Review
Interplay among viral antigens, cellular pathways and tumor microenvironment in the pathogenesis of EBV-driven lymphomas

https://doi.org/10.1016/j.semcancer.2013.07.005Get rights and content

Abstract

Epstein–Barr virus (EBV) is a ubiquitous human γ-herpes virus that has established an elegant strategy to persist as a life-long asymptomatic infection in memory B lymphocytes. EBV has potent transforming properties for B lymphocytes and it is pathogenically associated with a variety of lymphomas of B or NK/T cell origin. The viral latency programs expressed can hijack or deregulate cellular pathways critical for cell proliferation and survival, while impairing anti-viral immune responses. Similar effects may also be induced by EBV-encoded micro-RNAs, which may have a pathogenic role particularly in lymphomas showing a restricted expression of viral proteins. Of note, recent data have challenged the view that only the EBV latency is relevant for lymphomagenesis, suggesting that lytic EBV replication may also contribute to the development of EBV-associated lymphoproliferations. The recent advances in the elucidation of the mechanisms underlying EBV-induced cell transformation and immune evasion are providing the rationale for innovative and tailored treatment approaches for EBV-driven lymphomas.

Introduction

The Epstein–Barr virus (EBV) is a ubiquitous human gamma-herpesvirus able to establish a lifelong infection in more than 90% of individuals [1]. It was originally discovered in cultured Burkitt lymphoma cells in 1964 and in 1997 it has been included among the “carcinogenic agents” by the World Health Organization (WHO) on the basis of its direct transforming potential and close association with human malignancies, mainly lymphomas. Primary infection is usually asymptomatic and only when it is delayed until adolescence or adulthood, which occurs mainly in developed countries, it can cause a self-limiting lymphoproliferative disorder, known as infectious mononucleosis (IM). The main target of EBV infection is the B lymphocyte, although T, NK, and epithelial cells may be also infected in vivo, as indicated by the occurrence of EBV-carrying malignancies that originate from these cells. B lymphocytes are infected through interaction of the virus with the complement receptor-2/CD21 expressed at the cell surface. The infection is usually non-productive, latent, whereas intermittent reactivation and virus replication at epithelial surfaces allows the spreading of EBV to new hosts. Under physiological conditions, EBV-seropositive adults carry 1–50 EBV-infected B lymphocytes per million cells in the peripheral blood [2]. These resting memory B lymphocytes show no or limited expression of virally encoded genes [2], [3] and are considered the reservoir of EBV latency. EBV reaches the memory B cell compartment through its ability to elegantly exploit the B-cell differentiation pathway. After initial infection of naïve B lymphocytes, EBV may establish different programs of latency that may be sequentially expressed according to the type, differentiation and activation status of infected cells, finally reaching the memory B-cell compartment. Shortly after infection, EBV activates a program that includes the full set of latency proteins, the six EBV nuclear antigens (EBNAs) and three latent membrane proteins (LMP-1, LMP-2A, LMP-2B). This broad latency pattern (type III latency) activates and induces proliferation of B lymphocytes, which can be initiated in vitro in continuously growing lymphoblastoid cell lines (LCL). A second latency program (type II) occurs in the germinal centers of lymphoid follicles where EBV-carrying B lymphoblasts migrate, as normal B lymphocytes do after having encountered the cognate antigen [4]. These cells express only EBNA-1 and the LMPs (type II latency), a so-called “rescue” program that provides signals allowing EBV-infected cells to survive and differentiate into memory B lymphocytes [5]. Cells leaving the germinal center as resting memory B lymphocytes silence the expression of all EBV latency proteins (type 0 latency) or may express EBNA-1 upon cell division (type I latency), a strict requirement for replication of the viral genome and its maintenance within the cell. This elegant strategy allows EBV-carrying cells to escape immune surveillance while establishing lifelong persistence in humans. In several experimental systems, an additional latency program was detected in which the nuclear antigens EBNAs, but no LMP-1, are expressed. This program, denoted as type IIb latency, does not induce proliferation and thus probably does not contribute to tumor development [6].

In all forms of latency, EBV expresses the EBERs, small non-polyadenylated, non-coding double-strand RNAs, which may also contribute to EBV-driven B-cell immortalization [7]. EBV may activate the lytic replication program upon terminal differentiation of EBV-infected memory B lymphocytes into antibody-secreting plasma cells [5]. Although well equipped to promote the growth of B lymphocytes, EBV may drive the proliferation of these cells only transiently in immunocompetent hosts. This is due to the existence of a complex, strictly regulated immunological control involving various humoral and cellular effectors of immunity. Through the long time evolutionary adaptation in humans, the virus has evolved several potent mechanisms by which the type III B lymphocytes that express the “growth program” evade the immune response. This may also explain why only a limited proportion of EBV-seropositive individuals develop EBV-associated lymphomas, even in the setting of immune deficiency. In rare cases, therefore, a derangement of the normal differentiation pathway may prevent infected B lymphocytes from entering into a resting state, allowing thus EBV to fully manifest its transforming potential.

