Next Article in Journal
Recurrence of Upper Extremity Deep Vein Thrombosis Secondary to COVID-19
Next Article in Special Issue
The Role of Coinfections in the EBV–Host Broken Equilibrium
Previous Article in Journal
The Role of Non-Structural Protein NSs in the Pathogenesis of Severe Fever with Thrombocytopenia Syndrome
Previous Article in Special Issue
Inflammasome, the Constitutive Heterochromatin Machinery, and Replication of an Oncogenic Herpesvirus
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pathogenic Role of Epstein–Barr Virus in Lung Cancers

1
Department of Medicine, Tulane University Health Sciences Center, New Orleans, LA 70112, USA
2
National Research Center for the Control and Prevention of Infectious Diseases, Nagasaki University, Sakamoto, Nagasaki 852-8523, Japan
3
Department of Pathology and Laboratory Medicine, Tulane University Health Sciences Center and Tulane Cancer Center, New Orleans, LA 70112, USA
4
Institute of Translational Research, Ochsner Clinic Foundation, New Orleans, LA 70121, USA
*
Author to whom correspondence should be addressed.
These authors have contributed equally to this work.
Viruses 2021, 13(5), 877; https://doi.org/10.3390/v13050877
Submission received: 30 March 2021 / Revised: 27 April 2021 / Accepted: 7 May 2021 / Published: 11 May 2021
(This article belongs to the Special Issue Epstein Barr Virus)

Abstract

:
Human oncogenic viruses account for at least 12% of total cancer cases worldwide. Epstein–Barr virus (EBV) is the first identified human oncogenic virus and it alone causes ~200,000 cancer cases and ~1.8% of total cancer-related death annually. Over the past 40 years, increasing lines of evidence have supported a causal link between EBV infection and a subgroup of lung cancers (LCs). In this article, we review the current understanding of the EBV-LC association and the etiological role of EBV in lung carcinogenesis. We also discuss the clinical impact of the knowledge gained from previous research, challenges, and future directions in this field. Given the high clinical relevance of EBV-LC association, there is an urgent need for further investigation on this topic.

1. Lung Cancers

Lung cancers (LCs) are the number one killer among cancers in the U.S. and estimated to cause more than 131,000 deaths in 2021 [1]. With more than 200,000 annual cases in the U.S., LCs have remained as the second most common cancers in both men and women for many years [1]. Based on the histological features, LCs can be classified as small cell lung cancers (SCLCs) and non-small cell lung cancers (NSCLCs) according to the WHO 2015 guidelines [2]. NSCLCs account for ~85% of total LC cases and have two major histological sub-types (adenocarcinoma (LUAD) and squamous cell carcinoma (LUSC)) as well as several other rarer sub-types, including pulmonary lymphoepithelioma-like carcinoma (LELC) [2,3].
LC is a complex and multifactorial malignant disease with a currently unclear etiology. It is believed that genetic, epigenetic, and environmental factors all can contribute to lung cancer development. Among those known risk factors, smoking is a definitive oncogenic factor for LC development. Nevertheless, the incidence of LCs is only slowly declining even after the dramatic reduction of smoking through public health awareness campaigns. To date, only a small portion (10–20%) of smokers develop LCs [4]. With a lower smoking rate seen among females, it is estimated that at least 50% of female LC patients worldwide are never smokers. In addition, LCs are the seventh most common cause of cancer death in never smokers [5,6]. Together, this evidence indicates that other major etiological factors are responsible for LC development. The concept of infectious agents as a potential oncogenic trigger for the LC development has long been proposed. However, a causal link between most of microbial infections and LCs has not yet been established.

2. Epstein–Barr Virus

Epstein–Barr virus (EBV) is also known as human herpesvirus 4 and belongs to the gamma-herpesvirus subfamily, in the same family with Kaposi’s sarcoma-associated herpesvirus (KSHV). EBV is the first human oncogenic virus discovered in 1964, which currently accounts for ~200,000 annual cancer cases and ~1.8% of all cancer-related deaths globally [7,8,9,10,11]. EBV is etiologically associated with a number of human lymphoid and epithelial malignancies including Burkitt’s lymphoma (BL), Hodgkin’s disease (HD), extranodal nasal-type natural killer/T cell lymphoma (NKTL), post-transplant lymphoproliferative disease (PTLD) and lymphomas (e.g., diffuse large B-cell lymphoma) in immunocompromised individuals, nasopharyngeal carcinoma (NPC), gastric carcinoma (GC), as well as a subset of lung cancers (LCs) [7,8,9,12,13,14,15].
Like other herpesviruses, EBV employs two distinct stages to complete its life cycle: latency and lytic cycle (or lytic reactivation). After an initial, usually asymptomatic infection, EBV will establish a life-long persistent infection in the host. During latency, the EBV genomes remain as episomes and replicate in the S-phase. The replicated episomes are then partitioned into the daughter cells during cell division. In order to avoid host immune surveillance, only a limited repertoire of viral genes are expressed. Notably, abundant viral non-coding RNAs with low antigenicity are also synthesized to facilitate the viral replication. In response to certain physiological or pathological stimuli, the latent genome can be reactivated, characterized by a series of highly ordered events. The viral immediate-early gene products, such as Zta and Rta, will function as a molecular switch to turn on the entire lytic cascade and induce the expression of ~100 viral early and late genes. Ultimately, this leads to the production of new infectious virions [16,17].
The infectious virions can be transmitted through body fluids such as blood and urine, but they are mainly spread through saliva exchange. Like other human herpesviruses, except varicella-zoster virus (VZV), vaccine-based blockade of EBV infection is still in the development stage and not yet an option. It is estimated that ~90% of the global adult population are currently carrying EBV [8]. To date, the detailed mechanism of EBV replication in vivo is still largely unclear. Based on available limited evidence, different views of these replicative events have emerged. One view is that EBV may first enter the replication-permissive epithelial cells in the oral cavity where an active lytic replication occurs. The propagated infectious virions are then released from the initial epithelial cells to infect the nearby infiltrating B cells in which the full spectra of viral latency transcripts are synthesized (also known as type III latency) (Table 1). An alternative view is that EBV transmigrates across polarized human oral epithelial cells by apical to basolateral transcytosis without causing lytic replication and subsequently infects B cells [18]. The type III latency can activate B cells and lead to a transient expansion of the EBV-infected B cell pool. Most of infected B cells will be annihilated by the host immune surveillance. Only a subset of infected B cells can persist for the lifetime of the host. This is attributable to their expression of limited number of viral antigens. In these viral reservoir cells (i.e., memory B cells) either no (type 0 latency) or only one viral protein, EBV-encoded nuclear antigen (EBNA) 1 (type I latency) is expressed (Table 1). Furthermore, to permanently live with the hosts, new infectious virions need to be continuously produced to replenish the viral reservoir. It is believed that sporadic reactivation occurs in epithelial tissues as well as B cells differentiating into plasma cells [19,20], and the progeny virions are released to infect new host cells.
Accumulating evidence has shown that both viral latency and lytic cycle are required for EBV pathogenesis. There are approximately 100 open reading frames encoded by the EBV genome. Among them, some latent genes such as EBV-encoded nuclear antigen 1 (EBNA1) [21], EBV-encoded nuclear antigen 2 (EBNA2), EBV-encoded nuclear antigen 3C (EBNA3C), and latent membrane protein 1 (LMP1) have been shown to mediate viral oncogenesis in cell and/or animal models. These viral oncogene products can activate various tumor-associated pathways such as Notch and nuclear factor-kB (NF-kB) signalings. In addition to well-characterized viral protein-coding genes, EBV has been shown to utilize viral non-coding RNAs (ncRNAs) such as microRNAs (miRNAs), long non-coding RNAs (lncRNAs), small non-coding EBV-encoded RNAs (EBERs), as well as recently identified circular RNA (circRNA) to facilitate its life cycle and oncogenesis [22,23,24,25,26,27,28,29,30].

