microenvironment. TILs from NPC failed to produce IFN-gamma and to exert cytotoxicity when stimulated by lymphoblastoid cell lines (34). A more recent study demonstrated that both physical and pharmacologic mediated depletion of Tregs from PBMC enhances EBV-specific T cell responses in EBV-stimulated T cell lines generated from NPC (62).

A holistic immunotherapy strategy to target NPC must take into account the following:

  • Cancer specific factors
    • Genomic and proteomic differences between cancer and host, that are both EBV specific, but otherwise cancer genome specific too;
    • Presence of cancer-associated antigens, that are ordinarily poorly expressed in normal tissue, including the known cancer testis antigens;
    • Presence of immune-suppressive checkpoints on cancer cells;
    • Immunosuppressive factors in the tumor microenvironment such as but not limited to tumor hypoxia, immune-suppressive cytokine production, the presence of myeloid derived suppressor cells, and immunosuppressive regulatory T cells;
  • Host specific factors
    • HLA Class I and II type and expression that determines presentation of peptide sequences of intracellular proteins to various subsets of immune cells;
    • Immune cell population diversity and matching to tumor immune epitopes and other immunogenic cancer epitopes;
    • Dendritic cell function, presentation of tumor antigens, and interaction with immune cell subsets;
    • Host specific tumor permissive factors that have yet to be identified.

Immunotherapy strategies against NPC—overview

In our opinion, these strategies fall into two broad categories. The first category comprises strategies that aim to harness the host’s pre-existing anti-tumor capability that may be suppressed by tumor, or to augment the host’s innate ability to mount an immune response against tumor. This category of strategies assumes an innate pre-existing capacity to augment host immune response that the cancer may already have escaped, and aims to meaningfully directly impact the host immune system to mount an immune response against NPC, which represents an inflammatory cancer phenotype. Examples of these include immune checkpoint inhibitors anti-PD1, anti-PD-L1, anti-CTLA4, and anti-LAG3 antibodies to disinhibit the immune response against cancer, and cancer vaccines that attempt to stimulate and generate a host immune response.

The second category comprises therapeutic strategies that directly and preferentially target cancer cells. Chemotherapy and radiation can stimulate immunogenic cell death and this is increasingly being studied and understood for use with other immunotherapy strategies. Immune cells that target cancer cells directly include cytotoxic T lymphocytes and cytokine induced killer cells.


Host targeting agents

Immune checkpoint inhibitors

More than a fifth of patients with previously treated metastatic NPC showed an objective measurable response when treated with the pembrolizumab, according to a study reported at the 2015 European Cancer Congress. Pembrolizumab is a highly selective humanized monoclonal IgG4-kappa isotype antibody against PD-1 that is designed to block the negative immune regulatory signaling of the PD-1 receptor expressed by T cells (63). Two-thirds of patients in the study had some degree of reduction in target lesion size. The median duration of response was 10.8 months. The objective response rate with pembrolizumab in NPC was 22.2%, all partial responses. Another 15 patients had stable disease, resulting in a disease control rate of 77.8%. Forty one out of 44 patients screened for study had tumors that tested positive for PD-L1 expression. All but 2 of the patients had received at least 1 prior line of therapy for advanced disease, and a third of the patients had received 5 or more prior regimens. Median progression-free survival was 5.6 months. A recently opened trial uses Nivolumab (BMS), another anti-PD1 antibody, to treat patients with recurrent and/or metastatic NPC (NCT02339558). LAG3 represents another immune checkpoint that may confer immune escape. There is an ongoing Phase I clinical trial evaluating the safety and efficacy of an anti LAG3 antibody, LAG525 (Novartis), as a single agent and in combination with an anti-PD1 antibody, PDR001 (Novartis), in patients with advanced malignancies (NCCT02460224). This study includes NPC in its inclusion criteria.