Section snippets

EBV-associated B-cell lymphomas

EBV-associated B-cell lymphoproliferative disorders include Burkitt lymphoma (BL), classic Hodgkin's lymphoma (HL), post-transplant lymphoproliferative disorders (PTLD), HIV-associated lymphoproliferative disorders (including primary central nervous system lymphoma, diffuse large B-cell lymphoma (DLBCL) with immunoblastic morphology, KSHV-positive primary effusion lymphoma and its solid variant, and plasmablastic lymphoma), and other rare histotypes (including lymphomatoid granulomatosis,

Concluding remarks

Our knowledge on the multiple mechanisms exploited by EBV for the development of lymphoid malignancies has greatly improved over the last years, providing an increasing number of new therapeutic targets. Highly promising immune interventions are based on the possibility to generate specific cytotoxic T lymphocytes targeting the immunogenic latent EBV proteins [197], [198]. Significant clinical benefits were particularly registered in the management of PTLD, whereas the outcome of patients with

Conflict of interest

The authors declare that there are no conflicts of interest.

Acknowledgements

The study was supported by a grant from the Associazione Italiana per la Ricerca sul Cancro (contract 10301 to R.D.) and from the Swedish Cancer Society (Cancerfonden).

References (199)

  • K. Belhadj et al.

    Hepatosplenic gammadelta T-cell lymphoma is a rare clinicopathologic entity with poor outcome: report on a series of 21 patients

    Blood

    (2003)
  • G. Klein et al.

    Interaction of Epstein–Barr virus (EBV) with human B-lymphocytes

    Biochemical and Biophysical Research Communications

    (2010)
  • M.A. Calderwood et al.

    Epstein–Barr virus nuclear protein 3C binds to the N-terminal (NTD) and beta trefoil domains (BTD) of RBP/CSL; only the NTD interaction is essential for lymphoblastoid cell growth

    Virology

    (2011)
  • C.W. Dawson et al.

    The role of the EBV-encoded latent membrane proteins LMP1 and LMP2 in the pathogenesis of nasopharyngeal carcinoma (NPC)

    Seminars in Cancer Biology

    (2012)
  • D. Wang et al.

    An EBV membrane protein expressed in immortalized lymphocytes transforms established rodent cells

    Cell

    (1985)
  • N. Raab-Traub

    Novel mechanisms of EBV-induced oncogenesis

    Current Opinion in Virology

    (2012)
  • K. Jones et al.

    Expansion of EBNA1-specific effector T cells in posttransplantation lymphoproliferative disorders

    Blood

    (2010)
  • S. Gottschalk et al.

    An Epstein–Barr virus deletion mutant associated with fatal lymphoproliferative disease unresponsive to therapy with virus-specific CTLs

    Blood

    (2001)
  • T. Frisan et al.

    Local suppression of Epstein–Barr virus (EBV)-specific cytotoxicity in biopsies of EBV-positive Hodgkin's disease

    Blood

    (1995)
  • A.G. Eliopoulos et al.

    Activation of the p38 mitogen-activated protein kinase pathway by Epstein–Barr virus-encoded latent membrane protein 1 coregulates interleukin-6 and interleukin-8 production

    Journal of Biological Chemistry

    (1999)
  • J. Ouyang et al.

    Viral induction and targeted inhibition of galectin-1 in EBV+ posttransplant lymphoproliferative disorders

    Blood

    (2011)
  • M.J. Allday

    How does Epstein–Barr virus (EBV) complement the activation of Myc in the pathogenesis of Burkitt's lymphoma?

    Seminars in Cancer Biology

    (2009)
  • M.S. Lindstrom et al.

    Role of genetic and epigenetic changes in Burkitt lymphoma

    Seminars in Cancer Biology

    (2002)
  • N. Nagy et al.

    To the genesis of Burkitt lymphoma: regulation of apoptosis by EBNA-1 and SAP may determine the fate of Ig-myc translocation carrying B lymphocytes

    Seminars in Cancer Biology

    (2009)
  • V. Saridakis et al.