3. Presence of EBV in Lung Cancers

The human lung is not a sterile anatomic site and it is regularly in contact with the external environment [31]. A diversity of microorganisms including bacteria, yeasts, and viruses can be harbored in either healthy or diseased lung [31]. Some of those infectious microorganisms have long been speculated to contribute to lung carcinogenesis [32,33].
A possible association of EBV with LCs was first supported by the finding that some LC patients had a significantly higher level of anti-EBV IgA in the sera compared to the healthy control individuals [34]. Later on, the presence of EBV in the bronchoalveolar fluid from LC patients further suggests that the lung tissue may act as a major EBV reservoir [35]. In 1987, Begin and colleagues reported the first EBV-positive LC case [36]. In this case, a 40-year-old Asian female nonsmoker developed an uncommon type of nonkeratinizing lung squamous cell carcinoma. Histologically, this poorly differentiated epithelial tumor mimics the lymphoepithelioma-like carcinoma (LELC). In the following years, an increasing number of primary EBV-positive LELCs have been detected in the lung. Now, these subtypes of lung cancers are diagnosed as “non-small cell carcinoma, not otherwise specified” under the NSCLC group according to the 2015 World Health Organization classification [2]. These EBV-positive LELCs are characterized by poorly or undifferentiated squamoid or glandular carcinoma with intensive tumor infiltrating immune cells and preferentially occur in Asian patients [37,38,39,40].
In addition to the uncommon LELC subtype, emerging evidence has shown that EBV is also present in the tumor cells of conventional NSCLC subtypes including lung squamous-cell carcinomas (LUSC) and lung adenocarcinomas (LUAD) [41,42,43,44,45,46,47]. In addition to NSCLC, Chu and colleagues also detected EBV gene products such as EBNA1 and LMP1 in SCLC patients in the United States. However, it is unclear if the detected EBV products are from tumor-infiltrating EBV-positive (EBV(+)) B cells or the LC cells [48].
Here, we summarized the studies reporting the status and potential roles of EBV in lung carcinogenesis. Studies were identified by searching the PubMed database and only articles (including abstracts) published in English were reviewed. The latest literature search was performed in March 2021. Having carefully evaluated the extracted studies, we selected 42 relevant studies and the EBV status in LCs was summarized in Table 2 [12,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76].
Having reviewed the data, we found that the association of EBV and LCs shows significant differences based on tumor histology types and geographical sites where the studies were carried out. The reported incidence rates vary from 0 to 100% (Table 2). EBV positivity was more frequently observed in Asian LC patients with the LELC subtype than patients in other racial groups or having other LC subtypes.
The variation of incidence appears also affected by the sample sizes and methodologies used for EBV infection. Notably, the majority of the studies used traditional viral screening methods such as polymerase chain reaction (PCR), in situ hybridization (ISH), and immunohistochemistry (IHC). Due to the inherent limitations of these traditional screening methods (such as PCR priming issues, usage of inappropriate/biased detection markers, inconsistent thresholds for positivity, etc.), the accuracy of the incidence data was questionable. We reasoned that an ideal detection approach should cover the full sequence of LC cells and search for any present EBV genetic materials.
To date, the next-generation sequencing (NGS) technology has been successfully utilized to discover and interrogate various oncogenic pathogens. NGS uses an unbiased method to globally assess all the exogenous microorganisms within a tumor sample with high sensitivity and specificity. Several research teams, including ours, have successfully utilized NGS (e.g., RNA-seq) to interrogate exogenous pathogens associated with human cancers [12,14,15,77,78,79,80,81,82,83,84,85]. Furthermore, NGS technology has enabled us to not only discover new oncogenic pathogens, but also identify previous false discoveries.
In addition to the traditional methods, we utilized our unbiased RNA-seq based informatics approach to comprehensively interrogate the involvement of EBV in LCs in our recent work [12]. RNA-seq datasets of 1127 LC samples were analyzed by our NGS pipeline. Samples were mainly collected from LC patients in western countries. To our knowledge, this is the largest scale of screening work for EBV infection in LCs to date. We reasoned that the sample size should be sufficient for us to draw a definitive conclusion of the involvement of EBV in LCs. Our NGS analysis showed that four LC cases exhibit transcriptional active EBV infection. In addition to NGS, we also conducted ISH to examine the expression of EBV transcripts in LC cells. Strong EBV-encoded RNA (EBER) signals were observed in three out of 110 analyzed LC tissues. Notably, the EBER signals were detected in LC cells but not in the tumor-infiltrating immune cells.
The low EBV infection rate (~0.6%) observed in our study indicates that EBV is unlikely to play a significant role in the development of LCs in the western countries, but it may contribute to the development of a subset of LC cases [12]. In areas where EBV-associated cancers are endemic, such as Southeast Asia and sub-Saharan African, the link between EBV and LCs may be more prevalent. Another rationale for the observed low EBV incidence rate is that EBV may use the hit-and-run strategy to infect lung epithelial cells, which subsequently promotes the LC development [86]. Thus, the transient presence of EBV genomes may cause genetic scars in the infected cells, leading to a permanent alteration of host gene expression and lung carcinogenesis. In accordance, Hu et al. recently provided evidence showing that EBV may use a similar hit-and-run mechanism to promote breast cancer development [87].

4. Etiological Role of EBV in LC

4.1. EBV Transcriptome

EBV exhibits various latency programs in the infected lymphoid and epithelial cells (Table 1). The type of latency program can reflect how EBV interacts with its host cells and facilitates carcinogenesis. In our recent study, we also tried to elucidate EBV latency program in LCs by conducting the first comprehensive transcriptome analysis of the EBV(+) LCs [12]. Our sequencing study detected multiple viral latency products including abundant EBNA1, LMP1, LMP2A, LMP2B, as well as BamHI A rightward transcripts (BART) in EBV(+) LC tissues, which resembles a type II-like viral latency program [12].
The high level of BART is consistent with a true EBV latency since BART is more highly expressed in the infected epithelial cells than in B cells. Although previous studies have been unable to detect protein from endogenous BART (e.g., RPMS1 and A73) [88,89], the robust expression of these transcripts indicates a functional role in LCs, presumably as lncRNAs, which has been previously proposed in the EBV(+) GC [90]. Since many known lncRNAs function in molecular complexes that inhibit transcription, it is likely that BART can selectively repress cellular gene expression in EBV(+) LCs. BART also encodes at least 44 viral intronic microRNAs (miRNAs). The pathogenic roles of these BART miRNAs in the EBV’s life cycle and in EBV-associated cancers have also been characterized [91]. The high level of BART in LCs would facilitate a significant role in modulating the cellular phenotype by BART miRNAs in LCs. Furthermore, we also observed new transcript isoforms from two novel regions within the BamHI A loci (i.e., the region between exons 4 and 5, as well as between exons 6 and 7), which are likely initiated by a hidden promoter [12]. These new transcripts may similarly play a role in non-coding RNA-mediated modulation of cellular function.
Several novel alternative splicing events of LMP2A have been detected in various EBV-associated cancers [27,92,93]. In EBV(+) LCs, the classical splicing event between the first exon (exon 1A) and exon 2 of LMP2A was not detected [12]. Instead, a novel splicing event between splicing sites located within LMP2A exon 2 and RPMS1 exon 7 was detected [12]. Thus, together with all these findings, we speculated that the alternative splicing of LMP2A may be more common than we previously expected and it may play important roles in EBV life cycle and pathogenesis.

4.2. EBV-Associated Immunoevasion

In some EBV-associated cancers such as NPC and GC, EBV is known to alter the tumor immune microenvironment to facilitate the carcinogenesis [15,81]. In accordance, in the context of LCs, we also observed an increased immune cell infiltration in EBV(+) LCs [12]. Despite this heightened influx of immune cells, EBV(+) LC cells persist in the patients. It suggests that EBV(+) LCs may have successfully employed certain immunoevasion strategies to ensure virus/tumor survival.
Indeed, elevated levels of multiple immune checkpoint molecules, such as indoleamine 2,4-dioxygenase (IDO), programmed cell death 1 (PD-1), cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), lymphocyte-activation gene 3 (LAG3), B and T lymphocyte attenuator (BTLA), and v-domain Ig suppressor of T cell activation (VISTA) were also detected in EBV(+) LCs [12]. Further, several studies have reported that high levels of programmed death-ligand 1 (PD-L1) were observed in EBV(+) LCs [64,66]. These immune inhibitors may contribute to EBV-associated tumor immune tolerance. For example, IDO is one of the top EBV-induced immune inhibitors. IDO may inhibit the activities of cytotoxic T lymphocytes and NK cells by causing local tryptophan depletion in the tumor niche and thus enhance tumor survival [15,94,95,96].
Along this line, we also detected a high level of BNLF2a gene expression in the absence of significant expression of other viral lytic genes in the EBV(+) LCs [12]. BNLF2a is classified as an early lytic phase protein and can suppress immune detection of the EBV(+) cells by blocking viral antigen presentation to the major histocompatibility complex (MHC) class I molecules [81,97,98,99,100,101,102]. Interestingly, expression of BNLF2a has been reported in a good portion of EBV(+) GC tissues and cell lines [15,81]. Thus, it is possible that EBV infection may similarly promote LC immunoevasion by expressing BNLF2a.

4.3. EBV-Associated Alteration of Tumor Pathways

Unlike other EBV-associated cancers such as NPC and GC, the EBV-mediated alteration of cellular signaling pathways in lungs is poorly characterized. Having examined the cellular transcriptome of EBV(+) LCs, we found the activation of several EBV-associated oncogenic pathways and inhibition of multiple tumor suppressors [12]. It includes the alteration of G2/M and G1/S cell cycle control, the p53, HIPPO, and Sirtuin signaling pathways [12]. EBV might play a direct regulatory role in these tumor pathways. For example, the activation of breast cancer 1 (BRCA1) signaling may be triggered by EBV infection, since BRCA1 is an important signaling molecule in the innate sensing of herpesvirus DNA and EBV replication [103,104]. The activation of tumor necrosis factor receptor (TNFR) signaling in EBV(+) LCs is likely due to the expression of viral oncogene LMP1 which acts as a constitutively activated truncated form of the TNFR. Further, the activation of cyclin-dependent kinase 5 (CDK5) signaling is likely due to the expression of viral oncogene EBNA2 which enhances the expression of p35, a CDK5 activator.

