Abstract
Extranodal NK/T-cell lymphoma (ENKTL) is a rare aggressive extranodal non-Hodgkin lymphoma (NHL) universally associated with Epstein-Barr virus (EBV). ENKTL most commonly occurs in non-elderly immune competent males in Asia and South America. A number of antecedent lymphoproliferative disorders (LPDs) have been described in Asian and South American patients, but the majority of Caucasian ENKTL patients have no known preceding LPD or underlying immunodeficiency. Other than EBV, no environmental or extrinsic factor has been implicated in oncogenesis. The precise mechanisms by which EBV infects NK or T cells and the virus’ role in the pathogenesis of ENKTL have not been fully deciphered. However, a number of recent discoveries including disturbances in cell signaling and mutations in tumor suppressor genes have been identified, which are providing insights into the pathogenesis of ENKTL. In this review, we highlight the molecular, viral, and genetic underpinnings of ENKTL and discuss potential therapeutic implications.
Introduction
Early descriptions of destructive nasal tumors, consistent with extranodal NK/T-cell lymphoma (ENKTL), date back over 100 years (McBride, 1991; Woods, 1921), but the cell of origin in cases of “lethal midline granuloma” or “rhinitis gangrenosa progressiva” (other terms included polymorphic reticulosis or malignant midline reticulosis) was not identified until 1982 (Ishii et al., 1982). Ishii and colleagues showed that the malignant cells within these lesions reacted with antibodies against T-cell antigens but not B-cell antigens. Subsequently, antibodies against the CD56 antigen questioned the T-cell lineage of the malignant cells (Kanavaros et al., 1993; Suzumiya et al., 1994; Wong et al., 1992). Over the past 35 years, a body of evidence has emerged supporting the notion that the majority of these tumors are of natural killer (NK) cell origin, with germline T-cell receptor gene configurations (Ho et al., 1990; Wong et al., 1992). The explanation for the original finding describing this neoplasm as a T-cell lymphoma was due to the reactivity of polyclonal anti-CD3 antibodies against the cytoplasmic subunit (ε-chain) of the CD3 molecule, which is preserved in NK-cells (Chan et al., 1996), in contrast to surface CD3, which is typically expressed in mature T-cells but not NK-cells. As a result of the initial misunderstanding, this NHL was designated as an NK/T-cell lymphoma and formally incorporated into the World Health Organization (WHO) classification of hematologic malignancies in 1999. It is further defined as extranodal as approximately 75% of cases present in the upper aerodigestive tract (UADT) with frequent angioinvasion and necrosis. Extranasal (non-UADT) ENKTL most commonly involves the gastrointestinal tract, skin, testis, lung, and soft tissue. The neoplastic cells are known to contain cytotoxic granule associated proteins [granzyme B, perforin, and T-cell restricted intracellular antigen (TIA-1)] and EBV-encoded RNAs (EBERs) (Chan et al., 1994; Harabuchi et al., 1990; Kawa-Ha et al., 1989). Classically, the neoplastic cells are described as activated NK-cells with expression of CD2, CD56, cytoplasmic CD3ε+ (surface CD3-), and germline T-cell receptor (Swerdlow et al., 2016). Several reports have documented the expression of killer-cell immunoglobulin-like receptors (KIRs) further validating their NK-cell phenotype (Dukers et al., 2001). A small subset of cases demonstrates a cytotoxic T-cell phenotype (sCD3+, CD56- with α/β or less frequently γ/δ TCR expression) (Pongpruttipan et al., 2012). In the recently revised WHO Classification, “ENKTL, nasal type” remains the only category for this neoplasm irrespective of cell of origin or site of disease.
Predisposing Factors in ENKTL
ENKTL is male predominant (∼2:1) and more common in East Asia and Central/South America compared to North America and Europe. Within North America and Europe, ENKTL is more common among Asian Pacific Islander and non-white Hispanics. Thus, there appears to be a genetic predisposition for increased risk of ENKTL in these populations. In other EBV+ lymphoproliferative disorders (LPDs) higher rates of T/NK-cell infection occur in Asian cohorts (Cohen et al., 2011b). It has also been shown that particular human leukocyte antigen (HLA) subtypes affect the success of T-cell surveillance for EBV and thereby influence a recipient’s predisposition to EBV-associated lymphomas (Reshef et al., 2011). For example, Lustberg and colleagues showed that the HLA-B8 was associated with T-cell post-transplant lymphoproliferative disorder (PTLD) (Lustberg et al., 2015). In vitro studies suggest that ENKTL cells retain major histocompatibility (MHC) class I and class II antigen processing function and can be recognized by CD8+ and CD4+ T cells targeting proteins produced by EBV [i.e., latent membrane protein-2 (LMP2) and Epstein-Barr nuclear antigen-1 (EBNA1)]. It remains unclear if specific HLA subtypes are more common in ENKTL. We hypothesize that there are certain HLA subtypes more common in Asian males that account for the higher incidence in this cohort. The genetic susceptibility to ENKTL remains an interesting area of discovery in ENKTL along with other EBV-associated T/NK-cell LPDs.
Asian and U.S. cancer registry data show significantly lower rates of ENKTL in U.S. Asians compared to native Asians (Bassig et al., 2016). This suggests that in addition to increased genetic susceptibility in some races, there may be environmental factors more common in certain areas of the globe contributing to the increased incidence of ENKTL in the East. Co-factors such as chronic inflammation, co-infection(s), time of primary EBV infection, malaria (Rickinson, 2014), and HIV (Carbone et al., 2009), which are implicated as cofactors in the lymphomagenesis of EBV-associated Burkitt lymphoma, have been suggested (Rickinson, 2014). In ENKTL, there are no co-factors, other than EBV, that are clearly associated; however, a subset of ENKTL cases is preceded by chronic inflammation or an LPD. Well characterized antecedent LPDs include chronic active EBV (CAEBV) (Jones et al., 1988; Kawa-Ha et al., 1989; Kimura et al., 2001; Ohshima et al., 1998), hemophagocytic lymphohistiocytosis (HLH), hypersensitivity to mosquito bite (HMB) (Ishihara et al., 1997; Kawa et al., 2001), and hydroa vacciniforme-like lymphoma (HVLL) (Quintanilla-Martinez et al., 2013). Predominantly in patients from South America HVLL is reported to precede ENKTL (Quintanilla-Martinez et al., 2013). HMB is shown to precede several NK-cell LPDs, including NK-cell CAEBV and ENKTL (Kawa et al., 2001; Takahashi et al., 2011). Unifying links between this spectrum of LPDs are EBV infection and inflammation. Previously studied factors related to EBV+ T/NK-cell growth include interleukin-9 as an autocrine growth factor (Nagato et al., 2005; Yang et al., 2004), interleukin-10 as an enhancer of responsiveness to interleukin-2 (Harabuchi et al., 2009), the interferon gammainducible protein 10 as a promoter of tumor invasion (Moriai et al., 2009), and CD70 as a cell surface receptor for soluble CD27, which is a paracrine growth factor (Yoshino et al., 2013). This work underscores the importance of the microenvironment in ENKTL and dependence of a cross-talk with surrounding cell types for adequate EBV-infection of NK/T cells and subsequent growth and survival. This is exemplified by recent studies showing that monocytes, one of the major contributors to the nasal tumor infiltrate, deliver interleukin-15 to EBV-positive NK/T cells, inducing the EBV latent membrane protein-1 (LMP1) oncogene expression and secretion of the LMP1-inducible cytokine IP10 which works as a chemoattractant to drive monocyte recruitment (Ishii et al., 2012). In summary there remain several well described antecedent LPDs; however, they do not consistently and reproducibly result in progression to ENKTL. Thus, additional genetic, environmental, or microenvironmental triggers are likely relevant to progression to ENKTL.
The majority of EBV+ lymphomas are of B-cell origin and often associated with a pre-existing primary or acquired immune deficiency. For example, post-transplant lymphoproliferative disorder (PTLD) can result from EBV driven lymphoproliferation and evolution to lymphoma in patients receiving iatrogenic immunosuppression. “EBV+ diffuse large B-cell lymphoma (DLBCL) of the elderly” (changed to “EBV+ DLBCL, NOS” in 2016 WHO Classification) with a median age of 77 years is thought to develop secondary to age related immune senescence (Castillo et al., 2016). Whereas, the vast majority of ENKTL patients have no known underlying immune deficiency with a median age at diagnosis of 55 years. Recent reports show that EBV+ DLBCL can be seen in young individuals and may be associated with a permissive microenvironment with upregulation of programmed death ligand 1 (PD-L1) and increased indoleamine 2,3-dioxygenase (IDO) positive cell content (Beltran et al., 2011; Nicolae et al., 2015; Ok et al., 2015). Similarly, a subset of ENKTL express PD-L1 (Kim et al., 2016), which may be prognostic (Kim et al., 2016). Takahashi et al. (2011) showed that younger ENKTL patients (i.e., <50 years of age) more frequently had B symptoms, worse performance status, advanced stage, and more sites of extranodal involvement. Future studies should focus on improving our understanding of the tumor microenvironment, immune response to EBV, and clinically unapparent immune dysregulation, which may differ by age or cohort. Ultimately, it is possible that specific genetic alterations or immune defects may predispose these clinically immune competent individuals to EBV-associated neoplasms.
