Elsevier

Oral Oncology

Volume 84, September 2018, Pages 61-70
Oral Oncology

Review
Cellular-based immunotherapy in Epstein-Barr virus induced nasopharyngeal cancer

https://doi.org/10.1016/j.oraloncology.2018.07.011Get rights and content

Highlights

  • Up to 20% NPC patients developed relapse following definitive chemo-radiotherapy.

  • Presence of EBV-associated tumor antigens allows potential targeting using CBI.

  • CBI reported enhanced EBV-specific antitumor immune response with some benefit.

  • Immune priming via CBI may diminish previous acquired resistance to failed therapy.

  • Developments of novel CBI with combination therapy may improve cancer outcomes.

Abstract

Undifferentiated Nasopharyngeal carcinoma (NPC) is ubiquitously identified with the Epstein-Barr virus (EBV), making this cancer a suitable candidate for cellular-based immunotherapy (CBI) due to its expression of potentially targetable tumor-associated viral antigens. Various preclinical and clinical studies have explored the use of cytotoxic T cells (CTLs), tumor-infiltrating lymphocytes (TILs), natural killer (NK) cells, and dendritic cells (DCs) in the treatment of both refractory and locally advanced NPC with some success. Notably, immune-mediated antitumor effects were observed even in heavily pre-treated NPC patients, suggesting potential clinical benefit of CBI in this group of patients. These immune anti-tumor effects may be even more clinically evident when used as a first-line treatment, since there may not be an intense immunosuppressive environment which is typically encountered in refractory cancer patients. Additionally, CBI may exert an effect in priming the immune system and diminishing the cancer’s acquired resistance to exert a more robust response to previously failed chemotherapy. Although these results are encouraging, further refinements of clinical protocols to boost anti-tumor response and benefit a larger subset of patients proved necessary. Herein, we aim to review the rational of developing CBI in EBV-induced NPC and summarize its current applications in clinical studies.

Introduction

Nasopharyngeal carcinoma (NPC) occurs worldwide with approximately 87,000 incident cases and 51,000 deaths annually, representing about 0.7% of the global cancer burden [1]. A distinct racial and geographical variation is evident with a higher incidence rate in both southern China and Southeast Asia. This geographic predisposition is suggestive of both genetic and environmental risk factors in its pathogenesis. For instance, several at risk HLA genes such as the HLA-A subtype; and exposure to nitrosamines and nitrosamine precursors in the diet have been reported as risk factors of developing NPC. [2], [3], [4]. Polymorphisms in certain genes, such as CYP2E1, XRCC1, and hOGG1, have also been associated with an altered risk of developing NPC [5], [6]. For instance, CYP2E1 belongs to the cytochrome P450 family and is involved in the metabolic activation of nitrosamines and other pro-carcinogens into reactive intermediates capable of inducing DNA damage. Polymorphisms in CYP2E1, particularly the c2 variant (present in 20–25% of Asians as compared to less than 10% of Caucasians), results in a polymorphic base substitution which up-regulates CYP2E1 expression [5]. Conversely, XRCC1 and hOGG1 are DNA repair genes which play a role in the base excision repair (BER) pathway, and polymorphic variants of both XRCC1 (codon 194, 280 and 399) and hOGG1 (codon 326) have demonstrated decreased capacity for repair of DNA damage caused by nitrosamine compounds and oxidative stress [6], [7], [8]. Taken together, an individual with polymorphism in all three genes may succumb to greater DNA damage, thus increasing the risk of developing NPC.

Due to its radiosensitive nature, radiotherapy is the primary treatment modality for all stages of non-metastatic NPC; with concurrent chemo-radiotherapy being administered for locally advanced cases. This treatment paradigm attained more than 85% chance of loco-regional control and 80% survival [9], [10]. Despite positive clinical outcomes, a significant number of patients developed local regional failure (approximately 18%) or distant metastasis (approximately 17%), necessitating new treatment paradigms in this cancer [11]. Additionally, the close anatomical proximity of the nasopharynx to several critical structures including the brainstem, optic nerves, inner ears, and temporomandibular joint, precluded surgical salvage or radical re-radiation since severe radiation associated adverse effects like temporal lobe brain necrosis or carotid blow-out syndrome can occur [12], [13]. Therefore, there is a critical need for novel treatment modalities so as to minimize treatment-associated complications and to improve current survival rates in this cancer.