4.4. EBV-Associated Genomic Changes

So far, the mutational landscape has only been explored in the LELC subtype of EBV(+) LCs. Emerging evidence has shown that major driver mutations for EBV-negative (EBV(−)) LCs such as epidermal growth factor receptor (EGFR), Kirsten rat sarcoma (KRAS), mesenchymal epithelial transition factor (MET), anaplastic lymphoma kinase (ALK), and c-ros oncogene 1 (ROS1) are rarely observed in the EBV(+) LCs [62,105,106,107,108]. The observation is consistent with other EBV(+) cancers such as NPC and GC which also exhibit distinct mutation profiles compared to the uninfected tumors. It is speculated that the viral oncogenes may only compensate the oncogenic effects of certain cancer driver mutations, but additional mutations are still required for cancer formation.
Recently, Hong and colleagues utilized whole exome sequencing, targeted deep sequencing and single-nucleotide polymorphism array to survey the genomic landscapes of 91 EBV(+) LC samples [68]. They found that EBV(+) LCs show distinct genomic features from EBV(−) LCs, NK/T-cell lymphoma and EBV(+) GC but share similarities with EBV(+) NPC. Furthermore, EBV(+) LCs exhibit a low degree of somatic mutation but widespread copy number variations. A widespread signature two mutations were observed in EBV(+) LCs. This is likely caused by the overactivity of the AID/APOBEC family of cytidine deaminases. The APOBEC family proteins can serve as endogenous mutagens for oncogenesis. Genetic lesions were enriched in several oncogenic pathways including NF-kB and JAK/STAT. The frequently dysregulated NF-kB signaling could be hijacked by EBV to enhance cellular survival and facilitate viral persistence. Ubiquitous losses of type I interferon (IFN) genes were also seen in EBV(+) LCs, which likely impairs the production of anti-viral cytokine and IFN-dependent JAK/STAT activation. Together, the APOBEC family gene signature, deregulated NF-kB signaling, and loss of type I IFN genes may contribute to the EBV-induced lung carcinogenesis.
Meanwhile, Chau et al. conducted a whole genome sequencing analysis of 57 EBV(+) LCs with LELC subtypes [64]. They found that EBV(+) LCs showed a unique mutation profile with low incidence of p53 mutation as well as other major cancer driver mutations in RTK/RAS/RAF and PI3K/AKT/mTOR pathways but enriched for loss-of-function mutations in several inhibitors of NF-kB pathways. Overall, the mutation pattern is different from EBV(−) LCs, but rather resembles that of EBV(+) NPC.
In another study, Chen and colleagues did whole genome sequencing analyses of eight EBV(+) LC samples with LELC subtype [66]. The somatic mutation profiles were compared to the ones of 50 EBV(−) LUAD, 50 EBV(−) LUSC, and 26 EBV(+) NPC samples. They found that EBV(+) LCs showed a total of 14 frequently mutated genes, which is much lower than those in other cancer samples analyzed. A decreased number of gene copies was also widely observed in EBV(+) LCs, including tumor-associated genes such as zinc finger and BTB domain-containing 16 (ZBTB16), peroxisome proliferator activated receptor gamma (PPARG), and transforming growth factor beta receptor 2 (TGFBR2). It is speculated that the copy number loss may be particularly important for EBV-associated lung carcinogenesis.

4.5. Proposed Disease Model of EBV-Associated LCs

In general, tumor viruses are necessary, but not sufficient for their associated cancer development. In accordance with this information, we found that primary lung epithelial cells cannot be immortalized by EBV infection alone (Lin unpublished data). Thus, in the context of EBV-associated LCs, we postulated that EBV infection only contributes to a portion of the oncogenic events. Additional genetic and epigenetic mutations and co-factors such as chronic inflammation, immunosuppression, and environmental mutagens are also required for this multistep oncogenic process. We now favor a disease model in which the impact of EBV infection on the lung carcinogenesis might be a consequence of the aberrant establishment of viral latency in lung epithelial cells that have already undergone premalignant changes (Figure 1). In our proposed disease model, some pre-existing genetic alterations induced by pro-tumor signals (e.g., chronic inflammation) in precursor dysplastic lesions are important to support EBV infection and maintain Type II latency in the lung epithelia. Persistent EBV infection induces the expression of cellular and viral genes in the milieu of infected lesions, which subsequently activates a number of cancer hallmarks [109,110] and leads to lung carcinogenesis (Figure 1). To further elucidate the etiological role of EBV, future work is warranted to screen pre-malignant lung tissues for EBV infection. The observation of monoclonal proliferation of lung epithelial cells carrying latently infected EBV will strongly indicate the presence of EBV in the early stage of lung carcinogenesis.

5. Experimental Models for EBV(+) LC

Understanding of the EBV-lung epithelial cell interaction depends on appropriate in vitro and in vivo experimental models. A lung cancer cell line carrying natively infected EBV should be an ideal in vitro system to explore the complex interplay between EBV and the host cells. To our knowledge, such cell systems have not yet been reported. We thus utilized our NGS pipeline to analyze the RNA-seq data sets of 182 lung cancer cell lines from the Cancer Cell Line Encyclopedia (CCLE) cohort [111], including 129 NSCLC and 53 SCLC samples. Our data showed that none of the analyzed cells carries latently infected EBV (Lin unpublished data). Since EBV(+) LC cell models are not available, we have set out to infect the lung cancer cells using the well-established cell-to-cell EBV infection method by co-culturing EBV(+) Akata cells with lung cancer cells. After EBV infection, a lung cancer cell line carrying a type II latency was successfully established (Lin unpublished data), which should be a good model to monitor the direct interaction between EBV and lung epithelial cells. Furthermore, an animal model for EBV(+) LCs has not yet been reported. Future work to develop a suitable in vivo disease model (e.g., a patient-derived xenograft EBV(+) LC mouse model) will be surely warranted and it will be critical for understanding EBV-mediated immune evasion and infection during lung carcinogenesis.

6. Challenges and Future Directions

One remaining puzzle is how EBV gains access to the lung epithelium when the EBV life cycle is believed to occur in oral epithelial cells and B cells. Although we cannot rule out the possibility that lung epithelium may serve as a normal reservoir for EBV infection, another possible explanation is that some inflammation events preceding lung cancer may help the EBV infection. Since lung tissue is not a sterile environment, the constant interaction between pulmonary microbiota and lung tissue may elicit inflammation that subsequently attracts lymphocytes and certain lytic-inducing epithelium-derived extracellular vesicles [112,113] to facilitate EBV infection. Further, since EBV is constantly shed into the saliva, direct aspiration of EBV(+) saliva into the lung might also be a possible route for infection.
In contrast to the well-established route for EBV entry into B cells, it is still largely unclear how EBV enters epithelial cells. We reasoned that EBV may enter the lung epithelial cells through multiple mechanisms: (1) cell-to-cell mechanism through which EBV can enter lung epithelial cells by cell membrane contact or the formation of cell-in-cell structure of EBV-infected B cells and uninfected epithelial cells [114]; (2) Cell-free virus may enter cells by directly interacting with the EBV receptor (CD21) expressed on the respiratory epithelial cells; (3) Recent studies have shown that two novel receptors neuropilin 1 (NRP1) and ephrin A2 (EFNA2) can mediate EBV entry into nasopharyngeal epithelial cells. Since NRP1 are also expressed on the lung epithelial cells, NRP1 may facilitate viral internalization and membrane fusion by interacting with EBV glycoprotein gB [115,116]. Although EFNA2 has been shown to promote the cell-free virus entry into nasopharyngeal epithelial cells, EBV is less likely to use this route since EFNA2 is only weakly expressed in lung epithelial cells [117,118].
The observation that EBV(+) LCs are mostly seen in Asian populations and that the disease is more prevalent in NPC endemic regions raises the possibility that some viral genomic variations may contribute to the unusual geographic distribution of EBV(+) LCs. As a DNA virus, EBV has long been thought to have a highly conserved genome. However, increasing lines of evidence have shown that EBV genome polymorphism may contribute to its oncogenicity. For example, Feng and colleagues reported that a particular single nucleotide polymorphism in the EBV genome (SNP G155391A) leading to an RPMS1 variant is strongly associated with NPC but not other EBV-related malignancies [119]. It will be interesting to see if this particular RPMS1 variant also impacts the development of EBV(+) LCs.
The low survival rate of LCs partially results from the fact that LCs are largely diagnosed in their late/advanced stages when surgical treatment is no longer an option. However, since EBV itself can serve as a great biomarker, routine application of EBV serology or nucleic acid testing could offer early diagnosis of EBV(+) LCs during the surgically treatable stages of this disease. Additionally, the EBV testing could also be useful in monitoring treatment and providing prognostic information. Further, since EBV(+) LCs usually do not carry the EGFR mutation, which makes conventional tyrosine kinase inhibitor (TKI)-based therapy ineffective. Thus, stratification of cancer patients with EBV(+) LCs will help choose more efficient therapeutic regimen. For example, since EBV(+) LCs usually express higher levels of PD-L1 as well as other immune checkpoint molecules, the new immune checkpoint inhibitors may help improve the overall survival of EBV(+) LCs by preventing immune evasion of the EBV-infected tumor cells. In the future, EBV(+) LC patients may also benefit from next-generation drugs targeting the EBV gene products that exclusively expressed in the cancer cells.
Together, better understanding of EBV-associated lung carcinogenesis will provide important mechanistic basis for the development of personalized cancer therapy and diagnostics that will ultimately benefit many cancer patients.