NK/T-cells as a Target for EBV Infection
The first evidence that EBV infection was implicated in the progression to T or NK-cell neoplasms came from a report describing three patients (two of them adults) with clinical and serologic features of CAEBV who subsequently developed fatal T-cell lymphoma (Jones et al., 1988). Since then, the incidence of EBV T/NK-cell infection in published studies has been conflicting. Recently, a Japanese study compared the cellular target of EBV infection in the peripheral blood of patients with EBV-associated HLH, CAEBV, and infectious mononucleosis (IM). EBV infection was predominantly in CD8+ T-cells in EBV-associated HLH, whereas the dominant EBV infected cell populations were CD4+ T-cells and CD16+ NK-cells in CAEBV. In IM patients the predominant infected cell type was B-cells (Kasahara et al., 2001). In a study of a U.S. CAEBV patient cohort, EBV infected predominantly B-cells, whereby 58% (11/19) had a “B-cell disease” (Cohen et al., 2011b). With the exception of CAEBV, it’s unknown how frequently EBV infects T or NK-cells in non-Asian patients. These studies highlight the differences in rates of T/NK-cell infection across races. The reason for this remains an active area of investigations but could be related to genetic (i.e., HLA) differences, immune response, or variation in the viral genome. Interestingly, U.S. patients with CAEBV showed reduced NK-cell numbers and progressive loss of B-cells with hypogammaglobinemia; whereas patients with CAEBV from Japan had normal or increased numbers of NK cells (Cohen et al., 2011b), further highlighting the geodemographic differences in response to EBV infection. Irrespective of the differences in risk of T/NK-cell infection across races, it remains unclear how frequently EBV T/NK-cell infection progresses to T/NK-cell neoplasms.
Variations in the viral genome may predispose individuals to developing EBV-associated LPDs. EBV can be divided into two major types, mainly based on differences observed in EBNA genes (Dambaugh et al., 1984; Rowe et al., 1989). Recent work, including a large-scale sequencing study of EBV isolates from multiple tumor types and healthy carriers (Palser et al., 2015), suggests that while the distinction between EBV type 1 (EBV-1) and type 2 (EBV-2) is accurate and reproducible, the genomic diversity of EBV is greater than previously recognized (Chang et al., 2009). EBV-1 is more prevalent in the developed world (e.g., U.S., Europe, Asia) whereas EBV-2 is encountered more frequently in equatorial Africa. The impact that this diversity may have on the oncogenic properties of the virus remain unknown. EBV-1 readily transforms B-cells in culture, leading to the outgrowth of immortalized lymphoblastoid cell lines (LCL), while EBV-2 is poorly transforming (Dolan et al., 2006; Rowe et al., 1989). Recently, Coleman et al. (2015) showed that EBV-2 is able to efficiently infect CD8+ cytotoxic T-cells and induce proliferation and alter cytokine expression, although the relevance that this in vitro model may have on the development of ENKTL remains to be defined.
A recent French study in ENKTL patients detected EBV-1 in tumor and blood from all French natives (n=11), and EBV-2 in blood of two patients from Africa (corresponding tumor was not assessed). A control cohort without malignancy revealed only the presence of EBV-1 in blood (Halabi et al., 2016), which agrees with previously published results (Chiang et al., 1996; 1999; Kim et al., 2003; Kuo et al., 2004; Nagamine et al., 2007; Suzumiya et al., 1999). This study found a recurring 30 bp deletion (del30) in the LMP1 gene in 6 of 13 patients, which has been reported in 86–100% of Asian patients (Chiang et al., 1996; Kim et al., 2003; Kuo et al., 2004; Nagamine et al., 2007; Suzumiya et al., 1999; Tai et al., 2004). Other clonal variations were also reported, and in patients who achieved complete remission the wild-type form of LMP1 was more commonly detectable after treatment. Further investigation and comparison into clonal variations of EBV among U.S./European, Asian, and African cohorts at diagnosis and longitudinally after treatment is warranted. It’s unclear if these differences have any impact on the spectrum of clinical presentation observed in Asian populations compared to North American, European, South American, or African cases. Highlighted in a recently published update on the epidemiology and clinical characteristics of ENKTL, the clinical spectrum of disease described in North America and Europe is narrower, compared to Asia. This variability could be secondary to the small sample size in the West, publication bias, and the differences across studies in defining “extranodal NK/T-cell lymphoma” versus other EBV-associated T/NK-cell LPDs. In summary, it remains to be defined if genetic susceptibility or immune response across races, or if differences in the viral genome account for the higher rates of T/NK-cell infection in Asia.
Role of EBV in ENKTL
While the incidence of ENKTL and the antecedent LPD varies across the globe, EBV-encoded transcripts and proteins are universally detected in the neoplastic cells of all ENKTL patients, irrespective of race (Au et al., 2009; Chiang et al., 1996; Minarovits et al., 1994; van Gorp et al., 1996). Additionally EBV genomes in tumor lesions are demonstrably clonal (Minarovits et al., 1994; Tao et al., 1995). EBV is a ubiquitous lymphotropic gammaherpesvirus that infects >90% of the world population with a biological cycle of primary infection, latency, and lytic reactivation, whereby each latency type (I, II, or III) is characterized by a specific EBV transcriptional program, dictated in part by the level of immune competence of the host (Cohen et al., 2011a; Young and Rickinson, 2004). The demonstration that EBV most efficiently infects and transforms resting B-cells in vitro, using CD21 and HLA class II as coreceptors (Fingeroth et al., 1984; Li et al., 1997; Nemerow et al., 1985), and the observation that patients with IM have large numbers of circulating EBV-infected B-cells established the canonical view of EBV’s distinct tropism for B-cells. Furthermore, these observations contributed to the development of valuable models of primary infection, replication, latency, and life-long persistence (Babcock et al., 1998; Kurth et al., 2000). According to these models, EBV initially infects and replicates in the oropharynx via co-receptors expressed on B-cells (HLA class II, CD21, and beta 1 integrin) and epithelial cells (beta 1, αvβ6/8 integrins) and then, under the selective pressure of an effective cell-mediated immune response, turns off most of its genes and enters a state of latency, with resting memory B-cells being the primary reservoir. In ENKTL the tumor most commonly occurs at sites of primary EBV infection. It is hypothesized that EBV infection in ENKTL occurs while NK-cells are attempting to kill an EBV infected cell target (Tabiasco et al., 2003). The exact mechanism by which EBV infects T/NK-cells remains to be clarified.
The full spectrum of latent EBV genes expressed during the infection of B-cells includes six nuclear antigens [EBNA 1, 2, 3A, 3B, 3C, and leader protein (LP)], three latent membrane proteins (LMP1, 2A, and 2B), two small EBV-encoded RNA’s (EBER1 and EBER2), and three clusters of micro-RNAs (miRs). Available evidence suggests that EBV genome copy numbers within ENKTL biopsies typically number no more than 20 virions per cell, consistent with latent episomal infection, although higher loads indicative of lytic virus replication have been observed and may have prognostic relevance (Hsieh et al., 2007). EBV-encoded transcripts and proteins are detected usually with a latency program I, sometimes II (Asano et al., 2013; Matsuo and Drexler, 2003; Minarovits et al., 1994; Suzuki, 2014; Takahara et al., 2006; Tao et al., 1995; Xu et al., 2001). It has been proposed that quantitation or monitoring of EBV genome copy number in serum or whole blood is a prognostic and predictive biomarker (Au et al., 2004; Ito et al., 2012; Jaccard et al., 2011; Kanakry et al., 2016; Suzuki, 2014; 2011; Wang et al., 2012). However, since most quantitative PCR assays will detect both encapsulated virions and cell-free EBV DNA, viremia may simply reflect tumor shed DNA rather than virus replication. Therefore, the ideal prognostic and predictive EBV assay may vary by lymphoma subtype and the corresponding cell infected cell type. These observations require prospective evaluation alongside correlative virological studies, including in particular, a determination of the frequency and significance of EBV-harboring T and NK-cells in the peripheral blood at diagnosis and follow-up.