Section snippets

Epstein-Barr virus (EBV) association with NPC

Close association between NPC and the Epstein-Barr virus (EBV) infection is well established with numerous studies reporting an elevated anti-EBV antibodies titers and the presence of EBV DNA in nearly all endemic undifferentiated variant of NPC [14], [15]. EBV belongs to the γ-1 herpes virus family that infects more than 90% of the global population. In most individuals, infection occurs early during childhood and exists as lifelong asymptomatic infection which does not cause any pathological

EBV latent antigens

EBNA-1, LMP-1, and LMP-2A/B are among the EBV proteins expressed in NPC. While EBNA-1 is frequently expressed in all cases of undifferentiated NPC, expression of LMP-1 and LMP-2 is more heterogenous. EBNA-1 appears to be a dominant target for CD4+ T cells, due to the presence of a large glycine-alanine repeat domain within its protein sequence that prevents proteasomal breakdown of the molecule and its subsequent presentation by the HLA-class 1 pathway [29]. This reduces EBNA-1’s visibility to

EBV and host immunity

The host immune response against these EBV latent antigens appeared to be suboptimal due to several possible mechanisms. For example, highly immunogenic EBV latency genes (EBNA-2 and EBNA-3) are exclusively switched off in NPC in order to avoid immune recognition [74]. In place of that, several non-translated small RNAs (EBERs) are expressed which are undetectable by T cells. EBV also impairs both HLA-class I and II antigen presentation pathways through several EBV lytic and latent antigens [75]

Rational for CBI in NPC

Several factors suggest that CBI may be feasible and more efficacious in treating NPC. Firstly, EBV-infected NPC cells express structural proteins that are known targets for CD4+ and CD8+ T cells [29], [82]. Secondly, NPC cells express numerous immune regulatory molecules (CD40, CD70, CD80 and CD86) critical in mediating T cell activation [83]. Thirdly, strategies have been developed to selectively expand the appropriate immune cells and prime them with a pool of EBV antigens expressed in NPC,

Cytotoxic CD8+ T lymphocytes (CTLs)

In the last 16 years, numerous studies have shown some success of adoptive T cell therapy directed against EBV antigens in the treatment of NPC. This method involves ex vivo stimulation of autologous CD8+ T cells followed by intravenous reinfusion into the patient. There are several advantages offered by this treatment strategy as T cells possess high-avidity binding to EBV antigens and are able to undergo substantial in vitro expansion to attain clinical treatment numbers [84]. Moreover,

Therapeutic vaccination

Therapeutic vaccination may well represent a quicker method of stimulating the immune response via two developed strategies–either through the administration of EBV peptide-loaded dendritic cells (DCs) or via viral vectors modified to express EBV antigens. A summary of the clinical trials conducted using DC and viral vector vaccine is shown in Table 4.

DC vaccine

DCs are the most efficient antigen presenting cells (APCs) competent in the effective presentation of EBV antigens to CTLs, bridging the innate

Conclusion

Increase in fundamental understanding of immunological mechanisms in EBV-induced NPC carcinogenesis and their interactions with the host immune system will allow for improvement in immunotherapeutic strategies so as to improve patient outcomes. Presently, CBI has demonstrated acceptable tolerability and safety outcomes, with positive clinical benefit demonstrated especially in CTL-based therapies. Though encouraging, the benefit is limited to a small subset of patients. Further modification of

Funding

Chwee Ming Lim acknowledges the Transitional Award (Talent Development Program) from the National Medical Research Council Singapore. No other specific grant was obtained from funding agencies in the public, commercial, or not-for-profit sectors.

Conflict of interest

None declared.

References (116)

  • R.G. Caldwell et al.

    Epstein-Barr virus LMP2A drives B cell development and survival in the absence of normal B cell receptor signals

    Immunity

    (1998)
  • C. Mancao et al.