Cancer vaccines

Therapeutic cancer vaccines for NPC have historically targeted EBV antigens. A study using an LMP2 vaccine has been reported. Autologous monocyte-derived dendritic cells cultured from patients with NPC and matured with cytokines were pulsed with HLA-A1101, A2402, or B40011 restricted epitope peptides from EBV-LMP2, and injected into inguinal lymph nodes. This strategy generated an expansion in the LMP2-specific response in the peripheral blood in the majority of patients, and a partial clinical response in 2 of 16 patients enrolled in the study was seen (64). A more recent Phase II study evaluated the use of dendritic cells transduced with an adenovirus-DeltaLMP1-LMP2 vector given as five biweekly intradermal injections to sixteen heavily pretreated stage 4c NPC patients. This first-in-human study demonstrated the safety of this strategy. No increase was seen in the frequency of LMP1/2-specific T cells (65). Another clinical trial using the MVA-EL vaccine has provided evidence for the effectiveness of the direct administration of a poly-specific vaccine to generate LMP/EBNA1-specific CTL responses in patients. This recombinant vaccinia virus–based vaccine, which encodes a functionally inactive fusion protein containing the CD4 epitope-rich C-terminal half of EBNA1 and CD8 epitope-rich LMP2A could induce T-cell response in 80% of patients, in some cases boosting response to both CD4+ and CD8+ mediated immunity against EBNA1 and/or LMP2 (66). This vaccine is now being evaluated in a phase II trial involving patients who have detectable plasma EBV DNA after RT or who experience optimal response to palliative chemotherapy (NCT01094405).


Cancer targeting agents

Immunogenic cell death with chemotherapy and radiotherapy: concepts from studies in other cancers

Cancer cell death can be immunogenic or non-immunogenic. Immunogenic cell death (ICD) involves changes in the composition of the cell surface as well as release of soluble mediators that occurs in a defined temporal sequence. Endoplasmic reticulum stress and autophagy result in calretuculin (CRT) exposure in the outer leaflet of pre-apoptotic cancer cells. Additionally, these pre-apoptotic cells secrete ATP, and release nuclear protein HMGB1 as membranes become permeabilized during necrosis. CRT, ATP, and HMGB1 bind to CD91, P2RX7, and TLR4 respectively, facilitating the recruitment of dendritic cells in the tumor bed, and engulfment of tumor antigens by dendritic cells and optimal antigen presentation to T cells (67). Radiation is commonly used in NPC and is known to cause ICD accompanied by CRT exposure, ATP release, and HMGB1 release. The concept of immunogenic cell death may well underpin the rationale for strategies that combine standard treatments of chemotherapy, small molecule inhibitors, and radiation therapy with immunotherapy. Studies to characterize the capacity of these treatments to cause immunogenic cell death specifically in NPC are needed.

Cell based therapies

EBV is associated with several cancer namely, post-transplant lymphoproliferative disease (PTLD), Hodgkin lymphoma (HL), Burkitt lymphoma, tumors in HIV-infected patients, T cell lymphoma, NK/T cell lymphoma, gastric cancers, and NPC (5). Following primary infection, EBV persists for life as a latent infection which is controlled by cytotoxic T lymphocytes (CTL) (68). Adoptive immunotherapy was first developed for the treatment of PTLD and has now been successfully utilized for over ten years using autologous EBV-immortalized LCLs to stimulate the expansion of EBV-specific CTLs (69). CTL therapies in NPC were developed on the basis of this evidence.

CTL can be heterogeneous, primarily with regards to their differentiation status and homing properties. Following antigen encounter, a naïve or memory T cell will proliferate and acquire an increasing number of effector functions, resulting in fully differentiated effector cells which display the full array of effector functions (70,71). However, differentiation into effector cells significantly alters the trafficking properties of the T cell (72). There is now evidence that this change in homing properties can be tissue-specific, whereby stimulation in different lymphoid organs can influence trafficking to particular peripheral tissues (73,74). Effective immunotherapeutic treatment of NPC may be dependent upon the capacity to generate CTL that can home in to nasopharyngeal tissue and other sites of metastatic disease. It also remains to be elucidated what impact the differentiation status of CTL has upon survival post-transfer. Although terminal differentiation may generate greater effector function, poor survival of these T cells post-transfer may reduce the number of cells accessing tumor sites. There is evidence that less differentiated T cells retain a greater capacity to expand following antigen encounter in vivo and provide greater protection following transfer (75). Therefore, treatment with non-terminally differentiated CTL may have some benefit in prolonging their survival and proliferation capacity following adoptive transfer.