EBV is a highly adaptable tumor virus that can transform different cell types through constitutive activation of NF-kB, inhibition of apoptosis, activation of MYC, BCL2, and NOTCH1, and induction of extensive DNA methylation and genomic instability in the host cell. These effects are mediated by EBV latent proteins that function as transcriptional coactivators (EBNAs), signaling molecules (LMP-1, 2A), and epigenetic modifiers (EBNAs, LMP-1), affecting a broad range of transcriptional programs and pathways in the host cell. Furthermore, the immediate early (IE) gene BZLF1, which activates EBV’s lytic cycle, directly promotes B-cell lymphomagenesis and expression of BZLF1’s activator XBP1 is associated with poor outcomes in B-cell lymphoma. EBV is also known to influence the T-bet/GATA3 axis (Th1/Th2) in T-cells, which is associated with survival in PTCL (Iqbal et al., 2014). EBV causes upregulation of GATA3 expression in vitro (Siemer et al., 2008), and the EBV-encoded miRBART20-5p can inhibit T-bet translation potentially blocking differentiation towards Th1 lineage (Lin et al., 2013). The role of latent, lytic, and miRs in ENKTL are highlighted in Table 1.
Table 1.
Gene(s) | Primary Function | Major Findings (as it relates to gene expression in ENKTL) |
Reference(s) |
---|---|---|---|
EBER | In B cells, some evidence indicates that EBER expression protects against apoptosis and contributes to proliferation. | Thought to function via TLR3 to amplify the inflammatory response in HLH, CAEBV, and IM; unknown in ENKTL. | Iwakiri et al. J Exp Med, 2009. |
EBNA1 | Ensure faithful transmission of the circularized EBV episome to daughter cells by facilitating its replication during cell division. | Partial silencing in EBV+ NK cell line reduced cell proliferation. | Ian et al. Cancer Biol Ther, 2008. |
EBNA2 | DNA binding protein, interacts with cellular RBPJk. | Absent by IHC in tumors. | Chiang et al. Int J Cancer, 1996. |
EBNA3 | Affect transcription of viral and cellular genes. | Absent. | Chiang et al. Int J Cancer, 1996. |
LMP1 | Classic oncogene in B-cell transformation; modulator of cell signaling; induces a number of antiapoptotic proteins includes BCL2; functions to constitutively activate the TNF receptor and functionally resembles CD40, providing growth and differentiation signals to B-cells. | Expression is seen in the vast majority of cell lines but this does not mirror in vivo situation. Microenvironmental factors and cytokines (e.g., IL2, IL10) may be influential in expression in tumors. By IHC, some ENKTL tumors are LMP1-. | Chiang et al. Int J Cancer, 1996. |
LMP2 | Facilitate immortalization and lytic cycle but are not essential for B-cell transformation; may drive proliferation and survival of B-cells in the absence of BCR signaling. | Expression typically absent by IHC, although LMP2 specific CD8+ T-cells recognize and kill cell lines and induce clinical responses in patients. Subsequently, a novel LMP2 transcript was identified, which may serve as the target. | Fox et al. Blood, 2010. Chiang. Int J Cancer, 1996. |
BZLF | Immediate-early genes; expressed following lytic activation. | Negative ZEBRA IHC in tumors. | Chiang et al. Int J Cancer, 1996. |
BHRF1 miRNA cluster | No expression in cell lines. Detected in rare cells suggesting that lytic transcripts are most likely expressed by rare cells entering lytic cycle. | Chiang et al. Int J Cancer, 1996. | |
BART miRNA cluster | BART miRNAs are increased in cell lines. Mir-BART9 seems to influence expression of LMP1 and cell growth. mir-BART20-5p inhibits translation of T-bet in EBV-infected YT lymphoma cells of NK-cell origin. | Ramakrishnan et al. PLoS One, 2011. Lin et al. Am J Pathol, 2013. |
Gene Expression Profiling and Genomic Studies
In addition to the rarity of ENKTL, biopsy specimens are typically small and necrotic, and the availability of unfixed tissue for molecular genetic studies is limited. Thus, tumor derived cell lines remain a valuable resource. However, given the clinical and heterogeneous overlap between T/NK-cell LPDs and ENKTL, cell line data might be misleading. Table 2 highlights several ENKTL cell lines, which we believe, based on the clinical context and cell of origin, may reflect ENKTL as defined in 2016. Several “NK//T-cell lymphoma” cell lines are more likely to be other T/NK-cell LPDs as currently defined in 2016, such as CAEBV (e.g., KAI3, SNK10, SNK16, SNT13, SNT15), ANKL (KHYG1, IMC-1), and other subtypes of leukemia (NKL, YT). While it has been shown that many of the “NK/T-cell lymphoma” cell lines share genomic alterations, the ENKTL cell lines disclose overexpression of a number of genes related to growth factor activity, apoptosis, cell growth, signal transduction and cell adhesion in comparison to CAEBV-derived cell lines (Zhang et al., 2006). A model of pathogenesis in which the EBV oncoprotein LMP-1 induces the deregulation of p53, activation of C-MYC, and NFkB pathway, resulting in up-regulation of survivin has been proposed (Ng et al., 2011).
Table 2.
Cell Line | Patient Source | Cell Lineage |
---|---|---|
NKYS | 19 year old female from Japan | NK-cell, εCD3+, CD56+ |
SNK6 | 62 year old, nasal tumor from Japan | NK-cell, εCD3+, CD56+ |
NK92 | 50 year old non-nasal tumor from Caucasian | NK-cell, εCD3+, CD56+ |
SNK1 | 24 year old from Japan | NK-cell, εCD3+, CD56+ |
SNT8 | 48 year old, nasal tumor from Japan | γδT-cell, CD3+, CD56+ |
NK-S1 | Xenograft of nasal tumor from China (Loong et al. Leuk & Lymphoma, 2008) | εCD3+, TIA-1+, CD56- |
It must be emphasized, that most studies have examined small cohorts of cases, sometimes heterogeneous, and that many were conducted in the late 1990s or early 2000s, a period when the diagnostic criteria for ENKTL, had not yet been fully established. Table 3 highlights several previous genetic, epigenetic, and miRNA studies in ENKTL. The most frequent chromosomal losses are observed at 1p, 6q, 11q, 13q, and 17p, and the gains most commonly at 1q, 2q, 7q, 17q, and 20q. The frequently observed DNA losses at chromosomes 6q and 13q suggested the implication of tumor suppressor genes mapping to these loci, in the pathogenesis of ENKTL. In more recent array comparative genomic hybridization (CGH) studies, the deletion of chromosome 6q (6q21–6q25) was found in 40–50% of ENKTL cases (Huang et al., 2010; Iqbal et al., 2009; 2011; Ko et al., 2001; Nakashima et al., 2005; Siu et al., 1999; Sun et al., 2003; Taborelli et al., 2006).
Table 3.