    Epstein-Barr virus latent membrane protein 2A is a B-cell receptor mimic and essential for B-cell survival

    Blood

    (2007)
  • S.W. Tsao et al.

    The significance of LMP1 expression in nasopharyngeal carcinoma

    Sem Cancer Biol

    (2002)
  • N. Blake 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)
  • C. Wrzesinski et al.

    Less is more: lymphodepletion followed by hematopoietic stem cell transplant augments adoptive T-cell-based anti-tumor immunotherapy

    Curr Opin Immunol

    (2005)
  • A. Masmoudi et al.

    Epstein-Barr virus-targeted immunotherapy for nasopharyngeal carcinoma

    Cancer Treat Rev

    (2007)
  • K.C. Straathof et al.

    Treatment of nasopharyngeal carcinoma with Epstein-Barr virus–specific T lymphocytes

    Blood

    (2005)
  • C.U. Louis et al.

    Enhancing the in vivo expansion of adoptively transferred EBV-specific CTL with lymphodepleting CD45 monoclonal antibodies in NPC patients

    Blood

    (2009)
  • C.A. Klebanoff et al.

    Sinks, suppressors and antigen presenters: how lymphodepletion enhances T cell-mediated tumor immunotherapy

    Trends Immunol

    (2005)
  • S. Secondino et al.

    T-cell therapy for EBV-associated nasopharyngeal carcinoma: preparative lymphodepleting chemotherapy does not improve clinical results

    Ann Oncol

    (2012)
  • W.K. Chia et al.

    Adoptive T-cell transfer and chemotherapy in the first-line treatment of metastatic and/or locally recurrent nasopharyngeal carcinoma

    Mol Ther

    (2014)
  • J. Ferlay et al.

    Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012

    Int J Cancer

    (2015)
  • M.J. Simons et al.

    De-the GB. Immunogenetic aspects of nasopharyngeal carcinoma: I. differences in HL-A antigen profiles between patients and control groups

    Int J Cancer

    (1974)
  • C.C. Lu et al.

    Nasopharyngeal carcinoma-susceptibility locus is localized to a 132 kb segment containing HLA-A using high-resolution microsatellite mapping

    Int J Cancer

    (2005)
  • J.M. Yuan et al.

    Preserved foods in relation to risk of nasopharyngeal carcinoma in ShanghaiChina

    Int J Cancer

    (2000)
  • A. Hildesheim et al.

    CYP2E1 genetic polymorphisms and risk of nasopharyngeal carcinoma in Taiwan

    J Natl Cancer Inst

    (1997)
  • E.Y. Cho et al.

    Nasopharyngeal carcinoma and genetic polymorphisms of DNA repair enzymes XRCC1 and hOGG1

    Cancer Epidemiol Biomark Prev

    (2003)
  • Q. Li et al.

    Association of DNA base-excision repair XRCC1, OGG1 and APE1 gene polymorphisms with nasopharyngeal carcinoma susceptibility in a Chinese population

    Asian Pac J Cancer Prev

    (2013)
  • N. Laantri et al.

    XRCC1 and hOGG1 genes and risk of nasopharyngeal carcinoma in North African countries

    Mol Carcinog

    (2011)
  • Q.Y. Chen et al.

    Concurrent chemoradiotherapy vs radiotherapy alone in stage II nasopharyngeal carcinoma: phase III randomized trial

    J Natl Cancer Inst

    (2011)
  • H.W. Mak et al.

    Clinical outcome among nasopharyngeal cancer patients in a multi-ethnic society in Singapore

    PLoS One

    (2015)
  • J. Chen et al.

    Radiation induced temporal lobe necrosis in patients with nasopharyngeal carcinoma: a review of new avenues in its management

    Radiat Oncol (London, England)

    (2011)
  • K.C. Chen et al.

    Postirradiated carotid blowout syndrome in patients with nasopharyngeal carcinoma: a case-control study

    Head Neck

    (2015)
  • G. Henle et al.

    Epstein-Barr virus-specific IgA serum antibodies as an outstanding feature of nasopharyngeal carcinoma

    Inter J Cancer

    (1976)
  • J.C. Lin et al.