Author Contributions

Z.L., D.B. and R.A. contributed to the conceptualization and drafting/revising the manuscript. A.N., J.A., J.K. and L.L. contributed to conceptualization and manuscript revision. All authors have read and agreed to the published version of the manuscript.

Funding

Research reported in this publication was supported by the National Cancer Institute of the National Institutes of Health under Award Number R01CA261258 to Z.L. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. This work was also supported by a National Institutes of Health COBRE grant P20GM121288, a U.S. –Japan Cooperative Medical Sciences Program Collaborative Award from the National Institute of Allergy and Infectious Diseases and CRDF Global (grant number DAA3-19-65602-1), a Tulane school of medicine faculty research pilot grant, and a Carol Lavin Bernick faculty grant to Z.L. as well as the Japan Society for the Promotion of Science (grant number JP17K4567), the Takeda Science Foundation, the Shiseido Female Researcher Science, and Japanese Initiative for Progress of Research on Infectious Disease for global Epidemic (J-PRIDE) grant from Japan Agency for Medical Research and Development (grant number 20fm0208101j0004) to A.N.

Conflicts of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be constructed as a potential conflict of interest.

References

  1. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer statistics, 2021. CA Cancer J. Clin. 2021, 71, 7–33. [Google Scholar] [CrossRef] [PubMed]
  2. Travis, W.D.; Brambilla, E.; Nicholson, A.G.; Yatabe, Y.; Austin, J.H.M.; Beasley, M.B.; Chirieac, L.R.; Dacic, S.; Duhig, E.; Flieder, D.B.; et al. The 2015 world health organization classification of lung tumors: Impact of genetic, clinical and radiologic advances since the 2004 classification. J. Thorac. Oncol. 2015, 10, 1243–1260. [Google Scholar] [CrossRef] [Green Version]
  3. Zappa, C.; Mousa, S.A. Non-small cell lung cancer: Current treatment and future advances. Transl. Lung Cancer Res. 2016, 5, 288–300. [Google Scholar] [CrossRef] [Green Version]
  4. Thun, M.J.; Henley, S.J.; Calle, E.E. Tobacco use and cancer: An epidemiologic perspective for geneticists. Oncogene 2002, 21, 7307–7325. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Sun, S.; Schiller, J.H.; Gazdar, A.F. Lung cancer in never smokers—A different disease. Nat. Rev. Cancer 2007, 7, 778–790. [Google Scholar] [CrossRef]
  6. Cheng, T.Y.; Cramb, S.M.; Baade, P.D.; Youlden, D.R.; Nwogu, C.; Reid, M.E. The international epidemiology of lung cancer: Latest trends, disparities, and tumor characteristics. J. Thorac. Oncol. 2016, 11, 1653–1671. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Young, L.S.; Yap, L.F.; Murray, P.G. Epstein-barr virus: More than 50 years old and still providing surprises. Nat. Rev. Cancer 2016, 16, 789–802. [Google Scholar] [CrossRef] [PubMed]
  8. Rickinson, A.; Kieff, E. Epstein-barr virus. In Fields Virology, 5th ed.; Knipe, D., Howley, P., Eds.; Lippincott Williams & Wilkins: Philadelphia, PA, USA, 2007; pp. 2655–2700. [Google Scholar]
  9. Shannon-Lowe, C.; Rickinson, A. The global landscape of ebv-associated tumors. Front. Oncol. 2019, 9, 713. [Google Scholar] [CrossRef] [Green Version]
  10. de Martel, C.; Ferlay, J.; Franceschi, S.; Vignat, J.; Bray, F.; Forman, D.; Plummer, M. Global burden of cancers attributable to infections in 2008: A review and synthetic analysis. Lancet Oncol. 2012, 13, 607–615. [Google Scholar] [CrossRef]
  11. Khan, G.; Hashim, M.J. Global burden of deaths from epstein-barr virus attributable malignancies 1990–2010. Infect. Agent. Cancer 2014, 9, 38. [Google Scholar] [CrossRef] [Green Version]
  12. Kheir, F.; Zhao, M.; Strong, M.J.; Yu, Y.; Nanbo, A.; Flemington, E.K.; Morris, G.F.; Reiss, K.; Li, L.; Lin, Z. Detection of epstein-barr virus infection in non-small cell lung cancer. Cancers 2019, 11, 759. [Google Scholar] [CrossRef] [Green Version]
  13. De Sanjose, S.; Bosch, R.; Schouten, T.; Verkuijlen, S.; Nieters, A.; Foretova, L.; Maynadie, M.; Cocco, P.L.; Staines, A.; Becker, N.; et al. Epstein-barr virus infection and risk of lymphoma: Immunoblot analysis of antibody responses against ebv-related proteins in a large series of lymphoma subjects and matched controls. Int. J. Cancer 2007, 121, 1806–1812. [Google Scholar] [CrossRef]
  14. Strong, M.J.; O’Grady, T.; Lin, Z.; Xu, G.; Baddoo, M.; Parsons, C.; Zhang, K.; Taylor, C.M.; Flemington, E.K. Epstein-barr virus and human herpesvirus 6 detection in a non-hodgkin’s diffuse large b-cell lymphoma cohort by using rna sequencing. J. Virol. 2013, 87, 13059–13062. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Strong, M.J.; Xu, G.; Coco, J.; Baribault, C.; Vinay, D.S.; Lacey, M.R.; Strong, A.L.; Lehman, T.A.; Seddon, M.B.; Lin, Z.; et al. Differences in gastric carcinoma microenvironment stratify according to ebv infection intensity: Implications for possible immune adjuvant therapy. PLoS Pathog. 2013, 9, e1003341. [Google Scholar] [CrossRef] [PubMed]
  16. Zhao, M.; Nanbo, A.; Becnel, D.; Qin, Z.; Morris, G.F.; Li, L.; Lin, Z. Ubiquitin modification of the epstein-barr virus immediate early transactivator zta. J. Virol. 2020, 94. [Google Scholar] [CrossRef]
  17. Lin, Z.; Flemington, E. Regulation of ebv latency by viral lytic proteins. In Epstein-Barr Virus: Latency and Transformation; Robertson, E.S., Ed.; Caister Academic Press: London, UK, 2010; pp. 167–192. [Google Scholar]
  18. Tugizov, S.M.; Herrera, R.; Palefsky, J.M. Epstein-barr virus transcytosis through polarized oral epithelial cells. J. Virol. 2013, 87, 8179–8194. [Google Scholar] [CrossRef] [Green Version]
  19. Laichalk, L.L.; Thorley-Lawson, D.A. Terminal differentiation into plasma cells initiates the replicative cycle of epstein-barr virus in vivo. J. Virol. 2005, 79, 1296–1307. [Google Scholar] [CrossRef] [Green Version]
  20. Sun, C.C.; Thorley-Lawson, D.A. Plasma cell-specific transcription factor xbp-1s binds to and transactivates the epstein-barr virus bzlf1 promoter. J. Virol. 2007, 81, 13566–13577. [Google Scholar] [CrossRef] [Green Version]
  21. Wilson, J.B.; Manet, E.; Gruffat, H.; Busson, P.; Blondel, M.; Fahraeus, R. Ebna1: Oncogenic activity, immune evasion and biochemical functions provide targets for novel therapeutic strategies against epstein-barr virus- associated cancers. Cancers 2018, 10, 109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Moss, W.N.; Steitz, J.A. Genome-wide analyses of epstein-barr virus reveal conserved rna structures and a novel stable intronic sequence rna. BMC Genom. 2013, 14, 543. [Google Scholar] [CrossRef] [Green Version]
  23. Hutzinger, R.; Feederle, R.; Mrazek, J.; Schiefermeier, N.; Balwierz, P.J.; Zavolan, M.; Polacek, N.; Delecluse, H.J.; Huttenhofer, A. Expression and processing of a small nucleolar rna from the epstein-barr virus genome. PLoS Pathog. 2009, 5, e1000547. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. O’Grady, T.; Cao, S.; Strong, M.J.; Concha, M.; Wang, X.; Splinter Bondurant, S.; Adams, M.; Baddoo, M.; Srivastav, S.K.; Lin, Z.; et al. Global bidirectional transcription of the epstein-barr virus genome during reactivation. J. Virol. 2014, 88, 1604–1616. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. O’Grady, T.; Wang, X.; Honer Zu Bentrup, K.; Baddoo, M.; Concha, M.; Flemington, E.K. Global transcript structure resolution of high gene density genomes through multi-platform data integration. Nucleic Acids Res. 2016, 44, e145. [Google Scholar] [CrossRef] [PubMed]