    Structure of the p53 binding domain of HAUSP/USP7 bound to Epstein–Barr nuclear antigen 1 implications for EBV-mediated immortalization

    Molecular Cell

    (2005)
  • N. Yamamoto et al.

    Malignant transformation of B lymphoma cell line BJAB by Epstein–Barr virus-encoded small RNAs

    FEBS Letters

    (2000)
  • S.F. Fischer et al.

    Proapoptotic BH3-only protein Bim is essential for developmentally programmed death of germinal center-derived memory B cells and antibody-forming cells

    Blood

    (2007)
  • J.I. Cohen

    Epstein–Barr virus infection

    New England Journal of Medicine

    (2000)
  • E.M. Miyashita et al.

    Identification of the site of Epstein–Barr virus persistence in vivo as a resting B cell

    Journal of Virology

    (1997)
  • D.A. Thorley-Lawson

    Epstein–Barr virus: exploiting the immune system

    Nature Reviews Immunology

    (2001)
  • E. Klein et al.

    EBV genome carrying B lymphocytes that express the nuclear protein EBNA-2 but not LMP-1: type IIb latency

    Oncoimmunology

    (2013)
  • M. Yajima et al.

    Critical role of Epstein–Barr Virus (EBV)-encoded RNA in efficient EBV-induced B-lymphocyte growth transformation

    Journal of Virology

    (2005)
  • A. Carbone et al.

    EBV-associated lymphoproliferative disorders: classification and treatment

    Oncologist

    (2008)
  • I. Magrath

    Epidemiology: clues to the pathogenesis of Burkitt lymphoma

    British Journal of Haematology

    (2012)
  • E.S. Jaffe

    The 2008 WHO classification of lymphomas: implications for clinical practice and translational research

    Hematology. American Society of Hematology. Education Program

    (2009)
  • S. Swerdlow et al.

    World Health Organization classification of tumors, pathology and genetics of tumours of Hematopoietic and Lymphoid Tissues

    (2008)
  • K.J. Flavell et al.

    Hodgkin's disease and the Epstein–Barr virus

    Molecular Pathology

    (2000)
  • H. Hjalgrim et al.

    Infectious mononucleosis, childhood social environment, and risk of Hodgkin lymphoma

    Cancer Research

    (2007)
  • H.S. Khalidi et al.

    Detection of Epstein–Barr virus in the L & H cells of nodular lymphocyte predominance Hodgkin's disease: report of a case documented by immunohistochemical, in situ hybridization, and polymerase chain reaction methods

    American Journal of Clinical Pathology

    (1997)
  • C.F. McKhann

    Primary malignancy in patients undergoing immunosuppression for renal transplantation

    Transplantation

    (1969)
  • I. Penn et al.

    Malignant lymphomas in transplantation patients

    Transplantation Proceedings

    (1969)
  • M.P. Thompson et al.

    Epstein–Barr virus and cancer

    Clinical Cancer Research

    (2004)
  • J.E. Davis et al.

    Treatment options for post-transplant lymphoproliferative disorder and other Epstein–Barr virus-associated malignancies

    Tissue Antigens

    (2004)
  • M. Raphael et al.

    Lymphomas associated with infection by the human immune deficiency virus (HIV). World Health Organization Classification of Tumours, Pathology and Genetics of Tumours of Haematopoietic and Lymphoid Tissue Ed. by E.S. Jaffe, N.L. Harris, H. Stein, and J.W. Varidman

    (2001)
  • E. Cesarman et al.

    Kaposi's sarcoma-associated herpesvirus-like DNA sequences in AIDS-related body-cavity-based lymphomas

    New England Journal of Medicine

    (1995)
  • H.J. Delecluse et al.

    Epstein Barr virus-associated tumours: an update for the attention of the working pathologist

    Journal of Clinical Pathology

    (2007)
  • A. Carbone et al.

    AIDS-related plasmablastic lymphomas of the oral cavity and jaws: a diagnostic dilemma

    Annals of Otology, Rhinology and Laryngology

    (1999)
  • T. Oyama et al.

    Age-related EBV-associated B-cell lymphoproliferative disorders constitute a distinct clinicopathologic group: a study of 96 patients

    Clinical Cancer Research

    (2007)
  • P. Adam et al.

    Epstein–Barr virus-positive diffuse large B-cell lymphomas of the elderly

    Advances in Anatomic Pathology

    (2011)
  • H.H. Wong et al.

    Epstein–Barr virus positive diffuse large B-cell lymphoma of the elderly

    Leukemia and Lymphoma

    (2009)
  • Cited by (61)

    View all citing articles on Scopus
    View full text