    Quantification of plasma Epstein-Barr virus DNA in patients with advanced nasopharyngeal carcinoma

    New Engl J Med

    (2004)
  • H. zur Hausen et al.

    EBV DNA in biopsies of Burkitt tumours and anaplastic carcinomas of the nasopharynx

    Nature

    (1970)
  • M. Tokunaga et al.

    Epstein-Barr virus in gastric carcinoma

    Am J Pathol

    (1993)
  • K.J. Flavell et al.

    Hodgkin's disease and the Epstein-Barr virus

    Mol Pathol

    (2000)
  • C.D. Gregory et al.

    Different epstein-Barr virus-B cell interactions in phenotypically distinct clones of a Burkitt's lymphoma cell line

    J Gen Virol

    (1990)
  • M. Rowe et al.

    Differences in B cell growth phenotype reflect novel patterns of Epstein-Barr virus latent gene expression in Burkitt's lymphoma cells

    EMBO J

    (1987)
  • E.M. Deacon et al.

    Epstein-Barr virus and Hodgkin's disease: transcriptional analysis of virus latency in the malignant cells

    J Exp Med

    (1993)
  • L. Brooks et al.

    Epstein-Barr virus latent gene transcription in nasopharyngeal carcinoma cells: coexpression of EBNA1, LMP1, and LMP2 transcripts

    J Virol

    (1992)
  • E. Tse et al.

    Epstein Barr virus-associated lymphoproliferative diseases: the virus as a therapeutic target

    Exp Mol Med

    (2015)
  • M.R. Ghigna et al.

    Epstein-Barr virus infection and altered control of apoptotic pathways in posttransplant lymphoproliferative disorders

    Pathobiol: J Immunopathol Mol Cell Biol

    (2013)
  • R. Pathmanathan et al.

    Clonal proliferations of cells infected with Epstein-Barr virus in preinvasive lesions related to nasopharyngeal carcinoma

    New Engl J Med

    (1995)
  • L.S. Young et al.

    Identification of a human epithelial cell surface protein sharing an epitope with the C3d/Epstein-Barr virus receptor molecule of B lymphocytes

    Int J Cancer

    (1989)
  • C.T. Lin et al.

    The mechanism of Epstein-Barr virus infection in nasopharyngeal carcinoma cells

    Am J Pathol

    (1997)
  • R. Kuppers

    B cells under influence: transformation of B cells by Epstein-Barr virus

    Nat Rev Immunol

    (2003)
  • C. Munz et al.

    Human CD4(+) T lymphocytes consistently respond to the latent Epstein-Barr virus nuclear antigen EBNA1

    J Exp Med

    (2000)
  • S.P. Lee et al.

    CD8 T cell recognition of endogenously expressed epstein-barr virus nuclear antigen 1

    J Exp Med

    (2004)
  • Cited by (17)

    • Natural killer cells play an important role in virus infection control: Antiviral mechanism, subset expansion and clinical application

      2021, Clinical Immunology
      Citation Excerpt :

      NK cells have also attempted to be an alternative approach for adoptive cell therapy in NPC [73]. Due to the shorter lifespan of NK cells compared to cytotoxic T cells (CTLs), it is necessary for a suicide vector to control the over-expansion of transferred cells [73]. Presently, in a phase I/II clinical trials, high-activity NK cells and expanded NK cells combined with cetuximab are currently used to treat refractory NPC (NCT 02507154).

    • Monocytic Myeloid-Derived Suppressor Cells Underpin Resistance to Adoptive T Cell Therapy in Nasopharyngeal Carcinoma

      2021, Molecular Therapy
      Citation Excerpt :

      An alternative to standard chemotherapy regimens is the use of immunotherapeutic strategies, which focus on increasing the immune response to cancer-associated antigens. Experimental NPC immunotherapies have included dendritic cell (DC) vaccination,3–5 immune checkpoint blockade (ICB),6–9 and cytotoxic T-lymphocyte (CTL) infusion.10–16 NPC and other virally derived cancers should be an ideal candidate for immunotherapeutic strategies due to the presence of nonself, viral antigens, which are more immunogenic than neo-epitopes found in cancers that arise from inherited or de novo mutations.

    View all citing articles on Scopus
    View full text