  26. Cao, S.; Strong, M.J.; Wang, X.; Moss, W.N.; Concha, M.; Lin, Z.; O’Grady, T.; Baddoo, M.; Fewell, C.; Renne, R.; et al. High-throughput rna sequencing-based virome analysis of 50 lymphoma cell lines from the cancer cell line encyclopedia project. J. Virol. 2015, 89, 713–729. [Google Scholar] [CrossRef] [Green Version]
  27. Concha, M.; Wang, X.; Cao, S.; Baddoo, M.; Fewell, C.; Lin, Z.; Hulme, W.; Hedges, D.; McBride, J.; Flemington, E.K. Identification of new viral genes and transcript isoforms during epstein-barr virus reactivation using rna-seq. J. Virol. 2012, 86, 1458–1467. [Google Scholar] [CrossRef] [Green Version]
  28. Pfeffer, S.; Zavolan, M.; Grasser, F.A.; Chien, M.; Russo, J.J.; Ju, J.; John, B.; Enright, A.J.; Marks, D.; Sander, C.; et al. Identification of virus-encoded micrornas. Science 2004, 304, 734–736. [Google Scholar] [CrossRef]
  29. Cai, X.; Schafer, A.; Lu, S.; Bilello, J.P.; Desrosiers, R.C.; Edwards, R.; Raab-Traub, N.; Cullen, B.R. Epstein-barr virus micrornas are evolutionarily conserved and differentially expressed. PLoS Pathog. 2006, 2, e23. [Google Scholar] [CrossRef] [Green Version]
  30. Cao, S.; Moss, W.; O’Grady, T.; Concha, M.; Strong, M.J.; Wang, X.; Yu, Y.; Baddoo, M.; Zhang, K.; Fewell, C.; et al. New noncoding lytic transcripts derived from the epstein-barr virus latency origin of replication, orip, are hyperedited, bind the paraspeckle protein, nono/p54nrb, and support viral lytic transcription. J. Virol. 2015, 89, 7120–7132. [Google Scholar] [CrossRef] [Green Version]
  31. Khatiwada, S.; Subedi, A. Lung microbiome and coronavirus disease 2019 (COVID-19): Possible link and implications. Hum. Microb. J. 2020, 17, 100073. [Google Scholar] [CrossRef]
  32. Engels, E.A. Inflammation in the development of lung cancer: Epidemiological evidence. Expert Rev. Anticancer Ther. 2008, 8, 605–615. [Google Scholar] [CrossRef]
  33. Leroux, C.; Girard, N.; Cottin, V.; Greenland, T.; Mornex, J.F.; Archer, F. Jaagsiekte sheep retrovirus (jsrv): From virus to lung cancer in sheep. Vet. Res. 2007, 38, 211–228. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Desgranges, C.; de-The, G. Epstein-barr virus specific iga serum antibodies in nasopharyngeal and other respiratory carcinomas. Int. J. Cancer 1979, 24, 555–559. [Google Scholar] [CrossRef]
  35. Lung, M.L.; Lam, W.K.; So, S.Y.; Lam, W.P.; Chan, K.H.; Ng, M.H. Evidence that respiratory tract is major reservoir for epstein-barr virus. Lancet 1985, 1, 889–892. [Google Scholar] [CrossRef]
  36. Begin, L.R.; Eskandari, J.; Joncas, J.; Panasci, L. Epstein-barr virus related lymphoepithelioma-like carcinoma of lung. J. Surg. Oncol. 1987, 36, 280–283. [Google Scholar] [CrossRef]
  37. Wockel, W.; Hofler, G.; Popper, H.H.; Morresi, A. Lymphoepithelioma-like carcinoma of the lung. Pathol. Res. Pract. 1995, 191, 1170–1174. [Google Scholar] [CrossRef]
  38. Higashiyama, M.; Doi, O.; Kodama, K.; Yokouchi, H.; Tateishi, R.; Horiuchi, K.; Mishima, K. Lymphoepithelioma-like carcinoma of the lung: Analysis of two cases for epstein-barr virus infection. Hum. Pathol. 1995, 26, 1278–1282. [Google Scholar] [CrossRef]
  39. Chan, J.K.; Hui, P.K.; Tsang, W.Y.; Law, C.K.; Ma, C.C.; Yip, T.T.; Poon, Y.F. Primary lymphoepithelioma-like carcinoma of the lung. A clinicopathologic study of 11 cases. Cancer 1995, 76, 413–422. [Google Scholar] [CrossRef]
  40. Han, A.J.; Xiong, M.; Zong, Y.S. Association of epstein-barr virus with lymphoepithelioma-like carcinoma of the lung in southern china. Am. J. Clin. Pathol. 2000, 114, 220–226. [Google Scholar] [CrossRef]
  41. Gomez-Roman, J.J.; Martinez, M.N.; Fernandez, S.L.; Val-Bernal, J.F. Epstein-barr virus-associated adenocarcinomas and squamous-cell lung carcinomas. Mod. Pathol. 2009, 22, 530–537. [Google Scholar] [CrossRef] [Green Version]
  42. Chen, F.F.; Yan, J.J.; Lai, W.W.; Jin, Y.T.; Su, I.J. Epstein-barr virus-associated nonsmall cell lung carcinoma: Undifferentiated “lymphoepithelioma-like” carcinoma as a distinct entity with better prognosis. Cancer 1998, 82, 2334–2342. [Google Scholar] [CrossRef]
  43. Li, C.M.; Han, G.L.; Zhang, S.J. Detection of epstein-barr virus in lung carcinoma tissue by in situ hybridization. Zhonghua Shi Yan He Lin Chuang Bing Du Xue Za Zhi 2007, 21, 288–290. [Google Scholar]
  44. Kasai, K.; Sato, Y.; Kameya, T.; Inoue, H.; Yoshimura, H.; Kon, S.; Kikuchi, K. Incidence of latent infection of epstein-barr virus in lung cancers--an analysis of eber1 expression in lung cancers by in situ hybridization. J. Pathol. 1994, 174, 257–265. [Google Scholar] [CrossRef]
  45. Huber, M.; Pavlova, B.; Muhlberger, H.; Hollaus, P.; Lintner, F. Detection of the epstein-barr virus in primary adenocarcinoma of the lung with signet-ring cells. Virchows Arch. 2002, 441, 25–30. [Google Scholar] [CrossRef] [PubMed]
  46. Wang, S.; Xiong, H.; Yan, S.; Wu, N.; Lu, Z. Identification and characterization of epstein-barr virus genomes in lung carcinoma biopsy samples by next-generation sequencing technology. Sci. Rep. 2016, 6, 26156. [Google Scholar] [CrossRef] [Green Version]
  47. Jafarian, A.H.; Omidi-Ashrafi, A.; Mohamadian-Roshan, N.; Karimi-Shahri, M.; Ghazvini, K.; Boroumand-Noughabi, S. Association of epstein barr virus deoxyribonucleic acid with lung carcinoma. Indian J. Pathol. Microbiol. 2013, 56, 359–364. [Google Scholar] [PubMed]
  48. Chu, P.G.; Cerilli, L.; Chen, Y.Y.; Mills, S.E.; Weiss, L.M. Epstein-barr virus plays no role in the tumorigenesis of small-cell carcinoma of the lung. Mod. Pathol. 2004, 17, 158–164. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Butler, A.E.; Colby, T.V.; Weiss, L.; Lombard, C. Lymphoepithelioma-like carcinoma of the lung. Am. J. Surg. Pathol. 1989, 13, 632–639. [Google Scholar] [CrossRef] [PubMed]
  50. Gal, A.A.; Unger, E.R.; Koss, M.N.; Yen, T.S. Detection of epstein-barr virus in lymphoepithelioma-like carcinoma of the lung. Mod. Pathol. 1991, 4, 264–268. [Google Scholar] [PubMed]
  51. Miller, B.; Montgomery, C.; Watne, A.L.; Johnson, D.; Bailey, T.; Kowalski, R. Lymphoepithelioma-like carcinoma of the lung. J. Surg. Oncol. 1991, 48, 62–68. [Google Scholar] [CrossRef] [PubMed]
  52. Pittaluga, S.; Wong, M.P.; Chung, L.P.; Loke, S.L. Clonal epstein-barr virus in lymphoepithelioma-like carcinoma of the lung. Am. J. Surg. Pathol. 1993, 17, 678–682. [Google Scholar] [CrossRef]
  53. Wong, M.P.; Chung, L.P.; Yuen, S.T.; Leung, S.Y.; Chan, S.Y.; Wang, E.; Fu, K.H. In situ detection of epstein-barr virus in non-small cell lung carcinomas. J Pathol. 1995, 177, 233–240. [Google Scholar] [CrossRef] [PubMed]
  54. Ferrara, G.; Nappi, O. Lymphoepithelioma-like carcinoma of the lung. Two cases diagnosed in caucasian patients. Tumori 1995, 81, 144–147. [Google Scholar] [CrossRef] [PubMed]
  55. Conway, E.J.; Hudnall, S.D.; Lazarides, A.; Bahler, A.; Fraire, A.E.; Cagle, P.T. Absence of evidence for an etiologic role for epstein-barr virus in neoplasms of the lung and pleura. Mod. Pathol. 1996, 9, 491–495. [Google Scholar] [PubMed]