Authors | Methodology | Source | Findings |
---|---|---|---|
Yamaguchi et al. Cancer, 1995. | IHC and reverse transcription (RT) PCR | Ten Japanese patients with nasal NK/T-cell lymphoma | Nine of the 10 patients were P-glycoprotein positive by IHC. MDR1 mRNA was detected in all seven pts examined by RT-PCR. |
Wong et al. BJH, 1997. | Conventional karyotype | 7 CD56+ leukemia/lym-phoma (2 extranasal, 1 nasal, 3 ANKL, 1 blastoid leukemia/lymphoma) | Del(6)(q21q25) is a recurring abnormality. |
Siu et al. Am J Pathol, 1999. | CGH | Primary tumors from four nasal NK cell lymphomas, one nasal-type NK cell lym-phoma, and five NK cell lymphomas/leukemias | Deletions at 6q16-q27 (four cases), 13q14-q34 (three cases), 11q22-q25 (two cases), and 17p13 (two cases). |
Ko et al. Cytometry, 2001. | CGH | 7 nasal ENKTL (6 NK-ENKTL, 1 T-ENKTL) | Frequent DNA losses at 1p, 17p, and 12q and gains at 2q, 13q, and 10q. Infrequent loss of 6q comprise several candidate tumor suppressor genes (PRDM1, HACE1, FOX03, AIM1, ATG5). |
Nakashima et al. Genes Chromosomes Cancer, 2005. | Homemade array-based CGH | 10 aggressive NK-cell leukemia cases and 17 ENKTL, nasal type | Gain of 1q23.1–24.2 and 1q31.3–q44), and loss of 7p15.1–p22.3 and 17p13.1 occurred significantly more frequently in ENKTL. Gain of 2q, and loss of 6q16.1–q27, 11q22.3–q23.3, 5p14.1–p14.3, 5q34–q35.3, 1p36.23–p36.33, 2p16.1–p16.3, 4q12, and 4q31.3–q32.1 were nonsignificantly more common in ENKTL. |
Iqbal et al. Leukemia, 2009. | CGH & GEP | NK cell lines & 7 patients with NK-cell malignancies | PRDM1 was the most likely tumor promoting gene in del6q21. ATG5 and AIM1 may also participate in the tumor development and progression. |
Huang et al. Blood, 2010. | GEP | 9 tumors (1 T-cell ENKTL, 8 NK-cell ENKTL) & 2 cell lines (SNK6 and SNK8) | Compared to normal NK cells, tumors were closer to activated than resting cells and overexpressed several genes related to vascular biology, EBV induced genes, and PDGFRA. Integrative analysis also evidenced deregulation of the tumor suppressor HACE1 in the frequently deleted 6q21 region. |
Jiang et al. Nature Genetics, 2015. | Whole exome sequencing | 25 de novo ENKTL | Recurrent mutations were most frequently located in the RNA helicase gene DDX3X (20%), tumor suppressors (TP53 & MGA), JAK-STAT pathway molecules (STAT3 & STAT5B), and epigenetic modifiers (MLL2, ARID1A, EP300, & ASXL3). |
Kucuk et al. Clin Cancer Res, 2015. | Global promotor methylation analysis | 12 ENKTL and 7 NK cell lines (NK92, KHYG1, YT, SNK1, SNK6, NKYS, & KAI3) | Identified 95 genes with strong evidence for being silenced because of promotor methylation, including BCL2L11 (BIM), DAPK1, PTPN6 (SHP1), TET2, SOCS6, and ASNS. |
Emerging Therapies
Anthracycline-based chemotherapy (i.e., CHOP) has now been shown to be largely ineffective, in part due to high levels of expression of P-glycoprotein (Wang et al., 2008). Additionally, the SMILE (dexamethasone, methotrexate, ifosfamide, L-asparaginase, etoposide) regimen has demonstrated significant efficacy, and has become standard–particularly in patients with advanced disease (Kwong et al., 2012; Yamaguchi et al., 2011). However, numerous novel therapies are promising in ENKTL. Brentuximab vedotin is an anti-CD30 antibody conjugate, which is efficacious in CD30-positive lymphomas (Younes et al., 2010). CD30 is expressed in about 70% of ENKTL (Sabattini et al., 2013). Lenalidomide is an immunomodulatory analog (IMiDs) with activity in lymphoid malignancies, including T-cell lymphomas, primarily through immune modulation (Kritharis et al., 2015; Toumishey et al., 2015). Further studies are required to examine the effectiveness of these agents in ENKTL. Additional immunomodulatory strategies include use of checkpoint inhibitors. As a single agent in an unselected cohort, checkpoint blockade was not very effective in T-cell neoplasms; however, EBV upregulates PD-L1 and in ENKTL checkpoint blockade has been more promising (Kwong et al., 2017). Antigen-specific T-cells targeting immunodominant viral antigens from EBV have been used with dramatic success to treat EBV-associated PTLD (Doubrovina et al., 2012; Heslop et al., 2010). Patients with ENKTL are associated with type I/II EBV latency, where only weakly immunogenic EBV antigens LMP1, LMP2, and EBNA1 are expressed (Fox et al., 2010). Despite this, of six ENKTL patients treated with LMP-CTLs, four had complete responses, which remained in remission at a median of 3.1 years after CTL infusion (Bollard et al., 2014). This study indicates autologous T-cells directed to the LMP antigens can induce durable complete responses without significant toxicity. Development of EBV-CTLs against EBNA1 or novel methods of upregulating LMP1 may be better strategies in ENKTL. Unfortunately, EBV-CTLs require week(s) to produce and are thus often not available. A donor derived “off the shelf” bank of EBV-CTLs is being developed (Hanley et al., 2012), and if the risk of graft versus host can be minimized, it will be an attractive option. Additional, targeted therapies include JAK inhibitors, and mTOR inhibitors. Ultimately, combinatorial or sequential strategies that optimally exploit genomic, viral, and immunologic properties of the tumor will succeed.
Footnotes
Disclosure
The authors report no conflicts of interest.
References
- Asano N, Kato S, Nakamura S. Epstein-Barr virus-associated natural killer/T-cell lymphomas: best practice and research. Clin Haematol. 2013;26(1):15–21. doi: 10.1016/j.beha.2013.04.002. [DOI] [PubMed] [Google Scholar]
- Au WY, Pang A, Choy C, Chim CS, Kwong YL. Quantification of circulating Epstein-Barr virus (EBV) DNA in the diagnosis and monitoring of natural killer cell and EBV-positive lymphomas in immunocompetent patients. Blood. 2004;104(1):243–249. doi: 10.1182/blood-2003-12-4197. [DOI] [PubMed] [Google Scholar]
- Au WY, Weisenburger DD, Intragumtornchai T, Nakamura S, Kim WS, Sng I, Vose J, Armitage JO, Liang R. Clinical differences between nasal and extranasal natural killer/T-cell lymphoma: a study of 136 cases from the International Peripheral T-Cell Lymphoma Project. Blood. 2009;113(17):3931–3937. doi: 10.1182/blood-2008-10-185256. [DOI] [PubMed] [Google Scholar]
- Babcock GJ, Decker LL, Volk M, Thorley-Lawson DA. EBV persistence in memory B cells in vivo. Immunity. 1998;9(3):395–404. doi: 10.1016/s1074-7613(00)80622-6. [DOI] [PubMed] [Google Scholar]
- Bassig BA, Au WY, Mang O, Ngan R, Morton LM, Ip DK, Hu W, Zheng T, Seow WJ, Xu J, Lan Q, Rothman N. Subtype-specific incidence rates of lymphoid malignancies in Hong Kong compared to the United States, 2001–2010. Cancer Epidemiol. 2016;42:15–23. doi: 10.1016/j.canep.2016.02.007. [DOI] [PubMed] [Google Scholar]
- Beltran BE, Morales D, Quinones P, Medeiros LJ, Miranda RN, Castillo JJ. EBV-positive diffuse large b-cell lymphoma in young immunocompetent individuals. Clin Lymphoma Myeloma Leuk. 2011;11(6):512–516. doi: 10.1016/j.clml.2011.07.003. [DOI] [PubMed] [Google Scholar]
- Bollard CM, Gottschalk S, Torrano V, Diouf O, Ku S, Hazrat Y, Carrum G, Ramos C, Fayad L, Shpall EJ, Pro B, Liu H, Wu MF, Lee D, Sheehan AM, Zu Y, Gee AP, Brenner MK, Heslop HE, Rooney CM. Sustained complete responses in patients with lymphoma receiving autologous cytotoxic T lymphocytes targeting Epstein-Barr virus latent membrane proteins. J Clin Oncol. 2014;32(8):798–808. doi: 10.1200/JCO.2013.51.5304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carbone A, Cesarman E, Spina M, Gloghini A, Schulz TF. HIV-associated lymphomas and gamma-herpesviruses. Blood. 2009;113(6):1213–1224. doi: 10.1182/blood-2008-09-180315. [DOI] [PubMed] [Google Scholar]