Current strategies used to generate CTL that rely upon long-term in vitro cultures will generate cells with a late-stage effector phenotype. Lymphodepletion prior to adoptive transfer may provide another mechanism to enhance survival and proliferation of transferred CTL. In addition to the benefits associated with the removal of Treg cells, there is evidence that lymphodepletion can enhance the efficacy of CTL-based therapy by removing T cells which compete for homeostatic cytokines, such as IL-15 and IL-7, and thus creating ‘space’ in the lymphoid system to accommodate transferred T cells (76,77). However, some recent observations have suggested that whilst lymphodepletion may promote T cell engraftment (78) it may not improve the clinical outcome following T cell therapy (79). Our group had previously shown that a delayed graft-versus-NPC effect was demonstrable in three of 21 heavily-pretreated advanced NPC patients who received a conditioning regimen of subablative cyclophosphamide, in vivo T cell lymphodepletion with iv thymoglobuline and thymic irradiation followed by sibling HLA-matched and one-antigen mismatched allogeneic peripheral blood stem cells. The delayed objective responses were coincident with rising donor haematopoietic chimerism and better survivors correlated with chronic graft-versus-host disease. These results indicated to us that a potentially powerful immune alloresponse was operative against even bulky, progressing, and chemoresistant NPC disease (80).

We proceeded to conduct and complete a phase II trial exploring the role of cytoreductive chemotherapy followed by autologous CTL in previously untreated patients with advanced EBV-associated NPC. The patients received four cycles of gemcitabine and carboplatin followed by six doses of EBV-specific T cells (81).This combination therapy was well tolerated and resulted in an encouraging response rate of 71.4% with 3 complete and 22 partial responses. Moreover, the median overall survival of 29.9 months and the 2- and 3-year overall survival rates at 62.9% and 37.1%, respectively, were significantly higher than those observed in historical controls receiving chemotherapy alone (11–22 months). The study was the first in which a chemotherapy regimen followed by a planned cell-therapy is given as frontline therapy for any cancer, allowing timely delivery of adequate CTL cells following chemotherapy completion. The study also had a high overall completion rate, with 35 of the 38 enrolled patients receiving the planned consolidation with EBV-specific T cells with no attendant grade III or IV toxicities with CTL therapy. A multicenter Phase III randomized control trial (NCT02578641) using this protocol is underway.

The Italian group had previously treated ten advanced NPC patients progressing after conventional therapy, using autologous EBV-specific T cells generated from EBV-infected LCLs as antigen presenting cells to stimulate a polyclonal response to latent EBV antigens. They observed partial responses in two patients and stable disease in four others (82). The Baylor group previously observed 10 responses in 15 patients treated with active disease (5 complete responses, 2 partial responses, and 3 with stable disease) (83,84). An additional eight patients were treated in their second or subsequent remission, and five remained free of disease with follow-up of six years. Both groups have attempted to improve these results by pretreating patients with lymphodepletion using either chemotherapy with cyclophosphamide and fludarabine (79) or CD45-depleting antibodies (78) but neither added approach improved the overall response rate. In the studies by both groups, the LCL-induced EBV-specific T cells contain T cell clones that target all nine latent-cycle antigens of EBV as well as some of the virus’s lytic antigens. The majority of the T cells, however, are responding to the most immunogenic antigens, including EBNA3 and the lytic-cycle antigens such as BZLF1, which are not expressed by EBV-infected NPC cells. Instead, the tumor cells express antigens associated with the type II latency pattern, including LMP1, LMP2, EBNA1, and BARF, which are less immunogenic and are present at a lower frequency in polyclonal LCL-induced EBV-specific T cells. It is therefore notable that both groups have identified an association between measurable benefit of EBV-specific T cells and the presence in the product of LMP2-reactive clones that expand in the patient after infusion (82,83). This observation was also seen in the Phase II study reported by Chia et al., who showed a strong association of benefit with specificity for EBV-LMP2 in the infused line (P=0.04) (81).

Hence current studies are enriching lines for cells that recognize the EBV antigens expressed in NPC and other type II latency tumors, using either overlapping peptide pools pulsed on dendritic cells (85) or an adenoviral construct termed AdE1-LMPpoly that encodes EBNA1 fused to CD8+ T cell epitopes from LMP1 and LMP2 to stimulate T cells (86). The second approach has been tested in 16 patients with recurrent and metastatic NPC who received EBV-specific T cells generated by stimulation with AdE1-LMPpoly. After adoptive transfer, there was a transient increase in the frequency of T cells responding to LMP1, LMP2, and EBNA1. The median overall survival of these patients was 523 days, compared with 220 days in patients who did not receive T cells (86).

Currently, several novel strategies to improve the activity of CTL in NPC are being explored in clinical trials. MALTED is testing closely matched allogeneic CTL (NCT01447056), and RESIST-NPC is testing CTL cell that additionally express Dominant Negative Receptor that confers them resistance to TGFbeta, a factor secreted by cancer cells that confers immune suppression to CTL and allows immune escape (NCT02065362).

Cytokine-induced killer (CIK) cells represent a heterogeneous population of immune cells that have been expanded from peripheral blood mononuclear cells using cytokines. These have shown in vitro killing in a variety of cancers (87). NPC patients who received autologous CIK cell transfusion in combination with gemcitabine plus cisplatin chemotherapy had a higher overall survival and progression-free survival rates than patients with gemcitabine plus cisplatin chemotherapy (88) CIKs have also demonstrated tumor killing capacity against putative cancer stem cells of nasopharyngeal cancer, in pre-clinical models. This was demonstrated to be mediated somewhat via NKG2D–ligands as blocking by anti-NKG2D antibody significantly but partially abrogated CIK cell-mediated cytolysis against putative NPC cancer stem cells (89).


Future directions

The broad and potent responses of immune checkpoint inhibitors in a wide variety of tumors, is deepening our understanding of tumor immunogenicity and spearheading a resurgent interest in immunotherapy for NPC. As the complex interplay of EBV and NPC continues to be unraveled, it is likely that immunotherapeutic strategies will merge into mainstream clinical practice and offer durable remissions in patients with advanced NPC who are this day incurable.


Acknowledgements

None.


Footnote

Conflicts of Interest: The authors have no conflicts of interest to declare.


References

  1. Global Burden of Disease Cancer Collaboration, Fitzmaurice C, Dicker D, et al. The Global Burden of Cancer 2013. JAMA Oncol 2015;1:505-27. [Crossref] [PubMed]
  2. Jin Y, Shi YX, Cai XY, et al. Comparison of five cisplatin-based regimens frequently used as the first-line protocols in metastatic nasopharyngeal carcinoma. J Cancer Res Clin Oncol 2012;138:1717-25. [Crossref] [PubMed]
  3. Al-Sarraf M, LeBlanc M, Giri PG, et al. Chemoradiotherapy versus radiotherapy in patients with advanced nasopharyngeal cancer: phase III randomized Intergroup study 0099. J Clin Oncol 1998;16:1310-7. [PubMed]
  4. Wee J, Tan EH, Tai BC, et al. Randomized trial of radiotherapy versus concurrent chemoradiotherapy followed by adjuvant chemotherapy in patients with American Joint Committee on Cancer/International Union against cancer stage III and IV nasopharyngeal cancer of the endemic variety. J Clin Oncol 2005;23:6730-8. [Crossref] [PubMed]
  5. Anagnostopoulos I, Hummel M. Epstein-Barr virus in tumours. Histopathology 1996;29:297-315. [Crossref] [PubMed]
  6. Young LS, Rickinson AB. Epstein-Barr virus: 40 years on. Nat Rev Cancer 2004;4:757-68. [Crossref] [PubMed]
  7. Young LS, Dawson CW, Clark D, et al. Epstein-Barr virus gene expression in nasopharyngeal carcinoma. J Gen Virol 1988;69:1051-65. [Crossref] [PubMed]
  8. Brooks L, Yao QY, Rickinson AB, et al. Epstein-Barr virus latent gene transcription in nasopharyngeal carcinoma cells: coexpression of EBNA1, LMP1, and LMP2 transcripts. J Virol 1992;66:2689-97. [PubMed]
  9. Sam CK, Brooks LA, Niedobitek G, et al. Analysis of Epstein-Barr virus infection in nasopharyngeal biopsies from a group at high risk of nasopharyngeal carcinoma. Int J Cancer 1993;53:957-62. [Crossref] [PubMed]
  10. Gilligan KJ, Rajadurai P, Lin JC, et al. Expression of the Epstein-Barr virus BamHI A fragment in nasopharyngeal carcinoma: evidence for a viral protein expressed in vivo. J Virol 1991;65:6252-9. [PubMed]
  11. Decaussin G, Sbih-Lammali F, de Turenne-Tessier M, et al. Expression of BARF1 gene encoded by Epstein-Barr virus in nasopharyngeal carcinoma biopsies. Cancer Res 2000;60:5584-8. [PubMed]
  12. 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;76:82-8. [Crossref] [PubMed]
  13. Lee SP. Nasopharyngeal carcinoma and the EBV-specific T cell response: prospects for immunotherapy. Semin Cancer Biol 2002;12:463-71. [Crossref] [PubMed]
  14. Hislop AD, Taylor GS, Sauce D, et al. Cellular responses to viral infection in humans: lessons from Epstein-Barr virus. Annu Rev Immunol 2007;25:587-617. [Crossref] [PubMed]
  15. Khanna R, Tellam J, Duraiswamy J, et al. Immunotherapeutic strategies for EBV-associated malignancies. Trends Mol Med 2001;7:270-6. [Crossref] [PubMed]
  16. Smith C, Beagley L, Khanna R. Acquisition of polyfunctionality by Epstein-Barr virus-specific CD8+ T cells correlates with increased resistance to galectin-1-mediated suppression. J Virol 2009;83:6192-8. [Crossref] [PubMed]
  17. Smith C, Wakisaka N, Crough T, et al. Discerning regulation of cis- and trans-presentation of CD8+ T-cell epitopes by EBV-encoded oncogene LMP-1 through self-aggregation. Blood 2009;113:6148-52. [Crossref] [PubMed]
  18. Li HP, Chang YS. Epstein-Barr virus latent membrane protein 1: structure and functions. J Biomed Sci 2003;10:490-504. [Crossref] [PubMed]
  19. Thorley-Lawson DA. Epstein-Barr virus: exploiting the immune system. Nat Rev Immunol 2001;1:75-82. [Crossref] [PubMed]
  20. Wang D, Liebowitz D, Kieff E. An EBV membrane protein expressed in immortalized lymphocytes transforms established rodent cells. Cell 1985;43:831-40. [Crossref] [PubMed]
  21. Kaye KM, Izumi KM, Kieff E. Epstein-Barr virus latent membrane protein 1 is essential for B-lymphocyte growth transformation. Proc Natl Acad Sci U S A 1993;90:9150-4. [Crossref] [PubMed]
  22. Longnecker R. Epstein-Barr virus latency: LMP2, a regulator or means for Epstein-Barr virus persistence? Adv Cancer Res 2000;79:175-200. [Crossref] [PubMed]
  23. Scholle F, Bendt KM, Raab-Traub N. Epstein-Barr virus LMP2A transforms epithelial cells, inhibits cell differentiation, and activates Akt. J Virol 2000;74:10681-9. [Crossref] [PubMed]
  24. Rickinson AB, Moss DJ. Human cytotoxic T lymphocyte responses to Epstein-Barr virus infection. Annu Rev Immunol 1997;15:405-31. [Crossref] [PubMed]
  25. Yin Y, Manoury B, Fahraeus R. Self-inhibition of synthesis and antigen presentation by Epstein-Barr virus-encoded EBNA1. Science 2003;301:1371-4. [Crossref] [PubMed]
  26. Tellam J, Fogg MH, Rist M, et al. Influence of translation efficiency of homologous viral proteins on the endogenous presentation of CD8+ T cell epitopes. J Exp Med 2007;204:525-32. [Crossref] [PubMed]
  27. Tellam J, Smith C, Rist M, et al. Regulation of protein translation through mRNA structure influences MHC class I loading and T cell recognition. Proc Natl Acad Sci U S A 2008;105:9319-24. [Crossref] [PubMed]
  28. Blake N, Lee S, Redchenko I, et al. Human CD8+ T cell responses to EBV EBNA1: HLA class I presentation of the (Gly-Ala)-containing protein requires exogenous processing. Immunity 1997;7:791-802. [Crossref] [PubMed]
  29. Tellam J, Connolly G, Green KJ, et al. Endogenous presentation of CD8+ T cell epitopes from Epstein-Barr virus-encoded nuclear antigen 1. J Exp Med 2004;199:1421-31. [Crossref] [PubMed]
  30. Lee SP, Brooks JM, Al-Jarrah H, et al. CD8 T cell recognition of endogenously expressed epstein-barr virus nuclear antigen 1. J Exp Med 2004;199:1409-20. [Crossref] [PubMed]
  31. Voo KS, Fu T, Wang HY, et al. Evidence for the presentation of major histocompatibility complex class I-restricted Epstein-Barr virus nuclear antigen 1 peptides to CD8+ T lymphocytes. J Exp Med 2004;199:459-70. [Crossref] [PubMed]
  32. 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;58:310-4. [PubMed]
  33. Yao Y, Minter HA, Chen X, et al. Heterogeneity of HLA and EBER expression in Epstein-Barr virus-associated nasopharyngeal carcinoma. Int J Cancer 2000;88:949-55. [Crossref] [PubMed]
  34. 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:e1122. [Crossref] [PubMed]
  35. Chin YM, Mushiroda T, Takahashi A, et al. HLA-A SNPs and amino acid variants are associated with nasopharyngeal carcinoma in Malaysian Chinese. Int J Cancer 2015;136:678-87. [PubMed]
  36. Tian W, Zhu FM, Wang WY, et al. Sequence-based typing of HLA-A gene in 930 patients with nasopharyngeal carcinoma in Hunan province, southern China. Tissue Antigens 2015;86:15-20. [Crossref] [PubMed]
  37. Shen Y, Zhang S, Sun R, et al. Understanding the interplay between host immunity and Epstein-Barr virus in NPC patients. Emerg Microbes Infect 2015;4:e20. [Crossref] [PubMed]
  38. Tang YL, Lu JH, Cao L, et al. Genetic variations of EBV-LMP1 from nasopharyngeal carcinoma biopsies: potential loss of T cell epitopes. Braz J Med Biol Res 2008;41:110-6. [Crossref] [PubMed]
  39. Horst D, Burrows SR, Gatherer D, et al. Epstein-Barr virus isolates retain their capacity to evade T cell immunity through BNLF2a despite extensive sequence variation. J Virol 2012;86:572-7. [Crossref] [PubMed]
  40. Tsai MH, Raykova A, Klinke O, et al. Spontaneous lytic replication and epitheliotropism define an Epstein-Barr virus strain found in carcinomas. Cell Rep 2013;5:458-70. [Crossref] [PubMed]
  41. Lin DC, Meng X, Hazawa M, et al. The genomic landscape of nasopharyngeal carcinoma. Nat Genet 2014;46:866-71. [Crossref] [PubMed]
  42. Yang L, Liu B, Qiu F, et al. The effect of functional MAPKAPK2 copy number variation CNV-30450 on elevating nasopharyngeal carcinoma risk is modulated by EBV infection. Carcinogenesis 2014;35:46-52. [Crossref] [PubMed]
  43. Niller HH, Tarnai Z, Decsi G, et al. Role of epigenetics in EBV regulation and pathogenesis. Future Microbiol 2014;9:747-56. [Crossref] [PubMed]
  44. Manzo T, Heslop HE, Rooney CM. Antigen-specific T cell therapies for cancer. Hum Mol Genet 2015;24:R67-73. [Crossref] [PubMed]
  45. Barber DL, Wherry EJ, Masopust D, et al. Restoring function in exhausted CD8 T cells during chronic viral infection. Nature 2006;439:682-7. [Crossref] [PubMed]
  46. Flies DB, Sandler BJ, Sznol M, et al. Blockade of the B7-H1/PD-1 pathway for cancer immunotherapy. Yale J Biol Med 2011;84:409-21. [PubMed]
  47. Wherry EJ. T cell exhaustion. Nat Immunol 2011;12:492-9. [Crossref] [PubMed]
  48. Mazanet MM, Hughes CC. B7-H1 is expressed by human endothelial cells and suppresses T cell cytokine synthesis. J Immunol 2002;169:3581-8. [Crossref] [PubMed]
  49. Carreno BM, Collins M. The B7 family of ligands and its receptors: new pathways for costimulation and inhibition of immune responses. Annu Rev Immunol 2002;20:29-53. [Crossref] [PubMed]
  50. Greenwald RJ, Freeman GJ, Sharpe AH. The B7 family revisited. Annu Rev Immunol. 2005;23:515-48. [Crossref] [PubMed]
  51. Pentcheva-Hoang T, Corse E, Allison JP. Negative regulators of T-cell activation: potential targets for therapeutic intervention in cancer, autoimmune disease, and persistent infections. Immunol Rev 2009;229:67-87. [Crossref] [PubMed]
  52. Ahmadzadeh M, Johnson LA, Heemskerk B, et al. Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood 2009;114:1537-44. [Crossref] [PubMed]
  53. Ott PA, Hodi FS. The B7-H1/PD-1 pathway in cancers associated with infections and inflammation: opportunities for therapeutic intervention. Chin Clin Oncol 2013;2:7. [PubMed]
  54. Chen BJ, Chapuy B, Ouyang J, et al. PD-L1 expression is characteristic of a subset of aggressive B-cell lymphomas and virus-associated malignancies. Clin Cancer Res 2013;19:3462-73. [Crossref] [PubMed]
  55. Zhang F, Liu Z, Cui Y, et al. The clinical significance of the expression of costimulatory molecule PD-L1 in nasopharyngeal carcinoma. Lin Chung Er Bi Yan Hou Tou Jing Wai Ke Za Zhi 2008;22:408-10. [PubMed]
  56. Hsu MC, Hsiao JR, Chang KC, et al. Increase of programmed death-1-expressing intratumoral CD8 T cells predicts a poor prognosis for nasopharyngeal carcinoma. Mod Pathol 2010;23:1393-403. [Crossref] [PubMed]
  57. 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 U S A 1987;84:6262-6. [Crossref] [PubMed]
  58. 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;59:1599-605. [PubMed]
  59. 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;32:42-9. [Crossref] [PubMed]
  60. 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;206:68-75. [Crossref] [PubMed]
  61. 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;96:617-22. [Crossref] [PubMed]
  62. Fogg M, Murphy JR, Lorch J, et al. Therapeutic targeting of regulatory T cells enhances tumor-specific CD8+ T cell responses in Epstein-Barr virus associated nasopharyngeal carcinoma. Virology 2013;441:107-13. [Crossref] [PubMed]
  63. Hamid O, Robert C, Daud A, et al. Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N Engl J Med 2013;369:134-44. [Crossref] [PubMed]
  64. Lin CL, Lo WF, Lee TH, et al. Immunization with Epstein-Barr Virus (EBV) peptide-pulsed dendritic cells induces functional CD8+ T-cell immunity and may lead to tumor regression in patients with EBV-positive nasopharyngeal carcinoma. Cancer Res 2002;62:6952-8. [PubMed]
  65. Chia WK, Wang WW, Teo M, et al. A phase II study evaluating the safety and efficacy of an adenovirus-DeltaLMP1-LMP2 transduced dendritic cell vaccine in patients with advanced metastatic nasopharyngeal carcinoma. Ann Oncol 2012;23:997-1005. [Crossref] [PubMed]
  66. Hui EP, Taylor GS, Jia H, 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. [Crossref] [PubMed]
  67. Kroemer G, Galluzzi L, Kepp O, et al. Immunogenic cell death in cancer therapy. Annu Rev Immunol 2013;31:51-72. [Crossref] [PubMed]
  68. Khanna R, Burrows SR. Role of cytotoxic T lymphocytes in Epstein-Barr virus-associated diseases. Annu Rev Microbiol 2000;54:19-48. [Crossref] [PubMed]
  69. Haque T, Taylor C, Wilkie GM, et al. Complete regression of posttransplant lymphoproliferative disease using partially HLA-matched Epstein Barr virus-specific cytotoxic T cells. Transplantation 2001;72:1399-402. [Crossref] [PubMed]
  70. Seder RA, Ahmed R. Similarities and differences in CD4+ and CD8+ effector and memory T cell generation. Nat Immunol 2003;4:835-42. [Crossref] [PubMed]
  71. Wherry EJ, Ahmed R. Memory CD8 T-cell differentiation during viral infection. J Virol 2004;78:5535-45. [Crossref] [PubMed]
  72. Klebanoff CA, Gattinoni L, Torabi-Parizi P, et al. Central memory self/tumor-reactive CD8+ T cells confer superior antitumor immunity compared with effector memory T cells. Proc Natl Acad Sci U S A 2005;102:9571-6. [Crossref] [PubMed]
  73. Klonowski KD, Marzo AL, Williams KJ, et al. CD8 T cell recall responses are regulated by the tissue tropism of the memory cell and pathogen. J Immunol 2006;177:6738-46. [Crossref] [PubMed]
  74. Klonowski KD, Williams KJ, Marzo AL, et al. Dynamics of blood-borne CD8 memory T cell migration in vivo. Immunity 2004;20:551-62. [Crossref] [PubMed]
  75. Gattinoni L, Klebanoff CA, Palmer DC, et al. Acquisition of full effector function in vitro paradoxically impairs the in vivo antitumor efficacy of adoptively transferred CD8+ T cells. J Clin Invest 2005;115:1616-26. [Crossref] [PubMed]
  76. Gattinoni L, Finkelstein SE, Klebanoff CA, et al. Removal of homeostatic cytokine sinks by lymphodepletion enhances the efficacy of adoptively transferred tumor-specific CD8+ T cells. J Exp Med 2005;202:907-12. [Crossref] [PubMed]
  77. Muranski P, Boni A, Wrzesinski C, et al. Increased intensity lymphodepletion and adoptive immunotherapy--how far can we go? Nat Clin Pract Oncol 2006;3:668-81. [Crossref] [PubMed]
  78. Louis CU, Straathof K, Bollard CM, et al. Enhancing the in vivo expansion of adoptively transferred EBV-specific CTL with lymphodepleting CD45 monoclonal antibodies in NPC patients. Blood 2009;113:2442-50. [Crossref] [PubMed]
  79. Secondino S, Zecca M, Licitra L, et al. T-cell therapy for EBV-associated nasopharyngeal carcinoma: preparative lymphodepleting chemotherapy does not improve clinical results. Ann Oncol 2012;23:435-41. [Crossref] [PubMed]
  80. Toh HC, Chia WK, Sun L, et al. Graft-vs-tumor effect in patients with advanced nasopharyngeal cancer treated with nonmyeloablative allogeneic PBSC transplantation. Bone Marrow Transplant 2011;46:573-9. [Crossref] [PubMed]
  81. Chia WK, Teo M, Wang WW, et al. Adoptive T-cell transfer and chemotherapy in the first-line treatment of metastatic and/or locally recurrent nasopharyngeal carcinoma. Mol Ther 2014;22:132-9. [Crossref] [PubMed]
  82. Comoli P, Pedrazzoli P, Maccario R, et al. Cell therapy of stage IV nasopharyngeal carcinoma with autologous Epstein-Barr virus-targeted cytotoxic T lymphocytes. J Clin Oncol 2005;23:8942-9. [Crossref] [PubMed]
  83. Louis CU, Straathof K, Bollard CM, et al. Adoptive transfer of EBV-specific T cells results in sustained clinical responses in patients with locoregional nasopharyngeal carcinoma. J Immunother 2010;33:983-90. [Crossref] [PubMed]
  84. Straathof KC, Bollard CM, Popat U, et al. Treatment of nasopharyngeal carcinoma with Epstein-Barr virus--specific T lymphocytes. Blood 2005;105:1898-904. [Crossref] [PubMed]
  85. Ngo MC, Ando J, Leen AM, et al. Complementation of antigen-presenting cells to generate T lymphocytes with broad target specificity. J Immunother 2014;37:193-203. [Crossref] [PubMed]
  86. Smith C, Tsang J, Beagley L, et al. Effective treatment of metastatic forms of Epstein-Barr virus-associated nasopharyngeal carcinoma with a novel adenovirus-based adoptive immunotherapy. Cancer Res 2012;72:1116-25. [Crossref] [PubMed]
  87. Sangiolo D. Cytokine induced killer cells as promising immunotherapy for solid tumors. J Cancer 2011;2:363-8. [Crossref] [PubMed]
  88. Li JJ, Gu MF, Pan K, et al. Autologous cytokine-induced killer cell transfusion in combination with gemcitabine plus cisplatin regimen chemotherapy for metastatic nasopharyngeal carcinoma. J Immunother 2012;35:189-95. [Crossref] [PubMed]
  89. Wei F, Rong XX, Xie RY, et al. Cytokine-induced killer cells efficiently kill stem-like cancer cells of nasopharyngeal carcinoma via the NKG2D-ligands recognition. Oncotarget 2015;6:35023-39. [PubMed]
Cite this article as: Jain A, Chia WK, Toh HC. Immunotherapy for nasopharyngeal cancer—a review. Chin Clin Oncol 2016;5(2):22. doi: 10.21037/cco.2016.03.08

Download Citation