  56. Frank, M.W.; Shields, T.W.; Joob, A.W.; Kies, M.S.; Sturgis, C.D.; Yeldandi, A.; Cribbins, A.J.; Fullerton, D.A. Lymphoepithelioma-like carcinoma of the lung. Ann. Thorac. Surg. 1997, 64, 1162–1164. [Google Scholar] [CrossRef]
  57. Curcio, L.D.; Cohen, J.S.; Grannis, F.W., Jr.; Paz, I.B.; Chilcote, R.; Weiss, L.M. Primary lymphoepithelioma-like carcinoma of the lung in a child. Report of an epstein-barr virus-related neoplasm. Chest 1997, 111, 250–251. [Google Scholar] [CrossRef]
  58. Kasai, K.; Kon, S.; Sato, N.; Muraishi, K.; Yoshida, H.; Nakai, N.; Hamakawa, H.; Itoh, C.; Yamaoka, S. Case report of lymphoepithelioma-like carcinoma of the lung--lymphoid population consisting of cytotoxic t cells in resting state. Pathol. Res. Pract. 1999, 195, 773–779. [Google Scholar] [CrossRef]
  59. Muraishi, K.; Kon, S.; Yosida, H.; Hamakawa, H.; Nakai, N.; Itoh, C.; Yamaoka, S.; Kasai, K. lymphoepithelioma-like carcinoma of the lung. Nihon Kokyuki Gakkai Zasshi 1999, 37, 565–570. [Google Scholar] [PubMed]
  60. Castro, C.Y.; Ostrowski, M.L.; Barrios, R.; Green, L.K.; Popper, H.H.; Powell, S.; Cagle, P.T.; Ro, J.Y. Relationship between epstein-barr virus and lymphoepithelioma-like carcinoma of the lung: A clinicopathologic study of 6 cases and review of the literature. Hum. Pathol. 2001, 32, 863–872. [Google Scholar] [CrossRef] [PubMed]
  61. Ngan, R.K.; Yip, T.T.; Cheng, W.W.; Chan, J.K.; Cho, W.C.; Ma, V.W.; Wan, K.K.; Au, S.K.; Law, C.K.; Lau, W.H. Circulating epstein-barr virus DNA in serum of patients with lymphoepithelioma-like carcinoma of the lung: A potential surrogate marker for monitoring disease. Clin. Cancer Res. 2002, 8, 986–994. [Google Scholar]
  62. Chang, Y.L.; Wu, C.T.; Shih, J.Y.; Lee, Y.C. New aspects in clinicopathologic and oncogene studies of 23 pulmonary lymphoepithelioma-like carcinomas. Am. J. Surg. Pathol. 2002, 26, 715–723. [Google Scholar] [CrossRef]
  63. Brouchet, L.; Valmary, S.; Dahan, M.; Didier, A.; Galateau-Salle, F.; Brousset, P.; Degano, B. Detection of oncogenic virus genomes and gene products in lung carcinoma. Br. J. Cancer 2005, 92, 743–746. [Google Scholar] [CrossRef] [Green Version]
  64. Chau, S.L.; Tong, J.H.; Chow, C.; Kwan, J.S.; Lung, R.W.; Chung, L.Y.; Tin, E.K.; Wong, S.S.; Cheung, A.H.; Lau, R.W.; et al. Distinct molecular landscape of epstein-barr virus associated pulmonary lymphoepithelioma-like carcinoma revealed by genomic sequencing. Cancers 2020, 12, 2065. [Google Scholar] [CrossRef] [PubMed]
  65. Chen, B.; Chen, X.; Zhou, P.; Yang, L.; Ren, J.; Yang, X.; Li, W. Primary pulmonary lymphoepithelioma-like carcinoma: A rare type of lung cancer with a favorable outcome in comparison to squamous carcinoma. Respir. Res. 2019, 20, 262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Chen, B.; Zhang, Y.; Dai, S.; Zhou, P.; Luo, W.; Wang, Z.; Chen, X.; Cheng, P.; Zheng, G.; Ren, J.; et al. Molecular characteristics of primary pulmonary lymphoepithelioma-like carcinoma based on integrated genomic analyses. Signal. Transduct. Target. Ther. 2021, 6, 6. [Google Scholar] [CrossRef] [PubMed]
  67. Firincioglulari, A.; Akinci Ozyurek, B.; Erdogan, Y.; Incekara, F.; Yilmaz, E.; Ozaydin, H.E. Lymphoepithelioma-like carcinoma of the lung: A rare case report and review of the literature. Tuberk. Toraks 2020, 68, 453–457. [Google Scholar] [CrossRef] [PubMed]
  68. Hong, S.; Liu, D.; Luo, S.; Fang, W.; Zhan, J.; Fu, S.; Zhang, Y.; Wu, X.; Zhou, H.; Chen, X.; et al. The genomic landscape of epstein-barr virus-associated pulmonary lymphoepithelioma-like carcinoma. Nat. Commun. 2019, 10, 3108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Huang, Y.C.; Hsueh, C.; Ho, S.Y.; Liao, C.Y. Lymphoepithelioma-like carcinoma of the lung: An unusual case and literature review. Case Rep. Pulmonol. 2013, 2013, 143405. [Google Scholar] [CrossRef]
  70. Kobayashi, M.; Ito, M.; Sano, K.; Honda, T.; Nakayama, J. Pulmonary lymphoepithelioma-like carcinoma: Predominant infiltration of tumor-associated cytotoxic t lymphocytes might represent the enhanced tumor immunity. Intern. Med. 2004, 43, 323–326. [Google Scholar] [CrossRef] [Green Version]
  71. Koshiol, J.; Gulley, M.L.; Zhao, Y.; Rubagotti, M.; Marincola, F.M.; Rotunno, M.; Tang, W.; Bergen, A.W.; Bertazzi, P.A.; Roy, D.; et al. Epstein-barr virus micrornas and lung cancer. Br. J. Cancer 2011, 105, 320–326. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Lim, W.T.; Chuah, K.L.; Leong, S.S.; Tan, E.H.; Toh, C.K. Assessment of human papillomavirus and epstein-barr virus in lung adenocarcinoma. Oncol. Rep. 2009, 21, 971–975. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Sasaki, A.; Kato, T.; Ujiie, H.; Cho, Y.; Sato, M.; Kaji, M. Primary pulmonary lymphoepithelioma-like carcinoma with positive expression of epstein-barr virus and pd-l1: A case report. Int. J. Surg. Case Rep. 2021, 79, 431–435. [Google Scholar] [CrossRef] [PubMed]
  74. Tanaka, S.; Chen, F.; Date, H. Pulmonary lymphoepithelioma-like carcinoma with rapid progression. Gen. Thorac. Cardiovasc. Surg. 2012, 60, 164–167. [Google Scholar] [CrossRef] [PubMed]
  75. Wong, J.F.; Teo, M.C. Case report: Lymphoepithelial-like carcinoma of the lung—A chronic disease? World J. Surg. Oncol. 2012, 10, 91. [Google Scholar] [CrossRef] [Green Version]
  76. Yoshino, N.; Kubokura, H.; Yamauchi, S.; Ohaki, Y.; Koizumi, K.; Shimizu, K. Lymphoepithelioma-like carcinoma of the lung: Case in which the patient has been followed up for 7 years postoperatively. JPN J. Thorac. Cardiovasc. Surg. 2005, 53, 653–656. [Google Scholar] [CrossRef]
  77. Lin, Z.; Xu, G.; Deng, N.; Taylor, C.; Zhu, D.; Flemington, E.K. Quantitative and qualitative rna-seq-based evaluation of epstein-barr virus transcription in type i latency burkitt’s lymphoma cells. J. Virol. 2010, 84, 13053–13058. [Google Scholar] [CrossRef] [Green Version]
  78. Lin, Z.; Puetter, A.; Coco, J.; Xu, G.; Strong, M.J.; Wang, X.; Fewell, C.; Baddoo, M.; Taylor, C.; Flemington, E.K. Detection of murine leukemia virus in the epstein-barr virus-positive human b-cell line jy, using a computational rna-seq-based exogenous agent detection pipeline, parses. J. Virol. 2012, 86, 2970–2977. [Google Scholar] [CrossRef] [Green Version]
  79. Lin, Z.; Wang, X.; Strong, M.J.; Concha, M.; Baddoo, M.; Xu, G.; Baribault, C.; Fewell, C.; Hulme, W.; Hedges, D.; et al. Whole-genome sequencing of the akata and mutu epstein-barr virus strains. J. Virol. 2013, 87, 1172–1182. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Strong, M.J.; Baddoo, M.; Nanbo, A.; Xu, M.; Puetter, A.; Lin, Z. Comprehensive high-throughput rna sequencing analysis reveals contamination of multiple nasopharyngeal carcinoma cell lines with hela cell genomes. J. Virol. 2014, 88, 10696–10704. [Google Scholar] [CrossRef] [Green Version]
  81. Strong, M.J.; Laskow, T.; Nakhoul, H.; Blanchard, E.; Liu, Y.; Wang, X.; Baddoo, M.; Lin, Z.; Yin, Q.; Flemington, E.K. Latent expression of the epstein-barr virus (ebv)-encoded major histocompatibility complex class i tap inhibitor, bnlf2a, in ebv-positive gastric carcinomas. J. Virol. 2015, 89, 10110–10114. [Google Scholar] [CrossRef] [Green Version]
  82. Strong, M.J.; Blanchard, E.t.; Lin, Z.; Morris, C.A.; Baddoo, M.; Taylor, C.M.; Ware, M.L.; Flemington, E.K. A comprehensive next generation sequencing-based virome assessment in brain tissue suggests no major virus-tumor association. Acta Neuropathol. Commun. 2016, 4, 71. [Google Scholar] [CrossRef] [Green Version]