- Castillo JJ, Beltran BE, Miranda RN, Young KH, Chavez JC, Sotomayor EM. EBV-positive diffuse large B-cell lymphoma of the elderly: 2016 update on diagnosis, risk-stratification, and management. Am J Hematol. 2016;91(5):529–537. doi: 10.1002/ajh.24370. [DOI] [PubMed] [Google Scholar]
- Chan JK, Tsang WY, Ng CS. Clarification of CD3 immunoreactivity in nasal T/natural killer cell lymphomas: the neoplastic cells are often CD3 epsilon+ Blood. 1996;87(2):839–841. [PubMed] [Google Scholar]
- Chan JK, Yip TT, Tsang WY, Ng CS, Lau WH, Poon YF, Wong CC, Ma VW. Detection of Epstein-Barr viral RNA in malignant lymphomas of the upper aerodigestive tract. Am J Surg Pathol. 1994;18(9):938–946. doi: 10.1097/00000478-199409000-00009. [DOI] [PubMed] [Google Scholar]
- Chang CM, Yu KJ, Mbulaiteye SM, Hildesheim A, Bhatia K. The extent of genetic diversity of Epstein-Barr virus and its geographic and disease patterns: a need for reappraisal. Virus Res. 2009;143(2):209–221. doi: 10.1016/j.virusres.2009.07.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chiang AK, Tao Q, Srivastava G, Ho FC. Nasal NK- and T-cell lymphomas share the same type of Epstein-Barr virus latency as nasopharyngeal carcinoma and Hodgkin’s disease. Int J Cancer. 1996;68(3):285–290. doi: 10.1002/(SICI)1097-0215(19961104)68:3<285::AID-IJC3>3.0.CO;2-Y. [DOI] [PubMed] [Google Scholar]
- Chiang AK, Wong KY, Liang AC, Srivastava G. Comparative analysis of Epstein-Barr virus gene polymorphisms in nasal T/NK-cell lymphomas and normal nasal tissues: implications on virus strain selection in malignancy. Int J Cancer. 1999;80(3):356–364. doi: 10.1002/(sici)1097-0215(19990129)80:3<356::aid-ijc4>3.0.co;2-d. [DOI] [PubMed] [Google Scholar]
- Cohen JI, Fauci AS, Varmus H, Nabel GJ. Epstein-Barr virus: an important vaccine target for cancer prevention. Sci Transl Med. 2011a;3(107):107fs107. doi: 10.1126/scitranslmed.3002878. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cohen JI, Jaffe ES, Dale JK, Pittaluga S, Heslop HE, Rooney CM, Gottschalk S, Bollard CM, Rao VK, Marques A, Burbelo PD, Turk SP, Fulton R, Wayne AS, Little RF, Cairo MS, El-Mallawany NK, Fowler D, Sportes C, Bishop MR, et al. Characterization and treatment of chronic active Epstein-Barr virus disease: a 28-year experience in the United States. Blood. 2011b;117(22):5835–5849. doi: 10.1182/blood-2010-11-316745. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Coleman CB, Wohlford EM, Smith NA, King CA, Ritchie JA, Baresel PC, Kimura H, Rochford R. Epstein-Barr virus type 2 latently infects T cells, inducing an atypical activation characterized by expression of lymphotactic cytokines. J Virol. 2015;89(4):2301–2312. doi: 10.1128/JVI.03001-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dambaugh T, Hennessy K, Chamnankit L, Kieff E. U2 region of Epstein-Barr virus DNA may encode Epstein-Barr nuclear antigen 2. Proc Natl Acad Sci. 1984;81(23):7632–7636. doi: 10.1073/pnas.81.23.7632. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dolan A, Addison C, Gatherer D, Davison AJ, Mcgeoch DJ. The genome of Epstein-Barr virus type 2 strain AG876. Virology. 2006;350(1):164–170. doi: 10.1016/j.virol.2006.01.015. [DOI] [PubMed] [Google Scholar]
- Doubrovina E, Oflaz-Sozmen B, Prockop SE, Kernan NA, Abramson S, Teruya-Feldstein J, Hedvat C, Chou JF, Heller G, Barker JN, Boulad F, Castro-Malaspina H, George D, Jakubowski A, Koehne G, Papadopoulos EB, Scaradavou A, Small TN, Khalaf R, Young JW, et al. Adoptive immunotherapy with unselected or EBV-specific T cells for biopsy-proven EBV+ lymphomas after allogeneic hematopoietic cell transplantation. Blood. 2012;119(11):2644–2656. doi: 10.1182/blood-2011-08-371971. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dukers DF, Vermeer MH, Jaspars LH, Sander CA, Flaig MJ, Vos W, Willemze R, Meijer CJ. Expression of killer cell inhibitory receptors is restricted to true NK cell lymphomas and a subset of intestinal enteropathy-type T cell lymphomas with a cytotoxic phenotype. J Clin Pathol. 2001;54(3):224–228. doi: 10.1136/jcp.54.3.224. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fingeroth JD, Weis JJ, Tedder TF, Strominger JL, Biro PA, Fearon DT. Epstein-Barr virus receptor of human B lymphocytes is the C3d receptor CR2. Proc Natl Acad Sci. 1984;81(14):4510–4514. doi: 10.1073/pnas.81.14.4510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fox CP, Haigh TA, Taylor GS, Long HM, Lee SP, Shannon-Lowe C, O’connor S, Bollard CM, Iqbal J, Chan WC, Rickinson AB, Bell AI, Rowe M. A novel latent membrane 2 transcript expressed in Epstein-Barr virus-positive NK- and T-cell lymphoproliferative disease encodes a target for cellular immunotherapy. Blood. 2010;116(19):3695–3704. doi: 10.1182/blood-2010-06-292268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Halabi MA, Jaccard A, Moulinas R, Bahri R, Al Mouhammad H, Mammari N, Feuillard J, Ranger-Rogez S. Clonal deleted latent membrane protein 1 variants of Epstein-Barr virus are predominant in European extranodal NK/T lymphomas and disappear during successful treatment. Int J Cancer. 2016;139(4):793–802. doi: 10.1002/ijc.30128. [DOI] [PubMed] [Google Scholar]
- Hanley PJ, Lam S, Shpall EJ, Bollard CM. Expanding cytotoxic T lymphocytes from umbilical cord blood that target cytomegalovirus, Epstein-Barr virus, and adenovirus. J Vis Exp. 2012;(63):e3627. doi: 10.3791/3627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Harabuchi Y, Takahara M, Kishibe K, Moriai S, Nagato T, Ishii H. Nasal natural killer (NK)/T-cell lymphoma: clinical, histological, virological, and genetic features. Int J Clin Oncol. 2009;14(3):181–190. doi: 10.1007/s10147-009-0882-7. [DOI] [PubMed] [Google Scholar]
- Harabuchi Y, Yamanaka N, Kataura A, Imai S, Kinoshita T, Mizuno F, Osato T. Epstein-Barr virus in nasal T-cell lymphomas in patients with lethal midline granuloma. Lancet. 1990;335(8682):128–130. doi: 10.1016/0140-6736(90)90002-m. [DOI] [PubMed] [Google Scholar]
- Heslop HE, Slobod KS, Pule MA, Hale GA, Rousseau A, Smith CA, Bollard CM, Liu H, Wu MF, Rochester RJ, Amrolia PJ, Hurwitz JL, Brenner MK, Rooney CM. Long-term outcome of EBV-specific T-cell infusions to prevent or treat EBV-related lymphoproliferative disease in transplant recipients. Blood. 2010;115(5):925–935. doi: 10.1182/blood-2009-08-239186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ho FC, Srivastava G, Loke SL, Fu KH, Leung BP, Liang R, Choy D. Presence of Epstein-Barr virus DNA in nasal lymphomas of B and ‘T’ cell type. Hematol Oncol. 1990;8(5):271–281. doi: 10.1002/hon.2900080505. [DOI] [PubMed] [Google Scholar]
- Hsieh PP, Tung CL, Chan AB, Liao JB, Wang JS, Tseng HH, Su HH, Chang KC, Chang CC. EBV viral load in tumor tissue is an important prognostic indicator for nasal NK/T-cell lymphoma. Am J Clin Pathol. 2007;128(4):579–584. doi: 10.1309/MN4Y8HLQWKD9NB5E. [DOI] [PubMed] [Google Scholar]
- Huang Y, De Reynies A, De Leval L, Ghazi B, Martin-Garcia N, Travert M, Bosq J, Briere J, Petit B, Thomas E, Coppo P, Marafioti T, Emile JF, Delfau-Larue MH, Schmitt C, Gaulard P. Gene expression profiling identifies emerging oncogenic pathways operating in extranodal NK/T-cell lymphoma, nasal type. Blood. 2010;115(6):1226–1237. doi: 10.1182/blood-2009-05-221275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Iqbal J, Kucuk C, Deleeuw RJ, Srivastava G, Tam W, Geng H, Klinkebiel D, Christman JK, Patel K, Cao K, Shen L, Dybkaer K, Tsui IF, Ali H, Shimizu N, Au WY, Lam WL, Chan WC. Genomic analyses reveal global functional alterations that promote tumor growth and novel tumor suppressor genes in natural killer-cell malignancies. Leukemia. 2009;23(6):1139–1151. doi: 10.1038/leu.2009.3. [DOI] [PubMed] [Google Scholar]
- Iqbal J, Weisenburger DD, Chowdhury A, Tsai MY, Srivastava G, Greiner TC, Kucuk C, Deffenbacher K, Vose J, Smith L, Au WY, Nakamura S, Seto M, Delabie J, Berger F, Loong F, Ko YH, Sng I, Liu X, Loughran TP, et al. Natural killer cell lymphoma shares strikingly similar molecular features with a group of non-hepatosplenic gammadelta T-cell lymphoma and is highly sensitive to a novel aurora kinase A inhibitor in vitro. Leukemia. 2011;25(2):348–358. doi: 10.1038/leu.2010.255. [DOI] [PubMed] [Google Scholar]
- Iqbal J, Wright G, Wang C, Rosenwald A, Gascoyne RD, Weisenburger DD, Greiner TC, Smith L, Guo S, Wilcox RA, Teh BT, Lim ST, Tan SY, Rimsza LM, Jaffe ES, Campo E, Martinez A, Delabie J, Braziel RM, Cook JR, et al. Gene expression signatures delineate biological and prognostic subgroups in peripheral T-cell lymphoma. Blood. 2014;123(19):2915–2923. doi: 10.1182/blood-2013-11-536359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ishihara S, Okada S, Wakiguchi H, Kurashige T, Hirai K, Kawa-Ha K. Clonal lymphoproliferation following chronic active Epstein-Barr virus infection and hypersensitivity to mosquito bites. Am J Hematol. 1997;54(4):276–281. doi: 10.1002/(sici)1096-8652(199704)54:4<276::aid-ajh3>3.0.co;2-s. [DOI] [PubMed] [Google Scholar]
- Ishii H, Takahara M, Nagato T, Kis LL, Nagy N, Kishibe K, Harabuchi Y, Klein E. Monocytes enhance cell proliferation and LMP1 expression of nasal natural killer/T-cell lymphoma cells by cell contact-dependent interaction through membrane-bound IL-15. Int J Cancer. 2012;130(1):48–58. doi: 10.1002/ijc.25969. [DOI] [PubMed] [Google Scholar]
- Ishii Y, Yamanaka N, Ogawa K, Yoshida Y, Takami T, Matsuura A, Isago H, Kataura A, Kikuchi K. Nasal T-cell lymphoma as a type of so-called “lethal midline granuloma”. Cancer. 1982;50(11):2336–2344. doi: 10.1002/1097-0142(19821201)50:11<2336::aid-cncr2820501120>3.0.co;2-c. [DOI] [PubMed] [Google Scholar]
- Ito Y, Kimura H, Maeda Y, Hashimoto C, Ishida F, Izutsu K, Fukushima N, Isobe Y, Takizawa J, Hasegawa Y, Kobayashi H, Okamura S, Kobayashi H, Yamaguchi M, Suzumiya J, Hyo R, Nakamura S, Kawa K, Oshimi K, Suzuki R. Pretreatment EBV-DNA copy number is predictive of response and toxicities to SMILE chemotherapy for extranodal NK/T-cell lymphoma, nasal type. Clin Cancer Res. 2012;18(15):4183–4190. doi: 10.1158/1078-0432.CCR-12-1064. [DOI] [PubMed] [Google Scholar]
- Jaccard A, Gachard N, Marin B, Rogez S, Audrain M, Suarez F, Tilly H, Morschhauser F, Thieblemont C, Ysebaert L, Devidas A, Petit B, De Leval L, Gaulard P, Feuillard J, Bordessoule D, Hermine O. Efficacy of L-asparaginase with methotrexate and dexamethasone (AspaMetDex regimen) in patients with refractory or relapsing extranodal NK/T-cell lymphoma, a phase 2 study. Blood. 2011;117(6):1834–1839. doi: 10.1182/blood-2010-09-307454. [DOI] [PubMed] [Google Scholar]
- Jones JF, Shurin S, Abramowsky C, Tubbs RR, Sciotto CG, Wahl R, Sands J, Gottman D, Katz BZ, Sklar J. T-cell lymphomas containing Epstein-Barr viral DNA in patients with chronic Epstein-Barr virus infections. N Engl J Med. 1988;318(12):733–741. doi: 10.1056/NEJM198803243181203. [DOI] [PubMed] [Google Scholar]
- Kanakry JA, Hegde AM, Durand CM, Massie AB, Greer AE, Ambinder RF, Valsamakis A. The clinical significance of EBV DNA in the plasma and peripheral blood mononuclear cells of patients with or without EBV diseases. Blood. 2016;127(16):2007–2017. doi: 10.1182/blood-2015-09-672030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kanavaros P, Lescs MC, Briere J, Divine M, Galateau F, Joab I, Bosq J, Farcet JP, Reyes F, Gaulard P. Nasal T-cell lymphoma: a clinicopathologic entity associated with peculiar phenotype and with Epstein-Barr virus. Blood. 1993;81(10):2688–2695. [PubMed] [Google Scholar]
- Kasahara Y, Yachie A, Takei K, Kanegane C, Okada K, Ohta K, Seki H, Igarashi N, Maruhashi K, Katayama K, Katoh E, Terao G, Sakiyama Y, Koizumi S. Differential cellular targets of Epstein-Barr virus (EBV) infection between acute EBV-associated hemophagocytic lymphohistiocytosis and chronic active EBV infection. Blood. 2001;98(6):1882–1888. doi: 10.1182/blood.v98.6.1882. [DOI] [PubMed] [Google Scholar]
- Kawa K, Okamura T, Yagi K, Takeuchi M, Nakayama M, Inoue M. Mosquito allergy and Epstein-Barr virus-associated T/natural killer-cell lymphoproliferative disease. Blood. 2001;98(10):3173–3174. doi: 10.1182/blood.v98.10.3173. [DOI] [PubMed] [Google Scholar]
- Kawa-Ha K, Ishihara S, Ninomiya T, Yumura-Yagi K, Hara J, Murayama F, Tawa A, Hirai K. CD3-negative lymphoproliferative disease of granular lymphocytes containing Epstein-Barr viral DNA. J Clin Invest. 1989;84(1):51–55. doi: 10.1172/JCI114168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim JE, Kim YA, Jeon YK, Park SS, Heo DS, Kim CW. Comparative analysis of NK/T-cell lymphoma and peripheral T-cell lymphoma in Korea: Clinicopathological correlations and analysis of EBV strain type and 30-bp deletion variant LMP1. Pathol Int. 2003;53(11):735–743. doi: 10.1046/j.1320-5463.2003.01552.x. [DOI] [PubMed] [Google Scholar]
- Kim WY, Jung HY, Nam SJ, Kim TM, Heo DS, Kim CW, Jeon YK. Expression of programmed cell death ligand 1 (PD-L1) in advanced stage EBV-associated extranodal NK/T cell lymphoma is associated with better prognosis. Virchows Archiv. 2016 doi: 10.1007/s00428-016-2011-0. [DOI] [PubMed] [Google Scholar]
- Kimura H, Hoshino Y, Kanegane H, Tsuge I, Okamura T, Kawa K, Morishima T. Clinical and virologic characteristics of chronic active Epstein-Barr virus infection. Blood. 2001;98(2):280–286. doi: 10.1182/blood.v98.2.280. [DOI] [PubMed] [Google Scholar]
- Ko YH, Choi KE, Han JH, Kim JM, Ree HJ. Comparative genomic hybridization study of nasal-type NK/T-cell lymphoma. Cytometry. 2001;46(2):85–91. doi: 10.1002/cyto.1069. [DOI] [PubMed] [Google Scholar]
- Kritharis A, Coyle M, Sharma J, Evens AM. Lenalidomide in non-Hodgkin lymphoma: biological perspectives and therapeutic opportunities. Blood. 2015;125(16):2471–2476. doi: 10.1182/blood-2014-11-567792. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kuo TT, Shih LY, Tsang NM. Nasal NK/T cell lymphoma in Taiwan: a clinicopathologic study of 22 cases, with analysis of histologic subtypes, Epstein-Barr virus LMP-1 gene association, and treatment modalities. Int J Surg Pathol. 2004;12(4):375–387. doi: 10.1177/106689690401200410. [DOI] [PubMed] [Google Scholar]
- Kurth J, Spieker T, Wustrow J, Strickler GJ, Hansmann LM, Rajewsky K, Kuppers R. EBV-infected B cells in infectious mononucleosis: viral strategies for spreading in the B cell compartment and establishing latency. Immunity. 2000;13(4):485–495. doi: 10.1016/s1074-7613(00)00048-0. [DOI] [PubMed] [Google Scholar]
- Kwong YL, Chan TS, Tan D, Kim SJ, Poon LM, Mow B, Khong PL, Loong F, Au-Yeung R, Iqbal J, Phipps C, Tse E. PD1 blockade with pembrolizumab is highly effective in relapsed or refractory NK/T-cell lymphoma failing L-asparaginase. Blood. 2017 doi: 10.1182/blood-2016-12-756841. [DOI] [PubMed] [Google Scholar]
- Kwong YL, Kim WS, Lim ST, Kim SJ, Tang T, Tse E, Leung AY, Chim CS. SMILE for natural killer/T-cell lymphoma: analysis of safety and efficacy from the Asia Lymphoma Study Group. Blood. 2012;120(15):2973–2980. doi: 10.1182/blood-2012-05-431460. [DOI] [PubMed] [Google Scholar]
- Li Q, Spriggs MK, Kovats S, Turk SM, Comeau MR, Nepom B, Hutt-Fletcher LM. Epstein-Barr virus uses HLA class II as a cofactor for infection of B lymphocytes. J Virol. 1997;71(6):4657–4662. doi: 10.1128/jvi.71.6.4657-4662.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lin TC, Liu TY, Hsu SM, Lin CW. Epstein-Barr virus-encoded miR-BART20-5p inhibits T-bet translation with secondary sup-pression of p53 in invasive nasal NK/T-cell lymphoma. Am J Pathol. 2013;182(5):1865–1875. doi: 10.1016/j.ajpath.2013.01.025. [DOI] [PubMed] [Google Scholar]
- Lustberg ME, Pelletier RP, Porcu P, Martin SI, Quinion CD, Geyer SM, Caligiuri MA, Baiocchi RA. Human leukocyte antigen type and posttransplant lymphoproliferative disorder. Transplantation. 2015;99(6):1220–1225. doi: 10.1097/TP.0000000000000487. [DOI] [PubMed] [Google Scholar]
- Matsuo Y, Drexler HG. Immunoprofiling of cell lines derived from natural killer-cell and natural killer-like T-cell leukemia-lymphoma. Leuk. Res. 2003;27(10):935–945. doi: 10.1016/s0145-2126(03)00024-9. [DOI] [PubMed] [Google Scholar]
- Mcbride P. Photographs of a case of rapid destruction of the nose and face. 1897. J Laryngol Otol. 1991;105(12):1120. doi: 10.1017/s0022215100118407. [DOI] [PubMed] [Google Scholar]
- Minarovits J, Hu LF, Imai S, Harabuchi Y, Kataura A, Minarovits-Kormuta S, Osato T, Klein G. Clonality, expression and methylation patterns of the Epstein-Barr virus genomes in lethal midline granulomas classified as peripheral angiocentric T cell lymphomas. J Gen Virol. 1994;75(Pt 1):77–84. doi: 10.1099/0022-1317-75-1-77. [DOI] [PubMed] [Google Scholar]
- Moriai S, Takahara M, Ogino T, Nagato T, Kishibe K, Ishii H, Katayama A, Shimizu N, Harabuchi Y. Production of interferon-{gamma}-inducible protein-10 and its role as an autocrine invasion factor in nasal natural killer/T-cell lymphoma cells. Clin Cancer Res. 2009;15(22):6771–6779. doi: 10.1158/1078-0432.CCR-09-1052. [DOI] [PubMed] [Google Scholar]
- Nagamine M, Takahara M, Kishibe K, Nagato T, Ishii H, Bandoh N, Ogino T, Harabuchi Y. Sequence variations of Epstein-Barr virus LMP1 gene in nasal NK/T-cell lymphoma. Virus Genes. 2007;34(1):47–54. doi: 10.1007/s11262-006-0008-5. [DOI] [PubMed] [Google Scholar]
- Nagato T, Kobayashi H, Kishibe K, Takahara M, Ogino T, Ishii H, Oikawa K, Aoki N, Sato K, Kimura S, Shimizu N, Tateno M, Harabuchi Y. Expression of interleukin-9 in nasal natural killer/T-cell lymphoma cell lines and patients. Clin Cancer Res. 2005;11(23):8250–8257. doi: 10.1158/1078-0432.CCR-05-1426. [DOI] [PubMed] [Google Scholar]
- Nakashima Y, Tagawa H, Suzuki R, Karnan S, Karube K, Ohshima K, Muta K, Nawata H, Morishima Y, Nakamura S, Seto M. Genome-wide array-based comparative genomic hybridization of natural killer cell lymphoma/leukemia: different genomic alteration patterns of aggressive NK-cell leukemia and extranodal Nk/T-cell lymphoma, nasal type. Genes Chromosomes Cancer. 2005;44(3):247–255. doi: 10.1002/gcc.20245. [DOI] [PubMed] [Google Scholar]
- Nemerow GR, Wolfert R, Mcnaughton ME, Cooper NR. Identification and characterization of the Epstein-Barr virus receptor on human B lymphocytes and its relationship to the C3d complement receptor (CR2) J Virol. 1985;55(2):347–351. doi: 10.1128/jvi.55.2.347-351.1985. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ng SB, Selvarajan V, Huang G, Zhou J, Feldman AL, Law M, Kwong YL, Shimizu N, Kagami Y, Aozasa K, Salto-Tellez M, Chng WJ. Activated oncogenic pathways and therapeutic targets in extranodal nasal-type NK/T cell lymphoma revealed by gene expression profiling. J Pathol. 2011;223(4):496–510. doi: 10.1002/path.2823. [DOI] [PubMed] [Google Scholar]
- Nicolae A, Pittaluga S, Abdullah S, Steinberg SM, Pham TA, Davies-Hill T, Xi L, Raffeld M, Jaffe ES. EBV-positive large B-cell lymphomas in young patients: a nodal lymphoma with evidence for a tolerogenic immune environment. Blood. 2015;126(7):863–872. doi: 10.1182/blood-2015-02-630632. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ohshima K, Suzumiya J, Sugihara M, Nagafuchi S, Ohga S, Kikuchi M. Clinicopathological study of severe chronic active Epstein-Barr virus infection that developed in association with lymphoproliferative disorder and/or hemophagocytic syndrome. Pathol Int. 1998;48(12):934–943. doi: 10.1111/j.1440-1827.1998.tb03864.x. [DOI] [PubMed] [Google Scholar]
- Ok CY, Ye Q, Li L, Manyam GC, Deng L, Goswami RR, Wang X, Montes-Moreno S, Visco C, Tzankov A, Dybkaer K, Zhang L, Abramson J, Sohani AR, Chiu A, Orazi A, Zu Y, Bhagat G, Richards KL, Hsi ED, et al. Age cutoff for Epstein-Barr virus-positive diffuse large B-cell lymphoma-is it necessary? Oncotarget. 2015;6(16):13933–13945. doi: 10.18632/oncotarget.4324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Palser AL, Grayson NE, White RE, Corton C, Correia S, Ba Abdullah MM, Watson SJ, Cotten M, Arrand JR, Murray PG, Allday MJ, Rickinson AB, Young LS, Farrell PJ, Kellam P. Genome diversity of Epstein-Barr virus from multiple tumor types and normal infection. J Virol. 2015;89(10):5222–5237. doi: 10.1128/JVI.03614-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pongpruttipan T, Sukpanichnant S, Assanasen T, Wannakrairot P, Boonsakan P, Kanoksil W, Kayasut K, Mitarnun W, Khuhapinant A, Bunworasate U, Puavilai T, Bedavanija A, Garcia-Herrera A, Campo E, Cook JR, Choi J, Swerdlow SH. Extranodal NK/T-cell lymphoma, nasal type, includes cases of natural killer cell and alphabeta, gammadelta, and alphabeta/gammadelta T-cell origin: a comprehensive clinicopathologic and phenotypic study. Am J Surg Pathol. 2012;36(4):481–499. doi: 10.1097/PAS.0b013e31824433d8. [DOI] [PubMed] [Google Scholar]
- Quintanilla-Martinez L, Ridaura C, Nagl F, Saez-De-Ocariz M, Duran-Mckinster C, Ruiz-Maldonado R, Alderete G, Grube P, Lome-Maldonado C, Bonzheim I, Fend F. Hydroa vacciniforme-like lymphoma: a chronic EBV+ lymphoproliferative disorder with risk to develop a systemic lymphoma. Blood. 2013;122(18):3101–3110. doi: 10.1182/blood-2013-05-502203. [DOI] [PubMed] [Google Scholar]
- Reshef R, Luskin MR, Kamoun M, Vardhanabhuti S, Tomaszewski JE, Stadtmauer EA, Porter DL, Heitjan DF, Tsai De E. Association of HLA polymorphisms with post-transplant lymphoproliferative disorder in solid-organ transplant recipients. Am J Tranplant. 2011;11(4):817–825. doi: 10.1111/j.1600-6143.2011.03454.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rickinson AB. Co-infections, inflammation and oncogenesis: future directions for EBV research. Sem Cancer Biol. 2014;26:99–115. doi: 10.1016/j.semcancer.2014.04.004. [DOI] [PubMed] [Google Scholar]
- Rowe M, Young LS, Cadwallader K, Petti L, Kieff E, Rickinson AB. Distinction between Epstein-Barr virus type A (EBNA 2A) and type B (EBNA 2B) isolates extends to the EBNA 3 family of nuclear proteins. J Virol. 1989;63(3):1031–1039. doi: 10.1128/jvi.63.3.1031-1039.1989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sabattini E, Pizzi M, Tabanelli V, Baldin P, Sacchetti CS, Agostinelli C, Zinzani PL, Pileri SA. CD30 expression in peripheral T-cell lymphomas. Haematologica. 2013;98(8):e81–82. doi: 10.3324/haematol.2013.084913. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Siemer D, Kurth J, Lang S, Lehnerdt G, Stanelle J, Kuppers R. EBV transformation overrides gene expression patterns of B cell differentiation stages. Mol Immunol. 2008;45(11):3133–3141. doi: 10.1016/j.molimm.2008.03.002. [DOI] [PubMed] [Google Scholar]
- Siu LL, Wong KF, Chan JK, Kwong YL. Comparative genomic hybridization analysis of natural killer cell lymphoma/leukemia. Recognition of consistent patterns of genetic alterations. Am J Pathol. 1999;155(5):1419–1425. doi: 10.1016/S0002-9440(10)65454-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sun HS, Su IJ, Lin YC, Chen JS, Fang SY. A 2.6 Mb interval on chromosome 6q25.2-q25.3 is commonly deleted in human nasal natural killer/T-cell lymphoma. Br J Haematol. 2003;122(4):590–599. doi: 10.1046/j.1365-2141.2003.04419.x. [DOI] [PubMed] [Google Scholar]
- Suzuki R. Pathogenesis and treatment of extranodal natural killer/T-cell lymphoma. Sem Hematol. 2014;51(1):42–51. doi: 10.1053/j.seminhematol.2013.11.007. [DOI] [PubMed] [Google Scholar]
- Suzuki R, Yamaguchi M, Izutsu K, Yamamoto G, Takada K, Harabuchi Y, Isobe Y, Gomyo H, Koike T, Okamoto M, Hyo R, Suzumiya J, Nakamura S, Kawa K, Oshimi K, Group NK-CTS. Prospective measurement of Epstein-Barr virus-DNA in plasma and peripheral blood mononuclear cells of extranodal NK/T-cell lymphoma, nasal type. Blood. 2011;118(23):6018–6022. doi: 10.1182/blood-2011-05-354142. [DOI] [PubMed] [Google Scholar]
- Suzumiya J, Ohshima K, Takeshita M, Kanda M, Kawasaki C, Kimura N, Tamura K, Kikuchi M. Nasal lymphomas in Japan: a high prevalence of Epstein-Barr virus type A and deletion within the latent membrane protein gene. Leuk. Lymphoma. 1999;35(5–6):567–578. doi: 10.1080/10428199909169621. [DOI] [PubMed] [Google Scholar]
- Suzumiya J, Takeshita M, Kimura N, Kikuchi M, Uchida T, Hisano S, Eura Y, Kozuru M, Nomura Y, Tomita K, et al. Expression of adult and fetal natural killer cell markers in sinonasal lymphomas. Blood. 1994;83(8):2255–2260. [PubMed] [Google Scholar]
- Swerdlow SH, Campo E, Pileri SA, Harris NL, Stein H, Siebert R, Advani R, Ghielmini M, Salles GA, Zelenetz AD, Jaffe ES. The 2016 revision of the World Health Organization classification of lymphoid neoplasms. Blood. 2016;127(20):2375–2390. doi: 10.1182/blood-2016-01-643569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tabiasco J, Vercellone A, Meggetto F, Hudrisier D, Brousset P, Fournie JJ. Acquisition of viral receptor by NK cells through immunological synapse. J Immunol (Baltimore, MD) 2003;170(12):5993–5998. doi: 10.4049/jimmunol.170.12.5993. [DOI] [PubMed] [Google Scholar]
- Taborelli M, Tibiletti MG, Martin V, Pozzi B, Bertoni F, Capella C. Chromosome band 6q deletion pattern in malignant lymphomas. Cancer Genet Cytogenet. 2006;165(2):106–113. doi: 10.1016/j.cancergencyto.2005.06.025. [DOI] [PubMed] [Google Scholar]
- Tai YC, Kim LH, Peh SC. High frequency of EBV association and 30-bp deletion in the LMP-1 gene in CD56 lymphomas of the upper aerodigestive tract. Pathol Int. 2004;54(3):158–166. doi: 10.1111/j.1440-1827.2003.01602.x. [DOI] [PubMed] [Google Scholar]
- Takahara M, Kis LL, Nagy N, Liu A, Harabuchi Y, Klein G, Klein E. Concomitant increase of LMP1 and CD25 (IL-2-receptor alpha) expression induced by IL-10 in the EBV-positive NK lines SNK6 and KAI3. Int J Cancer. 2006;119(12):2775–2783. doi: 10.1002/ijc.22139. [DOI] [PubMed] [Google Scholar]
- Takahashi E, Ohshima K, Kimura H, Hara K, Suzuki R, Kawa K, Eimoto T, Nakamura S. Clinicopathological analysis of the age-related differences in patients with Epstein-Barr virus (EBV)-associated extranasal natural killer (NK)/T-cell lymphoma with reference to the relationship with aggressive NK cell leukaemia and chronic active EBV infection-associated lymphoproliferative disorders. Histopathology. 2011;59(4):660–671. doi: 10.1111/j.1365-2559.2011.03976.x. [DOI] [PubMed] [Google Scholar]
- Tao Q, Ho FC, Loke SL, Srivastava G. Epstein-Barr virus is localized in the tumour cells of nasal lymphomas of NK, T or B cell type. Int J Cancer. 1995;60(3):315–320. doi: 10.1002/ijc.2910600306. [DOI] [PubMed] [Google Scholar]
- Toumishey E, Prasad A, Dueck G, Chua N, Finch D, Johnston J, Van Der Jagt R, Stewart D, White D, Belch A, Reiman T. Final report of a phase 2 clinical trial of lenalidomide monotherapy for patients with T-cell lymphoma. Cancer. 2015;121(5):716–723. doi: 10.1002/cncr.29103. [DOI] [PubMed] [Google Scholar]
- Van Gorp J, Brink A, Oudejans JJ, Van Den Brule AJ, Van Den Tweel JG, Jiwa NM, De Bruin PC, Meijer CJ. Expression of Epstein-Barr virus encoded latent genes in nasal T cell lymphomas. J Clin Pathol. 1996;49(1):72–76. doi: 10.1136/jcp.49.1.72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang B, Li XQ, Ma X, Hong X, Lu H, Guo Y. Immunohistochemical expression and clinical significance of P-glycoprotein in previously untreated extranodal NK/T-cell lymphoma, nasal type. Am J Hematol. 2008;83(10):795–799. doi: 10.1002/ajh.21256. [DOI] [PubMed] [Google Scholar]
- Wang ZY, Liu QF, Wang H, Jin J, Wang WH, Wang SL, Song YW, Liu YP, Fang H, Ren H, Wu RY, Chen B, Zhang XM, Lu NN, Zhou LQ, Li YX. Clinical implications of plasma Epstein-Barr virus DNA in early-stage extranodal nasal-type NK/T-cell lymphoma patients receiving primary radiotherapy. Blood. 2012;120(10):2003–2010. doi: 10.1182/blood-2012-06-435024. [DOI] [PubMed] [Google Scholar]
- Wong KF, Chan JK, Ng CS, Lee KC, Tsang WY, Cheung MM. CD56 (NKH1)-positive hematolymphoid malignancies: an aggressive neoplasm featuring frequent cutaneous/mucosal involvement, cytoplasmic azurophilic granules, and angiocentricity. Hum Pathol. 1992;23(7):798–804. doi: 10.1016/0046-8177(92)90350-c. [DOI] [PubMed] [Google Scholar]
- Woods R. Observations on malignant granuloma of the nose. Br Med J. 1921;2(3159):65–66.61. doi: 10.1136/bmj.2.3159.65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu ZG, Iwatsuki K, Oyama N, Ohtsuka M, Satoh M, Kikuchi S, Akiba H, Kaneko F. The latency pattern of Epstein-Barr virus infection and viral IL-10 expression in cutaneous natural killer/T-cell lymphomas. Br J Cancer. 2001;84(7):920–925. doi: 10.1054/bjoc.2000.1687. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yamaguchi M, Kwong YL, Kim WS, Maeda Y, Hashimoto C, Suh C, Izutsu K, Ishida F, Isobe Y, Sueoka E, Suzumiya J, Kodama T, Kimura H, Hyo R, Nakamura S, Oshimi K, Suzuki R. Phase II study of SMILE chemotherapy for newly diagnosed stage IV, relapsed, or refractory extranodal natural killer (NK)/T-cell lymphoma, nasal type: the NK-Cell Tumor Study Group study. J Clin Oncol. 2011;29(33):4410–4416. doi: 10.1200/JCO.2011.35.6287. [DOI] [PubMed] [Google Scholar]
- Yang L, Aozasa K, Oshimi K, Takada K. Epstein-Barr virus (EBV)-encoded RNA promotes growth of EBV-infected T cells through interleukin-9 induction. Cancer Res. 2004;64(15):5332–5337. doi: 10.1158/0008-5472.CAN-04-0733. [DOI] [PubMed] [Google Scholar]
- Yoshino K, Kishibe K, Nagato T, Ueda S, Komabayashi Y, Takahara M, Harabuchi Y. Expression of CD70 in nasal natural killer/T cell lymphoma cell lines and patients; its role for cell proliferation through binding to soluble CD27. Br J Haematol. 2013;160(3):331–342. doi: 10.1111/bjh.12136. [DOI] [PubMed] [Google Scholar]
- Younes A, Bartlett NL, Leonard JP, Kennedy DA, Lynch CM, Sievers EL, Forero-Torres A. Brentuximab vedotin (SGN-35) for relapsed CD30-positive lymphomas. N Engl J Med. 2010;363(19):1812–1821. doi: 10.1056/NEJMoa1002965. [DOI] [PubMed] [Google Scholar]
- Young LS, Rickinson AB. Epstein-Barr virus: 40 years on. Nat Rev Cancer. 2004;4(10):757–768. doi: 10.1038/nrc1452. [DOI] [PubMed] [Google Scholar]
- Zhang Y, Ohyashiki JH, Takaku T, Shimizu N, Ohyashiki K. Transcriptional profiling of Epstein-Barr virus (EBV) genes and host cellular genes in nasal NK/T-cell lymphoma and chronic active EBV infection. Br J Cancer. 2006;94(4):599–608. doi: 10.1038/sj.bjc.6602968. [DOI] [PMC free article] [PubMed] [Google Scholar]