  83. Feng, H.; Shuda, M.; Chang, Y.; Moore, P.S. Clonal integration of a polyomavirus in human merkel cell carcinoma. Science 2008, 319, 1096–1100. [Google Scholar] [CrossRef] [Green Version]
  84. Castellarin, M.; Warren, R.L.; Freeman, J.D.; Dreolini, L.; Krzywinski, M.; Strauss, J.; Barnes, R.; Watson, P.; Allen-Vercoe, E.; Moore, R.A.; et al. Fusobacterium nucleatum infection is prevalent in human colorectal carcinoma. Genome Res. 2012, 22, 299–306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Kostic, A.D.; Gevers, D.; Pedamallu, C.S.; Michaud, M.; Duke, F.; Earl, A.M.; Ojesina, A.I.; Jung, J.; Bass, A.J.; Tabernero, J.; et al. Genomic analysis identifies association of fusobacterium with colorectal carcinoma. Genome Res. 2012, 22, 292–298. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Ambinder, R.F. Gammaherpesviruses and “hit-and-run” oncogenesis. Am. J. Pathol. 2000, 156, 1–3. [Google Scholar] [CrossRef]
  87. Hu, H.; Luo, M.L.; Desmedt, C.; Nabavi, S.; Yadegarynia, S.; Hong, A.; Konstantinopoulos, P.A.; Gabrielson, E.; Hines-Boykin, R.; Pihan, G.; et al. Epstein-barr virus infection of mammary epithelial cells promotes malignant transformation. EBioMedicine 2016, 9, 148–160. [Google Scholar] [CrossRef] [Green Version]
  88. Al-Mozaini, M.; Bodelon, G.; Karstegl, C.E.; Jin, B.; Al-Ahdal, M.; Farrell, P.J. Epstein-barr virus bart gene expression. J. Gen. Virol. 2009, 90, 307–316. [Google Scholar] [CrossRef]
  89. Smith, P.R.; de Jesus, O.; Turner, D.; Hollyoake, M.; Karstegl, C.E.; Griffin, B.E.; Karran, L.; Wang, Y.; Hayward, S.D.; Farrell, P.J. Structure and coding content of cst (bart) family rnas of epstein-barr virus. J. Virol. 2000, 74, 3082–3092. [Google Scholar] [CrossRef] [Green Version]
  90. Marquitz, A.R.; Mathur, A.; Edwards, R.H.; Raab-Traub, N. Host gene expression is regulated by two types of noncoding rnas transcribed from the epstein-barr virus bamhi a rightward transcript region. J. Virol. 2015, 89, 11256–11268. [Google Scholar] [CrossRef] [Green Version]
  91. Lin, Z.; Flemington, E.K. Mirnas in the pathogenesis of oncogenic human viruses. Cancer Lett. 2011, 305, 186–199. [Google Scholar] [CrossRef] [Green Version]
  92. Fox, C.P.; Haigh, T.A.; Taylor, G.S.; Long, H.M.; Lee, S.P.; Shannon-Lowe, C.; O’Connor, S.; Bollard, C.M.; Iqbal, J.; Chan, W.C.; et al. 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, 3695–3704. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Cen, O.; Longnecker, R. Latent membrane protein 2 (lmp2). Curr. Top. Microbiol. Immunol. 2015, 391, 151–180. [Google Scholar]
  94. Hwu, P.; Du, M.X.; Lapointe, R.; Do, M.; Taylor, M.W.; Young, H.A. Indoleamine 2,3-dioxygenase production by human dendritic cells results in the inhibition of t cell proliferation. J. Immunol. 2000, 164, 3596–3599. [Google Scholar] [CrossRef] [PubMed]
  95. Munn, D.H.; Shafizadeh, E.; Attwood, J.T.; Bondarev, I.; Pashine, A.; Mellor, A.L. Inhibition of t cell proliferation by macrophage tryptophan catabolism. J. Exp. Med. 1999, 189, 1363–1372. [Google Scholar] [CrossRef] [PubMed]
  96. Uyttenhove, C.; Pilotte, L.; Theate, I.; Stroobant, V.; Colau, D.; Parmentier, N.; Boon, T.; Van den Eynde, B.J. Evidence for a tumoral immune resistance mechanism based on tryptophan degradation by indoleamine 2,3-dioxygenase. Nat. Med. 2003, 9, 1269–1274. [Google Scholar] [CrossRef]
  97. Bell, M.J.; Abbott, R.J.; Croft, N.P.; Hislop, A.D.; Burrows, S.R. An hla-a2-restricted t-cell epitope mapped to the bnlf2a immune evasion protein of epstein-barr virus that inhibits tap. J. Virol. 2009, 83, 2783–2788. [Google Scholar] [CrossRef] [Green Version]
  98. Horst, D.; van Leeuwen, D.; Croft, N.P.; Garstka, M.A.; Hislop, A.D.; Kremmer, E.; Rickinson, A.B.; Wiertz, E.J.; Ressing, M.E. Specific targeting of the ebv lytic phase protein bnlf2a to the transporter associated with antigen processing results in impairment of hla class i-restricted antigen presentation. J. Immunol. 2009, 182, 2313–2324. [Google Scholar] [CrossRef] [Green Version]
  99. Croft, N.P.; Shannon-Lowe, C.; Bell, A.I.; Horst, D.; Kremmer, E.; Ressing, M.E.; Wiertz, E.J.; Middeldorp, J.M.; Rowe, M.; Rickinson, A.B.; et al. Stage-specific inhibition of mhc class i presentation by the epstein-barr virus bnlf2a protein during virus lytic cycle. PLoS Pathog. 2009, 5, e1000490. [Google Scholar] [CrossRef] [PubMed]
  100. Horst, D.; Favaloro, V.; Vilardi, F.; van Leeuwen, H.C.; Garstka, M.A.; Hislop, A.D.; Rabu, C.; Kremmer, E.; Rickinson, A.B.; High, S.; et al. Ebv protein bnlf2a exploits host tail-anchored protein integration machinery to inhibit tap. J. Immunol. 2011, 186, 3594–3605. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Wycisk, A.I.; Lin, J.; Loch, S.; Hobohm, K.; Funke, J.; Wieneke, R.; Koch, J.; Skach, W.R.; Mayerhofer, P.U.; Tampe, R. Epstein-barr viral bnlf2a protein hijacks the tail-anchored protein insertion machinery to block antigen processing by the transport complex tap. J. Biol. Chem. 2011, 286, 41402–41412. [Google Scholar] [CrossRef] [Green Version]
  102. Thorley-Lawson, D.A.; Gross, A. Persistence of the epstein-barr virus and the origins of associated lymphomas. N. Engl. J. Med. 2004, 350, 1328–1337. [Google Scholar] [CrossRef]
  103. Dutta, D.; Dutta, S.; Veettil, M.V.; Roy, A.; Ansari, M.A.; Iqbal, J.; Chikoti, L.; Kumar, B.; Johnson, K.E.; Chandran, B. Brca1 regulates ifi16 mediated nuclear innate sensing of herpes viral DNA and subsequent induction of the innate inflammasome and interferon-beta responses. PLoS Pathog. 2015, 11, e1005030. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Liao, G.; Huang, J.; Fixman, E.D.; Hayward, S.D. The epstein-barr virus replication protein bblf2/3 provides an origin-tethering function through interaction with the zinc finger DNA binding protein zbrk1 and the kap-1 corepressor. J. Virol. 2005, 79, 245–256. [Google Scholar] [CrossRef] [Green Version]
  105. Chang, Y.L.; Yang, C.Y.; Lin, M.W.; Wu, C.T.; Yang, P.C. Pd-l1 is highly expressed in lung lymphoepithelioma-like carcinoma: A potential rationale for immunotherapy. Lung Cancer 2015, 88, 254–259. [Google Scholar] [CrossRef] [PubMed]
  106. Yin, K.; Feng, H.B.; Li, L.L.; Chen, Y.; Xie, Z.; Lv, Z.Y.; Guo, W.B.; Lu, D.X.; Yang, X.N.; Yan, W.Q.; et al. Low frequency of mutation of epidermal growth factor receptor (egfr) and arrangement of anaplastic lymphoma kinase (alk) in primary pulmonary lymphoepithelioma-like carcinoma. Thorac. Cancer 2020, 11, 346–352. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Tam, I.Y.; Chung, L.P.; Suen, W.S.; Wang, E.; Wong, M.C.; Ho, K.K.; Lam, W.K.; Chiu, S.W.; Girard, L.; Minna, J.D.; et al. Distinct epidermal growth factor receptor and kras mutation patterns in non-small cell lung cancer patients with different tobacco exposure and clinicopathologic features. Clin. Cancer Res. 2006, 12, 1647–1653. [Google Scholar] [CrossRef] [Green Version]
  108. Xie, Z.; Liu, L.; Lin, X.; Xie, X.; Gu, Y.; Liu, M.; Zhang, J.; Ouyang, M.; Lizaso, A.; Zhang, H.; et al. A multicenter analysis of genomic profiles and pd-l1 expression of primary lymphoepithelioma-like carcinoma of the lung. Mod. Pathol. 2020, 33, 626–638. [Google Scholar] [CrossRef] [Green Version]
  109. Hanahan, D.; Weinberg, R.A. The hallmarks of cancer. Cell 2000, 100, 57–70. [Google Scholar] [CrossRef] [Green Version]
  110. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [Green Version]
  111. Barretina, J.; Caponigro, G.; Stransky, N.; Venkatesan, K.; Margolin, A.A.; Kim, S.; Wilson, C.J.; Lehar, J.; Kryukov, G.V.; Sonkin, D.; et al. The cancer cell line encyclopedia enables predictive modelling of anticancer drug sensitivity. Nature 2012, 483, 603–607. [Google Scholar] [CrossRef]
  112. Zhao, M.; Nanbo, A.; Sun, L.; Lin, Z. Extracellular vesicles in epstein-barr virus’ life cycle and pathogenesis. Microorganisms 2019, 7, 48. [Google Scholar] [CrossRef] [Green Version]
  113. Lin, Z.; Swan, K.; Zhang, X.; Cao, S.; Brett, Z.; Drury, S.; Strong, M.J.; Fewell, C.; Puetter, A.; Wang, X.; et al. Secreted oral epithelial cell membrane vesicles induce epstein-barr virus reactivation in latently infected b cells. J. Virol. 2016, 90, 3469–3479. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Ni, C.; Chen, Y.; Zeng, M.; Pei, R.; Du, Y.; Tang, L.; Wang, M.; Hu, Y.; Zhu, H.; He, M.; et al. In-cell infection: A novel pathway for epstein-barr virus infection mediated by cell-in-cell structures. Cell Res. 2015, 25, 785–800. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Wang, H.B.; Zhang, H.; Zhang, J.P.; Li, Y.; Zhao, B.; Feng, G.K.; Du, Y.; Xiong, D.; Zhong, Q.; Liu, W.L.; et al. Neuropilin 1 is an entry factor that promotes ebv infection of nasopharyngeal epithelial cells. Nat. Commun. 2015, 6, 6240. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Xiong, D.; Du, Y.; Wang, H.B.; Zhao, B.; Zhang, H.; Li, Y.; Hu, L.J.; Cao, J.Y.; Zhong, Q.; Liu, W.L.; et al. Nonmuscle myosin heavy chain iia mediates epstein-barr virus infection of nasopharyngeal epithelial cells. Proc. Natl. Acad. Sci. USA 2015, 112, 11036–11041. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Chen, J.; Sathiyamoorthy, K.; Zhang, X.; Schaller, S.; Perez White, B.E.; Jardetzky, T.S.; Longnecker, R. Ephrin receptor a2 is a functional entry receptor for epstein-barr virus. Nat. Microbiol. 2018, 3, 172–180. [Google Scholar] [CrossRef] [PubMed]
  118. Zhang, H.; Li, Y.; Wang, H.B.; Zhang, A.; Chen, M.L.; Fang, Z.X.; Dong, X.D.; Li, S.B.; Du, Y.; Xiong, D.; et al. Ephrin receptor a2 is an epithelial cell receptor for epstein-barr virus entry. Nat. Microbiol. 2018, 3, 1–8. [Google Scholar] [CrossRef]
  119. Feng, F.T.; Cui, Q.; Liu, W.S.; Guo, Y.M.; Feng, Q.S.; Chen, L.Z.; Xu, M.; Luo, B.; Li, D.J.; Hu, L.F.; et al. A single nucleotide polymorphism in the epstein-barr virus genome is strongly associated with a high risk of nasopharyngeal carcinoma. Chin. J. Cancer 2015, 34, 563–572. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Proposed disease model of EBV-associated LCs. Some pre-existing genetic alterations induced by pro-tumor signals (e.g., chronic inflammation) in precursor dysplastic lesions are important to support EBV infection and maintain Type II latency in the lung epithelia. In the milieu of EBV-infected lesions, expression of cellular genes such as immune checkpoint molecules and viral genes including EBNA1, LMP1, LMP2, BNLF2a, and EBV ncRNAs can activate a number of cancer hallmarks (highlighted in colored box), which leads to lung carcinogenesis. Viral genes are depicted in blue and host genes in red. Activation of cancer hallmarks is predicted based on experimental data obtained from other well-established EBV-associated cancers.
Figure 1. Proposed disease model of EBV-associated LCs. Some pre-existing genetic alterations induced by pro-tumor signals (e.g., chronic inflammation) in precursor dysplastic lesions are important to support EBV infection and maintain Type II latency in the lung epithelia. In the milieu of EBV-infected lesions, expression of cellular genes such as immune checkpoint molecules and viral genes including EBNA1, LMP1, LMP2, BNLF2a, and EBV ncRNAs can activate a number of cancer hallmarks (highlighted in colored box), which leads to lung carcinogenesis. Viral genes are depicted in blue and host genes in red. Activation of cancer hallmarks is predicted based on experimental data obtained from other well-established EBV-associated cancers.
Viruses 13 00877 g001
Table 1. EBV gene expression in different types of latency.
Table 1. EBV gene expression in different types of latency.
Latency TypesEBV GenesExamples of EBV Associated Cancers
0EBER1, EBER2, RPMS1, viral miRNAsMemory B cells in EBV(+) individuals
IEBER1, EBER2, RPMS1, viral miRNAs, and EBNA1Burkitt’s lymphoma
IIEBER1, EBER2, RPMS1, viral miRNAs, EBNA1, LMP1, LMP2A, and LMP2BNasopharyngeal carcinoma Lung cancer
IIIEBER1, EBER2, RPMS1, viral miRNAs, EBNA1, EBNA2, EBNA3A, EBNA3B, EBNA3C, EBNA-LP, LMP1, LMP2A, and LMP2BAIDS-associated lymphoma
Table 2. Association between EBV infection and lung cancers.
Table 2. Association between EBV infection and lung cancers.
Tumor TypesNumber of Total CasesNumber of EBV(+) CasesEBV Incidence Rates (%)Detection MethodsGeographical SitesReferences
LELC11100.0SerologyAmerica[36]
LELC4375.0ISH, SerologyAmerica[49]
LELC11100.0ISH, Serology, PCRAmerica[50]
LELC100.0ISHAmerica[51]
LELC55100.0ISH, Southern BlotAsia[52]
NSCLC/SCLC8056.3ISH, IHC, PCRAsia[44]
NSCLC16795.4ISH, Southern Blot, IHCAsia[53]
LELC11100.0ISH, Serology, PCREurope[37]
LELC1111100.0ISHAsia[39]
LELC22100.0ISH, PCR, IHCAsia[38]
LELC200.0ISHEurope[54]
NSCLC13000.0ISHAmerica[55]
LELC100.0IHCAmerica[56]
LELC11100.0ISHAmerica[57]
NSCLC127118.7ISH, IHCAsia[42]
NSCLC55100.0ISHAsia[58]
LELC11100.0ISH, PCRAsia[59]
NSCLC513058.8ISH, IHCAsia[40]
LELC600.0ISHAmerica[60]
LUAD3133.3ISH, PCR, IHCEurope[45]
LELC1111100.0ISH, Serology, PCRAsia[61]
LELC2323100.0SerologyAsia[62]
SCLC2314.3ISH, IHCAmerica[48]
LELC11100.0ISHAsia[70]
NSCLC/SCLC12200.0ISH, PCR, IHCEurope[63]
LELC100.0ISHAsia[76]
NSCLC1083633.3ISHAsia[43]
NSCLC191263.2ISH, PCR, IHCEurope[41]
LUAD11000.0ISHAsia[72]
NSCLC48714.6PCR, MicroarrayAmerica[71]
LELC11100.0ISH, PCR, SerologyAsia[74]
LELC11100.0ISHAsia[75]
LELC11100.0ISHAsia[69]
NSCLC/SCLC48510.4PCR, IHCAsia[47]
NSCLC6646.1ISH, NGSAsia[46]
NSCLC112770.6NGS, ISHAmerica, Europe, Asia[12]
NSCLC1763318.8ISH, PCRAsia[65]
LELC150150100.0ISHAsia[68]
LELC5757100.0ISHAsia[64]
LELC11100.0ISH, PCRAsia, Europe[67]
LELC88100.0NGSAsia[66]
LELC11100.0ISHAsia[73]
LELC: pulmonary lymphoepithelioma-like carcinoma; NSCLC: non-small cell lung cancer; SCLC: small cell lung cancer; in situ hybridization (ISH); polymerase chain reaction (PCR); immunohistochemistry (IHC); next-generation sequencing (NGS).
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Becnel, D.; Abdelghani, R.; Nanbo, A.; Avilala, J.; Kahn, J.; Li, L.; Lin, Z. Pathogenic Role of Epstein–Barr Virus in Lung Cancers. Viruses 2021, 13, 877. https://doi.org/10.3390/v13050877

AMA Style

Becnel D, Abdelghani R, Nanbo A, Avilala J, Kahn J, Li L, Lin Z. Pathogenic Role of Epstein–Barr Virus in Lung Cancers. Viruses. 2021; 13(5):877. https://doi.org/10.3390/v13050877

Chicago/Turabian Style

Becnel, David, Ramsy Abdelghani, Asuka Nanbo, Janardhan Avilala, Jacob Kahn, Li Li, and Zhen Lin. 2021. "Pathogenic Role of Epstein–Barr Virus in Lung Cancers" Viruses 13, no. 5: 877. https://doi.org/10.3390/v13050877

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop